Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Mol Metab ; 53: 101331, 2021 11.
Article in English | MEDLINE | ID: mdl-34474167

ABSTRACT

BACKGROUND: The cycle of feeding and fasting is fundamental to life and closely coordinated with changes of metabolic programs. During extended starvation, ketogenesis coupled with fatty acid oxidation in the liver supplies ketone bodies to extrahepatic tissues as the major form of fuel. In this study, we demonstrated that PAQR9, a member of the progesterone and adipoQ receptor family, has a regulatory role on hepatic ketogenesis. METHODS: We analyzed the phenotype of Paqr9-deleted mice. We also used biochemical methods to investigate the interaction of PAQR9 with PPARα and HUWE1, an E3 ubiquitin ligase. RESULTS: The expression of Paqr9 was decreased during fasting partly depending on PPARγ. The overall phenotype of the mice was not altered by Paqr9 deletion under normal chow feeding. However, fasting-induced ketogenesis and fatty acid oxidation were attenuated by Paqr9 deletion. Mechanistically, Paqr9 deletion decreased protein stability of PPARα via enhancing its poly-ubiquitination. PAQR9 competed with HUWE1 for interaction with PPARα, thus preventing ubiquitin-mediated degradation of PPARα. CONCLUSION: Our study reveals that PAQR9 impacts starvation-mediated metabolic changes in the liver via post-translational regulation of PPARα.


Subject(s)
Fasting/metabolism , Fatty Acids/metabolism , Ketone Bodies/metabolism , PPAR alpha/metabolism , Receptors, Progesterone/metabolism , Animals , Humans , Mice , Protein Stability , Receptors, Progesterone/deficiency
2.
Cell Commun Signal ; 19(1): 42, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33832499

ABSTRACT

BACKGROUND: Progesterone receptor membrane component 1 (Pgrmc1) is a non-classical progesterone receptor associated with the development of the mammary gland and xenograft-induced breast cancer. Importantly, Pgrmc1 is associated with the expression of estrogen receptor alpha and can be used for predicting the prognosis of breast cancer. Whether the genetic deletion of Pgrmc1 affects the progression of breast cancer is still unclear. METHODS: We used MMTV-PyMT transgenic mice that spontaneously develop breast tumors. In backcrossed FVB Pgrmc1 knockout (KO) mice, we monitored the development of the primary tumor and lung metastasis. In MCF-7 and MDA-MB-231 tumor cell lines, the migratory activity was evaluated after Pgrmc1 knockdown. RESULTS: There was no significant difference in the development of breast cancer in terms of tumor size at 13 weeks of age between WT and Pgrmc1 KO mice. However, Pgrmc1 KO mice had a significantly longer survival duration compared with WT mice. Furthermore, Pgrmc1 KO mice exhibited a significantly lower degree of lung metastasis. Compared with those of WT mice, the tumors of Pgrmc1 KO mice had a low expression of focal adhesion kinase and epithelial-mesenchymal transition markers. PGRMC1 knockdown resulted in a significantly reduced migration rate in breast cancer cell lines. CONCLUSIONS: Pgrmc1 KO mice with breast cancer had a prolonged survival, which was accompanied by a low degree of lung metastasis. PGRMC1 showed a significant role in the migration of breast cancer cells, and may serve as a potential therapeutic target in breast cancer. Video Abstract.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement , Membrane Proteins/deficiency , Receptors, Progesterone/deficiency , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival , Female , Gene Deletion , Humans , Lung Neoplasms/secondary , Male , Membrane Proteins/metabolism , Mice, Knockout , Neoplasm Metastasis , Receptors, Progesterone/metabolism
3.
Biochem Biophys Res Commun ; 550: 151-157, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33706098

ABSTRACT

Endometriosis is a disorder in which endometrial cells normally limited to the lining of the uterus proliferate outside the uterine cavity and can cause pelvic pain and infertility. ARID1A levels are significantly reduced in the eutopic endometrium from women with endometriosis. Uterine specific Arid1a knock-out mice were infertile due to loss of epithelial progesterone receptor (PGR) signaling. However, the functional association of ARID1A and PGR in endometriosis has not been studied. We examined the expression patterns and co-localization of ARID1A and PGR in eutopic endometrium from women with and without endometriosis using immunostaining and Western blot analysis. ARID1A and PGR proteins co-localized in the epithelium during the proliferative and the early secretory phases. Our immunoprecipitation analysis and proximity ligation assay (PLA) revealed physical interaction between ARID1A and PGR-A but not PGR-B in the mouse and human endometrium. ARID1A levels positively correlated with PGR levels in the eutopic endometrium of women with endometriosis. Our results bring new perspectives on the molecular mechanisms involved in endometrial receptivity and progesterone resistance in endometriosis. The interrelationship between ARID1A and PGR may contribute to explaining the non-receptive endometrium in endometriosis-related infertility.


Subject(s)
DNA-Binding Proteins/metabolism , Endometriosis/metabolism , Endometrium/metabolism , Receptors, Progesterone/metabolism , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/deficiency , Endometriosis/pathology , Endometrium/pathology , Female , HEK293 Cells , Humans , Immunoprecipitation , Mice , Protein Binding , Receptors, Progesterone/deficiency , Transcription Factors/deficiency
4.
Biomed Res Int ; 2020: 8823270, 2020.
Article in English | MEDLINE | ID: mdl-33134390

ABSTRACT

It is unclear whether the methyltransferase-like 14 (METTL14) protein promotes or suppresses cancer growth. We examined the association between METTL14 expression, cancer progression, and patient prognosis in a total of 398 breast cancer tissue specimens. Significantly fewer cancer tissue specimens compared with normal breast tissue expressed high levels of METTL14 (52.8% vs. 75.0%). METTL14 expression was negatively associated with tumor grade and positively associated with patient age, estrogen, and progesterone receptor status. High METTL14 expression was more common in luminal A and luminal B tissue (75.9% and 60.8%, respectively), compared with human epidermal growth factor receptor 2- (HER2-) enriched and triple-negative breast cancer (TNBC) samples (38.2% and 18.6%, respectively). In multiple logistic regression analysis, independent predictors of METTL14 expression in breast cancer included higher tumor grade (odds ratio (OR) = 0.494, 95% confidence interval (CI): 0.289-0.844; P = 0.010), TNBC subtype (OR = 0.109, 95% CI: 0.054-0.222; P < 0.001), and HER2-enriched subtype (OR = 0.298, 95% CI: 0.156-0.567; P < 0.001). No clear relationship was observed between patient prognosis and METTL14 expression. It appears that downregulated METTL14 expression in breast cancer is associated with tumor grade and molecular classification.


Subject(s)
Breast Neoplasms/genetics , Methyltransferases/genetics , Receptor, ErbB-2/genetics , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Triple Negative Breast Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Case-Control Studies , Female , Humans , Methyltransferases/metabolism , Middle Aged , Neoplasm Grading , Neoplasm Staging , Receptor, ErbB-2/deficiency , Receptors, Estrogen/deficiency , Receptors, Progesterone/deficiency , Receptors, Progesterone/metabolism , Survival Analysis , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology
5.
J Endocrinol ; 244(1): 111-122, 2020 01.
Article in English | MEDLINE | ID: mdl-31585440

ABSTRACT

The progesterone receptor (PR, encoded by Pgr) plays essential roles in reproduction. Female mice lacking the PR are infertile, due to the loss of the protein's functions in the brain, ovary, and uterus. PR is also expressed in pituitary gonadotrope cells, but its specific role therein has not been assessed in vivo. We therefore generated gonadotrope-specific Pgr conditional knockout mice (cKO) using the Cre-LoxP system. Overall, both female and male cKO mice appeared phenotypically normal. cKO females displayed regular estrous cycles (vaginal cytology) and normal fertility (litter size and frequency). Reproductive organ weights were comparable between wild-type and cKO mice of both sexes, as were production and secretion of the gonadotropins, LH and FSH, with one exception. On the afternoon of proestrus, the amplitude of the LH surge was blunted in cKO females relative to controls. Contrary to predictions of earlier models, this did not appear to derive from impaired GnRH self-priming. Collectively, these data indicate that PR function in gonadotropes may be limited to regulation of LH surge amplitude in female mice via a currently unknown mechanism.


Subject(s)
Estrous Cycle/genetics , Gonadotrophs/metabolism , Luteinizing Hormone/metabolism , Receptors, Progesterone/deficiency , Animals , Female , Follicle Stimulating Hormone , Gonadotropins/metabolism , Male , Mice , Mice, Knockout
6.
Gen Comp Endocrinol ; 285: 113275, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31536721

ABSTRACT

The progestin receptor membrane components (Pgrmcs) contain two paralogs, Pgrmc1 and Pgrmc2. Our previous research into single knockout of Pgrmc1 or Pgrmc2 suggests that Pgrmc1 and Pgrmc2 regulate membrane progestin receptor or steroid synthesis and therefore female fertility in zebrafish. Additional roles of Pgrmcs may not be determined in using single Pgrmc knockouts due to compensatory roles between Pgrmc1 and Pgrmc2. To address this question, we crossed single knockout pgrmc1 (pgrmc1-/-) with pgrmc2 (pgrmc2-/-), and generated double knockouts for both pgrmc1 and pgrmc2 (pgrmc1/2-/-) in a vertebrate model, zebrafish. In addition to the delayed oocyte maturation and reduced female fertility, significant reduced ovulation was found in double knockout (pgrmc1/2-/-) in vivo, though not detected in either single knockout of Pgrmc (pgrmc1-/- or pgrmc2-/-). We also found significant down regulation of nuclear progestin receptor (Pgr) protein expression only in pgrmc1/2-/-, which was most likely the cause of reduced ovulation. Lower protein expression of Pgr also resulted in reduced expression of metalloproteinase in pgrmc1/2-/-. With this study, we have provided new evidence for the physiological functions of Pgrmcs in the regulation of female fertility by regulation of ovulation, likely via regulation of Pgr, which affects regulation of metalloproteinase expression and oocyte ovulation.


Subject(s)
Cell Nucleus/metabolism , Down-Regulation , Gene Knockout Techniques , Infertility/genetics , Membrane Proteins/deficiency , Receptors, Progesterone/genetics , Zebrafish Proteins/deficiency , Zebrafish/genetics , Animals , Female , Membrane Proteins/metabolism , Metalloproteases/metabolism , Oocytes/metabolism , Oogenesis , Ovary/metabolism , Ovulation , Receptors, Progesterone/deficiency , Receptors, Progesterone/metabolism , Zebrafish/metabolism , Zebrafish Proteins/metabolism
7.
Nature ; 576(7785): 138-142, 2019 12.
Article in English | MEDLINE | ID: mdl-31748741

ABSTRACT

Haem is an essential prosthetic group of numerous proteins and a central signalling molecule in many physiologic processes1,2. The chemical reactivity of haem means that a network of intracellular chaperone proteins is required to avert the cytotoxic effects of free haem, but the constituents of such trafficking pathways are unknown3,4. Haem synthesis is completed in mitochondria, with ferrochelatase adding iron to protoporphyrin IX. How this vital but highly reactive metabolite is delivered from mitochondria to haemoproteins throughout the cell remains poorly defined3,4. Here we show that progesterone receptor membrane component 2 (PGRMC2) is required for delivery of labile, or signalling haem, to the nucleus. Deletion of PGMRC2 in brown fat, which has a high demand for haem, reduced labile haem in the nucleus and increased stability of the haem-responsive transcriptional repressors Rev-Erbα and BACH1. Ensuing alterations in gene expression caused severe mitochondrial defects that rendered adipose-specific PGRMC2-null mice unable to activate adaptive thermogenesis and prone to greater metabolic deterioration when fed a high-fat diet. By contrast, obese-diabetic mice treated with a small-molecule PGRMC2 activator showed substantial improvement of diabetic features. These studies uncover a role for PGRMC2 in intracellular haem transport, reveal the influence of adipose tissue haem dynamics on physiology and suggest that modulation of PGRMC2 may revert obesity-linked defects in adipocytes.


Subject(s)
Adipocytes/metabolism , Heme/metabolism , Membrane Proteins/metabolism , Receptors, Progesterone/metabolism , Animals , Homeostasis , Humans , Intracellular Space/metabolism , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Molecular Chaperones/metabolism , Receptors, Progesterone/deficiency , Receptors, Progesterone/genetics , Transcription, Genetic
8.
PLoS Genet ; 14(11): e1007787, 2018 11.
Article in English | MEDLINE | ID: mdl-30452456

ABSTRACT

Successful embryo implantation requires a receptive endometrium. Poor uterine receptivity can account for implantation failure in women who experience recurrent pregnancy loss or multiple rounds of unsuccessful in vitro fertilization cycles. Here, we demonstrate that the transcription factor Forkhead Box O1 (FOXO1) is a critical regulator of endometrial receptivity in vivo. Uterine ablation of Foxo1 using the progesterone receptor Cre (PgrCre) mouse model resulted in infertility due to altered epithelial cell polarity and apoptosis, preventing the embryo from penetrating the luminal epithelium. Analysis of the uterine transcriptome after Foxo1 ablation identified alterations in gene expression for transcripts involved in the activation of cell invasion, molecular transport, apoptosis, ß-catenin (CTNNB1) signaling pathway, and an increase in PGR signaling. The increase of PGR signaling was due to PGR expression being retained in the uterine epithelium during the window of receptivity. Constitutive expression of epithelial PGR during this receptive period inhibited expression of FOXO1 in the nucleus of the uterine epithelium. The reciprocal expression of PGR and FOXO1 was conserved in human endometrial samples during the proliferative and secretory phase. This demonstrates that expression of FOXO1 and the loss of PGR during the window of receptivity are interrelated and critical for embryo implantation.


Subject(s)
Embryo Implantation/genetics , Embryo Implantation/physiology , Endometrium/metabolism , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Animals , Cell Nucleus/metabolism , Cell Polarity/genetics , Cell Polarity/physiology , Decidua/physiology , Endometrium/cytology , Female , Forkhead Box Protein O1/deficiency , Gene Expression Profiling , Humans , Mice , Mice, Knockout , Pregnancy , Receptors, Progesterone/deficiency , Signal Transduction
9.
Sci Rep ; 8(1): 15711, 2018 10 24.
Article in English | MEDLINE | ID: mdl-30356113

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) results from triglyceride accumulation within the liver and some of them advances to non-alcoholic steatohepatitis (NASH). It is important to note that in NAFLD development, hepatic de novo lipogenesis (DNL) derives from excess carbohydrates and fats under a condition of excess energy through ß-oxidation. As a main regulator for DNL, sterol regulatory element-binding protein 1 (Srebp-1) forms complex with progesterone receptor membrane component 1 (Pgrmc1). To investigate whether Pgrmc1 may have a notable effect on DNL via SREBP-1 activation, we generated Pgrmc1 knockout (KO) mice and fed a high fat diet for one month. High-fat-fed Pgrmc1 KO mice showed a substantial increase in levels of hepatic TG accumulation, and they were predisposed to NAFLD when compared to WT mice. Loss of Pgrmc1 increased mature SREBP-1 protein level, suggesting that induction of hepatic steatosis in Pgrmc1 KO mice might be triggered by de novo lipogenesis. Moreover, Pgrmc1 KO mice were also more vulnerable to early stage of NASH, showing high levels of alanine aminotransferase, obesity-linked pro-inflammatory cytokines, and fibrosis markers. This is interesting because Pgrmc1 involves with the first step in regulating the hepatic de novo lipogenesis under an excess energy condition.


Subject(s)
Lipogenesis , Membrane Proteins/deficiency , Non-alcoholic Fatty Liver Disease/etiology , Receptors, Progesterone/deficiency , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Diet, High-Fat , Membrane Proteins/metabolism , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/pathology , Receptors, Progesterone/metabolism , Triglycerides/metabolism
10.
Biol Reprod ; 99(4): 864-876, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29741588

ABSTRACT

Preterm birth (PTB), parturition prior to 37 weeks' gestation, is the leading cause of neonatal mortality. The causes of spontaneous PTB are poorly understood; however, recent studies suggest that this condition may arise as a consequence of the parental fetal environment. Specifically, we previously demonstrated that developmental exposure of male mice (F1 animals) to the environmental endocrine disruptor 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) was associated with reduced sperm quantity/quality in adulthood and control female partners frequently delivered preterm. Reproductive defects persisted in the F2 and F3 descendants, and spontaneous PTB was common. Reproductive changes in the F3 males, the first generation without direct TCDD exposure, suggest the occurrence of epigenetic alterations in the sperm, which have the potential to impact placental development. Herein, we conducted an epigenetic microarray analysis of control and F1 male-derived placentae, which identified 2171 differentially methylated regions, including the progesterone receptor (Pgr) and insulin-like growth factor (Igf2). To assess if Pgr and Igf2 DNA methylation changes were present in sperm and persist in future generations, we assessed methylation and expression of these genes in F1/F3 sperm and F3-derived placentae. Although alterations in methylation and gene expression were observed, in most tissues, only Pgr reached statistical significance. Despite the modest gene expression changes in Igf2, offspring of F1 and F3 males consistently exhibited IUGR. Taken together, our data indicate that paternal developmental TCDD exposure is associated with transgenerational placental dysfunction, suggesting epigenetic modifications within the sperm have occurred. An evaluation of additional genes and alternative epigenetic mechanisms is warranted.


Subject(s)
Epigenesis, Genetic , Insulin-Like Growth Factor II/genetics , Paternal Exposure/adverse effects , Placenta/metabolism , Receptors, Progesterone/genetics , Spermatozoa/metabolism , Animals , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Disease Models, Animal , Endocrine Disruptors/toxicity , Epigenesis, Genetic/drug effects , Female , Fetal Growth Retardation/etiology , Insulin-Like Growth Factor II/deficiency , Insulin-Like Growth Factor II/metabolism , Male , Mice , Mice, Inbred C57BL , Placentation/genetics , Polychlorinated Dibenzodioxins/toxicity , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Progesterone/deficiency , Receptors, Progesterone/metabolism
11.
Int J Mol Sci ; 19(2)2018 Feb 02.
Article in English | MEDLINE | ID: mdl-29393914

ABSTRACT

Breast cancer is the most common malignancy in women and the second leading cause of cancer death in women. Triple negative breast cancer (TNBC) subtype is a breast cancer subset without ER (estrogen receptor), PR (progesterone receptor) and HER2 (human epidermal growth factor receptor 2) expression, limiting treatment options and presenting a poorer survival rate. Thus, we investigated whether histone deacetylation inhibitor (HDACi) could be used as potential anti-cancer therapy on breast cancer cells. In this study, we found TNBC and HER2-enriched breast cancers are extremely sensitive to Panobinostat, Belinostat of HDACi via experiments of cell viability assay, apoptotic marker identification and flow cytometry measurement. On the other hand, we developed a bioluminescence-based live cell non-invasive apoptosis detection sensor (NIADS) detection system to evaluate the quantitative and kinetic analyses of apoptotic cell death by HDAC treatment on breast cancer cells. In addition, the use of HDACi may also contribute a synergic anti-cancer effect with co-treatment of chemotherapeutic agent such as doxorubicin on TNBC cells (MDA-MB-231), but not in breast normal epithelia cells (MCF-10A), providing therapeutic benefits against breast tumor in the clinic.


Subject(s)
Antineoplastic Agents/pharmacology , Biological Assay , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Sulfonamides/pharmacology , Cell Line, Tumor , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Flow Cytometry , Histone Deacetylases/metabolism , Humans , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Panobinostat , Receptor, ErbB-2/deficiency , Receptor, ErbB-2/genetics , Receptors, Estrogen/deficiency , Receptors, Estrogen/genetics , Receptors, Progesterone/deficiency , Receptors, Progesterone/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
12.
Mol Hum Reprod ; 23(9): 646-653, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28911214

ABSTRACT

STUDY QUESTION: Is Growth Regulation by Estrogen in Breast Cancer 1 (GREB1) required for progesterone-driven endometrial stromal cell decidualization? SUMMARY ANSWER: GREB1 is a novel progesterone-responsive gene required for progesterone-driven human endometrial stromal cell (HESC) decidualization. WHAT IS KNOWN ALREADY: Successful establishment of pregnancy requires HESCs to transform from fibroblastic to epithelioid cells in a process called decidualization. This process depends on the hormone progesterone, but the molecular mechanisms by which it occurs have not been determined. STUDY DESIGN, SIZE, DURATION: Primary and transformed HESCs in which GREB1 expression was knocked down were decidualized in culture for up to 6 days. Wild-type and progesterone receptor (PR) knockout mice were treated with progesterone, and their uteri were assessed for levels of GREB1 expression. PARTICIPANTS/MATERIALS, SETTING, METHODS: Analysis of previous data included data mining of expression profile data sets and in silico transcription factor-binding analysis. Endometrial biopsies obtained from healthy women of reproductive age during the proliferative phase (Days 8-12) of their menstrual cycle were used for isolating HESCs. Experiments were carried out with early passage (no more than four passages) HESCs isolated from at least three subjects. Transcript levels of decidualization markers prolactin (PRL) and insulin-like growth factor-binding protein-1 (IGFBP-1) were detected by quantitative RT-PCR as readouts for HESC decidualization. Cells were also imaged by phase-contrast microscopy. To assess the requirement for GREB1, PR and SRC-2, cells were transfected with specifically targeted small interfering RNAs. Results are shown as mean and SE from three replicates of one representative patient-derived primary endometrial cell line. Experiments were also conducted with transformed HESCs. MAIN RESULTS AND THE ROLE OF CHANCE: Progesterone treatment of mice and transformed HESCs led to an ~5-fold (5.6 ± 0.81, P < 0.05, and 5.2 ± 0.26, P < 0.01, respectively) increase in GREB1 transcript levels. This increase was significantly reduced in the uteri of PR knock-out mice (P < 0.01), in HESCs treated with the PR antagonist RU486 (P < 0.01), or in HESCs in which PR expression was knocked down (P < 0.05). When GREB1 expression was knocked down, progesterone-driven decidualization markers in both immortalized and primary HESCs was significantly reduced (P < 0.05 and P < 0.01). Finally, GREB1 knock down signficantly reduced expression of the PR target genes WNT4 and FOXOA1 (P < 0.05 and P < 0.01, respectively). LARGE SCALE DATA: This study used the Nuclear Receptor Signaling Atlas. LIMITATIONS, REASONS FOR CAUTION: Although in vitro cell culture studies indicate that GREB1 is required for endoemtrial decidualization, the in vivo role of GREB1 in endometrial function and dysfunction should be assessed by using knock-out mouse models. WIDER IMPLICATIONS OF THE FINDINGS: Identification and functional analysis of GREB1 as a key molecular mediator of decidualization may lead to improved diagnosis and clinical management of women with peri-implantation loss due to inadequate endometrial decidualization. STUDY FUNDING AND COMPETING INTEREST(S): This research was funded in part by: a National Institutes of Health (NIH)/ National Institute of Child Health and Human Development (NICHD) grant (R00 HD080742) and Washington University School of Medicine start-up funds to R.K., an NIH/NICHD grant (RO1 HD-07857) to B.W.O.M., and a NIH/NICHD grant (R01 HD-042311) to J.P.L. The authors declare no conflicts of interests.


Subject(s)
Decidua/drug effects , Gene Expression Regulation, Developmental , Neoplasm Proteins/genetics , Progesterone/pharmacology , Receptors, Progesterone/genetics , Stromal Cells/drug effects , Animals , Cell Differentiation , Decidua/cytology , Decidua/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor Binding Protein 1/metabolism , Mice , Mice, Knockout , Mifepristone/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Nuclear Receptor Coactivator 2/antagonists & inhibitors , Nuclear Receptor Coactivator 2/genetics , Nuclear Receptor Coactivator 2/metabolism , Pregnancy , Primary Cell Culture , Prolactin/genetics , Prolactin/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/deficiency , Signal Transduction , Stromal Cells/cytology , Stromal Cells/metabolism , Wnt4 Protein/genetics , Wnt4 Protein/metabolism
13.
Reprod Domest Anim ; 52(1): 174-178, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27862412

ABSTRACT

A three-year-old intact female Old English sheepdog was presented for evaluation of infertility. A uterine biopsy was performed during dioestrus, and the microscopic appearance was inconsistent with progesterone stimulation; the glands were sparse, simple and failed to show coiling, while the glandular epithelium was cuboidal instead of columnar. There was very little evidence of glandular activity. Due to the inappropriate appearance of the glands for the stage of the cycle, immunohistochemistry for progesterone receptors was performed. No progesterone receptor-positive immunoreactivity was identified in the endometrial luminal epithelium, glandular epithelium or stroma. Weak intranuclear immunoreactivity was identified within the smooth muscle cells of the myometrium. The absence of progesterone receptors within the endometrial glands is the most likely explanation for the abnormal appearance of the endometrium and for this bitch's infertility. To our knowledge, this is the first report of endometrial progesterone receptor absence in a bitch.


Subject(s)
Endometrium/pathology , Infertility, Female/veterinary , Progesterone/blood , Receptors, Progesterone/deficiency , Animals , Dogs , Female , Immunohistochemistry , Infertility, Female/etiology , Pregnancy
14.
FEBS Lett ; 590(24): 4606-4616, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27859250

ABSTRACT

Development of targeted therapies for triple-negative breast cancer (TNBC, a more aggressive subtype) is an unmet medical need. We analyzed data from 887 patients with invasive breast cancer and observed that increased Wnt and histone deacetylase (HDAC) activities are associated with estrogen receptor 1 (ESR1) and progesterone receptor (PGR) repression, poor survival, and increased relapse. The inverse correlation between Wnt signaling and repression of ESR1 and PGR expression was found to be magnified in cancer stem cell (CSC) subpopulations in TNBC cell lines. Cosuppression of Wnt, HDAC, and ESR1 using clinically relevant low-dose inhibitors effectively repressed both bulk and CSC subpopulations and converted CSCs to non-CSCs in TNBC cells without affecting MCF-10A mammary epithelial cells.


Subject(s)
Antineoplastic Agents/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Histone Deacetylases/genetics , Neoplastic Stem Cells/drug effects , Triple Negative Breast Neoplasms/genetics , Wnt Proteins/antagonists & inhibitors , CD24 Antigen/genetics , CD24 Antigen/metabolism , Cell Line , Cell Proliferation/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor, ErbB-2/deficiency , Receptor, ErbB-2/genetics , Receptors, Progesterone/deficiency , Receptors, Progesterone/genetics , Signal Transduction , Survival Analysis , Tamoxifen/pharmacology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , Valproic Acid/pharmacology , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/antagonists & inhibitors , beta Catenin/genetics , beta Catenin/metabolism
15.
Curr Mol Med ; 16(3): 276-87, 2016.
Article in English | MEDLINE | ID: mdl-26917262

ABSTRACT

Endometriosis is a major cause of infertility and pelvic pain, affecting more than 10% of reproductive-aged women. Progesterone resistance has been observed in the endometrium of women with this disease, as evidenced by alterations in progesterone-responsive gene and protein expression. cAMPResponse Element-Binding 3-like protein 1 (Creb3l1) has previously been identified as a progesterone receptor (PR) target gene in mouse uterus via high density DNA microarray analysis. However, CREB3L1 function has not been studied in the context of endometriosis and uterine biology. In this study, we validated progesterone (P4) regulation of Creb3l1 in the uteri of wild-type and progesterone receptor knockout (PRKO) mice. Furthermore, we observed that CREB3L1 expression was significantly higher in secretory phase human endometrium compared to proliferative phase and that CREB3L1 expression was significantly decreased in the endometrium of women with endometriosis. Lastly, by transfecting CREB3L1 siRNA into cultured human endometrial stromal cells (hESCs) prior to hormonal induction of in vitro decidualization, we showed that CREB3L1 is required for the decidualization process. Interestingly, phosphorylation of ERK1/2, critical factor for decidualization, was also significantly reduced in CREB3L1-silenced hESCs. It is known that hESCs from patients with endometriosis show impaired decidualization and that dysregulation of the P4-PR signaling axis is linked to a variety of endometrial diseases including infertility and endometriosis. Therefore, these results suggest that CREB3L1 is required for decidualization in mice and humans and may be linked to the pathogenesis of endometriosis in a P4-dependent manner.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Endometriosis/genetics , Endometrium/metabolism , Nerve Tissue Proteins/genetics , Progesterone/pharmacology , Receptors, Progesterone/genetics , Adult , Animals , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Cyclic AMP Response Element-Binding Protein/metabolism , Endometriosis/metabolism , Endometriosis/pathology , Endometriosis/surgery , Endometrium/pathology , Female , Gene Expression Regulation , Humans , Hysterectomy , Menstrual Cycle/drug effects , Menstrual Cycle/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Progesterone/deficiency , Signal Transduction , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology
16.
PLoS One ; 11(1): e0145647, 2016.
Article in English | MEDLINE | ID: mdl-26751376

ABSTRACT

MTAP is a ubiquitously expressed gene important for adenine and methionine salvage. The gene is located at 9p21, a chromosome region often deleted in breast carcinomas, similar to CDKN2A, a recognized tumor suppressor gene. Several research groups have shown that MTAP acts as a tumor suppressor, and some therapeutic approaches were proposed based on a tumors´ MTAP status. We analyzed MTAP and CDKN2A gene (RT-qPCR) and protein (western-blotting) expression in seven breast cancer cell lines and evaluated their promoter methylation patterns to better characterize the contribution of these genes to breast cancer. Cytotoxicity assays with inhibitors of de novo adenine synthesis (5-FU, AZA and MTX) after MTAP gene knockdown showed an increased sensitivity, mainly to 5-FU. MTAP expression was also evaluated in two groups of samples from breast cancer patients, fresh tumors and paired normal breast tissue, and from formalin-fixed paraffin embedded (FFPE) core breast cancer samples diagnosed as Luminal-A tumors and triple negative breast tumors (TNBC). The difference of MTAP expression between fresh tumors and normal tissues was not statistically significant. However, MTAP expression was significantly higher in Luminal-A breast tumors than in TNBC, suggesting the lack of expression in more aggressive breast tumors and the possibility of using the new approaches based on MTAP status in TNBC.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Carcinoma, Ductal, Breast/genetics , Carcinoma, Lobular/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression Regulation, Neoplastic , Purine-Nucleoside Phosphorylase/genetics , Triple Negative Breast Neoplasms/genetics , Adenocarcinoma, Mucinous/drug therapy , Adenocarcinoma, Mucinous/metabolism , Adenocarcinoma, Mucinous/pathology , Antineoplastic Agents/pharmacology , Azacitidine/pharmacology , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/drug therapy , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/pathology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Methylation , Estrogen Receptor alpha/deficiency , Estrogen Receptor alpha/genetics , Female , Fluorouracil/pharmacology , Humans , Lymphatic Metastasis , Methotrexate/pharmacology , Organ Specificity , Promoter Regions, Genetic , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Purine-Nucleoside Phosphorylase/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor, ErbB-2/deficiency , Receptor, ErbB-2/genetics , Receptors, Progesterone/deficiency , Receptors, Progesterone/genetics , Signal Transduction , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
17.
Appl Immunohistochem Mol Morphol ; 24(1): 42-50, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26200837

ABSTRACT

Receptor activator of NF-κB (RANK) and its ligand, RANKL, are essential for osteoclastogenesis and modulate osteolytic bone metastasis. The RANKL/RANK system is also fundamental for mammary gland development and plays a potential role in breast carcinogenesis. c-Src, a nonreceptor tyrosine kinase downstream of RANK, is overexpressed in most breast cancers and plays a key role in several transduction pathways. The aim of the study was to examine the expression of these molecules in tissue microarrays constructed from 62 advanced breast cancers and 10 breast cancers controls (no metastasis after follow-up). Significantly higher levels of RANK and lower levels of RANKL were found in triple-negative (ER-/PR-/HER2-) tumors when compared with luminal subtypes, whereas their levels in the HER2 subtype were quantitatively in between. RANK expression was significantly associated with tumor grade/differentiation by multivariate analysis. Despite their high expression in bone, neither molecule in primary tumors seemed to be related to a bone-seeking phenotype. Rather, they were significantly correlated with a brain-metastatic phenotype. RANKL and RANK were significantly associated with survival outcomes. Further, Src expression showed a significantly positive linear relationship with RANK, suggesting a potential mechanism of the RANKL-RANK axis in regulating breast cancer cell differentiation and antiapoptosis. Thus, these molecules may be potential therapeutic targets, especially in triple-negative tumors, for which the only systemic treatment option is cytotoxic chemotherapy.


Subject(s)
Bone Neoplasms/diagnosis , Brain Neoplasms/diagnosis , Breast Neoplasms/diagnosis , Carcinoma, Ductal, Breast/diagnosis , RANK Ligand/genetics , Receptor Activator of Nuclear Factor-kappa B/genetics , src-Family Kinases/genetics , Adult , Aged , Aged, 80 and over , Bone Neoplasms/genetics , Bone Neoplasms/mortality , Bone Neoplasms/secondary , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/secondary , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/mortality , Carcinoma, Ductal, Breast/secondary , Cell Differentiation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Estrogen Receptor alpha/deficiency , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Neoplasm Grading , Neoplasm Staging , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptor, ErbB-2/deficiency , Receptor, ErbB-2/genetics , Receptors, Progesterone/deficiency , Receptors, Progesterone/genetics , Signal Transduction , Survival Analysis , Tissue Array Analysis , src-Family Kinases/metabolism
18.
PLoS One ; 10(10): e0139490, 2015.
Article in English | MEDLINE | ID: mdl-26431032

ABSTRACT

The effect of progesterone on bone remains elusive. We previously reported that global progesterone receptor (PR) knockout mice displayed high bone mass phenotype, suggesting that PR influences bone growth and modeling. Recently, Mx1+ cells were characterized to be mesenchymal stem cell-like pluripotent Cells. The aim of this study was to evaluate whether the PR in Mx1+ cells regulates osteogenesis. Using the Mx1-Cre;mT/mG reporter mouse model, we found that the calvarial cells exhibited minimal background Mx1-Cre activity prior to Cre activation by IFNα treatment as compared to the bone marrow stromal cells. IFNα treatment significantly activated Mx1-Cre in the calvarial cells. When the PR gene was deleted in the Mx1-Cre;PR-flox calvarial cells in vitro, significantly higher levels of expression of osteoblast maturation marker genes (RUNX2, Osteocalcin, and Dmp1) and osteogenic potential were detected. The PR-deficient calvariae exhibited greater bone volume, especially in the males. Although Mx1-Cre activity could be induced on the bone surface in vivo, the Mx1+ cells did not differentiate into osteocytes in long bones. Bone volumes at the distal femurs and the bone turnover marker serum Osteocalcin were similar between the Mx1-Cre;PR-flox mutant mice and the corresponding wild types in both sexes. In conclusion, our data demonstrates that blocking progesterone signaling via PRs in calvarial Mx1+ cells promoted osteoblast differentiation in the calvaria. Mx1+ was expressed by heterogeneous cells in bone marrow and did not differentiate into osteocyte during long bone development in vivo. Selectively inactivating the PR gene in Mx1+ cells affected the membrane bone formation but did not affect peripheral skeletal homeostasis.


Subject(s)
Femur/pathology , Gene Knockout Techniques , Interferon-alpha/pharmacology , Mesenchymal Stem Cells/cytology , Myxovirus Resistance Proteins/genetics , Osteoblasts/pathology , Osteogenesis/physiology , Pluripotent Stem Cells/cytology , Promoter Regions, Genetic/drug effects , Receptors, Progesterone/deficiency , Skull/pathology , Animals , Biomarkers , Bone Marrow/pathology , Cells, Cultured , Chondrocytes/pathology , Crosses, Genetic , Female , Gene Expression Regulation, Developmental , Genes, Reporter , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Growth Plate/pathology , Integrases , Luminescent Proteins/analysis , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Organ Specificity , Osteogenesis/drug effects , Pluripotent Stem Cells/metabolism , Progesterone/physiology , Receptors, Progesterone/genetics , Receptors, Progesterone/physiology , Recombinant Fusion Proteins/metabolism , Sex Characteristics , Red Fluorescent Protein
19.
Sci Rep ; 5: 14943, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26456840

ABSTRACT

Migraine headache is often timed with the menstrual cycle. Some studies have reported reduced risk of breast cancer in migraineurs but most of those did not distinguish menstrually-related from non-menstrually-related migraine. To examine the possible associations between breast cancer and migraine overall and between cancer subcategories and the two migraine subtypes, we used a cohort study of 50,884 women whose sister had breast cancer and a sister-matched case-control study including 1,418 young-onset (<50 years) breast cancer cases. We analyzed the two studies individually and also in tandem via a hybrid Cox model, examining subcategories of breast cancer in relation to menstrually-related and non-menstrually-related migraine. History of migraine was not associated with breast cancer overall. Migraine showed an inverse association with ductal carcinoma in situ (HR = 0.77; 95% CI (0.62,0.96)). Also, women with non-menstrually-related migraine had increased risk (HR = 1.30, 95% CI (0.93,1.81)) while women with menstrually-related migraine had decreased risk (HR = 0.63, 95% CI (0.42,0.96)) of hormone-receptor-negative (ER-/PR-) cancer, with a significant contrast in estimated effects (P = 0.005). While replication of these subset-based findings will be needed, effect specificity could suggest that while migraine has little overall association with breast cancer, menstrual migraine may be associated with reduced risk of ER-/PR- breast cancer.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Migraine Disorders/genetics , Triple Negative Breast Neoplasms/genetics , Adult , Aged , Breast Neoplasms/complications , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/complications , Carcinoma, Intraductal, Noninfiltrating/pathology , Case-Control Studies , Female , Gene Expression , Humans , Menstruation , Middle Aged , Migraine Disorders/complications , Migraine Disorders/pathology , Proportional Hazards Models , Receptor, ErbB-2/deficiency , Receptor, ErbB-2/genetics , Receptors, Estrogen/deficiency , Receptors, Estrogen/genetics , Receptors, Progesterone/deficiency , Receptors, Progesterone/genetics , Retrospective Studies , Siblings , Triple Negative Breast Neoplasms/complications , Triple Negative Breast Neoplasms/pathology
20.
Breast Cancer Res Treat ; 153(1): 219-34, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26253945

ABSTRACT

DNA methylation at the 5 position of cytosine (5 mC) is an epigenetic hallmark in cancer. The 5 mC can be converted to 5-hydroxymethylcytosine (5 hmC) through a ten-eleven-translocation (TET). We investigated the impact of 5 mC, 5 hmC, TET1, and TET2 on tumorigenesis and prognosis of breast cancer. Immunohistochemistry was used to assess the levels of 5 mC, 5 hmC, TET1, and TET2 in the corresponding tumor adjacent normal (n = 309), ductal carcinoma in situ (DCIS, n = 120), and invasive ductal carcinoma (IDC, n = 309) tissues for 309 breast ductal carcinoma patients. 5 mC, 5 hmC, TET1-n, and TET2-n were significantly decreased during DCIS and IDC progression. In IDC, the decrease of 5 hmC was correlated with the cytoplasmic mislocalization of TET1 (p < 0.001) as well as poor disease-specific survival (DSS) (adjusted hazard ratio [AHR] 1.95, p = 0.003) and disease-free survival (DFS) (AHR 1.91, p = 0.006). The combined decrease of 5 mC and 5 hmC was correlated with worse DSS (AHR 2.19, p = 0.008) and DFS (AHR 1.99, p = 0.036). Stratification analysis revealed that the low level of 5 mC was associated with poor DSS (AHR 1.89, p = 0.044) and DFS (AHR 2.02, p = 0.035) for the ER/PR-positive subtype. Conversely, the low level of 5 hmC was associated with worse DSS (AHR 2.77, p = 0.002) and DFS (AHR 2.69, p = 0.006) for the ER/PR-negative subtype. The decreases of 5 mC, 5 hmC, TET1-n, and TET2-n were biomarkers of tumor development. The global reduction of 5 hmC was a poor prognostic factor for IDC, especially for ER/PR-negative subtype.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cytosine/analogs & derivatives , DNA Methylation , 5-Methylcytosine/analogs & derivatives , Adult , Aged , Breast Neoplasms/epidemiology , Breast Neoplasms/pathology , Cytosine/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases , Female , Gene Expression , Humans , Immunohistochemistry , Middle Aged , Mixed Function Oxygenases , Neoplasm Grading , Neoplasm Staging , Prognosis , Protein Transport , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptors, Estrogen/deficiency , Receptors, Progesterone/deficiency , Risk Factors , Survival Analysis , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...