Abstract

Uterine fibroids (UFs) are the most prevalent gynecologic neoplasm, affecting 70-80% of women over their lifespan. Although UFs are benign they can become life-threatening and require invasive surgeries such as myomectomy and hysterectomy. Notwithstanding the significant negative influence UFs have on female reproductive health, very little is known about early events that initiate tumor development. Several risk factors for UFs have been identified including vitamin D deficiency, inflammation, DNA repair deficiency, and environmental exposures to endocrine-disrupting chemicals (EDCs). EDCs have come under scrutiny recently due to their role in UF development. Epidemiologic studies have found an association between increased risk for early UF diagnosis and in utero EDC exposure. Environmental exposure to EDCs during uterine development increases UF incidence in a UF animal model. Notably, several studies demonstrated that abnormal myometrial stem cells (MMSCs) are the cell origin for UFs development. Our recent studies demonstrated that early-life EDC exposure reprogrammed the MMSCs toward a pro-fibroid landscape and altered the DNA repair and inflammation pathways. Notably, Vitamin D3 (VITD3) as a natural compound shrank the UF growth concomitantly with the reversion of several abnormal biological pathways and ameliorated the developmental exposure-induced DNA damage and pro-inflammation pathway in primed MMSCs. This review highlights and emphasizes the importance of multiple pathway interactions in the context of hypovitaminosis D at the MMSCs level and provides proof-of-concept information that can help develop a safe, long-term, durable, and non-surgical therapeutic option for UFs.

Highlights

  • Uterine fibroids (UFs, AKA: leiomyoma) are the most widespread pelvic tumors in women of reproductive age

  • Al-Hendy group showed that Vitamin D3 (VITD3) deficiency had been connected with rising sex steroid receptors and proliferation-related gene expression increasing fibrosis and immunosuppression by Tregs expansion increased inflammation and DNA damage accumulation in murine myometrium, leading to increased risk of tumor development due to attenuation of the homology-dependent doublestrand breaks (DSBs) repair[132]

  • In addition to the effect of VITD3 on the UFs and UF cells, recently El-Kafas et al have shown for the first time that VITD3 treatment improves DNA repair machinery load in myometrial stem cells (MMSCs) early-life exposed to DES and upregulates the expression of DNA repair sensors genes MRN (MRE11, RAD50, Nijmegen breakage syndrome 1 (NBS1)), mediator’s genes (CHEK1, CHEK2, and BRCA1) and DSB effectors genes (RAD51, BRCA2), and downregulate the DNA damage sensor γ-H2AX

Read more

Summary

Section D: Clinical Pharmacy & Pharmacology

Endocrine-Disrupting Chemicals and Vitamin D Deficiency in the Pathogenesis of Uterine Fibroids.

INTRODUCTION
Findings
CONCLUSIONS
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call