Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Hum Reprod ; 29(7): 1400-12, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24781425

ABSTRACT

STUDY QUESTION: Can administration of a prostaglandin (PG) E2 receptor 2 (PTGER2) antagonist prevent pregnancy in adult female monkeys by blocking periovulatory events in the follicle without altering menstrual cyclicity or general health? SUMMARY ANSWER: This is the first study to demonstrate that a PTGER2 antagonist can serve as an effective non-hormonal contraceptive in primates. WHAT IS KNOWN ALREADY: The requirement for PGE2 in ovulation and the release of an oocyte surrounded by expanded cumulus cells (cumulus-oocyte expansion; C-OE) was established through the generation of PTGS2 and PTGER2 null-mutant mice. A critical role for PGE2 in primate ovulation is supported by evidence that intrafollicular injection of indomethacin in rhesus monkeys suppressed follicle rupture, whereas co-injection of PGE2 with indomethacin resulted in ovulation. STUDY DESIGN, SIZE, DURATION: First, controlled ovulation protocols were performed in adult, female rhesus monkeys to analyze the mRNA levels for genes encoding PGE2 synthesis and signaling components in the naturally selected pre-ovulatory follicle at different times after the ovulatory hCG stimulus (0, 12, 24, 36 h pre-ovulation; 36 h post-ovulation, n = 3-4/time point). Second, controlled ovarian stimulation cycles were utilized to obtain multiple cumulus-oocyte complexes (COCs) from rhesus monkeys to evaluate the role of PGE2 in C-OE in vitro (n = 3-4 animals/treatment; ≥3 COCs/animal/treatment). Third, adult cycling female cynomolgus macaques were randomly assigned (n = 10/group) to vehicle (control) or PTGER2 antagonist (BAY06) groups to perform a contraceptive trial. After the first treatment cycle, a male of proven fertility was introduced into each group and they remained housed together for the duration of the 5-month contraceptive trial that was followed by a post-treatment reversibility trial. PARTICIPANTS/MATERIALS, SETTING, METHODS: Quantitative real-time PCR, COC culture and expansion, immunofluorescence/confocal microscopy, enzyme immunoassay, contraceptive trial, ultrasonography, complete blood counts, serum biochemistry tests and blood lipid profiles. MAIN RESULTS AND THE ROLE OF CHANCE: Several mRNAs encoding proteins involved in PGE2 synthesis, metabolism and signaling increase (P < 0.05) in the periovulatory follicle after administration of an ovulatory hCG bolus. PGE2 signaling through PTGER2 induces cumulus cell expansion and production of hyaluronic acid, which are critical events for fertilization. Moreover, chronic administration of a selective PTGER2 antagonist resulted in a significant (P < 0.05 versus vehicle-treated controls) contraceptive effect without altering steroid hormone patterns or menstrual cyclicity during a 5-months contraceptive trial. Fertility recovered as early as 1 month after ending treatment. LIMITATIONS, REASONS FOR CAUTION: This is a proof-of-concept study in a non-human primate model. Further investigations are warranted to elucidate the mechanism(s) of PTGER2 antagonist action in the primate ovary. Although PTGER2 antagonist treatment did not produce any obvious undesirable effects, improvements in the mode of administration, as well as the efficacy of these compounds, are necessary to consider such a contraceptive for women. WIDER IMPLICATIONS OF THE FINDINGS: Monitoring as well as improving the efficacy and safety of female contraceptives is an important public health activity. Even though hormonal contraceptives are effective for women, concerns remain regarding their side-effects and long-term use because of the widespread actions of such steroidal products in many tissues. Moreover, some women cannot take hormones for medical reasons. Thus, development of non-hormonal contraceptives for women is warranted. STUDY FUNDING/COMPETING INTEREST(S): Supported by Bayer HealthCare Pharmaceuticals, The Eunice Kennedy Shriver NICHD Contraceptive Development and Research Center (U54 HD055744), NIH Office of the Director (Oregon National Primate Research Center P51 OD011092), and a Lalor Foundation Postdoctoral Basic Research Fellowship (MCP). The use of the Leica confocal was supported by grant number S10RR024585. Some of the authors (N.B., A.R., K.-H.F., U.F., B.B. and B.L.) are employees of Bayer Healthcare Pharma.


Subject(s)
Contraception/methods , Contraceptive Agents/therapeutic use , Receptors, Prostaglandin E/antagonists & inhibitors , Animals , Female , Gene Expression Regulation , Indomethacin/therapeutic use , Macaca , Macaca fascicularis , Oocytes/drug effects , Ovarian Follicle/drug effects , Ovary/drug effects , Ovulation/drug effects , Pregnancy , Pregnancy, Animal , RNA, Messenger/metabolism , Time Factors
2.
Handb Exp Pharmacol ; (214): 543-87, 2012.
Article in English | MEDLINE | ID: mdl-23027466

ABSTRACT

Sex steroid receptors are ligand-triggered transcription factors. Oestrogen, progesterone and androgen receptors form, together with the glucocorticoid and mineralocorticoid receptors, a subgroup of the superfamily of nuclear receptors. They share a common mode of action, namely translating a hormone-i.e. a small-molecule signal-from outside to changes in gene expression and cell fate, and thereby represent "natural" pharmacological targets.For pharmacological therapy, these receptors have originally been addressed by hormones and synthetic hormone analogues in order to overcome pathologies related to deficiencies in the natural ligands. Another major use for female sex hormone receptor modulators is oral contraception, i.e. birth control.On the other side, blocking the activity of sex steroid receptors has become an established way to treat hormone-dependent malignancies, such as breast and prostate cancer.In this review, we will discuss how the experience gained from the classical pharmacology of these receptors and their molecular similarities led to new options for the treatment of gender-specific diseases and highlight recent progress in medicinal chemistry of sex hormone-modulating drugs.


Subject(s)
Hormone Antagonists/therapeutic use , Receptors, Androgen/drug effects , Receptors, Estrogen/drug effects , Receptors, Progesterone/drug effects , Breast Neoplasms/drug therapy , Female , Humans , Male , Prostatic Neoplasms/drug therapy , Receptors, Androgen/physiology , Receptors, Estrogen/physiology , Receptors, Progesterone/physiology , Selective Estrogen Receptor Modulators/therapeutic use
3.
Carcinogenesis ; 32(11): 1675-83, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21856997

ABSTRACT

Epidemiological data indicate that intake of estrogens and isoflavones may be beneficial for the prevention of colorectal cancer (CRC). Based on this data, the aim of the study was to investigate estrogen receptor (ER) subtype-specific effects on intestinal homeostasis. Ovariectomized (OVX) female Wistar rats were either treated with 17ß-estradiol (4 µg/kg body wt/day) (E2), an ERα-specific agonist (ALPHA) (10 µg/kg body wt/day), an ERß-specific agonist (BETA) (100 µg/kg body wt/day) or genistein (GEN) (10 mg/kg body wt/day) for three weeks. Vehicle-treated OVX and SHAM animals and those cotreated with BETA and the pure antiestrogen Fulvestrant (ICI 182780) (100 µg/kg body wt/day and 3 mg/kg body wt/day) served as controls. GEN and BETA treatment but not E2 and ALPHA administration reduced proliferation in ileal and colonic mucosa cells. The rate of apoptosis in the small intestine and colon was increased by treatment with BETA and GEN, but not by E2. BETA induced antiproliferative and proapoptotic activity also in SHAM animals. The effects were antagonized by the pure antiestrogen Fulvestrant. Polymerase chain reaction gene array analysis revealed that BETA resulted in the downregulation of the oncogene transformation-related protein 63 (p63). Our data indicate that activation of the ERß by specific ERß agonists and GEN induces antiproliferative and proapoptotic effects in the intestinal tract. This observation can be taken as an indication that intake of GEN and specific ERß agonists may protect the ileal and colonic epithelium from tumor development via modulation of tissue homeostasis.


Subject(s)
Anticarcinogenic Agents/pharmacology , Cell Proliferation/drug effects , Estrogen Receptor alpha/agonists , Estrogen Receptor beta/agonists , Genistein/pharmacology , Intestine, Large/drug effects , Intestine, Small/drug effects , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Blotting, Western , Drug Synergism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Estrogens/pharmacology , Female , Fulvestrant , Gene Expression Profiling , Immunoenzyme Techniques , Intestine, Large/metabolism , Intestine, Small/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction
4.
J Steroid Biochem Mol Biol ; 120(1): 53-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20347979

ABSTRACT

The age-related decline in ovarian sex hormone production following the onset of menopause alters skeletal muscle metabolic, structural and functional characteristics. The myosin heavy chain (MHC) expression pattern defines skeletal muscle contraction velocity and is therefore an important factor in skeletal muscle function. The present study was designed to examine the effects of 17beta estradiol (E2), estrogen receptor (ER) subtype selective agonists (ERalpha, ERbeta) or genistein (Gen) following ovary removal (OVX) in female Wistar rats in combination with a high intensity treadmill-based exercise protocol (Ex) or normal cage-based activity (NoEx) on MHC protein expression patterns in the slow fiber type m.Soleus (Sol) and the fast fiber type m.Gastrocnemius (Gas). Gen and E2 in the Sol significantly stimulated MHC-I expression relative to OVX only in the absence of exercise (NoEx). MHC-IIb expression in the Gas was significantly increased relative to OVX in Gen Ex and E2 Ex and NoEx groups. The estrogenic effects in the Sol and Gas were both predominantly mediated via ERbeta pathways, since the ERbeta agonist induced greater MHC increases than OVX or ERalpha. We therefore propose that high intensity exercise in combination with exposure to E2, Gen, ERalpha or ERbeta agonists in OVX rats exerts differential effects on MHC expression in skeletal muscles composed of mainly slow type I MHC (Sol) or fast type II MHC (Gas). In summary, the data shows that MHC composition is affected by estrogens and exercise in a fiber type specific manner and that these effects are mainly mediated by ER-beta. This is of great importance with respect to skeletal muscle health and potential treatment with ER selective agonists.


Subject(s)
Estradiol/pharmacology , Estrogen Receptor beta/metabolism , Genistein/pharmacology , Muscle, Skeletal/metabolism , Myosin Heavy Chains/metabolism , Animals , Female , Ovariectomy , Random Allocation , Rats , Rats, Wistar , Time Factors
5.
Arthritis Rheum ; 62(4): 1017-25, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20131258

ABSTRACT

OBJECTIVE: A number of experimental observations have associated elevated estrogen levels with amelioration of inflammation. The involvement of estrogen and estrogen receptor (ER) isotypes in the regulation of inflammation in males is not well understood. In this study, we used specific ERalpha and ERbeta agonists in male mice deficient in estrogen because of a deletion of aromatase (aromatase-knockout [ArKO] mice) to investigate ER isotype utilization in estrogen regulation of inflammation. METHODS: Lipopolysaccharide (LPS)-induced cytokine expression and antigen-induced arthritis (AIA) were investigated in male ArKO and WT littermate mice, as well as in response to selective agonists of ERalpha (16alpha-LE2) and ERbeta (8beta-VE2). The therapeutic effect of selective ER agonists was also examined in mice with collagen-induced arthritis (CIA). RESULTS: Estrogen deficiency in ArKO mice was associated with significant increases in LPS-induced serum interleukin-6 (IL-6), tumor necrosis factor, monocyte chemotactic protein 1, and interferon-gamma levels, which were significantly abrogated by administration of 16alpha-LE2, but not 8beta-VE2. In contrast, both 16alpha-LE2 and 8beta-VE2 significantly increased LPS-induced IL-10 levels. Estrogen deficiency was also associated with significant exacerbation of AIA and antigen-specific T cell proliferation, which was reversed by administration of either 16alpha-LE2 or 8beta-VE2. ArKO mice showed increased antigen-specific T cell proliferation in response to immunization with type II collagen (CII). Administration of 16alpha-LE2, but not 8beta-VE2, significantly reduced the severity of CIA, which was associated with inhibition of anti-CII-specific IgG. CONCLUSION: These data indicate that endogenous estrogen plays an essential inhibitory role in inflammation in male mice and that ERalpha is the dominant receptor that mediates these effects.


Subject(s)
Cytokines/genetics , Estrogen Receptor alpha/physiology , Animals , Aromatase/deficiency , Aromatase/genetics , Arthritis/blood , Arthritis/chemically induced , Crosses, Genetic , Cytokines/blood , Estradiol/blood , Estrogen Receptor beta/physiology , Estrogens/physiology , Exons/genetics , Female , Homeostasis , Humans , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Reference Values , Sequence Deletion
6.
Bone ; 43(5): 849-55, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18708178

ABSTRACT

The functions of estrogen receptors (ER) alpha and beta (ER-alpha and beta) in bone and fat tissue are not precisely described. Therefore we studied the effects of a specific ERalpha and ERbeta agonist in bone and fat of ovariectomized (ovx) rats and compared them with the effects of estradiol (E2). Animals were s.c. injected for 4-weeks with 3 doses of the ERalpha agonist 16alpha-LE2 or the ERbeta agonist 8beta-VE2 or with E2. The intermediate doses were antagonized by an additional daily treatment with ICI (1.53mg). Bone and fat parameters were evaluated by quantitative computer tomography (qCT). Estrogen regulated hormones were also measured. Uterine weights were stimulated; serum LH and leptin levels suppressed E2 and the ERalpha agonist. Density of the cancellous metaphyseal structures of the tibia was reduced in the controls which was prevented by E2 and the ERalpha agonist. Endosteal surface, endosteal, periosteal circumferences and fat depots were largest in the controls and the ERbeta treated animals and lowest in the E2 and the 16alpha-LE2 injected ovx rats. Osteocalcin and the CrossLaps were highest in the ovx controls and reduced by E2 and the ERalpha agonist. Serum osteocalcin was stimulated by the ERbeta agonist. The strain strength index (SSI) in relation to the bodyweight - an indicator of bone elasticity - was lowest in controls and increased dose dependently in the E2 and in the ERalpha treated animals. Most effects in the uterus, serum and bone were antagonized by ICI. Most effects in the bone and fat were exerted by mechanisms involving the ERalpha but the ERbeta agonist appears to stimulate osteoblasts.


Subject(s)
Adipose Tissue/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Tibia , Animals , Bone Density , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Estradiol/analogs & derivatives , Estradiol/metabolism , Estrogen Antagonists/metabolism , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/agonists , Estrogen Receptor beta/genetics , Female , Fulvestrant , Organ Size , Osteocalcin/blood , Ovariectomy , Rats , Rats, Sprague-Dawley , Tibia/anatomy & histology , Tibia/metabolism , Uterus/anatomy & histology
7.
Mol Cell Endocrinol ; 291(1-2): 104-8, 2008 Sep 10.
Article in English | MEDLINE | ID: mdl-18433985

ABSTRACT

To further elucidate the processes involved in the physiology of bone-protection by estrogens, ovariectomized (OVX) rats were treated subcutaneously with 17beta-estradiol (E(2)), the ERalpha-specific agonist (16alpha-LE2) and the ERbeta-specific agonist (8beta-VE2). OVX and intact animals served as controls. Biomarkers of bone-formation (osteocalcin (OC), osteopontin (OPN)) and bone-resorption (telopeptides of collagen type I (CTx), pyridinoline cross-links (Pyd)) were quantified. Bone mineral density was measured by computed tomography. OVX-induced bone loss could be antagonized by subcutaneous administration of 17beta-estradiol and 16alpha-LE2. Serum levels of CTx, OC and OPN were significantly elevated in OVX compared to intact animals and reduced by 17beta-estradiol and 16alpha-LE2. Treatment of OVX rats with 8beta-VE2 did not affect bone mineral density (BMD) or bone-marker serum levels. Taken together, the complex expression pattern of bone-markers in OVX rats following subcutaneous administration of ER subtype-specific agonists indicates that 17beta-estradiol exerts its bone-protective effects by modulating the activity of osteoclasts and osteoblasts via ERalpha.


Subject(s)
Biomarkers/metabolism , Bone and Bones/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Animals , Bone Density , Estradiol/metabolism , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/agonists , Estrogen Receptor beta/genetics , Female , Homeostasis , Molecular Structure , Organ Size , Ovariectomy , Random Allocation , Rats , Uterus/anatomy & histology , Uterus/metabolism
8.
Br J Pharmacol ; 153(7): 1432-7, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18246095

ABSTRACT

BACKGROUND AND PURPOSE: Studies with oestrogen receptoralpha (ERalpha)- and ERbeta-selective compounds have already shown that the effects of 17beta-estradiol (E2) on body weight, movement drive and bone-protection are mediated via ERalpha. This study was based on the hypothesis that activation of ERbeta may antagonize ERalpha-mediated effects and designed to investigate potential effects of ERalpha/ERbeta heterodimers. EXPERIMENTAL APPROACH: Ovariectomized (OVX) female Wistar rats were treated with combinations of the ERalpha-specific agonist 16alpha-LE2 (ALPHA; 1 and 10 microg kg(-1) d(-1)), the ERbeta-specific agonist 8beta-VE2 (BETA; 100 microg kg(-1) d(-1)), the phytoestrogen, genistein (10 mg kg(-1) d(-1)) and with the anti-oestrogen compound, ICI 182,780 (3 mg kg(-1) d(-1)) for three weeks. The combined effects of the substances on body weight increase, tibial bone mineral density (BMD) and the influence on running wheel activity (RWA) were investigated. KEY RESULTS: OVX-induced body weight increase was reduced by co-administration of genistein and BETA. Co-application of BETA or genistein with ALPHA had no effect on ALPHA-mediated bone-protection. The RWA of OVX animals was significantly reduced by treatment with genistein but stimulated by application of ALPHA. The stimulatory effect of ALPHA on RWA could be antagonized by co-treatment with the pure antioestrogen ICI 182,780 but also by co-administration of genistein or BETA. CONCLUSIONS AND IMPLICATIONS: Our results indicate that activation of ERbeta may modulate ERalpha-mediated physiological effects in vivo. The observation that substances with selective affinity for ERbeta are able to antagonize distinct physiological functions, like RWA, may be of great relevance to the pharmaceutical use of such drugs.


Subject(s)
Estrogen Receptor alpha/agonists , Estrogen Receptor beta/agonists , Estrogens/pharmacology , Animals , Body Weight/drug effects , Bone Density/drug effects , Dose-Response Relationship, Drug , Drug Therapy, Combination , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Female , Fulvestrant , Genistein/pharmacology , Ligands , Motor Activity/drug effects , Ovariectomy , Phytoestrogens/pharmacology , Rats , Rats, Wistar
9.
Neurosci Lett ; 417(1): 95-9, 2007 Apr 24.
Article in English | MEDLINE | ID: mdl-17398000

ABSTRACT

Sex steroids have potent effects on mood, mental state and cognition. Our previous findings and those of others suggest that these effects may be due at least in part to estradiol actions on central serotonergic mechanisms. Specifically, estradiol-17beta in its acute positive feedback mode for gonadotropin release in the female rat induces expression of the genes for the 5-hydroxytryptamine(2A) receptor (5-HT(2A)R) and the serotonin transporter (SERT) in the dorsal raphe nucleus (DRN). This is accompanied by an increase in the densities of 5-HT(2A)R and the SERT in forebrain regions which in the human are concerned with the control of mood, mental state, cognition and emotion. Here we report that raloxifene, a benzothiophene and selective estrogen receptor modulator (SERM), completely blocked estradiol stimulation of brain 5-HT(2A)R and SERT expression in acutely ovariectomized rats. Raloxifene also blocked the estrogen-induced surge of luteinizing hormone. Treatment of acutely ovariectomized rats with raloxifene alone increased the density of SERT sites in the mid-frontal cortex and decreased the density of 5-HT(2A)R in the posterior olfactory tubercle. The inhibitory effects of raloxifene on acute estrogen-induction of central serotonergic mechanisms were similar to those of tamoxifen even though there are major differences between the two SERMs in their affinity for the two estrogen receptor subtypes and their actions on the uterus. These findings provide robust evidence that estradiol induction of the 5-HT(2A)R and the SERT in brain is mediated by nuclear estrogen receptors. Our data may provide the basis for obtaining a better understanding of the effects of sex steroids on mood and mental state in the human and the possible rational development of congeners of sex steroids for the treatment of mental disorders.


Subject(s)
Brain/drug effects , Estradiol/pharmacology , Raloxifene Hydrochloride/pharmacology , Receptor, Serotonin, 5-HT2A/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Serotonin Plasma Membrane Transport Proteins/metabolism , Animals , Brain/anatomy & histology , Brain/metabolism , Brain Chemistry/drug effects , Brain Chemistry/physiology , Estradiol/therapeutic use , Female , Luteinizing Hormone/metabolism , Mood Disorders/drug therapy , Mood Disorders/metabolism , Mood Disorders/physiopathology , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT2A/genetics , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Tamoxifen/pharmacology
10.
Bone ; 40(6): 1529-35, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17383949

ABSTRACT

Reduced estrogen levels occurring during menopause in women are accompanied by a variety of disorders, e.g. hot flushes, depressions, osteoporosis, increase in body weight and reduced movement drive. The phytoestrogen genistein (GEN) has been demonstrated to have a significant bone-protective potency. In order to study the ER subtype-specific effects of this phytoestrogen on bone in an animal model, ovariectomized (OVX) female Wistar rats were either treated with 17beta-estradiol (E(2)) (4 microg/kg/day), the ER alpha-specific agonist (ALPHA) 16 alpha-LE(2) (10 microg/kg/day), the ER beta-specific agonist (BETA) 8 beta-VE(2) (100 microg/kg/day) or GEN (10 mg/kg/day) for 3 weeks. Vehicle-treated OVX animals served as controls. All animals had the opportunity of voluntary wheel running. Movement activity, changes of body weight and trabecular bone mineral density (BMD) in the tibia were analyzed. E(2) and ALPHA treatment, but not treatment with BETA, significantly increased the movement activity of OVX rats. Treatment with GEN resulted in a significant decrease of movement activity as compared to OVX animals. Bone mineral density in the trabecular area of the tibia and the expression of bone morphogenetic protein-2 (BMP-2) were significantly reduced in OVX- and BETA-treated rats as compared to rats substituted with E(2), ALPHA and GEN. The bone-protective effect of ALPHA was antagonized by co-treatment with the pure antiestrogen Faslodex (ICI). In order to distinguish hormone-dependent effects from those of exercise, we performed an additional experiment where the animals had no opportunity of wheel running. The results demonstrate that physically inactive rats have a stronger decrease of bone mineral density than physically active animals. Very surprisingly, our data demonstrate that GEN has no bone-protective activity in the absence of physical activity. In contrast, ALPHA and E(2) are bone-protective in the presence and absence of physical activity. In conclusion, our data provide evidence that the effects of E(2) on body weight, movement drive and protection of bone mineral density are mediated via ER alpha, whereas activation of ER beta has only a limited effect. Our data also indicate that the bone-protective effects of GEN may be mediated via ER alpha-dependent mechanisms and that physical activity has a strong impact on the bone-protective potency of this phytoestrogen.


Subject(s)
Estrogen Receptor alpha/metabolism , Genistein/pharmacology , Motor Activity/physiology , Phytoestrogens/pharmacology , Phytotherapy , Animals , Bone Density/drug effects , Dose-Response Relationship, Drug , Female , Genistein/administration & dosage , Ovariectomy , Phytoestrogens/administration & dosage , Rats , Rats, Wistar
11.
Article in English | MEDLINE | ID: mdl-18540572

ABSTRACT

Postmenopausal women that still have an uterus and suffer from hot flushes are treated with combinations of estrogens and progestins. Whereas estrogens are indispensable for treating postmenopausal symptoms, progestins are added to counteract the proliferative activity of estrogens on uterine epithelial cells. However, in the mammary gland, progestins, given together with estrogens, stimulate the proliferation of mammary epithelial cells. Therefore, progestins with reduced proliferative activity in the mammary gland would be of advantage for hormone therapy of postmenopausal women. In order to identify progestins with better tissue-selectivity, we exploited the activation of different signal transduction pathways by the classical progesterone receptor. We demonstrated that progestins with reduced non-genomic versus genomic activity in vitro show a better dissociation of uterine versus mammary gland effects in vivo than medroxyprogesterone acetate (MPA), a synthetic progestin that is widely used in hormone therapy.


Subject(s)
Genomics , Progestins/physiology , Animals , Cell Proliferation , Epithelial Cells/cytology , Female , Humans , Mammary Glands, Human/cytology , Pregnancy , Receptors, Progesterone/physiology , Uterus/cytology
12.
Article in English | MEDLINE | ID: mdl-17824170

ABSTRACT

The multiple actions of estrogen in mammalian physiology are brought about, on a molecular level, by several signaling pathways, and mediated by at least two receptors-estrogen receptor (ER) alpha and beta. Analysis of knock-out mice devoid of either or both receptor isoforms revealed the essential function of estrogen receptor alpha in female reproduction, as ERalpha deficiency leads to a complex endocrine phenotype, severe disturbances in several reproductive organs, and infertility. This reflects the many actions of estrogen in female reproductive endocrinology. To carry the understanding of estrogen action to a cellular resolution, modern genetic technologies can be employed, including artificial chromosome-based transgenesis and conditional gene targeting. The combination of these techniques yields mouse models that lack ERalpha in specific cell types of the body. Using cell-type-specific ERalpha mutants, it could be shown that ERa in neurons is essential for the luteinizing hormone (LH) surge that triggers ovulation. Studies using ERalpha and ERbeta-selective agonists reveal that ERalpha activation is sufficient to induce an ovulatory hormonal stimulus. Thus, genetic analysis and selective pharmacological tools can complement each other in the molecular and cellular dissection of hormone receptor function in vivo.


Subject(s)
Receptors, Estrogen/physiology , Signal Transduction/physiology , Alleles , Animals , Hepatocytes/metabolism , Humans , Mice , Mice, Knockout , Organ Specificity , Receptors, Estrogen/genetics
13.
Article in English | MEDLINE | ID: mdl-17824173

ABSTRACT

Gender differences in the development of cardiovascular disease suggested for a protective function of estrogens in heart disease. The negative or neutral outcome of clinical trials on hormone replacement therapy provides clear evidence that the role of female sex hormones in the cardiovascular system is more complex than previously thought. In particular, the function of estrogens can not be understood without detailed knowledge on the specific function of both estrogen receptor subtypes in the heart and in the vasculature. In here, we review recent studies on subtype selective ERalpha and ERbeta agonists in different animal models of hypertension, cardiac hypertrophy and vascular inflammation. The results indicate that the activation of specific ER subtypes confers specific as well as redundant protective effects in hypertensive heart disease that might ultimately translate into novel treatment options for hypertensive heart disease.


Subject(s)
Cardiovascular System/drug effects , Estrogen Receptor alpha/agonists , Estrogen Receptor beta/agonists , Selective Estrogen Receptor Modulators/pharmacology , Animals , Cardiomegaly/drug therapy , Estrogen Receptor alpha/physiology , Estrogen Receptor beta/physiology , Humans , Hypertension/drug therapy , Rats , Rats, Inbred SHR
14.
Article in English | MEDLINE | ID: mdl-17824175

ABSTRACT

Androgens are known regulators of the growth and differentiation of the prostate gland and are effective during development and maturity as well as in disease. The role of estrogens is less well characterized, but dual direct and indirect actions on prostate growth and differentiation have been demonstrated, facilitated via both ERalpha, and ERbeta. Previous studies using animal models to determine the role of ERbeta in the prostate have been problematic due to the centrally mediated responses to estrogen administration via ERalpha that can lower androgen levels and lead to epithelial regression, thereby masking any direct effects on the prostate mediated by ERbeta. Our alternate approach was to use the estrogen-deficient aromatase knockout (ArKO) mouse and the method of tissue recombination to provide new insight into estrogen action on prostate growth and pathology. Firstly, utilizing homo- and heterotypic tissue recombinants, we demonstrate that stromal aromatase deficiency results in the induction of hyperplasia in previously normal prostatic epithelium and that this response is the result of local changes to the paracrine interaction between stroma and epithelium. Secondly, using tissue recombination and an ERbeta-specific agonist, we demonstrate that the activation of ERbeta results in an anti-proliferative response that is not influenced by alterations to systemic androgen levels or activation of ERalpha. Finally, using intact ArKO mice this study demonstrates that the administration of an ERbeta-specific agonist abrogates existing hyperplastic epithelial pathology specifically in the prostate but an ERbeta-specific agonist does not. Therefore, in the absence of stromal aromatase gene expression, epithelial proliferation, leading to prostatic hypertrophy and hyperplasia, may result from a combination of androgenic stimulation of proliferation and failed activation of ERbeta by locally synthesized estrogens. These data demonstrate essential and beneficial effects of estrogens that are necessary for normal growth of the prostate and distinguish them from those that adversely alter prostate growth and differentiation. This indicates the potential of antiandrogens and SERMS, as opposed to aromatase inhibitors, for the management of prostate hyperplasia and hypertrophy.


Subject(s)
Estrogen Receptor alpha/physiology , Estrogen Receptor beta/physiology , Prostate/growth & development , Selective Estrogen Receptor Modulators/pharmacology , Animals , Aromatase/physiology , Humans , Ligands , Male , Mice , Mice, Knockout , Prostatic Hyperplasia/etiology
15.
Article in English | MEDLINE | ID: mdl-17824177

ABSTRACT

Different molecular mechanisms mediate the diverse biological effects of estrogens. The classical genomic mechanism is based on the function of the ER as a ligand-dependent transcription factor that binds to estrogen-response elements (EREs) in promoters of target genes to initiate gene expression. These direct genomic effects play a prominent role in the regulation of reproductive function. In contrast, nongenomic effects mediated by the classical ER have been demonstrated to activate PI3K, leading to the activation of endothelial NOS (eNOS) and hence vasorelaxation. Pathway-selective ER ligands might represent a novel option for hormone replacement therapy. Here we describe the identification and in vitro characterization of tool compounds that bind the ER reasonably well but exhibit low transcriptional activity on ERE-driven promoters. However, these compounds behave as potent stimulators of PI3K/Akt activation in vitro and lead to aortic vessel relaxation, a mechanism that is thought to be driven by nongenomic ER action. In a second set of experiments, we analyze how the in vitro pathway selectivity translates into the in vivo situation. We examine our tool compounds in comparison to estradiol and estren in the following paradigms: bone protection, uterine growth assays, and mammary gland assays.


Subject(s)
Estrogen Receptor alpha/physiology , Selective Estrogen Receptor Modulators/pharmacology , Signal Transduction/drug effects , Animals , Female , Humans , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/growth & development , Osteoporosis/prevention & control , Ovariectomy , Uterus/growth & development
18.
J Biol Chem ; 275(21): 15885-94, 2000 May 26.
Article in English | MEDLINE | ID: mdl-10748166

ABSTRACT

Clinical studies have shown that estrogen replacement therapy (ERT) reduces the incidence and severity of osteoporosis and cardiovascular disease in postmenopausal women. However, long term estrogen treatment also increases the risk of endometrial and breast cancer. The selective estrogen receptor (ER) modulators (SERMs) tamoxifen and raloxifene, cause antagonistic and agonistic responses when bound to the ER. Their predominantly antagonistic actions in the mammary gland form the rationale for their therapeutic utility in estrogen-responsive breast cancer, while their agonistic estrogen-like effects in bone and the cardiovascular system make them candidates for ERT regimens. Of these two SERMs, raloxifene is preferred because it has markedly less uterine-stimulatory activity than either estrogen or tamoxifen. To identify additional SERMs, a method to classify compounds based on differential gene expression modulation was developed. By analysis of 24 different combinations of genes and cells, a selected set of assays that permitted discrimination between estrogen, tamoxifen, raloxifene, and the pure ER antagonist ICI164384 was generated. This assay panel was employed to measure the activity of 38 compounds, and the gene expression fingerprints (GEFs) obtained for each compound were used to classify all compounds into eight groups. The compound's GEF predicted its uterine-stimulatory activity. One group of compounds was evaluated for activity in attenuating bone loss in ovariectomized rats. Most compounds with similar GEFs had similar in vivo activities, thereby suggesting that GEF-based screens could be useful in predicting a compound's in vivo pharmacological profile.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Animals , Bone Density/drug effects , Endometrium/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Replacement Therapy , Estrogens/pharmacology , Female , Humans , Polyunsaturated Alkamides , Protein Binding , RNA, Messenger/metabolism , Raloxifene Hydrochloride/pharmacology , Raloxifene Hydrochloride/therapeutic use , Rats , Rats, Wistar , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , Transcortin/genetics , Tumor Cells, Cultured
19.
J Med Chem ; 43(26): 5010-6, 2000 Dec 28.
Article in English | MEDLINE | ID: mdl-11150172

ABSTRACT

Herein we describe the chemical synthesis and pharmacological characterization of a novel, highly potent progesterone receptor (PR) antagonist, ZK 230211. The introduction of a 17alpha-pentafluorethyl side chain in the D-ring of the steroid skeleton allowed the combination of high antiprogestagenic activity with little or no other endocrinological effects. In contrast to many other antiprogestins, ZK 230211 did not convert to an agonist in the presence of protein kinase A (PKA) activators and showed high antiprogestagenic activity on both PR isoforms PR-A and PR-B. This high antiprogestagenic activity could also be demonstrated in several in vivo models. Furthermore, this compound displayed only marginal antiglucocorticoid effects. In tumor models ZK 230211 exhibited strong antiproliferative action. The pharmacological properties of ZK 230211 may prove useful in the treatment of endometriosis, leiomyomas, breast cancer, and in hormone replacement therapy.


Subject(s)
Estrenes/chemical synthesis , Hormone Antagonists/chemical synthesis , Receptors, Progesterone/antagonists & inhibitors , Abortifacient Agents/chemical synthesis , Abortifacient Agents/metabolism , Abortifacient Agents/pharmacology , Adrenalectomy , Androgen Antagonists/chemical synthesis , Androgen Antagonists/metabolism , Androgen Antagonists/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Binding, Competitive , Castration , Cell Line , Estrenes/metabolism , Estrenes/pharmacology , Female , Glucocorticoids/antagonists & inhibitors , Gonanes/pharmacology , Hormone Antagonists/metabolism , Hormone Antagonists/pharmacology , Ligands , Male , Mammary Neoplasms, Experimental/drug therapy , Mifepristone/pharmacology , Progesterone/antagonists & inhibitors , Progesterone/pharmacology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Rabbits , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Progesterone/metabolism , Transcriptional Activation
20.
Brain Res Mol Brain Res ; 73(1-2): 119-28, 1999 Nov 10.
Article in English | MEDLINE | ID: mdl-10581405

ABSTRACT

Estradiol-17beta (E(2)), in its positive feedback mode for gonadotropin release in the female rat, induces expression of the genes for the 5-hydroxytryptamine(2A) receptor (5-HT(2A)R) and the serotonin transporter (SERT) in the dorsal raphe nucleus (DRN) with a concomitant increase in the densities of 5-HT(2A)R and the SERT in rat forebrain. The forebrain regions affected are those which, in humans, are concerned with the control of mood, mental state, cognition and emotion. Here we have used the mixed estradiol agonist/antagonist, tamoxifen, to determine whether this action of estradiol is mediated by cytoplasmic estradiol receptors. Acute treatment ( approximately 32 h) of ovariectomized rats with estradiol benzoate (EB) increased significantly the amount of 5-HT(2A)R mRNA and SERT mRNA in the DRN and the densities of 5-HT(2A)R and SERT binding sites in the forebrain. These effects of EB were completely blocked by tamoxifen. Treatment with tamoxifen alone had no effect on either gene expression or the density of binding sites. Together, these data show that tamoxifen acts as a pure estradiol antagonist with respect to serotonergic mechanisms in brain. Detailed analysis of the effects of estradiol and tamoxifen on the DRN showed that SERT gene expression is constitutive only in the posterior DRN; in the anterior DRN, SERT gene expression appears to depend upon estrogen induction which is blocked by tamoxifen. Our findings strongly suggest that estradiol receptors are involved in mediating estradiol action on central serotonergic mechanisms and are relevant for our understanding of the effects of antiestrogens as well as estradiol on mood, mental state and cognition.


Subject(s)
Brain/drug effects , Carrier Proteins/metabolism , Estradiol/pharmacology , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Nerve Tissue Proteins , RNA, Messenger/drug effects , Receptors, Serotonin/metabolism , Tamoxifen/pharmacology , Animals , Binding Sites/drug effects , Binding, Competitive/drug effects , Brain/metabolism , Carrier Proteins/genetics , Estradiol/analogs & derivatives , Estrogen Antagonists/pharmacology , Estrogen Replacement Therapy , Female , In Situ Hybridization , Luteinizing Hormone/blood , Luteinizing Hormone/drug effects , Membrane Glycoproteins/genetics , Organ Size/drug effects , Ovariectomy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Radioligand Assay , Raphe Nuclei/drug effects , Raphe Nuclei/metabolism , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT2A , Receptors, Serotonin/genetics , Serotonin Plasma Membrane Transport Proteins , Tritium , Uterus/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...