Open Access System for Information Sharing

Login Library

 

Conference
Cited 0 time in webofscience Cited 0 time in scopus
Metadata Downloads

3D Bioprinting of Insulin-Producing Cell Aggregates-Derived from Human Pluripotent Stem Cells with Pancreatic Tissue-Derived Bioink

Title
3D Bioprinting of Insulin-Producing Cell Aggregates-Derived from Human Pluripotent Stem Cells with Pancreatic Tissue-Derived Bioink
Authors
MYUNGJI, KIMDONG, GYU HWANGCHO, SEUNGYEONKIMJAEWOOKJANG, JIN AHIn kyoung ShimSong Cheol Kim
Date Issued
2020-08-28
Publisher
한국조직공학 재생의학회 학술대회
Abstract
Native pancreatic islets are clustered with diverse endocrine cells as functional units and surrounded by microvasculature networks communicating with neighboring cells for releasing hormones in response to glucose stimulation. 3D bioprinting technology has emerged as an promising strategy for the fabrication of engineered pancreatic tissue constructs because it enables implementation of complex tissue microarchitecture by precise positioning of multiple cells within functional bioink. In this study, we fabricated a pancreatic tissue construct through 3D aggregate bioprinting method using tissue-derived bioink with stem cell-derived insulin producing cells (IPC) to replicate the structural and functional features of native human pancreatic tissue. Selection of suitable bioink is a first step towards building functional 3D pancreatic tissue constructs. In previous study, we suggested pancreatic tissue derived-decellularized extracellular matrix (pdECM) as a attractive printable material to recapitulate native pancreatic cell niche. Here, we investigated representative constituents of pdECM bioink through proteomic analysis to evaluate that pdECM bioink can provide sufficient microenviromental cues. Additionally, functional gene classification regarding matrix-mediated characteristics were observed via gene ontology (GO) analysis. Collagen type VI was most abundant protein in pdECM bioink and other crucial ECM proteins for cell-matrix interactions were also enriched compared to the collagen bioink. Furthermore, we differentiated human embryonic stem cells (hESC) into IPC via four-stage protocol to generate functional human pancreatic cells. The differentiated cells we obtained at each stage were characterized by gene expression profile and flow cytometry analysis. Key markers of beta cells including PDX1, NKX6.1, and insulin were highly expressed after stage 3. After generation of IPC, printing conditions regarding the size of IPC aggregates were optimized for mimicking native pancreatic tissue geometry. In addition, we demonstrated interaction capacity between IPC aggregates with co-culture condition using human umbilical vein endothelial cells (HUVEC) and human mesenchymal stem cells (hMSC). Immunofluorescent staining results of printed constructs revealed that rapid induction of IPC aggregates networks can occur in tri-culture condition. The developed 3D human pancreatic tissue constructs will be able to broaden the application of in vitro disease models of diabetes and transplantable constructs for in vivo study.
URI
https://oasis.postech.ac.kr/handle/2014.oak/105998
Article Type
Conference
Citation
제21차 한국조직공학 재생의학회 학술대회, page. 101 - 101, 2020-08-28
Files in This Item:
There are no files associated with this item.

qr_code

  • mendeley

Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.

Related Researcher

Researcher

장진아JANG, JIN AH
Dept of Mechanical Enginrg
Read more

Views & Downloads

Browse