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:

Targeted pharmacological therapy restores β-cell function for diabetes remission

An Author Correction to this article was published on 14 April 2020

This article has been updated

Abstract

Dedifferentiation of insulin-secreting β cells in the islets of Langerhans has been proposed to be a major mechanism of β-cell dysfunction. Whether dedifferentiated β cells can be targeted by pharmacological intervention for diabetes remission, and ways in which this could be accomplished, are unknown as yet. Here we report the use of streptozotocin-induced diabetes to study β-cell dedifferentiation in mice. Single-cell RNA sequencing (scRNA-seq) of islets identified markers and pathways associated with β-cell dedifferentiation and dysfunction. Single and combinatorial pharmacology further show that insulin treatment triggers insulin receptor pathway activation in β cells and restores maturation and function for diabetes remission. Additional β-cell selective delivery of oestrogen by Glucagon-like peptide-1 (GLP-1–oestrogen conjugate) decreases daily insulin requirements by 60%, triggers oestrogen-specific activation of the endoplasmic-reticulum-associated protein degradation system, and further increases β-cell survival and regeneration. GLP-1–oestrogen also protects human β cells against cytokine-induced dysfunction. This study not only describes mechanisms of β-cell dedifferentiation and regeneration, but also reveals pharmacological entry points to target dedifferentiated β cells for diabetes remission.

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: GLP-1–oestrogen and PEG–insulin treatment regenerates functional β-cell mass.
Fig. 2: Sustained effects of GLP-1–oestrogen to ameliorate mSTZ diabetes in mice.
Fig. 3: GLP-1–oestrogen improves function of human micro-islets.
Fig. 4: β-cell dedifferentiation in mSTZ-diabetic mice.
Fig. 5: mSTZ-derived β cells are similar to immature β cells from embryonic or immature islets.
Fig. 6: β-cell redifferentiation upon insulin and GLP-1–oestrogen treatment.
Fig. 7: Treatment specific effects of β-cell regeneration.
Fig. 8: Origin and fate of treated endocrine cells.

Similar content being viewed by others

Data availability

Custom python scripts written for performing scRNA-seq data analysis are available in a github repository (https://github.com/theislab/pancreas-targeted_pharmacology). Versions of packages that might influence numerical results are indicated in the scripts. Raw data and gene expression matrices of scRNA-seq are deposited in GEO under the accession number GSE128565. Source data for Figs. 13 and 7 and Extended Data Figs. 15 and 10 are provided with the paper.

Change history

References

  1. Matveyenko, A. V. & Butler, P. C. Relationship between β-cell mass and diabetes onset. Diabetes Obes. Metab. 10, 23–31 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Herold, K. C. et al. An anti-CD3 antibody, teplizumab, in relatives at risk for type 1 diabetes. N. Engl. J. Med. 381, 603–613 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Harrison, L. B., Adams-Huet, B., Raskin, P. & Lingvay, I. β-cell function preservation after 3.5 years of intensive diabetes therapy. Diabetes Care 35, 1406–1412 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Chen, H.-S. et al. Beneficial effects of insulin on glycemic control and beta-cell function in newly diagnosed type 2 diabetes with severe hyperglycemia after short-term intensive insulin therapy. Diabetes Care 31, 1927–1932 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. The Diabetes Control and Complications Trial Research Group. Effect of intensive therapy on residual β-cell function in patients with type 1 diabetes in the diabetes control and complications trial: a randomized, controlled trial. Ann. Intern. Med. 128, 517–523.

  6. Weng, J. et al. Effect of intensive insulin therapy on β-cell function and glycaemic control in patients with newly diagnosed type 2 diabetes: a multicentre randomised parallel-group trial. The Lancet 371, 1753–1760 (2008).

    Article  CAS  Google Scholar 

  7. Alvarsson, M. et al. Beneficial effects of insulin versus sulphonylurea on insulin secretion and metabolic control in recently diagnosed type 2 diabetic patients. Diabetes Care 26, 2231–2237 (2003).

    Article  CAS  PubMed  Google Scholar 

  8. Rui, J. et al. β cells that resist immunological attack develop during progression of autoimmune diabetes in NOD mice. Cell Metab. 25, 727–738 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Talchai, C., Xuan, S., Lin, H. V., Sussel, L. & Accili, D. Pancreatic β cell dedifferentiation as a mechanism of diabetic β cell failure. Cell 150, 1223–1234 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cinti, F. et al. Evidence of β-cell dedifferentiation in human type 2 diabetes. J. Clin. Endocrinol. Metab. 101, 1044–1054 (2016).

    Article  CAS  PubMed  Google Scholar 

  11. Like, A. A. & Rossini, A. A. Streptozotocin-induced pancreatic insulitis: new model of diabetes mellitus. Science 193, 415–417 (1976).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Chera, S. et al. Diabetes recovery by age-dependent conversion of pancreatic δ-cells into insulin producers. Nature 514, 503–507 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Brereton, M. F. et al. Reversible changes in pancreatic islet structure and function produced by elevated blood glucose. Nat. Commun. 5, 4639 (2014).

    Article  CAS  PubMed  Google Scholar 

  15. Wang, Z., York, N. W., Nichols, C. G. & Remedi, M. S. Pancreatic β cell dedifferentiation in diabetes and redifferentiation following insulin therapy. Cell Metab. 19, 872–882 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Tiano, J. P. & Mauvais-Jarvis, F. Importance of oestrogen receptors to preserve functional β-cell mass in diabetes. Nat. Rev. Endocrinol. 8, 342–351 (2012).

    Article  CAS  PubMed  Google Scholar 

  17. Chon, S. & Gautier, J.-F. An update on the effect of incretin-based therapies on β-cell function and mass. Diabetes Metab. J. 40, 99–114 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Marso, S. P. et al. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N. Engl. J. Med. 375, 1834–1844 (2016).

    Article  CAS  PubMed  Google Scholar 

  19. Finan, B. et al. Targeted estrogen delivery reverses the metabolic syndrome. Nat. Med. 18, 1847–1856 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Clemmensen, C. et al. Emerging hormonal-based combination pharmacotherapies for the treatment of metabolic diseases. Nat. Rev. Endocrinol. 15, 90–104 (2019).

    Article  PubMed  Google Scholar 

  21. Bastidas-Ponce, A. et al. Foxa2 and Pdx1 cooperatively regulate postnatal maturation of pancreatic β-cells. Mol. Metab. 6, 524–534 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Blum, B. et al. Functional beta-cell maturation is marked by an increased glucose threshold and by expression of urocortin 3. Nat. Biotechnol. 30, 261–264 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Nishimura, W. et al. A switch from MafB to MafA expression accompanies differentiation to pancreatic beta-cells. Dev. Biol. 293, 526–539 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Bader, E. et al. Identification of proliferative and mature β-cells in the islets of Langerhans. Nature 535, 430–434 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Roscioni, S. S., Migliorini, A., Gegg, M. & Lickert, H. Impact of islet architecture on β-cell heterogeneity, plasticity and function. Nat. Rev. Endocrinol. 12, 695–709 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Ediger, B. N. et al. Islet-1 is essential for pancreatic β-cell function. Diabetes 63, 4206–4217 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Gao, T. et al. Pdx1 maintains β cell identity and function by repressing an α cell program. Cell Metab. 19, 259–271 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Gu, C. et al. Pancreatic β cells require NeuroD to achieve and maintain functional maturity. Cell Metab. 11, 298–310 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Gutiérrez, G. D. et al. Pancreatic β cell identity requires continual repression of non–β cell programs. J. Clin. Invest. 127, 244–259 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Swisa, A. et al. PAX6 maintains β cell identity by repressing genes of alternative islet cell types. J. Clin. Invest. 127, 230–243 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Taylor, B. L., Liu, F.-F. & Sander, M. Nkx6.1 is essential for maintaining the functional state of pancreatic beta cells. Cell Rep. 4, 1262–1275 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kim-Muller, J. Y. et al. Aldehyde dehydrogenase 1a3 defines a subset of failing pancreatic β cells in diabetic mice. Nat. Commun. 7, 12631 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Dahan, T. et al. Pancreatic β-cells express the fetal islet hormone gastrin in rodent and human diabetes. Diabetes 66, 426–436 (2017).

    Article  CAS  PubMed  Google Scholar 

  34. Solimena, M. et al. Systems biology of the IMIDIA biobank from organ donors and pancreatectomised patients defines a novel transcriptomic signature of islets from individuals with type 2 diabetes. Diabetologia 61, 641–657 (2018).

    Article  CAS  PubMed  Google Scholar 

  35. Camunas-Soler, J.et al. Pancreas patch-seq links physiologic dysfunction in diabetes to single-cell transcriptomic phenotypes. Preprint at bioRxiv https://doi.org/10.1101/555110 (2019).

  36. Weir, G. C. & Bonner-Weir, S. Islet β cell mass in diabetes and how it relates to function, birth, and death: islet β cell mass in diabetes. Ann. N. Y. Acad. Sci. 1281, 92–105 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Qiu, W.-L. et al. Deciphering pancreatic islet β cell and α cell maturation pathways and characteristic features at the single-cell level. Cell Metab. 25, 1194–1205.e4 (2017).

    Article  CAS  PubMed  Google Scholar 

  38. Taniguchi, C. M., Emanuelli, B. & Kahn, C. R. Critical nodes in signalling pathways: insights into insulin action. Nat. Rev. Mol. Cell Biol. 7, 85–96 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Rowlands, J., Heng, J., Newsholme, P. & Carlessi, R. Pleiotropic effects of GLP-1 and analogs on cell signaling, metabolism, and function. Front. Endocrinol. 9, 672 (2018).

    Article  Google Scholar 

  40. Segars, J. H. & Driggers, P. H. Estrogen action and cytoplasmic signaling cascades. Part I: membrane-associated signaling complexes. Trends Endocrinol. Metab. 13, 349–354 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hancock, M. L. et al. Insulin receptor associates with promoters genome-wide and regulates gene expression. Cell 177, 722–736.e22 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kulkarni, R. N. et al. Altered function of insulin receptor substrate-1–deficient mouse islets and cultured β-cell lines. J. Clin. Invest. 104, R69–R75 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ueki, K. et al. Total insulin and IGF-I resistance in pancreatic β cells causes overt diabetes. Nat. Genet. 38, 583–588 (2006).

    Article  CAS  PubMed  Google Scholar 

  44. Fonseca, S. G., Gromada, J. & Urano, F. Endoplasmic reticulum stress and pancreatic β-cell death. Trends Endocrinol. Metab. 22, 266–274 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Sims, E. K. et al. Elevations in the fasting serum proinsulin-to-C-peptide ratio precede the onset of type 1 diabetes. Diabetes Care 39, 1519–1526 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Xu, B. et al. Estrogens promote misfolded proinsulin degradation to protect insulin production and delay diabetes. Cell Rep. 24, 181–196 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Tiwari, A. et al. SDF2L1 interacts with the ER-associated degradation machinery and retards the degradation of mutant proinsulin in pancreatic β-cells. J. Cell Sci. 126, 1962–1968 (2013).

    CAS  PubMed  Google Scholar 

  48. Ho, D. V. & Chan, J. Y. Induction of Herpud1 expression by ER stress is regulated by Nrf1. FEBS Lett. 589, 615–620 (2015).

    Article  CAS  PubMed  Google Scholar 

  49. Wong, N., Morahan, G., Stathopoulos, M., Proietto, J. & Andrikopoulos, S. A novel mechanism regulating insulin secretion involving Herpud1 in mice. Diabetologia 56, 1569–1576 (2013).

    Article  CAS  PubMed  Google Scholar 

  50. Belmont, P. J. et al. Roles for endoplasmic reticulum-associated degradation and the novel endoplasmic reticulum stress response gene Derlin-3 in the ischemic heart. Circ. Res. 106, 307–316 (2010).

    Article  CAS  PubMed  Google Scholar 

  51. Zhu, D. et al. Single-cell transcriptome analysis reveals estrogen signaling coordinately augments one-carbon, polyamine, and purine synthesis in breast cancer. Cell Rep. 25, 2285–2298.e4 (2018).

    Article  CAS  PubMed  Google Scholar 

  52. Torrent, M., Chalancon, G., de Groot, N. S., Wuster, A. & Madan Babu, M. Cells alter their tRNA abundance to selectively regulate protein synthesis during stress conditions. Sci. Signal. 11, eaat6409 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Xu, G. et al. Downregulation of GLP-1 and GIP receptor expression by hyperglycemia: possible contribution to impaired incretin effects in diabetes. Diabetes 56, 1551–1558 (2007).

    Article  CAS  PubMed  Google Scholar 

  54. Fritsche, A., Stefan, N., Hardt, E., Häring, H. & Stumvoll, M. Characterisation of beta-cell dysfunction of impaired glucose tolerance: evidence for impairment of incretin-induced insulin secretion. Diabetologia 43, 852–858 (2000).

    Article  CAS  PubMed  Google Scholar 

  55. Kjems, L. L., Holst, J. J., Vølund, A. & Madsbad, S. The influence of GLP-1 on glucose-stimulated insulin secretion: effects on beta-cell sensitivity in type 2 and nondiabetic subjects. Diabetes 52, 380–386 (2003).

    Article  CAS  PubMed  Google Scholar 

  56. Jonas, J. C. et al. Chronic hyperglycemia triggers loss of pancreatic beta cell differentiation in an animal model of diabetes. J. Biol. Chem. 274, 14112–14121 (1999).

    Article  CAS  PubMed  Google Scholar 

  57. Keenan, H. A. et al. Residual insulin production and pancreatic β-cell turnover after 50 years of diabetes: Joslin Medalist Study. Diabetes 59, 2846–2853 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Lam, C. J., ChatterjeeA., ShenE., Cox, A. R. & Kushner, J. A. Low-level insulin content within abundant non-á islet endocrine cells in long-standing type 1 diabetes. Diabetes 68, 598–608 (2019).

    Article  CAS  PubMed  Google Scholar 

  59. Seiron, P. et al. Characterisation of the endocrine pancreas in type 1 diabetes: islet size is maintained but islet number is markedly reduced. J. Pathol. Clin. Res. 5, 248–255 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Waaseth, M. et al. Hormone replacement therapy use and plasma levels of sex hormones in the Norwegian Women and Cancer postgenome cohort: a cross-sectional analysis. BMC Womens Health 8, 1 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. 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 

  63. Büttner, M., Miao, Z., Wolf, F. A., Teichmann, S. A. & Theis, F. J. A test metric for assessing single-cell RNA-seq batch correction. Nat. Methods 16, 43–49 (2019).

    Article  PubMed  CAS  Google Scholar 

  64. Wolf, F. A. et al. PAGA: graph abstraction reconciles clustering with trajectory inference through a topology preserving map of single cells. Genome Biol. 20, 59 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Blondel, V. D., Guillaume, J.-L., Lambiotte, R. & Lefebvre, E. Fast unfolding of communities in large networks. J. Stat. Mech. Theory Exp. 2008, P10008 (2008).

    Article  Google Scholar 

  66. Becht, E. et al. Dimensionality reduction for visualizing single-cell data using UMAP. Nat. Biotechnol. 37, 38–44 (2018).

    Article  CAS  Google Scholar 

  67. Chiang, M.-K. & Melton, D. A. Single-cell transcript analysis of pancreas development. Dev. Cell 4, 383–393 (2003).

    Article  CAS  PubMed  Google Scholar 

  68. Katsuta, H. et al. Single pancreatic beta cells co-express multiple islet hormone genes in mice. Diabetologia 53, 128–138 (2010).

    Article  CAS  PubMed  Google Scholar 

  69. Alpert, S., Hanahan, D. & Teitelman, G. Hybrid insulin genes reveal a developmental lineage for pancreatic endocrine cells and imply a relationship with neurons. Cell 53, 295–308 (1988).

    Article  CAS  PubMed  Google Scholar 

  70. Zheng, G. X. Y. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Wolock, S. L., Lopez, R. & Klein, A. M. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 8, 281–291.e9 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Kowalczyk, M. S. et al. Single-cell RNA-seq reveals changes in cell cycle and differentiation programs upon aging of hematopoietic stem cells. Genome Res. 25, 1860–1872 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. 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 

  74. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Law, C. W., Chen, Y., Shi, W. & Smyth, G. K. voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 15, R29 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  77. Soneson, C. & Robinson, M. D. Bias, robustness and scalability in single-cell differential expression analysis. Nat. Methods 15, 255–261 (2018).

    Article  CAS  PubMed  Google Scholar 

  78. Kuleshov, M. V. et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44, W90–W97 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Haghverdi, L., Büttner, M., Wolf, F. A., Buettner, F. & Theis, F. J. Diffusion pseudotime robustly reconstructs lineage branching. Nat. Methods 13, 845–848 (2016).

    Article  CAS  PubMed  Google Scholar 

  80. Tritschler, S. et al. Concepts and limitations for learning developmental trajectories from single cell genomics. Development 146, dev170506 (2019).

    Article  PubMed  Google Scholar 

  81. Polański, K. et al. BBKNN: fast batch alignment of single cell transcriptomes. Bioinformatics 36, 964–965 (2019).

    Google Scholar 

  82. La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank L. Müller, L. Sehrer, E. Malogajski and M. Kilian from the Helmholtz Diabetes Center in Munich for excellent assistance with in vivo mouse experiments. We thank J. Jaki, C. Salinno, F. Volta, J. Beckenbauer, A. Savoca and R. Fimmen for excellent assistance with in vitro experiments. We thank C. Pyke and P. Gottrup Mortensen for providing the GLP-1R antibody. We thank V. Bergen, M. Lücken, L. Simon and D. Fischer for fruitful discussions on the computational analysis. This work was supported in part by funding to M.H.T. from the Alexander von Humboldt Foundation and the Initiative and Networking Fund of the Helmholtz Association and funding by the European Research Council (AdG HypoFlam grant no. 695054). In addition, this work was supported by funds from the Helmholtz future topic ‘Aging and Metabolic programming’, the Helmholtz Alliance ICEMED, the Helmholtz Initiative on Personalised Medicine, iMed, by the Helmholtz Association, and the Helmholtz cross-programme topic ‘Metabolic Dysfunction’. M.A.S.-G. was funded by a Marie Sklodowska-Curie individual Fellowship (grant no. 706965, GCG-T3 Dyslipidemia). F.J.T. acknowledges support by the BMBF (grant no. 01IS18036A and grant no. 01IS18053A), by the German Research Foundation (DFG) within the Collaborative Research Centre 1243, Subproject A17, by the Helmholtz Association (Incubator grant sparse2big, grant no. ZT-I-0007) and by the Chan Zuckerberg Initiative DAF (advised fund of Silicon Valley Community Foundation, 182835).

Author information

Authors and Affiliations

Authors

Contributions

S.S. performed in vivo and ex vivo rodent experiments, pancreas histology, analysed and interpreted all data, interpreted scRNA-seq data, and wrote the manuscript. A.B-P. performed ex vivo rodent experiments, pancreas histology, analysed and interpreted data, and cowrote the manuscript. S.T. analysed and interpreted scRNA-seq data and cowrote the manuscript. M.B. performed ex vivo experiments and helped to draft the manuscript. A.B. performed ex vivo experiments and helped to prepare the single-cell suspensions for scRNA-seq. M.A.S.-G. performed in vivo experiments. M.T-M. performed ex vivo experiments. M.K., K.F., S.J, and A.H. performed in vivo experiments and helped to interpret data. E.B. and S.R. performed ex vivo experiments and helped to interpret data. S.U. helped to interpret data. A.F. conducted and analysed automatic pancreatic histology. B.Y. and A.N. performed, analysed, and interpreted human micro-islet experiments. C.B.J. designed, analysed, interpreted and supervised the rat study, interpreted in vivo data and helped to write the manuscript. M.C. designed and oversaw human micro-islet experiments and helped to interpret data. B.Y. synthesised and characterised compounds. B.F. designed the in vivo rodent experiment, synthesised and characterised compounds, interpreted the data, and helped to write the manuscript. R.D.D. and M.H.T conceptualised and interpreted all studies and helped to write the manuscript. F.J.T. conceptualised, supervised, and interpreted the scRNA-seq analysis and helped to write the manuscript. S.M.H, T.D.M, and H.L. conceptualised, designed, supervised, and interpreted all studies and wrote the manuscript.

Corresponding authors

Correspondence to Fabian J. Theis, Susanna M. Hofmann, Timo D. Müller or Heiko Lickert.

Ethics declarations

Competing interests

C.B.J., M.C., B.Y, B.F., and R.D.D. are current employees of Novo Nordisk. Novo Nordisk has licensed from Indiana University intellectual property pertaining to this report. M.H.T. serves as a scientific advisory board member of ERX Pharmaceuticals, Inc., Cambridge, MA. F.J.T. reports receiving consulting fees from Roche Diagnostics GmbH and Cellarity Inc., and ownership interest in Cellarity, Inc. and Dermagnostix. S.T. reports receiving consulting fees from Cellarity, Inc. The Institute for Diabetes and Obesity receives research support from Novo Nordisk. All other authors declare no conflict of interest.

Additional information

Peer review information Primary Handling Editor: Elena Bellafante.

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

Extended data

Extended Data Fig. 1 Remaining β cells lose cell identity 10 days after last STZ injection.

Effects of either vehicle or the mSTZ treatment on a, fasting blood glucose (No STZ: n = 20, mSTZ: n = 107; unpaired two-sided t-test; t = 14.64, df = 125), b, pancreatic islets histology (No STZ: 179 islets of n = 3 mice, mSTZ: 182, n = 3; unpaired two-sided t-test; β: t = 11.44, df = 358; α: t = 10.98, df = 356; δ: t = 4.27, df = 338; images are representative from no STZ n = 3 and mSTZ n = 3 mice), c, the insulin positive area within pancreatic sections (No STZ: 27 sections of n = 3 mice; STZ: 27, n = 3; unpaired two-sided t- test; t = 3.646, df = 52), d, the proliferation (No STZ: 58 islets of n = 3 mice; STZ: 69, n = 3) and e, apoptosis rate in β cells (No STZ: 46 islets of n = 3 mice; STZ: 42, n = 3; unpaired two-sided t-test, t = 3.955, df = 86), f, the expression of β-cell functional marker Ucn3 and Glut2 (images are representative of dataset plotted in b from no STZ n = 3 and mSTZ n = 3 mice), g, the homoeostatic model assessment of β-cell function (HOMA-β) (No STZ: n = 20, STZ: n = 107; unpaired two-sided t-test; t = 20.65, df = 124) and h, the ratio of fasting C-peptide to fasting blood glucose (No STZ: n = 20, STZ: n = 106; unpaired two-sided t-test; t = 14.03, df = 122). Boxplots covering all data points are depicted. Line indicates the median. Scale bar, 50 μm. Scale bar zoom-in, 20 µm.

Source data

Extended Data Fig. 2 Benefits of polypharmcotherapy to ameliorate mSTZ diabetes in mice.

Effect of treatment with indicated compounds and doses on a, fasting plasma insulin levels at week 12 of treatment (mSTZ-vehicle, n = 12; oestrogen, n = 10; GLP-1, n = 11; GLP-1/oestrogen, n = 11; one-way ANOVA with Tukey post-hoc; F (3, 39) = 10.66) and b, body weight in the end of the study (no STZ-vehicle, n = 12; mSTZ-vehicle, n = 13; GLP-1, n = 11; oestrogen, n = 11; GLP-1/oestrogen, n = 11; PEG-insulin, n = 9; GLP-1/oestrogen and PEG-insulin, n = 10; unpaired two-sided t-test; t = 2.436, df = 17). (c, d) Comparison of PEG-insulin and GLP-1/oestrogen plus PEG-insulin co-treated mice. c, Blood glucose after intraperitoneal glucose (0.5 g/kg) at week 12 (no STZ-vehicle, n = 12; PEG-insulin, n = 9; GLP-1/oestrogen and PEG-insulin, n = 10; one-way ANOVA with Tukey post-hoc (F (2, 27) = 24.71)). d, Pancreatic insulin content in the end of the study (no STZ-vehicle, n = 4; PEG-insulin, n = 4; GLP-1/oestrogen and PEG-insulin, n = 4; unpaired two-sided t-test; t = 4.534, df = 6). All data are mean ± SEM.

Source data

Extended Data Fig. 3 Tissue specificity and β-cell selectivity of the GLP-1/oestrogen conjugate.

(a, b) Treatment of female OVX Sprague-Dawley rats. a, Study scheme. b, Dry uterus weight. Data are mean ± SEM. N = 8 female rats per group. One-way ANOVA with Tukey post-hoc (F (4, 34) = 44.89). (c-g) Treatment of male FVFPBFDHom mice. (c) FACS gating strategy of dispersed endocrine cells based on granularity (Side Scatter Cell (SSC)) and PBF (405 nm) intensity. d, qPCR analysis confirmed sorting strategy of endocrine cells. Data are values of sorted cells from n = 2 mice. e, Study scheme; FVFPBFDHom male mice were treated with vehicle (n = 7), oestrogen (n = 5), GLP-1 (n = 9), or GLP-1/oestrogen (n = 11) at the indicated doses for four weeks. f, Fasting blood glucose. Data are mean ± SEM. g, Sorted endocrine cell populations after treatment (vehicle (n = 4, cells of n = 2 mice each were pooled), oestrogen (n = 4, cells of n = 2 mice each were pooled), GLP-1 (n = 5, cells of n = 2 and n = 3 mice were pooled), or GLP-1/oestrogen (n = 6, islets of n = 3 mice each were pooled)). Data are mean ± SD.

Source data

Extended Data Fig. 4 Viability and cell death of human micro-islets.

Measurement of human micro-islet viability and cell death with and without cytokine exposure and in the present of different compounds at the indicated doses. (a) ATP content and (b) Caspase 3/7 activity of human micro-islets. a, N = 6 micro-islets of n = 3 human donors for each condition. Boxplot of all data points. Line indicates the median. #P indicates P-value to vehicle no stress condition. *P indicates P-value to vehicle cytokine exposure. No stress versus cytokines stress by unpaired two-sided t-test (t = 1.756, df = 33). Otherwise one-way ANOVA with donor as random effect followed by Tukey post-hoc (Flow dose(4, 81) = 6.68; Fmedium dose(4, 82) = 10.68; Fhigh dose(4, 81) = 6.30). b, N = 3–4 micro-islets of n = 3 human donors for each condition. Boxplot of all data points. Line indicates the median. #P indicates P-value to vehicle no stress condition. *P indicates P-value to vehicle cytokine exposure. No stress versus cytokines stress by unpaired two-sided t-test (t = 2.567, df = 22). Otherwise one-way ANOVA with donor as random effect followed by Tukey post-hoc (Flow dose(4, 52) = 5.58; Fmedium dose(4, 52) = 4.44; Fhigh dose(4,53) = 4.23).

Source data

Extended Data Fig. 5 Physiological characteristics of mice used for scRNA-seq.

Representative mice (n = 3) of each treatment were used for scRNA-seq. a, Fasting glucose levels. One-way ANOVA with Tukey post-hoc test among mSTZ, oestrogen, GLP-1 and GLP- 1/oestrogen treated mice (F (3, 8) = 21.23). One-way ANOVA with Tukey post-hoc among no STZ, GLP-1/oestrogen, PEG-insulin, and co-treated mice (F (3, 8) = 94.06). b, Fasting C-peptide levels. One-way ANOVA with Tukey post-hoc test among STZ, oestrogen, GLP-1 and GLP- 1/oestrogen treated mice (F (3, 8) = 9.073). All data are mean ± SEM.

Source data

Extended Data Fig. 6 β-cell heterogeneity in healthy mice.

a, Endocrine cell annotation is based on the hormone expression of insulin (Ins), glucagon (Gcg), somatostatin (Sst), and pancreatic polypeptide (PP). b, UMAP plot showing all endocrine cells (7578 cells in total) from healthy mice. The cell number and proportion of each endocrine cluster is indicated. c, Redefined clustering of the Ins+ β cells revealed two main β-cell subpopulations. d, Expression changes of genes from selected pathways along a pseudotime trajectory from β2- to β1 cells. β1-cells were downsampled to 1000 cells for better visualisation. e, GO term and KEGG pathway enrichment analysis of up- (log(fold change) > 0.25) and downregulated (log(fold change) < -0.25) genes in β2- (278 cells) compared to β1 cells (5380 cells). Cells were pooled from n = 3 mice. Representative terms from Supplementary table 2 are depicted. We used limma-trend to find differentially expressed genes (M&M). Gene enrichment was done with EnrichR using Fisher’s exact test to identify regulated ontologies/pathways (M&M). f, Violin plots showing the distribution of the expression of proliferation and β-cell maturation genes suggesting an immature phenotype of cycling β-cells. Accordingly, 16/403 of the β2 cells, whereas only 2/5319 of the mature β1 cells were classified as cycling (M&M). Cells were pooled from n = 3 mice. Violin shows the distribution as a kernel density estimate fit. Points in violin interior show individual data points. Boxplot in violin interior shows median, quartile and whisker values. g, Measured proportion and expected doublet frequency of polyhormonal cell clusters. Expected doublet frequency is calculated given a doublet rate of 10% (M&M). h, Boxplot displaying the doublet score distribution of mono- and polyhormonal cell clusters. A high score indicates a high doublet probability. Cells were pooled from n = 3 mice. Boxplot shows the quartile values and extreme values. Whiskers extend to 1.5 IQRs of the lower and upper quartile. Outliers are displayed individually.

Extended Data Fig. 7 β-cell dedifferentiation in mSTZ-diabetic mice.

(ad), Volcano plots showing differential expression and its significance (-log10(adjusted p-Value), limma-trend) for each gene in (a) β-, (b) α-, (c) PP-, and (d) δ-cells from mSTZ treated versus healthy mice. Red line indicates thresholds used on significance level and gene expression change. Significantly regulated genes are highlighted in black. Genes significantly regulated in only one cell type but not the others are highlighted in blue. p-values were correct for multiple testing using BH. Cells were pooled from no STZ (n = 3) and mSTZ-vehicle (n = 3) treated mice. (e, f) GO term and KEGG pathway enrichment analysis of up- (log(fold change) > 0.25) and downregulated (log(fold change) < -0.25) genes in (e) α- and (f) δ cells in mSTZ treated versus healthy mice. We used limma-trend to find differentially expressed genes (M&M). Gene enrichment was done with EnrichR using Fisher’s exact test to identify regulated ontologies/pathways (M&M). Cells were pooled from no STZ (n = 3) and mSTZ-vehicle (n = 3) treated mice. (g, h) Comparison between dysregulated genes in mSTZ-β cells in mice with (g) data from RNA-seq of human T2D pancreata and (h) from scRNA-seq of human T1D β cells. Gene names of overlapping genes and identified dedifferentiation markers in Fig. 4e) are listed. i, Violin plots showing the distribution of the expression of endocrine developmental genes in beta cells of mSTZ treated and healthy mice. Violin shows the distribution as a kernel density estimate fit. Points in violin interior show individual data points. Boxplot in violin interior shows median, quartile and whisker values. Cells were pooled from no STZ (n = 3) and mSTZ-vehicle (n = 3) treated mice.

Extended Data Fig. 8 Common and distinct pathways of embryonic and dedifferentiated β cells.

Gene ontologies (Pvalue < 0.0001) and KEGG pathways (Pvalue < 0.05) that are commonly and specifically (a) down- and (b) upregulated in embryonic and mSTZ-derived β-cells. Representative terms from Supplementary Table 4 are depicted. Gene enrichment was done with EnrichR using Fisher’s exact test to identify regulated ontologies/pathways (M&M). Cells were pooled from mSTZ-vehicle (n = 3) treated mice.

Extended Data Fig. 9 Effects on endocrine cells of different treatments.

UMAP plot of all endocrine cells after 100 days of treatment showing endocrine cell distribution in each individual treatment. Total cell number for (a) mSTZ diabetic mice 5001, for (b) oestrogen treated mice 4889, for (c) GLP-1 treated mice 3874, for (d) GLP-1/oestrogen treated mice 5201, for (e) PEG-insulin treated mice 3217, and for (f) GLP-1/oestrogen (GLP-1/E) and PEG-insulin (PEG-ins) co-treated mice 3276. Values indicate the proportions of each cell cluster. Cells of n = 3 mice for each treatment were pooled.

Extended Data Fig. 10 β-cell maturation after compound treatment.

a, Immunohistochemical analysis of Ucn3 expression during the course of the study. Scale bar, 50 μm. Day 0: Images are representative of no STZ-vehicle (n = 3) and mSTZ-vehicle (n = 3) treated mice. Day 25: Images are representative of no STZ-vehicle (n = 3), mSTZ-vehicle (n = 3), GLP-1/oestrogen (n = 3), PEG-insulin (n = 3), and GLP-1/oestrogen and PEG-insulin (n = 3) co-treated mice. Day 100: Images are representative of no STZ-vehicle (n = 3), mSTZ-vehicle (n = 3), GLP-1/oestrogen (n = 2), PEG-insulin (n = 2), and GLP-1/oestrogen and PEG-insulin (n = 3) co-treated mice. b, Plasma proinsulin/C-peptide ration in the end of the study (no STZ-vehicle, n = 8; mSTZ-vehicle, n = 8; oestrogen, n = 6; GLP-1, n = 5; GLP-1/oestrogen, n = 6; PEG-insulin, n = 6, GLP-1/oestrogen and PEG-insulin, n = 6; one-way ANOVA with Tukey post-hoc: F(6, 36) = 8.12). Data are mean ± SEM. c, Representative staining for insulin and Sel1l after 25 days of treatment. Arrow indicates Sel1l + insulin + -cells, which were especially found in GLP-1/oestrogen and PEG-insulin co-treated mice. Sel1l + insulin–cells (arrow head) were more common in mSTZ-diabetic and PEG-insulin treated mice. Images are representative of no STZ-vehicle (n = 3), mSTZ-vehicle (n = 3), PEG-insulin (n = 3), and GLP-1/oestrogen + PEG-insulin (n = 3) co-treated mice. Scale bar, 20μm. d, Expression of selected ER stress and ERAD-associated genes by scRNA-seq at study end.

Source data

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Tables 1–5

Source data

Source Data Fig. 1

Statistical Source Data for each panel

Source Data Fig. 2

Statistical Source Data for each panel

Source Data Fig. 3

Statistical Source Data for each panel

Source Data Fig. 7

Statistical Source Data for each panel

Source Data Extended Data Fig. 1

Statistical Source Data for each panel

Source Data Extended Data Fig. 2

Statistical Source Data for each panel

Source Data Extended Data Fig. 3

Statistical Source Data for each panel

Source Data Extended Data Fig. 4

Statistical Source Data for each panel

Source Data Extended Data Fig. 5

Statistical Source Data for each panel

Source Data Extended Data Fig. 10

Statistical Source Data for each panel

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sachs, S., Bastidas-Ponce, A., Tritschler, S. et al. Targeted pharmacological therapy restores β-cell function for diabetes remission. Nat Metab 2, 192–209 (2020). https://doi.org/10.1038/s42255-020-0171-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-020-0171-3

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research