51
|
Mitsumori K. POSSIBLE MECHANISM ON ENHANCED CARCINOGENESIS OF GENOTOXIC CARCINOGENS AND UNSOLVED MECHANISMS ON LESSER CARCINOGENIC SUSCEPTIBILITY TO SOME CARCINOGENS IN RASH2 MICE. J Toxicol Sci 2003; 28:371-83. [PMID: 14746341 DOI: 10.2131/jts.28.371] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The rasH2 mice are hemizygous transgenic mice carrying the human prototype c-Ha-ras gene with its own promoter region, and have been used in 6-month short-term carcinogenicity tests for pharmaceutical drugs in accordance with the recommendation of the International Conference on Harmonization of Technical Requirements of Pharmaceuticals for Human Use (ICH). Based on the validation studies, it has been recognized that they are very susceptible to genotoxic carcinogens. To elucidate the mechanism of the enhanced carcinogenesis, spontaneous and chemically induced tumors in rasH2 mice have been subjected to molecular analyses, but the results have thus far been equivocal. This article focuses on the possible molecular mechanism of enhanced carcinogenesis in rasH2 mice, based on the results of a search in the literature. In addition, there are several reports suggesting lesser carcinogenic susceptibility of rasH2 mice to some carcinogens: Malignant lymphomas were induced by treatment with phenolphthalein in heterozygous p53 knockout mice, but not in rasH2 mice, and ethinylestradiol, uterine tumor promoter, resulted in depression of uterine proliferative lesions in rasH2 mice. In this review, the possible mechanisms of why rasH2 mice were less sensitive for these carcinogens are also discussed.
Collapse
Affiliation(s)
- Kunitoshi Mitsumori
- Laboratory of Veterinary Pathology, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509
| |
Collapse
|
52
|
Abstract
The decisive events in the development of decidual cells (DC) are presented through examples of human and rodent decidua. Human decidua is formed by large decidual cells (LDC), endometrial granulated cells (eGC), and small decidual cells. The LDC form the main type of decidual membranes, which determine the morphological characteristics of the decidua as a tissue. Immediate precursor cells of LDC are located below the basement membrane of the uterine epithelium before and during implantation. At the next stage of differentiation, LDC acquire a spindle-like shape. Rodent LDC form an epithelium-like structure with gland properties at the terminal stage of differentiation. The single-cell structure of human decidua is a derivative of the epithelial organization of rodent decidua. Spindle-like rat LDC are characterized by a high level of protein, RNA, and DNA synthesis and by intensive proliferation. At the beginning of pregnancy, a cell proliferation predominates over cell loss. By Days 12-13 of rat pregnancy LDC loss reaches 80% per day. Terminally differentiated LDC (tLDC) disappear from decidua due to apoptosis. Apoptosis of tLDC and the exhaustion of their precursors account for the disappearance of LDC in the middle of rat pregnancy. Human term decidua is composed of living cells. Human LDC (hLDC) comprise the largest part of human decidual cells (hLDC). hLDC account for 60-90% of hDC but their relative amount can decrease to 35% in the case of significant cell loss under unfavorable conditions. A decrease of LDC is not accompanied by DC proliferation. The lack of ability of decidua to compensate for DC loss suggests DC is a growing type of cell population without cambial cells. LDC function largely by blebbing and budding. Human and rat endometrial granulated cells (eGC) are characterized by a low level of natural killer (NK) activity and a high level of natural suppressor (NS) activity. The combination of NK and NS properties is characteristic of the eGC immunoregulatory function. Other functions of decidua include control of inflammation and trophoblast growth and expansion in the uterus. The life span of decidual cells is limited by pregnancy.
Collapse
Affiliation(s)
- V M Mikhailov
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg 194064, Russia
| |
Collapse
|
53
|
Jakacka M, Ito M, Martinson F, Ishikawa T, Lee EJ, Jameson JL. An estrogen receptor (ER)alpha deoxyribonucleic acid-binding domain knock-in mutation provides evidence for nonclassical ER pathway signaling in vivo. Mol Endocrinol 2002; 16:2188-201. [PMID: 12351685 DOI: 10.1210/me.2001-0174] [Citation(s) in RCA: 151] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We created a nonclassical estrogen receptor (ER) knock-in mouse model by introducing a mutation that selectively eliminates classical ER signaling through estrogen response elements, while preserving the nonclassical ER pathway. Heterozygous nonclassical ER knock-in (NERKI) females are infertile. Their ovaries contain no corpora lutea, reflecting a defect in ovulation, and the stromal cells contain lipid droplets, suggesting altered steroidogenesis. The uteri are enlarged with evidence of cystic endometrial hyperplasia, and the mammary glands are hypoplastic. These phenotypic features indicate differential ER effects on growth and development in various estrogen-responsive tissues. These findings suggest that nonclassical ER signaling pathways play an important physiological role in the development and function of the reproductive system.
Collapse
Affiliation(s)
- Monika Jakacka
- Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Medical School, Chicago, Illinois 60611, USA
| | | | | | | | | | | |
Collapse
|
54
|
Cytokine and growth factor network in human endometrium. Immunol Allergy Clin North Am 2002. [DOI: 10.1016/s0889-8561(02)00018-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
55
|
Lim H, Song H, Paria BC, Reese J, Das SK, Dey SK. Molecules in blastocyst implantation: uterine and embryonic perspectives. VITAMINS AND HORMONES 2002; 64:43-76. [PMID: 11898397 DOI: 10.1016/s0083-6729(02)64002-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Synchronized development of the embryo to the active stage of the blastocyst, differentiation of the uterus to the receptive state, and a "cross talk" between the blastocyst and uterine luminal epithelium are essential to the process of implantation. In spite of considerable accumulation of information and the present state of the knowledge, our understanding of the definitive mechanisms that regulate these events remains elusive. Although there are species variations in the process of implantation, many basic similarities do exist among various species. This review focuses on specific aspects of the implantation process in mice with the hope that many of the findings will be relevant to the process in humans. To establish signaling mechanisms of embryo-uterine interactions during implantation, studies on both embryonic and uterine consequences are required to generate more meaningful information. Due to ethical restriction and experimental limitation, it is difficult to generate such information in humans. This review has attempted to provide a comprehensive, but not complete, narration of a number of embryonic and uterine factors that are involved in the process of implantation in autocrine, paracrine, and/or juxtacrine manners in mice at the physiological, cellular, molecular, and genetic levels.
Collapse
Affiliation(s)
- Hyunjung Lim
- Department of Molecular and Integrative Physiology, Ralph L. Smith Research Center, University of Kansas Medical Center, Kansas City, Kansas 66160-7338, USA
| | | | | | | | | | | |
Collapse
|
56
|
Baran N, Kelly PA, Binart N. Characterization of a prolactin-regulated gene in reproductive tissues using the prolactin receptor knockout mouse model. Biol Reprod 2002; 66:1210-8. [PMID: 11906943 DOI: 10.1095/biolreprod66.4.1210] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Prolactin (PRL) exerts pleiotropic physiological effects in various cells and tissues, although it is mainly considered as a regulator of reproduction and cell growth. Null mutation of the prolactin receptor (PRLR) gene leads to female sterility due to a failure of embryo implantation. Using this mouse model and the method of mRNA differential display, we identified PRL target genes that are regulated during the peri-implantation period. We characterized 1 among the 45 isolated genes, UA-3, which is regulated in the uterus as well as in the ovary during early pregnancy. This gene corresponds to a P311 mouse cDNA that was originally identified for its high expression in late-stage embryonic brain and adult cerebellum. We report here that UA-3 is present in numerous tissues as well as in ovary and uterus at the site of blastocyst apposition, and that its expression is hormonally regulated. Moreover, in situ hybridization reveals high expression in ovarian granulosa cells and in uterine epithelium. Recently, it has been suggested that P311 expression is tightly regulated at several levels by mechanisms that control cellular growth, transformation, motility, or a combination of these. Taken together, these results suggest that P311 could be involved in these processes during pregnancy, although its function remains to be clearly established.
Collapse
Affiliation(s)
- Nathalie Baran
- INSERM Unité 344, Endocrinologie Moléculaire, Faculté de Médecine Necker Enfants Malades, 156 rue de Vaugirard, 75730 Paris Cedex 15, France
| | | | | |
Collapse
|
57
|
Bigsby RM. Control of growth and differentiation of the endometrium: the role of tissue interactions. Ann N Y Acad Sci 2002; 955:110-7; discussion 118, 396-406. [PMID: 11949941 DOI: 10.1111/j.1749-6632.2002.tb02771.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Early work with neonatal mice showed that estrogen receptor-negative uterine epithelium responded to estrogen treatment. Since the underlying mesenchymal cells were estrogen receptor-positive, it was suggested that these cells mediated the hormonal response through elaboration of a paracrine factor. Cell culture work showed that mesenchymal cells produced soluble factors that stimulate uterine epithelium, but hormonal regulation was absent or minimal. The paracrine hypothesis of estrogen action has been proved by the use of tissue recombinant studies in which epithelium from estrogen receptor-alpha knockout mice was combined with wild-type mesenchyme; estrogen stimulated the ER alpha-negative epithelium if the underlying stromal cells were receptor-positive. Also, it is hypothesized that there is a reciprocal paracrine interaction during stimulation with progesterone and estrogen. Accordingly, under progesterone dominance, the epithelium elaborates factors that direct the underlying stroma to proliferate when estrogen is administered. Although this hypothesis needs further testing, it has been shown that the uterine epithelium is required for stromal responsiveness to hormones. The question arises: What are the factors that mediate the effects of the steroid hormones in the uterus? Several peptide growth factors are regulated by estrogen and/or progesterone. Use of knockout animals will allow a determination of the role that these factors play in the uterus. However, ablation of many of these growth factor genes has proved lethal to the newborn animals, making it impossible to study hormonal effects using standard techniques. Tissue xenograft and tissue recombination studies offer a means of defining the role of specific growth factors in uterine physiology.
Collapse
Affiliation(s)
- Robert M Bigsby
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis 46202-5121, USA.
| |
Collapse
|
58
|
Afonso S, Tovar C, Romagnano L, Babiarz B. Control and expression of cystatin C by mouse decidual cultures. Mol Reprod Dev 2002; 61:155-63. [PMID: 11803549 DOI: 10.1002/mrd.1142] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
During mouse embryo implantation, trophoblast invasion is controlled in part by a balance of trophoblast-derived proteinases and uterine decidual proteinase inhibitors. Our work has focused on cystatin C, the secreted inhibitor of cathepsins B and L. We have previously shown that cystatin C is synthesized by the uterine decidua and localized to the cells in close contact with the trophoblast during implantation in vivo. In the work reported here we have established that decidualizing cultures show a similar upregulation of cystatin C. Using Northern and Western blotting and immunolocalization techniques both cystatin C mRNA and secreted protein increased with the morphological differentiation of stromal or decidual capsule cultures. In an effort to understand the regulation of cystatin C expression, decidual cells were analyzed under various culture conditions. Cystatin C expression was upregulated by increased cell density and by the presence of serum in the media. The growth factors TGF-beta(1) and EGF were found to induce cystatin C to levels comparable to serum stimulation. Co-culture with ectoplacental cones (EPCs) likewise induced expression and resulted in the localization of cystatin at the decidua:trophoblast interface. This work shows that decidualizing cultures are a good system to study cystatin C expression and that the expression is controlled in part by TGF-beta(1) and EGF signaling.
Collapse
Affiliation(s)
- Suzanne Afonso
- Department of Cell, Developmental and Neurobiology, Nelson Labs, Busch Campus, Rutgers University, Piscataway, New Jersey 08855, USA
| | | | | | | |
Collapse
|
59
|
Gao Z, Matsuo H, Wang Y, Nakago S, Maruo T. Up-regulation by IGF-I of proliferating cell nuclear antigen and Bcl-2 protein expression in human uterine leiomyoma cells. J Clin Endocrinol Metab 2001; 86:5593-9. [PMID: 11701740 DOI: 10.1210/jcem.86.11.8008] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IGF-I has been reported to play a role in regulating proliferation of human leiomyoma cells. There is, however, little evidence to suggest that IGF-I inhibits apoptosis in the leiomyoma cells. The present study was conducted to elucidate whether IGF-I affects apoptosis and Bcl-2 protein expression, an apoptosis-inhibiting gene product, in cultured leiomyoma cells. In addition, we examined the effect of IGF-I on proliferating cell nuclear antigen (PCNA) expression in cultured leiomyoma cells. Isolated human leiomyoma cells were subcultured in phenol red-free DMEM supplemented with 10% FBS for 120 h and then stepped down to serum-free conditions for an additional 72 h in the absence or presence of graded concentrations of IGF-I (1.0, 10, and 100 ng/ml). The effects of IGF-I on Bcl-2 protein and PCNA expression in cultured leiomyoma cells were assessed by Western immunoblot analysis and immunocytochemical staining, whereas the effects of IGF-I on the cell viability and apoptosis of the cultured cells were determined by 3-(4,5-dimethylatriazol-2-yl)-2,5diphenyltetrasodium bromide (MTT) assay and terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine 5'-triphosphate nick end labeling assay, respectively. Immunocytochemical staining demonstrated that IGF-I treatment resulted in the increase in PCNA labeling index in cultured leiomyoma cells in a dose-dependent manner. Immunoblot analysis of proteins extracted from the cultured leiomyoma cells revealed that the addition of IGF-I (10 and 100 ng/ml) significantly increased the expression of 35-kDa immunoreactive PCNA and 26-kDa Bcl-2 protein, compared with those in control cultures. Cell survival and proliferation of cultured leiomyoma cells, assessed by MTT assay, was significantly augmented by IGF-I treatment, compared with those of control cultures. Terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine 5'-triphosphate nick end labeling assay showed that the apoptosis-positive rate of leiomyoma cells treated with IGF-I was significantly decreased, compared with that in control cultures. The present results suggest that IGF-I plays crucial roles in leiomyoma cell growth, not only in promoting the proliferative potential by up-regulation of PCNA expression but also in down-regulating apoptosis by up-regulation of Bcl-2 protein expression in leiomyoma cells.
Collapse
Affiliation(s)
- Z Gao
- Department of Obstetrics and Gynecology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | | | | | | | | |
Collapse
|
60
|
Ma W, Tan J, Matsumoto H, Robert B, Abrahamson DR, Das SK, Dey SK. Adult tissue angiogenesis: evidence for negative regulation by estrogen in the uterus. Mol Endocrinol 2001; 15:1983-92. [PMID: 11682628 DOI: 10.1210/mend.15.11.0734] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Increased uterine vascular permeability and angiogenesis are two major events of embryo implantation and placentation during pregnancy. These latter processes require coordinated, uterine-specific interactions between progesterone (P4) and estrogen (E) signaling. Although roles of these steroids have long been suspected, definitive functions of E and/or P4 in uterine angiogenesis still remain elusive. We have therefore exploited the availability of reporter and mutant mice to explore the regulation of angiogenesis in response to steroid hormonal changes in vivo. We present here molecular, genetic, physiological, and pharmacological evidence that E and P4 have different effects in vivo: E promotes uterine vascular permeability but profoundly inhibits angiogenesis, whereas P4 stimulates angiogenesis with little effect on vascular permeability. These effects of E and P4 are mediated by differential spatiotemporal expression of proangiogenic factors in the uterus.
Collapse
MESH Headings
- Animals
- Endothelial Growth Factors/genetics
- Endothelial Growth Factors/metabolism
- Estradiol/analogs & derivatives
- Estradiol/metabolism
- Estradiol/pharmacology
- Estrogen Antagonists/pharmacology
- Estrogen Receptor alpha
- Female
- Fulvestrant
- Hormone Antagonists/pharmacology
- Lymphokines/drug effects
- Lymphokines/genetics
- Lymphokines/metabolism
- Male
- Mice
- Mice, Mutant Strains
- Mifepristone/pharmacology
- Neovascularization, Physiologic/drug effects
- Neovascularization, Physiologic/physiology
- Progesterone/metabolism
- Progesterone/pharmacology
- Receptor Protein-Tyrosine Kinases/drug effects
- Receptor Protein-Tyrosine Kinases/genetics
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Receptors, Growth Factor/drug effects
- Receptors, Growth Factor/genetics
- Receptors, Growth Factor/metabolism
- Receptors, Prolactin/genetics
- Receptors, Prolactin/metabolism
- Receptors, Vascular Endothelial Growth Factor
- Uterus/blood supply
- Uterus/drug effects
- Uterus/physiology
- Vascular Endothelial Growth Factor A
- Vascular Endothelial Growth Factors
Collapse
Affiliation(s)
- W Ma
- Department of Molecular and Integrative Physiology, Ralph L. Smith Research Center, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | | | | | | | |
Collapse
|
61
|
Klonisch T, Wolf P, Hombach-Klonisch S, Vogt S, Kuechenhoff A, Tetens F, Fischer B. Epidermal growth factor-like ligands and erbB genes in the peri-implantation rabbit uterus and blastocyst. Biol Reprod 2001; 64:1835-44. [PMID: 11369616 DOI: 10.1095/biolreprod64.6.1835] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Molecular cloning of the partial cDNA coding sequences of the four erbB receptors and the epidermal growth factor (EGF)-like ligands EGF, transforming growth factor alpha (TGF), and heparin-binding EGF (HB-EGF) has provided the basis for a comprehensive analysis of the spatiotemporal expression pattern of the EGF receptor/ligand system during the peri-implantation period in the rabbit. Employing nonradioactive in situ hybridization and immunolocalization, we observed differential expression of erbB1-erbB3 within the trophectoderm of the blastocyst. ErbB1 was strongly expressed in the cytotrophoblast but was downregulated upon syncytium formation. ErbB3 was a product of both the cyto- and syncytiotrophoblast. Despite the expression of erbB2 mRNA, the trophectoderm was devoid of immunoreactive ErbB2. ErbB4 gene activity was exclusively detected in the trophoblast at midpregnancy. The luminal and glandular epithelium and stroma of the nonpregnant, pseudopregnant, and pregnant rabbit uterus at Day 6 of gestation also expressed ErbB1-ErbB3. In the peri-implantation period, gene activities of erbB1-erbB3 were upregulated upon decidualization. At the site of implantation, uterine luminal epithelial cells apposing the preimplantation blastocyst displayed a distinct membrane immunolocalization of ErbB2, identifying the uterine epithelium as target for EGF, TGFalpha, and HB-EGF derived from both the embryonic trophectoderm and the uterine epithelium. In the luminal epithelium at the antimesometrial uterine site, HB-EGF gene activity was upregulated at the time of blastocyst attachment, but this upregulation was not reflected in an increase in immunoreactive HB-EGF. The detection of tyrosine phosphorylated ErbB2 in the rabbit placenta indicated the presence of a functional ErbB/EGF-like system in the pregnant rabbit uterus. This study provides strong evidence for a role of the ErbB/EGF-like system in embryo/maternal interactions during the peri-implantation period in the rabbit.
Collapse
Affiliation(s)
- T Klonisch
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University, D-06097 Halle/Saale, Germany.
| | | | | | | | | | | | | |
Collapse
|
62
|
Kim JG, Vallet JL, Christenson RK. Characterization of uterine epidermal growth factor during early pregnancy in pigs. Domest Anim Endocrinol 2001; 20:253-65. [PMID: 11518619 DOI: 10.1016/s0739-7240(01)00097-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Genomic research has identified a quantitative trait locus for uterine capacity, a component trait contributing to litter size, on porcine chromosome 8. The epidermal growth factor (EGF) gene, on porcine chromosome 8, may influence uterine capacity because of its growth-promoting activities. Using reverse transcription-polymerase chain reaction (PCR) and iterative screening of a porcine reproductive tissue cDNA library, 4932 bp cDNA sequence coding for porcine EGF precursor was obtained. The predicted protein sequence of the EGF precursor contained 1214 amino acids, similar to human EGF precursor (1207 amino acids, 81% identity). Curiously, the sequence of the mature peptide was less homologous between species than other regions of EGF precursor. The presence of conserved regions outside the mature peptide may suggest that these regions are functionally important. Expression of EGF mRNA in the endometrium of White crossbred gilts (n = 3 to 5 each) was determined by Northern blotting using 20 microg of total RNA from endometrium of D 10, 13, and 15 cyclic, and D 10, 13, 15, 20, 30, and 40 of pregnant gilts. A 3342 bp probe from EGF precursor was used. The bands corresponding to EGF mRNA were quantified by densitometry and results were analyzed by ANOVA. EGF mRNA expression decreased significantly from D 13 to 15 of the cycle and pregnancy (P = 0.04), and from D 30 to 40 of pregnancy (P = 0.01). These findings show that EGF mRNA expression is temporally regulated during the cycle and early pregnancy, and this pattern of gene expression may be important during early conceptus development.
Collapse
Affiliation(s)
- J G Kim
- USDA-ARS, Roman L. Hruska U.S. Meat Animal Research Center, P. O. Box 166, Clay Center, NE 68933-0166, USA
| | | | | |
Collapse
|
63
|
Sahlin L, Meikle A, Tasende C, Lindberg M, Masironi B, Eriksson H. Regulation of insulin-like growth factor-I and thioredoxin expression by estradiol in the reproductive tract of the prepubertal female lamb. J Steroid Biochem Mol Biol 2001; 77:123-8. [PMID: 11377977 DOI: 10.1016/s0960-0760(01)00042-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Estradiol (E(2)) has been shown to be an important uterine growth promoting molecule in the ovariectomized (ovx) rat, which increases the mRNA levels of insulin-like growth factor-I (IGF-I) and the redox enzyme thioredoxin. The aim of this study was to explore the role of E(2) in the regulation of IGF-I and thioredoxin in the reproductive tract of the prepubertal female lamb. Twenty 3-month-old lambs were treated with i.m. injections of E(2) at 24 h intervals. The animals were sacrificed 12 or 24 h after the last injection, and 72 h was the longest treatment period. The mRNA levels of thioredoxin and IGF-I were determined by a solution hybridization technique. There was a 5-fold increase in the cervical IGF-I mRNA level 12 h after the first E(2) injection. The uterine IGF-I mRNA level was doubled after 12 h and this increase was maintained during the rest of the experimental period. The IGF-I mRNA level in the oviducts was more than doubled 12 and 24 h after the E(2) injection, then the level decreased towards the initial level. The thioredoxin mRNA level in the cervix was increased 4-fold after 24 h, whereas no significant effect was seen in the uterus. The thioredoxin mRNA level in the oviduct was more than doubled 12 and 24 h after the first E(2) injection. Thus, estradiol regulates the expression of IGF-I and thioredoxin in the reproductive tract of prepubertal lambs.
Collapse
Affiliation(s)
- L Sahlin
- Department of Woman and Child Health, Division for Reproductive Endocrinology, Karolinska Hospital, L5:01, S-171 76 Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|
64
|
Kurachi O, Matsuo H, Samoto T, Maruo T. Tumor necrosis factor-alpha expression in human uterine leiomyoma and its down-regulation by progesterone. J Clin Endocrinol Metab 2001; 86:2275-80. [PMID: 11344239 DOI: 10.1210/jcem.86.5.7469] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although tumor necrosis factor-alpha (TNFalpha) has been shown mainly to inhibit proliferation and induce apoptosis in a variety of cells, no information is available regarding whether human leiomyoma cells express TNFalpha. In the present study, we examined the expression of TNFalpha in leiomyomas, in comparison with that in the adjacent normal myometrium, using immunohistochemical staining and Western immunoblot analysis with a polyclonal antibody to human TNFalpha. Furthermore, we investigated the effect of sex steroid hormones on TNFalpha expression in leiomyoma cells cultured under serum-free, phenol red-free conditions. Immunohistochemical staining showed that TNFalpha expression in leiomyoma cells was higher than that in the adjacent normal myometrial cells, being more abundant in the proliferative phase than in the secretory, progesterone (P4)-dominated, phase of the menstrual cycle. TNFalpha expression in leiomyoma cells in pregnant uterus was scarce. Western immunoblot analyses of leiomyoma and normal myometrial tissue extracts revealed that TNFalpha, with a molecular mass of 17.3 kDa, was abundantly present in leiomyoma tissue extracts, relative to normal myometrial tissue extracts, and that TNFalpha expression in leiomyoma cells was most abundant in the proliferative phase of the menstrual cycle, less abundant in the secretory phase, and least abundant in pregnant uterus; whereas no such changes in TNFalpha expression were noted in the normal myometrium. In monolayer cultures of uterine leiomyoma cells under serum-free conditions, addition of P4 (3.18 x 10(-7) mol/L) resulted in a decrease in TNFalpha expression in the cells, relative to that in control cultures, whereas treatment with 17beta-estradiol (3.67 x 10(-8) mol/L) did not affect the TNFalpha expression in the cells. The concentrations of sex steroids used were within the physiological tissue concentrations noted in leiomyoma and myometrium. The present results suggest that the abundant expression of TNFalpha may be a molecular basis characteristic of leiomyomas in the human uterus and that P4 may play a vital role in down-regulating the expression of TNFalpha in human uterine leiomyoma.
Collapse
Affiliation(s)
- O Kurachi
- Department of Obstetrics and Gynecology, Kobe University School of Medicine, Kobe 650-0017, Japan
| | | | | | | |
Collapse
|
65
|
Környei JL, Oszter A, Kovács KA, Vértes Z, Komlósi KM, Göcze PM, Vértes M. Anti-mitogenic action of opioid peptides on epidermal growth factor-stimulated uterine cells. Eur J Pharmacol 2001; 414:155-63. [PMID: 11239915 DOI: 10.1016/s0014-2999(01)00790-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Endogenous opioid peptides are negative regulators of estradiol-induced uterine cell proliferation. To investigate the possible molecular target site(s) of their anti-mitogenic action, we examined the effect of opioid peptides on epidermal growth factor-induced cell proliferation both in uterine primary cell cultures prepared from adult rats and in human myometrial smooth muscle cell lines. Epidermal growth factor (EGF) significantly increased cell density in both types of cultured monolayers. This EGF-induced stimulation of cell proliferation was blocked by [D-Met(2)-Pro(5)]enkephalinamide in a time-dependent, receptor-mediated manner. The effective concentrations were within the physiological nanomolar range. Enkephalinamide did not have any effect on the basal rate of proliferation of the uterine cells. Our results on this novel physiological cross-talk suggest that shared step(s) of the mechanism of action of estradiol and EGF might be targeted by opioid peptides and not the general machinery of cell proliferation.
Collapse
Affiliation(s)
- J L Környei
- Institute of Physiology, Pécs University, Medical School, Szigeti str. 12, Pécs H-7643, Hungary.
| | | | | | | | | | | | | |
Collapse
|
66
|
Kurita T, Wang YZ, Donjacour AA, Zhao C, Lydon JP, O'Malley BW, Isaacs JT, Dahiya R, Cunha GR. Paracrine regulation of apoptosis by steroid hormones in the male and female reproductive system. Cell Death Differ 2001; 8:192-200. [PMID: 11313721 DOI: 10.1038/sj.cdd.4400797] [Citation(s) in RCA: 149] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2000] [Revised: 08/30/2000] [Accepted: 10/11/2000] [Indexed: 11/09/2022] Open
Abstract
In males, androgens are essential in maintaining the integrity of the prostate. Androgen-ablation induces apoptosis of the prostatic epithelium. In females, ovariectomy induces apoptosis in uterine epithelium while progesterone inhibits this process. The objective of this study was to determine whether androgen and progesterone inhibit apoptosis, respectively, in mouse prostatic and uterine epithelia via steroid receptors in the epithelium or in the stroma. To address this question, prostatic tissue recombinants were prepared with rat urogenital sinus mesenchyme plus bladder epithelium from wild-type or testicular feminization mutant (Tfm) mice. Thus, prostatic tissue was generated having androgen receptor (AR) in both epithelium and stroma or in the stroma only. Castration of hosts induced apoptosis in the AR-negative Tfm prostatic epithelium with an epithelial apoptotic index virtually identical to prostatic tissue recombinants containing wild-type epithelium. Moreover, this castration-induced prostatic epithelial apoptosis was blocked by testosterone and dihydrotestosterone in both wild-type and Tfm prostatic tissue recombinants. Likewise, uterine tissue recombinants were prepared in which epithelium and/or stroma was devoid of progesterone receptor (PR) by using uterine epithelium and stroma of wild-type and PR knockout mice. Progesterone inhibited uterine epithelial apoptosis only in tissue recombinants prepared with PR-positive stroma. The PR status of the epithelium did not affect epithelial apoptotic index. Therefore, the apoptosis in prostatic and uterine epithelia is regulated by androgen and progesterone via stromal AR and PR, respectively. In both cases, epithelial AR or PR is not required for hormonal regulation of epithelial apoptosis in prostatic and uterine epithelium.
Collapse
Affiliation(s)
- T Kurita
- Department of Anatomy, University of California, San Francisco 94143, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Monroe DG, Jin DF, Sanders MM. Estrogen opposes the apoptotic effects of bone morphogenetic protein 7 on tissue remodeling. Mol Cell Biol 2000; 20:4626-34. [PMID: 10848589 PMCID: PMC85866 DOI: 10.1128/mcb.20.13.4626-4634.2000] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Interactions between estrogen and growth factor signaling pathways at the level of gene expression play important roles in the function of reproductive tissues. For example, estrogen regulates transforming growth factor beta (TGFbeta) in the uterus during the proliferative phase of the mammalian reproductive cycle. Bone morphogenetic protein 7 (BMP-7), a member of the TGFbeta superfamily, is also involved in the development and function of reproductive tissues. However, relatively few studies have addressed the expression of BMP-7 in reproductive tissues, and the role of BMP-7 remains unclear. As part of an ongoing effort to understand how estrogen represses gene expression and to study its interactions with other signaling pathways, chick BMP-7 (cBMP-7) was cloned. cBMP-7 mRNA levels are repressed threefold within 8 h following estrogen treatment in the chick oviduct, an extremely estrogen-responsive reproductive tissue. This regulation occurs at the transcriptional level. Estrogen has a protective role in many tissues, and withdrawal from estrogen often leads to tissue regression; however, the mechanisms mediating regression of the oviduct remain unknown. Terminal transferase-mediated end-labeling and DNA laddering assays demonstrated that regression of the oviduct during estrogen withdrawal involves apoptosis, which is a novel observation. cBMP-7 mRNA levels during estrogen withdrawal increase concurrently with the apoptotic index of the oviduct. Furthermore, addition of purified BMP-7 induces apoptosis in primary oviduct cells. This report demonstrates that the function of BMP-7 in the oviduct involves the induction of apoptosis and that estrogen plays an important role in opposing this function.
Collapse
Affiliation(s)
- D G Monroe
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
68
|
Sirbasku DA, Moreno-Cuevas JE. Estrogen mitogenic action. ii. negative regulation of the steroid hormone-responsive growth of cell lines derived from human and rodent target tissue tumors and conceptual implications. In Vitro Cell Dev Biol Anim 2000; 36:428-46. [PMID: 11039494 DOI: 10.1290/1071-2690(2000)036<0428:emainr>2.0.co;2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In an accompanying report (Moreno-Cuevas, J. E.; Sirbasku, D. A., In Vitro Cell. Dev. Biol.; 2000), we demonstrated 80-fold estrogen mitogenic effects with MTW9/PL2 rat mammary tumor cells in cultures supplemented with charcoal-dextran-treated serum. All sera tested contained an estrogen reversible inhibitor(s). The purpose of this report is to extend those observations to additional sex steroid-responsive human and rodent cell lines. Every line tested showed a biphasic response to hormone-depleted serum. Concentrations of < or = 10% (v/v) promoted substantive growth. At higher concentrations, serum was progressively inhibitory. With estrogen receptor-positive (ER+) human breast cancer cells, rat pituitary tumor cells, and Syrian hamster kidney tumor cells, 50% (v/v) serum caused significant inhibition, which was reversed by very low physiologic concentrations of estrogens. This same pattern was observed with the steroid hormone-responsive LNCaP human prostatic carcinoma cells. Because steroid hormone mitogenic effects are now easily demonstrable using our new methods, the identification of positive results has nullified our original endocrine estromedin hypothesis. We also evaluated autocrine/paracrine growth factor models of estrogen-responsive growth. We asked if insulin-like growth factors I and II, insulin, transforming growth factor alpha, or epidermal growth factor substituted for the positive effects of estrogens. Growth factors did not reverse the serum-caused inhibition. We asked also if transforming growth factor beta (TGFP) substituted for the serum-borne inhibitor. TGFbeta did not substitute. Altogether, our results are most consistent with the concept of a unique serum-borne inhibitor as has been proposed in the estrocolyone model. However, the aspect of the estrocolyone model related to steroid hormone mechanism of action requires more evaluation. The effects of sex steroids at picomolar concentrations may reflect mediation via inhibitor "activated" intracellular signaling pathways.
Collapse
Affiliation(s)
- D A Sirbasku
- The University of Texas-Houston Health Science Center, 77225-0036, USA.
| | | |
Collapse
|
69
|
Stewart CL, Cullinan EB. Preimplantation development of the mammalian embryo and its regulation by growth factors. DEVELOPMENTAL GENETICS 2000; 21:91-101. [PMID: 9291585 DOI: 10.1002/(sici)1520-6408(1997)21:1<91::aid-dvg11>3.0.co;2-d] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Preimplantation development in mammals involves both the development of the embryo and the preparation of the uterus in anticipation of blastocysts implantation. Preparation of the uterus for implantation is primarily under the control of the ovarian sex steroids, estrogen and progesterone. Increasing evidence is revealing that their effects on cell proliferation and differentiation in the uterus are medicated by locally produced growth factors and cytokines. In contrast, preimplantation development of the embryo to the blastocyst stage appears to be independent of exogenous growth factors. Implantation, the point at which the blastocyst forms a more intimate association with the maternal tissues, is regulated by the uterine expression of leukemia inhibitory factor (LIF). LIF is required both to promote embryo attachment and for decidualization of the uterus. In the absence of LIF, neither of these events occur. Uterine expression of LIF at the time of implantation has been described in many species, suggesting that LIF may be of general significance in regulating embryo implantation in mammals.
Collapse
Affiliation(s)
- C L Stewart
- Laboratory of Cancer and Developmental Biology, NCI-Frederick Cancer Research and Development Center, MD 21702, USA
| | | |
Collapse
|
70
|
Wood GW, Hausmann EH, Choudhuri R, Dileepan KN. Expression and regulation of histidine decarboxylase mRNA expression in the uterus during pregnancy in the mouse. Cytokine 2000; 12:622-9. [PMID: 10843737 DOI: 10.1006/cyto.2000.0667] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It has been hypothesized that hormonally regulated histamine production plays a role in preparation of the uterus for implantation. Histidine decarboxylase (HDC) is the rate-limiting enzyme for histamine production. The current study was designed to determine intrauterine expression of HDC mRNA expression during pregnancy in the mouse. High levels of HDC mRNA expression were observed in the preimplantation mouse uterus with peak expression occurring on day 4. High levels of HDC mRNA expression were also detected in the post-implantation uterus. In an effort to determine whether HDC mRNA is regulated by pro-inflammatory cytokines, the HDC mRNA pattern was compared to intrauterine expression of mRNA's for interleukin-1alpha (IL-1alpha), IL-1beta, macrophage chemotactic protein-1 (MCP-1) and RANTES (regulated on activation, normal T expressed and secreted) during the peri-implantation period. IL-1beta, MCP-1 and RANTES mRNA levels were increased in the uterus on days 1-2 and on days 4-5. Increased expression of IL-1alpha mRNA was observed on days 1-2 and days 5-7. There was no clear relationship between HDC mRNA expression and cytokine/chemokine mRNA expression. Progesterone-stimulated intrauterine expression of HDC mRNA. Intrauterine cytokine/chemokine mRNA was also hormonally regulated. This data allowed the possibility that one or more of these pro-inflammatory cytokines could be involved in regulating intrauterine HDC mRNA production. Recombinant IL-1alpha, IL-1beta, MCP-1 and RANTES all failed to induce HDC mRNA expression in the preimplantation uterus in a mouse pseudopregnancy model. At the same time, IL-1beta induced the expression of mRNA for each of the four cytokines/chemokines. Despite the fact that these were also produced in the uterus during pregnancy and were hormonally regulated, none of these cytokines induced intrauterine HDC mRNA expression. The data suggest that progesterone is involved in the regulation of HDC mRNA expression in the preimplantation uterus, but IL-1alpha/beta, MCP-1 and RANTES, which have been reported to regulate histamine synthesis during inflammatory processes, do not appear to play a role.
Collapse
Affiliation(s)
- G W Wood
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.
| | | | | | | |
Collapse
|
71
|
Uchide T, Masuda H, Lee YS, Makiyama Y, Mitsui Y, Saida K. Fluctuating gene expression and localized cellular distribution of vasoactive intestinal contractor (VIC) in mouse uterus. J Histochem Cytochem 2000; 48:699-707. [PMID: 10769054 DOI: 10.1177/002215540004800514] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
To understand the physiological roles of vasoactive intestinal contractor (VIC) and endothelin-2 (ET-2) in the uterus, we examined the expression levels of VIC mRNA by real-time quantitative reverse transcription-linked polymerase chain reaction (RT-PCR) and characterized the cellular distribution of VIC peptide and mRNA by immunostaining and in situ hybridization in mouse uterus. In pregnant mouse uterus, VIC mRNA expression changed considerably between Days 10.5 and 12.5 of pregnancy. The expression levels were significantly (p<0.05) higher (approximately fivefold) in the later stage of pregnancy (Days 12.5-17.5) than in the earlier stage (Days 7.5-10.5). In nonpregnant uterus, VIC mRNA expression was significantly (p <0.05) higher (approximately threefold) in proestrus and estrus than in diestrus. Immunohistochemical studies demonstrated the presence of VIC peptide in endometrial epithelial cells, myometrial cells, and vascular smooth muscle cells during the estrous cycle and pregnancy and after parturition. Notably, myometrial cells showed dominant immunostaining in proestrus and estrus, in the later pregnancy stage, and in the early postpartum period, analogous to the expression pattern of VIC mRNA. In situ hybridization confirmed localization of VIC mRNA in myometrial cells. These findings suggest that VIC may play an important role in the function of myometrial cells.
Collapse
Affiliation(s)
- T Uchide
- National Institute of Bioscience and Human-Technology, Agency of Industrial Science and Technology, Tsukuba, Ibaraki, Japan
| | | | | | | | | | | |
Collapse
|
72
|
Nie GY, Li Y, Hampton AL, Salamonsen LA, Clements JA, Findlay JK. Identification of monoclonal nonspecific suppressor factor beta (mNSFbeta) as one of the genes differentially expressed at implantation sites compared to interimplantation sites in the mouse uterus. Mol Reprod Dev 2000; 55:351-63. [PMID: 10694741 DOI: 10.1002/(sici)1098-2795(200004)55:4<351::aid-mrd1>3.0.co;2-l] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Successful implantation requires synchronous development of and active dialogue between the maternal endometrium and the implanting blastocyst. While it is well established that appropriate maternal steroid hormones are essential for endometrial preparation for implantation, the molecular events at the actual site of implantation are still little understood. The aims of our studies were to identify genes explicitly expressed or repressed at the sites of implantation by utilising RNA differential display (DDPCR), and to establish the roles of these genes in the implantation process in a mouse model. Ten bands unique in implantation sites compared to interimplantation sites were identified by DDPCR and subsequently confirmed by Northern blotting. One of these bands contained a cDNA fragment that was highly homologous to mouse monoclonal nonspecific suppressor factor beta (MNSFbeta) or Fau. The full cDNA sequence of this gene, obtained by screening a lambdagt11 cDNA library, was essentially the same as MNSFbeta, except that it had much longer 5' untranslated region. Interestingly, both Northern and immunohistochemical analysis showed that the expression of this gene was much lower in implantation sites compared to interimplantation sites on day 4.5 of pregnancy, when embryos first attach to the uterus and initiate implantation, and on day 5.5, when implantation has advanced. These results suggest a role for MNSF during implantation and early pregnancy, possibly through regulating the proliferation and/or differentiation of uterine stromal cells. It may also be involved in the selective production of TH2-type cytokines in implantation sites to regulate the immune system at the maternal-fetal interface.
Collapse
Affiliation(s)
- G Y Nie
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia.
| | | | | | | | | | | |
Collapse
|
73
|
Yee D, Lee AV. Crosstalk between the insulin-like growth factors and estrogens in breast cancer. J Mammary Gland Biol Neoplasia 2000; 5:107-15. [PMID: 10791773 DOI: 10.1023/a:1009575518338] [Citation(s) in RCA: 187] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Once it was recognized that breast tumor growth was stimulated by estrogens, successful therapeutic strategies based on depriving the tumor of this hormone were developed. Since the growth stimulatory properties of the estrogens are governed by the estrogen receptor (ER), understanding the mechanisms that activate ER are highly relevant. In addition to estrogens, peptide growth factors can also activate the ER. The insulin-like growth factors (IGFs) are potent mitogens for ER-positive breast cancer cell lines. This review will examine the evidence for interaction between these two pathways. The IGFs can activate the ER, while ER transcriptionally regulates genes required for IGF action. Moreover, blockade of ER function can inhibit IGF-mediated mitogenesis and interruption of IGF action can similarly inhibit estrogenic stimulation of breast cancer cells. Taken together, these observations suggest that the two growth regulatory pathways are tightly linked and that a further understanding of the mechanism of this crosstalk could lead to new therapeutic strategies in breast cancer.
Collapse
Affiliation(s)
- D Yee
- Department of Medicine, University of Minnesota Cancer Center, Minneapolis 55455, USA.
| | | |
Collapse
|
74
|
Nie GY, Li Y, Wang J, Minoura H, Findlay JK, Salamonsen LA. Complex regulation of calcium-binding protein D9k (calbindin-D(9k)) in the mouse uterus during early pregnancy and at the site of embryo implantation. Biol Reprod 2000; 62:27-36. [PMID: 10611064 DOI: 10.1095/biolreprod62.1.27] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Establishment of receptive endometrium is essential for implantation. Our aim was to identify and characterize genes uniquely regulated at the sites of implantation in mouse uterus by RNA differential display polymerase chain reaction (DDPCR). One of the gene fragments identified was 86% homologous to rat calcium-binding protein D9k (calbindin-D(9k)); the mouse counterpart had not then been cloned, but subsequently an mRNA sequence of mouse calbindin-D(9k) became available in GenBank (accession number: AF028071). This sequence is 99% homologous to the DDPCR-derived gene tag but has a shorter 3' end. Reverse transcription-polymerase chain reaction (RT-PCR) was performed using the sequence of 3' end of the DDPCR product and the 5' end of AF028071, and a full cDNA was obtained. This gene was primarily up-regulated by progesterone, but not by estrogen. It was further increased by the combination of the two steroids. Expression of calbindin-D(9k) was overall increased in the uterus during early pregnancy, but the level was significantly lower in implantation compared to interimplantation sites on Days 4.5 and 5.5 of pregnancy, becoming barely detectable in both sites after Day 6.5. In situ hybridization localized this mRNA predominantly in the luminal epithelium of the pregnant uterus. The complex regulation of calbindin-D(9k) in mouse uterus suggests an important role for this protein during pregnancy.
Collapse
Affiliation(s)
- G Y Nie
- Prince Henry's Institute of Medical Research, Clayton, Victoria 3168, Australia.
| | | | | | | | | | | |
Collapse
|
75
|
Abstract
Previous studies in our laboratory demonstrated the presence of sialomucin complex (SMC)/Muc4 covering the rat uterine luminal epithelium. SMC/Muc4 expression in the uterus is regulated by estrogen and progesterone and lost at the time of receptivity. In contrast to this hormonal regulation at the uterine luminal surface, SMC/Muc4 in the uterine glandular epithelium, oviduct, cervix, and vagina was constitutively expressed at all stages of the estrous cycle. Furthermore, SMC was expressed in the cervix and vagina of the ovariectomized rat, even though it is not found in the uterine luminal epithelium. Both soluble and membrane-bound forms of SMC were present in these tissues. Immunohistochemical analyses showed distinctive localization patterns of SMC in the various tissues during the estrous cycle. Moreover, the previously unreported expression of SMC/Muc4 in the isthmus, ampulla, and infundibulum of the oviduct suggests potential functions in gamete development. These results indicate that SMC/Muc4 is expressed in most tissues of the female reproductive tract, in which it may have multiple functions. However, hormonal regulation appears to be restricted to the uterine luminal epithelium.
Collapse
Affiliation(s)
- N Idris
- Department of Cell Biology and Anatomy, University of Miami School of Medicine, Miami, Florida 33101, USA
| | | |
Collapse
|
76
|
Ribeiro ML, Perez Martinez S, Farina M, Ogando D, Gimeno M, Franchi A. The effect of epidermal growth factor on prostaglandin synthesis of oestrogenized rat uterus is mediated by nitric oxide. Prostaglandins Leukot Essent Fatty Acids 1999; 61:353-8. [PMID: 10718107 DOI: 10.1054/plef.1999.0110] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We examined the possible relationship between cytokines, nitric oxide and prostaglandins (PGs) in the estrogenized rat uterus. Results indicate that epidermal growth factor (EGF) enhances the synthesis of prostaglandins in estrogenized rat uteri and induces the augmentation of nitric oxide (NO) production in this tissue by stimulating iNOS. While the effect of EGF is abolished by L-NMMA, an NO antagonist, the NS-398, a cyclooxygenase-II (COX-II) inhibitor, prevents the augmentation of prostanoids induced by EGF. These results suggest that there is an interaction among EGF, NO and PGs and that in this interrelationship are involved COX-II and iNOS. This mechanism might be important during implantation and labor.
Collapse
|
77
|
Chan CS, Harvey MB, Clements JA. Temporal and tissue-specific expression of kallikrein (Klk) genes and identification of a novel Klk messenger ribonucleic acid transcript during early development in the mouse. Biol Reprod 1999; 61:621-8. [PMID: 10456837 DOI: 10.1095/biolreprod61.3.621] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The kallikreins are a multigene family of serine proteases that act on a diverse number of substrates, including several growth factors and extracellular matrix (ECM) glycoproteins and proteinases. Recently, this family has been implicated in the process of early development and embryo implantation. In this study, we used reverse transcription-polymerase chain reaction with gene-specific primers and Southern hybridization to elucidate the temporal and tissue-specific expression patterns of the mouse kallikreins Klk1, Klk3, Klk5, Klk9, and Klk21 during early development in the embryo, uterus, and decidua. We observed the expression of Klk1 (tissue kallikrein), Klk3 (gamma-nerve growth factor), Klk9 (epidermal growth factor-binding protein), and Klk21 in the early conceptus (until the 2-cell stage). Only Klk21 continued to be expressed in the blastocyst until Day 7.5 of pregnancy. Expression of Klk9 reappeared at Day 7.5 and was consistently detected until Day 11, the last day studied; Klk1 was again expressed in the embryo from Day 9.5, with decreased levels by Day 11. In contrast, in the uterus or decidua, there was no expression of Klk1 until Day 7.5, when mRNA transcripts were abundant; transcripts then decreased in the Day 9.5 and Day 11 uterus. Expression of Klk21 in the uterus and decidua displayed a similar pattern but was detected at much lower levels. Interestingly, a novel Klk21-like mRNA was also detected in uterine tissue samples but not in embryonic samples; Klk3, Klk5, and Klk9 were not consistently expressed in the uterus or decidua over this time. This is the first report of the expression of specific kallikreins during early development. The distinct gene- and tissue-specific expression patterns presented in this study, in conjunction with the well-characterized roles of kallikreins in regulation of protein activation, ECM degradation, and proliferative events, suggests the involvement of the kallikrein gene family during early development.
Collapse
Affiliation(s)
- C S Chan
- Centre for Molecular Biotechnology, School of Life Sciences, Queensland University of Technology, Brisbane, Queensland 4001, Australia
| | | | | |
Collapse
|
78
|
Gerstenberg C, Allen WR, Stewart F. Factors controlling epidermal growth factor (EGF) gene expression in the endometrium of the mare. Mol Reprod Dev 1999; 53:255-65. [PMID: 10369386 DOI: 10.1002/(sici)1098-2795(199907)53:3<255::aid-mrd1>3.0.co;2-d] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Previous studies showed a dramatic increase in EGF gene expression in the endometrial glands of pregnant mares around day 40 after ovulation. To investigate how the steroid hormones of pregnancy might regulate this expression, in situ hybridization was used to monitor the levels of EGF mRNA in endometrial biopsies obtained from seasonally anoestrous or ovariectomised mares given exogenous progesterone and oestrogen, alone or in combination, for up to 46 days. Biopsies were also taken from mares during the non-pregnant cycle, during normal pregnancies and pregnancies compromised by endometrial pathology (endometriosis) or because of incompatible extraspecific embryo transfers (donkey-in-horse pregnancies). Only a few samples showed weak EGF expression during the late luteal phase of the oestrous cycle. During normal pregnancy, the previously observed dramatic increase of expression after day 40 of gestation was confirmed. Although aged mares suffering from endometriosis and mares carrying an extraspecific donkey conceptus showed the same increase of EGF mRNA in normal glands, this was virtually absent from gland cross-sections compromised due to inflammatory or fibrotic changes. Administration of various doses and combinations of progesterone and oestrogen for < 35 days yielded negative or only weakly positive hybridization results, whereas progesterone alone for > or = 40 days upregulated EGF expression strongly irrespective of additional treatment with oestrogen. This is the first experimental evidence that EGF expression in the endometrium can be induced by progesterone alone. The requirement for prolonged progesterone priming is of considerable interest in the context of the unusually late stage of gestation at which placental attachment commences in equids.
Collapse
Affiliation(s)
- C Gerstenberg
- University of Cambridge Department of Clinical Veterinary Medicine, Newmarket, Suffolk, United Kingdom
| | | | | |
Collapse
|
79
|
Abstract
All scientific investigations begin with distinct objectives: first is the hypothesis upon which studies are undertaken to disprove, and second is the overall aim of obtaining further information, from which future and more precise hypotheses may be drawn. Studies focusing on the generation and use of gene-targeted animal models also apply these goals and may be loosely categorized into sequential phases that become apparent as the use of the model progresses. Initial studies of knockout models often focus on the plausibility of the model based on prior knowledge and whether the generation of an animal lacking the particular gene will prove lethal or not. Upon the successful generation of a knockout, confirmatory studies are undertaken to corroborate previously established hypotheses of the function of the disrupted gene product. As these studies continue, observations of unpredicted phenotypes or, more likely, the lack of a phenotype that was expected based on models put forth from past investigations are noted. Often the surprising phenotype is due to the loss of a gene product that is downstream from the functions of the disrupted gene, whereas the lack of an expected phenotype may be due to compensatory roles filled by alternate mechanisms. As the descriptive studies of the knockout continue, use of the model is often shifted to the role as a unique research reagent, to be used in studies that 1) were not previously possible in a wild-type model; 2) aimed at finding related proteins or pathways whose existence or functions were previously masked; or 3) the subsequent effects of the gene disruption on related physiological and biochemical systems. The alpha ERKO mice continue to satisfy the confirmatory role of a knockout quite well. As summarized in Table 4, the phenotypes observed in the alpha ERKO due to estrogen insensitivity have definitively illustrated several roles that were previously believed to be dependent on functional ER alpha, including 1) the proliferative and differentiative actions critical to the function of the adult female reproductive tract and mammary gland; 2) as an obligatory component in growth factor signaling in the uterus and mammary gland; 3) as the principal steroid involved in negative regulation of gonadotropin gene transcription and LH levels in the hypothalamic-pituitary axis; 4) as a positive regulator of PR expression in several tissues; 5) in the positive regulation of PRL synthesis and secretion from the pituitary; 6) as a promotional factor in oncogene-induced mammary neoplasia; and 7) as a crucial component in the differentiation and activation of several behaviors in both the female and male. The list of unpredictable phenotypes in the alpha ERKO must begin with the observation that generation of an animal lacking a functional ER alpha gene was successful and produced animals of both sexes that exhibit a life span comparable to wild-type. The successful generation of beta ERKO mice suggests that this receptor is also not essential to survival and was most likely not a compensatory factor in the survival of the alpha ERKO. In support of this is our recent successful generation of double knockout, or alpha beta ERKO mice of both sexes. The precise defects in certain components of male reproduction, including the production of abnormal sperm and the loss of intromission and ejaculatory responses that were observed in the alpha ERKO, were quite surprising. In turn, certain estrogen pathways in the alpha ERKO female appear intact or unaffected, such as the ability of the uterus to successfully exhibit a progesterone-induced decidualization response, and the possible maintenance of an LH surge system in the hypothalamus. [ABSTRACT TRUNCATED]
Collapse
Affiliation(s)
- J F Couse
- Receptor Biology Section, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | |
Collapse
|
80
|
Chen D, Ganapathy P, Zhu LJ, Xu X, Li Q, Bagchi IC, Bagchi MK. Potential regulation of membrane trafficking by estrogen receptor alpha via induction of rab11 in uterine glands during implantation. Mol Endocrinol 1999; 13:993-1004. [PMID: 10379897 DOI: 10.1210/mend.13.6.0287] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The steroid hormone estrogen profoundly influences the early events in the uterus leading to embryo implantation. It is thought that estrogen triggers the expression of a unique set of genes in the preimplantation endometrium that in turn control implantation. To identify these estrogen-induced genes, we used a delayed implantation model system in which embryo attachment to endometrium is dependent on estrogen administration. Using a mRNA differential display (DD) method, we isolated a number of cDNAs representing mRNAs whose expression is either turned on or turned off in response to an implantation-inducing dose of estrogen. We identified one of these cDNAs as that encoding rab11, a p21ras-like GTP-binding protein (G protein), which functions in the targeting of transport vesicles to the plasma membrane. In normal pregnant rats, rab11 mRNA was expressed at low levels on days 1-2 of pregnancy, but its expression was markedly enhanced (approximately 6- to 8-fold) between days 3-5 immediately before implantation. In situ hybridization and immunocytochemistry revealed that rab11 expression in the uterus was predominantly in the glandular epithelium. In ovariectomized rats, the expression of rab11 mRNA was induced in the endometrium in response to estrogen. To determine whether this effect of estrogen was mediated through its nuclear receptors, we examined rab11 expression in a transformed endometrial cell line, Ishikawa. In transient transfection experiments, we observed that overexpression of estrogen receptor (ER) alpha or beta induced endogenous rab11 mRNA in a hormone-dependent manner. ER bound to an antagonist, ICI 182,780, failed to activate this gene expression. These findings, together with the observation that ER alpha but not ER beta is detected in the glands of the preimplantation uterus, indicate that rab11 is one of the proteins that are specifically induced by estrogen-complexed ER alpha in rat endometrium at the onset of implantation. Our results imply that estrogen, which induces the synthesis of many growth factors and their receptors and other secretory proteins that are thought to be critical for implantation, may also facilitate their transport to the membrane and/or secretion by stimulating the expression of rab11, a component of the membrane-trafficking pathway. This study therefore provides novel insights into the diverse cellular mechanisms by which estrogen, acting via its nuclear receptors, may influence blastocyst implantation.
Collapse
Affiliation(s)
- D Chen
- Population Council and The Rockefeller University, New York, New York 10021, USA
| | | | | | | | | | | | | |
Collapse
|
81
|
Mendoza-Rodríguez CA, Camacho-Arroyo I, García GA, Cerbón MA. Variations of progesterone receptor and c-fos gene expression in the rat uterus after treatment with norethisterone and its A-ring reduced metabolites. Contraception 1999; 59:339-43. [PMID: 10494488 DOI: 10.1016/s0010-7824(99)00039-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
It has been suggested that some contraceptive derivatives of 19-nor-testosterone possess estrogenic activity that may facilitate the development of breast cancer. The aim of this work was to investigate the estrogenic properties of norethisterone (NET) and its A-ring-reduced derivatives by determining progesterone receptor (PR) and c-fos mRNA content of two estrogen-regulated genes in the uterus of ovariectomized rats. mRNA content was evaluated by Northern blot 1-6 h after 17 beta-estradiol administration. The highest PR and c-fos mRNA content was observed 3 h and 2 h after 17 beta-estradiol administration, respectively. NET did not modify either PR or c-fos mRNA content. In contrast, 5 alpha- and 3 beta, 5 alpha-NET significantly increased mRNA content of both genes. The increase in c-fos mRNA content induced by these reduced compounds was lower than that found with estradiol treatment. The overall results indicate that NET administration can indirectly induce estrogenic effects through the action of its 5 alpha-dihydro and 3 beta, 5 alpha-tetrahydro derivatives.
Collapse
|
82
|
Lavigne MC, Ramwell PW, Clarke R. Growth and phenotypic characterization of porcine coronary artery smooth muscle cells. In Vitro Cell Dev Biol Anim 1999; 35:136-43. [PMID: 10476909 DOI: 10.1007/s11626-999-0015-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Vascular smooth muscle cell (VSMC) proliferation significantly contributes to atherosclerotic plaque formation and limits the success rate of percutaneous transluminal coronary angioplasty. We derived a population of porcine coronary artery SMCs to characterize VSMC proliferation and phenotype in preparation to study the molecular actions of VSMC mitogens and antiproliferative agents. Growth assays were designed to minimize the estrogen content in the culture medium, since this steroid hormone significantly influences VSMC growth and the expression of VSMC mitogens and their receptors. Culture conditions were identified such that this criterion was achieved while maintaining a significant VSMC growth rate. Cells cultured in serum-free medium, regardless of growth factor supplements, did not remain adherent to a plastic culture substrate, nor did they proliferate. Dextran-coated charcoal (DCC)-treated sera, including fetal bovine, calf, and porcine, supported VSMC adhesion, but not growth. Whole fetal bovine serum (FBS) produced the best proliferative response. A type-I collagen-coated culture surface significantly enhanced VSMC growth, but only in culture medium containing non-DCC-treated FBS. Flow cytometry analyses confirmed the mitogenic effects of this substrate. The VSMCs exhibited a morphological change on type-I collagen, but this was not accompanied by a change in VSMC phenotype. Our data indicate that culture of these porcine coronary artery SMCs in 2.5% FBS plus 10 ng platelet-derived growth factor-BB per ml in phenol red-free medium on type-I collagen may be the optimal conditions for studying the molecular aspects of VSMC mitogens and antiproliferative agents.
Collapse
Affiliation(s)
- M C Lavigne
- Department of Physiology and Biophysics, Georgetown University Medical Center, Washington, DC 20007, USA
| | | | | |
Collapse
|
83
|
Hirzel DJ, Wang J, Das SK, Dey SK, Mead RA. Changes in uterine expression of leukemia inhibitory factor during pregnancy in the Western spotted skunk. Biol Reprod 1999; 60:484-92. [PMID: 9916018 DOI: 10.1095/biolreprod60.2.484] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Mutation of the leukemia inhibitory factor (LIF) gene results in reproductive failure in LIF -/- mice due to an inability to implant their blastocysts. This condition is reversed by infusion of LIF or by transferral of embryos to pseudopregnant, wild-type mice. This led us to hypothesize that embryonic diapause in the spotted skunk is due to insufficient uterine expression of LIF whereas resumption of development and implantation are associated with increased LIF expression. We also investigated the hormonal control of LIF expression. Uterine concentrations of LIF mRNA were determined by quantitative reverse transcription-polymerase chain reaction. Changes in cell-specific localization of LIF mRNA and protein were determined by in situ hybridization and immunocytochemistry. LIF mRNA was detected but was not abundant during embryonic diapause; it then increased when blastocysts resumed development and remained elevated prior to implantation. LIF mRNA and protein could not be localized in the uterus during embryonic diapause but were quite apparent in luminal and glandular epithelium during blastocyst activation. Prolactin, progesterone, and estradiol failed to increase uterine concentrations of LIF mRNA above those in ovariectomized controls. These data are consistent with the initial hypothesis and suggest that LIF may somehow be involved in preparing the uterus for implantation in the spotted skunk.
Collapse
Affiliation(s)
- D J Hirzel
- Department of Biological Sciences, University of Idaho, Moscow, Idaho 83844, USA
| | | | | | | | | |
Collapse
|
84
|
Curtis SH, Korach KS. Steroid Receptor Knockout Models: Phenotypes and Responses Illustrate Interactions between Receptor Signaling Pathways in Vivo. ADVANCES IN PHARMACOLOGY 1999; 47:357-80. [PMID: 10582092 DOI: 10.1016/s1054-3589(08)60117-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
|
85
|
Sidhu SS, Kimber SJ. Hormonal control of H-type alpha(1-2)fucosyltransferase messenger ribonucleic acid in the mouse uterus. Biol Reprod 1999; 60:147-57. [PMID: 9858499 DOI: 10.1095/biolreprod60.1.147] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The H epitope, an alpha(1-2)fucosylated carbohydrate structure, has been implicated in initial attachment of the murine blastocyst to luminal uterine epithelial cells in vitro. In this study, the expression of the H-type alpha(1-2)fucosyltransferase (FUT1) gene was examined in endometrium of mice. Northern blotting of luminal epithelial RNA identified a single 6.2-kilobase transcript. In situ hybridization studies showed a signal for FUT1 mRNA on Days 1-3 of pregnancy in glands and luminal epithelium. The signal diminished by Day 4 and could not be detected on Day 5 of pregnancy. The in situ signal in endometrial epithelia was highest at estrus and metestrus and was absent at diestrus. Estrogen treatment after ovariectomy gave strong FUT1 mRNA expression in epithelia, but with progesterone, progesterone + estrogen, or vehicle, no message could be detected. A semiquantitative reverse transcription-polymerase chain reaction (PCR) analysis of FUT1 mRNA from luminal epithelium generated large amounts of PCR product on Day 1 of pregnancy; this diminished on Days 2, 3, and 4, and the product was barely detectable on Day 5. A kinetic analysis of FUT1 activity on Day 1 of pregnancy suggested a single enzyme with a Michaelis-Menten constant (Km) of 0.29 mM towards phenyl-beta-D-galactoside and of 1.75 mM towards Galbeta(1-3)GalNAc. These results suggest that expression of the H epitope is regulated at the level of FUT1 transcription and that transcription is stimulated by estrogen in the endometrial epithelium.
Collapse
Affiliation(s)
- S S Sidhu
- School of Biological Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | | |
Collapse
|
86
|
Lim H, Das SK, Dey SK. erbB genes in the mouse uterus: cell-specific signaling by epidermal growth factor (EGF) family of growth factors during implantation. Dev Biol 1998; 204:97-110. [PMID: 9851845 DOI: 10.1006/dbio.1998.9072] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
We previously described spatiotemporal expression of various epidermal growth factor (EGF)-like ligands and receptor subtypes, ErbB1 and ErbB2, during the peri-implantation period. To better understand the roles of these ligands and their possible signaling schemes in implantation, it is important to define the status of all the ligands and receptor subtypes in the uterus/embryo. No information is available about uterine and embryonic status of ErbB3 or ErbB4 during implantation. We cloned mouse erbB3 and erbB4 cDNAs and examined their expression and bioactivity in the peri-implantation uterus (days 1-8). Two erbB3 (cytoplasmic and extracellular) and three erbB4 (two cytoplasmic and one extracellular) clones were generated. Both forms of the erbB3 clone showed similar transcript profiles, while different transcript profiles were obtained with erbB4 clones. The steady-state levels of erbB3 and erbB4 mRNAs in whole uterine poly(A)+ RNA samples showed little changes during the peri-implantation period, while their unique cell-specific accumulation was noted. erbB3 is predominantly expressed in the epithelial cells, although decidual and embryonic cells also accumulate this mRNA. In contrast, the erbB4 mRNA is primarily expressed in the submyometrial stroma and myometrial connective tissues during this period. Additionally, the extracellular form of the erbB4 clone detected signals in a subpopulation of stromal cells. Autophosphorylation and immunoprecipitation studies provided evidence that uterine ErbB3 and ErbB4 are biologically active. This study provides a comprehensive analysis of possible ligand-receptor signaling schemes for EGF-like ligands in implantation.
Collapse
Affiliation(s)
- H Lim
- Department of Molecular and Integrative Physiology, Ralph L. Smith Research Center, University of Kansas Medical Center, Kansas City, Kansas, 66160-7338, USA
| | | | | |
Collapse
|
87
|
Flores JM, Sánchez MA, García P, Sánchez B, Nieto A. Immunohistochemical localization of epidermal growth factor, transforming growth factor-alpha and growth factor-beta s in the caprine peri-implantation period. Theriogenology 1998; 50:931-44. [PMID: 10734465 DOI: 10.1016/s0093-691x(98)00197-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Control over the action of steroid hormones in the uterus and conceptus during the initial period of gestation appears to be regulated locally by growth factors. This study involved immunohistochemical detection of epidermal growth factor (EGF), transforming growth factor-alpha (TGF-alpha) and transforming growth factor-beta s (TGF-beta s), to determine their role in the caprine peri-implantation period. Epidermal growth factor was expressed in the luminal and glandular endometrial epithelium of goats on all days studied (Days 22 to 30 post coitum), but it was not detected in trophoblastic cells or in other embryonic structures. Between Days 22 and 30 post coitum, TGF-alpha was detected in the epithelial cells and superficial stroma of the uterus and in the trophoendodermic cells of the embryo. Transforming growth factor-beta s expression, observed in the endometrium, embryo and extraembryonic membranes on Day 22 post coitum, decreased by Day 24 post coitum and disappeared in the embryo by Day 30 post coitum, while remaining in the other structures. The presence of these growth factors during the peri-implantation period in the goat suggests their participation in proliferation and differentiation phenomena which occur during implantation and embryonic development.
Collapse
Affiliation(s)
- J M Flores
- Department of Animal Pathology II, Veterinary School Complutense University, Madrid, Spain
| | | | | | | | | |
Collapse
|
88
|
Jaber L, Kan FW. Non-identical distribution pattern of epidermal growth factor and platelet-derived growth factor in the mouse uterus during the oestrous cycle and early pregnancy. THE HISTOCHEMICAL JOURNAL 1998; 30:711-22. [PMID: 9873998 DOI: 10.1023/a:1003441904274] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
In the present study, we examined by immunohistochemistry the cell-specific distribution of epidermal growth factor (EGF) and platelet-derived growth factor (PDGF) in the mouse uterus during the oestrous cycle and throughout the first 7 days of pregnancy. Paraffin-embedded tissue samples were immunostained using the avidin-biotin peroxidase technique and then examined by light microscopy. Our results showed that immunostaining for EGF was detected in the stroma but not in the luminal or glandular epithelium. A high concentration of EGF was detected in the stroma around the time of embryo implantation at days 3, 4 and 5 of pregnancy. The implanted embryo at day 7 of gestation showed immunostaining for EGF between the ectoderm and endoderm layers. The cell distribution pattern for PDGF was found to be different from that observed with EGF. Luminal and glandular epithelia displayed PDGF immunostaining throughout the first 7 days of pregnancy, with the highest intensity at days 4 and 5 of gestation. In contrast, no immunostaining was observed in the luminal and glandular epithelia at post-oestrus, dioestrus and pro-oestrus stages. However, a weak reaction started to appear at oestrus. The embryo at the blastocyst stage displayed a strong immunoreaction for antibody against PDGF. In addition, the decidual boundary zone surrounding the implanted embryo at days 5, 6 and 7 of gestation also showed an immunostaining for PDGF. The present observations demonstrate clearly the presence of EGF and PDGF in the mouse uterus in high concentrations at the peri-implantation period. Thus, our results, together with what is known about the effect of EGF and PDGF in controlling the growth, differentiation and activation of a variety of cell types, suggest a possible role for these growth factors during the preparation of the endometrium for implantation in controlling the proliferation activity of stromal and/or epithelial cells.
Collapse
Affiliation(s)
- L Jaber
- Department of Anatomy and Cell Biology, Faculty of Medicine, Queen's University, Kingston, Ontario, Canada
| | | |
Collapse
|
89
|
Nikolova Z, Djonov V, Zuercher G, Andres AC, Ziemiecki A. Cell-type specific and estrogen dependent expression of the receptor tyrosine kinase EphB4 and its ligand ephrin-B2 during mammary gland morphogenesis. J Cell Sci 1998; 111 ( Pt 18):2741-51. [PMID: 9718367 DOI: 10.1242/jcs.111.18.2741] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Morphogenesis of the mammary gland occurs mainly during adult life and is dependent on a complex interplay of hormonal, cell-cell and cell-matrix interactions. The molecular mechanisms involved in pattern formation of the mammary epithelium in adult life are poorly understood. Recently, several members of the Eph family of receptor tyrosine kinases and their ligands have been shown to participate in pattern formation during embryogenesis and conceivably may fulfill similar functions during adult morphogenesis. We have investigated the expression of a member of this family, EphB4, and its cognate ligand, ephrin-B2, during normal and malignant mouse mammary morphogenesis. A spatially, temporarily and hormonally coordinated expression of both the receptor and ligand was observed. The receptor was predominantly localized in the myoepithelial cells surrounding the ducts and alveoli whereas ligand expression was limited to the luminal epithelial cells. Expression of both was induced at the onset of gland morphogenesis at puberty and was differentially regulated during the estrus cycle. Ovariectomy of pre-pubertal or adult females abolished the expression of both receptor and ligand and administration of estrogen alone was sufficient to restore their normal expression. Disruption of the balanced expression was observed during experimental mouse mammary carcinogenesis. Ligand expression was lost at the onset of tumorigenesis and receptor expression shifted from myoepithelial to epithelial cells with progressive malignancy. These results implicate both the EphB4 receptor and its ligand ephrin-B2 in the hormone dependent morphogenesis of the mammary gland. Furthermore, their deregulated expression may contribute to mammary carcinogenesis.
Collapse
Affiliation(s)
- Z Nikolova
- Department of Clinical Research, University of Berne, Tiefenaustrasse 120, CH-3004 Berne, Switzerland
| | | | | | | | | |
Collapse
|
90
|
Onohara Y, Harada T, Tanikawa M, Iwabe T, Yoshioka H, Taniguchi F, Mitsunari M, Tsudo T, Terakawa N. Autocrine effects of transforming growth factor-alpha on the development of preimplantation mouse embryos. J Assist Reprod Genet 1998; 15:395-402. [PMID: 9673886 PMCID: PMC3455022 DOI: 10.1023/a:1022541218487] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
PURPOSE We wished to explore the role of transforming growth factor (TGF)-alpha in mouse embryonic development. METHODS We examined the gene expression of TGF-alpha and epidermal growth factor receptor (EGFR) in mouse blastocysts by the reverse transcription-polymerase chain reaction and evaluated the effects of TGF-alpha on the development of preimplantation mouse embryos using TGF-alpha antisense oligodeoxynucleotide. Mouse teratocarcinoma F9 cells were also a subject of this study. RESULTS Gene transcripts of TGF-alpha and EGFR were present in both blastocysts and F9 cells. TGF-alpha significantly stimulated the rate of blastocoel expansion in early-cavitating blastocysts and the proliferation of F9 cells. Northern blot analysis showed that TGF-alpha gene expression in F9 cells was markedly suppressed in the presence of TGF-alpha antisense oligodeoxynucleotide. TGF-alpha antisense oligonucleotide significantly reduced the rate of blastocoel expansion and the growth of F9 cells. The inhibitory effects of TGF-alpha antisense oligonucleotide on blastocysts and F9 cells were reversed by the addition of TGF-alpha. CONCLUSIONS The present observations suggest that TGF-alpha acts as an autocrine factor in the development of preimplantation mouse embryos.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal
- Blastocyst/chemistry
- Blastocyst/drug effects
- Blastocyst/physiology
- Blotting, Northern
- Chorionic Gonadotropin/pharmacology
- Chorionic Gonadotropin/therapeutic use
- Electrophoresis, Agar Gel
- ErbB Receptors/genetics
- ErbB Receptors/metabolism
- Female
- Gene Expression Regulation, Developmental
- Male
- Mice
- Oligonucleotides, Antisense/metabolism
- Oligonucleotides, Antisense/pharmacology
- Polymerase Chain Reaction
- Pregnancy
- RNA, Messenger/analysis
- RNA, Messenger/chemistry
- RNA, Messenger/metabolism
- Teratocarcinoma
- Transcription, Genetic
- Transforming Growth Factor alpha/genetics
- Transforming Growth Factor alpha/metabolism
- Transforming Growth Factor alpha/physiology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Y Onohara
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Shimomura Y, Matsuo H, Samoto T, Maruo T. Up-regulation by progesterone of proliferating cell nuclear antigen and epidermal growth factor expression in human uterine leiomyoma. J Clin Endocrinol Metab 1998; 83:2192-8. [PMID: 9626159 DOI: 10.1210/jcem.83.6.4879] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Uterine leiomyoma is the most common smooth muscle cell tumor of the myometrium. Estrogen and progesterone (P4) are believed to be physiological regulators of leiomyoma growth. We recently showed that Bcl-2 protein, an apoptosis-inhibiting gene product, was abundantly expressed in leiomyoma relative to its expression in the normal myometrium and that Bcl-2 protein expression in cultured leiomyoma cells was up-regulated by P4, but down-regulated by 17 beta-estradiol (E2). To further characterize the molecular mechanism of sex steroidal regulation of leiomyoma growth, we examined the effect of menstrual phase on proliferating cell nuclear antigen (PCNA) expression in leiomyoma and investigated whether sex steroids could influence PCNA expression in leiomyoma cells cultured under serum-free conditions by immunoblot and immunohistochemical analyses. As epidermal growth factor (EGF) has been shown to mediate estrogen action and to play a crucial role in regulating leiomyoma growth, we also investigated the effects of sex steroids on the expression of EGF and EGF receptor (EGF-R) in cultured leiomyoma cells. The PCNA labeling index in leiomyomas was much greater in the secretory, P4-dominated, phase than in the proliferative phase of the menstrual cycle and was significantly higher than that in the adjacent normal myometrium throughout the menstrual cycle. In monolayer cultures of leiomyoma cells, the addition of either E2 (10 ng/mL) or P4 (100 ng/mL) resulted in an increase in PCNA expression in the cells compared to that in control cultures, whereas in monolayer cultures of myometrial cells, the addition of E2 augmented PCNA expression in the cells, but P4 did not. Immunoblot analysis of proteins extracted from cultured leiomyoma cells revealed that leiomyoma cells contained immunoreactive EGF with a molecular mass of 133 kDa and that the addition of P4 resulted in a remarkable increase in the expression of 133- and 71-kDa immunoreactive EGF in the cells compared to that in control cultures, whereas the addition of E2 resulted in a somewhat lower expression of immunoreactive EGF in the cells. Furthermore, immunocytochemical analysis with a monoclonal antibody to human EGF-R demonstrated that the treatment with E2 augmented EGF-R expression in the cells compared to that in untreated cells, but P4 did not. The concentrations of sex steroids used were within the physiological tissue concentrations found in leiomyomas and myometria. These results indicate that P4 up-regulates the expression of PCNA and immunoreactive EGF in leiomyoma cells, whereas E2 up-regulates the expression of PCNA and EGF-R in those cells. As it is evident that EGF plays a crucial role as a local factor in regulating leiomyoma growth, the P4-induced increase in PCNA expression in leiomyoma cells may be mediated by P4-induced enhanced expression of EGF-like proteins in the cells, whereas the E2-induced increase in PCNA expression in leiomyoma cells may be mediated by E2-induced enhanced expression of EGF-R in those cells. It is, therefore, conceivable that P4 and E2 act in combination to stimulate the proliferative potential of leiomyoma cells through the induction of EGF-like proteins and EGF-R expression in uterine leiomyoma.
Collapse
Affiliation(s)
- Y Shimomura
- Department of Obstetrics and Gynecology, Kobe University School of Medicine, Japan
| | | | | | | |
Collapse
|
92
|
Wiczyk HP, Grow DR, Adams LA, O'Shea DL, Reece MT. Pelvic adhesions contain sex steroid receptors and produce angiogenesis growth factors. Fertil Steril 1998; 69:511-6. [PMID: 9531888 DOI: 10.1016/s0015-0282(97)00529-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To evaluate female pelvic adhesion tissue for the presence of estrogen receptor (ER), progesterone receptor (PR), basic fibroblastic growth factor (basic-FGF), and vascular endothelial growth factor (VEGF). DESIGN Descriptive study. SETTING Patients at a tertiary medical center. PATIENTS Female reproductive age patients undergoing gynecologic surgery who were not receiving hormonal therapy. INTERVENTIONS Female reproductive tract peritoneal adhesion tissue was excised, frozen, and sent for immunohistologic evaluation. MAIN OUTCOME MEASURE Presence of ER, PR, basic-FGF, and VEGF in adhesion tissue. RESULTS Nineteen of 19 specimens were positive for PR; 16 of 19 specimens were positive for ER, which was present in a variety of the different cell types constituting adhesion. Vascular endothelial growth factor and basic-FGF were detected in endothelial cells of blood vessels supplying this tissue as well as in mesothelial cells. CONCLUSION Adhesion tissue contains ER, PR, and growth factors that may be important in the genesis of the permanent fibrovascular bands between pelvic organs. This supports the theoretical possibility of hormonal manipulation of these tissues to negatively influence postoperative pelvic adhesion formation.
Collapse
Affiliation(s)
- H P Wiczyk
- Department of Obstetrics and Gynecology, Tufts University School of Medicine, Springfield, Massachusetts, USA
| | | | | | | | | |
Collapse
|
93
|
Abstract
HB-EGF is a heparin-binding member of the EGF family that was initially identified in the conditioned medium of human macrophages. Soluble mature HB-EGF is proteolytically processed from a larger membrane-anchored precursor and is a potent mitogen and chemotactic factor for fibroblasts, smooth muscle cells but not endothelial cells. HB-EGF activates two EGF receptor subtypes, HER1 and HER4 and binds to cell surface HSPG. The transmembrane form of HB-EGF is a juxtacrine growth and adhesion factor and is uniquely the receptor for diphtheria toxin. HB-EGF gene expression is highly regulated, for example by cytokines, growth factors, and transcription factors such as MyoD. HB-EGF has been implicated as a participant in a variety of normal physiological processes such as blastocyst implantation and wound healing, and in pathological processes such as tumor growth, SMC hyperplasia and atherosclerosis.
Collapse
Affiliation(s)
- G Raab
- Department of Surgery, Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
94
|
Das SK, Das N, Wang J, Lim H, Schryver B, Plowman GD, Dey SK. Expression of betacellulin and epiregulin genes in the mouse uterus temporally by the blastocyst solely at the site of its apposition is coincident with the "window" of implantation. Dev Biol 1997; 190:178-90. [PMID: 9344537 DOI: 10.1006/dbio.1997.8694] [Citation(s) in RCA: 95] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In the mouse, the process of implantation is initiated by the attachment reaction between the blastocyst trophectoderm and uterine luminal epithelium that occurs at 2200-2300 h on day 4 (day 1 = vaginal plug) of pregnancy. Several members of the EGF family are considered important in embryo-uterine interactions during implantation. This investigation demonstrates that the expression of two additions to the family, betacellulin and epiregulin, are exquisitely restricted to the mouse uterine luminal epithelium and underlying stroma adjacent to the implanting blastocyst. These genes are not expressed during progesterone-maintained delayed implantation, but are rapidly switched on in the uterus surrounding the implanting blastocyst following termination of the delay by estrogen. These results provide evidence that expression of betacellulin and epiregulin in the uterus requires the presence of an active blastocyst and suggest an involvement of these growth factors in the process of implantation.
Collapse
Affiliation(s)
- S K Das
- Ralph L. Smith Research Center, University of Kansas Medical Center, Kansas City, Kansas 66160-7338, USA
| | | | | | | | | | | | | |
Collapse
|
95
|
Saji M, Taga M, Matsui H, Suyama K, Kurogi K, Minaguchi H. Gene expression and specific binding of platelet-derived growth factor and its effect on DNA synthesis in human decidual cells. Mol Cell Endocrinol 1997; 132:73-80. [PMID: 9324048 DOI: 10.1016/s0303-7207(97)00122-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
To clarify the biological significance of platelet-derived growth factor (PDGF) in human decidual cell function, which is important for the maintenance of pregnancy, we investigated gene expression of the PDGF subunits, PDGF-A and PDGF-B, specific binding of the PDGF isoform, and the effect of PDGF dimers on DNA synthesis in human decidual cells. We detected in decidua from early pregnancy the expected DNA bands of PDGF-A and PDGF-B by reverse transcriptase-polymerase chain reaction (RT-PCR) as well as mRNAs of each PDGF subunit by Northern blot hybridization, demonstrating that both PDGF subunits exist in this tissue. Scatchard plot analysis showed that decidual cells had both PDGF-alpha and PDGF-beta receptors. PDGF-AA, -AB and -BB stimulated [3H]-thymidine incorporation in cultured decidual cells in a dose-dependent manner. These results indicate the importance of PDGF in human decidua.
Collapse
Affiliation(s)
- M Saji
- Department of Obstetrics and Gynecology, Yokohama City University School of Medicine, Yokohama, Japan
| | | | | | | | | | | |
Collapse
|
96
|
Hanada K, Hattori MA, Fujihara N. Improvement of Rat Embryo Implantation by Epidermal Growth Factor. JOURNAL OF APPLIED ANIMAL RESEARCH 1997. [DOI: 10.1080/09712119.1997.9706188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
97
|
Kudoh M, Susaki Y, Ideyama Y, Nanya T, Mori M, Shikama H. Inhibitory effects of a novel aromatase inhibitor, YM511, on growth of endometrial explants and insulin-like growth factor-I gene expression in rats with experimental endometriosis. J Steroid Biochem Mol Biol 1997; 63:75-80. [PMID: 9449208 DOI: 10.1016/s0960-0760(97)00074-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Estrogen deprivation therapy effectively prevents progress of endometriosis but the precise mechanism by which estrogen stimulates growth of endometriotic implants is still unknown. We examined effects of hypoestrogenic state induced by ovariectomy, gonadotropin-releasing hormone agonist (leuprolide) or aromatase inhibitor (YM511), on growth of experimental endometrial explant, a section of endometrium transplanted under the renal capsule, in rats. Ovariectomy gradually reduced the volume of endometrial explants for 21 days. YM511 (0.1 mg/kg) and leuprolide (1 mg/rat) completely reduced volume of endometrial explants but they differed widely in the onset of inhibitory action. YM511 prevented growth of explants on day 4 but leuprolide had no inhibitory effect until day 15. YM511 dose-dependently reduced volume of endometrial explants and its minimum effective dose was 0.04 mg/kg. Insulin-like growth factor-I (IGF-I) mRNA expression in endometrial explant and uterus was examined on day 4. YM511 decreased IGF-I expression in endometrial explant and uterus by 58% and 48%, respectively. Reductions of the extent of IGF-I expression by YM511 and ovariectomy were comparable. A significant correlation between the volume and IGF-I mRNA expression in endometrial explant suggests that local expression of this gene may play an important role in stimulating growth of endometrial explants.
Collapse
Affiliation(s)
- M Kudoh
- Metabolic Diseases Research, Institute for Drug Discovery Research, Yamanouchi Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | | | | | | | | | | |
Collapse
|
98
|
|
99
|
Harada T, Fujikawa T, Yoshida S, Onohara Y, Tanikawa M, Terakawa N. Expression of transforming growth factor alpha (TGF-alpha) gene in mouse embryonic development. J Assist Reprod Genet 1997; 14:262-9. [PMID: 9147239 PMCID: PMC3454719 DOI: 10.1007/bf02765827] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
PURPOSE The expression of genes for TGF-alpha, epidermal growth factor (EGF), and the EGF receptor (EGFR) in mouse blastocysts was evaluated by the reverse transcription-polymerase chain reaction (RT-PCR). We evaluated the effects of TGF-alpha and EGF on the development of mouse embryo prior to implantation. RESULTS The results revealed the presence of transcripts of TGF-alpha and EGFR. However, EGF mRNA was not observed in repeated experiments. None of these growth factors influenced the rate of development from the two-cell stage to the blastocyst stage when added to the culture medium. These effects were further examined on measuring the incorporation of tritiated thymidine and leucine, providing indices of the synthesis of DNA and protein, respectively. A concentration of only 0.1 ng/ml of TGF-alpha, which shares a cell surface receptor with EGF, stimulated the synthesis of both DNA and protein. EGF at a concentration of 10 ng/ml stimulated the synthesis of DNA and protein by blastocysts. To explore autocrine effects of TGF-alpha on the rate of blastocoel expansion, TGF-alpha antisense oligodeoxynucleotides was used to reduce expression of the TGF-alpha gene. TGF-alpha at a concentration of 0.1 ng/ml stimulates the rate of blastocoel expansion in early cavitating mouse blastocysts. In contrast, TGF-alpha antisense oligonucleotides significantly reduced the rate of expansion. CONCLUSIONS Our present observations suggest that TGF-alpha/EGF and the EGFR may be involved in regulating embryonic development. In particular, TGF-alpha may serve as an autocrine factor in the regulation of embryonic development.
Collapse
Affiliation(s)
- T Harada
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Japan
| | | | | | | | | | | |
Collapse
|
100
|
Taga M, Sakakibara H, Suyama K, Ikeda M, Minaguchi H. Gene expression of transforming growth factor-alpha in human endometrium during decidualization. J Assist Reprod Genet 1997; 14:218-22. [PMID: 9130070 PMCID: PMC3454694 DOI: 10.1007/bf02766113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
PURPOSE We previously reported the gene expression of epidermal growth factor (EGF) in the process of decidualization in the human endometrium. This study was undertaken to investigate the biological significance of transforming growth factor-alpha (TGF-alpha), which shares a significant sequence homology with EGF, in the regulation of decidualization. METHODS The gene encoding TGF-alpha was analyzed by Northern blot hybridization in nonpregnant human endometria and decidua from 6 to 8 weeks of gestation as well as in cultured stromal cells with or without medroxyprogesterone acetate. RESULTS No transcript was detected in proliferative and secretory endometrium, whereas a transcript of 4.8 kb, which was in agreement with the size of human prepro-TGF-alpha mRNA previously reported, was clearly detected in decidua. Transcript of 4.8 kb was also detected in vitro in medroxyprogesterone acetate (MPA)-induced decidual cells, while no TGF-alpha transcript was found in endometrial stromal cells cultured without MPA. CONCLUSIONS These results suggest that the gene expression of TGF-alpha in uterine stromal cells is enhanced by stimulation from sex steroids and that TGF-alpha, like EGF, functions as one of the regulatory factors for decidualization in the human uterus.
Collapse
Affiliation(s)
- M Taga
- Department of Obstetrics and Gynecology, Yokohama City University School of Medicine, Japan
| | | | | | | | | |
Collapse
|