Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Eur J Obstet Gynecol Reprod Biol ; 294: 217-221, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38301500

ABSTRACT

OBJECTIVE: Recent studies have suggested that endometriosis could be the result of excessive activation of signal transducer and activator of transcription 3 (STAT3), which is associated with the regulation of essential cellular mechanisms such as proliferation, invasion, and apoptosis. That finding implies that regulating STAT3 activation could play a key role in treating endometriosis. In the present study, we aimed to evaluate whether the anti-endometriotic effects of dienogest is mediated by the regulation of STAT3 activation. STUDY DESIGN: STAT3 activation was evaluated in normal endometrial and ovarian endometriotic tissues obtained from patients with/without preoperative dienogest treatment. A subsequent in vitro analysis with endometriotic cyst stromal cells (ECSCs) was used to confirm the direct influence of dienogest in STAT3 activation. RESULT: STAT3 activation is significantly higher in endometriotic tissues from non-treated patients than in normal endometrial tissues, and that difference is reversed by preoperative administration of dienogest. Similarly, the inhibitory effects of dienogest on STAT3 activation are demonstrated by in vitro results showing that dienogest treatment significantly inhibits IL-6-stimulated STAT3 activation in cultured ECSCs. That inhibition was accompanied by decreased expression of proliferative (PCNA), invasive (MMP-2), and anti-apoptotic (BCL-2) proteins. Furthermore, downregulating STAT3 activity with siRNA decreased PCNA, MMP-2, and BCL-2 expression in IL-6-treated ECSCs. CONCLUSION: Dienogest inhibits STAT3 activation in ECSCs, which affects their proliferation, invasiveness, and apoptosis.


Subject(s)
Endometriosis , Nandrolone/analogs & derivatives , Female , Humans , Endometriosis/genetics , Matrix Metalloproteinase 2 , STAT3 Transcription Factor/metabolism , Interleukin-6/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/pharmacology , Stromal Cells/metabolism , Endometrium/metabolism
2.
Mol Hum Reprod ; 28(4)2022 04 01.
Article in English | MEDLINE | ID: mdl-35333355

ABSTRACT

The NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome is a cytosolic multi-protein complex that induces inflammation and is known to be regulated negatively by autophagy. Previous studies reported an abnormal induction of autophagy linked to progesterone resistance in human endometriotic cells. Therefore, an aberrant autophagy induction response to progesterone might contribute to the altered inflammatory response observed in endometriotic tissues. To evaluate this hypothesis, we elucidate whether regulation of the NLRP3 inflammasome by ovarian steroids is mediated by autophagy in human endometrial stromal cells (normal endometrial stromal cells (NESCs)) from patients with uterine leiomyoma (presumed normal) and whether abnormal autophagy induction in endometriotic cyst stromal cells (ECSCs) affects NLRP3 inflammasome-induced interleukin-1ß (IL-1ß) production. Our results show that estrogen enhanced NLRP3 inflammasome activation in NESCs, resulting in increased IL-1ß production. Progesterone decreased NLRP3 inflammasome activity with an increase in autophagy induction in estrogen-treated NESCs. Inhibition of NLRP3 inflammasome activity by progesterone was blocked by autophagy inhibition. However, progesterone failed to change NLRP3 inflammasome activity and autophagy induction in estrogen-treated ECSCs. In contrast, dienogest, a specific progesterone receptor agonist, reduced NLRP3 inflammasome-mediated IL-1ß production through autophagy induction in ECSCs. Furthermore, autophagy induction was decreased and NLRP3 inflammasome activity was increased in endometriotic tissues, which was reversed by preoperative administration of dienogest. In conclusion, our results suggest that progesterone inhibits NLRP3 inflammasome activation through autophagy in endometrial stromal cells. However, this inhibitory effect is attenuated in endometriotic stromal cells due to an aberrant autophagic response to progesterone, which could lead to an altered inflammatory response in endometriosis.


Subject(s)
Cysts , Endometriosis , Autophagy , Endometriosis/genetics , Female , Humans , Inflammasomes/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Progesterone/pharmacology , Stromal Cells
3.
Mol Hum Reprod ; 27(2)2021 02 05.
Article in English | MEDLINE | ID: mdl-33507306

ABSTRACT

Endoplasmic reticulum (ER) stress serves as a key modulator of the inflammatory response by controlling nuclear factor-kappaB (NF-κB) signaling. Previous studies from our laboratory have reported an abnormal induction of ER stress linked to progesterone resistance in human endometriotic cells. Therefore, an aberrant ER stress response to progesterone might contribute to the altered inflammatory response observed in endometriotic tissues. To evaluate this hypothesis, we investigated whether ER stress is involved in regulation of NF-κB in endometrial stromal cells and whether induction of aberrant ER stress in endometriotic stromal cells affects pro-inflammatory cytokine production. We found that tunicamycin-induced ER stress inhibited NF-κB activation and pro-inflammatory cytokine (IL-6 and COX2) production in TNF-α- or IL-1ß-treated normal endometrial stromal cells (NECSs). Tunicamycin increased the expression of A20 and C/EBPß, which are negative regulators of NF-κB, and this increase inhibited NF-κB activity in NESCs incubated with TNF-α or IL-1ß. Similarly, progesterone increased A20 and C/EBPß expression through upregulation of ER stress in NESCs, resulting in inhibition of NF-κB activity and IL-6 and COX2 production. However, progesterone had no significant effects on induction of ER stress, A20 or C/EBPß expression, NF-κB activity or IL-6 or COX2 production in ovarian endometriotic cyst stromal cells (ECSCs). In contrast, upregulation of ER stress by tunicamycin significantly reduced IL-6 and COX2 production by inhibiting NF-κB activity in ECSCs. In conclusion, our results suggest that NF-κB activity in endometriotic stromal cells was not inhibited because of an aberrant ER stress response to progesterone, resulting in an increase in pro-inflammatory cytokine production.


Subject(s)
Endometriosis/metabolism , Endometrium/drug effects , Endoplasmic Reticulum Stress/drug effects , NF-kappa B/metabolism , Progesterone/pharmacology , Stromal Cells/drug effects , CCAAT-Enhancer-Binding Protein-beta/metabolism , Case-Control Studies , Cells, Cultured , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Endometriosis/pathology , Endometrium/metabolism , Endometrium/pathology , Endoplasmic Reticulum Chaperone BiP , Female , Heat-Shock Proteins/metabolism , Humans , Inflammation Mediators/metabolism , Signal Transduction , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism
4.
Mol Hum Reprod ; 26(1): 30-39, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31814016

ABSTRACT

Dienogest, a specific progesterone receptor agonist, is used in the treatment of endometriosis. However, it is still unclear as to the mechanisms of therapeutic effects on endometriosis. Our recent study showed that endometriosis may be the result of aberrant endoplasmic reticulum (ER) stress induction due to progesterone resistance. This finding suggests that the regulation of ER stress induction may play a key role in treatment of endometriosis. Therefore, the anti-endometriotic effects of dienogest may be mediated by regulation of ER stress. To test this hypothesis, we elucidate whether dienogest affects endometriotic stromal cell apoptosis, proliferation and invasiveness by modulating ER stress-induced CCAAT/enhancer-binding protein homologous protein (CHOP) expression. Specifically, PRKR-like ER kinase (PERK)/eukaryotic initiation factor 2α (eIF2α)/activating transcription factor 4 (ATF4), inositol-requiring kinase 1 (IRE1)/TNF receptor-associated factor 2 (TRAF2)/apoptosis signal-regulating kinase 1 (ASK1)/c-Jun N-terminal kinase (JNK) signaling, and downstream CHOP were evaluated to determine the involved ER stress-mediated regulation mechanism of CHOP expression. Our results show that progesterone treatment did not have any significant effects on ER stress, apoptosis, proliferation, and invasion in estrogen-treated endometriotic cyst stromal cells (ECSCs). However, dienogest treatment upregulated the induction of ER stress. It also led to increased apoptosis, and decreased proliferation and invasiveness. These dienogest-induced changes in apoptosis, proliferation and invasiveness were reversed by the ER stress inhibitor salubrinal. Furthermore, dienogest-induced ER stress increased CHOP expression through activation of both PERK/elf2α/ATF4 and IRE1/TRAF2/ASK1/JNK signaling. This upregulation was blocked by transfection with PERK and IRE1 siRNA, which decreased apoptosis and increased the proliferation and invasiveness of dienogest-treated ECSCs. Taken together, our findings indicate that dienogest enhances ER stress induction in endometriotic stromal cells, which affects apoptosis, proliferation and invasiveness via CHOP upregulation.


Subject(s)
Cysts/drug therapy , Endometriosis/drug therapy , Endoplasmic Reticulum Stress/drug effects , Hormone Antagonists/pharmacology , Nandrolone/analogs & derivatives , Transcription Factor CHOP/genetics , eIF-2 Kinase/genetics , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Adult , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cysts/genetics , Cysts/metabolism , Cysts/pathology , Endometriosis/genetics , Endometriosis/metabolism , Endometriosis/pathology , Endometrium/drug effects , Endometrium/metabolism , Endometrium/pathology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/genetics , Endoribonucleases/metabolism , Female , Gene Expression Regulation , Humans , MAP Kinase Kinase Kinase 5/genetics , MAP Kinase Kinase Kinase 5/metabolism , MAP Kinase Signaling System , Nandrolone/pharmacology , Progesterone/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , Transcription Factor CHOP/agonists , Transcription Factor CHOP/metabolism , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/metabolism
5.
Mol Hum Reprod ; 25(3): 101-110, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30657961

ABSTRACT

Endoplasmic reticulum (ER) stress is known to reduce invasiveness in some cancer cells by inhibiting the AKT/mTOR pathway. A previous study from our laboratory suggested that ER stress is promoted by progesterone in human endometrial cells, which suggests that progesterone may inhibit endometrial cell invasiveness by up-regulating ER stress. Therefore, aberrant ER stress in response to progesterone may contribute to the altered invasiveness found in endometriotic tissues. To test this hypothesis, we elucidate whether ER stress is involved in regulation of human endometrial cell invasiveness through the AKT/mTOR pathway and if this involvement is associated with altered invasiveness in endometriotic cells. Specifically, we sought to determine the effects of ER stress on AKT/mTOR pathway by evaluating ER stress-mediated CHOP/TRIB3 signaling, a negative regulator of AKT. We found that ER stress marker GRP78 expression increased with CHOP and TRIB3 expression in normal endometrial stromal cells (NESCs) treated with tunicamycin, and this increase was accompanied by decreased AKT and mTOR activity and cellular invasiveness. Similarly, progesterone increased GRP78, CHOP and TRIB3 expression in NESCs. Subsequently, inhibition of AKT and mTOR activity decreased cellular invasiveness. This progesterone-induced decrease in cellular invasiveness was reversed by inhibition of ER stress. In contrast, progesterone did not change CHOP, TRIB3, AKT, mTOR or invasiveness in endometriotic cyst stromal cells. In contrast to normal endometrium, endometriotic tissues showed no changes in CHOP, TRIB3 and invasion-related proteins (MMP2 and MMP9) expression throughout the menstrual cycle. Taken together, our findings indicate that abnormal ER stress response to progesterone increased endometriotic stromal cell invasiveness via the AKT/mTOR pathway.


Subject(s)
Endometriosis/metabolism , Endoplasmic Reticulum Stress/physiology , Stromal Cells/metabolism , Blotting, Western , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Endometriosis/genetics , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/genetics , Female , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism
6.
Reproduction ; 153(1): 11-21, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-30390418

ABSTRACT

In some human cancer cells, PTEN (phosphatase and tensin homolog deleted on chromosome 10) is known to regulate autophagy induction positively through the inhibition of PI3K/AKT pathway, leading to the activation of mTOR, a major negative regulator of autophagy. Recent studies reported that PTEN expression is abnormally decreased in endometriotic lesions. In endometriosis, abnormal PTEN expression may contribute to the alteration of endometrial cell autophagy, which may affect apoptosis because endometrial cell autophagy is directly involved in the regulation of apoptosis. To test this hypothesis, we evaluated the involvement of PTEN in the regulation of autophagy induction in human normal endometrial stromal cells (NESCs). In addition, we sought to determine whether aberrant PTEN expression in endometriotic cyst stromal cells (ECSCs) is associated with autophagy dysregulation, and a subsequent decrease in apoptosis. Our results show that PTEN expression was enhanced by progesterone treatment in NESCs. Subsequently, autophagy and apoptosis induction increased through the inhibition of AKT and mTOR activity. This progesterone-induced increase in apoptosis was reversed by the inhibition of autophagy induction using either mifepristone (progesterone receptor modulator) or PTEN inhibitor. In contrast, progesterone had no significant effects on PTEN expression, AKT, mTOR activity, autophagy or apoptosis in ECSCs. Furthermore, in contrast to normal eutopic endometrium, endometriotic tissues have constant PTEN expression, autophagy and apoptosis throughout the menstrual cycle. In conclusion, our results suggest abnormal PTEN expression in response to progesterone was observed in ECSCs, which led to the dysregulation of autophagy induction via AKT/mTOR signalling and a subsequent decrease in apoptosis.

7.
Fertil Steril ; 104(3): 655-64.e1, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26051103

ABSTRACT

OBJECTIVE: To elucidate the therapeutic mechanisms of progestin and the effects of progesterone and progestin (dienogest) on autophagy induction and regulation in endometriotic cells, specifically the effects of progesterone and dienogest on the phosphoinositide-3/protein kinase B (PI3K-AKT) and mitogen-activated protein kinase kinases 1 and 2 (MEK1/2)/extracellular-signal-regulated kinase 1/2 (ERK1/2) pathways, which activate mammalian target of rapamycin (mTOR), a major negative regulator of autophagy. DESIGN: In vitro study using human endometriotic cyst stromal cells (ECSCs). SETTING: University medical center. PATIENT(S): Fifteen patients with ovarian endometrioma. INTERVENTION(S): ECSCs treated with progesterone or dienogest. MAIN OUTCOME MEASURE(S): Autophagy as measured by the expression of the microtubule-associated protein light chain 3-II (LC3-II) and autophagosome formation, and levels of AKT, ERK1/2, and mTOR activity to quantify the phosphorylation of AKT, ERK1/2, and S6K (the downstream target of mTOR). RESULT(S): Progesterone treatment had not statistically significant effect on LC3-II expression, autophagosome formation, or phosphorylation of AKT, ERK1/2, or S6K in estrogen-treated ECSCs. However, dienogest treatment up-regulated LC3-II expression and stimulated autophagosome formation. These effects were accompanied by decreased activation of AKT, ERK1/2, and S6K. Furthermore, incubation of ECSCs with AKT and ERK1/2 inhibitors, which mimicked dienogest-mediated inhibition of AKT and ERK1/2 activity, suppressed S6K activity, followed by an increase in LC3-II expression. In addition, cotreatment with dienogest and 3-methyladenine (autophagy inhibitor) decreased the levels of apoptosis of ECSCs compared with the single treatment with dienogest. CONCLUSION(S): Our results suggest that dienogest treatment of endometriotic cells suppresses AKT and ERK1/2 activity, thereby in turn inhibiting mTOR, inducing autophagy, and promoting apoptosis.


Subject(s)
Autophagy/drug effects , Endometriosis/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nandrolone/analogs & derivatives , Ovarian Cysts/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Stromal Cells/drug effects , TOR Serine-Threonine Kinases/metabolism , Apoptosis/drug effects , Cells, Cultured , Endometriosis/pathology , Enzyme Activation , Female , Humans , Nandrolone/pharmacology , Ovarian Cysts/pathology , Phosphorylation , Progesterone/pharmacology , Signal Transduction/drug effects , Stromal Cells/enzymology , Stromal Cells/pathology
8.
Mol Hum Reprod ; 20(10): 972-80, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25107837

ABSTRACT

Autophagy is known to be regulated by the phosphoinositide-3 kinase (PI3K)-protein kinase B (AKT) and/or mitogen-activated protein kinase 1/2 (MEK1/2)-extracellular signal-regulated kinase 1/2 (ERK1/2) pathways, leading to activation of mammalian target of rapamycin (mTOR), a major negative regulator of autophagy. However, some reports have also suggested that autophagic regulation by the PI3K-AKT and/or MEK1/2-ERK1/2 pathways may not be mediated by mTOR activity, and there is no direct evidence of the involvement of these pathways in luteal cell autophagy regulation. To elucidate the luteal cell-specific regulatory mechanisms of autophagy induction during corpus luteum (CL) regression, we evaluated whether luteal cell autophagy is regulated by the PI3K-AKT pathway and/or MEK1/2-ERK1/2 pathway and if this regulation is mediated by mTOR. We found that autophagy induction increased despite mTOR activation in luteal cells cultured with prostaglandin F2α (PGF2α), an important mediator of CL regression, suggesting that PGF2α-induced autophagy is independent of mTOR regulation. We also found that PGF2α-induced autophagy was not mediated by AKT activity, because AKT inhibition using a PI3K inhibitor (wortmannin) did not change autophagy induction or mTOR activity. In contrast, ERK1/2 activity increased in PGF2α-treated luteal cells, as did the levels of autophagy induction despite increased mTOR activity. Furthermore, PGF2α-mediated up-regulation of luteal cell autophagy was reversed by addition of ERK1/2 inhibitors, despite a decrease in mTOR activity. These in vitro results suggest that luteal cell autophagy is induced by increased ERK1/2 activity during CL regression, and is independent of mTOR activity. This finding was further supported by in vivo experiments in a pseudo-pregnant rat model, which showed that induction of luteal cell autophagy increased during luteal stage progression and that this was accompanied by increased ERK1/2 and mTOR activity. Taken together, our findings indicate that activation of ERK1/2 is a key event in the induction of luteal cell autophagy during CL regression which is not associated with mTOR regulation.


Subject(s)
Autophagy/physiology , Corpus Luteum/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Luteal Cells/metabolism , TOR Serine-Threonine Kinases/metabolism , Androstadienes/pharmacology , Animals , Autophagy/drug effects , Dinoprost/pharmacology , Extracellular Signal-Regulated MAP Kinases/biosynthesis , Female , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Pregnancy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Wortmannin
9.
Mol Hum Reprod ; 20(4): 309-17, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24319109

ABSTRACT

Mammalian target of rapamycin (mTOR) is known to be a major negative regulator of autophagy. Recent studies have shown that mTOR activity is abnormally increased in endometriotic lesions. In endometriosis, abnormal mTOR activity may contribute to the alteration of endometrial cell autophagy, which may affect apoptosis because endometrial cell autophagy is directly involved in the regulation of apoptosis. To test this hypothesis, we investigated whether endometrial cell autophagy is altered by aberrant mTOR activity and is associated with apoptosis in ovarian endometriotic cysts. Our results show that endometrial cell autophagy induction was increased by mTOR inhibition as the menstrual cycle progresses in the normal endometrium, and that it is correlated with apoptosis. However, in endometriotic tissues from ovarian endometriotic cysts, autophagy, mTOR activity and apoptosis were constant throughout the menstrual cycle, suggesting that a constant level of autophagy is maintained by disinhibition of mTOR activity during the menstrual cycle in endometriotic tissues and is related to decreased apoptosis. Indeed, compared with normal endometrium, increased mTOR activity during the secretory phase in endometriotic tissues inhibited autophagy and apoptosis induction. In addition, to determine the direct effect of autophagy induction mediated by mTOR on endometriotic cell apoptosis, endometriotic cells were treated with rapamacin (mTOR inhibitor) with and without 3-methyladenine (3-MA, autophagy inhibitor). Although rapamycin treatment induced autophagy and led to apoptosis promotion, the pro-apoptotic effect of rapamycin was reversed by the addition of 3-MA, suggesting that mTOR inhibition promotes endometriotic cell apoptosis via autophagy induction. In conclusion, our results suggest that aberrant mTOR activity in ovarian endometriotic cysts leads to alteration of endometrial cell autophagy, which is associated with abnormal apoptosis.


Subject(s)
Apoptosis/genetics , Autophagy/genetics , Cysts/genetics , Endometriosis/genetics , TOR Serine-Threonine Kinases/genetics , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Cysts/metabolism , Cysts/pathology , Endometriosis/metabolism , Endometriosis/pathology , Endometrium/metabolism , Endometrium/pathology , Female , Gene Expression Regulation , Humans , Menstrual Cycle/physiology , Primary Cell Culture , Sirolimus/pharmacology , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , TOR Serine-Threonine Kinases/metabolism
10.
Reproduction ; 147(1): 73-80, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24131573

ABSTRACT

In this study, we examined whether granulosa cell autophagy during follicular development and atresia was regulated by the class I phosphoinositide-3 kinase/protein kinase B (AKT) pathway, which is known to control the activity of mammalian target of rapamycin (mTOR), a major negative regulator of autophagy. Ovaries and granulosa cells were obtained using an established gonadotropin-primed immature rat model that induces follicular development and atresia. Autophagy was evaluated by measuring the expression level of microtubule-associated protein light chain 3-II (LC3-II) using western blots and immunohistochemistry. The activity of AKT and mTOR was also examined by observing the phosphorylation of AKT and ribosomal protein S6 kinase (S6K) respectively. After gonadotropin injection, LC3-II expression was suppressed and phosphorylation of AKT and S6K increased in rat granulosa cells. By contrast, gonadotropin withdrawal by metabolic clearance promoted LC3-II expression and decreased phosphorylation of AKT and S6K. In addition, in-vitro FSH treatment of rat granulosa cells also indicated inhibition of LC3-II expression accompanied by a marked increase in phosphorylation of AKT and S6K. Inhibition of AKT phosphorylation using AKT inhibitor VIII suppressed FSH-mediated phosphorylation of S6K, followed by an increase in LC3-II expression. Furthermore, co-treatment with FSH and AKT inhibitor increased the levels of apoptosis and cell death of granulosa cells compared with the single treatment with FSH. Taken together, our findings indicated that AKT-mediated activation of mTOR suppresses granulosa cell autophagy during follicular development and is involved in the regulation of apoptotic cell death.


Subject(s)
Autophagy/physiology , Granulosa Cells/metabolism , Ovarian Follicle/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Apoptosis/physiology , Female , Ovarian Follicle/growth & development , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Rats , Signal Transduction/physiology
11.
Biol Reprod ; 86(3): 70, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22088918

ABSTRACT

Autophagy appears to play an important role in the normal development and maintenance of homeostasis in a variety of tissues, including the female reproductive tract. However, the role of autophagy and the association between autophagy and apoptosis in cyclic remodeling of the human endometrium have not been described. Therefore, we investigated the involvement of autophagy during the human endometrial cycle and its association with apoptosis. Endometrial samples were obtained from 15 premenopausal, nonpregnant women who underwent hysterectomies for benign gynecological reasons. The autophagy-associated protein, microtubule-associated protein 1 light chain 3 alpha (MAP1LC3A), was immunolocalized, and its expression level was measured by Western blot analysis. Apoptosis was evaluated by measuring the expression level of cleaved caspase 3 protein. MAP1LC3A protein was primarily expressed within the endometrial glandular cells and increased during the secretory phase. The expression level of the membrane-bound form of MAP1LC3A (MAP1LC3A-II) also increased as the menstrual cycle progressed, reaching a maximum level during the late secretory phase. This pattern coincided with the expression of cleaved caspase 3. Furthermore, expression of MAP1LC3A-II and cleaved caspase 3 increased in the in vitro-cultured endometrial cancer cells when estrogen and/or progesterone were withdrawn from the culture media to mimic physiological hormonal changes. These findings suggest that endometrial cell autophagy is directly involved in the cyclic remodeling of the human endometrium and is correlated with apoptosis. In addition, we inhibited autophagic processes using 3-methyladenine (3-MA) or bafilomycin A1 (Baf A1) to evaluate the role of autophagy in apoptosis induction in endometrial cancer cells. While the inhibition of autophagosome formation using 3-MA did not decrease apoptosis or cell death, the inhibition of autophagosome degradation by fusion with lysosomes using Baf A1 increased apoptosis and cell death, suggesting that the accumulation of autophagosomes induces apoptosis. Furthermore, Baf A1-induced apoptotic cell death was decreased by the apoptosis inhibitor N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (Z-VAD-FMK). In conclusion, these results indicate that autophagy is involved in the endometrial cell cycle affecting apoptosis and is most prominent during the late secretory phase.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Cell Cycle/physiology , Endometrium/physiology , Homeostasis/physiology , Adenine/analogs & derivatives , Adenine/pharmacology , Adult , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Caspase 3/physiology , Cell Cycle/drug effects , Cells, Cultured , Endometrium/cytology , Endometrium/drug effects , Female , Homeostasis/drug effects , Humans , Macrolides/pharmacology , Menstrual Cycle/physiology , Microtubule-Associated Proteins/physiology , Middle Aged , Proto-Oncogene Proteins c-bcl-2 , bcl-2-Associated X Protein/physiology
12.
Biol Reprod ; 85(3): 465-72, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21593476

ABSTRACT

Autophagy is associated with luteal cells death during regression of the corpus luteum (CL) in some species. However, the involvement of autophagy or the association between autophagy and apoptosis in CL regression are largely unknown. Therefore, we investigated the role of autophagy in CL regression and its association with apoptosis. Ovaries were obtained from pseudopregnant rats at Days 2 (early), 7 (mid-), and 14 and 20 (late-luteal stage) of the pseudopregnancy; autophagy-associated protein (microtuble-associated protein light chain 3 [LC3]) was immunolocalized and its expression level was measured. Luteal cell apoptosis was evaluated by measuring cleaved caspase 3 expression. LC3 expression increased slightly from early to mid-luteal stage, with maximal levels detected at the late-luteal stage in steroidogenic luteal cells. The expression level of the membrane form of LC3 (LC3-II) also increased during luteal stage progression, and reached a maximum at the end point of late-luteal stage (Day 20). This pattern coincided with cleaved caspase 3 expression. Furthermore, LC3-II expression increased, as did levels of cleaved caspase 3 in luteal cells cultured with prostaglandin F(2alpha) known to induce CL regression. These findings suggest that luteal cell autophagy is directly involved in CL regression, and is correlated with increased apoptosis. In addition, autophagic processes were inhibited using 3-methyladenine or bafilomycin A1 to evaluate the role of autophagy in apoptosis induction. Inhibition of autophagosome degradation by fusion with lysosomes (bafilomycin A1) increased apoptosis and cell death. Furthermore, inhibition of autophagosome formation (3-methyladenine) decreased apoptosis and cell death, suggesting that the accumulation of autophagosomes induces luteal cell apoptosis. In conclusion, these results indicate that autophagy is involved in rat luteal cell death through apoptosis, and is most prominent during CL regression.


Subject(s)
Apoptosis , Autophagy , Corpus Luteum/physiology , Animals , Corpus Luteum/ultrastructure , Dinoprost/metabolism , Female , Phagosomes/physiology , Phagosomes/ultrastructure , Pseudopregnancy/metabolism , Rats , Rats, Sprague-Dawley
13.
Fertil Steril ; 95(4): 1482-6, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-20630503

ABSTRACT

This study evaluated the effect of autophagosome accumulation on apoptotic cell death in granulosa cells from developing follicles. Our results indicate that the accumulation of autophagosomes induces apoptotic cell death of granulosa cells through decreased bcl-2 expression and subsequent caspase activation.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Granulosa Cells/pathology , Lysosomes/pathology , Animals , Cell Death/physiology , Female , Granulosa Cells/metabolism , Lysosomes/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Rats , Rats, Sprague-Dawley
14.
Fertil Steril ; 91(3): 744-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18321491

ABSTRACT

OBJECTIVE: To explain the unexpected low response to GnRH antagonist protocol in reproductive women with normal baseline hormone profiles. DESIGN: Retrospective study. SETTING: University hospital. PATIENT(S): Twenty-five women undergoing their first IVF cycle. INTERVENTION(S): Follicular fluid (FF) from large follicles (>15 mm) was obtained during oocyte retrieval from unexpected low responders (n = 13, group A) and 12 age-matched normal responders (n = 12, group B). MAIN OUTCOME MEASURE(S): The FF markers known to reflect follicle environment (insulin-like growth factor [IGF] II, IGF-binding protein 4, müllerian-inhibiting substance, pregnancy-associated plasma protein A, soluble Fas, and vascular endothelial growth factor [VEGF]) were analyzed by ELISA. RESULT(S): The baseline characteristics (age, day 3 serum LH, FSH, E(2), duration and dose of r-FSH, GnRH antagonist) were not different between the two groups. The number of large follicles, oocytes retrieved, and serum E(2) levels on the day of hCG injection were significantly higher in group B. Whereas the other follicular markers did not differ between the two groups, VEGF was significantly higher in group A. In addition, the VEGF concentration showed an inverse correlation with the total number of oocytes retrieved. CONCLUSION(S): The unexpected low response in women with normal basal hormone profiles, during GnRH antagonist protocol, was associated with altered follicular VEGF expression.


Subject(s)
Fertility Agents, Female/administration & dosage , Fertilization in Vitro , Follicular Fluid/metabolism , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/administration & dosage , Hormones/blood , Ovulation Induction/methods , Vascular Endothelial Growth Factor A/metabolism , Adult , Chorionic Gonadotropin/administration & dosage , Estradiol/blood , Female , Follicle Stimulating Hormone, Human/administration & dosage , Follicle Stimulating Hormone, Human/blood , Gonadotropin-Releasing Hormone/administration & dosage , Gonadotropin-Releasing Hormone/analogs & derivatives , Humans , Luteinizing Hormone/blood , Oocyte Retrieval , Pancuronium/administration & dosage , Pancuronium/analogs & derivatives , Pregnancy , Pregnancy Rate , Progesterone/blood , Retrospective Studies , Treatment Outcome
15.
Cryobiology ; 57(3): 209-15, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18789915

ABSTRACT

Cryopreservation of ovarian tissue has been reported to delay the development of preantral follicles by temporary suppression of granulosa cell proliferation during in vitro culture. This delay might be overcome by treatment with activin A and/or IGF-I, known to stimulate granulosa cell proliferation. However, the effects of these growth factors, on delayed follicle development induced by ovarian tissue cryopreservation, have not been evaluated. Therefore, we studied the effects of activin A and/or IGF-I on granulosa cell proliferation and follicle development in preantral follicles isolated from mouse cryopreserved ovarian tissues. The preantral follicles isolated from fresh ovarian tissues were cultured with control medium (CM) for 10 days. The preantral follicles isolated from cryopreserved ovarian tissues were cultured with CM and with CM+activin A (100 ng/ml), IGF-I (50 ng/ml) or activin A+IGF-I added for 10, 12 and 14 days. The follicles were stimulated with hCG at the end of culture. The granulosa cell proliferation was evaluated by measuring the PCNA expression and the follicle development assessed by comparing the follicle diameter and oocyte maturation. The expressed level of PCNA was significantly decreased in the cryopreserved preantral follicles cultured with CM, compared to the fresh group (p<0.05), but increased to the level of the fresh group by the addition of activin A, IGF-I or activin A and IGF-I. The maximum follicle diameter and oocyte maturation rate were obtained in the fresh group after 10 days of culture, while the diameter and oocyte maturation rate of cryopreserved preantral follicles reached similar levels after 14 days. Under conditions of CM with added activin A or activin A+IGF-I, both the diameter and oocyte maturation rate of the cryopreserved preantral follicles improved to the levels of the fresh group after 12 days. However, the stimulatory effect was not different in comparisons between activin A and activin A+IGF-I. In addition, adding activin A significantly increased the survival rate of cryopreserved preantral follicles compared to IGF-I (p<0.05). On the other hand, the diameter of the cryopreserved preantral follicles did not increase to the level of the fresh group by addition of IGF-I, although the oocyte maturation rate was improved to the level of fresh group after 12 days of culture. These findings indicate that activin A has a greater stimulatory effect on the growth of preantral follicles than does IGF-I. In conclusion, the addition of activin A to culture media stimulated granulosa cell proliferation to overcome the delay in culture of the development of preantral follicles isolated from cryopreserved mouse ovaries.


Subject(s)
Activins/pharmacology , Cryopreservation/methods , Insulin-Like Growth Factor I/pharmacology , Ovarian Follicle/drug effects , Animals , Cell Survival , Cells, Cultured , Female , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Mice , Oocytes/cytology , Oocytes/drug effects , Oocytes/growth & development , Ovarian Follicle/cytology , Ovarian Follicle/growth & development , Proliferating Cell Nuclear Antigen/metabolism
16.
Cryobiology ; 56(1): 36-42, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18155187

ABSTRACT

Cryopreservation of ovarian tissue has been reported to delay the development of preantral follicles during in vitro culture, but the mechanism causing this impairment has not been brought to light. In order to elucidate the underlying mechanism of delayed follicular development, we evaluated the effects of cryopreservation on the proliferation of granulosa cells during culture of mouse preantral follicles, as a sufficient population of granulosa cells is critical for normal follicular development. Additionally the initial cell death of granulosa cells was estimated immediately after cryopreservation. The ovarian tissues obtained from 12-day-old female mice were cryopreservation by vitrification. The granulosa cell proliferation was evaluated by measuring the PCNA expression and the expression of cell cycle regulators such as cyclin D2, CDK4, cyclin E and CDK2 in preantral follicles isolated from fresh and cryopreserved ovarian tissues that were cultured for 48 h. The viability of granulosa cells was evaluated by measuring the proportion of necrotic areas. The granulosa cell proliferation of the cryopreserved preantral follicles was decreased significantly compared to that of the fresh controls at 0 and 24h after culture (P<0.05), and this was increased to the control levels after 48 h of culture. The expressions of cyclin D2, Cdk4, cyclin E and Cdk2 were also decreased in the cryopreserved ovarian tissues at 0 and 24h after culture (P<0.05), but they were increased to the control levels after 48 h of culture. The proportion of the necrotic area was significantly higher in cryopreserved preantral follicles compared to that of the fresh preantral follicles (P<0.05). This suggests that cryopreservation of ovarian tissues may delay the preantral follicle development by temporary suppressing the granulosa cell proliferation through the cell cycle regulators (cyclin D2, Cdk4, cyclin E and Cdk2) and by granulosa cell death immediately after warming.


Subject(s)
Cell Proliferation , Cryopreservation , Granulosa Cells/physiology , Ovarian Follicle/cytology , Ovary/cytology , Animals , Cell Survival , Cyclin D2 , Cyclin E/biosynthesis , Cyclin-Dependent Kinase 2/biosynthesis , Cyclin-Dependent Kinase 4/biosynthesis , Cyclins/biosynthesis , Female , Granulosa Cells/pathology , Mice , Necrosis/pathology , Ovarian Follicle/metabolism , Proliferating Cell Nuclear Antigen/biosynthesis
17.
Cryobiology ; 54(1): 55-62, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17196581

ABSTRACT

The cryopreservation of ovarian tissue has been reported to affect the development of preantral follicles. However, the effect of cryopreservation of ovarian tissue on the development of primordial follicles remains to be elucidated. This study was conducted to evaluate the effect of cryopreservation on the development of frozen-thawed mouse primordial follicles. One-day-old mouse ovaries were cryopreserved by either slow-freezing or a vitrification method. The development of primordial follicles was evaluated histologically and also with markers for follicle development such as: GDF-9, inhibin-alpha subunit and ZP3 in fresh and frozen-thawed ovaries cultured for five days. The proportion of apoptotic and necrotic areas was analyzed in fresh and frozen-thawed ovaries at one and five days after culture, in order to examine the viability of ovarian cells that influence primordial follicle development. The development rate of primordial follicles was significantly lower in slow-frozen and vitrified ovaries than the fresh controls after five days of in vitro culture (P<0.05). The mRNA expression for all developmental markers was slightly decreased in the frozen-thawed ovaries; this difference was not significant. The proportion of apoptosis was significantly increased in the slow-frozen and vitrified ovaries compared to the fresh ovaries at one day (P<0.05); however, there was no difference at five days after culture. The proportion of the area of necrosis was significantly higher in slow-frozen and vitrified ovaries compared to the fresh ovaries at one and five days after culture (P<0.05). Our preliminary data suggest that ovarian tissue cryopreservation using slow-freezing and vitrification methods inhibits development of primordial follicles. This may be caused by the death of ovarian cells through apoptosis and necrosis after cryopreservation.


Subject(s)
Cryopreservation/methods , Ovarian Follicle/growth & development , Ovary , Animals , Cryopreservation/veterinary , Female , Immunohistochemistry , Mice , Mice, Inbred ICR , Ovary/anatomy & histology , Ovary/physiology , RNA, Messenger/metabolism , Staining and Labeling , Time Factors
18.
Eur J Obstet Gynecol Reprod Biol ; 125(2): 233-8, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16443318

ABSTRACT

OBJECTIVES: This study was conducted to evaluate the involvement of apoptosis in the freeze-thaw process and to investigate the anti-apoptotic effect of vascular endothelial growth factor (VEGF) in the frozen-thawed granulosa cells. STUDY DESIGN: Isolated rat granulosa cells were cultured, frozen-thawed, and were cultured for 24h. Cell viabilities (by Trypan blue exclusion test) and apoptotic patterns (by Annexin-V/propidium iodide (PI) Double-Staining) were determined at each step. Apoptotic cell death was confirmed by following DNA degradation and caspase-3 activity. RESULTS: After freeze-thaw process and 24h of culture, reductions in the cellular viabilities and increases in the number of cells containing degraded DNA were lower in the VEGF pretreated group than in the control group (p<0.05). In the VEGF pretreated group, increases in the proportions of late apoptotic cells [Annexin-V (+)/PI (+)] were significantly lower and caspase-3 expression was prevented immediate after thawing (p<0.05). Furthermore, increases in the proportions of early apoptotic cells [Annexin-V (+)/PI (-)] and reductions in the proportions of viable cells [Annexin-V (-)/PI (-)] were significantly lower in the VEGF pretreated group after culture for 24h (p<0.05). Of the different doses of VEGF pretreated, 50ng/ml was found to be most effective with respect to protecting frozen-thawed granulosa cells from cryoinjury. CONCLUSION: Granulosa cell damage induced by cryopreservation is mediated, at least in part, by an apoptotic process. Our preliminary results suggest that VEGF treatment before freeze-thaw process reduces rat ovarian granulosa cell damage by inhibiting apoptosis.


Subject(s)
Apoptosis/drug effects , Cryopreservation , Granulosa Cells/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Animals , Cell Culture Techniques/methods , Cell Survival/drug effects , Cell Survival/physiology , Female , Flow Cytometry/methods , Granulosa Cells/physiology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology
19.
Mol Reprod Dev ; 69(3): 289-95, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15349840

ABSTRACT

This study was conducted to investigate the effect of recipient activation time on the chromatin structure and development of bovine nuclear transfer embryos. Serum-starved skin cells were electrofused to enucleated oocytes, activated 1-5 hr after fusion, and cultured in vitro. Some fused eggs were fixed at each time point after fusion without activation, or 3 or 7 hr after activation. Some nocodazole treated zygotes were fixed to analyze their chromosome constitutions. The proportion of eggs with a morphologically normal premature chromosome condensation (PCC) state increased 1-2 hr after fusion. Whereas eggs with elongated chromosome plate increased as activation time was prolonged to 3 hr, and 5 hr after fusion, 58.1% of eggs showed more than two scattered chromosome sets. The proportion of eggs with a single chromatin mass (40.6-56.7%) significantly increased when eggs were activated within 2.5 hr after fusion (P < 0.05). Only 23.3% of reconstituted embryos activated 5 hr after fusion formed one pronucleus-like structure (PN), whereas, 64.5-78.3% of embryos activated 1-2.5 hr after fusion formed one PN. The proportion of embryos with normal chromosome constitutions decreased as activation time was prolonged. Development rates to the blastocyst stage were higher in eggs activated within 2 hr after fusion (17.3-21.7%) compared to those of others (0-8.6%, P < 0.05). The result of the present study suggests that activation time can affect the chromatin structure and in vitro development of bovine nuclear transfer embryos.


Subject(s)
Animals, Genetically Modified , Cell Nucleus/physiology , Chromatin Assembly and Disassembly/physiology , Chromatin/physiology , Embryo, Mammalian/physiology , Nuclear Transfer Techniques , Animals , Cattle , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...