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.

  • Letter
  • Published:

Implantation underneath the abdominal anterior rectus sheath enables effective and functional engraftment of stem-cell-derived islets

This article has been updated

Abstract

Human pluripotent stem cell-derived islets (hPSC islets) are a promising alternative to primary human islets for the treatment of insulin-deficient diabetes. We previously demonstrated the feasibility of this approach in nonhuman primates; however, the therapeutic effects of hPSC islets can be limited by the maladaptive processes at the transplantation site. Here, we demonstrate successful implantation of hPSC-derived islets in a new transplantation site in the abdomen, the subanterior rectus sheath, in eight nonhuman primates (five male and three female). In this proof-of-principle study, we find that hPSC islets survive and gradually mature after transplantation, leading to improved glycemic control in diabetic primates. Notably, C-peptide secretion responds to meal challenge from 6 weeks post-transplantation (wpt), with stimulation indices comparable to those of native islets. The average post-prandial C-peptide level reaches approximately 2.0 ng ml−1 from 8 wpt, which is five times higher than the peak value we previously obtained after portal vein infusion of hPSC islets and was associated with a decrease of glycated hemoglobin levels by 44% at 12 wpt. Although additional studies in larger cohorts involving long-term follow-up of transplants are needed, our results indicate that the subanterior rectus sheath supports functional maturation and maintenance of hPSC islets, suggesting that it warrants further exploration as a transplantation target site in the context of for hPSC-based cell-replacement therapies.

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: Evaluation of hPSC-islet grafts in three extraperitoneal transplantation sites at 1 and 4 weeks after transplantation.
Fig. 2: subanterior rectus sheath transplantation of hPSC islets resulted in improvement of glycemic control in immunosuppressed diabetic rhesus macaques.
Fig. 3: C-peptide secretion of diabetic rhesus macaques with hPSC islets transplantation under the anterior rectus sheath.
Fig. 4: Characterization of subanterior rectus sheath hPSC-islet grafts at 13 wpt.

Similar content being viewed by others

Data availability

Spatial transcriptomic sequencing data have been deposited in the CNGB Nucleotide Sequence Archive of China National GeneBank DataBase with accession number CNP0003618 (https://db.cngb.org/search/project/CNP0003618/). Other experimental data, materials or reagents are available upon request and will be released subject to a material transfer agreement. Source data are provided with this paper.

Change history

  • 16 January 2023

    In the version of the article initially published, there was a unit error in Figure 3a–d,i–l, where “ng dl1” appeared rather than “ng ml1”, as now appears in the corrected HTML and PDF versions of the article.

References

  1. Du, Y. et al. Human pluripotent stem-cell-derived islets ameliorate diabetes in non-human primates. Nat. Med. 28, 272–282 (2022).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  3. 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 

  4. Balboa, D. et al. Functional, metabolic and transcriptional maturation of human pancreatic islets derived from stem cells. Nat. Biotechnol. https://doi.org/10.1038/s41587-022-01219-z (2022).

  5. Grapin-Botton, A. & Ludwig, B. Stem cell-derived β cells go in monkeys. Cell Stem Cell 29, 500–502 (2022).

    Article  CAS  Google Scholar 

  6. Rickels, M. R. & Robertson, R. P. Pancreatic Islet transplantation in humans: recent progress and future directions. Endocr. Rev. 40, 631–668 (2019).

    Article  Google Scholar 

  7. McCall, M. & Shapiro, A. M. Update on islet transplantation. Cold Spring Harb. Perspect. Med 2, a007823 (2012).

    Article  Google Scholar 

  8. Brusko, T. M., Russ, H. A. & Stabler, C. L. Strategies for durable β cell replacement in type 1 diabetes. Science 373, 516–522 (2021).

    Article  CAS  Google Scholar 

  9. Shapiro, A. M. et al. The portal immunosuppressive storm: relevance to islet transplantation? Therapeutic Drug Monit. 27, 35–37 (2005).

    Article  Google Scholar 

  10. Cantarelli, E. & Piemonti, L. Alternative transplantation sites for pancreatic islet grafts. Curr. Diab. Rep. 11, 364–374 (2011).

    Article  Google Scholar 

  11. Cunha, G. R. & Baskin, L. Use of sub-renal capsule transplantation in developmental biology. Differ. Res. Biol. Diversity 91, 4–9 (2016).

    Article  CAS  Google Scholar 

  12. Buckingham, M. Skeletal muscle formation in vertebrates. Curr. Opin. Genet. Dev. 11, 440–448 (2001).

    Article  CAS  Google Scholar 

  13. Wigmore, P. M. & Evans, D. J. Molecular and cellular mechanisms involved in the generation of fiber diversity during myogenesis. Int. Rev. Cytol. 216, 175–232 (2002).

    Article  CAS  Google Scholar 

  14. Sevensma, K.E., Leavitt, L. & Pihl, K.D. Anatomy, Abdomen and Pelvis, Rectus Sheath. (StatPearls Publishing, 2022).

  15. Yu, M. et al. Islet transplantation in the subcutaneous space achieves long-term euglycaemia in preclinical models of type 1 diabetes. Nat. Metab. 2, 1013–1020 (2020).

    Article  CAS  Google Scholar 

  16. Bertuzzi, F., Colussi, G., Lauterio, A. & De Carlis, L. Intramuscular islet allotransplantation in type 1 diabetes mellitus. Eur. Rev. Med. Pharmacol. Sci. 22, 1731–1736 (2018).

    CAS  Google Scholar 

  17. Sakata, N. et al. Strategy for clinical setting in intramuscular and subcutaneous islet transplantation. Diabetes/Metab. Res. Rev. 30, 1–10 (2014).

    Article  Google Scholar 

  18. Rafael, E. et al. Intramuscular autotransplantation of pancreatic islets in a 7-year-old child: a 2-year follow-up. Am. J. Transplant. 8, 458–462 (2008).

    Article  CAS  Google Scholar 

  19. Guan, J. et al. Chemical reprogramming of human somatic cells to pluripotent stem cells. Nature 605, 325–331 (2022).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  21. Kaneto, H. et al. PDX-1 and MafA play a crucial role in pancreatic β-cell differentiation and maintenance of mature β-cell function. Endocr. J. 55, 235–252 (2008).

    Article  CAS  Google Scholar 

  22. Zhu, H., Yu, L., He, Y. & Wang, B. Nonhuman primate models of type 1 diabetes mellitus for islet transplantation. J. Diabetes Res. 2014, 785948 (2014).

    Article  Google Scholar 

  23. Shin, J. S. et al. Long-term control of diabetes in immunosuppressed nonhuman primates (NHP) by the transplantation of adult porcine islets. Am. J. Transplant. 15, 2837–2850 (2015).

    Article  CAS  Google Scholar 

  24. Sherwani, S. I., Khan, H. A., Ekhzaimy, A., Masood, A. & Sakharkar, M. K. Significance of HbA1c test in diagnosis and prognosis of diabetic patients. Biomark. Insights 11, 95–104 (2016).

    Article  CAS  Google Scholar 

  25. American Diabetes, A. 2. Classification and diagnosis of diabetes: standards of medical care in diabetes 2021. Diabetes Care 44, S15–S33 (2021).

    Article  Google Scholar 

  26. Bottino, R. et al. Recovery of endogenous β-cell function in nonhuman primates after chemical diabetes induction and islet transplantation. Diabetes 58, 442–447 (2009).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  28. Leighton, E., Sainsbury, C. A. & Jones, G. C. A practical review of C-peptide testing in diabetes. Diabetes Ther. 8, 475–487 (2017).

    Article  CAS  Google Scholar 

  29. Jones, A. G. & Hattersley, A. T. The clinical utility of C-peptide measurement in the care of patients with diabetes. Diabet. Med. 30, 803–817 (2013).

    Article  CAS  Google Scholar 

  30. Matsumoto, S. & Shimoda, M. Current situation of clinical islet transplantation from allogeneic toward xenogeneic. J. Diabetes 12, 733–741 (2020).

    Article  Google Scholar 

  31. Graham, M. L., Bellin, M. D., Papas, K. K., Hering, B. J. & Schuurman, H. J. Species incompatibilities in the pig-to-macaque islet xenotransplant model affect transplant outcome: a comparison with allotransplantation. Xenotransplantation 18, 328–342 (2011).

    Article  Google Scholar 

  32. Kim, J. M. et al. Induction, management, and complications of streptozotocin-induced diabetes mellitus in rhesus monkeys. Xenotransplantation 23, 472–478 (2016).

    Article  Google Scholar 

  33. Graham, M. L. et al. Refining the high-dose streptozotocin-induced diabetic non-human primate model: an evaluation of risk factors and outcomes. Exp. Biol. Med. 236, 1218–1230 (2011).

    Article  CAS  Google Scholar 

  34. Silver, B. et al. EADSG guidelines: insulin therapy in diabetes. Diabetes Ther. 9, 449–492 (2018).

    Article  CAS  Google Scholar 

  35. Chen, A. et al. Spatiotemporal transcriptomic atlas of mouse organogenesis using DNA nanoball-patterned arrays. Cell 185, 1777–1792 (2022).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by National Natural Science Foundation of China (32288102 to H.D.); National Science and Technology Innovation 2030 Major Program (2021ZD0200900 to X.P.); CAMS Innovation Fund for Medical Sciences (CIFMS,2021-I2M-1-024 to X.P.); Key R&D Program of Zhejiang (2022C03SA170260 to Y.D.). We thank J. Lou for the discussion and advice on the transplantation site for human islets. Q. Yao at the Department of Clinical Pharmacology of the First Affiliated Hospital of Kunming Medical University for therapeutic drug monitoring; B. Xie at the Key Laboratory of Organ Development and Regeneration of Zhejiang Province for confocal image acquisition. We thank J. Vaughan at the Salk Institute for Biological Studies and M. Huising at the University of California, Davis for their kind provision of the UCN3 antibody. We thank C. Wang for animal research ethics guidance and J. Cao for clinical assistance with the macaques. We thank X. Yuan for his original drawings in Extended Data Fig. 1. We thank B. Liu, L. Wang, J Guan, Y. Fu and L. Cheng for discussions in the course of the preparation of this manuscript. We thank all authors of our previous nonhuman primate study1, which laid the foundation for this study.

Author information

Authors and Affiliations

Authors

Contributions

H.D., Y.D. and X.P. supervised the project. H.D. and Y.D. conceived the experimental design. Y.D., S.S., S.L. and D.S. wrote the manuscript. Z.L., D.S., S.L., Z. L. and W.Y. performed most of the experiments on nonhuman primates. S.W. and Y.W. generated hPSC islets for transplantation. Z.L., Y.W., Y.Z. and S.L. recovered and qualified cells for transplantation. Z.L., J.Y., Y.S., Z.W., X.F., W.S. and H.L. performed the characterization of hPSC-islet grafts. S.W. and Z.S. designed the treatment plan of diabetic recipients. W.S. and Z.L. gave advice on the transplantation site for hPSC islets. Y.L. performed routine blood and biochemical tests.

Corresponding authors

Correspondence to Xiaozhong Peng, Yuanyuan Du or Hongkui Deng.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks Gordon Weir and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Christoph Schmitt, in collaboration with the Nature Metabolism team.

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 Schematic of hPSC-islet transplantation strategies.

a, hPSC islets were transplanted into three extraperitoneal sites for comparison. b, Under the guidance of ultrasound imaging, hPSC islets were transplanted into the subanterior rectus sheath using a puncture needle. c, hPSC-islet was delivered by eight injections, with four parallel injections on each side of the abdominis rectus.

Extended Data Fig. 2 Immunostaining with human cell marker and islet enriched transcription factors in hPSC-islet grafts in three extraperitoneal transplantation sites at 1 week post-transplantation.

a, Representative immunohistochemical staining of human cell marker Stem121 in human grafts in Monkey 1 and 2, which results showed the morphology and structure of residual human grafts in three tested transplantation sites at 1 wpt. Scale bars, 400 μm (Monkey 1) and 200 μm (Monkey 2); for the enlarged view (indicated with green border), 50 μm (Monkey 1) and 25 μm (Monkey 2). Similar results were obtained from five independent grafts. b-c, Representative immunofluorescence staining of C-peptide and β cell specific transcription factor PDX1 (b) or NKX2.2 (c) in human grafts in Monkey 1. Scale bar, 50 μm. Similar results were obtained from five independent grafts. d, Quantitative assessment of the expression of islet-enriched transcription factors (PDX1, NKX6.1 and NKX2.2) in human grafts in three indicated sites (n = 5 independent grafts). Data presented as mean ± SEM. Two-tailed t-test was used to determine significance between groups and P value was indicated above the bars.

Source data

Extended Data Fig. 3 Evaluation of hPSC-islet grafts in Monkey 3 and 4 at 4 weeks post-transplantation.

a, Representative immunohistochemical staining of human cell marker Stem121 in three indicated graft sites, which results showed the morphology and structure of residual human grafts in three tested transplantation sites at 4 wpt. Scale bars, 400 μm; for the enlarged view (indicated with green border), 50 μm. Similar results were obtained from five independent grafts. b, Representative immunofluorescence staining of islet hormones in subanterior rectus sheath grafts. Scale bar, 50 μm. Similar results were obtained from five independent grafts. c, Representative immunofluorescence staining of pancreatic transcription factors (TFs) and UCN3 in subanterior rectus sheath grafts in Monkey 3. Scale bar, 50 μm. Similar results were obtained from five independent grafts. d, Representative immunohistochemical staining of endothelial marker CD31 in subanterior rectus sheath grafts. Scale bars, 100 μm; for the enlarged view (indicated with green border), 25 μm. Similar results were obtained from five independent grafts.

Extended Data Fig. 4 Evaluation of hPSC-islet grafts in Monkey 3 and 4 at 4 weeks post-transplantation.

a, Representative immunofluorescence staining of pancreatic transcription factors (TFs) and UCN3 in subanterior rectus sheath grafts in Monkey 4. Scale bar, 50 μm. Similar results were obtained from five independent grafts. b, Representative immunohistochemical staining of T cell marker CD3, B cell marker CD20 and macrophage marker CD68 in subanterior rectus sheath grafts (left). Monkey spleen or liver sections were used as control. The quantitative analysis of immunological infiltration in hPSC-islet grafts (n = 10 independent grafts) (right). Data presented as mean ± SEM. Scale bars, 100 μm.

Source data

Extended Data Fig. 5 Subanterior rectus sheath transplantation of hPSC islets improved glycemic control in immunosuppressed diabetic rhesus macaques.

a-d, Daily fasting blood glucose levels of diabetic macaque recipients before STZ treatment (gray), before (brown) and after (black) subanterior rectus sheath transplantation of hPSC islets (infusion procedure conducted at day 0). e-h, Blood glucose levels of diabetic monkeys at 1 month before transplantation (Pre-Tx) and 3 months after sub-rectus sheath transplantation (9 - 12 wpt) (n = 28 independent values). i, C-peptide secretion levels of four diabetic monkeys (Monkey 5 - 8) before transplantation (Pre-Tx), 6 weeks (6 wpt) and 3 months (3 mpt) after sub-rectus sheath transplantation (n = 4 monkeys). The C-peptide secretion level of diabetic monkeys at 3 months after intraportal infusion of hPSC islets (Du et al., 3 mpt) was cited for comparison (n = 4 monkeys). Data presented as mean ± SEM. Two-tailed t-test was used to determine significance between groups and P value was indicated above the bars.

Source data

Extended Data Fig. 6 Characterization of subanterior rectus sheath hPSC-islet grafts in Monkey 6-8 at 13 wpt.

a-f, Representative immunofluorescence staining of islet hormones, pancreatic transcription factors (PDX1, NKX6.1, NKX2.2 and MAFA) and UCN3 in subanterior rectus sheath grafts. Scale bar, 50 μm. Similar results were obtained from five independent grafts. g, Representative immunohistochemical staining of endothelial marker CD31 in subanterior rectus sheath grafts. Scale bars, 100 μm; for the enlarged view (indicated with green border), 25 μm. Similar results were obtained from five independent grafts.

Extended Data Fig. 7 The in situ gene expression analysis of hPSC-islet grafts under anterior rectus sheath in Monkey 5 and 6 using Stereo-seq.

a-b, Hematoxylin and eosin (H&E) staining and immunohistochemical staining of human cell marker Stem121 indicated the human grafts in rectus abdominis tissues of Monkey 5 (a) and 6 (b). Scale bars, 400 μm. c-f, The expression of islet-enriched hormones and transcription factors (c and d) and functional genes (e and f) in rectus abdominis sections of Monkey 5 (c and e) and 6 (d and f). OXPHOS, oxidative phosphorylation. The results of spatial transcriptomics analysis were reproduced on two independent recipient macaques. Similar human grafts (Stem121 positive) could be obtained from five independent sections.

Extended Data Fig. 8 Characterization of hPSC-islet grafts of Monkey 5 and 6 at 12 weeks after sub-rectus sheath transplantation.

a-b, Representative immunofluorescence staining of PCSK1 (a) and PCSK2 (b) in subanterior rectus sheath grafts and human islets. Scale bar, 50 μm. Similar results were obtained from five independent grafts. c, Vessel density in hPSC-islet grafts and native human islets (n = 10 independent grafts). Two-tailed t-test was used to determine significance between groups and P value was indicated above the bars. d, Representative immunohistochemical staining of T cell marker CD3, B cell marker CD20 and macrophage marker CD68 in subanterior rectus sheath grafts (left). Monkey spleen or liver sections were used as control. The quantitative analysis of immunological infiltration in hPSC-islet grafts (n = 10 independent grafts) (right). Scale bars, 100 μm. Data presented as mean ± SEM. Two-tailed t-test was used without adjustments for multiple comparisons to determine significance between groups and P value was indicated above the bars.

Source data

Extended Data Fig. 9 Gross anatomy, histological and immunological analysis of native pancreas of STZ-treated recipient monkeys.

a, Gross anatomy and H&E staining of pancreas sections of recipient monkeys, with islet structures outlined in white. Scale bar, 100 μm. Similar results were obtained from five independent sections. b, C-peptide staining of pancreas sections of STZ-treated recipient monkeys. Scale bars, 200 μm; for the enlarged view (indicated with green border), 50 μm. Similar results were obtained from five independent sections. c-e, Representative immunofluorescence staining of CHGA (c), islet hormones (d) and CK19 and Proinsulin (e) in pancreas sections of STZ-treated recipient Monkeys. Scale bar, 50 μm. Similar results were obtained from five independent sections.

Extended Data Fig. 10 Ultrasound examination and postmortem examination of major organs in transplanted diabetic macaques.

Gross anatomy (a, d, g and j), H&E staining (b, e, h and k) and ultrasound examination (c, f, i and l) of major organs of Monkey 5 - 8. Scale bar, 200 μm. n.a., not available. Similar results of H&E staining were obtained from five independent sections.

Supplementary information

Supplementary Information

Supplementary Figs. 1–11 and Supplementary Tables 1–10.

Reporting Summary

Supplementary Video 1

The procedure of subanterior rectus sheath transplantation under the guidance of ultrasound.

Supplementary Data 1

Statistical Source Data for Supplementary Fig. 1

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 Extended Data Fig. 2

Statistical Source Data.

Source Data Extended Data Fig. 4

Statistical Source Data.

Source Data Extended Data Fig. 5

Statistical Source Data.

Source Data Extended Data Fig. 8

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

Liang, Z., Sun, D., Lu, S. et al. Implantation underneath the abdominal anterior rectus sheath enables effective and functional engraftment of stem-cell-derived islets. Nat Metab 5, 29–40 (2023). https://doi.org/10.1038/s42255-022-00713-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-022-00713-7

This article is cited by

Search

Quick links

Nature Briefing

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

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