Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Cell Cycle ; 17(8): 1007-1013, 2018.
Article in English | MEDLINE | ID: mdl-29912628

ABSTRACT

In an attempt to explore the early developmental arrest in embryos from polycystic ovarian syndrome (PCOS) patients, we sequenced the transcriptome profiles of PCOS arrested 2-cell embryos, non-PCOS arrested 2-cell embryos and non-arrested 2-cell embryos using single-cell RNA-Seq technique. Differential expression analysis was performed using the DEGSeq R package. Gene Ontology (GO) enrichment was analyzed using the GOseq R package. Data revealed 62 differentially expressed genes between non-PCOS arrested and PCOS arrested embryos and 2217 differentially expressed genes between PCOS arrested and non-arrested 2-cell embryos. A total of 49 differently expressed genes (DEGs) were annotated with GO terms in the up-regulated genes between PCOS arrested and non-PCOS arrested embryos after GO enrichment. A total of 29 DEGs were annotated with GO terms in the down-regulated genes between PCOS arrested and non-arrested 2-cell embryos after GO enrichment. These data can provide a reference for screening specific genes involved in the arrest of PCOS embryos.


Subject(s)
Embryo, Mammalian/pathology , Gene Expression Profiling , Polycystic Ovary Syndrome/embryology , Polycystic Ovary Syndrome/genetics , Cell Cycle/genetics , Down-Regulation/genetics , Female , Gene Ontology , Humans , Molecular Sequence Annotation , Up-Regulation/genetics
2.
Reprod Biol Endocrinol ; 15(1): 61, 2017 Aug 08.
Article in English | MEDLINE | ID: mdl-28789693

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS), whose aetiology is unknown, is predominately a familial syndrome but confirmation of candidate genes has proved elusive. The developmental hypothesis for the origin of PCOS suggests that exposure of the fetus to excess androgens influences imprinting, leading to altered genetic expression in adult life. The aim of this pilot study was to examine whether the female fetus of a mother with PCOS is indeed exposed to excess androgens. METHODS: Using sebum production in the newborn as a surrogate for exposure to excess androgens during pregnancy thisprospective case control studyexamined whether neonatal sebum excretion is greater in female infants born to PCOS mothers compared to non-PCOS. Women with known PCOS (all 3 Rotterdam criteria) (n = 9) and non-PCOS controls (n = 12), with a female fetus, were recruited at 24 weeks pregnancy and serum testosterone estimated. Sebum was measured using Sebutape® for 30 and 60 min within 24 h of birth, at 1 week, 4-6 weeks and 6 months after birth in both mother and child. Sebum excretion was measured in mother and child in the same site at each time frame and consistently. All semi-quantitative sebum excretion estimations were compared (t-test) between the two groups and correlated with testosterone concentrations during pregnancy. RESULTS: In this pilot study, 21 women completed the 6 month examination period (PCOS group (n = 9) and controls (n = 12). Mean testosterone was 6.2 nmol/L (normal <3.1 nmol/L) in PCOS mothers and 2.75 nmol/L in controls at 24 weeks pregnancy. At all time frames, the results of sebum excretion at 30 and 60 min were consistent. The sebum excretion of mothers in both groups was fairly constant from birth throughout 6 months. All babies were born between 37 and 41 weeks gestational age. Six of nine newborns had detectable sebum excretion at birth in the PCOS mothers group compared to 1 of 12 in the controls (P = 0.01). CONCLUSIONS: These results suggest that women with PCOS could hyper-expose their fetus to androgens in-utero and that this may be detected using a simple novel test within 24 h of birth to predict development of PCOS in adult life and induce research to eliminate its symptoms. TRIAL REGISTRATION: NCT 02654548 .Clinical Trials UK.Retrospectively registered 11/1/16.


Subject(s)
Polycystic Ovary Syndrome/complications , Prenatal Exposure Delayed Effects , Sebum/metabolism , Androgens/metabolism , Female , Genomic Imprinting , Humans , Infant, Newborn , Pilot Projects , Polycystic Ovary Syndrome/diagnosis , Polycystic Ovary Syndrome/embryology , Pregnancy
3.
PLoS One ; 10(7): e0132113, 2015.
Article in English | MEDLINE | ID: mdl-26148093

ABSTRACT

Increased maternal androgen exposure during pregnancy programmes a polycystic ovary syndrome (PCOS)-like condition, with metabolic dysfunction, in adult female offspring. Other in utero exposures associated with the development of insulin resistance, such as intrauterine growth restriction and exposure to prenatal glucocorticoids, are associated with altered fetal gluconeogenesis. We therefore aimed to assess the effect of maternal androgenisation on the expression of PEPCK and G6PC in the ovine fetus. Pregnant Scottish Greyface sheep were treated with twice weekly testosterone propionate (TP; 100mg) or vehicle control from day 62 to day 102 of gestation. At day 90 and day 112 fetal plasma and liver and kidney tissue was collected for analysis. PEPCK and G6PC expression were analysed by quantitative RT-PCR, immunohistochemistry and western blotting. PEPCK and G6PC were localised to fetal hepatocytes but maternal androgens had no effect on female or male fetuses. PEPCK and G6PC were also localised to the renal tubules and renal PEPCK (P<0.01) and G6PC (P = 0.057) were lower in females after prenatal androgenisation with no change in male fetuses. These tissue and sex specific observations could not be explained by alterations in fetal insulin or cortisol. The sexual dimorphism may be related to the increase in circulating estrogen (P<0.01) and testosterone (P<0.001) in females but not males. The tissue specific effects may be related to the increased expression of ESR1 (P<0.01) and AR (P<0.05) in the kidney when compared to the fetal liver. After discontinuation of maternal androgenisation female fetal kidney PEPCK expression normalised. These data further highlight the fetal and sexual dimorphic effects of maternal androgenisation, an antecedent to adult disease and the plasticity of fetal development.


Subject(s)
Androgens/adverse effects , Gluconeogenesis/drug effects , Kidney/embryology , Polycystic Ovary Syndrome/embryology , Prenatal Exposure Delayed Effects/metabolism , Testosterone Propionate/adverse effects , Androgens/pharmacology , Animals , Disease Models, Animal , Female , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Glucose-6-Phosphatase/biosynthesis , Hepatocytes/metabolism , Humans , Kidney/pathology , Male , Phosphoenolpyruvate Carboxykinase (ATP)/biosynthesis , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/pathology , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/pathology , Sheep , Testosterone Propionate/pharmacology
4.
PLoS One ; 8(2): e56263, 2013.
Article in English | MEDLINE | ID: mdl-23457541

ABSTRACT

Using an ovine model of polycystic ovary syndrome (PCOS), (pregnant ewes injected with testosterone propionate (TP) (100 mg twice weekly) from day (d)62 to d102 of d147 gestation (maternal injection - MI-TP)), we previously reported female offspring with normal glucose tolerance but hyperinsulinemia. We therefore examined insulin signalling and pancreatic morphology in these offspring using quantitative (Q) RT-PCR and western blotting. In addition the fetal pancreatic responses to MI-TP, and androgenic and estrogenic contributions to such responses (direct fetal injection (FI) of TP (20 mg) or diethylstilbestrol (DES) (20 mg) at d62 and d82 gestation) were assessed at d90 gestation. Fetal plasma was assayed for insulin, testosterone and estradiol, pancreatic tissue was cultured, and expression of key ß-cell developmental genes was assessed by QRT-PCR. In female d62MI-TP offspring insulin signalling was unaltered but there was a pancreatic phenotype with increased numbers of ß-cells (P<0.05). The fetal pancreas expressed androgen receptors in islets and genes involved in ß-cell development and function (PDX1, IGF1R, INSR and INS) were up-regulated in female fetuses after d62MI-TP treatment (P<0.05-0.01). In addition the d62MI-TP pancreas showed increased insulin secretion under euglycaemic conditions (P<0.05) in vitro. The same effects were not seen in the male fetal pancreas or when MI-TP was started at d30, before the male programming window. As d62MI-TP increased both fetal plasma testosterone (P<0.05) and estradiol concentrations (P<0.05) we assessed the relative contribution of androgens and estrogens. FI-TP (commencing d62) (not FI-DES treatment) caused elevated basal insulin secretion in vitro and the genes altered by d62MI-TP treatment were similarly altered by FI-TP but not FI-DES. In conclusion, androgen over-exposure alters fetal pancreatic development and ß-cell numbers in offspring. These data suggest that that there may be a primary pancreatic phenotype in models of PCOS, and that there may be a distinct male and female pancreas.


Subject(s)
Androgens/pharmacology , Fetus/drug effects , Fetus/embryology , Pancreas/drug effects , Pancreas/embryology , Polycystic Ovary Syndrome/embryology , Androgens/administration & dosage , Animals , Estradiol/metabolism , Estrogens/pharmacology , Female , Fetus/metabolism , Fetus/pathology , Gene Expression Regulation, Developmental/drug effects , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Maternal Exposure/adverse effects , Pancreas/metabolism , Pancreas/pathology , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Pregnancy , Sheep , Signal Transduction/drug effects , Testosterone Propionate/administration & dosage , Testosterone Propionate/pharmacology
5.
Mol Cell Endocrinol ; 373(1-2): 21-8, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23370180

ABSTRACT

With close genomic and phenotypic similarity to humans, nonhuman primate models provide comprehensive epigenetic mimics of polycystic ovary syndrome (PCOS), suggesting early life targeting for prevention. Fetal exposure to testosterone (T), of all nonhuman primate emulations, provides the closest PCOS-like phenotypes, with early-to-mid gestation T-exposed female rhesus monkeys exhibiting adult reproductive, endocrinological and metabolic dysfunctional traits that are co-pathologies of PCOS. Late gestational T exposure, while inducing adult ovarian hyperandrogenism and menstrual abnormalities, has less dysfunctional metabolic accompaniment. Fetal exposures to dihydrotestosterone (DHT) or diethylstilbestrol (DES) suggest androgenic and estrogenic aspects of fetal programming. Neonatal exposure to T produces no PCOS-like outcome, while continuous T treatment of juvenile females causes precocious weight gain and early menarche (high T), or high LH and weight gain (moderate T). Acute T exposure of adult females generates polyfollicular ovaries, while chronic T exposure induces subtle menstrual irregularities without metabolic dysfunction.


Subject(s)
Disease Models, Animal , Polycystic Ovary Syndrome/metabolism , Age Factors , Animals , Animals, Newborn , Female , Humans , Insulin Resistance , Phenotype , Polycystic Ovary Syndrome/embryology , Pregnancy , Primates , Testosterone/physiology
6.
Hum Reprod Update ; 17(2): 210-27, 2011.
Article in English | MEDLINE | ID: mdl-20961922

ABSTRACT

BACKGROUND: There is now compelling evidence that long-term health and physiological function are modified by events that occur early in life and involve interactions between the genome and the developmental environment. That reproductive function may similarly be influenced by early life events has been established in selected human populations, and investigations into underlying mechanisms are the subject of current animal studies. METHODS: No systematic literature search was conducted. This review highlights early life influences on reproduction with a particular focus on nutritional impacts, and provides a brief overview with reference to some key studies in both the human and animal literature. We highlight the controversies, current unanswered questions and mechanisms underlying the association between the early life environment and long-term reproductive function. RESULTS AND CONCLUSIONS: Currently, the impact of early life events on reproductive health and disease risk is poorly understood. It is clear, however, that nutrition spanning the entire developmental lifespan plays an integral role. Improved insight into the underlying mechanisms is likely to have significant implications for our current understanding of reproductive disorders, and therefore for the health and reproductive potential of future generations.


Subject(s)
Prenatal Nutritional Physiological Phenomena , Reproductive Physiological Phenomena , Adolescent , Adult , Animals , Environmental Exposure , Female , Humans , Menopause/physiology , Models, Animal , Obesity/complications , Obesity/epidemiology , Polycystic Ovary Syndrome/embryology , Polycystic Ovary Syndrome/epidemiology , Pregnancy , Puberty/physiology , Puberty, Precocious/epidemiology , Rats
7.
Hum Reprod Update ; 17(1): 17-33, 2011.
Article in English | MEDLINE | ID: mdl-20639519

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is a common metabolic dysfunction and heterogeneous endocrine disorder in women of reproductive age. Although patients with PCOS are typically characterized by increased numbers of oocytes retrieved during IVF, they are often of poor quality, leading to lower fertilization, cleavage and implantation rates, and a higher miscarriage rate. METHODS: For this review, we searched the database MEDLINE (1950 to January 2010) and Google for all full texts and/or abstract articles published in English with content related to oocyte maturation and embryo developmental competence. RESULTS: The search showed that alteration of many factors may directly or indirectly impair the competence of maturating oocytes through endocrine and local paracrine/autocrine actions, resulting in a lower pregnancy rate in patients with PCOS. The extra-ovarian factors identified included gonadotrophins, hyperandrogenemia and hyperinsulinemia, although intra-ovarian factors included members of the epidermal, fibroblast, insulin-like and neurotrophin families of growth factors, as well as the cytokines. CONCLUSIONS: Any abnormality in the extra- and/or intra-ovarian factors may negatively affect the granulosa cell-oocyte interaction, oocyte maturation and potential embryonic developmental competence, contributing to unsuccessful outcomes for patients with PCOS who are undergoing assisted reproduction.


Subject(s)
Embryonic Development , Oocytes/growth & development , Ovary/physiopathology , Polycystic Ovary Syndrome/embryology , Female , Follicle Stimulating Hormone/deficiency , Humans , Hyperandrogenism/complications , Hyperinsulinism/complications , Intercellular Signaling Peptides and Proteins/physiology , Luteinizing Hormone/metabolism , Oocytes/metabolism , Paracrine Communication , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/metabolism , Pregnancy
8.
Endocr Dev ; 13: 145-158, 2008.
Article in English | MEDLINE | ID: mdl-18493139

ABSTRACT

Adrenal androgen excess is found in adult female rhesus monkeys previously exposed to androgen treatment during early gestation. In adulthood, such prenatally androgenized female monkeys exhibit elevated basal circulating levels of dehydroepiandrosterone sulfate (DHEAS), typical of polycystic ovary syndrome (PCOS) women with adrenal androgen excess. Further androgen and glucocorticoid abnormalities in PA female monkeys are revealed by acute ACTH stimulation: DHEA, androstenedione and corticosterone responses are all elevated compared to responses in controls. Pioglitazone treatment, however, diminishes circulating DHEAS responses to ACTH in both prenatally androgenized and control female monkeys, while increasing the 17-hydroxyprogesterone response and reducing the DHEA to 17-hydroxyprogesterone ratio. Since 60-min post-ACTH serum values for 17-hydroxyprogesterone correlate negatively with basal serum insulin levels (all female monkeys on pioglitazone and placebo treatment combined), while similar DHEAS values correlate positively with basal serum insulin levels, circulating insulin levels may preferentially support adrenal androgen biosynthesis in both prenatally androgenized and control female rhesus monkeys. Overall, our findings suggest that differentiation of the monkey adrenal cortex in a hyperandrogenic fetal environment may permanently upregulate adult adrenal androgen biosynthesis through specific elevation of 17,20-lyase activity in the zona fasciculata-reticularis. As adult prenatally androgenized female rhesus monkeys closely emulate PCOS-like symptoms, excess fetal androgen programming may contribute to adult adrenal androgen excess in women with PCOS.


Subject(s)
Disease Models, Animal , Fetal Development/physiology , Hyperandrogenism/embryology , Polycystic Ovary Syndrome/embryology , Primates , Adrenal Glands/embryology , Adrenal Glands/metabolism , Androgens/metabolism , Animals , Animals, Newborn , Female , Fetal Diseases/pathology , Gestational Age , Humans , Hyperandrogenism/pathology , Insulin/physiology , Macaca mulatta/embryology , Models, Biological , Polycystic Ovary Syndrome/metabolism , Primates/embryology
9.
Biol Reprod ; 79(1): 154-63, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18385445

ABSTRACT

Experimentally induced fetal androgen excess induces polycystic ovary syndrome-like traits in adult female rhesus monkeys (Macaca mulatta). Developmental changes leading to this endocrinopathy are not known. We therefore studied 15 time-mated, gravid female rhesus monkeys with known female fetuses. Nine dams received daily s.c. injections of 15 mg of testosterone propionate (TP), and six received injections of oil vehicle (control) from 40 through 80 days of gestation (term, 165 days; range, +/-10 days). All fetuses were delivered by cesarean section using established methods at term. Ultrasound-guided fetal blood sample collection and peripheral venous sample collection of dams and subsequent infants enabled determination of circulating levels of steroid hormones, LH and FSH. The TP injections elevated serum testosterone and androstenedione levels in the dams and prenatally androgenized (PA) fetuses. After cessation of TP injections, testosterone levels returned to values within the reference range for animals in these age groups, whereas serum androstenedione levels in PA infants were elevated. The TP injections did not increase estrogen levels in the dams or the PA fetuses or infants, yet conjugated estrogen levels were elevated in the TP-injected dams. Serum levels of LH and FSH were elevated in late-gestation PA fetuses, and LH levels were elevated in PA infants. These studies suggest that experimentally induced fetal androgen excess increases gonadotropin secretion in PA female fetuses and infants and elevates endogenous androgen levels in PA infants. Thus, in this nonhuman primate model, differential programming of the fetal hypothalamo-pituitary unit with concomitant hyperandrogenism provides evidence to suggest developmental origins of LH and androgen excess in adulthood.


Subject(s)
Androgens/pharmacology , Endocrine System/physiopathology , Fetus/drug effects , Polycystic Ovary Syndrome/chemically induced , Prenatal Exposure Delayed Effects/chemically induced , Virilism/chemically induced , Androgens/blood , Animals , Animals, Newborn , Birth Weight/drug effects , Estrogens/blood , Female , Fetus/pathology , Luteinizing Hormone/blood , Macaca mulatta , Polycystic Ovary Syndrome/embryology , Polycystic Ovary Syndrome/pathology , Polycystic Ovary Syndrome/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects/blood , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/physiopathology , Prenatal Exposure Delayed Effects/veterinary , Progesterone/blood , Virilism/blood , Virilism/pathology , Virilism/physiopathology
10.
Med Hypotheses ; 70(3): 638-42, 2008.
Article in English | MEDLINE | ID: mdl-17764855

ABSTRACT

Polycystic ovary syndrome (PCOS) is one of the most common, yet heterogeneous and complex, endocrine disorders in women of reproductive age. Although the aetiology of PCOS remains uncertain, emerging evidence has indicated that exposure of the female fetus to the hyperandrogenism milieu in utero may result in PCOS phenotype after birth. Such a phenomenon has been formulated as the fetal origin of PCOS, which intends to give a possible explanation for PCOS aetiology. Given that the epigenetic modifications are usually involved in the development and inheritance of many adult diseases with fetal origin, we propose a hypothesis here referred to as "epigenetic abnormality underlying the fetal origin of PCOS". It states that in utero hyperandrogenism exposure may disturb the epigenetic reprogramming in fetal reproductive tissue, thereby resulting in postnatal POCS phenotype in women of reproductive age. Meanwhile, the incomplete erasure of such epigenetic abnormality in germ cells after fertilization may promote the transgenerational inherence of POCS. Thus, this epigenetic abnormality hypothesis has established a novel mechanism for PCOS development and inheritance. If verified, our hypothesis would open new avenues for the possible intervention at the critical period of prenatal life to prevent PCOS development and inheritance in adult women. Moreover, analysis of the epigenetic phenotypes and identification of specific epigenetic changes may help develop new tools for monitoring fetal development under an in utero hyperandrogenism environment.


Subject(s)
Chromosome Aberrations/embryology , Polycystic Ovary Syndrome/embryology , Polycystic Ovary Syndrome/physiopathology , Adult , Female , Fertilization , Germ Cells/pathology , Humans , Polycystic Ovary Syndrome/complications , Pregnancy
11.
Hum Reprod ; 22(2): 395-400, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17062583

ABSTRACT

BACKGROUND: In 1998, we revealed a sequence departing from prenatal growth restraint in girls and evolving, through precocious pubarche (PP) in mid-childhood, towards anovulatory and hyperinsulinaemic hyperandrogenism. The latter condition fulfilled the criteria for the diagnosis of polycystic ovary syndrome (PCOS), which was then defined independently of the presence of polycystic ovaries (PCOs). Since 2003, the diagnosis of PCOS has been extended by adding PCO as an alternative criterion. We verified longitudinally over 28 +or- 2 years the prevalence of PCO and its potential relationship to growth before birth in a group of post-PP women (n=14, mean age=28 years; body mass index=24.3 kg/m2) belonging to the original cohort of 35 girls in whom the PP-PCOS sequence was described. METHODS: Endocrine-metabolic variables, body composition (by dual-energy X-ray absorptiometry), carotid intima-media thickness (IMT) and ovarian morphology by transvaginal ultrasonography were assessed in all women. RESULTS: Post-PP women with a birthweight (BW) in the lowest quartile, when compared with post-PP women with a higher BW, had smaller ovaries (mean volume=4.0 versus 9.0 ml; P=0.004) and a much lower prevalence of PCO (0 versus 67%; P=0.006). The remaining variables were similar between BW subgroups. CONCLUSIONS: The presence of a PCO morphology in women with a PP history was found to relate to prenatal growth. It would be of interest to verify whether a similar relationship exists in anovulatory and/or hyperandrogenic women without PP history.


Subject(s)
Polycystic Ovary Syndrome/embryology , Puberty, Precocious/physiopathology , Adolescent , Adult , Birth Weight , Carotid Arteries/diagnostic imaging , Child , Female , Humans , Ovary/diagnostic imaging , Ultrasonography, Doppler
12.
Clin Endocrinol (Oxf) ; 65(5): 648-54, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17054468

ABSTRACT

OBJECTIVE: The pathogenesis of human pituitary adenomas remains unclear, but we report a case of FSH-secreting pituitary adenoma whose monohormonal phenotype suggests it was of fetal origin. PATIENTS: A 28-year-old woman presented with abdominal discomfort and irregular menses, enlarged multicystic ovaries and elevated serum oestradiol, with sustained high-normal FSH and low LH levels. MEASUREMENTS: Endocrine studies were performed before and after curative surgery, with assessment of tumour hormone secretion in vitro, and immunostaining of tumour tissue for a series of gonadotrope proteins. RESULTS: Immunocytochemistry showed that tumour cells were monohormonal for FSH. Normal components of gonadotrope signalling pathways were expressed, including oestrogen receptor-alpha, activin receptors, secretogranin-II and chromogranin-A. beta-glycan, the putative inhibin coreceptor, was absent. Tumour culture in vitro confirmed secretion of FSH with minimal LH, that was unsuppressed by oestradiol or inhibin-A. Human fetal pituitary tissue contained FSH-only cells at 18 weeks gestation, whereas normal adult pituitary tissue contained only bihormonal gonadotropes. CONCLUSIONS: We propose that this pituitary adenoma represents an indolent tumour of monohormonal fetal gonadotrope cells that originated early in gestation. Pituitary tumours may therefore arise from abnormal persistence of fetal cell types, with extremely slow growth over many years until reaching a size threshold to generate an endocrine syndrome. Understanding fetal pituitary architecture and function may be more informative for new insights into pituitary tumour pathogenesis than classical theories of cancer biology that invoke unrestrained cell proliferation.


Subject(s)
Adenoma/embryology , Gonadotrophs/metabolism , Pituitary Neoplasms/embryology , Adenoma/complications , Adenoma/metabolism , Adult , Estradiol/blood , Female , Follicle Stimulating Hormone/analysis , Follicle Stimulating Hormone/blood , Follicle Stimulating Hormone/metabolism , Humans , Immunohistochemistry/methods , Immunoradiometric Assay/methods , Luteinizing Hormone/blood , Pituitary Gland, Anterior/embryology , Pituitary Gland, Anterior/metabolism , Pituitary Neoplasms/complications , Pituitary Neoplasms/metabolism , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/embryology , Polycystic Ovary Syndrome/etiology , Tissue Culture Techniques
13.
J Endocrinol ; 174(1): 1-5, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12098657

ABSTRACT

Polycystic ovary syndrome (PCOS) is a common but complex endocrine disorder and is a major cause of anovulation and consequent subfertility. It is also associated with a metabolic disturbance, characterized by hyperinsulinaemia and insulin resistance that carries an increased risk of type 2 diabetes in later life. Despite its prevalence little is known about its aetiology, but there is increasing evidence for an important genetic involvement. On the basis of experimental observations in the prenatally androgenized sheep and rhesus monkey, and supported by data from human studies, we propose that the clinical and biochemical features of PCOS can arise as a consequence of genetically determined hypersecretion of androgens by the ovary during, or very likely long before, puberty. The resulting hyperandrogenism results in 'programming' of the hypothalamic-pituitary unit to favour excess LH secretion, and encourages preferential abdominal adiposity that predisposes to insulin resistance. The severity of hyperinsulinaemia and insulin resistance (which has a profound influence on the phenotype of PCOS) is further influenced by both genetic factors (such as polymorphism in the insulin gene regulatory region) and environmental factors, notably obesity. This hypothesis therefore suggests a unifying, 'linear' model to explain the aetiology of the heterogeneous phenotype.


Subject(s)
Polycystic Ovary Syndrome/etiology , Androgens/biosynthesis , Androgens/physiology , Animals , Female , Humans , Insulin/metabolism , Insulin Resistance/physiology , Luteinizing Hormone/metabolism , Macaca mulatta , Ovarian Diseases/metabolism , Polycystic Ovary Syndrome/embryology , Pregnancy , Prenatal Exposure Delayed Effects , Sheep
16.
Lancet ; 350(9085): 1131-5, 1997 Oct 18.
Article in English | MEDLINE | ID: mdl-9343501

ABSTRACT

BACKGROUND: Polycystic ovaries are a common disorder associated with menstrual irregularities, subfertility, hirsutism, acne, and a range of endocrine abnormalities, including high concentrations of plasma luteinising hormone (LH) and excessive androgen production. The pathophysiology is not understood. We investigated whether the disorder originates during intrauterine life. METHODS: We examined 235 women aged 40-42 years who were born in Sheffield, UK. We related the prevalence of polycystic ovaries and the plasma concentrations of gonadotropin hormones and androgens to the women's body size at birth, and the length of gestation. FINDINGS: 49 (21%) of the women had polycystic ovaries. We defined two groups of women with the disorder, which correspond to the two groups that commonly present clinically. The first group comprised obese women who were androgenised, with higher than normal concentrations of plasma LH and testosterone. These women had above-average birthweight and were born to overweight mothers. The second group comprised women of normal weight who had high plasma LH, but normal testosterone concentrations. These women were born after term (40 weeks' gestation). INTERPRETATION: The two common forms of polycystic ovary syndrome have different origins in intrauterine life. Obese, hirsute women with polycystic ovaries have higher than normal ovarian secretion of androgens that are associated with high birthweight and maternal obesity. Thin women with polycystic ovaries have altered hypothalamic control of LH release resulting from prolonged gestation.


Subject(s)
Birth Weight , Embryonic and Fetal Development , Polycystic Ovary Syndrome/embryology , Adult , Body Weight , Case-Control Studies , England/epidemiology , Female , Gestational Age , Humans , Infant, Newborn , Luteinizing Hormone/blood , Polycystic Ovary Syndrome/epidemiology , Pregnancy , Prevalence , Testosterone/blood
SELECTION OF CITATIONS
SEARCH DETAIL