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:

Disruption of the Uty epigenetic regulator locus in hematopoietic cells phenocopies the profibrotic attributes of Y chromosome loss in heart failure

Abstract

Heart failure affects millions of people worldwide, with men exhibiting a higher incidence than women. Our previous work has shown that mosaic loss of the Y chromosome (LOY) in leukocytes is causally associated with an increased risk for heart failure. Here, we show that LOY macrophages from the failing hearts of humans with dilated cardiomyopathy exhibit widespread changes in gene expression that correlate with cardiac fibroblast activation. Moreover, we identify the ubiquitously transcribed tetratricopeptide Y-linked (Uty) gene in leukocytes as a causal locus for an accelerated progression of heart failure in male mice with LOY. We demonstrate that Uty disruption leads to epigenetic alterations in both monocytes and macrophages, increasing the propensity of differentiation into profibrotic macrophages. Treatment with a transforming growth factor-β-neutralizing antibody prevented the cardiac pathology associated with Uty deficiency in leukocytes. These findings shed light on the mechanisms that contribute to the higher incidence of heart failure in men.

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: Hematopoietic LOY in human DCM scRNA-Seq datasets.
Fig. 2: Deficiency of Y chromosome genes in hematopoietic cells exacerbates cardiac dysfunction in response to pressure overload.
Fig. 3: Hematopoietic Uty is a potential gene candidate that contributes to LOY-mediated deterioration of cardiac function.
Fig. 4: Uty disruption in hematopoietic cells accelerates cardiac dysfunction in response to pressure overload.
Fig. 5: Multimodal single-cell analysis reveals the profibrotic chromatin accessibility signatures of recruited UtyGT cardiac monocytes and macrophages.
Fig. 6: Inhibition of TGFβ reverses cardiac dysfunction after TAC in mice with hematopoietic Uty deficiency.

Similar content being viewed by others

Data availability

All data supporting the findings in this study are included in the main article and associated files. Single-cell multiomics raw sequencing data and processed data files used to generate and perform analyses of control and UtyGT cardiac leukocytes are available in the NCBI GEO repository under accession no. GSE241486. Additional analyses used datasets available in the NCBI GEO repository under accession nos. GSE183852 and GSE145154 and the Broad Institute Single Cell Portal under accession no. SCP1303. Source data are provided with this paper.

Code availability

Code used to analyze datasets was generated from available vignettes and tutorials. This code is available at https://github.com/nicholaschavkin/UTY_HF.

References

  1. Thompson, D. J. et al. Genetic predisposition to mosaic Y chromosome loss in blood. Nature 575, 652–657 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Jacobs, P. A. et al. Change of human chromosome count distributions with age: evidence for a sex difference. Nature 197, 1080–1081 (1963).

    Article  ADS  CAS  PubMed  Google Scholar 

  3. Pierre, R. V. & Hoagland, H. C. Age-associated aneuploidy: loss of Y chromosome from human bone marrow cells with aging. Cancer 30, 889–894 (1972).

    Article  CAS  PubMed  Google Scholar 

  4. Colaco, S. & Modi, D. Genetics of the human Y chromosome and its association with male infertility. Reprod. Biol. Endocrinol. 16, 14 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Forsberg, L. A. et al. Mosaic loss of chromosome Y in peripheral blood is associated with shorter survival and higher risk of cancer. Nat. Genet. 46, 624–628 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Haitjema, S. et al. Loss of Y chromosome in blood is associated with major cardiovascular events during follow-up in men after carotid endarterectomy. Circ. Cardiovasc. Genet. 10, e001544 (2017).

    Article  CAS  PubMed  Google Scholar 

  7. Mas-Peiro, S. et al. Mosaic loss of Y chromosome in monocytes is associated with lower survival after transcatheter aortic valve replacement. Eur. Heart J. 44, 1943–1952 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Sano, S. et al. Hematopoietic loss of Y chromosome leads to cardiac fibrosis and heart failure mortality. Science 377, 292–297 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  9. Dumanski, J. P. et al. Immune cells lacking Y chromosome show dysregulation of autosomal gene expression. Cell. Mol. Life Sci. 78, 4019–4033 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Chaffin, M. et al. Single-nucleus profiling of human dilated and hypertrophic cardiomyopathy. Nature 608, 174–180 (2022).

    Article  ADS  CAS  PubMed  Google Scholar 

  11. Koenig, A. L. et al. Single-cell transcriptomics reveals cell-type-specific diversification in human heart failure. Nat. Cardiovasc. Res. 1, 263–280 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Rao, M. et al. Resolving the intertwining of inflammation and fibrosis in human heart failure at single-cell level. Basic Res. Cardiol. 116, 55 (2021).

    Article  ADS  PubMed  Google Scholar 

  13. Daseke, M. J. 2nd et al. Cardiac fibroblast activation during myocardial infarction wound healing: fibroblast polarization after MI. Matrix Biol. 91–92, 109–116 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Mouton, A. J. et al. Fibroblast polarization over the myocardial infarction time continuum shifts roles from inflammation to angiogenesis. Basic Res. Cardiol. 114, 6 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Burgoyne, P. S. & Arnold, A. P. A primer on the use of mouse models for identifying direct sex chromosome effects that cause sex differences in non-gonadal tissues. Biol. Sex Differ. 7, 68 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Chavkin, N. W. et al. The cell surface receptors Ror1/2 control cardiac myofibroblast differentiation. J. Am. Heart Assoc. 10, e019904 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Wang, Y. et al. Wnt5a-mediated neutrophil recruitment has an obligatory role in pressure overload-induced cardiac dysfunction. Circulation 140, 487–499 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Soh, Y. Q. S. et al. Sequencing the mouse Y chromosome reveals convergent gene acquisition and amplification on both sex chromosomes. Cell 159, 800–813 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Skelly, D. A. et al. Single-cell transcriptional profiling reveals cellular diversity and intercommunication in the mouse heart. Cell Rep. 22, 600–610 (2018).

    Article  CAS  PubMed  Google Scholar 

  20. Sano, S. et al. Lentiviral CRISPR/Cas9-mediated genome editing for the study of hematopoietic cells in disease models. J. Vis. Exp. https://doi.org/10.3791/59977 (2019).

    Article  PubMed  Google Scholar 

  21. Chu, V. T. et al. Efficient CRISPR-mediated mutagenesis in primary immune cells using CrispRGold and a C57BL/6 Cas9 transgenic mouse line. Proc. Natl Acad. Sci. USA 113, 12514–12519 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  22. Shpargel, K. B., Sengoku, T., Yokoyama, S. & Magnuson, T. UTX and UTY demonstrate histone demethylase-independent function in mouse embryonic development. PLoS Genet. 8, e1002964 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Park, E. et al. Bone marrow transplantation procedures in mice to study clonal hematopoiesis. J. Vis. Exp. https://doi.org/10.3791/61875 (2021).

    Article  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  25. Matsumura, T. et al. An azoospermic factor gene, Ddx3y and its paralog, Ddx3x are dispensable in germ cells for male fertility. J. Reprod. Dev. 65, 121–128 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Shi, B. et al. UTX condensation underlies its tumour-suppressive activity. Nature 597, 726–731 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Walport, L. J. et al. Human UTY(KDM6C) is a male-specific Nϵ-methyl lysyl demethylase. J. Biol. Chem. 289, 18302–18313 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Conlon, F. L. & Arnold, A. P. Sex chromosome mechanisms in cardiac development and disease. Nat. Cardiovasc. Res. 2, 340–350 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Sano, S., Thel, M. C. & Walsh, K. Mosaic loss of Y chromosome in white blood cells: its impact on men’s health. Physiology (Bethesda) 38, 0 (2023).

    PubMed  Google Scholar 

  30. Li, R. & Zhu, J. Effects of aneuploidy on cell behaviour and function. Nat. Rev. Mol. Cell Biol. 23, 250–265 (2022).

    Article  PubMed  Google Scholar 

  31. Zhang, Q. et al. Mosaic loss of chromosome Y promotes leukemogenesis and clonal hematopoiesis. JCI Insight 7, e153768 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Abdel-Hafiz, H. A. et al. Y chromosome loss in cancer drives growth by evasion of adaptive immunity. Nature 619, 624–631 (2023).

    Article  ADS  CAS  PubMed  Google Scholar 

  33. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Sano, S. & Walsh, K. Development of a mouse model of hematopoietic loss of Y chromosome. Bio-protocol 13, e4729 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Wang, Y. et al. Murine models of clonal haematopoiesis to assess mechanisms of cardiovascular disease. Cardiovasc. Res. 118, 1413–1432 (2022).

    Article  CAS  PubMed  Google Scholar 

  36. Stuart, T. et al. Single-cell chromatin state analysis with Signac. Nat. Methods 18, 1333–1341 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Feng, J. et al. Identifying ChIP–seq enrichment using MACS. Nat. Protoc. 7, 1728–1740 (2012).

    Article  CAS  PubMed  Google Scholar 

  38. van Dijk, D. et al. Recovering gene interactions from single-cell data using data diffusion. Cell 174, 716–729 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by National Institutes of Health (NIH) grants AG073249 and HL142650 and National Aeronautics and Space Administration grant 80NSSC21K0549 to K.W.; NIH grant HL152174 to S.S. and K.W.; the University of Virginia Medical Scientist Training Program (T32GM007267) to J.D.C.; American Heart Association grant 23CDA1054358 to N.W.C.; Grant-in-Aid for Research Activity Start-up (21K20879) to S.S.; Grant-in-Aid for Scientific Research (22K08162) to S.S.; Grant for Basic Research of the Japanese Heart Failure Society to S.S.; Grant for Basic Research of the Japanese Circulation Society to S.S.; Research Grant of the Japan Cardiovascular Research Foundation to S.S.; Research Grant of the SENSHIN Medical Research Foundation to S.S.; Research Grant of the MSD Life Science Foundation to S.S.; Novartis research grant to S.S.; Research Grant of the Kondou Kinen Medical Foundation to S.S.; and the Bayer Scholarship for Cardiovascular Research to S.S. We would like to thank X. Chen and H. Hrncir (Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA) for technical assistance. We acknowledge the use of Servier Medical Art, as Fig. 5a was partially generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license. Some illustrations (Fig. 3a and Supplementary Fig. 1) were created with BioRender.com. This research was made possible by the University of Virginia Flow Cytometry Core (RRID: SCR_017829) and Genome Analysis and Technology Core (RRID: SCR_018883).

Author information

Authors and Affiliations

Authors

Contributions

S.S. and K.W. designed the experiments. K.H., N.W.C., Yohei A., Y.W., H.O., Y.Y., M.A.E., J.D.C., M.C.T., M.S., E.M.-Y., Yuka A., H.D., A.H.P. and S.S. performed the experiments. Y.W. performed statistical analyses. A.P.A., B.D.G. and K.K.H. provided scientific advice. S.S., N.W.C. and K.W. wrote the paper. All authors approved the final version of the paper.

Corresponding authors

Correspondence to Soichi Sano or Kenneth Walsh.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cardiovascular Research thanks Tim Stuart, Alan Tall 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

Extended Data Fig. 1 Hematopoietic LOY in human dilated cardiomyopathy scRNA-Seq datasets.

a. UMAP dimensionality reduction of annotated cardiac cells from integrated scRNA-Seq datasets (Koenig NCVR 2022, Rao BRC 2021, Chaffin Nature 2022). b. Expression of cell type-specific genes used for annotation. c. UMAP of annotated cardiac leukocytes. d. Expression of cell type-specific genes used for annotation. e. Total number of control and LOY leukocytes within each leukocyte subtype (gray = control, red = LOY). f. UMAP of annotated cardiac fibroblasts with expression of cell type-specific genes used for myofibroblast annotation. g. Linear correlation of myofibroblast percentage within each patient and LOY percentage within each patient, presented by disease.

Source data

Extended Data Fig. 2 Hematopoietic cells with Y chromosome gene deficiency do not display fitness advantage in vivo.

Mice underwent partial (50%) bone marrow reconstitution with XY* and XY*X cells following lethal irradiation. After 4 weeks and 8 weeks of recovery, flow cytometric analysis of peripheral blood was performed. (XY* n = 7, XY*X n = 7). Data are presented as mean values +/- SEM. WBC; white blood cells.

Source data

Extended Data Fig. 3 Hematopoietic LOY in human healthy cardiac scRNA-Seq datasets.

a. UMAP dimensionality reduction of annotated cardiac cells from integrated scRNA-Seq datasets of healthy male patients. b-c. Expression of markers for immune cells (PTPRC), monocytes/macrophages (CD68, CD14), and Y chromosome genes (UTY, DDX3Y, KDM5D), visualized on (B) UMAP dimensionality reduction plots, and (C) relative expression between cell types.

Source data

Extended Data Fig. 4 CRISPR/Cas9-mediated gene disruption of Y chromosome genes in bone marrow lineage-negative cells.

a. Gating strategy used for the Fluorescence-Activated Cell Sorting (FACS) of peripheral blood cells. Cells transduced with lentivirus transduced cells are designed to express turbo red fluorescent protein (tRFP). b-e. The results of Tracking of Indels by Decomposition (TIDE) analysis of sorted cells revealed the presence of multiple insertions and deletions in Ddx3y (b), Uty (c), Kdm5d (d), Eif2s3y (e) genes. f. Percentage of tRFP-positive cells in blood following BMT in the different gene-editing screens (n = 10 for all conditions, statistical analysis by Student’s t test). Data are presented as mean values +/- SEM.

Source data

Extended Data Fig. 5 Hematopoietic parameters remain unaffected by CRISPR/Cas9-mediated disruption of hematopoietic Y chromosome genes (Ddx3y, Kdm5d, Uty, Eif2s3y).

White blood cell count, hemoglobin concentration, and platelet count were assessed at 4 months (a, b, d) and 1 month (c) post-bone marrow transplant (n = 10 for all conditions). Data are presented as mean values +/- SEM. WBC; white blood cell, HGB; hemoglobin, PLT; platelet.

Source data

Extended Data Fig. 6 CRISPR/Cas9-mediated disruption of hematopoietic Eif2s3y, Ddx3y, or Kdm5d genes do not promote cardiac dysfunction after TAC.

a, c and e. Sequential echocardiographic analysis of KO mice and control mice after TAC at the indicated time points (a: control, n = 6; Eif2s3y-KO, n = 6, c: control n = 10, Ddx3y-KO n = 8, e: control n = 25, Kdm5d-KO n = 23). b, d and f. Heart weight (HW) and lung weight (LW) relative to tibia length (TL) at 28 days after TAC procedure (b-HW/TL; control, n = 5; Eif2s3y-KO, n = 6, b-LW/TL; control, n = 5; Eif2s3y-KO, n = 6, d-HW/TL: control n = 9, Ddx3y-KO n = 8, d-LW/TL: control n = 8, Ddx3y-KO n = 8, f-HW/TL: control n = 24, Kdm5d-KO n = 21, f-LW/TL: control n = 7, Kdm5d-KO n = 7). Data are presented as mean values +/- SEM.

Source data

Extended Data Fig. 7 Uty expression and X chromosome paralogs in hematopoietic cells of UtyGT mice.

a. Transcript levels of Uty in total bone marrow cells (BM, wild-type n = 3, UtyGT n = 3, Student’s t test) and whole peripheral blood cells (PB, wild-type n = 5, UtyGT n = 5, Student’s t test). b. Transcript levels of Utx, Ddx3x, Eif2s3x, and Kdm5c in BM cells (n = 3 for all conditions). After TAC, lung weight, body weight and blood cell counts in wild-type mice and UtyGT mice were determined. c. Lung weight relative to tibial length at 0 and 28 days after TAC procedure. (Day 0: wild-type n = 5, UtyGT n = 5, Day 28: wild-type n = 10, UtyGT n = 10). d. Body weight over the time course after TAC (wild-type n = 10, UtyGT n = 10). LW; lung weight, TL: tibial length, BW; body weight. e. Flow cytometry was performed at baseline and 1 month post-TAC to determine cell counts in peripheral blood (Pre: Control n = 9, UtyGT n = 9, 1 M after TAC: Control n = 8, UtyGT n = 6, statistical analysis by two-way ANOVA post hoc Tukey). Data are presented as mean values +/- SEM, p-values: N.S.=not significant, *<0.05, **<0.01. BM; bone marrow, PB; peripheral blood; WBC; white blood cells.

Source data

Extended Data Fig. 8 Transgenic-mediated Ddx3y-KO of hematopoietic cells does not promote cardiac dysfunction after TAC.

a. Schematic of experimental procedure. b. Transcript levels of the Y chromosome genes (Eif2s3y, Ddx3y, Kdm5d, Uty) in bone marrow cells (wild-type n = 3, Ddx3y-KO n = 3). c. Transcript levels of the X chromosome gene homologues (Eif2s3x, Ddx3x, Kdm5c, Utx) in white blood cells isolated from bone marrow from either group of mice (wild-type n = 3, Ddx3y-KO n = 3). d. Sequential echocardiographic analysis of mice transplanted with wild-type or Ddx3y-KO cells. Repeated measurement was performed at the indicated time points after TAC operation (wild-type, n = 10; Ddx3y-KO, n = 8). e. Heart weight (HW) and lung weight (LW) relative to tibial length (TL) at 4 weeks after TAC procedure (HW/TL; control, n = 10; Ddx3y-KO, n = 8, LW/TL; wild-type, n = 10; Ddx3y-KO, n = 8). f. Transcript levels of heart failure markers in heart tissue at four weeks after TAC operation (wild-type, n = 10; Ddx3y-KO, n = 8). Statistical analyses were performed using 2-way ANOVA with Sidak’s multiple comparison tests d. and two-sided unpaired Student’s t test (b, c, e, f). TAC; transverse aortic constriction, FS; fractional shortening, LVPWTd; left ventricular posterior wall thickness at end-diastole, LVDs; left ventricular diameter at end-systole, LVDd; left ventricular diameter at end-diastole. Dots in all panels represent individual samples. Data are shown as mean ± SEM.

Source data

Extended Data Fig. 9 Multi-modal single-cell omics of recruited UtyGT cardiac leukocytes.

a. Quality control metrics for single cell muiltiomics dataset: RNA reads per cell (nCount_RNA), ATAC reads per cell (nCount_ATAC), quantified peaks per cell (nCount_peaks), enrichment of ATAC reads near transcriptional start sites (TSS Enrichment), nucleosome banding pattern (nucleosome_signal) (box plots: min 25%, max 75%, middle median, whiskers 5%-95%, Control n = 8156, UtyGT n = 9169). b. DNA accessibility coverage plot for the Uty locus in Control and UTYGT samples showing ATAC reads and calculated peaks. c. Expression of known genes that are enriched in specific leukocyte cell types used to annotate clusters of single cells within the single-cell multiomics dataset. d. UMAP dimensionality reduction of wild-type and UtyGT cardiac leukocytes based on differential chromatin availability around genes. e. Scoring of each cell on increased chromatin availability around genes associated with fibrotic macrophages (Fibrotic Score) or inflammatory macrophages (Inflammatory Score), plotted by UMAP position.

Source data

Extended Data Fig. 10 Abundance of monocytes and macrophages in the hearts from control and UtyGT mice after TAC.

a. Relative abundance of monocytes and macrophages in datasets from control and UtyGT hearts quantified by percentage of total cell number. b. Flow cytometric analysis of macrophage counts in heart tissue 28 days after TAC. The absolute numbers of cells were normalized by tissue weight (Sham: wild-type n = 5, UtyGT n = 5; TAC: wild-type n = 5, UtyGT n = 5, data are presented as mean values +/- SEM, two-way ANOVA post hoc Tukey, p-value: **<0.01, ***<0.001, ****<0.0001). c. Variation in expression of each gene between Control and UtyGT monocytes and macrophages plotted as -Log10(p-value) versus Log2(Fold Change) (Wilcoxon Rank Sum statistical test). d, e. Gene Ontology terms significantly enriched in significant differentially expressed genes monocytes and macrophages (d: enriched in UtyGT cells, e: enriched in Control cells).

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1–5 and Fig. 1.

Reporting Summary

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical 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

Horitani, K., Chavkin, N.W., Arai, Y. et al. Disruption of the Uty epigenetic regulator locus in hematopoietic cells phenocopies the profibrotic attributes of Y chromosome loss in heart failure. Nat Cardiovasc Res 3, 343–355 (2024). https://doi.org/10.1038/s44161-024-00441-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s44161-024-00441-z

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing