Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Hum Exp Toxicol ; 39(11): 1518-1527, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32519556

ABSTRACT

Ethephon, a member of the organophosphorus compounds, is one of the most widely used plant growth regulators for artificial ripening. Although million pounds of this chemical is being used annually, the knowledge regarding its molecular toxicity is yet not sufficient. The purpose of this study was to evaluate the potential developmental toxicity of ethephon using embryonic stem cell model. The mouse embryonic stem cells (mESCs) were exposed to various concentrations of ethephon and the viability, cell cycle alteration and changes in the gene expression profile were evaluated using high-throughput RNA sequencing. Further, the effect of ethephon on neural differentiation potential was examined. The results showed that ethephon at noncytotoxic doses induced cell cycle arrest in mESCs. Gene ontology enrichment analysis showed that terms related to cell fate and organismal development, including neuron fate commitment, embryo development and cardiac cell differentiation, were markedly enriched in ethephon-treated cells. Neural induction of mESCs in the presence of ethephon was inhibited and the expression of neural genes was decreased in differentiated cells. Results obtained from this work clearly demonstrate that ethephon affects the gene expression profile of undifferentiated mESCs and prevents neural differentiation. Therefore, more caution against the frequent application of ethephon is advised.


Subject(s)
Mouse Embryonic Stem Cells/drug effects , Organophosphorus Compounds/toxicity , Plant Growth Regulators/toxicity , Transcriptome/drug effects , Animals , Cell Cycle/drug effects , Cell Survival/drug effects , Cells, Cultured , Mice , Mouse Embryonic Stem Cells/metabolism , Neurogenesis/drug effects
2.
Hum Exp Toxicol ; 35(8): 839-50, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26519479

ABSTRACT

BACKGROUND: Chlorpyrifos (CP) is an organophosphorus pesticide that induces oxidative stress through the production of free radicals and depletes intracellular antioxidant reserves. In this study, the efficacy of three antioxidants (melatonin, coenzyme Q10 (CoQ10), and vinpocetine) on alleviation of toxic effects of CP was evaluated. MATERIALS AND METHODS: Cytotoxicity of CP, in the presence or absence of effective doses of melatonin, CoQ10, and vinpocetine, was determined in human peripheral blood lymphocytes after 72-h exposure. The levels of acetylcholinesterase (AChE) activity along with tumor necrosis factor α (TNF-α), as inflammatory index, were measured. Further, the viability and oxidative stress markers including cellular mitochondrial activity, cell death modes (apoptosis vs. necrosis), total antioxidant power (TAP), total thiol molecules (TTM), lipid peroxidation (LPO), and myeloperoxidase (MPO) activity were measured. RESULTS: CoQ10 and also the combination of the three antioxidants were the most notable in opposing toxicity of CP and led to increasing TAP and TTM; improvement of AChE activity; and lowering LPO, MPO, TNF-α, and apoptosis compared to CP alone. CONCLUSION: CP toxicity overwhelms the intracellular antioxidant defense mechanisms. Exogenous supplementation with antioxidants, such as the ones we have investigated, seems to be effective in the prevention of cytotoxicity of CP.


Subject(s)
Antioxidants/pharmacology , Chlorpyrifos/toxicity , Environmental Pollutants/toxicity , Lymphocytes/drug effects , Melatonin/pharmacology , Ubiquinone/analogs & derivatives , Vinca Alkaloids/pharmacology , Antioxidants/metabolism , Cell Culture Techniques , Cells, Cultured , Free Radicals/metabolism , Humans , Lymphocytes/metabolism , Oxidative Stress/drug effects , Ubiquinone/pharmacology
3.
Theriogenology ; 78(3): 620-31, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22541319

ABSTRACT

The objective was to evaluate ovarian functionality and oxidative response in hyperandrogenism-induced polycystic ovary (PCO) and the protective effects of immunomodulator drug (IMOD), an electromagnetically-treated, selenium-based, herbal medicine. Daily oral administration of letrozole (1 mg/kg) for 21 consecutive days induced ovarian cysts in female rats. An effective dose of IMOD (30 mg/kg per day) was given intraperitoneally for 21 days. Biomarkers of ovarian function, serum concentrations of estradiol, progesterone, testosterone, and ovarian prostaglandin-E (PGE), were analyzed. To determine the role of oxidative stress (OS) in hyperandrogenism-induced PCO, concentrations of cellular lipid peroxidation (LPO), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), peroxynitrite (ONOO), and tumor necrosis factor (TNF)-α as a marker of inflammation and apoptosis were measured in serum and ovaries. Letrozole-induced PCO resulted in significant increases in concentrations of lipid peroxidation and peroxynitrite in serum and ovary, but significantly decreased superoxide dismutase, catalase, and glutathione peroxidase. Serum concentrations of testosterone and TNF-α, and ovarian prostaglandin-E were increased (P < 0.001) in animals with cysts versus control, whereas estradiol and progesterone were decreased (P < 0.01 and P < 0.001, respectively). When compared with controls, letrozole induced irregular cycles and PCO characterized by a high incidence of subcapsular ovarian cysts with a diminished granulosa cell layer, luteinized granulosa cells in the cyst wall, significantly more atretic preantral and antral follicles, and absence of CL. There were almost no intact primary, secondary, and tertiary follicles in PCO rats. All end points assessed were significantly improved by IMOD and reached close to normal levels. In conclusion, the present study provided evidence that toxic free radicals and TNF-α were involved in the pathogenesis of PCO; furthermore, IMOD prevented ovarian histopathologic, endocrine, and biochemical alterations induced by hyperandrogenism.


Subject(s)
Antioxidants/administration & dosage , Hyperandrogenism/complications , Plant Extracts/administration & dosage , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/prevention & control , Selenium/administration & dosage , Animals , Antioxidants/analysis , Biomarkers/analysis , Estradiol/blood , Female , Hyperandrogenism/chemically induced , Letrozole , Lipid Peroxidation/drug effects , Nitriles/administration & dosage , Ovary/chemistry , Oxidative Stress/drug effects , Polycystic Ovary Syndrome/etiology , Progesterone/blood , Prostaglandins E/analysis , Rats , Rats, Wistar , Testosterone/blood , Triazoles/administration & dosage , Tumor Necrosis Factor-alpha/blood
4.
Hum Exp Toxicol ; 31(9): 887-97, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22076494

ABSTRACT

This study aimed to investigate the possible relationship between ovarian functionality and the oxidative response during cystogenesis induced by hyperandrogenization with letrozole and examine protective effect of the peroxisome proliferator-activated receptor gamma (PPAR-γ) agonist, pioglitazone (PIO), in polycystic ovary (PCO). Ovarian cysts were induced by oral administration of letrozol (1 mg/kg/day) for 21 consecutive days in the female rats. Effective dose of PIO (20 mg/kg/day) was administrated orally for 21 days. Serum estradiol (E), progesterone (P), testosterone (T), and the ovarian immunomodulator prostaglandin E (PGE) were analyzed as biomarkers of ovarian function. To determine the role of oxidative stress in PCO, the level of cellular lipid peroxidation (LPO), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), and peroxynitrite (ONOO), and tumor necrosis factor alpha (TNF-α) as a marker of inflammation and apoptosis were measured in serum and the ovaries. Letrozole-induced PCO in rats exhibited a significant increase in LPO and ONOO in serum and ovary while significantly decreased serum and ovarian SOD, CAT, and GPx. Serum T and TNF-α, and ovarian PGE were increased in animals with cysts compared with healthy controls, while E and P diminished. When compared to control group, letrozole-treated group showed irregular sexual cycles, polycystic ovaries characterized by high incidence of sub-capsular ovarian cyst with diminished or scant granulosa cell layer, increased number of atretic pre-antral and antral follicles and absence of corpus luteum. There were almost no primary, secondary, and tertiary follicles observed in PCO rats. All measured parameters were improved by PIO and reached close to normal levels. The present study further supports the role of oxidative/nitrosative stress and infiammatory responses in the pathogenesis of letrozole-induced hyperandrogenic PCO rats. Results indicate that PIO is able to exert direct antioxidative and anti-inflammatory effects on the endocrine, biochemical, and pathological alterations independent of its possible effects mediated via increased insulin sensitivity in hyperandrogenized PCO.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Oxidative Stress/drug effects , Polycystic Ovary Syndrome/drug therapy , Thiazolidinediones/therapeutic use , Tumor Necrosis Factor-alpha/metabolism , Animals , Aromatase Inhibitors , Estradiol/blood , Female , Hypoglycemic Agents/therapeutic use , Letrozole , Lipid Peroxidation/drug effects , Nitriles , PPAR gamma/agonists , Pioglitazone , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Progesterone/blood , Prostaglandins E/blood , Rats , Rats, Wistar , Testosterone/blood , Triazoles
SELECTION OF CITATIONS
SEARCH DETAIL
...