首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
The human breast cancer cell line MCF-7 has been adapted to long-term growth at 0.5 and 0.05% fetal bovine serum. Free cytoplasmic and filled nuclear estrogen receptors were found in cells grown at 5, 0.5, and 0.05% fetal bovine serum. Cells grown in medium with 0.05% dextran-charcoal-treated serum contained cytoplasmic receptors but no filled nuclear receptors, indicating that this medium did not contain biologically active concentrations of estrogen. Addition of estradiol to the medium translocated the cytoplasmic receptor to the nucleus and stimulated progesterone receptor synthesis but did not increase the growth rate. The antiestrogen tamoxifen (TAM) inhibited cell growth at all serum concentrations investigated, at least in part by a reduction of the growth fraction. The sensitivity to TAM was highest at low serum concentrations. The effect of TAM could be reversed by estradiol at TAM concentrations of 10(-6) M or lower. It is concluded that estradiol does not have a direct growth-stimulatory effect on our MCF-7 cells in monolayer cultures although the cells contain functional estrogen receptors and growth of the cells in athymic mice requires estrogens. TAM has an estrogen-competitive, inhibitory effect on the growth of MCF-7 cells at concentrations lower than 10(-6) M. At higher concentrations, the growth inhibition is unspecific and noncompetitive by estradiol.  相似文献   

2.
We have examined the properties of the estrogen receptor and progesterone receptor in MDA-MB-134 human breast cells and have evaluated the effects of estrogen on cell proliferation and progesterone receptor levels in these cells as indices of hormonal sensitivity. These cells contain high levels of estrogen receptor (approximately 1.5 pmol/mg DNA) and low levels of progesterone receptor (0.15 pmol/mg DNA). More than 80% of the estrogen receptor is found in the nuclear fraction in the absence of estrogen, and the Kd of the receptor for estradiol is approximately 1.5 X 10(-10) M. Upon exposure to estradiol, the receptors become occupied, but there is no processing or apparent decrease in either nuclear or total cellular estrogen receptor content, as can be seen in MCF-7 human breast cancer cells. The nuclear estrogen receptor sediments as a 4.6 S species on high salt sucrose gradients, and it can be detected on sodium dodecyl sulfate-polyacrylamide gel immunoblot analysis as a species of molecular weight 65,000, identical to that of the MCF-7 estrogen receptor, using the monoclonal antibodies D75P3 gamma and H222Sp gamma prepared against the MCF-7 estrogen receptor. The estrogen receptor shows binding selectivity for estrogens and antiestrogens, and its affinity for ligands follows the order diethylstilbestrol (190%) greater than estradiol (100%) greater than estriol (13%) greater than tamoxifen (3%), as expected for estrogen receptor. Hence the receptor appears normal in many of its physicochemical properties and in terms of its binding affinity and specificity for estrogens and antiestrogens. Control cells contain low levels of progesterone receptor that display high affinity (Kd = 6 X 10(-9) M) for the synthetic progestin R5020, but exposure to estradiol (10(-11)-10(-7)M) fails to increase cellular progesterone receptor levels. In contrast, estradiol markedly stimulates the rate of cell proliferation, while tamoxifen suppresses the growth of control and of estradiol treated cells. Hence, our data show that these cells, which contain substantial levels of estrogen receptor, respond to estrogen with enhanced cell proliferation but fail to have their progesterone receptor level modulated by estradiol. These cells represent an interesting and unusual situation in which estrogenic regulation of proliferation and the stimulation of progesterone receptor are dissociated. These cells should prove useful in further evaluation of estrogenic regulation of cell proliferation and specific protein synthesis in human breast cancer.  相似文献   

3.
Although antiestrogen therapy is effective in the treatment of hormone-responsive breast tumors, approximately 40% of the patients with estrogen receptor-positive tumors fail to respond to antiestrogens. To better understand the mechanisms by which antiestrogens inhibit the growth of hormone-dependent breast cancers, we have investigated the physicochemical properties and binding characteristics of the estrogen receptors with estradiol and antiestrogens and the occurrence of estrogen-noncompetible antiestrogen binding sites in two estrogen-sensitive but tamoxifen-growth-resistant estrogen receptor-positive MCF-7 cell variant clones, R3-98 and R27. In the variant cells, estradiol (10(-8) M) significantly stimulates cell proliferation as in the parent MCF-7 cells, but the antiestrogen tamoxifen (10(-6) M) has no significant effect on growth of the variant cells, whereas antiestrogen strongly inhibits proliferation of the parent MCF-7 cells. All three cell types contain high concentrations of estrogen receptor (150 to 250 fmol/mg protein), and competition binding analysis shows that the relative binding affinity of a series of compounds for estrogen receptor is similar among the three cell types with the affinity of trans-hydroxytamoxifen greater than estradiol greater than alpha-[4-pyrrolidinoethoxy]phenyl-4-hydroxy-alpha'-nitrostilben e greater than tamoxifen. Salt-extracted nuclear receptor complexes prepared from the three cell types showed similar sedimentation behavior on 0.4 M KCl-containing sucrose gradients with [3H]estradiol-labeled receptor complexes sedimenting at 4.2S, whereas receptors complexed with either of the antiestrogens trans-[3H]-hydroxytamoxifen or [3H]alpha-[4-pyrrolidinoethoxy]phenyl-4-hydroxy-alpha'-nitrosti lbene sediment at 5.5S. In all 3 cell types, the nuclear receptor forms react with an estrogen receptor monoclonal antibody, D547Sp gamma, to form complexes which sediment at 8.5S. The nuclear estrogen receptors from the parental MCF-7 and the two variant cells, when covalently labeled with [3H]-tamoxifen aziridine in intact cells and then salt extracted have identical molecular weights of approximately 62,000, as determined by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. The covalently labeled nuclear and cytosol receptors in these 3 cell lines also show identical migration in 8 M urea polyacrylamide isoelectric focusing gels consistent with a predominant receptor species of isoelectric point approximately 5.7.(ABSTRACT TRUNCATED AT 400 WORDS)  相似文献   

4.
A substantial proportion of human breast cancers contain estrogen receptors, and it is believed that the growth of some of these tumors and their synthesis of specific proteins are stimulated by estrogens. Since natural estrogens, such as estradiol, react reversibly with estrogen receptors, it was of interest to determine the biological consequences that would result from very strong, possibly irreversible interaction of an estradiol-based ligand with the estrogen receptor of breast cancer cells. For these studies, we have examined the receptor interactions and biological character of 11 beta- chloromethylestradiol (CME) and 11 beta- bromomethylestradiol (BME) as potential estradiol-based affinity labeling ligands in MCF-7 human breast cancer cells which contain high levels of estrogen receptors. The apparent relative binding affinities of CME and BME for MCF-7 estrogen receptor measured by competitive binding assay are 230 and 15%, respectively, whereas the affinity of estradiol is considered 100%. Incubation of receptor preparations from MCF-7 cells or rat uteri with CME at 21 degrees results in a concentration- and time-dependent decrease in receptor content measured by exchange assays with [3H]estradiol. This may be due to covalent attachment of CME to receptor and is termed "inactivation." Inactivation of 80 to 85% of the receptors occurs within 30 min at 21 degrees by exposure to 5 or 20 nM CME, with 2 nM giving 20 to 40% inactivation. This receptor inactivation is prevented by preincubation with 2000 nM estradiol, indicating that the interaction of CME is occurring at the estradiol binding site on the receptor. MCF-7 cells incubated with 20 nM CME show a rapid loss of cytosol receptor sites and no accumulation of receptors detectable by exchange assay in the nucleus, while 20 nM estradiol shows nuclear localization of receptor. BME, in contrast, inactivates only a portion (approximately 40%) of estrogen receptors. CME and BME both behave as estrogen agonists. They stimulate the proliferation of MCF-7 cells and increase cellular progesterone receptor content and plasminogen activator activity. CME is at least as potent as estradiol on a molar basis in increasing all of these activities, while BME shows a biopotency of only 1% of that of estradiol or CME. Hence, although CME reacts very strongly and apparently irreversibly with estrogen receptors in MCF-7 cells, it still behaves as a potent estrogen agonist.  相似文献   

5.
We have examined the effect of short-term and long-term growth in the absence of estrogens on the proliferation rate and estrogen and antiestrogen responsiveness of MCF-7 human breast cancer cells. The removal of phenol red, the pH indicator in tissue culture medium that is weakly estrogenic (Y. Berthois et al., Proc. Natl. Acad. Sci. USA, 83:2496-2500, 1986), immediately slows the cell proliferation rate, and MCF-7 cells grown in phenol red-free medium with charcoal dextran-treated serum for periods up to 1 mo maintain this reduced rate of cell proliferation. In these short-term phenol red-withdrawn cells, estradiol stimulates proliferation markedly and reproducibly, and antiestrogens inhibit estrogen-stimulated proliferation. Antiestrogens by themselves appear as partial agonists/antagonists; at low concentrations they stimulate proliferation weakly, but they show no stimulation at the high concentrations where they fully inhibit estrogen-stimulated proliferation. In contrast to the short-term phenol red-withdrawn cells, cells maintained for several months (5 to 6 mo) in the apparently complete absence of estrogens (no phenol red, with charcoal dextran-treated calf serum) show a markedly increased basal rate of cell proliferation; estradiol is unable to increase this rate of proliferation further, but antiestrogens are able to decrease proliferation. This change in growth pattern is associated with a 3-fold increase in cellular estrogen receptor levels. Despite their differing basal growth rates, cells grown in either the short-term (less than 1 mo) or long-term (greater than 6 mo) absence of estrogens both have progesterone receptor levels that are very low and, in both cases, estradiol increases progesterone receptor levels markedly. Thus, under long-term estrogen-free conditions, there is a dissociation between the stimulation of cell proliferation and of specific protein synthesis (progesterone receptor) by estrogen. The increase in the cell proliferation rate observed in cells grown in the long-term absence of estrogen may reflect altered regulation of growth factor production or altered sensitivity to growth factors in the medium or produced by the cells themselves. Hence, these breast cancer cells adapt significantly to long-term growth in estrogen-free conditions, an observation that may be relevant to understanding the growth of hormone-responsive human breast cancers in vivo.  相似文献   

6.
Androstene-3beta, 17alpha-diol (17alpha-AED) inhibits DNA synthesis and induces apoptosis in several myeloid cancer cell lines. The purpose of this study was to determine if 17alpha-AED inhibition of human breast carcinoma cell proliferation is dependent on the estrogen or androgen receptor. At concentrations of 12.5 to 50 x 10(-9) M 17alpha-AED inhibited the proliferation of ZR75-1, estrogen receptor-positive (ER+) cells, by 54% to 68%. Further, 17alpha-AED inhibited MDA-MB231, estrogen receptor-negative (ER-) cells, by 33.6% to 56.0%. The inhibitory effect was dose dependent with a minimal effective inhibitory dose at 12.5x10(-9) M for both cell lines. Both 17beta-AED and estradiol potentiate the inhibitory effect of 17alpha-AED on ER+ cells at lower doses (3.13 to 6.25 x 10(-9) M) where 17alpha-AED alone was not inhibitory. The inhibitory action of 17alpha-AED on human mammary carcinomas appears to be independent of either the alpha estrogen or the androgen receptors.  相似文献   

7.
Summary Antiestrogens have proven to be effective in controlling the growth of hormone-responsive breast cancers. At the concentrations of antiestrogens achieved in the blood of breast cancer patients taking antiestrogens (up to 2 × 10–6 M), antiestrogens selectively inhibit the proliferation of estrogen receptor-containing breast cancer cells, and this inhibition is reversible by estradiol. Antiestrogens also inhibit estrogen-stimulation of several specific protein synthetic activities in breast cancer cells, including increases in plasminogen activator activity, progesterone receptor levels and production of several secreted glycoproteins and intracellular proteins.Antiestrogens bind with high affinity to the estrogen receptor and to additional microsomal binding sites to which estrogens do not bind. These latter sites, called antiestrogen binding sites (AEBS), are present in equal concentrations in estrogen receptor-positive and -negative breast cancer cells and are present in a wide variety of tissues, with highest concentrations being found in the liver. The antiestrogenic and growth suppressive potencies of a variety of antiestrogens correlate best with their affinity for estrogen receptor and not with affinity for AEBS.Antiestrogens undergo bioactivation and metabolismin vivo and hydroxylated forms of the antiestrogen have markedly enhanced affinities for the estrogen receptor. Detailed studies with high affinity radiolabelled antiestrogens indicate that antiestrogens induce important conformational changes in receptor that are reflected in the enhanced maintenance of a 5 S form of the estrogen receptor complex; reduced interaction with DNA; and altered activation and dissociation kinetics of the antiestrogen-estrogen receptor complex. These conformational changes effected by antiestrogens likely result in different interactions with chromatin, causing altered cell proliferation and protein synthesis.Analyses of the rates of synthesis and turnover of the estrogen receptor through pulse-chase experiments utilizing the covalently attaching antiestrogen, tamoxifen aziridine, and studies employing dense amino acid labeling of estrogen receptor reveal that the antiestrogen-occupied receptor is degraded at a rate (t 1/2 = 4 h) similar to that of the control unoccupied receptor. Hence, antiestrogens do not prevent estrogen receptor synthesis and they do not either accelerate or block estrogen receptor degradation.Our findings raise serious doubts about the role of the AEBS in mediating directly the growth suppressive actions of antiestrogens, and suggest that interaction with the estrogen receptor is most likely the mechanism underlying the growth-inhibitory effects of antiestrogens. At present, the role of the AEBS in the actions of antiestrogens or in possible antiestrogen metabolism remains unclear.  相似文献   

8.
9.
Human mammary carcinoma cell lines (MCF-7) were analysed for their hormone sensitivity before and after transfection with a v-Ha-ras oncogene or with a neomycin-resistance gene followed by selection in vitro or in vivo. Our aim was to test how the expression of the ras oncogene would influence the estradiol sensitivity of MCF-7 cells. In culture, MCF-7 cells expressing the viral p21 oncogene product, as compared to parental MCF-7 cells and their control derivatives, showed lower levels of a 67-kDa estrogen receptor. Progesterone receptors, however, remained sensitive to up-regulation by estrogens. The oncogene-expressing cells were less sensitive than all controls to stimulation of proliferation by 10(-8)M estradiol or to inhibition of proliferation by 2-CH3-4-OH tamoxifen, and this was not dependent upon the type of culture medium used. After s.c. or i.p. injection into female athymic nude mice, ovariectomized or left intact, the growth of MCF-7 cells expressing the ras oncogene product and of all control cells was sensitive to stimulation by estrogen supplementation. Conversely, cell lines derived from tumors generated with long latency in untreated athymic nude mice by v-ras-expressing MCF-7 cells showed efficient formation of quickly growing tumors in the absence of estrogen supplementation. No differences were observed in invasion and metastasis of the different MCF-7 cell types injected into athymic nude mice that were supplemented with estrogens or not.  相似文献   

10.
Dehydroepiandrosterone, an adrenal hormone derived from cholesterol, can be metabolized to estrogens (estradiol) and androgens (testosterone). In this study, we evaluated whether the antiproliferative effect induced by dehydroepiandrosterone in MCF-7 cells (an estrogen-dependent breast cancer cell line) is direct, or indirect, through its conversion to estradiol or testosterone. Although dehydroepiandrosterone had an antiproliferative effect at supraphysiologic concentrations, when it was used at physiologic concentrations, it increased the proliferation of MCF-7 cells. 17Beta-estradiol induced an increase in MCF-7 cell proliferation at physiologic concentrations, whereas testosterone had a weak inhibitory effect at 100 microM. Dehydroepiandrosterone sulfate (its inactive sulfate ester) had no effect upon the cell cycle. Dehydroepiandrosterone-induced antiproliferative and proliferative effects were not blocked by inhibitors of androgen or estrogen receptors, thus indicating that its effect is secondary to a direct interaction with a "putative" receptor rather than a conversion into steroid hormones. These results suggest that dehydroepiandrosterone could be used at supraphysiologic concentrations in the treatment of breast cancer.  相似文献   

11.
The human breast tumor cloned cell lines T47D-A8 and All are estrogen dependent for cell proliferation in the nude mouse model. In contrast, these cells multiplied at similar rates when grown in serum-free cultures, regardless of the presence of 17 beta-estradiol (3 X 10(-11) to 3 X 10(-8) M estradiol). Addition of 10% charcoal-dextran stripped human female serum to the culture medium resulted in a marked inhibition of cell proliferation. The addition of 3 X 10(-11) M estradiol overcame the inhibitory effect of serum. Similar results were obtained with the human breast tumor C7MCF7 cell line. Both cell lines contain similar estrophilin levels. The Kd of the estrophilin-estradiol complex was 0.39 X 10(-10) M for C7MCF7 cells and 4.4 X 10(-10) M for T47D-A11 cells. Maximal cell yields were achieved at 5 X 10(-12) M free estradiol levels in 10% charcoal-dextran stripped serum supplemented medium. These data are compatible with the following interpretation: (a) estradiol-sensitive cells are inhibited from proliferating by a serum-borne factor; and (b) estradiol neutralizes this inhibitory effect. This mechanism seems not to be mediated by estradiol binding to the cellular estrophilins because (a) the free estradiol levels needed for maximal response are significantly lower than the estrophilin Kds, and (b) maximal proliferation rates occur at similar estradiol concentrations for these three cell lines, regardless of the binding properties of their estrophilins.  相似文献   

12.
In this report, we have studied the effect of epidermal growth factor (EGF) on the proliferation of breast cancer cell lines, as well as the modulation of estrogen -and epidermal growth factor - receptor levels by EGF treatment. We have observed that all the cell lines analysed were stimulated by EGF in low serum containing media. The MCF-7AZ cell line, its H-ras transfected MCF-7AZ TD5 variant and the MCF-7 cells, all of them containing a relatively low number of epidermal growth factor receptors, were growth stimulated in a dose-dependent manner by 10(-9) M to 10(-8) M EGF. The MDA-MB 231, A431 and BT20 cell lines that express higher receptor levels were stimulated with relatively low concentrations of epidermal growth factor (5 x 10(-13) M to 10(-11) M). However, A431 and BT20 cells were shown to be growth-inhibited in the presence of higher EGF concentrations (10(-10) M to 10(-8) M). We also observed that EGF down-regulated epidermal growth factor receptor while it up-regulated estrogen receptor. In addition, Scatchard analysis of radiolabeled EGF binding on cell surface demonstrated that the concentrations of growth factor necessary to occupy a given number of epidermal growth factor receptors are inversely correlated with the total level of these receptors. Our findings suggest that the mitogenic effect of epidermal growth factor on cell proliferation is a function of the quantity of EGF-occupied receptors.  相似文献   

13.
Alkaline phosphatase activity in human endometrial cancer cells of the estrogen-responsive Ishikawa line was markedly stimulated (3-20-fold in 4 days) by estrogens, 5 alpha-dihydrotestosterone, and dehydroepiandrosterone but not by testosterone, medroxyprogesterone acetate, glucocorticoids, several peptide hormones, prostaglandins, or growth factors. Maximum responses to estradiol were obtained at concentrations between 10(-9) and 10(-7) M; at 10(-8) M estradiol, the highest activity was reached 48-72 h after addition of the hormone. A linear relationship between enzyme activity at 48 h and the length of exposure to the hormone was observed. Dibutyryl cyclic guanosine 3':5'-monophosphate, but not dibutyryl cyclic adenosine 3':5'-monophosphate enhanced alkaline phosphatase activity and acted synergistically with estradiol. trans-4-Monohydroxytamoxifen completely antagonized the stimulatory effect of estradiol and had no agonistic activity. Dihydrotestosterone and dehydroepiandrosterone appear to exert their effects, at least in part, by interacting with estrogen receptors, since the simultaneous presence in the medium of monohydroxytamoxifen abolished their influence on alkaline phosphatase activity. The specific antiandrogen monohydroxyflutamide partially antagonized the effect of these hormones, suggesting that their action involved androgenic mechanisms as well. Exposure to elevated temperature and to specific inhibitors identified alkaline phosphatase of Ishikawa cells as a placental-type isoenzyme, thus contrasting with the nonplacental type found in glandular epithelial cells of normal endometrium and in another human endometrial cancer cell line, HEC-50. This study extends our previous observations of estrogen responsiveness in the Ishikawa cell line. In addition to the previously reported stimulatory effects on growth and progesterone receptor levels, we are now describing the stimulation by estrogens and C19 steroids of an enzyme, alkaline phosphatase, which can be used as a convenient end point to examine mechanisms of hormonal action.  相似文献   

14.
Summary The estrogen receptor that is present in many breast tumors provides a mechanism for the concentration of estrogens. Hence, it should be possible to effect a selective, receptor-mediated killing of estrogen receptor-rich tumor cells with a suitable cytotoxic estrogen derivative. Because the dose per cell that can be delivered by a receptor-mediated process is limited by the capacity of the estrogen receptor system (ca. 1,000–10,000 molecules per cell), the cytotoxic moiety of these derivatives needs to be directed at a target where an effective cell kill can be achieved with this limited dose; DNA appears to be the most suitable target. The estrogen derivatives that contain DNA-alkylating and cross-linking groups and retain high affinity for the estrogen receptor should be carried selectively to the nucleus. There, it is hoped that they would react with DNA with reasonable efficiency. Definitive tests of receptor-mediated cytotoxicity in vitro could be performed in cultures of human breast tumor cells that are receptor positive (e.g., MCF-7) or receptor negative (e.g., MDA-MB-231). These tests would involve comparison of the potency of the cytotoxic estrogens with that of cytotoxic control compounds, both in the presence and absence of estradiol, in order to correct for possible differences in the sensitivity of the different cell lines to cytotoxic agents and to metabolic effects of estradiol. These agents would then be tested in experimental mammary tumor systems in the rat. Our current understanding of the characteristics of estrogen receptor binding, and the availability of suitable systems for studying these agents in vitro and in vivo, make a definitive test of selective cytotoxicity, mediated by the estrogen receptor, a timely and sensible endeavor.  相似文献   

15.
Effects of tamoxifen on telomerase activity in breast carcinoma cell lines   总被引:5,自引:0,他引:5  
Aldous WK  Marean AJ  DeHart MJ  Matej LA  Moore KH 《Cancer》1999,85(7):1523-1529
  相似文献   

16.
M Harris  T Zacharewski  S Safe 《Cancer research》1990,50(12):3579-3584
Treatment of MCF-7 human breast cancer cells with 17 beta-[3H]estradiol resulted in a rapid accumulation of occupied nuclear estrogen receptor complex in which levels were maximized within 1 h and decreased after 3 h. Pretreatment of the cells with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) 6 or 12 h prior to the addition of the radiolabeled hormone resulted in a 38 and 63% reduction in the levels of the occupied nuclear estrogen receptor, respectively, whereas addition of TCDD 1 h prior to the radioligand did not cause any significant change in the levels of the occupied nuclear receptor using velocity sedimentation analysis. Moreover, it was also shown with estrogen receptor antibodies that the TCDD-mediated dose-dependent decrease in occupied nuclear receptor levels was paralleled by a comparable decrease in immunoreactive protein at concentrations of 10(-8) to 10(-11) M TCDD. The reduction in levels of the occupied nuclear estrogen receptor was not due to increased estradiol metabolism since a significant reduction was observed at TCDD concentrations (10(-11) to 10(-13) M) which do not induce cytochrome P-450-dependent monooxygenase enzyme activities in MCF-7 cells. Treatment of the MCF-7 cells with actinomycin D or cycloheximide resulted in a greater than 2-fold increase in levels of the occupied nuclear estrogen receptor, and cotreatment of the cells with both TCDD and these inhibitors significantly decreased levels of the nuclear estrogen receptor complex. The structure-activity relationships for TCDD and several congeners were similar for both the reduction of occupied nuclear estrogen receptor levels and several aryl hydrocarbon (Ah) receptor agonist activities, and these results support a role of the Ah receptor in these processes.  相似文献   

17.
Effects of estrogens on the cytoplasmic microtubule networkwere examined by the indirect immunofluorescence method usinganti-ß-tubulin antibody. Estradiol, a naturally occurringestrogen, decreased the amount of cytoplasmic microtubule fibersduring interphase in the human breast cancer cell lines MCF-7and MDA-MB-231. Since MDA-MB-231 is an estrogen receptor-negativecell line, estradiol-induced microtubule disruption seems tobe independent of estradiol binding to receptors. The effectiveconcentration of estradiol required for induction of microtubuledisruption in 50% of the cells (EC50) was 81 µM for MCF-7cells and 82 µM for MDA-MB-231 cells. A synthetic estrogen,diethylstilbestrol, also induced a decrease in microtubule fibers,with an EC50 value of 48 µM for MCF-7 cells and 50 µMfor MDA-MB-231 cells. When estrogentreated and microtubule-depolymerizedcells were washed and the medium was replaced with fresh, intracellularmicrotubule networks reappeared within 3 h. When MCF-7 cellswere cultured for 4 days with estradiol (50 µM), cellgrowth was completely inhibited. However, estrone affected themicrotubule network and cell proliferation only slightly. Theseresults suggest that estradiol-induced microtubule disruptionis closely related to its inhibitory effect on cell growth.  相似文献   

18.
The effects of estradiol and two synthetic progestins (ORG2058 and R5020) on the expression of the high-affinity, metastasis-associated laminin receptor in two human breast carcinoma cell lines were examined. The T47D cell line contains estrogen and progesterone receptors, but the MDA-MB 231 cell line lacks both receptors. Treatment of T47D cells with 10(-9) M estradiol alone results in a three-fold increase (P less than or equal to .05) in the steady-state level of laminin receptor mRNA determined by RNA blot analysis as well as in cell-surface, laminin receptor expression that is evaluated by immunofluorescence. No effects of estradiol on the receptor-negative MDA-MB 231 cells were observed. Untreated and steroid-treated MDA-MB 231 cells had higher levels of laminin receptor mRNA than did untreated or estradiol-treated T47D cells. A more dramatic increase (five-fold; P less than or equal to .005) of mRNA and cell-surface expression in T47D cells was observed after treatment with estradiol plus 10(-8) M progestin or with progestin alone. Estradiol treatment also increased chemotaxis and haptotaxis of T47D cells but not of MDA-MB 231 cells to laminin; it had no effect on the attachment of these latter cells to laminin. Interestingly, treatment with estradiol plus progestin or progestin alone significantly increased the attachment of T47D cells to laminin but did not have an effect on either haptotaxis or chemotaxis to laminin. These results suggest that the various cell-laminin interactions are mediated by different mechanisms. The augmentation of laminin receptor mRNA by estrogen and progesterone treatment in hormone receptor-positive cells, but not in cells that lack these receptors, may relate functionally to the difference in the clinical aggressiveness between classes of breast cancers.  相似文献   

19.
High dose estrogen therapy has been used effectively in the treatment of human breast cancer. To understand the mechanisms involved, the effects of high concentrations (5-100 microM) of estrogens were studied in estrogen receptor (ER) positive (T-47D and MCF-7) and ER negative (MDA-MB-330) human breast cancer cell lines in vitro. Inhibition of cellular proliferation was seen with the synthetic estrogen diethylstilbestrol (DES) at concentrations greater than 10 microM in each of the three cell lines. In T-47D cells DES was shown by clonogenic survival assays to be cytotoxic. This effect was evident in both plateau phase and exponentially growing cultures, in contrast to the effects of the antiestrogen tamoxifen, which has minimal effects on plateau phase cells. The effects of DES on the proliferation of exponentially growing cultures were accompanied by changes in cell cycle parameters which included an increase in the percentages of S-phase, G2 + M, and polyploid cells and a corresponding decrease in the percentage of G0-G1 cells. These changes, which contrasted with the known effects of tamoxifen, were not seen in the non- or slowly cycling plateau phase T-47D cells. Such results are consistent with two mechanisms of action of high dose estrogen in vitro: a cell cycle phase-specific effect and cell cycle-independent cytotoxicity. The stereoisomers 17 alpha-estradiol and 17 beta-estradiol had similar potency to DES in inhibiting cell proliferation and inducing these changes in cell cycle parameters in both MCF-7 and MDA-MB-330 cells. The high-dose estrogen effect was ligand specific in that estrone and estriol were less potent than DES, 17 alpha-estradiol and 17 beta-estradiol in inhibiting cell proliferation, and the characteristic cell cycle changes were produced only by concentrations of estriol greater than 75 microM and not at all by estrone at concentrations up to 100 microM. The androgens testosterone and dihydrotestosterone were similar in effect to estrone. The cell cycle changes associated with estrogen-induced growth inhibition in vitro are identical to those observed during regression of ER positive but not ER negative human tumor xenografts in nude mice. However, the role of ER in mediating estrogen-induced regression of ER positive tumors in vivo remains undefined.  相似文献   

20.
Development of resistance to hormonal therapy in breast cancer is frequently associated with a decline or loss of cellular estrogen receptors. Agents which up-regulate the receptor may reduce the incidence of hormonal resistance. Antiestrogens at concentrations ranging from 0.1 to 1 microM produced a 2- to 4-fold increase of estrogen receptors in MCF-7 and T-47D breast cancer cells. This increase, which occurred as early as 3 h and was sustained throughout the 4 days of continuous exposure to tamoxifen, was primarily due to an enhancement in receptor synthesis.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号