Thyroid hormone promotes postnatal rat pancreatic β-cell development and glucose-responsive insulin secretion through MAFA

C Aguayo-Mazzucato, AM Zavacki, A Marinelarena… - Diabetes, 2013 - Am Diabetes Assoc
C Aguayo-Mazzucato, AM Zavacki, A Marinelarena, J Hollister-Lock, I El Khattabi, A Marsili…
Diabetes, 2013Am Diabetes Assoc
Neonatal β cells do not secrete glucose-responsive insulin and are considered immature.
We previously showed the transcription factor MAFA is key for the functional maturation of β
cells, but the physiological regulators of this process are unknown. Here we show that
postnatal rat β cells express thyroid hormone (TH) receptor isoforms and deiodinases in an
age-dependent pattern as glucose responsiveness develops. In vivo neonatal
triiodothyronine supplementation and TH inhibition, respectively, accelerated and delayed …
Neonatal β cells do not secrete glucose-responsive insulin and are considered immature. We previously showed the transcription factor MAFA is key for the functional maturation of β cells, but the physiological regulators of this process are unknown. Here we show that postnatal rat β cells express thyroid hormone (TH) receptor isoforms and deiodinases in an age-dependent pattern as glucose responsiveness develops. In vivo neonatal triiodothyronine supplementation and TH inhibition, respectively, accelerated and delayed metabolic development. In vitro exposure of immature islets to triiodothyronine enhanced the expression of Mafa, the secretion of glucose-responsive insulin, and the proportion of responsive cells, all of which are effects that were abolished in the presence of dominant-negative Mafa. Using chromatin immunoprecipitation and electrophoretic mobility shift assay, we show that TH has a direct receptor-ligand interaction with the Mafa promoter and, using a luciferase reporter, that this interaction was functional. Thus, TH can be considered a physiological regulator of functional maturation of β cells via its induction of Mafa.
Am Diabetes Assoc