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:

Age-associated clonal B cells drive B cell lymphoma in mice

Abstract

Although cancer is an age-related disease, how the processes of aging contribute to cancer progression is not well understood. In this study, we uncovered how mouse B cell lymphoma develops as a consequence of a naturally aged system. We show here that this malignancy is associated with an age-associated clonal B cell (ACBC) population that likely originates from age-associated B cells. Driven by c-Myc activation, promoter hypermethylation and somatic mutations, IgM+ ACBCs clonally expand independently of germinal centers and show increased biological age. ACBCs become self-sufficient and support malignancy when transferred into young recipients. Inhibition of mTOR or c-Myc in old mice attenuates pre-malignant changes in B cells during aging. Although the etiology of mouse and human B cell lymphomas is considered distinct, epigenetic changes in transformed mouse B cells are enriched for changes observed in human B cell lymphomas. Together, our findings characterize the spontaneous progression of cancer during aging through both cell-intrinsic and microenvironmental changes and suggest interventions for its prevention.

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: B cells increase in size and undergo clonal expansion driven by somatic mutations and epigenetic alterations during aging.
Fig. 2: Aged B cells acquire an epigenetic program of malignant cells, and scRNA-seq reveals them as clonal B cell populations.
Fig. 3: Longitudinal analysis and transplantation experiments reveal ACBCs as a cancerous B cell population that leads to animal fatalities, and co-culture experiments point to their origin.
Fig. 4: Myc deficiency and rapamycin treatment inhibit cell size increase and age-related clonal expansion of B cells.

Similar content being viewed by others

Data availability

Raw reads for RNA sequencing, whole-exome sequencing and reduced-representation bisulfite sequencing are available at the Sequence Read Archive (PRJNA694093). Raw and normalized proteomics data are provided in Supplementary Table 5. All other data supporting the findings of the study are available from the corresponding author upon reasonable request.

Code availability

Code can be found by the following GitHub link: https://github.com/AnastasiaShind/AnastasiaShind/tree/AnastasiaShind-NatAging-Bcells.

References

  1. Niccoli, T. & Partridge, L. Ageing as a risk factor for disease. Curr. Biol. 22, R741–R752 (2012).

    Article  PubMed  CAS  Google Scholar 

  2. SEER Cancer Statistics Review (CSR) 1975–2017. National Cancer Institute https://seer.cancer.gov/archive/csr/1975_2017/browse_csr.php?sectionSEL=4&pageSEL=sect_04_table.17 (2019).

  3. Zenin, A. et al. Identification of 12 genetic loci associated with human healthspan. Commun. Biol. 2, 41 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Martincorena, I. et al. Somatic mutant clones colonize the human esophagus with age. Science 362, 911–917 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Yizhak, K. et al. RNA sequence analysis reveals macroscopic somatic clonal expansion across normal tissues. Science 364, eaaw0726 (2019).

  6. Yokoyama, A. et al. Age-related remodelling of oesophageal epithelia by mutated cancer drivers. Nature 565, 312–317 (2019).

    Article  PubMed  CAS  Google Scholar 

  7. Fane, M. & Weeraratna, A. T. How the ageing microenvironment influences tumour progression. Nat. Rev. Cancer 20, 89–106 (2020).

    Article  PubMed  CAS  Google Scholar 

  8. Gabay, M., Li, Y. & Felsher, D. W. MYC activation is a hallmark of cancer initiation and maintenance. Cold Spring Harb. Perspect. Med. 4, a014241 (2014).

  9. Adams, J. M. et al. The c-myc oncogene driven by immunoglobulin enhancers induces lymphoid malignancy in transgenic mice. Nature 318, 533–538 (1985).

    Article  PubMed  CAS  Google Scholar 

  10. Harris, A. W. et al. The E mu-myc transgenic mouse. A model for high-incidence spontaneous lymphoma and leukemia of early B cells. J. Exp. Med. 167, 353–371 (1988).

    Article  PubMed  CAS  Google Scholar 

  11. Lefebure, M. et al. Genomic characterisation of Eμ-Myc mouse lymphomas identifies Bcor as a Myc co-operative tumour-suppressor gene. Nat. Commun. 8, 14581 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Hofmann, J. W. et al. Reduced expression of MYC increases longevity and enhances healthspan. Cell 160, 477–488 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Simpson, P. & Morata, G. Differential mitotic rates and patterns of growth in compartments in the Drosophila wing. Dev. Biol. 85, 299–308 (1981).

    Article  PubMed  CAS  Google Scholar 

  14. Clavería, C., Giovinazzo, G., Sierra, R. & Torres, M. Myc-driven endogenous cell competition in the early mammalian embryo. Nature 500, 39–44 (2013).

    Article  PubMed  Google Scholar 

  15. Muñoz-Martín, N., Sierra, R., Schimmang, T., Villa Del Campo, C. & Torres, M. Myc is dispensable for cardiomyocyte development but rescues Mycn-deficient hearts through functional replacement and cell competition. Development 146, dev170753 (2019).

  16. Mayle, A. et al. Dnmt3a loss predisposes murine hematopoietic stem cells to malignant transformation. Blood 125, 629–638 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  18. Wang, Y. et al. Tet2-mediated clonal hematopoiesis in nonconditioned mice accelerates age-associated cardiac dysfunction. JCI Insight 5, e135204 (2020).

  19. Saghafinia, S., Mina, M., Riggi, N., Hanahan, D. & Ciriello, G. Pan-cancer landscape of aberrant DNA methylation across human tumors. Cell Rep. 25, 1066–1080 (2018).

    Article  PubMed  CAS  Google Scholar 

  20. Coussens, L. M. & Werb, Z. Inflammation and cancer. Nature 420, 860–867 (2002).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Grivennikov, S. I., Greten, F. R. & Karin, M. Immunity, inflammation, and cancer. Cell 140, 883–899 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Hao, Y., O’Neill, P., Naradikian, M. S., Scholz, J. L. & Cancro, M. P. A B-cell subset uniquely responsive to innate stimuli accumulates in aged mice. Blood 118, 1294–1304 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Zheng, Y. et al. A human circulating immune cell landscape in aging and COVID-19. Protein Cell 11, 740–770 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Rubtsova, K., Rubtsov, A. V., van Dyk, L. F., Kappler, J. W. & Marrack, P. T-box transcription factor T-bet, a key player in a unique type of B-cell activation essential for effective viral clearance. Proc. Natl Acad. Sci. USA 110, E3216–E3224 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Rubtsov, A. V. et al. Toll-like receptor 7 (TLR7)-driven accumulation of a novel CD11c+ B-cell population is important for the development of autoimmunity. Blood 118, 1305–1315 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Moir, S. et al. Evidence for HIV-associated B cell exhaustion in a dysfunctional memory B cell compartment in HIV-infected viremic individuals. J. Exp. Med. 205, 1797–1805 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Wu, C. et al. Lupus-associated atypical memory B cells are mTORC1-hyperactivated and functionally dysregulated. Ann. Rheum. Dis. 78, 1090–1100 (2019).

    Article  PubMed  CAS  Google Scholar 

  28. Brayton, C. F., Treuting, P. M. & Ward, J. M. Pathobiology of aging mice and GEM: background strains and experimental design. Vet. Pathol. 49, 85–105 (2012).

    Article  PubMed  CAS  Google Scholar 

  29. Turturro, A., Duffy, P., Hass, B., Kodell, R. & Hart, R. Survival characteristics and age-adjusted disease incidences in C57BL/6 mice fed a commonly used cereal-based diet modulated by dietary restriction. J. Gerontol. A Biol. Sci. Med. Sci. 57, B379–389 (2002).

  30. Consortium, G. T. The Genotype-Tissue Expression (GTEx) project. Nat. Genet. 45, 580–585 (2013).

    Article  Google Scholar 

  31. Petkovich, D. A. et al. Using DNA methylation profiling to evaluate biological age and longevity interventions. Cell Metab. 25, 954–960 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Yang, Z. et al. Correlation of an epigenetic mitotic clock with cancer risk. Genome Biol. 17, 205 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Zhou, W. et al. DNA methylation loss in late-replicating domains is linked to mitotic cell division. Nat. Genet. 50, 591–602 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Esteller, M., Corn, P. G., Baylin, S. B. & Herman, J. G. A gene hypermethylation profile of human cancer. Cancer Res. 61, 3225–3229 (2001).

    PubMed  CAS  Google Scholar 

  35. Herman, J. G. & Baylin, S. B. Gene silencing in cancer in association with promoter hypermethylation. N. Engl. J. Med. 349, 2042–2054 (2003).

    Article  PubMed  CAS  Google Scholar 

  36. Kimmel, J. C. et al. Murine single-cell RNA-seq reveals cell-identity- and tissue-specific trajectories of aging. Genome Res. 29, 2088–2103 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Tabula Muris, C. A single-cell transcriptomic atlas characterizes ageing tissues in the mouse. Nature 583, 590–595 (2020).

    Article  Google Scholar 

  38. Keller, B. et al. The expansion of human T-bethighCD21low B cells is T cell dependent. Sci. Immunol. 6, eabh0891 (2021).

    Article  PubMed  CAS  Google Scholar 

  39. Ren, X. et al. COVID-19 immune features revealed by a large-scale single-cell transcriptome atlas. Cell 184, 1895–1913 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Sun, L. et al. Rapamycin targets STAT3 and impacts c-Myc to suppress tumor growth. Cell Chem. Biol. 29, 373–385 (2021).

  41. Greenough, A. & Dave, S. S. New clues to the molecular pathogenesis of Burkitt lymphoma revealed through next-generation sequencing. Curr. Opin. Hematol. 21, 326–332 (2014).

    Article  PubMed  CAS  Google Scholar 

  42. Richter, J. et al. Recurrent mutation of the ID3 gene in Burkitt lymphoma identified by integrated genome, exome and transcriptome sequencing. Nat. Genet. 44, 1316–1320 (2012).

    Article  PubMed  CAS  Google Scholar 

  43. Love, C., Remple, R., Li, G. & Dave, S. ID3 loss in vivo synergizes with MYC-driven oncogenesis in Burkitt lymphoma. Blood 126, 3906 (2015).

  44. Chen, S. et al. Id3 orchestrates germinal center B cell development. Mol. Cell Biol. 36, 2543–2552 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Knox, J. J. et al. T-bet+ B cells are induced by human viral infections and dominate the HIV gp140 response. JCI Insight 2, e92943 (2017).

  46. Atallah-Yunes, S. A., Murphy, D. J. & Noy, A. HIV-associated Burkitt lymphoma. Lancet Haematol. 7, e594–e600 (2020).

    Article  PubMed  Google Scholar 

  47. Holla, P. et al. Shared transcriptional profiles of atypical B cells suggest common drivers of expansion and function in malaria, HIV, and autoimmunity. Sci. Adv. 7, eabg8384 (2021).

  48. Zintzaras, E., Voulgarelis, M. & Moutsopoulos, H. M. The risk of lymphoma development in autoimmune diseases: a meta-analysis. Arch. Intern. Med. 165, 2337–2344 (2005).

    Article  PubMed  Google Scholar 

  49. Meyer, N. & Penn, L. Z. Reflecting on 25 years with MYC. Nat. Rev. Cancer 8, 976–990 (2008).

    Article  PubMed  CAS  Google Scholar 

  50. Miller, R. A. et al. Rapamycin-mediated lifespan increase in mice is dose and sex dependent and metabolically distinct from dietary restriction. Aging Cell 13, 468–477 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  52. Li, H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. Preprint at https://arxiv.org/abs/1303.3997 (2013).

  53. Li, H. et al. The sequence alignment/map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Van der Auwera, G. A. et al. From FastQ data to high confidence variant calls: the genome analysis toolkit best practices pipeline. Curr. Protoc. Bioinformatics 43, 11.10.11–11.10.33 (2013).

    Google Scholar 

  55. McLaren, W. et al. The Ensembl Variant Effect Predictor. Genome Biol. 17, 122 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Li, J. et al. TMTpro reagents: a set of isobaric labeling mass tags enables simultaneous proteome-wide measurements across 16 samples. Nat. Methods 17, 399–404 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Navarrete-Perea, J., Yu, Q., Gygi, S. P. & Paulo, J. A. Streamlined tandem mass tag (SL-TMT) protocol: an efficient strategy for quantitative (phospho)proteome profiling using tandem mass tag-synchronous precursor selection-MS3. J. Proteome Res. 17, 2226–2236 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Paulo, J. A., Navarrete-Perea, J., Erickson, A. R., Knott, J. & Gygi, S. P. An internal standard for assessing phosphopeptide recovery from metal ion/oxide enrichment strategies. J. Am. Soc. Mass Spectrom. 29, 1505–1511 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Paulo, J. A. et al. Quantitative mass spectrometry-based multiplexing compares the abundance of 5000 S. cerevisiae proteins across 10 carbon sources. J. Proteom. 148, 85–93 (2016).

    Article  CAS  Google Scholar 

  60. Schweppe, D. K., Rusin, S. F., Gygi, S. P. & Paulo, J. A. Optimized workflow for multiplexed phosphorylation analysis of TMT-labeled peptides using high-field asymmetric waveform ion mobility spectrometry. J. Proteome Res. 19, 554–560 (2020).

    Article  PubMed  CAS  Google Scholar 

  61. Gygi, J. P. et al. Web-based search tool for visualizing instrument performance using the triple knockout (TKO) proteome standard. J. Proteome Res. 18, 687–693 (2019).

    Article  PubMed  CAS  Google Scholar 

  62. Paulo, J. A., O’Connell, J. D. & Gygi, S. P. A triple knockout (TKO) proteomics standard for diagnosing ion interference in isobaric labeling experiments. J. Am. Soc. Mass Spectrom. 27, 1620–1625 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Elias, J. E. & Gygi, S. P. Target-decoy search strategy for mass spectrometry-based proteomics. Methods Mol. Biol. 604, 55–71 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Elias, J. E. & Gygi, S. P. Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry. Nat. Methods 4, 207–214 (2007).

    Article  PubMed  CAS  Google Scholar 

  65. Beausoleil, S. A., Villén, J., Gerber, S. A., Rush, J. & Gygi, S. P. A probability-based approach for high-throughput protein phosphorylation analysis and site localization. Nat. Biotechnol. 24, 1285–1292 (2006).

    Article  PubMed  CAS  Google Scholar 

  66. McCarthy, D. J., Chen, Y. & Smyth, G. K. Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res. 40, 4288–4297 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Krug, K. et al. A curated resource for phosphosite-specific signature analysis. Mol. Cell Proteom. 18, 576–593 (2019).

    Article  CAS  Google Scholar 

  68. Wirbel, J., Cutillas, P. & Saez-Rodriguez, J. Phosphoproteomics-based profiling of kinase activities in cancer cells. Methods Mol. Biol. 1711, 103–132 (2018).

    Article  PubMed  CAS  Google Scholar 

  69. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Wang, Z., Hu, J., Johnson, W. E. & Campbell, J. D. scruff: an R/Bioconductor package for preprocessing single-cell RNA-sequencing data. BMC Bioinformatics 20, 222 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. John, C. R. et al. M3C: Monte Carlo reference-based consensus clustering. Sci Rep. 10, 1816 (2020).

  73. Gayoso, A. et al. A Python library for probabilistic analysis of single-cell omics data. Nat. Biotechnol. 40, 163–166 (2022).

    Article  PubMed  CAS  Google Scholar 

  74. Van de Sande, B. et al. A scalable SCENIC workflow for single-cell gene regulatory network analysis. Nat. Protoc. 15, 2247–2276 (2020).

    Article  PubMed  Google Scholar 

  75. Bolotin, D. A. et al. MiXCR: software for comprehensive adaptive immunity profiling. Nat. Methods 12, 380–381 (2015).

    Article  PubMed  CAS  Google Scholar 

  76. Shugay, M. et al. VDJtools: unifying post-analysis of T cell receptor repertoires. PLoS Comput Biol. 11, e1004503 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  77. Schindelin, J., Rueden, C. T., Hiner, M. C. & Eliceiri, K. W. The ImageJ ecosystem: an open platform for biomedical image analysis. Mol. Reprod. Dev. 82, 518–529 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Meer, M. V., Podolskiy, D. I., Tyshkovskiy, A. & Gladyshev, V. N. A whole lifespan mouse multi-tissue DNA methylation clock. eLife 7, e40675 (2018).

  79. Krueger, F. & Andrews, S. R. Bismark: a flexible aligner and methylation caller for bisulfite-seq applications. Bioinformatics 27, 1571–1572 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Wang, M. & Lemos, B. Ribosomal DNA harbors an evolutionarily conserved clock of biological aging. Genome Res. 29, 325–333 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Tyshkovskiy, A. et al. Identification and application of gene expression signatures associated with lifespan extension. Cell Metab. 30, 573–593 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Street, K. et al. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genomics 19, 477 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  83. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  84. Yu, G., Wang, L.-G., Han, Y. & He, Q.-Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Andreatta, M. & Carmona, S. J. UCell: robust and scalable single-cell gene signature scoring. Comput Struct. Biotechnol. J. 19, 3796–3798 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank members of the Gladyshev laboratory for discussions. This study was supported by the Max Kade Foundation to J.P.C., National Institute on Aging grants to V.N.G. and National Institutes of Health R01DC020190 and R01DC017166 grants to A.A.I.

Author information

Authors and Affiliations

Authors

Contributions

J.P.C. and A.V.S. conceived the project, designed and performed experiments, analyzed the data and drafted the manuscript. A.B., O.S.S., J.A.P., C.K., A.P.P., M.M., M.V.M., M.G., E.L., R.M., Y.H., K.J., A.T., A.K. and G.L. performed experiments, analyzed the data and revised the manuscript. G.L., S.P.G., A.A.I. and J.M.S. provided research materials, assisted with experimental design and revised the manuscript. J.P.M. interpreted the data, designed experiments and provided research materials. V.N.G. supervised the study, obtained funding and revised the manuscript.

Corresponding author

Correspondence to Vadim N. Gladyshev.

Ethics declarations

Competing interests

After the initial submission of this paper, A.V.S. had a change in employment status (Retro Biosciences). M.V.M. is currently employed by Altos Labs. Experimental work was completed before these employment changes. R.M. is a paid employee of and has equity interests in Retro Biosciences. The other authors declare no competing interests.

Peer review

Peer review information

Nature Aging thanks the anonymous reviewers for their contributions 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

Extended Data Fig. 1 Age-related changes in cell size and immune cell composition in C57BL/6 mice.

(a) Number of immune cells in the spleens of young, old and old lymphoma-bearing mice as measured by FACS and calculated based on the total number of cells in the spleen. (b) Mean size of T cells or (c) myeloid cells in the spleens as measured by FACS forward scatter. (d) Number of B cells in the spleens of young, old and old lymphoma-bearing mice as measured by FACS and calculated based on the total number of cells in the spleen. (e) Mean cell size in the spleens of young, old and old lymphoma-bearing mice as measured by FACS normalized to T cell size from the same donors. (f) Gating strategy for total B cells, follicular, marginal zone, CD21-CD23-, germinal center B cells and plasma cells. (g) Correlation between age-related phenotypes of B cells measured in old C57BL/6 mice. Scale bar is a Pearson correlation coefficient. Asterisks indicate statistical significance. **** p-value < 0.0001, *** p-value < 0.005, ** p-value < 0.01, * p-value < 0.05. (h) Similarity score of the transcriptomic signature of mouse cancerous B cells versus publicly available datasets of different human and mouse cancers (left plot). Enrichment scores for the Hallmark MYC targets pathway for human and mouse cancers (right plot). (i) Correlation between the similarity score from (h) on the left plot and enrichment score (NES) from (h) on the right plot. R is a Pearson correlation coefficient. (j) Enrichment of differentially methylated promoters found in B cells from old or lymphoma-bearing mice against those found in human cancers. ‘Mouse lymphoma up’ and ‘Mouse lymphoma down’ gene lists were created from our DNA methylation dataset and used as positive control in the analysis. P-values were calculated by Clusterprofiler package GSEA function that used a permutation test. (k) Enrichment of genes mutated in mouse lymphoma against these mutated in human cancers. ‘Mouse lymphoma’ gene list was created from our genome sequencing dataset and used as a positive control in the analysis. P-values were calculated by Clusterprofiler package GSEA function that used a permutation test. P-values for all other plots were calculated with a two-sided Student t-test unless otherwise specified. Data distribution was assumed to be normal but this was not formally tested. Each dot is the data from one mouse.

Extended Data Fig. 2 Mouse and human B cells clonally expand with age, which leads to reduced Ig diversity.

(a) Correlation between B-cell size as measured by FACS and the size of top B-cell clone as measured based on reconstruction of CDR3 regions from RNA-seq data to estimate Ig (ChaoE_Mean) from FACS-sorted B cells from young and old mice. (b) (left) Proportion of somatic mutations per nucleotide within the CDR3 region in all clones compared to largest clones. (right) IgH isotype distribution across young, old and lymphoma samples. Light blue means ‘not detected’. (c) Correlation between top clone size (Ig) estimated from CDR3 sequencing and top clone size estimated by VAF from whole exome sequences. (d) Correlation of clone size for the same clones reconstructed from RNA and from genomic sequencing of CDR3 regions. (e) Scheme of the analysis of sorted splenic B cells from 27 months old mice by RNA-seq, Immunoseq and WES/WGS. Liver WES/WGS were used as a control to exclude germline somatic mutations. (f) Number of somatic mutations per sample identified with whole exome (upper plot) or genome (bottom, left plot) sequencing. (g) (h) (i) (j) and (k) reconstructed mutational signatures of total somatic mutations in B cells of old mice. (l) Scheme of the analysis of the GTEx dataset. RNA sequences from spleen and blood were used to reconstruct CDR3 regions, estimate Ig diversity (chaoE mean), and WGS of blood from the same subjects was used to predict somatic mutations. (l) Schematic representation of the samples used. Spleen RNA-seq data from the GTEx project was utilized to reconstruct CDR3 diversity (chaoE_mean). Similarly, blood RNA-seq data from the same source was used to assess CDR3 diversity (chaoE_mean) and additionally, WGS data was employed for somatic mutation prediction. (m) Ig diversity (chaoE_mean) in spleens of young and old GTEx subjects calculated from RNA-seq data. P-value was calculated with a two-sided Student t-test. (n) Correlation between Ig diversity (chaoE_mean) in blood of GTEx subjects and their age, in years. (o) Correlation between top clone (WES) and Ig clone sizes (sum of top 5 CDR3 clones) in blood of GTEx subjects. R is a Pearson correlation coefficient in each plot, unless otherwise specified. All boxplots were plotted with geom_boxplot function grom ggplot2 package in R. One side of the box represents the 25th percentile, the other end of the box represents the 75th percentile, line in the middle represents median, length of whiskers is between largest and smallest data points that lay within 1.5 to 3 time the interquartile range.

Extended Data Fig. 3 Clonal expansions of B cells are supported by c-Myc and DNA methylation alterations.

(a) Mean DNA promoter methylation levels for genes encoding c-Myc targets. (b) Correlation between mean B cell size and the rDNA methylation age. (c) Correlation between the DNA methylation age calculated by the blood epigenetic clock36 and the size of top B cell clones. (d) Correlation of top Ig clone size and mean global DNA methylation of promoters and gene bodies in B cells of old mice. (e) c-Myc fold change in young, old and lymphoma FACS-sorted B cells normalized for DNA concentration and measured by qRT-PCR, p-values were calculated with a two-sided Student t-test. Each dot is a mouse in the young group, n = 6. Old and lymphoma groups contain a mixture of samples from regular-sized B cells and enlarged B cells that come from seven mice in the old group and three mice in the lymphoma group. (f) Correlation between Ptpn1 gene expression and mean DNA methylation of its promoter, dots are sized by size of the top clone of B cells in a sample. Correlations between two variables were evaluated using Pearson’s correlation coefficients (g) Same as (f) but for Id3 and Mitf genes. R is a Pearson correlation coefficient whenever R is indicated. Each dot is an independent biological replica. All boxplots were plotted with geom_boxplot function grom ggplot2 package in R, One side of the box represents the 25th percentile, the other end of the box represents the 75th percentile, line in the middle represents median, length of whiskers is between largest and smallest data points that lay within 1.5 to 3 times the interquartile range.

Extended Data Fig. 4 Single-cell RNA sequencing uncovers a clonal B-cell population and its relation to age-associated B cells (ABC).

(a) Enrichment plots of gene expression changes in ABC based on bulk public RNA-seq datasets for signatures from age-related B cell populations based on single-cell RNA-seq. (b) Correlation between clonality signature score estimated for each cell and clone size (% of reconstructed IgK) to which it belongs using two publicly available datasets36,37. (c) UMAP plots of single-cell RNA-seq of the spleens of old mice from the combined Kimmel dataset36 and Tabula Muris Senis dataset37 (left plot). Immune cell enrichment in mitotic genes clustered according to G1, G2M and S phases (right plot). (d) Proportion of cells predicted to be in G1, G2M or S mitotic phases across B cell populations. (e) c-Myc and Tbx21 fold change to Actnb measured with qRT-PCR in splenic follicular B cells, ABC and ACBC FACS-sorted from 25–27-month-old mice, p-value was calculated with a one-sided Student t-test. Data distribution was assumed to be normal but this was not formally tested. Each dot is data from one mouse. (f) Gating strategy for ABC and ACBC populations. (g) Cell sizes measured by FSC of follicular and ACBC in the spleens of young (n = 4) and old (n = 10) mice. (h) Representative images of follicular and ACBC cells immunolabeled for c-Myc and for DNA with DAPI (Scale bar - 10 μm). Quantification of cell sizes and mean c-Myc intensity for follicular and ACBC estimated from confocal images for two independent experiments. (i) Predicted interaction of immune cells by CellChat from single-cell RNA-seq of spleens from four mice coming from two datasets36,37. All boxplots were plotted with geom_boxplot function grom ggplot2 package in R, One side of the box represents the 25th percentile, the other end of the box represents the 75th percentile, line in the middle represents median, length of whiskers is between largest and smallest data points that lay within 1.5 to 3 time the interquartile range.

Extended Data Fig. 5 Age-associated clonal B cells infiltrate peripheral tissues.

(a) UMAP plots of single-cell RNA-seq from many combined tissues across mouse lifespan in months. Data was obtained from Tabula Muris Senis dataset37 (b) ACBC signature enrichment in B-cell populations in a multitude of tissues such as Brown adipose tissue (BAT), Gonadal Adipose Tissue (GAT), Kidney, Liver, Lung, Mesenteric adipose tissue (MAT), Bone marrow, Subcutaneous adipose tissue (SCAT) and spleen. All data was extracted from Tabula Muris Senis dataset37. Cell signature scores were performed by UCell and statistical analysis was performed based on Mann Whitney U statistic85.

Extended Data Fig. 6 Increased B cell clonality and enrichment for ABC and ACBC signatures in aged human donors.

(a) UMAPs showing ABC, ACBC and Myc targets enrichment across age gradient. (b) UMAPs showing the distribution of cells with ‘no’ and ‘yes’ for B cell clonality (c) IgH isotype distribution in ACBC enriched B cells. Single-cell RNA sequencing data was acquired from39 and the corresponding metadata from the Gene Expression Omnibus (GEO) database (GSE158055). Cells with available BCR sequences (‘BCR single cell sequencing’ == ‘Yes’) were selected from 19 healthy donors and retained only B cells or cells with an assigned BCR clone ID.

Extended Data Fig. 7 Longitudinal analysis reveals B-cell size and CD21CD23 B cells as predictors of lifespan, and co-culture experiments point to their origin.

(a) Schematic representation of the longitudinal experiment. Blood was collected from the tails of C57BL/6 female mice across seven timepoints for FACS analysis. (b) and (c) changes in blood cell composition with age starting with 25 mice at 20 months of age, and 11 mice surviving by 31 months of age. (d) c-Myc transcript levels measured by RT-PCR in young (YF) and old follicular (OF) B cells incubated with i) age-associated B cells (ABC) from old spleens, ii) immune cells (CD45+) from young spleens excluding B cells (yEE), or iii) immune cells (CD45+) from old spleens excluding B cells (oEE). Results shown are from three independent experiments, three biological replicas of mice in two experiments, and four in the third experiment. P-values for all other plots were calculated with a two-sided Student t-test. Data distribution was assumed to be normal but this was not formally tested. Follicular and ABC as well as cells excluding CD19+ cells (EE) were sorted from different spleens to recover enough cells per replica. (e) Boxplot of log2FC of ABC (darkred) and ACBC (darkblue) upregulated genes in WT B cells and the same genes in CD22KO GC B cells from light zone (LZ, pale red and marine green) and dark zone (DZ, red and cyan) of CD22KO mice (Supplementary Table 2, PMID: 52943874). Each dot represents a gene from either the ABC or ACBC signature. Wilcoxon non-parametric test for comparing means of independent samples with the greater alternative, **** p-value < 0.0001, *** p-value < 0.005, ** p-value < 0.01, * p-value < 0.05. All boxplots were plotted with geom_boxplot function grom ggplot2 package in R. One side of the box represents the 25th percentile, the other end of the box represents the 75th percentile, line in the middle represents median, length of whiskers is between largest and smallest data points that lay within 1.5 to 3 time the interquartile range.

Extended Data Fig. 8 Effects of c-Myc deficiency and overexpression on immune cell composition and premalignant phenotypes of B cells.

(a) c-Myc expression fold change normalized to DNA concentration in B cells, (b) spleen weight normalized by body weight, and (c) the RNA to DNA concentration ratio in B cells, (d–f) splenic cell sizes from Myc+/− mice and their wild type (Wt) siblings. (g) Cell composition of immune cells (CD45+) and B-cell populations (h, i) in the spleens of old Wt and Myc+/− mice, gated as in Extended Data Fig. 1f,g. (j) Analysis of Emu-Myc mice. (k) Total RNA/DNA ratio in FACS-sorted splenic B cells from non-carrier (NC) controls and Emu-Myc mice. n = 3 mice in each Wt and Eu-Myc groups, Eu-Myc points are a mixture of regular-sized B cells and enlarged B cells summing up to two samples per mouse. (l) Mean size of B and T cells measured by FACS in the spleens of NC and Emu-Myc mice. (m) Mean cell size of B-cell populations measured by FACS in the bone marrow of NC and Emu-Myc mice. (n) Ig diversity (chaoE_mean) of splenic B cells from NC and Emu-Myc mice, calculated from RNA-seq data using mixcr and VDJtools software, n = 3 mice in each Wt and Eu-Myc groups, Eu-Myc points are a mixture of regular-sized B cells and enlarged B cells summing up to two samples per mouse. All boxplots were plotted with geom_boxplot function grom ggplot2 package in R. One side of the box represents the 25th percentile, the other end of the box represents the 75th percentile and represent interquartile range, line in the middle of the box represents median, length of whiskers is between largest and smallest data points that lay within 1.5 to 3 times the interquartile range. All p-values were calculated using one-sided T-test. Each dot is a mouse unless specified otherwise.

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Tables 1–5.

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

Castro, J.P., Shindyapina, A.V., Barbieri, A. et al. Age-associated clonal B cells drive B cell lymphoma in mice. Nat Aging (2024). https://doi.org/10.1038/s43587-024-00671-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s43587-024-00671-7

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer