Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Mol Cell Endocrinol ; 577: 112044, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37580010

ABSTRACT

Tissue kallikrein 1 (Klk1) is a serine protease that degrades several proteins including insulin-like growth factor binding protein-3 and extracellular matrix molecules. Klk1 mRNA expression in the mouse uterus was increased by estradiol-17ß (E2). The present study aimed to clarify the regulatory mechanism for Klk1 expression by estrogen. The promoter analysis of the 5'-flanking region of Klk1 showed that the minimal promoter of Klk1 existed in the -136/+24 region, and the estrogen-responsive region in the -433/-136 region. Tamoxifen increased Klk1 mRNA expression and the promoter activity, suggesting the involvement of AP-1 sites. Site-directed mutagenesis for the putative AP-1 sites in the -433/-136 region showed that the two putative AP-1 sites were involved in the regulation of Klk1 expression. Binding of estrogen receptor α (ERα) to the -433/-136 region was revealed by Chip assay. These results indicated that ERα bound the two putative AP-1 sites and transactivated Klk1 in the mouse uterus.

2.
Sci Rep ; 12(1): 17472, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36302800

ABSTRACT

Studies in genetically modified mice establish that essential roles of endogenous neuromedin U (NMU) are anorexigenic function and metabolic regulation, indicating that NMU is expected to be a potential target for anti-obesity agents. However, in central administration experiments in rats, inconsistent results have been obtained, and the essential role of NMU energy metabolism in rats remain unclear. This study aims to elucidate the role of endogenous NMU in rats. We generated NMU knockout (KO) rats that unexpectedly showed no difference in body weight, adiposity, circulating metabolic markers, body temperature, locomotor activity, and food consumption in both normal and high fat chow feeding. Furthermore, unlike reported in mice, expressions of Nmu and NMU receptor type 2 (Nmur2) mRNA were hardly detectable in the rat hypothalamic nuclei regulating feeding and energy metabolism, including the arcuate nucleus and paraventricular nucleus, while Nmu was expressed in pars tuberalis and Nmur2 was expressed in the ependymal cell layer of the third ventricle. These results indicate that the species-specific expression pattern of Nmu and Nmur2 may allow NMU to have distinct functions across species, and that endogenous NMU does not function as an anorexigenic hormone in rats.


Subject(s)
Neuropeptides , Peptide Hormones , Rats , Animals , Mice , Receptors, Neurotransmitter/genetics , Receptors, Neurotransmitter/metabolism , Neuropeptides/metabolism , Body Weight/physiology , Eating
3.
Gen Comp Endocrinol ; 306: 113752, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33711314

ABSTRACT

The adrenomedullin (AM) family is involved in diverse biological functions, including cardiovascular regulation and body fluid homeostasis, in multiple vertebrate lineages. The AM family consists of AM1, AM2, and AM5 in tetrapods, and the receptor for mammalian AMs has been identified as the complex of calcitonin receptor-like receptor (CLR) and receptor activity-modifying protein 2 (RAMP2) or RAMP3. However, the receptors for AM in amphibians have not been identified. In this study, we identified the cDNAs encoding calcrl (clr), ramp2, and ramp3 receptor components from the western clawed frog (Xenopus tropicalis). Messenger RNAs of amphibian clr and ramp2 were highly expressed in the heart, whereas that of ramp3 was highly expressed in the whole blood. In HEK293T cells expressing clr-ramp2, cAMP response element luciferase (CRE-Luc) reporter activity was activated by am1. In HEK293T cells expressing clr-ramp3, CRE-Luc reporter activity was increased by the treatment with am2 at the lowest dose, but with am5 and am1 at higher dose. Our results provided new insights into the roles of AM family peptides through CLR-RAMP receptor complexes in the tetrapods.


Subject(s)
Adrenomedullin , Peptide Hormones , Receptors, Calcitonin , Adrenomedullin/genetics , Animals , Calcitonin Receptor-Like Protein/genetics , HEK293 Cells , Humans , Receptor Activity-Modifying Protein 2/genetics , Receptor Activity-Modifying Protein 3/genetics , Receptors, Adrenomedullin/genetics , Receptors, Calcitonin/genetics , Xenopus
4.
Mol Cell Endocrinol ; 496: 110518, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31344393

ABSTRACT

Neuromedin U (NMU) shows circadian expression in the rat pars tuberalis (PT), and is known to be suppressed by melatonin. Here we examined the involvement of adenosine in the regulation of Nmu expression. We found that the rat PT expressed adenosine receptor A2b and that an adenosine receptor agonist, NECA, stimulated Nmu expression in brain slice cultures. In vitro promoter assays revealed that NECA stimulated Nmu promoter activity via a cAMP response element (CRE) in the presence of adenosine receptor A2b. NECA also increased the levels of phosphorylated CRE-binding protein. These findings suggest that adenosine stimulates Nmu expression by activating the cAMP signaling pathway through adenosine receptor A2b in the rat PT. This is the first report to demonstrate that Nmu expression in the PT is regulated by adenosine, which acts as an intravital central metabolic signal, in addition to melatonin, which acts as an external photoperiodic environmental signal.


Subject(s)
Adenosine/pharmacology , Gene Expression Regulation/drug effects , Neuropeptides/biosynthesis , Pituitary Gland/metabolism , RNA, Messenger/biosynthesis , Second Messenger Systems/drug effects , Animals , Cyclic AMP/metabolism , Male , Pituitary Gland/cytology , Rats , Rats, Inbred F344 , Receptor, Adenosine A2B/metabolism
5.
Cell Tissue Res ; 375(3): 743-754, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30377784

ABSTRACT

We previously demonstrated that female Runx3 knockout (Runx3-/-) mice were anovulatory and their uteri were atrophic and that Runx3 mRNA was expressed in granulosa cells. To clarify how Runx3 regulates folliculogenesis and ovulation, we examine the effects of Runx3 knockout on the gene expression of growth factors associated with folliculogenesis and enzymes associated with steroidogenesis. In Runx3-/- mouse ovaries, the numbers of primary and antral follicles were lower than those in wild-type (wt) mice at 3 weeks of age, indicating that the loss of Runx3 affects folliculogenesis. The expression of genes encoding activin and inhibin subunits (Inha, Inhba and Inhbb) was also decreased in ovaries from the Runx3-/- mice compared with that in wt mice. Moreover, the expression of the genes Cyp11a1 and Cyp19a1 encoding steroidogenic enzymes was also decreased. In cultured granulosa cells from 3-week-old mouse ovaries, Cyp19a1 mRNA levels were lower in Runx3-/- mice than those in wt mice. Follicle-stimulating hormone (FSH) treatment increased Cyp19a1 mRNA levels in both wt and Runx3-/- granulosa cells in culture but the mRNA level in Runx3-/- granulosa cells was lower than that in wt ones, indicating that granulosa cells could not fully function in the absence of Runx3. At 3 weeks of age, gonadotropin α subunit, FSHß subunit and luteinizing hormone (LH) ß subunit mRNA levels were decreased in Runx3-/- mice. These findings suggest that Runx3 plays a key role in female reproduction by regulating folliculogenesis and steroidogenesis in granulosa cells.


Subject(s)
Core Binding Factor Alpha 3 Subunit/metabolism , Granulosa Cells/metabolism , Organogenesis , Steroids/biosynthesis , Animals , Core Binding Factor Alpha 3 Subunit/deficiency , Estradiol/biosynthesis , Female , Follicle Stimulating Hormone/pharmacology , Gene Expression Regulation, Developmental/drug effects , Granulosa Cells/drug effects , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice, Inbred BALB C , Organogenesis/drug effects , Progesterone/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Gonadotropin/genetics , Receptors, Gonadotropin/metabolism
6.
Gen Comp Endocrinol ; 272: 109-116, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30594591

ABSTRACT

Chicken early (EF) and late feathering (LF) are sex-linked phenotypes conferred by wild-type k+ and dominant K alleles on chromosome Z, respectively. Besides prolactin (PRL) receptor (PRLR) and sperm flagellar 2 (SPEF2) genes, the K allele contains a fusion gene in which partially duplicated PRLR (dPRLR) and SPEF2 (dSPEF2) genes are linked in a tail-to-tail manner. The causative dPRLR gene encodes a C-terminal truncated receptor. LF chickens have short or no primaries at hatching; however, their feather growth rate is higher than that of EF chickens. This study aimed to elucidate the molecular basis of the K allele's biphasic effect on feather development. By 3'RACE and RT-PCR analyses, we demonstrated that dSPEF2 gene transcription occurred beyond all coding exons of the dPRLR gene on the opposite strand and that dPRLR mRNA was less abundant than PRLR mRNA. In addition, a 5'UTR splice variant (SPV) of PRL receptor mRNAs was increased in LF chickens. In vitro expression analysis of 5'UTR linked to the luciferase reporter gene revealed higher translation efficiency of SPV. RT-qPCR showed that the dPRLR mRNA level was higher in embryos; conversely, SPV was higher in hatched chickens, as was dSPEF2 mRNA. These findings suggest that the K allele inhibits feather development at the fetal stage by expressing dPRLR to attenuate PRLR function and promotes feather growth after hatching by increasing PRLR through dSPEF2 mRNA expression. Increased SPV may cause greater feather growth than that in EF chickens by increasing the availability of PRLR homodimers and enhancing PRL signaling.


Subject(s)
Chickens/metabolism , Feathers/metabolism , Receptors, Prolactin/metabolism , Animals , Female
7.
J Reprod Dev ; 62(5): 479-486, 2016 Oct 18.
Article in English | MEDLINE | ID: mdl-27301496

ABSTRACT

We previously demonstrated that the Runx3 transcription factor is expressed in the hypothalami, pituitaries, and ovaries of mice, and that Runx3 knockout (Runx3-/-) mice are anovulatory and their uteri are atrophic. Runx3 mRNA expression was detected in the granulosa cells of ovarian follicles, and in the anteroventral periventricular nucleus (AVPV) and arcuate nucleus (ARC). In the present study, we examined the effects of Runx3 knockout on the gene expression of enzymes associated with steroidogenesis. We found decreased Cyp11a1 mRNA expression in Runx3-/- mouse ovaries compared with that in wild-type (wt) mouse ovaries at the age of 8 weeks. In situ hybridization analysis showed that the percentages of Cyp11a1 mRNA-expressing theca cells in follicles of Runx3-/- mice were decreased compared with those of wt mice. In accord with the alterations in Runx3-/- mouse ovaries, Kiss1 mRNA levels in ARC were increased, whereas mRNA levels of kisspeptin in AVPV were decreased, and gonadotropin-releasing hormone in the preoptic area and follicle-stimulating hormone ß subunit gene were increased in Runx3-/- mice. Following an ovarian transplantation experiment between Runx3-/- mice and wt mice, corpora lutea were observed when ovaries from Runx3-/- mice were transplanted into wt mice, but not when those from wt mice were transplanted into Runx3-/- mice, suggesting that Runx3 in the hypothalamo-pituitary system may drive gonadotropin release to induce ovulation in the ovary. These findings indicate that Runx3 plays a crucial role in the hypothalamo-pituitary-gonadal axis.


Subject(s)
Core Binding Factor Alpha 3 Subunit/physiology , Ovary/physiology , Ovulation/physiology , Animals , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Core Binding Factor Alpha 3 Subunit/genetics , Female , Gonadotropins/metabolism , Granulosa Cells/cytology , Hypothalamo-Hypophyseal System , Hypothalamus/metabolism , Hypothalamus, Anterior/physiology , In Situ Hybridization , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Ovarian Follicle/physiology , RNA, Messenger/metabolism , Steroids/chemistry , Transcription Factors/metabolism
8.
Gene ; 562(1): 62-9, 2015 May 10.
Article in English | MEDLINE | ID: mdl-25701401

ABSTRACT

Melanocortin receptor 3 (MC3R) is expressed in the hypothalamus and pituitary in humans and rodents, and is involved in the control of feeding, energy metabolism, and pituitary function. In the mouse pituitary, MC3R is detected in mammotrophs. This study aimed to clarify the regulatory mechanism for Mc3r expression in the mouse pituitary. The promoter activities of reporter constructs for the MC3R gene 5'-flanking region up to -4000 bp (transcription initiation site designated as +1) were analyzed. The promoter activity significantly increased in the -86/+109 construct, but decreased in the -38/+109 construct, indicating that the minimal promoter required for basal expression of Mc3r is located in the -86/+109 region. Putative binding sites for transcription factors AP-1 and ATF4 were found in the 5'-flanking region of Mc3r. Site-directed mutation or deletion of these sites affected the promoter activities. In gel-shift assays with a nuclear extract of mouse anterior pituitary cells, band-shifts were detected for both sites after the addition of the nuclear extract, and were decreased in the presence of excess unlabeled probe competitors. These results indicated that both sites were involved in the regulation of Mc3r expression in anterior pituitary cells. Estradiol-17ß treatment increased the Mc3r promoter activity, indicating that the gene is regulated by estradiol-17ß. In conclusion, we have demonstrated the minimum promoter region required for Mc3r expression, and identified two binding sites for AP-1 and ATF4 and in the 5' upstream-flanking region of Mc3r that are essential for Mc3r expression.


Subject(s)
Activating Transcription Factor 4/genetics , Pituitary Gland, Anterior/metabolism , Promoter Regions, Genetic , Receptor, Melanocortin, Type 3/genetics , Transcription Factor AP-1/genetics , 5' Flanking Region , Activating Transcription Factor 4/metabolism , Animals , Binding Sites , Estradiol/pharmacology , Gene Expression Regulation , Genes, Reporter , HEK293 Cells , Humans , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Inbred ICR , Pituitary Gland, Anterior/drug effects , Protein Binding , Receptor, Melanocortin, Type 3/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism
9.
Gene ; 542(1): 23-8, 2014 May 25.
Article in English | MEDLINE | ID: mdl-24631266

ABSTRACT

Feathers are elaborate skin appendages shared by birds and theropod dinosaurs that have hierarchical branching of the rachis, barbs, and barbules. Feather filaments consist of ß-keratins encoded by multiple genes, most of which are located in tandem arrays on chromosomes 2, 25, and 27 in chicken. The expansion of the genes is thought to have contributed to feather evolution; however, it is unclear how the individual genes are involved in feather formation. The aim of the present study was to identify feather keratin genes involved in the formation of barbules. Using a combination of microarray analysis, reverse-transcription polymerase chain reaction, and in situ hybridization, we found an uncharacterized keratin gene on chromosome 7 that was expressed specifically in barbule cells in regenerating chicken feathers. We have named the gene barbule specific keratin 1 (BlSK1). The BlSK1 gene structure was similar to the gene structure of previously characterized feather keratin genes, and consisted of a non-coding leader exon, an intron, and an exon with an open reading frame (ORF). The ORF was predicted to encode a 98 aa long protein, which shared 59% identity with feather keratin B. Orthologs of BlSK1 were found in the genomes of other avian species, including turkey, duck, zebra finch, and flycatcher, in regions that shared synteny with chromosome 7 of chicken. Interestingly, BlSK1 was expressed in feather follicles that generated pennaceous barbules but not in follicles that generated plumulaceous barbules. These results suggested that the composition of feather keratins probably varies depending on the structure of the feather filaments and, that individual feather keratin genes may be involved in building different portions and/or types of feathers in chicken.


Subject(s)
Chickens/genetics , Feathers/cytology , Feathers/metabolism , Gene Expression Regulation , beta-Keratins/genetics , Animals , Base Sequence , Biological Evolution , Estradiol/pharmacology , Female , Male , Oligonucleotide Array Sequence Analysis , Open Reading Frames/genetics , RNA, Messenger/biosynthesis , Sequence Alignment
10.
J Reprod Dev ; 60(3): 216-23, 2014.
Article in English | MEDLINE | ID: mdl-24670778

ABSTRACT

Insulin-like growth factor 1 (IGF-1) is involved in regulations of reproductive functions in rats and mice. IGF-1 expression is regulated by estrogen in several reproductive organs including the uterus and ovary. Two types of estrogen receptor (ERα and ERß) are expressed in mouse uteri and ovaries, and it is unclear whether they differently mediate IGF-1 gene transcription. To clarify the roles of ERα and ERß, mouse endometrial stromal cells and ovarian granulosa cells were treated with ligands specific for individual estrogen receptors. In endometrial stromal cells, propyl-pyrazole-triol (PPT; ERα-selective agonist) increased Igf1 mRNA expression, which was suppressed by methyl-piperidino-pyrazole (MPP, ERα-selective antagonist), while diarylpropionitrile (DPN, ERß-potency selective agonist) increased Igf1 mRNA expression, which was inhibited by MPP but not by 4-[2-phenyl-5,7-bis(trifluoromethyl)pyrazolo[1,5-α]pyrimidin-3-yl]phenol (PHTPP; ERß antagonist). PHTPP enhanced the DPN-induced increase in Igf1 mRNA expression. In ovarian granulosa cells, E2 and DPN decreased Igf1 mRNA expression, whereas PPT did not affect Igf1 mRNA levels. In these cells, PHTPP inhibited the DPN-induced decrease in Igf1 mRNA expression. These results suggest that ERα facilitates Igf1 transcription, whereas ERß appears to inhibit Igf1 gene transcription in mouse endometrial stromal cells and ovarian granulosa cells.


Subject(s)
Endometrium/metabolism , Estrogen Receptor alpha/physiology , Estrogen Receptor beta/physiology , Granulosa Cells/metabolism , Insulin-Like Growth Factor I/genetics , Stromal Cells/metabolism , Animals , Cells, Cultured , Endometrium/drug effects , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor beta/agonists , Estrogen Receptor beta/antagonists & inhibitors , Estrogens/pharmacology , Female , Gene Expression Regulation/drug effects , Granulosa Cells/drug effects , Humans , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Inbred ICR , Ovary/cytology , Ovary/drug effects , Ovary/metabolism , Stromal Cells/drug effects
11.
J Reprod Dev ; 59(4): 398-404, 2013.
Article in English | MEDLINE | ID: mdl-23719562

ABSTRACT

Reproductive functions decline with the onset of diabetes in female mice. Diabetic mice have smaller uteri with an underdeveloped endometrium, suggesting diminished estrogen-induced growth. We aimed to clarify the changes in the estrous cycle and in insulin-like growth factor 1 (IGF1) expression in the uteri of streptozotocin (STZ)-treated diabetic mice, because IGF1 is one of the main growth factors involved in estrogen-induced uterine growth. ICR female mice were intraperitoneally administered STZ (10 mg/100 g BW), and blood glucose levels were determined. Mice with blood glucose levels > 200 mg/dl were classified as diabetic mice. The onset of diabetes was associated with acyclic estrous cycles. Diabetes was also induced with STZ in ovariectomized mice. Uterine Igf1 mRNA levels were reduced in ovariectomized STZ-treated diabetic mice. Estrogen is known to stimulate Igf1 mRNA expression in the uterus, but estrogen action was abolished in the uteri of STZ-treated diabetic mice. mRNA expressions of estrogen receptor α (ERα) and steroid hormone receptor coactivators (SRC-1/Ncoa1, SRC-2/Ncoa2, SRC-3/Ncoa3 and CBP/p300/Crebbp) were reduced in the uteri of ovariectomized STZ-treated diabetic mice. The present study demonstrates that diabetes induces a decline in female reproductive functions in mice. Igf1 expression in ovariectomized diabetic female mice was decreased, and decreased responsiveness to estrogen in the uteri of diabetic mice is probably associated with a reduction in ERα and steroid receptor coactivator mRNA expression.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Estrous Cycle/metabolism , Gene Expression Regulation/physiology , Uterus/metabolism , Animals , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/genetics , Female , Insulin-Like Growth Factor I/biosynthesis , Mice , Mice, Inbred ICR , Nuclear Receptor Coactivators/biosynthesis , Nuclear Receptor Coactivators/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Streptozocin/administration & dosage
12.
J Reprod Dev ; 58(5): 592-8, 2012.
Article in English | MEDLINE | ID: mdl-22785323

ABSTRACT

Runx3 is a transcription factor that belongs to the Runx family. We studied the localization of Runx3 mRNA in the mouse uterus, and its function in the mouse endometrium using Runx3 knockout (Runx3(-/-)) mice. Runx3 mRNA was detected in the endometrial luminal epithelial cells, glandular epithelial cells and stromal cells below the epithelial cell layer on the luminal side. The uteri of Runx3(-/-) mice were smaller than those of wt mice. The endometrial layer and uterine glands of Runx3(-/-) mice were less developed than those of wild-type mice, and the endometrial stromal layer was thinner. Transforming growth factor ß1 and ß3 (TGFß1 and ß3) mRNA levels in endometrial stromal cells of Runx3(-/-) mice were low compared with those of wild-type mice. Estradiol-17ß (E2) increased Tgfb2 mRNA levels in endometrial stromal cells of Runx3(-/-) mice, but not in those of wild-type mice. E2 increased epidermal growth factor (EGF) mRNA levels in endometrial stromal cells of wild-type mice, but did not increase those of Runx3(-/-) mice. The diminished Tgfb1 and Tgfb3 mRNA expressions may lead to the reduced proliferation of endometrial stromal cells. Alterations of E2-associated expressions of Tgfb2 and Egf mRNA in endometrial stromal cells of Runx3(-/-) mice may be associated with suppression of E2-dependent endometrial epithelial cell proliferation in Runx3(-/-) mice. Thus, Runx3 is likely to be a regulatory factor responsible for endometrial growth.


Subject(s)
Core Binding Factor Alpha 3 Subunit/metabolism , Endometrium/growth & development , Endometrium/metabolism , Gene Expression Regulation, Developmental , Animals , Cell Proliferation , Cells, Cultured , Core Binding Factor Alpha 3 Subunit/genetics , Crosses, Genetic , Endometrium/cytology , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Estradiol/metabolism , Female , Mice , Mice, Inbred BALB C , Mice, Knockout , Organ Size , Organogenesis , Ovary/cytology , Ovary/growth & development , Ovary/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta3/genetics , Transforming Growth Factor beta3/metabolism , Uterus/cytology , Uterus/growth & development , Uterus/metabolism
13.
Zoolog Sci ; 29(6): 377-83, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22639808

ABSTRACT

Transforming growth factor-α (TGFα) is thought to be involved in the regulation of endometrial cells. We investigated Tgfa mRNA expression, and the effects of TGFα on DNA-synthesis and gene expression of insulin-like growth factor 1 (IGF1), IGF binding protein-3 (IGFBP3) and IGF1 receptor in the mouse endometrial cells, because IGF1 is involved in estrogen-induced growth of endometrial cells. We also investigated the role of TGFα on matrix metalloproteinase (MMP) expression, as MMPs are involved both in tissue remodeling during cell proliferation and in enhancement of IGF1 signaling through the degradation of IGFBP3. Tgfa mRNA expression was detected in endometrial luminal and glandular epithelial cells, and stromal cells. Tgfa mRNA signals did not appear to change in endometrial luminal epithelial cells, but signals in glandular epithelial cells were higher at diestrus 1, 2 and proestrus, and the number of stromal cells showing strong signals appeared to increase at diestrus 1 and 2. Endometrial epithelial and stromal cells were treated with estradiol-17ß (E2) or progesterone (P4). E2 or P4 stimulated Tgfa mRNA expression in stromal cells. TGFα stimulated DNA synthesis in endometrial epithelial and stromal cells, while E2 and P4 stimulated DNA synthesis in stromal cells. In stromal cells, TGFα, at as low as 1 ng/ml, decreased Igfbp3 and Mmp9 mRNA levels, while high dose (10 ng/ml) of TGFα decreased Igf1 mRNA level and increased Mmp3 mRNA level. These results imply that TGFα stimulates proliferation of endometrial stromal cells through multiple mechanisms, including its regulation of Igfbp3 and Mmp3 transcription.


Subject(s)
Endometrium/cytology , Stromal Cells/metabolism , Transforming Growth Factor alpha/metabolism , Aging , Animals , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Estradiol/pharmacology , Estrous Cycle/physiology , Female , Gene Expression Regulation/physiology , Matrix Metalloproteinases, Secreted/genetics , Matrix Metalloproteinases, Secreted/metabolism , Mice , Progesterone/pharmacology , RNA/genetics , RNA/metabolism , Stromal Cells/drug effects , Transforming Growth Factor alpha/genetics
14.
Gen Comp Endocrinol ; 178(2): 180-4, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22634956

ABSTRACT

Pit-1 is a POU-domain transcription factor that promotes growth hormone (GH), prolactin (PRL), and thyroid-stimulating hormone ß subunit (TSHß) gene expression in the pituitary gland. Alternative splicing of Pit-1 gene transcripts has been shown to give rise to several variants with discrete transactivation properties. Recently, we identified a mouse Pit-1 w that is generated by alternative promoter usage and is expressed in a variety of tissues including the testis. Using a combination of reverse-transcription polymerase chain reaction analyses and luciferase reporter gene assays, we investigated the possible role of Pit-1 w in the mouse testis. In postnatal testicular development, the expression of Pit-1 w mRNA was significantly up-regulated between 18 and 20 days after birth when the numbers of secondary spermatocytes and spermatids have been reported to increase in mice. The PRL mRNA, but not the mRNAs for GH or TSHß, showed intratesticular expression patterns that were similar to those of the Pit-1 w mRNA. In experimental unilaterally cryptorchid testes of adult mice, spermatid numbers were extremely low and the expression levels of both the Pit-1 w and PRL mRNAs dropped dramatically. Furthermore, in the luciferase reporter gene assays, we found that Pit-1 w specifically transactivated the PRL promoter but had no effect on the promoters of GH or TSHß. These results suggested that Pit-1 w could be involved in the paracrine/autocrine system in mice and may be necessary for normal testicular function via its possible role in regulating PRL expression in testicular germ cells. This is the first report demonstrating the possible role of Pit-1 w in mammals.


Subject(s)
Prolactin/genetics , Testis/metabolism , Transcription Factor Pit-1/genetics , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Germ Cells/cytology , Germ Cells/metabolism , Humans , Male , Mice , Mice, Inbred ICR , Pituitary Gland/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Testis/cytology , Thyrotropin, beta Subunit/genetics
15.
Gen Comp Endocrinol ; 177(2): 231-7, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22554923

ABSTRACT

Brilliant plumage is typical of male birds, thus sexual plumage dichromatism is seen in many avian species; however, the molecular mechanism underlying this remains unclear. The agouti signaling protein (ASIP) is a paracrine factor that stimulates yellow/red pigment (pheomelanin) synthesis and inhibits black/brown pigment (eumelanin) synthesis in follicular melanocytes. In mammals, the distal promoter of the ASIP gene acts exclusively on the ventral side of the body to create a countershading pigmentation pattern by stimulating pheomelanin synthesis in the ventrum. Here, we examined the role of the distal ASIP promoter in controlling estrogen-dependent sexual dichromatism in chickens. Reverse-transcription polymerase chain reaction analyses revealed that ASIP class 1 mRNAs transcribed by the distal promoter were expressed exclusively on the ventral side of chicks and adult females displaying countershading. In showy adult males, the ASIP class 1 mRNAs were expressed in gold-colored ornamental feathers grown on the back. In the presence of estrogen, males molted into female-like plumage and ASIP class 1 mRNAs expression was altered to female patterns. These results suggest that the distal ASIP promoter produces countershading in chicks and adult females, similar to the ventral-specific ASIP promoter in mammals. In addition, the class 1 promoter plays an important role for creating sexual plumage dichromatism controlled by estrogen. This is the first evidence for a pigmentation gene having been modified in its expression during evolution to develop phenotypic diversity between individuals of different sexes.


Subject(s)
Agouti Signaling Protein/genetics , Chickens , Pigmentation/genetics , Promoter Regions, Genetic/genetics , Sex Differentiation/genetics , Agouti Signaling Protein/metabolism , Agouti Signaling Protein/physiology , Animals , Chickens/genetics , Chickens/growth & development , Chickens/metabolism , Chickens/physiology , Conserved Sequence/genetics , Estrogens/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Male , Models, Biological , Phenotype , Pigmentation/drug effects , Promoter Regions, Genetic/drug effects , Sex Differentiation/drug effects , Sexual Maturation/drug effects , Sexual Maturation/genetics , Sexual Maturation/physiology
16.
Gen Comp Endocrinol ; 175(3): 495-9, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22202606

ABSTRACT

Hair and feather pigmentation is mainly determined by the distribution of two kinds of melanin, eumelanin and pheomelanin, which produce brown to black and yellow to red colorations, respectively. The agouti signaling protein (ASIP) acts as an antagonist or an inverse agonist of the melanocortin 1 receptor (MC1R), a G protein-coupled receptor for α-melanocyte-stimulating hormone (α-MSH). This antagonism of the MC1R by ASIP on melanocytes initiates a switch of melanin synthesis from eumelanogenesis to pheomelanogenesis in mammals. In the present study, we isolated multiple ASIP mRNA variants generated by alternative splicing and promoters in chicken feather follicles. The mRNA variants showed a discrete tissue distribution. However, mRNAs were expressed predominantly in the feather pulp of follicles. Paralleling mRNA distribution, ASIP immunoreactivity was observed in feather pulp. Interestingly, ASIP was stained with pheomelanin but not eumelanin in pulp areas that face developing barbs. We suggest that the elaborate color pattern of individual feathers is formed in part by the antagonistic action of ASIP that is produced by multiple mRNA variants in chicken feather follicles.


Subject(s)
Agouti Signaling Protein/physiology , Chickens/physiology , Feathers/physiology , Pigmentation/physiology , Agouti Signaling Protein/genetics , Animals , Melanins/physiology , Paracrine Communication/physiology , RNA, Messenger/physiology
17.
Zoolog Sci ; 28(11): 790-801, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22035301

ABSTRACT

In our laboratory, a single autosomal recessive mutation in a phenotype similar to ruby-eye (ru/Hps6(ru)) or ruby-eye 2 (ru2/Hps5(ru2)) spontaneously occurred in siblings of C57BL/10JHir (+/+, black) mice in 2006. RT-PCR analysis revealed that this novel mutation, named ru2(d)/Hps5(ru2-d), exhibited frameshift by 997G deletion in the Hps5 gene. To clarify the mechanism of the hypopigmentation, the characteristics of proliferation and differentiation of ru2(d)/ru2(d) epidermal melanoblasts and melanocytes cultured in a serum-free medium were investigated. The proliferation of ru2(d)/ru2(d) melanoblasts and melanocytes did not differ from that of +/+ melanoblasts and melanocytes. However, the differentiation of ru2(d)/ru2(d) melanocytes was greatly inhibited. Tyrosinase (TYR) activity, expression of TYR, TYR-related protein 1 (TRP1) and TRP2 (dopachrome tautomerase, DCT), eumelanin synthesis, and the number of stage IV melanosomes markedly decreased in ru2(d)/ru2(d) melanocytes. However, excess L-tyrosine (Tyr) added to culture media from initiation of the primary culture rescued the reduced differentiation through increase in TYR activity, expression of TYR, TRP1, TRP2 and Kit, eumelanin synthesis, and stage IV melanosomes. L-Tyr injected into ru2(d)/ru2(d) mice also stimulated melanocyte differentiation. These results suggest that the ru2(d) allele inhibits melanocyte differentiation, and that its impaired differentiation is rescued by excess Tyr.


Subject(s)
Gene Deletion , Melanocytes/cytology , Melanocytes/metabolism , Proteins/metabolism , Tyrosine/pharmacology , Animals , Cell Differentiation , Cells, Cultured , Culture Media , Dihydroxyphenylalanine/metabolism , Gene Expression Regulation/physiology , Immunohistochemistry , Melanins/metabolism , Mice , Monophenol Monooxygenase/genetics , Monophenol Monooxygenase/metabolism , Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
18.
Gen Comp Endocrinol ; 173(2): 289-94, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21745476

ABSTRACT

Pit-1 is a pituitary-specific transcription factor responsible for pituitary development and hormone expression in mammals. Alternative splicing of Pit-1 gene transcripts has been shown to give rise to several variants with discrete transactivation properties; however, those arising from alternative promoters such as avian Pit-1 w have not yet been identified in mammals. Here, comparative genomics analysis followed by reverse transcription-polymerase chain reaction (RT-PCR) and rapid amplification of 5' cDNA ends (5'RACE) were used in identifying Pit-1 w mRNA in the mouse pituitary. The mouse Pit-1 w mRNA is generated by using an alternative promoter located in the first intron, as with chicken Pit-1 w, and is expressed in a wide variety of tissues besides the pituitary. In the testis, Pit-1 w is expressed as the predominant variant and a protein of 33 kDa. During the first wave of spermatogenesis, expression of Pit-1 w mRNA at substantial levels was observed from 3 weeks, but not at 1 or 2 weeks after birth. A combination of immunohistochemistry and in situ hybridization detected Pit-1 mRNA and Pit-1 immunoreactivity in the spermatogonia, spermatocytes, and spermatids in the testis of adult mice. Because secondary spermatocytes and haploid spermatids increase in number between 18 and 20 days after birth in mice, it is possible that mouse Pit-1 w plays a role in spermatogenesis. This is the first report demonstrating the expression of Pit-1 variants arising from alternative promoters in mammals.


Subject(s)
Spermatogenesis/physiology , Testis/metabolism , Transcription Factor Pit-1/metabolism , Alternative Splicing/genetics , Animals , Blotting, Western , Female , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Spermatids/metabolism , Spermatocytes/metabolism , Spermatogenesis/genetics , Transcription Factor Pit-1/genetics
19.
Gen Comp Endocrinol ; 173(2): 248-52, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21703269

ABSTRACT

In the present study, we expressed chicken (ch) Pit-1α (chPit-1α) and chPit-1γin vitro to compare the roles of chPit-1s in the transcription of the chicken growth hormone (chGH) gene. Both green fluorescence protein (GFP)-fused chPit-1γ and GFP-fused chPit-1α were localized in the nuclei of COS-7 cells. In a luciferase reporter gene assay, both chPit-1α and chPit-1γ transactivated the chGH promoter, and chPit-1α showed a more potent effect than chPit-1γ. On the other hand, an increase of cellular cAMP induced by forskolin promoted transactivation of the chGH gene with chPit-1α and chPit-1γ to similar extents. These results suggest that chPit-1γ may modulate the basal promoter activity of the chGH gene to the same degree as chPit-1α; however, a structural difference observed at the N-terminus transactivation domains in chPit-1α and chPit-1γ could be associated with the efficiency of basal activation of the chGH promoter.


Subject(s)
Growth Hormone/genetics , Protein Isoforms/metabolism , Transcription Factor Pit-1/metabolism , Animals , COS Cells , Cell Nucleus/metabolism , Chickens , Chlorocebus aethiops , Green Fluorescent Proteins , Promoter Regions, Genetic/genetics , Protein Isoforms/genetics , Transcription Factor Pit-1/genetics
20.
Gen Comp Endocrinol ; 171(1): 46-51, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21187100

ABSTRACT

Feather coloration in chickens mainly depends on melanin produced by melanocytes located in the feather follicles. The melanocortin 1 receptor (MC1R) on follicular melanocytes regulates melanin synthesis; however, the source of the melanocortins that interact with the receptors remains unclear. In this study, we examine the potential expression of melanocortins and characterize the mRNAs for the precursor pro-opiomelanocortin (POMC) in chicken feather follicles. Reverse transcription-polymerase chain reaction (RT-PCR) revealed the expression of mRNAs for POMC, prohormone convertase 1 (PC1) and PC2, and western blotting detected adrenocorticotropic hormone (ACTH)-related products of POMC processing in feather follicles, suggesting that melanocortins are produced locally in the tissues of chickens. A combination of 5'RACE (rapid amplification of cDNA 5' end), 3'RACE and RT-PCR analyzes identified two classes of POMC mRNA, class a and class b, which encode the same full-length POMC protein but have different non-coding leader exons. Class a mRNAs were expressed specifically in feather follicles, whereas class b mRNAs were expressed in the pituitary, hypothalamus, and various peripheral tissues that we examined. Within the feather follicles, the class a mRNAs were distributed in epidermal layers from middle to distal locations, whereas the class b mRNAs were mainly expressed in pulp at proximal locations. Our findings suggest that feather pigmentation is regulated by locally produced melanocortins, and indicate that the melanocortins encoded by the different classes of POMC mRNAs may play different intra-follicular roles in chickens. This is the first report that demonstrates alternative promoter usage generating different full-length POMC mRNAs in vertebrates.


Subject(s)
Epidermis/metabolism , Feathers/metabolism , Pro-Opiomelanocortin/metabolism , Promoter Regions, Genetic/genetics , Animals , Chickens , Immunoblotting , Immunohistochemistry , Pro-Opiomelanocortin/genetics , RNA, Messenger , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL