GLI3 resides at the intersection of hedgehog and androgen action to promote male sex differentiation

Autor: Samantha R. Lewis, Elena M. Kaftanovskaya, Stephanie Winske, Chad M. Vezina, Martin J. Cohn, Anbarasi Kothandapani, Anna Baines, Jessica Noel, Abbey Zacharski, Emily M. Merton, Alexander I. Agoulnik, Joan S. Jorgensen, Kyle Krellwitz
Jazyk: angličtina
Rok vydání: 2020
Předmět:
Male
Cancer Research
Embryology
Sex Differentiation
QH426-470
Biochemistry
Mice
0302 clinical medicine
Cell Signaling
Cryptorchidism
Medicine and Health Sciences
Morphogenesis
Insulin
Testosterone
Testes
Lipid Hormones
Testicular Hormones
Genetics (clinical)
0303 health sciences
Sexual Differentiation
Leydig cell
Gene Expression Regulation
Developmental

Leydig Cells
Cell Differentiation
Hedgehog signaling pathway
medicine.anatomical_structure
embryonic structures
Androgens
Male sex differentiation
Anatomy
Genital Anatomy
Signal Transduction
Research Article
medicine.medical_specialty
animal structures
medicine.drug_class
Mice
Transgenic

Nerve Tissue Proteins
Biology
03 medical and health sciences
Zinc Finger Protein Gli3
Internal medicine
medicine
Genetics
Animals
Humans
Hedgehog Proteins
Molecular Biology
Hedgehog
Ecology
Evolution
Behavior and Systematics

030304 developmental biology
Sexual differentiation
Embryos
Reproductive System
Proteins
Biology and Life Sciences
Cell Biology
Androgen
Hormones
Disease Models
Animal

Endocrinology
Mutation
Hedgehog Signaling
Androgen insufficiency
030217 neurology & neurosurgery
Developmental Biology
Zdroj: PLoS Genetics, Vol 16, Iss 6, p e1008810 (2020)
PLoS Genetics
ISSN: 1553-7404
1553-7390
Popis: Urogenital tract abnormalities are among the most common congenital defects in humans. Male urogenital development requires Hedgehog-GLI signaling and testicular hormones, but how these pathways interact is unclear. We found that Gli3XtJ mutant mice exhibit cryptorchidism and hypospadias due to local effects of GLI3 loss and systemic effects of testicular hormone deficiency. Fetal Leydig cells, the sole source of these hormones in developing testis, were reduced in numbers in Gli3XtJ testes, and their functional identity diminished over time. Androgen supplementation partially rescued testicular descent but not hypospadias in Gli3XtJ mutants, decoupling local effects of GLI3 loss from systemic effects of androgen insufficiency. Reintroduction of GLI3 activator (GLI3A) into Gli3XtJ testes restored expression of Hedgehog pathway and steroidogenic genes. Together, our results show a novel function for the activated form of GLI3 that translates Hedgehog signals to reinforce fetal Leydig cell identity and stimulate timely INSL3 and testosterone synthesis in the developing testis. In turn, exquisite timing and concentrations of testosterone are required to work alongside local GLI3 activity to control development of a functionally integrated male urogenital tract.
Author summary Disorders in male sex differentiation (DSD) are among the most common defects in all live births, yet in many cases, pediatric patient families are reluctant to address the issue and endure lifelong consequences. Urogenital tract development, as in many organ systems, depends on exquisite timing among layers of a number of signaling pathways. Here, we show that interactions between the hedgehog and androgen signaling pathways are required for the development of internal and external male sex characteristics, but results for each tissue is distinct. This new knowledge will aid in discovering the means by which congenital malformations might occur, identify potential developmental targets that might be vulnerable to environmental exposures, and promote new ideas for how they might be prevented.
Databáze: OpenAIRE