Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Pharmacol Rep ; 71(1): 96-104, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30508725

ABSTRACT

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.


Subject(s)
Dehydroepiandrosterone , Hyperandrogenism/prevention & control , Ovarian Follicle/drug effects , PPAR gamma/agonists , Pioglitazone/pharmacology , Animals , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Disease Models, Animal , Estradiol/metabolism , Female , Hyperandrogenism/chemically induced , Hyperandrogenism/metabolism , Hyperandrogenism/physiopathology , Inflammation Mediators/metabolism , Ligands , Ovarian Follicle/metabolism , Ovarian Follicle/pathology , Ovarian Follicle/physiopathology , Oxidative Stress/drug effects , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Testosterone/metabolism
2.
Gynecol Endocrinol ; 33(3): 185-187, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28102088

ABSTRACT

Most frequent causes of androgenic manifestation are Cushing's syndrome, PCO, benign and malignant androgen-secreting non adrenal tumors and iatrogenic hirsutism. Hyperplasia or neoplasms of ectopic adrenocortical gland are rare. We report a case of a 63-year old female with hirsutism and alopecia. Laboratory data highlighted increased levels of androgens. Diagnostic imaging revealed normal morphology of adrenocortical gland and ovaries. In view of the clinical picture and suspected diagnosis of extra-adrenal cause, she underwent bilateral salpingo-oophorectomy. Histologic examination showed an ectopic adrenal gland with adenoma in the ovarian and peri-ovarian tissue. At six months of follow up, the patients has no sign of hyperandrogenism. In case of hyperandrogenism in postmenopausal women and in the absence of the adrenocortical gland abnormality, ovarian origin should be considered in the differential diagnosis.


Subject(s)
Adenoma/diagnosis , Hyperandrogenism/etiology , Ovarian Neoplasms/diagnosis , Adenoma/pathology , Adenoma/physiopathology , Adenoma/surgery , Alopecia/etiology , Alopecia/prevention & control , Diagnosis, Differential , Female , Hirsutism/etiology , Hirsutism/prevention & control , Humans , Hyperandrogenism/physiopathology , Hyperandrogenism/prevention & control , Middle Aged , Ovarian Neoplasms/pathology , Ovarian Neoplasms/physiopathology , Ovarian Neoplasms/surgery , Ovariectomy , Rome , Salpingectomy , Treatment Outcome
3.
Endocrinology ; 157(1): 382-94, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26465200

ABSTRACT

Hyperandrogenism is the central feature of polycystic ovary syndrome (PCOS). Due to the intricate relationship between hyperandrogenism and insulin resistance in PCOS, 50%-70% of these patients also present with hyperinsulinemia. Metformin, an insulin sensitizer, has been used to reduce insulin resistance and improve fertility in women with PCOS. In previous work, we have noted that a dietary medium-chain fatty acid, decanoic acid (DA), improves glucose tolerance and lipid profile in a mouse model of diabetes. Here, we report for the first time that DA, like metformin, inhibits androgen biosynthesis in NCI-H295R steroidogenic cells by regulating the enzyme 3ß-hydroxysteroid dehydrogenase/Δ5-Δ4-isomerase type 2 (HSD3B2). The inhibitory effect on HSD3B2 and androgen production required cAMP stimulation, suggesting a mechanistic action via the cAMP-stimulated pathway. Specifically, both DA and metformin reduced cAMP-enhanced recruitment of the orphan nuclear receptor Nur77 to the HSD3B2 promoter, coupled with decreased transcription and protein expression of HSD3B2. In a letrozole-induced PCOS rat model, treatment with DA or metformin reduced serum-free testosterone, lowered fasting insulin, and restored estrous cyclicity. In addition, DA treatment lowered serum total testosterone and decreased HSD3B2 protein expression in the adrenals and ovaries. We conclude that DA inhibits androgen biosynthesis via mechanisms resulting in the suppression of HSD3B2 expression, an effect consistently observed both in vitro and in vivo. The efficacy of DA in reversing the endocrine and metabolic abnormalities of the letrozole-induced PCOS rat model are promising, raising the possibility that diets including DA could be beneficial for the management of both hyperandrogenism and insulin resistance in PCOS.


Subject(s)
Decanoic Acids/therapeutic use , Dietary Fats/therapeutic use , Disease Models, Animal , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Polycystic Ovary Syndrome/diet therapy , Progesterone Reductase/antagonists & inhibitors , Promoter Regions, Genetic , Adrenal Cortex/enzymology , Adrenal Cortex/metabolism , Adrenal Glands/enzymology , Adrenal Glands/metabolism , Androgens/analysis , Androgens/chemistry , Androgens/metabolism , Animals , Cell Line , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/metabolism , Decanoic Acids/metabolism , Dietary Fats/metabolism , Enzyme Repression , Female , Humans , Hyperandrogenism/etiology , Hyperandrogenism/prevention & control , Insulin Resistance , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Ovary/enzymology , Ovary/metabolism , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Polycystic Ovary Syndrome/physiopathology , Progesterone Reductase/genetics , Progesterone Reductase/metabolism , Random Allocation , Rats, Wistar
4.
Endocrinology ; 156(11): 4071-80, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26322372

ABSTRACT

This study was designed to differentiate the contributions of hyperandrogenism, insulin resistance (IR), and body weight to the development of endothelial dysfunction in polycystic ovary syndrome and determine the effectiveness of insulin sensitization and antiandrogenic therapy after the establishment of vascular and metabolic dysfunction using a rat model of polycystic ovary syndrome. We hypothesized that the observed endothelial dysfunction was a direct steroidal effect, as opposed to changes in insulin sensitivity or body weight. Prepubertal female rats were randomized to the implantation of a pellet containing DHT or sham procedure. In phase 1, DHT-exposed animals were randomized to pair feeding to prevent weight gain or metformin, an insulin-sensitizing agent, from 5 to 14 weeks. In phase 2, DHT-exposed animals were randomized to treatment with metformin or flutamide, a nonsteroidal androgen receptor blocker from 12 to 16 weeks. Endothelial function was assessed by the vasodilatory response of preconstricted arteries to acetylcholine. Serum steroid levels were analyzed in phase 1 animals. Fasting blood glucose and plasma insulin were analyzed and homeostasis model assessment index calculated in all animals. Our data confirm the presence of endothelial dysfunction as well as increased body weight, hypertension, hyperinsulinemia, and greater IR among DHT-treated animals. Even when normal weight was maintained through pair feeding, endothelial dysfunction, hyperinsulinemia, and IR still developed. Furthermore, despite weight gain, treatment with metformin and flutamide improved insulin sensitivity and blood pressure and restored normal endothelial function. Therefore, the observed endothelial dysfunction is most likely a direct result of hyperandrogenism-induced reductions in insulin sensitivity, as opposed to weight gain.


Subject(s)
Body Weight/physiology , Endothelium, Vascular/physiopathology , Hyperandrogenism/physiopathology , Insulin Resistance/physiology , Polycystic Ovary Syndrome/physiopathology , Androgen Antagonists/pharmacology , Androgens/pharmacology , Animals , Arteries/drug effects , Arteries/physiopathology , Blood Pressure/drug effects , Blood Pressure/physiology , Body Weight/drug effects , Dihydrotestosterone/pharmacology , Endothelium, Vascular/drug effects , Female , Flutamide/pharmacology , Hyperandrogenism/prevention & control , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Random Allocation , Rats, Wistar , Time Factors , Vasodilation/drug effects , Vasodilation/physiology
5.
Endocrinology ; 156(4): 1441-52, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25643156

ABSTRACT

Polycystic ovary syndrome (PCOS) is associated with reproductive, endocrine, and metabolic abnormalities. Because hyperandrogenism is the most consistent PCOS feature, we used wild-type (WT) and androgen receptor (AR) knockout (ARKO) mice, together with a mouse model of PCOS, to investigate the contribution of genomic AR-mediated actions in the development of PCOS traits. PCOS features were induced by prenatal exposure to dihydrotestosterone (250 µg) or oil vehicle (control) on days 16-18 of gestation in WT, heterozygote, and homozygote ARKO mice. DHT treatment of WT mice induced ovarian cysts (100% vs 0%), disrupted estrous cycles (42% vs 100% cycling), and led to fewer corpora lutea (5.0±0.4 vs 9.8±1.8). However, diestrus serum LH and FSH, and estradiol-induced-negative feedback as well as hypothalamic expression of kisspeptin, neurokinin B, and dynorphin, were unaffected by DHT treatment in WT mice. DHT-treated WT mice exhibited a more than 48% increase in adipocyte area but without changes in body fat. In contrast, heterozygous and homozygous ARKO mice exposed to DHT maintained comparable ovarian (histo)morphology, estrous cycling, and corpora lutea numbers, without any increase in adipocyte size. These findings provide strong evidence that genomic AR signaling is an important mediator in the development of these PCOS traits with a dose dependency that allows even AR haplosufficiency to prevent induction by prenatal androgenization of PCOS features in adult life.


Subject(s)
Hyperandrogenism/prevention & control , Polycystic Ovary Syndrome/prevention & control , Prenatal Exposure Delayed Effects/prevention & control , Receptors, Androgen/metabolism , Adipose Tissue/metabolism , Androgens , Animals , Disease Models, Animal , Estrous Cycle , Female , Hyperandrogenism/chemically induced , Hyperandrogenism/metabolism , Mice , Mice, Knockout , Ovary/metabolism , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Receptors, Androgen/genetics
6.
Gynecol Endocrinol ; 31(4): 332-6, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25558892

ABSTRACT

OBJECTIVES: To evaluate different oral contraceptive pill (OCP) pretreatment associated differential in-vitro fertilization (IVF)/intracytoplasmic sperm injection (ICSI) outcomes of polycystic ovary syndrome (PCOS) patients and explore enhanced hormonal balance induced by the pretreatment. METHODS: This retrospective study included 500 PCOS women and 565 normal ovulating counterparts undergoing IVF/ICSI. The PCOS patients were divided into three groups based on the OCP pretreatment regimens: non-OCP (without OCP pretreatment), unsuccessive OCP (the period of successive pretreatment ≤2 months) and successive OCP (the period of successive pretreatment ≥3 months) groups. Comprehensive hormonal and ultra-sonographic assessments were performed before/after IVF pretreatment. Confounding factors affecting pregnancy outcomes were analyzed with logistic regression. RESULTS: PCOS patients with significant endocrine disorders had reduced implantation and pregnancy rates and increased miscarriage rate. Successive, not unsuccessive OCP pretreatment, significantly improved the implantation and pregnancy rates, and reduced the incidence of monotocous small-for-gestational age infants, which was accompanied by remarkably decreased hyperandrogenism and antral follicles. CONCLUSION: PCOS is an independent risk factor for poor IVF outcome. Successive, not unsuccessive, OCP cyclical pretreatment could improve pregnancy outcome of PCOS patients, associated with reduction of hyperandrogenism and antral follicle excess.


Subject(s)
Contraceptives, Oral, Sequential/therapeutic use , Fertilization in Vitro , Hyperandrogenism/prevention & control , Infertility, Female/therapy , Ovarian Hyperstimulation Syndrome/prevention & control , Ovulation Induction/adverse effects , Polycystic Ovary Syndrome/drug therapy , Adult , China/epidemiology , Female , Fetal Growth Retardation/etiology , Fetal Growth Retardation/prevention & control , Hospitals, University , Humans , Hyperandrogenism/etiology , Infertility, Female/etiology , Outpatient Clinics, Hospital , Ovarian Hyperstimulation Syndrome/etiology , Polycystic Ovary Syndrome/physiopathology , Pregnancy , Pregnancy Rate , Retrospective Studies , Sperm Injections, Intracytoplasmic , Statistics as Topic , Young Adult
7.
Reprod Biol Endocrinol ; 12: 98, 2014 Oct 11.
Article in English | MEDLINE | ID: mdl-25304843

ABSTRACT

BACKGROUND: The use of insulin-sensitizing drugs has been shown to improve both the reproductive and the metabolic aspects of PCOS. However, the mechanisms by which metformin exerts its effects in PCOS are still not completely understood. There is growing evidence of a direct effect of metformin on ovarian steroidogenesis, independent of its effects on insulin sensitivity. METHODS: We evaluated the short-term effects of metformin compared to placebo on basal and LH- stimulated androgen secretion as well as on hormonal and metabolic parameters in 19 women with PCOS during a four-day randomized, double-blinded placebo-controlled clinical trial. In a three month follow-up evaluation, we investigated the longer-term therapeutic effects of metformin on ovulation, metabolic and endocrine parameters. RESULTS: Compared to placebo, 2 days of metformin was associated with a borderline significant reduction in the free androgen index (FAI) (p = 0.05) and with a reduction in the serum concentration of LH-stimulated testosterone (T) (p = 0.03). Following three months of use, a decline in serum T was observed, independent of changes in weight, metabolic parameters, or insulin sensitivity. CONCLUSIONS: In women with PCOS, Metformin induces a prompt decrease in LH-stimulated T secretion after only several days of use. This action precedes the medication's effects on insulin sensitivity or weight loss.


Subject(s)
Adrenal Cortex/drug effects , Hypoglycemic Agents/therapeutic use , Insulin Resistance , Luteinizing Hormone/antagonists & inhibitors , Metformin/therapeutic use , Polycystic Ovary Syndrome/drug therapy , Testosterone/metabolism , Adrenal Cortex/metabolism , Adrenal Cortex/physiopathology , Adult , Body Mass Index , Double-Blind Method , Female , Follow-Up Studies , Humans , Hyperandrogenism/etiology , Hyperandrogenism/prevention & control , Hyperinsulinism/etiology , Hyperinsulinism/prevention & control , Hypoglycemic Agents/adverse effects , Luteinizing Hormone/genetics , Metformin/adverse effects , Overweight/complications , Ovulation/drug effects , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Testosterone/blood , Weight Gain/drug effects , Young Adult
8.
Gynecol Endocrinol ; 30(3): 205-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24351072

ABSTRACT

Myo-inositol and D-chiro-inositol are capable of improving the ovarian function and metabolism of polycystic ovary syndrome (PCOS) patients. The aim of this work is to compare the effects of myo-inositol and D-chiro-inositol in PCOS. We enrolled 50 patients, with homogeneous bio-physical features, affected by PCOS and menstrual irregularities, and we randomly divided them into two groups: 25 were treated with 4 g of myo-inositol/die plus 400 mcg of folic acid/die orally for six months, 25 with 1 g of D-chiro-inositol/die plus 400 mcg of folic acid/die orally for six months. We analyzed in both groups pre-treatment and post-treatment BMI, systolic and diastolic blood pressure, Ferriman-Gallwey score, Cremoncini score, serum LH, LH/FSH ratio, total and free testosterone, dehydroepiandrosterone sulfate (DHEA-S), Δ-4-androstenedione, SHBG, prolactin, glucose/immunoreactive insulin (IRI) ratio, homeostatic model assessment (HOMA) index, and the resumption of regular menstrual cycles. Both the isoforms of inositol were effective in improving ovarian function and metabolism in patients with PCOS, although myo-inositol showed the most marked effect on the metabolic profile, whereas D-chiro-inositol reduced hyperandrogenism better.


Subject(s)
Dietary Supplements , Hyperandrogenism/prevention & control , Inositol/therapeutic use , Insulin Resistance , Menstruation Disturbances/prevention & control , Ovary/physiopathology , Polycystic Ovary Syndrome/diet therapy , Adolescent , Adult , Amenorrhea/etiology , Amenorrhea/prevention & control , Double-Blind Method , Female , Folic Acid/therapeutic use , Follicle Stimulating Hormone/blood , Humans , Hyperandrogenism/etiology , Hypertension/etiology , Hypertension/prevention & control , Inositol/chemistry , Italy , Luteinizing Hormone/blood , Menstruation Disturbances/etiology , Metrorrhagia/etiology , Metrorrhagia/prevention & control , Oligomenorrhea/etiology , Oligomenorrhea/prevention & control , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Stereoisomerism , Young Adult
9.
Endokrynol Pol ; 64(5): 409-14, 2013.
Article in English | MEDLINE | ID: mdl-24186599

ABSTRACT

The latest list of reimbursed medicines includes, as a new addition, metformin for the treatment of polycystic ovary syndrome (PCOS), which is extremely important for practicing physicians. While this paper briefly summarises the current state of knowledge on PCOS, its main aim is to remind the reader about the effectiveness of metformin in women with PCOS in controlling glycaemia, increasing tissue sensitivity to insulin and affecting endothelial function, vascular inflammation, lipid profile and other risk factors of atherosclerosis, which suggests its cardioprotective effects. The paper also discusses the clinical effect of metformin relative to hyperandrogenism, menstrual cycle disorders and ovulation induction. The paper concludes with an algorithm for the diagnosis and management of PCOS.


Subject(s)
Metformin/therapeutic use , Polycystic Ovary Syndrome/drug therapy , Algorithms , Female , Humans , Hyperandrogenism/complications , Hyperandrogenism/prevention & control , Hypoglycemic Agents/therapeutic use , Off-Label Use , Ovulation Induction , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/diagnosis
10.
J Clin Endocrinol Metab ; 98(12): 4798-807, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24152688

ABSTRACT

CONTEXT: Statins have been shown to improve hyperandrogenism in women with polycystic ovary syndrome (PCOS). However, their use has also been associated with impairment of glucose metabolism and an increased risk of type 2 diabetes mellitus. Because women with PCOS are prone to disturbances in glucose metabolism, statin therapy could also have negative effects. OBJECTIVE: Our objective was to explore the effects of atorvastatin therapy on hormonal and metabolic parameters in women with PCOS. DESIGN AND SETTING: We conducted a randomized, double-blind, placebo-controlled 6-month follow-up study conducted at Oulu University Hospital, Finland. PATIENTS: Women with PCOS (Rotterdam criteria) were treated with atorvastatin (20 mg/d, n = 15) or placebo (n = 13) for 6 months. INTERVENTIONS: Fasting serum samples were collected at baseline and at 3 and 6 months. Oral and iv glucose tolerance tests were performed at 0 and 6 months. MAIN OUTCOME MEASURES: Androgen secretion and glucose metabolism were measured. RESULTS: Fasting levels and area under the curve of insulin increased significantly and insulin sensitivity (insulinogenic and Matsuda indexes) decreased during 6 months of atorvastatin therapy. Serum levels of dehydroepiandrosterone sulfate decreased in the atorvastatin group, whereas no change was observed in serum testosterone levels. Levels of C-reactive protein, total and low-density lipoprotein-cholesterol, and triglycerides decreased significantly during statin therapy. CONCLUSIONS: Atorvastatin therapy improves chronic inflammation and lipid profile, but it impairs insulin sensitivity in women with PCOS. Because women with PCOS have an increased risk of developing type 2 diabetes mellitus, the results suggest that statin therapy should be initiated on the basis of generally accepted criteria and individual risk assessment of cardiovascular disease, and not only because of PCOS.


Subject(s)
Heptanoic Acids/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Insulin Resistance , Polycystic Ovary Syndrome/drug therapy , Pyrroles/adverse effects , Adult , Androgen Antagonists/adverse effects , Androgen Antagonists/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Atorvastatin , Body Mass Index , Double-Blind Method , Female , Follow-Up Studies , Glucose Intolerance/chemically induced , Glucose Tolerance Test , Heptanoic Acids/therapeutic use , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hyperandrogenism/etiology , Hyperandrogenism/prevention & control , Hyperinsulinism/chemically induced , Hyperlipidemias/etiology , Hyperlipidemias/prevention & control , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Middle Aged , Overweight/complications , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/immunology , Polycystic Ovary Syndrome/physiopathology , Pyrroles/therapeutic use
11.
Gynecol Endocrinol ; 29(3): 246-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23194004

ABSTRACT

OBJECTIVE: To evaluate the relationship between serum anti-Mullerian hormone levels (AMH) and insulin resistance (IR) before and after meformin treatment and to compare AMH levels of polycystic ovary syndrome (PCOS) women in the early follicular phase. METHODS: Twenty PCOS women with IR, taking metformin 1500 mg/day for 8 weeks, and 16 non-PCOS controls were enrolled in this longitudinal study. Serum levels of AMH, insulin, glucose, testosterone, and quantitative insulin check index (QUICKI), were assessed before and after treatment in PCOS group. RESULTS: AMH levels were higher in untreated PCOS (p < 0.0001), as were luteinizing hormone (LH) (p = 0.0004), testosterone (p = 0.0017) as well as 17-hydroxyprogesterone (p = 0.03). PCOS women show positive correlation between AMH and testosterone (R = 0.83; p < 0.0001) only prior to treatment. Metformin treatment, lead to a significant decrease in serum insulin (p = 0.0132) and testosterone (p = 0.0017) levels. However, no alteration in AMH levels was observed after treatment. CONCLUSION: Despite the improvement of metabolic parameters and the reduction of androgen levels, AMH levels did not change after metformin treatment. Maybe, the dose, and possibly the time of use, of metformin are factors associated with the reduction of AMH levels.


Subject(s)
Anti-Mullerian Hormone/blood , Hyperandrogenism/prevention & control , Hyperinsulinism/prevention & control , Hypoglycemic Agents/therapeutic use , Insulin Resistance , Metformin/therapeutic use , Polycystic Ovary Syndrome/drug therapy , 17-alpha-Hydroxyprogesterone/blood , Adolescent , Adult , Female , Follicular Phase , Humans , Hyperandrogenism/etiology , Hyperinsulinism/etiology , Insulin/blood , Longitudinal Studies , Luteinizing Hormone/blood , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Prospective Studies , Testosterone/blood , Up-Regulation/drug effects , Young Adult
12.
Gynecol Endocrinol ; 29(3): 242-5, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23194076

ABSTRACT

The present study investigates the combined effect of diet, physical exercise and Orlistat for 24 weeks, on serum anti-Müllerian hormone (AMH) levels in overweight and obese women with polycystic ovary syndrome (PCOS) and in overweight and obese controls. Sixty-one (61) selected women with PCOS and 20 overweight and obese controls followed an energy-restricted diet, physical exercise plus Orlistat administration (120 mg, 3 times per day) for 24 weeks. At baseline, week 12 and week 24, serum levels of AMH, FSH, LH, PRL, androgens, sex hormone-binding globulin (SHBG), glucose, and insulin were measured and Free Androgen Index (FAI) and Insulin Resistance (IR) indices were calculated. In PCOS women, serum AMH levels increased after 12 and 24 weeks of treatment. After 12 weeks LH and SHBG were increased, while Testosterone decreased. After 12 and 24 weeks, FAI was decreased and all indices of IR were significantly improved. We concluded that in overweight and obese women with PCOS Orlistat administration, combined with diet and physical exercise, for 24 weeks, resulted in significant weight loss, improvement of hyperandrogenism and insulin sensitivity, and increased serum AMH levels.


Subject(s)
Anti-Mullerian Hormone/blood , Anti-Obesity Agents/therapeutic use , Diet, Reducing , Exercise , Lactones/therapeutic use , Overweight/diet therapy , Polycystic Ovary Syndrome/drug therapy , Adult , Body Mass Index , Combined Modality Therapy , Female , Humans , Hyperandrogenism/etiology , Hyperandrogenism/prevention & control , Insulin Resistance , Luteinizing Hormone/blood , Obesity/complications , Obesity/diet therapy , Obesity/therapy , Orlistat , Overweight/complications , Overweight/therapy , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Testosterone/blood , Up-Regulation/drug effects , Weight Loss/drug effects , Young Adult
13.
Gynecol Endocrinol ; 28(12): 965-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22780885

ABSTRACT

OBJECTIVE: To assess effects of vitamin D and Calcium (Ca) on hormonal and metabolic milieu of polycystic ovary syndrome (PCOS). DESIGN: Single arm open label trial. METHODS: Twelve overweight and vitamin D deficient women with PCOS underwent a 2 hour oral glucose tolerance testing at baseline and following 3-month supplementation with vitamin D (daily dose of 3533 IU, increased to 8533 IU after the first five participants) and 530 mg elemental Ca daily. MAIN OUTCOME MEASURES: Blood pressure (BP), plasma glucose, insulin, total testosterone (T) androstenedione (A), sex hormone binding globulin, lifestyle parameters were assessed at baseline and following 3-month intervention. Insulin resistance (IR) and area under the curve for glucose and insulin were computed; paired analyses were conducted. RESULTS: Improved serum 25OHD (p < 0.001) and reductions in total T (p = 0.036) and A (p = 0.090) levels were noted following 3-month supplementation, compared to baseline. Significant lowering in BP parameters was seen in participants with baseline BP ≥ 120/80 mmHg (n = 8) and in those with baseline serum 25OHD ≤20 ng/ml (n = 9). Parameters of glucose homeostasis and IR remained unchanged (p > 0.05). CONCLUSIONS: Androgen and BP profiles improved followed three month intervention, suggesting therapeutic implications of vitamin D and Ca in overweight and vitamin D deficient women with PCOS.


Subject(s)
Calcium, Dietary/therapeutic use , Cholecalciferol/therapeutic use , Dietary Supplements , Ergocalciferols/therapeutic use , Overweight/complications , Polycystic Ovary Syndrome/diet therapy , Vitamin D Deficiency/diet therapy , 25-Hydroxyvitamin D 2/blood , Adult , Body Mass Index , Calcifediol/blood , Calcium, Dietary/adverse effects , Cholecalciferol/administration & dosage , Cholecalciferol/adverse effects , Cholecalciferol/metabolism , Cohort Studies , Dietary Supplements/adverse effects , Ergocalciferols/administration & dosage , Ergocalciferols/adverse effects , Ergocalciferols/metabolism , Female , Humans , Hyperandrogenism/etiology , Hyperandrogenism/prevention & control , Hypertension/etiology , Hypertension/prevention & control , Patient Dropouts , Pilot Projects , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Testosterone Congeners/blood , Vitamin D Deficiency/complications , Young Adult
14.
Gynecol Endocrinol ; 28(8): 606-10, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22329763

ABSTRACT

We studied (1) the effects of oral contraceptive pills (OCPs) on hirsutism, hormonal and metabolic variables in 49 polycystic ovary syndrome patients without metabolic comorbidities and (2) the effect of 17-hydroxysteroid dehydrogenase type 5 gene polymorphism (-71A/G HSD17B5 SNP) on the response to OCP treatment. Mean age was 21.9 ± 6.5 years. Patients received monophasic OCP (20 µg ethinyl estradiol plus 75 µg gestodene), 21/28 days per cycle, during 6 months; 32 patients with severe hirsutism also received spironolactone 100 mg. The frequencies of HSD17B5 genotypes were: AA = 0.49 (55.1%), AG = 0.42 (30.6%) and GG = 0.09 (14.3%). After 6 months, body mass index and waist circumference remained unchanged regardless of the presence of allele G. A slight reduction (p < 0.05) was noted in systolic blood pressure (p < 0.05) and luteinizing hormone levels, whereas a slight increase (p < 0.05) was noted in lipids. Total testosterone and hirsutism score declined, while sex hormone binding globulin increased after OCP treatment (p < 0.05). None of these changes were associated with genotype. Insulin and homeostasis model assessment remained unchanged after treatment and did not vary according to the presence of allele G. OCP seems to ameliorate androgenic symptoms without compromising metabolic parameters. The -71A/G SNP of HSD17B5 gene did not contribute to the improvements observed.


Subject(s)
Contraceptives, Oral, Combined/therapeutic use , Ethinyl Estradiol/therapeutic use , Hirsutism/prevention & control , Norpregnenes/therapeutic use , Polycystic Ovary Syndrome/drug therapy , Sex Hormone-Binding Globulin/analysis , Testosterone/blood , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Adolescent , Adult , Aldo-Keto Reductase Family 1 Member C3 , Brazil , Contraceptives, Oral, Combined/adverse effects , Drug Therapy, Combination , Ethinyl Estradiol/adverse effects , Female , Genetic Association Studies , Hirsutism/drug therapy , Hirsutism/etiology , Hirsutism/physiopathology , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Hydroxyprostaglandin Dehydrogenases/metabolism , Hyperandrogenism/etiology , Hyperandrogenism/prevention & control , Mineralocorticoid Receptor Antagonists/therapeutic use , Norpregnenes/adverse effects , Pilot Projects , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Polymorphism, Single Nucleotide , Severity of Illness Index , Spironolactone/therapeutic use , Young Adult
15.
Eur J Obstet Gynecol Reprod Biol ; 159(1): 127-31, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21831508

ABSTRACT

OBJECTIVE: To evaluate the clinical, endocrine and metabolic effects of metformin and N-acetyl-cysteine (NAC) in patients with polycystic ovary syndrome (PCOS). STUDY DESIGN: In this prospective trial, 100 women with PCOS were randomly divided to receive metformin (500 mg p.o. three times daily) or NAC (600 mg p.o. three times daily) for 24 weeks. Hyperandrogenism, lipid profiles, hirsutism scores, menstrual irregularity, insulin sensitivity and tumour necrosis factor-α (TNF-α) levels were measured at baseline and after the treatment period. RESULTS: Both treatments resulted in a significant decrease in body mass index, hirsutism score, fasting insulin, HOMA index, free testosterone and menstrual irregularity compared with baseline values, and both treatments had equal efficacy. NAC led to a significant decrease in both total cholesterol and low-density lipoprotein levels, whereas metformin only led to a decrease in total cholesterol level. Although TNF-α levels increased following treatment for both groups, the difference from baseline was not significant. CONCLUSIONS: Metformin and NAC appear to have comparable effects on hyperandrogenism, hyperinsulinaemia and menstrual irregularity in women with PCOS. The effects of metformin and NAC on insulin sensitivity are not associated with TNF-α.


Subject(s)
Acetylcysteine/therapeutic use , Antioxidants/therapeutic use , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Polycystic Ovary Syndrome/drug therapy , Acetylcysteine/adverse effects , Adolescent , Adult , Anticholesteremic Agents/adverse effects , Anticholesteremic Agents/therapeutic use , Antioxidants/adverse effects , Body Mass Index , Female , Hirsutism/etiology , Hirsutism/prevention & control , Humans , Hyperandrogenism/etiology , Hyperandrogenism/prevention & control , Hypercholesterolemia/etiology , Hypercholesterolemia/prevention & control , Hyperinsulinism/etiology , Hyperinsulinism/prevention & control , Hypoglycemic Agents/adverse effects , Insulin Resistance , Menstruation Disturbances/etiology , Menstruation Disturbances/prevention & control , Metformin/adverse effects , Patient Dropouts , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Severity of Illness Index , Tumor Necrosis Factor-alpha/blood , Young Adult
16.
J Clin Endocrinol Metab ; 96(8): E1262-7, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21632811

ABSTRACT

CONTEXT: Girls with a combined history of low(-normal) birth weight (LBW) and precocious pubarche (PP) are at high risk to develop polycystic ovary syndrome (PCOS). OBJECTIVE: The objective of the study was to compare the capacity of early vs. late metformin treatment to prevent adolescent PCOS. DESIGN: This was a randomized, open-label study over 7 yr. SETTING: The study was conducted at a university hospital. PATIENTS: Thirty-eight LBW-PP girls were followed up from the mean age 8 until age 15 yr. INTERVENTION: Early metformin (study yr 1-4; age 8-12 yr) vs. late metformin (yr 6; age 13-14 yr). MAIN OUTCOME MEASURES: Measures included height; weight; hirsutism score; menstrual cycle; endocrine-metabolic screening (fasting; follicular phase); C-reactive protein; body composition (absorptiometry); abdominal fat partitioning (magnetic resonance imaging); ovarian morphology (ultrasound); PCOS (National Institutes of Health and Androgen Excess Society definitions) after yr 7 (all girls thus untreated for at least 1 yr). RESULTS: None of the girls dropped out of the study. At age 15 yr, early-metformin girls were taller (4 cm), were in a less proinflammatory state, and had less central fat due to reductions in visceral and hepatic fat. Hirsutism, androgen excess, oligomenorrhea, and PCOS were between 2- and 8-fold more prevalent in late- than early-treated girls. Abdominal adiposity was the first variable to diverge (at age 8-10 yr) between girls without vs. with PCOS at age 15 yr. CONCLUSIONS: In LBW-PP girls, early metformin therapy was found to prevent or delay the development of hirsutism, androgen excess, oligomenorrhea, and PCOS more effectively than late metformin. The time window of late childhood and early puberty may be more critical for the development, and thus for the prevention, of adolescent PCOS than the first years beyond menarche.


Subject(s)
Hirsutism/drug therapy , Hyperandrogenism/drug therapy , Metformin/administration & dosage , Oligomenorrhea/drug therapy , Polycystic Ovary Syndrome/prevention & control , Puberty, Precocious/drug therapy , Adolescent , Child , Female , Follow-Up Studies , Hirsutism/epidemiology , Hirsutism/prevention & control , Humans , Hyperandrogenism/epidemiology , Hyperandrogenism/prevention & control , Hypoglycemic Agents/administration & dosage , Menarche , Oligomenorrhea/epidemiology , Oligomenorrhea/prevention & control , Polycystic Ovary Syndrome/epidemiology , Prevalence , Puberty, Precocious/epidemiology , Risk Factors , Treatment Outcome
18.
Clin Endocrinol (Oxf) ; 73(3): 286-90, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20455892

ABSTRACT

Management of severe insulin resistance (IR) is a major clinical challenge in many patients with obesity or lipodystrophy, and also in rarer patients with proven or suspected genetic defects in the insulin receptor or downstream signalling. The latter group can present at any time between birth and early adult life, with a variable clinical course broadly correlated with the severity of IR. Primary insulin signalling defects are usually associated with poor weight gain rather than obesity. Initially, extreme hyperinsulinaemia produces ovarian enlargement and hyperandrogenism in women, and often fasting or postprandial hypoglycaemia. However, any hypoglycaemia gradually evolves into insulin-resistant hyperglycaemia when beta cell function declines. Optimal management of these complex disorders depends on early diagnosis and appropriate targeting of both high and low glucose levels. In newborns, continuous nasogastric feeding may reduce harmful glycaemic fluctuations, and in older patients, acarbose may mitigate postprandial hypoglycaemia. Insulin sensitization, initially with metformin but later with trials of additional agents such as thiazolidinediones, is the mainstay of early therapy, but insulin replacement, eventually with very high doses, is required once diabetes has supervened. Preliminary data suggest that rhIGF-1 can improve survival in infants with the most severe insulin receptor defects and also improve beta cell function in older patients with milder receptoropathies. The utility of newer therapies such as glucagon-like peptide-1 agonists and dipeptidyl peptidase-IV inhibitors remains untested in this condition. Thus, management of these patients remains largely empirical, and there is a pressing need to collate data centrally to optimize treatment algorithms.


Subject(s)
Hypoglycemia/prevention & control , Insulin Resistance , Insulin-Like Growth Factor I/therapeutic use , Adolescent , Adult , Blood Glucose/metabolism , Diabetes Complications/blood , Female , Glycated Hemoglobin/metabolism , Humans , Hyperandrogenism/complications , Hyperandrogenism/prevention & control , Hyperinsulinism/complications , Hyperinsulinism/prevention & control , Hypoglycemia/complications , Hypoglycemia/diagnosis , Infant, Newborn , Insulin/blood , Insulin-Like Growth Factor I/genetics , Mutation , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/complications , Receptor, Insulin/genetics , Recombinant Proteins/therapeutic use , Syndrome , Treatment Outcome
19.
BJOG ; 117(2): 150-5, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20002395

ABSTRACT

Polycystic ovary syndrome (PCOS) is the most common endocrinopathy among reproductive-aged women, and it typically presents during adolescence. The objective of this review is to describe the clinical manifestations of PCOS in adolescent girls and the underlying basis for the altered reproductive physiology. Recognising adolescents at risk for PCOS and taking the appropriate steps to reduce circulating androgen levels is critical in reducing the clinical symptomatology of this disorder, and the development of adulthood infertility, diabetes, and metabolic syndrome in patients with PCOS.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Hyperandrogenism/metabolism , Menstruation Disturbances/physiopathology , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Puberty/physiology , Adolescent , Adult , Androgens/metabolism , Anovulation/metabolism , Anti-Mullerian Hormone/metabolism , Child , Dehydroepiandrosterone/metabolism , Female , Follicle Stimulating Hormone/metabolism , Humans , Hyperandrogenism/prevention & control , Hyperinsulinism/metabolism , Luteinizing Hormone/drug effects , Luteinizing Hormone/metabolism , Menstrual Cycle/metabolism , Menstruation Disturbances/metabolism , Obesity/metabolism , Ovary/diagnostic imaging , Ovary/pathology , Ovary/physiopathology , Polycystic Ovary Syndrome/diagnostic imaging , Progesterone/pharmacology , Progesterone/physiology , Risk Factors , Sex Hormone-Binding Globulin/metabolism , Testosterone/blood , Ultrasonography
20.
Endocr Pract ; 13(6): 601-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17954415

ABSTRACT

OBJECTIVE: To ascertain an association between the a priori known insulin resistance caused by antipsychotic agents and divalproex and adrenal hyperandrogenism and to determine whether the associated hyperandrogenism is reversible with insulin sensitizers. METHODS: We studied 26 consecutive psychiatric inpatients (22 women and 4 men) receiving the aforementioned medications, who were referred to us for a consultation. They ranged in age from 19 to 79 years and had a mean body mass index (SEM) of 32.35 +/- 1.26 kg/m2. Between 8 AM and 9 AM, blood samples were collected for 17-hydroxyprogesterone, 17-hydroxypregnenolone, androstenedione, dehydroepiandrosterone (DHEA), DHEA sulfate, 11-deoxycortisol, luteinizing hormone and follicle-stimulating hormone (in reproductive age women), estrone, estradiol (in reproductive age women), free testosterone (in women), deoxycorticosterone, and sex hormone-binding globulin (SHBG), which were measured by radioimmunoassay, after chromatography if necessary. For intact, premenopausal women, measurement of the abnormal steroid metabolite or SHBG level was repeated during prednisone therapy (5 mg at bedtime) to document the likely adrenal origin of the abnormality. Men, women who had undergone bilateral oophorectomy, and postmenopausal women had hyperandrogenism of adrenal origin by default. Clinical features included central obesity, acanthosis, hirsutism, alopecia, type 2 diabetes mellitus, and oligomenorrhea. RESULTS: We found reversed estrone/estradiol ratios in 4 patients, decreased SHBG in 4, increased 17-hydroxy-pregnenolone in 8, increased 17-hydroxyprogesterone in 2, increased deoxycorticosterone in 2, increased DHEA sulfate in 1, increased 11-deoxycortisol in 4, increased androstenedione in 1, and reversed ratios of luteinizin hormone to follicle-stimulating hormone in 2. The bio-chemical abnormalities were corrected in 8 of 8 patients receiving metformin and in 2 of 2 patients receiving rosiglitazone. CONCLUSION: Insulin resistance caused by antipsychotic agents and divalproex is associated with adrenal hyperandrogenism. Metformin and rosiglitazone correct the biochemical abnormalities detected without compromising their psychotropic effect. Adrenal androgen synthesis may be increased by hyperinsulinemia-induced hyperphosphorylation of P450c17 alpha, resulting in an increase in its 17,20-lyase activity, which magnifies the effects of any distal steroidogenic enzyme defects. Treatment with metformin or rosiglitazone prevents excess adrenal androgen synthesis.


Subject(s)
Antipsychotic Agents/adverse effects , Hyperandrogenism/prevention & control , Metformin/therapeutic use , Thiazolidinediones/therapeutic use , Valproic Acid/adverse effects , 17-alpha-Hydroxyprogesterone/blood , Adolescent , Adrenal Glands/drug effects , Adrenal Glands/metabolism , Adrenal Glands/pathology , Adult , Aged , Antipsychotic Agents/therapeutic use , Cortodoxone/blood , Dehydroepiandrosterone/blood , Endocrine Disruptors/adverse effects , Estradiol/blood , Estrone/blood , Female , Follicle Stimulating Hormone/blood , Humans , Hyperandrogenism/blood , Hyperandrogenism/chemically induced , Hypoglycemic Agents/therapeutic use , Insulin Resistance , Luteinizing Hormone/blood , Male , Middle Aged , Radioimmunoassay , Rosiglitazone , Testosterone/blood , Valproic Acid/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...