Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2022 Apr;29(4):1357-1367.
doi: 10.1007/s43032-021-00700-5. Epub 2021 Oct 15.

Aberrant H19 Expression Disrupts Ovarian Cyp17 and Testosterone Production and Is Associated with Polycystic Ovary Syndrome in Women

Affiliations

Aberrant H19 Expression Disrupts Ovarian Cyp17 and Testosterone Production and Is Associated with Polycystic Ovary Syndrome in Women

Zhaojuan Chen et al. Reprod Sci. 2022 Apr.

Abstract

As one of the most common endocrine disorders affecting women, polycystic ovary syndrome (PCOS) is associated with serious conditions including anovulation, endometrial cancer, infertility, hyperandrogenemia, and an increased risk for obesity and metabolic derangements. One contributing etiology to the pathophysiology of hyperandrogenemia associated with PCOS is an intrinsic alteration in ovarian steroidogenesis, leading to enhanced synthesis of androgens including testosterone. Studies have suggested that the increased testosterone synthesis seen in PCOS is driven in part by increased activity of CYP17A1, the rate-limiting enzyme for the formation of androgens in the gonads and adrenal cortex, which represents a critical factor driving enhanced testosterone secretion in PCOS. In this work, we evaluated the hypothesis that dysregulation of the noncoding RNA H19 results in aberrant CYP17 and testosterone production. To achieve this, we measured Cyp17 in ovarian tissues of H19 knockout mice, and quantified serum testosterone levels, in comparison with wild-type controls. We also evaluated circulating and ovarian H19 expression and correlated results with the presence or absence of PCOS in a group of women undergoing evaluation and treatment for infertility. We found that the loss of H19 in a mouse model results in decreased ovarian Cyp17, along with decreased serum testosterone in female mice. Moreover, utilizing serum samples and cumulus cells from women with PCOS, we showed that circulating and ovarian levels of H19 are increased in women with PCOS compared to controls. Findings from our multimodal experimental strategy, involving both a mouse model of dysregulated H19 expression and clinical serum and ovarian cellular samples from women with PCOS, suggest that the loss of H19 may disrupt androgen production via a Cyp17-mediated mechanism. Conversely, excess H19 may play a role in the pathogenesis of PCOS-associated hyperandrogenemia.

Keywords: H19; Noncoding RNA; PCOS; ncRNA.

PubMed Disclaimer

Conflict of interest statement

Conflict of Interest

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Decreased CYP17α levels in ovaries of H19KO female mice. To evaluate CYP17α expression, RNA extraction, cDNA synthesis, and RT-qPCR were performed on ovarian tissue from 8-week-old H19KO and WT female mice (n = 5 per group). a Mean relative Cyp17α expression levels by real-time PCR are shown. Cyp17α expression was decreased in ovaries from H19KO female mice compared to that from WT. Beta-actin was used as a control. b Western blot analysis was performed to measure CYP17α protein levels, using GAPDH as control. c Quantification of Western blot showing decreased CYP17α protein in ovary compared to that in WT. All data were analyzed using Student’s t-test. *p = 0.05; ***p < 0.001. Error bars represent one SEM. P, proestrus; E, estrus; M, metestrus; D, diestrus
Fig. 2
Fig. 2
Serum quantification of hormone levels in female mice. Serum hormone levels were quantified via retroorbital blood collection in H19KO and WT female mice at each estrus cycle stage. Serum testosterone (T) levels were lower in the estrus stage in female H19KO mice compared to those in WT mice (Fig. 2a; 20.66 ng/dL vs 33.42 ng/dL; p < 0.01). Serum corticosterone concentrations were similar across estrus cycle stage in H19KO and WT mice. All data were analyzed using two-way ANOVA with correction for multiple comparisons. **p < 0.005. Error bars represent one standard error of the mean (SEM)
Fig. 3
Fig. 3
Serum H19 expression is increased in women with PCOS. Blood samples from 35 patients (19 controls diagnosed with male and/or tubal factor infertility and 16 women with hyperandrogenic PCOS) undergoing infertility evaluation were collected in the early follicular phase (days 2–4 of the menstrual cycle) and evaluated for H19 expression. The bar graph shows the expression level of H19 in women with PCOS, presented as fold change relative to women without PCOS (e.g., male and tubal factor controls). Relative H19 expression increased 2.5-fold in women with PCOS compared to that in women with male/tubal factor infertility (p < 0.0005)
Fig. 4
Fig. 4
Cumulus cell expression of H19 in women with PCOS versus controls (male/tubal factor). A total of 39 women undergoing in vitro fertilization (29 non-PCOS women undergoing IVF for male factor infertility and 10 women with PCOS) were evaluated. At the time of oocyte retrieval, cumulus cells were collected, total RNA was isolated, and cDNA was synthesized by reverse transcription. RNA levels were analyzed using quantitative real-time PCR. H19 RNA levels were normalized to those of β-actin and presented as relative expression levels using the comparative CT method. Box plot demonstrates increased relative H19 expression (2.8-fold) in cumulus cells of PCOS patients undergoing IVF, compared to non-PCOS patients (p < 0.05)

Similar articles

Cited by

References

    1. Bozdag G, Mumusoglu S, Zengin D, Karabulut E, Yildiz BO. The prevalence and phenotypic features of polycystic ovary syndrome: a systematic review and meta-analysis. Hum Reprod. 2016;31(12):2841–55. 10.1093/humrep/dew218. - DOI - PubMed
    1. Randeva HS, Tan BK, Weickert MO, et al. Cardiometabolic aspects of the polycystic ovary syndrome. Endocr Rev. 2012;33(5):812–41. 10.1210/er.2012-1003. - DOI - PMC - PubMed
    1. Gottschau M, Kjaer SK, Jensen A, Munk C, Mellemkjaer L. Risk of cancer among women with polycystic ovary syndrome: a Danish cohort study. Gynecol Oncol. 2015;136(1):99–103. 10.1016/j.ygyno.2014.11.012. - DOI - PubMed
    1. Azziz R, Woods KS, Reyna R, Key TJ, Knochenhauer ES, Yildiz BO. The prevalence and features of the polycystic ovary syndrome in an unselected population. J Clin Endocrinol Metab. 2004;89(6):2745–9. 10.1210/jc.2003-032046. - DOI - PubMed
    1. Lerchbaum E, Schwetz V, Rabe T, Giuliani A, Obermayer-Pietsch B. Hyperandrogenemia in polycystic ovary syndrome: exploration of the role of free testosterone and androstenedione in metabolic phenotype. PLoS ONE. 2014;9(10):e108263–e108263. 10.1371/journal.pone.0108263. - DOI - PMC - PubMed

Publication types

-