Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
1.
Cytokine ; 179: 156639, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38733946

RESUMEN

AIMS: Polycystic ovarian syndrome (PCOS) is one of the most common (about 5-20%) reproductive disorders in women of reproductive age; it is characterized by polycystic ovaries, hyperandrogenism, and oligo/ anovulation. The levels and expression of ovarian adipokines are deregulated in the PCOS. Apelin is an adipokine that acts through its receptor (APJ) and is known to express in the various tissues including the ovary. It has also been suggested that apelin and APJ could be targeted as therapeutic adjuncts for the management of PCOS. However, no study has been conducted on the management of PCOS by targeting the apelin system. Thus, we aimed to evaluate its impact on combating PCOS-associated ovarian pathogenesis. METHODS: The current work employed a letrozole-induced-hyperandrogenism PCOS-like mice model to investigate the effects of apelin13 and APJ, antagonist ML221. The PCOS model was induced by oral administration of letrozole (1 mg/kg) for 21 days. A total of four experimental groups were made, control, PCOS control, PCOS + aplein13, and PCOS + ML221. The treatment of apelin13 and ML221 was given from day 22 for two weeks. KEY FINDINGS: The letrozole-induced PCOS-like features such as hyperandrogenism, cystic follicle, decreased corpus luteum, elevated levels of LH/FSH ratio, and up-regulation of ovarian AR expression were ameliorated by apelin13 and ML221 treatment. However, the PCOS-augmented oxidative stress and apoptosis were suppressed by apelin 13 treatments only. ML221 treatment still showed elevated oxidative stress and stimulated apoptosis as reflected by decreased antioxidant enzymes and increased active caspase3 and Bax expression. The expression of ERs was elevated in all groups except control. Furthermore, the PCOS model showed elevated expression of APJ and apelin13 treatment down-regulated its own receptor. Overall, observing the ovarian histology, corpus luteum formation, and decreased androgen levels by both apelin13 and ML221 showed ameliorative effects on the cystic ovary. SIGNIFICANCE: Despite the similar morphological observation of ovarian histology, apelin13 and ML221 exhibited opposite effects on oxidative stress and apoptosis. Therefore, apelin13 (which down-regulates APJ) and ML221 (an APJ antagonist) may have suppressed APJ signalling, which would account for our findings on the mitigation of polycystic ovarian syndrome. In conclusion, both apelin13 and ML221 mediated mitigation have different mechanisms, which need further investigation.


Asunto(s)
Receptores de Apelina , Apelina , Letrozol , Ovario , Síndrome del Ovario Poliquístico , Letrozol/farmacología , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Animales , Femenino , Receptores de Apelina/metabolismo , Ratones , Apelina/metabolismo , Ovario/metabolismo , Ovario/patología , Ovario/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Hiperandrogenismo/metabolismo , Hiperandrogenismo/inducido químicamente , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad
2.
BMJ Open ; 13(11): e073619, 2023 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-37989369

RESUMEN

OBJECTIVE: Serum prokineticin-1 (s-PROK1) in the second and third trimester of pregnancy is positively correlated to preeclampsia, intrauterine growth restriction (IUGR) and preterm delivery. Women with polycystic ovary syndrome (PCOS) are prone to these adverse pregnancy outcomes. However, the contribution of PROK1 to the development of pregnancy complications and the effect of metformin and hyperandrogenism on s-PROK1 in PCOS have not been studied previously. DESIGN: This work is a post hoc analysis of two prospective, randomised, placebo-controlled trials. SETTING: Pregnant women with PCOS were included from 11 study centres in Norway. PARTICIPANTS: From 313 women, 264 participated in the present study after exclusions due to dropouts or insufficient serum samples. INTERVENTION: Women with PCOS were randomly administered with metformin or placebo, from first trimester to delivery. PRIMARY AND SECONDARY OUTCOME MEASURES: s-PROK1 was analysed using ELISA at gestational week 19 and related to pregnancy complications, fasting insulin levels, homoeostatic model assessment for insulin resistance (HOMA-IR), testosterone, or androstenedione levels, metformin use, PCOS phenotype and hyperandrogenism. RESULTS: Maternal s-PROK1 in the second trimester did not predict pregnancy-induced hypertension, pre-eclampsia or late miscarriage/preterm delivery in women with PCOS. However, s-PROK1 was lower in women who used metformin before inclusion, both in those randomised to metformin and to placebo, compared with those who did not. s-PROK1 was also lower in those who used metformin both at conception and during pregnancy compared with those who used metformin from inclusion or did not use metformin at all. s-PROK1 was lower in hyperandrogenic compared with normo-androgenic women with PCOS. CONCLUSIONS: Maternal s-PROK1 in the second trimester did not predict pregnancy complications in PCOS. Those who used metformin at conception and/or during pregnancy had lower s-PROK1. PCOS women with hyperandrogenism exhibited lower s-PROK1 compared with normo-adrogenic phenotypes. TRIAL REGISTRATION NUMBER: NCT03259919 and NCT00159536.


Asunto(s)
Hormonas Gastrointestinales , Hiperandrogenismo , Metformina , Síndrome del Ovario Poliquístico , Preeclampsia , Complicaciones del Embarazo , Nacimiento Prematuro , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina , Recién Nacido , Femenino , Embarazo , Humanos , Metformina/uso terapéutico , Síndrome del Ovario Poliquístico/complicaciones , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Hiperandrogenismo/inducido químicamente , Hiperandrogenismo/complicaciones , Hiperandrogenismo/tratamiento farmacológico , Estudios Prospectivos , Complicaciones del Embarazo/tratamiento farmacológico , Complicaciones del Embarazo/inducido químicamente , Preeclampsia/tratamiento farmacológico , Hormonas Gastrointestinales/uso terapéutico
3.
Sci Rep ; 13(1): 2682, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36792653

RESUMEN

Adverse in-utero insults during fetal life alters offspring's developmental trajectory, including that of the cardiovascular system. Gestational hyperandrogenism is once such adverse in-utero insult. Gestational testosterone (T)-treatment, an environment of gestational hyperandrogenism, manifests as hypertension and pathological left ventricular (LV) remodeling in adult ovine offspring. Furthermore, sexual dimorphism is noted in cardiomyocyte number and morphology in fetal life and at birth. This study investigated transcriptional changes and potential biomarkers of prenatal T excess-induced adverse cardiac programming. Genome-wide coding and non-coding (nc) RNA expression were compared between prenatal T-treated (T propionate 100 mg intramuscular twice weekly from days 30 to 90 of gestation; Term: 147 days) and control ovine LV at day 90 fetus in both sexes. Prenatal T induced differential expression of mRNAs in the LV of female (2 down, 5 up) and male (3 down, 1 up) (FDR < 0.05, absolute log2 fold change > 0.5); pathways analysis demonstrated 205 pathways unique to the female, 382 unique to the male and 23 common pathways. In the male, analysis of ncRNA showed differential regulation of 15 lncRNAs (14 down, 1 up) and 27 snoRNAs (26 down and 1 up). These findings suggest sexual dimorphic modulation of cardiac coding and ncRNA with gestational T excess.


Asunto(s)
Hiperandrogenismo , Efectos Tardíos de la Exposición Prenatal , Embarazo , Humanos , Ovinos , Animales , Femenino , Masculino , Ventrículos Cardíacos/metabolismo , Hiperandrogenismo/inducido químicamente , Testosterona/farmacología , Feto/metabolismo , Miocitos Cardíacos/metabolismo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente
4.
Front Endocrinol (Lausanne) ; 13: 868572, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35757434

RESUMEN

Obesity, altered glucose homeostasis, hyperinsulinism, and reproductive dysfunction develops in female humans and mammals with hyperandrogenism. We previously reported that low dose dihydrotestosterone (DHT) administration results in metabolic and reproductive dysfunction in the absence of obesity in female mice, and conditional knock-out of the androgen receptor (Ar) in the liver (LivARKO) protects female mice from DHT-induced glucose intolerance and hyperinsulinemia. Since altered metabolic function will regulate reproduction, and liver plays a pivotal role in the reversible regulation of reproductive function, we sought to determine the reproductive phenotype of LivARKO mice under normal and hyperandrogenemic conditions. Using Cre/Lox technology, we deleted the Ar in the liver, and we observed LivARKO female mice have normal puberty timing, cyclicity and reproductive function. After DHT treatment, like control mice, LivARKO experience altered estrous cycling, reduced numbers of corpus lutea, and infertility. Liver Ar is not involved in hyperandrogenemia-induced reproductive dysfunction. The reproductive dysfunction in the DHT-treated LivARKO lean females with normal glucose homeostasis indicates that androgen-induced reproductive dysfunction is independent from metabolic dysfunction.


Asunto(s)
Hiperandrogenismo , Síndrome del Ovario Poliquístico , Receptores Androgénicos , Reproducción , Animales , Dihidrotestosterona/farmacología , Femenino , Glucosa/metabolismo , Humanos , Hiperandrogenismo/inducido químicamente , Hiperandrogenismo/genética , Hiperandrogenismo/metabolismo , Hiperandrogenismo/fisiopatología , Hígado/metabolismo , Ratones , Obesidad/metabolismo , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/metabolismo , Receptores Androgénicos/deficiencia , Receptores Androgénicos/metabolismo , Reproducción/fisiología
5.
Steroids ; 177: 108936, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34752810

RESUMEN

Polycystic ovary syndrome (PCOS) is a multi-factorial endocrine disorder associated with hyperandrogenism. Dehydroepiandrosterone (DHEA) administration to prepubertal rats stimulates androgen biosynthesis and generation of the PCOS model. The present study aimed to evaluate the anti-androgenic effects of quercetin (Q) in comparison with metformin (MET) on hyperandrogenism and ovarian dysfunction in a DHEA-induced PCOS rat model. After induction of PCOS, female rats were allocated into six groups with 7 rats in each group: normal control; PCOS (DHEA), MET (25 mg/kg, oral administration), Q (25 mg/kg, oral administration), DHEA + MET (25 mg/kg, oral administration), and DHEA + Q (25 mg/kg, oral administration) for 28 days. MET and Q individually reduced body weight, serum free testosterone (T) and luteinizing hormone (LH), and LH/follicle-stimulating hormone (FSH) ratio in the PCOS rats. Both treatments elevated estradiol (E2) level, ovarian aromatase protein content, and E2/free T ratio in the PCOS rats. Additionally, MET and Q increased preantral, antral, and preovulatory follicles and corpora lutea counts, while both treatments decreased atretic follicle count and eliminated the formation of cysts in the PCOS rats. MET and Q reduced ovarian Bax and elevated Bcl-2 protein abundance in the PCOS rats. Our study revealed that Q is as effective as MET in reducing hyperandrogenism via decreasing free T level and improving hypothalamic-pituitary-ovarian axis function. The results suggest that MET and Q may enhance E2 concentration, ovarian aromatase protein content, folliculogenesis, and decrease atresia via attenuation of hyperandrogenism in PCOS rats.


Asunto(s)
Antagonistas de Andrógenos/farmacología , Modelos Animales de Enfermedad , Hiperandrogenismo/tratamiento farmacológico , Ovario/efectos de los fármacos , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Quercetina/farmacología , Antagonistas de Andrógenos/química , Animales , Deshidroepiandrosterona , Femenino , Hiperandrogenismo/inducido químicamente , Hiperandrogenismo/metabolismo , Ovario/metabolismo , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/metabolismo , Quercetina/química , Ratas , Ratas Wistar
6.
Life Sci ; 276: 119409, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-33781825

RESUMEN

Polycystic ovary syndrome is a common reproductive disorder in the female of reproductive age, which is characterized by hyperandrogenism, insulin resistance, cystic ovary and infertility. The level of pro-inflammatory adipokine, visfatin is elevated in PCOS conditions in human and animal. In this study, letrozole induced hyperandrogenised PCOS mice model have been used to unravel the effects of visfatin inhibition. The results showed that letrozole induced hyperandrogenisation significantly (p < 0.05) elevates ovarian visfatin concentration from 66.03 ± 1.77 to 112.08 ± 3.7 ng/ml, and visfatin expression to 2.5 fold (p < 0.05) compared to control. Visfatin inhibition in PCOS by FK866 has significantly (p < 0.05) suppressed the secretion of androgens, androstenedione (from 0.329 ± 0.07 to 0.097 ± 0.01 ng/ml) and testosterone levels (from 0.045 ± 0.003 to 0.014 ± 0.0009 ng/ml). Ovarian histology showed that visfatin inhibition suppressed cyst formation and promotes corpus luteum formation. Visfatin inhibition has suppressed apoptosis and increases the expression of BCL2 along with increase in the proliferation (GCNA expression elevated). Visfatin inhibition has increased ovarian glucose content (from 167.05 ± 8.5 to 210 ± 7 mg/dl), along with increase in ovarian GLUT8 expression. In vitro study has also supported the in vivo findings where FK866 treatment significantly (p < 0.05) suppressed testosterone (control-3.84 ± 0.44 ng/ml, 1 nM FK866-2.02 ± 0.048 ng/ml, 10 nM FK866-1.74 ± 0.20 ng/ml) and androstenedione (control-4.68 ± 0.91 ng/ml, 1 nM FK866-3.38 ± 0.27 ng/ml, 10 nM FK866-4.55 ± 0.83 ng/ml) production from PCOS ovary. In conclusion, this is first report, which showed that visfatin inhibition by FK866 in hyperandrogenised mice ameliorates pathogenesis of PCOS. Thus, it may be suggested that visfatin inhibition could have a therapeutic potential in PCOS management along with other intervention.


Asunto(s)
Acrilamidas/farmacología , Citocinas/antagonistas & inhibidores , Modelos Animales de Enfermedad , Hiperandrogenismo/tratamiento farmacológico , Letrozol/toxicidad , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Piperidinas/farmacología , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Andrógenos/metabolismo , Animales , Glucemia/metabolismo , Femenino , Hiperandrogenismo/inducido químicamente , Hiperandrogenismo/metabolismo , Hiperandrogenismo/patología , Resistencia a la Insulina , Ratones , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología
7.
Gynecol Endocrinol ; 37(1): 72-77, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32988245

RESUMEN

BACKGROUND: Increasing evidences have proposed that kisspeptins may be involved in polycystic ovary syndrome (PCOS) including hyperandrogenism. This work aimed to investigate the effect of kisspeptin in hyperandrogenism induced by testosterone. METHODS: The most suitable concentration of testosterone to induce hyperandrogenism was determined by detecting the mRNA changes of kisspeptin and macrophages pro-inflammatory cytokines. The role of kisspeptin in hyperandrogenism was investigated by RT-PCR of kisspeptin and pro-inflammatory cytokines, by CCK-8 of cell viability, by Annexin V-FITC/PI staining followed by flow cytometry of apoptosis, by ELISA of pro-inflammatory cytokines and by Western blot of kisspeptin and antiapoptotic Bcl-2 and proapoptotic Bax. RESULTS: We found that testosterone elevated kisspeptin, pro-inflammatory cytokines expressions and nitrite release in excessive androgen stimulated macrophages and further inhibited the macrophages cell viability and increased apoptosis. Kisspeptin knockdown reversed the tendency caused by testosterone and decreased apoptosis in macrophages treated with testosterone. Moreover, mRNA and protein expression levels of Bcl-2 and Bax were assessed. We showed a reduction in Bcl-2 mRNA and protein expression levels and an overexpression of Bax mRNA and protein expression levels. Kiss1 silencing reversed Bcl-2 and Bax expressions. CONCLUSION: Kisspeptin inactivation confers resistance in hyperandrogenism by inhibiting pro-inflammatory cytokines expressions and apoptosis. Our results may help to comprehend the role of kisspeptin in the mechanisms of hyperandrogenism.


Asunto(s)
Citocinas/metabolismo , Hiperandrogenismo/metabolismo , Kisspeptinas/metabolismo , Macrófagos/metabolismo , Nitritos/metabolismo , Animales , Apoptosis , Supervivencia Celular , Femenino , Técnicas de Silenciamiento del Gen , Hiperandrogenismo/inducido químicamente , Hiperandrogenismo/inmunología , Kisspeptinas/genética , Ratas Sprague-Dawley , Bazo/inmunología , Testosterona
8.
Metabolism ; 107: 154241, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32304754

RESUMEN

BACKGROUND: Hyperandrogenism is one of the major characteristics of polycystic ovary syndrome (PCOS). Abnormal miR-125b-5p expression has been documented in multiple diseases, but whether miR-125b-5p is associated with aberrant steroidogenesis in preantral follicles remains unknown. METHODS: Steriod hormone concentrations and miR-125b-5p expression were measured in clinical serum samples from PCOS patients. Using a mouse preantral follicle culture model and a letrozole-induced PCOS mouse model, we investigated the mechanism underlying miR-125b-5p regulation of androgen and oestrogen secretion. RESULTS: The decreased miR-125b-5p expression was observed in the sera from hyperandrogenic PCOS (HA-PCOS) patients. In mouse preantral follicles, inhibiting miR-125b-5p increased the expression of androgen synthesis-related genes and stimulated the secretion of testosterone, while simultaneously downregulating oestrogen synthesis-related genes and decreasing oestradiol release. Ectopically expressed miR-125b-5p reversed the effects on steroidogenesis-related gene expression and hormone release. Mechanistic studies identified Pak3 as a direct target of miR-125b-5p. Furthermore, inhibiting miR-125b-5p facilitated the activation of ERK1/2 in mouse preantral follicles, while inhibiting Pak3 abrogated this activating effect. These results were recapitulated in letrozole-induced PCOS mouse ovaries. Of note, inhibiting PAK3 antagonised the positive effect of miR-125b-5p siRNA on the expressions of androgen synthesis-related enzymes and testosterone secretion. Luteinizing hormone (LH) inhibited miR-125b-5p expression, and stimulated Pak3 expression. CONCLUSION: High serum LH concentrations in PCOS patients repress miR-125b-5p expression, which further increases Pak3 expression, leading to activation of ERK1/2 signalling, thus stimulating the expression of androgen synthesis-related enzymes and testosterone secretion in HA-PCOS.


Asunto(s)
MicroARNs/genética , Folículo Ovárico/metabolismo , Esteroides/biosíntesis , Andrógenos/biosíntesis , Andrógenos/genética , Animales , Estradiol/metabolismo , Estrógenos/biosíntesis , Estrógenos/genética , Femenino , Regulación de la Expresión Génica/genética , Hiperandrogenismo/inducido químicamente , Hiperandrogenismo/metabolismo , Letrozol , Hormona Luteinizante/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Endogámicos C57BL , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/metabolismo , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo
9.
Medicina (Kaunas) ; 56(3)2020 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-32120970

RESUMEN

Background and Objectives: Polycystic ovary syndrome (PCOS) is one of the most prevalent disorders among women of reproductive age. It is considered as a pro-inflammatory state with chronic low-grade inflammation, one of the key factors contributing to the pathogenesis of this disorder. Polycystic ovary is a well-established criterion for PCOS. The present investigation aimed at finding the role of hyperandrogenism, the most important feature of PCOS, in the development of this inflammatory state. To address this problem, we adopted a model system that developed polycystic ovary morphology (PCOM), which could be most effectively used in order to study the role of non-aromatizable androgen in inflammation in PCOS. Materials and Methods: Six rats were used to induce PCOM in 21-days-old female Wistar albino rats by using a pre-determined release of dihydrotestosterone (DHT), a potent non-aromatizable androgen, achieved by implanting a DHT osmotic pump, which is designed to release a daily dose of 83 µg. Results: After 90 days, the rats displayed irregular estrous cycles and multiple ovarian cysts similar to human PCOS. Elevated serum inflammatory markers such as tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß), and the presence of a necrotic lesion in the liver, osteoclast in the femur, multinucleated giant cells and lymphocytes in the ovary based on histopathological observation of DHT-treated rats clearly indicated the onset of inflammation in the hyperandrogenic state. Our results show no significant alterations in serum hormones such as luteinizing hormone (LH), follicle stimulating hormone (FSH), insulin, and cortisol between control and hyperandrogenised rats. DHT was significantly elevated as compared to control. mRNA studies showed an increased expression level of TNF-α and IL-1ß, further, the mRNA expression of urocortin 1 (Ucn-1) was stupendously elevated in the liver of hyperandrogenised rats. Conclusions: Thus, results from this study provide: (1) a good PCOM model system in order to study the inflammatory changes in PCOS aspects, (2) alteration of inflammatory markers in PCOM rats that could be either due to its direct effect or by the regulation of various inflammatory genes and markers in the liver of hyperandrogenic state suggesting the regulatory role of DHT, and (3) alteration in stress-related protein in the liver of PCOM rats.


Asunto(s)
Citocinas/sangre , Dihidrotestosterona/efectos adversos , Hiperandrogenismo/metabolismo , Mediadores de Inflamación/sangre , Síndrome del Ovario Poliquístico/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Hormona Folículo Estimulante/sangre , Hiperandrogenismo/inducido químicamente , Insulina/sangre , Hígado/metabolismo , Hormona Luteinizante/sangre , Síndrome del Ovario Poliquístico/complicaciones , Ratas , Ratas Wistar , Urocortinas/metabolismo
10.
J Endocrinol ; 245(2): 291-300, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32171180

RESUMEN

Polycystic ovary syndrome (PCOS), one of the most common female endocrine disorder, is a prevalent cause of infertility. Hyperandrogenism is a key feature in PCOS and is correlated with increased expression of VEGF and cytokines in the ovaries. We have previously shown that pigment epithelium-derived factor (PEDF), an endogenous protein, presents potent anti-angiogenic and anti-inflammatory activities in the ovary and negates the effects of cytokines and VEGF. Additionally, PEDF plays a role in both pathophysiology and treatment of ovarian-hyperstimulation syndrome (OHSS), frequently seen in PCOS patients. We established hyperandrogenic-PCOS models, both in vivo, using mice exposed prenatally to dihydrotestosterone (DHT) and, in vitro, using human primary granulosa cells (hpGCs) and human granulosa cell line (KGN). In PCOS-induced mice, the mRNA levels of I l-6, V egf and Amh were higher than those of control; yet, treatment with rPEDF decreased these levels. Moreover, treating OHSS-induced PCOS-mice with rPEDF alleviated all OHSS symptoms. Stimulation of hpGCs with DHT resulted in downregulation of PEDF mRNA expression, concomitantly with a significant increase in IL-6 and IL-8 mRNAs expression. However, co-stimulation of DHT with rPEDF attenuated the increase in cytokines expression. The anti-inflammatory effect of PEDF was found to be mediated via PPARγ pathway. Our findings suggest that rPEDF treatment may normalize the ovarian angiogenic-inflammatory imbalance, induced by PCOS-associated hyperandrogenism. Moreover, the therapeutic potency of PEDF in preventing OHSS symptomes offers a rationale for using PEDF as novel physiological treatment for PCOS sequels.


Asunto(s)
Antiinflamatorios/metabolismo , Proteínas del Ojo/metabolismo , Hiperandrogenismo/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Serpinas/metabolismo , Transducción de Señal/fisiología , Animales , Línea Celular , Dihidrotestosterona , Modelos Animales de Enfermedad , Femenino , Células de la Granulosa/metabolismo , Humanos , Hiperandrogenismo/inducido químicamente , Hiperandrogenismo/complicaciones , Ratones , Ovario/metabolismo , Síndrome del Ovario Poliquístico/inducido químicamente
11.
J Endocrinol ; 245(2): 281-289, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32163914

RESUMEN

Polycystic ovary syndrome (PCOS) is a complex syndrome involving both endocrine and metabolic disorders. Gut microbiota and the intestinal immune factor IL-22 play an important role in the pathogenesis of PCOS. However, the therapeutic role of IL-22 in high androgen-induced PCOS mice is not clear. We aimed to determine the therapeutic effects of IL-22 on the DHEA-induced PCOS mouse model and to explore the possible mechanism of IL-22 in regulating hyperandrogenism-associated PCOS. Insulin resistance levels and ovarian functions were investigated in DHEA-induced PCOS mice with or without additional IL-22 treatment. We found that IL-22 could reverse insulin resistance, disturbed estrous cycle, abnormal ovary morphology, and decreased embryo number in DHEA mice. Mechanistically, IL-22 upregulated the browning of white adipose tissue in DHEA mice. This study demonstrated that IL-22-associated browning of white adipose tissue regulated insulin sensitivity and ovarian functions in PCOS, suggesting that IL-22 may be of value for the treatment of PCOS with a hyperandrogenism phenotype.


Asunto(s)
Interleucinas/farmacología , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Andrógenos , Animales , Modelos Animales de Enfermedad , Ciclo Estral/efectos de los fármacos , Femenino , Hiperandrogenismo/inducido químicamente , Hiperandrogenismo/complicaciones , Resistencia a la Insulina/fisiología , Ratones , Ovario/fisiopatología , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/fisiopatología , Interleucina-22
12.
Sci Rep ; 9(1): 19232, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31848372

RESUMEN

Polycystic ovary syndrome (PCOS) is a major reproductive disorder that is responsible for 80% of anovulatory infertility and that is associated with hyperandrogenemia, increased risk of obesity, and white adipose tissue (WAT) dysfunction. We have previously demonstrated that the combination of chronic testosterone (T) treatment and an obesogenic Western-style diet (WSD) exerts synergistic functional effects on WAT, leading to increased lipid accumulation in visceral adipocytes by an unknown mechanism. In this study, we examined the whole-genome transcriptional response in visceral WAT to T and WSD, alone and in combination. We observed a synergistic effect of T and WSD on gene expression, resulting in upregulation of lipid storage genes concomitant with adipocyte hypertrophy. Because DNA methylation is known to be associated with body fat distribution and the etiology of PCOS, we conducted whole-genome DNA methylation analysis of visceral WAT. While only a fraction of differentially expressed genes also exhibited differential DNA methylation, in silico analysis showed that differentially methylated regions were enriched in transcription factor binding motifs, suggesting a potential gene regulatory role for these regions. In summary, this study demonstrates that hyperandrogenemia alone does not induce global transcriptional and epigenetic response in young female macaques unless combined with an obesogenic diet.


Asunto(s)
Metilación de ADN , Dieta Occidental/efectos adversos , Hiperandrogenismo/metabolismo , Grasa Intraabdominal/metabolismo , Obesidad/metabolismo , Transcripción Genética , Animales , Femenino , Hiperandrogenismo/inducido químicamente , Hiperandrogenismo/patología , Grasa Intraabdominal/patología , Macaca mulatta , Obesidad/inducido químicamente , Obesidad/patología , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología , Testosterona/efectos adversos , Testosterona/farmacología
13.
PLoS One ; 14(5): e0216951, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31083690

RESUMEN

Hyperandrogenism is a risk factor of cerebrovascular diseases as androgens can alter markedly the regulation of cerebrovascular tone. We examined the combined impact of androgen excess and vitamin D deficiency (VDD), a common co-morbidity in hyperandrogenic disorders, on remodeling and testosterone-induced vascular responses of anterior cerebral arteries (ACA) in order to evaluate the interplay between androgens and VDD in the cerebral vasculature. Male and female Wistar rats were either fed with vitamin D deficient or vitamin D supplemented diet. Half of the female animals from both groups received transdermal testosterone treatment. After 8 weeks, vessel lumen, wall thickness and testosterone-induced vascular tone of isolated ACA were determined using pressure microangiometry and histological examination. Androgen receptor protein expression in the wall of cerebral arteries was examined using immunohistochemistry. In female rats only combined VDD and testosterone treatment decreased the lumen and increased the wall thickness of ACA. In males, however VDD by itself was able to decrease the lumen and increase the wall thickness. Vascular reactivity showed similar alterations: in females, testosterone constricted the ACA only after combined VDD and hyperandrogenism, whereas in males VDD resulted in increased testosterone-induced contractions in spite of decreased androgen receptor expression. In conclusion, a marked interplay between hyperandrogenism and VDD results in inward remodeling and enhanced testosterone-induced constrictions of cerebral arteries, which might compromise the cerebral circulation and thus, increase the risk of stroke in the long term. In addition, the early cerebrovascular manifestation of VDD appears to require androgen excess and thus, depends on gender.


Asunto(s)
Andrógenos/efectos adversos , Hiperandrogenismo/fisiopatología , Accidente Cerebrovascular/fisiopatología , Testosterona/efectos adversos , Deficiencia de Vitamina D/fisiopatología , Administración Oral , Andrógenos/administración & dosificación , Andrógenos/sangre , Animales , Arteria Cerebral Anterior , Dieta , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Hiperandrogenismo/sangre , Hiperandrogenismo/inducido químicamente , Hiperandrogenismo/complicaciones , Masculino , Ratas , Ratas Wistar , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Riesgo , Factores Sexuales , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/inducido químicamente , Accidente Cerebrovascular/etiología , Testosterona/administración & dosificación , Testosterona/sangre , Vasoconstricción/efectos de los fármacos , Vitamina D/administración & dosificación , Deficiencia de Vitamina D/sangre , Deficiencia de Vitamina D/inducido químicamente , Deficiencia de Vitamina D/complicaciones
14.
Reprod Fertil Dev ; 31(10): 1597-1606, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31142430

RESUMEN

Polycystic ovary syndrome (PCOS) is an endocrine disorder with a high prevalence in women of childbearing age. To date, there is no method of efficiently diagnosing PCOS and curing it completely because its pathomechanism remains unclear. Here, we investigated whether metabolic abnormalities maintain the hyperandrogenism and PCOS-like ovaries and whether the symptoms induced by excess androgen are treatable. We ceased the abnormal dihydrotestosterone (DHT) stimulation to determine changes in PCOS-like mice. After ceasing DHT stimulation, the ovarian morphology and gene expression recovered from the DHT-stimulated status. However, after cessation of DHT stimulation, the hypertrophy of adipose tissues and hepatic steatosis were not significantly restored, and fat accumulation-related gene expression and serum metabolic markers in the mice were altered. These findings showed that the reproductive dysfunction was obviously relieved, but because the metabolic abnormalities were not relieved after the cessation of excess androgen for 30 days, it appears that the latter may not maintain the former.


Asunto(s)
Dihidrotestosterona/administración & dosificación , Dihidrotestosterona/efectos adversos , Hiperandrogenismo/inducido químicamente , Síndrome Metabólico/inducido químicamente , Síndrome Metabólico/patología , Síndrome del Ovario Poliquístico/inducido químicamente , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/patología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Expresión Génica/efectos de los fármacos , Hiperandrogenismo/sangre , Hiperandrogenismo/genética , Síndrome Metabólico/sangre , Síndrome Metabólico/genética , Ratones , Ratones Endogámicos C57BL , Ovario/efectos de los fármacos , Ovario/patología , Síndrome del Ovario Poliquístico/sangre , Síndrome del Ovario Poliquístico/genética , Estimulación Química , Factores de Tiempo
15.
Endocrinology ; 160(6): 1377-1393, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30951142

RESUMEN

Ovarian theca androgen production is regulated by the pituitary LH and intrafollicular factors. Enhanced androgen biosynthesis by theca cells contributes to polycystic ovary syndrome (PCOS) in women, but the ovarian consequences of elevated androgens are not completely understood. Our study documents the molecular events that are altered in the theca and stromal cells of mice exposed to high androgen levels, using the nonaromatizable androgen DHT. Changes in ovarian morphology and function were observed not only in follicles, but also in the stromal compartment. Genome-wide microarray analyses revealed marked changes in the ovarian transcriptome of DHT-treated females within 1 week. Particularly striking was the increased expression of vascular cell adhesion molecule 1 (Vcam1) specifically in the NR2F2/COUPTF-II lineage theca cells, not granulosa cells, of growing follicles and throughout the stroma of the androgen-treated mice. This response was mediated by androgen receptors (ARs) present in theca and stromal cells. Human theca-derived cultures expressed both ARs and NR2F2 that were nuclear. VCAM1 mRNA and protein were higher in PCOS-derived theca cells compared with control theca and reduced markedly by the AR antagonist flutamide. In the DHT-treated mice, VCAM1 was transiently induced by equine chorionic gonadotropin, when androgen and estrogen biosynthesis peak in preovulatory follicles, and was potently suppressed by a superovulatory dose of human chorionic gonadotropin. High levels of VCAM1 in the theca and interstitial cells of DHT-treated mice and in adult Leydig cells indicate that there may be novel functions for VCAM1 in reproductive tissues, including the gonads.


Asunto(s)
Dihidrotestosterona , Hiperandrogenismo/metabolismo , Folículo Ovárico/metabolismo , Ovario/metabolismo , Células del Estroma/metabolismo , Células Tecales/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Animales , Factor de Transcripción COUP II/metabolismo , Femenino , Hiperandrogenismo/inducido químicamente , Ratones , Receptores Androgénicos/metabolismo
16.
J Pediatr Endocrinol Metab ; 32(5): 461-470, 2019 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-31005950

RESUMEN

Background Hyperandrogenism with or without polycystic ovarian syndrome is seen in adolescents with type 1 diabetes (T1D), especially those with suboptimal control. Objective To assess the effect of metformin on hyperandrogenism and ovarian function in adolescents with T1D. Methods This prospective study included 28 T1D females showing signs of hyperandrogenism. History taking (detailed diabetes history and menstrual history) and anthropometric measurements (weight, height, body mass index [BMI], waist and hip circumference) were initially performed, and then the patients were assessed for the manifestations of hyperandrogenism (acne, hirsutism as well as pelvic ultrasound [U/S] for ovarian morphology). Biochemical evaluation for ovulation (progesterone assessment during the luteal phase), sex steroids (estradiol, testosterone, dehydroepiandrosterone sulfate [DHEAS] and androstenedione), prolactin, glycemic control (hemoglobin A1c [HbA1c]) and gonadotropin levels (follicle stimulating hormone [FSH] and luteinizing hormone [LH]) was done. Patients were subjected to 500 mg metformin twice daily orally for 1 year, and then the patients were re-evaluated for clinical and biochemical parameters. Results Metformin therapy resulted in a significant reduction in weight (p = 0.001), BMI (p = 0.002), acne (p = 0.008), hirsutism score (0.007), LH (p = 0.008), testosterone (p < 0.001) and androstenedione levels (p = 0.028) in adolescent girls with T1D. Regarding menstrual irregularities, there was a significant reduction in the number of patients with oligomenorrhea (68%) with a p value of <0.001. However, there were no significant reduction in the daily insulin requirements (p = 0.782) or HbA1c (p = 0.068). Nausea and/or abdominal pain were the commonly reported side effects of metformin (64%). Conclusions Metformin as an insulin sensitizing agent improved the BMI and cycle regularity together with clinical and biochemical hyperandrogenism in T1D adolescent girls. However, it did not improve their glycemic control.


Asunto(s)
Biomarcadores/análisis , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Hiperandrogenismo/inducido químicamente , Hipoglucemiantes/efectos adversos , Metformina/efectos adversos , Adolescente , Adulto , Niño , Diabetes Mellitus Tipo 1/patología , Femenino , Estudios de Seguimiento , Humanos , Pronóstico , Estudios Prospectivos , Adulto Joven
17.
Reprod Sci ; 26(5): 657-668, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-29929445

RESUMEN

The effects of androgens on the uterus have been poorly studied and they need to be clarified to understand why androgen excess, such as observed in women with polycystic ovary syndrome (PCOS), is a risk factor for the development of endometrial hyperplasia, cancer, and infertility. Thus, uterine histomorphology in a PCOS experimental model was evaluated. Beginning at weaning, female rats were injected daily with dehydroepiandrosterone (DHEA, 6 mg/100 g body weight) or vehicle (sesame oil) for 20 consecutive days. On postnatal day 41 (PND41), DHEA-treated animals showed high serum testosterone levels. In addition, uterine histological analysis showed a significant increase in luminal epithelial height and glandular density without changes in cell proliferation. The thickness of the subepithelial stroma and myometrium also increased in these animals. The effect of DHEA on uterine thickness was accompanied by a significant reduction in cell density in both tissue compartments (subepithelial stroma and myometrium). Cell proliferation was not altered in the myometrium, whereas a decrease in the proliferative activity was seen at PND41 in the subepithelial stroma of DHEA animals. The analysis of the extracellular space showed that the changes in the thickness of the subepithelial stroma and myometrium were related to an increase in the organization of collagen fibers and water imbibition. The latter was associated with higher aquaporin 3 and 8 expression. This study provides evidence to further the understanding of PCOS-associated hyperandrogenism effects on uterine architecture. This could have implications for the regulation of uterine function and the development of uterine lesions.


Asunto(s)
Hiperandrogenismo/patología , Síndrome del Ovario Poliquístico/patología , Útero/patología , Animales , Deshidroepiandrosterona/administración & dosificación , Modelos Animales de Enfermedad , Receptor alfa de Estrógeno/metabolismo , Femenino , Hiperandrogenismo/inducido químicamente , Hiperandrogenismo/metabolismo , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/metabolismo , Ratas Wistar , Receptores Androgénicos/metabolismo , Útero/metabolismo
18.
Pharmacol Rep ; 71(1): 96-104, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30508725

RESUMEN

BACKGROUND: Peroxisome proliferator-activated receptor gamma (PPARG) is a nuclear factor that may act on the early development of ovarian follicles and on follicular steroidogenesis. However, the exact mechanism of PPARG action remains unknown. We have previously found that androgen excess alters early ovarian function and the PPARG system. The aim of the present study was to evaluate whether PPARG activation (using the synthetic ligand pioglitazone (PGZ)) ameliorates the alterations in early ovarian function induced by androgen excess. METHODS: Female prepubertal rats were treated with equine chorionic gonadotropin (eCG) to induce folliculogenesis, together with dehydroepiandrosterone (DHEA) to induce hyperandrogenism and/or PGZ to evaluate PPARG activation. We assessed i) very early ovarian folliculogenesis, ii) PPARG activation, iii) ovarian steroidogenic enzymes, iv) the estradiol/testosterone ratio, v) the ovarian inflammatory status and vi) oxidative stress. RESULTS: PGZ prevented the inactivation of ovarian PPARG induced by androgen excess by increasing PPARG itself and the gene expression of PPARG-coactivator 1 alpha (PGC1A), and by decreasing the gene expression of nuclear co-repressor (NCOR). PGZ also prevented the altered ovarian steroidogenesis, pro-inflammatory status and oxidative stress induced by androgen excess. CONCLUSIONS: Our findings suggest that PPARG activation plays important roles in modulating early ovarian function, and highlight the importance of understanding the role(s) of PPARG activation in the ovary, and the possible involvement in the treatment of ovarian pathologies, and/or the impact in regulating/improving fertility.


Asunto(s)
Deshidroepiandrosterona , Hiperandrogenismo/prevención & control , Folículo Ovárico/efectos de los fármacos , PPAR gamma/agonistas , Pioglitazona/farmacología , Animales , Proteínas Co-Represoras/genética , Proteínas Co-Represoras/metabolismo , Modelos Animales de Enfermedad , Estradiol/metabolismo , Femenino , Hiperandrogenismo/inducido químicamente , Hiperandrogenismo/metabolismo , Hiperandrogenismo/fisiopatología , Mediadores de Inflamación/metabolismo , Ligandos , Folículo Ovárico/metabolismo , Folículo Ovárico/patología , Folículo Ovárico/fisiopatología , Estrés Oxidativo/efectos de los fármacos , PPAR gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Testosterona/metabolismo
19.
Biosci Rep ; 39(1)2019 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-30573529

RESUMEN

Shaoyao-Gancao Decoction (SGD) has been widely used for the treatment of gynopathy. The present study aimed to evaluate the therapeutic effect and potential mechanism of SGD on hyperandrogenism in polycystic ovary syndrome (PCOS) rats. In the present work, SGD was orally administrated to the PCOS rats at the dose of 12.5, 25, and 50 g/kg/d for 14 consecutive days. UPLC-MS/MS was performed to identify the main chemical components of SGD. Body weight, ovarian weight, cystic dilating follicles, and serum levels of steroid hormones were tested to evaluate the therapeutic effect of SGD. In order to further clarify the underlying mechanism, we also measured mRNA and the protein levels of NF-κB, NF-κB p65, P-NF-κB p65, and IκB by RT-qPCR and Western blotting techniques. Our results showed that SGD treatment significantly alleviated hyperandrogenism in PCOS rats as evidenced by reduced serum levels of T and increased E2 and FSH levels. In addition, SGD effectively reduced the phosphorylation of NF-κB p65 and increased the expression of IκB. Results of the present study demonstrated that SGD could ameliorate hyperandrogenism in PCOS rats, and the potential mechanism may relate to the NF-κB pathway.


Asunto(s)
Medicamentos Herbarios Chinos/administración & dosificación , Hiperandrogenismo/tratamiento farmacológico , FN-kappa B/genética , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hiperandrogenismo/inducido químicamente , Hiperandrogenismo/genética , Hiperandrogenismo/patología , Quinasa I-kappa B/genética , Letrozol/toxicidad , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/patología , Ratas , Factor de Transcripción ReIA/genética
20.
Endocrinology ; 159(12): 4056-4064, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30376052

RESUMEN

The main clinical feature associated with hyperandrogenism in polycystic ovary syndrome (PCOS) in humans is hirsutism, where hair increases its length, pigmentation, and particularly its diameter. Currently, it is not known whether PCOS animal models also exhibit changes in the hair. Therefore, the aim of this study was to explore the wool characteristics in sheep prenatally androgenized (PA) with testosterone propionate. After 4 and 13 months of life, wool was collected from the top of the shoulder of both females and males (both androgenized and controls). The offspring sheep were followed for up to 19 months of life to evaluate testosterone and androstenedione serum levels by ultra-high-performance liquid chromatography-tandem mass spectrometry, determine insulin and glucose response to intravenous glucose tolerance test, and address estrus cyclicity during the second breeding season. PA male animals showed a reduction in wool fiber diameter at 4 months of age compared with controls (P = 0.02) but not at 13 months, whereas PA females showed increased hair diameter at 13 months (P = 0.002), with no difference at 4 months. No substantial changes in other hair parameters (length, color, and medullation) were identified. In addition, increased levels of serum testosterone were observed in PA female sheep compared with controls at 12 months (P = 0.03). Our results indicate for the first time, to our knowledge, that changes in wool fiber diameter observed in PA ewes replicate, at the translational level, the increase in hair diameter in hirsute women with PCOS.


Asunto(s)
Andrógenos , Modelos Animales de Enfermedad , Hirsutismo , Síndrome del Ovario Poliquístico , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Ovinos , Virilismo/inducido químicamente , Animales , Femenino , Prueba de Tolerancia a la Glucosa , Hirsutismo/sangre , Hirsutismo/inducido químicamente , Hirsutismo/complicaciones , Hirsutismo/patología , Hiperandrogenismo/sangre , Hiperandrogenismo/inducido químicamente , Hiperandrogenismo/patología , Masculino , Síndrome del Ovario Poliquístico/sangre , Síndrome del Ovario Poliquístico/complicaciones , Síndrome del Ovario Poliquístico/patología , Embarazo , Efectos Tardíos de la Exposición Prenatal/sangre , Efectos Tardíos de la Exposición Prenatal/patología , Propionato de Testosterona , Virilismo/sangre , Virilismo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...