2020
DOI: 10.1096/fj.201902384r
|View full text |Cite
|
Sign up to set email alerts
|

The lack of HSD17B3 in male mice results in disturbed Leydig cell maturation and endocrine imbalance akin to humans with HSD17B3 deficiency

Abstract: Hydroxysteroid (17β) dehydrogenase type 3 (HSD17B3) deficiency causes a disorder of sex development in humans, where affected males are born with female-appearing external genitalia, but are virilized during puberty. The hormonal disturbances observed in the Hsd17b3 knockout mice (HSD17B3KO), generated in the present study, mimic those found in patients with HSD17B3 mutations. Identical to affected humans, serum T in the adult HSD17B3KO mice was within the normal range, while a striking increase was detected i… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
3
1
1

Citation Types

0
24
0

Year Published

2021
2021
2024
2024

Publication Types

Select...
8

Relationship

0
8

Authors

Journals

citations
Cited by 9 publications
(24 citation statements)
references
References 50 publications
(58 reference statements)
0
24
0
Order By: Relevance
“…HSD17B3 is the last enzyme in the synthesis of T and responsible for the conversion of A‐dione into T. In AR Lmon/Y testis, this final step is affected, resulting in an accumulation of A‐dione. Similar to AR Lmon/Y , it was recently shown that a HSD17B3 knockout mouse model also has very high circulating A‐dione levels, in combination with elevated serum T values, indicating that other enzymes can compensate for HSD17B3 in T synthesis (Rebourcet et al , 2020; Sipilä et al , 2020). In contrast, the disruption of the HPG axis in the AR −/Y mice leads to a more severe testicular phenotype: while LH levels are increased compared with WT males, T synthesis remains low although the defective LC cells still have some response as evidenced by the circulating A‐dione (De Gendt et al , 2004) and (Fig 3B).…”
Section: Discussionmentioning
confidence: 98%
“…HSD17B3 is the last enzyme in the synthesis of T and responsible for the conversion of A‐dione into T. In AR Lmon/Y testis, this final step is affected, resulting in an accumulation of A‐dione. Similar to AR Lmon/Y , it was recently shown that a HSD17B3 knockout mouse model also has very high circulating A‐dione levels, in combination with elevated serum T values, indicating that other enzymes can compensate for HSD17B3 in T synthesis (Rebourcet et al , 2020; Sipilä et al , 2020). In contrast, the disruption of the HPG axis in the AR −/Y mice leads to a more severe testicular phenotype: while LH levels are increased compared with WT males, T synthesis remains low although the defective LC cells still have some response as evidenced by the circulating A‐dione (De Gendt et al , 2004) and (Fig 3B).…”
Section: Discussionmentioning
confidence: 98%
“…Although the physiological significance of this activity is unclear, HSD17B3 may contribute to testicular E2 production. In support of this hypothesis, testicular E2 levels in Hsd17b3 KO mice were found to be decreased, despite E1 levels being increased [11]. Estrogen-estrogen receptor (ER) pathways are essential for spermatogenesis [37][38][39].…”
Section: Discussionmentioning
confidence: 91%
“…In addition to humans, it was recently reported in DSD dogs that truncation of the encoded protein was caused by 2-bp deletion of the HSD17B3 gene [10]. Furthermore, male Hsd17b3KO mice were reported to show a delay in puberty, owing to an abnormal A4/T ratio [11]. These findings indicate that the HSD17B3 gene plays important roles in sex differentiation in various mammalian species.…”
Section: Discussionmentioning
confidence: 96%
See 1 more Smart Citation
“…Since the in silico model establishment of the HSD17B3 deficiency in mice, two independent research groups have now reported the phenotypes of independently derived Hsd17b3 -deficient mouse lines [ 17 , 34 ]. These models were designed to mimic the human mutation causing HSD17B3 deficiency by blocking the conversion of androstenedione to the active androgen testosterone, subsequently causing disruption to the canonical androgen production pathway.…”
Section: Transgenic Mouse Models Of Hsd17b3 Deficiencymentioning
confidence: 99%