Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
1.
Mol Hum Reprod ; 27(7)2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34057472

RESUMO

During follicular development, a few dominant follicles develop to large antral dominant follicles, whereas the remaining follicles undergo atretic degeneration. Because vascularization on the follicular surface is a morphological feature of dominant follicles, we previously classified these follicles as vascularized follicles (VFs) and non-VFs (NVFs). In NVFs, progesterone producing genes were expressed similarly to that in VFs; however, the progesterone concentration in follicular fluid was low in large NVFs. Therefore, we estimated that progesterone is converted to cortisol, which induces the loss of follicular functions. In this study, we comparative analyzed the expression of genes for progesterone converting enzymes (Cytochrome (CYP)11B1, CYP21A2, Hydroxysteroid (HSD)11B2) and cortisol receptor (NR3C1) in VF and NVF granulosa cells. In NVFs, expression of cortisol producing genes (CYP11B1 and CYP21A2) was higher than in VFs. Expression of the gene for the cortisol metabolizing enzyme HSD11B2 in NVFs was significantly lower than in VFs. In NVFs, accompanied by increasing cortisol concentration in follicular fluid, apoptosis of granulosa and cumulus cells was observed. Cultivation with FSH and metyrapone (a CYP11B1 inhibitor) of NVF cumulus-oocyte complexes inhibited apoptosis of cumulus cells and induced cumulus cell proliferation and oocyte maturation. Cortisol-induced CYP11B1 and CYP21A2 expression, whereas FSH-induced HSD11B2 mRNA expression in VF granulosa cells in the presence of cortisol. Furthermore, an addition of 18ß-glycyrrhetinic acid (18-GA; a HSD17B2 inhibitor) to cortisol and FSH-containing medium increased apoptosis of VF granulosa cells. These results suggested that cortisol is a stimulatory factor that induces follicular atresia; furthermore, inhibition of cortisol production by FSH might increase the number of healthy preovulatory follicles in pigs.


Assuntos
Hormônio Foliculoestimulante/farmacologia , Atresia Folicular/efeitos dos fármacos , Hidrocortisona/farmacologia , 11-beta-Hidroxiesteroide Desidrogenases/biossíntese , 11-beta-Hidroxiesteroide Desidrogenases/genética , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Células do Cúmulo/efeitos dos fármacos , Células do Cúmulo/metabolismo , Indução Enzimática , Feminino , Hormônio Foliculoestimulante/fisiologia , Líquido Folicular/química , Regulação da Expressão Gênica , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Hidrocortisona/análise , Hidrocortisona/fisiologia , Metirapona/farmacologia , Modelos Biológicos , Progesterona/metabolismo , Receptores de Glucocorticoides/biossíntese , Receptores de Glucocorticoides/genética , Esteroide 11-beta-Hidroxilase/biossíntese , Esteroide 11-beta-Hidroxilase/genética , Esteroide 21-Hidroxilase/biossíntese , Esteroide 21-Hidroxilase/genética , Suínos
2.
Oxid Med Cell Longev ; 2020: 3648040, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33294120

RESUMO

Increased follicular atresia occurs with aging and results in reduced fecundity in laying chickens. Therefore, relieving follicular atresia of aging poultry is a crucial measure to maintain sustained high laying performance. As an antiaging agent, metformin was reported to play important roles in preventing aging in diverse animals. In this study, the physiological state of the prehierarchical follicles in the peak-laying hens (D280) and aged hens (D580) was compared, followed with exploration for the possible capacity of metformin in delaying atresia of the prehierarchical follicles in the aged D580 hens. Results showed that the capacity of yolk deposition within follicles declined with aging, and the point of endoplasmic reticulum- (ER-) mitochondrion contact decreased in the ultrastructure of the follicular cells. Meanwhile, the expression of apoptosis signaling genes was increased in the atretic small white follicles. Subsequently, the H2O2-induced follicular atresia model was established to evaluate the enhancing capacity of metformin on yolk deposition and inhibition of apoptosis in the atretic small white follicles. Metformin inhibited apoptosis through regulating cooperation of the mitochondrion-associated ER membranes and the insulin (PI3K/AKT) signaling pathway. Furthermore, metformin regulated calcium ion homeostasis to relieve ER-stress and inhibited release of mitochondrion apoptosis factors (BAD and caspase). Additionally, metformin activated PI3K/AKT that suppressed activation of BAD (downstream of the insulin signaling pathway) in the atretic follicles. Further, serum estrogen level and liver estrogen receptor-α expression were increased after dietary metformin supplementation in D580 hens. These results indicated that administration of dietary metformin activated the PI3K/AKT and calcium signaling pathway and enhanced yolk deposition to prevent chicken follicular atresia.


Assuntos
Envelhecimento/fisiologia , Sinalização do Cálcio/efeitos dos fármacos , Atresia Folicular/efeitos dos fármacos , Metformina/farmacologia , Animais , Caspases/metabolismo , Galinhas/metabolismo , Feminino , Atresia Folicular/fisiologia , Células da Granulosa/metabolismo , Peróxido de Hidrogênio/metabolismo , Folículo Ovariano/citologia , Folículo Ovariano/metabolismo , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo
3.
Artigo em Inglês | MEDLINE | ID: mdl-33198925

RESUMO

Methoxychlor (MXC), an organo-chlorine insecticide, is a reproductive toxicant in females, causing apoptosis-mediated follicular atresia. To elucidate the potentials of Methoxychlor as a geno-toxicant, granulosa cells of healthy antral follicles, exposed to MXC and antioxidant, N-acetyl-l-cysteine, were studied by the terminal deoxynucleotidyltransferase-dUTP nick end-labelling and single-cell gel electrophoresis (comet) assays. MXC caused DNA fragmentation, as revealed by the increased incidence of dark brown condensed TUNEL positive cells in contrast with lightly brown TUNEL negative cells with maximum TUNEL positive cells were observed in 100 µg/mL MXC treated groups. Quantitatively, maximum geno-toxicity was exhibited at highest MXC treatment with percent tail DNA as 17.87 ± 0.85, 41.16 ± 3.94, and 47.73 ± 3.71 in comparison with control (0.65 ± 0.03, 2.91 ± 0.27, and 7.16 ± 1.39) after 24, 48 and 72 h exposure duration, respectively. MXC treated groups exhibited Type 1-Type 3 comets as compared to Type 0 comets in control groups. Supplementation of NAC led to significant (p < 0.05) decline in geno-toxicity in MXC treated groups with maximum amelioration observed at 5 and 10 mM. Consequently, increased DNA damage attributed to the granulosa cells apoptosis in response to Methoxychlor exposure was significantly combated by NAC supplementation, preventing the geno-toxicity induced cyto-toxicity in GCs.


Assuntos
Acetilcisteína/farmacologia , Apoptose/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Metoxicloro/toxicidade , Animais , Ensaio Cometa , Feminino , Atresia Folicular/efeitos dos fármacos , Sequestradores de Radicais Livres/farmacologia , Cabras , Inseticidas/toxicidade , Folículo Ovariano/citologia , Análise de Célula Única/métodos
4.
Int J Mol Med ; 46(5): 1923-1937, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32901866

RESUMO

It has been suggested that oxidative stress involving reactive oxygen species (ROS) induces granulosa cell apoptosis, leading to follicular atresia, and that T­lymphokine­activated killer cell­originated protein kinase (TOPK) suppresses cancer cell apoptosis induced by several stimuli. However, it remains to be determined whether TOPK affects oxidative stress­induced granulosa cell apoptosis. The present study demonstrates that TOPK inhibition increases human granulosa COV434 cell apoptosis induced by hydrogen peroxide (H2O2). Co­treatment with the TOPK inhibitor, OTS514, in combination with H2O2 increased p53 acetylation and its expression, whereas it decreased Sirtuin 1 (SIRT1) expression, contributing to the promotion of apoptosis. In addition, the SIRT1 activator, resveratrol, or the SIRT1 inhibitor, Ex527, reduced or elevated H2O2­induced COV434 cell apoptosis, respectively. Furthermore, the p53 inhibitor, Pifithrin­µ, diminished the augmentation in poly(ADP­ribose) polymerase (PARP) cleavage induced by OTS514 plus H2O2, while the Mdm2 antagonist, Nutlin 3, increased PARP cleavage. Moreover, OTS514 further decreased the SIRT1 transcriptional activity decreased by H2O2, but promoted the H2O2­induced p53 or p21 transcriptional activity. Notably, the expression of exogenous p53 reduced SIRT1 transcriptional activity. Taken together, the findings of the present study demonstrate that TOPK inhibition promotes p53­mediated granulosa cell apoptosis through SIRT1 downregulation in response to H2O2. Therefore, it can be concluded that TOPK suppresses H2O2­induced apoptosis through the modulation of the p53/SIRT1 axis, suggesting a potential role of TOPK in the regulation of human granulosa cell apoptosis, leading to the promotion of abnormal follicular development.


Assuntos
Apoptose/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Sirtuína 1/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Acetilação/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Feminino , Atresia Folicular/efeitos dos fármacos , Atresia Folicular/metabolismo , Células da Granulosa/metabolismo , Células da Granulosa/patologia , Humanos , Peróxido de Hidrogênio/farmacologia , Poli(ADP-Ribose) Polimerases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transcrição Gênica/efeitos dos fármacos
5.
Neurotoxicology ; 79: 164-176, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32407858

RESUMO

Bisphenol-A (BPA) is an estrogenic chemical extensively used in industrial and household applications. The present study was conducted to investigate the effect of chronic exposure to BPA on the adult female neuroendocrine system. Herein, we found that expose of adult female mice to BPA (50 µg/kg) by oral gavage for 60 days (BPA mice) prolonged diestrus and decreased serum 17ß-estradiol (E2) concentration by reducing the number of antral follicles and corpora luteum. In comparison with controls, the levels of serum luteinizing hormone (LH), follicle stimulating hormone (FSH), hypothalamic gonadotrophin releasing hormone (GnRH) and the expression of kisspeptin in anteroventral periventricular nucleus (AVPV) decreased in BPA mice, which could be reversed by injecting kisspeptin-10 (i.c.v.). Treatment with BPA or estrogen receptor α (ERα) antagonist MPP, but not ERß antagonist PHTPP inhibited E2-induced AVPV-kisspeptin expression in ovariectomized mice. Use of ERα agonist PPT rather than ERß agonist DPN enhanced AVPV-kisspepetin expression, which decreased after treatment with BPA. The amplitude of the proestrus LH surge decreased in mice exposed to BPA, but was recovered by administering kisspeptin-10. The present study provides in vivo evidence that chronic exposure to a low dose of BPA suppressed ERα-induced activation of AVPV-kisspeptin neurons, leading to prolonged diestrus and reduced ovulation in adult female mice.


Assuntos
Compostos Benzidrílicos/toxicidade , Diestro/efeitos dos fármacos , Disruptores Endócrinos/toxicidade , Atresia Folicular/efeitos dos fármacos , Hipotálamo Anterior/efeitos dos fármacos , Kisspeptinas/metabolismo , Neurônios/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Fenóis/toxicidade , Animais , Apoptose/efeitos dos fármacos , Diestro/metabolismo , Regulação para Baixo , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/metabolismo , Feminino , Hipotálamo Anterior/metabolismo , Camundongos Endogâmicos ICR , Neurônios/metabolismo , Folículo Ovariano/patologia , Ovariectomia , Ovulação/efeitos dos fármacos , Transdução de Sinais , Fatores de Tempo
6.
Toxicol Appl Pharmacol ; 381: 114714, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31437492

RESUMO

Ovarian toxicity and infertility are major side effects of cancer therapy in young female cancer patients. We and others have previously demonstrated that doxorubicin (DOX), one of the most widely used chemotherapeutic chemicals, has a dose-dependent toxicity on growing follicles. However, it is not fully understood if the primordial follicles are the direct or indirect target of DOX. Using both prepubertal and young adult female mouse models, we comprehensively investigated the effect of DOX on all developmental stages of follicles, determined the impact of DOX on primordial follicle survival, activation, and development, as well as compared the impact of age on DOX-induced ovarian toxicity. Twenty-one-day-old CD-1 female mice were intraperitoneally injected with PBS or clinically relevant dose of DOX at 10 mg/kg once. Results indicated that DOX primarily damaged granulosa cells in growing follicles and oocytes in primordial follicles and DOX-induced growing follicle apoptosis was associated with the primordial follicle overactivation. Using the 5-day-old female mice with a more uniform primordial follicle population, our data revealed that DOX also directly promoted primordial follicle death and the DNA damage-TAp63α-C-CASP3 pathway was involved in DOX-induced primordial follicle oocyte apoptosis. Compared to 21-day- and 8-week-old female mice that were treated with the same dose of DOX, the 5-day-old mice had the most severe primordial follicle loss as well as the least degree of primordial follicle overactivation. Taken together, these results demonstrate that DOX obliterates mouse ovarian reserve through both primordial follicle atresia and overactivation and the DOX-induced ovarian toxicity is age dependent.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Doxorrubicina/efeitos adversos , Atresia Folicular/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Reserva Ovariana/efeitos dos fármacos , Animais , Dano ao DNA , Feminino , Camundongos , Folículo Ovariano/patologia
7.
Mol Reprod Dev ; 86(5): 566-575, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30806494

RESUMO

Ovarian follicular development is a complex process that requires codevelopment of the perifollicular vascular network, which is closely regulated by angiogenic factors, gonadotropins, sex steroids, and their metabolites. To detect the effects of vascular endothelial growth factor 120 (VEGF120), follicle-stimulating hormone (FSH), and 2-methoxyestradiol (2ME2) on follicular angiogenesis during development and atresia, we treated sexually immature and mature female mice with VEGF120, FSH, 2ME2, and FSH receptor (FSHR) antagonist singly or in combination via intraperitoneal injection. The number of follicles and their perifollicular angiogenesis and atresia rates at different developmental stages were examined in paraffin sections after hematoxylin and eosin staining. The results showed that the exogenous factors have specific and precise effects on developmental, angiogenesis, and atresia processes in follicles of different sizes in mature and immature mice. Perifollicular angiogenesis was regulated by VEGFA and closely related to follicular development and atresia. 2ME2 affected angiogenesis through VEGFA and might regulate atresia directly. FSH might control VEGFA function via both transcriptional and posttranscriptional mechanisms because FSHR was required for achieving VEGFA functions at all the follicular development stages. The present study presents insights into the mechanisms of FSH, 2ME2, and VEGFA in follicular development and disorders and provides a foundation for the development of new therapeutic strategies.


Assuntos
2-Metoxiestradiol/farmacologia , Hormônio Foliculoestimulante/farmacologia , Atresia Folicular , Neovascularização Fisiológica/efeitos dos fármacos , Folículo Ovariano , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Feminino , Atresia Folicular/efeitos dos fármacos , Atresia Folicular/metabolismo , Camundongos , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/metabolismo
8.
Toxicol Sci ; 166(1): 97-107, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30085306

RESUMO

With increasing improvements in cancer survival rates, it is critical to reduce the significant long-term side effects that afflict patients following treatment. For women, consequences of chemotherapy-induced damage to the reproductive system include infertility and premature menopause, which adversely effects cognition, mood, cardiovascular, bone, and sexual health, and increases the risk of early mortality. These long-term effects impact patient's life quality and highlight a significant and on-going burden on the health system after treatment. However, the precise mechanisms through which chemotherapeutic agents induce ovarian damage and primordial follicle depletion remain to be characterized. Hence, preventing the development of effective pharmacological methods to preserve fertility and improve quality of life after treatment. The chemotherapeutic agent 5-Fluorouracil (5FU) is not deemed cytotoxic to the ovary, however, risks to long-term fertility after multiple doses are not known. Therefore, we sought to evaluate the impact of 3, weekly doses of 5FU treatment on the ovary. Using a mouse model enabled accurate histomorphometric analysis of follicle numbers and ovarian structure and function, to accurately assess cumulative impact of 5FU on the ovary. This study clearly demonstrated that multidose 5FU treatment resulted in dramatic and progressive atresia of growing follicles and a profound decrease in ovarian volume due to reduced corpus luteum counts. However, primordial follicle numbers were unaffected. Thus, 5FU is unlikely to cause permanent infertility when administered to women of pre or reproductive age. Furthermore, this study suggests that depletion of the growing follicle population is insufficient to stimulate follicle activation and primordial follicle depletion.


Assuntos
Antimetabólitos Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Dano ao DNA , Fluoruracila/toxicidade , Atresia Folicular/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Animais , Antimetabólitos Antineoplásicos/administração & dosagem , Apoptose/genética , Corpo Lúteo/efeitos dos fármacos , Corpo Lúteo/crescimento & desenvolvimento , Corpo Lúteo/patologia , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Fluoruracila/administração & dosagem , Atresia Folicular/genética , Injeções Intraperitoneais , Camundongos Endogâmicos C57BL , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/patologia
9.
Cell Death Dis ; 9(6): 618, 2018 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-29795269

RESUMO

Female gametes are stored in the ovary in structures called primordial follicles, the supply of which is non-renewable. It is well established that DNA-damaging cancer treatments can deplete the ovarian reserve of primordial follicles, causing premature ovarian failure and infertility. The precise mechanisms underlying this chemotherapy-driven follicle loss are unclear, and this has limited the development of targeted ovarian-protective agents. To address this fundamental knowledge gap, we used gene deletion mouse models to examine the role of the DNA damage-induced pro-apoptotic protein, PUMA, and its transcriptional activator TAp63, in primordial follicle depletion caused by treatment with cyclophosphamide or cisplatin. Cyclophosphamide caused almost complete destruction of the primordial follicle pool in adult wild-type (WT) mice, and a significant destructive effect was also observed for cisplatin. In striking contrast, Puma-/- mice retained 100% of their primordial follicles following either genotoxic treatment. Furthermore, elimination of PUMA alone completely preserved fertility in cyclophosphamide-treated mice, indicating that oocytes rescued from DNA damage-induced death can repair themselves sufficiently to support reproductive function and offspring health. Primordial follicles were also protected in TAp63-/- mice following cisplatin treatment, but not cyclophosphamide, suggesting mechanistic differences in the induction of apoptosis and depletion of the ovarian reserve in response to these different chemotherapies. These studies identify PUMA as a crucial effector of apoptosis responsible for depletion of primordial follicles following exposure to cyclophosphamide or cisplatin, and this indicates that inhibition of PUMA may be an effective ovarian-protective strategy during cancer treatment in women.


Assuntos
Antineoplásicos/efeitos adversos , Proteínas Reguladoras de Apoptose/deficiência , Dano ao DNA , Fertilidade , Reserva Ovariana/fisiologia , Proteínas Supressoras de Tumor/deficiência , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Cisplatino/efeitos adversos , Ciclofosfamida/efeitos adversos , Feminino , Atresia Folicular/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/patologia , Reserva Ovariana/efeitos dos fármacos , Fosfoproteínas/deficiência , Fosfoproteínas/metabolismo , Transativadores/deficiência , Transativadores/metabolismo , Proteínas Supressoras de Tumor/metabolismo
10.
J Assist Reprod Genet ; 35(6): 1053-1060, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29589294

RESUMO

PURPOSE: Undesirable side effects of cancer treatments are common and include damage to the ovary, and depletion of the follicle reserve, which if severe enough, can lead to infertility and early menopause. Antimetabolite drugs, such as 5-fluorouracil (5-FU), are not considered to be detrimental to the ovary, but the ovotoxicity of 5-FU has not been evaluated in any detail. The purpose of this study was to evaluate the effects of 5-FU on follicle number. METHODS: In this study, adult female C57Bl6 mice (n = 4-6 animals/group) received a single dose of saline or 5-FU (150 mg/kg) and markers of ovarian damage and follicle depletion were assessed 12 h and 7 days later. RESULTS: Exposure to 5-FU did not alter primordial and primary follicle numbers. Atresia of secondary and antral follicles was increased significantly 12 h after 5-FU treatment, but atresia rates returned to levels similar to that of saline treated controls at 7 days. The number of corpora lutea were reduced 7 days after exposure to 5-FU, possibly as a consequence of earlier follicular atresia. CONCLUSIONS: These findings suggest that a single dose of 5-FU is mildly ovotoxic, but any effects on ovarian function are likely transient because the primordial follicle population is not depleted. Collectively, these data support the notion that 5-FU is unlikely to impact on the long-term fertility of women.


Assuntos
Antimetabólitos Antineoplásicos/toxicidade , Fluoruracila/toxicidade , Atresia Folicular/efeitos dos fármacos , Folículo Ovariano/patologia , Animais , Relação Dose-Resposta a Droga , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Folículo Ovariano/efeitos dos fármacos
11.
Eur J Obstet Gynecol Reprod Biol ; 222: 19-24, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29331755

RESUMO

OBJECTIVE(S): We aimed to analyze if anti-apoptotic agent sphingosine-1-phosphate offers protection against in vitro follicle atresia during culture of human ovarian cortical samples. STUDY DESIGN: A translational research study of ex-vivo and in-vitro models of human ovarian tissue. MATERIAL AND METHODS: Ovarian cortical tissue fragments (1 × 0.5 cm) were obtained from young patients (n = 15 mean age ±â€¯SD: 29.4 ±â€¯2.5) undergoing laparoscopic excision of benign ovarian cysts. The samples were cultured for 4 days in 24-well format culture plate using conventional culture techniques. S1P was added to culture media at 200 and 400 µM concentrations. At the end of culture period the samples were processed for both histomorphological assessment and detection of apoptosis with immunohistochemistry and western blot methods using apoptosis marker cleaved caspase-3. In vitro estradiol (E2) and AMH productions of the samples were measured with ELISA. Follicle counts were expressed as the mean number of follicles per mm2. RESULTS: The mean numbers of primordial and secondary follicles were 3.2 ±â€¯0.4 and 0.7 ±â€¯0.2 respectively, in the fresh fixed uncultured samples. After four days of culture their numbers were significantly decreased to 0.8 ±â€¯0.2 (p < 0.01) and 0.1 ±â€¯0.05 (p < 0.05) respectively, in the control samples cultured without S1P compared to fresh fixed samples. S1P treatment decreased follicle atresia and significantly higher number of primordials (2.3 ±â€¯0.3, p < 0.01) and secondary follicles (0.5 ±â€¯0.1, p < 0.05) survived in the samples after 4 day culture period compared to those cultured without S1P. In line with this there was dose-dependent decrease in the protein expression of cleaved caspase-3 on western blot and in the number of apoptotic follicles stained positive for cleaved caspase-3 on immunohistochemistry in the samples incubated with S1P at 200 and 400 µM concentrations. Furthermore, those samples incubated with S1P produced significantly higher amounts of E2 (2339 ±â€¯321 vs. 1156 ±â€¯125 pg/mL respectively, p < 0.01) compared to control samples. CONCLUSIONS: These results suggest that S1P promotes follicle survival in human ovarian cortical samples in vitro.


Assuntos
Apoptose/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Folículo Ovariano/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Esfingosina/análogos & derivados , Adulto , Hormônio Antimülleriano/metabolismo , Biomarcadores/metabolismo , Western Blotting , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Estradiol/metabolismo , Feminino , Atresia Folicular/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Cistos Ovarianos/patologia , Cistos Ovarianos/cirurgia , Folículo Ovariano/metabolismo , Folículo Ovariano/patologia , Proteólise/efeitos dos fármacos , Esfingosina/farmacologia , Técnicas de Cultura de Tecidos
12.
Food Chem Toxicol ; 112: 19-25, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29258955

RESUMO

Given the importance of bisphenol A (BPA) as a xenoestrogen and its potential effects on human and animal health, we evaluated BPA exposure's short-term effects on follicular development, yolk protein vitellogenin (VTG) production and aromatase expression in female zebrafish. Histological modifications were observed along with increased presence of atretic follicles. Whole-body VTG concentration increased with the dose of BPA exposure. In contrast, expression of Cyp19a mRNA in the ovaries of BPA-exposed fish exhibited an apparent non-monotonic response curve, marked by downregulation at 1 µg/L BPA, upregulation at 10 µg/L BPA, and a return to downregulation at 100 µg/L BPA and higher doses. Ovaries only exhibited significant increases in follicular atresia and VTG concentration after exposure to 100 µg/L BPA and higher doses. Ovarian histopathology, aromatase Cyp19a transcript levels and whole-body VTG protein abundance may be good biomarkers for early detection of environmental BPA exposure.


Assuntos
Compostos Benzidrílicos/toxicidade , Disruptores Endócrinos/toxicidade , Atresia Folicular/efeitos dos fármacos , Ovário/efeitos dos fármacos , Fenóis/toxicidade , Poluentes Químicos da Água/toxicidade , Peixe-Zebra , Animais , Aromatase/genética , Biomarcadores/metabolismo , Relação Dose-Resposta a Droga , Regulação para Baixo , Monitoramento Ambiental/métodos , Estradiol/metabolismo , Feminino , Ovário/enzimologia , Ovário/patologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Regulação para Cima , Vitelogeninas/metabolismo , Proteínas de Peixe-Zebra/genética
13.
Biol Reprod ; 97(3): 449-465, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-29024988

RESUMO

Chemotherapy can cause early menopause or infertility in women and have a profound negative impact on the quality of life of young female cancer survivors. Various factors are known to influence the risk of chemotherapy-induced ovarian failure, including the drug dose and treatment duration; however, the scheduling of dose administration has not yet been evaluated as an independent risk factor. We hypothesized that low-dose metronomic (LDM) chemotherapy scheduling would be less detrimental to ovarian function than the traditional maximum tolerated dose (MTD) strategy. In vitro, MTD cyclophosphamide exposure resulted in decreased proliferation and increased granulosa cell apoptosis, while cells treated with LDM cyclophosphamide were not different from untreated controls. Treatments of MTD cyclophosphamide induced high levels of follicle atresia and enhanced follicle recruitment in mice. In contrast, LDM delivery of an equivalent dose of cyclophosphamide reduced growing follicle numbers, but was not associated with higher levels of follicle atresia or recruitment. MTD cyclophosphamide induced significant vascular disruption and DNA damage in vivo, while LDM chemotherapy with equal cumulative amounts of cyclophosphamide was not different from controls. MTD chemotherapy also had a negative effect on mouse-fertility outcomes. Our findings suggest that LDM scheduling could potentially minimize the long-term effects of cyclophosphamide on female fertility by preventing follicle depletion from enhanced activation.


Assuntos
Antineoplásicos Alquilantes/administração & dosagem , Antineoplásicos Alquilantes/toxicidade , Ciclofosfamida/administração & dosagem , Ciclofosfamida/toxicidade , Fertilidade/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Ovário/efeitos dos fármacos , Animais , Vasos Sanguíneos/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dano ao DNA , Sistemas de Liberação de Medicamentos , Ciclo Estral/efeitos dos fármacos , Feminino , Atresia Folicular/efeitos dos fármacos , Células da Granulosa/ultraestrutura , Dose Máxima Tolerável , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Ratos
14.
Biosci Trends ; 11(4): 427-438, 2017 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-28717062

RESUMO

Female fertility declines with age as the number of ovarian follicles decreases and aneuploidy increases. Degradation of the cohesin complex might be responsible for age-related aneuploidy. Dehydroepiandrosterone (DHEA) can improve the ovarian reserve and reduce the rate of aneuploidy, but the relationship between DHEA and cohesin levels in oocytes is still unknown. The aim of the current study was to evaluate the effect of the supplement DHEA on ovarian function, including the number of follicles and cohesin levels in oocytes. C57BL/6J mice at 3 weeks, 6 weeks, 12 weeks, 6 months, and 10 months of age were used to obtain a systematic view into follicle apoptosis and cohesin levels in oocytes. Nine-month-old C57BL/6J mice were administered saline (n = 5), 17ß-estradiol (100 µg/kg per day, n = 5), or DHEA (5mg/Kg per day, n = 5). After 4 weeks, aged mice were weighed and sacrificed, and ovarian tissue samples were prepared. Anti-VASA staining and HE staining were used to count the number of follicles. Anti-γH2AX staining and TUNEL were used to measure follicle apoptosis and immunofluorescent staining was used to detect the levels of three oocyte cohesin subunits: REC8, SMC1ß, and SMC3. Administration of the supplements 17ß-estradiol and DHEA to aged mice increased the number of primordial and primary follicles and decreased the age-related apoptosis of follicles. Levels of the cohesin subunits REC8 and SMC1ß declined with age, but DHEA and 17ß-estradiol tended to delay that decline. The supplement DHEA increased the number of primordial and primary follicles in aged mice by inhibiting follicle apoptosis and tended to delay the decrease in cohesin levels in oocytes.


Assuntos
Envelhecimento/metabolismo , Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Desidroepiandrosterona/farmacologia , Oócitos/citologia , Oócitos/metabolismo , Animais , Hormônio Antimülleriano/sangue , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Desidroepiandrosterona/administração & dosagem , Estradiol/sangue , Feminino , Atresia Folicular/sangue , Atresia Folicular/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Complexos Multiproteicos/metabolismo , Oócitos/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/metabolismo , Subunidades Proteicas/metabolismo , Coesinas
15.
Environ Toxicol ; 32(10): 2244-2255, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28646569

RESUMO

Present study was performed in order to update the possible mechanism(s), involving in nanosilver particles (NSPs)-induced detrimental impacts in ovarian tissue. For this purpose, 24 mature female rats were divided into control and 0.5, 1, 5 mg/kg NSPs-received groups (intraperitoneally, for 35 days). Follicular growth and atresia, ovarian total antioxidant capacity (TAC), malondialdehyde (MDA), superoxide dismutase (SOD) contents, serum estrogen (E2 ) level and macrophages infiltration were investigated. Moreover, ovarian angiogenesis, cellular mRNA damage and cytochrome aromatase CYP19 expression were analyzed. The NSPs enhanced follicular atresia diminished E2, reduced TAC and SOD level, elevated MDA content and up-regulated macrophages infiltration. Cellular mRNA damage, impaired angiogenesis and diminished CYP19 expression were revealed in NSPs-received groups. Therefore NSPs by down-regulating aromatization, reduce E2 synthesis which then it leads to impaired angiogenesis. The impaired angiogenesis in turn down-regulates ovarian antioxidant status, which partially enhances follicular atresia by triggering lipid peroxidation and mRNA damage.


Assuntos
Aromatase/metabolismo , Atresia Folicular/efeitos dos fármacos , Nanopartículas Metálicas/toxicidade , Ovário/efeitos dos fármacos , Estresse Oxidativo , Prata/toxicidade , Animais , Antioxidantes/metabolismo , Estradiol/sangue , Feminino , Peroxidação de Lipídeos/efeitos dos fármacos , Malondialdeído/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Ovário/irrigação sanguínea , Ovário/metabolismo , RNA Mensageiro/metabolismo , Ratos Wistar , Superóxido Dismutase/metabolismo , Testosterona/sangue
16.
Mol Cell Endocrinol ; 433: 56-65, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27235859

RESUMO

Anti-Müllerian hormone (AMH) is a member of the transforming growth factor-ß superfamily and plays a crucial role in testicular and ovarian functions. In clinical practice, AMH is used as a diagnostic and/or prognostic marker in women in association with ovulation induction and in various pathophysiological conditions. Despite widespread clinical use of AMH, our mechanistic understanding of AMH actions in regulating follicular development is limited. Using a mouse model, we in this study report that in vivo AMH treatment while stalls follicular development and inhibits ovulation, also prevents follicular atresia. We further show that these AMH actions are mediated through induction of two miRNAs, miR-181a and miR-181b, which regulate various aspects of FSH signaling and follicular growth, ultimately affecting downstream gene expression and folliculogenesis. We also report that in this mouse model AMH pre-treatment prior to superovulation improves oocyte yield. These studies, therefore, offer new mechanistic insight into AMH actions in folliculogenesis and point toward potential utilization of AMH as a therapeutic agent.


Assuntos
Hormônio Antimülleriano/farmacologia , Folículo Ovariano/efeitos dos fármacos , Animais , Estradiol/metabolismo , Feminino , Hormônio Foliculoestimulante/metabolismo , Atresia Folicular/efeitos dos fármacos , Atresia Folicular/metabolismo , Glicoproteínas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Folículo Ovariano/metabolismo , Indução da Ovulação/métodos , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Superovulação/efeitos dos fármacos
17.
Toxicol Appl Pharmacol ; 303: 65-78, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27129868

RESUMO

Resveratrol (RVT), a polyphenolic component in grapes and red wine, has been known for its cytoprotective actions against several diseases. However, beneficial effects of RVT against early exposure to endocrine disrupting chemicals (EDCs) have not been understood. EDCs are linked to several ovarian diseases such as premature ovarian failure, polycystic ovary syndrome, early menopause and infertility in women. Hexavalent chromium (CrVI) is a heavy metal EDC, and widely used in >50 industries. Environmental contamination with CrVI in the US is rapidly increasing, predisposing the human to several illnesses including cancers and still birth. Our lab has been involved in determining the molecular mechanism of CrVI-induced female infertility and intervention strategies to mitigate CrVI effects. Lactating mother rats were exposed to CrVI (50ppm potassium dichromate) from postpartum days 1-21 through drinking water with or without RVT (10mg/kg body wt., through oral gavage daily). During this time, F1 females received respective treatments through mother's milk. On postnatal day (PND) 25, blood and the ovary, kidney and liver were collected from the F1 females for analyses. CrVI increased atresia of follicles by increasing cytochrome C and cleaved caspase-3; decreasing antiapoptotic proteins; decreasing estradiol (E2) biosynthesis and enhancing metabolic clearance of E2, increasing oxidative stress and decreasing endogenous antioxidants. RVT mitigated the effects of CrVI by upregulating cell survival proteins and AOXs; and restored E2 levels by inhibiting hydroxylation, glucuronidation and sulphation of E2. This is the first study to report the protective effects of RVT against any toxicant in the ovary.


Assuntos
Cromo/toxicidade , Ovário/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Estilbenos/farmacologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Carcinógenos/toxicidade , Catalase/metabolismo , Poluentes Ambientais/toxicidade , Estradiol/sangue , Estradiol/metabolismo , Feminino , Atresia Folicular/efeitos dos fármacos , Glutationa Peroxidase/metabolismo , Rim/efeitos dos fármacos , Rim/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Oócitos/efeitos dos fármacos , Oócitos/patologia , Ovário/patologia , Gravidez , Progesterona/sangue , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Resveratrol , Superóxido Dismutase/metabolismo , Testosterona/sangue
18.
Sci Rep ; 6: 23120, 2016 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-26979400

RESUMO

Chemotherapy (CTx)-induced premature ovarian failure (POF) in woman remains clinically irreversible. Amniotic fluid stem cells (AFSCs) have shown the potential to treat CTx-induced POF; however, the underlying mechanism is unclear. Here we demonstrate that AFSC-derived exosomes recapitulate the anti-apoptotic effect of AFSCs on CTx-damaged granulosa cells (GCs), which are vital for the growth of ovarian follicles. AFSC-derived exosomes prevent ovarian follicular atresia in CTx-treated mice via the delivery of microRNAs in which both miR-146a and miR-10a are highly enriched and their potential target genes are critical to apoptosis. The down-regulation of these two miRNAs in AFSC-derived exosomes attenuates the anti-apoptotic effect on CTx-damaged GCs in vitro. Further, the administration of these miRNAs recapitulates the effects both in vitro and in vivo, in which miR-10a contributes a dominant influence. Our findings illustrate that miR-10a has potential as a novel therapeutic agent for the treatment of POF.


Assuntos
MicroRNAs/uso terapêutico , Folículo Ovariano/fisiologia , Insuficiência Ovariana Primária/prevenção & controle , Células-Tronco/metabolismo , Líquido Amniótico/citologia , Animais , Antineoplásicos Alquilantes/efeitos adversos , Apoptose , Sequência de Bases , Sítios de Ligação , Sobrevivência Celular , Células Cultivadas , Técnicas de Cocultura , Ciclofosfamida/efeitos adversos , Exossomos/metabolismo , Feminino , Atresia Folicular/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Camundongos Endogâmicos ICR , MicroRNAs/metabolismo , Folículo Ovariano/efeitos dos fármacos , Insuficiência Ovariana Primária/induzido quimicamente , Interferência de RNA , Transplante de Células-Tronco
19.
Mol Cell Endocrinol ; 409: 103-12, 2015 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-25817543

RESUMO

The intronic microRNA let-7g controls cell differentiation and proliferation during angiogenesis and oncogenesis. Here, we demonstrate that let-7g regulates granulosa cell (GC) apoptosis and follicular atresia in the pig ovary. Bioinformatics analyses and luciferase reporter assays showed that transforming growth factor-ß type 1 receptor (TGFBR1) is a let-7g target. Overexpression of let-7g induced apoptosis of porcine GCs in vitro and repressed the mRNA and protein levels of TGFBR1, as well as the level of phosphorylated SMAD3 (p-SMAD3) protein. RNA interference-mediated knockdown of TGFBR1 and inhibitor LY2157299-mediated blocking of TGFBR1 significantly increased the rate of apoptosis of GCs and Caspase-3 activity. In addition, treatment of porcine GCs with TGF-ß1 reduced the level of let-7g and increased the levels of the TGFBR1 mRNA and proteins significantly. Overall, these results demonstrate that let-7g regulates the apoptosis of GCs in the pig ovary by targeting TGFBR1 and down-regulating the TGF-ß signaling pathway.


Assuntos
Atresia Folicular/genética , Células da Granulosa/fisiologia , MicroRNAs/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Feminino , Atresia Folicular/efeitos dos fármacos , Atresia Folicular/metabolismo , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Fosforilação , Pirazóis/farmacologia , Quinolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/metabolismo , Suínos , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/farmacologia
20.
Biol Reprod ; 92(3): 67, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25568306

RESUMO

Environmental exposure to endocrine-disrupting chemicals (EDCs) is one cause of premature ovarian failure (POF). Hexavalent chromium (CrVI) is a heavy metal EDC widely used in more than 50 industries, including chrome plating, welding, wood processing, and tanneries. Recent data from U.S. Environmental Protection Agency indicate increased levels of Cr in drinking water from several American cities, which potentially predispose residents to various health problems. Recently, we demonstrated that gestational exposure to CrVI caused POF in F1 offspring. The current study was performed to identify the molecular mechanism behind CrVI-induced POF. Pregnant rats were treated with 25 ppm of potassium dichromate from Gestational Day (GD) 9.5 to GD 14.5 through drinking water, and the fetuses were exposed to CrVI through transplacental transfer. Ovaries were removed from the fetuses or pups on Embryonic Day (ED) 15.5, ED 17.5, Postnatal Day (PND) 1, PND 4, or PND 25, and various analyses were performed. Results showed that gestational exposure to CrVI: 1) increased germ cell/oocyte apoptosis and advanced germ cell nest (GCN) breakdown; 2) increased X-prolyl aminopeptidase (Xpnpep) 2, a POF marker in humans, during GCN breakdown; 3) decreased Xpnpep2 during postnatal follicle development; and 4) increased colocalization of Xpnpep2 with Col3 and Col4. We also found that Xpnpep2 inversely regulated the expression of Col1, Col3, and Col4 in all the developmental stages studied. Thus, CrVI advanced GCN breakdown and increased follicle atresia in F1 female progeny by targeting Xpnpep2.


Assuntos
Aminopeptidases/fisiologia , Cromo/efeitos adversos , Cromo/farmacologia , Fase Folicular/efeitos dos fármacos , Óvulo/efeitos dos fármacos , Insuficiência Ovariana Primária/induzido quimicamente , Insuficiência Ovariana Primária/fisiopatologia , Animais , Apoptose/efeitos dos fármacos , Carcinógenos Ambientais/efeitos adversos , Carcinógenos Ambientais/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Colágeno Tipo I/fisiologia , Colágeno Tipo III/fisiologia , Colágeno Tipo IV/fisiologia , Modelos Animais de Doenças , Feminino , Atresia Folicular/efeitos dos fármacos , Atresia Folicular/fisiologia , Fase Folicular/fisiologia , Ovário/efeitos dos fármacos , Ovário/fisiologia , Óvulo/fisiologia , Gravidez , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA