Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Radiat Res ; 194(5): 557-565, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33045089

ABSTRACT

Astronauts participating in prolonged space missions constitute a population of individuals who are at an increased risk for cataractogenesis due to exposure to densely ionizing charged particles. Using a rat model, we have previously shown that after irradiation of eyes with either low-linear energy transfer (LET) 60Co γ rays or high-LET 56Fe particles, the rate of progression of anterior and posterior subcapsular cataracts was significantly greater in ovariectomized females implanted with 17-ß-estradiol (E2) compared to ovariectomized or intact rats. However, our additional low-LET studies indicated that cataractogenesis may be a modifiable late effect, since we have shown that the modulation of cataractogenesis is dependent upon the timing of administration of E2. Interestingly, we found that E2 protected against cataractogenesis induced by low-LET radiation, but only if administered after the exposure; if administered prior to and after irradiation, for the entire period of observation, then E2 enhanced progression and incidence of cataracts. Since most radioprotectors tested to date are unsuccessful in protecting against the effects of high-LET radiation, we wished to determine whether the protection mediated by E2 against radiation cataractogenesis induced by low-LET radiation would also be observed after high-LET irradiation. Female 56-day-old Sprague-Dawley rats were treated with E2 at various times relative to the time of single-eye irradiation with 2 Gy of 56Fe ions. We found that administration of E2 before irradiation and throughout the lifetime of the rat enhanced cataractogenesis compared to ovariectomized animals. The enhancing effect was slightly reduced when estrogen was removed after irradiation. However, in contrast to what we observed after γ-ray irradiation, there was no inhibition of cataractogenesis if E2 was administered only after 56Fe-ion irradiation. We conclude that protection against cataractogenesis by estrogen is dependent upon the type and ionization density of radiation that the lens was exposed to. The lack of inhibition of radiation cataractogenesis in rats that receive E2 treatment after high-LET irradiation may be attributed to the qualitative differences in the types of DNA damage induced with high-LET radiation compared to low-LET radiation or how damage may be modified at the DNA or tissue level after irradiation.


Subject(s)
Cataract/prevention & control , Cobalt Radioisotopes , Estradiol/therapeutic use , Gamma Rays/adverse effects , Heavy Ions/adverse effects , Iron , Radiation Injuries, Experimental/prevention & control , Aerospace Medicine , Animals , Cataract/etiology , Drug Administration Schedule , Drug Implants , Estradiol/administration & dosage , Incidence , Linear Energy Transfer , Ovariectomy , Rats , Rats, Sprague-Dawley
2.
Biol Reprod ; 92(5): 122, 2015 May.
Article in English | MEDLINE | ID: mdl-25882702

ABSTRACT

Progesterone (P4) and the synthetic glucocorticoid dexamethasone (Dex) inhibit luminal epithelial (LE) proliferation in neonatal mouse uteri. This study determined the roles of progesterone receptor and estrogen receptor 1 (PR and ESR1, respectively) in P4- and Dex-induced inhibition of LE proliferation using PR knockout (PRKO) and Esr1 knockout (Esr1KO) mice. Wild-type (WT), heterozygous, and homozygous PRKO female pups were injected with vehicle, P4 (40 µg/g body weight), or Dex (4 or 40 µg/g body weight) on Postnatal Day 5, then 24 h later immunostained for markers of cell proliferation. In WT and heterozygous mice, P4 sharply reduced LE proliferation, and Dex produced dose-responsive decreases equaling those of P4 at the higher dose. Critically, although both doses of Dex similarly decreased proliferation compared to vehicle-treated PRKOs, treatment of PRKO pups with the high Dex dose (40 µg/g) did not inhibit LE as much as treatments of WT mice with this Dex dose or with P4. Stromal proliferation was stimulated by P4 in WT but not PRKO mice, and Dex did not alter stromal proliferation. Uteri of all genotypes strongly expressed glucocorticoid receptor (GR), demonstrating that impaired Dex effects in PRKOs did not reflect GR deficiency. Furthermore, inhibition of LE proliferation by Dex (40 µg/g body weight) in Esr1KO mice was normal, so this process does not involve ESR1. In summary, inhibitory Dex effects on LE proliferation occur partially through non-PR-mediated mechanisms, presumably GR, as indicated by Dex inhibition of LE proliferation in PRKOs. However, maximal inhibitory Dex effects on uterine LE proliferation are not seen in PRKO mice with even high Dex, indicating that maximal Dex effects in WT mice also involve PR.


Subject(s)
Dexamethasone/pharmacology , Epithelial Cells/drug effects , Progesterone/pharmacology , Receptors, Progesterone/metabolism , Uterus/physiology , Animals , Animals, Newborn , Epithelial Cells/physiology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation , Mice , Mice, Knockout , Receptors, Progesterone/genetics , Uterus/cytology , Uterus/drug effects
3.
Radiat Res ; 181(6): 578-83, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24811864

ABSTRACT

The detonation of an improvised nuclear device during a radiological terrorist attack could result in the exposure of thousands of civilians and first responders to lethal or potentially lethal doses of ionizing radiation (IR). There is a major effort in the United States to develop phamacological mitigators of radiation lethality that would be effective particularly if administered after irradiation. We show here that giving female C57BL/6 mice a subcutaneous surgical incision after whole body exposure to an LD50/30 X-ray dose protects against radiation lethality and increases survival from 50% to over 90% (P = 0.0001). The increase in survival, at least in part, appears to be due to enhanced recovery of hematopoiesis, notably red blood cells, neutrophils and platelets. While a definitive mechanism has yet to be elucidated, we propose that this approach may be used to identify potentially novel mechanisms and pathways that could aid in the development of novel pharmacological radiation countermeasures.


Subject(s)
Hematopoiesis/physiology , Radiation Injuries, Experimental/drug therapy , Whole-Body Irradiation , Animals , Blood Platelets/radiation effects , Erythrocytes/radiation effects , Female , Humans , Mice , Neutrophils/radiation effects , Radiation Injuries, Experimental/pathology , Survival Analysis
4.
Gynecol Oncol ; 131(1): 198-206, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23851291

ABSTRACT

OBJECTIVE: The objective of this study is to determine whether an altered DNA replication process is responsible for some of genetic damage observed in ovarian cancer. METHODS: The replication fidelity of the DNA synthetic process was evaluated in both malignant and non-malignant human ovarian cells. The types of replication errors produced were identified. In addition, kinetic analyses of the efficiency of ovarian cancer DNA polymerases for misincorporating nucleotides were performed. RESULTS: We report for the first time that ovarian cancer cells harbor an error promoting DNA replication apparatus which contributes to the decrease in DNA synthetic fidelity exhibited by these cells. Our study also shows that the decrease in DNA replication fidelity was not a result of an increased DNA replication activity. In addition, it was observed that the higher rate of DNA replication errors does not result in significant differences in the type of DNA replication-errors made during the DNA replication process; just the relative abundance. A detailed kinetic analysis of the efficiency of misincorporating nucleotides demonstrated that the DNA polymerases within the ovarian cancer cells exhibited a significant propensity for creating purine-pyrimidine nucleotide mismatches relative to non-malignant ovarian cells, while being only slightly more efficient at incorrectly pairing a purine nucleotide with a purine nucleotide. CONCLUSIONS: All together, these data suggest that the systematic analysis of the DNA replication process in ovarian cancer could uncover information on some of the molecular mechanisms that drive the accumulation of genetic damage, and probably contribute to the pathogenesis of the disease.


Subject(s)
Carcinoma/genetics , DNA Replication , DNA, Neoplasm/biosynthesis , DNA-Directed DNA Polymerase , Multienzyme Complexes , Mutation , Ovarian Neoplasms/genetics , Cell Line, Tumor , DNA-Directed DNA Polymerase/metabolism , Deoxyadenine Nucleotides/metabolism , Female , Humans , Kinetics , Lac Operon/genetics , Ovary/cytology
5.
Radiat Res ; 178(4): 260-5, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22880623

ABSTRACT

Age at the time of exposure to sparsely ionizing radiation has been established as a key determinant of radiation cataractogenesis. However, while some reports suggest that the lenses of the young are hypersensitive, data from older studies are often conflicting and somewhat difficult to interpret when the radioresponse of young lenses is compared to that of adult lenses. Moreover, the mechanism of the age-response function for radiation cataractogenesis has yet to be identified. Since steroid sex hormones, notably estradiol, appear to play a role in age-related cataractogenesis, we hypothesized that the age response for radiation cataractogenesis could be dictated by estradiol status. We recently showed that exposure to high-linear energy transfer (LET) radiation resulted in a reduced latent period for, and enhanced progression of cataracts in rats that were 1 year old at the time of exposure compared to those that were 56 days old. However, the enhanced sensitivity of older lenses compared to younger lenses was independent of estradiol status. In the current study, we found that for 1-year-old rats exposed to 10 Gy of low-LET (60)Co γ rays, the rate of increase in the development of posterior and anterior subcapsular cataracts was higher in older ovary-intact rats compared to young rats. However, cataracts were detected much earlier in ovary-intact 56-day-old rats compared to 1-year-old rats, regardless of their treatment groups (ovary-intact, ovariectomized, or ovariectomized and implanted with capsules containing estradiol). Thus, despite a consistent estradiol response (potentiating effect of estrogen) within a given age group, the differences between the radiation response of old and young lenses cannot be accounted for solely by estradiol status.


Subject(s)
Cataract/etiology , Estradiol/physiology , Radiation Injuries, Experimental/etiology , Age Factors , Animals , Estradiol/blood , Female , Linear Energy Transfer , Rats , Rats, Sprague-Dawley
6.
Carcinogenesis ; 32(8): 1162-6, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21606321

ABSTRACT

Mice treated neonatally with diethylnitrosamine (DEN) develop liver tumors in a male-dominant manner, reflecting the male bias in human hepatocellular carcinoma. Evidence suggests that estrogen, androgen, prolactin (PRL) and growth hormone (GH) modify liver tumorigenesis. We determined the roles of estrogen receptor-α (ERα) and prolactin receptor (PRLR) using receptor null mice, ERαKO (C57Bl/6J) and PRLR-KO (129Ola-X-C57BL/6), in the neonatal-DEN model of liver tumorigenesis. In both mouse strains, females had reduced tumorigenesis compared with males (P < 0.01), regardless of ERα or PRLR status. Tumorigenesis was not affected by ovariectomy in C57Bl/6J mice but it was increased by ovariectomy in the mixed strain, 129Ola-X-C57BL/6, regardless of PRLR status. ERαKO males had 47% fewer tumors than ERα wild-type males (P < 0.01). On the other hand, estradiol treatment protected against tumorigenesis in males only in the presence of ERα. As evidenced by liver gene expression, lack of ERα did not alter the pattern of GH secretion in males but resulted in the male GH pattern in females. These observations indicate that ERα is not required for lower tumorigenesis in females, but it is required for the protective effects of exogenously delivered estradiol. Unexpectedly, the results indicate that ERα plays a role in promotion of liver tumors in males. In addition, it can be concluded that sex differences in liver tumorigenesis cannot be explained by the sexually dimorphic pattern of GH secretion. The results also rule out PRL as the mediator of the protective effect of the ovaries.


Subject(s)
Diethylnitrosamine/toxicity , Estrogen Receptor alpha/physiology , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/prevention & control , Receptors, Prolactin/physiology , Animals , Estradiol/pharmacology , Female , Gene Expression/drug effects , Growth Hormone/metabolism , Humans , Liver Neoplasms, Experimental/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovariectomy , Prolactin/pharmacology
7.
Radiat Res ; 175(1): 37-43, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21175345

ABSTRACT

Astronauts participating in extended lunar missions or the projected mission to Mars would likely be exposed to significant doses of high-linear energy transfer (LET) heavy energetic charged (HZE) particles. Exposure to even relatively low doses of such space radiation may result in a reduced latent period for and an increased incidence of lens opacification. However, the determinants of cataractogenesis induced by densely ionizing radiation have not been clearly elucidated. In the current study, we show that age at the time of exposure is a key determinant of cataractogenesis in rats whose eyes have been exposed to 2 Gy of (56)Fe ions. The rate of progression of cataractogenesis was significantly greater in the irradiated eyes of 1-year-old rats compared to young (56-day-old) rats. Furthermore, older ovariectomized rats that received exogenous estrogen treatment (17-ß-estradiol) commencing 1 week prior to irradiation and continuing throughout the period of observation of up to approximately 600 days after irradiation showed an increased incidence of cataracts and faster progression of opacification compared to intact rats with endogenous estrogen or ovariectomized rats. The same potentiating effect (higher incidence, reduced latent period) was observed for irradiated eyes of young rats. Modulation of estrogen status in the 1-year-old animals (e.g., removal of estrogen by ovariectomy or continuous exposure to estrogen) did not increase the latent period or reduce the incidence to that of intact 56-day-old rats. Since the rapid onset and progression of cataracts in 1-year-old compared to 56-day-old rats was independent of estrogen status, we conclude that estrogen cannot account for the age-dependent differences in cataractogenesis induced by high-LET radiation.


Subject(s)
Cataract/etiology , Estrogens/physiology , Radiation Injuries, Experimental/etiology , Age Factors , Animals , Female , Humans , Linear Energy Transfer , Rats , Rats, Sprague-Dawley , Space Flight , Species Specificity
8.
Epigenetics ; 5(8): 716-29, 2010.
Article in English | MEDLINE | ID: mdl-20676026

ABSTRACT

"Epigenetic plasticity" refers to the capability of mammalian cells to alter their differentiation status via chromatin remodeling-associated alterations in gene expression. While epigenetic plasticity has been best associated with lineage commitment of embryonic stem cells, recent studies have demonstrated chromatin remodeling even in terminally differentiated normal cells, and advanced-stage melanoma and breast cancer cells, in context-dependent responses to alterations in their microenvironment. In the current study, we extend this attribute of epigenetic plasticity to aggressive ovarian cancer cells, by using an integrative approach to associate cellular phenotypes with chromatin modifications ("ChIP-chip") and mRNA and microRNA expression. While we identified numerous gene promoters possessing the well-known "bivalent mark" of H3K27me3/H3K4me2, we also report 14 distinct, lesser-known bi-, tri-, and tetravalent combinations of activating and repressive chromatin modifications, in platinum-resistant CP70 ovarian cancer cells. The vast majority (>90%) of all the histone marks studied localized to regions within 2000 bp of transcription start sites, supporting a role in gene regulation. Upon a simple alteration in the microenvironment, transition from two- to three-dimensional culture, an increase (17% to 38%) in repressive-only marked promoters was observed, concomitant with a decrease (31% to 21%) in multivalent (i.e., juxtaposed permissive and repressive histone marked) promoters. Like embryonic/tissue stem and other (non-ovarian) carcinoma cells, ovarian cancer cell epigenetic plasticity reflects an inherent transcriptional flexibility for context-responsive alterations in phenotype. It is possible that this plasticity could be therapeutically exploited for the management of this lethal gynecologic malignancy.


Subject(s)
Chromatin Assembly and Disassembly , Drug Resistance, Neoplasm , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/metabolism , Female , Histones/genetics , Histones/metabolism , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Ovarian Neoplasms/genetics , Promoter Regions, Genetic/genetics , Transcription, Genetic/genetics , Tumor Cells, Cultured
9.
Radiat Res ; 173(2): 191-6, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20095851

ABSTRACT

Planning for long-duration manned lunar and interplanetary missions requires an understanding of radiation-induced cataractogenesis. Previously, it was demonstrated that low-linear energy transfer (LET) irradiation with 10 Gy of (60)Co gamma rays resulted in an increased incidence of cataracts in male rats compared to female rats. This gender difference was not due to differences in estrogen, since male rats treated with the major secreted estrogen 17-beta-estradiol (E2) showed an identical increase compared to untreated males. We now compare the incidence and rate of progression of cataracts induced by high-LET radiation in male and female Sprague-Dawley rats. Rats received a single dose of 1 Gy of 600 MeV (56)Fe ions. Lens opacification was measured at 2-4-week intervals with a slit lamp. The incidence and rate of progression of radiation-induced cataracts was significantly increased in the animals in which estrogen was available from endogenous or exogenous sources. Male rats with E2 capsules implanted had significantly higher rates of progression compared to male rats with empty capsules implanted (P = 0.025) but not compared to the intact female rats. These results contrast with data obtained after low-LET irradiation and suggest the possibility that the different types of damage caused by high- and low-LET radiation may be influenced differentially by steroid sex hormones.


Subject(s)
Cataract/etiology , Estradiol/pharmacology , Linear Energy Transfer , Sex Factors , Animals , Cataract/pathology , Disease Progression , Female , Male , Rats , Rats, Sprague-Dawley
10.
Cancer Res ; 69(24): 9192-201, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19951993

ABSTRACT

Tissue transglutaminase (TG2), an enzyme that catalyzes Ca(2+)-dependent aggregation and polymerization of proteins, is overexpressed in ovarian cancer cells and tumors. We previously reported that TG2 facilitates tumor dissemination using an i.p. xenograft model. Here we show that TG2 modulates epithelial-to-mesenchymal transition (EMT), contributing to increased ovarian cancer cell invasiveness and tumor metastasis. By using stable knockdown and overexpression in epithelial ovarian cancer cells, we show that TG2 induces a mesenchymal phenotype, characterized by cadherin switch and invasive behavior in a Matrigel matrix. This is mediated at the transcriptional level by altering the expression levels and function of several transcriptional repressors, including Zeb1. One mechanism through which TG2 induces Zeb1 is by activating the nuclear factor-kappaB complex. The effects of TG2 on ovarian cancer cell phenotype and invasiveness translate into increased tumor formation and metastasis in vivo, as assessed by an orthotopic ovarian xenograft model. Highly expressed in ovarian tumors, TG2 promotes EMT and enhances ovarian tumor metastasis by activating oncogenic signaling.


Subject(s)
Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Transglutaminases/biosynthesis , Animals , Ascites/pathology , Cadherins/biosynthesis , Cadherins/deficiency , Cadherins/genetics , Cell Line, Tumor , Disease Progression , Epithelial Cells/pathology , Female , GTP-Binding Proteins , Homeodomain Proteins/genetics , Humans , Mesoderm/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/metabolism , Ovarian Neoplasms/genetics , Peritoneal Neoplasms/secondary , Protein Glutamine gamma Glutamyltransferase 2 , Transcription Factors/genetics , Transcription, Genetic , Transfection , Transglutaminases/genetics , Transplantation, Heterologous , Zinc Finger E-box-Binding Homeobox 1
11.
J Am Assoc Lab Anim Sci ; 48(5): 517-20, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19807973

ABSTRACT

This case report describes the unanticipated development of pyometra in Brown Norway rats after treatment with estrogen. Sprague Dawley and Brown Norway rats were ovariectomized and randomly assigned to treatment groups (subcutaneous implantation of either a capsule containing 20 mg 17beta-estradiol or an empty capsule, as a control). After irradiation of only the right eye, the rats were followed for several months in an attempt to determine the effects of estrogen on radiation cataractogenesis and investigate potential strain differences in this phenomenon. However, all Brown Norway rats that received estradiol treatment developed pyometra, whereas none the Sprague Dawley or control Brown Norway rats did. This case demonstrates the potential adverse effects of exogenous estrogen therapy, which are strain-specific in the rat. Caution should be taken when designing estrogen-related experiments involving Brown Norway rats and other potentially sensitive strains.


Subject(s)
Estrogens/adverse effects , Pyometra/veterinary , Rats , Rodent Diseases/etiology , Animals , Cataract/drug therapy , Estrogens/administration & dosage , Estrogens/therapeutic use , Eye/radiation effects , Female , Ovariectomy , Pyometra/etiology , Pyometra/microbiology , Rats, Sprague-Dawley , Rodent Diseases/microbiology , Species Specificity , Uterus/drug effects
12.
Radiat Res ; 172(1): 129-33, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19580515

ABSTRACT

Radiation cataractogenesis is an important consideration for radiotherapy patients and for astronauts. Data in the literature suggest that gender and/or estrogen may play a role in the incidence of age-related cataracts. However, few data exist on the effect of gender on radiation-induced cataractogenesis. We compared the incidence and rate of progression of cataracts induced by ionizing radiation in male and female Sprague-Dawley rats. Male rats were implanted with either an empty silastic capsule or a capsule containing 17-beta-estradiol. Ovary-intact female rats were implanted with empty capsules. All rats received a single dose of 10 Gy (60Co gamma rays) to the right eye only. Lens opacification was measured at 2-4-week intervals with a slit lamp. The incidence of radiation-induced cataracts was significantly increased in male rats compared to female rats (P=0.034). There was no difference in the rate of cataract progression between the three groups. Our data suggest there is a gender-related difference in radiation-induced cataractogenesis, but the increased incidence of radiation cataractogenesis in male rats compared to female rats cannot be attributed to estrogen levels, since there was no difference in cataract incidence between male rats implanted with empty capsules and those implanted with capsules containing 17-beta-estradiol.


Subject(s)
Cataract/etiology , Cataract/pathology , Estradiol/metabolism , Radiation Injuries, Experimental/complications , Radiation Injuries, Experimental/pathology , Sex Characteristics , Animals , Disease Progression , Female , Gamma Rays/adverse effects , Lens, Crystalline/pathology , Lens, Crystalline/radiation effects , Linear Models , Male , Rats , Rats, Sprague-Dawley
13.
Invest Ophthalmol Vis Sci ; 50(7): 3304-10, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19255148

ABSTRACT

PURPOSE: Epidemiologic data on the effects of female sex hormones in cataract formation are conflicting. With the use of a rat model of radiation-induced cataractogenesis, it was found that estrogen can either enhance or inhibit the progression of radiation cataracts, depending on when the hormone is administered. The present study was performed to further define radiation-hormone interactions during cataractogenesis. METHODS: In one experiment, rats were left ovary-intact or ovariectomized and were then irradiated with 2.5, 5, 10, or 15 Gy to one eye. In another experiment, ovariectomized rats were treated continuously with three different doses of estradiol through a slow-release capsule implanted subcutaneously, after which one eye was irradiated with 15 Gy. In all animals, cataract formation was followed by slit lamp examination at regular intervals. RESULTS: Latency to identification of cataracts decreased exponentially with increasing radiation dose. The presence of ovaries enhanced cataractogenesis when the eye was irradiated with 15 Gy, but there was no difference between ovary-intact and ovariectomized rats that were irradiated at lower doses. In ovariectomized rats irradiated with 15 Gy, estradiol increased the rate of progression of cataracts in a dose-dependent manner. The rate of cataract progression increased linearly with increasing estradiol dose; there was no sign of saturation at high estradiol doses, as would be expected from a receptor-mediated effect. CONCLUSIONS: Ovarian hormones enhance radiation-induced cataract formation; hormone supplementation experiments indicate that estrogen is responsible for this effect. The data suggest that the enhancing effect of estradiol is not mediated by its receptor, but this requires further study.


Subject(s)
Cataract/metabolism , Estrogens/metabolism , Gamma Rays/adverse effects , Lens, Crystalline/radiation effects , Ovary/physiology , Radiation Injuries, Experimental/metabolism , Animals , Cataract/etiology , Cataract/pathology , Disease Progression , Dose-Response Relationship, Radiation , Estradiol/administration & dosage , Female , Ovariectomy , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/pathology , Rats , Rats, Sprague-Dawley
14.
Environ Health Perspect ; 117(1): 93-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19165393

ABSTRACT

BACKGROUND: A previous study from our laboratory showed that polybrominated diphenyl ethers (PBDEs) were metabolized to hydroxylated PBDEs (HO-PBDEs) in mice and that para-HO-PBDEs were the most abundant and, potentially, the most toxic metabolites. OBJECTIVE: The goal of this study was to determine the concentrations of HO-PBDEs in blood from pregnant women, who had not been intentionally or occupationally exposed to these flame retardants, and from their newborn babies. METHODS: Twenty human blood samples were obtained from a hospital in Indianapolis, Indiana, and analyzed for both PBDEs and HO-PBDEs using electron-capture negative-ionization gas chromatographic mass spectrometry. RESULTS: The metabolite pattern of HO-PBDEs in human blood was quite different from that found in mice; 5-HO-BDE-47 and 6-HO-BDE-47 were the most abundant metabolites of BDE-47, and 5'-HO-BDE-99 and 6'-HO-BDE-99 were the most abundant metabolites of BDE-99. The relative concentrations between precursor and corresponding metabolites indicated that BDE-99 was more likely to be metabolized than BDE-47 and BDE-100. In addition, three bromophenols were also detected as products of the cleavage of the diphenyl ether bond. The ratio of total hydroxylated metabolites relative to their PBDE precursors ranged from 0.10 to 2.8, indicating that hydroxylated metabolites of PBDEs were accumulated in human blood. CONCLUSIONS: The quite different PBDE metabolite pattern observed in humans versus mice indicates that different enzymes might be involved in the metabolic process. Although the levels of HO-PBDE metabolites found in human blood were low, these metabolites seemed to be accumulating.


Subject(s)
Polybrominated Biphenyls/blood , Animals , Ethers , Gas Chromatography-Mass Spectrometry , Humans , Hydroxylation , Mice , United States
15.
Environ Health Perspect ; 116(10): 1315-21, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18941571

ABSTRACT

BACKGROUND: Polybrominated diphenyl ethers (PBDEs) are widely found in the environment and are suspected endocrine disruptors. We previously identified six hydroxylated metabolites of PBDE (OH-PBDEs) in treated mice. OBJECTIVE: We tested the hypothesis that OH-PBDEs would interact with and alter activity of estrogen receptor-alpha (ER-alpha). METHODS: We tested estrogenicity using two assays: 3H-estradiol (3H-E2) displacement from recombinant ER-alpha and induction of reporter gene (ERE-luciferase) in cultured cells. We incubated the PBDE mixture DE-71 with rat liver microsomes and tested the resultant metabolite mixture for estrogenic activity. We also determined relative estrogenic potential of individual hydroxylated PBDE congeners. RESULTS: Reporter gene activity was increased by DE-71 that had been subjected to microsomal metabolism. DE-71 did not displace E2 from ER-alpha, but all six of the OH-PBDE metabolites did. para-Hydroxylated metabolites displayed a 10- to 30-fold higher affinity for ER-alpha compared with ortho-hydroxylated PBDEs, and one produced a maximal effect 30% higher than that produced by E2. Coadministration of E2 and DE-71, or certain of its metabolites, yielded reporter activity greater than either chemical alone. Two ortho-OH-PBDEs were antiestrogenic in the reporter assay. CONCLUSIONS: The observations--that the DE-71 mixture did not displace 3H-E2 from ER-alpha while the hydroxylated metabolites did-suggest that the weak estrogenic effects of DE-71 are due to metabolic activation of individual congeners. However, the behavior of DE-71 and its metabolites, when co-administered with E2, suggest a secondary, undetermined mechanism from classical ER-alpha activation.


Subject(s)
Estrogen Receptor alpha/drug effects , Phenyl Ethers/toxicity , Polybrominated Biphenyls/toxicity , Animals , Biotransformation , Estrogen Receptor alpha/metabolism , Genes, Reporter , Halogenated Diphenyl Ethers , Hydroxylation , Ligands , Microsomes, Liver/drug effects , Phenyl Ethers/metabolism , Phenyl Ethers/pharmacokinetics , Polybrominated Biphenyls/metabolism , Polybrominated Biphenyls/pharmacokinetics , Rats , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism
16.
Mol Cancer Ther ; 7(7): 1993-2002, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18645009

ABSTRACT

Epithelial ovarian cancer (EOC) arises from the epithelial layer covering the surface of ovaries and i.p. metastasis is commonly observed at diagnosis. Sphingosine-1-phosphate (S1P), a bioactive lipid signaling molecule, is potentially involved in EOC tumorigenesis. We have found that S1P is elevated in human EOC ascites. We show that physiologically relevant concentrations of S1P stimulate migration and invasion of EOC cells but inhibit migration of human ovarian surface epithelial (HOSE) cells. In addition, S1P inhibits lysophosphatidic acid (LPA)-induced cell migration in HOSE but not in EOC cells. We have provided the first line of evidence that the expression levels of S1P receptor subtypes are not the only determinants for how cells respond to S1P. Although S1P(1) is expressed and functional in HOSE cells, the inhibitory effect mediated by S1P(2) is dominant in those cells. The cellular preexisting stress fibers are also important determinants for the migratory response to S1P. Differential S1P-induced morphology changes are noted in EOC and HOSE cells. Preexisting stress fibers in HOSE cells are further enhanced by S1P treatment, resulting in the negative migratory response to S1P. By contrast, EOC cells lost stress fibers and S1P treatment induces filopodium-like structures at cell edges, which correlates with increased cell motility. In addition, inhibition of the protein kinase C pathway is likely to be involved in the inhibitory effect of S1P on LPA-induced cell migration in HOSE cells. These findings are important for the development of new therapeutics targeting S1P and LPA in EOC.


Subject(s)
Cell Movement/drug effects , Epithelial Cells/pathology , Lysophospholipids/pharmacology , Ovarian Neoplasms/pathology , Sphingosine/analogs & derivatives , Ascites/pathology , Cell Line, Tumor , Cell Shape/drug effects , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Female , Humans , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Ovarian Neoplasms/enzymology , Protein Kinase C/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction/drug effects , Sphingosine/pharmacology , Stress Fibers/drug effects , Stress Fibers/pathology
17.
Mol Cancer Ther ; 7(7): 2096-108, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18645020

ABSTRACT

Breast cancer resistance to the antiestrogens tamoxifen (OHT) and fulvestrant is accompanied by alterations in both estrogen-dependent and estrogen-independent signaling pathways. Consequently, effective inhibition of both pathways may be necessary to block proliferation of antiestrogen-resistant breast cancer cells. In this study, we examined the effects of apigenin, a dietary plant flavonoid with potential anticancer properties, on estrogen-responsive, antiestrogen-sensitive MCF7 breast cancer cells and two MCF7 sublines with acquired resistance to either OHT or fulvestrant. We found that apigenin can function as both an estrogen and an antiestrogen in a dose-dependent manner. At low concentrations (1 mumol/L), apigenin stimulated MCF7 cell growth but had no effect on the antiestrogen-resistant MCF7 sublines. In contrast, at high concentrations (>10 mumol/L), the drug inhibited growth of MCF7 cells and the antiestrogen-resistant sublines, and the combination of apigenin with either OHT or fulvestrant showed synergistic, growth-inhibitory effects on both antiestrogen-sensitive and antiestrogen-resistant breast cancer cells. To further elucidate the molecular mechanism of apigenin as either an estrogen or an antiestrogen, effects of the drug on estrogen receptor-alpha (ERalpha); transactivation activity, mobility, stability, and ERalpha-coactivator interactions were investigated. Low-dose apigenin enhanced receptor transcriptional activity by promoting interaction between ERalpha and its coactivator amplified in breast cancer-1. However, higher doses (>10 mumol/L) of apigenin inhibited ERalpha mobility (as determined by fluorescence recovery after photobleaching assays), down-regulated ERalpha and amplified in breast cancer-1 expression levels, and inhibited multiple protein kinases, including p38, protein kinase A, mitogen-activated protein kinase, and AKT. Collectively, these results show that apigenin can function as both an antiestrogen and a protein kinase inhibitor with activity against breast cancer cells with acquired resistance to OHT or fulvestrant. We conclude that apigenin, through its ability to target both ERalpha-dependent and ERalpha-independent pathways, holds promise as a new therapeutic agent against antiestrogen-resistant breast cancer.


Subject(s)
Apigenin/pharmacology , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha/metabolism , Breast Neoplasms/enzymology , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Drug Synergism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Female , Fluorescence Recovery After Photobleaching , Fulvestrant , Genes, Reporter , Histone Acetyltransferases/metabolism , Humans , Hydroxytestosterones/pharmacology , Nuclear Receptor Coactivator 3 , Protein Binding/drug effects , Protein Kinases/metabolism , Protein Transport/drug effects , Trans-Activators/metabolism , Transcription, Genetic/drug effects
18.
Endocr Relat Cancer ; 15(2): 475-83, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18509000

ABSTRACT

Numerous epidemiological observations point to sex differences in lung cancer etiology and progression. The present study was aimed at understanding the bases of these sex differences. To test the effect of estradiol on tumor progression, we used a mouse model based on conditional Kras expression and concurrent deletion of Tp53 following inhalation of an adenoviral vector expressing Cre recombinase (AdeCre). Ovariectomized females and males were treated with estradiol via a continuous-release capsule. Tumor multiplicity, tumor volume, and histological grade were determined at 10 weeks after AdeCre administration. Cell proliferation was monitored by Ki67 immunohistochemistry at 4 and 10 weeks after AdeCre administration. At 10 weeks, female mice had more than twice the number of tumors evident on the surface of the lungs than male mice; ovariectomy eliminated this sex difference. The estrogen treatment significantly increased tumor number and volume in ovariectomized females and in males. Histological character of the tumors ranged from adenoma to adenocarcinoma. Ovary-intact females exhibited higher grade tumors than ovariectomized females or males. Progression to higher histological grade was stimulated by estrogen in male mice but not in ovariectomized females. At 10 weeks after AdeCre administration, tumor cell Ki67-labeling varied widely, precluding assessment of an estrogen effect; however, at 4 weeks, Ki67 labeling of lung parenchymal cells was increased 3.5-fold by estrogen. In conclusion, estrogen acts as a promoter for lung adenocarcinoma in a genetically defined lung cancer model; estrogen-induced cell proliferation in the oncogene-initiated cells is likely to play a role in this tumor promoter activity.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Estrogens/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Animals , Disease Models, Animal , Disease Progression , Female , Immunoblotting , Integrases/genetics , Male , Mice , Mice, Transgenic , Ovariectomy , Proto-Oncogene Proteins p21(ras)/genetics , Receptors, Estrogen/metabolism , Sex Characteristics , Tumor Suppressor Protein p53/genetics
19.
Environ Health Perspect ; 116(5): 605-11, 2008 May.
Article in English | MEDLINE | ID: mdl-18470304

ABSTRACT

BACKGROUND: Polybrominated diphenyl ethers (PBDEs) are widely found in the environment, and they may act as endocrine disruptors. OBJECTIVE: Our goal in this study was to test the PBDE mixture DE-71 for estrogenic activity. METHODS: We used proliferation of cultured breast cancer cells (MCF-7) and trophic effects in the reproductive tracts of ovariectomized mice as estrogen bioassays. DE-71 was administered to mice by subcutaneous injection (sc) or oral gavage (po), alone or in combination with estradiol, for 3 or 34 days. Liver weights and cytochrome P450 enzyme activities were also measured. RESULTS: DE-71 increased MCF-7 cell proliferation, and this was prevented by antiestrogen. DE-71 cotreatment reduced the effect of estradiol in MCF-7 cells. In the mouse 3-day assay, DE-71 administered alone had no effect on uterine weight, uterine epithelial height (UEH), or vaginal epithelial thickness (VET); however, when DE-71 was administered as a cotreatment, it potentiated estradiol's effect on uterine weight. DE-71 administered sc to BALB/c mice for 34 days slightly increased UEH and VET, and attenuated the estradiol-induced increase in UEH; these effects were not seen in BALB/c mice treated po or in C57BL/6 mice treated sc. DE-71 increased liver weight in BALB/c, C57BL/6, and estrogen receptor-alpha knockout mice. We also found an increase in liver cytochrome P450 1A (CYP1A) and CYP2B activities when DE-71 was administered po, but only CYP2B increased after sc treatment. CONCLUSION: DE-71 behaves as a weak estrogen. In mice, the treatment route and duration determined if DE-71 was estrogenic. BALB/c mice are more susceptible to DE-71 effects in estrogen target tissues than C57BL/6 mice. DE-71 increased liver weight independently of estrogen receptor-alpha.


Subject(s)
Estrogen Receptor Modulators/pharmacology , Phenyl Ethers/pharmacology , Polybrominated Biphenyls/pharmacology , Animals , Biological Assay , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cytochrome P-450 Enzyme System/analysis , Endocrine Disruptors , Estradiol/administration & dosage , Female , Halogenated Diphenyl Ethers , Liver/cytology , Liver/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovariectomy , Phenyl Ethers/administration & dosage , Polybrominated Biphenyls/administration & dosage
20.
J Mol Endocrinol ; 40(5): 199-210, 2008 May.
Article in English | MEDLINE | ID: mdl-18434427

ABSTRACT

A splicing variant of rat striatin-3 (rSTRN3gamma) was found to associate with estrogen receptor-alpha (ERalpha) in a ligand-dependent manner. In two-hybrid and pull-down analyses, estradiol induced an interaction between rSTRN3gamma and ERalpha. STRN3gamma protein was found in nuclear extracts from rat uterus and human cell lines. Overexpression of rSTRN3gamma induced a decrease in ERalpha transcriptional activity but had no effect on ERbeta activity. Immunoprecipitation analyses showed that rSTRN3gamma interacts with both the ERalpha and the catalytic subunit of protein phosphatase 2A (PP2A(C)). The transrepressor action of rSTRN3gamma was overcome by okadaic acid, an inhibitor of PP2A(C), and by cotransfection of PP2A(C) siRNA. rSTRN3gamma caused dephosphorylation of ERalpha at serine 118 and this was abrogated by okadaic acid. ERalpha lacking phosphorylation sites at either serine 118 or 167 was insensitive to the corepressor action of rSTRN3gamma. These observations suggest that an rSTRN3gamma-PP2A(C) complex is recruited to agonist-activated ERalpha, thereby leading to its dephosphorylation and inhibiting transcription.


Subject(s)
Autoantigens/physiology , Calmodulin-Binding Proteins/physiology , Estrogen Antagonists/metabolism , Protein Phosphatase 2/metabolism , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Amino Acid Sequence , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Female , HeLa Cells , Humans , Mice , Molecular Sequence Data , Phosphorylation , Protein Isoforms/physiology , Rats , Receptors, Estrogen/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...