301
|
Bigsby RM, Caperell-Grant A, Berry N, Nephew K, Lubahn D. Estrogen induces a systemic growth factor through an estrogen receptor-alpha-dependent mechanism. Biol Reprod 2003; 70:178-83. [PMID: 13679309 DOI: 10.1095/biolreprod.103.021337] [Citation(s) in RCA: 21] [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
Estrogen induces proliferation of uterine epithelium through a paracrine action of estrogen receptor (ERalpha) in the underlying stroma. In ovariectomized mice primed with progesterone, estrogen stimulates proliferation in both the epithelium and the stroma. We set out to test whether a paracrine mode of action is involved in estrogen-induced proliferation of the uterine stroma. Epithelial and mesenchymal tissues derived from uteri of neonatal ERalpha null mice (ERalphaKO) or wild-type mice were separated and recombined in all four possible configurations (ERalpha+ or ERalpha- epithelium with ERalpha+ or ERalpha- mesenchyme) and grafted into female athymic mice. After 5 wk, hosts were ovariectomized and challenged with hormone treatment, and cellular proliferation was monitored by thymidine autoradiography. Results showed that, although the full response of the epithelium was dependent on an ERalpha-positive mesenchyme, stromal cell proliferation was independent of tissue ERalpha. This latter observation suggests that the response of the stroma was due to a systemic factor induced in the ERalpha-positive hosts. To test this possibility, pieces of whole uterus from neonatal wild-type or ERalphaKO mice were grafted into syngeneic wild-type or ERalphaKO hosts. In these whole-uterus grafts, estradiol stimulated ERalphaKO uterine stroma when they were grown in wild-type hosts but not when grown in ERalphaKO hosts. The epithelium of whole-uterus ERalphaKO grafts did not respond to estrogen, regardless of the host phenotype. These observations suggest that treatment of progesterone-primed mice with estradiol stimulates production of a systemic factor that is capable of inducing uterine stromal cell proliferation and that this systemic factor is produced by an ERalpha-dependent mechanism.
Collapse
Affiliation(s)
- Robert M Bigsby
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, Indiana 46202-5121, USA.
| | | | | | | | | |
Collapse
|
302
|
Grant KS, Wira CR. Effect of mouse uterine stromal cells on epithelial cell transepithelial resistance (TER) and TNFalpha and TGFbeta release in culture. Biol Reprod 2003; 69:1091-8. [PMID: 12773432 DOI: 10.1095/biolreprod.103.015495] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Recognizing that uterine stromal cells regulate several uterine epithelial cell function(s), the current study was undertaken to more fully define cell-cell communication in the uterus and to examine the role of uterine stromal cells in regulating epithelial cell monolayer integrity and cytokine release. Uterine epithelial and stromal cells from adult intact mice were isolated and cultured separately on cell culture inserts and/or in culture plates. Epithelial cells, which reach confluence as indicated by high transepithelial resistance (TER > 1000 ohms/well), preferentially release transforming growth factor-beta (TGFbeta) into the basolateral chamber ( approximately 70% > apical) and tumor necrosis factor-alpha (TNFalpha) into the apical compartment ( approximately 30% > basolateral). When epithelial cells on cell culture inserts were transferred to plates containing stromal cells, coculture for 24-48 h increased epithelial cell TER ( approximately 70% higher than control) and decreased TNFalpha release into both the apical and basolateral chambers ( approximately 30%-50%). In contrast, TGFbeta release was not affected by the presence of stromal cells. In other studies, the effects of stromal cells on epithelial cell TER and TNFalpha release persisted for 5-7 days following the removal of stromal cells and were also seen in coculture studies in which conditioned stromal media (CSM) was placed in the basolateral chamber. These studies indicate that uterine stromal cells produce a soluble factor(s) that regulates epithelial cell TER and release of TNFalpha without effecting TGFbeta release. These results suggest that uterine stromal cells communicate with epithelial cells via a soluble factor(s) to maintain uterine integrity and epithelial secretory function.
Collapse
Affiliation(s)
- Katherine S Grant
- Department of Physiology, Dartmouth Medical School, Lebanon, New Hampshire 03756, USA.
| | | |
Collapse
|
303
|
Jiang F, Saunders BO, Haller E, Livingston S, Nicosia SV, Bai W. Conditionally immortal ovarian cell lines for investigating the influence of ovarian stroma on the estrogen sensitivity and tumorigenicity of ovarian surface epithelial cells. In Vitro Cell Dev Biol Anim 2003; 39:304-12. [PMID: 12964904 DOI: 10.1290/1543-706x(2003)039<0304:cioclf>2.0.co;2] [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: 11/11/2022]
Abstract
The tendency of the ovarian surface epithelium (OSE) to undergo metaplastic and morphogenetic changes during the life cycle, at variance with the adjacent peritoneal mesothelial cells, suggests that its biology may be regulated by underlying ovarian stromal cues. However, little is known about the role that the ovarian stroma plays in the pathobiology of the OSE, largely because of the lack of a suitable in vitro model. Here, we describe the establishment and characterization of conditionally immortalized ovarian stromal and surface epithelial cell lines from H-2K(b)-tsA58 transgenic mice that carry the thermolabile mutant of SV-40 large T antigen under the control of an interferon-gamma (IFN-gamma)-inducible promoter. These cells express functional T antigens, grow continuously under permissive conditions at 33 degrees C in the presence of IFN-gamma, and stop dividing when the activity and expression of the tumor antigen is suppressed by restrictive conditions without IFN-gamma at 39 degrees C. Morphological, immunohistochemical, and ultrastructural analyses show that conditionally immortal OSE cells form cobblestone-like monolayers, express cytokeratin and vimentin, contain several microvilli, and develop tight junctions, whereas stromal cells are spindle-like, express vimentin but not cytokeratin, and contain rare microvilli, thus exhibiting epithelial and stromal phenotypes, respectively. At variance with the reported behavior of rat epithelial cells, conditionally immortal mouse epithelial cells are not spontaneously transformed after continuous culture in vitro. More importantly, conditioned media from stromal cells cultured under permissive conditions increase the specific activity of the endogenous estrogen receptor in BG-1 human ovarian epithelial cancer cells and promote these cells' anchorage-independent growth, suggesting the paracrine influence of a stromal factor. In addition, stromal cells cultured under restrictive conditions retain this growth-stimulatory activity, which, therefore, appears to be independent of T antigen expression. These established cell lines should provide a useful in vitro model system for studying the role of cellular interactions in OSE cell growth and tumorigenesis.
Collapse
Affiliation(s)
- Feng Jiang
- Department of Pathology, University of South Florida College of Medicine and H. Lee Moffitt Cancer Center, Tampa, Florida 33612-4799, USA
| | | | | | | | | | | |
Collapse
|
304
|
Wira CR, Rossoll RM. Oestradiol regulation of antigen presentation by uterine stromal cells: role of transforming growth factor-beta production by epithelial cells in mediating antigen-presenting cell function. Immunology 2003; 109:398-406. [PMID: 12807486 PMCID: PMC1782972 DOI: 10.1046/j.1365-2567.2003.01670.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2003] [Revised: 03/27/2003] [Accepted: 04/09/2003] [Indexed: 11/20/2022] Open
Abstract
We have previously shown that oestradiol treatment of ovariectomized rats for 3 days inhibits antigen presentation by uterine stromal cells at a time when oestradiol increases the numbers of antigen-presenting cells (APC) in the uterine stroma. In the present study, we found that oestradiol treatment for 1 day is sufficient to inhibit antigen presentation by stromal cells. To define the mechanism(s) of this inhibition, we examined the effect of cytokines and found that exogenous transforming growth factor-beta (TGF-beta) inhibits antigen presentation when stromal cells from saline- but not oestradiol-treated animals are incubated with ovalbumin (OVA)-specific T cells and OVA. In contrast, antigen presentation by uterine epithelial cells was not affected by TGF-beta. In other studies, the acute inhibitory effect of oestradiol (1 day) on stromal antigen presentation is fully reversed when anti-TGF-beta antibody is added to the culture media. When given for 3 days, oestradiol inhibition of antigen presentation is partially reversed by anti-TGF-beta antibody at a time when antibodies to tumour necrosis factor-alpha and interleukin-10 have no effect. To determine whether uterine epithelial cells produce TGF-beta, epithelial cells were grown to confluence on transwell inserts. Our findings indicate that uterine epithelial cells produce biologically active TGF-beta which is preferentially released basolaterally in the direction of underlying stromal cells. When oestradiol is given to ovariectomized rats 1 day before sacrifice, TGF-beta production by epithelial cells increases within 24 hr in culture, relative to saline controls. Taken together, these results suggest that oestradiol inhibition of stromal cell antigen presentation is mediated through the stimulatory effect of oestradiol on TGF-beta production by epithelial cells.
Collapse
Affiliation(s)
- Charles R Wira
- Department of Physiology, Dartmouth Medical School, Lebanon, NH 03756-0001, USA.
| | | |
Collapse
|
305
|
Bukovsky A, Caudle MR, Cekanova M, Fernando RI, Wimalasena J, Foster JS, Henley DC, Elder RF. Placental expression of estrogen receptor beta and its hormone binding variant--comparison with estrogen receptor alpha and a role for estrogen receptors in asymmetric division and differentiation of estrogen-dependent cells. Reprod Biol Endocrinol 2003; 1:36. [PMID: 12740031 PMCID: PMC155643 DOI: 10.1186/1477-7827-1-36] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2003] [Accepted: 04/15/2003] [Indexed: 11/29/2022] Open
Abstract
During human pregnancy, the production of 17-beta-estradiol (E2) rises steadily to eighty fold at term, and placenta has been found to specifically bind estrogens. We have recently demonstrated the expression of estrogen receptor alpha (ER-alpha) protein in human placenta and its localization in villous cytotrophoblast (CT), vascular pericytes, and amniotic fibroblasts. In vitro, E2 stimulated development of large syncytiotrophoblast (ST) aggregates. In the present study we utilized ER-beta affinity purified polyclonal (N19:sc6820) and ER-alpha monoclonal (clone h-151) antibodies. Western blot analysis revealed a single approximately 52 kDa ER-beta band in chorionic villi (CV) protein extracts. In CV, strong cytoplasmic ER-beta immunoreactivity was confined to ST. Dual color immunohistochemistry revealed asymmetric segregation of ER-alpha in dividing villous CT cells. Prior to separation, the cell nuclei more distant from ST exhibited high ER-alpha, while cell nuclei associated with ST showed diminution of ER-alpha and appearance of ER-beta. In trophoblast cultures, development of ST aggregates was associated with diminution of ER-alpha and appearance of ER-beta immunoreactivity. ER-beta was also detected in endothelial cells, amniotic epithelial cells and fibroblasts, extravillous trophoblast (nuclear and cytoplasmic) and decidual cells (cytoplasmic only). In addition, CFK-E12 (E12) and CWK-F12 (F12) monoclonal antibodies, which recognize approximately 64 kDa ER-beta with hormone binding domain, showed nuclear-specific reactivity with villous ST, extravillous trophoblast, and amniotic epithelium and fibroblasts. Western blot analysis indicated abundant expression of a approximately 64 kDa ER-beta variant in trophoblast cultures, significantly higher when compared to the chorionic villi and freshly isolated trophoblast cell protein extracts. This is the first report on ER-beta expression in human placenta and cultured trophoblast. Our data indicate that during trophoblast differentiation, the ER-alpha is associated with a less, and ER-beta with the more differentiated state. Enhanced expression of approximately 64 kDa ER-beta variant in trophoblast cultures suggests a unique role of ER-beta hormone binding domain in the regulation of trophoblast differentiation. Our data also indicate that asymmetric segregation of ER-alpha may play a role in asymmetric division of estrogen-dependent cells.
Collapse
Affiliation(s)
- Antonin Bukovsky
- Laboratory for Development, Differentiation and Cancer, Department of Obstetrics and Gynecology, The University of Tennessee Graduate School of Medicine, Knoxville, Tennessee 37920, USA
| | - Michael R Caudle
- Laboratory for Development, Differentiation and Cancer, Department of Obstetrics and Gynecology, The University of Tennessee Graduate School of Medicine, Knoxville, Tennessee 37920, USA
| | - Maria Cekanova
- Laboratory for Development, Differentiation and Cancer, Department of Obstetrics and Gynecology, The University of Tennessee Graduate School of Medicine, Knoxville, Tennessee 37920, USA
| | - Romaine I Fernando
- Laboratory for Development, Differentiation and Cancer, Department of Obstetrics and Gynecology, The University of Tennessee Graduate School of Medicine, Knoxville, Tennessee 37920, USA
| | - Jay Wimalasena
- Laboratory for Development, Differentiation and Cancer, Department of Obstetrics and Gynecology, The University of Tennessee Graduate School of Medicine, Knoxville, Tennessee 37920, USA
| | - James S Foster
- Laboratory for Development, Differentiation and Cancer, Department of Obstetrics and Gynecology, The University of Tennessee Graduate School of Medicine, Knoxville, Tennessee 37920, USA
| | - Donald C Henley
- Laboratory for Development, Differentiation and Cancer, Department of Obstetrics and Gynecology, The University of Tennessee Graduate School of Medicine, Knoxville, Tennessee 37920, USA
| | - Robert F Elder
- Laboratory for Development, Differentiation and Cancer, Department of Obstetrics and Gynecology, The University of Tennessee Graduate School of Medicine, Knoxville, Tennessee 37920, USA
| |
Collapse
|
306
|
Chauhan H, Abraham A, Phillips JRA, Pringle JH, Walker RA, Jones JL. There is more than one kind of myofibroblast: analysis of CD34 expression in benign, in situ, and invasive breast lesions. J Clin Pathol 2003; 56:271-6. [PMID: 12663638 PMCID: PMC1769930 DOI: 10.1136/jcp.56.4.271] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
AIMS Smooth muscle actin (SMA) positive myofibroblasts have been implicated in tumour invasion; however, acquisition of SMA is not limited to peritumorous fibroblasts and other changes in fibroblasts may be more specifically related to the malignant environment. CD34 is a sialomucin expressed by normal breast fibroblasts but lost in invasive carcinomas. The aim of this study was to establish the relation between CD34 and SMA expression in breast fibroblasts and to analyse whether loss of CD34 is specific for invasive disease. METHODS Immunohistochemistry for CD34 and SMA was performed on 135 cases including 10 normal, 10 fibroadenomas, 40 infiltrating ductal carcinomas, 55 cases of ductal carcinoma in situ (DCIS), and 20 radial scar/complex sclerosing lesions. The relation between staining pattern and histopathological features was recorded as positive, negative, or reduced. RESULTS Fibroblasts around all normal duct-lobule units and those showing epithelial hyperplasia were CD34 positive and mainly SMA negative. In fibroadenomas, fibroblasts retained CD34 but acquired SMA expression. In contrast, fibroblasts around invasive carcinoma were CD34 positive and SMA negative. In DCIS, loss of CD34 was significantly more frequent in high grade tumours than in low or intermediate grade ones (p < 0.001). The acquisition of SMA was seen more frequently than the loss of CD34, particularly in non-high grade DCIS. In all radial scars, fibroblasts were SMA positive but CD34 negative, and a similar pattern was seen in stromal cells in areas of fibrosis following core biopsy. CONCLUSIONS These results show that SMA positive myofibroblasts exhibit variable expression of CD34, indicating that these markers are not coordinately controlled. Loss of CD34 is strongly related to the malignant phenotype, in both invasive and preinvasive disease, but is not entirely specific because radial scar fibroblasts and fibroblasts in reactive fibrosis exhibit a similar phenotype. The functional relevance of altered CD34 expression is unclear but the very focal changes implicate local signalling mechanisms probably of epithelial origin.
Collapse
Affiliation(s)
- H Chauhan
- Department of Pathology, University Hospital of Leicester NHS Trust, Leicester LE2 7LX, UK
| | | | | | | | | | | |
Collapse
|
307
|
Yamazawa K, Matsui H, Seki K, Sekiya S. A case-control study of endometrial cancer after antipsychotics exposure in premenopausal women. Oncology 2003; 64:116-23. [PMID: 12566908 DOI: 10.1159/000067769] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Most endometrial cancers are related to hormonal imbalance, and antipsychotics are a common cause of hyperprolactinemia. We investigated the possible relation between the use of antipsychotics and the risk of endometrial cancer. METHODS A case-control study was conducted on premenopausal women at the Chiba University Hospital between 1989 and 2000. The cases were 41 patients with histologically confirmed endometrial cancer. For each case, there were 3 controls matched by age and date of visit. Subsequently 123 women without endometrial cancer were enrolled as controls. Detailed information including obesity, parity, hypertension, diabetes mellitus, use of exogenous estrogen and use of antipsychotics was analyzed. Odds ratios, two-sided p values and 95% confidence intervals were estimated by a conditional logistic regression analysis. Radioimmunoassays of serum luteinizing hormone, follicle-stimulating hormone, prolactin, estradiol and progesterone were performed in 4 of 5 patients with endometrial cancer who were using antipsychotics. RESULTS Use of antipsychotics, diabetes mellitus and obesity were identified as independent variables with risk estimates of 5.4, 9.3 and 4.9, respectively. Serum progesterone, estradiol and prolactin levels ranged from 0.4 to 0.7 ng/ml, 32 to 110 pg/ml and 34 to 258 ng/ml, respectively. CONCLUSIONS Use of antipsychotics is a risk factor for endometrial cancer, and hyperprolactinemia associated with antipsychotics may impart the risk of endometrial cancer in premenopausal women.
Collapse
Affiliation(s)
- Koji Yamazawa
- Department of Reproductive Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | | | | | | |
Collapse
|
308
|
Sukjumlong S, Kaeoket K, Dalin AM, Persson E. Immunohistochemical studies on oestrogen receptor alpha (ER alpha) and the proliferative marker Ki-67 in the sow uterus at different stages of the oestrous cycle. Reprod Domest Anim 2003; 38:5-12. [PMID: 12535323 DOI: 10.1046/j.1439-0531.2003.00383.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In order to better understand physiological changes during the different stages of the oestrous cycle, immunohistochemistry was used in the present study to investigate the distribution of oestrogen receptor alpha (ER alpha) as well as the proliferative marker Ki-67, in the sow uterus during the oestrous cycle. Uterine samples were collected from multiparous sows with normal reproductive performance at selected stages of the oestrous cycle: at late dioestrus (d 17), prooestrus (d 19), oestrous (d 1), early dioestrus (d 4) and dioestrus (d 11-12), respectively. The tissue samples were fixed in 10% formaldehyde, embedded in paraffin and subjected to immunohistochemistry using monoclonal antibodies against ER alpha (C-311) and Ki-67 (MM-1). In general, the immunostaining of both ER alpha and Ki-67 was confined to nuclei of the target cells. Variations were seen, not only at the different stages of the oestrous cycle, but also in the different tissue compartments of the uterus. In the epithelia, the strongest ER alpha staining and highest amount of positive Ki-67 cells were found at early dioestrus. In the myometrium, the highest levels of staining of both ER alpha and Ki-67 positive cells were found at pro-oestrus and oestrus. For the proliferative marker, Ki-67, no positive cells were found at dioestrus and late dioestrus in the epithelium and myometrium. In the connective tissue stroma (subepithelial layer), the highest number of ER alpha positive cells were found at oestrus, which was significantly different compared with other stages (p< or = 0.05), whereas the levels of Ki-67 positive cells were relatively low and did not differ between the stages examined. Significant correlations between the number of ER alpha positive cells in the stroma and Ki-67 positive cells in the epithelia were observed. This suggests indirect regulatory mechanisms on epithelial proliferation via ER alpha in the stroma. In conclusion, these findings in the sow uterus show that the presence of ER alpha as well as Ki-67 protein varies not only between different stages of the oestrous cycle but also between different tissue compartments of the uterus. These findings indicate various regulatory mechanisms and stress the importance of localising ER alpha and proliferating cells in different uterine tissues.
Collapse
Affiliation(s)
- S Sukjumlong
- Department of Anatomy and Histology, Centre for Reproductive Biology in Uppsala (CRU), Faculty of Veterinary Medicine, Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden
| | | | | | | |
Collapse
|
309
|
Stavreus-Evers A, Hovatta O, Eriksson H, Landgren BM. Development and characterization of an endometrial tissue culture system. Reprod Biomed Online 2003; 7:243-9. [PMID: 14567900 DOI: 10.1016/s1472-6483(10)61759-2] [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: 01/09/2023]
Abstract
A culture system has been developed for pieces of whole endometrium, comprising many different cell types. The system maintains the integrity of all compartments important for tissue function and allows functional studies of the human endometrium. Endometrial biopsy strips were collected from fertile women. The strips were cut into small pieces and cultured for 8 h on dishes pre-coated with Matrigel. The oxygen concentration was kept low (5%), since it was shown to be beneficial for the culture in preliminary studies. Scanning electron microscopy and immunohistochemical detection of proliferating cell nuclear antigen (PCNA), oestrogen receptor alpha (ERalpha), progesterone receptor (PR) A + B and B and TUNEL were used to characterize the endometrium. During culture, the luminal and glandular epithelial cells began to cover the surface injured when cutting the sample, suggesting a healing process. The luminal epithelial surface appeared normal after culture. There was an increased sensitivity to culture after the implantation window. This new method for culture of endometrial strips in vitro has proven successful and may be used for functional studies of the human endometrium.
Collapse
Affiliation(s)
- Anneli Stavreus-Evers
- Division of Reproductive Endocrinology, Department of Woman and Child Health, Karolinska Hospital, 171 76 Stockholm, Sweden.
| | | | | | | |
Collapse
|
310
|
Hastings JM, Licence DR, Burton GJ, Charnock-Jones DS, Smith SK. Soluble vascular endothelial growth factor receptor 1 inhibits edema and epithelial proliferation induced by 17beta-estradiol in the mouse uterus. Endocrinology 2003; 144:326-34. [PMID: 12488361 DOI: 10.1210/en.2002-220641] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The uterine response to 17beta-estradiol (E2) includes increased water retention, enhanced vascular permeability, DNA and RNA synthesis, and increased cellular mitosis. We have used the natural antagonist of vascular endothelial growth factor A (VEGF-A), sflt-1 (soluble form of flt-1), to determine whether the edematous and proliferative effects of E2 in the uterus are mediated by VEGF-A. Female BALB/c mice were ovariectomized and treated with E2 (10 micro g/kg) in the absence or presence of sflt-1 (0.8 and 4.0 mg/kg) for 24 h. E2 induced increases in uterine mass from 25.3 to 36.8 mg, in total cross-sectional uterine area from 771 to 1133 micro m(2), in cross-sectional endometrial area from 268 to 569 micro m(2), and in the mitotic index of lumenal epithelial cells from 0% to 53%. Antagonism with sflt-1 reduced the E2-induced increases in total uterine area to 779 micro m(2), endometrial area to 398 micro m(2) and the mitotic index of lumenal epithelial cells to 25%, but the E2-induced increase in uterine mass was not significantly reduced. From these data we conclude that the edematous response and proliferation of lumenal epithelial cells in the murine uterus are mediated in part through VEGF-A. These data suggest that sflt-1 could be a useful anti-VEGF-A agent and may be effective in modifying uterine biology.
Collapse
Affiliation(s)
- Julie M Hastings
- Reproductive Molecular Research Group, Department of Pathology, University of Cambridge, Cambridge, United Kingdom CB2 1QP
| | | | | | | | | |
Collapse
|
311
|
Blackwell DM, Speth RC, Mirando MA. Morphometric analysis of the uterine endometrium of swine on days 12 and 16 postestrus. THE ANATOMICAL RECORD. PART A, DISCOVERIES IN MOLECULAR, CELLULAR, AND EVOLUTIONARY BIOLOGY 2003; 270:59-66. [PMID: 12494490 DOI: 10.1002/ar.a.10182] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The uterine endometrium of swine is comprised of luminal epithelial, glandular epithelial, and stromal cells that secrete the luteolysin, prostaglandin F(2alpha) (PGF(2alpha)), during late diestrus. However, which of these cells contribute the most to luteolytic PGF(2alpha) secretion is unknown because the cellular composition of the endometrium has not been quantified. Therefore, this study quantified the cellular composition of the endometrium on days 12 and 16 postestrus by histologic and morphometric analyses. On day 12, the endometrium consisted predominantly of stromal cells (47% of total cell quantity) and glandular epithelial cells (37%), whereas luminal epithelial cells represented only 16% of the total of the three cell types. The number of glandular epithelial cells tended to increase (P < 0.10) between days 12 and 16, such that they comprised 45% of the endometrium by day 16, while the number of stromal and luminal cells did not change and accounted for 45% and 10% of the cells, respectively. Luminal epithelial cells had a 58% greater cross-sectional area (P < 0.001) than glandular epithelial cells, whereas glandular epithelial cells had a 22% greater area (P < 0.001) than stromal cells. Glandular epithelial cells decreased (P < 0.001) in cross-sectional area between days 12 and 16, whereas the area of luminal epithelial and stromal cells remained unchanged. These results indicate that the porcine endometrium is comprised predominantly of stromal and glandular epithelial cells that are likely to contribute substantially to endometrial PGF(2alpha) secretion during luteolysis. The contribution of glandular epithelium to luteolytic PGF(2alpha) secretion probably increases during diestrus as the number of these cells increases.
Collapse
Affiliation(s)
- Dianna M Blackwell
- Department of Animal Sciences, Washington State University, Pullman, Washington, USA
| | | | | |
Collapse
|
312
|
Pillai SB, Jones JM, Koos RD. Treatment of rats with 17beta-estradiol or relaxin rapidly inhibits uterine estrogen receptor beta1 and beta2 messenger ribonucleic acid levels. Biol Reprod 2002; 67:1919-26. [PMID: 12444070 DOI: 10.1095/biolreprod.102.003392] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Estrogen regulates the growth and differentiation of the uterus via binding to estrogen receptors (ERs), members of the nuclear receptor family of transcription factors. Two forms of ER exist: ERalpha and ERbeta. The former is a well-characterized mediator of estrogen-induced transcription, but the function of the latter is unclear. Recent in vitro studies suggest that both splicing forms of ERbeta expressed in rat tissues, beta1 and beta2, may function as inhibitors of ERalpha transcriptional activity. To gain insight into the role of ERbeta in estrogen action, we examined the effects of estrogen and relaxin, a ligand-independent activator of ERs, on the expression of ERbeta1 and ERbeta2 mRNA in the uterus in vivo. Eighteen-day-old female rats were ovariectomized and, after recovery, treated with 17beta-estradiol, relaxin, or vehicle. Quantitative reverse transcription-polymerase chain reaction analyses of uterine RNA from estrogen-treated animals revealed marked decreases in the steady-state levels of the mRNAs for both ERbeta1 and ERbeta2 at 3, 6, and 24 h after treatment. Relaxin induced a similar effect. Neither hormone had any significant effect on ERalpha mRNA levels. To determine if endogenous estrogen exerts this effect, we examined the expression of ERbetas in the uterus during the estrous cycle. Levels of both isoforms were highest at diestrus (low estrogen), were significantly lower at early proestrus (rising estrogen), reached a nadir during late proestrus (peak estrogen), and rebounded at estrus (declining estrogen). These data suggest that down-regulation of ERbeta expression may be required for estrogen to exert its full trophic effects on the uterus.
Collapse
Affiliation(s)
- Suresh B Pillai
- Department of Physiology, University of Maryland School of Medicine, Baltimore 21201, USA
| | | | | |
Collapse
|
313
|
Abstract
Prostatic development is induced by androgens acting via mesenchymal-epithelial interactions. Androgens elicit their morphogenetic effects by acting through androgen receptors (ARs) in urogenital sinus mesenchyme (UGM), which induces prostatic epithelial development. In adulthood reciprocal homeostatic stromal-epithelial interactions maintain functional differentiation and growth-quiescence. Testosterone plus estradiol (T+E2) have been shown to induce prostatic carcinogenesis in animal models. Thus, tissue recombinant studies were undertaken to explore the mechanisms of prostatic carcinogenesis in BPH-1 cells in which ARs and estrogen receptors (ERs) are undetectable. For this purpose, BPH-1 cells were combined with UGM, and the UGM+BPH-1 recombinants were grafted to adult male hosts. Solid branched epithelial cords and ductal structures formed in untreated UGM+BPH-1 recombinants. Growth was modest, and tumors did not develop. UGM+BPH-1 recombinants treated with T+E2 formed invasive carcinomas. BPH-1 cells lack ARs and ERs, whereas rat UGM expresses both of these receptors. These data show that immortalized nontumorigenic human prostatic epithelial cells can undergo hormonal carcinogenesis in response to T+E2 stimulation via paracrine mechanisms and demonstrate that the stromal environment plays an important role in mediating hormonal carcinogenesis. During prostatic carcinogenesis the stroma undergoes progressive loss of smooth muscle with the appearance of carcinoma-associated fibroblasts (CAF). This altered stroma was tested for its ability to promote carcinogenesis of nontumorigenic but immortalized human prostatic epithelial cells (BPH-1). CAF+BPH-1 tissue recombinants formed large carcinomas. In contrast, recombinants composed of normal prostatic stroma+BPH-1 cells exhibited minimal growth. This stroma-induced malignant transformation was associated with additional genetic alterations and changes in gene expression. Thus, alteration in the stromal microenvironment was sufficient to promote malignant transformation of human prostatic epithelial cells.
Collapse
Affiliation(s)
- Gerald R Cunha
- Departments of Anatomy and Urology, University of California, San Francisco, CA 94143-0452, USA.
| | | | | |
Collapse
|
314
|
Curtis Hewitt S, Goulding EH, Eddy EM, Korach KS. Studies Using the Estrogen Receptor α Knockout Uterus Demonstrate That Implantation but Not Decidualization-Associated Signaling Is Estrogen Dependent. Biol Reprod 2002; 67:1268-77. [PMID: 12297545 DOI: 10.1095/biolreprod67.4.1268] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Ovarian hormonal signaling is essential for proper functioning of the uterus in the establishment of pregnancy. Previous studies have demonstrated that decidualization, a stromal transformation that occurs in response to embryo implantation, can be elicited in the uterus of estrogen receptor alpha knockout (alphaERKO) mice in the absence of the estrogen dependence normally seen in wild-type (WT) mice for this response. While the alphaERKO stromal compartment demonstrated the necessary decidual response, embryo implantation is a process initiated in the epithelial layer, a uterine component that lacks estrogen responsiveness in the alphaERKO. To determine if the alphaERKO uterus would be competent for implantation, donor embryos were transferred into the uterine lumen of WT and alphaERKO females that had been ovariectomized and treated with exogenous estradiol and progesterone to mimic early pregnancy. No implantation occurred in the alphaERKO, while implantation sites containing live embryos were seen in similarly treated WT uteri, indicating that functional estrogen receptor alpha (ERalpha) is required for implantation. Previous observations of estrogen-independent decidualization in the alphaERKO prompted investigation of the mechanism leading to estrogen independence of this process. The disruption of progesterone receptor (PR), Hoxa10, Cox2, or LIF in transgenic mice results in the loss of decidualization response. Therefore, the expression of these genes was studied in WT and alphaERKO uteri by comparing expression following vehicle, progesterone alone (P), or estradiol priming followed by progesterone with nidatory estradiol (E+Pe) and by comparing expression following the above hormonal manipulations in addition to luminal infusion of oil used previously as decidualization-initiating stimulus. The whole-uterus level of PR and Hoxa10 mRNAs did not vary; however, the PR protein was induced in the stroma 24 h after oil infusion. Interestingly, in the WT, this induction was most apparent in samples receiving E+Pe, while in the alphaERKO samples, the induction occurred independent of any hormone priming. Cox2 protein and mRNA increased in both WT and alphaERKO samples 2 h after oil infusion in all three of the treatment groups. In the WT samples, Cox2 levels remained elevated 24 h after oil infusion only in the E+Pe treatment group; however, the elevated Cox2 was seen in samples taken 24 h after oil infusion in all three alphaERKO treatment groups. The alphaERKO uterine tissue appeared to sustain more extensive damage when examined 24 h after oil infusion. Severe trauma, such as crushing of the uterine tissue, has previously been shown to remove the requirement for nidatory estradiol for deciduomas to develop, indicating that the greater susceptibility of alphaERKO uterine tissue to damage from intraluminal oil infusion is contributing to decidualization in the absence of ERalpha. Leukemia inhibitory factor (LIF) mRNA was also induced following estradiol treatment in the WT, but also following oil infusion in WT samples that were not treated with estradiol. In contrast, estradiol does not induce LIF mRNA in the alphaERKO, but oil infusion leads to a robust increase in LIF in all alphaERKO sample groups. LIF binds and activates its membrane receptor, which initiates responses including the phosphorylation and nuclear translocation of Stat3 transcription factor. Thus, Stat3 phosphorylation was studied in WT and alphaERKO samples and found to be induced following oil infusion in all samples. Together, these and previous observations illustrate that estrogen is essential for epithelial proliferation and embryo implantation and that estrogen is dispensable for stromal decidualization in the alphaERKO, as the essential genes and signals required for the response are still induced.
Collapse
Affiliation(s)
- Sylvia Curtis Hewitt
- Laboratory of Reproductive and Developmental Toxicology, NIEHS, NIH, Research Triangle Park, North Carolina 27709, USA
| | | | | | | |
Collapse
|
315
|
Hendry WJ, Sheehan DM, Khan SA, May JV. Developing a laboratory animal model for perinatal endocrine disruption: the hamster chronicles. Exp Biol Med (Maywood) 2002; 227:709-23. [PMID: 12324652 DOI: 10.1177/153537020222700904] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
At the biomedical, regulatory, and public level, considerable concern surrounds the concept that inappropriate exposure to endocrine-disrupting chemicals, especially during the prenatal and/or neonatal period, may disrupt normal reproductive tract development and adult function. The intent of this review was to 1. Describe some unique advantages of the hamster for perinatal endocrine disruptor (ED) studies, 2. Summarize the morphological and molecular consequences of exposure to the established perinatal ED, diethylstilbestrol, in the female and male hamster, 3. Present some new, histomorphological insight into the process of neonatal diethylstilbestrol-induced disruption in the hamster uterus, and 4. Introduce recent efforts and future plans to evaluate the potency and mechanism of action of other putative EDs in the hamster experimental system. Taken together, the findings indicate that the hamster represents a unique and sensitive in vivo system to probe the phenomenon of endocrine disruption. The spectrum of candidate endpoints includes developmental toxicity, neoplasia, and more subtle endpoints of reproductive dysfunction.
Collapse
Affiliation(s)
- William J Hendry
- Department of Biological Sciences, Wichita State University, Kansas 67260-0026, USA.
| | | | | | | |
Collapse
|
316
|
Carnahan KG, Uzumcu M, Hu J, Sample GL, Braileanu GT, Mirando MA. Oxytocin stimulates secretion of prostaglandin F(2alpha) from endometrial cells of swine in the presence of progesterone. Domest Anim Endocrinol 2002; 23:435-45. [PMID: 12206876 DOI: 10.1016/s0739-7240(02)00176-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Oxytocin (OT) stimulates endometrial secretion of prostaglandin (PG) F(2 alpha) during corpus luteum regression in swine but there is differential responsiveness to OT among endometrial cell types. To determine if progesterone influenced responsiveness of luminal epithelial, glandular epithelial, and stromal cells to 100 nM OT during luteolysis in swine, cells were isolated from endometrium of 15 gilts by differential enzymatic digestion and sieve filtration on day 16 postestrus and cultured continuously in the presence of 0, 10 or 100 nM progesterone. For phospholipase C (PLC) activity and PGF(2 alpha) secretion, stromal cells were most responsive to OT (P<0.01) in the absence of progesterone, whereas luminal epithelial cells were unresponsive and glandular epithelial cells displayed an intermediate response to OT (P<0.09). Progesterone enhanced PLC activity linearly in glandular epithelial cells (P<0.05) and influenced it quadratically in stromal cells (P=0.05). The effect of OT and progesterone on PLC activity in luminal epithelial cells was not significant, and progesterone did not increase PLC activity in response to OT in any cell type. Culture in the presence of progesterone, enhanced PGF(2 alpha) secretion in response to OT in luminal epithelial cells (P<0.05) but not in glandular epithelial or stromal cells. Progesterone also increased overall PGF(2 alpha) release from glandular epithelial (P<0.05) and stromal cells (P<0.06) across both levels of OT treatment. These results indicate that progesterone enhanced PGF(2 alpha) secretion from luminal epithelial cells in response to OT and increased basal PGF(2 alpha) release from glandular epithelial and stromal cells.
Collapse
Affiliation(s)
- K G Carnahan
- Department of Animal Sciences, Center for Reproductive Biology, Washington State University, Pullman, WA 99164-6353, USA
| | | | | | | | | | | |
Collapse
|
317
|
Weihua Z, Ekman J, Almkvist A, Saji S, Wang L, Warner M, Gustafsson JA. Involvement of androgen receptor in 17beta-estradiol-induced cell proliferation in rat uterus. Biol Reprod 2002; 67:616-23. [PMID: 12135905 DOI: 10.1095/biolreprod67.2.616] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Although it is known that, in the uterus, estrogen receptor alpha (ERalpha) is involved in proliferation and progesterone receptor in differentiation, the role of the two other gonadal-hormone receptors expressed in the uterus, androgen receptor (AR) and estrogen receptor beta (ERbeta), remains undefined. In this study, the involvement of AR in 17beta-estradiol (E(2))-induced cellular proliferation in the immature rat uterus was investigated. AR levels were low in the untreated immature uterus, but 24 h after treatment of rats with E(2), there was an increase in the levels of AR and of two androgen-regulated genes, IGF-I and Crisp (cysteine-rich secretory protein). As expected, E(2) induced proliferation of luminal epithelial cells. These actions of E(2) were all blocked by both the antiestrogen tamoxifen and the antiandrogen flutamide. The E(2)-induced AR was found by immunohistochemistry to be localized exclusively in the stroma, mainly in the myometrium, where it colocalized with ERalpha but not with ERbeta. ERbeta, detected with two different ERbeta-specific antibodies, was expressed in both stromal and epithelial cells either alone or together with ERalpha. Treatment with E(2) caused down-regulation of ERalpha and ERbeta in the epithelium. The data suggest that, in E(2)-induced epithelial cell proliferation, ERalpha induces stromal AR and AR amplifies the ERalpha signal by induction of IGF-I. Because AR is never expressed in cells with ERbeta, it is unlikely that ERbeta signaling is involved in this pathway. These results indicate an important role for AR in proliferation of the uterus, where estrogen and androgen do not represent separate pathways but are sequential steps in one pathway.
Collapse
Affiliation(s)
- Zhang Weihua
- Departments of Medical Nutrition and Bioscience, Karolinska Institute, Novum, Huddinge, Stockholm S-141 86, Sweden.
| | | | | | | | | | | | | |
Collapse
|
318
|
Lessey BA, Gui Y, Apparao KBC, Young SL, Mulholland J. Regulated expression of heparin-binding EGF-like growth factor (HB-EGF) in the human endometrium: a potential paracrine role during implantation. Mol Reprod Dev 2002; 62:446-55. [PMID: 12112577 DOI: 10.1002/mrd.10129] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Heparin-binding epidermal growth factor (HB-EGF) is a recently identified member of the EGF growth factor family found to be expressed in the uterus of both mouse and human at the time of implantation. In the present study, we investigated the expression patterns of HB-EGF in normal cycling endometrium and compared its expression with the fertility-associated endometrial epithelial biomarkers alpha(v)beta(3) integrin, leukemia inhibitory factor (LIF) and homeobox gene, HOXA-10. RNase protection assay (RPA) using RNA made from endometrium collected from different phases of the menstrual cycle demonstrated increased HB-EGF expression during the mid-secretory phase, a pattern similar to, but slightly preceding the expression of alpha(v)beta(3) integrin and HOXA-10. In vitro studies demonstrated stimulation of HB-EGF expression by estradiol-17beta (E(2)) and progesterone (P(4)) alone or in combination in stromal cells. Combined treatment with E(2) + P(4) was, however, required to stimulate epithelial HB-EGF expression. In vitro experiments demonstrated the ability of HB-EGF to stimulate epithelial expression of the key endometrial proteins including LIF, HOXA-10, and the beta(3) integrin subunit. Each has previously been demonstrated to be an important epithelial biomarker expressed during the implantation window. In addition, conditioned media from endometrial stromal cells treated with E(2) + P(4) + relaxin mimicked the stimulatory effect of HB-EGF on epithelial expression of the beta(3) integrin subunit. The stimulatory effect of the stromal-conditioned medium was blocked by antibodies that neutralize a known receptor for HB-EGF. These data suggest that uterine receptivity may be regulated in part by the stromal-derived HB-EGF.
Collapse
Affiliation(s)
- Bruce A Lessey
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
| | | | | | | | | |
Collapse
|
319
|
Day JK, Bauer AM, DesBordes C, Zhuang Y, Kim BE, Newton LG, Nehra V, Forsee KM, MacDonald RS, Besch-Williford C, Huang THM, Lubahn DB. Genistein alters methylation patterns in mice. J Nutr 2002; 132:2419S-2423S. [PMID: 12163704 DOI: 10.1093/jn/132.8.2419s] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In this study we examine the effect of the phytoestrogen genistein on DNA methylation. DNA methylation is thought to inhibit transcription of genes by regulating alterations in chromatin structure. Estrogenic compounds have been reported to regulate DNA methylation in a small number of studies. Additionally, phytoestrogens are believed to affect progression of some human diseases, such as estrogen-dependent cancers, osteoporosis and cardiovascular disease. Specifically, our working hypothesis is that certain soy phytoestrogens, such as genistein, may be involved in preventing the development of certain prostate and mammary cancers by maintaining a protective DNA methylation profile. The objective of the present study is to use mouse differential methylation hybridization (DMH) arrays to test for changes in the methylation status of the cytosine guanine dinucleotide (CpG) islands in the mouse genome by examining how these methylation patterns are affected by genistein. Male mice were fed a casein-based diet (control) or the same diet containing 300 mg genistein/kg according to one of four regimens: control diet for 4 wk, genistein diet for 4 wk, control diet for 2 wk followed by genistein diet for 2 wk and genistein diet for 2 wk followed by control diet for 2 wk. DNA from liver, brain and prostate were then screened with DMH arrays. Clones with methylation differences were sequenced and compared with known sequences. In conclusion, consumption of genistein diet was positively correlated with changes in prostate DNA methylation at CpG islands of specific mouse genes.
Collapse
Affiliation(s)
- J Kevin Day
- Department of Biochemistry, University of Missouri, Columbia 65211, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
320
|
Kawashima K, Yamakawa K, Takahashi W, Takizawa S, Yin P, Sugiyama N, Kanba S, Arita J. The estrogen-occupied estrogen receptor functions as a negative regulator to inhibit cell proliferation induced by insulin/IGF-1: a cell context-specific antimitogenic action of estradiol on rat lactotrophs in culture. Endocrinology 2002; 143:2750-8. [PMID: 12072410 DOI: 10.1210/endo.143.7.8915] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Estrogens stimulate cell proliferation in typical estrogen-responsive tissues including the anterior pituitary gland. Here we report that 17-beta estradiol (E2) has estrogen receptor-mediated mitogenic and antimitogenic actions on rat lactotrophs in primary culture, depending on the cell context. E2 did not affect basal proliferation at 2 d after treatment, but it increased it at 4 d. Insulin markedly increased proliferative activity, which was inhibited by simultaneous treatment with E2, even after only 2 d of treatment. This antimitogenic action on insulin-induced proliferation was also observed with other estrogens but not with nonestrogenic steroids. Treatment with antiestrogens in combination with E2 antagonized both the mitogenic and antimitogenic actions of E2. Antiestrogen treatment alone inhibited basal proliferation, and it mimicked the inhibitory action of E2 on insulin-induced proliferation with less potency. In parallel with cell proliferation, an insulin-induced increase in the cell number of cyclin D1-immunoreactive lactotrophs was inhibited by E2 treatment. Although the antimitogenic action of E2 was seen with a wide range of doses of insulin or IGF-1, proliferation was stimulated rather than inhibited by E2 when cells were treated with serum or forskolin/isobutylmethylxanthine instead of insulin, indicating a mitogen-specific, but not proliferative activity-dependent, inhibition by E2. The results of estrogen-occupied estrogen receptors as negative regulators of proliferation suggest a novel interaction between estrogen and growth factors in the regulation of proliferation in estrogen-responsive cells.
Collapse
Affiliation(s)
- Kengo Kawashima
- Department of Physiology, Yamanashi Medical University, Japan
| | | | | | | | | | | | | | | |
Collapse
|
321
|
Zumbado M, Boada LD, Torres S, Monterde JG, Díaz-Chico BN, Afonso JL, Cabrera JJ, Blanco A. Evaluation of acute hepatotoxic effects exerted by environmental estrogens nonylphenol and 4-octylphenol in immature male rats. Toxicology 2002; 175:49-62. [PMID: 12049835 DOI: 10.1016/s0300-483x(02)00046-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nonylphenol (NP) and 4-Octylphenol (4OP) have shown estrogenic properties both in vivo and in vitro. Researchers have known for years that estrogens induce a wide number of hepatotoxic actions in rodents. In order to study the acute hepatic effects exerted by NP and 4OP on rat liver the following endpoints were evaluated: relative liver weight (RLW), morphology, cell cycle and ploidy status, monooxygenase enzymes content and levels of both, cytosolic estrogen receptor (cER) and microsomal binding sites for estrogens (mEBS). Immature male Sprague-Dawley rats were injected intraperitoneally (i.p.) with 60 mg/kg of NP or 4OP for 1, 5 or 10 days. Despite the fact that RLW of the animals was not modified but any treatment, the histopathological study revealed the presence of an increase in the percentage of both, mitotic activity and Ki-67-labeling index (LI) in the livers from animals treated with alkylphenols in absence of any degenerative lesion. Furthermore, all the livers from alkylphenols-treated groups showed the presence of abnormal mitosis and c-mitosis. Although the levels of both, cER and cytochrome P450 (Cyt. P450) were not affected by any treatment, concentration of the mEBS was decreased after 10 days of treatment with alkylphenols. These findings taken together suggest that the exposition to alkylphenols induce cell proliferation and spindle disturbances and that these compounds are capable of modulating the expression of putative membrane receptors for estrogens.
Collapse
Affiliation(s)
- Manuel Zumbado
- Laboratory of Toxicology, Department of Clinical Sciences, Health Sciences Center and Faculty of Veterinary Medicine, University of Las Palmas de Gran Canaria, PO Box 550, 35080 Las Palmas de Gran Canaria, Canary Islands, Spain
| | | | | | | | | | | | | | | |
Collapse
|
322
|
Vermeirsch H, Van den Broeck W, Coryn M, Simoens P. Immunohistochemical detection of androgen receptors in the canine uterus throughout the estrus cycle. Theriogenology 2002; 57:2203-16. [PMID: 12141570 DOI: 10.1016/s0093-691x(02)00908-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Serum androgen levels in the bitch increase during proestrus and remain elevated until metestrus. To find out whether androgens can have a direct impact on the canine uterus, androgen receptors (AR) were identified immunohistochemically in uterine tissue. Androgen receptor distribution in the uterine horns, body and cervix was described during different cycle stages, during pregnancy and in the postpartum period. Nuclear staining for AR was observed in cells of the surface epithelium, glandular ducts, basal glands and stroma of the endometrium, and in myometrial smooth muscle cells. In addition, cytoplasmic staining was observed in epithelial cells from proestrus to early metestrus, when the cells were secretory active, and in stroma cells during pregnancy, suggesting a role for androgens in decidualization. During pregnancy and in the postpartum period nuclear staining for AR was nearly absent. During the estrus cycle stroma cells stained with higher intensities for AR than epithelial cells, supporting the idea that stroma cells mediate some effects of steroid hormones on epithelial cells in the genital tract. In contrast with earlier findings on estrogen receptor-alpha and progesterone receptors, no significant changes in androgen receptor expression were observed during the estrus cycle. Few correlations were found between the staining for AR and serum levels of the sex steroids. The present findings suggest that there is a basal expression of AR in the canine uterus throughout the estrus cycle that may not be influenced by sex steroid hormones.
Collapse
|
323
|
Engelhardt H, Croy BA, King GJ. Conceptus influences the distribution of uterine leukocytes during early porcine pregnancy. Biol Reprod 2002; 66:1875-80. [PMID: 12021075 DOI: 10.1095/biolreprod66.6.1875] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Pregnancy in humans and rodents is associated with dramatic changes in leukocyte populations within the uterus. In these species, recruitment of leukocytes, mostly natural killer (NK) lymphocytes, accompanies decidualization of endometrial stroma even in the absence of pregnancy. In the pig, a nondecidualizing species, the predominant lymphocytes in the pregnant uterus are T and/or NK cells, but their distribution relative to embryonic attachment sites has not been reported. The objective of this study was to compare the abundance of leukocytes in porcine endometrium in contact with trophoblast with that between attachment sites during the early postattachment period. Uteri were recovered on Days 15-17 (n = 4), 18 and 19 (n = 4), 21 and 22 (n = 5), and 25-27 (n = 2) of gestation and from cycling pigs during the luteal phase (Day 15; n = 3). Leukocytes were identified in uterus obtained at versus between attachment sites using an antibody reactive with all leukocytes (CD44). In all pregnant animals, leukocytes were diffusely scattered throughout the endometrial stroma but were rare or absent in the luminal epithelium. Leukocyte density was approximately 3-fold greater in endometrium in contact with conceptuses than in endometrium between attachment sites throughout the early postattachment period. Leukocyte density during the luteal phase was similar to that between attachment sites, suggesting that leukocyte recruitment was a localized response to the embryo. The ability of an individual porcine conceptus to recruit maternal leukocytes to the adjacent stroma may be a vital step in early placental development and embryo survival.
Collapse
Affiliation(s)
- Heidi Engelhardt
- Department of Animal and Poultry Science, University of Guelph, Guelph, Ontario, Canada N1G 2W1.
| | | | | |
Collapse
|
324
|
Yoshida M, Takenaka A, Katsuda SI, Kurokawa Y, Maekawa A. Neonatal exposure to p-tert-octylphenol causes abnormal expression of estrogen receptor alpha and subsequent alteration of cell proliferating activity in the developing Donryu rat uterus. Toxicol Pathol 2002; 30:357-64. [PMID: 12051553 DOI: 10.1080/01926230252929936] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
In the present study, we investigated immunohistochemically the time-course alterations in estrogen receptor alpha (ER) expression and cell proliferating activity in the developing uteri of Donryu rats exposed neonatally to a high dose p-tert-octylphenol (OP), an endocrine disrupting chemical (EDC). OP-treatment (sc injections of 100 mg/kg, every other day from postnatal days 1 to 15) induced an early and enhanced ER expression in the luminal epithelium compared with age-matched controls from postnatal day (PND) 10, and increased proliferating cell nuclear antigen (PCNA) positive cells up to PND21. At PND28, ER expression in the luminal epithelium of the OP-treated group was decreased, in association with decline in the luminal epithelial areas. PND14, the second week of life, is coincident with the normal time for differentiation when the luminal epithelium invaginates into the stroma to form uterine glands. OP-treatment, however, delayed and inhibited gland-formation, and suppressed ER expression in the invaginated-luminal and glandular epithelium at this time. These results indicate that ER expression in these sites is strongly linked with cell proliferating activity. In stromal cells, ER was expressed from PND6 in both groups without any PCNA positive cells, but significantly lower values were noted in the OP-treated group up to PND10. Our immunohistochemical investigation did not reveal any abnormalities in expression of the proto-oncogene c-fos, mitotic inhibitor p21, or epidermal growth factor antigen, although the apoptotic index in the luminal epithelium was slightly increased in the OP-treated group. These results demonstrate neonatal effects of a high dose of OP, already detectable at PND10, with early and enhanced ER expression, resulting in increase of cell proliferative activity in the luminal epithelium, though expression in the glandular epithelium was suppressed in relation to inhibited gland-genesis. The present study thus suggests that neonatal exposure to high doses of EDCs with estrogenic activity can induce abnormal differentiation in the developing rat uteri via abnormal ER expression and subsequent alteration of cell proliferating activity.
Collapse
Affiliation(s)
- Midori Yoshida
- Department of Pathology, Sasaki Institute, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
325
|
Abstract
Endometrial receptivity towards embryo implantation is a complex process that involves the ovary, endometrium and embryo. The dialog between the ovary and the endometrium provides the hormonal stimulus for establishment of a successful pregnancy. The hormones estrogen and progesterone act in concert to stimulate the expression of key molecules necessary for embryos to attach and invade. It is thought that initial attachment of the embryo involves cell adhesion events. The best characterized cell adhesion molecule on the luminal surface of the endometrium is the alphavbeta3 integrin. Its ligand osteopontin (OPN) is co-localized with alphavbeta3 and may play a role in endometrial or embryo signaling or facilitate embryo attachment to the apical surface prior to invasion. Surprising new evidence suggests that these two proteins are differentially regulated. Acting directly on endometrial epithelium, progesterone stimulates OPN expression. Using a stromal-mediated paracrine mechanism, HB-EGF or other EGF molecules appear to stimulate epithelial alphavbeta3 expression. In this article, we review what is known about these two pathways.
Collapse
Affiliation(s)
- Bruce A Lessey
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
326
|
Nie R, Zhou Q, Jassim E, Saunders PTK, Hess RA. Differential expression of estrogen receptors alpha and beta in the reproductive tracts of adult male dogs and cats. Biol Reprod 2002; 66:1161-8. [PMID: 11906937 DOI: 10.1095/biolreprod66.4.1161] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Expression of estrogen receptors (ERs) in the reproductive tracts of adult male dogs and cats has not been reported. In the present study, ERalpha and ERbeta were localized by immunohistochemistry using ER-specific antibodies. ERalpha was found in interstitial cells and peritubular myoid cells in the dog testis, but only in interstitial cells of the cat. In rete testis of the dog, epithelial cells were positive for ERalpha staining, but in the cat, rete testis epithelium was only weakly positive. In efferent ductules of the dog, both ciliated and nonciliated cells stained intensely positive. In the cat, ciliated epithelial cells were less stained than nonciliated epithelial cells. Epithelial cells in dog epididymis and vas deferens were negative for ERalpha. In the cat, except for the initial region of caput epididymis, ERalpha staining was positive in the epithelial cells of epididymis and vas deferens. Multiple cell types of dog and cat testes stained positive for ERbeta. In rete testis and efferent ductules, epithelial cells were weakly positive for ERbeta. Most epithelial cells of the epididymis and vas deferens exhibited a strong positive staining in both species. In addition, double staining was used to demonstrate colocalization of both ERalpha and ERbeta in efferent ductules of both species. The specificity of antibodies was demonstrated by Western blot analysis. This study reveals a differential localization of ERalpha and ERbeta in male dog and cat reproductive tracts, demonstrating more intensive expression of ERbeta than ERalpha. However, as in other species, the efferent ductules remained the region of highest concentration of ERalpha.
Collapse
Affiliation(s)
- Rong Nie
- Department of Veterinary Biosciences, University of Illinois, 2001 S. Lincoln, Urbana, IL 61802, USA
| | | | | | | | | |
Collapse
|
327
|
Abstract
The endometrium undergoes characteristic histologic changes during the menstrual cycle as it prepares for embryo implantation. Historic and current data suggest the presence of a defined period of maximal uterine receptivity during the mid-secretory phase occurring between days 7 and 10 postovulation. In recent years, we and others have sought to define biochemical markers of receptivity that might be used to better understand this time of endometrial differentiation. Based on the work with cell adhesion molecules, we have discovered three different integrins that are only coexpressed during this time in the cycle when embryos will successfully implant. By studying the regulation of one of these, the alpha(v)beta3 integrin, and its extracellular matrix ligand, osteopontin (OPN), we have defined two separate regulatory pathways that may regulate endometrial receptivity. While alpha(v)beta3 expression appears to be stimulated by EGF or heparin-binding EGF, osteopontin is stimulated by progesterone. We now believe the former pathway is a paracrine-mediated signal, while the latter is a direct effect of progesterone on the estrogen-primed endometrial epithelium. In women with endometriosis, it appears that alpha(v)beta3 expression is reduced, while OPN expression is unaffected. Interestingly, binding of OPN to the surface epithelium appears quite limited when alpha(v)beta3 expression is lacking. Such evidence continues to reinforce the notion that endometrium from some women with endometriosis is dysfunctional and may account for the reduction in cycle fecundity noted in this group of patients.
Collapse
Affiliation(s)
- Bruce A Lessey
- Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, 27599, USA.
| |
Collapse
|
328
|
DeMayo FJ, Zhao B, Takamoto N, Tsai SY. Mechanisms of action of estrogen and progesterone. Ann N Y Acad Sci 2002; 955:48-59; discussion 86-8, 396-406. [PMID: 11949965 DOI: 10.1111/j.1749-6632.2002.tb02765.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Estrogen and progesterone are steroid hormones that play a pivotal role in the regulation of mammalian reproduction. One primary action of these hormones is to regulate the development and function of the uterus. These hormones act by regulating the transcription of specific genes in the uterus. The actions of these hormones are mediated by their specific hormone receptors. These receptors are nuclear transcription factors, whose transcriptional regulatory activity is mediated by the binding of the specific steroid to these molecules. Once these receptors bind hormone, they can bind to specific cis-acting sequences in the promoter region of responsive genes and regulate transcription of these genes. In the regulation of transcription, these receptors interact with specific cofactors to activate the transcriptional machinery. A second gene family, the Steroid Receptor Coactivator (SRC) family, has been identified that serves to modulate the transcriptional activity of the hormone receptors. To date, three members of the SRC family have been identified. During the last decade, gene targeting technology has been used to identify the role of these receptors in the regulation of reproduction and uterine biology.
Collapse
Affiliation(s)
- Francesco J DeMayo
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | | | |
Collapse
|
329
|
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: 1.3] [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
|
330
|
Affiliation(s)
- Yasunori YOSHIMURA
- Department of Obstetrics and Gynecology, Keio University School of Medicine
| |
Collapse
|
331
|
Yamashita S. Ontogeny of Estrogen Receptor (ER) .ALPHA. and .BETA. in the Female Reproductive Tissues of Mice Neonatally Exposed to Diethylstilbestrol. Acta Histochem Cytochem 2002. [DOI: 10.1267/ahc.35.395] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
332
|
Yang S, Fang Z, Gurates B, Tamura M, Miller J, Ferrer K, Bulun SE. Stromal PRs mediate induction of 17beta-hydroxysteroid dehydrogenase type 2 expression in human endometrial epithelium: a paracrine mechanism for inactivation of E2. Mol Endocrinol 2001; 15:2093-105. [PMID: 11731611 DOI: 10.1210/mend.15.12.0742] [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: 11/19/2022] Open
Abstract
Progesterone stimulates the expression of 17beta-hydroxysteroid dehydrogenase (HSD) type 2, which catalyzes the conversion of the potent estrogen, E2, to an inactive form, estrone, in epithelial cells of human endometrial tissue. Various effects of progesterone on uterine epithelium have recently been shown to be mediated by stromal PRs in mice. We describe herein a critical paracrine mechanism whereby progesterone induction of 17beta-HSD type 2 enzyme activity, transcript levels, and promoter activity in human endometrial epithelial cells are mediated primarily by PR in endometrial stromal cells. Medium conditioned with progestin-pretreated human endometrial stromal cells robustly increased 17beta-HSD type 2 enzyme activity (2-fold) and mRNA levels (13.2-fold) in Ishikawa malignant endometrial epithelial cells. In contrast, direct progestin treatment of Ishikawa epithelial cells gave rise to much smaller increases in enzyme activity (1.2-fold) and mRNA levels (4-fold). These results suggest that progesterone- dependent paracrine factors arising from stromal cells are primarily responsible for the induction of epithelial 17beta-HSD type 2 expression in the endometrium. We transfected serial deletion mutants of the -1,244 bp 5'-flanking region of the 17beta-HSD type 2 gene into Ishikawa cells. No progesterone response elements could be identified upstream of the 17beta-HSD type 2 promoter. Stromal PR-dependent induction of the 17beta-HSD type 2 promoter was mediated by a critical regulatory region mapped to the -200/-100 bp sequence. Direct treatment of Ishikawa cells with progestin gave rise to a maximal increase in the activity of -200 bp/Luciferase construct only by 1.2-fold, whereas medium conditioned by progestin-pretreated endometrial stromal cells increased promoter activity up to 2.4-fold in a time- and concentration-dependent manner. The stimulatory effect of medium conditioned by progestin-pretreated stromal cells was enhanced strikingly by increasing stromal cell PR levels with the addition of estrogen. This epithelial-stromal interaction was specific for endometrial epithelial cells, since 17beta-HSD type 2 could not be induced in malignant breast epithelial cells by media conditioned with progestin-treated breast or endometrial stromal cells. In conclusion, progesterone regulates the conversion of biologically active E2 to estrone by inducing the 17beta-HSD type 2 enzyme in human endometrial epithelium primarily via PR in stromal cells, which secrete factors that induce transcription mediated primarily by the -200/-100 bp 5'-regulatory region of the 17beta-HSD type 2 promoter.
Collapse
Affiliation(s)
- S Yang
- Division of Reproductive Endocrinology, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | |
Collapse
|
333
|
Abstract
The balance between cell proliferation and programmed cell death (apoptosis) determines body patterns during animal development and controls compartment sizes, tissue architecture and remodeling. The removal of primordial structures by apoptosis allows the organism to develop sex specifically and to adapt for novel functions at later stages; apoptosis also limits the size of evolving structures. It is a ubiquitous function that is essential for all cells. Although inappropriate regulation or execution of apoptosis leads to disease, such as cancer, there is now evidence for its great therapeutic potential. This would be particularly true if apoptosis could be targeted at defined cell compartments, rather than acting ubiquitously like chemotherapy. Here, we discuss the potential of nuclear receptor ligands, many of which act through their cognate receptors in defined body compartments as modulators of cell life and death, with special emphasis on the molecular pathways by which these receptors affect cell-cycle progression, survival and apoptosis.
Collapse
Affiliation(s)
- L Altucci
- Dipartimento di Patologia Generale e Oncologia, Seconda Università degli Studi di Napoli, Centro Sperimentale S. Andrea delle Dame, Via De Crecchio 7, 80138 Naples, Italy
| | | |
Collapse
|
334
|
Kurita T, Cooke PS, Cunha GR. Epithelial-stromal tissue interaction in paramesonephric (Müllerian) epithelial differentiation. Dev Biol 2001; 240:194-211. [PMID: 11784056 DOI: 10.1006/dbio.2001.0458] [Citation(s) in RCA: 133] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
During organogenesis, the middle to caudal portion of Müllerian epithelium differentiates into uterine and vaginal epithelia in females. Functional differentiation of uterine and vaginal epithelia occurs in adulthood, and is regulated by 17beta-estradiol (E(2)) and progesterone. In this report, the roles of mesenchyme/stroma in differentiation of uterine and vaginal epithelia were studied in tissue recombination experiments. At birth, Müllerian epithelium was negative for uterine and vaginal epithelial markers. Tissue recombinant experiments showed that uterine and vaginal gene expression patterns were induced in neonatal Müllerian epithelium by the respective mesenchymes. Differentiated adult uterine and vaginal epithelia did not change their original gene expression in response to heterotypic mesenchymal induction. In the adult vagina, E(2) induced expression of involucrin, a CCAAT/enhancer-binding protein beta and cytokeratin 1 via estrogen receptor alpha (ERalpha). Tissue recombination experiments with wild-type and ERalpha knockout mice demonstrated that epithelial gene expression is regulated by E(2) via epithelial-stromal tissue interactions. Uterine/vaginal heterotypic tissue recombinations demonstrated that functional differentiation of uterine and vaginal epithelia required organ-specific stromal factors. In contrast, stromal signals regulating epithelial proliferation appeared to be nonspecific in the uterus and vagina.
Collapse
Affiliation(s)
- T Kurita
- Department of Anatomy, University of California, San Francisco, California 94143-0452, USA
| | | | | |
Collapse
|
335
|
Gray CA, Bartol FF, Tarleton BJ, Wiley AA, Johnson GA, Bazer FW, Spencer TE. Developmental biology of uterine glands. Biol Reprod 2001; 65:1311-23. [PMID: 11673245 DOI: 10.1095/biolreprod65.5.1311] [Citation(s) in RCA: 334] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
All mammalian uteri contain endometrial glands that synthesize or transport and secrete substances essential for survival and development of the conceptus (embryo/fetus and associated extraembryonic membranes). In rodents, uterine secretory products of the endometrial glands are unequivocally required for establishment of uterine receptivity and conceptus implantation. Analyses of the ovine uterine gland knockout model support a primary role for endometrial glands and, by default, their secretions in peri-implantation conceptus survival and development. Uterine adenogenesis is the process whereby endometrial glands develop. In humans, this process begins in the fetus, continues postnatally, and is completed during puberty. In contrast, endometrial adenogenesis is primarily a postnatal event in sheep, pigs, and rodents. Typically, endometrial adenogenesis involves differentiation and budding of glandular epithelium from luminal epithelium, followed by invagination and extensive tubular coiling and branching morphogenesis throughout the uterine stroma to the myometrium. This process requires site-specific alterations in cell proliferation and extracellular matrix (ECM) remodeling as well as paracrine cell-cell and cell-ECM interactions that support the actions of specific hormones and growth factors. Studies of uterine development in neonatal ungulates implicate prolactin, estradiol-17 beta, and their receptors in mechanisms regulating endometrial adenogenesis. These same hormones appear to regulate endometrial gland morphogenesis in menstruating primates and humans during reconstruction of the functionalis from the basalis endometrium after menses. In sheep and pigs, extensive endometrial gland hyperplasia and hypertrophy occur during gestation, presumably to provide increasing histotrophic support for conceptus growth and development. In the rabbit, sheep, and pig, a servomechanism is proposed to regulate endometrial gland development and differentiated function during pregnancy that involves sequential actions of ovarian steroid hormones, pregnancy recognition signals, and lactogenic hormones from the pituitary or placenta. That disruption of uterine development during critical organizational periods can alter the functional capacity and embryotrophic potential of the adult uterus reinforces the importance of understanding the developmental biology of uterine glands. Unexplained high rates of peri-implantation embryonic loss in humans and livestock may reflect defects in endometrial gland morphogenesis due to genetic errors, epigenetic influences of endocrine disruptors, and pathological lesions.
Collapse
Affiliation(s)
- C A Gray
- Center for Animal Biotechnology and Genomics, Department of Animal Science, Texas A&M University, College Station, Texas 77843, USA
| | | | | | | | | | | | | |
Collapse
|
336
|
Rhen T, Crews D. Distribution of androgen and estrogen receptor mRNA in the brain and reproductive tissues of the leopard gecko, Eublepharis macularius. J Comp Neurol 2001; 437:385-97. [PMID: 11503141 DOI: 10.1002/cne.1290] [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: 11/09/2022]
Abstract
Incubation temperature during embryonic development determines gonadal sex in the leopard gecko, Eublepharis macularius. In addition, both incubation temperature and gonadal sex influence behavioral responses to androgen and estrogen treatments in adulthood. Although these findings suggest that temperature and sex steroids act upon a common neural substrate to influence behavior, it is unclear where temperature and hormone effects are integrated. To begin to address this question, we identified areas of the leopard gecko brain that express androgen receptor (AR) and estrogen receptor (ER) mRNA. We gonadectomized adult female and male geckos from an incubation temperature that produces a female-biased sex ratio and another temperature that produces a male-biased sex ratio. Females and males from both temperatures were then treated with equivalent levels of various sex steroids. Region-specific patterns of AR mRNA expression and ER mRNA expression were observed upon hybridization of radiolabeled (35S) cRNA probes to thin sections of reproductive tissues (male hemipenes and female oviduct) and brain. Labeling for AR mRNA was very intense in the epithelium, but not within the body, of the male hemipenes. In contrast, expression of ER mRNA was prominent in most of the oviduct but not in the luminal epithelium. Within the brain, labeling for AR mRNA was conspicuous in the anterior olfactory nucleus, the lateral septum, the medial preoptic area, the periventricular preoptic area, the external nucleus of the amygdala, the anterior hypothalamus, the ventromedial hypothalamus, the premammillary nucleus, and the caudal portion of the periventricular nucleus of the hypothalamus. Expression of ER mRNA was sparse in the septum and was prominent in the ventromedial hypothalamus, the caudal portion of the periventricular nucleus of the hypothalamus, and a group of cells near the torus semicircularis. Many of these brain regions have been implicated in the regulation of hormone-dependent, sex-typical reproductive and agonistic behaviors in other vertebrates. Consequently, these nuclei are likely to control such behaviors in the leopard gecko and also are candidate neural substrates for mediating temperature effects on behavior.
Collapse
Affiliation(s)
- T Rhen
- Section of Integrative Biology, School of Biological Sciences, University of Texas at Austin, Austin, Texas 78712, USA.
| | | |
Collapse
|
337
|
Kimmins S, MacLaren LA. Oestrous cycle and pregnancy effects on the distribution of oestrogen and progesterone receptors in bovine endometrium. Placenta 2001; 22:742-8. [PMID: 11597195 DOI: 10.1053/plac.2001.0708] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Reproductive cyclicity and pregnancy require cell and tissue specific expression of oestrogen (OR) and progesterone receptors (PR). To determine the effects of cycle day and early pregnancy (days 16-30) on the distribution of OR and PR, uteri were collected from cycling (n=39), pregnant (n=14), and steroid-treated ovariectomized cattle (n=10). Immunohistochemistry was used to localize OR and PR, and staining intensity was scored. Moderate staining for the PR was detected in the stroma at proestrus (days 17-20). Expression increased through oestrous, reaching maximal levels by metestrus (days 1-6) (P< 0.05). During diestrus (days 7-16), PR expression was downregulated (P< 0.05). Progesterone receptor expression was detected in the luminal epithelium in one of three animals each at days 3 and 6 of the oestrous cycle. In pregnant animals only, caruncular stroma showed reduced PR staining compared to intercaruncular stroma (P< 0.05). Reactivity to the OR antibody in cycling animals was detected in the stroma and glandular epithelium. Levels increased through proestrus and oestrous, were maximal during metestrus (P< 0.05), and decreased during diestrus (P< 0.05). Interestingly, there was transient upregulation in the luminal epithelium on day 16 or day 14 (P< 0.05). This was not evident at day 16 of pregnancy, although some OR reactivity was present in stroma and glands. By day 18 of pregnancy OR were undetectable (P< 0.05). Oestrogen-treated ovariectomized animals had increased OR and PR expression in comparison to progesterone treated cows (P< 0.05). Untreated ovariectomized cows showed constitutive steroid receptor expression. These results support the widely accepted role for sex steroids in the regulation of their receptors and indicate that there is paracrine regulation of steroid receptor expression in the bovine uterus.
Collapse
Affiliation(s)
- S Kimmins
- Department of Plant and Animal Sciences, Nova Scotia Agricultural College, Truro, Nova Scotia, Canada, B2N 5E3
| | | |
Collapse
|
338
|
Tarleton BJ, Wiley AA, Bartol FF. Neonatal estradiol exposure alters uterine morphology and endometrial transcriptional activity in prepubertal gilts. Domest Anim Endocrinol 2001; 21:111-25. [PMID: 11585701 DOI: 10.1016/s0739-7240(01)00106-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Porcine endometrial development between birth (postnatal day = PND 0) and PND 56 involves differentiation of glandular epithelium (GE) from luminal epithelium (LE) and estrogen receptor-alpha (ER) expression. Juvenile ER architecture evolves after birth, as stroma and nascent GE first express ER. Mature ER architecture is evident after PND 30, when stroma, GE and LE are ER-positive. When administered during discrete periods between PND 0 and 56, effects of estradiol-17beta valerate (EV) on the neonatal porcine uterus relate to endometrial ER architecture. Transient EV exposure from birth reduces embryo survival in pregnant adult gilts. Effects of EV, administered as juvenile endometrial ER architecture develops (P1, PND 0-13), or after mature ER architecture is established (P2, PND 42-55), were evaluated in uteri from gilts treated with corn oil or EV in P1 or P2 and hysterectomized on PND 100 without additional steroids (NSt), on PND 102 after EV on PND100-101 (EV2), or on PND 117 after EV2 followed by progesterone on PND 102-116 (EP). Neonatal EV reduced uterine weight (P < 0.02), size (P < 0.01), luminal protein content (P < 0.07), and percent incorporation of 3H-leucine into nondialyzable endometrial products in vitro (P < 0.01). Group (NSt, EV2, EP) -specific treatment effects detected for endometrial ER, progesterone receptor, uteroferrin, and/or retinol binding protein mRNA levels were frequently related to period (P1,P2). Results support the idea that estrogen-sensitive postnatal organizational events, including those defined, in part, by endometrial ER architecture, are likely components of genetic and epigenetic programs governing uterine morphogenesis and ontogeny of endometrial function in the pig.
Collapse
Affiliation(s)
- B J Tarleton
- Department of Animal and Dairy Sciences, Program in Cell and Molecular Biosciences, Auburn University, Auburn, Alabama 36849-5415, USA
| | | | | |
Collapse
|
339
|
Lundeen SG, Zhang Z, Zhu Y, Carver JM, Winneker RC. Rat uterine complement C3 expression as a model for progesterone receptor modulators: characterization of the new progestin trimegestone. J Steroid Biochem Mol Biol 2001; 78:137-43. [PMID: 11566438 DOI: 10.1016/s0960-0760(01)00091-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Progestins have a wide variety of activities in female reproduction. There are also pharmacological applications for progestins, including hormone replacement therapy and contraception. Here we report the development and characterization of the rat uterine complement component C3 mRNA as a molecular target for the evaluation of the antiestrogenic activity of progestins in the uterus. In this assay, ethinyl estradiol (EE) is used to stimulate C3 expression and progestins are then evaluated for their ability to inhibit this expression. The three reference progestins, progesterone (P4), levonorgestrel (LNG), and medroxyprogesterone acetate (MPA) blocked the increase in C3 mRNA levels induced by EE. Dexamethasone (DEX) and 17alpha-methyl testosterone did not inhibit the estrogen induced C3 mRNA levels; in fact, DEX caused a further increase in C3 mRNA levels. Finally, the antiprogestin RU486 was able to block the MPA inhibition of C3 message. RU486, like DEX, caused an increase in C3 mRNA levels above that of estrogen treatment alone. The model was also used to evaluate trimegestone (TMG), a new steroidal progestin, that has been shown to be a potent and selective progesterone receptor agonist. The activity of TMG in the rat uterine decidualization and ovulation inhibition assays was similar to MPA. However, in the C3 model, TMG caused a dose-dependent inhibition of the EE induced C3 message and was approximately five-fold more potent in this model than MPA (EC(50) of 4.7 microg/kg and 26.5 microg/kg, respectively). Therefore, TMG was a more potent antagonist of estrogenic activity in the uterine endometrium than any of the reference progestins tested and therefore may be more effective in protecting the endometrium in hormone replacement therapy.
Collapse
Affiliation(s)
- S G Lundeen
- Endocrinology Division, Women's Health Research Institute, Wyeth-Ayerst Research, 145 King of Prussia Road, Radnor, PA 19087 USA.
| | | | | | | | | |
Collapse
|
340
|
Weihua Z, Makela S, Andersson LC, Salmi S, Saji S, Webster JI, Jensen EV, Nilsson S, Warner M, Gustafsson JA. A role for estrogen receptor beta in the regulation of growth of the ventral prostate. Proc Natl Acad Sci U S A 2001; 98:6330-5. [PMID: 11371645 PMCID: PMC33468 DOI: 10.1073/pnas.111150898] [Citation(s) in RCA: 335] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In normal rats and mice, immunostaining with specific antibodies revealed that nuclei of most prostatic epithelial cells harbor estrogen receptor beta (ERbeta). In rat ventral prostate, 530- and 549-aa isoforms of the receptor were identified. These sediment in the 4S region of low-salt sucrose gradients, indicating that prostatic ERbeta does not contain the same protein chaperones that are associated with ERalpha. Estradiol (E(2)) binding and ERbeta immunoreactivity coincide on the gradient, with no indication of ERalpha. In prostates from mice in which the ERbeta gene has been inactivated (BERKO), androgen receptor (AR) levels are elevated, and the tissue contains multiple hyperplastic foci. Most epithelial cells express the proliferation antigen Ki-67. In contrast, prostatic epithelium from wild-type littermates is single layered with no hyperplasia, and very few cells express Ki-67. Rat ventral prostate contains an estrogenic component, which comigrates on HPLC with the testosterone metabolite 5alpha-androstane-3beta,17beta-diol (3betaAdiol). This compound, which competes with E(2) for binding to ERbeta and elicits an estrogenic response in the aorta but not in the pituitary, decreases the AR content in prostates of wild-type mice but does not affect the elevated levels seen in ERbeta knockout (BERKO) mice. Thus ERbeta, probably as a complex with 3betaAdiol, is involved in regulating the AR content of the rodent prostate and in restraining epithelial growth. These findings suggest that ligands specific for ERbeta may be useful in the prevention and/or clinical management of prostatic hyperplasia and neoplasia.
Collapse
Affiliation(s)
- Z Weihua
- Department of Medical Nutrition, Karolinska Institute, Novum, Huddinge, S-141 86 Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
341
|
Rosenfeld CS, Roberts RM, Lubahn DB. Estrogen receptor- and aromatase-deficient mice provide insight into the roles of estrogen within the ovary and uterus. Mol Reprod Dev 2001; 59:336-46. [PMID: 11424220 DOI: 10.1002/mrd.1039] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Estrogen receptor (ER)- (alpha, beta, and both alpha and beta) and aromatase (Ar) knockout (KO) mice have been created to assess the biological effects of estrogens. This review article discusses the ovarian and uterine phenotypes of these mice. The data obtained have confirmed some older inferences about how the steroid acts, but have also revealed some unexpected aspects of estrogen action. Mol. Reprod. Dev. 59:336-346, 2001.
Collapse
Affiliation(s)
- C S Rosenfeld
- Department of Animal Sciences, University of Missouri, Columbia, Missouri 65211, USA
| | | | | |
Collapse
|
342
|
Arnold JT, Kaufman DG, Seppälä M, Lessey BA. Endometrial stromal cells regulate epithelial cell growth in vitro: a new co-culture model. Hum Reprod 2001; 16:836-45. [PMID: 11331626 DOI: 10.1093/humrep/16.5.836] [Citation(s) in RCA: 186] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The regulation of epithelial cell function and morphogenesis by the paracrine effectors from the mesenchyme or stroma has been well established using in-vivo studies. A more complete understanding of these relationships has been delayed due, in part, to a lack of appropriate co-culture models. In this study, we describe a co-culture model which demonstrates that normal paracrine relationships can be reconstituted in vitro and that human endometrial stromal cells regulate both growth and differentiation of primary human endometrial epithelial cells. Interesting differences in the proliferation of stromal and epithelial cells were noted in response to the basement membrane extract, Matrigel((R)). Exposure of stromal cells to Matrigel((R)) enhanced the paracrine capacity of these cells in vitro. When epithelial cells were co-cultured in contact with stromal cells embedded in Matrigel((R)), epithelial cell growth was inhibited by 65-80% compared to controls. Stromal cells in contact with Matrigel((R)) also regulated epithelial cell differentiation, as shown by induction of glycodelin expression. These co-culture studies show great promise as a method to investigate the cellular interactions between endometrial stromal and epithelial cells and their environment and to understand the molecular basis for the regulation of normal growth and differentiation of cells within complex tissues such as the endometrium.
Collapse
Affiliation(s)
- J T Arnold
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | | | | |
Collapse
|
343
|
Osborne LJ, Tonissen KF, Tang VH, Clarke FM. Expression and localisation of thioredoxin in mouse reproductive tissues during the oestrous cycle. Mol Reprod Dev 2001; 58:359-67. [PMID: 11241771 DOI: 10.1002/1098-2795(20010401)58:4<359::aid-mrd2>3.0.co;2-k] [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: 11/09/2022]
Abstract
Thioredoxin expression within the reproductive tissues of the female mouse was analysed during the oestrous cycle stages of dioestrus, oestrus and metoestrus by Western blot analyses and immunocytochemistry. From Western blot analyses the expression of thioredoxin was found to be increased in oestrus compared to dioestrus and metoestrus. Localisation of thioredoxin within the reproductive organs of the mouse during the oestrous cycle has shown that the expression of thioredoxin is specific for distinct areas within the reproductive organs. These areas are the stratified squamous epithelium of the vagina, the simple columnar epithelium and the uterine glands of the uterus, the ciliated columnar epithelium of the oviduct, the corpus lutea, the interstitial cells and the secondary follicles of the ovary. The discrete cellular localisation and oestrous dependence of thioredoxin expression are suggestive of specific roles in various reproductive processes.
Collapse
Affiliation(s)
- L J Osborne
- School of Biomolecular and Biomedical Science, Faculty of Science, Griffith University, Nathan, Qld, Australia
| | | | | | | |
Collapse
|
344
|
Yamashita S. Histochemistry and cytochemistry of nuclear receptors. PROGRESS IN HISTOCHEMISTRY AND CYTOCHEMISTRY 2001; 36:91-176. [PMID: 11213555 DOI: 10.1016/s0079-6336(01)80004-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Receptors of steroid hormones, thyroid hormones and several kinds of vitamins have been shown to act as nuclear transcription factors and to form a nuclear receptor (NR) family. Histochemical techniques including autoradiography using radio-labeled ligands, immunohistochemistry and in situ hybridization histochemistry, have displayed that target cells of these receptors are distributed not only in the classical target organs but also widely in a variety of tissues; these techniques can demonstrate the presence of receptor proteins and mRNAs, even though they are expressed in a small cell population of tissues. On the other hand, many studies have been performed to demonstrate the interaction between NRs and nuclear and cytoplasmic proteins, and to clarify the mechanism of transcriptional regulation through NRs in artificial conditions which are created in gene transfer experiments or under cell-free conditions. Some data coincide with those obtained from histochemical techniques, however, some histochemical data do not support the results of studies in vitro. This review focuses on the following topics: histochemical methodologies to detect NRs, the distribution and function of NRs in the tissues, the intracellular and intranuclear localization of NRs, roles of gonadal steroid receptors and their ligands on developing tissues including cell communications such as mesenchymal-stromal interaction, and the interaction between other cellular components and NRs. In addition, the agreement and disagreement between the results of histochemical studies and those from the experiments in the model systems or in vitro are discussed.
Collapse
MESH Headings
- Animals
- Female
- Green Fluorescent Proteins
- Histocytochemistry
- Humans
- Immunohistochemistry
- Luminescent Proteins
- Male
- Microscopy, Immunoelectron
- Receptors, Androgen/analysis
- Receptors, Androgen/physiology
- Receptors, Cytoplasmic and Nuclear/analysis
- Receptors, Cytoplasmic and Nuclear/physiology
- Receptors, Estrogen/analysis
- Receptors, Estrogen/physiology
- Receptors, Glucocorticoid/analysis
- Receptors, Glucocorticoid/physiology
- Receptors, Progesterone/analysis
- Receptors, Progesterone/physiology
- Receptors, Steroid/analysis
- Receptors, Steroid/physiology
Collapse
Affiliation(s)
- S Yamashita
- Keio Junior College of Nursing, 35-Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| |
Collapse
|
345
|
Pierro E, Minici F, Alesiani O, Miceli F, Proto C, Screpanti I, Mancuso S, Lanzone A. Stromal-epithelial interactions modulate estrogen responsiveness in normal human endometrium. Biol Reprod 2001; 64:831-8. [PMID: 11207198 DOI: 10.1095/biolreprod64.3.831] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The coculture of endometrial epithelial cells (EEC) with stromal cells (ESC) allows achievement of an improved in vitro system for studying interactions between cells via soluble signals. The purpose of this study was to investigate whether 17beta-estradiol and insulin can induce proliferation of EEC through ESC-secreted factors. No evidence of estrogen-induced EEC proliferation has been reported so far in the conventional culture methods. To this end, we used an in vitro bicameral coculture model where human EEC were grown on extracellular matrix-coated inserts applied in dishes containing ESC. Proliferation was assessed by tritiated thymidine incorporation. Homogeneity of endometrial cell populations was ascertained immunocytochemically. 17beta-estradiol did not induce any proliferative effect on EEC cultured alone. Endometrial epithelial cell proliferation was significantly enhanced in EEC/ESC cocultures; moreover, it was further increased by 17beta-estradiol addition. Insulin increased proliferation in EEC cultured alone, but again the effect was more pronounced in EEC/ESC cocultures. Coincubation of 17beta-estradiol and an antibody against insulin-like growth factor I (IGF I) led to neutralization of ESC-mediated EEC proliferation. This work provides evidence that the effect of 17beta-estradiol on human EEC proliferation may be mediated at least in part through ESC-secreted IGF I. We also showed that insulin effect is also partially due to ESC activation.
Collapse
Affiliation(s)
- E Pierro
- Unità Operativa di Ginecologia ed Ostetricia, Ospedale G.B. Grassi, Roma 00121, Italy
| | | | | | | | | | | | | | | |
Collapse
|
346
|
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: 137] [Impact Index Per Article: 5.7] [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
|
347
|
Risbridger G, Wang H, Young P, Kurita T, Wang YZ, Lubahn D, Gustafsson JA, Cunha G, Wong YZ. Evidence that epithelial and mesenchymal estrogen receptor-alpha mediates effects of estrogen on prostatic epithelium. Dev Biol 2001; 229:432-42. [PMID: 11150243 DOI: 10.1006/dbio.2000.9994] [Citation(s) in RCA: 128] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In combination with androgens, estrogens can induce aberrant growth and malignancy of the prostate gland. Estrogen action is mediated through two receptor subtypes: estrogen receptors alpha (ERalpha) and beta (ERbeta). Wild-type (wt) and transgenic mice lacking a functional ERalpha (alphaERKO) or ERbeta (betaERKO) were treated with the synthetic estrogen diethylstilbestrol (DES). DES induced prostatic squamous metaplasia (SQM) in wt and betaERKO but not in alphaERKO mice, indicating an essential role for ERalpha, but not ERbeta, in the induction of SQM of prostatic epithelium. In order to determine the respective roles of epithelial and stromal ERalpha in this response, the following tissue recombinants were constructed with prostatic epithelia (E) and stroma (S) from wt and ERKO mice: wt-S+wt-E, alphaERKO-S+alphaERKO-E, wt-S+alphaERKO-E, and alphaERKO-S+wt-E. A metaplastic response to DES was observed in wt-S+wt-E tissue recombinants. This response to DES involved multilayering of basal epithelial cells, expression of cytokeratin 10, and up-regulation of the progesterone receptor. Tissue recombinants containing alphaERKO-E and/or -S (alphaERKO-S+alphaERKO-E, wt-S+alphaERKO-E, and alphaERKO-S+wt-E) failed to respond to DES. Therefore, full and uniform epithelial SQM requires ERalpha in the epithelium and stroma. These results provide a novel insight into the cell-cell interactions mediating estrogen action in the prostate via ERalpha.
Collapse
Affiliation(s)
- G Risbridger
- Institute of Reproduction & Development, Monash University, Melbourne, Victoria, 3168, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
348
|
Bai W, Oliveros-Saunders B, Wang Q, Acevedo-Duncan ME, Nicosia SV. Estrogen stimulation of ovarian surface epithelial cell proliferation. In Vitro Cell Dev Biol Anim 2000; 36:657-66. [PMID: 11229598 DOI: 10.1290/1071-2690(2000)036<0657:esoose>2.0.co;2] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Ovarian cancer is the leading cause of gynecological cancer mortality, and 85-90% of this malignancy originates from the ovarian surface epithelium (OSE). The etiology of ovarian epithelial cancer is unknown, but a role for estrogens has been suspected. However, the effect of estrogens on OSE cell proliferation remains to be determined. Using the rabbit model, our studies have demonstrated that 17beta-estradiol stimulates OSE cell proliferation and the formation of a papillary ovarian surface morphology similar to that seen in human ovarian serous neoplasms of low malignant potential. Immunohistochemical staining of ovarian tissue sections with an antibody to the estrogen receptor alpha demonstrates its expression in both OSE cells and stromal interstitial cells. In primary ovarian cell cultures, the proliferative response of the epithelial cells to 17beta-estradiol depends on the expression of the estrogen receptor alpha in the epithelial cells. However, when the epithelial cells are grown together with ovarian stromal cells, their proliferative response to this hormone is greatly enhanced, suggesting the involvement of stromal-epithelial interactions. These studies suggest a role for estrogens and the estrogen receptor alpha in OSE growth.
Collapse
Affiliation(s)
- W Bai
- Department of Pathology, University of South Florida College of Medicine and H. Lee Moffitt Cancer Center and Research Institute, Tampa 33612-4799, USA
| | | | | | | | | |
Collapse
|
349
|
Abstract
Reproductive tissues respond to steroid hormones and thus are particularly vulnerable to the effects of exogenous steroid 'mimic' compounds (endocrine disrupters). One such endocrine disrupter, diethylstilbestrol (DES), is linked to gynecological cancers and changes in uterine structure that reduce or completely abrogate reproductive competence. Until recently, little was known about the identity of target genes and signaling pathways involved in pathologies linked to endocrine disrupters such as DES. We outline genetic, cellular and molecular roles for patterning genes, with emphasis on homeobox and Wnt genes. There is evidence that changes in the expression of Wnt and homeogenes underlie many of the defects induced by DES. Data obtained from murine systems will likely apply to a broad spectrum of gynecological pathologies involving abnormal cell behaviors ranging from fibroids to malignant tumors. Knowledge garnered from modern molecular genetics should lead to progress in the emerging field of molecular gynecology.
Collapse
Affiliation(s)
- J Kitajewski
- Department of Pathology and OB/GYN, Columbia University, New York, New York, USA
| | | |
Collapse
|
350
|
Zhao L, Samuel CS, Tregear GW, Beck F, Wintour EM. Collagen studies in late pregnant relaxin null mice. Biol Reprod 2000; 63:697-703. [PMID: 10952910 DOI: 10.1095/biolreprod63.3.697] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The relaxin knockout (rlx -/-) mouse was used to assess the effect, during pregnancy, of relaxin with regard to water, collagen content, growth, and morphology of the nipple (N), vagina (V), uterus, cervix (C), pubic symphysis (PS), and mammary gland (MG). The results presented here indicate that during pregnancy, relaxin increases the growth of the N, C, V, and PS. Large increases in water content in the PS (20%) occurred in pregnant (Day 18.5) wild-type (rlx +/+) mice but not in rlx -/- animals. This indicates that in the PS, relaxin might increase the concentration of a water-retaining extracellular matrix component (hyaluronate). In the pregnant rlx +/+ mouse, collagen content decreased significantly in the N and V but not in other tissues. There were no significant changes in the rlx -/- mouse. This contrasts with findings in the rat, in which relaxin has been found to cause decreases in collagen concentrations in the V, C, and PS. Histological analysis showed that the collagen stain was more condensed in the tissues (V, C, PS, N, and MG) of rlx -/- mice than in those of rlx +/+ mice. This phenomenon indicates that the failure of collagen degradation and lack of growth in the N underlie the inability of the rlx -/- mice to feed their young, as reported previously. Vaginal and cervical luminal epithelia, which proliferated markedly in the rlx +/+ pregnant mice, remained relatively atrophic in the rlx -/- mice. As proliferation and differentiation of uterine and vaginal epithelia are thought to be induced by a paracrine stromal factor that acts upon estrogen stimulation, our results indicate that relaxin may be this paracrine factor.
Collapse
Affiliation(s)
- L Zhao
- Howard Florey Institute of Experimental Physiology and Medicine, The University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | | | | | |
Collapse
|