Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 198
Filter
1.
Pharmacol Ther ; 209: 107502, 2020 05.
Article in English | MEDLINE | ID: mdl-32061922

ABSTRACT

Increasing evidence of interdependence between G protein-coupled receptors and receptor tyrosine kinase signaling pathways has prompted reevaluation of crosstalk between these receptors in disease and therapy. Investigations into thyroid-stimulating hormone (TSH) and insulin-like growth factor 1 (IGF1) receptor crosstalk, and its application to the clinic have in particular shown recent progress. In this review, we summarize current insights into the mechanism of TSH/IGF1 receptor crosstalk. We discuss evidence that crosstalk is one of the underlying causes of TSHR-based disease and the feasibility of using combinations of TSH receptor and IGF1 receptor antagonists to increase the therapeutic index for the treatment of Graves' hyperthyroidism and Graves' ophthalmopathy.


Subject(s)
Graves Ophthalmopathy/metabolism , Receptor Cross-Talk/physiology , Receptor, IGF Type 1/metabolism , Receptors, Thyrotropin/metabolism , Thyrotropin/metabolism , Animals , Autoantibodies/drug effects , Autoantibodies/metabolism , Graves Ophthalmopathy/drug therapy , Hormone Antagonists/administration & dosage , Hormone Antagonists/metabolism , Humans , Receptor Cross-Talk/drug effects , Receptor, IGF Type 1/antagonists & inhibitors , Receptors, Thyrotropin/antagonists & inhibitors , Thyrotropin/antagonists & inhibitors
3.
J Pharm Biomed Anal ; 154: 108-115, 2018 May 30.
Article in English | MEDLINE | ID: mdl-29544105

ABSTRACT

Mifepristone (RU486) is developed originally as a contraceptive used by hundreds of millions of women world-wide, and also reported as a safe and long-term psychotic depressant, or as a cancer chemotherapeutic agent used by both sexes. In our preliminary study aimed at developing mifepristone as a cancer metastatic chemopreventive, we coincidentally observed that blood mifepristone concentrations in female rats seem to be higher than those in male ones post administration. To substantiate if the pharmacokinetic differences between sexes exist, we established a fast UPLC-MS/MS method to determine mifepristone concentrations in plasma, and analyzed blood concentrations of mifepristone over time in rats and dogs of both sexes. Mifepristone in plasma or incubation liquid was recovered by liquid-liquid extraction using 1 mL of ethyl acetate. Chromatographic separation was performed on a C18 column at 35 °C, with a gradient elution consisting of methanol and water containing 0.1% (v/v) formic acid at a flow rate of 0.3 mL/min. And pharmacokinetic parameters such as elimination half-life, and mean residence time were calculated by using the non-compartmental pharmacokinetics data analysis software. In this work, administrations of mifepristone to rats and beagle dogs revealed that the plasma concentrations of mifepristone (AUC, Cmax) were significantly higher (P < 0.05) in females than that in males. In vitro liver microsomal incubation experiments showed that the metabolic rate of mifepristone in males was higher than that in females, which was consistent with the results of in vivo experiments. In general, we first found the sex-related differences about pharmacokinetic properties of mifepristone and revealed the metabolism difference of hepatic microsomal enzyme is the main reason.


Subject(s)
Antipsychotic Agents/pharmacokinetics , Hormone Antagonists/pharmacokinetics , Mifepristone/pharmacokinetics , Animals , Antipsychotic Agents/metabolism , Chromatography, High Pressure Liquid/methods , Dogs , Female , Half-Life , Hormone Antagonists/metabolism , Male , Microsomes, Liver , Mifepristone/metabolism , Models, Animal , Rats , Rats, Sprague-Dawley , Sex Factors , Tandem Mass Spectrometry/methods
4.
Toxicol Sci ; 160(2): 205-216, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28973306

ABSTRACT

A broad range of pesticides have been reported to interfere with the normal function of the thyroid endocrine system. However, the precise mechanism(s) of action has not yet been thoroughly elucidated. In this study, 21 pesticides were assessed for their binding interactions and the potential to disrupt thyroid homeostasis. In the GH3 luciferase reporter gene assays, 5 of the pesticides tested had agonistic effects in the order of procymidone > imidacloprid > mancozeb > fluroxypyr > atrazine. 11 pesticides inhibited luciferase activity of T3 to varying degrees, demonstrating their antagonistic activity. And there are 4 pesticides showed mixed effects when treated with different concentrations. Surface plasmon resonance (SPR) biosensor technique was used to directly measure the binding interactions of these pesticides to the human thyroid hormone receptor (hTR). 13 pesticides were observed to bind directly with TR, with a KD ranging from 4.80E-08 M to 9.44E-07 M. The association and disassociation of the hTR/pesticide complex revealed 2 distinctive binding modes between the agonists and antagonists. At the same time, a different binding mode was displayed by the pesticides showed mix agonist and antagonist activity. In addition, the molecular docking simulation analyses indicated that the interaction energy calculated by CDOCKER for the agonists and antagonists correlated well with the KD values measured by the surface plasmon resonance assay. These results help to explain the differences of the TR activities of these tested pesticides.


Subject(s)
Endocrine Disruptors/toxicity , Fungicides, Industrial/toxicity , Herbicides/toxicity , Hormone Antagonists/toxicity , Insecticides/toxicity , Pituitary Neoplasms/metabolism , Thyroid Hormone Receptors alpha/drug effects , Thyroid Hormone Receptors beta/drug effects , Animals , Binding Sites , Biosensing Techniques , Cell Line, Tumor , Dose-Response Relationship, Drug , Endocrine Disruptors/chemistry , Endocrine Disruptors/metabolism , Fungicides, Industrial/chemistry , Fungicides, Industrial/metabolism , Herbicides/chemistry , Herbicides/metabolism , Hormone Antagonists/chemistry , Hormone Antagonists/metabolism , Humans , Insecticides/chemistry , Insecticides/metabolism , Kinetics , Ligands , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/genetics , Molecular Docking Simulation , Pituitary Neoplasms/genetics , Protein Binding , Protein Conformation , Rats , Risk Assessment , Structure-Activity Relationship , Surface Plasmon Resonance , Thyroid Hormone Receptors alpha/chemistry , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors beta/chemistry , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Receptors beta/metabolism , Transfection
5.
Physiol Res ; 66(Suppl 3): S295-S303, 2017 09 26.
Article in English | MEDLINE | ID: mdl-28948813

ABSTRACT

Resistance to steroid hormones presents a serious problem with respect to their mass use in therapy. It may be caused genetically by mutation of genes involved in hormonal signaling, not only steroid receptors, but also other players in the signaling cascade as co-regulators and other nuclear factors, mediating the hormone-born signal. Another possibility is acquired resistance which may develop under long-term steroid treatment, of which a particular case is down regulation of the receptors. In the review recent knowledge is summarized on the mechanism of main steroid hormone action, pointing to already proven or potential sites causing steroid resistance. We have attempted to address following questions: 1) What does stay behind differences among patients as to their response to the (anti)steroid treatment? 2) Why do various tissues/cells respond differently to the same steroid hormone though they contain the same receptors? 3) Are such differences genetically dependent? The main attention was devoted to glucocorticoids as the most frequently used steroid therapeutics. Further, androgen insensitivity is discussed with a particular attention to acquired resistance to androgen deprivation therapy of prostate cancer. Finally the potential causes are outlined of breast and related cancer(s) resistance to antiestrogen therapy.


Subject(s)
Drug Resistance/physiology , Hormone Antagonists/metabolism , Hormone Antagonists/pharmacology , Receptors, Steroid/metabolism , Steroids/pharmacology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Drug Resistance/drug effects , Female , Glucocorticoids/metabolism , Glucocorticoids/pharmacology , Glucocorticoids/therapeutic use , Gonadal Steroid Hormones/metabolism , Gonadal Steroid Hormones/pharmacology , Gonadal Steroid Hormones/therapeutic use , Hormone Antagonists/therapeutic use , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Steroids/metabolism , Steroids/therapeutic use
6.
Sci Rep ; 6: 34710, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27725717

ABSTRACT

Preterm labor caused by uterine contractions is a major contributor to neonatal morbidity and mortality. Treatment intended to reduce uterine contractions include tocolytic agents, such as indomethacin. Unfortunately, clinically used tocolytics are frequently inefficient and cross the placenta causing fetal side effects. Here we show for the first time in obstetrics the use of a targeted nanoparticle directed to the pregnant uterus and loaded with a tocolytic for reducing its placental passage and sustaining its efficacy. Nanoliposomes encapsulating indomethacin and decorated with clinically used oxytocin receptor antagonist were designed and evaluated in-vitro, ex-vivo and in-vivo. The proposed approach resulted in targeting uterine cells in-vitro, inhibiting uterine contractions ex-vivo, while doubling uterine drug concentration, decreasing fetal levels, and maintaining the preterm birth rate in vivo in a pregnant mouse model. This promising approach opens new horizons for drug development in obstetrics that could greatly impact preterm birth, which currently has no successful treatments.


Subject(s)
Indomethacin/pharmacology , Liposomes/administration & dosage , Molecular Targeted Therapy/methods , Nanostructures/administration & dosage , Obstetric Labor, Premature/prevention & control , Premature Birth/prevention & control , Tocolytic Agents/pharmacology , Uterus/drug effects , Animals , Disease Models, Animal , Drug Compounding , Female , Gene Expression , Hormone Antagonists/chemistry , Hormone Antagonists/metabolism , Humans , Indomethacin/pharmacokinetics , Liposomes/chemistry , Mice , Nanostructures/chemistry , Placenta/metabolism , Pregnancy , Protein Binding , Receptors, Oxytocin/metabolism , Tocolytic Agents/pharmacokinetics , Uterine Contraction/drug effects , Uterus/metabolism , Vasotocin/analogs & derivatives , Vasotocin/chemistry , Vasotocin/metabolism
7.
J Pept Sci ; 21(7): 569-76, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25851250

ABSTRACT

To increase the selectivity of chemotherapeutic agents, receptor-mediated tumor-targeting approaches have been developed. Here, degarelix [Ac-D-Nal-D-Cpa-D-Pal-Ser-Aph(L-Hor)-D-Aph(Cbm)-Leu-ILys-Pro-D-Ala-NH2], a gonadotropin-releasing hormone antagonist, was employed as a targeting moiety for paclitaxel (PTX). Five PTX-degarelix conjugates were synthesized, in which PTX was attached via disulfide bond to the different position in the degarelix sequence. All of the PTX-degarelix conjugates exhibited a half-life greater than 10 h determined in human serum. A fluorometric imaging plate reader assay showed that the conjugates LK-MY-9 and LK-MY-10 had an antagonism efficacy similar to that of degarelix. The in vitro cytostatic effects of the conjugates were determined by a (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) (MTS) assay, and the 50% inhibitory concentration value of the conjugates on 3T3 mouse embryonic fibroblast cells were one order of magnitude higher than the 50% inhibitory concentration values of the conjugates on MCF-7 human breast cancer cells and HT-29 human colon cancer cells. Receptor saturation tests further demonstrated that pre-incubation of the cells with degarelix reduced the efficacy of LK-MY-10 in a concentration-dependent manner. In conclusion, degarelix is a valid and stable moiety that has great potential for targeting chemotherapy drugs.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Glycoconjugates/chemical synthesis , Hormone Antagonists/chemistry , Oligopeptides/chemistry , Paclitaxel/chemistry , Receptors, LHRH/antagonists & inhibitors , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cell Survival/drug effects , Disulfides/chemistry , Glycoconjugates/pharmacology , HT29 Cells , Hormone Antagonists/metabolism , Hormone Antagonists/pharmacology , Humans , Inhibitory Concentration 50 , MCF-7 Cells , Mice , Molecular Targeted Therapy , NIH 3T3 Cells , Oligopeptides/metabolism , Oligopeptides/pharmacology , Paclitaxel/pharmacology , Protein Binding , Receptors, LHRH/chemistry , Receptors, LHRH/metabolism
8.
Toxicol Lett ; 226(2): 132-9, 2014 Apr 21.
Article in English | MEDLINE | ID: mdl-24518828

ABSTRACT

Glucocorticoids are an essential part of the endocrine system that is responsible for a variety of functions such as regulation of immune activity, appropriate brain function, and fetal development. Disturbance of glucocorticoid signaling can lead to various cardiovascular, inflammatory, and autoimmune diseases, so the identification of chemicals that can modulate activity of the glucocorticoid receptor (GR) is crucial. In this study, molecular docking was utilized to find new agonists and antagonists of the GR. The best hits were further tested on the in vitro model of MDA-kb2 cells expressing luciferase activity in a GR-dependent manner. Nine new potential modulators of the receptor, belonging to six structurally diverse classes, were identified. Six of them, tetramethrin and cypermethrin, diethyl hexyl phthalate and diphenyl isophthalate, naphthol AS-OL and dicumyl peroxide, induced luciferase activity; while the other three, bisphenol P, bisphenol M, and Antioxidant 425, suppressed luciferase activity. Of the nine potential GR modulators, only bisphenol M displayed appreciable binding affinity for the receptor.


Subject(s)
Endocrine Disruptors/toxicity , Gene Expression Regulation/drug effects , Genes, Reporter , Hormone Antagonists/toxicity , Luciferases/biosynthesis , Molecular Docking Simulation , Receptors, Glucocorticoid/drug effects , Binding Sites , Binding, Competitive , Cell Line , Databases, Chemical , Dose-Response Relationship, Drug , Endocrine Disruptors/chemistry , Endocrine Disruptors/metabolism , Hormone Antagonists/chemistry , Hormone Antagonists/metabolism , Humans , Luciferases/genetics , Molecular Structure , Protein Conformation , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/metabolism , Transfection
9.
PLoS One ; 8(5): e64610, 2013.
Article in English | MEDLINE | ID: mdl-23724068

ABSTRACT

Alternative splicing of genes generates novel mRNAs, leading to the evolution of new functional proteins. Cholecystokinin (CCK) induces the release of pancreatic enzymes and the contraction of the gallbladder to promote the digestion of fat and proteins. CCK activates two G-protein-coupled receptors, CCKA and CCKB. Here, we showed that a CCKsv (splicing variant), originated de novo during Catarrhini evolution by including a portion of intronic sequence of the CCK gene, encodes novel C-terminal peptide sequence followed by a new poly-adenylation signal. CCKsv is expressed in many human tissues and likely a secreted peptide retaining the original signal peptide and the N-terminal proteolytic processing signal, together with novel C-terminal sequences. Although CCKsv cannot activate CCK receptors, it partially inhibits the CRE- or SRF-driven reporter activities stimulated by wide type CCK-8 mediated by both CCK receptors. Co-treatment with CCKsv also partially antagonizes Ewing tumor cell growth stimulated by CCK-8. Our study provides an example of new peptide hormone antagonist evolution in primates.


Subject(s)
Alternative Splicing/genetics , Evolution, Molecular , Hormone Antagonists/metabolism , Primates/genetics , Alternative Splicing/drug effects , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cell Proliferation/drug effects , Cholecystokinin/chemistry , Cholecystokinin/genetics , Cholecystokinin/metabolism , Hormone Antagonists/pharmacology , Humans , Introns/genetics , Molecular Sequence Data , Organ Specificity/genetics , Receptors, Cholecystokinin/metabolism , Sarcoma, Ewing/pathology
10.
Bioconjug Chem ; 24(5): 766-71, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23600997

ABSTRACT

We employed molecular modeling to design and then synthesize fluorescent ligands for the human progesterone receptor. Boron dipyrromethene (BODIPY) or tetramethylrhodamine were conjugated to the progesterone receptor antagonist RU486 (Mifepristone) through an extended hydrophilic linker. The fluorescent ligands demonstrated comparable bioactivity to the parent antagonist in live cells and triggered nuclear translocation of the receptor in a specific manner. The BODIPY labeled ligand was applied to investigate the dependency of progesterone receptor nuclear translocation on partner proteins and to show that functional heat shock protein 90 but not immunophilin FKBP52 activity is essential. A tissue distribution study indicated that the fluorescent ligand preferentially accumulates in tissues that express high levels of the receptor in vivo. The design and properties of the BODIPY-labeled RU486 make it a potential candidate for in vivo imaging of PR by positron emission tomography through incorporation of (18)F into the BODIPY core.


Subject(s)
Boron Compounds/analysis , Fluorescent Dyes/analysis , Hormone Antagonists/analysis , Mifepristone/analysis , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/metabolism , Boron Compounds/metabolism , Breast/cytology , Breast/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival , Female , Fluorescent Dyes/metabolism , Hormone Antagonists/metabolism , Humans , Mifepristone/metabolism , Models, Molecular , Optical Imaging , Receptors, Progesterone/analysis
11.
Anal Biochem ; 436(1): 1-9, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23333588

ABSTRACT

The pleiotropic cytokine hormone leptin, by activating its receptor OB-R, plays a major role in many biological processes, including energy homeostasis, immune function, and cell survival and proliferation. Abnormal leptin action is associated with obesity, autoimmune diseases, and cancer. The pharmacological characterization of OB-R and the development of synthetic OB-R ligands are still in their infancy because currently available binding assays are not compatible with ligand saturation binding experiments and high-throughput screening (HTS) approaches. We have developed here a novel homogeneous time-resolved fluorescence-based binding assay that overcomes these limitations. In this assay, fluorescently labeled leptin or leptin antagonist binds to the SNAP-tagged OB-R covalently labeled with terbium cryptate (Tb). Successful binding is monitored by measuring the energy transfer between the Tb energy donor and the fluorescently labeled leptin energy acceptor. Ligand binding saturation experiments revealed high-affinity dissociation constants in the subnanomolar range with an excellent signal-to-noise ratio. The assay performed in a 384-well format shows high specificity and reproducibility, making it perfectly compatible with HTS applications to identify new OB-R agonists or antagonists. In addition, fluorescently labeled leptin and SNAP-tagged OB-R will be valuable tools for monitoring leptin and OB-R trafficking in cells and tissues.


Subject(s)
Fluorescence , Receptors, Leptin/metabolism , Alkaline Phosphatase/chemistry , Alkaline Phosphatase/metabolism , Binding Sites/drug effects , Cells, Cultured , HEK293 Cells , High-Throughput Screening Assays , Hormone Antagonists/chemistry , Hormone Antagonists/metabolism , Hormone Antagonists/pharmacology , Humans , Leptin/antagonists & inhibitors , Leptin/chemistry , Leptin/metabolism , Ligands , Protein Binding , Receptors, Leptin/analysis , Reproducibility of Results , Time Factors
12.
Am J Physiol Endocrinol Metab ; 302(3): E356-64, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22094470

ABSTRACT

Since anterior pituitary expresses prolactin receptors, prolactin secreted by lactotropes could exert autocrine or paracrine actions on anterior pituitary cells. In fact, it has been observed that prolactin inhibits its own expression by lactotropes. Our hypothesis is that prolactin participates in the control of anterior pituitary cell turnover. In the present study, we explored the action of prolactin on proliferation and apoptosis of anterior pituitary cells and its effect on the expression of the prolactin receptor. To determine the activity of endogenous prolactin, we evaluated the effect of the competitive prolactin receptor antagonist Δ1-9-G129R-hPRL in vivo, using transgenic mice that constitutively and systemically express this antagonist. The weight of the pituitary gland and the anterior pituitary proliferation index, determined by BrdU incorporation, were higher in transgenic mice expressing the antagonist than in wild-type littermates. In addition, blockade of prolactin receptor in vitro by Δ1-9-G129R-hPRL increased proliferation and inhibited apoptosis of somatolactotrope GH3 cells and of primary cultures of male rat anterior pituitary cells, including lactotropes. These results suggest that prolactin acts as an autocrine/paracrine antiproliferative and proapoptotic factor in the anterior pituitary gland. In addition, anterior pituitary expression of the long isoform of the prolactin receptor, measured by real-time PCR, increased about 10-fold in transgenic mice expressing the prolactin receptor antagonist, whereas only a modest increase in the S3 short-isoform expression was observed. These results suggest that endogenous prolactin may regulate its own biological actions in the anterior pituitary by inhibiting the expression of the long isoform of the prolactin receptor. In conclusion, our observations suggest that prolactin is involved in the maintenance of physiological cell renewal in the anterior pituitary. Alterations in this physiological role of prolactin could contribute to pituitary tumor development.


Subject(s)
Cell Proliferation/drug effects , Gene Expression Regulation , Hormone Antagonists/pharmacology , Pituitary Gland, Anterior/metabolism , Prolactin/analogs & derivatives , Prolactin/physiology , Receptors, Prolactin/metabolism , Animals , Apoptosis/drug effects , Cell Line , Cells, Cultured , Gene Expression Regulation/drug effects , Hormone Antagonists/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Organ Size , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/drug effects , Pituitary Gland, Anterior/pathology , Prolactin/antagonists & inhibitors , Prolactin/genetics , Prolactin/metabolism , Prolactin/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Prolactin/antagonists & inhibitors , Receptors, Prolactin/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Signal Transduction/drug effects
13.
Horm Metab Res ; 43(11): 816-20, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22009378

ABSTRACT

Epithelial ovarian carcinoma is the leading cause of cancer-related deaths among women with gynecologic malignancies. Antagonists of the growth hormone-releasing hormone (GHRH) have been shown to inhibit growth of various cancers through endocrine, autocrine, and paracrine mechanisms. In this study, we have investigated the effects of GHRH antagonists (GHRHa) in ES-2 human clear cell ovarian cancer and in UCI-107 human serous ovarian cancer in vitro and in vivo. We evaluated the expression of mRNA for GHRH receptor, the binding to GHRH receptors, in specimens of ES-2 ovarian cancer. We evaluated also the in vitro effects of GHRHa on ES-2 cells and the in vivo effect of 2 different GHRHa on ES-2 and UCI-107 tumors. Nude mice bearing xenografts on ES-2 and UCI-107 ovarian cancer were treated with JMR-132 and MZ-J-7-118, respectively. Tumor growth was compared to control. ES-2 cells expressed mRNA for the functional splice variant SV1 of the GHRH receptor. JMR-132 inhibited cell proliferation in vitro by 42% and 18% at 10 and 1 µM concentration, respectively. Specific high affinity receptors for GHRH were detected in ES-2 cancer samples. In vivo daily subcutaneous injections of GHRHa significantly reduced tumor growth compared to a control group in both animal models. Our results indicate that GHRHa such as JMR-132 and MZ-J-7-118 can inhibit the growth of human ovarian cancer. The efficacy of GHRHa in ovarian cancer should be assessed in clinical trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/therapeutic use , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Hormone Antagonists/metabolism , Hormone Antagonists/pharmacology , Humans , Mice , Mice, Nude , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Random Allocation , Sermorelin/analogs & derivatives , Sermorelin/pharmacology , Sermorelin/therapeutic use , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
14.
Biomed Res ; 32(5): 321-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22033301

ABSTRACT

Ciliary beat frequency (CBF) was measured in slice preparations of the Fallopian tube fimbria, using videomicroscopy with a high-speed (500 Hz) camera in guinea pigs that were treated with ß-oestradiol benzoate (ßE2B) and medroxy progesterone (mPRG). In non-ovulating guinea pigs at 4 weeks of age, the CBF of the fimbria was high (17.8 Hz). In sexually mature guinea pigs (12-16 weeks of age) with constant ovulation, the CBF varied from 12 Hz to 16 Hz. The in vivo administration of both ICI-182,780 (a blocker of ßE2 receptors) and mifepristone (a blocker of PRG receptors) induced high CBF (17.4 Hz). The administration of ßE2B at a low (3.2 mg/kg/day) or high (32 mg/kg/day) dose decreased the CBF to 14.5 Hz or 11 Hz, respectively. ICI-182,780 abolished the ßE2B-induced changes in CBF and decreased CBF to 12 Hz. The administration of mPRG (6.4 mg/kg/day) decreased CBF to 12.5 Hz. Mifepristone abolished this mPRG-induced decrease in CBF and maintained the CBF at 15 Hz. However, administering both ßE2B and mPRG increased CBF to 17.5 Hz, suggesting that ßE2B inhibits mPRG actions and vice versa. To confirm the interactions between ßE2B and mPRG, we administered both ßE2B and mPRG to guinea pigs that were pretreated for 1.5 days with either mPRG (6.4 mg/kg/day) or ßE2B (3.2 mg/kg/day). Prior treatment with ßE2B or mPRG prevented the increase in CBF that was otherwise by ßE2B plus mPRG, and maintained the CBF at 14.5 Hz or 13 Hz, respectively. The administration of ßE2B plus mPRG still induced the expression of PRG receptors, indicating that the highest CBF is not the result of no expression of the receptors. In the beating cilia of the fimbria, the signals that are activated by the ßE2 and PRG receptors are proposed to antagonize each other in regulating the frequency.


Subject(s)
Estradiol/analogs & derivatives , Fallopian Tubes/drug effects , Fallopian Tubes/physiology , Medroxyprogesterone/pharmacology , Ovary/physiology , Animals , Cilia/drug effects , Cilia/physiology , Drug Interactions , Estradiol/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Fornix, Brain/metabolism , Guinea Pigs , Hormone Antagonists/metabolism , Humans , In Vitro Techniques , Medroxyprogesterone/metabolism , Receptors, Progesterone/metabolism
15.
Bioorg Med Chem Lett ; 21(19): 5684-7, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21885275

ABSTRACT

The V1a receptor has emerged as an attractive target for a range of indications including Raynaud's disease and dysmenorrhoea. As part of an effort to discover a new class of orally active V1a antagonist, we optimised a highly lipophilic, metabolically unstable lead into a range of potent, selective and metabolically stable V1a antagonists. In this communication, we demonstrate the series-dependent effect of limiting the number of rotatable bonds in order to decrease Cytochrome P450-mediated metabolism. This effort culminated in the discovery of PF-184563, a novel, selective V1a antagonist with excellent in vitro and in vivo properties.


Subject(s)
Antidiuretic Hormone Receptor Antagonists , Benzodiazepines/chemical synthesis , Benzodiazepines/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Drug Design , Drug Discovery , Dysmenorrhea/drug therapy , Hormone Antagonists/chemical synthesis , Hormone Antagonists/pharmacology , Triazoles/chemical synthesis , Triazoles/pharmacology , Benzodiazepines/chemistry , Benzodiazepines/metabolism , Drug Stability , Female , Hormone Antagonists/chemistry , Hormone Antagonists/metabolism , Humans , Microsomes/physiology , Molecular Structure , Triazoles/chemistry , Triazoles/metabolism
16.
Brain Res Bull ; 84(6): 425-9, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21272620

ABSTRACT

An increasing number of studies suggest that nicotine/tobacco addiction is modulated by ovarian hormones. The levels of estrogen and progesterone appear to be important in the success of quit attempts and smoking cessation. In women smokers with the diagnosis or risk of breast cancer, the estrogen receptor modulator tamoxifen (TAM) is widely used, and even though the detrimental health effects of smoking are known, this vulnerable group has difficulty quitting and continues to smoke. The current study tested the effect of the estrogen receptor modulator TAM and the progesterone receptor antagonist mifepriston (RU486) on nicotine-induced conditioned place preference (CPP) in adult female rats. A three chambered CPP apparatus was used and nicotine was paired with the initially non-preferred chamber. Rats received nicotine or saline and hormone receptor modulators (vehicle, TAM, RU486) in a 2×3 experimental design. We have previously shown that nicotine induces CPP in male Sprague-Dawley rats but not in females. Our results show that while nicotine alone does not induce CPP in female rats, rats treated with TAM exhibit nicotine-induced CPP. Although RU486 has an aversive effect when applied alone, this is ameliorated by nicotine. These results confirm the role of ovarian hormone receptors in nicotine-induced CPP and may have clinical implications for developing more efficient smoking cessation approaches in women smokers.


Subject(s)
Conditioning, Psychological/drug effects , Mifepristone/metabolism , Mifepristone/pharmacology , Nicotine/metabolism , Nicotine/pharmacology , Tamoxifen/metabolism , Tamoxifen/pharmacology , Animals , Female , Hormone Antagonists/metabolism , Hormone Antagonists/pharmacology , Humans , Male , Nicotinic Agonists/metabolism , Nicotinic Agonists/pharmacology , Rats , Rats, Sprague-Dawley , Selective Estrogen Receptor Modulators/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Smoking Cessation , Tobacco Use Disorder
17.
Brain Behav Evol ; 75(4): 282-91, 2010.
Article in English | MEDLINE | ID: mdl-20693783

ABSTRACT

The neuropeptide arginine vasotocin (AVT) is well known to modulate both aggression and affiliation, yet few studies relate individual behavioral state to a quantitative assessment of AVT distribution in the brain. Here, using a wild population of beaugregory damselfish, Stegastes leucostictus, we assess: (1) the effect of AVT on courtship, and (2) with reference to our previous study on AVT modulation of aggression in this species, the relationship between AVT-like immunoreactive (ir) fiber distribution in the forebrain's preoptic area and individual courtship and aggression levels. Exogenous AVT did not affect courtship, yet Manning compound, an arginine vasopressin (AVP) V1a receptor antagonist, significantly lowered but did not eradicate courtship. Consistent with AVT's known facilitation of aggression in this species, the density of AVT-ir fibers in the preoptic area was significantly negatively correlated to aggression. Our findings match similar behavioral and immunoreactive patterns of neuropeptide secretion in other taxa. Unlike aggression, preoptic AVT-ir fiber density was not significantly correlated to individual courtship levels. The results suggest a differential involvement of preoptic AVT neurons and/or their receptors in supporting the expression of aggression and courtship.


Subject(s)
Aggression/physiology , Courtship , Neurons/cytology , Perciformes/physiology , Sexual Behavior, Animal/physiology , Vasotocin/metabolism , Animals , Antidiuretic Hormone Receptor Antagonists , Arginine Vasopressin/analogs & derivatives , Arginine Vasopressin/metabolism , Female , Hormone Antagonists/metabolism , Male , Neural Pathways/cytology , Neural Pathways/metabolism , Neurons/metabolism , Oxytocin/analogs & derivatives , Oxytocin/metabolism , Perciformes/anatomy & histology , Preoptic Area/cytology , Preoptic Area/metabolism , Prosencephalon/cytology , Prosencephalon/metabolism , Receptors, Vasopressin/metabolism
18.
J Comp Neurol ; 518(16): 3359-80, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20575072

ABSTRACT

The fruitfly, Drosophila, is dependent on its olfactory sense in food search and reproduction. Processing of odorant information takes place in the antennal lobes, the primary olfactory center in the insect brain. Besides classical neurotransmitters, earlier studies have indicated the presence of a few neuropeptides in the olfactory system. In the present study we made an extensive analysis of the expression of neuropeptides in the Drosophila antennal lobes by direct profiling using matrix-assisted laser desorption/ionization-time-of-flight (MALDI-TOF) mass spectrometry and immunocytochemistry. Neuropeptides from seven different precursor genes were unambiguously identified and their localization in neurons was subsequently revealed by immunocytochemistry. These were short neuropeptide F, tachykinin related peptide, allatostatin A, myoinhibitory peptide, SIFamide, IPNamide, and myosuppressin. The neuropeptides were expressed in subsets of olfactory sensory cells and different populations of local interneurons and extrinsic (centrifugal) neurons. In some neuron types neuropeptides were colocalized with classical neurotransmitters. Our findings suggest a huge complexity in peptidergic signaling in different circuits of the antennal lobe.


Subject(s)
Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila melanogaster/anatomy & histology , Neuropeptides/chemistry , Neuropeptides/metabolism , Amino Acid Sequence , Animals , Brain/anatomy & histology , Brain/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Female , Hormone Antagonists/metabolism , Interneurons/cytology , Interneurons/metabolism , Male , Molecular Sequence Data , Neuropeptides/genetics , Olfactory Pathways/anatomy & histology , Olfactory Pathways/physiology , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/metabolism , Signal Transduction/physiology , Smell/physiology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
19.
Inflamm Res ; 59(12): 1053-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20574769

ABSTRACT

OBJECTIVE: To explore the site of action of maprotiline, as an atypical antidepressant, on carrageenan-induced paw edema. SUBJECTS: Male Wistar rats were used. METHODS: Firstly, the anti-inflammatory effect of systemic maprotiline (12.5, 25 and 50 mg kg(-1)) was assessed using a paw edema model. Secondly, different doses of maprotiline were administrated intracerebroventricularly, intrathecally and locally before carrageenan challenge. Finally, we tried to reverse the anti-inflammatory effect of maprotiline by propranolol (10 mg kg(-1)), prazosin (4 mg kg(-1)), yohimbine (10 mg kg(-1)), naloxone (4 mg kg(-1)) and mifepristone (5 mg kg(-1)). RESULTS: Systemic, intracerebroventricular and subplantar application of maprotiline significantly inhibited peripheral edema, but intrathecal maprotiline did not alter the degree of paw swelling. The applied antagonists failed to change the anti-inflammatory activity of maprotiline. CONCLUSION: These results demonstrate that maprotiline has a potent anti-inflammatory effect and this effect is linked to the peripheral and supraspinal actions of the drug.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antidepressive Agents, Second-Generation/therapeutic use , Carrageenan/pharmacology , Edema , Maprotiline/therapeutic use , Adrenergic alpha-1 Receptor Antagonists/metabolism , Adrenergic alpha-2 Receptor Antagonists/metabolism , Adrenergic beta-Antagonists/metabolism , Animals , Anti-Inflammatory Agents/metabolism , Antidepressive Agents, Second-Generation/metabolism , Edema/chemically induced , Edema/drug therapy , Hormone Antagonists/metabolism , Indomethacin/metabolism , Indomethacin/therapeutic use , Injections, Spinal , Male , Maprotiline/metabolism , Mifepristone/metabolism , Naloxone/metabolism , Narcotic Antagonists/metabolism , Prazosin/metabolism , Propranolol/metabolism , Rats , Rats, Wistar , Yohimbine/metabolism
20.
J Comp Neurol ; 518(9): 1391-417, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20187126

ABSTRACT

Information transmission and processing in the brain is achieved through a small family of chemical neurotransmitters and neuromodulators and a very large family of neuropeptides. In order to understand neural networks in the brain it will be necessary, therefore, to understand the connectivity, morphology, and distribution of peptidergic neurons, and to elucidate their function in the brain. In this study we characterize the distribution of substances related to Dip-allatostatin I in the honeybee brain, which belongs to the allatostatin-A (AST) peptide family sharing the conserved c-terminal sequence -YXFGL-NH(2). We found about 500 AST-immunoreactive (ASTir) neurons in the brain, scattered in 18 groups that varied in their precise location across individuals. Almost all areas of the brain were innervated by ASTir fibers. Most ASTir neurites formed networks within functionally distinct areas, e.g., the antennal lobes, the mushroom bodies, or the optic lobes, indicating local functions of the peptide. A small number of very large neurons had widespread arborizations and neurites were found in the corpora cardiaca and in the cervical connectives, suggesting that AST also has global functions. We double-stained AST and GABA and found that a subset of ASTir neurons were GABA-immunoreactive (GABAir). Double staining AST with backfills of olfactory receptor neurons or mass fills of neurons in the antennal lobes and in the mushroom bodies allowed a more fine-grained description of ASTir networks. Together, this first comprehensive description of AST in the bee brain suggests a diverse functional role of AST, including local and global computational tasks.


Subject(s)
Bees/metabolism , Hormone Antagonists/metabolism , Neural Pathways , Neurons/metabolism , Neuropeptides/metabolism , Amino Acid Sequence , Animals , Bees/anatomy & histology , Brain/cytology , Brain/metabolism , Ganglia, Invertebrate/cytology , Ganglia, Invertebrate/metabolism , Immunohistochemistry , Molecular Sequence Data , Mushroom Bodies/cytology , Mushroom Bodies/metabolism , Neural Pathways/anatomy & histology , Neural Pathways/metabolism , Neurons/cytology , Neuropeptides/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...