Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Hum Reprod ; 30(5)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38603629

RESUMEN

Polycystic ovary syndrome (PCOS) is one of the most common endocrine disorders in women of reproductive age, but its pathology has not been fully characterized and the optimal treatment strategy remains unclear. Cellular senescence is a permanent state of cell-cycle arrest that can be induced by multiple stresses. Senescent cells contribute to the pathogenesis of various diseases, owing to an alteration in secretory profile, termed 'senescence-associated secretory phenotype' (SASP), including with respect to pro-inflammatory cytokines. Senolytics, a class of drugs that selectively eliminate senescent cells, are now being used clinically, and a combination of dasatinib and quercetin (DQ) has been extensively used as a senolytic. We aimed to investigate whether cellular senescence is involved in the pathology of PCOS and whether DQ treatment has beneficial effects in patients with PCOS. We obtained ovaries from patients with or without PCOS, and established a mouse model of PCOS by injecting dehydroepiandrosterone. The expression of the senescence markers p16INK4a, p21, p53, γH2AX, and senescence-associated ß-galactosidase and the SASP-related factor interleukin-6 was significantly higher in the ovaries of patients with PCOS and PCOS mice than in controls. To evaluate the effects of hyperandrogenism and DQ on cellular senescence in vitro, we stimulated cultured human granulosa cells (GCs) with testosterone and treated them with DQ. The expression of markers of senescence and a SASP-related factor was increased by testosterone, and DQ reduced this increase. DQ reduced the expression of markers of senescence and a SASP-related factor in the ovaries of PCOS mice and improved their morphology. These results indicate that cellular senescence occurs in PCOS. Hyperandrogenism causes cellular senescence in GCs in PCOS, and senolytic treatment reduces the accumulation of senescent GCs and improves ovarian morphology under hyperandrogenism. Thus, DQ might represent a novel therapy for PCOS.


Asunto(s)
Senescencia Celular , Células de la Granulosa , Síndrome del Ovario Poliquístico , Quercetina , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología , Femenino , Senescencia Celular/efectos de los fármacos , Humanos , Animales , Células de la Granulosa/metabolismo , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/patología , Quercetina/farmacología , Ratones , Fenotipo Secretor Asociado a la Senescencia , Adulto , Dasatinib/farmacología , Modelos Animales de Enfermedad , Senoterapéuticos/farmacología , Hiperandrogenismo/patología , Hiperandrogenismo/metabolismo , Interleucina-6/metabolismo , Deshidroepiandrosterona/farmacología
2.
Int J Mol Sci ; 24(24)2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-38139022

RESUMEN

Young female cancer patients can develop chemotherapy-induced primary ovarian insufficiency (POI). Cyclophosphamide (Cy) is one of the most widely used chemotherapies and has the highest risk of damaging the ovaries. Recent studies elucidated the pivotal roles of cellular senescence, which is characterized by permanent cell growth arrest, in the pathologies of various diseases. Moreover, several promising senolytics, including dasatinib and quercetin (DQ), which remove senescent cells, are being developed. In the present study, we investigated whether cellular senescence is involved in Cy-induced POI and whether DQ treatment rescues Cy-induced ovarian damage. Expression of the cellular senescence markers p16, p21, p53, and γH2AX was upregulated in granulosa cells of POI mice and in human granulosa cells treated with Cy, which was abrogated by DQ treatment. The administration of Cy decreased the numbers of primordial and primary follicles, with a concomitant increase in the ratio of growing to dormant follicles, which was partially rescued by DQ. Moreover, DQ treatment significantly improved the response to ovulation induction and fertility in POI mice by extending reproductive life. Thus, cellular senescence plays critical roles in Cy-induced POI, and targeting senescent cells with senolytics, such as DQ, might be a promising strategy to protect against Cy-induced ovarian damage.


Asunto(s)
Insuficiencia Ovárica Primaria , Humanos , Ratones , Femenino , Animales , Insuficiencia Ovárica Primaria/patología , Senoterapéuticos , Ciclofosfamida/toxicidad , Dasatinib/efectos adversos , Senescencia Celular
3.
FASEB J ; 35(11): e21971, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34653284

RESUMEN

It has been recently recognized that prenatal androgen exposure is involved in the development of polycystic ovary syndrome (PCOS) in adulthood. In addition, the gut microbiome in adult patients and rodents with PCOS differs from that of healthy individuals. Moreover, recent studies have suggested that the gut microbiome may play a causative role in the pathogenesis of PCOS. We wondered whether prenatal androgen exposure induces gut microbial dysbiosis early in life and is associated with the development of PCOS in later life. To test this hypothesis, we studied the development of PCOS-like phenotypes in prenatally androgenized (PNA) female mice and compared the gut microbiome of PNA and control offspring from 4 to 16 weeks of age. PNA offspring showed a reproductive phenotype from 6 weeks and a metabolic phenotype from 12 weeks of age. The α-diversity of the gut microbiome of the PNA group was higher at 8 weeks and lower at 12 and 16 weeks of age, and the ß-diversity differed from control at 8 weeks. However, a significant difference in the composition of gut microbiome between the PNA and control groups was already apparent at 4 weeks. Allobaculum and Roseburia were less abundant in PNA offspring, and may therefore be targets for future interventional studies. In conclusion, abnormalities in the gut microbiome appear as early as or even before PCOS-like phenotypes develop in PNA mice. Thus, the gut microbiome in early life is a potential target for the prevention of PCOS in later life.


Asunto(s)
Andrógenos/metabolismo , Microbioma Gastrointestinal , Síndrome del Ovario Poliquístico , Efectos Tardíos de la Exposición Prenatal/microbiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/microbiología , Embarazo
4.
Mol Hum Reprod ; 27(3)2021 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-33493289

RESUMEN

Recent studies have uncovered the critical role of aryl hydrocarbon receptor (AHR) in various diseases, including obesity and cancer progression, independent of its previously identified role as a receptor for endocrine-disrupting chemicals (EDCs). We previously showed that endoplasmic reticulum (ER) stress, a newly recognized local factor in the follicular microenvironment, is activated in granulosa cells from patients with polycystic ovary syndrome (PCOS) and a mouse model of the disease. By affecting diverse functions of granulosa cells, ER stress contributes to PCOS pathology. We hypothesized that expression of AHR and activation of its downstream signaling were upregulated by ER stress in granulosa cells, irrespective of the presence of EDCs, thereby promoting PCOS pathogenesis. In this study, we found that AHR, AHR nuclear translocator (ARNT), and AHR target gene cytochrome P450 1B1 (CYP1B1) were upregulated in the granulosa cells of PCOS patients and model mice. We examined CYP1B1 as a representative AHR target gene. AHR and ARNT were upregulated by ER stress in human granulosa-lutein cells (GLCs), resulting in an increase in the expression and activity of CYP1B1. Administration of the AHR antagonist CH223191 to PCOS mice restored estrous cycling and decreased the number of atretic antral follicles, concomitant with downregulation of AHR and CYP1B1 in granulosa cells. Taken together, our findings indicate that AHR activated by ER stress in the follicular microenvironment contributes to PCOS pathology, and that AHR represents a novel therapeutic target for PCOS.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Estrés del Retículo Endoplásmico , Células de la Granulosa/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Adulto , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Compuestos Azo/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Estudios de Casos y Controles , Células Cultivadas , Citocromo P-450 CYP1B1/metabolismo , Modelos Animales de Enfermedad , Ciclo Estral/metabolismo , Femenino , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/patología , Humanos , Ratones Endogámicos BALB C , Persona de Mediana Edad , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/patología , Pirazoles/farmacología , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Receptores de Hidrocarburo de Aril/genética , Transducción de Señal , Regulación hacia Arriba , Adulto Joven
5.
Artículo en Inglés | MEDLINE | ID: mdl-32911575

RESUMEN

We here describe a case of the prolapse of pedunculated submucosal leiomyoma through the cervix during the treatment of a gonadotropin-releasing hormone (GnRH) antagonist relugolix. We also present the literature review of the cases of leiomyoma prolapse during GnRH modulators. A 55-year-old woman with atypical vaginal bleeding diagnosed submucosal uterine fibroid 6 cm in diameter, and daily oral administration of relugolix was conducted. On the 35th day of the administration, heavy vaginal bleeding suddenly occurred due to leiomyoma prolapse. Finally, she underwent abdominal hysterectomy to treat heavy bleeding. To date, six cases of leiomyoma prolapse during GnRH modulators have been reported, in which all the previous cases were treated with GnRH agonists. This is the first case report of leiomyoma prolapse during GnRH antagonist treatment. Notably, leiomyoma prolapse is a possible common adverse effect of GnRH modulators for the treatment of submucosal leiomyoma, which is caused by rapid decrease in its volume.

6.
J Obstet Gynaecol Res ; 44(2): 191-198, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29316073

RESUMEN

Endometriosis is characterized by the implantation and growth of endometriotic tissues outside the uterus. It is widely accepted the theory that endometriosis is caused by the implantation of endometrial tissue from retrograde menstruation; however, retrograde menstruation occurs in almost all women and other factors are required for the establishment of endometriosis, such as cell survival, cell invasion, angiogenesis, and cell growth. Immune factors in the local environment may, therefore, contribute to the formation and progression of endometriosis. Current evidence supports the involvement of immune cells in the pathogenesis of endometriosis. Peritoneal neutrophils and macrophages secrete biochemical factors that help endometriotic cell growth and invasion, and angiogenesis. Peritoneal macrophages and NK cells in endometriosis have limited capability of eliminating endometrial cells in the peritoneal cavity. An imbalance of T cell subsets leads to aberrant cytokine secretions and inflammation that results in the growth of endometriosis lesions. It is still uncertain whether these immune cells have a role in the initial cause and/or stimulate actions that enhance disease; however, in either case, modulating the actions of these cells may prevent initiation or disease progression. Further studies are needed to deepen the understanding of the pathology of endometriosis and to develop novel management approaches of benefit to women suffering from this disease.


Asunto(s)
Células Dendríticas/patología , Endometriosis/inmunología , Linfocitos/patología , Macrófagos/patología , Neutrófilos/patología , Endometriosis/patología , Femenino , Humanos
7.
J Obstet Gynaecol Res ; 40(3): 770-8, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24320086

RESUMEN

AIM: Endometriosis is a chronic inflammatory disease. Sirtuin 1 (SIRT1) plays a role in regulation of inflammation. The role of SIRT1 in endometriosis remains unknown. We here addressed the anti-inflammatory effects of SIRT1 on endometriosis. METHODS: The expression of SIRT1 in human ovarian endometriomas and eutopic endometria were examined using immunohistochemistry and reverse transcription polymerase chain reaction (RT-PCR). Endometriotic stromal cells (ESC) obtained from endometriomas were exposed to either resveratrol or sirtinol, an activator or inhibitor of sirtuins, respectively, and tumor necrosis factor (TNF)-α-induced interleukin (IL)-8 release from the ESC was assessed at mRNA and protein levels. RESULTS: Both immunochemistry and RT-PCR demonstrated that SIRT1 was expressed in ESC and normal endometrial stromal cells. Resveratrol suppressed TNF-α-induced IL-8 release from the ESC in a dose-dependent manner while sirtinol increased IL-8 release. CONCLUSION: These opposing effects of SIRT1-related agents suggest that IL-8 release from the ESC is modulated through the SIRT1 pathway. Resveratrol may have the potential to ameliorate local inflammation in endometriomas.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Endometriosis/tratamiento farmacológico , Endometrio/efectos de los fármacos , Enfermedades del Ovario/tratamiento farmacológico , Sirtuina 1/metabolismo , Estilbenos/farmacología , Células del Estroma/efectos de los fármacos , Adulto , Benzamidas/farmacología , Células Cultivadas , Endometriosis/inmunología , Endometriosis/metabolismo , Endometriosis/patología , Endometrio/inmunología , Endometrio/metabolismo , Endometrio/patología , Activadores de Enzimas/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Interleucina-8/metabolismo , Naftoles/farmacología , Enfermedades del Ovario/inmunología , Enfermedades del Ovario/metabolismo , Enfermedades del Ovario/patología , Ovario/efectos de los fármacos , Ovario/inmunología , Ovario/metabolismo , Ovario/patología , Resveratrol , Transducción de Señal/efectos de los fármacos , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/química , Sirtuina 1/genética , Células del Estroma/inmunología , Células del Estroma/metabolismo , Células del Estroma/patología
8.
Reprod Toxicol ; 125: 108571, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38458359

RESUMEN

Ulipristal (UPA), a selective progesterone receptor modulator, has both agonistic and antagonistic effects on progesterone receptors. UPA suppresses ovulation by inhibiting the luteinizing hormone (LH) surge from the pituitary gland; however, the direct effect of UPA on ovarian tissue remains poorly studied. In the present study, we examined the effects of UPA on the ovaries of rats. Rats were treated for 28 days with UPA, and the effects of UPA on ovarian tissue were examined histologically and the expression of antioxidant genes and cell death markers were also investigated. UPA treatment increased the number of primordial follicles at each treatment group, primordial follicles increased at all dose levels, but the size/magnitude of the effect decreased with the increasing dose. The number of primary and antral follicles tended to increase with increasing UPA levels. Furthermore, the decrease in primary follicle number could be attributed to the exhaustion of follicles, but the examination of proliferation markers, oxidative stress markers, and cell death markers revealed no remarkable toxic effects on ovarian tissues. These results suggest that UPA treatment promotes follicle development at each stage but inhibits ovulation by suppressing the LH surge, resulting in an increase in atretic follicles or unruptured luteinized cysts. These results suggest that UPA may not have both toxic effects on the ovary and a direct local effect on ovarian follicles, but we should be careful about the effects of prolonged UPA treatment in patients with uterine fibroids on their future fecundity.


Asunto(s)
Norpregnadienos , Ovario , Inhibición de la Ovulación , Humanos , Femenino , Ratas , Animales , Folículo Ovárico , Ovulación , Hormona Luteinizante , Progesterona/farmacología
9.
Front Endocrinol (Lausanne) ; 15: 1346084, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38572478

RESUMEN

Objective: A Mediterranean dietary pattern, sleeping habits, physical activity, and lifestyle appear to affect reproductive health. There are few reports about whether fertility-specific quality of life (QOL) is linked to infertility treatment outcomes. The aim of this study is to investigate when lifestyle factors and fertility-specific QOL are comprehensively considered, which factors influence assisted reproductive technology (ART) outcomes. Methods: This prospective cohort includes 291 women undergoing a first ART treatment at multiple centers in Japan and was designed to evaluate the influence of diet, physical activity, sleeping pattern, computer use duration, and fertility-specific quality of life tool (FertiQoL) score on ART treatment outcomes using a questionnaire. The primary endpoint was the good-quality blastocyst rate per oocyte retrieval and the secondary endpoints were a positive pregnancy test and gestational sac (GS) detection. Results: The good-quality blastocyst rate per oocyte retrieval tended to be negatively associated with frequent fish consumption. After all embryo transfer (ET) cycles, a positive pregnancy test tended to be positively associated with longer sleep and longer computer use (OR = 1.6, 95% CI = 0.9-2.7 and OR = 1.7, CI = 1.0-2.8, respectively) and negatively associated with a smoking partner (OR = 0.6, CI = 0.3-1.0). GS detection was positively and significantly associated with frequent olive oil intake and longer computer use (OR = 1.7, CI = 1.0-3.0 and OR = 1.7, CI = 1.0-3.0, respectively). After ET cycles with a single blastocyst, a positive pregnancy test was positively and significantly associated with longer computer use (OR = 2.0, CI = 1.1-3.7), while GS detection was significantly more likely in women with longer computer use (OR = 2.1, CI = 1.1-3.8) and tended to be more likely in women with a higher FertiQoL Total scaled treatment score (OR = 1.8, CI = 1.0-3.3). p < 0.05 was considered statistically significant and 0.05 ≤ p <0.01 as tendency. Conclusions: Olive oil may be an important factor in dietary habits. Fertility-specific QOL and smoking cessation guidance for partners are important for infertile couples.


Asunto(s)
Infertilidad , Calidad de Vida , Humanos , Embarazo , Femenino , Estudios Prospectivos , Aceite de Oliva , Fertilidad , Fertilización In Vitro , Infertilidad/terapia , Estilo de Vida
10.
Front Cell Dev Biol ; 12: 1365624, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38590777

RESUMEN

The gut microbiome is implicated in the pathogenesis of polycystic ovary syndrome (PCOS), and prenatal androgen exposure is involved in the development of PCOS in later life. Our previous study of a mouse model of PCOS induced by prenatal dihydrotestosterone (DHT) exposure showed that the reproductive phenotype of PCOS appears from puberty, followed by the appearance of the metabolic phenotype after young adulthood, while changes in the gut microbiota was already apparent before puberty. To determine whether the prenatal or postnatal nurturing environment primarily contributes to these changes that characterize prenatally androgenized (PNA) offspring, we used a cross-fostering model to evaluate the effects of changes in the postnatal early-life environment of PNA offspring on the development of PCOS-like phenotypes and alterations in the gut microbiota in later life. Female PNA offspring fostered by normal dams (exposed to an abnormal prenatal environment only, fostered PNA) exhibited less marked PCOS-like phenotypes than PNA offspring, especially with respect to the metabolic phenotype. The gut microbiota of the fostered PNA offspring was similar to that of controls before adolescence, but differences between the fostered PNA and control groups became apparent after young adulthood. In conclusion, both prenatal androgen exposure and the postnatal early-life environment created by the DHT injection of mothers contribute to the development of PCOS-like phenotypes and the alterations in the gut microbiota that characterize PNA offspring. Thus, both the pre- and postnatal environments represent targets for the prevention of PCOS and the associated alteration in the gut microbiota in later life.

11.
J Ovarian Res ; 16(1): 182, 2023 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-37653540

RESUMEN

BACKGROUND: Most women with anovulatory infertility show polycystic ovarian syndrome (PCOS), and androgen excess is known as a key factor involved in pathogenicity of PCOS. However, the mechanism of follicular developmental arrest in PCOS is not completely understood. The reproductive function of Neuropeptide Y (NPY) in the ovary during folliculogenesis was previously reported; NPY function in apoptosis and proliferation of granulosa cells (GCs) is follicular-stage dependent. The objective of this study was to investigate the role of NPY in ovarian follicular development and the pathogenesis of PCOS. METHODS: To simulate the PCOS phenotype using a rat model, 21-day old Sprague Dawley rats were implanted with dihydrotestosterone (DHT) capsule (83 µg/day) and euthanized after 28 days. mRNA and protein content of NPY and its receptors were assessed in GCs from DHT treated rats using RT-qPCR and Western blot, respectively. Proliferation and apoptosis of GCs was assessed using Ki67- and TUNEL assays. Finally, NPY levels were measured in human follicular fluid (FF) from matched PCOS and non-PCOS patients using ELISA. RESULTS: GCs from DHT treated rats (PCOS-GCs) contained significantly less NPY protein and Npy mRNA by 0.16- and 0.56-fold, respectively, and more NPY receptor type 2 and 5 protein by 2.21- and 3.17-fold, respectively, when compared to sham control. Addition of recombinant NPY to PCOS-GCs culture did not alter Ki67-positive but significantly decreased TUNEL-positive cells by 0.65-fold, but not to baseline levels. There was no significant difference in NPY levels in FF between PCOS and non-PCOS subjects. CONCLUSIONS: These results indicate that DHT modulates expression of NPY and its receptors, NPY decreases DHT-induced GCs apoptosis. That alterations in NPY's function might be involved in follicular developmental failure of PCOS.


Asunto(s)
Neuropéptido Y , Síndrome del Ovario Poliquístico , Animales , Femenino , Humanos , Ratas , Apoptosis , Dihidrotestosterona , Células de la Granulosa , Antígeno Ki-67 , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Ratas Sprague-Dawley , ARN Mensajero
12.
Front Endocrinol (Lausanne) ; 14: 1124405, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36875481

RESUMEN

Polycystic ovary syndrome (PCOS) is the most common endocrine disorder among reproductive-age women, affecting up to 15% of women in this group, and the most common cause of anovulatory infertility. Although its etiology remains unclear, recent research has revealed the critical role of endoplasmic reticulum (ER) stress in the pathophysiology of PCOS. ER stress is defined as a condition in which unfolded or misfolded proteins accumulate in the ER because of an imbalance in the demand for protein folding and the protein-folding capacity of the ER. ER stress results in the activation of several signal transduction cascades, collectively termed the unfolded protein response (UPR), which regulates various cellular activities. In principle, the UPR restores homeostasis and keeps the cell alive. However, if the ER stress cannot be resolved, it induces programmed cell death. ER stress has recently been recognized to play diverse roles in both physiological and pathological conditions of the ovary. In this review, we summarize current knowledge of the roles of ER stress in the pathogenesis of PCOS. ER stress pathways are activated in the ovaries of both a mouse model of PCOS and in humans, and local hyperandrogenism in the follicular microenvironment associated with PCOS is responsible for activating these. The activation of ER stress contributes to the pathophysiology of PCOS through multiple effects in granulosa cells. Finally, we discuss the potential for ER stress to serve as a novel therapeutic target for PCOS.


Asunto(s)
Síndrome del Ovario Poliquístico , Animales , Ratones , Humanos , Femenino , Estrés del Retículo Endoplásmico , Respuesta de Proteína Desplegada , Apoptosis , Microambiente Tumoral
13.
J Ovarian Res ; 16(1): 74, 2023 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-37046285

RESUMEN

Polycystic ovarian syndrome (PCOS) is a complex multi-factorial syndrome associated with androgen excess and anovulatory infertility. In the current study, we investigated the role of dihydrotestosterone-induced exosomal miR-379-5p release in determining the destiny of the developing follicles. Our hypothesis was that androgen regulates granulosa cell miR-379-5p content by facilitating its exosomal release in a follicular-stage dependent manner, a process which determines granulosa cell fate. Compared to human non-PCOS subjects, individuals with PCOS exhibit higher follicular fluid free testosterone levels, lower exosomal miR-379-5p content and granulosa cell proliferation. Androgenized rats exhibited lower granulosa cell miR-379-5p but higher phosphoinositide-dependent kinase-1 (PDK1; a miR-379-5p target) content and proliferation. Androgen reduced granulosa cell miR-379-5p content by increasing its exosomal release in preantral follicles, but not in antral follicles in vitro. Studies with an exosomal release inhibitor confirmed that androgen-induced exosomal miR-379-5p release decreased granulosa cell miR-379-5p content and proliferation. Ovarian overexpression of miR-379-5p suppressed granulosa cell proliferation, and basal and androgen-induced preantral follicle growth in vivo. These findings suggest that increased exosomal miR-379-5p release in granulosa cells is a proliferative response to androgenic stimulation specific for the preantral stage of follicle development and that dysregulation of this response at the antral stage is associated with follicular growth arrest, as observed in human PCOS.


Asunto(s)
MicroARNs , Síndrome del Ovario Poliquístico , Femenino , Humanos , Ratas , Animales , Andrógenos/farmacología , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/genética , Células de la Granulosa , MicroARNs/genética
14.
Hum Reprod ; 27(10): 3028-35, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22888172

RESUMEN

STUDY QUESTION: Is thymic stromal lymphopoietin (TSLP) involved in the pathophysiology of endometriosis? SUMMARY ANSWER: TSLP is up-regulated by interleukin (IL)-1ß and may be involved in the development of endometriosis. WHAT IS KNOWN ALREADY: Endometriosis is a chronic inflammatory disease in which the Th2 immune response is activated and has been suggested to promote the disease. TSLP is a master cytokine that drive Th2 immune response. STUDY DESIGN, SIZE, DURATION: A laboratory study. PARTICIPANTS/MATERIALS, SETTING, METHODS: Primary cultures of endometrioma stromal cells (ESCs) were treated with IL-1ß, a typical inflammatory cytokine associated with endometriosis. Gene expression of TSLP in ESCs and secretion of TSLP protein from ESCs were studied using quantitative PCR and a specific ELISA. Interferon γ (IFNγ), a typical Th1 cytokine, and IL-4, a typical Th2 cytokine, were added to the culture to evaluate their effect on the IL-1ß-induced secretion of TSLP. Inhibitors of p38 mitogen-activated protein kinase (MAPK), p42/44 MAPK and stress-activated protein kinase/Jun amino-terminal kinase (SAPK/JNK) were added to the culture to examine intracellular signals involved in IL-1ß-induced TSLP secretion. The expression of TSLP in endometrioma tissue was examined by immunohistochemistry. The concentration of TSLP in the serum and peritoneal fluid (PF) of women with or without endometriosis was measured with a specific ELISA. MAIN RESULTS AND THE ROLE OF CHANCE: IL-1ß stimulated the expression of TSLP mRNA and secretion of TSLP protein from ESCs. IL-4 enhanced the IL-1ß-induced TSLP secretion from ESCs, while IFNγ reduced it. Inhibitors of p42/44 MAPK, p38 MAPK and SAPK/JNK suppressed the IL-1ß-induced secretion of TSLP from ESCs. Positive immunostaining of TSLP was observed in the stroma of endometrioma tissue. TSLP concentrations in the serum and PF were both higher in women with endometriosis compared with those without endometriosis. LIMITATIONS, REASONS FOR CAUTION: The present study was only in vitro. The samples used for culture were endometrioma tissues, not including other types of endometriosis. Therefore, the present findings should be interpreted with caution. WIDER IMPLICATIONS OF THE FINDINGS: This study provided new insights in the Th2 immune response-related mechanism in endometriosis. STUDY FUNDING: This study is partly supported by grants from the Ministry of Health, Labour and Welfare, and the Ministry of Education, Culture, Sports, Science and Technology. The authors have no conflicts of interest to declare.


Asunto(s)
Citocinas/metabolismo , Endometriosis/etiología , Interleucina-1beta/farmacología , Células del Estroma/efectos de los fármacos , Células Cultivadas , Citocinas/genética , Citocinas/fisiología , Endometriosis/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Interferón gamma/farmacología , Interleucina-4/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/farmacología , Proteína Quinasa 1 Activada por Mitógenos/farmacología , Reacción en Cadena de la Polimerasa , Células del Estroma/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Linfopoyetina del Estroma Tímico
15.
Biomolecules ; 12(8)2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-36008931

RESUMEN

Endoplasmic reticulum (ER) stress activated in granulosa cells contributes to the pathophysiology of polycystic ovary syndrome (PCOS). In addition, recent studies have demonstrated that Notch signaling plays multiple roles in the ovary via cell-to-cell interactions. We hypothesized that ER stress activated in granulosa cells of antral follicles in PCOS induces Notch signaling in these cells, and that activated Notch signaling induces aberrant cumulus-oocyte complex (COC) expansion. Expression of Notch2 and Notch-target transcription factors was increased in granulosa cells of PCOS patients and model mice. ER stress increased expression of Notch2 and Notch-target transcription factors in cultured human granulosa-lutein cells (GLCs). Inhibition of Notch signaling abrogated ER stress-induced expression of genes associated with COC expansion in cultured human GLCs, as well as ER stress-enhanced expansion of cumulus cells in cultured murine COCs. Furthermore, inhibition of Notch signaling reduced the areas of COCs in PCOS model mice with activated ER stress in the ovary, indicating that Notch signaling regulates COC expansion in vivo. Our findings suggest that Notch2 signaling is activated in granulosa cells in PCOS and regulates COC expansion. It remains to be elucidated whether aberrant COC expansion induced by the ER stress-Notch pathway is associated with ovulatory dysfunction in PCOS patients.


Asunto(s)
Síndrome del Ovario Poliquístico , Animales , Células del Cúmulo/metabolismo , Estrés del Retículo Endoplásmico , Femenino , Humanos , Ratones , Oocitos/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Factores de Transcripción/metabolismo
16.
J Ovarian Res ; 13(1): 5, 2020 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-31915051

RESUMEN

BACKGROUND: The complex regulatory mechanism involved in ovarian follicular development is not completely understood. Neuronal neuropeptide Y (NPY) is involved in the regulation of feeding behavior, energy homeostasis, and reproduction behavior, while its function in ovarian follicular development is not clear. The objective of this study was to investigate if and how NPY regulates follicle development in the ovary. METHODS: All experiments were performed using Sprague Dawley rats. To understand NPY expression pattern at different stages of follicular development, NPY content was assessed using immunohistochemistry in individual follicles. NPY and its receptors expression pattern were evaluated in granulosa cells isolated from preantral (PA), early antral (EA) and late antral follicles (LAF). The influence of NPY on granulosa cell proliferation and apoptosis were further assessed in vitro, using Ki67- and TUNEL-positivity assays. To investigate whether NPY induced-proliferation in EA granulosa cells is mediated through the activation of NPY receptor Y5 (NPY5R) and Mitogen-activated protein kinase (MEK) signal pathway, EA granulosa cells were treated with NPY5R antagonist (CGP71683) and MEK inhibitors (PD98059 and U0126), and Ki67-positive cells were assessed. RESULTS: NPY protein expression was follicular stage-dependent and cell type-specific. NPY signal intensity in EA was higher than those in PA and LAF. Antral granulosa cells showed the highest signal intensity compared to mural granulosa cells, cumulus cells and theca cells. Granulosa cells NPY protein content and mRNA abundance were higher in EA than in LAF. NPY receptor contents in granulosa cells were follicular stage-dependent. While NPY reduced apoptosis of EA granulosa cells, it increased the proliferation through NPY5R and MEK pathway. In contrast, in LAF granulosa cells, NPY reduced proliferation and increased the number of apoptotic cells, with no significant effects on PA granulosa cells. CONCLUSION: This study is the first to evaluate the intraovarian role of NPY in granulosa cells at various stage of follicular development. These results indicate that NPY regulates granulosa cells proliferation and apoptosis in a follicular stage-dependent and autocrine manner. NPY may play a role in pathogenesis of ovarian follicular disorders.


Asunto(s)
Apoptosis/genética , Proliferación Celular/genética , Regulación de la Expresión Génica , Células de la Granulosa/metabolismo , Neuropéptido Y/genética , Folículo Ovárico/metabolismo , Animales , Western Blotting , Células Cultivadas , Femenino , Células de la Granulosa/citología , Sistema de Señalización de MAP Quinasas , Neuropéptido Y/metabolismo , Folículo Ovárico/citología , Ovario/citología , Ovario/metabolismo , Ratas Sprague-Dawley , Receptores de Neuropéptido Y/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Endocrinology ; 161(2)2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32020188

RESUMEN

Polycystic ovary syndrome (PCOS) is associated with hyperandrogenism, and we previously found that androgens activate endoplasmic reticulum (ER) stress in granulosa cells from patients with PCOS. In addition, recent studies demonstrated the accumulation of advanced glycation end products (AGEs) in granulosa cells from PCOS patients, which contribute to the pathology. Therefore, we hypothesized that androgens upregulate the receptor for AGEs (RAGE) expression in granulosa cells by activating ER stress, thereby increasing the accumulation of AGEs in these cells and contributing to the pathology. In the present study, we show that testosterone increases RAGE expression and AGE accumulation in cultured human granulosa-lutein cells (GLCs), and this is reduced by pretreatment with tauroursodeoxycholic acid (TUDCA), an ER stress inhibitor in clinical use. Knockdown of the transcription factor C/EBP homologous protein (CHOP), an unfolded protein response factor activated by ER stress, inhibits testosterone-induced RAGE expression and AGE accumulation. The expression of RAGE and the accumulation of AGEs are upregulated in granulosa cells from PCOS patients and dehydroepiandrosterone-induced PCOS mice. Administration of the RAGE inhibitor FPS-ZM1 or TUDCA to PCOS mice reduces RAGE expression and AGE accumulation in granulosa cells, improves their estrous cycle, and reduces the number of atretic antral follicles. In summary, our findings indicate that hyperandrogenism in PCOS increases the expression of RAGE and accumulation of AGEs in the ovary by activating ER stress, and that targeting the AGE-RAGE system, either by using a RAGE inhibitor or a clinically available ER stress inhibitor, may represent a novel approach to PCOS therapy.


Asunto(s)
Estrés del Retículo Endoplásmico , Productos Finales de Glicación Avanzada/metabolismo , Células de la Granulosa/metabolismo , Hiperandrogenismo/metabolismo , Síndrome del Ovario Poliquístico/etiología , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Animales , Benzamidas/uso terapéutico , Estudios de Casos y Controles , Células Cultivadas , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Ratones Endogámicos BALB C , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Síndrome del Ovario Poliquístico/metabolismo , Ácido Tauroquenodesoxicólico/uso terapéutico , Testosterona
18.
Reprod Sci ; 26(8): 1105-1110, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30326781

RESUMEN

OBJECTIVE: The aim of this study was to clarify the expression of Ninj1 in endometriosis and adenomyosis lesions, and its inductive factor in human endometriotic stromal cells (ESCs). BACKGROUND: Nerve injury-induced protein 1 (Ninj1) is a molecule originally identified in dorsal root ganglion neurons and Schwann cells after nerve injury and promotes neurite outgrowth. The aim of this study was to clarify the expression of Ninj1 in endometriosis and adenomyosis lesions, and its inductive factor in human endometriotic stromal cells (ESCs). MATERIALS AND METHODS: Tissues were obtained with consent from patients diagnosed with ovarian endometrioma (n = 15 in total), peritoneal endometriosis (n = 5), adenomyosis (n = 5), and other gynecological disorders (n = 5, control) during surgery. Immunohistochemistry was conducted in order to detect Ninj1 protein expression in the lesion of endometriosis, adenomyosis, and eutopic endometrium. Nerve fibers in the ovarian endometrioma were detected by positive staining of PGP-9.5. To evaluate the effects of IL-1ß on Ninj1 gene expression in endometriosis, ESCs isolated from ovarian endometrioma (n = 5) were treated with IL-1ß (5 ng/mL) for 3 or 6 hours. Messenger RNA (mRNA) expression for Ninj1 was examined using quantitative RT-PCR. RESULTS: The Ninj1 protein was expressed by ovarian endometrioma, peritoneal endometriotic, and adenomyotic tissue. Nerve fibers were found in the areas of positive staining for Ninj1 in ovarian endometrioma. IL-1ß, an indicator of inflammation in endometriosis, significantly increased Ninj1 mRNA expression by ESC. CONCLUSION: Our study demonstrates that Ninj1 is expressed in endometriosis and adenomyosis and is induced by the inflammatory stimuli. Given the neurogenetic property of Ninj1, our results imply that Ninj1, induced by inflammation in endometriosis lesion, may contribute to the pathogenesis of pain symptoms characteristic of endometriosis.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Endometriosis/metabolismo , Endometrio/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Enfermedades del Ovario/metabolismo , Enfermedades Peritoneales/metabolismo , Células del Estroma/metabolismo , Adenomiosis/metabolismo , Endometrio/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Inflamación/metabolismo , Interleucina-1beta/farmacología , Células del Estroma/efectos de los fármacos
19.
Fertil Steril ; 107(1): 167-173.e2, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27793384

RESUMEN

OBJECTIVES: To characterize peritoneal dendritic cells (DCs) in endometriosis and to clarify their role in its etiology. DESIGN: Experimental. SETTING: University hospital. PATIENT(S): Sixty-three women (35 patients with endometriosis and 28 control women) who had undergone laparoscopic surgery. INTERVENTION(S): Peritoneal DCs from endometriosis and control samples were analyzed for the expression of cell surface markers. Monocyte-derived dendritic cells (Mo-DCs) were cultured with dead endometrial stromal cells (dESCs) to investigate changes in phagocytic activity and cytokine expression. MAIN OUTCOME MEASURE(S): Cell surface markers and cytokine expression and identification with the use of flow cytometry or reverse-transcription polymerase chain reaction (RT-PCR). Changes in cytokine expression and phagocytic activity of Mo-DCs cultured with dESCs and d-mannan were measured with the use of flow cytometry and RT-PCR. RESULT(S): The proportion of mannose receptor (MR)-positive myeloid DC type 1 was higher in endometriosis samples than in control samples. The blocking of MR reduced phagocytosis of dESCs by Mo-DCs. Mo-DCs cultured with dESCs expressed higher levels of interleukin (IL) 1ß and IL-6 than control samples. CONCLUSION(S): Peritoneal DCs in endometriosis tissue express high levels of MR, which promotes phagocytosis of dead endometrial cells and thereby contributes to the etiology of endometriosis.


Asunto(s)
Células Dendríticas/metabolismo , Endometriosis/metabolismo , Lectinas Tipo C/metabolismo , Lectinas de Unión a Manosa/metabolismo , Receptores de Superficie Celular/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Endometriosis/diagnóstico , Endometriosis/inmunología , Endometriosis/patología , Femenino , Hospitales Universitarios , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Lectinas Tipo C/efectos de los fármacos , Mananos/farmacología , Receptor de Manosa , Lectinas de Unión a Manosa/efectos de los fármacos , Cavidad Peritoneal/citología , Fagocitosis , Receptores de Superficie Celular/efectos de los fármacos , Células del Estroma/inmunología , Células del Estroma/patología
20.
J Reprod Immunol ; 119: 44-48, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28064076

RESUMEN

Drospirenone has been used as a progestin in oral contraceptives with ethinyl estradiol (DRSP/EE) and is expected to regulate endometriosis, however, the direct effects of drospirenone on endometriosis have not been clarified. The aim of this study was to evaluate the anti-inflammatory, anti-angiogenic and anti-neurogenic effects of drospirenone on endometriotic stromal cells (ESC). ESC isolated from endometriotic tissues were obtained from patients during laparoscopic surgery for ovarian endometriosis. ESC were exposed to IL-1ß and cultured in the absence or presence of drospirenone. mRNA expression was evaluated using quantitative RT-PCR, and protein was measured using ELISAs. To evaluate the effect of drospirenone on progesterone receptor (PR) and mineralocorticoid receptor (MR), ESC were transfected with siRNA against PR (siPR) and MR (siMR), and cultured in the presence or absence of drospirenone. Drospirenone significantly decreased IL-6, IL-8, VEGF and NGF mRNA expression by ESC. Drospirenone (10-5M) significantly decreased IL-6 secretion and 10-7M drospirenone decreased IL-8 and VEGF secretion. Knockdown of PR, but not MR, negated the effects of drospirenone. In summary, this study demonstrates that drospirenone has anti-inflammatory, anti-angiogenic and anti-neurogenic effects on ESC and these effects are mediated by PR. These drospirenone effects may contribute to the regulatory effects of drospirenone-containing oral contraceptives on endometriosis.


Asunto(s)
Androstenos/farmacología , Antiinflamatorios/farmacología , Endometriosis/tratamiento farmacológico , Endometrio/patología , Células del Estroma/efectos de los fármacos , Adulto , Androstenos/uso terapéutico , Antiinflamatorios/uso terapéutico , Células Cultivadas , Anticonceptivos Orales/uso terapéutico , Citocinas/genética , Citocinas/metabolismo , Etinilestradiol/uso terapéutico , Femenino , Humanos , Factor de Crecimiento Nervioso/genética , Factor de Crecimiento Nervioso/metabolismo , ARN Interferente Pequeño/genética , Receptores de Mineralocorticoides/genética , Receptores de Mineralocorticoides/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Células del Estroma/inmunología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA