Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Rev Bras Ginecol Obstet ; 43(6): 480-486, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34318473

ABSTRACT

The process of ovulation involves multiple and iterrelated genetic, biochemical, and morphological events: cessation of the proliferation of granulosa cells, resumption of oocyte meiosis, expansion of cumulus cell-oocyte complexes, digestion of the follicle wall, and extrusion of the metaphase-II oocyte. The present narrative review examines these interrelated steps in detail. The combined or isolated roles of the follicle-stimulating hormone (FSH) and luteinizing hormone (LH) are highlighted. Genes indiced by the FSH genes are relevant in the cumulus expansion, and LH-induced genes are critical for the resumption of meiosis and digestion of the follicle wall. A non-human model for follicle-wall digestion and oocyte release was provided.


O processo de ovulação envolve modificações genéticas, bioquímicas e morfológicas múltiplas e interrelacionadas: suspensão da proliferação das células da granulosa, reinício da meiose do oócito, expansão das células do complexo cumulus-oócito, digestão da parede folicular, e extrusão do oócito. Esta revisão narrativa examina em detalhes cada um desses eventos e os principais genes e proteínas envolvidos. Mais importante, a ação combinada ou isolada do hormônio folículo-estimulante (HFE) e do hormônio luteinizante (HL) é destacada. Detalha-se o papel do HFE na expansão do cumulus e do HL na digestão da parede folicular, permitindo a extrusão do oócito na superfície ovariana. Proveu-se um modelo não humano para explicar a digestão da parede folicular.


Subject(s)
Luteinizing Hormone/physiology , Ovulation/physiology , Animals , Cumulus Cells/physiology , Female , Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone/physiology , Granulosa Cells/physiology , Humans , Luteinizing Hormone/genetics , Meiosis/genetics , Meiosis/physiology , Models, Animal , Oocytes/growth & development , Ovarian Follicle/growth & development , Ovulation/genetics , Signal Transduction
2.
Rev. bras. ginecol. obstet ; Rev. bras. ginecol. obstet;43(6): 480-486, June 2021. graf
Article in English | LILACS | ID: biblio-1341135

ABSTRACT

Abstract The process of ovulation involves multiple and iterrelated genetic, biochemical, and morphological events: cessation of the proliferation of granulosa cells, resumption of oocyte meiosis, expansion of cumulus cell-oocyte complexes, digestion of the follicle wall, and extrusion of the metaphase-II oocyte. The present narrative review examines these interrelated steps in detail. The combined or isolated roles of the folliclestimulating hormone (FSH) and luteinizing hormone (LH) are highlighted. Genes indiced by the FSH genes are relevant in the cumulus expansion, and LH-induced genes are critical for the resumption ofmeiosis and digestion of the follicle wall. A nonhuman model for follicle-wall digestion and oocyte release was provided.


Resumo O processo de ovulação envolve modificações genéticas, bioquímicas e morfológicas múltiplas e interrelacionadas: suspensão da proliferação das células da granulosa, reinício da meiose do oócito, expansão das células do complexo cumulus-oócito, digestão da parede folicular, e extrusão do oócito. Esta revisão narrativa examina em detalhes cada um desses eventos e os principais genes e proteínas envolvidos. Mais importante, a ação combinada ou isolada do hormônio folículo-estimulante (HFE) e do hormônio luteinizante (HL) é destacada. Detalha-se o papel do HFE na expansão do cumulus e do HL na digestão da parede folicular, permitindo a extrusão do oócito na superfície ovariana. Proveu-se um modelo não humano para explicar a digestão da parede folicular.


Subject(s)
Humans , Animals , Female , Ovulation/physiology , Luteinizing Hormone/physiology , Oocytes/growth & development , Ovulation/genetics , Luteinizing Hormone/genetics , Signal Transduction , Models, Animal , Cumulus Cells/physiology , Follicle Stimulating Hormone/physiology , Follicle Stimulating Hormone/genetics , Ovarian Follicle/growth & development , Granulosa Cells/physiology , Meiosis/physiology , Meiosis/genetics
3.
Article in English | MEDLINE | ID: mdl-33385526

ABSTRACT

In this study, we measured aluminum (Al) bioconcentration in the brain, ovaries, and liver of Oreochromis niloticus females, and analyzed the effects of exposure to Al and acidic pH on the gene expression of follicle-stimulating hormone (ßfsh) and luteinizing hormone (ßlh) in these animals. Mature females were divided into 4 groups, thus being maintained for 96 h in one of the following conditions: control at neutral pH (Ctr); Al at neutral pH (Al); acidic pH (Ac), and Al at acidic pH (Al-Ac). pH alone did not influence Al bioconcentration in the brain. The animals from the Al-Ac group bioconcentrated more Al in the ovaries than those from the Al group, while no differences were observed in the liver. Aluminum bioconcentration was higher in the brain than in the liver and ovaries in Al-exposed animals (Al and Al-Ac), and higher in the brain than in the ovaries in the Ctr and Ac groups. The liver bioconcentrates more Al than the ovaries in the females from the Ctr and Ac groups. Aluminum and/or acidic pH did not alter ßfsh gene expression, while ßlh gene expression decreased in females from the Al group. Aluminum acted as an endocrine disruptor, suggesting deleterious effects in reproduction that could result in ovulation failure. Aluminum can act directly and/or indirectly in the pituitary, affecting ovarian steroidogenesis and altering the reproductive endocrine axis of mature O. niloticus females in an acute period of exposure.


Subject(s)
Aluminum/toxicity , Cichlids , Follicle Stimulating Hormone/metabolism , Luteinizing Hormone/metabolism , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Animals , Female , Follicle Stimulating Hormone/genetics , Gene Expression Regulation/drug effects , Hydrogen-Ion Concentration , Luteinizing Hormone/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
Sci Rep ; 10(1): 12840, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32732973

ABSTRACT

Currently, the generation of cell lines for the production of recombinant proteins has the limitation of unstable gene expression due to the repeat-induced gene silencing or the loss of transgene copies resulting from recombination events. In this work, we developed a new strategy based on the sequential insertion of transgenes for generating stable clones producing high levels of a chimeric human follicle-stimulating hormone (hscFSH). Gene insertion was done by transducing HEK-293 cells with a lentiviral vector containing a bicistronic transcriptional unit for expressing hscFSH and GFP genes. Clone selection was performed by flow cytometry coupled to cell sorting, and the GFP gene was further removed by CRE-mediated site-specific recombination. High-producing clones of hscFSH were obtained after three rounds of lentiviral transduction. Expression levels increased in a step-wise manner from 7 to 23 pg/cell/day, with a relatively constant rate of 7 pg/cell/day in each round of transduction. The GFP gene was successfully removed from the cell genome without disturbing the hscFSH gene expression. Clones generated using this approach showed stable expression levels for more than two years. This is the first report describing the sequential insertion of transgenes as an alternative for increasing the expression levels of transformed cell lines. The methodology described here could notably impact on biotechnological industry by improving the capacity of mammalian cells to produce biopharmaceuticals.


Subject(s)
Follicle Stimulating Hormone/biosynthesis , Mutagenesis, Insertional/methods , Transgenes/genetics , Biotechnology/methods , Clone Cells , Flow Cytometry/methods , Follicle Stimulating Hormone/genetics , Gene Expression , Genetic Vectors , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Lentivirus/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Transduction, Genetic
5.
Genet Test Mol Biomarkers ; 23(1): 39-44, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30585745

ABSTRACT

BACKGROUND: Follicle-stimulating hormone (FSH) is essential to the hypothalamic-pituitary-gonadal axis, playing a key role in human reproduction. It is a heterodimer comprised of a hormone-specific ß-chain (FSH-ß) that is associated with an α-chain. It exerts its biological activities by binding to the FSH receptor (FSHR). The ß-subunit, which is encoded by the FSHB gene, is responsible for ensuring binding specificity to the FSHR. There is a promoter polymorphism in this gene, c.-211G>T (rs10835638), upstream of the transcription start site; and in vitro studies have reported that the T allele decreases FSHB transcription in gonadotrophic cells. AIMS: Investigate the possible effects of the FSHB c.-211G/T polymorphism on hormonal profile and in in vitro fertilization (IVF)/intracytoplasmic sperm injection outcomes in normoovulatory Brazilian women. METHODS: A cross-sectional study of 140 women (median age = 33 years [CI: 32-34]) with infertility mainly caused by male (n = 85) or tuboperitoneal (n = 55) factors. In this study we evaluated FSH, estradiol, luteinizing hormone (LH), progesterone, prolactin and anti-Mullerian hormone levels, and antral follicle counting (AFC). Genotyping was performed using the TaqMan real-time polymerase chain reaction methodology. RESULTS: The wild-type allele G was found in 86.4% and the polymorphic allele T in 13.6% of the women respectively. The TT genotype was not found in any women. Women carrying the GT genotype had a poorer response more frequently to controlled ovarian hyperstimulation when compared to individuals with the GG genotype (47.4% vs. 26.5%, p = 0.010), higher LH levels (3.1 IU/mL vs. 2.4 IU/mL, p = <0.001), lower AFC (8.0 vs. 10.0, p = 0.03), oocytes retrieved (3.0 vs. 5.0, p = 0.03), MII (3.0 vs. 4.0, p = 0.02), and embryos (2.0 vs. 3.0, p = 0.02). Despite these findings, no difference was observed in pregnancy rate. CONCLUSION: Our findings suggest that the FSHB c.-211G/T polymorphism may modestly alter some aspects of the female reproductive system, but they are not associated with significantly different IVF outcomes.


Subject(s)
Carrier Proteins/genetics , Glycopeptides/genetics , Adult , Alleles , Anti-Mullerian Hormone/genetics , Brazil , Cross-Sectional Studies , Female , Fertilization in Vitro , Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone/metabolism , Follicle Stimulating Hormone, beta Subunit/genetics , Gene Frequency/genetics , Genotype , Humans , Luteinizing Hormone/genetics , Polymorphism, Single Nucleotide/genetics , Pregnancy , Pregnancy Rate , Promoter Regions, Genetic/genetics , Receptors, FSH , Reproductive Health
6.
Rev. bras. ciênc. avic ; 21(2): 1-8, 2019. tab, ilus, graf
Article in English | VETINDEX | ID: biblio-1490630

ABSTRACT

There is increasing evidence that bone morphogenetic protein 6 (BMP6) plays critical roles in regulating various stages of ovarian follicle development in mammals. However, the mechanisms of regulation of BMP6 in the chicken ovary remain unclear. In this study, mRNA and protein expression level of BMP6 in chicken ovarian follicles at different development stages were determined by qRT-PCR and western blot separately. Different concentrations of BMP6 protein and FSH were added to the culture medium, and the effects to proliferation of granulose cells were detected, further effect on expression pattern of progesterone synthesis associated genes were also analyzed by qRT-PCR and Western blotting and the secretion of progesterone was detected by ELISA. The results showed that mRNA and protein expression level of BMP6 increased significantly in the follicle with the development of follicle (p<0.05) and reached a peak at F1 follicle. Adding concentration of 50ng/ml and 100ng/ml of BMP6 protein promoted significantly the proliferation of granulosa cells (p<0.05), as well as up-regulated the expression of Steroid hormone synthesis acute regulatory protein (StAR) and 3β-hydroxysteroid dehydrogenase (3β-HSD) genes in mRNA and protein level. Meanwhile, the secretion of progesterone was significantly higher in the group that added BMP6 and FSH separately than blank control group (p<0.05) and reached a peak in the group that both added BMP6 and FSH. Collectively, these findings highlight that BMP6 is associated with proliferation of follicular cells and the synthesis of progesterone, which indicated that it took an important role in the follicular development of chicken.


Subject(s)
Animals , Granulosa Cells , Ovarian Follicle/growth & development , Chickens/growth & development , Chickens/genetics , Follicle Stimulating Hormone/analysis , Follicle Stimulating Hormone/genetics , Progesterone/analysis , Progesterone/genetics , /analysis , /genetics , China
7.
R. bras. Ci. avíc. ; 21(2): 1-8, 2019. tab, ilus, graf
Article in English | VETINDEX | ID: vti-23263

ABSTRACT

There is increasing evidence that bone morphogenetic protein 6 (BMP6) plays critical roles in regulating various stages of ovarian follicle development in mammals. However, the mechanisms of regulation of BMP6 in the chicken ovary remain unclear. In this study, mRNA and protein expression level of BMP6 in chicken ovarian follicles at different development stages were determined by qRT-PCR and western blot separately. Different concentrations of BMP6 protein and FSH were added to the culture medium, and the effects to proliferation of granulose cells were detected, further effect on expression pattern of progesterone synthesis associated genes were also analyzed by qRT-PCR and Western blotting and the secretion of progesterone was detected by ELISA. The results showed that mRNA and protein expression level of BMP6 increased significantly in the follicle with the development of follicle (p<0.05) and reached a peak at F1 follicle. Adding concentration of 50ng/ml and 100ng/ml of BMP6 protein promoted significantly the proliferation of granulosa cells (p<0.05), as well as up-regulated the expression of Steroid hormone synthesis acute regulatory protein (StAR) and 3β-hydroxysteroid dehydrogenase (3β-HSD) genes in mRNA and protein level. Meanwhile, the secretion of progesterone was significantly higher in the group that added BMP6 and FSH separately than blank control group (p<0.05) and reached a peak in the group that both added BMP6 and FSH. Collectively, these findings highlight that BMP6 is associated with proliferation of follicular cells and the synthesis of progesterone, which indicated that it took an important role in the follicular development of chicken.(AU)


Subject(s)
Animals , Chickens/growth & development , Chickens/genetics , Bone Morphogenetic Protein 6/analysis , Bone Morphogenetic Protein 6/genetics , Granulosa Cells , Progesterone/analysis , Progesterone/genetics , Ovarian Follicle/growth & development , Follicle Stimulating Hormone/analysis , Follicle Stimulating Hormone/genetics , China
8.
PLoS One ; 13(11): e0207526, 2018.
Article in English | MEDLINE | ID: mdl-30462715

ABSTRACT

Follicle-stimulating hormone receptor (FSHR) is a G-protein coupled receptor (GPCR) and a prototype of the glycoprotein hormone receptors subfamily of GPCRs. Structural data of the FSHR ectodomain in complex with follicle-stimulating hormone suggests a "pull and lift" activation mechanism that triggers a conformational change on the seven α-helix transmembrane domain (TMD). To analyze the conformational changes of the FSHR TMD resulting from sequence variants associated with reproductive impairment in humans, we set up a computational approach combining helix modeling and molecular simulation methods to generate conformational ensembles of the receptor at room (300 K) and physiological (310 K) temperatures. We examined the receptor dynamics in an explicit membrane environment of polyunsaturated phospholipids and solvent water molecules. The analysis of the conformational dynamics of the functional (N680 and S680) and dysfunctional (mutations at D408) variants of the FSHR allowed us to validate the FSHR-TMD model. Functional variants display a concerted motion of flexible intracellular regions at TMD helices 5 and 6. Disruption of side chain interactions and conformational dynamics were detected upon mutation at D408 when replaced with alanine, arginine, or tyrosine. Dynamical network analysis confirmed that TMD helices 2 and 5 may share communication pathways in the functional FSHR variants, whereas no connectivity was detected in the dysfunctional mutants, indicating that the global dynamics of the FSHR was sensitive to mutations at amino acid residue 408, a key position apparently linked to misfolding and variable cell surface plasma membrane expression of FSHRs with distinct mutations at this position.


Subject(s)
Amino Acids/chemistry , Follicle Stimulating Hormone/chemistry , Molecular Conformation , Receptors, FSH/chemistry , Alanine/chemistry , Alanine/genetics , Amino Acid Sequence/genetics , Amino Acids/genetics , Computer Simulation , Follicle Stimulating Hormone/genetics , Humans , Lipid Bilayers/chemistry , Lipids/chemistry , Lipids/genetics , Molecular Dynamics Simulation , Point Mutation , Protein Conformation, alpha-Helical , Protein Folding , Receptors, FSH/genetics , Signal Transduction
9.
PLoS One ; 13(7): e0197894, 2018.
Article in English | MEDLINE | ID: mdl-30063719

ABSTRACT

BACKGROUND: Anti-Müllerian hormone (AMH) is expressed by granulosa cells of developing follicles and plays an inhibiting role in the cyclic process of follicular recruitment by determining follicle-stimulating hormone threshold levels. Knowledge of AMH expression in the porcine ovary is important to understand the reproductive efficiency in female pigs. RESEARCH AIM: In the present study we investigated the expression of AMH during follicular development in prepubertal and adult female pigs by immunohistochemistry, laser capture micro-dissection and RT-qPCR. RESULTS AND CONCLUSION: Although in many aspects the immunohistochemical localization of AMH in the porcine ovary does not differ from other species, there are also some striking differences. As in most species, AMH appears for the first time during porcine follicular development in the fusiform granulosa cells of recruited primordial follicles and continues to be present in granulosa cells up to the antral stage. By the time follicles reach the pre-ovulatory stage, AMH staining intensity increases significantly, and both protein and gene expression is not restricted to granulosa cells; theca cells now also express AMH. AMH continues to be expressed after ovulation in the luteal cells of the corpus luteum, a phenomenon unique to the porcine ovary. The physiological function of AMH in the corpus luteum is at present not clear. One can speculate that it may contribute to the regulation of the cyclic recruitment of small antral follicles. By avoiding premature exhaustion of the ovarian follicular reserve, AMH may contribute to optimization of reproductive performance in female pigs.


Subject(s)
Anti-Mullerian Hormone/genetics , Corpus Luteum/metabolism , Follicle Stimulating Hormone/genetics , Genetic Fitness , Granulosa Cells/metabolism , Theca Cells/metabolism , Animals , Anti-Mullerian Hormone/metabolism , Corpus Luteum/cytology , Female , Follicle Stimulating Hormone/metabolism , Gene Expression Regulation, Developmental , Granulosa Cells/cytology , Immunohistochemistry , Ovulation/genetics , Pregnancy , Swine , Theca Cells/cytology
10.
J Cell Biochem ; 119(12): 10131-10142, 2018 12.
Article in English | MEDLINE | ID: mdl-30129147

ABSTRACT

Resveratrol (RSV), a polyphenolic compound largely found in red grape skin, has been used as a nutritional supplement as it exhibits beneficial health effects, such as anticancer, cardioprotective, antiaging, and anti-inflammatory. Particularly, it has been shown that it participates in the mechanisms involved in cell proliferation. Sirtuin 1 (SIRT1) is considered a well-known RSV effector. Noteworthy, Sirt1-knockout animals are infertile. The aim of this study was, first, to determine whether RSV has any effect on Sertoli cell proliferation and, second, whether SIRT1, a putative target of RSV, is present in immature Sertoli cells. Sertoli cell cultures obtained from 8-day-old rats, which actively proliferate, were treated with RSV (10 and 50 µM) under basal and follicle-stimulating hormone (FSH)-stimulated conditions. Bromodeoxyuridine (BrdU) incorporation and the expression of cyclins D1, D2, D3, E1, and E2 and the Cip/Kip cell cycle inhibitors p21Cip and p27Kip were analyzed. RSV decreased BrdU incorporation and cyclins D1, D2, E1, and E2 expression and increased p21Cip and p27Kip messenger RNA (mRNA) levels. RSV also decreased FSH-stimulated BrdU incorporation and cyclins D1 and D2 mRNA levels. The effect of RSV on cMYC was also analyzed. RSV treatment did not modify basal and FSH-stimulated cMyc expression; however, it inhibited basal and FSH-stimulated cMYC transcriptional activity, suggesting a role of cMYC in RSV effects. Additionally, Sirt1 was detected in immature Sertoli cells. Altogether, these results suggest that RSV possibly, by activating SIRT1 and regulating cMYC transcriptional activity, participates in the regulation of immature Sertoli cell proliferation.


Subject(s)
Cell Proliferation/drug effects , Proto-Oncogene Proteins c-myc/genetics , Resveratrol/pharmacology , Sertoli Cells/drug effects , Sirtuin 1/genetics , Animals , Cells, Cultured , Follicle Stimulating Hormone/genetics , Gene Expression Regulation/drug effects , Male , Neoplasm Proteins/genetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Sertoli Cells/pathology
11.
J Assist Reprod Genet ; 35(8): 1483-1488, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29916099

ABSTRACT

PURPOSE: Primary ovarian insufficiency (POI) is a clinical condition observed in women younger than 40 years of age, characterized by amenorrhea, hypoestrogenism, high levels of follicle-stimulating hormone (FSH), and infertility. Mutations in some master regulators of the development, maturation, and maintenance of ovarian follicles such as BMP15, FSHR, FOXL2, and GDF9 have been suggested as etiological factors in the development of POI. The aim of this study, the first in the Mexican population, is to evaluate the presence of mutations or polymorphisms in these four candidate genes. METHODS: In a sample of 20 Mexican patients with idiopathic POI, we looked for and analyzed genetic variants in BMP15, FSHR, FOXL2, and GDF9 genes. RESULTS: We observed two polymorphisms: a coding change, c.919G>A (p.Ala307Thr), in the FSHR gene and a synonymous variant, c.447C>T (p.Thr149Thr), in the GDF9 gene. These two variants have been reported previously as polymorphisms (rs6165 and rs254286, respectively). We observed no significant difference associated with POI in the patients when compared with a healthy control group (p > 0.05). Also, no exonic variants were found for the genes BMP15 and FOXL2 in the individuals tested. CONCLUSIONS: The lack of association of the evaluated genes in this sample of Mexican women is consistent with the complex genetic etiology of POI that is observed across cohorts studied thus far.


Subject(s)
Bone Morphogenetic Protein 15/genetics , Forkhead Box Protein L2/genetics , Genetic Association Studies , Primary Ovarian Insufficiency/genetics , Adult , Female , Follicle Stimulating Hormone/genetics , Genetic Predisposition to Disease , Genotype , Growth Differentiation Factor 9/genetics , Humans , Mexico/epidemiology , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Polymorphism, Single Nucleotide/genetics , Pregnancy , Primary Ovarian Insufficiency/epidemiology , Primary Ovarian Insufficiency/physiopathology , Receptors, FSH/genetics
12.
DNA Cell Biol ; 37(7): 600-608, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29746152

ABSTRACT

Over the past decades, an increase has been described in exposure to environmental toxins; consequently, a series of studies has been carried out with the aim of identifying problems associated with health. One of the main risk factors is exposure to heavy metals. The adverse effects that these compounds exert on health are quite complex and difficult to elucidate, in that they act at different levels and there are various signaling pathways that are implicated in the mechanisms of damage. The Sertoli cells plays a role of vital importance during the process of spermatogenesis, and it has been identified as one of the principal targets of heavy metals. In the present review, cadmium, lead, and arsenic are broached as altering the physiology of the Sertoli cells, citing mechanisms that have been cited in the literature.


Subject(s)
Arsenic/toxicity , Cadmium/toxicity , Environmental Pollutants/toxicity , Gene Expression Regulation/drug effects , Lead/toxicity , Sertoli Cells/drug effects , Apoptosis/drug effects , Apoptosis/genetics , Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone/metabolism , Humans , Luteinizing Hormone/genetics , Luteinizing Hormone/metabolism , Male , Sertoli Cells/cytology , Sertoli Cells/metabolism , Signal Transduction , Spermatogenesis/drug effects , Spermatogenesis/genetics , Testosterone/antagonists & inhibitors , Testosterone/genetics , Testosterone/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Endocrine ; 59(1): 191-202, 2018 01.
Article in English | MEDLINE | ID: mdl-29210006

ABSTRACT

Hypothyroidism and thyrotoxicosis produce adverse effects in male reproduction by unknown mechanisms. We investigated whether triiodothyronine (T3) modulates luteinizing hormone (LH) and follicle stimulating hormone (FSH) synthesis/secretion, by inducing different thyroid states. In hypothyroidism, the content of Lhb and Fshb mRNAs was increased, while their association to ribosomes and the protein content were reduced and the serum LH and FSH concentrations were augmented and decreased, respectively. Thyrotoxicosis reduced Lhb mRNA and LH serum concentration, and increased Lhb mRNA translational rate. The Fshb mRNA content and its association to ribosomes were also increased, whereas FSH serum concentrations were comparable to euthyroid levels. Acute T3 treatment decreased the total content of Lhb and Fshb mRNAs, and increased their association to ribosomes, as well as the LHB and FSHB contents in secretory granules. This study shows that T3 acts on gonadotrophs, resulting in direct effects on LH and FSH synthesis/secretion of male rats, suggesting that some reproductive disorders observed in men may be associated with thyroid hormone imbalances.


Subject(s)
Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone/metabolism , Luteinizing Hormone/genetics , Luteinizing Hormone/metabolism , Triiodothyronine/pharmacology , Animals , Gene Expression/drug effects , Hypothyroidism/genetics , Hypothyroidism/metabolism , Male , RNA, Messenger/metabolism , Rats , Rats, Wistar , Thyroidectomy
14.
J Clin Endocrinol Metab ; 102(5): 1740-1749, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28323923

ABSTRACT

Context: Although genetic factors play a pivotal role in male pubertal timing, genome-wide association studies have identified only a few loci. Genetic variation of follicle-stimulating hormone (FSH) action affects adult reproductive parameters and female pubertal timing. Objective: To investigate whether genetic variation affecting FSH action is associated with onset of puberty in boys. Design: Cross-sectional and longitudinal study of two cohorts of healthy boys. Setting: This was a population-based study. Patients or Other Participants: Danish (n = 1130) and Chilean (n = 424) boys were followed through puberty and genotyped for FSHB c.-211G>T, FSHR c.-29A>G, and FSHR c.2039G>A. Main Outcome Measures: Clinical pubertal staging including orchidometry, anthropometry, and serum gonadotropin levels. Results: Although the cohorts differed markedly (e.g., body composition and genotype frequencies), genetic variation affecting FSH production (FSHB c.-211G>T) was associated with age at pubertal onset, as assessed by testicular enlargement, in both cohorts. The effect appeared further modified by coexistence of genetic variation affecting FSH sensitivity (FSHR c.-29G>A): After correcting for body mass index (BMI), boys with a ligand-receptor variant combination resulting in weak FSH action (i.e., FSHB c.-211GT/TT and FSHR c.-29AA) entered puberty 0.64 years [95% confidence interval (CI), 0.12 to 1.17 years; Denmark] and 0.94 years (95% CI, 0.00 to 1.88 years; Chile) later than boys with the most effective FSH action. Effects explained 1.7% (Denmark) and 1.5% (Chile) of the variance. In addition, BMI z score was negatively associated with pubertal timing (ß = -0.35 years in both cohorts), explaining 17.2% (Denmark) and 7.2% (Chile) of the variance. Conclusion: In two ethnically distinct populations, we independently identified an association of two genetic loci with male pubertal timing.


Subject(s)
Follicle Stimulating Hormone, beta Subunit/genetics , Puberty/genetics , Receptors, FSH/genetics , Testis/growth & development , Adolescent , Age Factors , Child , Child, Preschool , Chile , Cross-Sectional Studies , Denmark , Follicle Stimulating Hormone/genetics , Genetic Variation , Genotype , Humans , Longitudinal Studies , Male , Polymorphism, Single Nucleotide , Young Adult
15.
Reprod Fertil Dev ; 29(6): 1144-1154, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27166082

ABSTRACT

The aim of the present study was to evaluate the effect of anti-Müllerian hormone (AMH), with and without FSH, on the in vitro development of isolated caprine preantral follicles, as well as follicular steroid production and mRNA levels of AMH, hormone receptors (AMH and FSH), CYP19A1 (cytochrome P450, family 19, subfamily A, polypeptide 1), CYP17 (cytochrome P450, family 17, subfamily A, polypeptide 1), HSD3B (3-beta-hydroxysteroid dehydrogenase) and Myc (myelocytomatosis oncogene). Isolated secondary follicles were cultured in minimum essential medium alpha (α-MEM+) alone or supplemented with 50ng mL-1 AMH and/or 100ng mL-1 FSH added sequentially on different days of culture. Follicles were cultured for a total of 18 days, with different media during the first (Days 0-9) and second (Days 10-18) halves of the culture period, resulting in six treatment groups, as follows: α-MEM+/α-MEM+, FSH/FSH, AMH/AMH, AMH+FSH/AMH+FSH, AMH/FSH, and FSH/AMH. Follicle development was evaluated on the basis of follicular growth, oocyte maturation and steroid secretion. There was a decrease in follicular growth rate in the AMH, AMH+FSH and AMH/FSH treatment groups compared with α-MEM+ and FSH treatment groups (P<0.05). However, the different culture conditions had no effect on rates of meiotic resumption and steroid secretion (P>0.05). Moreover, follicles cultured in the presence of FSH had lower levels of AMH receptor type II (AMHRII) mRNA compared with non-cultured control (freshly isolated follicles), and the AMH and AMH/FSH treatment groups. In conclusion, AMH reduces the follicular growth rate of isolated goat preantral follicles in vitro without affecting follicular survival.


Subject(s)
Anti-Mullerian Hormone/metabolism , Follicle Stimulating Hormone/metabolism , Gene Expression Regulation, Developmental , Oogenesis , Ovarian Follicle/metabolism , Receptors, FSH/agonists , Receptors, Peptide/agonists , Receptors, Transforming Growth Factor beta/agonists , Abattoirs , Animals , Anti-Mullerian Hormone/genetics , Anti-Mullerian Hormone/pharmacology , Brazil , Cattle , Cell Proliferation/drug effects , Cell Size/drug effects , Cell Survival/drug effects , Crosses, Genetic , Estradiol/metabolism , Female , Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone/pharmacology , Gene Expression Regulation, Developmental/drug effects , Goats , Humans , Oogenesis/drug effects , Ovarian Follicle/cytology , Ovarian Follicle/drug effects , Progesterone/metabolism , Receptors, FSH/genetics , Receptors, FSH/metabolism , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Testosterone/metabolism , Tissue Culture Techniques
16.
Einstein (Säo Paulo) ; 14(2): 231-234,
Article in English | LILACS | ID: lil-788039

ABSTRACT

ABSTRACT The ovarian hyperstimulation syndrome is the combination of increased ovarian volume, due to the presence of multiple cysts and vascular hyperpermeability, with subsequent hypovolemia and hemoconcentration. We report a case of spontaneous syndrome in a singleton pregnancy. This was a spontaneous pregnancy with 12 weeks of gestational age. The pregnancy was uneventful until 11 weeks of gestational age. After that, the pregnant woman complained of progressive abdominal distention associated with abdominal discomfort. She did not report other symptoms. In the first trimester, a routine ultrasonography showed enlarged ovaries, multiples cysts and ascites. Upon admission, the patient was hemodynamically stable, her serum β-hCG was 24,487mIU/mL, thyroid-stimulating hormone was 2.2µUI/mL and free T4 was 1.8ng/dL. All results were within normal parameters. However, levels of estradiol were high (10,562pg/mL). During hospitalization, she received albumin, furosemide and prophylactic dose of enoxaparin. The patient was discharged on the sixth hospital day.


RESUMO A síndrome de hiperestimulação ovariana é a combinação do aumento dos ovários, devido à presença de múltiplos cistos e de hiperpermeabilidade vascular, com subsequente hipovolemia e hemoconcentração. Relata-se um caso de síndrome espontânea em uma gestação única. Trata-se de gravidez espontânea com 12 semanas de idade gestacional. A gravidez ocorreu sem intercorrências até 11 semanas de idade gestacional. Após, a gestante passou a se queixar de distensão abdominal progressiva, associada com desconforto abdominal. A paciente não relatava outros sintomas. A ultrassonografia de rotina no primeiro trimestre mostrou ovários aumentados com múltiplos cistos e ascite. No momento da internação, a paciente apresentava-se hemodinamicamente estável, com β-hCG sérico de 24.487mUI/mL, hormônio estimulante da tireoide de 2,2µUI/m e T4 livre de 1,8ng/dL, ou seja, valores dentro dos parâmetros normais. Porém, os níveis de estradiol estavam elevados (10.562pg/mL). Durante a internação, a paciente recebeu albumina, furosemida e enoxaparina profilática. A alta hospitalar ocorreu no sexto dia de internação.


Subject(s)
Humans , Female , Pregnancy , Adult , Pregnancy Complications/physiopathology , Ovarian Hyperstimulation Syndrome/physiopathology , Pregnancy Complications/etiology , Pregnancy Complications/blood , Pregnancy Trimester, First , Gestational Age , Ovarian Hyperstimulation Syndrome/etiology , Ovarian Hyperstimulation Syndrome/blood , Estradiol/blood , Follicle Stimulating Hormone/genetics , Mutation
17.
Einstein (Sao Paulo) ; 14(2): 231-4, 2016 May 24.
Article in English, Portuguese | MEDLINE | ID: mdl-27223308

ABSTRACT

The ovarian hyperstimulation syndrome is the combination of increased ovarian volume, due to the presence of multiple cysts and vascular hyperpermeability, with subsequent hypovolemia and hemoconcentration. We report a case of spontaneous syndrome in a singleton pregnancy. This was a spontaneous pregnancy with 12 weeks of gestational age. The pregnancy was uneventful until 11 weeks of gestational age. After that, the pregnant woman complained of progressive abdominal distention associated with abdominal discomfort. She did not report other symptoms. In the first trimester, a routine ultrasonography showed enlarged ovaries, multiples cysts and ascites. Upon admission, the patient was hemodynamically stable, her serum ß-hCG was 24,487mIU/mL, thyroid-stimulating hormone was 2.2µUI/mL and free T4 was 1.8ng/dL. All results were within normal parameters. However, levels of estradiol were high (10,562pg/mL). During hospitalization, she received albumin, furosemide and prophylactic dose of enoxaparin. The patient was discharged on the sixth hospital day. RESUMO A síndrome de hiperestimulação ovariana é a combinação do aumento dos ovários, devido à presença de múltiplos cistos e de hiperpermeabilidade vascular, com subsequente hipovolemia e hemoconcentração. Relata-se um caso de síndrome espontânea em uma gestação única. Trata-se de gravidez espontânea com 12 semanas de idade gestacional. A gravidez ocorreu sem intercorrências até 11 semanas de idade gestacional. Após, a gestante passou a se queixar de distensão abdominal progressiva, associada com desconforto abdominal. A paciente não relatava outros sintomas. A ultrassonografia de rotina no primeiro trimestre mostrou ovários aumentados com múltiplos cistos e ascite. No momento da internação, a paciente apresentava-se hemodinamicamente estável, com ß-hCG sérico de 24.487mUI/mL, hormônio estimulante da tireoide de 2,2µUI/m e T4 livre de 1,8ng/dL, ou seja, valores dentro dos parâmetros normais. Porém, os níveis de estradiol estavam elevados (10.562pg/mL). Durante a internação, a paciente recebeu albumina, furosemida e enoxaparina profilática. A alta hospitalar ocorreu no sexto dia de internação.


Subject(s)
Ovarian Hyperstimulation Syndrome/physiopathology , Pregnancy Complications/physiopathology , Adult , Estradiol/blood , Female , Follicle Stimulating Hormone/genetics , Gestational Age , Humans , Mutation , Ovarian Hyperstimulation Syndrome/blood , Ovarian Hyperstimulation Syndrome/etiology , Pregnancy , Pregnancy Complications/blood , Pregnancy Complications/etiology , Pregnancy Trimester, First
18.
Genet Mol Res ; 14(3): 8252-61, 2015 Jul 27.
Article in English | MEDLINE | ID: mdl-26345751

ABSTRACT

Follicle-stimulating hormone (FSH), a glycoprotein secreted by the anterior pituitary, can regulate ovarian function through the FSH receptor (FSHR). To evaluate the effects of the FSHR gene on reproductive traits in pigs, polymorphisms in exon 10 of the FSHR gene were observed by polymerase chain reaction-single-strand conformation polymorphism, and 3 single nucleotide polymorphisms (C1491T, G1885A, and C1977T) in exon 10 of the porcine FSHR gene, and 3 genotypes (AA, AB, and BB) for C1491T and 2 haplotypes (D and E) for G1885A and C1977T were identified. Further analysis of single nucleotide polymorphism genotypes associated with reproductive traits including total number born (TNB) and number born alive (NBA) was carried out in 3 pig populations including Berkshire, Wannan Black (a Chinese indigenous pig breed), and BW pigs (two-way crossbred pigs produced from Berkshire ♂ and Wannan Black pig ♀). The results showed that the TNB and NBA of Wannan Black pigs with the AB genotype were significantly higher than in AA genotype sows (P < 0.01) in multiparity sows and all parities. The TNB and NBA of Berkshire pigs with the DE genotype were significantly higher than the DD and EE genotype sows (P < 0.01) in gilts, sows and all parities. Overall, TNB and NBA from the 3 identified genotypes was DE > DD > EE. The results showed that polymorphisms in exon 10 of the FSHR gene had a significant effect on litter size traits of Wannan Black and Berkshire pigs. These results can be applied for marker-assisted selection in the 2 swine breeds.


Subject(s)
Litter Size/genetics , Receptors, FSH/genetics , Reproduction/genetics , Swine/genetics , Animals , Female , Follicle Stimulating Hormone/genetics , Gene Frequency , Genotype , Polymorphism, Restriction Fragment Length , Polymorphism, Single Nucleotide
19.
Fish Physiol Biochem ; 41(4): 843-52, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25869217

ABSTRACT

Neuropeptide Y (NPY) is considered the most potent orexigenic peptide, increasing before meal time and during fasting. In teleost, most studies on NPY action upon growth hormone (GH) and luteinizing hormone (LH) were conducted in females or group of animals without sex discrimination. The aim of this study was to evaluate whether NPY modulates the expression and release of GH and gonadotropins in both sexes of Cichlasoma dimerus. By double-label immunofluorescence, we first determined the association between NPY fibers and pituitary cells. In addition, we performed in vitro studies to evaluate the effect of NPY on GH and gonadotropins expression by real-time PCR, and release by Western blot, in males and females separately. Contacts between NPY fibers and GH and follicle-stimulating hormone (FSH)-producing cells were detected, indicating possible functional relationships. We observed an increase in GH release in the culture medium at 2 nM for males (p = 0.043) and 20 nM for females (p = 0.028). Pituitary FSH release was stimulated at 20 nM (p = 0.026) and 200 nM (p = 0.033) for males and females, respectively. Finally, NPY only increased ß-LH mRNA expression at 20 nM in females (p = 0.028) and its release at 2 nM (p = 0.049) and 200 nM for males (p = 0.005) and 200 nM for females (p = 0.018). In conclusion, NPY acts as a GH-, LH- and FSH-releasing factor, in a dose- and sex-dependent way.


Subject(s)
Cichlids/metabolism , Follicle Stimulating Hormone/metabolism , Growth Hormone/metabolism , Luteinizing Hormone/metabolism , Neuropeptide Y/pharmacology , Pituitary Gland/drug effects , Animals , Female , Follicle Stimulating Hormone/genetics , Growth Hormone/genetics , Luteinizing Hormone/genetics , Male , Pituitary Gland/metabolism
20.
Cell Physiol Biochem ; 35(4): 1401-12, 2015.
Article in English | MEDLINE | ID: mdl-25790842

ABSTRACT

BACKGROUND: In human assisted reproduction, the ovarian response to exogenous recombinant Follicle-stimulating Hormone (FSH) therapy is variable and difficult to predict. The standard protocol of ovarian hyperstimulation can result in satisfactory response; however, an unsatisfactory response necessitates FSH dose adjustment or results in ovarian hyperstimulation syndrome (OHSS). Polymorphisms in AMH and AMHR2 genes appear to affect hormone biological activities, thus affecting follicle recruitment and development, leading to infertility. We aimed to evaluate AMH and AMHR2 polymorphisms in infertile women, and correlate those findings with AMH, FSH and estradiol serum level response to controlled ovarian hyperstimulation (COH), as well as assisted reproduction outcomes. METHODS: A cross-sectional study comprising 186 infertile women that underwent one cycle of high complexity assisted reproductive treatment. Blood samples were collected and a TaqMan assay was used for AMH G146T/rs10407022 and AMHR2 A-482G/rs2002555, A10G/rs11170555, C1749G/rs2071558 and G4952A/rs3741664 genotyping, and FSH, estradiol and AMH levels were measured. The findings were correlated to human reproduction outcomes. RESULTS: AMH rs10407022 and AMHR2 rs2002555 polymorphisms were not associated with hormonal measurements, whereas AMHR2 rs11170555 and rs3741664 were positively associated with AMH, estradiol and FSH levels. The genotype distribution of AMH and AMHR2 genes according to Controlled Ovarian Hyperstimulation did not show a positive association. However, an association with AFC, degree of oocyte maturation (allele G of AMHR2 rs2071558) the number of embryos produced (alleles T and G of AMH rs10407022 and AMHR2 rs2002555, respectively) and frozen embryo (allele G of AMHR2 rs11170555) were found to be statistically associated. Considering COH, serum AMH and AFC were a positive predictor to OHSS. Regarding serum AMH and assisted reproduction outcomes, a positive correlation with all variables studied was found. Comparing AFC and AMH as predictors of human reproduction outcomes, the AFC was less effective than serum AMH. Considering pregnancy rates, no marker was positively associated. CONCLUSION: AMHR2 polymorphisms were associated with estradiol, AMH and FSH measurements, as well as number and quality of embryos, while AMH polymorphisms was associated with number of embryos produced. Serum AMH was correlated with nearly all variables analyzed in assisted reproductive treatment, demonstrating that it represents a better biomarker of OHSS and human reproduction outcomes compared to AMH and AMHR2 polymorphisms.


Subject(s)
Anti-Mullerian Hormone/genetics , Infertility, Female/genetics , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/genetics , Adult , Alleles , Anti-Mullerian Hormone/blood , Cross-Sectional Studies , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone/pharmacology , Genotype , Humans , Infertility, Female/pathology , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Polymorphism, Single Nucleotide , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL