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:

MYCL-mediated reprogramming expands pancreatic insulin-producing cells

Abstract

β cells have a limited capacity for regeneration, which predisposes towards diabetes. Here, we show that, of the MYC family members, Mycl plays a key role in proliferation of pancreatic endocrine cells. Genetic ablation of Mycl causes a reduction in the proliferation of pancreatic endocrine cells in neonatal mice. By contrast, the expression of Mycl in adult mice stimulates the proliferation of β and α cells, and the cells persist after withdrawal of Mycl expression. A subset of the expanded α cells give rise to insulin-producing cells after this withdrawal. Transient Mycl expression in vivo is sufficient to normalize the hyperglycaemia of diabetic mice. In vitro expression of Mycl similarly provokes active replication in islet cells, even in those from aged mice. Finally, we show that MYCL stimulates the division of human adult cadaveric islet cells. Our results demonstrate that the induction of Mycl alone expands the functional β-cell population, which may provide a regenerative strategy for β cells.

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: Role of Mycl in the proliferation of pancreatic endocrine cells in neonatal mice.
Fig. 2: Forced expression of Mycl causes the expansion of adult pancreatic islets.
Fig. 3: scRNA-seq analysis reveals the division of hormone-expressing islet cells.
Fig. 4: Lineage tracing and functional analysis during the Mycl-induced expansion of INS-producing cells in vivo.
Fig. 5: Mycl stimulates the division of mature hormone-expressing islet cells in vitro.
Fig. 6: MYCL induction stimulates the division of human hormone-producing islet cells.

Similar content being viewed by others

Data availability

The Gene Expression Omnibus accession numbers for the RNA-seq data and the scRNA-seq data reported in this paper are GSE178726 and GSE163947, respectively. The data that support the plots within this paper and other findings of this study are available from the corresponding author on reasonable request.

References

  1. Slack, J. M. Developmental biology of the pancreas. Development 121, 1569–1580 (1995).

    Article  CAS  PubMed  Google Scholar 

  2. Donath, M. Y. & Halban, P. A. Decreased beta-cell mass in diabetes: significance, mechanisms and therapeutic implications. Diabetologia 47, 581–589 (2004).

    Article  CAS  PubMed  Google Scholar 

  3. Cnop, M. et al. The long lifespan and low turnover of human islet beta cells estimated by mathematical modelling of lipofuscin accumulation. Diabetologia 53, 321–330 (2010).

    Article  CAS  PubMed  Google Scholar 

  4. Teta, M., Long, S. Y., Wartschow, L. M., Rankin, M. M. & Kushner, J. A. Very slow turnover of beta-cells in aged adult mice. Diabetes 54, 2557–2567 (2005).

    Article  CAS  PubMed  Google Scholar 

  5. Bonner-Weir, S., Trent, D. F. & Weir, G. C. Partial pancreatectomy in the rat and subsequent defect in glucose-induced insulin release. J. Clin. Invest. 71, 1544–1553 (1983).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Aguayo-Mazzucato, C. & Bonner-Weir, S. Pancreatic beta cell regeneration as a possible therapy for diabetes. Cell Metab. 27, 57–67 (2018).

    Article  CAS  PubMed  Google Scholar 

  7. van der Meulen, T. et al. Virgin beta cells persist throughout life at a neogenic niche within pancreatic islets. Cell Metab. 25, 911–926 e916 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Wang, D. et al. Long-term expansion of pancreatic islet organoids from resident Procr(+) progenitors. Cell 180, 1198–1211 e1119 (2020).

    Article  CAS  PubMed  Google Scholar 

  9. Zhou, Q. & Melton, D. A. Pancreas regeneration. Nature 557, 351–358 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wang, P. et al. Diabetes mellitus–advances and challenges in human beta-cell proliferation. Nat. Rev. Endocrinol. 11, 201–212 (2015).

    Article  CAS  PubMed  Google Scholar 

  11. Laybutt, D. R. et al. Overexpression of c-Myc in beta-cells of transgenic mice causes proliferation and apoptosis, downregulation of insulin gene expression, and diabetes. Diabetes 51, 1793–1804 (2002).

    Article  CAS  PubMed  Google Scholar 

  12. Rosselot, C. et al. The many lives of Myc in the pancreatic beta-cell. J. Biol. Chem. https://doi.org/10.1074/jbc.REV120.011149 (2020).

  13. Pelengaris, S., Khan, M. & Evan, G. I. Suppression of Myc-induced apoptosis in beta cells exposes multiple oncogenic properties of Myc and triggers carcinogenic progression. Cell 109, 321–334 (2002).

    Article  CAS  PubMed  Google Scholar 

  14. Wang, P. et al. A high-throughput chemical screen reveals that harmine-mediated inhibition of DYRK1A increases human pancreatic beta cell replication. Nat. Med. 21, 383–388 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Puri, S. et al. Replication confers beta cell immaturity. Nat. Commun. 9, 485 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Collombat, P. et al. The ectopic expression of Pax4 in the mouse pancreas converts progenitor cells into alpha and subsequently beta cells. Cell 138, 449–462 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Courtney, M. et al. The inactivation of Arx in pancreatic alpha-cells triggers their neogenesis and conversion into functional beta-like cells. PLoS Genet. 9, e1003934 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Ben-Othman, N. et al. Long-term GABA administration induces alpha cell-mediated beta-like cell neogenesis. Cell 168, 73–85 e11 (2017).

    Article  CAS  PubMed  Google Scholar 

  19. Li, J. et al. Artemisinins target GABAA receptor signaling and impair alpha cell identity. Cell 168, 86–100 e115 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. van der Meulen, T. et al. Artemether does not turn alpha cells into beta cells. Cell Metab. 27, 218–225 e214 (2018).

    Article  PubMed  CAS  Google Scholar 

  21. Ackermann, A. M., Moss, N. G. & Kaestner, K. H. GABA and artesunate do not induce pancreatic alpha-to-beta cell transdifferentiation in vivo. Cell Metab. 28, 787–792 e783 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Byrnes, L. E. et al. Lineage dynamics of murine pancreatic development at single-cell resolution. Nat. Commun. 9, 3922 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Qiu, W. L. et al. Deciphering pancreatic islet beta cell and alpha cell maturation pathways and characteristic features at the single-cell level. Cell Metab. 25, 1194–1205 e1194 (2017).

    Article  CAS  PubMed  Google Scholar 

  24. Yu, X. X. et al. Dynamics of chromatin marks and the role of JMJD3 during pancreatic endocrine cell fate commitment. Development 145, https://doi.org/10.1242/dev.163162 (2018).

  25. Lu, T. T. et al. The polycomb-dependent epigenome controls beta cell dysfunction, dedifferentiation, and diabetes. Cell Metab. 27, 1294–1308 e1297 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Hatton, K. S. et al. Expression and activity of L-Myc in normal mouse development. Mol. Cell. Biol. 16, 1794–1804 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Beard, C., Hochedlinger, K., Plath, K., Wutz, A. & Jaenisch, R. Efficient method to generate single-copy transgenic mice by site-specific integration in embryonic stem cells. Genesis 44, 23–28 (2006).

    Article  CAS  PubMed  Google Scholar 

  28. Ohnishi, K. et al. Premature termination of reprogramming in vivo leads to cancer development through altered epigenetic regulation. Cell 156, 663–677 (2014).

    Article  CAS  PubMed  Google Scholar 

  29. Rane, S. G. et al. Loss of Cdk4 expression causes insulin-deficient diabetes and Cdk4 activation results in beta-islet cell hyperplasia. Nat. Genet. 22, 44–52 (1999).

    Article  CAS  PubMed  Google Scholar 

  30. Muzumdar, M. D., Tasic, B., Miyamichi, K., Li, L. & Luo, L. A global double-fluorescent Cre reporter mouse. Genesis 45, 593–605 (2007).

    Article  CAS  PubMed  Google Scholar 

  31. Thorens, B. et al. Ins1(Cre) knock-in mice for beta cell-specific gene recombination. Diabetologia 58, 558–565 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Kim, S. J., Nian, C., Doudet, D. J. & McIntosh, C. H. Inhibition of dipeptidyl peptidase IV with sitagliptin (MK0431) prolongs islet graft survival in streptozotocin-induced diabetic mice. Diabetes 57, 1331–1339 (2008).

    Article  CAS  PubMed  Google Scholar 

  33. Zhang, N. et al. Elevated vascular endothelial growth factor production in islets improves islet graft vascularization. Diabetes 53, 963–970 (2004).

    Article  CAS  PubMed  Google Scholar 

  34. Alvarez-Dominguez, J. R. et al. Circadian entrainment triggers maturation of human in vitro islets. Cell Stem Cell 26, 108–122 e110 (2020).

    Article  CAS  PubMed  Google Scholar 

  35. Thorel, F. et al. Conversion of adult pancreatic alpha-cells to beta-cells after extreme beta-cell loss. Nature 464, 1149–1154 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Furuyama, K. et al. Diabetes relief in mice by glucose-sensing insulin-secreting human alpha-cells. Nature 567, 43–48 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Xiao, X. et al. Endogenous reprogramming of alpha cells into beta cells, induced by viral gene therapy, reverses autoimmune diabetes. Cell Stem Cell 22, 78–90 e74 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Birrer, M. J. et al. L-myc cooperates with ras to transform primary rat embryo fibroblasts. Mol. Cell. Biol. 8, 2668–2673 (1988).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Nakagawa, M., Takizawa, N., Narita, M., Ichisaka, T. & Yamanaka, S. Promotion of direct reprogramming by transformation-deficient Myc. Proc. Natl Acad. Sci. USA 107, 14152–14157 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Kim, D. W. et al. Genetic requirement for Mycl and efficacy of RNA Pol I inhibition in mouse models of small cell lung cancer. Genes Dev. 30, 1289–1299 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Weintraub, H. The MyoD family and myogenesis: redundancy, networks, and thresholds. Cell 75, 1241–1244 (1993).

    Article  CAS  PubMed  Google Scholar 

  42. Addis, R. C. & Epstein, J. A. Induced regeneration–the progress and promise of direct reprogramming for heart repair. Nat. Med. 19, 829–836 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Guo, Z. et al. In vivo direct reprogramming of reactive glial cells into functional neurons after brain injury and in an Alzheimer’s disease model. Cell Stem Cell 14, 188–202 (2014).

    Article  CAS  PubMed  Google Scholar 

  44. Zhou, Q., Brown, J., Kanarek, A., Rajagopal, J. & Melton, D. A. In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature 455, 627–632 (2008).

    Article  CAS  PubMed  Google Scholar 

  45. Talchai, C., Xuan, S., Kitamura, T., DePinho, R. A. & Accili, D. Generation of functional insulin-producing cells in the gut by Foxo1 ablation. Nat. Genet. 44, 406–412 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Luo, C. et al. Global DNA methylation remodeling during direct reprogramming of fibroblasts to neurons. eLife 8, https://doi.org/10.7554/eLife.40197 (2019).

  47. Hering, B. J. et al. Phase 3 trial of transplantation of human islets in type 1 diabetes complicated by severe hypoglycemia. Diabetes Care 39, 1230–1240 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Yagi, M. et al. Derivation of ground-state female ES cells maintaining gamete-derived DNA methylation. Nature 548, 224–227 (2017).

    Article  CAS  PubMed  Google Scholar 

  49. Kaneko, T., Sakuma, T., Yamamoto, T. & Mashimo, T. Simple knockout by electroporation of engineered endonucleases into intact rat embryos. Sci. Rep. 4, 6382 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Porat, S. et al. Control of pancreatic beta cell regeneration by glucose metabolism. Cell Metab. 13, 440–449 (2011).

    Article  CAS  PubMed  Google Scholar 

  51. Boj, S. F. et al. Organoid models of human and mouse ductal pancreatic cancer. Cell 160, 324–338 (2015).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  53. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. https://doi.org/10.14806/ej.17.1.200 (2011).

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. Yagi and K. Ito for their critical reading of the manuscript. We are grateful to Y. Yamada, R. Sakamoto, M. Kikuchi and M. Baba for technical assistance. Yasuhiro Y. and T.Y. were supported by AMED-CREST (grant no. 21gm1110004h9905), the Japan Agency for Medical Research and Development (AMED). Yasuhiro Y. was also supported in part by P-CREATE, AMED (grant no. 21cm0106203h0006) and JSPS KAKENHI (grant nos. 18H04026, 20H05384). The Institute for the Advanced Study of Human Biology (ASHBi) is supported by the World Premier International Research Center Initiative (WPI), MEXT, Japan. We thank to Single-CellGenome Information Analysis Core (SignAC) in ASHBi for the RNA sequence analysis.

Author information

Authors and Affiliations

Authors

Contributions

M.H. and Yasuhiro Y. designed and conceived the study and wrote the paper. M.H., Y.S., N.S., J.T., Yosuke Y., T.U. and M.O. generated mice, performed experiments, and generated RNA-seq and scRNA-seq libraries. H.S. performed the transmission electron microscopy analysis. S.T., M. Kato and J.N. provided technical instructions for experiments with regard to diabetes. M. Kabata, S.O. and T.Y. analysed RNA-seq and scRNA-seq data in mouse and human.

Corresponding author

Correspondence to Yasuhiro Yamada.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks Patrick A Carroll and the other, anonymous, reviewers for their contribution to the peer review of this work. Isabella Samuelson was the Primary Handling Editor.

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 Expression of MYC family genes during mouse pancreatic development.

a. scRNA-seq of E14.5 pancreatic epithelial cells. scRNA-seq data were obtained from GSE101099. b. Mycl expression in the scRNA-seq analysis of embryonic pancreatic endocrine cells at various time points. Red bars indicate the mean expression levels. scRNA-seq data were obtained from GSE115931. c. Mycl expression in the scRNA-seq analysis of β- and α-cells at various time points. Red bars indicate the mean expression levels. scRNA-seq data were obtained from GSE87375. d. ChIP-seq analysis for H3K27ac in pancreatic endocrine cells. Enrichment of H3K27ac at the Mycl locus was apparent only in E17.5 Ins1+ cells. The Chga locus is shown as a control. ChIP-seq data were obtained from GSE84324 and GSE110648. e. Generation of the Mycl knockout mouse. Genetic aberrations in two independent Mycl knockout mouse lines (nos. 56 and #949) are shown. f. In situ detection of Mycl transcripts. Lymphocytes from Mycl knockout mice (no. 949) displayed no detectable Mycl expression. Scale bars: 100 μm. g. Quantification of Ki67+ cell ratio in hormone-producing cells from P0 Mycl knockout mice (no. 949). Data are presented as means ± s.d. Numbers in parentheses are the numbers of mice analysed. Total numbers of analysed cells are: 963 INS+ and 295 GCG+ cells in +/+ mice; 1,028 INS+ and 418 GCG+ cells in +/- mice; 769 INS+ and 374 GCG+ cells in -/- mice. One-way ANOVA, followed by the Tukey–Kramer post-hoc test. h. IPGTT of Mycl knockout mice (no. 949) at 4 weeks of age. Data are presented as means ± s.d. Numbers in parentheses are the numbers of mice analysed. n.s.: not significant; two-way ANOVA, followed by the Tukey-Kramer post-hoc test.

Source data

Extended Data Fig. 2 Systemic induction of MYC family genes in mice.

a. ESCs with inducible alleles for MYC family genes. mCherry signals are detectable in ESCs after Dox treatment. Scale bars: 300 μm. b. qRT-PCR analysis of MYC family genes and mCherry in ESCs. MYC family genes and mCherry were induced by Dox treatment. Data are presented as means ± s.d. of biological triplicates. The mean expression levels in Dox ON cells were set to 1. Unpaired two-tailed t-test. c. Systemic induction of Myc expression in mice. mCherry expression was detectable in most organs after Dox administration in the drinking water for 3 days (2 mg/mL, with sucrose). Scale bars: 1 mm.

Extended Data Fig. 3 Tumour development after systemic induction of MYC family genes in mice.

a. Tumour development in Myc- and Mycn-induced mice. Four-week-old chimeric mice were treated with Dox for 4 weeks. Dashed lines indicate the areas of liver tumours. Arrows indicate intestinal tumours. Mycl-induced mice did not develop tumours in the liver, intestine, stomach, lung, or spleen. Scale bars: 500 μm, except for the intestine (200 μm). b. Incidence of liver tumours and intestinal dysplasia in mice after the induction of MYC family gene expression. Numbers in parentheses are the numbers of mice analysed. Some of Myc-induced mice were sacrificed before the termination of the experiment because of the highly morbid phenotype. c. Ki67 staining for various organs of Mycl ON mice (Fig. 2f). Eight-week-old mice were given Dox for 8 weeks. mCherry expression represents Mycl-expressing cells. Scale bars: 100 μm (Lung, Liver, and Kidney), 200 μm (Intestine). d. Quantification of the Ki67+ cell ratio in each organ of Mycl ON mice. The quantification in each image is shown. Data are presented as means ± s.d. Numbers in parentheses are the numbers of mice analysed. Total numbers of analysed cells are: 4,683 (No Dox) and 3,881 (Mycl ON) in the kidney; 3,182 (No Dox) and 2,228 (Mycl ON) in the liver; 1,966 (No Dox) and 1,390 (Mycl ON) in the lung. e. Expression of mCherry in liver tissues 48 hours after daily intraperitoneal injections of Dox (0.2 ml/day; 1.0 mg/mL) in RNA-seq analysis. f. Gene ontology (GO) term enrichments of indicated clusters in Fig. 2e. GO terms and the p-value (Fisher’s exact test) are shown. DAVID Bioinformatics Resources (v6.8) was used for the analysis. g. Left panel: RNA-seq analysis of liver tissues after induction of either Mycl or Myc for 48 hours. Heatmap showing changes in expression levels of downregulated genes by either Mycl or Myc induction (fold change>2, P<0.05 [FDR-adjusted Wald Test]). Right panel: GO term enrichments of indicated Clusters. GO terms and the p-value (Fisher’s exact test) are shown.

Extended Data Fig. 4 scRNA-seq analysis of murine islet cells after the induction of Mycl expression.

a. Histology of the pancreas in adult control, Mycl ON and Mycl ON-OFF mice. Enlarged islets on histological sections are observed in both Mycl ON and Mycl ON-OFF mice. Scale bars: 1 mm. b. Proportion of hormone-positive area in islets of adult control, Mycl ON and Mycl ON-OFF mice. The quantification in each mouse is shown. Data are presented as means ± s.d. Numbers in parentheses are the numbers of mice analysed. Total numbers of analysed islets are: 52 (control), 60 (Mycl ON), and 41 (Mycl ON-OFF) for INS and SST expression; 48 (control), 58 (Mycl ON), and 55 (Mycl ON-OFF) for GCG and PP expression. c. Expression levels and distribution of representative marker genes for each cell type on a UMAP plot in Fig. 3c. d. Dot plot showing the expression of cell cycle-related genes in islet cells. e. RT-qPCR for representative genes that exhibit the altered expression levels after Mycl induction in scRNA-seq analysis. Isolated islets from control (No Dox) and Mycl ON mice were used in this analysis. Data are presented as means ± s.d. of biological triplicates. Numbers in parentheses are the numbers of mice analysed. The mean expression level of Mycl, Stmn1, and Cdk4 in Mycl ON islets and that of Mafa in control islets was set to 1, respectively. Unpaired two-tailed t test. f. Expression levels and distribution of genes associated with immature islet cells. g. Expression levels and distribution of genes associated with common pancreatic progenitor cells.

Extended Data Fig. 5 scRNA-seq analysis of murine islet cells after the induction of Mycl expression.

a. Expression levels and distribution of representative marker genes for each cell type on a UMAP plot in Fig. 3f. b. UMAP visualization of islet cells from E12.5, E14.5, E17.5, adult control, Mycl ON and Mycl ON-OFF mice. scRNA-seq data of embryonic endocrine cells (GSE101099) were used for the analysis. c. Expression levels and distribution of genes associated with immature islet cells. d. Expression levels and distribution of proliferation-related genes.

Extended Data Fig. 6 scRNA-seq analysis of murine islet cells after the induction of Mycl expression.

Pancreatic islets isolated from Mycl-induced chimeric mice (n=2, male; Dox treatment for 8 weeks; 16 weeks of age) (Fig. 2f) were dissociated into single-cell suspensions in a TripLE solution. ESCs containing Rosa26-M2rtTA; Col1a1::tetO-Mycl-ires-mCherry with a Gcg reporter were utilised to generate the chimeric mice for the scRNA-seq analysis. Islets from the chimeric mice contained both mCherry+ Mycl-induced cells and non-induced cells. a. UMAP visualisation of 7,001 mouse islet cells from Mycl ON chimeric mice. A putative doublet cluster was removed. n=4,307 cells in replicate 1 and 2,694 cells in replicate 2. The UMAP visualisation is coloured for cluster identity, and annotated on the basis of marker gene expression. Expression levels and distribution of mCherry (Mycl) as well as representative marker genes for each cell type are shown. b. Left: Expression levels and distribution of Mki67 and Cdk4. Right: Dot plot showing the expression of cell cycle-related genes in islet cells. c. Expression levels of mature cell-related genes in each cluster. d. Dot plot showing the expression of immature cell-related genes in islet cells.

Extended Data Fig. 7 Lineage tracing analysis of islet cells after the induction of Mycl expression.

a. Schematic representation of the Gcg targeting allele and the generation of Gcg-CreERT2 ESCs. The mTmG reporter-containing ESCs were used for the targeting. b. Schematic illustration of the genomic construct containing the Mycl-inducible allele and reporter alleles for use in mice. c. Seven-week-old mice were injected with Tamoxifen (200 mg/kg) for 3 consecutive days and analysed at 8 weeks of age for reporter expression. Reporter-labelled cells exclusively expressed INS or GCG in Ins1-CreERT2; mTmG mice and Gcg-CreERT2; mTmG mice, respectively. For α-cell tracing, chimeric mice were obtained by injecting Gcg-CreERT2; mTmG ESCs into blastocysts. Scale bars: 20 μm. n=4 mice examined for each reporter. d. Schematic illustration of the experimental protocol for lineage tracing analysis during Mycl-induced expansion of islet cells in adult mice. Chimeric mice were used for α-cell tracing. e. Proportion of the islets containing mGFP+ cells in each mouse for Fig. 4b. Data are presented as means ± s.d. Numbers in parentheses are the numbers of mice analysed. f. Linage tracing analysis of mature β-cells in Mycl ON and Mycl ON-OFF islets for SST and PP expression. Scale bars: Scale bars: 20 μm. g. Linage tracing analysis of mature α-cells in Mycl ON and Mycl ON-OFF islets for SST and PP expression. Chimeric mice were used in all the α-cell tracing experiments in vivo. Scale bars: Scale bars: 20 μm. h. Quantification of SST-positive (left) and PP-positive (right) cells in mGFP+ cells. The quantification in each mouse is shown. Data are presented as means ± s.d. Numbers in parentheses are the numbers of mice analysed. Total numbers of traced mGFP+ cells are: 208 (control), 254 (Mycl ON), and 433 (Mycl ON-OFF) in mice with an Ins1 reporter for SST expression; 205 (control), 248 (Mycl ON), and 429 (Mycl ON-OFF) in mice with an Ins1 reporter for PP expression; 176 (control), 891 (Mycl ON), and 1,105 (Mycl ON-OFF) in mice with a Gcg reporter for SST expression; 203 (control), 855 (Mycl ON), and 1,274 (Mycl ON-OFF) in mice with a Gcg reporter for PP expression.

Extended Data Fig. 8 In vitro expansion of adult islet cells after the induction of Mcyl expression.

a. Lineage tracing analysis of mature α-cells in Mycl ON-OFF islets. A subset of α-cell reporter-labelled cells did not express GCG or SST. Scale bars: 50 μm (large panels), 20 μm (small panels). b. Left: Expression levels and distribution of Arx on a UMAP plot. Right: Expression levels of Arx in α-cells. Black bars in a white box indicate median expression levels. The white bars and lines represent the IQR and the lower/upper adjacent values (±1.5 IQR), respectively. Wilcoxon Rank Sum test. c. Body masses of mice during and after the induction of Mycl expression. Data are presented as means ± s.d. Numbers in parentheses indicate the numbers of mice analysed. n.s.: not significant; one-way ANOVA and Holm–Sidak’s post-hoc test. d. Islet cell clusters after the induction of Myc or Mycl expression. Expansion of mCherry+ clusters at Day 7. No Dox: Mycl-inducible islets without Dox treatment. Scale bars: 50 μm. n=10 mice were examined. e. Ki67+ and cleaved caspase 3+ cells among the INS+ cells 7 days after the induction of either Myc or Mycl commenced. No Dox: Mycl-inducible islets without Dox treatment. Scale bars: 20 μm. Quantification of the Ki67+ and cleaved caspase 3+ cell ratios, relative to the number of INS+ cells, in each mouse is shown. Data are presented as means ± s.d. Numbers in parentheses are the numbers of mice analysed. Total numbers of analysed INS+ cells are: 1,185 (No Dox), 1,001 (Myc-induced), and 1,566 (Mycl-induced) for the Ki67 analysis; 1,176 (No Dox), 1,246 (Myc-induced), and 1,168 (Mycl-induced) for the cleaved caspase 3 analysis. One-way ANOVA, followed by the Tukey-Kramer post-hoc test. f. Left: Schematic representation of the Ins1 targeting allele for the generation of the Ins1-ires-CreERT2 allele. Right: β-cell labelling in Ins1-ires-CreERT2; mTmG chimeric mice. Note that mGFP+ cells exclusively expressed INS. Scale bars: 20 μm. n=4 mice were examined. g. Schematic illustration of the protocol for lineage tracing analysis in vitro. Chimeric mice were used for in vitro lineage tracing analysis. h. Lineage tracing analysis of mature β- and α-cells. mGFP+ cells exclusively expressed the original hormone after induction of Mycl in vitro. Scale bars: 20 μm. n=4 mice examined for each tracing analysis.

Extended Data Fig. 9 In vitro expansion of functional islet cells.

a. Lineage tracing analysis of mature α-cells 7 days after transplantation into the kidney, following the Mycl-induced expansion of islet cell clusters for 7 days. n=4 mice examined. b. Repeated passage of islet cluster cells in vitro. Expanded islet clusters were passaged after dissociation into small clusters and continuously expanded after the second passage, but did not obviously do so after the fourth passage. Scale bars: 50 μm. n=3 mice examined. c. Islet cell clusters after the induction of Mycl expression in RPMI 1640 (D-glucose; 2.0 g/L) with 10% FBS in Matrigel. The expansion of mCherry+ cells is observed even in a low glucose medium. Scale bars: 300 μm. n=4 mice examined. d. Schematic illustration of the genomic construct of the lentivirus containing a Mycl-inducible system. e. Schematic illustration of the experimental protocol for lentivirus-mediated induction of Mycl expression in isolated islet cells from aged mice in vitro. f. Schematic illustration of the experimental protocol for the assessment of insulin secretion. g. Histology of the removed kidney. The transplanted cells included INS+ and SST+ cells. Scale bars: 300 μm. n=3 mice examined.

Extended Data Fig. 10 scRNA-seq analysis of human islet cells after the induction of MYCL expression.

a. Schematic illustration of an experimental protocol for the lentivirus-mediated induction of MYCL in human cadaveric islet cells in vitro. b. mCherry (MYCL)+ islet cells from donors 2 and 3, displaying staining for Ki67. Scale bars: 20 μm. Numbers of analysed cells are shown in Fig. 6e. c. UMAP visualisation of human cadaveric islet cells after the induction of MYCL, Mycl, or zsGreenDR expression. A putative doublet cluster was removed. n=816 MYCL-induced cells, 1,110 Mycl-induced cells and 962 for zsGreenDR-induced cells. d. Expression levels and distribution of mCherry for each transgene. e. Expression levels and distribution of representative marker genes for pancreatic cell types.

Supplementary information

Reporting Summary

Source Data Extended Data Fig. 1

Uncropped scans of gels in Extended Data Fig. 1e.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hirano, M., So, Y., Tsunekawa, S. et al. MYCL-mediated reprogramming expands pancreatic insulin-producing cells. Nat Metab 4, 254–268 (2022). https://doi.org/10.1038/s42255-022-00530-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-022-00530-y

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