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.

Interspecies organogenesis generates autologous functional islets

Abstract

Islet transplantation is an established therapy for diabetes. We have previously shown that rat pancreata can be created from rat pluripotent stem cells (PSCs) in mice through interspecies blastocyst complementation. Although they were functional and composed of rat-derived cells, the resulting pancreata were of mouse size, rendering them insufficient for isolating the numbers of islets required to treat diabetes in a rat model. Here, by performing the reverse experiment, injecting mouse PSCs into Pdx-1-deficient rat blastocysts, we generated rat-sized pancreata composed of mouse-PSC-derived cells. Islets subsequently prepared from these mouse–rat chimaeric pancreata were transplanted into mice with streptozotocin-induced diabetes. The transplanted islets successfully normalized and maintained host blood glucose levels for over 370 days in the absence of immunosuppression (excluding the first 5 days after transplant). These data provide proof-of-principle evidence for the therapeutic potential of PSC-derived islets generated by blastocyst complementation in a xenogeneic host.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Generation of apancreatic rats by TALEN-mediated mutagenesis.
Figure 2: Generation of rat pancreata by blastocyst complementation.
Figure 3: Generation of mouse pancreata in rat by interspecies blastocyst complementation.
Figure 4: Transplantation of mouseR islets into mice with drug-induced diabetes.

Similar content being viewed by others

References

  1. Pagliuca, F. W. et al. Generation of functional human pancreatic β cells in vitro. Cell 159, 428–439 (2014)

    Article  CAS  Google Scholar 

  2. Yusa, K. et al. Targeted gene correction of α1-antitrypsin deficiency in induced pluripotent stem cells. Nature 478, 391–394 (2011)

    Article  CAS  ADS  Google Scholar 

  3. Shi, Y., Kirwan, P., Smith, J., Robinson, H. P. C. & Livesey, F. J. Human cerebral cortex development from pluripotent stem cells to functional excitatory synapses. Nat. Neurosci. 15, 477–486 (2012)

    Article  CAS  Google Scholar 

  4. Nakamura, S. et al. Expandable megakaryocyte cell lines enable clinically applicable generation of platelets from human induced pluripotent stem cells. Cell Stem Cell 14, 535–548 (2014)

    Article  CAS  Google Scholar 

  5. Sun, N. et al. Patient-specific induced pluripotent stem cells as a model for familial dilated cardiomyopathy. Sci. Transl. Med. 4, 130ra47 (2012)

    Article  Google Scholar 

  6. Völkner, M. et al. Retinal organoids from pluripotent stem cells efficiently recapitulate retinogenesis. Stem Cell Rep. 6, 525–538 (2016)

    Article  Google Scholar 

  7. Lancaster, M. A. et al. Cerebral organoids model human brain development and microcephaly. Nature 501, 373–379 (2013)

    Article  CAS  ADS  Google Scholar 

  8. Spence, J. R. et al. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature 470, 105–109 (2011)

    Article  ADS  Google Scholar 

  9. Takebe, T. et al. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature 499, 481–484 (2013)

    Article  CAS  ADS  Google Scholar 

  10. Takasato, M. et al. Directing human embryonic stem cell differentiation towards a renal lineage generates a self-organizing kidney. Nat. Cell Biol. 16, 118–126 (2014)

    Article  CAS  Google Scholar 

  11. Kobayashi, T. et al. Generation of rat pancreas in mouse by interspecific blastocyst injection of pluripotent stem cells. Cell 142, 787–799 (2010)

    Article  CAS  Google Scholar 

  12. Rashid, T., Kobayashi, T. & Nakauchi, H. Revisiting the flight of Icarus: making human organs from PSCs with large animal chimeras. Cell Stem Cell 15, 406–409 (2014)

    Article  CAS  Google Scholar 

  13. Shapiro, A. M. J. et al. International trial of the Edmonton protocol for islet transplantation. N. Engl. J. Med. 355, 1318–1330 (2006)

    Article  CAS  Google Scholar 

  14. Rezania, A. et al. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat. Biotechnol. 32, 1121–1133 (2014)

    Article  CAS  Google Scholar 

  15. Vegas, A. J. et al. Long-term glycemic control using polymer-encapsulated human stem cell-derived β cells in immune-competent mice. Nat. Med. 22, 306–311 (2016)

    Article  CAS  Google Scholar 

  16. Stoffers, D. A., Ferrer, J., Clarke, W. L. & Habener, J. F. Early-onset type-II diabetes mellitus (MODY4) linked to IPF1. Nat. Genet. 17, 138–139 (1997)

    Article  CAS  Google Scholar 

  17. Ahlgren, U., Jonsson, J., Jonsson, L., Simu, K. & Edlund, H. β-cell-specific inactivation of the mouse Ipf1/Pdx1 gene results in loss of the β-cell phenotype and maturity onset diabetes. Genes Dev. 12, 1763–1768 (1998)

    Article  CAS  Google Scholar 

  18. Takahashi, R., Hirabayashi, M. & Ueda, M. Production of transgenic rats using cryopreserved pronuclear-stage zygotes. Transgenic Res. 8, 397–400 (1999)

    Article  CAS  Google Scholar 

  19. Grau, J., Boch, J. & Posch, S. TALENoffer: genome-wide TALEN off-target prediction. Bioinformatics 29, 2931–2932 (2013)

    Article  CAS  Google Scholar 

  20. Oliver-Krasinski, J. M. et al. The diabetes gene Pdx1 regulates the transcriptional network of pancreatic endocrine progenitor cells in mice. J. Clin. Invest. 119, 1888–1898 (2009)

    Article  CAS  Google Scholar 

  21. Moede, T., Leibiger, B., Pour, H. G., Berggren, P. & Leibiger, I. B. Identification of a nuclear localization signal, RRMKWKK, in the homeodomain transcription factor PDX-1. FEBS Lett. 461, 229–234 (1999)

    Article  CAS  Google Scholar 

  22. Rodriguez-Diaz, R. et al. α cells secrete acetylcholine as a non-neuronal paracrine signal priming beta cell function in humans. Nat. Med. 17, 888–892 (2011)

    Article  CAS  Google Scholar 

  23. Balboa, D. & Otonkoski, T. Human pluripotent stem cell based islet models for diabetes research. Best Pract. Res. Clin. Endocrinol. Metab. 29, 899–909 (2015)

    Article  CAS  Google Scholar 

  24. Sutherland, D. E. R. et al. Islet autotransplant outcomes after total pancreatectomy: a contrast to islet allograft outcomes. Transplantation 86, 1799–1802 (2008)

    Article  Google Scholar 

  25. Bennet, W. et al. Incompatibility between human blood and isolated islets of Langerhans: a finding with implications for clinical intraportal islet transplantation? Diabetes 48, 1907–1914 (1999)

    Article  CAS  Google Scholar 

  26. Stanger, B. Z., Tanaka, A. J. & Melton, D. A. Organ size is limited by the number of embryonic progenitor cells in the pancreas but not the liver. Nature 445, 886–891 (2007)

    Article  CAS  ADS  Google Scholar 

  27. Camargo, F. D. et al. YAP1 increases organ size and expands undifferentiated progenitor cells. Curr. Biol. 17, 2054–2060 (2007)

    Article  CAS  Google Scholar 

  28. Yimlamai, D. et al. Hippo pathway activity influences liver cell fate. Cell 157, 1324–1338 (2014)

    Article  CAS  Google Scholar 

  29. Barry, E. R. et al. Restriction of intestinal stem cell expansion and the regenerative response by YAP. Nature 493, 106–110 (2013)

    Article  ADS  Google Scholar 

  30. Agulnick, A. D. et al. Insulin-producing endocrine cells differentiated in vitro from human embryonic stem cells function in macroencapsulation devices in vivo. Stem Cells Transl. Med. 4, 1214–1222 (2015)

    Article  CAS  Google Scholar 

  31. Hirabayashi, M. et al. Ability of tetraploid rat blastocysts to support fetal development after complementation with embryonic stem cells. Mol. Reprod. Dev. 79, 402–412 (2012)

    Article  CAS  Google Scholar 

  32. Yamaguchi, T. et al. Development of an all-in-one inducible lentiviral vector for gene specific analysis of reprogramming. PLoS One 7, e41007 (2012)

    Article  CAS  ADS  Google Scholar 

  33. Yamaguchi, T., Hamanaka, S. & Nakauchi, H. The generation and maintenance of rat induced pluripotent stem cells. Methods Mol. Biol. 1210, 143–150 (2014)

    Article  Google Scholar 

Download references

Acknowledgements

We thank A. Oshima and H. Tsukui for technical support, K. Okada for secretarial support, H. Nagashima and M. Watanabe for advice in preparing the manuscript, A. S. Knisely for reading of the manuscript and Japan Insulin-Dependent Diabetes Mellitus (IDDM) Network for continuous support. This work was supported by grants from Japan Science and Technology Agency, Exploratory Research for Advanced Technology, Leading Advanced Projects for medical innovation, Japan Agency for Medical Research and Development, Japan Society for the Promotion of Science, KAKENHI Grant Number 26460358, research grant for type 1 diabetes, Japan IDDM Network and California Institute for Regenerative Medicine.

Author information

Authors and Affiliations

Authors

Contributions

T.Y. and H.S. designed, performed, and analysed all experiments and wrote the manuscript. M.I.-K., T.G., H.H., M.S., T.K., A.Y., and A.U. performed embryo manipulation. N.M. performed data analysis. Y.O. performed histopathological analysis. S.H. performed establishment of iPSCs. H.M. performed data analysis. D.T.R. wrote the manuscript. M.H. performed embryo manipulation and data analysis. H.N. designed the study and wrote the manuscript.

Corresponding author

Correspondence to Hiromitsu Nakauchi.

Ethics declarations

Competing interests

H.N. is a co-founder and shareholder of iCELL Inc., ChimaERA Corporation and ReproCELL Inc.

Additional information

Reviewer Information Nature thanks H. Lickert, Q. Zhou and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Extended data figures and tables

Extended Data Figure 1 The birth rate of Pdx1mu/mu rats and phenotype of Pdx1+/mu rats.

a, Results of mating Pdx1+/muA with Pdx1+/muB. Wild-type, Pdx1+/mu and Pdx1mu/mu rats were born in the mendelian ratio (Χ2 = 2, P = 0.37 by chi-squared test). b, Results of GTTs in Pdx1+/mu. MODY-like hyper-glycaemia were observed in Pdx1+/muA (2 of 6 rats) and Pdx1+/muB (3 of 6 rats). c, Amino acid sequences of Pdx1muA, Pdx1muB and wild-type Pdx1, which are predicted from full-length cDNA derived from mRNA in duodenum of Pdx1muA/muB or Pdx1+/+ rats.

Source data

Extended Data Figure 2 Immunofluorescence photomicrographs of rES-cell-derived pancreata generated in Pdx1mu/mu chimaeric rats.

a, Pancreata sections were stained with antibodies against rat glucagon, rat insulin, rat somatostatin, rat CK19 and rat α-amylase. Scale bars, 100 μm. b, Quantitative analysis of sections of pancreata. Percentages of EGFP-expressing areas in areas that were positive for insulin, glucagon, somatostatin, CK19 or α-amylase were analysed by image J software (n = 3). The mean values ± s.d. were obtained from 3 biological replicates. c, The area under glucose curve (AUC glucose), calculated from GTT data in Fig. 2e. The mean values ± s.d. were obtained from 2 (Pdx1mu/mu + rES cells, WT chimaera), 3 (WT), or 4 (Pdx+/mu + rES cells) independent experiments (P = 0.24 Pdx1+/mu + rESCs versus Pdx1mu/mu + rESCs; P = 0.88 Pdx1mu/mu + rESCs versus WT; P = 0.08 WT chimaera versus WT; P = 0.19 WT chimaera versus Pdx1+/mu + rESCs; Student’s t-test).

Source data

Extended Data Figure 3 Immunofluorescence photomicrographs of miPSC-derived-pancreata generated in Pdx1mu/mu chimaeric rats.

a, Pancreata sections were stained with antibodies against mouse glucagon, mouse insulin, mouse somatostatin, mouse CK19 and mouse α-amylase. Scale bars, 100 μm. b, Quantitative analysis of sections of pancreata. Percentages of EGFP-expressing areas in areas that were positive for insulin, glucagon, somatostatin, CK19 or α-amylase were analysed by Image J software (n = 3). The mean values ± s.d. were obtained from 3 biological replicates. c, AUC glucose, calculated from GTT data in Fig. 3e. The mean values ± s.d. were obtained from 3 (Pdx+/mu + mPSCs; WT), or 6 (Pdxmu/mu + mPSCs) independent experiments (P = 0.20 Pdx1+/mu + mPSCs versus Pdx1mu/mu + mPSCs; P = 0.14 Pdx1mu/mu + mPSCs versus WT; Student’s t-test).

Source data

Extended Data Figure 4 Clinical biochemistry and histologic observations in Pdx1mu/mu chimaeric rat with diabetic-like symptoms.

a, GTTs results, 6 weeks and 10 weeks after birth, in Pdx1mu/mu chimaeric rats that possess mPSC-derived pancreata (chimaeras 183 and 186). Blood sampling at the same time points as in Fig. 3e. Chimaera 186 showed diabetic-like symptoms. b, Photomicrographs of pancreata of Pdx1mu/mu chimaeric rat (chimaera 186) (left) and C57BL/6N mouse pancreata (right). IL, islet of Langerhans; PD, pancreatic duct.

Source data

Extended Data Figure 5 Injection studies of pancreatic duct through common bile duct.

PBS-containing trypan blue was injected from the duodenum into the common bile duct (see diagram, bottom) of a wild-type Wistar rat (upper left, before injection; lower left, after injection), Pdx1+/mu chimaeric rat (upper middle, before injection; lower middle, after injection), and a Pdx1mu/mu chimaeric rat (upper right, before injection; lower right, after injection). PBS diffused throughout the pancreata in wild-type rat and Pdx1+/mu chimaeric rat, whereas, in the Pdx1mu/mu chimaeric rat, the PBS was retained in common bile duct and flowed backward to duodenum.

Extended Data Figure 6 Analysis of transplanted islets.

a, Bright-field (top) and fluorescent (bottom) images of hemi-nephrectomized kidney with (right) or without (left) transplanted islets. b, Transplanted islets, kidney capsule; anti-CD3 and –CD11b, hematoxylin counterstain. Islets lie inside the dotted line (Scale bars: 100 nm). c, Immunohistological analysis of engrafted islets under kidney capsule. Sections were stained with antibody against EGFP, insulin, glucagon and somatostatin and with DAPI. Scale bars, 100 nm. d, Quantitative analysis of sections of engrafted islets. Ratio of insulin-, glucagon- and somatostatin-positive cells were analysed by Image J software (n = 3). The mean values ± s.d. were obtained from 3 biological replicates (P = 0.93 insulin+ area in mouseR islets versus in WT mouse islets; P = 0.89 glucagon+ area in mouseR islets versus in WT mouse islets; P = 0.77 somatostatin+ area in mouseR islets versus in WT mouse islets; Student’s t-test). e, Top, FACS diagrams, dispersed small samples of WT rat kidney (left), WT mouse kidney (middle) and mouse kidney with transplanted islets (right). Cells were stained with fluorophore-tagged antibodies against mouse and rat CD31 (mCD31 and rCD31, respectively). Bottom, FACS diagram, EGFP expression by dispersed cells of mouse kidney with transplanted islets (right) (n = 3).

Extended Data Figure 7 Analysis of mouseR islets transplanted mice.

a, AUC glucose, calculated from GTT data in Fig. 4d. The mean values ± s.d. were obtained from 3 independent experiments (P < 0.01 A, B or C versus D, E or F, Student’s t-test). b, Nonfasting mouse c-peptide level (pmol l−1) in serum from Pdx1muA/muB + mPSCs chimaera, mouse transplanted with mouseR islets, and C57BL/6 mouse. The lowest value of the x axis represent the lowest limit of detection. The mean values ± s.d. were obtained from 3 biological replicates except for Pdx1mu/mu + mPSCs that was from 2 biological replicates (P < 0.01 Pdx1mu/mu + mPSCs chimaera and mouseR islets transplanted mouse versus STZ treated mouse; P < 0.01 STZ treated mouse versus WT mouse; Student’s t-test). c, Nonfasting rat c-peptide level (pmol l−1) in serum of Pdx1muA/muB + miPSCs chimaera, mouse transplanted with mouseR islets, C57BL/6 mouse and Wistar rat. Values are mean ± s.d. N.D., not detected. The lowest value of the x axis represent the lowest limit of detection. The mean values ± s.d. were obtained from 3 biological replicates, except for Pdx1mu/mu + mPSCs, for which they were obtained from 2 biological replicates.

Source data

Extended Data Table 1 Sequence analysis of predicted off-target sites of Pdx1 TALEN

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yamaguchi, T., Sato, H., Kato-Itoh, M. et al. Interspecies organogenesis generates autologous functional islets. Nature 542, 191–196 (2017). https://doi.org/10.1038/nature21070

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature21070

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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