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










Publication year range
1.
Ecotoxicol Environ Saf ; 193: 110371, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32114246

ABSTRACT

Medroxyprogesterone acetate (MPA) is a widely used synthetic progestin in contraception pills and hormone replacement therapy. However, its effects on eye growth and development and function were largely unknown. In this study, the transcription of genes in the Notch signaling pathway and the visual cycle network were evaluated after chronic MPA exposure at 4.32 (L), 42.0 (M), and 424 (H) ng L-1 for 120 days in zebrafish. Meanwhile, the histology of the eyes was also examined. Transcriptional results showed that MPA at all three concentrations significantly increased the transcription of notch1a, dll4, jag1a, ctbp1 and rbpjb (key genes in the Notch signaling pathway) in the eyes of females. The up-regulation of noth1a, ctbp1 and kat2b was also observed in the eyes of males exposed to MPA at 424 ng L-1. In the visual cycle pathway, MPA increased the transcription of opn1sw1, opn1sw2, arr3a and rpe65a in the eyes of females from the M and H treatments. Histopathological analysis showed that exposure to 42.0 ng L-1 of MPA increased the thicknesses of inner nuclear layer in females and outer segment in males. Moreover, exposure to 424 ng L-1 of MPA increased the lens diameter in females. These results indicated that chronic MPA exposure affected the transcription of genes in the Notch signaling and in the visual cycle pathways, resulting in overgrowth of the eyes and interference of the eye functions. This study suggests that MPA pose a risk to fitness and survival of zebrafish in areas where MPA contamination exists.


Subject(s)
Contraceptive Agents, Hormonal/toxicity , Eye/drug effects , Medroxyprogesterone Acetate/toxicity , Animals , Eye/growth & development , Eye/pathology , Female , Male , Receptors, Notch/metabolism , Retina , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish/metabolism
2.
Molecules ; 24(8)2019 Apr 19.
Article in English | MEDLINE | ID: mdl-31010220

ABSTRACT

Uterine leiomyomas, also known as fibroids, are benign neoplasms of the uterus and have a high incidence rate in women of reproductive age. Hysterectomy or myomectomy is the initial treatment, but fibroids will recur if the patient is still exposed to similar risk factors. Therefore, developing new therapeutic strategies are urgently necessary. In this study, the anti-proliferation effects of each fraction of adlay seeds were evaluated in uterine leiomyomas, and we identified the potential phytochemical compounds. We found that the ethyl acetate fraction of adlay hull (AHE-ea) appeared to be highly efficient in the anti-proliferation of rat uterine leiomyoma ELT3 cells and primary human uterine leiomyoma (hUL) cells. The proliferation of primary human normal uterine smooth muscle (UtSMC) and normal uterine myometrial (hUM) cells were also suppressed by AHE-ea. Two phytosterols, stigmasterol and ß-sitosterol, were identified from AHE-ea fraction. Mice treated with AHE-ea and stigmasterol alone demonstrated reduced diethylstilbestrol/medroxyprogesterone 17-acetate (DES/MPA)-induced uterine myometrial hyperplasia, which is the critical step for the development of leiomyoma. Taken together, our results suggest that the AHE-ea fraction could be considered as a natural plant-based medicine in the prevention or treatment of uterine leiomyoma growth.


Subject(s)
Coix/chemistry , Leiomyoma/prevention & control , Plant Extracts/therapeutic use , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Diethylstilbestrol/toxicity , Female , Humans , Leiomyoma/drug therapy , Medroxyprogesterone Acetate/toxicity , Mice , Phosphorylation , Rats , Uterine Neoplasms/chemically induced , Uterine Neoplasms/drug therapy , Uterine Neoplasms/prevention & control
3.
Oncotarget ; 8(12): 18640-18656, 2017 Mar 21.
Article in English | MEDLINE | ID: mdl-28416734

ABSTRACT

Drugs that target the Renin-Angiotensin System (RAS) have recently come into focus for their potential utility as cancer treatments. The use of Angiotensin Receptor Blockers (ARBs) and Angiotensin-Converting Enzyme (ACE) Inhibitors (ACEIs) to manage hypertension in cancer patients is correlated with improved survival outcomes for renal, prostate, breast and small cell lung cancer. Previous studies demonstrate that the Angiotensin Receptor Type I (AT1R) is linked to breast cancer pathogenesis, with unbiased analysis of gene-expression studies identifying significant up-regulation of AGTR1, the gene encoding AT1R in ER+ve/HER2-ve tumors correlating with poor prognosis. However, there is no evidence, so far, of the functional contribution of AT1R to breast tumorigenesis. We explored the potential therapeutic benefit of ARB in a carcinogen-induced mouse model of breast cancer and clarified the mechanisms associated with its success.Mammary tumors were induced with 7,12-dimethylbenz[α]antracene (DMBA) and medroxyprogesterone acetate (MPA) in female wild type mice and the effects of the ARB, Losartan treatment assessed in a preventative setting (n = 15 per group). Tumor histopathology was characterised by immunohistochemistry, real-time qPCR to detect gene expression signatures, and tumor cytokine levels measured with quantitative bioplex assays. AT1R was detected with radiolabelled ligand binding assays in fresh frozen tumor samples.We showed that therapeutic inhibition of AT1R, with Losartan, resulted in a significant reduction in tumor burden; and no mammary tumor incidence in 20% of animals. We observed a significant reduction in tumor progression from DCIS to invasive cancer with Losartan treatment. This was associated with reduced tumor cell proliferation and a significant reduction in IL-6, pSTAT3 and TNFα levels. Analysis of tumor immune cell infiltrates, however, demonstrated no significant differences in the recruitment of lymphocytes or tumour-associated macrophages in Losartan or vehicle-treated mammary tumors.Analysis of AT1R expression with radiolabelled ligand binding assays in human breast cancer biopsies showed high AT1R levels in 30% of invasive ductal carcinomas analysed. Furthermore, analysis of the TCGA database identified that high AT1R expression to be associated with luminal breast cancer subtype.Our in vivo data and analysis of human invasive ductal carcinoma samples identify the AT1R is a potential therapeutic target in breast cancer, with the availability of a range of well-tolerated inhibitors currently used in clinics. We describe a novel signalling pathway critical in breast tumorigenesis, that may provide new therapeutic avenues to complement current treatments.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/drug therapy , Disease Progression , Losartan/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Receptor, Angiotensin, Type 1/metabolism , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Biopsy , Carcinogenesis/metabolism , Carcinoma, Intraductal, Noninfiltrating/chemically induced , Carcinoma, Intraductal, Noninfiltrating/immunology , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Proliferation/drug effects , Female , Humans , Immunohistochemistry , Interleukin-6/metabolism , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Medroxyprogesterone Acetate/toxicity , Mice , Neoplasm Invasiveness , Phosphorylation , Real-Time Polymerase Chain Reaction , Renin-Angiotensin System/drug effects , STAT3 Transcription Factor/metabolism , Signal Transduction , Tumor Burden/drug effects , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
4.
Oncol Rep ; 35(2): 825-32, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26719029

ABSTRACT

Postmenopausal women undergoing hormone-replacement therapy containing both progestins and estrogens are at an increased risk of developing breast cancer compared with women taking estrogen alone. We recently demonstrated that medroxyprogesterone acetate, a progestin commonly used for hormone-replacement therapy, accelerates development of mammary carcinogenesis in 7,12-dimethylbenz(a)anthracene­treated Sprague-Dawley rats. Synthetic antiprogestins used to block the deleterious effects of progestins, are themselves associated with toxic side-effects. In order to circumvent this, we used the aforementioned model to identify less toxic natural compounds that may prevent the development of progestin-accelerated tumors. Luteolin, a naturally-occurring flavonoid commonly found in fruits and vegetables, has previously been shown to possess anticancer properties. In our studies, both low (1 mg/kg) and high (25 mg/kg) doses of luteolin significantly suppressed progestin-dependent increases in tumor incidence, while increasing tumor latency and reducing the occurrence of large (>300 mm3) mammary tumors. However, an intermediate dose of luteolin (10 mg/kg), while suppressing the development of large tumors, did not affect either tumor incidence or latency. Immunohistochemical analysis of tumor tissues revealed that all concentrations of luteolin (1, 10, and 25 mg/kg) significantly reduced levels of VEGF within tumors. The suppressive effects of luteolin on tumor incidence and volume, together with its ability to reduce VEGF and blood vessels, persisted even after treatment was terminated. This suggests that luteolin possesses anti­angiogenic properties which could mechanistically explain its capacity to control tumor progression. Thus luteolin may be a valuable, non-toxic, naturally-occurring anticancer compound which may potentially be used to combat progestin-accelerated mammary tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Luteolin/pharmacology , Mammary Neoplasms, Experimental/pathology , Medroxyprogesterone Acetate/toxicity , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Estrogen Replacement Therapy/adverse effects , Female , Immunohistochemistry , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/biosynthesis
5.
Reproduction ; 150(6): 497-505, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26424807

ABSTRACT

The 3-month injectable contraceptive medroxyprogesterone acetate (MPA; Depo-Provera) is a synthetic progestin that protects against pregnancy by suppressing ovulation. Studies have focused on the resumption of ovulation after MPA-treatment cessation but neglected potential long-term effects of MPA exposure on future successful reproduction. MPA is frequently administered to adolescent girls; however, long-term fertility effects of adolescent MPA exposure have not been explored. We investigated fertility after extended MPA exposure in a species of old world primate, the sooty mangabey (Cercocebus atys). Female sooty mangabeys (n=31) received chronic MPA-treatment for 4-8 years. At MPA-treatment onset, subjects were either parous adults (n=14) or nulliparous adolescents (n=17), with adolescent-treated subjects being further divided into those who had reached first ovulation (n=10) and those who had not (n=7). After MPA-treatment cessation, adolescent-treated females had a significantly higher incidence of stillbirth than did age-matched and parity-matched controls, whereas adult-treated females did not differ from their matched controls. Females placed on MPA-treatment prior to first ovulation had a significantly higher incidence of stillbirth post-treatment than did females placed on MPA-treatment after first ovulation. Diabetic females had an increased incidence of stillbirth as compared to nondiabetic females; however, when controlling for diabetes, MPA exposure prior to first ovulation was still a significant positive predictor of stillbirth. These findings suggest that the post-treatment fertility effects of chronic MPA exposure vary with the developmental timing of treatment onset and raise concern about the use of MPA as a contraceptive for adolescent girls.


Subject(s)
Contraceptive Agents, Female/toxicity , Fertility/drug effects , Medroxyprogesterone Acetate/toxicity , Pregnancy Complications/chemically induced , Sexual Maturation , Stillbirth , Age Factors , Animals , Cercocebus atys , Diabetes Mellitus/physiopathology , Female , Models, Animal , Ovulation/drug effects , Pregnancy , Pregnancy Complications/physiopathology , Risk Assessment , Risk Factors , Time Factors
6.
Environ Sci Technol ; 49(7): 4636-45, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25742325

ABSTRACT

Medroxyprogesterone acetate (MPA) and dydrogesterone (DDG) are synthetic progestins widely used in human and veterinary medicine. Although aquatic organisms are exposed to them through wastewater and animal farm runoff, very little is known about their effects in the environment. Here we provide a comprehensive analysis of the responses of zebrafish (Danio rerio) to MPA, DDG, and their binary mixtures at measured concentrations between 4.5 and 1663 ng/L. DDG and both mixtures impaired reproductive capacities (egg production) of breeding pairs and led to histological alterations of ovaries and testes and increased gonadosomatic index. Transcriptional analysis of up to 28 genes belonging to different pathways demonstrated alterations in steroid hormone receptors, steroidogenesis enzymes, and specifically, the circadian rhythm genes, in different organs of adult zebrafish and eleuthero-embryos. Alterations occurred even at environmentally relevant concentrations of 4.5-4.8 ng/L MPA, DDG and the mixture in eleuthero-embryos and at 43-89 ng/L in adult zebrafish. Additionally, the mixtures displayed additive effects in most but not all parameters in adults and eleuthero-embryos, suggesting concentration addition. Our data suggest that MPA and DDG and their mixtures induce multiple transcriptional responses at environmentally relevant concentrations and adverse effects on reproduction and gonad histology at higher levels.


Subject(s)
Dydrogesterone/toxicity , Gene Expression Regulation/drug effects , Medroxyprogesterone Acetate/toxicity , Reproduction/drug effects , Water Pollutants, Chemical/toxicity , Zebrafish/physiology , Animals , Dose-Response Relationship, Drug , Female , Male , Ovary/drug effects , Random Allocation , Testis/drug effects , Zebrafish/genetics
7.
Toxicol Pathol ; 39(5): 867-78, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21670166

ABSTRACT

The role of estrogens during myocardial ischemia has been extensively studied. However, effects of a standard hormone replacement therapy including 17ß-estradiol (E2) combined with medroxyprogesterone acetate (MPA) have not been assessed, and this combination could have contributed to the negative outcomes of the clinical studies on hormone replacement. We hypothesized that adding MPA to an E2 treatment would aggravate chronic heart failure after experimental myocardial infarction (MI). To address this issue, we evaluated clinical signs of heart failure as well as left ventricular (LV) dysfunction and remodeling in ovariectomized rats subjected to chronic MI receiving E2 or E2 plus MPA. After eight weeks MI E2 showed no effects. Adding MPA to E2 aggravated LV remodeling and dysfunction as judged by increased heart weight, elevated myocyte cross-sectional areas, increased elevated left ventricle end diastolic pressure, and decreased LV fractional shortening. Impaired LV function in rats receiving MPA plus E2 was associated with increased cardiac reactive oxygen species generation and myocardial expression levels of NADPH oxidase subunits. These results support the interpretation that adding MPA to an E2 treatment complicates cardiovascular injury damage post-MI and therefore contributes to explain the adverse outcome of prospective clinical studies.


Subject(s)
Medroxyprogesterone Acetate/toxicity , Myocardial Infarction/physiopathology , Oxidative Stress/drug effects , Ventricular Dysfunction, Left/chemically induced , Ventricular Remodeling/drug effects , Analysis of Variance , Animals , Blood Pressure/drug effects , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Cardiomegaly/pathology , Chronic Disease , Electrocardiography , Estradiol/pharmacology , Female , Heart Rate/drug effects , Histocytochemistry , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , NADPH Oxidases/metabolism , Ovariectomy , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology
8.
Mol Cell Biol ; 30(23): 5456-72, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20876300

ABSTRACT

Progesterone receptor (PR) and ErbB-2 bidirectional cross talk participates in breast cancer development. Here, we identified a new mechanism of the PR and ErbB-2 interaction involving the PR induction of ErbB-2 nuclear translocation and the assembly of a transcriptional complex in which ErbB-2 acts as a coactivator of Stat3. We also highlighted that the function of ErbB-2 as a Stat3 coactivator drives progestin-induced cyclin D1 promoter activation. Notably, PR is also recruited together with Stat3 and ErbB-2 to the cyclin D1 promoter, unraveling a new and unexpected nonclassical PR genomic mechanism. The assembly of the nuclear Stat3/ErbB-2 transcriptional complex plays a key role in the proliferation of breast tumors with functional PR and ErbB-2. Our findings reveal a novel therapeutic intervention for PR- and ErbB-2-positive breast tumors via the specific blockage of ErbB-2 nuclear translocation.


Subject(s)
Breast Neoplasms/etiology , Breast Neoplasms/metabolism , Mammary Neoplasms, Experimental/etiology , Mammary Neoplasms, Experimental/metabolism , Receptor, ErbB-2/metabolism , Receptors, Progesterone/metabolism , STAT3 Transcription Factor/metabolism , Trans-Activators/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Female , Gene Knockdown Techniques , Genes, bcl-1 , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Medroxyprogesterone Acetate/toxicity , Mice , Mice, Inbred BALB C , Progestins/toxicity , Promoter Regions, Genetic , RNA, Small Interfering/genetics , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/genetics , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Signal Transduction , Transcription, Genetic/drug effects
9.
Toxicol Pathol ; 37(7): 969-82, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19841131

ABSTRACT

UNLABELLED: Elevated mineralocorticoid levels and female sex hormones have been shown to confer opposing effects on renal injury, but their combined effects are still unknown. OBJECTIVE: Identify the function of estrogens and of different synthetic progestins on aldosterone salt-mediated renal disease. METHODS: The role of 17beta-estradiol, medroxyprogesterone acetate (MPA), and drospirenone during renal injury was studied in Wistar rats subjected to uni-nephrectomy plus aldosterone salt treatment. RESULTS: Aldo-salt treatment of intact, ovariectomized, and estradiol-treated female rats resulted in remnant kidney hypertrophy without structural damage. Co-treatment with MPA, but not with drospirenone, increased kidney hypertrophy, fluid turnover, sodium retention, and potassium excretion. Medroxyprogesterone acetate also caused glomerular, vascular, tubular, and interstitial lesions that were accompanied by increased blood pressure and enhanced NADPH oxidase (p67phox) and sodium channel (alpha-ENaC) expression. Drospirenone, a progestin with anti-mineralocorticoid function, and spironolactone prevented kidney hypertrophy, hypertension, and sodium retention. Drospirenone and spironolactone also increased renal angiotensin II type 2 receptor expression and relieved aldosterone-induced suppression of serum angiotensin II levels. CONCLUSION: The choice of specific synthetic progestins has profound implications on the development of kidney injury and renal gene expression under conditions of elevated aldosterone serum levels and salt intake.


Subject(s)
Androstenes/pharmacology , Estradiol/toxicity , Kidney Diseases/chemically induced , Medroxyprogesterone Acetate/toxicity , Progesterone Congeners/pharmacology , Aldosterone/metabolism , Aldosterone/toxicity , Androstenes/metabolism , Animals , Blood Pressure/drug effects , Epithelial Sodium Channels/metabolism , Estradiol/chemistry , Estradiol/metabolism , Female , Hypertrophy/chemically induced , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Medroxyprogesterone Acetate/metabolism , Oxidative Stress/drug effects , Progesterone Congeners/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sodium Chloride/toxicity , Spironolactone/metabolism , Spironolactone/pharmacology
10.
J Toxicol Sci ; 34 Suppl 1: SP23-9, 2009.
Article in English | MEDLINE | ID: mdl-19265286

ABSTRACT

As a part of the collaborative study to evaluate the relationship between histopathological changes of the ovary and functional changes in female fertility, 2- and 4-week repeated-dose toxicity studies and a female fertility study were conducted using female Crl:CD(SD) rats using a synthetic progestagen of medroxyprogesterone acetate (MPA) as a test compound. MPA was administered to female rats by gavage at 0, 0.4, 2.0 and 10 mg/kg/day for 2 and 4 weeks to assess the histopathological changes of ovaries and to pregnant rats at the same doses from 2 weeks prior to mating until day 7 of gestation to examine female fertility. The number of non-pregnant female rats with irregular estrous cycle increased in number and there was a decrease in weight of ovaries was observed at doses > or = 2.0 mg/kg in the 2- and 4-week-treatment groups. The histopathological examination revealed an increased number of large atretic follicles and decreases in currently formed corpora lutea and previously-formed large or small ones were observed at the same doses in the 2- and 4-week treatment groups. In female fertility study, the number of animals with an irregular estrous cycle and elongation of mean estrous cycle increased at 0.4 mg/kg, with no changes in fertility. A decreased number of copulating animals and a decreased gestation rate with low preimplantation loss were observed in the 2.0 mg/kg-treatment group and no copulation was observed in the group treated with 10 mg/kg. Based on these results, changes in fertility induced by MPA correlated well with histopathological changes of ovaries after 2 and 4 weeks of treatment, which suggests that a 2 weeks administration period is sufficient to detect ovarian toxicity of MPA with repeated dosing


Subject(s)
Contraceptive Agents, Female/toxicity , Fertility/drug effects , Medroxyprogesterone Acetate/toxicity , Ovary/drug effects , Administration, Oral , Animals , Biomarkers/metabolism , Contraceptive Agents, Female/administration & dosage , Drug Administration Schedule , Embryo, Mammalian/drug effects , Embryonic Development/drug effects , Estrous Cycle/drug effects , Estrous Cycle/physiology , Female , Fertility/physiology , Japan , Medroxyprogesterone Acetate/administration & dosage , No-Observed-Adverse-Effect Level , Ovary/metabolism , Ovary/pathology , Pregnancy , Proliferating Cell Nuclear Antigen/metabolism , Public-Private Sector Partnerships , Rats , Rats, Sprague-Dawley , Societies, Scientific
11.
Endocr Relat Cancer ; 16(2): 333-50, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19190078

ABSTRACT

More than 60% of all breast neoplasias are ductal carcinomas expressing estrogen (ER) and progesterone receptors (PR). By contrast, most of the spontaneous, chemically or mouse mammary tumor virus induced tumors, as well as tumors arising in genetically modified mice do not express hormone receptors. We developed a model of breast cancer in which the administration of medroxyprogesterone acetate to BALB/c female mice induces mammary ductal carcinomas with a mean latency of 52 weeks and an incidence of about 80%. These tumors are hormone-dependent (HD), metastatic, express both ER and PR, and are maintained by syngeneic transplants. The model has been further refined to include mammary carcinomas that evolve through different stages of hormone dependence, as well as several hormone-responsive cell lines. In this review, we describe the main features of this tumor model, highlighting the role of PR as a trigger of key signaling pathways mediating tumor growth. In addition, we discuss the relevance of this model in comparison with other presently used breast cancer models pointing out its advantages and limitations and how, this model may be suitable to unravel key questions in breast cancer.


Subject(s)
Carcinoma, Ductal/metabolism , Mammary Neoplasms, Experimental/metabolism , Neoplasms, Hormone-Dependent/metabolism , Receptors, Progesterone/physiology , Animals , Antineoplastic Agents, Hormonal/toxicity , Female , Mammary Neoplasms, Experimental/pathology , Medroxyprogesterone Acetate/toxicity , Mice , Mice, Inbred BALB C , Receptors, Estrogen/metabolism
12.
Br J Pharmacol ; 158(8): 1951-60, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20050187

ABSTRACT

BACKGROUND AND PURPOSE: The risk for cardiovascular events including venous and arterial disease and stroke is elevated after treatment with estrogen and medroxyprogesterone acetate (MPA) in postmenopausal women. Here, we have investigated the effect of MPA on arterial thrombosis and atherosclerosis in a murine model of atherosclerosis. EXPERIMENTAL APPROACH: Apolipoprotein E (ApoE)-/- mice were bilaterally ovariectomized and treated with placebo, MPA (27.7 microg day(-1)) and MPA + 17-beta-oestradiol (E2; 1.1 microg day(-1)) for 90 days, on a Western-type diet. Thrombotic response was measured in a photothrombosis model, platelet activation by fluorescence activated cell sorting (FACS) analysis (CD62P) and thrombin generation by the endogenous thrombin potential (ETP). Furthermore, aortic plaque burden and aortic root plaque composition were determined. KEY RESULTS: MPA and MPA + E2-treated animals showed an aggravated thrombotic response shown by significantly reduced time to stable occlusion. The pro-thrombotic effect of MPA was paralleled by increased ETP whereas platelet activation was not affected. Furthermore, MPA + E2 reduced the number of cells positive for alpha-smooth muscle actin and increased hyaluronan in the plaque matrix. Interestingly, total plaque burden was reduced by MPA but unchanged by MPA + E2. CONCLUSION AND IMPLICATIONS: Long-term treatment with MPA and MPA + E2 increased arterial thrombosis despite inhibitory effects of MPA on atherosclerosis in ApoE-deficient mice. Increased thrombin formation, reduced smooth muscle content and remodelling of non-collagenous plaque matrix may be involved in the pro-thrombotic effects. Thus, MPA exhibits differential effects on arterial thrombosis and on atherosclerosis.


Subject(s)
Atherosclerosis/etiology , Estradiol/toxicity , Medroxyprogesterone Acetate/toxicity , Thrombosis/etiology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/toxicity , Apolipoproteins E/genetics , Atherosclerosis/physiopathology , Disease Models, Animal , Drug Therapy, Combination , Estradiol/pharmacology , Estrogens/pharmacology , Estrogens/toxicity , Medroxyprogesterone Acetate/pharmacology , Mice , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Ovariectomy , Platelet Activation/drug effects , Thrombin/metabolism , Thrombosis/physiopathology
13.
Life Sci ; 80(3): 212-8, 2006 Dec 23.
Article in English | MEDLINE | ID: mdl-17023004

ABSTRACT

Medroxyprogesterone acetate was studied at three different concentrations (1, 5 and 10 microM), for its genotoxic effects in human peripheral blood lymphocyte culture using chromosomal aberrations and sister chromatid exchanges as parameters. Duplicate peripheral blood cultures were treated with three different concentrations (1, 5 and 10 microM) of medroxyprogesterone acetate. The study was carried out both in the absence as well as in the presence of metabolic activation (S9 mix) with and without NADP. Medroxyprogesterone acetate was found genotoxic at 5 and 10 microM in the presence of S9 mix with NADP. To study the possible mechanism of the genotoxicity of medroxyprogesterone acetate, superoxide dismutase and catalase at different doses were used separately and in combination with 10 microM of medroxyprogesterone at different doses in the presence of S9 mix with NADP. Superoxide dismutase treatment results in an increase of the genotoxic damage but catalase treatment reduce the genotoxic damage of medroxyprogesterone acetate. Catalase treatment in combination with superoxide dismutase also results in the further reduction of the genotoxic damage. The results of the present study reveal that medroxyprogesterone acetate is genotoxic only in the presence of metabolic activation (S9 mix) with NADP. Treatments with superoxide dismutase and catalase suggests the possible generation of reactive oxygen species by redox cycling of various forms of quinones, similar to estrogens, that are the results of aromatic hydroxylation by cytochrome P450s.


Subject(s)
Contraceptives, Oral, Hormonal/toxicity , Cytochrome P-450 Enzyme System/metabolism , Lymphocytes/enzymology , Medroxyprogesterone Acetate/toxicity , Mutagens/toxicity , Reactive Oxygen Species/metabolism , Antioxidants/toxicity , Catalase/toxicity , Cells, Cultured , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Activation/drug effects , Humans , Lymphocytes/pathology , Medroxyprogesterone Acetate/agonists , Mutagenicity Tests , NADP/toxicity , Oxidation-Reduction/drug effects , Superoxide Dismutase/toxicity
14.
Clin Exp Allergy ; 33(10): 1457-63, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14519155

ABSTRACT

BACKGROUND: Sex hormones might affect the severity and evolution of bronchial asthma. From existing literature, there exists, however, no convincing evidence for either exacerbation or improvement of allergic symptoms by progesterone. OBJECTIVE: This study was aimed to explore the effect of exogenously administered progesterone in a mouse model of allergic asthma. METHODS: BALB/c mice were sensitized to ovalbumin (OVA) by intraperitoneal injections with OVA followed by chronic inhalation of nebulized OVA or physiologic saline (Sal). Medroxyprogesterone acetate or placebo was instilled daily into the oesophagus before and during the inhalatory OVA challenge phase. RESULTS: Progesterone worsened allergic airway inflammation in OVA-challenged mice, as evidenced by enhanced bronchial responsiveness to inhaled metacholine and increased bronchial eosinophilia. Elevated airway eosinophilia corresponded with higher bronchial and systemic IL-5 levels in the progesterone group. The ratio of IL-4/IFN-gamma levels in bronchoalveolar lavage fluid and numbers of eosinophil colony-forming units in the bone marrow were also elevated in the latter group. Progesterone, however, did not influence allergen-specific IgE production, nor did it affect bronchial responses in Sal-challenged mice. CONCLUSION: Our data show that exogenously administered progesterone aggravates the phenotype of eosinophilic airway inflammation in mice by enhancing systemic IL-5 production. Progesterone also increases bronchial hyper-reactivity.


Subject(s)
Asthma/chemically induced , Bronchial Hyperreactivity/chemically induced , Eosinophilia/chemically induced , Medroxyprogesterone Acetate/toxicity , Animals , Asthma/pathology , Asthma/physiopathology , Bone Marrow/drug effects , Bone Marrow/pathology , Bronchoalveolar Lavage Fluid/immunology , Cytokines/biosynthesis , Eosinophilia/pathology , Eosinophilia/physiopathology , Immunoglobulin E/blood , Male , Mice , Mice, Inbred BALB C , Ovalbumin/immunology
15.
Clin Exp Metastasis ; 20(2): 111-20, 2003.
Article in English | MEDLINE | ID: mdl-12705632

ABSTRACT

We have developed a series of novel mammary epithelial cell lines from tumors arising in strain 129 mice, with the ultimate goal of evaluating the role of host factors in the development of bone metastases. Mammary tumors were induced in mice with subcutaneously implanted medroxyprogesterone acetate (MPA) pellets followed by administration of DMBA by oral gavage. Mammary tumor development was efficient in the 129 strain and was independent of osteopontin (OPN) expression. Epithelial cell lines were isolated from these tumors; surprisingly, these cells did not form tumors upon inoculation into the mammary fat pad of syngeneic mice, even when MPA was present. One OPN-deficient cell line was selected for further study; full transformation of these cells required expression of both polyoma middle T and activated ras. These doubly transfected cells, 1029 GP+Er3, grew in soft agar, and formed hormone-independent tumors efficiently in the mammary fat pad that spontaneously metastasized to several soft tissue sites but not to the bone. Derivatives of these cells were isolated from tumors arising in the fat pad and from a lung metastasis (r3T and r3L, respectively): these cells formed tumors more rapidly in the fat pad than the parental GP+Er3 cells. Upon left ventricle injection, the r3T and r3L cells formed osteolytic bone metastases in 129 mice, with few metastases seen in other organs. These tumors filled the marrow cavity, and caused extensive destruction of both cortical and trabecular bone. Intriguingly, in an alternative syngeneic host, (129xC57B1/6) F1, osteolytic bone metastases were not seen on x-ray; instead extensive liver metastasis was present in these mice, indicating that genetic factors in these two strains regulate tumor cell homing and distribution during metastasis. These cell lines provide an important new tool in the study of bone metastasis, particularly in elucidating the role of host factors in the development of these lesions, as the 129 mouse strain is frequently used for genetic manipulations in the mouse.


Subject(s)
Bone Neoplasms/secondary , Epithelial Cells/pathology , Mammary Neoplasms, Experimental/pathology , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/immunology , Antigens, Polyomavirus Transforming/metabolism , Antineoplastic Agents, Hormonal/toxicity , Bone Neoplasms/pathology , Carcinogens/toxicity , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Female , Genes, ras/physiology , Heart Ventricles , Humans , Keratins/metabolism , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/secondary , Male , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/genetics , Medroxyprogesterone Acetate/toxicity , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Neoplastic Cells, Circulating/pathology , Osteolysis , Osteopontin , Retroviridae , Sialoglycoproteins/deficiency , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , Transfection , Tumor Cells, Cultured
17.
Cell Biol Toxicol ; 16(6): 385-90, 2000.
Article in English | MEDLINE | ID: mdl-11254164

ABSTRACT

The effect of medroxyprogesterone acetate (MPA) on the mitotic activity of bone marrow and testis during chemotherapy was investigated experimentally in an animal study. A total of 120 male Swiss albino mice were included in this study. Six groups were formed, each consisting of 20 mice. Low-dose MPA (LD-MPA) (15 mg/kg), high-dose MPA (HD-MPA) (100 mg/kg), LD-MPA plus cyclophosphamide (CP) (65 mg/kg), HD-MPA plus CP (65 mg/kg), and CP (65 mg/kg) were administered to the test groups and no drug was administered to the control group. Bone marrow samples and testis were examined for mitotic activity rate (MAR) on days 0, 18, 22, 26, and 30. In groups with regimens containing CP, MAR of hematopoietic cells in bone marrow was suppressed significantly (p<0.05). There was no difference in MAR of hematopoietic cells in bone marrow between the groups given MPA or not (p>0.05). Mitotic activity rate of the testis cells was significantly suppressed in groups with regimens containing MPA (p<0.05). In conclusion, MPA inhibited mitotic activity of testis, but there was no effect on the mitotic activity of bone marrow. These data do not seem to confirm the hypothesis of a myeloprotective effect of MPA.


Subject(s)
Bone Marrow Cells/drug effects , Medroxyprogesterone Acetate/toxicity , Progesterone Congeners/toxicity , Seminiferous Epithelium/drug effects , Animals , Bone Marrow Cells/cytology , Hematopoiesis/drug effects , Male , Mice , Mitosis/drug effects , Resting Phase, Cell Cycle/drug effects , Seminiferous Epithelium/cytology
18.
Oncogene ; 18(46): 6370-9, 1999 Nov 04.
Article in English | MEDLINE | ID: mdl-10597237

ABSTRACT

The present study addressed links between progestin and heregulin (HRG) signaling pathways in mammary tumors. An experimental model of hormonal carcinogenesis, in which the synthetic progestin medroxyprogesterone acetate (MPA) induced mammary adenocarcinomas in female Balb/c mice, was used. MPA induced an in vivo up-regulation of HRG mRNA expression in progestin-dependent (HD) tumor lines. Mammary tumor progression to a progestin-independent (HI) phenotype was accompanied by a high constitutive expression of HRG. The HRG message arose from the tumor epithelial cells. Primary cultures of malignant epithelial cells from a HD tumor line were used to investigate HRG involvement on cell proliferation. HRG induced a potent proliferative effect on these cells and potentiated MPA mitogenic effects. Blocking endogenous HRG synthesis by antisense oligodeoxynucleotides (ASODNs) to HRG mRNA inhibited MPA-induced cell growth, indicating that HRG acts as a mediator of MPA-induced growth. High levels of ErbB-2 and ErbB-3 expression and low ErbB-4 levels were found in HD cells. Treatment of these cells with either MPA or HRG resulted in tyrosine phosphorylation of both ErbB-2 and ErbB-3. Furthermore, both HRG and MPA proliferative effects were abolished when cells were treated with ASODNs to ErbB-2 mRNA, providing evidence for a critical role of ErbB-2 in HRG-induced growth. Finally, blocking type I insulin-like growth factor receptor (IGF-IR) expression with ASODN resulted in the complete inhibition of HRG proliferative effect, demonstrating that a functional IGF-IR is required for HRG mitogenic activity. These results provide the first evidence of interactions between progestins and HRB/ErbB signal transduction pathways in mammary cancer and the first demonstration that IGF-IR is required for HRG proliferative effects.


Subject(s)
Adenocarcinoma/genetics , Carcinogens/toxicity , Mammary Neoplasms, Experimental/genetics , Medroxyprogesterone Acetate/toxicity , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/genetics , Neuregulin-1/physiology , Progestins , Receptor, IGF Type 1/physiology , Signal Transduction/drug effects , Adenocarcinoma/chemically induced , Animals , Base Sequence , Cell Division/drug effects , DNA, Antisense/pharmacology , Female , Insulin-Like Growth Factor I/metabolism , Mammary Neoplasms, Experimental/chemically induced , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Neoplasm Proteins/genetics , Neoplasms, Hormone-Dependent/chemically induced , Neuregulin-1/genetics , RNA, Messenger/antagonists & inhibitors , RNA, Neoplasm/antagonists & inhibitors , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/genetics , Receptor, ErbB-2/physiology , Recombinant Proteins/metabolism , Tumor Cells, Cultured
19.
J Steroid Biochem Mol Biol ; 68(1-2): 11-21, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10215033

ABSTRACT

We have developed a model of hormonal carcinogenesis in BALB/c female mice, in which MPA induced ductal mammary adenocarcinomas, expressing high levels of estrogen and progesterone receptors (ER and PR). A series of tumor lines, retaining both PR and ER expression, were obtained from selected tumors, which are maintained by syngeneic passages. In this model progesterone behaves as the growth-stimulating hormone (progesterone-dependent or PD tumors), whereas estrogens induce tumor regression. Through selective treatments we were able to derive a series of progesterone-independent (PI) variants. These lines do not require progesterone treatment to grow in ovariectomized female BALB/c mice, but retain, however, the expression of ER and PR. The aim of this paper is to investigate a possible regulatory role of the progesterone receptor (PR) on PI tumor growth. ER and PR were detected by immunocytochemistry in all lines studied. They were also characterized using biochemical assays and Scatchard plots. No differences in Kd of PR or ER were detected in PI variants. AR or GR were not detected in tumor samples using biochemical assays. Estradiol (5 mg silastic pellet) induced complete tumor regression in all tumors tested. We also evaluated the effects of different antiprogestins on tumor growth. Onapristone (10 mg/kg/day) and mifepristone (4.5 mg/kg/day) were able to induce complete tumor regression. The antiandrogen flutamide (5 mg silastic pellet) had no effect on tumor growth in agreement with the lack of androgen receptors. We used an in vitro approach to corroborate that the antiprogestin-induced inhibition was not attributable to an intrinsic effect. Cultures of a selected PI line were treated with PR antisense oligodeoxynucleotides (ASPR) to inhibit in vitro cell proliferation. A significant decrease of 3H-thymidine uptake was observed in cells of a PI line growing in the presence of 2.5% charcoalized fetal calf serum and 0.8-20 microg/ml ASPR. It can be concluded that the PR pathway is an essential path in the growth stimulation of PI tumors.


Subject(s)
Adenocarcinoma/metabolism , Mammary Neoplasms, Experimental/metabolism , Receptors, Progesterone/metabolism , Adenocarcinoma/chemically induced , Adenocarcinoma/drug therapy , Androgens/metabolism , Animals , Binding Sites , Estradiol/pharmacology , Female , Flutamide/pharmacology , Glucocorticoids/metabolism , Gonanes/pharmacology , Hormone Antagonists/pharmacology , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/drug therapy , Medroxyprogesterone Acetate/toxicity , Mice , Mice, Inbred BALB C , Mifepristone/pharmacology , Neoplasms, Hormone-Dependent/chemically induced , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/metabolism , Oligonucleotides, Antisense/pharmacology , Ovariectomy , Receptors, Estrogen/metabolism , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/genetics , Thymidine/metabolism , Tumor Cells, Cultured
20.
Liver ; 18(5): 343-51, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9831364

ABSTRACT

AIMS/BACKGROUND: Estrogen is known to affect hepatobiliary function; however, it is unusual for high serum levels of estrogen to actually result in clinically detectable hyperbilirubinemia. Women affected by cholestatic jaundice during pregnancy share this genetic susceptibility with two Cricetulus hamsters, the Armenian hamster (Cricetulus migratorius) and the Chinese hamster (Cricetulus griseus). Nevertheless, the pathophysiologic process responsible for this estrogen induced icterus may be different in women and hamsters. The present study compares various facets of estrogen-induced icterus in these two closely related hamsters. METHODS: Hamsters were injected with various estrogens and the acute and chronic effects on liver were monitored by measuring changes in serum constituents and by observing changes in hepatic structure as seen grossly and by light and electron microscopy. RESULTS: In previous studies, hepatic tumors developed in most Armenian hamsters after chronic estrogen treatment, but in the present study, the livers of Chinese hamsters were remarkably free of neoplastic change under similar conditions. Also, when compared with the responses in the Armenian hamsters, signs of hepatic destruction and regeneration were less prevalent in estrogen-treated Chinese hamsters, and they were less susceptible to the effects of estrogen (because larger doses of estrogen were required to produce icterus and the bilirubin levels were lower and of shorter duration). In contrast to the findings in Armenian hamsters, bile canaliculi were severely affected in livers of estrogen-treated Chinese hamsters, and hepatic microvesicular steatosis, indicative of an unusual lipodystrophy caused by estrogen, was prominent. An additional lesion peculiar to the Chinese hamster was striking sinusoidal dilatation, which may be analogous to the oral contraceptive-induced sinusoidal dilatation in humans. CONCLUSIONS: Although these two hamster species are genetically similar, the genes activated by the estrogen receptor show remarkable heterogeneity when their respective livers are examined. Comparisons within these species may provide information about the specific gene activation responsible for particular pathologic events.


Subject(s)
Adenoma, Liver Cell/chemically induced , Carcinoma, Hepatocellular/chemically induced , Cricetulus , Estrogens/toxicity , Liver Neoplasms, Experimental/chemically induced , Liver/drug effects , Adenoma, Liver Cell/pathology , Animals , Bilirubin/blood , Carcinoma, Hepatocellular/pathology , Cricetinae , Diethylstilbestrol/toxicity , Female , Inclusion Bodies/drug effects , Inclusion Bodies/ultrastructure , Liver/pathology , Liver Neoplasms, Experimental/pathology , Male , Medroxyprogesterone Acetate/toxicity , Mifepristone/toxicity , Species Specificity , Tamoxifen/toxicity , Zeranol/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...