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:

p16High senescence restricts cellular plasticity during somatic cell reprogramming

Abstract

Despite advances in four-factor (4F)-induced reprogramming (4FR) in vitro and in vivo, how 4FR interconnects with senescence remains largely under investigated. Here, using genetic and chemical approaches to manipulate senescent cells, we show that removal of p16High cells resulted in the 4FR of somatic cells into totipotent-like stem cells. These cells expressed markers of both pluripotency and the two-cell embryonic state, readily formed implantation-competent blastoids and, following morula aggregation, contributed to embryonic and extraembryonic lineages. We identified senescence-dependent regulation of nicotinamide N-methyltransferase as a key mechanism controlling the S-adenosyl-l-methionine levels during 4FR that was required for expression of the two-cell genes and acquisition of an extraembryonic potential. Importantly, a partial 4F epigenetic reprogramming in old mice was able to reverse several markers of liver aging only in conjunction with the depletion of p16High cells. Our results show that the presence of p16High senescent cells limits cell plasticity, whereas their depletion can promote a totipotent-like state and histopathological tissue rejuvenation during 4F reprogramming.

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: p16High cells have a negative impact on iPS cell reprogramming.
Fig. 2: Removal of p16High cells during in vivo and in vitro iPS cell reprogramming leads to emergence of cells with totipotent features.
Fig. 3: Chemical removal of p16High cells during in vitro and in vivo reprogramming with senolytics D+Q leads to iPS cells with totipotent features.
Fig. 4: Depletion of senescent cells promotes iPS cell lines with epigenetic characteristics of ES and 2CLCs.
Fig. 5: Senescence-free iPS cell lines produce implantation-competent blastoids in vitro and contribute to extraembryonic lineage in vivo.
Fig. 6: NNMT-dependent regulation of SAM blocks transition into a totipotent-like state during iPS cell reprogramming.
Fig. 7: Removal of p16High cells improves liver plasticity during 4FPR in aging mice.

Similar content being viewed by others

Data availability

Sequencing raw data are deposited as sequence read archives and available under accession number PRJNA891955. The scRNA-seq data reported in this paper are deposited in the Genome Sequence Archive of the National Genomics Data Center, China National Center for Bioinformation/Beijing Institute of Genomics, Chinese Academy of Sciences (GSA: CRA010829) and are publicly accessible at https://ngdc.cncb.ac.cn/gsa. All original images are deposited with Mendeley (https://doi.org/10.17632/2xt47pcfwb.3). All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

References

  1. Hishida, T. et al. In vivo partial cellular reprogramming enhances liver plasticity and regeneration. Cell Rep. 39, 110730 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Reddy, P., Memczak, S. & Izpisua Belmonte, J. C. Unlocking tissue regenerative potential by epigenetic reprogramming. Cell Stem Cell 28, 5–7 (2021).

    Article  CAS  PubMed  Google Scholar 

  3. Ocampo, A. et al. In vivo amelioration of age-associated hallmarks by partial reprogramming. Cell 167, 1719–1733.e12 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Baker, D. J. et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 479, 232–236 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Baker, D. J. et al. Naturally occurring p16Ink4a-positive cells shorten healthy lifespan. Nature 530, 184–189 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Childs, B. G. et al. Senescent cells: an emerging target for diseases of ageing. Nat. Rev. Drug Discov. 16, 718–735 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Xu, M. et al. Senolytics improve physical function and increase lifespan in old age. Nat. Med. 24, 1246–1256 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Muñoz-Espín, D. et al. Programmed cell senescence during mammalian embryonic development. Cell 155, 1104–1118 (2013).

    Article  PubMed  Google Scholar 

  9. Storer, M. et al. Senescence is a developmental mechanism that contributes to embryonic growth and patterning. Cell 155, 1119–1130 (2013).

    Article  CAS  PubMed  Google Scholar 

  10. Li, H. et al. The Ink4/Arf locus is a barrier for iPS cell reprogramming. Nature 460, 1136–1139 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Banito, A. et al. Senescence impairs successful reprogramming to pluripotent stem cells. Genes Dev. 23, 2134–2139 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Utikal, J. et al. Immortalization eliminates a roadblock during cellular reprogramming into iPS cells. Nature 460, 1145–1148 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Mosteiro, L. et al. Tissue damage and senescence provide critical signals for cellular reprogramming in vivo. Science 354, aaf4445 (2016).

    Article  PubMed  Google Scholar 

  14. Wyatt, C. D. R. et al. A developmentally programmed splicing failure contributes to DNA damage-response attenuation during mammalian zygotic genome activation. Sci. Adv. 8, eabn4935 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Shen, H. et al. Mouse totipotent stem cells captured and maintained through spliceosomal repression. Cell 184, 2843–2859.e20 (2021).

    Article  CAS  PubMed  Google Scholar 

  16. Morgani, S. M. et al. Totipotent embryonic stem cells arise in ground-state culture conditions. Cell Rep. 3, 1945–1957 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Genet, M. & Torres-Padilla, M. E. The molecular and cellular features of 2-cell-like cells: a reference guide. Development 147, dev189688 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. Abad, M. et al. Reprogramming in vivo produces teratomas and iPS cells with totipotency features. Nature 502, 340–345 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. Grosse, L. et al. Defined p16High senescent cell types are indispensable for mouse healthspan. Cell Metab. 32, 87–99.e6 (2020).

    Article  CAS  PubMed  Google Scholar 

  20. Hall, B. M. et al. p16(Ink4a) and senescence-associated β-galactosidase can be induced in macrophages as part of a reversible response to physiological stimuli. Aging 9, 1867–1884 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Chen, T. et al. Embryonic stem cells promoting macrophage survival and function are crucial for teratoma development. Front. Immunol. 5, 93414–93430 (2014).

    Article  Google Scholar 

  22. Jachowicz, J. W. et al. LINE-1 activation after fertilization regulates global chromatin accessibility in the early mouse embryo. Nat. Genet. 49, 1502–1510 (2017).

    Article  CAS  PubMed  Google Scholar 

  23. Zhu, Y. et al. The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell 14, 644–658 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Zhu, Y. et al. Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell 15, 428–435 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ritschka, B. et al. The senotherapeutic drug ABT-737 disrupts aberrant p21 expression to restore liver regeneration in adult mice. Genes Dev. 34, 489–494 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Fragola, G. et al. Cell reprogramming requires silencing of a core subset of polycomb targets. PLoS Genet. 9, e1003292 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Yang, M. et al. Chemical-induced chromatin remodeling reprograms mouse ESCs to totipotent-like stem cells. Cell Stem Cell 29, 400–418.e13 (2022).

    Article  CAS  PubMed  Google Scholar 

  28. Huang, Y. et al. Stella modulates transcriptional and endogenous retrovirus programs during maternal-to-zygotic transition. eLife 6, e22345 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Sozen, B. et al. Self-organization of mouse stem cells into an extended potential blastoid. Dev. Cell 51, 698–712.e8 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Sozen, B. et al. Self-assembly of embryonic and two extra-embryonic stem cell types into gastrulating embryo-like structures. Nat. Cell Biol. 20, 979–989 (2018).

    Article  CAS  PubMed  Google Scholar 

  31. Li, R. et al. Generation of blastocyst-like structures from mouse embryonic and adult cell cultures. Cell 179, 687–702.e18 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Posfai, E. et al. Evaluating totipotency using criteria of increasing stringency. Nat. Cell Biol. 23, 49–60 (2021).

    Article  CAS  PubMed  Google Scholar 

  33. Martinez-Val, A. et al. Dissection of two routes to naïve pluripotency using different kinase inhibitors. Nat. Commun. 12, 1863 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zhao, J. et al. Metabolic remodelling during early mouse embryo development. Nat. Metab. 3, 1372–1384 (2021).

    Article  CAS  PubMed  Google Scholar 

  35. Dey, S. K. et al. Repurposing an adenine riboswitch into a fluorogenic imaging and sensing tag. Nat. Chem. Biol. 18, 180–190 (2022).

    Article  CAS  PubMed  Google Scholar 

  36. Roberti, A., Fernández, A. F. & Fraga, M. F. Nicotinamide N-methyltransferase: at the crossroads between cellular metabolism and epigenetic regulation. Mol. Metab. 45, 101165 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Riveiro, A. R. & Brickman, J. M. From pluripotency to totipotency: an experimentalist’s guide to cellular potency. Development 147, dev189845 (2020).

    Article  CAS  PubMed  Google Scholar 

  38. Vitullo, P., Sciamanna, I., Baiocchi, M., Sinibaldi-Vallebona, P. & Spadafora, C. LINE-1 retrotransposon copies are amplified during murine early embryo development. Mol. Reprod. Dev. 79, 118–127 (2012).

    Article  CAS  PubMed  Google Scholar 

  39. Garcia-Perez, J. L. et al. LINE-1 retrotransposition in human embryonic stem cells. Hum. Mol. Genet. 16, 1569–1577 (2007).

    Article  CAS  PubMed  Google Scholar 

  40. Richardson, S. R. et al. Heritable L1 retrotransposition in the mouse primordial germline and early embryo. Genome Res. 27, 1395–1405 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Chondronasiou, D. et al. Multi-omic rejuvenation of naturally aged tissues by a single cycle of transient reprogramming. Aging Cell 21, e13578 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Tanaka, S., Kunath, T., Hadjantonakis, A. K., Nagy, A. & Rossant, J. Promotion of trophoblast stem cell proliferation by FGF4. Science 282, 2072–2075 (1998).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

B.B.G. is a recipient of the ‘Vernadski bourse’ scholarship given by the French government and a postdoctoral fellowship from Ulysseus European University. Research in the laboratory of D.V.B. is supported by the Foundation FRM, ANR and INSERM programme ‘Agemed’. Research in the laboratory of S.S. is supported by the Agence Nationale de la Recherche, research in the laboratory of F.L. was supported by grants from the National Natural Science Foundation of China (31830061, 32030032) and research in the laboratory of O.N.D. was supported by RSF grant 19-75-20128. We thank M. Serrano and H. Li for providing i4F mouse strains19. We thank W. Li (Institute of Zoology, Institute for Stem Cell and Regeneration, Beijing) and L. Chao (Institute of Zoology, Institute for Stem Cell and Regeneration, Beijing) for their assistance in morula aggregation and transplantation. We thank S. R. Jaffrey (Department of Pharmacology, Weill Medical College, Cornell University, New York, NY, USA) for providing SAM sensor plasmids35. We acknowledge the ImagingCore Facility (PICMI), histology, genomic and animal housing facilities of IRCAN supported by le Cancéropole PACA, la Région PACA, le Conseil Départementale 06, l’INSERM, ARC, IBiSA and the Conseil Départemental 06 de la Région PACA.

Author information

Authors and Affiliations

Authors

Contributions

B.B.G., D.E., G.L., L.G., A.E., Z.K., A.K., B.K., C.M., E.L., F.L. and S.S. designed, conducted and analysed the experiments. D.E. and S.S. conducted the ChIP–seq and RNA-seq analyses, and G.L., Z.K. and F.L. conducted the scRNA-seq analysis. O.N.D. and C.G. provided methodological assistance, and D.V.B. secured funding, designed and supervised all the experiments, analysed the data and wrote the final version of the paper with input from all authors.

Corresponding author

Correspondence to Dmitry V. Bulavin.

Ethics declarations

Competing interests

B.B.G. and D.V.B. filed a patent application (PCT/EP2023/059341) related to this study. The other authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks Vera Gorbunova, Jichang Wang 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 Molecular aspects of p16 activation during in vitro and in vivo reprogramming.

a) Colocalization of p16High (EGFP +) cells with markers of DNA damage response (pATM, pKu80, pRad51) and cell cycle inhibitors p27 and p57. Positive controls for pATM, pKu80, pRad51, p27 derived by treatment DF with doxorubicin (500 nM, 24 h). Mouse placenta serves as a positive control for p57. Scale bars – 50 μm. b) Teratomas emerged during in vivo reprogramming in p16Cre;i4F;rtTA;mTmG mice produce all three germ layers. H&E staining shows the presence of Car, cartilage (mesoderm); ET, endodermal tube (endoderm); NR, neural rosette (ectoderm). Scale bar represents 100 μm. c) Immunofluorescent analysis of teratomas, obtained by in vivo reprogramming of p16Cre;i4F;rtTA;mTmG mouse co-stained for p16High and mesenchymal (Desmin, a-SMA), endothelial (Cd31), macrophagic (F4/80) and ectodermal (GFAP) markers. Scale bars – 50 μm. d) Immunofluorescent analysis of teratomas derived by grafting feeder-free p16Cre;i4F;rtTA;mTmg iPS cells into NSG mouse for p16High cells and mesenchymal markers Desmin and α-SMA, vascular endothelial marker Cd31, macrophage marker F4/80 and neuroectodermal marker Gfap. Scale bar – 50 μm.

Extended Data Fig. 2 Ablation of p16High senescent cells during in vivo reprogramming and distribution of Line1 and Mervl retroelements in mouse ES cells.

a) p16High cells depletion during in vivo reprogramming produce teratomas with all three germ layers. H&E staining of teratomas obtained from young (2-3-month-old) mice consists of derivatives of three germ layers: endoderm (endodermal tubes, ET); ectoderm (squamos epithelium, SE); mesoderm (bone nidus, BN). Scale bars – 100 μm. b) Immunofluorescent analysis of IOUD2 mouse ES cells for Mervl and Line-1 expression. Arrows indicate double positive cells. Scale bars – 20 μm.

Extended Data Fig. 3 Treatment of already established iPS cell line with D + Q does not expand cellular plasticity.

a) In vivo reprograming was induced with DOX in drinking water and 3 weeks later mice were administered with D + Q by gavaging or mock-treated. Analysis of p16High cells in developed teratoma showed a strong reduction in mice treated with D + Q. Scale bar - 200 μm. b) Analysis of macrophage infiltration in teratoma of a control mTmG mouse and mouse received D + Q. Scale bars 50 μm. c) H&E staining of teratoma obtained from mouse treated with D + Q. Mesoderm: smooth muscles, SM; ectoderm: neural rosette, NR; endoderm: endodermal tubes, ET. d) qPCR analysis of line-1 retroelement (total and two subclasses: md_a and md_t) and the 2C gene, zfp352, in control iPS cells and control iPS cells treated with D + Q after iPS line was established. Error bars represent SD, p-value evaluated by unpaired, two-tailed Student’s t-test with Welch’s correction, independent repeats, n = 3.

Source data

Extended Data Fig. 4 Deep sequencing reveals methylation pattern during iPS reprogramming in absence of p16High senescent cells.

a) Example genome browser view of H3K4me3 levels in fibroblasts on d12 of reprogramming. b) Unsupervised hierarchical clustering of H3K4me3 levels at peaks with differential coverage between samples (relative standard deviation between samples ≥0.5; clustering based on Ward’s minimum variance). c) Distributions of promoter H3K4me3 levels at fibroblast-specific genes, in fibroblasts before reprogramming (‘mTmG’ & ‘DTA’) and at d9 of reprogramming. Decrease in high-H3K4me3 promoters associated with reprogramming (‘mTmG’ vs ‘mTmG+dox’): p = 8.4 × 10−5, (‘DTA’ vs ‘DTA+dox’): p = 8.4 × 10—9; enhancement from DQ treatment (‘mTmG+DQ+dox’ vs ‘mTmG+dox’): p = 7.1 × 10−8; enhancement in DTA cells (‘DTA+dox’ vs ‘mTmG+dox’): p = 3.5 × 10−2 (Binomial test)). d) Example genome browser view of H3K27me3 levels in fibroblasts on d12 of reprogramming. e) Genome-wide extent of changes to H3K27me3 levels. Areas of pies correspond to the fraction of the genome exhibiting the indicated changes; blue/white slices depict the fraction among these that is unchanged (blue) or augmented (white) by depletion of senescent cells by DQ treatment. f) Genome-wide distribution of relative H3K27me3 levels in 10 kb bins, in normal fibroblasts (mTmG; x-axis) or in fibroblasts on d12 of reprogramming (mTmG+dox; y-axis). The black dotted line indictes the mean H3K27me3 level in during reprogramming corresponding to each level of coverage before reprogramming. g) Heatmap indicating the augmentation of H3K37me3 hypermethylation imparted by D + Q treatment during reprogramming (concordant results were obtained using DTA fibroblasts), in genomic regions as defined in (F): note that removal of senescent cells selectively promotes augmented H3K27me3 methylation at regions with low coverage in normal fibroblasts, and that already undergo increases in H3K27me3 during normal reprogramming. h) Example genome browser view of H3K27me3 (top tracks) and H3K4me3 (bottom tracks) levels in iPS cells. Red arrow highlights the H3K4me3 peak at the promoter of the Dppa3 (Stella) gene. For A-H the integration of all examined independent biological replicates (n = 3) examined. The accession number for genome-wide date is PRJNA891955. i) qPCR analysis of xkr9 and ripply2 mRNA in control and D + Q-treated iPS cells. p-value evaluated by unpaired, two-tailed Student’s t-test with Welch’s correction, independent repeats, n = 3.

Source data

Extended Data Fig. 5 iPS cells derived in senescence-free conditions exhibit totipotent-like features.

Analysis of Nanog and Gata6 protein content in blastoids, derived from a single cell of respective iPS cell line (Ctrl (mTmG), D + Q or DTA). Scale bar – 50 μm. b) Uncropped images of immunofluorescent analysis of 12 dpc placentas of chimeric embryo derived by aggregation of D + Q iPS with 8-cell morulas (related to Fig. 5d). Yellow boxes represent magnified part of images presented in Fig. 5d. Scale bars – 50 μm. c) Immunofluorescent analysis of 12 dpc placenta of chimeric embryo derived by aggregation of GFPpositive DTA iPS cells with 8-cell morulas. Scale bars - 50 μm. d) Violin plots showing the quality control results for single RNA-Seq analysis. e) Cell clusters in the 12.5dpc placental cells visualized by UMAP (left). Feature plots showing the expression of GFP for each cell cluster (right). f) Heatmap showing the DEGs (top 5 genes) for each cell cluster of GFP+ D + Q iPS-derived cells. Data plotted in graphs D-F is integration of all examined independent biological replicates (n = 3) examined. The accession number for scRNA-Seq data is CRA010829.

Extended Data Fig. 6 Maintenance of high SAM during iPS reprogramming level is crucial for the expansion of cellular plasticity to totipotent-like stage.

a) Analysis of SAM levels in control, D + Q, NNMTi and DTA iPS cells by immunofluorescent approach. Scale bar represents 50 μm. b) Quantification of SAM levels by ELISA during HEK293 cells after transfecting control and 2 different SAM sensor plasmids. c) Efficiency (mean ± SD) of mTmG iPS reprogramming in the presence of 5 mM cycloleucine, independent repeats, n = 3. d) Analysis of p16High cells (mean ± SD) on d12 and d15 of reprogramming in the population of control and SAM sensor 5.1-infected DF, n = 3. e) qPCR analysis of pluripotency genes oct4 and nanog in mTmG and DTA iPS lined established in the presence of cycloleucine, independent repeats, n = 3. f) Reprogramming efficiency of control (untreated) and treated with 10 μm SAM. Data represent as mean ± SD, independent repeats, n = 5. g) qPCR analysis of mervl retroelement activity and inducated 2 C markers in control iPS cells and iPS cells derived in presence of 10 μm SAM. Data represent as mean ± SD, independent repeats, n = 3. h) Reprogramming efficiency of control (untreated) and treated with Nnmt inhibitor (1 μm) JBSNF-000088 DF. Data plotted as mean ± SD, independent repeats, n = 3. i) Analysis of mervl, line-1 retroelement activity and 2 C marker tcstv3 of WT iPS cells, treated with NNMTi for 2 passages. Data plotted as mean ± SD, independent repeats, n = 3. j, l) Heatmaps of relative H3K27me3 levels at 10 kb genomic intervals (J) or at peaks (L) (rows) with differential coverage between samples (columns), with the major clusters (C1- C4) of similarly-behaving peaks indicated in each case. Heatmaps are normalized by rows. k) Unsupervised hierarchical clustering of H3K27me3 levels at 10 kb genomic intervals with differential coverage between samples. m) Distributions of H3K4me3 levels at gene promoters marked by H3K4me3 in fibroblasts before reprogramming. Decrease in high-H3K4me3 promoters associated with repgrogramming (‘mTmG’ vs ‘mTmG+dox’): p = 7.6 × 10−6; enhancement from DQ treatment (‘mTmG+DQ+dox’ vs ‘mTmG+dox’): p = 8.4 × 10−4; enhancement from NNMTi-treatment (‘mTmG+DQ+dox’ vs ‘mTmG+dox’): p = 3.2 × 10−4 (Binomial test)). For J-M the integration of all examined independent biological replicates (n = 3) examined.

Source data

Extended Data Fig. 7 Removal of p16High cells improves liver plasticity during partial 4F reprogramming in aging mice.

a) A scheme for 4F-induced rejuvenation experiments with different mouse strains as indicated. b) Immunofluorescent analysis of DNA damage marker 53bp1 in the liver of 2-month-old mouse (mTmG) and 18-month-old mice: p16Cre;mTmG (mTmG), p16Cre;DTA (DTA), p16Cre;i4F;mTmG (i4F), p16Cre;i4F;DTA (DTA+i4F). Quantification is shown in the graph ( ± SD). Scale bar – 20 μm. c) Immunofluorescent staining for endothelial marker Cd146 in liver samples as in (B). Percentage of the area covered by Cd146+ cells is shown in the graph. Scale bar – 100 μm. d) Immunofluorescent analysis for fibrosis marker Desmin in liver samples as in (B). Scale bar – 100 μm. For panels B-D p values estimated by Brown-Forsythe and Welch ANOVA test with Dunnett multiple comparison test and only difference between i4F and DTA+i4F is shown. Data presented as mean ± SD, independent repeats, n = 3.

Source data

Extended Data Fig. 8 A graphical abstract for the role of p16High senescent cells in reducing SAM availability during iPS reprogramming in vitro and in vivo.

p16High senescent cells induce expression of a SAM-consuming enzymes such as NNMT, lowering the level of SAM. This in turn reduces cellular plasticity during iPS reprogramming as cells no longer can achieve a totipotent-like state. Similarly, in vivo partial 4F reprogramming in mice significantly rejuvenates old livers only in combination with simultaneous removal of p16High senescent cells.

Extended Data Fig. 9 Example of gating strategy for the FACS of GFP+ cells isolated from mTmG mouse.

The general strategy for FACS-sorting of p16High cells is shown.

Supplementary information

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. 6

Statistical source data.

Source Data Fig. 7

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. 6

Statistical source data.

Source Data Extended Data Fig. 7

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

Grigorash, B.B., van Essen, D., Liang, G. et al. p16High senescence restricts cellular plasticity during somatic cell reprogramming. Nat Cell Biol 25, 1265–1278 (2023). https://doi.org/10.1038/s41556-023-01214-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-023-01214-9

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