Spermatogonial type 3 deiodinase regulates thyroid hormone target genes in developing testicular somatic cells

ME Martinez, CW Lary, AA Karaczyn… - …, 2019 - academic.oup.com
ME Martinez, CW Lary, AA Karaczyn, MD Griswold, A Hernandez
Endocrinology, 2019academic.oup.com
Premature overexposure to thyroid hormone causes profound effects on testis growth,
spermatogenesis, and male fertility. We used genetic mouse models of type 3 deiodinase
(DIO3) deficiency to determine the genetic programs affected by premature thyroid hormone
action and to define the role of DIO3 in regulating thyroid hormone economy in testicular
cells. Gene expression profiling in the neonatal testis of DIO3-deficient mice identified 5699
differentially expressed genes. Upregulated and downregulated genes were, respectively …
Abstract
Premature overexposure to thyroid hormone causes profound effects on testis growth, spermatogenesis, and male fertility. We used genetic mouse models of type 3 deiodinase (DIO3) deficiency to determine the genetic programs affected by premature thyroid hormone action and to define the role of DIO3 in regulating thyroid hormone economy in testicular cells. Gene expression profiling in the neonatal testis of DIO3-deficient mice identified 5699 differentially expressed genes. Upregulated and downregulated genes were, respectively, involved according to DAVID analysis with cell differentiation and proliferation. They included anti-Müllerian hormone and genes involved in the formation of the blood–testis barrier, which are specific to Sertoli cells (SCs). They also included steroidogenic genes, which are specific to Leydig cells. Comparison with published data sets of genes enriched in SCs and spermatogonia, and responsive to retinoic acid (RA), identified a subset of genes that were regulated similarly by RA and thyroid hormone. This subset of genes showed an expression bias, as they were downregulated when enriched in spermatogonia and upregulated when enriched in SCs. Furthermore, using a genetic approach, we found that DIO3 is not expressed in SCs, but spermatogonia-specific inactivation of DIO3 led to impaired testis growth, reduced SC number, decreased cell proliferation and, especially during neonatal development, altered gene expression specific to somatic cells. These findings indicate that spermatogonial DIO3 protects testicular cells from untimely thyroid hormone signaling and demonstrate a mechanism of cross-talk between somatic and germ cells in the neonatal testis that involves the regulation of thyroid hormone availability and action.
Oxford University Press