Abstract

BackgroundInsulin-producing cell clusters (IPCCs) have recently been generated in vitro from adipose tissue-derived stem cells (ASCs) to circumvent islet shortage. However, it is unknown how long they can survive upon transplantation, whether they are eventually rejected by recipients, and how their long-term survival can be induced to permanently cure type 1 diabetes. IPCC graft survival is critical for their clinical application and this issue must be systematically addressed prior to their in-depth clinical trials.Methodology/Principal FindingsHere we found that IPCC grafts that differentiated from murine ASCs in vitro, unlike their freshly isolated islet counterparts, did not survive long-term in syngeneic mice, suggesting that ASC-derived IPCCs have intrinsic survival disadvantage over freshly isolated islets. Indeed, β cells retrieved from IPCC syngrafts underwent faster apoptosis than their islet counterparts. However, blocking both Fas and TNF receptor death pathways inhibited their apoptosis and restored their long-term survival in syngeneic recipients. Furthermore, blocking CD40-CD154 costimulation and Fas/TNF signaling induced long-term IPCC allograft survival in overwhelming majority of recipients. Importantly, Fas-deficient IPCC allografts exhibited certain immune privilege and enjoyed long-term survival in diabetic NOD mice in the presence of CD28/CD40 joint blockade while their islet counterparts failed to do so.Conclusions/SignificanceLong-term survival of ASC-derived IPCC syngeneic grafts requires blocking Fas and TNF death pathways, whereas blocking both death pathways and CD28/CD40 costimulation is needed for long-term IPCC allograft survival in diabetic NOD mice. Our studies have important clinical implications for treating type 1 diabetes via ASC-derived IPCC transplantation.

Highlights

  • Pancreatic islet transplantation holds much promise for the cure of type 1 diabetes as transplantation of cadaveric islets is already conducted in the clinic to treat patients with type 1 diabetes

  • Several elegant studies have shown that Insulin-producing cell clusters (IPCCs) are generated in vitro from both human and murine adipose tissue-derived stem cells (ASCs) [14,15,16,17], suggesting that ASC-derived IPCCs may circumvent worldwide shortage of donor islets and eventually provide a cure for human type 1 diabetes

  • We found that ASCderived IPCCs have the intrinsic survival disadvantage over freshly isolated islets in syngeneic recipients

Read more

Summary

Introduction

Pancreatic islet transplantation holds much promise for the cure of type 1 diabetes as transplantation of cadaveric islets is already conducted in the clinic to treat patients with type 1 diabetes. Previous studies have shown that insulin-secreting cells are generated from embryonic stem cells [1,2,3,4,5,6,7] Their application in translational medicine could be limited because of ethical and legal concerns. Insulin-producing cell clusters (IPCCs) have recently been generated in vitro from adipose tissue-derived stem cells (ASCs) to circumvent islet shortage. It is unknown how long they can survive upon transplantation, whether they are eventually rejected by recipients, and how their long-term survival can be induced to permanently cure type 1 diabetes. IPCC graft survival is critical for their clinical application and this issue must be systematically addressed prior to their in-depth clinical trials

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call