130 results on '"Groome NP"'
Search Results
2. Demonstration of a non-steroidal, non-inhibin factor in the ovine corpus luteum of pregnancy that reduces pituitary responsiveness to GnRH-induced LH secretion in vitro
- Author
-
Fowler, PA, primary, Groome, NP, additional, and Al-Gubory, KH, additional
- Published
- 2003
- Full Text
- View/download PDF
3. Immunohistochemical localization of inhibin/activin alpha, betaA and betaB subunits and follistatin in bovine oocytes during in vitro maturation and fertilization
- Author
-
Silva, CC, primary, Groome, NP, additional, and Knight, PG, additional
- Published
- 2003
- Full Text
- View/download PDF
4. Differential effects of activin A on basal and gonadotrophin-induced secretion of inhibin A and progesterone by granulosa cells from preovulatory (F1-F3) chicken follicles
- Author
-
Lovell, TM, primary, Gladwell, RT, additional, Groome, NP, additional, and Knight, PG, additional
- Published
- 2002
- Full Text
- View/download PDF
5. Plasma FSH, inhibin A and inhibin isoforms containing pro- and -alphaC during winter anoestrus, spring transition and the breeding season in mares
- Author
-
Watson, ED, primary, Heald, M, additional, Tsigos, A, additional, Leask, R, additional, Steele, M, additional, Groome, NP, additional, and Riley, SC, additional
- Published
- 2002
- Full Text
- View/download PDF
6. Modulatory effects of gonadotrophins and insulin-like growth factor on the secretion of inhibin A and progesterone by granulosa cells from chicken preovulatory (F1-F3) follicles
- Author
-
Lovell, TM, primary, Gladwell, RT, additional, Groome, NP, additional, and Knight, PG, additional
- Published
- 2002
- Full Text
- View/download PDF
7. Presence of exon 5-deleted oestrogen receptor in human breast cancer: functional analysis and clinical significance
- Author
-
Desai, AJ, primary, Luqmani, YA, additional, Walters, JE, additional, Coope, RC, additional, Dagg, B, additional, Gomm, JJ, additional, Pace, PE, additional, Rees, CN, additional, Thirunavukkarasu, V, additional, Shousha, S, additional, Groome, NP, additional, Coombes, R, additional, and Ali, S, additional
- Published
- 1997
- Full Text
- View/download PDF
8. Serum activin A and follistatin concentrations during human pregnancy: a cross-sectional and longitudinal study.
- Author
-
O'Connor, AE, McFarlane, JR, Hayward, S, Yohkaichiya, T, Groome, NP, de Kretser, DM, O'Connor, A E, McFarlane, J R, Groome, N P, and de Kretser, D M
- Subjects
CARRIER proteins ,COMPARATIVE studies ,ENZYME-linked immunosorbent assay ,GESTATIONAL age ,GLYCOPROTEINS ,GROWTH factors ,LONGITUDINAL method ,RESEARCH methodology ,MEDICAL cooperation ,PEPTIDE hormones ,RADIOIMMUNOASSAY ,REFERENCE values ,RESEARCH ,EVALUATION research ,CROSS-sectional method - Abstract
Activin A, a dimer of the βA-subunit of inhibin, has been shown to have multiple biological activities and sites of production. Follistatin is a high-affinity binding protein for activin, which neutralizes its activity. This study provides the first data, using a cross-sectional design, on the measurement of both these proteins in the maternal circulation of a large cohort of women (6-39 weeks of gestation, n = 2-20 women/time point) during normal pregnancies, and confirms that similar patterns are seen in nine women studied longitudinally during pregnancy. The concentrations of total activin A were measured using a specific two-site enzyme-linked immunosorbent assay (ELISA), and a new radioimmunoassay for measuring total follistatin in serum utilizing dissociating reagents to eliminate the interference of activin is described. At 38-39 weeks gestation, both activin A and follistatin concentrations rose to a peak (4.59 ± 0.54 ng/ml and 72.7 ± 3.31 ng/ml, respectively). The activin A and follistatin concentrations were highly correlated both in the cross-sectional study (P < 0.0001) and in individual women in the longitudinal study (P < 0.05-0.0001). Concentrations of follistatin showed a greater increase in the second trimester of pregnancy relative to activin A concentrations. The parallel increase in the secretion of these two proteins throughout pregnancy probably reflects feto-placental secretion. [ABSTRACT FROM PUBLISHER]
- Published
- 1999
- Full Text
- View/download PDF
9. A longitudinal study of maternal serum inhibin-A, inhibin-B, activin-A, activin-AB, pro-alphaC and follistatin during pregnancy.
- Author
-
Fowler, PA, Evans, LE, Groome, NP, Templeton, A, Knight, PG, Fowler, P A, Evans, L W, Groome, N P, and Knight, P G
- Subjects
CARRIER proteins ,COMPARATIVE studies ,GLYCOPROTEINS ,GONADOTROPIN ,LONGITUDINAL method ,RESEARCH methodology ,MEDICAL cooperation ,PEPTIDE hormones ,RESEARCH ,STEROIDS ,TIME ,EVALUATION research - Abstract
Maternal serum concentrations of inhibin-A, inhibin-B, activin-A, activin-AB, pro-alphaC-related inhibin forms, total follistatin, steroids and gonadotrophins were measured longitudinally in six normal singleton pregnancies. Maternal venous blood was collected randomly during a spontaneous follicular phase prior to donor insemination, at 5, 7, 9, 11, 16, 20, 24, 28, 32 and 36 weeks after the first missed menses and in the early puerperium. Steroid and gonadotrophin profiles conformed to previous reports. While at week 5 of gestation inhibin-A, activin-A and follistatin concentrations were similar to those at the follicular phase, all three increased progressively (P < 0.001) to maximal concentrations in week 36: approximately 48-fold (3740 +/- 1349 ng inhibin-A/ml), approximately 22-fold (6109 +/- 1443 ng activin-A/ml) and approximately 10-fold (3563 +/- 418 ng follistatin/ml) higher. Pro-alphaC concentrations reached a maximum in weeks 5 (approximately 5-fold, P < 0.001) and 36 (1027 +/- 174 pg/ml, P < 0.01). Inhibin-B (71 +/- 23 pg/ml prior to pregnancy) was undetectable (<12 pg/ml) between week 5-16 of gestation but increased slightly in the third trimester (26 +/- 7 pg/ml in week 36). Activin-AB was undetectable throughout pregnancy. Post-partum concentrations of inhibin-A (41 +/- 12 ng/ml), inhibin-B (<12 pg/ml), activin-A (950 +/- 149 pg/ml), pro-alphaC (128 +/- 22 pg/ml) and follistatin (990 +/- 79 ng/ml) were substantially lower than at week 36 of gestation. The activin-A:follistatin ratio increased from 0.5 in week 5 to 1.8 in week 36, suggesting that more free activin-A is available in the maternal circulation during late pregnancy. [ABSTRACT FROM AUTHOR]
- Published
- 1998
- Full Text
- View/download PDF
10. Follistatin and activin A production by the male reproductive tract.
- Author
-
Anderson, RA, Evans, LW, Irvine, DS, McIntyre, MA, Groome, NP, Riley, SC, Anderson, R A, Evans, L W, Irvine, D S, McIntyre, M A, Groome, N P, and Riley, S C
- Subjects
CARRIER proteins ,MALE reproductive organs ,GLYCOPROTEINS ,IMMUNOHISTOCHEMISTRY ,PEPTIDE hormones ,SEMEN ,VASECTOMY - Abstract
Follistatin is a binding protein for the activin and inhibin family of hormones, regulating their biological activity. In the male reproductive tract, the interaction of these factors is likely to be involved in the regulation of the proliferation of several cell types. We have investigated the presence of follistatin and activin A in seminal plasma using specific immunoassays and have localized follistatin and activin/inhibin subunits in the adult human testis, prostate and seminal vesicle to establish their likely sources. High concentrations of immunoreactive follistatin were present in seminal plasma in normal men (mean 97.9 ng/ml; 1.43 ng/ml in peripheral plasma) and were similar in men with oligo/azoospermia and following vasectomy. Follistatin immunoreactivity was localized to both Leydig and Sertoli cells of the testis, and to epithelial cells of the prostate gland and seminal vesicle, which are likely to be the predominant sources of the hormone in seminal plasma. Activin A was also present in seminal plasma in normal men but was undetectable following vasectomy, thus deriving from the testis. Consistent with this finding, the βA-subunit was immunolocalized in Sertoli and Leydig cells but was not present in seminal vesicle or prostate gland. The functional significance of the high concentrations of follistatin secreted into seminal plasma by the prostate gland and/or seminal vesicle is uncertain, but they may regulate the biological activity of testis-derived activin A and inhibin B. [ABSTRACT FROM PUBLISHER]
- Published
- 1998
- Full Text
- View/download PDF
11. Follistatin and activin A in extra-embryonic coelomic and amniotic fluids and maternal serum in early pregnancy.
- Author
-
Riley, SC, Balfour, C, Wathen, NC, Chard, T, Evans, LW, Groome, NP, Wallace, EM, Riley, S C, Wathen, N C, Evans, L W, Groome, N P, and Wallace, E M
- Abstract
Follistatin is a specific binding protein which controls bioavailability of activins and inhibins which have an important role in fetal development. In the first trimester of pregnancy bioactive dimeric inhibins are found at high concentrations in the extra-embryonic coelomic fluid, but the distribution of follistatin and activins is not known. We have used recently developed immunoassays for follistatin, activin A and activin AB to determine their presence in the intrauterine compartments during early pregnancy. Follistatin was present in highest concentrations in the extra-embryonic coelomic fluid (11.72 ± 1.70 ng/ml; median ± SEM), with less in maternal serum (6.35 ± 4.58) and lowest amounts in amniotic fluid (0.97 ± 0.52). Follistatin concentrations in extra- embryonic coelomic fluid were highly correlated with both dimeric inhibin isoforms. Activin A was present in only barely detectable amounts in some samples of extra-embryonic coelomic fluid (41% of samples) and maternal serum (26%) and was undetectable in all amniotic fluid samples. Activin AB was undetectable in all compartments. The presence of follistatin in the amniotic and extra-embryonic coelomic fluids may regulate the availability of bioactive activins and inhibins which are released into the intrauterine compartments during the development of the fetus and placenta in early pregnancy. [ABSTRACT FROM PUBLISHER]
- Published
- 1998
- Full Text
- View/download PDF
12. Inhibin B in seminal plasma: testicular origin and relationship to spermatogenesis.
- Author
-
Anderson, RA, Irvine, DS, Balfour, C, Groome, NP, Riley, SC, Anderson, R A, Irvine, D S, Groome, N P, and Riley, S C
- Abstract
In men, inhibin B is the circulating isoform involved in the regulation of follicle stimulating hormone (FSH) secretion. Within the testis, inhibin B may have a role in Sertoli and germ cell interactions, thus secretion into seminal plasma may reflect seminiferous tubule function. Using specific immunoassays, inhibin B was present in seminal plasma in fertile men (n = 105) and in unselected men attending an infertility clinic (n = 174) with a wide range in concentration from undetectable (<15 pg/ml) up to 54,100 pg/ml (geometric mean 280 pg/ml). There was a highly significant correlation between seminal plasma inhibin B concentration and sperm concentration (r = 0.46, P < 0.001), but no correlation with percentages of spermatozoa with progressive motility or normal morphology. Inhibin A and isoforms containing pro and alphaC immunoreactivity were not detectable. In post-vasectomy seminal plasma samples (18 of 20) inhibin B was undetectable, indicating that the testis is the predominant source. In unselected men attending an infertility clinic, inhibin B was undetectable in 17% (present in remainder; maximum concentration 26,200 pg/ml; mean 263 pg/ml), with a highly significant correlation between seminal plasma inhibin B and sperm concentration (r = 0.55, P < 0.0001). In men with oligo/ azoospermia (sperm concentration <20 x 10(6)/ml), seminal plasma inhibin B concentrations were lower in those with elevated plasma FSH concentrations (mean values 42 and 205 pg/ml, P < 0.05). Inhibin alpha and betaB subunits were localized predominantly in Sertoli and Leydig cells, using immunohistochemistry. We conclude that inhibin B of testicular origin is present in normal human seminal plasma, but with a very wide range in concentration, and may reflect the functional state of the seminiferous epithelium. [ABSTRACT FROM AUTHOR]
- Published
- 1998
- Full Text
- View/download PDF
13. Evaluation of serum inhibin A as a surveillance marker after conservative management of tubal pregnancy.
- Author
-
D'Antona, D, Mamers, PM, Lowe, PJM, Balazs, N, Groome, NP, Wallace, EM, Mamers, P M, Lowe, P J, Groome, N P, and Wallace, E M
- Abstract
Tubal pregnancy is now commonly managed by laparoscopic salpingostomy or systemic methotrexate. A disadvantage of such conservative management is the need for appropriate follow-up, with serial measurement of serum concentrations of human chorionic gonadotrophin (HCG), to exclude persistent ectopic pregnancy (PEP). Concentrations of inhibin A, also a placental product, are significantly increased during pregnancy and the half-life of inhibin A is significantly shorter than that of HCG. To assess the suitability of inhibin A as a marker of PEP, we studied 16 women who had undergone surgery for a tubal pregnancy, measuring HCG and inhibin during follow-up. The mean ± SEM time taken to achieve non-pregnant concentrations of inhibin A was significantly shorter than for HCG (4.2 ± 0.8 days versus 21.6 ± 4.4 days respectively; P < 0.001 Wilcoxon signed rank test). However, in all women the inhibin A concentration increased rapidly after reaching a nadir, reflecting the return of ovarian function, complicating the interpretation of results. In four women inhibin A was almost undetectable preoperatively, while the corresponding HCG concentration was high. These data suggest that inhibin A will not be a useful marker for PEP but that it may provide a more accurate preoperative assessment of trophoblast viability than HCG, thereby improving management. [ABSTRACT FROM PUBLISHER]
- Published
- 1998
- Full Text
- View/download PDF
14. The anti-Müllerian hormone prodomain is displaced from the hormone/prodomain complex upon bivalent binding to the hormone receptor.
- Author
-
Cate RL, di Clemente N, Racine C, Groome NP, Pepinsky RB, and Whitty A
- Subjects
- Ligands, Protein Domains, Receptors, Transforming Growth Factor beta metabolism, Transforming Growth Factor beta metabolism, Anti-Mullerian Hormone metabolism, Peptide Hormones metabolism
- Abstract
Noncovalent complexes of transforming growth factor-β family growth/differentiation factors with their prodomains are classified as latent or active, depending on whether the complexes can bind their respective receptors. For the anti-Müllerian hormone (AMH), the hormone-prodomain complex is active, and the prodomain is displaced upon binding to its type II receptor, AMH receptor type-2 (AMHR2), on the cell surface. However, the mechanism by which this displacement occurs is unclear. Here, we used ELISA assays to measure the dependence of prodomain displacement on AMH concentration and analyzed results with respect to the behavior expected for reversible binding in combination with ligand-induced receptor dimerization. We found that, in solution, the prodomain has a high affinity for the growth factor (GF) (K
d = 0.4 pM). Binding of the AMH complex to a single AMHR2 molecule does not affect this Kd and does not induce prodomain displacement, indicating that the receptor binding site in the AMH complex is fully accessible to AMHR2. However, recruitment of a second AMHR2 molecule to bind the ligand bivalently leads to a 1000-fold increase in the Kd for the AMH complex, resulting in rapid release of the prodomain. Displacement occurs only if the AMHR2 is presented on a surface, indicating that prodomain displacement is caused by a conformational change in the GF induced by bivalent binding to AMHR2. In addition, we demonstrate that the bone morphogenetic protein 7 prodomain is displaced from the complex with its GF by a similar process, suggesting that this may represent a general mechanism for receptor-mediated prodomain displacement in this ligand family., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
15. Investigation of activin A in inflammatory responses of the testis and its role in the development of testicular fibrosis.
- Author
-
Kauerhof AC, Nicolas N, Bhushan S, Wahle E, Loveland KA, Fietz D, Bergmann M, Groome NP, Kliesch S, Schuppe HC, Pilatz A, Meinhardt A, Hedger MP, and Fijak M
- Subjects
- Animals, Collagen metabolism, Fibronectins metabolism, Fibrosis metabolism, Fibrosis pathology, Follistatin genetics, Follistatin metabolism, Humans, Infertility, Male pathology, Male, Mice, Orchitis pathology, Spermatogenesis, Testis pathology, Activins metabolism, Infertility, Male metabolism, Orchitis metabolism, Testis metabolism
- Abstract
Study Question: Does activin A contribute to testicular fibrosis under inflammatory conditions?, Summary Answer: Our results show that activin A and key fibrotic proteins are increased in human testicular biopsies with leukocytic infiltrates and impaired spermatogenesis and in murine experimental autoimmune orchitis (EAO) and that activin A stimulates fibrotic responses in peritubular cells (PTCs) and NIH 3T3 fibroblasts., What Is Known Already: Fibrosis is a feature of EAO. Activin A, a regulator of fibrosis, was increased in testes of mice with EAO and its expression correlated with severity of the disease., Study Design, Size, Duration: This is a cross-sectional and longitudinal study of adult mice immunized with testicular homogenate (TH) in adjuvant to induce EAO, collected at 30 (n = 6), 50 (n = 6) and 80 (n = 5) days after first immunization. Age-matched mice injected with adjuvant alone (n = 14) and untreated mice (n = 15) were included as controls. TH-immunized mice with elevated endogenous follistatin, injected with a non-replicative recombinant adeno-associated viral vector carrying a gene cassette of follistatin (rAAV-FST315; n = 3) or vector with an empty cassette (empty vector controls; n = 2) 30 days prior to the first immunization, as well as appropriate adjuvant (n = 2) and untreated (n = 2) controls, were also examined.Human testicular biopsies showing focal inflammatory lesions associated with impaired spermatogenesis (n = 7) were included. Biopsies showing intact spermatogenesis without inflammation, from obstructive azoospermia patients, served as controls (n = 7).Mouse primary PTC and NIH 3T3 fibroblasts were stimulated with activin A and follistatin 288 (FST288) to investigate the effect of activin A on the expression of fibrotic markers. Production of activin A by mouse primary Sertoli cells (SCs) was also investigated., Participants/materials, Setting, Methods: Testicular RNA and protein extracts collected from mice at days 30, 50 and 80 after first immunization were used for analysis of fibrotic marker genes and proteins, respectively. Total collagen was assessed by hydroxyproline assay and fibronectin; collagen I, III and IV, α-smooth muscle actin (α-SMA) expression and phosphorylation of suppressor of mothers against decapentaplegic (SMAD) family member 2 were measured by western blot. Immunofluorescence was used to detect fibronectin. Fibronectin (Fn), αSMA (Acta2), collagen I (Col1a2), III (Col3a1) and IV (Col4a1) mRNA in PTC and NIH 3T3 cells treated with activin A and/or FST288 were measured by quantitative RT-PCR (qRT-PCR). Activin A in SC following tumour necrosis factor (TNF) or FST288 stimulation was measured by ELISA. Human testicular biopsies were analysed by qRT-PCR for PTPRC (CD45) and activin A (INHBA), hydroxyproline assay and immunofluorescence., Main Results and the Role of Chance: Production of activin A by SC was stimulated by 25 and 50 ng/ml TNF (P < 0.01, P < 0.001, respectively) as compared to untreated cells. INHBA mRNA was increased in human testicular biopsies with leukocytic infiltrates and impaired spermatogenesis, compared with control biopsies (P < 0.05), accompanied by increased total collagen (P < 0.01) and fibronectin deposition. Total testicular collagen (P < 0.0001) and fibronectin protein expression (P < 0.05) were also increased in EAO, and fibronectin expression was correlated with the severity of the disease (r = 0.9028). In animals pre-treated with rAAV-FST315 prior to immunization with TH, protein expression of fibronectin was comparable to control. Stimulation of PTC and NIH 3T3 cells with activin A increased fibronectin mRNA (P < 0.05) and the production of collagen I (P < 0.001; P < 0.01) and fibronectin (P < 0.05). Moreover, activin A also increased collagen IV mRNA (P < 0.05) in PTC, while αSMA mRNA (P < 0.01) and protein (P < 0.0001) were significantly increased by activin A in NIH 3T3 cells., Large Scale Data: N/A., Limitations, Reasons for Caution: A limited number of human testicular specimens was available for the study. Part of the study was performed in vitro, including NIH 3T3 cells as a surrogate for testicular fibroblasts., Wider Implications of the Findings: Resident fibroblasts and PTC may contribute to the progression of testicular fibrosis following inflammation, and activin A is implicated as a key mediator of this process., Study Funding/competing Interest(s): This work was supported by the National Health and Medical Research Council of Australia, the Victorian Government's Operational Infrastructure Support Program and the International Research Training Group between Justus Liebig University (Giessen) and Monash University (Melbourne) (GRK 1871/1-2) on `Molecular pathogenesis on male reproductive disorders' funded by the Deutsche Forschungsgemeinschaft and Monash University. The authors declare no competing financial interests., (© The Author(s) 2019. Published by Oxford University Press on behalf of the European Society of Human Reproduction and Embryology. All rights reserved. For permissions, please e-mail: journals.permission@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
16. Weighting of orthostatic intolerance time measurements with standing difficulty score stratifies ME/CFS symptom severity and analyte detection.
- Author
-
Richardson AM, Lewis DP, Kita B, Ludlow H, Groome NP, Hedger MP, de Kretser DM, and Lidbury BA
- Subjects
- Activins blood, Adolescent, Adult, Aged, Biomarkers blood, Biomarkers urine, Case-Control Studies, Cohort Studies, Fatigue Syndrome, Chronic blood, Fatigue Syndrome, Chronic pathology, Female, Humans, Male, Middle Aged, Orthostatic Intolerance blood, Orthostatic Intolerance pathology, Time Factors, Young Adult, Fatigue Syndrome, Chronic complications, Fatigue Syndrome, Chronic physiopathology, Orthostatic Intolerance complications, Orthostatic Intolerance physiopathology, Posture, Severity of Illness Index
- Abstract
Background: Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is clinically defined and characterised by persistent disabling tiredness and exertional malaise, leading to functional impairment., Methods: This study introduces the weighted standing time (WST) as a proxy for ME/CFS severity, and investigates its behaviour in an Australian cohort. WST was calculated from standing time and subjective standing difficulty data, collected via orthostatic intolerance assessments. The distribution of WST for healthy controls and ME/CFS patients was correlated with the clinical criteria, as well as pathology and cytokine markers. Included in the WST cytokine analyses were activins A and B, cytokines causally linked to inflammation, and previously demonstrated to separate ME/CFS from healthy controls. Forty-five ME/CFS patients were recruited from the CFS Discovery Clinic (Victoria) between 2011 and 2013. Seventeen healthy controls were recruited concurrently and identically assessed., Results: WST distribution was significantly different between ME/CFS participants and controls, with six diagnostic criteria, five analytes and one cytokine also significantly different when comparing severity via WST. On direct comparison of ME/CFS to study controls, only serum activin B was significantly elevated, with no significant variation observed for a broad range of serum and urine markers, or other serum cytokines., Conclusions: The enhanced understanding of standing test behaviour to reflect orthostatic intolerance as a ME/CFS symptom, and the subsequent calculation of WST, will encourage the greater implementation of this simple test as a measure of ME/CFS diagnosis, and symptom severity, to the benefit of improved diagnosis and guidance for potential treatments.
- Published
- 2018
- Full Text
- View/download PDF
17. Nuclear and cytoplasmic expression of ERbeta1, ERbeta2, and ERbeta5 identifies distinct prognostic outcome for breast cancer patients.
- Author
-
Shaaban AM, Green AR, Karthik S, Alizadeh Y, Hughes TA, Harkins L, Ellis IO, Robertson JF, Paish EC, Saunders PT, Groome NP, and Speirs V
- Subjects
- BRCA1 Protein biosynthesis, Blotting, Western, Breast Neoplasms mortality, Disease-Free Survival, Female, Humans, Immunohistochemistry, Kaplan-Meier Estimate, Prognosis, Protein Isoforms biosynthesis, Receptors, Androgen biosynthesis, Receptors, Progesterone biosynthesis, Tissue Array Analysis, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Nucleus metabolism, Cytoplasm metabolism, Estrogen Receptor beta biosynthesis
- Abstract
Purpose: Previous conflicting results about the prognostic significance of estrogen receptor (ER)-beta in breast cancer may be explained by contribution of isoforms, of which five exist. Our aim was to elucidate the prognostic significance of ERbeta1, ERbeta2, and ERbeta5 by immunohistochemistry in a large cohort of breast carcinomas with long-term follow-up., Experimental Design: Tissue microarrays were stained with ERbeta1, ERbeta2, and ERbeta5 antibodies and scored as percentage of positive tumor cells and using the Allred system. Nuclear and cytoplasmic staining was evaluated and correlated with histopathologic characteristics, overall survival (OS), and disease-free survival (DFS)., Results: Nuclear ERbeta2 and ERbeta5, but not ERbeta1, significantly correlated with OS (P = 0.006, P = 0.039, and P = 0.099, respectively), and ERbeta2 additionally with DFS (P = 0.013). ERbeta2 also predicted response to endocrine therapy (P = 0.036); correlated positively with ERalpha, progesterone receptor, androgen receptor, and BRCA1; and correlated inversely with metastasis and vascular invasion. Tumors coexpressing ERbeta2 and ERalpha had better OS and DFS. Cytoplasmic ERbeta2 expression, alone or combined with nuclear staining, predicted significantly worse OS. Notably, patients with only cytoplasmic ERbeta2 expression had significantly worse outcome (P = 0.0014)., Conclusions: This is the first study elucidating the prognostic role of ERbeta1, ERbeta2, and ERbeta5 in a large breast cancer series. ERbeta2 is a powerful prognostic indicator in breast cancer, but nuclear and cytoplasmic expression differentially affect outcome. Measuring these in clinical breast cancer could provide a more comprehensive picture of patient outcome, complementing ERalpha.
- Published
- 2008
- Full Text
- View/download PDF
18. Serum levels of retinol-binding protein 4 and adiponectin in women with polycystic ovary syndrome: associations with visceral fat but no evidence for fat mass-independent effects on pathogenesis in this condition.
- Author
-
Barber TM, Hazell M, Christodoulides C, Golding SJ, Alvey C, Burling K, Vidal-Puig A, Groome NP, Wass JA, Franks S, and McCarthy MI
- Subjects
- Adiponectin physiology, Adult, Female, Humans, Insulin Resistance, Polycystic Ovary Syndrome blood, Retinol-Binding Proteins, Plasma physiology, Testosterone blood, Adiponectin blood, Intra-Abdominal Fat physiology, Polycystic Ovary Syndrome etiology, Retinol-Binding Proteins, Plasma analysis
- Abstract
Context: Insulin resistance, which associates with levels of retinol-binding protein 4 (RBP4) and adiponectin, is implicated in the development of polycystic ovary syndrome (PCOS)., Objective: The objective of the study was to explore the potential contribution of RBP4 and adiponectin in the etiology of PCOS and their relationships with specific fat depot measurements., Design: This was a cross-sectional study., Setting and Participants: Serum RBP4 and adiponectin levels were compared between 50 PCOS cases and 28 female controls (including 22 body mass index/fat mass-matched pairs) and correlated with specific fat depot (including visceral) axial magnetic resonance imaging cross-sectional area measurements. All subjects were of U.K. British/Irish origin., Main Outcome Measure(s): Serum levels of RBP4 (automated immunonephelometric assay) and adiponectin [immunoassay: total and high molecular weight (HMW)]. Data are reported as geometric mean (sd, range) and optionally adjusted for fat mass and age., Results: Between the 50 PCOS cases and 28 controls, serum RBP4 levels were indistinguishable [39.0 microg/ml (31.0, 49.0) vs. 41.6 microg/ml (32.7, 52.9), respectively, unadjusted P = 0.24; adjusted P = 0.55]. Total (and HMW) adiponectin levels were lower in PCOS cases [total adiponectin 19.9 microg/ml (14.2, 27.8) vs. 25.8 microg/ml (17.7, 37.7), respectively, unadjusted P = 2.4 x 10(-3); adjusted P = 0.10]. For the paired-sample analyzes, there were no differences in RBP4 (P = 0.09), total adiponectin (P = 0.06), HMW adiponectin (P =0.19), or HMW to total adiponectin ratio (P = 0.98). In PCOS cases, L4-visceral fat area was associated positively with RBP4 (r(2) = 0.34, P = 0.01) and negatively with HMW to total adiponectin ratio (r(2) = -0.44, P = 1.3 x 10(-3)). Controls showed similar relationships., Conclusions: Although associated with visceral fat, serum RBP4 and adiponectin levels do not play important, fat-mass-independent primary roles in the development of PCOS.
- Published
- 2008
- Full Text
- View/download PDF
19. Evaluation of the relationship between follicular fluid oxidative stress, ovarian hormones, and response to gonadotropin stimulation.
- Author
-
Appasamy M, Jauniaux E, Serhal P, Al-Qahtani A, Groome NP, and Muttukrishna S
- Subjects
- Activins metabolism, Adult, Anti-Mullerian Hormone metabolism, Antioxidants metabolism, Cross-Sectional Studies, Estradiol metabolism, Female, Fertilization in Vitro, Gonadal Hormones blood, Humans, Infertility, Female etiology, Infertility, Female metabolism, Infertility, Female physiopathology, Inhibins metabolism, Pregnancy, Pregnancy Outcome, Prospective Studies, Treatment Outcome, Fertility Agents, Female administration & dosage, Follicular Fluid metabolism, Gonadal Hormones metabolism, Gonadotropins administration & dosage, Infertility, Female therapy, Ovulation drug effects, Ovulation Induction methods, Oxidative Stress
- Abstract
Objective: To investigate the relationship between oxidative stress and the underlying causes of infertility, preovulatory ovarian hormones, and ovarian response to gonadotropin stimulation in patients undergoing assisted reproductive techniques., Design: Prospective, cross-sectional study., Setting: Assisted conception unit, university hospital., Patient(s): One hundred thirty women presenting with infertility, of the following types: male factor (n = 56), unexplained (n = 36), tubal factor (n = 16), polycystic ovary syndrome (n = 15), and endometriosis (n = 7)., Intervention(s): Follicular fluid (FF) and peripheral blood samples were collected at oocyte retrieval., Main Outcome Measure(s): Blood and FF samples were analyzed for inhibin A, inhibin B, activin A, anti-Müllerian hormone, and E(2) by using ELISA. Total antioxidant capacity (TAC) was measured in plasma and FF by using a calorimetric microplate assay., Result(s): There was no significant relationship between plasma or FF TAC and the underlying etiology of infertility. There was a statistically significant positive association between FF E(2) levels and TAC (r = 0.26). Higher antral follicle count, delta E(2) (day 3 E(2) minus day 2 E(2)), preovulatory serum anti-Müllerian hormone, inhibin B, and E(2) were associated with good ovarian response, whereas higher FF E(2) was associated with a statistically significant poor response. No significant direct relationship was observed between TAC and ovarian response as well as between TAC or any of the parameters measured and pregnancy outcome., Conclusion(s): Oxidative stress has an impact on the production of granulosa cell steroid hormones, in particular E(2), which is an important predictor of ovarian response. The positive association between FF E(2) and total antioxidant capacity suggests that E(2) may play a role in the ovarian antioxidant-oxidant balance.
- Published
- 2008
- Full Text
- View/download PDF
20. Human fetal testis Leydig cell disruption by exposure to the pesticide dieldrin at low concentrations.
- Author
-
Fowler PA, Abramovich DR, Haites NE, Cash P, Groome NP, Al-Qahtani A, Murray TJ, and Lea RG
- Subjects
- Cells, Cultured, Female, Gonadotropins metabolism, Humans, Immunohistochemistry methods, Male, Phosphoproteins biosynthesis, Pregnancy, Pregnancy Trimester, Second, Proteome, Testosterone biosynthesis, Dieldrin toxicity, Leydig Cells cytology, Leydig Cells drug effects, Pesticides toxicity, Testis drug effects, Testis embryology
- Abstract
Background: Declining human reproductive health over the last 60 years has been proposed to be due to effects of environmental chemicals, especially endocrine disrupting compounds, on fetal development. We investigated whether a model pesticide, dieldrin, at concentrations within both maternal circulation and environmental ranges (1 pmol/l = 0.0004 p.p.b. = 380.9 pg/l), could disrupt the human fetal testis., Methods: Human fetal testes were collected during the second trimester, a critical period of male sexual differentiation (development and masculinization). Testis explants were cultured for 24 h in the presence and absence of LH (10-1000 IU LH/l) and dieldrin (1 pmol and 1 nmol/l). Endocrine, immunohistological and proteome characteristics of the tissues were investigated., Results: Exposure to dieldrin reduced LH-induced testosterone secretion (P < 0.05) and tissue protein concentrations of LH receptor and steroid acute regulatory protein (P < 0.05). Dieldrin altered proteins associated with cancer, apoptosis, transcription and development. Wnt-2b was reduced 3-fold and immunolocalized to Leydig and Sertoli cells. Dieldrin also reversed some LH-induced changes in protein expression, supporting the conclusion that Leydig cell function is at risk from environmental chemicals., Conclusions: Our findings indicate that exposure to very low, biologically relevant, concentrations of environmental chemicals could affect the fetal human Leydig cell, reducing testosterone secretion and potentially leading to subtle dysregulation of reproductive development and adult fecundity.
- Published
- 2007
- Full Text
- View/download PDF
21. Rising follicle-stimulating hormone levels with age accelerate female reproductive failure.
- Author
-
McTavish KJ, Jimenez M, Walters KA, Spaliviero J, Groome NP, Themmen AP, Visser JA, Handelsman DJ, and Allan CM
- Subjects
- Animals, Female, Follicle Stimulating Hormone blood, Humans, Insulin genetics, Luteinizing Hormone blood, Mice, Mice, Transgenic, Ovarian Follicle drug effects, Ovarian Follicle physiology, Ovary physiology, Polymerase Chain Reaction, Promoter Regions, Genetic, Rats, Aging physiology, Fertility physiology, Follicle Stimulating Hormone physiology, Reproduction physiology
- Abstract
Rising serum FSH levels is one of the earliest signs of human female reproductive aging. Whether or not elevated FSH remains a passive reflection of a diminishing ovarian follicle pool or actively contributes to declining female fertility with age has not been established. We therefore investigated female reproduction in mice expressing progressively rising serum levels of transgenic human FSH (Tg-FSH, 2.5-10 IU/liter) independently of follicle depletion. We show that serum LH and estradiol levels and uterine size remained normal in Tg-FSH females, whereas ovarian weight and corpora lutea number were significantly increased up to 1.3- and 5-fold, respectively. Furthermore, the monotrophic FSH rise produced a striking biphasic effect on female fertility. Tg-FSH females less than 22 wk old delivered increased litter sizes, then beyond 23 wk, litter sizes decreased rapidly culminating in premature infertility despite continued ovary follicle development, and increased ovulation and uterine embryo implantation sites as well as normal serum levels of anti-Mullerian hormone, a marker of ovarian follicle reserve. We found that rising circulating Tg-FSH produced premature infertility by increasing embryo-fetal resorption and parturition failure with age. Thus, our Tg-FSH mice present a novel paradigm to investigate selective contributions of elevated FSH to age-related female infertility, which revealed that rising FSH levels, despite no exhaustion of ovarian reserve, actively accelerates female reproductive aging primarily by postimplantation reduction of embryo-fetal survival.
- Published
- 2007
- Full Text
- View/download PDF
22. Predictors of ovarian reserve in young women with breast cancer.
- Author
-
Lutchman Singh K, Muttukrishna S, Stein RC, McGarrigle HH, Patel A, Parikh B, Groome NP, Davies MC, and Chatterjee R
- Subjects
- Adult, Antineoplastic Agents adverse effects, Female, Humans, Ovarian Follicle drug effects, Ovarian Follicle pathology, Ovary drug effects, Breast Neoplasms drug therapy, Ovarian Function Tests, Ovary physiopathology
- Abstract
Ovarian reserve can be diminished following treatment for breast cancer. This study evaluated biochemical and biophysical parameters of ovarian reserve in these patients. Biochemical and biophysical tests of ovarian reserve were performed simultaneously in young (age 22-42 years), regularly menstruating women with breast cancer (n=22) and age-matched controls (n=24). All tests were performed before (baseline) and after transient ovarian stimulation in the early follicular phase. Patients were recruited both before and after completion of chemotherapy, with some patients being followed up prospectively. Serum samples were analysed for follicle-stimulating hormone (FSH), luteinising hormone (LH), oestradiol (E(2)), inhibins A and B, and antimullerian hormone (AMH). Biophysical (ultrasound) tests included ovarian volume, antral follicle count (AFC), ovarian stromal blood flow and uterine dimensions. Significant differences were revealed (when compared with the controls) for basal FSH (11.32+/-1.48 vs 6.62+/-0.42 mIU ml(-1), P<0.001), basal AMH (0.95+/-0.34 vs 7.89+/-1.62 ng ml(-1), P<0.001) and basal inhibin B (19.24+/-4.56 vs 83.61+/-13.45 pg ml(-1), P<0.001). Following transient ovarian stimulation, there were significant differences in the increment change (Delta) for inhibin B (3.02+/-2.3 vs 96.82+/-16.38 pg ml(-1), P<0.001) and E(2) (107.8+/-23.95 vs 283.2+/-40.34 pg ml(-1), P<0.01). AFC was the only biophysical parameter that was significantly different between patients and the controls (7.80+/-0.85 vs 16.77+/-1.11, P<0.001). Basal and stimulated biochemical (serum AMH, FSH, inhibin B and E(2)) and biophysical (AFC) tests may be potential markers of ovarian reserve in young women with breast cancer.
- Published
- 2007
- Full Text
- View/download PDF
23. The effects of chemotherapy and long-term gonadotrophin suppression on the ovarian reserve in premenopausal women with breast cancer.
- Author
-
Anderson RA, Themmen AP, Al-Qahtani A, Groome NP, and Cameron DA
- Subjects
- Adult, Antineoplastic Combined Chemotherapy Protocols adverse effects, Breast Neoplasms blood, Breast Neoplasms diagnostic imaging, Breast Neoplasms surgery, Female, Follicle Stimulating Hormone blood, Follicular Phase drug effects, Follicular Phase physiology, Humans, Luteinizing Hormone blood, Menstrual Cycle, Ovary diagnostic imaging, Ovary drug effects, Ovary physiopathology, Premenopause, Ultrasonography, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms drug therapy, Gonadotropins antagonists & inhibitors, Ovary physiology
- Abstract
Background: Reproductive function following cancer treatment is of increasing importance with improving survival rates. We therefore assessed the markers of the ovarian reserve in premenopausal women, to investigate and compare the effects of chemotherapy and long-term gonadotrophin withdrawal on ovarian function., Methods: Fifty premenopausal (age range 28-52 years) women with early breast cancer were recruited. Serum hormone and ovarian ultrasound measurements were taken before treatment and at intervals up to 1 year during chemotherapy or gonadotrophin suppressive therapy., Results: Pretreatment samples indicated a fall in anti-Müllerian hormone (AMH) concentration with age before changes in other hormone concentrations. AMH concentration showed a rapid and marked fall during chemotherapy, with undetectable concentrations in many women (P<0.0001). Inhibin B concentration showed a lesser fall (P<0.0001), whereas estradiol (E2) concentrations were maintained. Both antral follicle count (AFC) and ovarian volume fell (P<0.0001 and P<0.05 respectively). Regimens containing taxanes in addition to cyclophosphamide showed increased gonadotoxicity. Gonadotrophin suppression resulted in expected falls in E2 (P<0.05) and inhibin B (P<0.001) levels, but also resulted in a delayed fall in AMH level after 6 months (P<0.0001)., Conclusions: These data confirm the value of AMH concentration as an early indicator of ovarian ageing including assessment of chemotherapy-induced ovarian follicle loss. FSH and AMH concentration measurements may be useful for the comparison of ovarian toxicity of different chemotherapy regimens.
- Published
- 2006
- Full Text
- View/download PDF
24. Editorial: Anti-Müllerian hormone: Cinderella finds new admirers.
- Author
-
Al-Qahtani A and Groome NP
- Subjects
- Anti-Mullerian Hormone, Female, Humans, Glycoproteins blood, Menstrual Cycle blood, Testicular Hormones blood
- Published
- 2006
- Full Text
- View/download PDF
25. Serum anti-mullerian hormone levels reflect the size of the primordial follicle pool in mice.
- Author
-
Kevenaar ME, Meerasahib MF, Kramer P, van de Lang-Born BM, de Jong FH, Groome NP, Themmen AP, and Visser JA
- Subjects
- Aging, Animals, Anti-Mullerian Hormone, Female, Follicle Stimulating Hormone blood, Humans, Immunohistochemistry methods, Inhibins blood, Mice, Mice, Inbred C57BL, Sensitivity and Specificity, Enzyme-Linked Immunosorbent Assay methods, Glycoproteins blood, Granulosa Cells cytology, Ovarian Follicle cytology, Ovarian Follicle pathology, Testicular Hormones blood
- Abstract
Reproductive aging is the decline of female fertility with age. It is caused by the decrease in the number of growing follicles, resulting from primordial follicle pool depletion. Recently, we have shown that anti-Müllerian hormone (AMH) is produced by growing follicles, and studies in women indicate that serum AMH levels decrease with age and correlate with antral follicle count. However, whether serum AMH levels correlate directly with the size of the primordial follicle pool cannot be determined in women. In this work, we describe studies in mice in which we determined the dynamics of ovarian follicles during aging. Furthermore, we describe the development of a mouse AMH ELISA, allowing us to measure AMH levels in mice, for the first time. We observed that serum AMH levels decline with increasing age, whereas expression of AMH in individual growing follicles, studied by immunohistochemistry, did not change with age. Thus, the decline in serum AMH correlates directly with the decline in the number of growing follicles (r = 0.86, P < 0.0001). We observed that the number of growing follicles correlated with the number of primordial follicles (r = 0.93, P < 0.0001). Similarly, we found a strong correlation between AMH levels and number of primordial follicles (r = 0.83, P < 0.0001). In conclusion, serum AMH levels reflect the size of the primordial follicle pool in aging mice. Therefore, AMH is an excellent marker to assess the quantitative aspect of ovarian reserve, which may be useful for women at risk for early ovarian aging such as survivors of childhood cancers.
- Published
- 2006
- Full Text
- View/download PDF
26. Anti-müllerian hormone protein expression is reduced during the initial stages of follicle development in human polycystic ovaries.
- Author
-
Stubbs SA, Hardy K, Da Silva-Buttkus P, Stark J, Webber LJ, Flanagan AM, Themmen AP, Visser JA, Groome NP, and Franks S
- Subjects
- Adult, Anovulation genetics, Anovulation pathology, Anti-Mullerian Hormone, Cell Count, Female, Granulosa Cells physiology, Humans, Immunohistochemistry, Ovarian Follicle pathology, Ovary pathology, Polycystic Ovary Syndrome pathology, Glycoproteins biosynthesis, Glycoproteins genetics, Ovarian Follicle physiology, Polycystic Ovary Syndrome genetics, Polycystic Ovary Syndrome metabolism, Testicular Hormones biosynthesis, Testicular Hormones genetics
- Abstract
Context: Polycystic ovary syndrome, the most common cause of anovulatory infertility, is characterized by disordered folliculogenesis, notably increased progression from the primordial to the primary stages. This ovarian phenotype is similar to that observed in mice lacking anti-müllerian hormone (AMH)., Objective: The objective of this study is to investigate whether AMH is involved in accelerating the transition of follicles from primordial to primary stages in polycystic ovaries., Design: This study compares AMH expression in archive tissue from normal and polycystic ovaries., Setting: This is a laboratory-based study., Patients: Ovarian tissue from seven normoovulatory women and 16 women with polycystic ovaries (five of whom were anovulatory) was used in this study. Ovaries were classified by histology and with reference to menstrual cycle history and ultrasound., Main Outcome Measure: Presence and intensity of AMH expression in 1403 follicles was the main outcome measure., Results: AMH was observed from the primordial stage onward. AMH immunostaining was observed in significantly fewer primordial (P = 0.007) and transitional follicles (P = 0.001) in ovaries from anovulatory women with polycystic ovaries compared with women with regular cycles and either normal or polycystic ovaries. AMH-negative follicles had fewer pregranulosa cells in the largest cross-section of the follicle at both the primordial (median, four and six for AMH-negative and -positive follicles, respectively; P < 0.0001) and transitional stages (median six and nine; P < 0.0007) in normal tissue, and fewer at the transitional stage (median, seven and 11; P < 0.0001) in tissue from anovulatory women with polycystic ovaries. This suggests that AMH expression is associated with granulosa cell mitosis., Conclusions: These findings indicate a relative deficiency of AMH in primordial and transitional follicles in ovaries from anovulatory women with polycystic ovaries. This may contribute to disordered early follicle development in polycystic ovary syndrome.
- Published
- 2005
- Full Text
- View/download PDF
27. Follicular and hormonal dynamics during the estrous cycle in goats.
- Author
-
Medan MS, Watanabe G, Sasaki K, Groome NP, Sharawy S, and Taya K
- Subjects
- Animals, Dose-Response Relationship, Drug, Estradiol blood, Estrus, Female, Follicle Stimulating Hormone blood, Goats, Inhibins blood, Ovarian Follicle anatomy & histology, Ovarian Follicle metabolism, Ovarian Follicle physiology, Ovary diagnostic imaging, Ovulation, Time Factors, Ultrasonography methods, Estrous Cycle
- Abstract
Transrectal ultrasonography of ovaries was performed daily in 6 goats for 3 consecutive estrous cycles. Blood samples collected daily were measured for concentrations of FSH, inhibin A, and estradiol-17beta. Follicular and hormonal data were analyzed for associations between the follicular waves and hormonal concentrations. During the interovulatory intervals, follicular growth and regression occurred in a wave like pattern (2-5 waves), and the predominant patterns were three and four follicular waves. In addition, there was no significant difference among the diameters of dominant follicles during the growth phase of the follicular waves. The number of 3 mm follicles peaked on days 0, 7, and 11 in interovulatory intervals that had three follicular waves and on days -1, 5, 11, and 15 in those that had four follicular waves. Plasma concentrations of FSH increased around the day of follicular wave emergence and declined with the growth of follicles. Circulating FSH increased again concomitant with regression of dominant follicles in the anovulatory wave, whereas FSH levels remained low in the ovulatory wave. Inhibin A was negatively correlated with FSH, while it was positively correlated with estradiol-17beta, suggesting that inhibin A is a product of healthy growing follicles and that it contributes to the suppression of FSH secretion. In conclusion, the growth of ovarian follicles in goats exhibits a wave-like pattern, and follicular dominance is less apparent in goats. Moreover, inhibin A may be a key hormone for regulation of the follicular wave through suppression of FSH secretion in goats.
- Published
- 2005
- Full Text
- View/download PDF
28. Myostatin inhibits myogenesis and promotes adipogenesis in C3H 10T(1/2) mesenchymal multipotent cells.
- Author
-
Artaza JN, Bhasin S, Magee TR, Reisz-Porszasz S, Shen R, Groome NP, Meerasahib MF, and Gonzalez-Cadavid NF
- Subjects
- Adipocytes drug effects, Animals, Azacitidine pharmacology, Base Sequence, DNA Primers, Immunohistochemistry, Mesoderm physiology, Mice, Mice, Inbred C3H, MyoD Protein metabolism, Myoblasts cytology, Myoblasts physiology, Myostatin, Recombinant Proteins pharmacology, Reverse Transcriptase Polymerase Chain Reaction, Transforming Growth Factor beta genetics, Adipocytes cytology, Cell Differentiation drug effects, Mesoderm cytology, Transforming Growth Factor beta pharmacology
- Abstract
Inactivating mutations of the mammalian myostatin gene are associated with increased muscle mass and decreased fat mass; conversely, myostatin transgenic mice that overexpress myostatin in the skeletal muscle have decreased muscle mass and increased fat mass. We investigated the effects of recombinant myostatin protein and antimyostatin antibody on myogenic and adipogenic differentiation of mesenchymal multipotent cells. Accordingly, 10T(1/2) cells were incubated with 5'-azacytidine for 3 d to induce differentiation and then treated with a recombinant protein for myostatin (Mst) carboxy terminal 113 amino acids or a polyclonal anti-Mst antibody for 3, 7, and 14 d. Cells were also cotransfected with a Mst cDNA plasmid expressing the full-length 375-amino acid protein (pcDNA-Mst375) and the silencer RNAs for either Mst (pSil-Mst) or a random sequence (pSil-RS) for 3 or 7 d, and Mst expression was determined. Adipogenesis was evaluated by quantitative image analysis of fat cells before and after oil-red-O staining, immunocytochemistry of adiponectin, and Western blot for CCAAT/enhancer binding protein-alpha. Myogenesis was estimated by quantitative image analysis-immunocytochemistry for MyoD (Myo differentiation protein), myogenin, and myosin heavy chain type II, or by Western blot for myogenin. 5'-Azacytidine-mediated differentiation induced endogenous full-length Mst expression. Recombinant Mst carboxy terminal 113 amino acids inhibited both early and late markers of myogenesis and stimulated both early and late markers of adipogenesis, whereas the antibody against Mst exerted the reverse effects. Myogenin levels at 7 d after transfection of pcDNA-Mst375 were reduced as expected and elevated by pSil-Mst, which blocked efficiently Mst375 expression. In conclusion, myostatin promotes the differentiation of multipotent mesenchymal cells into the adipogenic lineage and inhibits myogenesis.
- Published
- 2005
- Full Text
- View/download PDF
29. Bone morphogenetic protein 15 and growth differentiation factor 9 co-operate to regulate granulosa cell function in ruminants.
- Author
-
McNatty KP, Juengel JL, Reader KL, Lun S, Myllymaa S, Lawrence SB, Western A, Meerasahib MF, Mottershead DG, Groome NP, Ritvos O, and Laitinen MP
- Subjects
- Animals, Cattle, Cell Culture Techniques, Drug Synergism, Female, Granulosa Cells drug effects, Growth Differentiation Factor 9, Inhibins analysis, Intercellular Signaling Peptides and Proteins metabolism, Progesterone analysis, Sheep, Species Specificity, Granulosa Cells metabolism, Inhibins biosynthesis, Intercellular Signaling Peptides and Proteins pharmacology, Progesterone biosynthesis, Ruminants metabolism
- Abstract
The oocyte-secreted polypeptide growth factors, growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15, also known as GDF9B) have both been shown to be essential for ovarian follicular development and ovulation rate. In addition, it is known from both in vivo and in vitro studies that these factors co-operate in some manner. To date, most studies examining the in vitro effects of these growth factors have used the rodent model. However, the evidence suggests that these growth factors have somewhat different roles between rodents and ruminants. Therefore, the objectives of these studies were to examine the effects of GDF9 and BMP15, alone and together, on the functions of ovine and bovine granulosa cells under in vitro conditions. Ovine (o)BMP15 given together with murine (m)GDF9 or oGDF9 was more potent in stimulating (3)H-thymidine incorporation by ovine granulosa cells compared with each growth factor alone. For bovine granulosa cells, there appeared to be little or no co-operativity between oBMP15 and oGDF9 as oBMP15 alone was as potent as any combination of the two growth factors in stimulating (3)H-thymidine uptake. The species of origin of GDF9 affected the progesterone response in ovine granulosa cells with mGDF9 stimulating and oGDF9 inhibiting progesterone production. Ovine BMP15 alone had no effect on progesterone production by ovine granulosa cells and these growth factors did not appear to co-operate. FSH-stimulated progesterone production by bovine granulosa cells was most potently inhibited when oBMP15 and murine or ovine GDF9 were administered together. As was observed for progesterone, the species of origin of GDF9 affected inhibin production by ovine granulosa cells where mGDF9 inhibited while oGDF9 stimulated production. Murine GDF9 also inhibited inhibin production from bovine granulosa cells. For both ovine and bovine granulosa cells, BMP15 alone had no effect on inhibin production and there did not appear to be any co-operation between GDF9 and BMP15. These results indicate that the effects of BMP15 and GDF9 varied with respect to the species of origin of the growth factor. Moreover, the effects of GDF9 and BMP15 together were often co-operative and not always the same as those observed for these growth factors alone.
- Published
- 2005
- Full Text
- View/download PDF
30. betaA- and betaC-activin, follistatin, activin receptor mRNA and betaC-activin peptide expression during rat liver regeneration.
- Author
-
Gold EJ, Zhang X, Wheatley AM, Mellor SL, Cranfield M, Risbridger GP, Groome NP, and Fleming JS
- Subjects
- Activin Receptors metabolism, Animals, Apoptosis, Body Weight, Hepatocytes cytology, Hepatocytes physiology, Inhibin-beta Subunits genetics, Male, Mitosis, Peptides genetics, Protein Isoforms genetics, Protein Subunits genetics, Random Allocation, Rats, Rats, Sprague-Dawley, Time Factors, Activin Receptors genetics, Follistatin metabolism, Inhibin-beta Subunits metabolism, Liver Regeneration physiology, Peptides metabolism, Protein Isoforms metabolism, Protein Subunits metabolism
- Abstract
The mRNA expression of two activin growth factor subunits (betaA- and betaC-activin), activin receptor subunits (ActRIIA, ActRIIB) and the activin-binding protein follistatin, and peptide expression of betaA-activin and betaC-activin subunits, were examined in regenerating rat liver after partial hepatectomy (PHx). Liver samples were collected from adult, male Sprague-Dawley rats, 12-240 h (n=3-5 rats per time point) after PHx or from sham-operated controls at the same time points. Hepatocyte mitosis and apoptosis were assessed histologically and by in situ cell death detection. RT and PCR were used to assess relative gene expression. betaA- and betaC-activin peptide immunoreactivity was assessed in liver and serum samples by western blotting, whereas cellular expression was investigated by immunohistochemistry, using specific monoclonal antibodies. betaA- and betaC-activin mRNA dropped to < 50% of sham control values 12 h after PHx and remained at this level until 168 h post-PHx, when betaA-activin expression increased to three times sham control values and betaC-activin mRNA returned to pre-PHx levels. A peak in follistatin expression was observed 24-48 h post-PHx, coincident with an increase in hepatocyte mitosis. No changes were observed in ActRIIA mRNA, whereas ActRIIB expression paralleled that of betaA-activin mRNA. betaC-activin immunoreactive homo- and heterodimers were observed in regenerating liver and serum. Mitotic hepatocytes frequently contained betaC-activin immunoreactivity, whereas apoptotic hepatocytes were often immunoreactive for betaA-activin. We conclude that betaA- and betaC-activin subunit proteins are autocrine growth regulators in regenerating liver and when expressed independently lead to hepatocyte apoptosis or mitosis in a subset of hepatocytes.
- Published
- 2005
- Full Text
- View/download PDF
31. Adenoviral gene transfer allows Smad-responsive gene promoter analyses and delineation of type I receptor usage of transforming growth factor-beta family ligands in cultured human granulosa luteal cells.
- Author
-
Kaivo-Oja N, Mottershead DG, Mazerbourg S, Myllymaa S, Duprat S, Gilchrist RB, Groome NP, Hsueh AJ, and Ritvos O
- Subjects
- Activins pharmacology, Bone Morphogenetic Protein 15, Cells, Cultured, Female, Growth Differentiation Factor 9, Humans, Inhibins biosynthesis, Intercellular Signaling Peptides and Proteins pharmacology, Ligands, Protein Serine-Threonine Kinases, Receptor, Transforming Growth Factor-beta Type I, Smad3 Protein, Activin Receptors, Type I physiology, Adenoviridae genetics, DNA-Binding Proteins metabolism, Gene Transfer, Horizontal, Luteal Cells metabolism, Promoter Regions, Genetic, Receptors, Transforming Growth Factor beta physiology, Trans-Activators metabolism, Transforming Growth Factor beta pharmacology
- Abstract
In the human ovary, cell growth and differentiation are regulated by members of the TGF-beta superfamily, including growth differentiation factor-9 (GDF9), TGF-beta, and activin. TGF-beta and activin are known to signal via Smad3 activation, and we have recently shown the involvement of Smad3 in cellular responses to GDF9. Recent studies with Smad3-deficient mice have also indicated a key role for this signaling mediator in ovarian folliculogenesis. We now demonstrate the use of a Smad3 reporter (CAGA-luciferase) adenovirus in primary cultures of human granulosa-luteal (hGL) cells to detect GDF9, TGF-beta, and activin responses. In rodent granulosa cells, TGF-beta and GDF9 signal through the TGF-beta type I receptor or activin receptor-like kinase 5 (Alk5), whereas the effect of activin is mediated though the activin type IB receptor, also known as Alk4. We now show that the GDF9 response in hGL cells is markedly potentiated upon overexpression of Alk5 by adenoviral gene transduction, as measured by the CAGA-luciferase reporter activity. A similar response to Alk5 overexpression was observed for TGF-beta, but not for activin. Adenoviral overexpression of the activin type IB receptor Alk4 in hGL cells specifically potentiated activin signaling, but not GDF9 or TGF-beta signaling. Alk5 overexpression in hGL cells also potentiated the GDF9 response when inhibin B production was used as the read-out. These results indicate that the CAGA-luciferase adenovirus can be used to study Smad3 signaling in primary cultures of human cells, and that adenoviral overexpression of wild-type receptors of the TGF-beta superfamily can be used to amplify the cellular response to ligands such as GDF9, TGF-beta, and activin. Furthermore, these studies indicate the involvement of Alk5 in GDF9 signaling in human cells and therefore, along with other recent studies, highlight how a limited number of type I and II receptors cooperate to generate specificity of action within the TGF-beta superfamily.
- Published
- 2005
- Full Text
- View/download PDF
32. The oocyte and its role in regulating ovulation rate: a new paradigm in reproductive biology.
- Author
-
McNatty KP, Moore LG, Hudson NL, Quirke LD, Lawrence SB, Reader K, Hanrahan JP, Smith P, Groome NP, Laitinen M, Ritvos O, and Juengel JL
- Subjects
- Animals, Bone Morphogenetic Protein 15, Female, Growth Differentiation Factor 9, Humans, Immunization, Intercellular Signaling Peptides and Proteins genetics, Intercellular Signaling Peptides and Proteins physiology, Mutation, Sheep, Structure-Activity Relationship, Growth Substances physiology, Mammals physiology, Oocytes physiology, Ovulation physiology
- Abstract
Ovulation rate in mammals is determined by a complex exchange of hormonal signals between the pituitary gland and the ovary and by a localised exchange of hormones within ovarian follicles between the oocyte and its adjacent somatic cells. From examination of inherited patterns of ovulation rate in sheep, point mutations have been identified in two oocyte-expressed genes, BMP15 (GDF9B) and GDF9. Animals heterozygous for any of these mutations have higher ovulation rates (that is, + 0.8-3) than wild-type contemporaries, whereas those homozygous for each of these mutations are sterile with ovarian follicular development disrupted during the preantral growth stages. Both GDF9 and BMP15 proteins are present in follicular fluid, indicating that they are secreted products. In vitro studies show that granulosa and/or cumulus cells are an important target for both growth factors. Multiple immunisations of sheep with BMP15 or GDF9 peptide protein conjugates show that both growth factors are essential for normal follicular growth and the maturation of preovulatory follicles. Short-term (that is, primary and booster) immunisation with a GDF9 or BMP15 peptide-protein conjugate has been shown to enhance ovulation rate and lamb production. In summary, recent studies of genetic mutations in sheep highlight the importance of oocyte-secreted factors in regulating ovulation rate, and these discoveries may help to explain why some mammals have a predisposition to produce two or more offspring rather than one.
- Published
- 2004
- Full Text
- View/download PDF
33. Immunoneutralization of growth differentiation factor 9 reveals it partially accounts for mouse oocyte mitogenic activity.
- Author
-
Gilchrist RB, Ritter LJ, Cranfield M, Jeffery LA, Amato F, Scott SJ, Myllymaa S, Kaivo-Oja N, Lankinen H, Mottershead DG, Groome NP, and Ritvos O
- Subjects
- Amino Acid Sequence, Animals, Bone Morphogenetic Protein 15, Female, Growth Differentiation Factor 9, Intercellular Signaling Peptides and Proteins chemistry, Mice, Mitogens chemistry, Mitogens immunology, Molecular Sequence Data, Oocytes metabolism, Protein Structure, Tertiary, Transforming Growth Factor beta genetics, Transforming Growth Factor beta immunology, Transforming Growth Factor beta metabolism, Antibodies, Monoclonal pharmacology, Intercellular Signaling Peptides and Proteins immunology, Intercellular Signaling Peptides and Proteins metabolism, Mitogens metabolism, Oocytes cytology
- Abstract
Paracrine factors secreted by oocytes play a pivotal role in promoting early ovarian follicle growth and in defining a morphogenic gradient in antral follicles, yet the exact identities of these oocyte factors remain unknown. This study was conducted to determine the extent to which the mitogenic activity of mouse oocytes can be attributed to growth differentiation factor 9 (GDF9). To do this, specific anti-human GDF9 monoclonal antibodies were generated. Based on epitope mapping and bioassays, a GDF9 neutralizing antibody, mAb-GDF9-53, was characterized with very low cross-reactivity with related transforming growth factor (TGF)beta superfamily members, including BMP15 (also called GDF9B). Pep-SPOT epitope mapping showed that mAb-GDF9-53 recognizes a short 4-aa sequence, and three-dimensional peptide modeling suggested that this binding motif lies at the C-terminal fingertip of mGDF9. As predicted by sequence alignments and modeling, the antibody detected recombinant GDF9, but not BMP15 in a Western blot and GDF9 protein in oocyte extract and oocyte-conditioned medium. In a mouse mural granulosa cell (MGC) bioassay, mAb-GDF9-53 completely abolished the mitogenic effects of GDF9, but had no effect on TGFbeta1 or activin A-stimulated MGC proliferation. An unrelated IgG at the same dose had no effect on GDF9 activity. This GDF9 neutralizing antibody was then tested in an established oocyte-secreted mitogen bioassay, where denuded oocytes cocultured with granulosa cells promote cell proliferation in a dose-dependent manner. The mAb-GDF9-53 dose dependently (0-160 microg/ml) decreased the mitogenic activity of oocytes but only by approximately 45% at the maximum dose of mAb. Just 5 microg/ml of mAb-GDF9-53 neutralized 90% of recombinant mGDF9 mitogenic activity, but only 15% of oocyte activity. Unlike mAb-GDF9-53, a TGFbeta pan-specific neutralizing antibody did not affect the mitogenic capacity of the oocyte, but completely neutralized TGF beta 1-induced DNA synthesis. This study has characterized a specific GDF9 neutralizing antibody. Our data provide the first direct evidence that the endogenous GDF9 protein is an important oocyte-secreted mitogen, but also show that GDF9 accounts for only part of total oocyte bioactivity.
- Published
- 2004
- Full Text
- View/download PDF
34. Nocturnal secretory dynamics of inhibin B and testosterone in pre- and peripubertal boys.
- Author
-
Crofton PM, Evans AE, Wallace AM, Groome NP, and Kelnar CJ
- Subjects
- Adolescent, Child, Follicle Stimulating Hormone, Human metabolism, Humans, Luteinizing Hormone metabolism, Male, Time Factors, Circadian Rhythm, Inhibins metabolism, Puberty metabolism, Testosterone metabolism
- Abstract
To investigate the secretory dynamics of testosterone and inhibin B, we collected samples every 20 min from 2000 h to 0800 h in 20 boys. Boys in group 1 (n = 5) were aged less than 8 yr, group 2 (n = 5) were aged more than 8 yr but 1.5 yr or more before pubertal onset, group 3 (n = 5) were studied 1.0 yr or less before pubertal onset, and group 4 (n = 5) were in early puberty. Testosterone increased after midnight in peripubertal boys, coinciding with the onset of LH pulsatility, and showed a pulsatile pattern in 6 of 10 of these boys. Cross-correlation analysis indicated significant temporal coupling between LH and testosterone. Inhibin B was higher in groups 3 and 4, compared with groups 1 and 2 (P < 0.01) and showed a downward trend overnight with no evidence of pulsatility and no evidence of short-term interactions with LH, FSH, or testosterone. Inhibin B and LH nocturnal means were both inversely correlated with time before pubertal onset (r(s) > or = -0.85, P < 0.01). Only LH nocturnal mean and amplitude, respectively, contributed independently to prediction of testosterone and inhibin B nocturnal means, explaining 71 and 65% of their variability. We conclude that both testosterone and inhibin B are related to nocturnal LH release in peripubertal boys but over different time scales.
- Published
- 2004
- Full Text
- View/download PDF
35. Anti-Müllerian hormone expression pattern in the human ovary: potential implications for initial and cyclic follicle recruitment.
- Author
-
Weenen C, Laven JS, Von Bergh AR, Cranfield M, Groome NP, Visser JA, Kramer P, Fauser BC, and Themmen AP
- Subjects
- Adolescent, Adult, Animals, Anti-Mullerian Hormone, Antibodies, Monoclonal, Blotting, Western, Female, Glycoproteins analysis, Glycoproteins immunology, Granulosa Cells cytology, Granulosa Cells metabolism, Humans, Immunohistochemistry, Male, Mice, Ovarian Follicle cytology, Ovarian Follicle growth & development, Staining and Labeling, Testicular Hormones analysis, Testicular Hormones immunology, Glycoproteins metabolism, Ovarian Follicle metabolism, Testicular Hormones metabolism
- Abstract
Anti-Müllerian hormone (AMH) is a member of the transforming growth factor-beta superfamily, which plays an important role in both ovarian primordial follicle recruitment and dominant follicle selection in mice. However, the role of AMH in folliculogenesis in humans has not been investigated in detail. In the present study, AMH expression was assessed using immunohistochemistry in ovarian sections, obtained from healthy regularly cycling women. To this end, a novel monoclonal antibody to human AMH was developed. AMH expression was not observed in primordial follicles, whereas 74% of the primary follicles showed at least a weak signal in the granulosa cells. The highest level of AMH expression was present in the granulosa cells of secondary, preantral and small antral follicles
- Published
- 2004
- Full Text
- View/download PDF
36. Endothelial cells and peripheral blood mononuclear cells are a potential source of extraplacental activin a in preeclampsia.
- Author
-
Tannetta DS, Muttukrishna S, Groome NP, Redman CW, and Sargent IL
- Subjects
- Cells, Cultured, Endothelium, Vascular drug effects, Endothelium, Vascular pathology, Endotoxins pharmacology, Escherichia coli, Female, Follistatin metabolism, Humans, Interleukin-1 pharmacology, Interleukin-6 pharmacology, Monocytes drug effects, Pregnancy, Recombinant Proteins pharmacology, Tumor Necrosis Factor-alpha pharmacology, Activins metabolism, Endothelium, Vascular metabolism, Inhibin-beta Subunits metabolism, Monocytes metabolism, Pre-Eclampsia metabolism
- Abstract
An excessive systemic inflammatory response, involving endothelial cells and leukocytes, underlies the maternal symptoms of preeclampsia. Activin A is raised in preeclampsia, suggesting a possible involvement in its pathophysiology. The placenta is the main source of activin A in normal pregnancy. We investigated whether peripheral blood mononuclear cells (PBMCs) and endothelium, activated by proinflammatory stimuli, were a potential source of activin A in preeclampsia. Both endotoxin and TNFalpha stimulated activin A secretion by PBMCs from nonpregnant, preeclamptic, and matched normal pregnant women (P < 0.05). Pregnancy increased the responsiveness of PBMCs to endotoxin (P < 0.05), whereas only the preeclamptic group were significantly more responsive to TNFalpha (P < 0.05). Human umbilical vein endothelial cells secreted activin A spontaneously and in response to TNFalpha (P < 0.05), but recombinant IL-1beta and IL-6 had no significant effect over the 72-h culture period. Inhibin A and follistatin were undetectable (<2 pg/ml and < 20 pg/ml, respectively) in PBMCs and human umbilical vein endothelial cell culture media. These data suggest that PBMCs and endothelium, activated by TNFalpha, could be extraplacental sources of activin A in preeclampsia. The pathological significance of increased activin A in preeclampsia is unknown, although it may have a role in the mechanisms underlying endothelium dysfunction.
- Published
- 2003
- Full Text
- View/download PDF
37. Activin betaC-subunit heterodimers provide a new mechanism of regulating activin levels in the prostate.
- Author
-
Mellor SL, Ball EM, O'Connor AE, Ethier JF, Cranfield M, Schmitt JF, Phillips DJ, Groome NP, and Risbridger GP
- Subjects
- Animals, CHO Cells, Cell Division drug effects, Cell Line, Cricetinae, Dimerization, Enzyme-Linked Immunosorbent Assay, Humans, Inhibin-beta Subunits genetics, Inhibin-beta Subunits metabolism, Inhibin-beta Subunits pharmacology, Male, Promoter Regions, Genetic drug effects, Promoter Regions, Genetic physiology, Sensitivity and Specificity, Tumor Cells, Cultured, Activins metabolism, Inhibin-beta Subunits physiology, Prostate metabolism
- Abstract
Activins are formed by dimerization of beta-subunits and, as members of the TGF-beta superfamily, have diverse roles as potent growth and differentiation factors. As the biological function of the activin C homodimer (betaC-betaC) is unknown, we sought to compare activin A (betaA-betaA), B (betaB-betaB), and C homodimer bioactivities and to investigate the consequences of activin betaC-subunit overexpression in prostate tumor cells. Exogenous activin A and B homodimers inhibited cell growth and activated activin-responsive promoters. In contrast, the activin C homodimer was unable to elicit these responses. We previously showed that the activin betaC-subunit heterodimerized with activin betaA in vitro to form activin AC. Therefore, we hypothesize that the activin betaC-subunit regulates the levels of bioactive activin A by the formation of activin AC heterodimers. To test this hypothesis, we measured activin AC heterodimer production using a novel specific two-site ELISA that we developed for this purpose. In the PC3 human prostate tumor cell line, activin betaC-subunit overexpression increased activin AC heterodimer levels, concomitantly reduced activin A levels, and decreased activin signaling. Overall, these data are consistent with a role for the activin betaC-subunit as a regulatory mechanism to reduce activin A secretion via intracellular heterodimerization.
- Published
- 2003
- Full Text
- View/download PDF
38. Ovarian dynamics and their associations with peripheral concentrations of gonadotropins, ovarian steroids, and inhibin during the estrous cycle in goats.
- Author
-
Medan MS, Watanabe G, Sasaki K, Sharawy S, Groome NP, and Taya K
- Subjects
- Animals, Corpus Luteum diagnostic imaging, Corpus Luteum physiology, Estradiol blood, Estrus blood, Female, Follicle Stimulating Hormone blood, Goats blood, Inhibins blood, Luteinizing Hormone blood, Ovarian Follicle diagnostic imaging, Ovarian Follicle physiology, Ovary diagnostic imaging, Progesterone blood, Ultrasonography, Estrus physiology, Goats physiology, Ovary physiology
- Abstract
Ovarian changes determined by daily transrectal ultrasound and its relationship with FSH, LH, estradiol-17beta, progesterone, and inhibin were investigated in six goats for three consecutive interovulatory intervals. Estrous cycles were synchronized using two injections of prostaglandin F2alpha analogue 11 days apart. All follicles 3 mm or greater in diameter and corpora lutea were measured daily. A follicular wave was defined as one or more follicles growing to 5 mm or greater in diameter. The day that the follicles reached 3 mm in diameter was defined as the day of wave emergence, and the first wave after ovulation was defined as wave 1. During the interovulatory interval (mean +/- SEM, 21.3 +/- 0.4 days; n = 18), follicular waves emerged at 0.3 +/- 0.5, 6.5 +/- 0.2, and 12.1 +/- 0.4 days for wave 1, wave 2, and wave 3, respectively, in goats with three waves of follicular development and at -0.6 +/- 0.3, 4.7 +/- 0.2, 9.4 +/- 0.5, and 13.4 +/- 0.5 days for wave 1, wave 2, wave 3, and wave 4, respectively, in goats with four waves of follicular development (Day 0 = the day of ovulation). The mean diameter of the largest follicle of the ovulatory wave was significantly larger than those of the largest follicles of the other waves. Corpora lutea could be identified ultrasonically at Day 3 postovulation and attained 12.1 +/- 0.3 mm in diameter on Day 8. Transient increases in plasma concentrations of FSH were detected around the day of follicular wave emergence. The level of FSH was negatively correlated with that of inhibin. These results demonstrated that follicular waves occurred in goats and that the predominant follicular wave pattern was four waves with ovulation from wave 4. These results also suggested that the emergence of follicular waves was closely associated with increased secretion of FSH.
- Published
- 2003
- Full Text
- View/download PDF
39. Plasma concentrations of inhibin a and follicle-stimulating hormone differ between cows with two or three waves of ovarian follicular development in a single estrous cycle.
- Author
-
Parker KI, Robertson DM, Groome NP, and Macmillan KL
- Subjects
- Animals, Cattle physiology, Estradiol blood, Female, Ovulation physiology, Progesterone blood, Regression Analysis, Cattle blood, Estrous Cycle physiology, Follicle Stimulating Hormone blood, Inhibins blood, Ovarian Follicle physiology
- Abstract
Patterns of ovarian follicle development were monitored daily in Holstein-Friesian cows that had two (n = 4) or three (n = 4) waves of ovarian follicle development during a single estrous cycle. The plasma from daily blood samples was used in assays for inhibin A, FSH, progesterone, and estradiol-17beta. Mean cycle lengths for cows with two and three waves were 21.8 and 25.3 days, respectively (P < 0.02). Although the average number of follicles >3-mm diameter on each pair of ovaries was similar for two- and three-wave cows on Days 2, 3, and 4 (Day 0 = day of ovulation; 8.6 vs. 9.6 follicles), there were more follicles >6-mm diameter on the ovaries of cows with two waves on Days 3 and 4. This difference was associated with a shorter interval from wave emergence to peak concentrations of inhibin A during the first wave in two-wave cows (2.0 vs. 3.8 days; P = 0.03) and with higher peak concentrations (474 vs. 332 pg/ml; P = 0.03). Differences in peak FSH concentrations were not significant (1.7 vs. 1.3 ng/ml; P = 0.10) and were inversely related to inhibin A concentrations. The peak concentrations of inhibin A and FSH in the second nonovulatory wave in the three-wave cows were similar to the low concentrations measured in the first wave (292 vs. 332 pg/ml of inhibin A, 1.3 vs. 1.3 ng/ml of FSH; P > 0.20). Average peak concentrations of inhibin A and FSH were similar during the ovulatory wave for cows with either two or three waves in a cycle (432 vs. 464 pg/ml of inhibin A, 2.3 vs. 2.1 ng/ml of FSH; P > 0.3). The lower concentrations of FSH during the emergence of the first follicular wave in cows with three-wave cycles may have reduced the rate of development of some of the follicles and reduced the concentrations of inhibin A. This pattern of lower concentrations of FSH and inhibin A was repeated in the second nonovulatory wave but not in the ovulatory wave. Subtle differences in the concentrations of these two hormones may underlie the mechanism that influences the number of waves of ovarian follicle development that occur during the bovine estrous cycle.
- Published
- 2003
- Full Text
- View/download PDF
40. Oocyte-mediated suppression of follicle-stimulating hormone- and insulin-like growth factor-induced secretion of steroids and inhibin-related proteins by bovine granulosa cells in vitro: possible role of transforming growth factor alpha.
- Author
-
Glister C, Groome NP, and Knight PG
- Subjects
- Animals, Cattle, Cell Count veterinary, Coculture Techniques, Epidermal Growth Factor biosynthesis, Epidermal Growth Factor metabolism, Estradiol metabolism, Female, Follicle Stimulating Hormone antagonists & inhibitors, Follistatin metabolism, Immunohistochemistry veterinary, Inhibins metabolism, Insulin-Like Growth Factor I antagonists & inhibitors, Microscopy, Confocal veterinary, Oocytes metabolism, Progesterone metabolism, Steroids metabolism, Transforming Growth Factor alpha biosynthesis, Transforming Growth Factor alpha metabolism, Activins metabolism, Follicle Stimulating Hormone pharmacology, Granulosa Cells metabolism, Inhibin-beta Subunits metabolism, Insulin-Like Growth Factor I pharmacology, Oocytes physiology, Steroids physiology, Transforming Growth Factor alpha physiology
- Abstract
The objective was to investigate the potential role of the oocyte in modulating proliferation and basal, FSH-induced and insulin-like growth factor (IGF)-induced secretion of inhibin A (inh A), activin A (act A), follistatin (FS), estradiol (E(2)), and progesterone (P(4)) by mural bovine granulosa cells. Cells from 4- to 6-mm follicles were cultured in serum-free medium containing insulin and androstenedione, and the effects of ovine FSH and IGF analogue (LR3-IGF-1) were tested alone and in the presence of denuded bovine oocytes (2, 8, or 20 per well). Medium was changed every 48 h, cultures were terminated after 144 h, and viable cell number was determined. Results are based on combined data from four independent cultures and are presented for the last time period only when responses were maximal. Both FSH and IGF increased (P < 0.001) secretion of inh A, act A, FS, E(2), and P(4) and raised cell number. In the absence of FSH or IGF, coculture with oocytes had no effect on any of the measured hormones, although cell number was increased up to 1.8-fold (P < 0.0001). Addition of oocytes to FSH-stimulated cells dose-dependently suppressed (P < 0.0001) inh A (6-fold maximum suppression), act A (5.5-fold), FS (3.6-fold), E(2) (4.6-fold), and P(4) (2.4-fold), with suppression increasing with FSH dose. Likewise, oocytes suppressed (P < 0.001) IGF-induced secretion of inh A, act A, FS, and E(2) (P < 0.05) but enhanced IGF-induced P(4) secretion (1.7-fold; P < 0.05). Given the similarity of these oocyte-mediated actions to those we observed previously following epidermal growth factor (EGF) treatment, we used immunocytochemistry to determine whether bovine oocytes express EGF or transforming growth factor (TGF) alpha. Intense staining with TGFalpha antibody (but not with EGF antibody) was detected in oocytes both before and after coculture. Experiments involving addition of TGFalpha to granulosa cells confirmed that the peptide mimicked the effects of oocytes on cell proliferation and on FSH- and IGF-induced hormone secretion. These experiments indicate that bovine oocytes secrete a factor(s) capable of modulating granulosa cell proliferation and responsiveness to FSH and IGF in terms of steroidogenesis and production of inhibin-related peptides, bovine oocytes express TGFalpha but not EGF, and TGFalpha is a prime candidate for mediating the actions of oocytes on bovine granulosa cells.
- Published
- 2003
- Full Text
- View/download PDF
41. Growth differentiation factor-9 induces Smad2 activation and inhibin B production in cultured human granulosa-luteal cells.
- Author
-
Kaivo-Oja N, Bondestam J, Kämäräinen M, Koskimies J, Vitt U, Cranfield M, Vuojolainen K, Kallio JP, Olkkonen VM, Hayashi M, Moustakas A, Groome NP, ten Dijke P, Hsueh AJ, and Ritvos O
- Subjects
- Adenoviridae genetics, Animals, Antibodies, Monoclonal, Bone Morphogenetic Protein 15, Cells, Cultured, DNA-Binding Proteins genetics, Dimerization, Female, Gene Expression Regulation, Viral, Growth Differentiation Factor 9, Humans, Inhibins chemistry, Intercellular Signaling Peptides and Proteins immunology, Luteal Cells drug effects, Mice, Phosphorylation, RNA, Messenger analysis, Rats, Recombinant Proteins pharmacology, Signal Transduction physiology, Smad2 Protein, Trans-Activators genetics, DNA-Binding Proteins metabolism, Inhibins genetics, Intercellular Signaling Peptides and Proteins pharmacology, Luteal Cells metabolism, Trans-Activators metabolism
- Abstract
The TGF beta family member growth differentiation factor-9 (GDF-9) is an oocyte-derived factor that is essential for mammalian ovarian folliculogenesis. GDF-9 mRNAs have been shown to be expressed in the human ovarian follicle from the primary follicle stage onward, and recombinant GDF-9 has been shown to promote human ovarian follicle growth in vitro. In this study with primary cultures of human granulosa-luteal (hGL) cells, we investigated whether recombinant GDF-9 activates components of the Smad signaling pathways known to be differentially activated by TGF beta and the bone morphogenetic proteins (BMPs). As with TGF beta, GDF-9 treatment caused the phosphorylation of endogenous 53-kDa proteins detected in Western blots with antiphospho-Smad2 antibodies (alpha PS2). However, unlike BMP-2, GDF-9 did not activate the phosphorylation of antiphospho-Smad1 antibody (alphaPS1)-immunoreactive proteins in hGL cells. Infection of hGL cells with an adenovirus expressing Smad2 (Ad-Smad2) confirmed that GDF-9 activates specifically phosphorylation of the Smad2 protein. Infection of hGL cells with Ad-Smad7, which expresses the inhibitory Smad7 protein, suppressed the levels of both GDF-9-induced endogenous and adenoviral alpha PS2-reactive proteins. Furthermore, GDF-9 increased the steady state levels of inhibin beta(B)-subunit mRNAs in hGL cells and strongly stimulated the secretion of dimeric inhibin B. Again, Ad-Smad7 blocked GDF-9-stimulated inhibin B production in a concentration-dependent manner. We identify here for the first time distinct molecular components of the GDF-9 signaling pathway in the human ovary. Our data suggest that GDF-9 mediates its effect through the pathway commonly activated by TGF beta and activin, but not that activated by many BMPs. The results are also consistent with the suggestion that in addition to endocrine control of inhibin production by gonadotropins, a local paracrine control of inhibin production is likely to occur via oocyte-derived factors in the human ovary.
- Published
- 2003
- Full Text
- View/download PDF
42. Differential expression of two estrogen receptor beta isoforms in the human fetal testis during the second trimester of pregnancy.
- Author
-
Gaskell TL, Robinson LL, Groome NP, Anderson RA, and Saunders PT
- Subjects
- Estrogen Receptor alpha, Estrogen Receptor beta, Female, Fetus chemistry, Fetus cytology, Fetus metabolism, Humans, Immunohistochemistry, Male, Microscopy, Confocal, Pregnancy Trimester, Second, Protein Isoforms genetics, Protein Isoforms metabolism, RNA, Messenger metabolism, Receptors, Estrogen genetics, Tissue Extracts metabolism, Pregnancy physiology, Receptors, Estrogen metabolism, Testis embryology
- Abstract
Testicular cancer is more common in individuals with disorders of the male reproductive tract. It has been suggested that inappropriate exposure to estrogens during fetal life may have an impact on maturation of testicular germ cells that are the cells of origin of the majority of testis cancers. The aim of the present study was to establish whether human fetal germ cells (gonocytes) are a potential target of estrogen action. To address this issue, we used RT-PCR and immunohistochemistry to examine the pattern of expression of estrogen receptors (ER alpha, ER beta, and ER beta 2 variant) in human fetal testes at 12-19 wk gestation. ER alpha, mRNA, and protein were not detected in any of the fetal testes. In contrast, using an antibody directed against the hinge domain of ER beta expression was detected in multiple testicular nuclei. RT-PCR with primers specific for full-length wild-type ER beta (ER beta 1) or the ER beta 2 variant formed by splicing of an alternative eighth exon, was performed on whole-tissue extracts and materials recovered by laser capture and revealed that mRNAs for both isoforms were expressed. Immunohistochemistry with isotype-specific monoclonal antibodies showed that ER beta 1 was low/undetectable in gonocytes, whereas these cells expressed the highest levels of ER beta 2, compared with other testicular cell types. Both ER beta 1 and ER beta 2 were detected in some but not all Sertoli cells, peritubular cells, and other interstitial cells including those tentatively identified as Leydig cells. Our immunohistochemical results demonstrate that during the second trimester, some but not all somatic cells within the human fetal testis express wild-type ER beta (ER beta 1) protein and/or the variant isoform of ER beta (ER beta 2) that lacks amino acids essential for binding of estradiol. ER beta 2 protein was readily detectable in fetal gonocytes, whereas ER beta 1 was not. We did not detect expression of ER alpha. The expression of ER beta 2, a variant proposed act as a dominant negative receptor, might prevent estrogen action in gonocytes. We suggest that during this period of fetal life, estrogenic ligands are most likely to act on somatic cells that contain ER beta 1 protein.
- Published
- 2003
- Full Text
- View/download PDF
43. Localization and secretion of inhibins in the equine fetal ovaries.
- Author
-
Tanaka Y, Taniyama H, Tsunoda N, Herath CB, Nakai R, Shinbo H, Nagamine N, Nambo Y, Nagata S, Watanabe G, Groome NP, and Taya K
- Subjects
- Animals, Female, Horses anatomy & histology, Inhibin-beta Subunits genetics, Inhibin-beta Subunits metabolism, Inhibins genetics, Maternal-Fetal Exchange, Organ Size, Ovary anatomy & histology, Pregnancy, RNA, Messenger genetics, RNA, Messenger metabolism, Fetus metabolism, Horses physiology, Inhibins metabolism, Ovary metabolism
- Abstract
To clarify the source of inhibins in equine female fetuses, concentrations of immunoreactive (ir-) inhibin, inhibin pro-alphaC, and inhibin A in both fetal and maternal circulation and in fetal ovaries were measured. In addition, the localization of inhibin alpha and inhibin/activin beta(A), and beta(B) subunits and the expression of inhibin alpha(A) and inhibin/activin beta(A) subunit mRNA in fetal ovaries were investigated using immunohistochemistry and in situ hybridization. Concentrations of circulating ir-inhibin, inhibin pro-alphaC, and inhibin A were remarkably more elevated in the fetal than in the maternal circulation between Days 100 and 250 of gestation. Fetal ovaries contained large amounts of ir-inhibin, inhibin pro-alphaC, and inhibin A. In contrast, these inhibin forms were undetectable in both the maternal ovaries and placenta. The inhibin alpha and inhibin/activin beta(A) and beta(B) subunit proteins were localized to enlarged interstitial cells of the equine fetal ovary. Expression of inhibin alpha and inhibin/activin beta(A) subunit mRNAs were also observed in the interstitial cells. We conclude that the main source of large amounts of inhibins in fetal circulation is interstitial cells of fetal ovary and is not of maternal origin. Furthermore, these inhibins may play some important physiological roles in the development of gonads in the equine fetus.
- Published
- 2003
- Full Text
- View/download PDF
44. Steroid receptor expression in uterine natural killer cells.
- Author
-
Henderson TA, Saunders PT, Moffett-King A, Groome NP, and Critchley HO
- Subjects
- CD56 Antigen metabolism, Computer Systems, Decidua metabolism, Endometrium metabolism, Estrogen Receptor alpha, Estrogen Receptor beta, Female, Humans, Immunohistochemistry, Pregnancy, Protein Isoforms genetics, Protein Isoforms metabolism, RNA, Messenger metabolism, Receptors, Estrogen genetics, Receptors, Estrogen metabolism, Receptors, Glucocorticoid genetics, Receptors, Glucocorticoid metabolism, Receptors, Progesterone genetics, Receptors, Progesterone metabolism, Receptors, Steroid genetics, Reverse Transcriptase Polymerase Chain Reaction, Uterus cytology, Killer Cells, Natural metabolism, Receptors, Steroid metabolism, Uterus metabolism
- Abstract
The endometrium contains a unique subset of uterine-specific natural killer (uNK) cells, the proposed functions of which include a role in decidualization, menstruation, and implantation. These cells increase in number during the mid-late secretory phase of the menstrual cycle and are also present in large numbers in early pregnancy. The cyclical nature of uNK cell appearance suggests hormonal regulation of these cells. To date, it has not been possible to localize either estrogen receptors (ERs) or progesterone receptors (PRs) to uNK cells. In the present study, we have investigated the steroid receptor expression of uNK cells, including not only ER alpha and PR but also wild-type ER beta 1, its variant form ER beta cx/beta 2, and glucocorticoid receptor (GR) using specific monoclonal antibodies and real-time quantitative RT-PCR. mRNA encoding ER alpha, PR, ER beta cx/beta 2, ER beta 1, and GR were identified in extracts of human endometrium across the menstrual cycle and in decidua. Quantitative real-time RT-PCR demonstrated an absence of ER alpha and PR mRNA in purified uNK cells. In contrast, mRNA for ER beta cx/beta 2, ER beta 1, and GR was present in uNK cells. ER alpha, PR, ER beta cx/beta 2, ER beta 1, and GR proteins were identified in endometrial and decidual biopsies. Colocalization using specific monoclonal antibodies confirmed that uNK cells were immunonegative for ER alpha and PR protein. These cells were also immunonegative for ER beta cx/beta 2 but did express ER beta 1 and GR proteins. These results raise the possibility that estrogens and glucocorticoids could be acting directly on uNK cells through ER beta and GR, respectively, to influence gene transcription in the endometrium and decidua.
- Published
- 2003
- Full Text
- View/download PDF
45. Growth differentiation factor 9 and bone morphogenetic protein 15 are essential for ovarian follicular development in sheep.
- Author
-
Juengel JL, Hudson NL, Heath DA, Smith P, Reader KL, Lawrence SB, O'Connell AR, Laitinen MP, Cranfield M, Groome NP, Ritvos O, and McNatty KP
- Subjects
- Animals, Antibodies blood, Antigens immunology, Estrus, Female, Growth Differentiation Factor 9, Hemocyanins immunology, Immunization, Immunization, Passive, Immunohistochemistry, Intercellular Signaling Peptides and Proteins analysis, Intercellular Signaling Peptides and Proteins immunology, Oocytes chemistry, Ovary chemistry, Ovulation, Progesterone blood, Intercellular Signaling Peptides and Proteins physiology, Ovarian Follicle physiology, Sheep physiology
- Abstract
The aim of this study was to test the hypothesis that both growth differential factor 9 (GDF9) and bone morphogenetic protein (BMP15; also known as GDF9B) are essential for normal ovarian follicular development in mammals with a low ovulation rate phenotype. Sheep (9-10 per group) were immunized with keyhole limpet hemocyanin (KLH; control), a GDF9-specific peptide conjugated to KLH (GDF9 peptide), a BMP15-specific peptide conjugated to KLH (BMP15 peptide), or the mature region of oBMP15 conjugated to KLH (oBMP15 mature protein) for a period of 7 mo and the effects of these treatments on various ovarian parameters such as ovarian follicular development, ovulation rate, and plasma progesterone concentrations evaluated. Also in the present study, we examined, by immunohistochemistry, the cellular localizations of GDF9 and BMP15 proteins in the ovaries of lambs. Both GDF9 and BMP15 proteins were localized specifically within ovarian follicles to the oocyte, thereby establishing for the sheep that the oocyte is the only intraovarian source of these growth factors. Immunization with either GDF9 peptide or BMP15 peptide caused anovulation in 7 of 10 and 9 of 10 ewes, respectively, when assessed at ovarian collection. Most ewes (7 of 10) immunized with oBMP15 mature protein had a least one observable estrus during the experimental period, and ovulation rate at this estrus was higher in these ewes compared with those immunized with KLH alone. In both the KLH-GDF9 peptide- and KLH-BMP15 peptide-treated ewes, histological examination of the ovaries at recovery (i.e., approximately 7 mo after the primary immunization) showed that most animals had few, if any, normal follicles beyond the primary (i.e., type 2) stage of development. In addition, abnormalities such as enlarged oocytes surrounded by a single layer of flattened and/or cuboidal granulosa cells or oocyte-free nodules of granulosa cells were often observed, especially in the anovulatory ewes. Passive immunization of ewes, each given 100 ml of a pool of plasma from the GDF9 peptide- or BMP15 peptide-immunized ewes at 4 days before induction of luteal regression also disrupted ovarian function. The ewes given the plasma against the GDF9 peptide formed 1-2 corpora lutea but 3 of 5 animals did not display normal luteal phase patterns of progesterone concentrations. The effect of plasma against the BMP15 peptide was more dramatic, with 4 of 5 animals failing to ovulate and 3 of 5 ewes lacking surface-visible antral follicles at laparoscopy. By contrast, administration of plasma against KLH did not affect ovulation rate or luteal function in any animal. In conclusion, these findings support the hypothesis that, in mammals with a low ovulation rate phenotype, both oocyte-derived GDF9 and BMP15 proteins are essential for normal follicular development, including both the early and later stages of growth.
- Published
- 2002
- Full Text
- View/download PDF
46. Inhibin B regulating follicle-stimulating hormone secretion during testicular recrudescence in the male golden hamster.
- Author
-
Jin W, Herath CB, Yoshida M, Arai KY, Saita E, Zhanquan S, Ren L, Watanabe G, Groome NP, and Taya K
- Subjects
- Animals, Cricetinae, Genitalia, Male anatomy & histology, Hormones blood, Immunohistochemistry, Male, Mesocricetus, Organ Size radiation effects, Osmolar Concentration, Proliferating Cell Nuclear Antigen metabolism, Sperm Motility radiation effects, Testis anatomy & histology, Follicle Stimulating Hormone metabolism, Inhibins physiology, Photoperiod, Testis physiology, Testis radiation effects
- Abstract
In the present study, to clarify whether inhibin affects follicle-stimulating hormone (FSH) secretion in the recrudescence of the male golden hamster, we used a recently developed specific enzyme-linked immunosorbent assay (ELISA) in order to measure 2 forms of inhibin molecules: inhibin B and inhibin pro-alphaC. In addition, we used the radioimmunoassay (RIA) to measure immunoreactive (ir-)inhibin, FSH, luteinizing hormone (LH), and testosterone. And finally, we used the proliferating cell nuclear antigen (PCNA) and computer-assisted sperm motion analysis (CASA) methods to ascertain how well spermatogenesis and sperm motility recover from the photoinhibition caused by exposure to a short-day (SD; 10-hour light: 14-hour dark) photoperiod. Animals were exposed to SD for 15 weeks, and then their testes were checked carefully and found to be completely regressed. Thereafter, those animals were transported to a long-day (LD; 14-hour light: 10-hour dark) photoperiod. Sampling was carried out at weeks 0 (exposed SD 15 weeks), 1, 2, 4, 6, 8, and 10. Plasma FSH rapidly increased and reached peak levels 2 weeks after transferral to the LD photoperiod and then declined to normal LD levels at week 6. Circulating ir-inhibin, inhibin B, and inhibin pro-alphaC rose to normal LD levels by week 4. A highly significant inverse correlation was observed between plasma FSH and inhibin B but not between FSH and either ir-inhibin or inhibin pro-alphaC. Plasma testosterone recovered to normal LD levels within 1 week. Sperm motility parameters were low until week 2 and recovered to normal LD levels from weeks 4 to 10. PCNA-labeled cells were confined to the spermatogenic cells of the seminiferous tubules, though Leydig and Sertoli cell nuclei were never stained for PCNA during the period studied. The number of pachytene spermatocytes and the diameter of seminiferous tubules increased in a time-dependent manner after transferral from SD to LD. In conclusion, these results suggest that 1) secretion of inhibin B may be stimulated by an early rise in FSH; 2) inhibin B suppresses FSH secretion from weeks 2 to 10, after transferral to the LD photoperiod; and 3) testes recrudescence is based on the increase in the number of sperm cells instead of the increase in the number of Sertoli and Leydig cells of the male golden hamster.
- Published
- 2002
47. Wild-type estrogen receptor (ERbeta1) and the splice variant (ERbetacx/beta2) are both expressed within the human endometrium throughout the normal menstrual cycle.
- Author
-
Critchley HO, Henderson TA, Kelly RW, Scobie GS, Evans LR, Groome NP, and Saunders PT
- Subjects
- Antibodies, Monoclonal, Cell Nucleus chemistry, Endometrium chemistry, Estrogen Receptor beta, Female, Gene Expression, Humans, Immunohistochemistry, Progesterone blood, RNA, Messenger analysis, Reverse Transcriptase Polymerase Chain Reaction, Alternative Splicing, Endometrium metabolism, Genetic Variation, Menstrual Cycle, Receptors, Estrogen genetics
- Abstract
Estrogen action is mediated via two subtypes of the estrogen receptor (ER), usually referred to as ERalpha and ERbeta. We have previously compared the spatial and temporal expressions of ERalpha and ERbeta proteins in human endometrium and reported that endothelial cells exclusively express ERbeta. In the present study we have extended our investigations to compare the pattern of expression of wild-type (ERbeta1) and a newly identified ERbeta variant isoform (ERbetacx/beta2) that lacks the ability to bind steroids. mRNAs encoding both ERbeta1 and ERbetacx/beta2 receptors were identified in human endometrial extracts by RT-PCR. Quantitative TaqMan R-TPCR demonstrated that levels of total mRNAs were increased significantly premenstrually as circulating progesterone levels declined. ERbeta1 and ERbetacx/beta2 proteins were identified within multiple cell types within the endometrium using isotype-specific monoclonal antibodies; immunoexpression of ERbetacx/beta2 appeared less intense than that of ERbeta1 in endometrial glandular epithelium and endothelial cells. Immunoexpression of ERbeta1 appeared unchanged throughout the menstrual cycle. In contrast, levels of ERbetacx/beta2-specific immunoreactivity were specifically reduced in gland cells within the functional layer, but not in those of the basal layer, in the midsecretory phase. It is possible that coexpression of ERbetacx/beta2 in cells containing ERbeta1 and/or ERalpha may modulate the effects of estrogens on the endometrium.
- Published
- 2002
- Full Text
- View/download PDF
48. ERbeta1 and the ERbeta2 splice variant (ERbetacx/beta2) are expressed in distinct cell populations in the adult human testis.
- Author
-
Saunders PT, Millar MR, Macpherson S, Irvine DS, Groome NP, Evans LR, Sharpe RM, and Scobie GA
- Subjects
- Adult, Antibodies, Monoclonal, Estrogen Receptor beta, Fluorescent Antibody Technique, Genetic Variation, Humans, Immunohistochemistry, Male, Protein Isoforms metabolism, Testis cytology, Tissue Distribution, DNA, Recombinant, Receptors, Estrogen genetics, Receptors, Estrogen metabolism, Testis metabolism
- Abstract
Estrogens can regulate germ cell function. Estrogen action is mediated via high affinity ERs; two subtypes (ERalpha and ERbeta) have been identified. We have shown previously that ERbeta is expressed in nuclei of multiple human testicular cells. A variant isoform of human (h) ERbeta (hERbetacx/2), formed by alternative splicing, has been identified in testicular cDNA libraries by two laboratories. The present study examined the expression of wild-type (ERbeta1) and variant (ERbeta2) beta receptors in human testes by 1) RT-PCR with isoform specific primers, and 2) single and double immunohistochemistry using monoclonal antibodies raised against peptides unique to the C termini of hERbeta1 and hERbeta2. PCR products specific for ERbeta1 and ERbeta2 were amplified from cDNA pools prepared from human testes and granulosa cells. On Western blots, the anti-ERbeta1 monoclonal antibody bound to recombinant ERbeta1 and the anti-ERbeta2 monoclonal to recombinant hERbeta2. Neither bound to the other ERbeta isoform nor to recombinant ERalpha. ERbeta1 and ERbeta2 proteins were both detected in human testis. Immunoexpression of ERbeta1 was most intense in pachytene spermatocytes and round spermatids, whereas low levels of expression were detected in Sertoli cells, spermatogonia, preleptotene, leptotene, zygotene, and diplotene spermatocytes. Highest levels of expression of ERbeta2 protein were detected in Sertoli cells and spermatogonia with low/variable expression in preleptotene, pachytene, and diplotene spermatocytes. No immunostaining was detected in elongating spermatids. Most interstitial cells expressed more ERbeta2 than ERbeta1. It is speculated that the cells most susceptible to modulation by estrogenic ligands are round spermatids in which levels of expression of ERbeta1 are high. In contrast, expression of ERbeta2, an isoform that may act as a dominant negative inhibitor of ER action, in Sertoli cells and spermatogonia, could protect these cells from adverse effects of estrogens.
- Published
- 2002
- Full Text
- View/download PDF
49. Women with poor response to IVF have lowered circulating gonadotrophin surge-attenuating factor (GnSAF) bioactivity during spontaneous and stimulated cycles.
- Author
-
Martinez F, Barri PN, Coroleu B, Tur R, Sorsa-Leslie T, Harris WJ, Groome NP, Knight PG, and Fowler PA
- Subjects
- Adult, Female, Gonadal Hormones, Gonadotropins blood, Humans, Infertility, Female blood, Inhibins blood, Ovulation Induction, Recombinant Proteins therapeutic use, Steroids blood, Treatment Failure, Fertilization in Vitro, Follicle Stimulating Hormone therapeutic use, Infertility, Female therapy, Menstrual Cycle physiology, Ovary drug effects, Proteins metabolism
- Abstract
Background: Up to 13% of IVF cancellations are due to poor responses during down-regulated cycles. Because premature luteinization occurs more frequently in older or "poor responder" patients, defective production of gonadotrophin surge-attenuating factor (GnSAF) may be involved., Methods: Nine women with normal previous IVF response (NORM) and 9 with previous poor IVF response (POOR) were monitored in a spontaneous cycle (blood samples: days 2, 7, 11, 15 and 20) and then stimulated with recombinant human FSH (rFSH) under GnRH agonist (blood samples: treatment days GnRH agonist + 2, GnRH agonist + 7, day of HCG administration and days HCG + 1 and HCG + 8). LH, FSH, estradiol, progesterone and inhibin-A and -B were assayed in individual samples while GnSAF bioactivity was determined in samples pooled according to day, cycle and IVF response., Results: During spontaneous cycles LH, steroids and inhibins were similar between NORM and POOR women, FSH was elevated in POOR women (4.9 +/- 0.3 versus 6.7 +/- 0.6 mIU/l, P < 0.01) and GnSAF bioactivity was detectable on days 2, 7 and 11 in NORM women only. During IVF cycles inhibin-A and -B rose more markedly in NORM than POOR women. Similarly GnSAF production peaked on day GnRH agonist + 7 in NORM women, but on the day of HCG administration in POOR women., Conclusions: Defects in ovarian responsiveness to FSH include reduced GnSAF production. This suggests that GnSAF should be investigated as a marker of ovarian reserve once an immunoassay becomes available.
- Published
- 2002
- Full Text
- View/download PDF
50. The testis as a major source of circulating inhibins in the male equine fetus during the second half of gestation.
- Author
-
Tanaka Y, Taniyama H, Tsunoda N, Shinbo H, Nagamine N, Nambo Y, Nagata S, Watanabe G, Herath CB, Groome NP, and Taya K
- Subjects
- Animals, Enzyme-Linked Immunosorbent Assay, Fetal Blood, Fetus metabolism, Immunohistochemistry, Inhibins blood, Male, Osmolar Concentration, Protein Precursors metabolism, Gestational Age, Horses embryology, Inhibins metabolism, Testis embryology
- Abstract
Immunolocalization of the inhibin (a) and inhibin/activin (beta3A and betaB) subunit proteins in equine fetal testes was investigated to determine the ability of the fetal testis to produce inhibins at 120, 150, 200, and 250 days of gestation. In addition, concentrations of immunoreactive (ir)-inhibin, inhibin pro-aC, and inhibin A in both the maternal and fetal circulation were measured. It was found that plasma concentrations of ir-inhibin, inhibin pro-alphaC, and inhibin A were much higher (P < .05) in the fetal than in the maternal circulation at any stage of gestation examined. Similarly, while fetal testicular homogenate contained increased amounts of inhibins, the inhibins were undetectable in homogenates of maternal ovaries and placentae. At 120 days of gestation, all 3 subunit proteins were localized to the interstitial cells, while the immunoreactivity for the inhibin/activin 3B subunit protein was also observed in Sertoli cells. The intensity of immunoreactivity for the 3 subunit proteins in interstitial cells increased as pregnancy advanced to day 200, and, at this stage, immunoreactivity for the inhibin alpha subunit protein was observed in the fetal testes in a pattern consistent with localization in Sertoli cells. Thus, the inhibin/activin betaA subunit protein was confined to interstitial cells during the gestational periods examined. We conclude that equine fetal testes secrete large amounts of inhibins, including dimeric inhibin A and possibly other dimeric forms, such as inhibin B and activins, into the fetal circulation. These results suggest that these proteins may play some important roles in the development of fetal testes during gestation.
- Published
- 2002
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.