Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Elife ; 122023 Dec 27.
Article in English | MEDLINE | ID: mdl-38150309

ABSTRACT

Annual cycles in daylength provide an initial predictive environmental cue that plants and animals use to time seasonal biology. Seasonal changes in photoperiodic information acts to entrain endogenous programs in physiology to optimize an animal's fitness. Attempts to identify the neural and molecular substrates of photoperiodic time measurement in birds have, to date, focused on blunt changes in light exposure during a restricted period of photoinducibility. The objectives of these studies were first to characterize a molecular seasonal clock in Japanese quail and second, to identify the key transcripts involved in endogenously generated interval timing that underlies photosensitivity in birds. We hypothesized that the mediobasal hypothalamus (MBH) provides the neuroendocrine control of photoperiod-induced changes in reproductive physiology, and that the pars distalis of the pituitary gland contains an endogenous internal timer for the short photoperiod-dependent development of reproductive photosensitivity. Here, we report distinct seasonal waveforms of transcript expression in the MBH, and pituitary gland and discovered the patterns were not synchronized across tissues. Follicle-stimulating hormone-ß (FSHß) expression increased during the simulated spring equinox, prior to photoinduced increases in prolactin, thyrotropin-stimulating hormone-ß, and testicular growth. Diurnal analyses of transcript expression showed sustained elevated levels of FSHß under conditions of the spring equinox, compared to autumnal equinox, short (<12L) and long (>12L) photoperiods. FSHß expression increased in quail held in non-stimulatory short photoperiod, indicative of the initiation of an endogenously programmed interval timer. These data identify that FSHß establishes a state of photosensitivity for the external coincidence timing of seasonal physiology. The independent regulation of FSHß expression provides an alternative pathway through which other supplementary environmental cues, such as temperature, can fine tune seasonal reproductive maturation and involution.


Subject(s)
Coturnix , Follicle Stimulating Hormone, beta Subunit , Photoperiod , Reproduction , Coturnix/physiology , Follicle Stimulating Hormone, beta Subunit/physiology , Seasons , Male , Animals
2.
Neuroscience ; 298: 211-9, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-25907444

ABSTRACT

The association between a fit body and a fit brain in children has led to a rise of behavioral and neuroscientific research. Yet, the relation of cardiorespiratory fitness on premotor neurocognitive preparation with early visual processing has received little attention. Here, 41 healthy, lower and higher fit preadolescent children were administered a modified version of the Eriksen flanker task while electroencephalography (EEG) and behavioral measures were recorded. Event-related potentials (ERPs) locked to the stimulus onset with an earlier than usual baseline (-900/-800 ms) allowed investigation of both the usual post-stimulus (i.e., the P1, N1 and P2) as well as the pre-stimulus ERP components, such as the Bereitschaftspotential (BP) and the prefrontal negativity (pN component). At the behavioral level, aerobic fitness was associated response accuracy, with higher fit children being more accurate than lower fit children. Fitness-related differences selectively emerged at prefrontal brain regions during response preparation, with larger pN amplitude for higher than lower fit children, and at early perceptual stages after stimulus onset, with larger P1 and N1 amplitudes in higher relative to lower fit children. Collectively, the results suggest that the benefits of being aerobically fit appear at the stage of cognitive preparation prior to stimulus presentation and the behavioral response during the performance of a task that challenges cognitive control. Further, it is likely that enhanced activity in prefrontal brain areas may improve cognitive control of visuo-motor tasks, allowing for stronger proactive inhibition and larger early allocation of selective attention resources on relevant external stimuli.


Subject(s)
Cognition/physiology , Evoked Potentials, Visual/physiology , Follicle Stimulating Hormone, beta Subunit/physiology , Intention , Physical Fitness/physiology , Protein Precursors/physiology , Visual Perception/physiology , Analysis of Variance , Brain Mapping , Child , Electroencephalography , Female , Humans , Male , Neuropsychological Tests , Photic Stimulation , Reaction Time
3.
PLoS One ; 9(4): e94244, 2014.
Article in English | MEDLINE | ID: mdl-24718625

ABSTRACT

Follicle-Stimulating Hormone Receptor (FSHR) -29G/A polymorphism (rs1394205) was reported to modulate gene expression and reproductive parameters in women, but data in men is limited. We aimed to bring evidence to the effect of FSHR -29G/A variants in men. In Baltic young male cohort (n = 982; Estonians, Latvians, Lithuanians; aged 20.2 ± 2.0 years), the FSHR -29 A-allele was significantly associated with higher serum FSH (linear regression: effect 0.27 IU/L; P = 0.0019, resistant to Bonferroni correction for multiple testing) and showed a non-significant trend for association with higher LH (0.19 IU/L) and total testosterone (0.93 nmol/L), but reduced Inhibin B (-7.84 pg/mL) and total testes volume (effect -1.00 mL). Next, we extended the study and tested the effect of FSHR gene haplotypes determined by the allelic combination of FSHR -29G/A and a well-studied variant c.2039 A/G (Asn680Ser, exon 10). Among the FSHR -29A/2039G haplotype carriers (A-Ser; haplotype-based linear regression), this genetic effect was enhanced for FSH (effect 0.40 IU/L), Inhibin B (-16.57 pg/mL) and total testes volume (-2.34 mL). Finally, we estimated the total contribution of three known FSH-action modulating SNPs (FSHB -211G/T; FSHR -29G/A, c.2039 A/G) to phenotypic variance in reproductive parameters among young men. The major FSH-action modulating SNPs explained together 2.3%, 1.4%, 1.0 and 1.1% of the measured variance in serum FSH, Inhibin B, testosterone and total testes volume, respectively. In contrast to the young male cohort, neither FSHR -29G/A nor FSHR haplotypes appeared to systematically modulate the reproductive physiology of oligozoospermic idiopathic infertile patients (n = 641, Estonians; aged 31.5 ± 6.0 years). In summary, this is the first study showing the significant effect of FSHR -29G/A on male serum FSH level. To account for the genetic effect of known common polymorphisms modulating FSH-action, we suggest haplotype-based analysis of FSHR SNPs (FSHR -29G/A, c.2039 A/G) in combination with FSHB -211G/T testing.


Subject(s)
Follicle Stimulating Hormone, Human/blood , Follicle Stimulating Hormone, beta Subunit/physiology , Inhibins/blood , Oligospermia/genetics , Polymorphism, Single Nucleotide , Receptors, FSH/physiology , Testis/pathology , Testosterone/blood , 5' Untranslated Regions/genetics , Alleles , Baltic States , Follicle Stimulating Hormone, beta Subunit/genetics , Genetic Variation , Haplotypes , Humans , Male , Oligospermia/blood , Oligospermia/ethnology , Organ Size , Phenotype , Receptors, FSH/genetics , Young Adult
4.
Eur J Endocrinol ; 170(3): R91-107, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24288354

ABSTRACT

OBJECTIVE: To assess the pharmacogenetic potential of FSH for infertility treatment. DESIGN: Review of the literature and genomic databases. METHODS: Single-nucleotide polymorphism (SNP) assessed: rs6166 (c.2039A>G, p.N680S), rs6165 (c.919A>G, p.T307A), rs1394205 (c.-29G>A) in FSHR, and rs10835638 (c.-211G>T) in FSHB. Literature search via PubMed. Blast analysis of genomic information available in the NCBI nucleotide database. Comparison of allele frequency and haplotype distribution using the http://spsmart.cesga.estool. RESULTS: All these SNPs appear first in Homo, result in reduced FSH action, and are present with variable frequencies and combinations worldwide. Stringent clinical studies demonstrate that the FSHR genotype influences serum FSH levels and gonadal response in both sexes. Serum FSH levels depend on the -211G>T SNP, influencing transcriptional activity of the FSHB promoter. Genotypes reducing FSH action are overrepresented in infertile subjects. CONCLUSIONS: Although the clinical relevance of the FSHR polymorphisms alone is limited, the combination of FSHR and FSHB genotypes has a much stronger impact than either one alone in both sexes. About 20% of people are carriers of the alleles associated with lower serum FSH levels/reduced FSHR expression or activity, possibly less favorable for reproduction. Prospective studies need to investigate whether stratification of infertile patients according to their FSHR-FSHB genotypes improves clinical efficacy of FSH treatment compared with the current, naïve approach. A relative enrichment of less favorable FSHR-FSHB genotypes may be related to changes in human reproductive strategies and be a marker of some health-related advantage at the cost of reduced fertility.


Subject(s)
Follicle Stimulating Hormone, beta Subunit/genetics , Receptors, FSH/genetics , Adult , Animals , Female , Follicle Stimulating Hormone, beta Subunit/blood , Follicle Stimulating Hormone, beta Subunit/physiology , Gene Frequency , Genetics, Population , Humans , Infertility/genetics , Male , Ovulation Induction/methods , Pharmacogenetics , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Receptors, FSH/physiology , Reproductive Techniques, Assisted
5.
Biochem Biophys Res Commun ; 422(1): 54-8, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22561017

ABSTRACT

A direct effect of FSH on bone turnover via stimulation of osteoclast formation has been reported. Here we show that monoclonal or polyclonal antibodies to FSH inhibit osteoclast formation induced by FSH to an extent similar to that noted in FSH receptor (FSHR) knockout cells. Furthermore, we document the amplification of FSHR cDNA from well-characterized human CD14+ osteoclast precursors and osteoclasts, and the direct sequencing of the PCR products to definitively establish the expression of FSHRs. At these sites, the FSHR was expressed predominantly as an isoform that omits exon 9, a linker between the FSH-binding region and a long, invariant signaling domain of the receptor. These data provide compelling evidence for expression of a FSH receptor isoform in osteoclasts and their precursors.


Subject(s)
Bone Resorption/metabolism , Follicle Stimulating Hormone, beta Subunit/physiology , Osteoclasts/physiology , Receptors, FSH/biosynthesis , Alternative Splicing , Animals , Antibodies, Monoclonal , Base Sequence , Cells, Cultured , Female , Follicle Stimulating Hormone, beta Subunit/antagonists & inhibitors , Humans , Mice , Molecular Sequence Data , Osteoclasts/drug effects , Ovary , Receptors, FSH/genetics , Transcription, Genetic
6.
Hum Reprod ; 23(9): 2160-6, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18567894

ABSTRACT

BACKGROUND: No polymorphisms affecting serum FSH levels have been described in the human FSHB gene. We have identified a potential regulatory single nucleotide polymorphism (SNP, rs10835638; G/T) 211 bp upstream from the FSHB mRNA transcription start-site, located within a highly conserved region among placental mammals. We aimed to determine the correlation of carrier status of rs10835638 alternative alleles with serum FSH level in men, and testicular and hormonal parameters. METHODS: A quantitative genetic association study using a cohort of healthy men (n = 554; age 19.2 +/- 1.7 years) visiting the Centre of Andrology, Tartu University Hospital, Estonia. RESULTS: Rs10835638 (allele frequencies: G 87.6%, T 12.4%) was significantly associated with serum FSH level (analysis of variance: F = 13.0, P = 0.0016, df = 1; regression testing for a linear trend: P = 0.0003). Subjects with the GG genotype exhibited higher FSH levels (3.37 +/- 1.79 IU/l, n = 423) compared with heterozygotes (2.84 +/- 1.54 IU/l, n = 125) (P = 0.0005), the group of T-allele carriers (GT+TT, 2.78 +/- 1.51 IU/l, n = 131) (P = 0.0005) and TT-homozygotes (2.02 +/- 0.81 IU/L, n = 6) (P = 0.031). Rs10835638 was also associated with significant (P < 0.05) reduction in free testosterone index and testes volume, but increased semen volume, sex hormone-binding globulin, serum testosterone and estradiol. LH and inhibin-B levels did not differ significantly between groups. CONCLUSIONS: The identification of a regulatory SNP in FSHB promoter paves the way to study the effect of constitutively low FSH on male health and fertility. As FSH contributes to follicular development and sex steroid production in women, the role of this FSHB variant in female reproductive success is still to be addressed.


Subject(s)
Follicle Stimulating Hormone, beta Subunit/genetics , Follicle Stimulating Hormone/blood , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Adolescent , Adult , Base Sequence , Cohort Studies , Conserved Sequence , Evolution, Molecular , Follicle Stimulating Hormone, beta Subunit/physiology , Genotype , Humans , Male
7.
Biol Reprod ; 74(6): 993-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16481592

ABSTRACT

The frequency of gonadotropin-releasing hormone (GNRH1, or GnRH) pulses secreted from the hypothalamus determine the ratios of the gonadotropin subunit genes luteinizing hormone beta (Lhb), follicle-stimulating hormone beta (Fshb) and the common alpha-glycoprotein subunit gene (Cga) transcribed in the anterior pituitaries of mammals. Fshb is preferentially transcribed at slower GNRH1 pulse frequencies, whereas Lhb and Cga are preferentially transcribed at more rapid pulse frequencies. Producing the gonadotropins in the correct proportions is critical for normal fertility. Currently, there is no definitive explanation for how GNRH1 pulses differentially activate gonadotropin subunit gene transcription. Several pathways may contribute to this regulation. For example, GNRH1-regulated GNRH1-receptor concentrations may lead to variable signaling pathway activation. Several signaling pathways are activated by GnRH, including mitogen-activated protein kinase, protein kinase C, calcium influx, and calcium-calmodulin kinase, and these may be preferentially regulated under certain conditions. In addition, some signaling proteins feed back to downregulate their own levels. Other arms of gonadotroph signaling appear to be regulated by synthesis, modification, and degradation of either transcription factors or regulatory proteins. Finally, the dynamic binding of proteins to the chromatin, and how that might be regulated by chromatin-modifying proteins, is addressed. Oscillations in expression, modification, and chromatin binding of the proteins involved in gonadotropin gene expression are likely a link between GNRH1 pulsatility and differential gonadotropin transcription.


Subject(s)
Gene Expression Regulation/physiology , Gonadotropin-Releasing Hormone/physiology , Gonadotropins/genetics , Transcription, Genetic/physiology , Animals , Chromatin/physiology , Follicle Stimulating Hormone, beta Subunit/genetics , Follicle Stimulating Hormone, beta Subunit/metabolism , Follicle Stimulating Hormone, beta Subunit/physiology , Glycoprotein Hormones, alpha Subunit/genetics , Glycoprotein Hormones, alpha Subunit/metabolism , Glycoprotein Hormones, alpha Subunit/physiology , Gonadotropins/metabolism , Gonadotropins/physiology , Gonads/physiology , Humans , Hypothalamo-Hypophyseal System/physiology , Hypothalamus/metabolism , Luteinizing Hormone, beta Subunit/genetics , Luteinizing Hormone, beta Subunit/metabolism , Luteinizing Hormone, beta Subunit/physiology , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Transcription, Genetic/genetics
8.
J Soc Gynecol Investig ; 12(4): 257-62, 2005 May.
Article in English | MEDLINE | ID: mdl-15866117

ABSTRACT

OBJECTIVE: This research was conducted to investigate the potential of mouse embryonic stem (ES) cells to differentiate in vitro into gonadotropes. METHODS: Undifferentiated ES cells were maintained on mitomycin C-inactivated fibroblasts in the presence of leukemia inhibitory factor (LIF). By a 5-day hanging drop culture devoid of them, ES cells were induced to form multidifferentiated structures called embryoid bodies (EBs). Reverse transcriptase-polymerase chain reaction (RT-PCR), Western blotting, and immunocytochemistry were used to analyze gene expression of gonadotrope markers in EBs at different time points during the culture. RESULTS: Homeo box gene expressed in ES cells (Hesx1), LIM homeobox protein 3 (Lhx3), paired like homeodomain factor 1 (Prop1), GATA binding protein 2 (GATA2), follicle-stimulating hormone beta (FSHbeta), and luteinizing hormone beta (LHbeta) mRNAs were detected at day 6 EBs and maintained throughout the culture to day 56. FSHbeta and LHbeta proteins were expressed in EBs from day 6 onward. Immunofluorescent labeling of FSHbeta and LHbeta showed that specific staining was restricted to the cytoplasm of some differentiated EB cells. With the prolongation of EB culture, the number of positive cells increased significantly. Both monohormonal and bihormonal cells were present, mainly in clusters within EBs and sparsely distributed among the outermost cells surrounding the EBs. CONCLUSION: These results indicate that mouse ES cells can give rise to mature gonadotrope-like cells in EBs. It also shows that EBs may serve as a novel model system to study the development and function of gonadotropes.


Subject(s)
Cell Differentiation , Follicle Stimulating Hormone, beta Subunit/biosynthesis , Luteinizing Hormone, beta Subunit/biosynthesis , Pituitary Gland/cytology , Stem Cells/physiology , Animals , Blotting, Western , Cell Culture Techniques , Follicle Stimulating Hormone, beta Subunit/physiology , Immunohistochemistry , Luteinizing Hormone, beta Subunit/physiology , Mice , Reverse Transcriptase Polymerase Chain Reaction
9.
Neuroendocrinology ; 80(3): 158-70, 2004.
Article in English | MEDLINE | ID: mdl-15583473

ABSTRACT

In this study we have isolated the follicle-stimulating hormone beta subunit gene from the Chinook salmon (csFSHbeta). This gene encodes for a protein that is highly similar to those isolated from other salmonids and shares all of the structural constraints seen in mammalian gonadotropins, including twelve conserved cysteines and a putative N-linked glycosylation site. The organization of the gene follows the conserved pattern regarding the numbers and positions of the introns, although the csFSHbeta gene contains a particularly large 6.2-kb first intron due to the inclusion of several transposon-like elements. Isolation of 1.2 kb of the 5' flanking region of the csFSHbeta gene and subsequent analysis in silico have revealed a number of putative elements which appear highly conserved in teleost FSHbeta gene promoters and are thus likely involved in basal and hormone-induced transcriptional regulation. The functionality of this 1.2-kb fragment in driving expression of a reporter gene and its response to GnRH was shown in gonadotropes, while the overexpression of AP-1 factors, Sf-1, estrogen receptor or Smad1 revealed that the promoter is responsive to these transcription factors. Our current study has opened the way for future analysis to verify the role of these factors in mediating hormonally induced transcription of this gene.


Subject(s)
5' Flanking Region/genetics , Follicle Stimulating Hormone, beta Subunit/genetics , Follicle Stimulating Hormone, beta Subunit/isolation & purification , Promoter Regions, Genetic/genetics , Salmon/genetics , 5' Flanking Region/physiology , Amino Acid Sequence , Animals , Base Sequence , DNA, Complementary/analysis , Female , Follicle Stimulating Hormone, beta Subunit/physiology , Molecular Sequence Data , Promoter Regions, Genetic/physiology , Sequence Homology, Amino Acid , Transcription, Genetic
10.
Mol Endocrinol ; 17(3): 318-32, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12554780

ABSTRACT

FSH is controlled by a variety of positive and negative stimuli, and the unique FSHbeta-subunit is a major target for this regulation. Activin is a key modulator of FSHbeta transcription and hormone secretion. The signal transduction pathway leading to FSH expression was previously unknown. Here, we show that the transcription factors Smad3 and Smad4 mediate activin-stimulated activity of the rat FSHbeta promoter in a pituitary-derived cell line, LbetaT2. Cells were transiently transfected with the rat FSHbeta promoter fused to a luciferase reporter gene (-338rFSHbeta-Luc), and a minimal activin-responsive region was identified. Transfection of Smad3, but not the highly related Smad2, led to a ligand-independent stimulation of the FSHbeta promoter activity. As expected, activin caused an additional increase of luciferase expression, which was blocked by cotreatment with follistatin. Although Smad4 alone had no effect on FSHbeta transcription, it significantly augmented Smad3 and activin-mediated stimulation of the promoter. A palindromic consensus Smad-binding element in the proximal promoter was found to bind Smad4, and elimination of the region resulted in a loss of activin-mediated FSHbeta transcription. The activin signaling pathway is conserved in a number of cells, but FSHbeta expression is restricted to gonadotropes. A pituitary-specific transcription factor necessary for activin-dependent induction of the FSHbeta promoter has been identified that permits FSHbeta expression in nongonadotrope cells. Pitx2 is a member of Pitx subfamily of bicoid-related homeodomain factors that is required for pituitary development and is present in the adult pituitary. This factor was transfected into LbetaT2 cells, where it caused up-regulation of basal and activin-mediated FSHbeta promoter activity. Furthermore, cotransfection of Pitx2c with Smad3 in kidney-derived TSA cells resulted in activin-regulated FSHbeta response, suggesting its important role in tissue-restricted regulation of FSHbeta by activin. A Pitx2c binding site was identified within the proximal promoter, and elimination of this region also resulted in a loss of activin-regulated FSHbeta promoter activity. Taken together, these studies suggest that the regulation of FSHbeta is dependent on activin-mediated signaling factors in concert with pituitary-derived nuclear regulatory proteins.


Subject(s)
Activins/physiology , Follicle Stimulating Hormone, beta Subunit/physiology , Transcriptional Activation/physiology , Xenopus Proteins , Animals , Base Sequence , Cell Line , Cells, Cultured , DNA-Binding Proteins/physiology , Electrophoretic Mobility Shift Assay , Follicle Stimulating Hormone, beta Subunit/biosynthesis , Follicle Stimulating Hormone, beta Subunit/genetics , Haplorhini , Homeodomain Proteins/physiology , Humans , Molecular Sequence Data , Pituitary Gland/physiology , Promoter Regions, Genetic/physiology , Rats , Signal Transduction/genetics , Signal Transduction/physiology , Smad3 Protein , Trans-Activators/physiology , Transcription, Genetic/physiology , Transfection
11.
Endocrinology ; 144(1): 138-45, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12488339

ABSTRACT

Previous studies have suggested that FSH may be involved in regulation of Leydig cell function. We have examined this directly using two mouse models with null mutations in either the FSH beta-subunit (FSHbetaKO mice) or the FSH receptor (FSHRKO mice). Circulating LH levels were normal in adult FSHbetaKO mice, but were significantly increased in FSHRKO mice. Intratesticular testosterone levels increased normally in FSHbetaKO mice from birth to adulthood, whereas testosterone levels in FSHRKO mice failed to increase normally after puberty and were significantly reduced in adult animals. This was associated with reduced levels of mRNA encoding cytochrome P450 side-chain cleavage, 3beta-hydroxysteroid dehydrogenase type VI, and steroidogenic acute regulatory protein in FSHRKO mice. Leydig cell number was normal in FSHbetaKO mice during development, but in FSHRKO mice Leydig cell number increased slowly after puberty and was significantly reduced in the adult animal. Transfection studies showed that the FSHR exhibits constitutive activity in the absence of agonist stimulation. The results indicate, therefore, that Sertoli cells regulate the development of Leydig cell number and that constitutive activity within the FSHR is sufficient to stimulate this process. The presence of the hormone itself is not required when circulating LH levels are adequate.


Subject(s)
Follicle Stimulating Hormone, beta Subunit/deficiency , Leydig Cells/physiology , Receptors, FSH/deficiency , Testis/growth & development , Animals , Cyclic AMP/metabolism , Follicle Stimulating Hormone, beta Subunit/genetics , Follicle Stimulating Hormone, beta Subunit/physiology , Gene Expression , Leydig Cells/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Point Mutation , Progesterone/biosynthesis , RNA, Messenger/analysis , Receptors, FSH/genetics , Receptors, FSH/physiology , Sertoli Cells/physiology , Testis/chemistry , Testosterone/analysis , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...