Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Sci Rep ; 12(1): 8624, 2022 05 21.
Article in English | MEDLINE | ID: mdl-35597810

ABSTRACT

The human luteinising hormone choriogonadotropin receptor (LHCGR) is a G-protein coupled receptor activated by both human chorionic gonadotropin (hCG) and luteinizing hormone (LH), two structurally related gonadotropins with essential roles in ovulation and maintenance of the corpus luteum. LHCGR expression predominates in ovarian tissues where it elicits functional responses through cyclic adenosine mononucleotide (cAMP), Ca2+ and extracellular signal-regulated kinase (ERK) signalling. LHCGR expression has also been localized to the human endometrium, with purported roles in decidualization and implantation. However, these observations are contentious. In this investigation, transcripts encoding LHCGR were undetectable in bulk RNA sequencing datasets from whole cycling endometrial tissue and cultured human endometrial stromal cells (EnSC). However, analysis of single-cell RNA sequencing data revealed cell-to-cell transcriptional heterogeneity, and we identified a small subpopulation of stromal cells with detectable LHCGR transcripts. In HEK-293 cells expressing recombinant LHCGR, both hCG and LH elicited robust cAMP, Ca2+ and ERK signals that were absent in wild-type HEK-293 cells. However, none of these responses were recapitulated in primary EnSC cultures. In addition, proliferation, viability and decidual transformation of EnSC were refractory to both hCG and LH, irrespective of treatment to induce differentiation. Although we challenge the assertion that LHCGR is expressed at a functionally active level in the human endometrium, the discovery of a discrete subpopulation of EnSC that express LHCGR transcripts may plausibly account for the conflicting evidence in the literature.


Subject(s)
Receptors, LH , Stromal Cells , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , HEK293 Cells , Humans , Receptors, G-Protein-Coupled , Receptors, LH/genetics , Receptors, LH/metabolism , Stromal Cells/metabolism
2.
Hum Reprod ; 35(1): 130-144, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31916571

ABSTRACT

STUDY QUESTION: Does an early proliferative phase endometrial biopsy harvested during ovarian stimulation harbour information predictive of the outcome following fresh embryo transfer (ET) in that same cycle? SUMMARY ANSWER: Transcriptome analysis of the whole-tissue endometrium did not reveal significant differential gene expression (DGE) in relation to the outcome; however, the secretome profile of isolated, cultured and in vitro decidualized endometrial stromal cells (EnSCs) varied significantly between patients who had a live birth compared to those with an implantation failure following fresh ET in the same cycle as the biopsy. WHAT IS KNOWN ALREADY: In the majority of endometrial receptivity research protocols, biopsies are harvested during the window of implantation (WOI). This, however, precludes ET in that same cycle, which is preferable as the endometrium has been shown to adapt over time. Endometrial biopsies taken during ovarian stimulation have been reported not to harm the chances of implantation, and in such biopsies DGE has been observed between women who achieve pregnancy versus those who do not. The impact of the endometrial proliferative phase on human embryo implantation remains unclear, but deserves further attention, especially since in luteal phase endometrial biopsies, a transcriptional signature predictive for repeated implantation failure has been associated with reduced cell proliferation, possibly indicating proliferative phase involvement. Isolation, culture and in vitro decidualization (IVD) of EnSCs is a frequently applied basic research technique to assess endometrial functioning, and a disordered EnSC secretome has previously been linked with failed implantation. STUDY DESIGN, SIZE, DURATION: This study was nested in a randomized controlled trial (RCT) investigating the effect of endometrial scratching during the early follicular phase of ovarian stimulation on clinical pregnancy rates after IVF/ICSI. Of the 96 endometrial biopsies available, after eliminating those without fresh ET and after extensive matching in order to minimize the risk of potential confounding, 18 samples were retained to study two clinical groups: nine biopsies of patients with a live birth versus nine biopsies of patients with an implantation failure, both following fresh ET performed in the same cycle as the biopsy. We studied the proliferative endometrium by analysing its transcriptome and by isolating, culturing and decidualizing EnSCs in vitro. We applied this latter technique for the first time on proliferative endometrial biopsies obtained during ovarian stimulation for in-cycle outcome prediction, in an attempt to overcome inter-cycle variability. PARTICIPANTS/MATERIALS, SETTING, METHODS: RNA-sequencing was performed for 18 individual whole-tissue endometrial biopsies on an Illumina HiSeq1500 machine. DGE was analysed three times using different approaches (DESeq2, EdgeR and the Wilcoxon rank-sum test, all in R). EnSC isolation and IVD was performed (for 2 and 4 days) for a subset of nine samples, after which media from undifferentiated and decidualized cultures were harvested, stored at -80°C and later assayed for 45 cytokines using a multiplex suspension bead immunoassay. The analysis was performed by partial least squares regression modelling. MAIN RESULTS AND THE ROLE OF CHANCE: After correction for multiple hypothesis testing, DGE analysis revealed no significant differences between endometrial samples from patients who had a live birth and those with an implantation failure following fresh ET. However secretome analysis after EnSC isolation and culture, showed two distinct clusters that clearly corresponded to the two clinical groups. Upon IVD, the secretome profiles shifted from that of undifferentiated cells but the difference between the two clinical groups remained yet were muted, suggesting convergence of cytokine profiles after decidualization. LIMITATIONS, REASONS FOR CAUTION: Caution is warranted due to the limited sample size of the study and the in vitro nature of the EnSC experiment. Validation on a larger scale is necessary, however, hard to fulfil given the very limited availability of in-cycle proliferative endometrial biopsies outside a RCT setting. WIDER IMPLICATIONS OF THE FINDINGS: These data support the hypothesis that the endometrium should be assessed not only during the WOI and that certain endometrial dysfunctionalities can probably be detected early in a cycle by making use of the proliferative phase. This insight opens new horizons for the development of endometrial tests, whether diagnostic or predictive of IVF/ICSI treatment outcome. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by Fonds Wetenschappelijk Onderzoek (FWO, Flanders, Belgium, 11M9415N, 1 524 417N), Wetenschappelijk Fonds Willy Gepts (WFWG G160, Universitair Ziekenhuis Brussel, Belgium) and the National Medicine Research Council (NMRC/CG/M003/2017, Singapore). There are no conflicts of interests. TRIAL REGISTRATION NUMBER: NCT02061228.


Subject(s)
Embryo Transfer , Sperm Injections, Intracytoplasmic , Belgium , Endometrium , Female , Humans , Pregnancy , Singapore
3.
Oncogene ; 38(25): 5111-5112, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30867566

ABSTRACT

In the published version of this article, the images for cytoplasmic and nuclear FGF7 in MDA-MB-231 cells were duplicated and mistaken for total FGF7 in SKBR-3 and MDA-MB-231 cells.

4.
Mol Hum Reprod ; 24(7): 375-387, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29846687

ABSTRACT

STUDY QUESTION: What are the changes in human embryos, in terms of morphology and gene expression, upon attachment to endometrial epithelial cells? SUMMARY ANSWER: Apposition and adhesion of human blastocysts to endometrial epithelial cells are predominantly initiated at the embryonic pole and these steps are associated with changes in expression of adhesion and extracellular matrix (ECM) genes in the embryo. WHAT IS KNOWN ALREADY: Both human and murine embryos have been co-cultured with Ishikawa cells, although embryonic gene expression associated with attachment has not yet been investigated in an in vitro implantation model. STUDY DESIGN, SIZE, DURATION: Vitrified human blastocysts were warmed and co-cultured for up to 48 h with Ishikawa cells, a model cell line for receptive endometrial epithelium. PARTICIPANTS/MATERIALS, SETTING, METHODS: Six days post-fertilization (6dpf) human embryos were co-cultured with Ishikawa cells for 12, 24 (7dpf) or 48 h (8dpf) and attachment rate and morphological development investigated. Expression of 84 adhesion and ECM genes was analysed by quantitative PCR. Immunofluorescence microscopy was used to assess the expression of three informative genes at the protein level. Data are reported on 145 human embryos. Mann-Whitney U was used for statistical analysis between two groups, with P < 0.05 considered significant. MAIN RESULTS AND THE ROLE OF CHANCE: The majority of embryos attached to Ishikawa cells at the level of the polar trophectoderm; 41% of co-cultured embryos were loosely attached after 12 h and 86% firmly attached after 24 h. Outgrowth of hCG-positive embryonic cells at 8dpf indicated differentiation of trophectoderm into invasive syncytiotrophoblast. Gene expression analysis was performed on loosely attached and unattached embryos co-cultured with Ishikawa cells for 12 h. In contrast to unattached embryos, loosely attached embryos expressed THBS1, TNC, COL12A1, CTNND2, ITGA3, ITGAV and LAMA3 and had significantly higher CD44 and TIMP1 transcript levels (P = 0.014 and P = 0.029, respectively). LAMA3, THBS1 and TNC expressions were validated at the protein level in firmly attached 7dpf embryos. Thrombospondin 1 (THBS1) resided in the cytoplasm of embryonic cells whereas laminin subunit alpha 3 (LAMA3) and tenascin C (TNC) were expressed on the cell surface of trophectoderm cells. Incubation with a neutralizing TNC antibody did not affect the rate of embryo attachment or hCG secretion. LARGE SCALE DATA: None. LIMITATIONS, REASONS FOR CAUTION: This in vitro study made use of an endometrial adenocarcinoma cell line to mimic receptive luminal epithelium. Also, the number of embryos was limited. Contamination of recovered embryos with Ishikawa cells was unlikely based on their differential gene expression profiles. WIDER IMPLICATIONS OF THE FINDINGS: Taken together, we provide a 'proof of concept' that initiation of the implantation process coincides with the induction of specific embryonic genes. Genome-wide expression profiling of a larger sample set may provide insights into the molecular embryonic pathways underlying successful or failed implantation. STUDY FUNDING AND COMPETING INTEREST(S): A.A. was supported by a grant from the 'Instituut voor Innovatie door Wetenschap en Technologie' (IWT, 121716, Flanders, Belgium). This work was supported by the 'Wetenschappelijk Fonds Willy Gepts' (WFWG G142 and G170, Universitair Ziekenhuis Brussel). The authors declare no conflict of interest.


Subject(s)
Blastocyst/metabolism , Cell Adhesion Molecules/genetics , Coculture Techniques/methods , Embryo Culture Techniques/methods , Embryo Implantation/genetics , Extracellular Matrix Proteins/genetics , Blastocyst/cytology , Blastocyst/physiology , Cell Adhesion/genetics , Cell Adhesion Molecules/metabolism , Cells, Cultured , Embryo, Mammalian , Embryonic Development/genetics , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Female , Gene Expression Regulation, Developmental , Humans
5.
Mol Endocrinol ; 29(10): 1454-67, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26280733

ABSTRACT

Progesterone (P4) maintains uterine quiescence during pregnancy and its functional withdrawal is associated with increased prostaglandin synthesis and the onset of labor. In primary human myometrial cells, the glucocorticoid receptor (GR) rather than the P4 receptor mediates P4 antagonism of IL-1ß-induced cyclooxygenase-2 (COX-2) expression, the rate-limiting enzyme in prostaglandin synthesis. We now report that P4 also acts via GR to induce MAPK phosphatase (MKP)-1 and knockdown of MKP-1 impairs the ability of P4 to repress IL-1ß-dependent COX-2 induction. Microarray analysis revealed that P4 repressed preferentially activator protein-1-responsive genes in response to IL-1ß. Consistent with these observations, we found that the ability of P4 to reduce c-Jun activation was lost upon GR as well as MKP-1 knockdown. Interestingly, c-Jun levels in human myometrial cells declined upon GR and MKP-1 knockdown, which suggests the presence of an activator protein-1 feedback loop. This is supported by our observation that c-Jun levels declined after an initial rise in primary myometrial cells treated with phorbol 12-myrisatate 13-acetate, a potent activator of c-Jun N-terminal kinase. Finally, we show that MKP-1 is an intermediate in P4-mediated repression of some but not all IL-1ß-responsive genes. For example, P4 repression of IL11 and IRAK3 was maintained upon MKP-1 knockdown. Taken together, the data show that P4 acts via GR to drive MKP-1 expression, which in turn inhibits IL-1ß-dependent c-Jun activation and COX-2 expression.


Subject(s)
Dual Specificity Phosphatase 1/metabolism , Inflammation/pathology , Myometrium/pathology , Progesterone/pharmacology , Transcription Factor AP-1/metabolism , Cyclooxygenase 2/metabolism , Feedback, Physiological/drug effects , Female , Gene Knockdown Techniques , Humans , Interleukin-1beta/pharmacology , Models, Biological , Myometrium/drug effects , Myometrium/metabolism , RNA, Small Interfering/metabolism , Receptors, Glucocorticoid/metabolism
6.
Placenta ; 36(4): 341-4, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25637411

ABSTRACT

The fetal endometrium becomes responsive to steroid hormones around the fourth month of pregnancy starting with an oestrogenic phase, which is followed late in pregnancy by a secretory phase. Based on post-mortem studies, the endometrium at birth is secretory in only one-third of neonates and proliferative in the remaining cases. Decidual or menstrual changes are rare in fetal endometrium despite high circulating steroid hormone levels, which drop rapidly after birth. Hence, acquisition of progesterone responsiveness appears to be dependent on endometrial maturation and relative immaturity may persist in a majority of girls until the menarche and early adolescence. Two major reproductive disorders have been linked with either advanced or delayed endometrial maturation. First, early-onset endometriosis may be caused by menstruation-like bleeding in the neonate, leading to tubal reflux and ectopic implantation of endometrial stem/progenitor cells. Second, persistence of partial progesterone resistance in adolescent girls may compromise deep placentation and account for the increased risk of major obstetrical syndromes, including preeclampsia, fetal growth retardation and preterm birth. The concept of neonatal origins of common reproductive disorders poses important research challenges but also subsumes potential new preventative strategies.


Subject(s)
Endometriosis/congenital , Endometrium/metabolism , Fetal Development , Models, Biological , Pregnancy Complications/etiology , Progesterone/metabolism , Adolescent , Adolescent Medicine/trends , Animals , Biomedical Research/trends , Endometriosis/immunology , Endometriosis/metabolism , Endometriosis/physiopathology , Endometrium/immunology , Female , Genital Diseases, Female/congenital , Genital Diseases, Female/immunology , Genital Diseases, Female/metabolism , Genital Diseases, Female/physiopathology , Humans , Infant, Newborn , Perinatology/trends , Placentation , Pregnancy , Pregnancy Complications/immunology , Pregnancy Complications/metabolism , Pregnancy Complications/prevention & control , Reproductive Medicine/trends
7.
Mol Hum Reprod ; 20(7): 591-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24674992

ABSTRACT

The pathogenesis of early-onset endometriosis has recently been revisited, sparked by the discovery of endometrial stem/progenitor cells and their possible role in endometriosis, and because maternal pregnancy hormone withdrawal following delivery induces uterine bleeding in the neonate. The neonatal uterus has a large cervix to corpus ratio which is functionally blocked with mucous, supporting the concept of retrograde shedding of neonatal endometrium. Only 5% show overt bleeding. Furthermore, the presence of endometriosis in pre-menarcheal girls and even in severe stage in adolescents supports the theory that early-onset endometriosis may originate from retrograde uterine bleeding soon after birth. Endometrial stem/progenitor cells have been identified in menstrual blood suggesting that they may also be shed during neonatal uterine bleeding. Thus, we hypothesized that stem/progenitor cells present in shedding endometrium may have a role in the pathogenesis of early-onset endometriosis through retrograde neonatal uterine bleeding. During the neonatal and pre-pubertal period, shed endometrial stem/progenitor cells are postulated to survive in the pelvic cavity in the absence of circulating estrogens supported by niche cells also shed during neonatal uterine bleeding. According to this hypothesis, during thelarche, under the influence of rising estrogen levels, endometrial stem/progenitor cells proliferate and establish ectopic endometrial lesions characteristic of endometriosis. This New Research Horizon review builds on recent discussions on the pathogenesis of early-onset endometriosis and raises new avenues for research into this costly condition.


Subject(s)
Adult Stem Cells/pathology , Endometriosis/etiology , Endometrium/pathology , Endometriosis/pathology , Female , Humans
8.
Mol Hum Reprod ; 20(2): 139-47, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24043696

ABSTRACT

The serum-and-glucocorticoid-inducible-kinase-1 (SGK1) is ubiquitously expressed and under genomic control by cell stress, hormones and further mediators. A most powerful stimulator of SGK1 expression is transforming growth factor TGFß1. SGK1 is activated by insulin and growth factors via phosphatidylinositol-3-kinase and the 3-phosphoinositide-dependent kinase PDK1. As shown recently, SGK1 increases the store-operated Ca(2+) entry (SOCE), which is accomplished by the pore-forming ion channel unit Orai. Most recent observations further revealed that SGK1 plays a critical role in the regulation of fertility. SGK1 is up-regulated in the luminal epithelium of women with unexplained infertility but down-regulated in decidualizing stromal cells of patients with recurrent pregnancy loss. The present study explored whether Orai1 is expressed in endometrium and sensitive to regulation by SGK1 and/or TGFß1. To this end, Orai1 protein abundance was determined by western blotting and SOCE by fura-2 fluorescence. As a result, Orai1 was expressed in human endometrium and in human endometrial Ishikawa cells. Orai1 expression and SOCE in Ishikawa cells were increased by transfection with constitutively active (S422D)SGK1 but not by transfection with inactive (K127N)SGK1. The difference of SOCE between (S422D)SGK1 and (K127N)SGK1-transfected cells was virtually abrogated in the presence of Orai1 inhibitor 2-aminoethoxydiphenyl borate (2-APB, 50 µM). Similar to (S422D)SGK1 transfection TGFß1 treatment up-regulated both Orai1 protein abundance and SOCE. In conclusion, Orai1 is expressed in the human endometrium and is up-regulated by SGK1 and TGFß1. The present observations thus uncover a novel element in SGK1-sensitive regulation of endometrial cells.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Endometrium/metabolism , Epithelial Cells/metabolism , Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta1/metabolism , Adult , Boron Compounds/pharmacology , Calcium Channels/genetics , Calcium Signaling , Endometrium/cytology , Endometrium/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Female , Gene Expression Regulation, Developmental , Humans , Immediate-Early Proteins/genetics , Ion Transport , ORAI1 Protein , Premenopause , Primary Cell Culture , Protein Serine-Threonine Kinases/genetics , Transfection , Transforming Growth Factor beta1/pharmacology
9.
Oncogene ; 33(34): 4316-29, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-24362530

ABSTRACT

The forkhead box transcription factor FOXM1 is an essential effector of G2/M-phase transition, mitosis and the DNA damage response. As such, it is frequently deregulated during tumorigenesis. Here we report that FOXM1 is dynamically modified by SUMO1 but not by SUMO2/3 at multiple sites. We show that FOXM1 SUMOylation is enhanced in MCF-7 breast cancer cells in response to treatment with epirubicin and mitotic inhibitors. Mutation of five consensus conjugation motifs yielded a SUMOylation-deficient mutant FOXM1. Conversely, fusion of the E2 ligase Ubc9 to FOXM1 generated an auto-SUMOylating mutant (FOXM1-Ubc9). Analysis of wild-type FOXM1 and mutants revealed that SUMOylation inhibits FOXM1 activity, promotes translocation to the cytoplasm and enhances APC/Cdh1-mediated ubiquitination and degradation. Further, expression of the SUMOylation-deficient mutant enhanced cell proliferation compared with wild-type FOXM1, whereas the FOXM1-Ubc9 fusion protein resulted in persistent cyclin B1 expression and slowed the time from mitotic entry to exit. In summary, our findings suggest that SUMOylation attenuates FOXM1 activity and causes mitotic delay in cytotoxic drug response.


Subject(s)
Forkhead Transcription Factors/metabolism , Mitosis , SUMO-1 Protein/metabolism , Sumoylation , Antibiotics, Antineoplastic/pharmacology , Antigens, CD , Binding Sites , Cadherins/metabolism , Cell Proliferation/drug effects , Cytoplasm/metabolism , Drug Resistance, Neoplasm , Epirubicin/pharmacology , Forkhead Box Protein M1 , G2 Phase Cell Cycle Checkpoints , HeLa Cells , Humans , MCF-7 Cells , Nocodazole/pharmacology , Protein Transport , Proteolysis
10.
Biochim Biophys Acta ; 1822(12): 1943-50, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22683339

ABSTRACT

Despite expanding global experience with advanced reproductive technologies, the majority of IVF attempts do not result in a successful pregnancy, foremost as a result of implantation failure. The process of embryo implantation, a remarkably dynamic and precisely controlled molecular and cellular event, appears inefficient in humans and is poorly understood. However, insights gained from clinical implantation failure, early pregnancy loss, and emerging techologies that enable molecular interrogation of endometrial-embryo interactions are unravelling this major limiting step in human reproduction. We review current molecular concepts thought to underlie implantation failure, consider the contribution of embryonic and endometrial factors, and discuss the clinical value of putative markers of impaired endometrial receptivity. Finally we highlight the nature of the dialogue between the maternal endometrium and the implanting embryo and discuss the concept of natural embryo selection. This article is part of a Special Issue entitled: Molecular Genetics of Human Reproductive Failure.


Subject(s)
Embryo Implantation/genetics , Abortion, Spontaneous , Animals , Endometrium/physiopathology , Female , Humans , Pregnancy
11.
Ultrasound Obstet Gynecol ; 39(6): 612-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21910147

ABSTRACT

Emerging evidence suggests that early embryo implantation is a more active maternal process than hitherto appreciated, involving active encapsulation of the implanting blastocyst by maternal decidual cells and coordinated changes in the underlying inner myometrium, known as the junctional zone. These concepts raise the possibility that early ultrasound markers predictive of adverse pregnancy outcome could be identified. In this review we assess the role of ultrasound in predicting the likelihood of different pregnancy outcomes and highlight potential novel markers that could be tested.


Subject(s)
Embryo Implantation , Endometrium/diagnostic imaging , Fertilization in Vitro/methods , Myometrium/diagnostic imaging , Uterine Artery/diagnostic imaging , Endometrium/blood supply , Female , Humans , Myometrium/blood supply , Predictive Value of Tests , Pregnancy , Pregnancy Outcome , Ultrasonography , Uterine Artery/physiopathology
12.
Facts Views Vis Obgyn ; 4(3): 33-43, 2012.
Article in English | MEDLINE | ID: mdl-24753902

ABSTRACT

Progesterone is central to many reproductive processes and is critical in regulating the menstrual cycle and maintaining pregnancy. We discuss here similarities in the molecular mechanisms that regulate the process of decidualisation in endometrial stromal cells and uterine quiescence in myometrial smooth muscle cells. We discuss recent evidence that the decidua may be an important mediator of progesterone actions in the onset of labor in mammalian species lacking progesterone withdrawal. These observations have relevance to recent clinical observations of the effect of progesterone administration in the prevention of preterm labor. We suggest that further research is required to understand the role of progesterone in maintaining the decidua in late pregnancy and particular focus should be given to the mechanisms that increase prostaglandin production in the uterus at term.

13.
Oncogene ; 31(14): 1845-58, 2012 Apr 05.
Article in English | MEDLINE | ID: mdl-21860419

ABSTRACT

Vascular endothelial growth factor (VEGF) has a central role in breast cancer development and progression, but the mechanisms that control its expression are poorly understood. Breast cancer tissue microarrays revealed an inverse correlation between the Forkhead transcription factor Forkhead box class O (FOXO)3a and VEGF expression. Using the lapatinib-sensitive breast cancer cell lines BT474 and SKBR3 as model systems, we tested the possibility that VEGF expression is negatively regulated by FOXO3a. Lapatinib treatment of BT474 or SKBR3 cells resulted in nuclear translocation and activation of FOXO3a, followed by a reduction in VEGF expression. Transient transfection and inducible expression experiments showed that FOXO3a represses the proximal VEGF promoter, whereas another Forkhead member, FOXM1, induces VEGF expression. Chromatin immunoprecipitation and oligonucleotide pull-down assays showed that both FOXO3a and FOXM1 bind a consensus Forkhead response element (FHRE) in the VEGF promoter. Upon lapatinib stimulation, activated FOXO3a displaces FOXM1 bound to the FHRE before recruiting histone deacetylase 2 (HDAC2) to the promoter, leading to decreased histones H3 and H4 acetylation, and concomitant transcriptional inhibition of VEGF. These results show that FOXO3a-dependent repression of target genes in breast cancer cells, such as VEGF, involves competitive displacement of DNA-bound FOXM1 and active recruitment of transcriptional repressor complexes.


Subject(s)
Breast Neoplasms/metabolism , Forkhead Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cell Line, Tumor , Female , Forkhead Box Protein M1 , Forkhead Box Protein O3 , Gene Expression Regulation, Neoplastic , Histone Deacetylase 2/metabolism , Humans , Lapatinib , Quinazolines/pharmacology
14.
Placenta ; 32(8): 592-7, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21641641

ABSTRACT

Human chorionic gonadotropin (hCG) is one of the earliest signals secreted by the implanting embryo. In addition to its well-known luteotropic function in early pregnancy, hCG also acts directly on decidualizing endometrium. Recently, we demonstrated that recombinant hCG (rhCG) prevented apoptosis in decidualizing human endometrial stromal cells (HESCs) exposed to oxidative stress. Two hCG preparations are widely used clinically: rhCG, produced by recombinant DNA technology, and urinay hCG (uhCG), extracted from urine of post-menopausal women. However, an analysis of the direct effects of rhCG and uhCG on the decidual phenotype of HESCs has not yet been done. In this study, we investigated the effects of uhCG and rhCG on the morphological and functional profiles of decidualizing HESCs. We demonstrate that neither rhCG nor uhCG alter the morphological appearance of the decidual HESC cultures, although rhCG but not uhCG attenuated prolactin expression, a major decidual marker protein. Moreover, rhCG, but not uhCG, protected decidualizing HESCs from oxidative cell death, mediated at least in part by two major mechanisms. First, rhCG, but not uhCG, enhances the expression of manganese superoxide dismutase, a cardinal enzyme in the cellular defense against oxidative damage. Second, rhCG signaling selectively limits activation of the apoptotic machinery in decidualizing HESCs by enhancing Bcl-2 expression whereas uhCG induces the expression of Fas ligand. Our results suggest that rhCG might be a preferable agent to protect the maternal decidua against oxidative damage in pregnancy, especially at the time of implantation and beyond.


Subject(s)
Chorionic Gonadotropin/pharmacology , Chorionic Gonadotropin/urine , Decidua/drug effects , Oxidative Stress/drug effects , Stromal Cells/metabolism , Adult , Decidua/cytology , Embryo Implantation/drug effects , Female , Humans , Middle Aged , Pregnancy , Prolactin/metabolism , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Stromal Cells/drug effects
15.
J Clin Endocrinol Metab ; 96(6): E1013-24, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21450990

ABSTRACT

CONTEXT: Progesterone administration reduces the risk of preterm labor in high-risk women with singleton pregnancies but has no effect in women with a multiple pregnancy. OBJECTIVE: We investigated whether progesterone is able to inhibit stretch-induced gene expression and/or whether stretch in turn inhibits progesterone action, perhaps providing an explanation for the functional progesterone withdrawal associated with human labor. METHODS AND RESULTS: In a series of in vitro studies using primary cultures of human myometrial cells, we found that preincubation with progesterone did not block stretch-induced ERK1/2 activation and cyclooxygenase-2 mRNA expression. Furthermore, we found that stretch did not alter the ability of progesterone to: 1) modulate progesterone-responsive gene expression; 2) activate a luciferase-linked progesterone response element; or 3) repress IL-1ß-driven cyclooxygenase-2 mRNA expression. We did find that stretch reduced the expression of progesterone receptor mRNA via nuclear factor κB activation but that this did not alter myometrial progesterone response. CONCLUSION: These data show that progesterone does not inhibit stretch-induced MAPK activation or gene expression, possibly explaining why progesterone is ineffective in the prevention of preterm labor in multiple pregnancy. Although stretch did reduce progesterone receptor expression in a nuclear factor κB-dependent manner, this was not sufficient to inhibit progesterone action, suggesting that it is not responsible for the functional progesterone withdrawal observed with the onset of human labor.


Subject(s)
Myometrium/metabolism , Progesterone/metabolism , Uterine Contraction/drug effects , Analysis of Variance , Blotting, Western , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression/drug effects , Humans , Myometrium/cytology , Myometrium/drug effects , Progesterone/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Uterine Contraction/physiology
16.
Acta Physiol (Oxf) ; 202(1): 39-45, 2011 May.
Article in English | MEDLINE | ID: mdl-21251236

ABSTRACT

AIM: Pregnancy is typically paralleled by substantial increase in maternal extracellular fluid volume, requiring net accumulation of water and NaCl. The positive water and salt balance is accomplished at least in part by increased uptake of salt secondary to enhanced salt appetite. Little is known about the underlying cellular mechanisms. Stimulation of salt appetite by mineralocorticoids, however, is known to be dependent on the serum- and glucocorticoid-inducible kinase SGK1. METHODS: To test for a role of SGK1 in the stimulation of salt appetite during pregnancy, fluid intake was recorded in pregnant SGK1 knockout mice (sgk1(-/-) ) and their wild type littermates (sgk1(+/+) ). The mice were offered two bottles, one with plain water and the other with isotonic saline. RESULTS: In early pregnancy, i.e. up to 10 days prior to parturition, the sgk1(+/+) mice displayed a significant preference for saline, whereas the sgk1(-/-) mice preferred water. Accordingly, the water intake was significantly smaller and saline intake was significantly larger in sgk1(+/+) mice than in sgk1(-/-) mice and the preference for water was significantly stronger in sgk1(-/-) mice than in sgk1(+/+) mice. Plasma aldosterone levels were higher in sgk1(-/-) mice than in sgk1(+/+) mice, a difference contrasting the enhanced salt appetite of sgk1(+/+) mice. CONCLUSIONS: SGK1 participates in the stimulation of salt appetite during pregnancy.


Subject(s)
Appetite , Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Sodium Chloride, Dietary , Aldosterone/blood , Animals , Drinking , Female , Immediate-Early Proteins/genetics , Mice , Mice, Knockout , Pregnancy , Protein Serine-Threonine Kinases/genetics
18.
Hum Reprod Update ; 15(1): 119-38, 2009.
Article in English | MEDLINE | ID: mdl-18936037

ABSTRACT

BACKGROUND: The steroid hormone progesterone is indispensable for mammalian procreation by controlling key female reproductive events that range from ovulation to implantation, maintenance of pregnancy and breast development. In addition to activating the progesterone receptors (PRs)-B and -A, members of the superfamily of ligand-dependent transcription factors, progesterone also elicits a variety of rapid signalling events independently of transcriptional or genomic regulation. This review covers our current knowledge on the mechanisms and relevance of non-genomic progesterone signalling in female reproduction. METHODS: PubMed was searched up to August 2008 for papers on progesterone actions in ovary/breast/endometrium/myometrium/brain, focusing primarily on non-genomic signalling mechanisms. RESULTS: Convergence and intertwining of rapid non-genomic events and the slower transcriptional actions critically determine the functional response to progesterone in the female reproductive system in a cell-type- and environment-specific manner. Several putative progesterone-binding moieties have been implicated in rapid signalling events, including the 'classical' PR and its variants, progesterone receptor membrane component 1, and the novel family of membrane progestin receptors. Progesterone and its metabolites have also been implicated in the allosteric regulation of several unrelated receptors, such as gamma-aminobutyric acid type A, oxytocin and sigma(1) receptors. CONCLUSIONS: Identification of the mechanisms and receptors that relay rapid progesterone signalling is an area of research fraught with difficulties and controversy. More in-depth characterization of the putative receptors is required before the non-genomic progesterone pathway in normal and pathological reproductive function can be targeted for pharmacological intervention.


Subject(s)
Progesterone/physiology , Reproduction/physiology , Brain/metabolism , Breast/metabolism , Endometrium/metabolism , Female , Genome, Human , Humans , Myometrium/metabolism , Ovary/metabolism , Progesterone/metabolism , Receptors, Progesterone/physiology , Signal Transduction
19.
Oncogene ; 27(1): 9-19, 2008 Jan 03.
Article in English | MEDLINE | ID: mdl-17599040

ABSTRACT

The forkhead transcription factor FOXO1, a downstream target of phosphatidylinositol-3-kinase/Akt signalling pathway, regulates cyclic differentiation and apoptosis in normal endometrium, but its role in endometrial carcinogenesis is unknown. Screening of endometrial cancer cell lines demonstrated that FOXO1 is expressed in HEC-1B cells, but not in Ishikawa cells, which in turn highly express the FOXO1 targeting E3-ubiquitin ligase Skp2. FOXO1 transcript levels were also lower in Ishikawa cells and treatment with the proteasomal inhibitor was insufficient to restore expression. Lack of FOXO1 expression in Ishikawa cells was not accounted for by differential promoter methylation or activity, but correlated with increased messenger RNA (mRNA) turnover. Comparative analysis demonstrated that HEC-1B cells proliferate slower, but are more resistant to paclitaxel-mediated cell death than Ishikawa cells, which were partially reversed upon silencing of FOXO1 in HEC-1B cells or its re-expression in Ishikawa cells. We further show that FOXO1 is required for the expression of the growth arrest- and DNA-damage-inducible gene GADD45alpha. Analysis of biopsy samples demonstrated a marked loss of FOXO1 and GADD45alpha mRNA and protein expression in endometrioid endometrial cancer compared to normal endometrium. Together, these observations suggest that loss of FOXO1 perturbs endometrial homeostasis, promotes uncontrolled cell proliferation and increases susceptibility to genotoxic insults.


Subject(s)
Carcinoma, Endometrioid/metabolism , Carcinoma, Endometrioid/pathology , Down-Regulation/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic/physiology , Carcinoma, Endometrioid/drug therapy , Carcinoma, Endometrioid/genetics , Cell Line, Tumor , Cell Proliferation , Down-Regulation/physiology , Drug Resistance, Neoplasm/genetics , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/physiology , Genomic Instability/genetics , Humans
20.
J Mol Endocrinol ; 36(3): 389-98, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16720711

ABSTRACT

The human endometrium undergoes cyclical waves of proliferation, differentiation and apoptosis in response to the rise and fall in ovarian oestradiol and progesterone levels. These hormonal responses in endometrial cells must be tightly kept in check to safeguard tissue homeostasis throughout reproductive life. The discovery that differentiating endometrium highly expresses the tumour suppressor p53, the forkhead transcription factor FOXO1, and promyelocytic leukaemia zinc finger protein (PLZF) has provided new insights into the molecular basis of life and death decisions in response to sex steroid hormones.


Subject(s)
Cell Death/physiology , Cell Survival/physiology , Endometrium/cytology , Progesterone/metabolism , Stromal Cells/physiology , Animals , Cyclic AMP/metabolism , DNA-Binding Proteins/metabolism , Endometrium/metabolism , Estradiol/metabolism , Female , Forkhead Transcription Factors/metabolism , Humans , Kruppel-Like Transcription Factors , Pregnancy , Promyelocytic Leukemia Zinc Finger Protein , Stromal Cells/cytology , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...