Acute and chronic inflammatory environments of the gastrointestinal tract like gastroesophageal reflux disease (GERD), intestinal graft vs. host disease (GVHD), acute rejection after small intestine transplantation, and IBD conditions like Ulcerative colitis (UC), are fairly common human diseases. Disease models based on human cell and tissue culture systems that recapitulate in vivo growth and differentiation patterns would enhance our understanding of disease progression and improve prevention and detection strategies. This is an important objective of this proposal. In these disorders, epithelial cel oxidative stress is a key pathogenic factor for disease progression. This oxidative stress is partly from endogenous enzymes (Cyclooxygenases and NADPH oxidases) that induce DNA damage and mutations, and alter DNA methylation patterns, which together contributes to the development of metaplasia and cancer. We have begun to explore this using organotypic culture systems to model Cox-2 in BE pathogenesis. When we ectopically express Cox-2 in normal human esophageal keratinocytes, we observe the development of intestinal mucin-filled cysts. We propose to extend this success in physiologically relevant directions. Our main objective will be to test the hypothesis that the organotypic culture systems can be modified to model acute and chronic oxidative stress in esophageal and intestinal epithelium that physiologically resembles in vivo events in GERD, BE, GVHD, small bowel transplant rejection, and UC. We propose to test this by pursuing the following Specific Aims: 1) Adapt the esophageal organotypic culture system to better model GERD esophagitis and progression of stem cells to metaplasia and dysplasia. 2) Develop organotypic and 3D multi-cellular culture systems to model inflammatory microenvironments of GVHD, small bowel transplant rejection, and IBD including UC. GERD/BE, IBD/neoplasia, and GVHD are important, relatively common conditions that place a significant burden on the US healthcare system. We propose to develop novel multi-cellular in vitro human tissue engineered models that are representative of the pathogenesis for these conditions. These models will be of enormous value, allowing us to test hypotheses and advance our understanding of these disorders rapidly, and would have a translational impact since pharmacologic inhibition of Cox-2 is well established. This work will greatly improve our ability to study, prevent, and treat Barrett's esophagus, IBD, and GVHD. It will also foster the development of novel therapeutic and preventive strategies that will improve patient care for these important clinical conditions.

Public Health Relevance

Chronic inflammatory conditions are an especially burdensome healthcare problem in the US. Not only must the pain and disability be managed, but patients and health-care providers must be vigilant for the long-term consequences of chronic inflammation, most prominently cancer. Two examples of this are acid reflux disease, which leads to metaplasia and cancer, and chronic inflammatory conditions of the intestine and colon like graft vs. host disease or ulcerative colitis, the latter which can also frequently progress to colon cancer. At present there is little known about the mechanisms giving rise to metaplasia and cancer in the setting of chronic inflammation. This proposal describes approaches to model and mechanistically explore the contributions of Cox-2, oxidative stress and DNA damage, inflammatory cells, and epithelial cells to the pathogenesis of metaplasia and cancer, and we anticipate our approaches will yield improved human cell culture models suitable for testing and novel therapeutic and preventive strategies that will improve patient care for these important clinical conditions.

Agency
National Institute of Health (NIH)
Institute
National Center for Advancing Translational Sciences (NCATS)
Type
Research Demonstration--Cooperative Agreements (U18)
Project #
1U18TR000536-01
Application #
8415397
Study Section
Special Emphasis Panel (ZRG1-BST-N (50))
Program Officer
Sutherland, Margaret L
Project Start
2012-07-24
Project End
2014-06-30
Budget Start
2012-07-24
Budget End
2013-06-30
Support Year
1
Fiscal Year
2012
Total Cost
$375,600
Indirect Cost
$128,100
Name
University of Pennsylvania
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
042250712
City
Philadelphia
State
PA
Country
United States
Zip Code
19104
Pattison, Amanda M; Blomain, Erik S; Merlino, Dante J et al. (2016) Intestinal Enteroids Model Guanylate Cyclase C-Dependent Secretion Induced by Heat-Stable Enterotoxins. Infect Immun 84:3083-91
Kong, Jianping; Whelan, Kelly A; Laczkó, Dorottya et al. (2016) Autophagy levels are elevated in barrett's esophagus and promote cell survival from acid and oxidative stress. Mol Carcinog 55:1526-1541
Hamilton, Kathryn E; Crissey, Mary Ann S; Lynch, John P et al. (2015) Culturing adult stem cells from mouse small intestinal crypts. Cold Spring Harb Protoc 2015:354-8
Kong, Jianping; Sai, Hong; Crissey, Mary Ann S et al. (2015) Immature myeloid progenitors promote disease progression in a mouse model of Barrett's-like metaplasia. Oncotarget 6:32980-3005
Nakagawa, Hiroshi; Whelan, Kelly; Lynch, John P (2015) Mechanisms of Barrett's oesophagus: intestinal differentiation, stem cells, and tissue models. Best Pract Res Clin Gastroenterol 29:3-16
Abrams, Julian A; Appelman, Henry D; Beer, David G et al. (2014) Barrett's Esophagus Translational Research Network (BETRNet): the pivotal role of multi-institutional collaboration in esophageal adenocarcinoma research. Gastroenterology 146:1586-90
Hartman, Kira G; Bortner Jr, James D; Falk, Gary W et al. (2014) Modeling human gastrointestinal inflammatory diseases using microphysiological culture systems. Exp Biol Med (Maywood) 239:1108-23
Vega, Maria E; Giroux, Véronique; Natsuizaka, Mitsuteru et al. (2014) Inhibition of Notch signaling enhances transdifferentiation of the esophageal squamous epithelium towards a Barrett's-like metaplasia via KLF4. Cell Cycle 13:3857-66
Magness, Scott T; Puthoff, Brent J; Crissey, Mary Ann et al. (2013) A multicenter study to standardize reporting and analyses of fluorescence-activated cell-sorted murine intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 305:G542-51
Hartman, Kira G; Bortner, James D; Falk, Gary W et al. (2013) Modeling inflammation and oxidative stress in gastrointestinal disease development using novel organotypic culture systems. Stem Cell Res Ther 4 Suppl 1:S5