Thyroid diseases are commonly caused by abnormal thyroid cell proliferation and differentiation, which can lead to various, possibly fatal complications. The mechanisms by which these diseases develop by are largely unknown and have yet to be discovered. Presently, the long-term goal of our parent grant is to elucidate the molecular mechanisms by which embryonic stem (ES) cells differentiate into definitive thyrocytes. Pluripotent ES cells, derived from the inner cell mass of early embryos, can both self-renew and differentiate into all cell types in the body and as such provide a key thyroid developmental model. The manipulation of the pluripotent potential of murine ES cells in vitro has allowed us to direct differentiation towards the thyroid lineage under the appropriate conditions. To expand upon this mouse model, we are also working to develop a similar model using NIH-registry human ES cells. We have recently developed a form of induced pluripotent stem (iPS) cell technology, which can reprogram adult skin fibroblasts into an ES cell-like state using the retrovirus-mediated transfection of four transcription factors (Oct4, Sox2, c-Myc, and Klf4). Our manipulated iPS cells are similar to ES cells in many respects, including the expression of main stem cell genes and proteins, embryoid body formation, teratomas formation, potency and differentiation. In this application, we will expand on human ES cell studies focused on the mechanisms regulating endoderm induction and its specification to thyroid lineage during ES cell differentiation. Furthermore, we propose to establish and propagate murine iPS cells from a mutant TSHR hypothyroid mouse model. The hypothyroid mice display severe congenital hypothyroidism and thyroid hyperplasia and provide an opportunity to explore the pathophysiological manifestations of this disease. The goal of Specific Aim 1 is to generate thyroid follicular cells from human ES cells. The goal of Specific Aim 2 is to produce and characterize murine iPS cells from normal and mutant TSHR fibroblasts. We will use our established methods to reprogram skin fibroblasts of normal and mutant TSHR mice into iPS cells by the ectopic expression of four transcription factors. We will test the hypothesis that the disease-specific iPS cells can be coaxed into thyroid follicular cells to explore the biological processes that lead to disease phenotypes. Finally, we will characterize the thyrocyte differentiation potential of these murine iPS cell lines in comparison to our well-established murine TSHR-/- ES cell lines by using in vitro and in vivo assays. More specifically, to initiate effort to new therapeutic approaches, we will use these disease-iPS cells as assay tools to validate the defects of iodide transport and thyroid hormone synthesis and secretion. The success of these aims will provide a number of therapeutic promises including: 1) the ability to establish patient-derived iPS cell lines as a research tool to model human disease, and 2) the opportunity to use patient-derived iPS cell lines to perform sophisticated testing of candidate therapeutics and screening assays.

Public Health Relevance

Thyroid diseases are commonly caused by abnormal thyroid cell proliferation and differentiation, which can lead to various, possibly fatal complications. The mechanisms by which these diseases develop by are largely unknown and have yet to be discovered. The goal of this project is to combine the advantages of embryonic stem cells and induced pluripotent stem cells with the powerful genetic tools available in mouse models to take a unique approach to thyroid development and disease.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK068057-09
Application #
8256760
Study Section
Integrative and Clinical Endocrinology and Reproduction Study Section (ICER)
Program Officer
Sato, Sheryl M
Project Start
2004-07-01
Project End
2014-03-31
Budget Start
2012-04-01
Budget End
2013-03-31
Support Year
9
Fiscal Year
2012
Total Cost
$299,471
Indirect Cost
$99,824
Name
Saint Louis University
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
050220722
City
Saint Louis
State
MO
Country
United States
Zip Code
63103
Li, Wen; Reeb, Ashley N; Sewell, William A et al. (2013) Phenotypic characterization of metastatic anaplastic thyroid cancer stem cells. PLoS One 8:e65095
Sewell, Will; Reeb, Ashley; Lin, Reigh-Yi (2013) An orthotopic mouse model of anaplastic thyroid carcinoma. J Vis Exp :
Arufe, Maria C; Lu, Min; Lin, Reigh-Yi (2009) Differentiation of murine embryonic stem cells to thyrocytes requires insulin and insulin-like growth factor-1. Biochem Biophys Res Commun 381:264-70
Friedman, Susan; Lu, Min; Schultz, Atara et al. (2009) CD133+ anaplastic thyroid cancer cells initiate tumors in immunodeficient mice and are regulated by thyrotropin. PLoS One 4:e5395
Lu, Min; Lin, Reigh-Yi (2008) TSH stimulates adipogenesis in mouse embryonic stem cells. J Endocrinol 196:159-69
Thomas, Dolly; Friedman, Susan; Lin, Reigh-Yi (2008) Thyroid stem cells: lessons from normal development and thyroid cancer. Endocr Relat Cancer 15:51-8
Lin, Reigh-Yi (2007) New insights into thyroid stem cells. Thyroid 17:1019-23
Arufe, Maria C; Lu, Min; Kubo, Atsushi et al. (2006) Directed differentiation of mouse embryonic stem cells into thyroid follicular cells. Endocrinology 147:3007-15