Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Ecotoxicol Environ Saf ; 206: 111190, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32871518

ABSTRACT

Radiotherapy represents a critical component in cancer treatment. However, premature ovarian failure (POF) is a major hurdle of deleterious off-target effects in young females, which, therefore, call for an effective radioprotective agent. The present study aimed to explore the molecular mechanism underlying the protective effects of N-acetyl-L-cysteine (NAC) against γ-radiation-provoked POF. Immature female Sprague-Dawley rats were orally-administered NAC (50 mg/kg) and were exposed to a single whole-body dose of 3.2 Gy ϒ-radiation. NAC administration remarkably reversed abnormal serum estradiol and anti-Müllerian hormone levels by 73% and 40%, respectively while ameliorating the histopathological and ultrastructural alterations-triggered by γ-radiation. Mechanistically, NAC alleviated radiation-induced oxidative damage through significantly increased glutathione peroxidase activity by 102% alongside with decreasing NADPH oxidase subunits (p22 and NOX4) gene expressions by 48% and 38%, respectively compared to the irradiated untreated group. Moreover, NAC administration achieved its therapeutic effect by inhibiting ovarian apoptosis-induced by radiation through downregulating p53 and Bax levels by 33% and 16%, respectively while increasing the Bcl-2 mRNA expression by 135%. Hence, the Bax/Bcl2 ratio and cytochrome c expression were subsequently reduced leading to decreased caspase 3 activity by 43%. Importantly, the anti-apoptotic property of NAC could be attributed to inactivation of MAPK signaling molecules; p38 and JNK, and enhancement of the ovarian vascular endothelial growth factor (VEGF) expression. Taken together, our results suggest that NAC can inhibit radiotherapy-induced POF while preserving ovarian function and structure through upregulating VEGF expression and suppressing NOX4/MAPK/p53 apoptotic signaling.


Subject(s)
Acetylcysteine/pharmacology , Gamma Rays/adverse effects , Ovary/drug effects , Primary Ovarian Insufficiency/prevention & control , Radiation-Protective Agents/pharmacology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Disease Models, Animal , Female , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/radiation effects , NADPH Oxidase 4/metabolism , Ovary/metabolism , Ovary/radiation effects , Ovary/ultrastructure , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Primary Ovarian Insufficiency/etiology , Primary Ovarian Insufficiency/metabolism , Primary Ovarian Insufficiency/pathology , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/metabolism
2.
Reprod Toxicol ; 43: 85-93, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24291358

ABSTRACT

The deleterious damage of reproductive function following radiotherapy is of increasing importance. In the present study, we investigated the impact of long-term sodium selenite (SS) treatment on radiotherapy-induced ovarian injury in a rat model. Two-week after radiation exposure vaginal cyclicity was arrested, and serum FSH level was elevated in irradiated female rats. SS significantly ameliorated ovarian and uterine oxidative stress induced by irradiation through decreasing the lipid peroxide level and increasing the glutathione level, and glutathione peroxidase activity. In the presence of SS, ovarian cytochrome c and caspase 3 expressions triggered by radiotherapy were decreased. SS significantly counteracted radiation-induced a widespread loss of ovarian follicles and caused further stimulation of follicular proliferation through enhancing PCNA expression. Despite such alteration in ovarian function, serum estradiol level did not change after irradiation, whereas SS significantly increased it. In conclusion, long-term SS treatment improved reproductive development, which was impaired by radiotherapy.


Subject(s)
Ovary/drug effects , Ovary/radiation effects , Radiation-Protective Agents/pharmacology , Sodium Selenite/pharmacology , Whole-Body Irradiation , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Estradiol/blood , Female , Fertility/drug effects , Fertility/radiation effects , Follicle Stimulating Hormone/blood , Ovary/pathology , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...