Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Biol Reprod ; 86(3): 86, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22190699

ABSTRACT

Prolactin (PRL), a pleiotropic hormone essential for maintenance of corpus luteum (CL) function and pregnancy, transduces its signal through two types of receptors, a short form (PRLR-S) and a long form (PRLR-L). Both types of receptors are expressed in the CL, yet their individual roles are not well defined. We have shown previously that female transgenic mice expressing only PRLR-S display total infertility characterized by defective follicular development and early degeneration of CL, suggesting that expression of PRLR-L is a prerequisite for normal follicular development and maintenance of CL. To determine whether PRLR-L alone is the sole receptor required to maintain normal CL formation, differentiation, and progesterone secretion, we generated two transgenic mice which express only PRLR-L, either ubiquitously (Tg-RL) or in a CL-specific manner (CL-RL). To generate CL-specific expression, we used the HSD17B7 promoter. We found both transgenic mice models cycled normally, displayed no apparent defect in follicular development, and had normal ovulation rates. The STAT5 signaling pathway, considered essential for luteinization and progesterone production, was activated by PRL in both transgenic mice models. However, soon after mating, Tg-RL and CL-RL mice showed early regression of CL, lack of progesterone production, and implantation failure that rendered them totally infertile. Embryo transfer studies demonstrated no embryo abnormalities, and supplementation with progesterone rescued implantation failure in these mice. Close observation revealed lack of luteinization and reduced expression of proteins involved in progesterone biosynthesis despite normal levels of LHCGR (LH-R), ESR1 (ER-alpha), CEBPB (C/EBP-beta) and CDKN1B (p27), proteins essential for luteinization. However, we found VEGFA, a key regulator of angiogenesis and vascularization, to be dramatically reduced in both Tg-RL and CL-RL mice. We also found collagen IV, a marker for the basal lamina of endothelial cells, aberrantly expressed and a discordant organization of endothelial cells in CL. Although luteinization did not occur in vivo, granulosa cells isolated from these mice luteinized in culture. Taken together, these results suggest that a vascularization defect in the CL may be responsible for lack of luteinization, progesterone production, and infertility in mice expressing only PRLR-L. This investigation therefore demonstrates that in contrast to earlier presumptions that PRLR-L alone is able to support normal CL formation and function, both isoforms of the PRL receptor are required in the CL for normal female fertility.


Subject(s)
Ovary/physiology , Receptors, Prolactin/chemistry , Receptors, Prolactin/physiology , Signal Transduction/physiology , Animals , Corpus Luteum/physiology , Female , Infertility, Female/metabolism , Infertility, Female/physiopathology , Mice , Mice, Transgenic , Models, Animal , Progesterone/metabolism , Protein Isoforms , Receptors, Prolactin/genetics , STAT Transcription Factors/physiology
2.
Mol Endocrinol ; 25(5): 754-66, 2011 May.
Article in English | MEDLINE | ID: mdl-21372145

ABSTRACT

Our laboratory has previously cloned and purified an ovarian protein found to be a novel 17ß-hydroxysteroid dehydrogenase type 7 enzyme (HSD17B7) (formerly prolactin receptor-associated protein) that converts the weak estrogen, estrone, to the highly potent estradiol. The regulation of this enzyme has not yet been explored. In this report, we show high expression of HSD17B7 in human ductal carcinoma and breast cancer cell lines and present evidence for a strong up-regulation of this enzyme by estradiol at the level of mRNA, protein expression, and promoter activity in MCF-7 cells. The effect of estradiol is mediated by estrogen receptor (ER)α, whereas ERß prevents this stimulation. ER antagonists, ICI 182,780 and 4-hydroxytamoxifen, prevent estradiol-induced stimulation of the endogenously expressed HSD17B7, suggesting that these inhibitors not only block estradiol action but also its production. We have identified a -185-bp region of the hsd17b7 promoter that is highly conserved among rat, mouse, and human and confers regulation by estradiol in MCF-7 cells. This region is devoid of a classical estradiol-response element but contains a nuclear factor 1 (NF1) site that is essential for estradiol action. We found that estradiol stimulates the recruitment and DNA binding of NF1 to this region of the hsd17b7 promoter. Furthermore, knockdown of NF1 family members, NF1B, NF1A, and NF1X, completely prevents induction of this gene by estradiol. In summary, our findings demonstrate that estradiol stimulates HSD17B7 transcriptional activity in breast cancer cells through a novel mechanism requiring NF1 and strongly suggest a positive feedback mechanism to increase local estradiol synthesis causing growth of estrogen-dependent breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Cell Cycle Proteins/genetics , Estradiol/biosynthesis , S100 Proteins/genetics , Animals , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Feedback, Physiological , Female , Gene Expression Regulation, Neoplastic , Humans , Immune Sera , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mice , NFI Transcription Factors/genetics , NFI Transcription Factors/metabolism , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Rabbits , Response Elements , S100 Calcium Binding Protein A6 , S100 Proteins/metabolism , Transcription, Genetic
3.
J Biol Chem ; 286(9): 7609-18, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21199871

ABSTRACT

Prolactin (PRL) is essential for normal reproduction and signals through two types of receptors, the short (PRL-RS) and long (PRL-RL) form. We have previously shown that transgenic mice expressing only PRL-RS (PRLR(-/-)RS) display abnormal follicular development and premature ovarian failure. Here, we report that MAPK, essential for normal follicular development, is critically inhibited by PRL in reproductive tissues of PRLR(-/-)RS mice. Consequently, the phosphorylation of MAPK downstream targets are also markedly inhibited by PRL without affecting immediate upstream kinases, suggesting involvement of MAPK specific phosphatase(s) in this inhibition. Similar results are obtained in a PRL-responsive ovary-derived cell line (GG-CL) that expresses only PRL-RS. However, we found the expression/activation of several known MAPK phosphatases not to be affected by PRL, suggesting a role of unidentified phosphatase(s). We detected a 27-kDa protein that binds to the intracellular domain of PRL-RS and identified it as dual specific phosphatase DUPD1. PRL does not induce expression of DUDP1 but represses its phosphorylation on Thr-155. We also show a physical association of this phosphatase with ERK1/2 and p38 MAPK. Using an in vitro phosphatase assay and overexpression studies, we established that DUPD1 is a MAPK phosphatase. Dual specific phosphatase inhibitors as well as siRNA to DUPD1, completely prevent PRL-mediated MAPK inhibition in ovarian cells. Our results strongly suggest that deactivation of MAPK by PRL/PRL-RS contributes to the severe ovarian defect in PRLR(-/-)RS mice and demonstrate the novel association of PRL-RS with DUPD1 and a role for this phosphatase in MAPK deactivation.


Subject(s)
Decidua/metabolism , Dual Specificity Phosphatase 1/metabolism , MAP Kinase Signaling System/physiology , Ovary/metabolism , Prolactin/metabolism , Receptors, Prolactin/metabolism , Animals , Cell Line , Decidua/cytology , Dual Specificity Phosphatase 1/genetics , Female , MAP Kinase Signaling System/drug effects , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Ovary/cytology , Ovary/growth & development , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/metabolism , Pregnancy , Primary Ovarian Insufficiency/metabolism , Primary Ovarian Insufficiency/physiopathology , Rats , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Reprod Biol Endocrinol ; 7: 87, 2009 Aug 24.
Article in English | MEDLINE | ID: mdl-19703295

ABSTRACT

BACKGROUND: It has been well established that prolactin (PRL) signals through the long form of its receptor (PRL-RL) and activates the Jak/Stat pathway for transcription of PRL target genes. However, signaling pathways mediated through the short PRL-R isoform (PRL-RS) remains controversial. Our recent finding that PRL signaling through PRL-RS represses two transcription factors critical for follicular development lead us to examine other putative PRL/PRL-RS target transcription factors in the decidua and ovary, two well-known target tissues of PRL action in reproduction. METHODS: In this investigation we used mice expressing PRL-RS on a PRL-R knockout background and a combo protein/DNA array to study the transcription factors regulated by PRL through PRL-RS only. RESULTS: We show that PRL activation of the PRL-RS receptor either stimulates or inhibits the DNA binding activity of a substantial number of transcription factors in the decidua as well as ovary. We found few transcription factors to be similarly regulated in both tissues, while most transcription factors are oppositely regulated by PRL in the decidua and ovary. In addition, some transcription factors are regulated by PRL only in the ovary or only in the decidua. Several of these transcription factors are involved in physiological pathways known to be regulated by PRL while others are novel. CONCLUSION: Our results clearly indicate that PRL does signal through PRL-RS in the decidua as well as the ovary, independently of PRL-RL, and activates/represses transcription factors in a tissue specific manner. This is the first report showing PRL/PRL-RS regulation of specific transcription factors. Many of these transcription factors were not previously known to be PRL targets, suggesting novel physiological roles for this hormone.


Subject(s)
DNA/metabolism , Decidua/drug effects , Ovary/drug effects , Prolactin/pharmacology , Receptors, Prolactin/metabolism , Transcription Factors/metabolism , Animals , CDX2 Transcription Factor , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , DNA/genetics , Decidua/metabolism , Electrophoretic Mobility Shift Assay , Eye Proteins/genetics , Eye Proteins/metabolism , Female , Gene Expression/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Mice, Transgenic , Ovary/metabolism , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Pregnancy , Prolactin/administration & dosage , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Prolactin/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Time Factors , Transcription Factors/genetics
5.
Endocrinology ; 150(7): 3327-35, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19342455

ABSTRACT

Prolactin (PRL) affects the development and function of the reproductive system by binding to two types of receptors, which differ by the size of their intracellular domain in rodents. Whereas the signaling pathway through the long form of the receptor (PRL-RL) is well characterized, signaling through the short form (PRL-RS) remains obscure. In this investigation, we examined transcription factors regulated by PRL in the ovary and decidua of mice expressing only PRL-RS in a PRL receptor null background. These mice provide a powerful in vivo model to study the selective signaling mechanism of PRL through PRL-RS independent of PRL-RL. We also examined the regulation of transcription factors in ovarian and uterine cell lines stably transfected with PRL-RS or PRL-RL. We focused our investigation on transcription factors similarly regulated in both these tissues and clearly established that signaling through PRL-RS does not activate the JaK/Stat in vivo but leads to severe down-regulation of Sp1 expression, DNA binding activity, and nuclear localization, events that appear to involve the calmodulin-dependent protein kinase pathway. Our in vivo and in culture data demonstrate that the PRL-RS activates a signaling pathway distinct from that of the PRL-RL.


Subject(s)
Prolactin/physiology , Receptors, Prolactin/physiology , STAT Transcription Factors/physiology , Signal Transduction/physiology , Sp1 Transcription Factor/physiology , Transcription Factors/metabolism , Animals , Cell Line , Decidua/drug effects , Decidua/physiology , Female , Janus Kinase 2/metabolism , Mice , Mice, Transgenic , Ovary/drug effects , Ovary/physiology , Rats , Receptors, Prolactin/genetics , Sp1 Transcription Factor/antagonists & inhibitors
6.
Mol Endocrinol ; 22(10): 2268-77, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18669642

ABSTRACT

Our laboratory has previously cloned and purified a protein named PRAP (prolactin receptor-associated protein) that was shown to be a novel 17beta-hydroxysteroid dehydrogenase (HSD) enzyme with dual activity. This enzyme, renamed HSD17B7 or PRAP/17beta-HSD7, converts estrone to estradiol and is also involved in cholesterol biosynthesis. The major site of its expression is the corpus luteum of a great number of species including rodents and humans. To examine the functional significance of HSD17B7 in pregnancy, we generated a knockout mouse model with targeted deletions of exons 1-4 of this gene. We anticipated a mouse with a severe fertility defect due to its inability to regulate estrogen levels during pregnancy. The heterozygous mutant mice are normal in their development and gross anatomy. The females cycle normally, and both male and female are fertile with normal litter size. To our surprise, the breeding of heterozygous mice yielded no viable HSD17B7 null mice. However, we found HSD17B7 null embryo alive in utero on d 8.5 and d 9.5. By d 10.5, the fetuses grow and suffer from severe brain malformation and heart defect. Because the brain depends on in situ cholesterol biosynthesis for its development beginning at d 10, the major cause of fetal death appears to be due to the cholesterol synthetic activity of this enzyme. By ablating HSD17B7 function, we have uncovered, in vivo, an important requirement for this enzyme during fetal development.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Embryo, Mammalian/physiology , Phosphoproteins/metabolism , 17-Hydroxysteroid Dehydrogenases/genetics , Animals , Embryo, Mammalian/anatomy & histology , Female , Gene Targeting , In Situ Hybridization , Male , Mice , Mice, Knockout , Phosphoproteins/genetics , Pregnancy , Tissue Distribution
7.
Mol Endocrinol ; 22(2): 513-22, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17975019

ABSTRACT

Prolactin (PRL) is a hormone with over 300 biological activities. Although the signaling pathway downstream of the long form of its receptor (RL) has been well characterized, little is known about PRL actions upon activation of the short form (RS). Here, we show that mice expressing only RS exhibit an ovarian phenotype of accelerated follicular recruitment followed by massive follicular death leading to premature ovarian failure. Consequently, RS-expressing ovaries of young adults are depleted of functional follicles and formed mostly by interstitium. We also show that activation of RS represses the expression of the transcription factor Forkhead box O3 (FOXO3) and that of the enzyme galactose-1-phosphate uridyltransferase (Galt), two proteins known to be essential for normal follicular development. Our finding that FOXO3 regulates the expression of Galt and enhances its transcriptional activity indicates that it is the repression of FOXO3 by PRL acting through RS that prevents Galt expression in the ovary and causes follicular death. Coexpression of RL with RS prevents PRL inhibition of Galt, and the ovarian defect is no longer seen in RS transgenic mice that coexpress RL, suggesting that RL prevents RS-induced ovarian impairment. In summary, we show that PRL signals through RS and causes, in the absence of RL, a severe ovarian pathology by repressing the expression of FOXO3 and that of its target gene Galt. We also provide evidence of a link between the premature ovarian failure seen in mice expressing RS and in mice with FOXO3 gene deletion as well as in human with Galt mutation.


Subject(s)
Forkhead Transcription Factors/metabolism , Ovary/metabolism , Prolactin/physiology , Receptors, Prolactin/physiology , Signal Transduction , Animals , Blotting, Western , Cell Line, Tumor , Electrophoretic Mobility Shift Assay , Female , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Genotype , Humans , Mice , Mice, Knockout , Mice, Transgenic , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Ovary/pathology , Prolactin/blood , Receptors, Prolactin/genetics , Receptors, Prolactin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , UTP-Hexose-1-Phosphate Uridylyltransferase/genetics , UTP-Hexose-1-Phosphate Uridylyltransferase/metabolism
8.
Endocrinology ; 146(6): 2807-16, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15731358

ABSTRACT

Prolactin receptor-associated protein (PRAP) originally cloned in our laboratory was shown to be a novel, luteal isoform of 17beta hydroxysteroid dehydrogenase 7 (17betaHSD7). In this study, we cloned the promoter region of rat PRAP/17betaHSD7 and investigated the mechanisms regulating both basal activity and LH-induced repression of this promoter. Truncated and site-specific mutants of PRAP/17betaHSD7 promoter identified two enhancer regions that contained highly conserved Sp1 binding site and bound Sp1 from nuclear extracts of both corpora lutea and a rat luteal cell line. Repression of PRAP/17betaHSD7 expression and promoter activity by human chorionic gonadotropin/forskolin was localized to a -52-bp proximal segment of the promoter. This region contained a conserved CCAAT site and bound nuclear factor Y; binding of this transcription factor was inhibited by human chorionic gonadotropin in vivo. Furthermore, mutation of the nuclear factor Y site in the -52-bp promoter-reporter construct abolished forskolin-mediated inhibition of the promoter in a rat luteal cell line. In summary, we have identified the promoter elements involved in the basal expression of PRAP/17betaHSD7. We have also found that LH-mediated repression of this gene is at the level of transcription and involves inhibition of nuclear factor YA binding to the CCAAT site within the proximal promoter.


Subject(s)
Phosphoproteins/genetics , Promoter Regions, Genetic/genetics , 5' Untranslated Regions/genetics , Animals , Base Sequence , Cells, Cultured , Cloning, Molecular , Corpus Luteum/cytology , Down-Regulation , Electrophoretic Mobility Shift Assay , Enhancer Elements, Genetic/genetics , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Luteinizing Hormone/metabolism , Luteinizing Hormone/pharmacology , Molecular Sequence Data , Phosphoproteins/physiology , Pregnancy , Rats , Rats, Sprague-Dawley , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...