To date, studies of human type 1 diabetes (T1D) have failed to provide a mechanistic understanding of the causes of the disease, largely because patients must be analyzed long after the autoimmune attack was initiated. Our ignorance of the key molecules and cells mediating the initiation and progression of human T1D may well underlie the paucity of significant new therapeutic interventions. Here we propose to reconstruct human T1D, using iPS-derived ? cells, thymic epithelial cells (TEC), and immune systems derived from T1D patients implanted in a novel immunodeficient mouse model based on the NOD-scid-IL2rgnull (NSG) strain. To accomplish our goal, we propose 2 aims.
Aim 1 will validate human immune and pancreatic beta cell functions in optimized immunodeficient mice (OPTI-MICE). Currently available OPTI-MICE will be improved using genetic techniques to: 1) enhance engraftment of human cells;2) allow for spontaneous and inducible hyperglycemia;3) support expression of human HLA alleles and cytokines;and 4) knockout mouse genes that impair human cell engraftment and function. This suite of improvements will be validated using implantation of fetal human stem cells and tissues.
Aim 2 will reconstruct human T1D in mice using cells derived from Type 1 diabetic iPS cells. These iPS cells will be used to produce the three key cell types: hematopoietic stem cells (HSC) that will generate immune systems, TEC, and ?-cells, all integral to the pathology of T1D. These cells will be derived through the use of directed differentiation and reprogramming strategies. We have been successful in generating functional human ? cells from human control and T1D patient iPS cells by a recently developed multi-step protocol for directed differentiation, providing a standardized and reproducible source of ? cells our studies. Functional human HSC will be generated using two technologies: 1) directed differentiation of iPS cells and 2) reprogramming of differentiated hematopoietic cells using defined factors to derive induced-HSCs. Functional human TEC will be generated using directed differentiation protocols similar to those used to achieve fully differentiated human ? cells. Each cell type will be subjected to rigorous analysis in vitro and in vivo to ensur full functionality. Differentiated cells derived from a single donor will be co-transplanted into OPTI-MICE, thus reconstituting an individual patient's disease in an animal model. Transplanted mice will be carefully monitored for the emergence of autoantibodies and autoreactive T-cells, and for destruction of ?-cells. This new model of human T1D will permit detailed observation, manipulation, and analysis of T1D as it progresses, enabling us to determine which cells and antigens initiate T1D. Such mechanistic insights will properly inform new approaches to curing, or even preventing, this disease. To accomplish our goal, we have assembled an interactive team of researchers formed using seed monies from the Helmsley Charitable Trust. We have now been working together and meeting regularly for over 5 years and have expertise in the relevant areas required to accomplish this project.

Public Health Relevance

To date, studies of human type 1 diabetes (T1D) have failed to provide a mechanistic understanding of the causes of the disease, largely because patients must by definition be analyzed long after the immune attack on their insulin-producing cells began. Here we propose to reconstruct human T1D, using patient derived pluripotent stem cells to generate insulin-secreting cells, autoreactive immune cells, and thymic tissue that educate the immune system to attack the insulin-producing cells, all of which will be transplanted into a novel mouse model that will permit the human disease to develop. This novel model will yield insights that will properly inform new approaches to curing, or even preventing, this disease.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
High Impact Research and Research Infrastructure Cooperative Agreement Programs—Multi-Yr Funding (UC4)
Project #
1UC4DK104218-01
Application #
8813948
Study Section
Special Emphasis Panel (ZDK1-GRB-J (O3))
Program Officer
Arreaza-Rubin, Guillermo
Project Start
2014-09-25
Project End
2019-06-30
Budget Start
2014-09-25
Budget End
2019-06-30
Support Year
1
Fiscal Year
2014
Total Cost
$4,109,951
Indirect Cost
$719,713
Name
University of Massachusetts Medical School Worcester
Department
Other Basic Sciences
Type
Schools of Medicine
DUNS #
603847393
City
Worcester
State
MA
Country
United States
Zip Code
01655
Mondal, Nandini; Dykstra, Brad; Lee, Jungmin et al. (2018) Distinct human ?(1,3)-fucosyltransferases drive Lewis-X/sialyl Lewis-X assembly in human cells. J Biol Chem 293:7300-7314
Saunders, Diane C; Brissova, Marcela; Phillips, Neil et al. (2018) Ectonucleoside Triphosphate Diphosphohydrolase-3 Antibody Targets Adult Human Pancreatic ? Cells for In Vitro and In Vivo Analysis. Cell Metab :
Kernfeld, Eric M; Genga, Ryan M J; Neherin, Kashfia et al. (2018) A Single-Cell Transcriptomic Atlas of Thymus Organogenesis Resolves Cell Types and Developmental Maturation. Immunity 48:1258-1270.e6
Durost, Philip A; Aryee, Ken-Edwin; Manzoor, Fatima et al. (2018) Gene Therapy with an Adeno-Associated Viral Vector Expressing Human Interleukin-2 Alters Immune System Homeostasis in Humanized Mice. Hum Gene Ther 29:352-365
Gawron, Melissa A; Duval, Mark; Carbone, Claudia et al. (2018) Human Anti-HIV-1 gp120 Monoclonal Antibodies with Neutralizing Activity Cloned from Humanized Mice Infected with HIV-1. J Immunol :
Roberts, Frederick R; Hupple, Clinton; Norowski, Elaine et al. (2017) Possible type 1 diabetes risk prediction: Using ultrasound imaging to assess pancreas inflammation in the inducible autoimmune diabetes BBDR model. PLoS One 12:e0178641
Verma, Mohit K; Clemens, Julia; Burzenski, Lisa et al. (2017) A novel hemolytic complement-sufficient NSG mouse model supports studies of complement-mediated antitumor activity in vivo. J Immunol Methods 446:47-53
Hosur, Vishnu; Low, Benjamin E; Avery, Cindy et al. (2017) Development of Humanized Mice in the Age of Genome Editing. J Cell Biochem 118:3043-3048
Walsh, Nicole C; Kenney, Laurie L; Jangalwe, Sonal et al. (2017) Humanized Mouse Models of Clinical Disease. Annu Rev Pathol 12:187-215
Nyalwidhe, Julius O; Gallagher, Glen R; Glenn, Lindsey M et al. (2017) Coxsackievirus-Induced Proteomic Alterations in Primary Human Islets Provide Insights for the Etiology of Diabetes. J Endocr Soc 1:1272-1286

Showing the most recent 10 out of 30 publications