Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Mol Hum Reprod ; 30(8)2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39037927

RESUMEN

EXOC5 is a crucial component of a large multi-subunit tethering complex, the exocyst complex, that is required for fusion of secretory vesicles with the plasma membrane. Exoc5 deleted mice die as early embryos. Therefore, to determine the role of EXOC5 in follicular and oocyte development, it was necessary to produce a conditional knockout (cKO), Zp3-Exoc5-cKO, in which Exoc5 was deleted only in oocytes. The first wave of folliculogenesis appeared histologically normal and progressed to the antral stage. However, after IVF with normal sperm, oocytes collected from the first wave (superovulated 21-day-old cKO mice) were shown to be developmentally incompetent. Adult follicular waves did not progress beyond the secondary follicle stage where they underwent apoptosis. Female cKO mice were infertile. Overall, these data suggest that the first wave of folliculogenesis is less sensitive to oocyte-specific loss of Exoc5, but the resulting gametes have reduced developmental competence. In contrast, subsequent waves of folliculogenesis require oocyte-specific Exoc5 for development past the preantral follicle stage. The Zp3-Exoc5-cKO mouse provides a model for disrupting folliculogenesis that also enables the separation between the first and subsequent waves of folliculogenesis.


Asunto(s)
Ratones Noqueados , Oocitos , Oogénesis , Folículo Ovárico , Animales , Femenino , Masculino , Ratones , Oocitos/metabolismo , Oogénesis/genética , Oogénesis/fisiología , Folículo Ovárico/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Glicoproteínas de la Zona Pelúcida/genética , Glicoproteínas de la Zona Pelúcida/metabolismo
2.
Reprod Biol Endocrinol ; 22(1): 51, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38671458

RESUMEN

BACKGROUND: Ovarian damage and follicle loss are major side effects of chemotherapy in young female patients with cancer. However, effective strategies to prevent these injuries are still lacking. The purpose of this study was to verify low-intensity pulsed ultrasound (LIPUS) can reduce ovarian injury caused by chemotherapy and to explore its underlying mechanisms in mice model. METHODS: The mice were randomly divided into the Control group, Cisplatin group, and Cisplatin + LIPUS group. The Cisplatin group and Cisplatin + LIPUS group were intraperitoneally injected with cisplatin every other day for a total of 10 injections, and the Control group was injected with saline. On the second day of each injection, the Cisplatin + LIPUS group received irradiation, whereas the other two groups received sham irradiation. We used a variety of biotechnologies to detect the differences in follicle count, granulosa cell apoptosis, fibrosis, transcriptome level, oxidative damage, and inflammation in differently treated mice. RESULT: LIPUS was able to reduce primordial follicle pool depletion induced by cisplatin and inhibit the apoptosis of granulosa cells. Transcriptomic results confirmed that LIPUS can reduce ovarian tissue injury. We demonstrated that LIPUS can relieve ovarian fibrosis by inhibiting TGF-ß1/Smads pathway. Meanwhile, it can reduce the oxidative damage and reduced the mRNA levels of proinflammatory cytokines caused by chemotherapy. CONCLUSION: LIPUS can reduce the toxic effects of chemotherapy drugs on ovaries, inhibit ovarian fibrosis, reduce the inflammatory response, and redcue the oxidative damage, reduce follicle depletion and to maintain the number of follicle pools.


Asunto(s)
Antineoplásicos , Cisplatino , Ovario , Ondas Ultrasónicas , Animales , Femenino , Ratones , Cisplatino/efectos adversos , Ovario/efectos de los fármacos , Ovario/efectos de la radiación , Ovario/patología , Antineoplásicos/efectos adversos , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/efectos de la radiación , Terapia por Ultrasonido/métodos
3.
Ecotoxicol Environ Saf ; 242: 113859, 2022 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-35816842

RESUMEN

Carbon tetrachloride (CCL4) is widely used as a chemical intermediate and as a feedstock in the production of chlorofluorocarbons. CCL4 is highly toxic in the liver, kidney, testicle, brain and other tissues. However, the effect of CCL4 on ovarian function has not been reported. In this study, we found that the mice treated with CCL4 showed decreased ovarian function with disturbed estrus cycle, decreased serum level of 17ß-estradiol and the reduced number of healthy follicles. Ovarian damage was accompanied by oxidative stress and the production of proinflammatory cytokines, especially interleukins. The indicators of oxidative stress, 4-Hydroxynonenal (4-HNE), 8-hydroxy-2´-deoxyguanosine (8-OHdG), 3-Nitrotyrosine (3-NT) and malondialdehyde (MDA), and the levels of proinflammatory cytokines IL-1α, IL-1ß, IL-6 and IL-11 were increased, while the antioxidants, including superoxide dismutase (SOD), nuclear factor erythroid2-related factor 2 (NRF2) and heme oxygenase-1 (HO-1), were decreased in the CCL4 group. In the CCL4 treated group, the results of Sirius Red staining, immunohistochemistry and qPCR indicated that proinflammatory cytokines caused further ovarian fibrosis. And CCL4 could also promote ovarian thecal cells to secrete inflammatory cytokines, resulting in fibrosis in vitro. In addition, CCL4 inhibited oocyte development and triggered oocyte apoptosis. In conclusion, CCL4 exposure causes ovarian damage by strong oxidative stress and the high expression of the proinflammatory cytokine mediated ovarian fibrosis.


Asunto(s)
Tetracloruro de Carbono , Enfermedad Hepática Inducida por Sustancias y Drogas , Animales , Antioxidantes/metabolismo , Tetracloruro de Carbono/metabolismo , Tetracloruro de Carbono/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Citocinas/metabolismo , Fibrosis , Hígado , Ratones , Estrés Oxidativo
4.
Reprod Med Biol ; 19(4): 415-424, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33071644

RESUMEN

PURPOSE: Ovarian vascular abnormality and ovarian fibrosis are observed in the low responder patients and aging mice. Vascularization and fibrosis are regulated by injury-repair system, such as wound. Thus, in this study, the authors tried to investigate the effect of the surgical treatment to ovarian surface with cutting on the functions of ovary in aging mouse model, gcNrg1KO. METHOD: The ovarian surface of gcNrg1KO was surgically cut, and then the ovary was returned inside of bursa ovarica. To assess the effect of cutting on fertility, mating test, smear analysis, and exogenous hormonal treatment were done. Additionally, the histological analysis was used for observing the remodeling of ovarian stroma after the surgical approach. RESULT: Ovarian fibrosis disappeared at 7 days after surgery. With the abrogation of fibrosis, the blood vessels were fluently observed around the follicles, and the follicular development was re-started. The responses against exogenous hormone were recovered at 21 days after the surgery, and estrous cycle and delivery were also recovered by the surgery and the fertility was maintained for 3 months. CONCLUSION: This cutting method of ovarian surface becomes a good option against low responder patients.

5.
J Microsc ; 261(3): 267-76, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26484626

RESUMEN

Some chemotherapeutic agents used for breast cancer (BC) treatment can induce severe side effects in the ovarian tissue. The combination of cyclophosphamide and docetaxel (TC) is widely used for BC treatment; however, its late effects in the ovary are not completely understood. The main purpose of this study was to evaluate the structural and ultrastructural alterations in the ovarian stroma induced by TC treatment. Wistar rats were divided into two groups: a control group and a TC group. They were euthanized 5 months after the end of treatment, and their plasma and ovaries were collected. Important alterations were noted. The serum estradiol level was significantly reduced in the TC group compared with the control group. Additionally, the number of apoptotic nuclei was higher in the TC group. The role of the inflammatory response in the development of ovarian damage was investigated, and we found an increased number of mast cells and increased expression of TNF-α in the TC group. The involvement of fibrosis was also investigated. The results showed that the TC group had increased expression levels of TGF-ß1, collagen type I (col-I) and collagen type III (col-III) compared with the control group. Ultrastructural analysis revealed the presence of collagen fibrils in the treated group and illustrated that the ovarian tissue architecture was more disorganized in this group than in the control group. The results from this study are important in the study of chemotherapy-induced ovarian failure and provide further insight into the mechanisms involved in the development of this disease.


Asunto(s)
Antineoplásicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Ciclofosfamida/efectos adversos , Neoplasias Mamarias Animales/tratamiento farmacológico , Ovario/efectos de los fármacos , Ovario/ultraestructura , Taxoides/efectos adversos , Útero/efectos de los fármacos , Animales , Antineoplásicos/uso terapéutico , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Ciclofosfamida/uso terapéutico , Docetaxel , Estradiol/sangre , Femenino , Microscopía Electrónica de Transmisión , Ratas , Ratas Wistar , Taxoides/uso terapéutico , Factor de Crecimiento Transformador beta1/metabolismo
6.
Cell Signal ; 122: 111334, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39102927

RESUMEN

OBJECTIVE: Chronic psychological stress is associated with impaired follicular development and ovarian dysfunction. Many aspects of this dysfunction and the underlying mechanisms remain unclear. Using a chronic unpredictable mild stress (CUMS) mouse model, we investigate the influence of chronic stress on ovarian function and explore potential mechanisms. METHODS: A CUMS mouse model was constructed over eight months, covering the period from sexual maturity to the onset of declining fertility in mice. At the end of the 2nd, 4th, 6th, and 8th months of exposure to CUMS, behavioral and physiological assays, including the sucrose preference test, tail suspension test, and serum corticosterone levels, were conducted to validate the effectiveness of the stress model. Fertility and ovarian function were assessed by analyzing the estrous cycle, number of offspring, sex hormone levels, follicle counts, granulosa cell proliferation and apoptosis, and the expression levels of fibrosis markers. Furthermore, proteomic analyses were performed on the ovaries to investigate the molecular mechanisms of ovarian fibrosis induced by CUMS. RESULTS: With continued CUMS exposure, there was a gradual decline in both the ovary-to-body weight ratio and the number of offspring. Moreover, the percentage of atretic follicles was notably higher in the CUMS-exposed groups compared to the control groups. It is noticeable that CUMS triggered granulosa cell apoptosis and halted proliferation. Additionally, increased expression of α-SMA and Collagen I in the ovaries of CUMS-exposed mice indicated that CUMS could induce ovarian fibrosis. Proteomic analysis provided insights into the activation of specific biological processes and molecules associated with fibrosis induced by chronic stress. CONCLUSIONS: Our results strongly suggest that exposure to CUMS induces ovarian fibrosis, which influences follicular development and ultimately contributes to fertility decline. These findings offer novel perspectives on the impact of chronic stress on ovarian dysfunction.


Asunto(s)
Fertilidad , Fibrosis , Ovario , Estrés Psicológico , Animales , Femenino , Ratones , Estrés Psicológico/complicaciones , Ovario/patología , Ovario/metabolismo , Apoptosis , Células de la Granulosa/metabolismo , Células de la Granulosa/patología , Modelos Animales de Enfermedad , Folículo Ovárico/metabolismo , Folículo Ovárico/patología , Proliferación Celular
7.
J Ovarian Res ; 17(1): 139, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38970048

RESUMEN

Ovarian fibrosis, characterized by the excessive proliferation of ovarian fibroblasts and the accumulation of extracellular matrix (ECM), serves as one of the primary causes of ovarian dysfunction. Despite the critical role of ovarian fibrosis in maintaining the normal physiological function of the mammalian ovaries, research on this condition has been greatly underestimated, which leads to a lack of clinical treatment options for ovarian dysfunction caused by fibrosis. This review synthesizes recent research on the molecular mechanisms of ovarian fibrosis, encompassing TGF-ß, extracellular matrix, inflammation, and other profibrotic factors contributing to abnormal ovarian fibrosis. Additionally, we summarize current treatment approaches for ovarian dysfunction targeting ovarian fibrosis, including antifibrotic drugs, stem cell transplantation, and exosomal therapies. The purpose of this review is to summarize the research progress on ovarian fibrosis and to propose potential therapeutic strategies targeting ovarian fibrosis for the treatment of ovarian dysfunction.


Asunto(s)
Fibrosis , Ovario , Humanos , Femenino , Ovario/patología , Ovario/metabolismo , Animales , Matriz Extracelular/metabolismo , Enfermedades del Ovario/metabolismo , Enfermedades del Ovario/patología , Enfermedades del Ovario/terapia , Terapia Molecular Dirigida , Factor de Crecimiento Transformador beta/metabolismo
8.
NanoImpact ; 34: 100507, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38663500

RESUMEN

Polystyrene microplastics, extensively considered endocrine disrupting chemicals, disturb the reproductive system of living organisms. Polycystic ovary syndrome (PCOS), the reproductive endocrinopathy, is longstanding concern due to its eternal impacts as reproductive disorder and infertility. Despite several reports in reproductive and endocrine toxicity, there is inadequate literature regarding the daily intake of polystyrene-microplastics via drinking water in causing PCOS and leading to ovarian fibrosis in long-term. The present study investigated whether daily consumption of polystyrene-microplastics at doses equivalent to human exposure can cause PCOS and progress to ovarian fibrosis, using female zebrafish as model. Resembling letrozole-PCOS zebrafish model, daily intake of polystyrene-microplastics displayed hallmark PCOS pathophysiology; like excess body weight and %Gonadosomatic index, decreased Follicle Stimulating Hormone and ß-estradiol, increased Luteinising Hormone, brain and ovarian Testosterone (39.3% and 75% respectively). Correspondingly, ovarian histology revealed more developing (stage I and II) oocytes and less mature oocytes alongwith cystic lesions; like follicular membrane disorganization, zona pellucida invagination, theca hypertrophy, basophilic granular accumulation and oocyte buddings. Lipid deposition in intestinal and ovarian tissues was evidenced and increased fasting blood glucose manifesting insulin resistance. The expression of PCOS biomarkers (tox3, dennd1a, fem1a) was significantly disturbed. Polystyrene microplastics played vital role in inducing PCOS further enhancing oxidative stress, which positively influences inflammation and aggravate ovarian mitophagy, shedding light on its ability to harshen PCOS into ovarian fibrosis, which is characterized by collagen deposition and upregulation of pro-fibrogenic biomarker genes. These findings illustrate the potential of daily microplastics intake via drinking water in triggering PCOS and its progression to ovarian fibrosis.


Asunto(s)
Agua Potable , Fibrosis , Microplásticos , Ovario , Síndrome del Ovario Poliquístico , Poliestirenos , Pez Cebra , Animales , Femenino , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/patología , Microplásticos/toxicidad , Microplásticos/efectos adversos , Poliestirenos/efectos adversos , Poliestirenos/toxicidad , Agua Potable/efectos adversos , Agua Potable/química , Ovario/patología , Ovario/efectos de los fármacos , Ovario/metabolismo , Fibrosis/inducido químicamente , Contaminantes Químicos del Agua/toxicidad , Contaminantes Químicos del Agua/efectos adversos , Modelos Animales de Enfermedad
9.
Sci Rep ; 14(1): 23178, 2024 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-39369073

RESUMEN

Previous studies have reported that senolytic drugs can reverse obesity-mediated accumulation of senescent cells in the ovary and protect against cisplatin-induced ovarian injury by removing senescent cells. Early intervention with ABT-263 has been shown to mitigate ovarian aging. However, it remains unknown whether treatment with ABT-263 could rejuvenate the aged ovary in reproductively old females. Therefore, the current study was aimed to investigate whether advanced age intervention with ABT-263 could ameliorate age-related decline in ovarian function. Fourteen 16-month-old mice with a C57/BL6 background were treated with ABT-263 (N = 7) or vehicle (N = 7) for two weeks. Mice were initially treated with ABT-263 (60 mg/kg/d) or vehicle for 7 consecutive days. After a 7-day break, the treatment was repeated for another 7 consecutive days. Six 2-month-old mice with C57BL/6 were used as a young control. The hormonal levels, estrus cycles, ovarian reserve, ovarian cell proliferation and apoptosis, ovarian fibrosis, and steroidogenic gene expression of ovarian stromal cells were evaluated. ABT-263 treatment did not rescue abnormal estrus cycles and sex hormonal levels, or inhibit the formation of multinucleated giant cells and ovarian stromal cell apoptosis in aged ovaries. However, it reduced ovarian fibrosis and preserved the steroidogenic gene expression of ovarian stromal cells in aged ovaries. Importantly, ABT-263 treatment further depleted ovarian follicles in aged mice. In conclusion, ABT-263 treatment accelerated the depletion of ovarian follicles in aged mice, suggesting that senolytic drugs for reproductively old female may adversely affect female fertility.


Asunto(s)
Envejecimiento , Compuestos de Anilina , Apoptosis , Ratones Endogámicos C57BL , Ovario , Sulfonamidas , Animales , Femenino , Ratones , Ovario/efectos de los fármacos , Ovario/metabolismo , Ovario/patología , Sulfonamidas/farmacología , Envejecimiento/efectos de los fármacos , Compuestos de Anilina/farmacología , Apoptosis/efectos de los fármacos , Senoterapéuticos/farmacología , Proliferación Celular/efectos de los fármacos , Reserva Ovárica/efectos de los fármacos , Ciclo Estral/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Células del Estroma/metabolismo , Células del Estroma/efectos de los fármacos
10.
J Mol Histol ; 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39261364

RESUMEN

Methyltransferase-like 3 (METTL3) is extensively reported to be involved in organ fibrosis. Ovarian fibrosis is a main characteristic of polycystic ovary syndrome (PCOS). However, the reaction mechanism of METTL3 in PCOS is poorly investigated. This paper was intended to reveal the role and the mechanism of METTL3 in PCOS. Animal and cell models of PCOS were induced by dehydroepiandrosterone (DHEA). H&E staining was performed to detect the pathological alterations in ovary tissues. Masson staining, immunofluorescence, along with western blot measured fibrosis both in vitro and in vivo. To evaluate estrous cycle, vaginal smear was performed. Lipid peroxidation and ferroptosis were evaluated by MDA assay kits, GSH assay kits, immunohistochemistry, Prussian blue staining and western blot. qRT-PCR and western blot were adopted to estimate METTL3 and GPX4 expression. The m6A and hormone secretion levels were respectively assessed by m6A RNA Methylation Quantitative Kit and corresponding kits. The interaction between METTL3 and GPX4 was testified by immunoprecipitation. The fibrosis and ferroptosis were aggravated and m6A and METTL3 expression were increased in ovarian tissues of DHEA-induced PCOS mice. METTL3 silencing alleviated pathological changes, affected hormone secretion level, and repressed fibrosis, lipid peroxidation and ferroptosis in the ovarian tissues of PCOS mice. In vitro, DHEA stimulation increased m6A and METTL3 expression and induced ferroptosis and fibrosis. METTL3 knockdown promoted GPX4 expression in DHEA-induced granulosa cells by m6A modification and restrained DHEA-induced fibrosis, lipid peroxidation and ferroptosis in granulosa cells via elevating GPX4. METTL3 silence inhibited ovarian fibrosis in PCOS, which was mediated through suppressing ferroptosis by upregulating GPX4 in m6A-dependent manner.

11.
FEBS Lett ; 597(21): 2643-2655, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37698355

RESUMEN

Ovarian fibrosis is a reproduction obstacle leading to female infertility in vertebrates, but the cause underlying the cellular events is unclear. Here, we found that the small adaptor protein SH3-domain-binding glutamate-rich protein like (Sh3bgrl) plays an important role in female reproduction in zebrafish. Two sh3bgrl mutant alleles that result in sh3bgrl depletion contribute to female spawning inability. Comparative transcriptome analysis revealed that sh3bgrl knockout mechanistically causes the upregulation of genes associated with extracellular matrix (ECM) and fiber generation in the zebrafish ovary. Consequently, extra ECM or fibers accumulate and are deposited in the ovary, resulting in eventual spawning inability. Our findings thus provide insights into understanding the underlying mechanism of infertility by ovarian fibrosis and provide a novel and valuable model to study female reproduction abnormality.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Ovario , Pez Cebra , Animales , Femenino , Proteínas Adaptadoras Transductoras de Señales/genética , Fibrosis , Pez Cebra/genética
12.
J Anim Sci ; 1012023 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-37061806

RESUMEN

An androgen excess ovarian micro-environment may limit follicle progression in sheep. Two populations of ewes with divergent follicular fluid androstenedione (A4) were identified in a flock in Jordan: High A4; (A4) ≥ 30 ng/mL, (N = 12) or Control A4 (Control); A4 ≤ 15 ng/mL; (N = 12). We hypothesized High A4 ewes would have increased steroidogenic enzyme mRNA abundance, inflammation, and follicular arrest. Messenger RNA abundance for steroidogenic enzymes StAR, CYP17A1, CYP11A1, and HSD3B1 were increased in theca cells while CYP17A1, CYP19A1, and HSD3B1 were increased in granulosa cells in High A4 ewes compared to Control. Gonadotropin receptor mRNA expression for LHCGR was increased in theca and FSHR in granulosa in High A4 ewes. Messenger RNA expression of FOS when reduced, increases expression of CYP17A1 which was observed in High A4 granulosa cells compared to Control. Furthermore, High A4 ewes had greater numbers of primordial follicles (P < 0.001) and fewer developing follicles compared to Control before, and after 7 d of culture, indicating follicular arrest was not alleviated by cortex culture. Increased fibrosis in the ovarian cortex was detected in High A4 ewes relative to Control (P < 0.001) suggesting increased inflammation and altered extracellular matrix deposition. Thus, this High A4 ewes population has similar characteristics to High A4 cows and women with polycystic ovary syndrome suggesting that naturally occurring androgen excess occurs in multiple species and may be a causative factor in follicular arrest and subsequent female sub- or infertility.


Excess androgen (androstenedione; A4) in ewes can result in ovarian follicular arrest and fibrosis contributing to anovulation in sheep. We have identified a naturally occurring ovarian A4 excess in a sheep population with similar characteristics to High A4 cows, both of which are similar to that in women with polycystic ovary syndrome indicating that several mammalian species experience naturally occurring androgen excess resulting in infertility or follicle arrest. Somatic cells, theca and granulosa, surrounding the egg in High A4 ewes had increased expression of steroidogenic enzymes, similar to that seen in High A4 cows, permitting more ovarian cells to manufacture androgens, which may be the cause of androgen excess. Thus, naturally occurring androgen-excess in domestic livestock females can be utilized as models to research the causes of androgen excess and determine the mechanisms that result in follicular arrest and sub- or infertility.


Asunto(s)
Enfermedades de los Bovinos , Enfermedades de las Ovejas , Femenino , Animales , Ovinos/genética , Bovinos , Andrógenos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Gonadotropina/genética , Células de la Granulosa/metabolismo , Complejos Multienzimáticos , Fibrosis
13.
Cell Rep ; 42(9): 113090, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37669164

RESUMEN

Polycystic ovary syndrome (PCOS) is an endocrine disorder and the main cause of anovulatory infertility, in which persistent activation of androgen receptor (AR) due to aberrant acetylation modifications of transcription is a potential trigger; however, the precise mechanisms of AR activation are poorly understood. In this study, AR activation in dehydroepiandrosterone- and letrozole-induced rat PCOS ovaries coincided with a marked increase of a chromatin acetylation "reader" BRD4. Further bioinformatic analysis showed that the AR promoter contained highly conserved binding motifs of BRD4 and HIF-1α. BRD4 and HIF-1α inducibly bound to the histone 3/4 acetylation-modified AR promoter, while administration of a BRD4-selective inhibitor JQ1 reduced the binding and AR transcription and improved the adverse expression of the core fibrotic mediators in PCOS ovaries and DHT-treated granulosa cells. Our data indicate that BRD4 upregulation and the resultant AR transcriptional activation constitute an important regulatory pathway that promotes ovarian fibrosis in PCOS.


Asunto(s)
Síndrome del Ovario Poliquístico , Receptores Androgénicos , Animales , Femenino , Humanos , Ratas , Proteínas de Ciclo Celular , Fibrosis , Proteínas Nucleares/genética , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Factores de Transcripción/genética
14.
DNA Cell Biol ; 41(4): 356-367, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35353637

RESUMEN

Polycystic ovary syndrome (PCOS) is one of the most common gynecological endocrine disorders, with sporadic ovulation, excessive androgens, and polycystic ovarian changes as the main clinical manifestations. Due to the high heterogeneity of its clinical manifestations, the discussion on its pathogenesis has not been unified. Current research has found that genetic factors, hyperandrogenism, chronic inflammation and oxidative stress, insulin resistance, and obesity are strongly associated with PCOS. Recently, when studying the specific mechanisms of the abovementioned factors in PCOS, the biological response process of endoplasmic reticulum stress (ERS) has gradually come to researchers' attention, and several studies have confirmed the involvement of ERS in the pathogenesis of PCOS and the improvement of a series of pathological manifestations of PCOS after the application of ERS inhibitors, which may be a new entry point for the treatment of PCOS. In this article, we review the relationship between ERS and various pathogenic factors of PCOS.


Asunto(s)
Hiperandrogenismo , Resistencia a la Insulina , Síndrome del Ovario Poliquístico , Estrés del Retículo Endoplásmico , Femenino , Humanos , Resistencia a la Insulina/genética , Masculino , Síndrome del Ovario Poliquístico/genética
15.
Aging (Albany NY) ; 13(5): 7084-7095, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33638949

RESUMEN

Polycystic ovarian syndrome (PCOS) is one of the most common reproductive endocrine disorders which are involved in complicated and unknown pathogenic mechanisms. Paeoniflorin (PAE) plays a significant anti-fibrotic role according to previous studies. The aim of the present study was to investigate the effect of PAE on ovarian fibrosis and its underlying mechanism in PCOS development. An animal model of PCOS was established by subcutaneous injection of 60mg/kg/d dehydroepiandrosterone (DHEA) for 35 consecutive days. Rats in PAE-L, PAE-M and PAE-H groups were administrated by gavage with PAE (20, 40, 80 mg/kg/d) for 4 weeks. Our results indicated that DHEA-induced PCOS rats showed similar phenotypes with PCOS patients. PAE could significantly block the DHEA-induced decline of ovary weight and organ coefficient, shorten the prolonged diestrus period, and regulate the irregular estrous cycle of PCOS rats. Moreover, PAE regulated reproductive hormone levels and improved ovarian fibrosis induced by DHEA. PAE treatment could also reduce the expression levels of TGF-ß1 and Smad3, and increase the expression levels of Smad7 and MMP2. In conclusion, PAE significantly attenuated the ovarian fibrosis in PCOS, which could be mediated by TGF-ß1/Smads signaling pathway. Herein, PAE can be used for the treatment of ovarian fibrosis in PCOS progression.


Asunto(s)
Glucósidos/uso terapéutico , Monoterpenos/uso terapéutico , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Deshidroepiandrosterona/farmacología , Modelos Animales de Enfermedad , Ciclo Estral/efectos de los fármacos , Femenino , Ovario/efectos de los fármacos , Ovario/metabolismo , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/metabolismo , Ratas , Ratas Sprague-Dawley
16.
Oncotarget ; 11(47): 4366-4370, 2020 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-33315987

RESUMEN

Ovarian aging is associated with significant changes in the structural organization of collagen, resulting in ovarian fibrosis. In many other tissues, fibrosis increases risks associated with tumorigenesis and metastasis. Thus, it is possible that ovarian fibrosis increases the risk of ovarian cancer by creating a microenvironment more permissive to tumor growth. In this research perspective, we review the impact of female reproduction on the development of ovarian fibrosis and the contributions of genetic and hormonal disruptions such as BRCA mutation, polycystic ovarian syndrome, and infertility to structural changes in the ovary and their relative risk of ovarian cancer. We also explore new fundamental questions in the field of ovarian fibrosis and possible prevention strategies such as metformin.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA