Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Reprod Sci ; 28(1): 252-262, 2021 01.
Article in English | MEDLINE | ID: mdl-32780361

ABSTRACT

In most mammals, labor is heralded by the withdrawal of progesterone. In humans, circulating progesterone levels increase as gestation advances while placental expression of progesterone receptor A (PR-A) declines. As a result of PR-A downregulation, the non-canonical NF-κB pathway is activated, an event implicated in triggering labor. Here, we sought to identify fetal-derived mediator(s) that represses placental PR-A in human placenta leading to activation of pro-labor signaling. Lipidomic profiling demonstrated enrichment of platelet-activating factor (PAF) in exosomes originating from the human fetus. Exposure of primary cytotrophoblasts to fetal exosomes from term pregnancies reduced PR-A expression by > 50%, and PAF also reduced PR-A message levels in a dose-dependent manner. Notably, fetal exosomes from preterm pregnancies had lower PAF levels and no effect on PR-A expression. Synthetic PAF-induced DNA methylation increases by 20% at the PR-A promoter, leading to recruitment of corepressors and downregulation of PR-A in cytotrophoblast. Furthermore, suppression of PR-A by PAF-stimulated expression of the pro-labor genes, corticotropin-releasing hormone (CRH) and cyclooxygenase-2 (COX-2), which was reversed by disruption of the DNA methyltransferases 3B and 3L. Taken together, PAF represents a novel fetal-derived candidate for initiation of labor by stimulating methylation and repression of PR-A and activating pro-labor signaling in trophoblast.


Subject(s)
Exosomes/metabolism , Fetus/metabolism , Labor, Obstetric/metabolism , Placenta/metabolism , Platelet Activating Factor/metabolism , Progesterone/metabolism , Receptors, Progesterone/metabolism , Cells, Cultured , DNA Methylation , Epigenesis, Genetic , Female , Gestational Age , Humans , Labor, Obstetric/genetics , Lipidomics , Pregnancy , Premature Birth/genetics , Premature Birth/metabolism , Premature Birth/physiopathology , Receptors, Progesterone/genetics , Signal Transduction
2.
Placenta ; 104: 8-15, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33197856

ABSTRACT

INTRODUCTION: The timing of parturition at end of human gestation may be controlled by fetal signals. The signaling molecules contributing to activation of human labor may be mediated by fetal exosomes. In this study, we focused on investigation of the role of microRNAs (miRNAs) derived from fetal exosomes in the regulation of human placental gene expression. METHODS: Using immunofluorescent labeling, array-based miRNA profiling assay, target prediction analysis, and conducting a variety of biochemical approaches including miRNA mimics, dual-luciferase, siRNA-mediated gene silencing, and immunohistochemical staining assay in primary trophoblast culture and formalin-fixed paraffin-embedded placental tissues, we examined whether fetal exosomal miRNAs can stimulate expression of proinflammatory mediators in human placenta. RESULTS: We showed placental uptake of exosomes derived from the umbilical artery, and found that 9 fetal exosomal miRNAs: let-7i-5p, miR-185-5p, miR-15b-5p, miR-376c-3p, miR-548d-5p, miR-92b-3p, miR-16-5p, and miR-1301-3p were significantly increased in placentas of women delivering following term labor compared to those delivering by Cesarean section in the late preterm period. Target prediction analysis identified miR-15b-5p of particular interest, because one of its predicted targets is Apelin, a potent inhibitor of proinflammatory mediators. We further found that miR-15b-5p repressed Apelin protein levels and activated pro-labor hormones and cytokines including IL-1, IL-6, IL-8, and TNF-α. DISCUSSION: These data suggest a potential fetal-to-placental signal that could play a role in the length of human gestation and onset of human labor.


Subject(s)
Apelin/metabolism , Cytokines/metabolism , Inflammation/metabolism , MicroRNAs/metabolism , Placenta/metabolism , Signal Transduction/physiology , Adult , Exosomes/metabolism , Female , Gene Expression Profiling , Humans , MicroRNAs/genetics , Pregnancy
3.
F S Sci ; 1(1): 67-77, 2020 Aug.
Article in English | MEDLINE | ID: mdl-35559741

ABSTRACT

OBJECTIVE: To characterize the expression and signaling of uterine GPR83 in vivo in the nonpregnant and pregnant mouse and in vitro in human endometrial and nonendometrial cells. DESIGN: Controlled laboratory study. SETTING: Not applicable. PATIENTS: Not applicable. INTERVENTIONS: None. MAIN OUTCOME MEASURES: Expression of uterine Gpr83 was determined by quantitative polymerase chain reaction throughout the estrous cycle and during early pregnancy in ovarian-stimulated and non-ovarian-stimulated mice and pregnant and pseudopregnant mice. Expression was also determined in ovariectomized mice after the administration of oil, E2, P4, or E2 + P4 and in stromal cells following 6 days of in vitro decidualization. GPR83 signaling was studied in human endometrial and embryonic kidney cell lines. Cells were treated by PEN, a GPR83 ligand, and PEN-induced extracellular signal-regulated kinase (ERK) phosphorylation was assayed under conditions that blocked Gαq/11 and/or ß-arrestin signaling. RESULTS: Uterine Gpr83 is expressed throughout the estrous cycle and during early pregnancy; expression increases dramatically at the time of uterine receptivity, embryo implantation, and stromal cell decidualization. In the ovariectomized mouse, hormone add-back reveals that Gpr83 expression is highly responsive to the combined treatment of E2 and P4, and studies in the ovarian-stimulated mouse show that expression is also very sensitive to changes in E2 and P4 and is therefore tightly regulated by E2 and P4. At the implantation site, expression is elevated up to D6 of pregnancy and then declines rapidly on D7 and D8, suggesting that if there is any involvement in decidualization, it is likely associated with primary but not secondary stromal cell decidualization. This premise was supported by the observation that stromal cell decidualization in vitro progresses with a decline in Gpr83 expression. In ERα/PR-expressing endometrial Ishikawa cells, GPR83 mediates PEN signals in a Gαq/11-dependent manner, and studies conducted in HEK 293 cells lacking ß-arrestin revealed that GPR83 also signals via a ß-arrestin-dependent manner. When signaling by either one or both pathways is downregulated, cells exhibit a major reduction in responsiveness to PEN treatment, demonstrating that signaling by both pathways is significant. CONCLUSION: We hypothesize that PEN/GPR83 signaling regulates uterine receptivity, embryo implantation, and primary stromal cell decidualization by coupling to Gαq/11- and ß-arrestin-dependent pathways.

4.
Sci Rep ; 9(1): 5519, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30940885

ABSTRACT

The non-canonical NF-κB signaling may be a central integrator of a placental clock that governs the length of human pregnancy. We sought to identify fetal signals that could activate this NF-κB pathway in the placenta, and in turn, contribute to the onset of labor. Proteomics analysis of exosomes purified from fetal cord arterial blood revealed a total of 328 proteins, among which 48 were more significantly abundant (p < 0.01) in samples from women who delivered following elective Cesarean-section at term (39 to 40 weeks of estimated gestational age, EGA) compared to those who had elective Cesarean deliveries near term (35 to 36 weeks of EGA). Computational, crystal structural, and gene functional analyses showed that one of these 48 proteins, C4BPA, binds to CD40 of placental villous trophoblast to activate p100 processing to p52, and in turn, pro-labor genes. These results suggest that fetal C4BPA-induced activation of non-canonical NF-κB in human placenta may play a critical role in processes of term or preterm labor.


Subject(s)
Complement C4b-Binding Protein/metabolism , Exosomes/metabolism , Lung/embryology , NF-kappa B p52 Subunit/metabolism , Placenta/metabolism , CD40 Antigens/metabolism , Complement C4b-Binding Protein/chemistry , Female , Fetal Blood/metabolism , Humans , Labor, Obstetric/metabolism , Lung/metabolism , Models, Molecular , NF-kappa B/metabolism , Pregnancy , Proteomics/methods , Signal Transduction
5.
Placenta ; 76: 40-50, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30642670

ABSTRACT

BACKGROUND: Chromatin alterations are important mediators of gene expression changes. We have recently shown that activated non-canonical NF-κB signaling (RelB/p52) recruits histone acetyltransferase CBP and deacetylase HDAC1 to selectively acetylate H3K9 (H3K9ac) to induce expression of corticotropin-releasing hormone (CRH) and prostaglandin-endoperoxide synthase-2 (PTGS2) in the human placenta. Both of these genes play a role in initiating parturition in human pregnancy. METHODS: We performed chromatin immunoprecipitation followed by gene sequencing (ChIP-seq) in primary term human cytotrophoblast (CTB) with use of antibodies to RelB, CBP, HDAC1 and H3K9ac. We further associated these chromatin alterations with gene expression changes from mid-trimester to term in CTB by RNA sequencing (RNA-seq). RESULTS: We detected a genome-wide differential gene enrichment between mid-trimester and term human placenta. Pathway analysis identified that cytokine-cytokine receptor interaction, NF-κB, and TNF are the leading pathways enriched in term placenta and associated with these chromatin alterations. DISCUSSIONS: Our analysis has provided the first-time characterization of the key players of human placental origin with molecular changes resulting from chromatin modifications, which could drive human labor.


Subject(s)
Histone Deacetylase 1/metabolism , Parturition/metabolism , Transcription Factor RelB/metabolism , Trophoblasts/metabolism , Chromatin Immunoprecipitation Sequencing , Female , Histone Acetyltransferases/metabolism , Humans , Pregnancy , Sequence Analysis, RNA , Signal Transduction
6.
Endocr Connect ; 7(12): 1380-1388, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30395535

ABSTRACT

Maternal vitamin D deficiency is linked to adverse pregnancy outcomes including spontaneous preterm birth (SPB). Placental corticotropin-releasing hormone (CRH) has been proposed to be part of a clock that governs the length of gestation in humans, with elevated maternal serum levels predicting early delivery. In this study, we test the hypothesis that vitamin D could contribute to the prevention of preterm labor by inhibiting CRH and other pro-labor mediators. The biological activity of vitamin D occurs via two pathways: non-genomic and genomic responses, both of which involve binding of 1,25-dihydroxyvitamin D (1,25(OH)2D), the active metabolite of vitamin D binding to the vitamin D receptor (VDR). By using chromatin immunoprecipitation followed by sequencing (ChIP-seq), we found that 1,25(OH)2D stimulates association of VDR with a number of miRNA genes including MIR181B2 and MIR26B, and their mature products miR-181b-5p and miR-26b-5p are predicted to target CRH and cyclooxygenase-2 (COX-2) mRNA at 3'-untranslated region (UTR), respectively. We performed RT-qPCR analysis to validate that expression of mature miR-181b-5p and miR-26b-5p in term human syncytiotrophoblast increased in response to treatment with 1,25(OH)2D. miR-181b-5p- or miR-26b-5p-mediated inhibition of CRH or COX-2 was further assessed by the use of miRNA mimics/inhibitors and a luciferase reporter assay. Taken together, this study has identified novel mechanisms by which vitamin D downregulates pro-labor genes and could lower the risk of preterm delivery.

7.
Sci Rep ; 8(1): 1657, 2018 01 26.
Article in English | MEDLINE | ID: mdl-29374256

ABSTRACT

The non-canonical NF-κB signaling (RelB/p52) pathway drives pro-labor genes in the human placenta, including corticotropin-releasing hormone (CRH) and cyclooxygenase-2 (COX-2), making this a potential therapeutic target to delay onset of labor. Here we sought to identify small molecule compounds from a pre-existing chemical library of orally active drugs that can inhibit this NF-κB signaling, and in turn, human placental CRH and COX-2 production. We used a cell-based assay coupled with a dual-luciferase reporter system to perform an in vitro screening of a small molecule library of 1,120 compounds for inhibition of the non-canonical NF-κB pathway. Cell toxicity studies and drug efflux transport MRP1 assays were used to further characterize the lead compounds. We have found that 14 drugs have selective inhibitory activity against lymphotoxin beta complex-induced activation of RelB/p52 in HEK293T cells, several of which also inhibited expression of CRH and COX-2 in human term trophoblast. We identified sulfapyridine and propranolol with activity against CRH and COX-2 that deserve further study. These drugs could serve as the basis for development of orally active drugs to affect length of gestation, first in an animal model, and then in clinical trials to prevent preterm birth during human pregnancy.


Subject(s)
Drug Evaluation, Preclinical , Propranolol/isolation & purification , Protein Kinase Inhibitors/isolation & purification , Protein Serine-Threonine Kinases/antagonists & inhibitors , Small Molecule Libraries , Sulfapyridine/isolation & purification , Tocolytic Agents/isolation & purification , Cells, Cultured , Corticotropin-Releasing Hormone/biosynthesis , Cyclooxygenase 2/biosynthesis , Female , Gene Expression Regulation/drug effects , Humans , Placenta , Pregnancy , Propranolol/pharmacology , Protein Kinase Inhibitors/pharmacology , Sulfapyridine/pharmacology , Tocolytic Agents/pharmacology , Trophoblasts/drug effects , NF-kappaB-Inducing Kinase
8.
PLoS One ; 11(1): e0147013, 2016.
Article in English | MEDLINE | ID: mdl-26751383

ABSTRACT

Women exposed to phthalates during pregnancy are at increased risk for delivering preterm, but the mechanism behind this relationship is unknown. Placental corticotropin-releasing hormone (CRH) and cyclooxygenase-2 (COX-2) are key mediators of parturition and are regulated by the non-canonical NF-kB (RelB/p52) signaling pathway. In this study, we demonstrate that one of the major phthalate metabolites, mono-(2-ethylhexyl)-phthalate (MEHP), increased CRH and COX-2 mRNA and protein abundance in a dose-dependent manner in primary cultures of cytotrophoblast. This was coupled with an increase in nuclear import of RelB/p52 and its association with the CRH and COX-2 promoters. Silencing of NF-kB inducing kinase, a central signaling component of the non-canonical NF-kB pathway, blocked MEHP-induced upregulation of CRH and COX-2. These results suggest a potential mechanism mediated by RelB/p52 by which phthalates could prematurely induce pro-labor gene activity and lead to preterm birth.


Subject(s)
Gene Expression Regulation , Labor, Obstetric/genetics , Labor, Obstetric/metabolism , Phthalic Acids/metabolism , Placenta/metabolism , Cell Nucleus/metabolism , Cesarean Section , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Female , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Humans , NF-kappa B/metabolism , Phthalic Acids/pharmacology , Placenta/drug effects , Pregnancy , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Transport , Transcription Factor RelB/metabolism , Trophoblasts/metabolism , Up-Regulation , NF-kappaB-Inducing Kinase
9.
Sci Signal ; 8(391): ra85, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26307012

ABSTRACT

Corticotropin-releasing hormone (CRH) produced in the placenta may be part of a clock that regulates the length of human gestation. Maternal plasma CRH abundance exponentially increases as pregnancy advances. Glucocorticoid stimulates CRH expression in full-term human placenta by promoting noncanonical (RelB/p52 heterodimer-mediated) nuclear factor κB (NF-κB) pathway activity. Using dexamethasone to mimic glucocorticoid exposure, we found that an epigenetic switch mediated the glucocorticoid-induced expression of CRH as gestation advances. The amount of acetylated histone H3 lysine 9 (H3K9) associated with the CRH promoter was greater in cytotrophoblasts from full-term placenta than in those from midterm placenta. Knocking down the lysine acetyltransferase CBP reduced H3K9 histone acetylation and prevented dexamethasone-induced CRH expression. Unexpectedly, knocking down the histone deacetylase HDAC1 or pharmacologically inhibiting type I and II HDACs also decreased the expression of CRH yet increased the acetylation of H3K9 and other histone regions. Both CBP and HDAC1 bound at the CRH promoter in a complex with the RelB/p52 heterodimer in a mutually dependent manner; knocking down any one factor in the complex prevented binding of the others as well as the dexamethasone-induced CRH expression. Our results suggest that glucocorticoids induce a transcription complex consisting of RelB/p52, CBP, and HDAC1 that triggers a dynamic acetylation-mediated epigenetic change to induce CRH expression in full-term human placenta.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Histones/metabolism , NF-kappa B p52 Subunit/metabolism , Placenta/metabolism , Pregnancy/metabolism , Transcription Factor RelB/metabolism , Acetylation/drug effects , Adolescent , Adult , Dexamethasone/pharmacology , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/physiology , Female , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Humans , Middle Aged
10.
J Clin Endocrinol Metab ; 99(2): E320-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24276461

ABSTRACT

CONTEXT: Progesterone (P4)contributes to the maintenance of human pregnancy, in part by inhibiting activity of the human pro-labor genes CRH and cyclooxygenase-2 (COX-2). However, the molecular mechanisms underlying the action of P4 remain poorly defined. We have shown that in human placenta, the constitutively activated noncanonical nuclear factor (NF)-κB pathway positively regulates CRH and COX-2, which is further stimulated by glucocorticoid receptor signaling. OBJECTIVE: We investigated the role of P4 receptor (PR) in the regulation of nuclear activity of v-rel avian reticuloendotheliosis viral oncogene homolog B (RelB)/NF-κB2 and, in turn, expression of placental CRH and COX-2. METHODS: We used a variety of techniques including gene silencing, ectopic expression, chromatin immunoprecipitation, Western blot, quantitative RT-PCR, and immunohistochemical staining assays in human placental tissues and primary culture of human cytotrophoblast. RESULTS: We identified PR isoform-A (PR-A) as the only isoform of PR produced in human placenta. PR-A levels were lower in term placenta than in midterm placenta. Depletion of PR-A by short interfering RNA derepressed inhibition of CRH and COX-2 by P4 and the synthetic progestin 17α-hydroxyprogesterone caproate. Overexpression of PR-A inhibited transcription of CRH and COX-2, which was further downregulated by treatment with P4 or 17α-hydroxyprogesterone caproate. Such an inhibition was mediated by a negative functional interaction of PR-A with the activity of RelB/NF-κB2. CONCLUSION: P4 inhibits the pro-labor genes CRH and COX-2 via PR-A repression of the noncanonical NF-κB signaling in human placenta. Characterization of these pathways may identify potential drug targets for prevention of preterm birth.


Subject(s)
NF-kappa B/metabolism , Placenta/metabolism , Protein Isoforms/metabolism , Receptors, Progesterone/metabolism , Signal Transduction/physiology , 17 alpha-Hydroxyprogesterone Caproate , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Down-Regulation/drug effects , Female , Humans , Hydroxyprogesterones/pharmacology , NF-kappa B/genetics , Placenta/drug effects , Pregnancy , Progesterone/pharmacology , Protein Isoforms/genetics , Receptors, Progesterone/genetics , Signal Transduction/drug effects , Up-Regulation/drug effects
11.
Mol Endocrinol ; 27(2): 203-11, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23239753

ABSTRACT

Our recent study demonstrated that constitutively activated RelB/NF-κB2 positively regulates the CRH in the human placenta. In the current study, we explored the role of the glucocorticoid receptor (GR) signaling in constitutive activation of the noncanonical NF-κB pathway. A glucocorticoid response element (GRE) motif search suggests that both NF-κB inducing kinase (NIK) and RelB genes, which are key regulators of the noncanonical NF-κB pathway, have a putative GRE within their promoter, approximately 1 kb upstream from the transcription start site. By using chromatin immunoprecipitation assay we identified that the GR and phosphorylated GR at Ser211 were associated with the GREs of both NIK and RelB. Dexamethasone stimulated expression of NIK, RelB, NF-κB2 as well as CRH and cyclooxygenase-2 (COX-2). Repression of GR by short interfering RNA resulted in inhibition of NIK, RelB, NF-κB2, CRH, and COX-2. In addition, depletion of GR attenuated glucocorticoid-mediated up-regulation of NIK, RelB, NF-κB2, CRH, and COX-2. Furthermore, siRNA specifically targeting NIK down-regulated CRH and COX-2. Taken together, these results suggest that constitutive activation of the noncanonical NF-κB pathway in term human placenta is driven by the GR signaling, which in turn up-regulates placental CRH and other NF-κB-responsive genes.


Subject(s)
NF-kappa B/metabolism , Placenta/metabolism , Receptors, Glucocorticoid/metabolism , Corticotropin-Releasing Hormone/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors , Dexamethasone/pharmacology , Down-Regulation , Enzyme Activation , Female , Humans , NF-kappa B p52 Subunit/genetics , NF-kappa B p52 Subunit/metabolism , Phosphorylation , Pregnancy , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Receptors, Glucocorticoid/genetics , Response Elements , Signal Transduction , Transcription Factor RelB/genetics , Transcription Factor RelB/metabolism , Transcription, Genetic , Trophoblasts , Up-Regulation , NF-kappaB-Inducing Kinase
12.
Mol Endocrinol ; 26(8): 1356-69, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22734038

ABSTRACT

Placental CRH may be part of a clock that governs the length of human gestation. The mechanism underlying differential regulation of CRH in the human placenta is poorly understood. We report here that constitutively activated RelB/nuclear factor-κB2 (NF-κB)-2 (p100/p52) acts as an endogenous stimulatory signal to regulate CRH by binding to an NF-κB enhancer of CRH gene promoter in the human placenta. Nuclear staining of NF-κB2 and RelB in villous syncytiotrophoblasts and cytotrophoblasts was coupled with cytoplasmic CRH in syncytial knots of cytotrophoblasts. Chromatin immunoprecipitation identified that CRH gene associated with both RelB and NF-κB2 (p52). Dexamethasone increased synthesis and nuclear translocation of RelB and NF-κB2 (p52) and their association with the CRH gene. In contrast, progesterone, a down-regulator of placental CRH, repressed NF-κB2 (p100) processing, nuclear translocation of RelB and NF-κB2 (p52), and their association with the CRH gene. Luciferase reporter assay determined that the NF-κB enhancer of CRH was sufficient to regulate transcriptional activity of a heterologous promoter in primary cytotrophoblasts. RNA interference-mediated repression of RelB or NF-κB2 resulted in significant inhibition of CRH at both transcriptional and translational levels and prevented the dexamethasone-mediated up-regulation of CRH transcription and translation. These results suggest that the noncanonical NF-κB pathway regulates CRH production in the human placenta and is responsible for the positive regulation of CRH by glucocorticoids.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , NF-kappa B p52 Subunit/physiology , Placenta/metabolism , Transcription Factor RelB/physiology , Analysis of Variance , Base Sequence , Cells, Cultured , Corticotropin-Releasing Hormone/genetics , Dexamethasone/pharmacology , Enhancer Elements, Genetic , Female , Gene Expression Regulation , Humans , Leupeptins/pharmacology , NF-kappa B p52 Subunit/metabolism , Placenta/cytology , Pregnancy , Progesterone/pharmacology , Progesterone/physiology , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Signal Transduction , Transcription Factor RelB/metabolism , Transcription, Genetic , Trophoblasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...