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:

TET deficiency perturbs mature B cell homeostasis and promotes oncogenesis associated with accumulation of G-quadruplex and R-loop structures

Subjects

Abstract

Enzymes of the TET family are methylcytosine dioxygenases that undergo frequent mutational or functional inactivation in human cancers. Recurrent loss-of-function mutations in TET proteins are frequent in human diffuse large B cell lymphoma (DLBCL). Here, we investigate the role of TET proteins in B cell homeostasis and development of B cell lymphomas with features of DLBCL. We show that deletion of Tet2 and Tet3 genes in mature B cells in mice perturbs B cell homeostasis and results in spontaneous development of germinal center (GC)-derived B cell lymphomas with increased G-quadruplexes and R-loops. At a genome-wide level, G-quadruplexes and R-loops were associated with increased DNA double-strand breaks (DSBs) at immunoglobulin switch regions. Deletion of the DNA methyltransferase DNMT1 in TET-deficient B cells prevented expansion of GC B cells, diminished the accumulation of G-quadruplexes and R-loops and delayed B lymphoma development, consistent with the opposing functions of DNMT and TET enzymes in DNA methylation and demethylation. Clustered regularly interspaced short palindromic repeats (CRISPR)-mediated depletion of nucleases and helicases that regulate G-quadruplexes and R-loops decreased the viability of TET-deficient B cells. Our studies suggest a molecular mechanism by which TET loss of function might predispose to the development of B cell malignancies.

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: TET deficiency in mature B cells causes B cell lymphoma.
Fig. 2: TET deficiency is associated with increased levels of G-quadruplexes and R-loops.
Fig. 3: Acute TET deletion is associated with increased levels of G-quadruplexes and R-loops.
Fig. 4: TET-deficient B cells show a genome-wide increase in G-quadruplexes and R-loops and increased translocations to immunoglobulin switch regions.
Fig. 5: Dnmt1 deletion delays oncogenesis in TET-deficient mice.

Similar content being viewed by others

Data availability

All genome-wide sequencing datasets have been deposited to the Gene Expression Omnibus (GEO) repository, accession number GSE161463. Any data and reagents will also be made available upon request. Source data are provided with this paper.

Code availability

The code used to process the next-generation sequencing datasets has been deposited in the GitHub repository at https://github.com/dsamanie7/Tet2-Tet3_DKO_CD19_cre.

References

  1. Tahiliani, M. et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 324, 930–935 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Ko, M. et al. Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2. Nature 468, 839–843 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Ito, S. et al. Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine. Science 333, 1300–1303 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Lio, C. W. et al. Tet2 and Tet3 cooperate with B-lineage transcription factors to regulate DNA modification and chromatin accessibility. eLife 5, e18290 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Orlanski, S. et al. Tissue-specific DNA demethylation is required for proper B-cell differentiation and function. Proc. Natl Acad. Sci. USA 113, 5018–5023 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Dominguez, P. M. et al. TET2 deficiency causes germinal center hyperplasia, impairs plasma cell differentiation, and promotes B-cell lymphomagenesis. Cancer Discov. 8, 1632–1653 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Lio, C. J. et al. TET enzymes augment activation-induced deaminase (AID) expression via 5-hydroxymethylcytosine modifications at the Aicda superenhancer. Sci Immunol 4, eaau7523 (2019)

  8. Rosikiewicz, W. et al. TET2 deficiency reprograms the germinal center B cell epigenome and silences genes linked to lymphomagenesis. Sci. Adv. 6, eaay5872 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lio, C. J. et al. TET methylcytosine oxidases: new insights from a decade of research. J. Biosci. 45, 21 (2020).

  10. Wu, H. & Zhang, Y. Reversing DNA methylation: mechanisms, genomics, and biological functions. Cell 156, 45–68 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Pastor, W. A., Aravind, L. & Rao, A. TETonic shift: biological roles of TET proteins in DNA demethylation and transcription. Nat. Rev. Mol. Cell Biol. 14, 341–356 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Rasmussen, K. D. & Helin, K. Role of TET enzymes in DNA methylation, development, and cancer. Genes Dev. 30, 733–750 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Cimmino, L., Abdel-Wahab, O., Levine, R. L. & Aifantis, I. TET family proteins and their role in stem cell differentiation and transformation. Cell Stem Cell 9, 193–204 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Huang, Y. & Rao, A. Connections between TET proteins and aberrant DNA modification in cancer. Trends Genet. 30, 464–474 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ko, M., An, J. & Rao, A. DNA methylation and hydroxymethylation in hematologic differentiation and transformation. Curr. Opin. Cell Biol. 37, 91–101 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lio, C. J., Yuita, H. & Rao, A. Dysregulation of the TET family of epigenetic regulators in lymphoid and myeloid malignancies. Blood 134, 1487–1497 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Reddy, A. et al. Genetic and functional drivers of diffuse large B cell lymphoma. Cell 171, 481–494 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Schmitz, R. et al. Genetics and pathogenesis of diffuse large B-cell lymphoma. N. Engl. J. Med. 378, 1396–1407 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Chapuy, B. et al. Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes. Nat. Med. 24, 679–690 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kafer, G. R. et al. 5-Hydroxymethylcytosine marks sites of DNA damage and promotes genome stability. Cell Rep. 14, 1283–1292 (2016).

    Article  CAS  PubMed  Google Scholar 

  21. Kharat, S. S. et al. Degradation of 5hmC-marked stalled replication forks by APE1 causes genomic instability. Sci. Signal. 13, eaba8091 (2020)

  22. Crossley, M. P., Bocek, M. & Cimprich, K. A. R-Loops as cellular regulators and genomic threats. Mol. Cell 73, 398–411 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Skourti-Stathaki, K. & Proudfoot, N. J. A double-edged sword: R loops as threats to genome integrity and powerful regulators of gene expression. Genes Dev. 28, 1384–1396 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hansel-Hertsch, R., Di Antonio, M. & Balasubramanian, S. DNA G-quadruplexes in the human genome: detection, functions and therapeutic potential. Nat. Rev. Mol. Cell Biol. 18, 279–284 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Rhodes, D. & Lipps, H. J. G-quadruplexes and their regulatory roles in biology. Nucleic Acids Res. 43, 8627–8637 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Sauer, M. & Paeschke, K. G-quadruplex unwinding helicases and their function in vivo. Biochem. Soc. Trans. 45, 1173–1182 (2017).

    Article  CAS  PubMed  Google Scholar 

  27. De Magis, A. et al. DNA damage and genome instability by G-quadruplex ligands are mediated by R loops in human cancer cells. Proc. Natl Acad. Sci. USA 116, 816–825 (2019).

    Article  PubMed  CAS  Google Scholar 

  28. Gray, L. T., Vallur, A. C., Eddy, J. & Maizels, N. G quadruplexes are genomewide targets of transcriptional helicases XPB and XPD. Nat. Chem. Biol. 10, 313–318 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Miglietta, G., Russo, M. & Capranico, G. G-quadruplex-R-loop interactions and the mechanism of anticancer G-quadruplex binders. Nucleic Acids Res. 48, 11942–11957 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Chedin, F. Nascent connections: R-loops and chromatin patterning. Trends Genet. 32, 828–838 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Niehrs, C. & Luke, B. Regulatory R-loops as facilitators of gene expression and genome stability. Nat. Rev. Mol. Cell Biol. 21, 167–178 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Sanz, L. A. et al. Prevalent, dynamic, and conserved R-loop structures associate with specific epigenomic signatures in mammals. Mol. Cell 63, 167–178 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. An, J. et al. Acute loss of TET function results in aggressive myeloid cancer in mice. Nat. Commun. 6, 10071 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. Cimmino, L. et al. TET1 is a tumor suppressor of hematopoietic malignancy. Nat. Immunol. 16, 653–662 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Tsagaratou, A. et al. TET proteins regulate the lineage specification and TCR-mediated expansion of iNKT cells. Nat. Immunol. 18, 45–53 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Mao, S. Q. et al. DNA G-quadruplex structures mold the DNA methylome. Nat. Struct. Mol. Biol. 25, 951–957 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Biffi, G., Tannahill, D., McCafferty, J. & Balasubramanian, S. Quantitative visualization of DNA G-quadruplex structures in human cells. Nat. Chem. 5, 182–186 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Muller, S. et al. Pyridostatin analogues promote telomere dysfunction and long-term growth inhibition in human cancer cells. Org. Biomol. Chem. 10, 6537–6546 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Sabharwal, N. C. et al. N-methylmesoporphyrin IX fluorescence as a reporter of strand orientation in guanine quadruplexes. FEBS J. 281, 1726–1737 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Nicoludis, J. M. et al. Optimized end-stacking provides specificity of N-methyl mesoporphyrin IX for human telomeric G-quadruplex DNA. J. Am. Chem. Soc. 134, 20446–20456 (2012).

    Article  CAS  PubMed  Google Scholar 

  41. Chen, L. et al. R-ChIP using inactive RNase H reveals dynamic coupling of R-loops with transcriptional pausing at gene promoters. Mol. Cell 68, 745–757 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Yan, Q., Shields, E. J., Bonasio, R. & Sarma, K. Mapping native R-loops genome-wide using a targeted nuclease approach. Cell Rep. 29, 1369–1380 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Crossley, M. P. et al. Catalytically inactive, purified RNase H1: a specific and sensitive probe for RNA–DNA hybrid imaging. J. Cell Biol. 220, e202101092 (2021).

  44. Boguslawski, S. J. et al. Characterization of monoclonal antibody to DNA.RNA and its application to immunodetection of hybrids. J. Immunol. Methods 89, 123–130 (1986).

    Article  CAS  PubMed  Google Scholar 

  45. Nojima, T. et al. In-vitro derived germinal centre B cells differentially generate memory B or plasma cells in vivo. Nat. Commun. 2, 465 (2011).

    Article  PubMed  CAS  Google Scholar 

  46. Lopez-Moyado, I. F. et al. Paradoxical association of TET loss of function with genome-wide DNA hypomethylation. Proc. Natl Acad. Sci. USA 116, 16933–16942 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Hansel-Hertsch, R. et al. G-quadruplex structures mark human regulatory chromatin. Nat. Genet. 48, 1267–1272 (2016).

    Article  CAS  PubMed  Google Scholar 

  48. Song, C. X. et al. Selective chemical labeling reveals the genome-wide distribution of 5-hydroxymethylcytosine. Nat. Biotechnol. 29, 68–72 (2011).

    Article  CAS  PubMed  Google Scholar 

  49. Hu, J. et al. Detecting DNA double-stranded breaks in mammalian genomes by linear amplification-mediated high-throughput genome-wide translocation sequencing. Nat. Protoc. 11, 853–871 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Shaknovich, R. et al. DNA methyltransferase 1 and DNA methylation patterning contribute to germinal center B-cell differentiation. Blood 118, 3559–3569 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Tanaka, S. et al. Tet2 and Tet3 in B cells are required to repress CD86 and prevent autoimmunity. Nat. Immunol. 21, 950–961 (2020).

    Article  CAS  PubMed  Google Scholar 

  52. Young, R. M. et al. Survival of human lymphoma cells requires B-cell receptor engagement by self-antigens. Proc. Natl Acad. Sci. USA 112, 13447–13454 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Nambiar, M. et al. Formation of a G-quadruplex at the BCL2 major breakpoint region of the t(14;18) translocation in follicular lymphoma. Nucleic Acids Res. 39, 936–948 (2011).

    Article  CAS  PubMed  Google Scholar 

  54. Rabkin, C. S., Hirt, C., Janz, S. & Dolken, G. t(14;18) translocations and risk of follicular lymphoma. J. Natl Cancer Inst. Monogr. 2008, 48–51, (2008)

  55. Qiao, Q. et al. AID recognizes structured DNA for class switch recombination. Mol. Cell 67, 361–373 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Xu, Y. Z. et al. Activation-induced cytidine deaminase localizes to G-quadruplex motifs at mutation hotspots in lymphoma. NAR Cancer 2, zcaa029 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Yewdell, W. T. et al. A hyper-IgM syndrome mutation in activation-induced cytidine deaminase disrupts G-quadruplex binding and genome-wide chromatin localization. Immunity 53, 952–970 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Zhang, W. & Xu, J. DNA methyltransferases and their roles in tumorigenesis. Biomark. Res. 5, 1 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Wang, Y. et al. G-quadruplex DNA drives genomic instability and represents a targetable molecular abnormality in ATRX-deficient malignant glioma. Nat. Commun. 10, 943 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Xu, H. et al. CX-5461 is a DNA G-quadruplex stabilizer with selective lethality in BRCA1/2 deficient tumours. Nat. Commun. 8, 14432 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Kang, J. et al. Simultaneous deletion of the methylcytosine oxidases Tet1 and Tet3 increases transcriptome variability in early embryogenesis. Proc. Natl Acad. Sci. USA 112, E4236–E4245 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Ko, M. et al. Ten-eleven-translocation 2 (TET2) negatively regulates homeostasis and differentiation of hematopoietic stem cells in mice. Proc. Natl Acad. Sci. USA 108, 14566–14571 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Chedin, F., Hartono, S. R., Sanz, L. A. & Vanoosthuyse, V. Best practices for the visualization, mapping, and manipulation of R-loops. EMBO J. 40, e106394 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Konig, F., Schubert, T. & Langst, G. The monoclonal S9.6 antibody exhibits highly variable binding affinities towards different R-loop sequences. PLoS ONE 12, e0178875 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Ramirez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  68. 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 

  69. Xi, Y. & Li, W. BSMAP: whole genome bisulfite sequence MAPping program. BMC Bioinformatics 10, 232 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  71. 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).

    PubMed  PubMed Central  Google Scholar 

  72. Bolotin, D. A. et al. Antigen receptor repertoire profiling from RNA-seq data. Nat. Biotechnol. 35, 908–911 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. 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 

  74. Krzywinski, M. et al. Circos: an information aesthetic for comparative genomics. Genome Res. 19, 1639–1645 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Kieffer-Kwon, K. R. et al. Myc regulates chromatin decompaction and nuclear architecture during B cell activation. Mol. Cell 67, 566–578 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank H. Yuita and I. Lopez-Moyado for generating the ERT2-cre Tet1fl/fl Tet2fl/fl Tet3fl/fl, ERT2-cre TKO mice, D. Kitamura at the Tokyo University of Science for sharing the 40LB cells, U. Basu and B. Laffleur at Columbia University for help with the HTGTS protocol, our collaborators at Cambridge Epigenetix (UK) for providing the 5hmC mapping kits, the LJI Flow Cytometry Core team (C. Kim, D. Hinz, C. Dillingham, M Haynes and S. Ellis) for help with cell sorting and the LJI next-generation sequencing core members (J. Day, S. Alarcon, H. Dose, K. Tanaguay and A. Hernandez) for help with sequencing. The BD FACSAria II is supported by the NIH (NIH S10OD016262, NIH S10RR027366), and our research used resources of the Advanced Light Source, which is a DOE Office of Science User Facility under contract number DE-AC02-05CH11231. The NovaSeq 6000 and the HiSeq 2500 were acquired through the Shared Instrumentation Grant (SIG) Program (S10) (NovaSeq 6000 S10OD025052 and HiSeq 2500 S10OD016262). K.S. acknowledges support from NIH grant DP2-NS105576. V.S. was supported by a Leukemia and Lymphoma Society Postdoctoral Fellowship (grant ID 5463-18) and currently by a K99/R00 award from the National Cancer Institute (grant ID CA248835). D.S.-C. and E.G.-A. were supported by University of California Institute for Mexico and the United States and El Consejo Nacional de Ciencia y Tecnología (UCMEXUS/CONACYT) pre-doctoral fellowship. This work is supported by the NIH grants R35 CA210043, R01 AI109842 and AI128589 to A.R. and K99/R00 CA248835, research funds from LLS grant 5463-18 and the Tullie and Rickey families SPARK award from LJI to V.S.

Author information

Authors and Affiliations

Authors

Contributions

V.S. and D.S.-C. acquired, analyzed and interpreted the data. Z.D. performed R-loop mapping experiments and helped with the interpretation of results. E.G.-A. and D.S.-C. performed the bioinformatics analysis. Q.Y. and K.S. provided the plasmids and suggestions for MapR. A.R. and V.S. supervised the studies, conceptualized the experiments and helped with data interpretation. V.S., D.S.-C. and A.R. wrote the manuscript. All authors were involved in reviewing and editing the manuscript.

Corresponding author

Correspondence to Anjana Rao.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Immunology thanks Jayanta Chaudhuri and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. L. A. Dempsey was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 TET deficiency leads to development of mature B cell lymphoma.

. a) H&E staining in spleens from 8 week-old Dfl and CD19 DKO mice The data is representative of 2 independent experiments. b) Kaplan-Meir curves displaying lymphoma-free survival of CD19 DKO (red) and control Dfl (blue) and CD19 Cre (green) mice. Y-axis, percent mice without lymphoma, defined by ≥ 3-fold increase in spleen weight and ≥ 2-fold increase in cellularity. c)f) Quantification of c) cell numbers, d) spleen weights, e) B cell numbers, f) percent of B cells in spleens of 9 week-old CD19 DKO (red) and control Dfl (blue) and, CD19 Cre (green) mice from at least 5 biological replicates and 4 independent experiments. g) Percent of B cells in spleens of CD19 DKO (red) and control Dfl (blue) and CD19 Cre (green) mice at different ages from 3 independent experiments. h) Quantification of cell numbers of CD4+ T, CD8+ T and CD4+ Tfh cells from spleens of 9 week-old CD19 DKO (red) and control Dfl (blue) mice from 3 biological replicates and 2 independent experiments. i) Percent of activated CD4+ and, CD8+ T cells, and CD4 Tfh cells from spleens of 9 week-old CD19 DKO (red) and control Dfl (blue) mice from 3 biological replicates and 2 independent experiments. j) and k) Flow cytometry plots of j) Activated CD62Llow (X-axis) and CD44+ (Y-axis) CD4+ and CD8+ T cells and k) CD4+ Tfh cells PD1+ (Y-axis) CXCR5+ (X-axis) from spleen of 9 week-old CD19 DKO (red) and control Dfl (blue) mice. Numbers represent frequency of gated populations. l) Scheme of adoptive transfer experiment. B cells from Dfl and CD19 DKO CD45.2 mice were transferred retro-orbitally into sub-lethally irradiated CD45.1 immunocompetent host mice. m) Kaplan-Meir curves displaying overall survival of CD45.1 host mice transplanted with B cells from Dfl or CD19 DKO mice. X-axis denotes time in weeks after transplantation. n) Enlarged spleens in CD45.1 host mice transplanted with CD19 DKO compared to Dfl B cells 8 weeks after adoptive transfer. o) Representative flow cytometry data from the spleens of CD45.1 host mice 8 weeks after transplantation with Dfl and CD19 DKO B cells. Frequencies of the CD45.1+ and CD45.2+ cell populations are shown. Statistical significance is calculated using the log-rank test for b) and m), one-way ANOVA for c)-f) and two-way ANOVA for g)-i). Error bars represent mean +/− standard deviation, ** p value ≤0.01 *** p value ≤0.001, **** p value ≤0.0001.

Extended Data Fig. 2 Expanded B cells from CD19 DKO mice have a germinal center (GC) origin.

. a), b) Gene set enrichment analysis (GSEA) plots showing enrichment for a GC B cell transcriptional signature in the transcriptional profile of CD19 DKO compared to Dfl B cells, using gene sets from a) GC versus follicular B cells and b) early GC versus late GC B cells. Y-axis denotes enrichment score. NES, Normalized enrichment score, FDR, False discovery rate. c) Representative flow cytometry data gated on splenic B cells from 8 week-old Dfl, CD19Cre (YFP+) and CD19 DKO (YFP+) mice. Numbers in the rectangles represent frequencies of GC B cells, identified as EFNB1+ (Y-axis) and IgDlow (X-axis). d) Representative flow cytometry data showing Ig isotype expression, gated on splenic B cells from 8 week-old Dfl, CD19Cre (YFP+) and CD19 DKO (YFP+) mice. Top, IgG1; middle, IgG2b; bottom, IgG3 X-axis shows expression of the default IgD isotype. Numbers represent frequencies of gated cell populations. e) Immunoblot showing AID expression in Dfl and CD19 DKO B cells (2 replicate experiments). Actin is used as a loading control. f) Relative fold-change in expression of μ and γ1 germline, Tet3, Irf4 and Myc transcripts measured by qRT-PCR in Dfl and CD19 DKO B cells from 2 biological replicates. g) Histogram (left panel) and bar-graph (right panel) showing staining with BCL6 antibody compared to isotype IgG controls in B cell from 8 week-old CD19Cre, Dfl and CD19 DKO mice from 3 independent experiments. h) and i) Bar plots displaying the proportions of (h) IgVH and (i) Igκ clonotypes (rearranged variable gene segments) from Dfl (blue) and CD19 DKO (red) B cells, identified from BCR repertoire analysis of RNA-Seq data. Y-axis represents the proportion of each clonotype. Each individual IgVH and Igκ clonotype is displayed using a different color in the bar plots. Numbers at the bottom represent the number of clonotypes identified in two independent replicates of Dfl (blue) and CD19 DKO (red) B cells. j) Representative flow cytometry data gated on Peyer’s patch B cells from 8 week-old Dfl and CD19 DKO mice. Numbers represent frequency of GC B cells, identified as FAS+ (Y-axis) and CD38 (X-axis). k) Quantification of GC B cell frequency in Peyer’s patches of Dfl (blue) and CD19 DKO (red) mice from 3 independent experiments. Statistical significance is calculated using two-tailed student t-test in f), k) and one-way ANOVA in g). Error bars represent mean +/− standard deviation in f), g) and k). ** p value ≤0.01.

Extended Data Fig. 3 TET deficiency is associated with increased levels of G-quadruplexes and R-loops.

. a) Flow cytometric detection of G-quadruplexes with BG4-Ig antibody or isotype IgG controls in primary B cells stimulated with 25 µg/ml LPS for 48 hours and treated with 10 µM pyridostatin (PDS, G4 ligand) for 24 hours. Numbers represent median fluorescence intensity. b) Quantification of median fluorescence intensity (MFI) of BG4-Ig signal from primary B cells treated with (red) or without (blue) PDS. Lines connect paired samples treated with or without PDS from 3 independent experiments. c) Fluorescence emission spectrum of NMM in the presence of a G4-forming oligonucleotide (oligo) from the human c-Kit gene promoter or a control oligo in which guanines in G4-forming regions (G-tracts) were mutated. d) Fluorescence enhancement over background (no oligos) for NMM at 610 nm in presence of known G4-forming oligos (Kit1, Kit2, Spb1) from the c-Kit gene locus or the telomeric repeat (Telo) or their respective mutated versions. e) G-quadruplex levels assessed by NMM or DMSO vehicle control (Veh) staining in untreated CH12 B cells or cells treated with 5 µM pyridostatin (PDS, G4 ligand) for 24 hours. Numbers represent median fluorescence intensity from 3 independent experiments. f) Flow cytometric detection of R-loops using V5-epitope-tagged recombinant RNASE H1 (rRNASE H1) in CH12 cells with or without RNASE H enzyme digestion during staining. Numbers represent median fluorescence intensity. g) Quantification of median fluorescence intensity (MFI) of R-loops (rRNASE H1) signal from CH12 cells with (red) or without (blue) RNASE H enzyme digestion. Lines connect paired samples with or without RNASE H digestion from 3 independent experiments. h) - m) Representative images (h, j, l) and quantification of mean fluorescence signal (i, k, m) of CD19cre and CD19 DKO YFP+ B cells stained with DAPI or propidium iodide and CD19, BG4-Ig (h, i), NMM (j, k) and rRNASE H1 (l, m) or respective controls using the AMNIS imagestream. Data are from two independent experiments. Statistical significance is calculated using paired student t-test in b) and g), two-tailed student t-test i), k) and m). Error bars represent mean +/− standard deviation, ** p value ≤0.01, **** p value ≤0.0001.

Extended Data Fig. 4 TET deficiency in multiple primary cell types is associated with increased DNA G-quadruplex structures.

. a) Representative flow cytometry data gated on splenic B cells from 8 week-old Cγ1Cre, Dfl and Cγ1 DKO mice 12 days after immunization with SRBCs. GC B cells are identified as FAS+ (Y-axis) and CD38 (X-axis). Numbers represent frequency of GC B cells. b), c) Quantification of (b) GC B cell frequencies and (c) absolute numbers of splenocytes from 8 week-old Cγ1Cre, Dfl and Cγ1 DKO mice 12 days after immunization with SRBCs from 3 independent experiments. d) Experimental design. ERT2Cre DKO or control Dfl mice were injected for 5 consecutive days with tamoxifen to induce Cre expression and TET deletion, then rested for 2 days. Splenic B cells were activated for 72 hours in vitro with LPS and IL-4 in the presence of 4-hydroxytamoxifen (4-OHT). e) G-quadruplex levels in naïve (left panel) and activated (right panel) B cells from tamoxifen-treated ERT2Cre DKO (YFP+) or control Dfl mice. Numbers represent median fluorescence intensity. f) Quantification of median fluorescence intensity (MFI) of NMM signal from naïve and activated B cells from ERT2Cre DKO (YFP+) or control Dfl mice from 3 independent experiments. g), i) G-quadruplex levels assessed by NMM or DMSO vehicle staining (Con) in (g) transferred CD45.2+ myeloid cells from ERT2Cre TKO (YFP+) or control Tfl mice, and (i) transferred CD45.2+ T cells from CD4Cre DKO (YFP+) or control Dfl mice. h), j) Quantification of median fluorescence intensity (MFI) of NMM signal in (h) transferred CD45.2+ myeloid cells from ERT2Cre TKO (YFP+) or control Tfl mice from 2 biological replicates and j) transferred CD45.2+ T cells from CD4Cre DKO (YFP + ) or control Dfl mice from 3 biological replicates. Statistical significance is calculated using one-way ANOVA in c), two-way f) and two-tailed student t-test in h) and j). Error bars represent mean +/− standard deviation, * p value ≤0.01, ** p value ≤0.005.

Extended Data Fig. 5 Increased apoptosis and DNA DSBs in TET-deficient B lymphoma cells depleted of enzymes that resolve G-quadruplexes and R-loops.

. a) Experimental design. Primary B cells from Dfl and CD19 DKO B cells were nucleofected with Cas9 RNPs loaded with sgRNAs against Rnase H1 or the known G4-binding helicases Atrx, Blm and Fancd2, then stimulated with 10 µg/ml LPS for 48 hours before assessing the frequency of apoptotic cells by flow cytometry for cleaved Caspase 3. b) Representative immunoblots showing decreased protein levels of ATRX, BLM, FANCD2 and RNASE H1 in CD19 DKO B cells nucleofected 48 hours earlier with the corresponding or CD4 Cas9 RNPs (Ctrl). The data is representative of at least 2 independent experiments. c) Representative flow cytometry plots quantifying percent apoptotic cells in Dfl and CD19 DKO B cells nucleofected with Cas9 RNPs. Y-axis, staining for cleaved Caspase 3; X-axis, forward scatter (FSC). d) Quantification of apoptosis, measured as percent of cells showing staining for cleaved Caspase 3, in cells nucleofected with Cas9 RNPs to Atrx, Blm, Fancd2, Rnase H from 3 biological replicates. e) Quantification of G-quadruplexes as NMM median fluorescence intensity (MFI) in Dfl and CD19 DKO B cells 48 hours after nucleofection with Cas9 RNPs. The signal is normalized to the NMM MFI of the same biological sample nucleofected with CD4 Cas9 RNPs (Ctrl) from 3 biological replicates. f) Quantification of DNA DSBs, assessed by γH2AX median fluorescence intensity (MFI) in Dfl and CD19 DKO B cells 48 hours after nucleofection with the indicated Cas9 RNPs from 3 biological replicates. The signal is normalized to the γH2AX MFI of the same biological sample nucleofected with control Cas9 RNP loaded with sgRNA against CD4 (Ctrl). g) Experimental design. Dfl and CD19 DKO B cells were treated for 2 days with the G-quadruplex stabilizing compound pyridostatin (PDS) prior to activation for 48 hours with LPS. h) Quantification of apoptosis, measured as percent of cells showing staining for cleaved caspase 3, in Dfl and CD19 DKO B cells cultured without (untreated) or with 10 µM PDS from 5 biological replicates and 3 independent experiments. i) Representative flow-cytometry plots quantifying percent apoptotic cells in Dfl and CD19 DKO B cells with or without PDS treatment. Y-axis, staining for cleaved caspase 3; X-axis, forward scatter (FSC). Statistical significance is calculated using two-way ANOVA. Error bars represent mean +/− standard error, * p value ≤0.05, ** p value ≤0.01, *** p value ≤0.0005 **** p value <0.0001.

Extended Data Fig. 6 TET deficiency is associated with genome-wide accumulation of G-quadruplexes and R-loops.

. a) Genome annotations of regions enriched for G-quadruplexes (G4) and R-loops (right bar) compared to their representation in the mouse genome (mm10) (left bar). b) Relative representation of different classes of motifs predicted to form G-quadruplexes (pG4) in control regions selected randomly from the genome (left bar) and regions enriched for G-quadruplexes and R-loops (right bar). c) Heat maps showing enrichment (RPM) for G-quadruplexes and R-loops in CD19 DKO and control B cells. The signal is plotted in a +/− 2 kb window from the center of the regions ordered based on decreasing intensity from top to bottom in the entire 4 kb window. R-loop signal is plotted after background subtraction of MNase-alone control. d) Profile histograms showing the signals for G-quadruplexes (G4) (RPM, reads per million), R-loops (RPM), WGBS (percent of 5mC + 5hmC/unmodified C) and 5hmC (RPM). The 9722 regions enriched for both G-quadruplexes and R-loops are divided into two categories – 6212 regions overlapping promoters (left panels) and 3510 regions not at promoters (right panels). Dashed grey lines indicate the center of the region and the 1 kb boundaries located on either side of the center. Blue and red lines show data from Dfl and CD19 DKO B cells, respectively. Asterisks represent statistical significance calculated by comparing the signals between Dfl and CD19 DKO B cells, either within the G-quadruplex and R-loop forming regions, the region to +/−1kb window or +/−1kb to 2 kb window for respective datasets. e) Profile histograms showing the 5hmC signal in Dfl (blue) and CD19 DKO (red) B cells in 23,467 regions identified as enriched for 5hmC signal. f) Violin plots quantifying enrichment (RPKM) of 5hmC signal in Dfl B cells in the +/−1 kb from G-quadruplex and R-loop forming regions at promoters, non-promoter regions and control regions randomly located in euchromatin (Hi-C A genomic compartment) from 2 biological replicates. g) Pie chart showing the differentially methylated regions (DMRs) in CD19 DKO compared to control Dfl B cells. Of a total of 6948 DMRs identified by WGBS, 1014 (15%) showed reduced DNA methylation (hypomethylation) and 5934 (85%) showed increased DNA methylation (hypermethylation). h) Box and whisker plots quantifying percent of 5mC + 5hmC/unmodified C (from WGBS) at and near the G4 and R-loop forming regions overlapping promoters and regions not overlapping promoters in Dfl (blue) and CD19 DKO (red) B cells from 2 biological replicates. The signal is plotted in three windows; window 1, within the G4 and R-loop regions; window 2, from region to +/− 1 kb on either side and; window 3, +/-1kb to +/−2kb on either side. i) Percent of 5mC + 5hmC/unmodified C (from WGBS) in random genomic regions of Dfl (blue) and CD19 DKO (red) B cells. j) Heatmaps of enrichment (RPM) for G-quadruplexes (left) and R-loops (right) in Dfl and CD19 DKO B cells, ordered in descending order of gene expression. k) MA plot (left) showing differentially expressed genes (DEGs) in CD19 DKO B cells. Red dots, upregulated DEGs; blue dots, downregulated DEGs; black dots, DEGs with G-quadruplexes and R-loops at their promoters (+/-1kb of TSS); yellow dots, non-DEGs with G-quadruplexes and R-loops at their promoters; grey dots, non-DEGs without G-quadruplexes and R-loops. The pie-chart (right) shows the percent of DEGs with (green) and without (brown) G-quadruplexes and R-loops at their promoters. Asterisks indicate statistical significance. Statistical significance is calculated using Kruskal-Wallis test and the ad hoc Dunn’s test in d), f) and h), Chi-square test in j). Boxes in box and whisker plots represent median (center) with 25th to 75 th percentile and whiskers represent maxima/minima. **** p value ≤0.0001 and ***** p value <0.000001.

Extended Data Fig. 7 Genome-wide analysis of TET deficient B cells.

. a)-c) Genome browser tracks showing the distribution of G-quadruplexes (G4), R-loops, RNA-Seq, WGBS, and 5hmC datasets for Dfl (blue tracks) and CD19 DKO (red tracks) B cells. Grey boxes indicate regions of interest. The blue arrows at the bottom show the location of the TSS and the direction of transcription. d) Circos plots to visually depict all translocations identified by HTGTS in Dfl and CD19 DKO replicates. Colored lines connect the Sµ bait with the translocation partner regions. Color scale represents the number of translocation partner regions identified in a 10 kb window. Translocations from two Dfl and CD19 DKO replicates are represented separately. e) Relative representation of different classes of motifs predicted to form G-quadruplexes (pG4) in control genomic regions selected randomly (right bar) and +/−300 bp from the center of translocation partner junctions identified from translocations in Dfl and CD19 DKO B cells (right bar). The numbers (n) of Dfl and CD19 DKO hits, and control regions are included in the plots. f) Density of AID motifs (WRCY/RGYW) in +/−300bp from the center of translocation partner junctions identified from translocations in Dfl and CD19 DKO B cells compared to control random regions in euchromatin (Hi-C A compartment) from 2 biological replicates. Statistical significance is calculated using the Wilcoxon signed rank test f), ***** p value <0.000001.

Extended Data Fig. 8 DNMT1 deletion delays oncogenesis in TET-deficient mice.

. a) Diagrammatic representation of the strategy used to confirm G-quadruplex binding. Nuclear lysates of activated B cells were incubated with biotin-conjugated single stranded G4- or non-G4-forming control oligonucleotides (Oligos) captured using streptavidin beads. b) Immunoblots showing flag-tagged BG4 (positive control), ATRX, BLM and DNMT1 proteins. Left lane, 1/10th input from nuclear lysates (1/10th Input); middle lane, proteins pulled down with G4 forming oligonucleotides; and right lane, proteins pulled down with non-G4 control oligonucleotides. The data is representative of at least 2 independent experiments. c) - d) Quantification of c) cell numbers, d) spleen weights, of 10 week-old Tfl (grey), CD19 Dnmt1 KO (purple), CD19 DKO (red) and CD19 TKO (brown) mice from 5 independent experiments. e) Enlarged spleen of 75-week-old CD19 TKO mice compared with Tfl control mice. f) Flow cytometric detection of G-quadruplexes with BG4-Ig antibody or isotype IgG controls in B cells from Tfl (YFP, grey), CD19 Dnmt1 KO (YFP+, purple), CD19 DKO (YFP+, red) and CD19 TKO (YFP+, brown) mice. g) – h) Quantification of median fluorescence intensity (MFI) of g) BG4-Ig signal, h) γH2AX signal from Tfl (YFP), CD19 Dnmt1 KO (YFP+), CD19 DKO (YFP+) and CD19 TKO (YFP+) B cells from 4 independent experiments. i), k) Flow cytometric detection of i) G-quadruplexes with BG4-Ig antibody or isotype IgG controls and k) R-loops using V5-epitope-tagged recombinant RNASE H1 (rRNASE H1) or IgG controls in GC B cells (Fas+) from Tfl (YFP, grey), CD19 DKO (YFP+, red) and CD19 TKO (YFP+, brown) mice. j), l) Quantification of median fluorescence intensity (MFI) of j) BG4-Ig signal and l) R-loops (rRNASE H1) signal in GC B cells (Fas+) from Tfl (YFP, grey), CD19 DKO (YFP+, red) and CD19 TKO (YFP+, brown) mice from 3 independent experiments. m) Model proposing functional interplay between TET and DNMT activities to limit GC B cell expansion. TET deficiency in B cells leads to increased G4 and R-loop structures and is associated with altered gene expression, DNA damage and development of B cell lymphoma. Statistical significance is calculated using one-way ANOVA in c), d), g), h), j) and l). Error bars represent mean +/− standard deviation, * p value ≤0.05, ** p value ≤0.01, *** p value ≤0.0005 **** p value <0.0001.

Extended Data Fig. 9 FACS gating strategy and original blots.

. a) Sequential gating strategy used for the flow cytometry analysis. The respective gate names are mentioned in the corresponding figures. b)-e) scanned immunoblots for Extended Data Fig. 2 (b), 5 (c) and 8 (d).

Supplementary information

Supplementary Information

Supplementary Table 1.

Reporting Summary

Supplementary Table

Supplementary Tables 2 and 3.

Source data

Source Data Fig. 1

Scanned western blots for Fig. 1i.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shukla, V., Samaniego-Castruita, D., Dong, Z. et al. TET deficiency perturbs mature B cell homeostasis and promotes oncogenesis associated with accumulation of G-quadruplex and R-loop structures. Nat Immunol 23, 99–108 (2022). https://doi.org/10.1038/s41590-021-01087-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-01087-w

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