The World Health Organization estimates that the global prevalence of diabetes in adults is 9%. Both Type 1 and Type 2 diabetes involve the issue of reduced ?-cell mass; subsequently a cure for diabetes must involve ?-cell replacement. Ideally, a cure would involve inducing regeneration via ?-cell neogenesis from endogenous pancreatic progenitors. For these reasons we are interested in explicating the process of ?-cell neogenesis, i.e., how ? cells are formed from progenitors in the pancreas. Unlike their mammalian counterparts, we have shown that zebrafish readily regenerate their ? cells following cell-specific ablation. Our goal is to identify the mechanisms behind the zebrafish?s capacity for ?-cell neogenesis. Such molecular pathways could then be pharmacologically exploited in humans to induce ?-cell neogenesis. We have recently made two discoveries critical to understanding how zebrafish so easily recover following ?- cell ablation. First, we identified the progenitor source for ?-cell neogenesis?namely a cell type called the centroacinar cell (CAC); second, we discovered that diminished activity of the Sox9b transcription factor leads to significantly accelerated regeneration. From these insights we hypothesized 1) Sox9b acts cell autonomously to maintain progenitor potency in adult CACs; 2) Diminishing Sox9b activity alters the behavior of CACs or their progeny in regeneration; and 3) The identification of downstream genes of SOX9 will elucidate molecular mechanisms that regulate ?-cell differentiation. By testing these hypotheses in three complementary yet independent aims, we expect to discover if the differences in regeneration between sox9b heterozygotes and wildtypes is due either to pre-existing differences in morphology or the behavior of the CACs during regeneration. Furthermore, we will use genomic approaches to identify the direct downstream transcriptional targets of Sox9 homologs because we expect these targets will be the mediators of the sox9b haploinsufficient phenotype. As part of our preliminary data we knocked down SOX9 levels in the human PANC-1 cell line, a surrogate for pancreatic progenitors, and identified affected transcript levels. We have also used chromatin immunoprecipitation and deep sequencing (ChIP-seq) to identify where SOX9 binds in the PANC-1 genome. Putting these results together has allowed us to find direct targets of SOX9 transcriptional activity in PANC-1 cells. We have identified interesting genes downstream of SOX9, such as EpCAM, and identified biological pathways that SOX9 controls, such as cilia function and Notch regulation. Greatly encouraged by our preliminary results we now aim to expand this analysis to find SOX9 targets during development and regeneration. By the end of this proposed work we expect a better understanding of SOX9 function and the discovery of potential therapeutic routes to alleviate ?-cell paucity in humans.

Public Health Relevance

This project focuses on the molecular mechanisms that regulate pancreatic progenitor differentiation into insulin-producing ? cells, and particularly the role of the transcription factor, SOX9. Our project will utilize the following: 1) the zebrafish as a model system, as it allows the study of differentiation during both development and regeneration; and 2) human cell lines, which facilitate studying regulation at a transcription level. Using a unique collection of tools, including transgenic reporters and mutant zebrafish, we will identify new targets and pathways that ultimately can be exploited to induce the production of insulin-producing cells and treat diabetes.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK080730-12
Application #
9922906
Study Section
Development - 1 Study Section (DEV1)
Program Officer
Zaghloul, Norann
Project Start
2008-07-01
Project End
2022-05-31
Budget Start
2020-06-01
Budget End
2021-05-31
Support Year
12
Fiscal Year
2020
Total Cost
Indirect Cost
Name
University of California Irvine
Department
Anatomy/Cell Biology
Type
Schools of Arts and Sciences
DUNS #
046705849
City
Irvine
State
CA
Country
United States
Zip Code
92617
Zhang, Danhua; Gates, Keith P; Barske, Lindsey et al. (2017) Endoderm Jagged induces liver and pancreas duct lineage in zebrafish. Nat Commun 8:769
Huang, Wei; Beer, Rebecca L; Delaspre, Fabien et al. (2016) Sox9b is a mediator of retinoic acid signaling restricting endocrine progenitor differentiation. Dev Biol 418:28-39
Beer, Rebecca L; Parsons, Michael J; Rovira, Meritxell (2016) Centroacinar cells: At the center of pancreas regeneration. Dev Biol 413:8-15
Delaspre, Fabien; Beer, Rebecca L; Rovira, Meritxell et al. (2015) Centroacinar Cells Are Progenitors That Contribute to Endocrine Pancreas Regeneration. Diabetes 64:3499-509
Park, J T; Johnson, N; Liu, S et al. (2015) Differential in vivo tumorigenicity of diverse KRAS mutations in vertebrate pancreas: A comprehensive survey. Oncogene 34:2801-6
Wang, Guangliang; Rajpurohit, Surendra K; Delaspre, Fabien et al. (2015) First quantitative high-throughput screen in zebrafish identifies novel pathways for increasing pancreatic ?-cell mass. Elife 4:
Wang, Yue J; Park, Joon T; Parsons, Michael J et al. (2015) Fate mapping of ptf1a-expressing cells during pancreatic organogenesis and regeneration in zebrafish. Dev Dyn 244:724-35
Huang, Wei; Wang, Guangliang; Delaspre, Fabien et al. (2014) Retinoic acid plays an evolutionarily conserved and biphasic role in pancreas development. Dev Biol 394:83-93
Subedi, Abhignya; Macurak, Michelle; Gee, Stephen T et al. (2014) Adoption of the Q transcriptional regulatory system for zebrafish transgenesis. Methods 66:433-40
Quillien, Aurelie; Moore, John C; Shin, Masahiro et al. (2014) Distinct Notch signaling outputs pattern the developing arterial system. Development 141:1544-52

Showing the most recent 10 out of 18 publications