Sa131 DEVELOPMENT OF A PERSONALIZED MODEL OF INTESTINAL FIBROSIS USING HUMAN INTESTINAL ORGANOIDS DERIVED FROM INDUCED PLURIPOTENT STEM CELLS

2021 ◽  
Vol 160 (6) ◽  
pp. S-431-S-432
Author(s):  
Hannah Q. Estrada ◽  
Shachi Patel ◽  
Shervin Rabizadeh ◽  
Stephan R. Targan ◽  
Robert J. Barrett
Author(s):  
Hannah Q Estrada ◽  
Shachi Patel ◽  
Shervin Rabizadeh ◽  
David Casero ◽  
Stephan R Targan ◽  
...  

Abstract Background Intestinal fibrosis is a serious complication of Crohn’s disease. Numerous cell types including intestinal epithelial and mesenchymal cells are implicated in this process, yet studies are hampered by the lack of personalized in vitro models. Human intestinal organoids (HIOs) derived from induced pluripotent stem cells (iPSCs) contain these cell types, and our goal was to determine the feasibility of utilizing these to develop a personalized intestinal fibrosis model. Methods iPSCs from 2 control individuals and 2 very early onset inflammatory bowel disease patients with stricturing complications were obtained and directed to form HIOs. Purified populations of epithelial and mesenchymal cells were derived from HIOs, and both types were treated with the profibrogenic cytokine transforming growth factor β (TGFβ). Quantitative polymerase chain reaction and RNA sequencing analysis were used to assay their responses. Results In iPSC-derived mesenchymal cells, there was a significant increase in the expression of profibrotic genes (Col1a1, Col5a1, and TIMP1) in response to TGFβ. RNA sequencing analysis identified further profibrotic genes and demonstrated differential responses to this cytokine in each of the 4 lines. Increases in profibrotic gene expression (Col1a1, FN, TIMP1) along with genes associated with epithelial-mesenchymal transition (vimentin and N-cadherin) were observed in TGFβ -treated epithelial cells. Conclusions We demonstrate the feasibility of utilizing iPSC-HIO technology to model intestinal fibrotic responses in vitro. This now permits the generation of near unlimited quantities of patient-specific cells that could be used to reveal cell- and environmental-specific mechanisms underpinning intestinal fibrosis.


2015 ◽  
Vol 83 (7) ◽  
pp. 2926-2934 ◽  
Author(s):  
Jessica L. Forbester ◽  
David Goulding ◽  
Ludovic Vallier ◽  
Nicholas Hannan ◽  
Christine Hale ◽  
...  

The intestinal mucosa forms the first line of defense against infections mediated by enteric pathogens such as salmonellae. Here we exploited intestinal “organoids” (iHOs) generated from human induced pluripotent stem cells (hIPSCs) to explore the interaction ofSalmonella entericaserovar Typhimurium with iHOs. Imaging and RNA sequencing were used to analyze these interactions, and clear changes in transcriptional signatures were detected, including altered patterns of cytokine expression after the exposure of iHOs to bacteria.S. Typhimurium microinjected into the lumen of iHOs was able to invade the epithelial barrier, with many bacteria residing withinSalmonella-containing vacuoles. AnS. TyphimuriuminvAmutant defective in theSalmonellapathogenicity island 1 invasion apparatus was less capable of invading the iHO epithelium. Hence, we provide evidence that hIPSC-derived organoids are a promising model of the intestinal epithelium for assessing interactions with enteric pathogens.


2017 ◽  
Vol 32 (1) ◽  
pp. 111-122 ◽  
Author(s):  
Kwang Bo Jung ◽  
Hana Lee ◽  
Ye Seul Son ◽  
Ji Hye Lee ◽  
Hyun‐Soo Cho ◽  
...  

2021 ◽  
Vol 27 (Supplement_1) ◽  
pp. S35-S35
Author(s):  
Hannah Estrada ◽  
Shachi Patel ◽  
Shervin Rabizadeh ◽  
Stephan Targan ◽  
Robert Barrett

Abstract Background Intestinal fibrosis is a serious complication of inflammatory bowel disease (IBD) with > 20% of Crohn’s disease patients developing this complication within 10 years of diagnosis. Despite improvements in anti-inflammatory medication, its incidence remains stubbornly high and thus far surgical intervention remains the only treatment option. Numerous cell types including intestinal epithelial and mesenchymal cells are implicated in this process, yet studies are hampered by the lack of personalized in vitro models. One potential avenue that would permit a personalized approach is to utilize human intestinal organoids (HIOs) derived from induced pluripotent stem cells (iPSCs). iPSCs can be generated from any individual, faithfully recapitulate the genetics of the host and can be directed to form HIOs that contain both epithelial and mesenchymal cells. Our goal was to determine the feasibility of utilizing iPSC-HIO technology to model intestinal fibrotic responses in vitro. Methods iPSCs from two control individuals and two very early onset-IBD (VEOIBD) patients with stricturing complications were obtained and directed to form HIOs. Given HIOs are heterogeneous in terms of size, shape and ratio of mesenchymal to epithelial cells, they were firstly dissociated to a single cell suspension and EpCAM was used to positively select for epithelial cells using magnetic activated cellular sorting. These EpCAM+ cells were then seeded onto transwells and EpCAM- cells were seeded as monolayers in 10% serum containing media. Both cell types were treated with the profibrotic cytokine TGFβ, and changes in the expression of selected genes were analyzed. Results iPSCs from all 4 individuals could be directed to form HIOs containing both epithelial (E-cadherin+) and mesenchymal (vimentin+) cells (see Fig. 1). In the TGFβ-treated mesenchymal cell population, expression of N-cadherin and Col1a1 was significantly increased in all four lines after 8 and 48hrs respectively, with the highest increase occurring in cells derived from VEOIBD patient 2 (see Table 1). In the TGFβ-treated epithelial cell population, Col1a1 and fibronectin expression was increased in all lines after 96hrs with the highest fold change occurring in cells derived from VEOIBD patient 1 (fibronectin) and 2 (Col1a1). Conclusion We demonstrate the feasibility of utilizing iPSC-HIO technology to model intestinal fibrotic responses in vitro. We show that iPSCs generated from all selected individuals could be directed to form HIOs and that responses to the profibrotic cytokine TGFβ can be examined in both intestinal epithelial and mesenchymal cells. This now permits the generation of near unlimited quantities of patient specific cells that could be used to reveal cell and environmental specific mechanisms underpinning intestinal fibrosis which may ultimately lead to personalized treatments. Fluorescent images of human intestinal organoids generated from A) Control 1, B) Control 2, C) VEOIBD patient 1 and D) VEOIBD patient 2 that were immunostained with vimentin (green), E-cadherin (red) and counterstainied with DAPI (blue). All images X20


2018 ◽  
Vol 46 (11) ◽  
pp. 1572-1580 ◽  
Author(s):  
Daichi Onozato ◽  
Misaki Yamashita ◽  
Anna Nakanishi ◽  
Takumi Akagawa ◽  
Yuriko Kida ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document