Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Cell Rep ; 36(8): 109579, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34433056

ABSTRACT

Ultraviolet (UV) light affects endocrinological and behavioral aspects of sexuality via an unknown mechanism. Here we discover that ultraviolet B (UVB) exposure enhances the levels of sex-steroid hormones and sexual behavior, which are mediated by the skin. In female mice, UVB exposure increases hypothalamus-pituitary-gonadal axis hormone levels, resulting in larger ovaries; extends estrus days; and increases anti-Mullerian hormone (AMH) expression. UVB exposure also enhances the sexual responsiveness and attractiveness of females and male-female interactions. Conditional knockout of p53 specifically in skin keratinocytes abolishes the effects of UVB. Thus, UVB triggers a skin-brain-gonadal axis through skin p53 activation. In humans, solar exposure enhances romantic passion in both genders and aggressiveness in men, as seen in analysis of individual questionaries, and positively correlates with testosterone level. Our findings suggest opportunities for treatment of sex-steroid-related dysfunctions.


Subject(s)
Anti-Mullerian Hormone/biosynthesis , Hypothalamo-Hypophyseal System/metabolism , Ovary/metabolism , Sexual Behavior/radiation effects , Skin/metabolism , Testosterone/biosynthesis , Ultraviolet Rays , Animals , Estrus/metabolism , Female , Gene Knockout Techniques , Keratinocytes/metabolism , Male , Mice
2.
Biol Reprod ; 103(3): 572-582, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32432313

ABSTRACT

In this study, we aimed to determine the origin of the difference, in terms of anti-Müllerian hormone production, existing between the bovine and porcine ovaries. We first confirmed by quantitative real-time-Polymerase-Chain Reaction, ELISA assay and immunohistochemistry that anti-Müllerian hormone mRNA and protein production are very low in porcine ovarian growing follicles compared to bovine ones. We then have transfected porcine and bovine granulosa cells with vectors containing the luciferase gene driven by the porcine or the bovine anti-Müllerian hormone promoter. These transfection experiments showed that the porcine anti-Müllerian hormone promoter is less active and less responsive to bone morphogenetic protein stimulations than the bovine promoter in both porcine and bovine cells. Moreover, bovine but not porcine granulosa cells were responsive to bone morphogenetic protein stimulation after transfection of a plasmidic construction including a strong response element to the bone morphogenetic proteins (12 repetitions of the GCCG sequence) upstream of the luciferase reporter gene. We also showed that SMAD6, an inhibitor of the SMAD1-5-8 pathway, is strongly expressed in porcine compared to the bovine granulosa cells. Overall, these results suggest that the low expression of anti-Müllerian hormone in porcine growing follicles is due to both a lack of activity/sensitivity of the porcine anti-Müllerian hormone promoter, and to the lack of responsiveness of porcine granulosa cells to bone morphogenetic protein signaling, potentially due to an overexpression of SMAD6 compared to bovine granulosa cells. We propose that the low levels of anti-Müllerian hormone in the pig would explain the poly-ovulatory phenotype in this species.


Subject(s)
Anti-Mullerian Hormone/biosynthesis , Granulosa Cells/metabolism , Ovary/metabolism , Animals , Anti-Mullerian Hormone/genetics , Bone Morphogenetic Proteins/biosynthesis , Cattle , Female , Gene Expression Regulation, Developmental , Immunohistochemistry , Ovary/cytology , Promoter Regions, Genetic , Signal Transduction/drug effects , Smad6 Protein/biosynthesis , Smad6 Protein/genetics , Species Specificity , Swine
3.
Prog Biophys Mol Biol ; 145: 10-18, 2019 08.
Article in English | MEDLINE | ID: mdl-30500339

ABSTRACT

It is known that LLLT has beneficial effects on several pathological conditions including wound healing, pain and inflammation. LLLT modulates biological processes, including cell proliferation, apoptosis and angiogenesis. In the present study, we examined the effect of local application of LLLT on follicular dynamics, ovarian reserve, AMH expression, progesterone levels, apoptosis, angiogenesis, and reproductive outcome in adult mice. LLLT (200 J/cm2) increased the percentage of primary and preantral follicles, whilst decreasing the percentage of corpora lutea compared to control ovaries. LLLT-treated ovaries did not exhibit any changes regarding the number of primordial follicles. We observed a higher percentage of AMH-positive follicles (in early stages of development) in LLLT-treated ovaries compared to control ovaries. LLLT reduced the P4 concentration and the apoptosis in early antral follicles compared to control ones. LLLT caused a reduction in the endothelial cell area and an increase in the periendothelial cell area in the ovary. Additionally, LLLT was able to improve oocyte quality. Our findings suggest that local application of LLLT modulates follicular dynamics by regulating apoptosis and the vascular stability in mouse ovary. In conclusion, these data indicate that LLLT might become a novel and useful tool in the treatment of several pathologies, including female reproductive disorders.


Subject(s)
Anti-Mullerian Hormone/biosynthesis , Apoptosis/radiation effects , Low-Level Light Therapy , Neovascularization, Physiologic/radiation effects , Ovary/radiation effects , Animals , Cell Line , Cell Proliferation/radiation effects , Corpus Luteum/radiation effects , Female , Fertilization in Vitro/radiation effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovarian Follicle/cytology , Ovarian Follicle/radiation effects , Ovary/blood supply , Ovary/cytology , Ovary/metabolism , Progesterone/biosynthesis , Superovulation/radiation effects
4.
Microsc Res Tech ; 82(6): 635-642, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30582244

ABSTRACT

Many studies have reported that human endometrial mesenchymal stem cells (HuMenSCs) are capable of repairing damaged tissues. The aim of the present study was to investigate the effects of HuMenSCs transplantation as a treatment modality in premature ovarian failure (POF) associated with chemotherapy-induced ovarian damage. HuMenSCs were isolated from menstrual blood samples of five women. After the in vitro culture of HuMenSCs, purity of the cells was assessed by cytometry using CD44, CD90, CD34, and CD45 FITC conjugate antibody. Twenty-four female Wistar rats were randomly divided into four groups: negative control, positive control, sham, and treatment groups. The rat models of POF used in our study were established by injecting busulfan intraperitoneally into the rats during the first estrus cycle. HuMenSCs were transplanted by injection via the tail vein into the POF-induced rats. Four weeks after POF induction, ovaries were collected and the levels of Amh, Fst, and Fshr expression in the granulosa cell (GC) layer, as well as plasma estradiol (E2) and progesterone (P4) levels were evaluated. Moreover, migration and localization of DiI-labeled HuMenSCs were detected, and the labeled cells were found to be localized in GCs layer of immature follicles. In addition to DiI-labelled HuMenSCs tracking, increased levels of expression of Amh and Fshr and Fst, and the high plasma levels of E2 and P4 confirmed that HuMenSC transplantation had a significant effect on follicle formation and ovulation in the treatment group compared with the negative control (POF) group.


Subject(s)
Cell Transplantation/methods , Granulosa Cells/physiology , Mesenchymal Stem Cells/physiology , Primary Ovarian Insufficiency/therapy , Animals , Anti-Mullerian Hormone/biosynthesis , Busulfan/administration & dosage , Disease Models, Animal , Female , Follicle Stimulating Hormone/biosynthesis , Gene Expression Profiling , Histocytochemistry , Humans , Injections, Intraperitoneal , Injections, Intravenous , Ovarian Follicle/pathology , Ovary/pathology , Ovary/physiology , Ovulation , Primary Ovarian Insufficiency/chemically induced , Rats, Wistar , Real-Time Polymerase Chain Reaction , Receptors, FSH/biosynthesis , Treatment Outcome
5.
Reprod Toxicol ; 80: 60-67, 2018 09.
Article in English | MEDLINE | ID: mdl-29969652

ABSTRACT

Humans are at daily risk by simultaneous exposures to a broad spectrum of man-made chemicals in the commercial products. Several classes of chemicals have been shown to alter follicle development and reduce fertility, leading to premature ovarian failure (POF) in mammals. We investigate the synergistic effects of 4-vinylcyclohexene diepoxide (VCD) and phthalate, including di(2-ethylhexyl) phthalate (DEHP), butyl benzyl phthalate (BBP) and di-n-butyl phthalate (DBP) on POF. Combination exposure with VCD and phthalate significantly reduced the numbers of primary follicles. The expressions of Amh and Sohlh2 were significantly decreased in the combination groups. Serum Amh levels were significantly lower in the combination groups. Additionally, serum levels of follicle-stimulating hormone were significantly increased in combination groups. Taken together, exposure to phthalates promotes the depletion of follicular follicles and consequently increases the risk of premature menopause, and combined exposure of phthalates and VCD to early menopausal women is likely to aggravate the POF syndrome.


Subject(s)
Cyclohexenes/toxicity , Endocrine Disruptors/toxicity , Estrous Cycle/drug effects , Ovarian Follicle/drug effects , Phthalic Acids/toxicity , Primary Ovarian Insufficiency/chemically induced , Vinyl Compounds/toxicity , Animals , Anti-Mullerian Hormone/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Drug Synergism , Female , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Primary Ovarian Insufficiency/metabolism , Rats, Sprague-Dawley
6.
J Comp Pathol ; 158: 22-31, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29422312

ABSTRACT

Anti-Müllerian hormone (AMH) is a homodimeric glycoprotein expressed exclusively in the gonads. This hormone is an important regulator of the early growth of follicles through inhibitory effects on the recruitment of primordial follicles into the pool of growing follicles and on granulosa cell proliferation. Cystic ovarian disease (COD) is an important disorder affecting the fertility of dairy cattle. In the present study, we evaluated the expression of AMH in granulosa cells and AMH secretion into follicular fluid in pre-ovulatory follicles from control cows, animals with spontaneously arising COD and during the development of the disease, at 5, 10 and 15 days of follicular persistence. To this end, after an oestrous synchronization protocol, low doses of progesterone was administered for 5, 10 and 15 days after the expected day of ovulation (day 0 of follicular persistence) in treated cows (groups P5, P10 and P15, respectively), using an intravaginal progesterone-releasing device. Results showed a decrease in the expression of AMH in granulosa cells throughout folliculogenesis (P <0.05) and in the spontaneously arising follicular cysts and persistent follicles related to the control group (P <0.05). There was also a higher concentration of AMH in the follicular fluid of persistent follicles at 10 and 15 days of follicular persistence (P <0.05). Together, these results may indicate an alteration in AMH expression and secretion, which occurs early in folliculogenesis and incipiently during the development of COD, and which could contribute to the recurrence of this disease in cattle.


Subject(s)
Anti-Mullerian Hormone/biosynthesis , Cattle Diseases/metabolism , Ovarian Cysts/veterinary , Animals , Cattle , Female
7.
Reprod Domest Anim ; 52 Suppl 2: 303-309, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27758004

ABSTRACT

During the last decade, analysis of anti-Müllerian hormone (AMH), highly conserved between mammalian species, has contributed to new information in reproductive endocrinology, due to clinically available diagnostic assays. AMH is produced solely in the gonads, in the Sertoli cells of testes and granulosa cells of the ovary, and thus offers possibilities to diagnose physiologic and pathologic conditions involving these organs. This article reviews indications for AMH analysis in cats and dogs, including diagnosing the presence of gonads, and granulosa or Sertoli cell tumours. Diagnostic challenges are addressed. One specific organ, the prostate, is commonly affected by pathologic changes in older dogs. A commercial assay for analysing canine prostatic specific esterase (CPSE) enables analysis of CPSE in clinical practice, of potential value in the workup of benign prostatic hyperplasia in male dogs. This is described in this review, as is a new method for analysis of steroids: liquid chromatography-tandem mass spectrometry LC-MS/MS. Steroids have since long been analysed in studies on reproduction, and LC-MS/MS has the advantage of allowing analysis of panels of multiple steroids from small sample volumes. Altogether, these available methods may give new insights into small animal reproduction and are valuable tools for the practicing veterinarian.


Subject(s)
Anti-Mullerian Hormone/blood , Cat Diseases/blood , Dog Diseases/blood , Reproduction , Animals , Anti-Mullerian Hormone/biosynthesis , Cats , Dogs , Esterases/analysis , Female , Genital Diseases, Female/diagnosis , Genital Diseases, Female/veterinary , Genital Diseases, Male/diagnosis , Genital Diseases, Male/veterinary , Granulosa Cells/metabolism , Male , Ovary/metabolism , Prostate/enzymology , Prostatic Hyperplasia/blood , Sertoli Cells/metabolism , Testis/metabolism
8.
J Assist Reprod Genet ; 33(12): 1665-1675, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27638727

ABSTRACT

PURPOSE: The main goals of this study were to investigate the expression of anti-Müllerian hormone (AMH) and its receptor (AMHR2) during follicular development in primates, and to evaluate the potential of AMH as a biomarker for follicle growth and oocyte maturation in vitro. METHODS: The mRNA and protein expression of AMH and AMHR2 were determined using isolated follicles and ovarian sections from rhesus macaques (n = 4) by real-time PCR and immunohistochemistry, respectively. Isolated secondary follicles were cultured individually. Follicle growth and media AMH concentrations were assessed by ELISA. The mRNA expression profiles, obtained from RNA sequencing, of in vitro- and in vivo-developed antral follicles were compared. Secondary follicles from additional animals (n = 35) were cultured. Follicle growth, oocyte maturation, and media AMH concentrations were evaluated for forecasting follicular development in vitro by AMH levels. RESULTS: AMH immunostaining was heterogeneous in the population of preantral follicles that were also stained for AMHR2. The mRNA expression profiles were comparable between in vivo- and in vitro-developed follicles. AMH levels produced by growing follicles were higher than those of nongrowing follicles in culture. With a cutoff value of 1.40 ng/ml, 85 % of nongrowing follicles could be identified while eliminating only 5 % of growing follicles. Growing follicles that generated metaphase II-stage oocytes secreted greater amounts of AMH than did those yielding immature germinal vesicle-stage oocytes. CONCLUSIONS: AMH, co-expressed with AMHR2, was produced heterogeneously by preantral follicles in macaques with levels correlated positively with follicle growth and oocyte maturation. AMH may serve as a biomarker for primate follicular development in vitro.


Subject(s)
Anti-Mullerian Hormone/biosynthesis , In Vitro Oocyte Maturation Techniques , Ovarian Follicle/metabolism , Receptors, Peptide/biosynthesis , Receptors, Transforming Growth Factor beta/biosynthesis , Animals , Anti-Mullerian Hormone/genetics , Biomarkers/metabolism , Estradiol/metabolism , Female , Follicle Stimulating Hormone/biosynthesis , Follicle Stimulating Hormone/genetics , Humans , Macaca mulatta , Oocytes/growth & development , Oocytes/metabolism , Oogenesis/genetics , Ovarian Follicle/growth & development , Progesterone/metabolism , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/genetics
9.
J Assist Reprod Genet ; 33(4): 535-43, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26879207

ABSTRACT

PURPOSE: The objective of this study was to test the hypothesis that ovarian kisspeptin (kiss1) and its receptor (kiss1r) expression are affected by age, obesity, and the age- and obesity-related chemokine monocyte chemoattractant protein-1 (MCP-1). METHODS: Ovaries from reproductive-aged and older C57BL/6J mice fed normal chow (NC) or high-fat (HF) diet, ovaries from age-matched young MCP-1 knockout and young control mice on NC, and finally, cumulus and mural granulosa cells (GCs) from women who underwent in vitro fertilization (IVF) were collected. Kiss1, kiss1r, anti-Mullerian hormone (AMH), and AMH receptor (AMHR-II) messenger RNA (mRNA) expression levels were quantified using real-time polymerase chain reaction (RT-PCR). RESULTS: In mouse ovaries, kiss1 and kiss1r mRNA levels were significantly higher in old compared to reproductive-aged mice, and diet-induced obesity did not alter kiss1 or kiss1r mRNA levels. Compared to young control mice, young MCP-1 knockout mice had significantly lower ovarian kiss1 mRNA but significantly higher AMH and AMHR-II mRNA levels. In human cumulus GCs, kiss1r mRNA levels were positively correlated with age but not with BMI. There was no expression of kiss1 mRNA in either cumulus or mural GCs. CONCLUSION: These data suggest a possible age-related physiologic role for the kisspeptinergic system in ovarian physiology. Additionally, the inflammatory MCP-1 may be associated with kiss1 and AMH genes, which are important in ovulation and folliculogenesis, respectively.


Subject(s)
Aging/genetics , Chemokine CCL2/genetics , Kisspeptins/biosynthesis , Obesity/genetics , Receptors, G-Protein-Coupled/biosynthesis , Aging/pathology , Animals , Anti-Mullerian Hormone/biosynthesis , Anti-Mullerian Hormone/genetics , Diet, High-Fat , Female , Fertilization in Vitro/methods , Gene Expression Regulation , Granulosa Cells/metabolism , Humans , Kisspeptins/genetics , Mice , Mice, Knockout , Obesity/pathology , Ovary/metabolism , RNA, Messenger/biosynthesis , Receptors, G-Protein-Coupled/genetics , Receptors, Kisspeptin-1 , Receptors, Peptide/biosynthesis , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/biosynthesis , Receptors, Transforming Growth Factor beta/genetics
10.
Sci Rep ; 5: 17323, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26612176

ABSTRACT

In vitro follicle growth is a potential approach to preserve fertility for young women who are facing a risk of premature ovarian failure (POF) caused by radiation or chemotherapy. Our two-step follicle culture strategy recapitulated the dynamic human follicle growth environment in vitro. Follicles developed from the preantral to antral stage, and, for the first time, produced meiotically competent metaphase II (MII) oocytes after in vitro maturation (IVM).


Subject(s)
Fertilization in Vitro , Oocytes/growth & development , Ovarian Follicle/growth & development , Tissue Culture Techniques/methods , Adult , Alginates/chemistry , Anti-Mullerian Hormone/biosynthesis , Anti-Mullerian Hormone/metabolism , Estradiol/biosynthesis , Estradiol/metabolism , Female , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Hydrogels/chemistry , Metaphase , Oocytes/cytology , Oocytes/metabolism , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Primary Ovarian Insufficiency/prevention & control , Progesterone/biosynthesis , Progesterone/metabolism , Tissue Culture Techniques/instrumentation
11.
PLoS One ; 10(5): e0127786, 2015.
Article in English | MEDLINE | ID: mdl-26024525

ABSTRACT

INTRODUCTION: In vitro activation and growth of primordial dormant follicles to produce fertilizable oocytes would provide a useful instrument for fertility preservation. The employment of Phosphatase and TENsin homolog (PTEN) inhibitors, in combination with Protein kinase B (Akt) stimulating molecules, has been previously employed to increase follicular activation through the stimulation of the PTEN-Akt pathway. METHODS: We aim to establish improved in vitro activation also for cancer patients whose ovarian tissue has already been cryopreserved. Fresh and previously cryopreserved human ovarian cortex were exposed to short-term, low-concentration and ovary-specific treatment with only a PTEN inhibitor. RESULTS: Our in vitro activation protocol enhances the activation mechanisms of primordial follicles in both fresh and cryopreserved samples, and enlarges growing populations without inducing apoptosis in either follicles or the surrounding stroma. Treatment augments estradiol secretion and restores the expression levels of the previously diminished Anti-Müllerian hormone by means of cryopreservation procedures. Genomic modulation of the relative expression of PTEN pathway genes was found in treated samples. CONCLUSION: The in vitro activation protocol offers new alternatives for patients with cryopreserved tissue as it increases the pool of viable activated follicles available for in vitro growth procedures. The combination of ovarian tissue cryopreservation and in vitro activation of primordial follicles, the main ovarian reserve component, will be a major advancement in fertility preservation.


Subject(s)
Anti-Mullerian Hormone/biosynthesis , Cryopreservation , Enzyme Inhibitors/pharmacology , Neoplasms , Organ Preservation , Ovarian Follicle/metabolism , PTEN Phosphohydrolase/antagonists & inhibitors , Female , Humans , PTEN Phosphohydrolase/biosynthesis , Tissue Survival
12.
PLoS One ; 9(6): e99393, 2014.
Article in English | MEDLINE | ID: mdl-24932501

ABSTRACT

Anti-mullerian hormone (AMH) is thought to reflect the growth of follicles and the ovarian function. However, the role of AMH in culture medium during in vitro maturation (IVM) on oocyte quality and subsequent development potential is unclear. The objective of this study is to investigate the effect of recombinant human AMH (rh-AMH) supplemented into IVM medium on oocyte quality. Cumulus-oocyte complexes (COCs) were obtained from ICR mice and cultured in vitro with the different concentrations (0-1,000 ng/ml) of rh-AMH. Following 16-18 h of culture, quantitative PCR and ELISA were performed to analyze GDF9 and BMP15 mRNA expression and protein production from the oocytes. Subsequently, in vitro fertilization (IVF) and early embryonic development were employed to further evaluate the quality of in vitro matured oocytes. The results showed that AMH was only expressed in cumulus cells but not in the oocytes. However, AMH most specific receptor, AMHR-II, was expressed in both oocytes and cumulus cells. The levels of GDF9 and BMP15 expression and blastocyst formation rate were significantly increased (p<0.05) when the IVM medium was supplemented with 100 ng/ml of rh-AMH. With AdH1-SiRNA/AMH for knocking down of AMH expression during IVM significantly reduced (p<0.05) the levels of GDF9 and BMP15 expression and blastocysts formation rate. These results suggest that AHM improves oocytes quality by up-regulating GDF9 and BMP15 expressions during IVM.


Subject(s)
Anti-Mullerian Hormone/pharmacology , Oocytes/drug effects , Animals , Anti-Mullerian Hormone/administration & dosage , Anti-Mullerian Hormone/biosynthesis , Blastocyst/drug effects , Bone Morphogenetic Protein 15/biosynthesis , Bone Morphogenetic Protein 15/genetics , Cells, Cultured , Culture Media/pharmacology , Cumulus Cells/drug effects , Cumulus Cells/metabolism , Dose-Response Relationship, Drug , Embryonic Development , Female , Fertilization in Vitro , Gene Knockdown Techniques , Growth Differentiation Factor 9/biosynthesis , Growth Differentiation Factor 9/genetics , Mice , Mice, Inbred ICR , Oocytes/cytology , Oocytes/metabolism , Polymerase Chain Reaction , RNA, Small Interfering/pharmacology , Receptors, Peptide/biosynthesis , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/biosynthesis , Receptors, Transforming Growth Factor beta/genetics , Recombinant Proteins/pharmacology
13.
J Assist Reprod Genet ; 31(10): 1295-301, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24938362

ABSTRACT

PURPOSE: We explored whether AMH, as a surrogate for oocyte supply, varies by FMR1 genotype in women diagnosed with diminished ovarian reserve (DOR), a subset of the Primary Ovarian Insufficiency phenotype. Research is inconsistent on the relationship between AMH and FMR1 repeat length, controlling for age. METHOD: Seventy-nine cycling women diagnosed with DOR, and without a family history of fragile X syndrome, provided blood for FMR1 and AMH testing. DOR was defined as elevated FSH and/or low AMH and/or low antral follicle count, with regular menses. FMR1 CGG repeats were stratified by the larger allele <35 repeats (n = 70) v. ≥35 repeats (n = 9). Quadratic and linear models were fit to predict log (AMH) controlling for age. The AMH sample used as the outcome variable was drawn at a later date than the diagnostic AMH. RESULTS: Serum AMH concentration median was 0.30 ng/mL; Ages ranged from 26-43 years. A quadratic model (including age(2)) did not show a relationship with FMR1 CGG level (p-value = 0.25). A linear model of log (AMH), corresponding to an exponential decline of AMH with increasing age, was significantly different, and had a steeper slope, for women with ≥ 35 CGG repeats than women with < 35 repeats (p = 0.035). CONCLUSION: Findings suggest a greater rate of follicular loss that starts at later ages in women with DOR and ≥ 35 CGG repeats.


Subject(s)
Anti-Mullerian Hormone/biosynthesis , Fragile X Mental Retardation Protein/genetics , Ovarian Reserve/genetics , Trinucleotide Repeat Expansion/genetics , Adult , Female , Follicle Stimulating Hormone/blood , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Genotype , Humans , Oocytes/metabolism , Ovary/metabolism , Primary Ovarian Insufficiency/blood , Primary Ovarian Insufficiency/genetics
14.
Biomed Res Int ; 2014: 875438, 2014.
Article in English | MEDLINE | ID: mdl-24707500

ABSTRACT

BACKGROUND: A delayed time to pregnancy was recently reported for women who had a loop electrosurgical excision procedure (LEEP) to remove cervical intraepithelial neoplasia (CIN) grade 2 or 3. The objective of the current study was to determine if treatment of CIN with LEEP is associated with decreased levels of anti-Müllerian hormone (AMH), a marker of ovarian reserve. METHODS: AMH levels were measured in 18 women treated with LEEP and 18 age-matched controls, who had colposcopy only and did not require LEEP. Cases and controls had their blood drawn at study entry time zero and again 6 months later. RESULTS: The mean AMH level decreased significantly from baseline to follow-up; however, no significant differences were observed when stratifying by LEEP status, suggesting that both groups experienced a similar decrease in AMH levels during the follow-up period. Although women treated with LEEP had lower overall AMH levels than controls at both baseline and follow-up, these differences were not statistically significant. CONCLUSION: Overall, the delayed time to pregnancy observed in women treated with LEEP is likely not due to a LEEP-associated decrease in ovarian reserve as measured by AMH; thus, other mechanism are responsible for the delayed time to pregnancy associated with LEEP.


Subject(s)
Anti-Mullerian Hormone/metabolism , Cervix Uteri/metabolism , Electrosurgery/adverse effects , Uterine Cervical Dysplasia/metabolism , Anti-Mullerian Hormone/biosynthesis , Cervix Uteri/pathology , Cervix Uteri/surgery , Female , Humans , Pregnancy , Uterine Cervical Dysplasia/pathology , Uterine Cervical Dysplasia/surgery
15.
Toxicology ; 321: 89-95, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24769059

ABSTRACT

Tamoxifen, as well as most endocrine-disrupting chemicals, affects the reproductive system and sexual development, but little is known about its disruption of the molecular pathways regulating mammalian sex determination. In fetal mice, the expression levels and pattern of key genes involved in controlling sexually dimorphic balance were analyzed both in vivo and in vitro by using whole-mount in situ hybridization and quantitative-PCR. Developmental tamoxifen exposure induced abnormal up-regulation of the testis differentiation marker Pdfgra in Leydig cells and of Sox9 and Fgf9 in Sertoli cells in XX gonad. Immunohistochemistry analysis confirmed the over-expression of SOX9 protein. Accordingly, the ovary development marker Foxl2 was depressed at both the mRNA and protein levels. The increase in testosterone and the reduction in 17ß-estradiol and progesterone were observed by using the in vitro assay with organotypic cultures. Taken together, results indicated that tamoxifen induced the ectopic expression of well-established sex-specific genes during the critical developmental period, thus resulting in abnormal testicular development in the XX gonad of mammals. This study facilitates a better understanding of the molecular mechanisms of antiestrogens and possibly of compounds that interrupt estrogen signaling by other modes of action, and the association with the pathogenesis of human sexual developmental disorders.


Subject(s)
Antineoplastic Agents, Hormonal/toxicity , Endocrine Disruptors , Ovary/growth & development , Sex Determination Processes/drug effects , Tamoxifen/toxicity , Testis/growth & development , Analysis of Variance , Animals , Anti-Mullerian Hormone/biosynthesis , Anti-Mullerian Hormone/genetics , Female , Forkhead Box Protein L2 , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Gonadal Steroid Hormones/biosynthesis , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Inbred ICR , Organ Culture Techniques , Ovary/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Receptor, Platelet-Derived Growth Factor beta/genetics , SOX9 Transcription Factor/biosynthesis , SOX9 Transcription Factor/genetics , Sex Differentiation/drug effects , Testis/drug effects
16.
Fertil Steril ; 101(5): 1353-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24613539

ABSTRACT

OBJECTIVE: To evaluate antimüllerian hormone (AMH) and AMH receptor II (AMHRII) mRNA and protein expression in endometrium and in ovarian or deep lesions of women with endometriosis. DESIGN: Prospective study. SETTING: University hospitals in Italy and Brazil. PATIENTS: Patients with endometriosis (n = 55) and healthy women (n = 45). INTERVENTIONS: Specimens of endometrium obtained by hysteroscopy from patients with endometriosis and from healthy control subjects; specimens of ovarian endometriosis (n = 29) or of deep endometriosis (n = 26) were collected by laparoscopy. Serum samples were collected in some endometriotic patients (n = 23) and healthy control subjects (n = 20). MAIN OUTCOME MEASURE(S): AMH and AMHRII mRNA levels were evaluated by quantitative reverse-transcription polymerase chain reaction and protein localization by immunohistochemistry. AMH levels in tissue homogenates and in serum were assessed by ELISA. RESULT(S): Endometrium from women with endometriosis showed higher AMH and AMHRII mRNA levels than control women, with no significant differences between proliferative and secretory phases. Specimens collected from ovarian or deep endometriosis showed the highest AMH and AMHRII mRNA expression. Immunolocalization study confirmed the high AMH and AMHRII protein expression in endometriotic lesions. No difference of serum AMH levels between the groups was found. CONCLUSION(S): The increased AMH and AMHRII mRNA and protein expression in endometrium and in endometriotic lesions suggests a possible involvement of AMH in endometriosis.


Subject(s)
Anti-Mullerian Hormone/biosynthesis , Endometriosis/diagnosis , Endometriosis/metabolism , Gene Expression Regulation , Receptors, Peptide/biosynthesis , Receptors, Transforming Growth Factor beta/biosynthesis , Adult , Anti-Mullerian Hormone/blood , Biomarkers/blood , Biomarkers/metabolism , Endometriosis/surgery , Endometrium/metabolism , Endometrium/pathology , Female , Humans , Prospective Studies , Receptors, Peptide/blood , Receptors, Transforming Growth Factor beta/blood , Young Adult
17.
Hum Reprod ; 29(1): 146-54, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24256992

ABSTRACT

STUDY QUESTION: What are the effects of exposure of ovarian tissue to dehydroepiandrosterone (DHEA) supplementation in vivo? SUMMARY ANSWER: DHEA exposure stimulates initiation of primordial follicles and development of gonadotrophin-responsive preantral/early antral follicles possibly mediated through promoting granulosa cell proliferation and enhancing anti-Mullerian hormone (AMH) expression. WHAT IS KNOWN ALREADY?: Ovarian ageing is a cause of subfertility and is associated with poor outcomes of IVF treatment and premature menopause. A few clinical studies have shown that DHEA can improve ovarian response and increase the chances of pregnancy after IVF treatment in women with a diminished ovarian reserve (DOR) suggesting DHEA may help to overcome the effect of ovarian ageing. However, there are no data about how DHEA acts on ovarian folliculogenesis. STUDY DESIGN, SIZE AND DURATION: A cortical autograft experimental model was conducted in six female sheep aged at least 24 months. The period of DHEA treatment in the animals lasted for 10 weeks. PARTICIPANTS/MATERIALS, SETTING, METHODS: All the animals were subjected to unilateral oophorectomy. Half of the ovary was fixed for histological analysis as a time-zero control. The remaining tissue was used to isolate patches of ovarian cortex which were autografted back onto the ovarian pedicle. The grafting procedure eradicated all growing follicles and synchronized early follicular development. After a 10-week treatment period with DHEA implants, the ewes were sacrificed and the graft and remaining ovary were harvested. Histological and immunohistochemistry (IHC) findings, accompanied with serum hormonal profiles were compared to determine the effect on the follicle population. MAIN RESULTS AND THE ROLE OF CHANCE: Higher proportions of the follicle population in the remaining ovary were observed to be in the antral stage after DHEA treatment. The observation coincided with an increase in the rate of primordial follicle initiation and preantral follicle development in cortical grafts and the remaining ovarian tissue, respectively. The IHC results indicated that DHEA increased the expression of both the proliferation marker (KI-67) in granulosa cells and the follicular AMH expression at the preantral and early antral follicle stages. LIMITATIONS, REASONS FOR CAUTION: The experimental design compared follicle populations before and after DHEA treatment within individual animals to allow changes over time to be detected against a background of high inter-animal variation. However, since no controls without DHEA were included, we cannot say what would have happened over time in its absence, and it is possible that other factors may have resulted in the changes in follicle development observed during the experiment. WIDER IMPLICATIONS OF THE FINDING: Our data supports the idea that DHEA might be a useful therapy to delay the effects of ovarian ageing. Therefore, it may have a role as an adjunct during IVF to improve ovarian response in women with DOR and as a treatment for premature ovarian insufficiency. STUDY FUNDING/COMPETING INTEREST(S): The research received finance support from the University of Nottingham. The authors declare no conflict of interest in this study.


Subject(s)
Dehydroepiandrosterone/pharmacology , Ovarian Follicle/drug effects , Animals , Anti-Mullerian Hormone/biosynthesis , Autografts , Dehydroepiandrosterone/administration & dosage , Drug Implants , Female , Ovarian Follicle/cytology , Ovarian Follicle/physiology , Ovary/drug effects , Ovary/transplantation , Pregnancy , Sheep, Domestic
18.
Endocrinology ; 154(10): 3931-6, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24025226

ABSTRACT

Müllerian inhibiting substance (MIS, also known as anti-Müllerian hormone), is a key factor of male sex differentiation in vertebrates. In amniotes, it is responsible for Müllerian duct regression in male embryos. In fish, despite the absence of Müllerian ducts, MIS is produced and controls germ cell proliferation during gonad differentiation. Here we show for the first time the presence of MIS in an amphibian species, Pleurodeles waltl. This is very astonishing because in caudate amphibians, Müllerian ducts do not regress in males. Phylogenetic analysis of MIS P. waltl ortholog revealed that the deduced protein segregates with MIS from other vertebrates and is clearly separated from other TGF-ß family members. In larvae, MIS mRNA was expressed at higher levels in the developing testes than in the ovaries. In the testis, MIS mRNA expression was located within the lobules that contain Sertoli cells. Besides, expression of MIS was modified in the case of sex reversal: it increased after masculinizing heat treatment and decreased after estradiol feminizing exposure. In addition to the data obtained recently in the fish medaka, our results suggest that the role of MIS on Müllerian ducts occurred secondarily during the course of evolution.


Subject(s)
Amphibian Proteins/metabolism , Anti-Mullerian Hormone/metabolism , Gene Expression Regulation, Developmental , Ovary/metabolism , Pleurodeles/physiology , Testis/metabolism , Amphibian Proteins/biosynthesis , Amphibian Proteins/chemistry , Amphibian Proteins/genetics , Animals , Anti-Mullerian Hormone/biosynthesis , Anti-Mullerian Hormone/chemistry , Anti-Mullerian Hormone/genetics , Female , In Situ Hybridization , Larva/growth & development , Larva/metabolism , Liver/growth & development , Liver/metabolism , Male , Metamorphosis, Biological , Mullerian Ducts/growth & development , Mullerian Ducts/metabolism , Organ Culture Techniques , Ovary/growth & development , Phylogeny , Protein Structure, Tertiary , RNA, Messenger/metabolism , Sertoli Cells/cytology , Sertoli Cells/metabolism , Sex Differentiation , Testis/cytology , Testis/growth & development
19.
Reprod Biol ; 13(3): 221-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24011193

ABSTRACT

Successful antral formation in vitro from bovine preantral follicles (145-170 µm) has been described previously, but antrum formation from the primary follicle (50-70 µm) has not yet been achieved in vitro. The aim of the study was to establish an optimal culture system supporting the growth and maturation of bovine primary follicles (50-70 µm) in vitro. Bovine primary follicles were cultured in a three-dimensional culture system for 13 or 21 days in alpha-minimum essential medium. Various treatments including follicle stimulating hormone (FSH), luteinizing hormone (LH), 17ß-estradiol (E2), basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF) were tested. The follicular diameter and antrum formation rate were recorded, and follicular maturation markers (P450 aromatase, CYP19A1; anti-Mullerian hormone, AMH; growth differentiation factor-9, GDF9; bone morphogenetic protein-15, BMP15; and type III transforming growth factor ß receptor, TGFßR3) were analyzed by real-time RT-PCR. After 21 days of culture under each treatment condition, the follicular diameter was significantly enlarged in the presence of FSH + LH + E2 + bFGF or FSH + LH + E2 + bFGF + EGF (p<0.05). An addition of 50 ng/ml bFGF or bFGF +25 ng/ml EGF initiated antrum formation by day 19 and day 17 of culture, and the antral cavity formation rate was 16.7% and 33.3% by 21 days of culture, respectively. The expression of follicular maturation markers (CYP19A1, AMH, GDF9, BMP15 and TGFßR3) was significantly altered. We conclude that addition of 50 ng/ml bFGF +25 ng/ml EGF to media containing FSH + LH + E2 turned out to be the most effective optimized culture conditions to support the growth and maturation of bovine primary follicles in vitro.


Subject(s)
Ovarian Follicle/physiology , Tissue Culture Techniques/methods , Animals , Anti-Mullerian Hormone/biosynthesis , Aromatase/biosynthesis , Bone Morphogenetic Protein 15/biosynthesis , Cattle , Culture Media , Estradiol/pharmacology , Female , Follicle Stimulating Hormone/pharmacology , Growth Differentiation Factor 9/biosynthesis , Luteinizing Hormone/pharmacology , Ovarian Follicle/drug effects
20.
J Assist Reprod Genet ; 30(9): 1239-44, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23737215

ABSTRACT

PURPOSE: Poor ovarian reserve and poor ovarian response presents a challenge to IVF centers. Dehydroepiandrosterone (DHEA) supplementation is increasingly being used by many IVF centers around the world in poor responders despite the lack of convincing data. We therefore examined the rationale for the use of DHEA in poor responders, address the relevant studies, present new data, and address its potential mechanisms of action. METHODS: All published articles on the role of DHEA in infertile women from 1990 to April 2013 were reviewed. RESULTS: Several studies have suggested an improvement in pregnancy rates with the use of DHEA. Potential mechanisms include improved follicular steroidogenesis, increased IGF-1, acting as a pre-hormone for follicular testosterone, reducing aneuploidy, and increasing AMH and antral follicle count. While the role of DHEA is intriguing, evidence-based recommendations are lacking. CONCLUSIONS: While nearly 25 % of IVF programs use DHEA currently, large randomized prospective trials are sorely needed. Until (and if) such trials are conducted, DHEA may be of benefit in suitable, well informed, and consented women with diminished ovarian reserve.


Subject(s)
Dehydroepiandrosterone/administration & dosage , Infertility, Female/drug therapy , Insulin-Like Growth Factor I/biosynthesis , Testosterone/biosynthesis , Adult , Anti-Mullerian Hormone/biosynthesis , Female , Fertilization in Vitro , Humans , Infertility, Female/pathology , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...