Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 332
Filter
1.
Curr Cardiol Rep ; 26(6): 483-495, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38568339

ABSTRACT

PURPOSE OF REVIEW: Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder in women of reproductive age. It has been associated with metabolic, reproductive, and psychiatric disorders. Despite its association with insulin resistance (IR) and cardiovascular disease (CVD) risk factors, the association between PCOS and CVD outcomes has been conflicting. This review reports the updated evidence between PCOS, insulin resistance, and CVD events. RECENT FINDINGS: IR is highly prevalent occurring in 50 to 95% of general and obese PCOS women. The etiology of PCOS involves IR and hyperandrogenism, which lead to CVD risk factors, subclinical CVD, and CVD outcomes. Multiple studies including meta-analysis confirmed a strong association between PCOS and CVD events including ischemic heart disease, stroke, atrial fibrillation, and diabetes, particularly among premenopausal women, and these associations were mediated by metabolic abnormalities. PCOS is highly familial and has substantial CVD risk and transgenerational effects regardless of obesity. A personalized approach to the CVD risk assessment and management of symptom manifestations should be conducted according to its phenotypes. Lifestyle modifications and reduction in environmental stressors should be encouraged for CVD prevention among PCOS women.


Subject(s)
Cardiovascular Diseases , Insulin Resistance , Polycystic Ovary Syndrome , Humans , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/physiopathology , Female , Cardiovascular Diseases/etiology , Obesity/complications , Obesity/physiopathology , Risk Factors , Risk Assessment , Heart Disease Risk Factors , Prevalence , Hyperandrogenism/complications , Hyperandrogenism/physiopathology
2.
Front Endocrinol (Lausanne) ; 13: 868572, 2022.
Article in English | MEDLINE | ID: mdl-35757434

ABSTRACT

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.


Subject(s)
Hyperandrogenism , Polycystic Ovary Syndrome , Receptors, Androgen , Reproduction , Animals , Dihydrotestosterone/pharmacology , Female , Glucose/metabolism , Humans , Hyperandrogenism/chemically induced , Hyperandrogenism/genetics , Hyperandrogenism/metabolism , Hyperandrogenism/physiopathology , Liver/metabolism , Mice , Obesity/metabolism , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Receptors, Androgen/deficiency , Receptors, Androgen/metabolism , Reproduction/physiology
3.
Gynecol Endocrinol ; 38(1): 2-9, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34787028

ABSTRACT

Polycystic ovary syndrome (PCOS) is a disease whose diagnosis is based on the detection of hyperandrogenism (HA) and ovulatory dysfunction. Women with PCOS frequently develop insulin resistance (IR), which generates a metabolic condition that involves a decrease in the action of insulin at the cellular level and is linked to compensatory hyperinsulinemia (HI). In PCOS, the ovary remains sensitive to the action of insulin. Additionally, it has been observed that the main effect of insulin in the ovary is the stimulation of androgen synthesis, resulting in HA, one of the fundamental characteristics of the PCOS. In this sense, the excess of androgens favors the development of IR, thus perpetuating the cycle of IR-HI-HA, and therefore PCOS. Moreover, mitochondrial dysfunction is present in PCOS patients and is a common feature in both IR and HA. This review places electron transfer as a key element in HA and IR development, with emphasis on the relationship between androgen biosynthesis and mitochondrial function. Indeed, metformin has been involved in repair mitochondrial dysfunction, decrease of oxidative stress, reduction of androgens levels and the enhancing of insulin sensitivity. Therefore, we propose that treatment with metformin could decrease HI and consequently HA, restoring, at least in part, the metabolic and hormonal disorders of PCOS.


Subject(s)
Feedback, Physiological/physiology , Hyperandrogenism/physiopathology , Insulin Resistance/physiology , Polycystic Ovary Syndrome/physiopathology , Androgens/biosynthesis , Electron Transport/physiology , Female , Humans , Hyperandrogenism/drug therapy , Hyperinsulinism/drug therapy , Insulin/physiology , Metformin/therapeutic use , Mitochondria/drug effects , Mitochondria/physiology , Ovary/metabolism
4.
Sci Rep ; 11(1): 22097, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34764381

ABSTRACT

Normoferritinemic women with functional hyperandrogenism show a mild iron overload. Iron excess, hyperandrogenism, and cardioautonomic dysfunction contribute to blood pressure (BP) abnormalities in these patients. Furthermore, combined oral contraceptives (COC) prescribed for hyperandrogenic symptoms may worse BP recordings. Iron depletion by phlebotomy appears to lower BP in other acquired iron overload conditions. We aimed to determine the effect of iron depletion on the office BP, ambulatory BP monitoring, and frequency of hypertension in patients with functional hyperandrogenism submitted to standard therapy with COC. We conducted a phase 2 randomized, controlled, parallel, open-label clinical trial (NCT02460445) in adult women with functional hyperandrogenism including hyperandrogenic polycystic ovary syndrome and idiopathic hyperandrogenism. After a 3-month run-in period of treatment with 35 µg ethinylestradiol plus 2 mg cyproterone acetate, participants were randomized (1:1) to three scheduled bloodlettings or observation for another 9 months. Main outcome measures were the changes in office BP, 24-h-ambulatory BP, and frequency of hypertension in both study arms. From June 2015 to June 2019, 33 women were included in the intention-to-treat analyses. We observed an increase in mean office systolic BP [mean of the differences (MD): 2.5 (0.3-4.8) mmHg] and night-time ambulatory systolic BP [MD 4.1 (1.4-6.8) mmHg] after 3 months on COC. The percentage of nocturnal BP non-dippers also increased, from 28.1 to 92.3% (P < 0.001). Office and ambulatory BP did not change throughout the experimental period of the trial, both when considering all women as a whole or as a function of the study arm. The frequency of the non-dipping pattern in BP decreased during the experimental period [OR 0.694 (0.577-0.835), P < 0.001], regardless of the study arm. Decreasing iron stores by scheduled bloodletting does not override the BP abnormalities caused by COC in women with functional hyperandrogenism.


Subject(s)
Blood Pressure/drug effects , Contraceptives, Oral, Combined/therapeutic use , Hyperandrogenism/drug therapy , Adult , Blood Pressure/physiology , Blood Pressure Monitoring, Ambulatory/methods , Bloodletting/methods , Cyproterone Acetate/therapeutic use , Drug Combinations , Ethinyl Estradiol/therapeutic use , Female , Humans , Hyperandrogenism/physiopathology , Hypertension/physiopathology , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/physiopathology , Young Adult
5.
Med Clin North Am ; 105(6): 1099-1116, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34688417

ABSTRACT

The approach to hyperandrogenism in women varies depending on the woman's age and severity of symptoms. Once tumorous hyperandrogenism is excluded, the most common cause is PCOS. Hirsutism is the most common presenting symptom. The woman's concern about her symptoms plays an important role in the management of disease. Although measurement of testosterone is useful in identifying an underlying cause, care must be taken when interpreting the less accurate assays that are available commercially. Surgical resection is curative in tumorous etiologies, whereas medical management is the mainstay for non-tumorous causes.


Subject(s)
Androgen Antagonists/therapeutic use , Hyperandrogenism/drug therapy , Hyperandrogenism/physiopathology , Age Factors , Female , Humans , Hyperandrogenism/diagnosis , Hyperandrogenism/etiology , Polycystic Ovary Syndrome/complications , Racial Groups , Testosterone/blood , Women's Health
6.
Cell Death Dis ; 12(11): 999, 2021 10 25.
Article in English | MEDLINE | ID: mdl-34697294

ABSTRACT

Duplication of MECP2 (methyl-CpG-binding protein 2) gene causes a serious neurological and developmental disorder called MECP2 duplication syndrome (MDS), which is usually found in males. A previous clinical study reported that MDS patient has precocious puberty with hyperandrogenism, suggesting increased MeCP2 may cause male hyperandrogenism. Here we use an MDS mouse model and confirm that MECP2 duplication significantly upregulates androgen levels. We show for the first time that MeCP2 is highly expressed in the Leydig cells of testis, where androgen is synthesized. Mechanistically, MECP2 duplication increases androgen synthesis and decreases androgen to estrogen conversion through either the upregulation of luteinizing hormone receptor (LHCGR) in testis, as a result of MeCP2 binds to G-quadruplex structure of Lhcgr promoter and recruits the transcription activator CREB1 or the downregulation of the expression of aromatase in testis by binding the CpG island of Rorα, an upstream regulator of aromatase. Taken together, we demonstrate that MeCP2 plays an important role in androgen synthesis, supporting a novel non-CNS function of MeCP2 in the process of sex hormone synthesis.


Subject(s)
Hyperandrogenism/genetics , Methyl-CpG-Binding Protein 2/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Receptors, LH/metabolism , Animals , Disease Models, Animal , Down-Regulation , Humans , Hyperandrogenism/physiopathology , Male , Mice , Up-Regulation
7.
Eur Rev Med Pharmacol Sci ; 25(11): 3947-3954, 2021 06.
Article in English | MEDLINE | ID: mdl-34156673

ABSTRACT

OBJECTIVE: Polycystic ovary syndrome (PCOS) is a complex endocrine and metabolic disorders, which may contribute to the development of cardiovascular diseases. The aim of our study was to evaluate the association between of 24-hour ambulatory blood pressure measurement (ABPM) and selected biochemical and anthropometric parameters in women with PCOS. PATIENTS AND METHODS: The study involved 153 Polish, Caucasian women with PCOS hospitalized in the Department of Endocrinology Gynecology from January 2018 to March 2020. All women had stable body mass during the 3-month period. ABPM was performed using a portable lightweight device with oscillometric technology accepted by International Protocol of European Society of Hypertension (ABPM, HolCARD CR-07, Poland). RESULTS: The first factor taken into consideration was the variability phenotypic subgroups of PCOS on the values of 24-hour ABPM. We revealed that the daytime and night-time systolic and diastolic blood pressure values were significantly higher in phenotype A subgroup than in other subgroups. Moreover, daytime and night-time systolic and diastolic blood pressure value as well as day-time heart ratio value were significantly higher in subgroup with than without hyperandrogenemia. The obese women with PCOS were characterized of the highest value of all night-time measurements among women with PCOS and normal weight, overweight or obesity. In addition, insulin resistance in the PCOS subgroup was associated with lower value of systolic, diastolic blood pressure and both at daytime and night-time heart rate value than in insulin sensitive PCOS subgroup. CONCLUSIONS: Hyperandrogenemia and obesity were the crucial influencing factors on 24-hour ABPM in the group of women with PCOS. In addition, hypertension, apart from visceral obesity, hyperinsulinemia and insulin resistance, could be considered as component of metabolic syndrome in women with PCOS.


Subject(s)
Hyperandrogenism/physiopathology , Hypertension/physiopathology , Obesity/physiopathology , Polycystic Ovary Syndrome/physiopathology , Adult , Blood Pressure , Blood Pressure Monitoring, Ambulatory , Female , Heart Rate , Hormones/blood , Humans , Hyperandrogenism/blood , Hypertension/blood , Insulin Resistance , Obesity/blood , Phenotype , Polycystic Ovary Syndrome/blood , Young Adult
8.
Fertil Steril ; 116(4): 1139-1146, 2021 10.
Article in English | MEDLINE | ID: mdl-34119324

ABSTRACT

OBJECTIVE: To study the relationship between circulating chemokine cysteine-cysteine motif ligand (CCL) 5 levels and cysteine-cysteine chemokine receptor type 5 (CCR5) expression in peripheral blood mononuclear cells (PBMCs) and adipose tissue with hyperandrogenism and insulin resistance in patients with polycystic ovary syndrome (PCOS). DESIGN: Case-control study. SETTING: University teaching hospital. PATIENT(S): Fifteen women with PCOS and 15 controls matched for body mass index and age were enrolled in this study. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Plasma levels of CCL3, CCL4, and CCL5 were determined using enzyme-linked immunosorbent assay kits, and omental adipose tissue and PBMCs were analyzed using real-time polymerase chain reaction to determine the expression level of CCR5 in participants. RESULT(S): Levels of CCL5 were significantly higher in women with PCOS. Expression of CCR5 in adipose tissue and PBMCs was significantly higher in women with PCOS compared with that in women in the control group. Cysteine-cysteine chemokine receptor type 5 expression also was upregulated in THP-1 cells after chronic exposure to testosterone. Levels of CCL5 had a significant positive correlation with testosterone levels in women with PCOS. Moreover, CCR5 showed a positive correlation with fasting glucose levels, homeostasis model insulin resistance index, and C-reactive protein. CONCLUSION(S): Increased levels of CCL5 and overexpression of CCR5 in PBMCs and adipose tissue are associated with hyperandrogenism and insulin resistance in women with PCOS. Additionally, CCR5 and CCL5 may be used as biomarkers in the pathogenesis of PCOS.


Subject(s)
Abdominal Fat/metabolism , Chemokine CCL5/metabolism , Hyperandrogenism/metabolism , Insulin Resistance , Leukocytes, Mononuclear/metabolism , Polycystic Ovary Syndrome/metabolism , Receptors, CCR5/metabolism , T-Lymphocytes/metabolism , Testosterone/blood , Abdominal Fat/immunology , Abdominal Fat/physiopathology , Adult , Blood Glucose/metabolism , C-Reactive Protein/analysis , Case-Control Studies , Female , Humans , Hyperandrogenism/diagnosis , Hyperandrogenism/immunology , Hyperandrogenism/physiopathology , Insulin/blood , Leukocytes, Mononuclear/immunology , Lymphocyte Activation , Omentum , Polycystic Ovary Syndrome/diagnosis , Polycystic Ovary Syndrome/immunology , Polycystic Ovary Syndrome/physiopathology , Receptors, CCR5/genetics , T-Lymphocytes/immunology , THP-1 Cells , Up-Regulation , Young Adult
9.
Reprod Sci ; 28(9): 2574-2581, 2021 09.
Article in English | MEDLINE | ID: mdl-33721298

ABSTRACT

We reported that consumption of a western-style diet (WSD) with and without hyperandrogenemia perturbed placental perfusion and altered levels of glucose transporter proteins in rhesus macaques. Based on that result, we hypothesized that placental glucose uptake would be dysregulated in this model. In this study, female rhesus macaques were assigned at puberty to one of four groups: subcutaneous cholesterol implants + standard chow diet (controls, C); testosterone implants + chow (T); cholesterol implants + a high-fat, WSD; and T+WSD. After ~6 years of treatment, animals were mated, and pregnancies were delivered by cesarean section at gestational day (G) 130 (the term is G168). Placental villous explants were immediately prepared for radiolabeled glucose assay. Linear glucose uptake was observed between 0 and 30 s. At 20 s, glucose uptake in placental villous explants did not differ across the four treatment groups with values as follows: C: 25.5 ± 6.33, T: 22.9 ± 0.404, WSD: 26.9.0 ± 3.71, and T+WSD: 33.0 ± 3.12 (mean ± SD expressed in pmol/mg). Unlike our prior experiment, glucose transporter expression was reduced in WSD placentas, and our in vitro functional assay did not demonstrate a difference in glucose uptake across the transporting epithelium of the placenta. Notably, maternal blood glucose levels were significantly elevated in animals chronically fed a WSD. This disparity may indicate differences in glucose utilization and metabolism by the placenta itself, as glucose transporter expression and circulating fetal glucose concentrations were comparable across all four groups in this pregnancy cohort.


Subject(s)
Blood Glucose/metabolism , Diet, Western/adverse effects , Hyperandrogenism/microbiology , Placenta/blood supply , Placenta/metabolism , Animal Feed , Animals , Disease Models, Animal , Female , Glucose Transport Proteins, Facilitative/genetics , Glucose Transport Proteins, Facilitative/metabolism , Hyperandrogenism/blood , Hyperandrogenism/physiopathology , Macaca mulatta , Maternal Nutritional Physiological Phenomena , Nutritive Value , Placental Circulation , Pregnancy , Time Factors
10.
J Ovarian Res ; 14(1): 31, 2021 Feb 12.
Article in English | MEDLINE | ID: mdl-33579321

ABSTRACT

BACKGROUND: To explore the efficacy of follitropin delta in ovarian stimulation of patients with the Rotterdam ESHRE/ASRM 2003 phenotypes of polycystic ovarian syndrome (PCOS) using a retrospective case series with an electronic file search in a reproductive medicine clinic. CASE PRESENTATION: Seventy-four patients with PCOS undergoing ovarian stimulation according to the individualized dosing algorithm of follitropin delta for in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI)/oocyte freezing were included. Follitropin delta resulted in a high number of pre-ovulatory follicles at the end of stimulation as expected in patients with PCOS. There was a large number of oocytes retrieved with an acceptable percentage of metaphase II (MII) oocytes. There were no cases of moderate or severe OHSS across all phenotypes. CONCLUSION: Follitropin delta, using the individualized dosing algorithm, appears to be a safe method of ovarian stimulation with a low risk of OHSS in PCOS patients without sacrificing successful stimulation outcomes.


Subject(s)
Anovulation/physiopathology , Follicle Stimulating Hormone, Human/therapeutic use , Hyperandrogenism/physiopathology , Infertility, Female/therapy , Ovarian Hyperstimulation Syndrome/epidemiology , Ovulation Induction/methods , Polycystic Ovary Syndrome/physiopathology , Adult , Aromatase Inhibitors/therapeutic use , Chorionic Gonadotropin/therapeutic use , Dopamine Agonists/therapeutic use , Female , Fertilization in Vitro , Gonadotropin-Releasing Hormone/agonists , Humans , Infertility, Female/complications , Oocyte Retrieval , Ovarian Hyperstimulation Syndrome/chemically induced , Ovarian Hyperstimulation Syndrome/prevention & control , Phenotype , Polycystic Ovary Syndrome/complications , Pregnancy , Pregnancy Rate , Recombinant Proteins/therapeutic use , Reproductive Control Agents/therapeutic use , Retrospective Studies , Sperm Injections, Intracytoplasmic , Tissue Preservation
11.
Hypertension ; 77(3): 1010-1019, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33517680

ABSTRACT

The purpose of this prospective, population-based cohort study was to evaluate the roles of polycystic ovary syndrome (PCOS), obesity, weight gain, and hyperandrogenemia in the development of hypertensive disorders of pregnancy (HDP) through fertile age both in PCOS and in non-PCOS women. The study population-NFBC1966 (Northern Finland Birth Cohort 1966)-allowed a long-term follow-up of women from age 14 until 46 years who developed HDP (n=408) or did not (n=3373). HDP diagnosis was confirmed by combining hospital discharge records, data from Finnish Medical Birth Registers, and the questionnaire data at age 46. Women with self-reported PCOS (srPCOS; n=279), defined by both oligo-amenorrhea and hirsutism at age 31 or with PCOS diagnosis by age 46, were compared with women without reported PCOS (n=1577). Women with srPCOS had an increased HDP risk (odds ratio, 1.56 [95% CI, 1.03-2.37]), but the association disappeared after adjustment for body mass index. In women with srPCOS and HDP, body mass index increased from age 14 to 46 significantly more than in srPCOS women without HDP (median [interquartile range], 9.82 [6.23-14.6] and 7.21 [4.16-10.5] kg/m2, respectively; P<0.001). Also, in non-PCOS women, the increase was higher in women with (7.54 [5.32-11.62] kg/m2; P<0.001) than without HDP (6.33 [3.90-9.33] kg/m2; P<0.001). Increase in waist circumference between ages 31 and 46 years was associated with HDP but not with PCOS. Hyperandrogenemia at 31 or 46 years did not associate with HDP (1.44 [0.98-2.11]). In conclusion, obesity, especially abdominal obesity, and weight gain from adolescence to age 46, but not srPCOS or hyperandrogenemia, were associated with an increased risk of HDP.


Subject(s)
Hyperandrogenism/physiopathology , Hypertension, Pregnancy-Induced/physiopathology , Obesity/physiopathology , Polycystic Ovary Syndrome/physiopathology , Weight Gain/physiology , Adolescent , Adult , Female , Follow-Up Studies , Humans , Hypertension, Pregnancy-Induced/diagnosis , Logistic Models , Middle Aged , Population Surveillance , Pregnancy , Prospective Studies , Risk Factors , Self Report , Young Adult
12.
Fertil Steril ; 115(6): 1557-1568, 2021 06.
Article in English | MEDLINE | ID: mdl-33602559

ABSTRACT

OBJECTIVE: To examine the relation of menstrual cyclicity abnormalities to hyperandrogenism (HA) and dynamic state insulin resistance (IR) in oligo-ovulatory women with polycystic ovary syndrome (PCOS). DESIGN: Prospective cross-sectional study. SETTING: Tertiary-care academic center. PATIENT(S): Fifty-seven women with PCOS (1990 National Institutes of Health criteria) and 57 healthy control women matched by body mass index (BMI). INTERVENTION(S): Short insulin tolerance test (ITT). MAIN OUTCOME MEASURE(S): Menstrual cyclicity, sex hormone-binding globulin (SHBG), measures of HA (i.e., modified Ferriman-Gallwey score, total and free testosterone, dehydroepiandrosterone sulfate), and the rate constant for plasma glucose disappearance (kITT) derived from the short ITT. RESULT(S): Adjusting for age, BMI, and ethnicity, the mean androgen measures were higher and SHBG trended lower, kITT was lower, and the prevalence of IR was higher in PCOS than in controls, independent of menstrual cyclicity. The optimal cutoff point for IR was set at kITT value of 3.57%/minute or lower. Overall, 79% of the women with PCOS had IR. To control further for the effect of ethnicity, a subgroup of 46 non-Hispanic white PCOS participants were studied; those who exhibited amenorrhea (n = 15) or oligomenorrhea (n = 19) had or tended toward having a lower kITT and a higher prevalence of IR than the women with PCOS and oligo-ovulatory eumenorrhea (n = 12). The kITT trended lower and the prevalence of IR trended higher in women with PCOS and amenorrhea than those with oligomenorrhea. The measures of SHBG and HA were similar across the three menstrual groups. CONCLUSION(S): Oligo-ovulatory women with PCOS and overt oligo/amenorrhea have greater degrees of IR but not HA when compared with oligo-ovulatory eumenorrheic women with PCOS, suggesting that IR and hyperinsulinemia but not HA play a role in determining the degree of menstrual dysfunction, which can be used as a clinical marker for the degree of IR in oligo-ovulatory PCOS.


Subject(s)
Hyperandrogenism/etiology , Insulin Resistance , Menstrual Cycle , Menstruation Disturbances/etiology , Polycystic Ovary Syndrome/complications , Adult , Biomarkers/blood , Blood Glucose/analysis , Case-Control Studies , Cross-Sectional Studies , Dehydroepiandrosterone Sulfate/blood , Female , Humans , Hyperandrogenism/blood , Hyperandrogenism/diagnosis , Hyperandrogenism/physiopathology , Menstruation Disturbances/blood , Menstruation Disturbances/diagnosis , Menstruation Disturbances/physiopathology , Ovulation , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/diagnosis , Polycystic Ovary Syndrome/physiopathology , Prospective Studies , Risk Factors , Severity of Illness Index , Sex Hormone-Binding Globulin/analysis , Testosterone/blood , Time Factors , Young Adult
13.
Pediatr Res ; 89(1): 118-126, 2021 01.
Article in English | MEDLINE | ID: mdl-32247282

ABSTRACT

BACKGROUND: We recently identified 35 women with polycystic ovarian syndrome (PCOS) who exhibited features of micronodular adrenocortical hyperplasia. Steroid hormone analysis can be more accurate using state-of-the-art ultra-performance convergence chromatography-tandem mass spectrometry (UPC2-MS/MS). We hypothesized that UPC2-MS/MS may be used to better define hormonally this distinct subgroup of patients with PCOS. METHODS: Plasma from PCOS patients (n = 35) and healthy volunteers (HVs, n = 19) who all received dexamethasone testing was analyzed. Samples were grouped per dexamethasone responses and followed by UPC2-MS/MS analysis. When insufficient, samples were pooled from patients with similar responses to allow quantification over the low end of the assay. RESULTS: The C11-oxy C19 (11ß-hydroxyandrostenedione, 11keto-androstenedione, 11ß-hydroxytestosterone, 11keto-testosterone):C19 (androstenedione, testosterone) steroid ratio was decreased by 1.75-fold in PCOS patients compared to HVs. Downstream steroid metabolites 11ß-hydroxyandrosterone and 11keto-androsterone were also measurable. The C11-oxy C21 steroids, 11-hydroxyprogesterone and 11keto-dihydroprogesterone levels, were 1.2- and 1.7-fold higher in PCOS patients compared to HVs, respectively. CONCLUSIONS: We hypothesized that UPC2-MS/MS may accurately quantify steroids, in vivo, and identify novel metabolites in a subgroup of patients with PCOS and adrenal abnormalities. Indeed, it appears that adrenal C11-oxy steroids have the potential of being used diagnostically to identify younger women and adolescents with PCOS who also have some evidence of micronodular adrenocortical hyperplasia. IMPACT: Adrenal C11-oxy steroids may be clinically important in identifying young patients with PCOS and adrenal abnormalities. The steroids presented in our manuscript have not yet been considered in the clinical setting so far, and we believe that this study could represent a first focused step towards the characterization of a distinct subgroup of women with PCOS who may in fact be treated differently than the average patient with PCOS. This paper can change the understanding of PCOS as one disorder: it is in fact a heterogeneous condition. In addition, for the subgroup of patients with PCOS associated with adrenocortical dysfunction, our paper provides novel hormonal markers that can be used diagnostically. Finally, the paper also adds to the basic pathophysiological understanding of adrenocortical-ovarian interactions in steroidogenesis of young women and adolescent girls with PCOS.


Subject(s)
Adrenal Cortex/metabolism , Chromatography, Liquid , Hyperandrogenism/blood , Polycystic Ovary Syndrome/blood , Steroids/blood , Tandem Mass Spectrometry , Adolescent , Adrenal Cortex/physiopathology , Adult , Biomarkers/blood , Case-Control Studies , Female , Humans , Hyperandrogenism/diagnosis , Hyperandrogenism/physiopathology , Polycystic Ovary Syndrome/diagnosis , Polycystic Ovary Syndrome/physiopathology , Prospective Studies , Young Adult
14.
Fertil Steril ; 116(1): 232-242, 2021 07.
Article in English | MEDLINE | ID: mdl-33341231

ABSTRACT

OBJECTIVE: To examine whether subcutaneous (SC) abdominal adipose stem cell differentiation into adipocytes in vitro predicts insulin sensitivity (Si) in vivo in normal-weight women with polycystic ovary syndrome (PCOS) and controls. DESIGN: Prospective cohort study. SETTING: Academic medical center. PATIENT(S): Eight normal-weight women with PCOS and 8 age- and body mass index-matched controls. INTERVENTION(S): Women underwent circulating hormone/metabolic determinations, intravenous glucose tolerance testing, total-body dual-energy x-ray absorptiometry, and SC abdominal fat biopsy. MAIN OUTCOME MEASURE(S): PPARγ and CEBPa gene expression and lipid content of adipocytes matured in vitro were compared between women with PCOS and control women, and correlated with patient characteristics, systemic Si, and adipose insulin resistance (adipose-IR). RESULT(S): Serum androgen levels, adipose-IR, and percentage of android fat were greater in women with PCOS than control women. Stem cell PPARγ and CEBPa gene expression increased maximally by day 12 without a female-type effect. In control cells, gene expression positively correlated with fasting serum insulin levels (both genes) and adipose-IR (CEBPa) and negatively correlated with Si (CEBPa). Conversely, CEBPa gene expression in PCOS cells negatively correlated with adipose-IR and serum free testosterone, whereas total lipid accumulation in these cells positively corelated with Si. CONCLUSION: In normal-weight women with PCOS, accelerated SC abdominal adipose stem cell differentiation into adipocytes in vitro favors Si in vivo, suggesting a role for hyperandrogenism in the evolution of metabolic thrift to enhance fat storage through increased cellular glucose uptake.


Subject(s)
Abdominal Fat/metabolism , Adipocytes/metabolism , Adipogenesis , Hyperandrogenism/metabolism , Insulin Resistance , Polycystic Ovary Syndrome/metabolism , Stem Cells/metabolism , Abdominal Fat/pathology , Abdominal Fat/physiopathology , Adipocytes/pathology , Adiposity , Adult , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Case-Control Studies , Cells, Cultured , Female , Humans , Hyperandrogenism/pathology , Hyperandrogenism/physiopathology , Ideal Body Weight , Lipid Metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Polycystic Ovary Syndrome/pathology , Polycystic Ovary Syndrome/physiopathology , Prospective Studies , Stem Cells/pathology , Time Factors , Young Adult
16.
Fertil Steril ; 113(6): 1275-1285.e2, 2020 06.
Article in English | MEDLINE | ID: mdl-32482257

ABSTRACT

OBJECTIVE: To investigate the placental morphology alterations and identify the clinical characteristics of women with polycystic ovary syndrome (PCOS) and their newborns. Pregnant women with PCOS (n = 12) and pregnant women without PCOS (n = 11) were recruited. Then, the placenta, maternal blood and cord blood were collected after delivery. DESIGN: Clinical observational study. SETTING: Not applicable. PATIENT(S): In the present study, pregnant women with PCOS and healthy pregnant women were recruited from the clinic of the Department of Obstetrics and Gynecology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, China, between February 2015 and October 2015. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): A proteomic analysis was performed on the placenta in women with PCOS and healthy women. RESULT(S): The maternal testosterone, androstenedione, dehydroepiandrosterone sulfate, free androgen index, cholesterol, apolipoprotein B, and apolipoprotein B/apolipoprotein A-I levels were significantly higher in the PCOS group than in the control group, and the offspring in the PCOS group had higher dehydroepiandrosterone sulfate, high-density lipoprotein, and cholesterol levels, when compared with the control group. The placenta in the PCOS group demonstrated infarction, calcification, and a greater intervillous space, when compared with the control group. A higher level of estrogen receptor-ß protein was observed in the placenta of women with PCOS, when compared with women without PCOS. A total of 258 proteins in the placenta were identified to be significantly different, when the PCOS and control groups were compared, and fibronectin 1 exhibited the closest relationship with other differential proteins. CONCLUSION(S): The overexposure to hyperandrogenism and hyperlipidemia affects the functions of the placenta, which are associated with the development of metabolic disorders in newborns.


Subject(s)
Androgens/blood , Fetal Blood/metabolism , Infant, Newborn, Diseases/etiology , Lipids/blood , Placenta/metabolism , Polycystic Ovary Syndrome/complications , Adult , Biomarkers/blood , Case-Control Studies , Female , Humans , Hyperandrogenism/blood , Hyperandrogenism/etiology , Hyperandrogenism/physiopathology , Hyperlipidemias/blood , Hyperlipidemias/etiology , Hyperlipidemias/physiopathology , Infant, Newborn , Infant, Newborn, Diseases/blood , Infant, Newborn, Diseases/physiopathology , Placenta/physiopathology , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/physiopathology , Pregnancy , Risk Factors , Up-Regulation
17.
Nurs Womens Health ; 24(2): 115-126, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32273076

ABSTRACT

Polycystic ovary syndrome (PCOS) is the most common endocrinopathy, affecting as many as 5% to 20% of women of reproductive age, depending on the diagnostic criteria applied. Features of PCOS include physiologic anovulation, hyperandrogenism, elevated luteinizing hormone, and increased gonadotropin-releasing hormone pulse frequency, which often manifest physically as acne and hirsutism. The clinical presentation of PCOS often mimics normal pubertal physiologic development, which may delay diagnosis and treatment of the condition in adolescent girls. A diagnosis of PCOS has life-long implications and is associated with increased risk for infertility, obesity, Type 2 diabetes, endometrial hyperplasia, uterine carcinoma, metabolic disorder, and cardiovascular disease. In this article, we provide an overview of clinical presentation, diagnostic criteria, health consequences, and current evidence-based clinical guidelines for the appropriate diagnosis and management of PCOS in adolescents.


Subject(s)
Polycystic Ovary Syndrome/complications , Adolescent , Anovulation/etiology , Anovulation/physiopathology , Female , Gonadotropin-Releasing Hormone/analysis , Humans , Hyperandrogenism/etiology , Hyperandrogenism/physiopathology , Insulin Resistance/physiology , Luteinizing Hormone/analysis , Obesity/complications , Obesity/physiopathology , Polycystic Ovary Syndrome/physiopathology
18.
Transl Res ; 219: 13-29, 2020 05.
Article in English | MEDLINE | ID: mdl-32119846

ABSTRACT

The mechanisms underlying metabolic and reproductive dysfunction caused by arrhythmic circadian clock and their involvement in polycystic ovary syndrome (PCOS) are not understood. Here, we addressed this issue using rats with constant light or darkness exposure for 8 weeks and human leukocytes and serum of PCOS and non-PCOS patients. Additionally, we utilized HepG2 cells and KGN cells to verify the molecular mechanisms. The arrhythmic expressions of circadian clock genes due to constant darkness induced the metabolic and reproductive hallmarks of PCOS in rats. After exposure to constant darkness, decreased brain and muscle ARNT-like protein 1 (BMAL1) promoted insulin resistance via glucose transporter 4 (GLUT4), and decreased period (PER) 1 and PER2 promoted androgen excess via insulin-like growth factor-binding protein 4 (IGFBP4) and sex hormone binding globulin (SHBG) in the liver. Hyperinsulinemia and hyperandrogenism shared a bidirectional link promoting aberrant expression of circadian genes and inducing apoptosis of ovarian granulosa cells. Notably, the altered expressions of circadian clock genes in darkness-treated rats matched those of PCOS patients. Furthermore, melatonin treatment relieved the hyperinsulinemia and hyperandrogenism of darkness-treated rats via BMAL1, PER1, and PER2. Restoring normal light/dark exposure for 2 weeks reversed these conditions via BMAL1. In conclusion, our findings elucidated the critical function of circadian clock genes, especially BMAL1, PER1, and PER2 in PCOS, which might aid the development of feasible preventive and therapeutic strategies for PCOS in women with biorhythm disorder.


Subject(s)
Circadian Clocks , Darkness , Hyperandrogenism/therapy , Insulin Resistance , Polycystic Ovary Syndrome/physiopathology , ARNTL Transcription Factors/metabolism , Animals , Apoptosis/physiology , Female , Granulosa Cells/pathology , Hyperandrogenism/physiopathology , Insulin/physiology , Liver/metabolism , Rats , Rats, Sprague-Dawley , Testosterone/physiology
19.
BJOG ; 127(8): 930-939, 2020 07.
Article in English | MEDLINE | ID: mdl-32048421

ABSTRACT

BACKGROUND: There is currently no concise systematic review or meta-analysis addressing cardio-metabolic risk factors in women experiencing infertility. OBJECTIVES: To determine whether infertile women have higher levels of cardiovascular risk factors compared with fertile women. SEARCH STRATEGY: We performed a systematic literature search using PubMed, Embase and CINAHL, Scopus and additional manual and bibliographic searches for relevant articles (end search date 6 November 2019). SELECTION CRITERIA: We selected studies that compared cardio-metabolic risk factors in fertile and infertile women of reproductive age. DATA COLLECTION AND ANALYSIS: At least two authors independently screened potentially eligible studies. MAIN RESULTS: There was an increased presence of several cardio-metabolic risk factors in infertile women compared with fertile women. Infertile women had statistically significant higher body mass index (BMI), increased total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C) and triglycerides (TG) compared with fertile women. Fasting glucose, fasting insulin, homeostatic model assessment of insulin resistance (HOMA-IR) and mean arterial pressure were not found to be different between fertile and infertile women. A subgroup analysis revealed that TC, fasting glucose and fasting insulin were increased, and high-density lipoprotein was decreased only in women with polycystic ovarian syndrome compared with fertile women, whereas BMI, TG and LDL-C were statistically significantly increased in women with any indication of infertility compared with fertile women. CONCLUSIONS: Infertile women have a higher level of cardio-metabolic risk factors compared with fertile women. This finding has clinical implications for infertile women in general, and those attempting to conceive through medically assisted reproduction. TWEETABLE ABSTRACT: Infertile women appear to have a higher level of cardio-metabolic risk factors compared with fertile women.


Subject(s)
Cardiovascular Diseases/physiopathology , Disease Susceptibility/physiopathology , Hyperandrogenism/physiopathology , Infertility, Female/physiopathology , Metabolic Syndrome/physiopathology , Polycystic Ovary Syndrome/physiopathology , Adult , Body Mass Index , Cardiovascular Diseases/blood , Cholesterol, LDL/blood , Disease Susceptibility/blood , Female , Humans , Hyperandrogenism/blood , Hyperandrogenism/complications , Infertility, Female/blood , Infertility, Female/etiology , Insulin Resistance/physiology , Metabolic Syndrome/blood , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/complications , Risk Factors , Triglycerides/blood
20.
BMC Endocr Disord ; 20(1): 3, 2020 Jan 06.
Article in English | MEDLINE | ID: mdl-31906930

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is an endocrine and metabolic disorder, and its pathogenesis is still under debate. Trimethylamine-N-oxide (TMAO) is a small, organic compound generated by the gut microbiome with a hypothesized relation to insulin resistance (IR) and low-grade inflammation in PCOS. By comparing plasma TMAO levels in non-PCOS participants and PCOS patients without hyperandrogenism (HA), we aimed to determine whether plasma TMAO levels correlate with PCOS without HA and to analyze their relationship with low-grade inflammation and IR. METHODS: A total of 27 PCOS patients without HA and 23 non-PCOS participants were enrolled in this study and subdivided into "nonobese" and "obese" arms for each group. Levels of plasma TMAO were quantified, and basic clinical characteristics and plasma biomarkers of inflammation were assessed. RESULTS: First, plasma TMAO levels, insulin levels and homeostatic model assessment of insulin resistance (HOMA-IR) values were higher in PCOS patients without HA, especially in the obese subgroup. Second, the levels of the inflammatory factors interleukin (IL)-17A, IL-18 and interferon gamma (IFN-γ) were significantly increased in obese PCOS patients without HA. Third, plasma TMAO levels were associated with body mass index (BMI) in the normal-weight groups, and the obese groups had higher fasting plasma insulin (FINS) and HOMA-IR values. Finally, logistic regression showed that the plasma levels of TMAO and luteinizing hormone/follicle-stimulating hormone (LH/FSH) were independent predictors of PCOS and indicated an increased risk of PCOS. CONCLUSIONS: Elevated plasma TMAO levels may be associated with the pathogenesis of PCOS without HA and correlated with increased systemic inflammation. Further studies are needed to determine the suitability of TMAO as a predictive biomarker and to identify possible therapies for PCOS.


Subject(s)
Biomarkers/blood , Hyperandrogenism/physiopathology , Methylamines/blood , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/epidemiology , Adult , Body Mass Index , Case-Control Studies , China/epidemiology , Female , Follow-Up Studies , Humans , Incidence , Insulin Resistance , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL