Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Development ; 150(22)2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37870089

ABSTRACT

Macroheterogeneity in follicle-stimulating hormone (FSH) ß-subunit N-glycosylation results in distinct FSH glycoforms. Hypoglycosylated FSH21 is the abundant and more bioactive form in pituitaries of females under 35 years of age, whereas fully glycosylated FSH24 is less bioactive and increases with age. To investigate whether the shift in FSH glycoform abundance contributes to the age-dependent decline in oocyte quality, the direct effects of FSH glycoforms on folliculogenesis and oocyte quality were determined using an encapsulated in vitro mouse follicle growth system. Long-term culture (10-12 days) with FSH21 (10 ng/ml) enhanced follicle growth, estradiol secretion and oocyte quality compared with FSH24 (10 ng/ml) treatment. FSH21 enhanced establishment of transzonal projections, gap junctions and cell-to-cell communication within 24 h in culture. Transient inhibition of FSH21-mediated bidirectional communication abrogated the positive effects of FSH21 on follicle growth, estradiol secretion and oocyte quality. Our data indicate that FSH21 promotes folliculogenesis and oocyte quality in vitro by increasing cell-to-cell communication early in folliculogenesis, and that the shift in in vivo abundance from FSH21 to FSH24 with reproductive aging may contribute to the age-dependent decline in oocyte quality.


Subject(s)
Follicle Stimulating Hormone , Oocytes , Female , Mice , Animals , Follicle Stimulating Hormone/pharmacology , Follicle Stimulating Hormone/physiology , Ovarian Follicle , Cell Communication , Estradiol/pharmacology
2.
Am J Physiol Endocrinol Metab ; 326(5): E626-E639, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38536037

ABSTRACT

Loss of ovarian function imparts increased susceptibility to obesity and metabolic disease. These effects are largely attributed to decreased estradiol (E2), but the role of increased follicle-stimulating hormone (FSH) in modulating energy balance has not been fully investigated. Previous work that blocked FSH binding to its receptor in mice suggested this hormone may play a part in modulating body weight and energy expenditure after ovariectomy (OVX). We used an alternate approach to isolate the individual and combined contributions of FSH and E2 in mediating energy imbalance and changes in tissue-level metabolic health. Female Wistar rats were ovariectomized and given the gonadotropin releasing hormone (GnRH) antagonist degarelix to suppress FSH production. E2 and FSH were then added back individually and in combination for a period of 3 wk. Energy balance, body mass composition, and transcriptomic profiles of individual tissues were obtained. In contrast to previous studies, suppression and replacement of FSH in our paradigm had no effect on body weight, body composition, food intake, or energy expenditure. We did, however, observe organ-specific effects of FSH that produced unique transcriptomic signatures of FSH in retroperitoneal white adipose tissue. These included reductions in biological processes related to lipogenesis and carbohydrate transport. In addition, rats administered FSH had reduced liver triglyceride concentration (P < 0.001), which correlated with FSH-induced changes at the transcriptomic level. Although not appearing to modulate energy balance after loss of ovarian function in rats, FSH may still impart tissue-specific effects in the liver and white adipose tissue that might affect the metabolic health of those organs.NEW & NOTEWORTHY We find no effect of follicle-stimulating hormone (FSH) on energy balance using a novel model in which rats are ovariectomized, subjected to gonadotropin-releasing hormone antagonism, and systematically given back FSH by osmotic pump. However, tissue-specific effects of FSH on adipose tissue and liver were observed in this study. These include unique transcriptomic signatures induced by the hormone and a stark reduction in hepatic triglyceride accumulation.


Subject(s)
Energy Metabolism , Estradiol , Follicle Stimulating Hormone , Ovariectomy , Rats, Wistar , Animals , Female , Energy Metabolism/drug effects , Rats , Follicle Stimulating Hormone/metabolism , Estradiol/pharmacology , Body Composition/drug effects , Body Weight/drug effects , Ovary/drug effects , Ovary/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects , Liver/metabolism , Liver/drug effects , Transcriptome/drug effects
3.
Biol Reprod ; 110(1): 198-210, 2024 Jan 13.
Article in English | MEDLINE | ID: mdl-37812459

ABSTRACT

Di(2-ethylhexyl) phthalate and diisononyl phthalate are widely used as plasticizers in polyvinyl chloride products. Short-term exposures to phthalates affect hormone levels, ovarian follicle populations, and ovarian gene expression. However, limited data exist regarding the effects of long-term exposure to phthalates on reproductive functions. Thus, this study tested the hypothesis that short-term and long-term exposure to di(2-ethylhexyl) phthalate or diisononyl phthalate disrupts follicle dynamics, ovarian and pituitary gene expression, and hormone levels in female mice. Adult CD-1 female mice were exposed to vehicle, di(2-ethylhexyl) phthalate, or diisononyl phthalate (0.15 ppm, 1.5 ppm, or 1500 ppm) via the chow for 1 or 6 months. Short-term exposure to di(2-ethylhexyl) phthalate (0.15 ppm) and diisononyl phthalate (1.5 ppm) decreased serum follicle-stimulating hormone levels compared to control. Long-term exposure to di(2-ethylhexyl) phthalate and diisononyl phthalate (1500 ppm) increased the percentage of primordial follicles and decreased the percentages of preantral and antral follicles compared to control. Both phthalates increased follicle-stimulating hormone levels (di(2-ethylhexyl) phthalate at 1500 ppm; diisononyl phthalate at 1.5 ppm) and decreased luteinizing hormone levels (di(2-ethylhexyl) phthalate at 0.15 and 1.5 ppm; diisononyl phthalate at 1.5 ppm and 1500 ppm) compared to control. Furthermore, both phthalates altered the expression of pituitary gonadotropin subunit genes (Cga, Fshb, and Lhb) and a transcription factor (Nr5a1) that regulates gonadotropin synthesis. These data indicate that long-term exposure to di(2-ethylhexyl) phthalate and diisononyl phthalate alters follicle growth dynamics in the ovary and the expression of gonadotropin subunit genes in the pituitary and consequently luteinizing hormone and follicle-stimulating hormone synthesis.


Subject(s)
Diethylhexyl Phthalate , Phthalic Acids , Mice , Animals , Female , Phthalic Acids/toxicity , Diethylhexyl Phthalate/toxicity , Ovarian Follicle/metabolism , Follicle Stimulating Hormone/pharmacology , Luteinizing Hormone/metabolism
4.
Nature ; 546(7656): 107-112, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28538730

ABSTRACT

Menopause is associated with bone loss and enhanced visceral adiposity. A polyclonal antibody that targets the ß-subunit of the pituitary hormone follicle-stimulating hormone (Fsh) increases bone mass in mice. Here, we report that this antibody sharply reduces adipose tissue in wild-type mice, phenocopying genetic haploinsufficiency for the Fsh receptor gene Fshr. The antibody also causes profound beiging, increases cellular mitochondrial density, activates brown adipose tissue and enhances thermogenesis. These actions result from the specific binding of the antibody to the ß-subunit of Fsh to block its action. Our studies uncover opportunities for simultaneously treating obesity and osteoporosis.


Subject(s)
Adipose Tissue/metabolism , Adiposity , Follicle Stimulating Hormone, beta Subunit/antagonists & inhibitors , Thermogenesis , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue/drug effects , Adipose Tissue, Beige/drug effects , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adiposity/drug effects , Animals , Antibodies/immunology , Antibodies/pharmacology , Diet, High-Fat/adverse effects , Female , Follicle Stimulating Hormone, beta Subunit/immunology , Haploinsufficiency , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Obesity/drug therapy , Obesity/prevention & control , Osteoporosis/drug therapy , Ovariectomy , Oxygen Consumption/drug effects , Receptors, FSH/antagonists & inhibitors , Receptors, FSH/genetics , Receptors, FSH/metabolism , Thermogenesis/drug effects , Uncoupling Protein 1/biosynthesis
5.
Proc Natl Acad Sci U S A ; 117(46): 28971-28979, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33127753

ABSTRACT

Blocking the action of FSH genetically or pharmacologically in mice reduces body fat, lowers serum cholesterol, and increases bone mass, making an anti-FSH agent a potential therapeutic for three global epidemics: obesity, osteoporosis, and hypercholesterolemia. Here, we report the generation, structure, and function of a first-in-class, fully humanized, epitope-specific FSH blocking antibody with a KD of 7 nM. Protein thermal shift, molecular dynamics, and fine mapping of the FSH-FSH receptor interface confirm stable binding of the Fab domain to two of five receptor-interacting residues of the FSHß subunit, which is sufficient to block its interaction with the FSH receptor. In doing so, the humanized antibody profoundly inhibited FSH action in cell-based assays, a prelude to further preclinical and clinical testing.


Subject(s)
Adipose Tissue/metabolism , Antibodies, Blocking/immunology , Bone and Bones/metabolism , Epitopes , Follicle Stimulating Hormone/immunology , Animals , Antibodies, Blocking/chemistry , Antibodies, Monoclonal , Bone Density , Female , Follicle Stimulating Hormone/chemistry , Follicle Stimulating Hormone, beta Subunit/immunology , Humans , Hypercholesterolemia , Mice , Mice, Inbred C57BL , Molecular Dynamics Simulation , Obesity , Osteoporosis , Receptors, FSH/metabolism
6.
Int J Mol Sci ; 23(24)2022 Dec 11.
Article in English | MEDLINE | ID: mdl-36555365

ABSTRACT

In the mouse, two distinct populations of Leydig cells arise during testis development. Fetal Leydig cells arise from a stem cell population and produce T required for masculinization. It is debated whether they persist in the adult testis. A second adult Leydig stem cell population gives rise to progenitor-immature-mature adult type Leydig cells that produce T in response to LH to maintain spermatogenesis. In testis of adult null male mice lacking either only LH (Lhb-/-) or LHR (Lhr-/-), mature Leydig cells are absent but fetal Leydig cells persist. Thus, it is not clear whether other ligands signal via LHRs in Lhb null mice or LH signals via other receptors in the absence of LHR in Lhr null mice. Moreover, it is not clear whether truncated LHR isoforms generated from the same Lhr gene promoter encode functionally relevant LH receptors. To determine the in vivo roles of LH-LHR signaling pathway in the Leydig cell lineage, we generated double null mutant mice lacking both LH Ligand and all forms of LHR. Phenotypic analysis indicated testis morpho-histological characteristics are identical among double null and single mutants which all showed poorly developed interstitium with a reduction in Leydig cell number and absence of late stage spermatids. Gene expression analyses confirmed that the majority of the T biosynthesis pathway enzyme-encoding mRNAs expressed in Leydig cells were all suppressed. Expression of thrombospondin-2, a fetal Leydig cell marker gene was upregulated in single and double null mutants indicating that fetal Leydig cells originate and develop independent of LH-LHR signaling pathway in vivo. Serum and intratesticular T levels were similarly suppressed in single and double mutants. Consequently, expression of AR-regulated genes in Sertoli and germ cells were similarly affected in single and double mutants without any evidence of any additive effect in the combined absence of both LH and LHR. Our studies unequivocally provide genetic evidence that in the mouse testis, fetal Leydig cells do not require LH-LHR signaling pathway and a one-to-one LH ligand-LHR signaling pathway exists in vivo to regulate adult Leydig cell lineage and spermatogenesis.


Subject(s)
Leydig Cells , Testis , Mice , Male , Animals , Leydig Cells/metabolism , Ligands , Testis/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , Signal Transduction , Testosterone/metabolism
7.
Biol Reprod ; 103(3): 455-470, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32406908

ABSTRACT

Folliculogenesis is a complex process that requires integration of autocrine, paracrine, and endocrine factors together with tightly regulated interactions between granulosa cells and oocytes for the growth and survival of healthy follicles. Culture of ovarian follicles is a powerful approach for investigating folliculogenesis and oogenesis in a tightly controlled environment. This method has not only enabled unprecedented insight into the fundamental biology of follicle development but also has far-reaching translational applications, including in fertility preservation for women whose ovarian follicles may be damaged by disease or its treatment or in wildlife conservation. Two- and three-dimensional follicle culture systems have been developed and are rapidly evolving. It is clear from a review of the literature on isolated follicle culture methods published over the past two decades (1980-2018) that protocols vary with respect to species examined, follicle isolation methods, culture techniques, culture media and nutrient and hormone supplementation, and experimental endpoints. Here we review the heterogeneity among these major variables of follicle culture protocols.


Subject(s)
Ovarian Follicle/growth & development , Animals , Culture Media , Female , Fertility Preservation , Hormones/pharmacology , Humans , Oocytes , Oogenesis , Organ Culture Techniques , Ovarian Follicle/drug effects , Ovarian Follicle/physiology , Pregnancy
8.
Biol Reprod ; 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38302090
9.
11.
Reproduction ; 158(6): 517-527, 2019 12.
Article in English | MEDLINE | ID: mdl-31600726

ABSTRACT

Female reproductive aging is characterized by a rise in follicle-stimulating hormone (FSH) levels during peri-menopause. N-linked glycans are co-translationally attached to the Asn7 and Asn24 residues on the FSHß subunit. Differences in the number of N-glycans on the FSHß subunit result in distinct glycoforms: hypo-glycosylated (FSH21/18, glycans absent on either Asn24 or Asn7, respectively) or fully-glycosylated (FSH24, glycans present on both Asn7 and Asn24). The relative abundance of FSH glycoforms changes with advanced reproductive age, shifting from predominantly FSH21/18 in younger women to FSH24 in older women. Previous in vitro studies in granulosa cell lines and in vivo studies using Fshb-null mice showed these glycoforms elicit differential bioactivities. However, the direct effects of FSH glycoforms on the mouse ovarian follicle have not yet been determined. In this study, we isolated secondary follicles from pre-pubertal mice and treated them with 20- or 100 ng/mL purified recombinant FSH glycoforms for 1 h or 18-20 h. Analysis of phosphorylated PKA substrates showed that glycoforms were bioactive in follicles following 1-h treatment, although differential bioactivity was only observed with the 100 ng/mL dose. Treatment of follicles with 100 ng/mL of each glycoform also induced distinct expression patterns of FSH-responsive genes as assessed by qPCR, consistent with differential function. Our results, therefore, indicate that FSH glycoforms are bioactive in isolated murine follicles.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Follicle Stimulating Hormone/metabolism , Gene Expression Regulation , Granulosa Cells/metabolism , Ovarian Follicle/metabolism , Protein Processing, Post-Translational , Animals , Female , Follicle Stimulating Hormone/genetics , Glycosylation , Granulosa Cells/cytology , Mice , Ovarian Follicle/cytology , Phosphorylation
12.
Biol Reprod ; 108(6): 850-853, 2023 06 09.
Article in English | MEDLINE | ID: mdl-37186133
13.
Biol Reprod ; 109(4): 381-385, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37738136
14.
Biol Reprod ; 97(4): 622-626, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-29036567

ABSTRACT

Pituitary gonadotropes synthesize and secrete follicle-stimulating hormone (FSH). FSH is a heterodimer that consists of an α- and ß-subunit. The α-subunit is common to other pituitary and placental glycoprotein hormones, and the ß-subunit is the hormone/receptor-specific subunit. Although the pituitary is the main tissue that accounts for circulating hormone, previous and recent reports indicate extra-pituitary sources of FSH production including mouse gonads, human stomach, prostate, umbilical cord vein endothelial cells, uterine myometrium, placenta, and chicken abdominal adipose tissue. Whether extra-pituitary derived FSH exerts any physiologically significant actions is not known. In this review, we have comprehensively analyzed the expression of mRNAs that encode mouse and human FSH subunits and also their corresponding expressed sequence tags in normal tissues, cancer cell lines, and primary tumors by public database mining. We propose criteria to assess the significance of individual FSH subunit or FSH dimer expression as well as genetic approaches to unambiguously define the physiological relevance of extra-pituitary FSH expression.


Subject(s)
Follicle Stimulating Hormone/metabolism , Pituitary Gland/physiology , Animals , Female , Humans
15.
Proc Natl Acad Sci U S A ; 111(15): 5735-40, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24706813

ABSTRACT

FSH and luteinizing hormone (LH) are secreted constitutively or in pulses, respectively, from pituitary gonadotropes in many vertebrates, and regulate ovarian function. The molecular basis for this evolutionarily conserved gonadotropin-specific secretion pattern is not understood. Here, we show that the carboxyterminal heptapeptide in LH is a gonadotropin-sorting determinant in vivo that directs pulsatile secretion. FSH containing this heptapeptide enters the regulated pathway in gonadotropes of transgenic mice, and is released in response to gonadotropin-releasing hormone, similar to LH. FSH released from the LH secretory pathway rescued ovarian defects in Fshb-null mice as efficiently as constitutively secreted FSH. Interestingly, the rerouted FSH enhanced ovarian follicle survival, caused a dramatic increase in number of ovulations, and prolonged female reproductive lifespan. Furthermore, the rerouted FSH vastly improved the in vivo fertilization competency of eggs, their subsequent development in vitro and when transplanted, the ability to produce offspring. Our study demonstrates the feasibility to fine-tune the target tissue responses by modifying the intracellular trafficking and secretory fate of a pituitary trophic hormone. The approach to interconvert the secretory fate of proteins in vivo has pathophysiological significance, and could explain the etiology of several hormone hyperstimulation and resistance syndromes.


Subject(s)
Biological Evolution , Follicle Stimulating Hormone/metabolism , Gonadotrophs/metabolism , Luteinizing Hormone/metabolism , Ovary/physiology , Signal Transduction/physiology , Analysis of Variance , Animals , Blotting, Western , Female , Fertility/physiology , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Immunoelectron , Ovarian Follicle/metabolism , Ovary/metabolism , Ovulation/metabolism , Real-Time Polymerase Chain Reaction
16.
J Biol Chem ; 290(5): 2699-714, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25525274

ABSTRACT

Pituitary gonadotropins follicle-stimulating hormone and luteinizing hormone are heterodimeric glycoproteins expressed in gonadotropes. They act on gonads and promote their development and functions including steroidogenesis and gametogenesis. Although transcriptional regulation of gonadotropin subunits has been well studied, the post-transcriptional regulation of gonadotropin subunits is not well understood. To test if microRNAs regulate the hormone-specific gonadotropin ß subunits in vivo, we deleted Dicer in gonadotropes by a Cre-lox genetic approach. We found that many of the DICER-dependent microRNAs, predicted in silico to bind gonadotropin ß subunit mRNAs, were suppressed in purified gonadotropes of mutant mice. Loss of DICER-dependent microRNAs in gonadotropes resulted in profound suppression of gonadotropin-ß subunit proteins and, consequently, the heterodimeric hormone secretion. In addition to suppression of basal levels, interestingly, the post-gonadectomy-induced rise in pituitary gonadotropin synthesis and secretion were both abolished in mutants, indicating a defective gonadal negative feedback control. Furthermore, mutants lacking Dicer in gonadotropes displayed severely reduced fertility and were rescued with exogenous hormones confirming that the fertility defects were secondary to suppressed gonadotropins. Our studies reveal that DICER-dependent microRNAs are essential for gonadotropin homeostasis and fertility in mice. Our studies also implicate microRNAs in gonadal feedback control of gonadotropin synthesis and secretion. Thus, DICER-dependent microRNAs confer a new layer of transcriptional and post-transcriptional regulation in gonadotropes to orchestrate the hypothalamus-pituitary-gonadal axis physiology.


Subject(s)
DEAD-box RNA Helicases/metabolism , Gonadotrophs/metabolism , Gonadotropins/metabolism , Ribonuclease III/metabolism , Animals , DEAD-box RNA Helicases/genetics , Female , Fertility/genetics , Fertility/physiology , Gonadotropins/genetics , Male , Mice , Mice, Knockout , MicroRNAs/genetics , Rats , Real-Time Polymerase Chain Reaction , Ribonuclease III/genetics
17.
Carcinogenesis ; 35(4): 795-806, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24265293

ABSTRACT

Prolactin (PRL) is a secretory cytokine produced by various tissues. Binding to the cognate PRL receptor (PRLR), it activates intracellular signaling via janus kinase (JAK), extracellular signal-regulated kinase (ERK) and signal transducer and activator of transcription (STAT) proteins. PRL regulates diverse activities under normal and abnormal conditions, including malignancies. Previous clinical data suggest serum PRL levels are elevated in colorectal cancer (CRC) patients. In this study, we first determined the expression of PRL and PRLR in colon cancer tissue and cell lines. Higher levels of PRLR expression were observed in the cancer cells and cell lines compared with normal colonic epithelial cells. Incubation of colon cancer cells with PRL-induced JAK2, STAT3 and ERK1/2 phosphorylation and increased expression of Jagged 1, which is a Notch-1 receptor ligand. Notch signaling regulates CRC stem cell population. We observed increased accumulation of the cleaved/active form of Notch-1 receptor (Notch intracellular domain) and increased expression of Notch responsive genes HEY1, HES1 and stem cell marker genes DCLK1, LGR5, ALDH1 and CD44. Finally, inhibiting PRL induced JAK2-STAT3 and JAK2-ERK1/2 using AG490 and PD98059, respectively, leads to complete abrogation of Notch signaling, suggesting a role for this pathway in regulating CRC stem cells. Together, our results demonstrate that cytokine signaling induced by PRL is active in colorectal cancers and may provide a novel target for therapeutic intervention.


Subject(s)
Colonic Neoplasms/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Janus Kinase 2/metabolism , Neoplastic Stem Cells/metabolism , Prolactin/metabolism , Receptors, Notch/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Base Sequence , Colonic Neoplasms/metabolism , DNA Primers , Humans , Phosphorylation , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL