Select your timezone:

Poster Session

Thursday October 26, 2023 - 16:30 to 18:00

Room: Foyer Area

P.19 Nanoencapsulation with T3 loaded polymersomes improves the function of neonatal porcine islet like cell clusters

Sang Hoon Lee, Korea

MGEN Solutions

Abstract

Nanoencapsulation with T3 loaded polymersomes improves the function of neonatal porcine islet like cell clusters

Sang Hoon Lee1,2, Min Se Kim2, Eun Jin Lee1, Sun Mi Ahn1, Hyun-Ouk Kim2, Jung-Taek Kang1.

1Biotechnology Research Institute, Mgensolution, Seoul, Korea; 2Department of Biotechnology and Bioengineering, Kangwon National University, Chuncheon, Korea

Introduction: Neonatal porcine islet like cell clusters (NPCCs) can be used to as a resource to treat type Ⅰ diabetes. To overcome a xenogenic immunogenicity of the NPCCs, nanoencapsulation of NPCCs have been tried. Previously, we confirmed the effects of immune protection in rodent models through nanoencapsulation of NPCCs using polyethylene glycol-b-polylactic acid based polymersomes (PSomes). In this study, to increase binding efficiency of PSomes on NPCCs, PSome surface was modified with N-hydroxysuccinimide (NHS) and Maleimide (Mal), in which can induce dual covalent bond for immunoisolation by binding the amine and thiol groups on NPCCs, respectively. Moreover, the triiodothyronine (T3) concerned with maturation of pancreatic β cells and islets differentiation derived stem cell, was loaded in PSomes. We evaluated whether T3 loaded PSomes improve the efficiency of nano-encapsulation and functionality of NPCCs.

Methods: NPCCs were isolated from neonatal porcine pancreas using enzyme digestion method and cultured during 8 days. And then, NPCCs were nanoencapsulated with T3-PSomes. The efficiency was measured by mean fluorescence intensity (MFI) of DiI using either fluorescence microscopy or confocal laser scanning microscopy. The functional assessment of nanoencapsulated NPCCs with T3-PSomes was evaluated by GSIS assay and qRT-PCR.

Results: The mean diameter of PSomes measured by dynamic light scattering showed that PSomes encapsulate conformally the surface of NPCCs and the MFI increases time-dependently. Nanoencapsulated with T3-PSomes didn’t affects the viability of NPCCs. Although the insulin secretion at high concentrations of glucose stimulation didn’t show a significant difference in the T3-PSome group compared with control group, insulin mRNA expression and normal insulin secretion were increased significantly in T3-PSome group, compared with control group. However, there was no significant difference in stimulation index among all groups.

Conclusions: Our study will provide the improved immunoisolation strategy through efficient nanoencapsulation of NPCCs through induction of dual covalent bond and will overcome the functional immaturity by stimulating differentiation of the NPCCs through a constant release of T3 loaded in PSomes. Also, our loading system will offer opportunity to use various maturation related chemical and functional reagent. Therefore, we expect that NPCCS can be used as the optimal source for islet xenotransplantation. Further studies are needed to demonstrate the immune protection effects of our nanoencapsulated NPCCs in human immune system.

References:

[1] Giwa, A. M. et al. Current understandings of the pathogenesis of type 1 diabetes: Genetics to environment. World J. Diabetes 11, 13–25 (2020).
[2] Strand, B. L., Coron, A. E. & Skjak-Braek, G. Current and future perspectives on alginate encapsulated pancreatic islet. Stem Cells Transl. Med. 6, 1053–1058 (2017).
[3] Cooper, D. K. C., Gollackner, B. & Sachs, D. H. Will the pig solve the transplantation backlog? Annu. Rev. Med. 53, 133–147 (2002).
[4] Van Der Windt, D. J. et al. Clinical islet xenotransplantation: How close are we? Diabetes 61, 3046–3055 (2012).
[5] Lu, T., Yang, B., Wang, R. & Qin, C. Xenotransplantation: Current Status in Preclinical Research. Front. Immunol. 10, (2020).
[6] Klymiuk, N., Ludwig, B., Seissler, J., Reichart, B. & Wolf, E. Current Concepts of Using Pigs as a Source for Beta-Cell Replacement Therapy of Type 1 Diabetes. Curr. Mol. Biol. Reports 2, 73–82 (2016).
[7] Hryhorowicz, M., Zeyland, J., Słomski, R. & Lipiński, D. Genetically Modified Pigs as Organ Donors for Xenotransplantation. Mol. Biotechnol. 59, 435–444 (2017).
[8] Jimenez-Vera, E., Davies, S., Phillips, P., O’Connell, P. J. & Hawthorne, W. J. Long-term cultured neonatal islet cell clusters demonstrate better outcomes for reversal of diabetes: In vivo and molecular profiles. Xenotransplantation 22, 114–123 (2015).
[9] Kim, H. O. et al. Cell-mimic polymersome-shielded islets for long-term immune protection of neonatal porcine islet-like cell clusters. J. Mater. Chem. B 8, 2476–2482 (2020).
[10] Lee, S. H., Kim, H. O. & Kang, J. T. Optimization of Nano-encapsulation on Neonatal Porcine Islet-like Cell Clusters Using Polymersomes. Nanoscale Res. Lett. 16, (2021).
[11] Tayebeh, Anajafi, S. M. Polymersome-based drug-delivery stratergies for cancer therapeutics. Ther. Deliv. 6, 521–534 (2015).
[12] Aguayo-Mazzucato, C. et al. MAFA and T3 drive maturation of both fetal human islets and insulin-producing cells differentiated from hESC. J. Clin. Endocrinol. Metab. 100, 3651–3659 (2015).
[13] 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).
[14] Pathak, S., Pham, T. T., Jeong, J. H. & Byun, Y. Immunoisolation of pancreatic islets via thin-layer surface modification. J. Control. Release 305, 176–193 (2019).

Organized by

Supported by

Hosted by


IPITA-IXA-CTRMS Joint Congress • San Diego, CA, USA • October 26-29, 2023
© 2024 IPITA-IXA-CTRMS 2023