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:

Rapid unleashing of macrophage efferocytic capacity via transcriptional pause release

Abstract

During development, inflammation or tissue injury, macrophages may successively engulf and process multiple apoptotic corpses via efferocytosis to achieve tissue homeostasis1. How macrophages may rapidly adapt their transcription to achieve continuous corpse uptake is incompletely understood. Transcriptional pause/release is an evolutionarily conserved mechanism, in which RNA polymerase (Pol) II initiates transcription for 20–60 nucleotides, is paused for minutes to hours and is then released to make full-length mRNA2. Here we show that macrophages, within minutes of corpse encounter, use transcriptional pause/release to unleash a rapid transcriptional response. For human and mouse macrophages, the Pol II pause/release was required for continuous efferocytosis in vitro and in vivo. Interestingly, blocking Pol II pause/release did not impede Fc receptor-mediated phagocytosis, yeast uptake or bacterial phagocytosis. Integration of data from three genomic approaches—precision nuclear run-on sequencing, RNA sequencing, and assay for transposase-accessible chromatin using sequencing (ATAC-seq)—on efferocytic macrophages at different time points revealed that Pol II pause/release controls expression of select transcription factors and downstream target genes. Mechanistic studies on transcription factor EGR3, prominently regulated by pause/release, uncovered EGR3-related reprogramming of other macrophage genes involved in cytoskeleton and corpse processing. Using lysosomal probes and a new genetic fluorescent reporter, we identify a role for pause/release in phagosome acidification during efferocytosis. Furthermore, microglia from egr3-deficient zebrafish embryos displayed reduced phagocytosis of apoptotic neurons and fewer maturing phagosomes, supporting defective corpse processing. Collectively, these data indicate that macrophages use Pol II pause/release as a mechanism to rapidly alter their transcriptional programs for efficient processing of the ingested apoptotic corpses and for successive efferocytosis.

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: Transcriptional pause/release contributes to efferocytosis.
Fig. 2: Importance of Pol II pause/release in continuous efferocytosis and corpse acidification.
Fig. 3: Rapid transcriptional upregulation of specific genes during efferocytosis links to pause/release.
Fig. 4: Pause/release-regulated transcription factor EGR3 affects corpse processing in vitro and in vivo.

Similar content being viewed by others

Data availability

All raw and processed sequencing data generated in this study have been deposited as SuperSeries in the NCBI Gene Expression Omnibus (GEO; https://www.ncbi.nlm.nih.gov/geo/) under accession number GSE253578. This SuperSeries comprises the following SubSeries: GSE253574 (ATAC-seq), GSE253576 (PRO-seq) and GSE25377 (RNA-seq). Publicly available datasets were used in this study: MSigDB resource: https://www.gsea-msigdb.org/gsea/msigdb/, UCSC Genome Browser annotation track database: https://genome.ucsc.edu/cgi-bin/hgTables, Jaspar database: https://jaspar.elixir.no and ENSEMBL: https://useast.ensembl.org/index.html. Gating strategy for efferocytosis (Supplementary Fig. 1) and raw, uncropped images of western blots (Supplementary Figs. 2 and 3) are provided in the Supplementary Information. Source data are provided with this paper.

Code availability

Custom codes used in this stud: for ATAC-seq, https://github.com/JetBrains-Research/chipseq-smk-pipeline; for PRO-seq https://github.com/AdelmanLab/NIH_scripts, https://github.com/lh3/seqtk and https://github.com/AdelmanLab/GetGeneAnnotation_GGA.

References

  1. Morioka, S., Maueröder, C. & Ravichandran, K. S. Living on the edge: efferocytosis at the interface of homeostasis and pathology. Immunity 50, 1149–1162 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Muse, G. W. et al. RNA polymerase is poised for activation across the genome. Nat. Genet. 39, 1507–1511 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Chawla, A. et al. A PPARy-LXR-ABCA1 pathway in macrophages is involved in cholesterol efflux and atherogenesis. Mol. Cell 7, 161–171 (2001).

    Article  CAS  PubMed  Google Scholar 

  4. Czimmerer, Z. & Nagy, L. Epigenomic regulation of macrophage polarization: where do the nuclear receptors belong? Immunol. Rev. 317, 152–165 (2023).

    Article  CAS  PubMed  Google Scholar 

  5. Boada-Romero, E., Martinez, J., Heckmann, B. L. & Green, D. R. The clearance of dead cells by efferocytosis. Nat. Rev. Mol. Cell Biol. 21, 398–414 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Morioka, S. et al. Chimeric efferocytic receptors improve apoptotic cell clearance and alleviate inflammation. Cell 185, 4887–4903.e4817 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Yurdagul, A. et al. Macrophage metabolism of apoptotic cell-derived arginine promotes continual efferocytosis and resolution of injury. Cell Metab. 31, 518–533.e510 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Core, L. J., Waterfall, J. J. & Lis, J. T. Nascent RNA sequencing reveals widespread pausing and divergent initiation at human promoters. Science 322, 1845–1848 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Gressel, S., Schwalb, B. & Cramer, P. The pause-initiation limit restricts transcription activation in human cells. Nat. Commun. 10, 3603 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Shao, W. & Zeitlinger, J. Paused RNA polymerase II inhibits new transcriptional initiation. Nat. Genet. 49, 1045–1051 (2017).

    Article  CAS  PubMed  Google Scholar 

  11. Abuhashem, A., Garg, V. & Hadjantonakis, A.-K. RNA polymerase II pausing in development: orchestrating transcription. Open Biol. 12, 210220 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Yu, L. et al. Negative elongation factor complex enables macrophage inflammatory responses by controlling anti-inflammatory gene expression. Nat. Commun. 11, 2286 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Yamaguchi, Y., Inukai, N., Narita, T., Wada, T. & Handa, H. Evidence that negative elongation factor represses transcription elongation through binding to a DRB sensitivity-inducing factor/RNA polymerase II complex and RNA. Mol. Cell. Biol. 22, 2918–2927 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Wei, P., Garber, M. E., Fang, S. M., Fischer, W. H. & Jones, K. A. A novel CDK9-associated C-type cyclin interacts directly with HIV-1 Tat and mediates its high-affinity, loop-specific binding to TAR RNA. Cell 92, 451–462 (1998).

    Article  CAS  PubMed  Google Scholar 

  15. Fujinaga, K. et al. Dynamics of human immunodeficiency virus transcription: P-TEFb phosphorylates RD and dissociates negative effectors from the transactivation response element. Mol. Cell. Biol. 24, 787–795 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kim, J. B. & Sharp, P. A. Positive transcription elongation factor B phosphorylates hSPT5 and RNA polymerase II carboxyl-terminal domain independently of cyclin-dependent kinase-activating kinase. J. Biol. Chem. 276, 12317–12323 (2001).

    Article  CAS  PubMed  Google Scholar 

  17. Henriques, T. et al. Stable pausing by RNA polymerase II provides an opportunity to target and integrate regulatory signals. Mol. Cell 52, 517–528 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. Buckley, M. S., Kwak, H., Zipfel, W. R. & Lis, J. T. Kinetics of promoter Pol II on Hsp70 reveal stable pausing and key insights into its regulation. Genes Dev. 28, 14–19 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lavin, Y. et al. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 159, 1312–1326 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Boettiger, A. N. & Levine, M. Synchronous and stochastic patterns of gene activation in the Drosophila embryo. Science 325, 471–473 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Saha, R. N. et al. Rapid activity-induced transcription of Arc and other IEGs relies on poised RNA polymerase II. Nat. Neurosci. 14, 848–856 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Rougvie, A. E. & Lis, J. T. The RNA polymerase II molecule at the 5’ end of the uninduced hsp70 gene of D. melanogaster is transcriptionally engaged. Cell 54, 795–804 (1988).

    Article  CAS  PubMed  Google Scholar 

  23. Mahat, D. B., Salamanca, H. H., Duarte, F. M., Danko, C. G. & Lis, J. T. Mammalian heat shock response and mechanisms underlying its genome-wide transcriptional regulation. Mol. Cell 62, 63–78 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Kwak, H., Fuda, N. J., Core, L. J. & Lis, J. T. Precise maps of RNA polymerase reveal how promoters direct initiation and pausing. Science 339, 950–953 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Lücking, U. et al. Identification of atuveciclib (BAY 1143572), the first highly selective, clinical PTEFb/CDK9 inhibitor for the treatment of cancer. ChemMedChem 12, 1776–1793 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Jonkers, I., Kwak, H. & Lis, J. T. Genome-wide dynamics of Pol II elongation and its interplay with promoter proximal pausing, chromatin, and exons. eLife 3, e02407 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Liang, K. et al. Targeting processive transcription elongation via SEC disruption for MYC-induced cancer therapy. Cell 175, 766–779.e717 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. DeBerardine, M., Booth, G. T., Versluis, P. P. & Lis, J. T. The NELF pausing checkpoint mediates the functional divergence of Cdk9. Nat. Commun. 14, 2762 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Aoi, Y. et al. SPT5 stabilization of promoter-proximal RNA polymerase II. Mol. Cell 81, 4413–4424.e4415 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Robb, C. M. et al. Chemically induced degradation of CDK9 by a proteolysis targeting chimera (PROTAC). Chem. Commun. 53, 7577–7580 (2017).

    Article  CAS  Google Scholar 

  31. Vos, S. M., Farnung, L., Urlaub, H. & Cramer, P. Structure of paused transcription complex Pol II–DSIF–NELF. Nature 560, 601–606 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Alexandra, G. C. et al. Evolution of promoter-proximal pausing enabled a new layer of transcription control. Preprint at bioRxiv https://doi.org/10.1101/2023.02.19.529146 (2023).

  33. Gilchrist, D. A. et al. Pausing of RNA polymerase II disrupts DNA-Specified nucleosome organization to enable precise gene regulation. Cell 143, 540–551 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Raymond, M. H. et al. Live cell tracking of macrophage efferocytosis during Drosophila embryo development in vivo. Science 375, 1182–1187 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Wang, Y. et al. Mitochondrial fission promotes the continued clearance of apoptotic cells by macrophages. Cell 171, 331–345.e322 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Lara Ordóñez, A. J., Fasiczka, R., Naaldijk, Y. & Hilfiker, S. Rab GTPases in Parkinson’s disease: a primer. Essays Biochem. 65, 961–974 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Proto, J. D. et al. Regulatory T cells promote macrophage efferocytosis during inflammation resolution. Immunity 49, 666–677.e666 (2018).

    Article  CAS  PubMed  Google Scholar 

  38. Maschalidi, S. et al. Targeting SLC7A11 improves efferocytosis by dendritic cells and wound healing in diabetes. Nature 606, 776–784 (2022).

    Article  CAS  PubMed  Google Scholar 

  39. Gerlach, B. D. et al. Efferocytosis induces macrophage proliferation to help resolve tissue injury. Cell Metab. 33, 2445–2463.e2448 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Meriwether, D. et al. Macrophage COX2 mediates efferocytosis, resolution reprogramming, and intestinal epithelial repair. Cell Mol. Gastroenterol. Hepatol. 13, 1095–1120 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Park, D. et al. BAI1 is an engulfment receptor for apoptotic cells upstream of the ELMO/Dock180/Rac module. Nature 450, 430–434 (2007).

    Article  CAS  PubMed  Google Scholar 

  42. Brahma, S. & Henikoff, S. The BAF chromatin remodeler synergizes with RNA polymerase II and transcription factors to evict nucleosomes. Nat. Genet. 56, 100–111 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Lambert, S. A. et al. The human transcription factors. Cell 172, 650–665 (2018).

    Article  CAS  PubMed  Google Scholar 

  44. Svaren, J. et al. NAB2, a corepressor of NGFI-A (Egr-1) and Krox20, is induced by proliferative and differentiative stimuli. Mol. Cell. Biol. 16, 3545–3553 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Swirnoff, A. H. et al. Nab1, a corepressor of NGFI-A (Egr-1), contains an active transcriptional repression domain. Mol. Cell. Biol. 18, 512–524 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Canchi, S. et al. Integrating gene and protein expression reveals perturbed functional networks in Alzheimer’s disease. Cell Rep. 28, 1103–1116.e1104 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Tourtellotte, W. & Milbrandt, J. Sensory ataxia and muscle spindle agenesis in mice lacking the transcription factor Egr3. Nat. Genet. 20, 87–91 (1998).

    Article  CAS  PubMed  Google Scholar 

  48. Casano, A. M., Albert, M. & Peri, F. Developmental apoptosis mediates entry and positioning of microglia in the zebrafish brain. Cell Rep. 16, 897–906 (2016).

    Article  CAS  PubMed  Google Scholar 

  49. Faronato, M. et al. DMXL2 drives epithelial to mesenchymal transition in hormonal therapy resistant breast cancer through Notch hyper-activation. Oncotarget 6, 22467–22479 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Wang, G. G. et al. Quantitative production of macrophages or neutrophils ex vivo using conditional Hoxb8. Nat. Methods 3, 287–293 (2006).

    Article  CAS  PubMed  Google Scholar 

  51. Corces, M. R. et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues. Nat. Methods 14, 959–962 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Köster, J. & Rahmann, S. Snakemake—a scalable bioinformatics workflow engine. Bioinformatics 28, 2520–2522 (2012).

    Article  PubMed  Google Scholar 

  53. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Ramírez, F., Dündar, F., Diehl, S., Grüning, B. A. & Manke, T. deepTools: a flexible platform for exploring deep-sequencing data. Nucleic Acids Res. 42, W187–W191 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Shpynov, O., Dievskii, A., Chernyatchik, R., Tsurinov, P. & Artyomov, M. N. Semi-supervised peak calling with SPAN and JBR genome browser. Bioinformatics 37, 4235–4237 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Zhang, Y. et al. Model-based Analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Ross-Innes, C. S. et al. Differential oestrogen receptor binding is associated with clinical outcome in breast cancer. Nature 481, 389–393 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Zhu, L. J. et al. ChIPpeakAnno: a Bioconductor package to annotate ChIP-seq and ChIP-chip data. BMC Bioinform. 11, 237 (2010).

    Article  Google Scholar 

  61. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  63. Danecek, P. et al. Twelve years of SAMtools and BCFtools. GigaScience 10, giab008 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

  65. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Julius, J. et al. A rapid, sensitive, scalable method for Precision Run-On sequencing (PRO-seq). Preprint at bioRxiv https://doi.org/10.1101/2020.05.18.102277 (2020).

  67. Reimer, K. A., Mimoso, C. A., Adelman, K. & Neugebauer, K. M. Co-transcriptional splicing regulates 3′ end cleavage during mammalian erythropoiesis. Mol. Cell 81, 998–1012.e1017 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Smith, T., Heger, A. & Sudbery, I. UMI-tools: modeling sequencing errors in Unique Molecular Identifiers to improve quantification accuracy. Genome Res. 27, 491–499 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Liberzon, A. et al. Molecular signatures database (MSigDB) 3.0. Bioinformatics 27, 1739–1740 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Nüsslein-Volhard, C. The zebrafish issue of Development. Development 139, 4099–4103 (2012).

    Article  PubMed  Google Scholar 

  72. Santos, D., Luzio, A. & Coimbra, A. M. Zebrafish sex differentiation and gonad development: a review on the impact of environmental factors. Aquat. Toxicol. 191, 141–163 (2017).

    Article  CAS  PubMed  Google Scholar 

  73. Kimmel, C. B., Ballard, W. W., Kimmel, S. R., Ullmann, B. & Schilling, T. F. Stages of embryonic development of the zebrafish. Dev. Dyn. 203, 253–310 (1995).

    Article  CAS  PubMed  Google Scholar 

  74. Peri, F. & Nüsslein-Volhard, C. Live imaging of neuronal degradation by microglia reveals a role for v0-ATPase a1 in phagosomal fusion in vivo. Cell 133, 916–927 (2008).

    Article  CAS  PubMed  Google Scholar 

  75. Gray, C. et al. Simultaneous intravital imaging of macrophage and neutrophil behaviour during inflammation using a novel transgenic zebrafish. Thromb. and Haemost. 105, 811–819 (2011).

    Article  CAS  Google Scholar 

  76. Villani, A. et al. Clearance by microglia depends on packaging of phagosomes into a unique cellular compartment. Dev. Cell 49, 77–88.e77 (2019).

    Article  CAS  PubMed  Google Scholar 

  77. Möller, K. et al. A role for the centrosome in regulating the rate of neuronal efferocytosis by microglia in vivo. eLife 11, e82094 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  78. Shiau, C. E., Kaufman, Z., Meireles, A. M. & Talbot, W. S. Differential requirement for irf8 in formation of embryonic and adult macrophages in zebrafish. PLoS ONE 10, e0117513 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  79. Wu, X., Shen, W., Zhang, B. & Meng, A. The genetic program of oocytes can be modified in vivo in the zebrafish ovary. J. Mol. Cell. Biol. 10, 479–493 (2018).

    Article  CAS  PubMed  Google Scholar 

  80. Kenyon, A. et al. Active nuclear transcriptome analysis reveals inflammasome-dependent mechanism for early neutrophil response to Mycobacterium marinum. Sci. Rep. 7, 6505 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of Ravichandran laboratory for input and critical reading of the manuscript; the Centre for Microscopy and Image Analysis, University of Zürich, for the Olympus IXplore SpinSR10 and the Leica SP8 inverse confocal laser scanning microscopes; and the Nascent Transcriptomics Core at Harvard Medical School for generating PRO-seq libraries and for assistance with data analysis. We thank the laboratory of Todd Fehniger for sourcing of human leukocytes from ImpactLife Blood Centers, and thank J. Milbrandt for providing Egr3-knockout mice. Schematics in Figs. 1a,d,e,g–j, 2a,d,g, 3a,c,f and 4a,d,h and Extended Data Figs. 3a,c,f, 4c, 5a and 6b,e,f were created using BioRender images as templates. T.T. was supported by a predoctoral fellowship from the American Heart Association (AHA 835593). K.S.R. was supported by NIAID (R01AI159551) and BJC Investigator Funds from the Washington University School of Medicine. S.K. was supported by an F31 fellowship from the NHLBI (1F31HL160134) and J.I.E. was supported by a fellowship from the NEI (EY031211). R.M.E. was supported by T32 Immunology Grant (T32AI007163). L.B. was supported by Emmanuel van der Schueren (EvdS) starter scholarship from Kom op tegen Kanker. E.B. and A.V. were supported by grants from the Swiss National Foundation grants (SNF310030_212794 and SNF31003A_182733).

Author information

Authors and Affiliations

Authors

Contributions

T.T. and K.S.R. designed all experiments and wrote the manuscript. T.T. performed most of the experiments. A.V., E.B. and F.P. assisted with the zebrafish experiments. S.R.G. assisted with the PRO-seq libraries. G.M.N. assisted with the PRO-seq analysis. O.S. and M.N.A. assisted with the ATAC-seq analysis. M.T. and P.Z. assisted with the RNA-seq analysis. G.C., S.K., R.M.E., J.S., Y.C., M.S., J.I.E. and L.B. assisted with various experiments. L.W.P. assisted with the generation of human PBMCs. M.R. assisted with the CharOFF construction.

Corresponding author

Correspondence to Kodi S. Ravichandran.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Edward Thorp and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Upregulated genes exhibit reduced pausing indices and pathway divergence during efferocytosis.

a, Gene Ontology pathway analysis (MSigDB) of upregulated and downregulated genes (determined by DESeq2, using the Wald test, padj<0.001) from the PRO-seq data at 45 min of efferocytosis (versus resting macrophages). b, Metagene plot of PRO-seq reads for the 331 genes upregulated in macrophages after 45 min efferocytosis (per Wald test) c, Heat map of PRO-seq signals around the proximal promoter region and gene body of the 331 genes upregulated in macrophages after 45 min efferocytosis. d, e, Cumulative distribution of pausing indices (from four independent experiments) of 331 upregulated genes (n-331) (d) and 105 randomly selected unchanged genes (e) as determined by DESeq2 and the Wald test (left). Pausing indices for each gene are compared between resting macrophages and macrophages after 45 min (top) or 90 min (bottom) of efferocytosis. Paired tow-tailed t-tests. ****p < 0.0001, ns: not significant. f, Upregulated genes (331) were divided into highly paused (PI > 4) and lowly paused genes (PI < 4), and pathway analysis performed via GSEA-MSigDB data base (https://www.gsea-msigdb.org/gsea/msigdb). In all figures, data are from four independent experimental replicates.

Source Data

Extended Data Fig. 2 Pause/release effects on efferocytosis versus Fc receptor-mediated phagocytosis.

a, Immunoblotting showing CDK9, Ser2P and β-Actin protein levels in macrophages treated with either DMSO or flavopiridol (10 µM) for 30 min. n = 3 independent experiments. b, Macrophages treated with DMSO or CDK9 inhibitors were incubated with TAMRA (pH-insensitive)-stained apoptotic Jurkat cells for 30 min. c, CDK9, Ser2P and β-Actin levels in macrophages treated with PROTAC-CDK9 (20 µM) for indicated times (left) or NVP-2 (concentrations indicated) for 30 min (right). n = 1 experiment. d, Efferocytosis quantified in macrophages with concurrent CDK9 inhibitors and apoptotic cells addition. n = 4 (left) and n = 3 (right) independent experiments. Paired two-tailed t-test. e, Uptake of anti-CD90.2-opsonized thymocytes by macrophages treated with NVP-2 (10 µM) or THAL-SNS-032 (20 µM) for 30 min. f, Editing efficiency in CRISPR/Cas9-generated NELFB- and NELFCD-deficient macrophages. n = 1 experiment. g, Efferocytosis kinetics were measured by live cell imaging (Incucyte) using WT and NELF-deficient macrophages incubated with CypHer5E-stained apoptotic Jurkat cells. Cytochalasin D was used as a control. Data are mean ± SEM. n = 4 independent experiments. Area under curve analysis and then unpaired two-tailed t-test with Welch’s correction. h, i, Continuous efferocytosis by human macrophages (left, n = 4 and right, n = 3 human donors of PBMCs; paired two-tailed t-test) (h) and mouse macrophages (i) treated with DMSO or CDK9 inhibitors. j, Second corpse uptake by mouse macrophages treated with CDK9 inhibitors and fed CypHer5E- and TAMRA-stained apoptotic Jurkat cells at a ratio of 1:1:1 apoptotic cell to phagocyte for 30 min. Double-positive macrophages have engulfed at least two corpses. n = 7 (left) and n = 6 (right) independent experiments. Paired two-tailed t-test. k, l, Continuous uptake of opsonized thymocytes in macrophages treated with DMSO or PROTAC-CDK9. d, g, h, j, Some of control samples were concurrently used in parallel experiments. b, e, i, l, In box and whiskers graphs (the center line denotes median, box edges encompass 25th to 75th percentiles, min to max points), dots show values from four independent experiments, unpaired two-tailed t-test. In all figures, *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.001, ns: not significant. Schematics in b,f,ik created with BioRender.com.

Source Data

Extended Data Fig. 3 Acidification and specific gene expression patterns during efferocytosis.

a, Continuous efferocytosis of CSFE+ (first round) and TAMRA+ (second round) apoptotic cells by macrophages pre-treated for 30 min with DMSO or bafilomycin A1. n = 4 independent experiments, paired two-tailed t-test. b, Relative expression (as determined by qPCR) of the indicated genes over time in macrophages during phagocytosis of synthetic beads. Data are mean ± SEM. c, PtdSer exposure was induced in RBCs using a calcium ionophore (A23187) and quantified by annexin V staining (left). The uptake of CypHer5E-stained PtdSer+ RBCs by mouse macrophages was assessed by flow cytometry (right) n = 4 independent experiments. Unpaired two-tailed t-test. d, Relative expression of EGR3 (as determined by qPCR) in human macrophages treated with DMSO or flavopiridol during efferocytosis. n:5 human donors of PBMCs; one-way ANOVA with Tukey’s multiple comparisons. a, c, d, In box and whiskers graphs, the center line denotes median, box edges encompass 25th to 75th percentiles, min to max points, and dots show independent data points. In all figures, *p < 0.05, **p < 0.01, ****p < 0.0001, ns: not significant. Schematics in a,c,d created with BioRender.com.

Source Data

Extended Data Fig. 4 EGR3 upregulation via Pol II pause/release during efferocytosis.

a, Heat map (derived from RNA-seq analysis) depicting differentially upregulated transcription factors (Wald test and corrected for multiple testing using the Benjamini and Hochberg method) at 45 min efferocytosis. Genes were ranked by fold-change. Data are from four independent experimental replicates. b, PRO-seq tracks (left) and relative pausing index (right) for Egr3 in resting macrophages and during efferocytosis. n = 4 independent experimental replicates. c, Schematic depicting ATAC-seq experiment in which mouse macrophages were fed apoptotic human Jurkat cells for 15 min, 30 min or 45 min of efferocytosis. n = 4 independent experimental replicates. d, Tracks of ATAC-seq showing chromatin accessibility in the promoter region of Egr3 in the resting state and during efferocytosis. Data derived from four independent experimental replicates. e, Relative expression of Egr3 in macrophages (as determined by qPCR) following Egr3 knockdown via shRNA (left) and Egr3 overexpression via retroviral vector (right). Data are presented as mean ± SEM. n = 3 independent experiments. f, Uptake of CypHer5E-stained apoptotic cells by wild type and Egr3-OE macrophages treated with either DMSO or flavopiridol. Two-way ANOVA with Tukey’s multiple comparisons. *p < 0.05 g, Fc receptor-mediated phagocytosis of CSFE-stained opsonized thymocytes by wild type, Egr3-deficient and Egr3-OE macrophages was assessed by flow cytometry after 30 min. Thymocytes opsonized with IgG was used as a control. One-way ANOVA with Tukey’s multiple comparisons. ns: non-significant. (f, g) Box and whiskers graphs (the center line denotes median, box edges encompass 25th to 75th percentiles, min to max points), and dots show values from four independent experiments. Schematics in c created with BioRender.com.

Source Data

Extended Data Fig. 5 egr3-deficient microglia in Zebrafish development exhibits impaired efferocytic capacity.

a, Schematic depicting the genome editing approach in zebrafish embryos (see method section for details). b, Percentage of injected embryos with confirmed deletion in exon 2 of egr3 across six independent experiments. CRISPANTs embryos (296 bp deletion) = 84.49%. Data are presented as mean ± s.d. N = 6 and 109 embryos analyzed. c, Immunoblotting (left) and quantification (right) of egr3 protein in control versus egr3 CRISPANTs embryos. n = 1 experiment. d, Representative dorsal images of non-injected irf8st95/st95 zebrafish embryo brain after Acridine Orange (AO) staining for apoptotic cells (top) and its segmentation (bottom). Quantification (right) of the basal levels of apoptosis in the optic tecta (OT) of non-injected irf8st95/st95 controls (N = 26) and egr3 CRISPANT (N = 23) embryos using Imaris automatic spot detection. Box and whiskers graphs (the center line denotes median, box edges encompass 25th to 75th percentiles, min to max points), unpaired two-tailed t-test. e, Representative images of a WT microglia at 3 days post fertilization (dpf) having the cytoplasm labelled in green (TgBAC(csf1ra:GAL4-VP16); Tg(UAS:nfsb-mCherry)) and freshly formed phagosomes labeled in magenta (Tg(UAS:mNeonGreen-Rab5)). The upper panel timepoint 1 (T1), in which the phagosome (arrowhead) is visible from the cell body (mCherry-green) but not yet in the Rab5 channel (mNeonGreen-magenta). T2 (4.5 min later) shows Rab5 recruitment to freshly formed phagosomes (arrowhead), indicating successful phagocytosis. Scale bar 10 µm. f, Quantification of phagocytic events (Rab5+ vesicles formed) in each microglia per hour counted. Violin plot, unpaired two-tailed t-test. g, Percentage of microglia having one or no successful phagocytic event (Rab5+ vesicles) per hour over the total. WT controls= 19.3%; egr3 CRISPANTs= 56.4%. e-g, T2 in WT control (N = 7 and n = 88) and egr3 CRISPANT (N = 7 and n = 101) where N=number of embryos and n=number of microglia cells quantified. h, Heat map of differential expression of EGR3-regulated genes that are associated with endosomal pathways. The Wald test corrected for multiple testing using the Benjamini and Hochberg method. padj<0.05. i, j, Knockdown efficiency of siRNAs targeting Dmxl2 and Elmo1. Unpaired two-tailed t-test. Three independent experiments, data are presented as mean ± SEM. k, Defective acidification in Dmxl2-deficient macrophages as shown by lysotracker staining during efferocytosis. n:3 independent experiments, paired two-tailed t-test. In all figures, *p < 0.05, **p < 0.01, ****p < 0.0001, ns: non-significant. Schematics in a created with BioRender.com.

Source Data

Extended Data Fig. 6 The pre-corpse internalization stages of efferocytosis impact Pol II release.

a, b, Defective acidification in Egr3-deficient macrophages as shown by lysotracker staining during efferocytosis (a) and GFP quenching in macrophages engulfing CharOFF cells (b). a, Box and whiskers graphs (the center line denotes median, box edges encompass 25th to 75th percentiles, min to max points). n = 4, paired two-tailed t-test. b, n = 3; ratio paired two-tailed t-test. c, Relative expression level of Egr1 and Egr3 (as determined by qPCR) in DMSO (n = 4) and flavopiridol (n = 3)-treated macrophages incubated with medium alone, supernatant from live Jurkat cells, and supernatant from apoptotic Jurkat cells for 30 min. Data are presented as mean ± SEM. Unpaired two-tailed t-test. d, e, Relative expression of Egr1 (n = 7) and Egr3 (n = 5) in CytoD (actin polymerization inhibitor)-treated macrophages with and without apoptotic cells (d) and DMSO or BMS (pan-TAM inhibitor) during efferocytosis (e). Paired two-tailed t-test. f, Relative expression of Egr3 in macrophages treated with either IgG (antibody control) or Axl and MerTK (20 nM) activating antibodies for 30 min. n:3, unpaired two-tailed t-test. In all figures, data points represent individual values for ‘n’ biologically independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001. Schematics in b,e,f created with BioRender.com.

Source Data

Supplementary information

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tufan, T., Comertpay, G., Villani, A. et al. Rapid unleashing of macrophage efferocytic capacity via transcriptional pause release. Nature 628, 408–415 (2024). https://doi.org/10.1038/s41586-024-07172-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-024-07172-y

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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