Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Rev Endocr Metab Disord ; 12(3): 141-51, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21340584

ABSTRACT

Here we review the emerging neurobiological understanding of the role of the brain's reward system in the regulation of body weight in health and in disease. Common obesity is characterized by the over-consumption of palatable/rewarding foods, reflecting an imbalance in the relative importance of hedonic versus homeostatic signals. The popular 'incentive salience theory' of food reward recognises not only a hedonic/pleasure component ('liking') but also an incentive motivation component ('wanting' or 'reward-seeking'). Central to the neurobiology of the reward mechanism is the mesoaccumbal dopamine system that confers incentive motivation not only for natural rewards such as food but also by artificial rewards (eg. addictive drugs). Indeed, this mesoaccumbal dopamine system receives and integrates information about the incentive (rewarding) value of foods with information about metabolic status. Problematic over-eating likely reflects a changing balance in the control exerted by hypothalamic versus reward circuits and/or it could reflect an allostatic shift in the hedonic set point for food reward. Certainly, for obesity to prevail, metabolic satiety signals such as leptin and insulin fail to regain control of appetitive brain networks, including those involved in food reward. On the other hand, metabolic control could reflect increased signalling by the stomach-derived orexigenic hormone, ghrelin. We have shown that ghrelin activates the mesoaccumbal dopamine system and that central ghrelin signalling is required for reward from both chemical drugs (eg alcohol) and also from palatable food. Future therapies for problematic over-eating and obesity may include drugs that interfere with incentive motivation, such as ghrelin antagonists.


Subject(s)
Body Weight/physiology , Brain/physiology , Dopamine/metabolism , Eating/physiology , Obesity/metabolism , Reward , Animals , Appetite/physiology , Food Preferences/physiology , Humans , Obesity/physiopathology
2.
Mol Cell Endocrinol ; 315(1-2): 121-30, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-19818377

ABSTRACT

Progesterone, acting via the nuclear progesterone receptor (PGR), reduces apoptosis in periovulatory granulosa cells, and is a likely mediator of the anti-atretic actions of LH. The underlying mechanisms, however, have not been clearly defined. In this study, we sought to identify progesterone-mediated transcriptional changes involved in apoptosis regulation. Granulosa cells from immature, gonadotropin-primed female rats were treated in vitro with 100 nM of the PGR antagonist Org 31710. Transcriptional effects were analyzed after 5 and 22 h of incubation using microarrays, and the expression of 85 genes was subsequently measured by quantitative PCR. Follow-up experiments focused on genes related to the functional group "apoptosis". We have identified novel, early gene targets of PGR that may be involved in the control of apoptosis and other biologically significant functions in periovulatory granulosa cells. This study expands our knowledge of events that occur during the processes of ovulation and luteinization.


Subject(s)
Estrenes/pharmacology , Furans/pharmacology , Granulosa Cells/drug effects , Granulosa Cells/physiology , Hormone Antagonists/pharmacology , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/metabolism , Transcription, Genetic/drug effects , Animals , Apoptosis , Cells, Cultured , Female , Gene Expression Profiling , Granulosa Cells/cytology , Luteinization/drug effects , Luteinization/physiology , Luteinizing Hormone/metabolism , Microarray Analysis , Ovulation/drug effects , Ovulation/physiology , Rats , Rats, Sprague-Dawley , Receptors, Progesterone/genetics
3.
Biol Reprod ; 80(6): 1160-7, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19208546

ABSTRACT

In this study, it was hypothesized that progesterone (P4) acts as a survival factor primarily by actions of the classic nuclear progesterone receptor (PGR) signaling pathway in rat periovulatory granulosa cells. Granulosa cells were isolated from immature female rats primed with equine chorionic gonadotropin/human chorionic gonadotropin and treated in vitro with PGR antagonists. As little as 10 nM of two different PGR antagonists (Org 31710 and RU 486) increased apoptosis measured as caspase 3/7 activity, which was reversed by cotreatment with the progestin R5020. Concurrently, P4 synthesis was decreased. Inhibition of P4 synthesis by cyanoketone similarly induced apoptosis but required greater inhibition of P4 synthesis than that seen after treatment with PGR antagonists. Therefore, the induction of apoptosis by PGR antagonists cannot be explained by decreased P4 synthesis alone. Low concentrations of R5020 also completely reversed the effects of cyanoketone. Inhibition of P4 synthesis was more effective in inducing apoptosis than treatment with PGR antagonists. However, cotreatment with PGR antagonists protected cells from the additional effects of cyanoketone, indicating partial agonist effects of the antagonists and a dominating role for PGR in P4-mediated regulation of apoptosis. Progesterone receptor membrane component 1 (PGRMC1) was expressed in granulosa cells; however, an anti-PGRMC1 antibody did not induce apoptosis in periovulatory granulosa cells. Neither anti-PGRMC1 nor P4 or cyanoketone affected apoptosis of immature granulosa cells. In conclusion, we show that P4 regulates apoptosis in periovulatory granulosa cells by acting via the classic nuclear receptor.


Subject(s)
Apoptosis/drug effects , Estrenes/pharmacology , Furans/pharmacology , Granulosa Cells/drug effects , Mifepristone/pharmacology , Receptors, Progesterone/antagonists & inhibitors , Animals , Antibodies, Blocking/pharmacology , Cyanoketone/pharmacology , Dose-Response Relationship, Drug , Female , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Membrane Proteins/metabolism , Progesterone/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Progesterone/immunology , Receptors, Progesterone/metabolism
4.
Mol Reprod Dev ; 74(10): 1317-26, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17342739

ABSTRACT

Progesterone is a survival factor in rat periovulatory granulosa cells. The mechanisms involved are unclear but progesterone receptor (PGR) antagonists have been shown to inhibit cholesterol synthesis and induce apoptosis. Furthermore, reports suggest that statins induce apoptosis by inhibition of protein isoprenylation. Statins inhibit the rate-limiting step of the cholesterol synthesis, thereby reducing availability of intermediates used for the post-translational isoprenylation process. It has been suggested that PGR antagonists in a similar manner induce apoptosis by decreasing cholesterol synthesis and thereby protein isoprenylation. In this study we hypothesized that the mechanism by which the nuclear PGR antagonist Org 31,710 induces apoptosis in rat periovulatory granulosa cells, is by decreasing cholesterol synthesis and thereby general cell protein isoprenylation. Incubation of isolated granulosa cells with Org 31,710 or simvastatin for 22 hr resulted in increased apoptosis and reduced cholesterol synthesis. However, simvastatin caused a substantial inhibition of cholesterol synthesis after 6 hr in culture without inducing apoptosis. In contrast, Org 31,710 had only a modest effect on cholesterol synthesis after 6 hr while it significantly induced apoptosis. Addition of isoprenylation substrates partially reversed apoptosis induced by simvastatin and to a lesser extent apoptosis induced by Org 31,710. In addition, and in contrast to Org 31,710, simvastatin caused a decrease in isoprenylation of a selected isoprenylation marker protein, the Ras-related protein RAB11. In conclusion, we demonstrate that the PGR antagonist inhibits cholesterol synthesis in granulosa cells but reduced protein isoprenylation is not the mediating mechanism of increased apoptosis as previously hypothesized.


Subject(s)
Apoptosis/drug effects , Estrenes/pharmacology , Furans/pharmacology , Granulosa Cells/drug effects , Protein Prenylation/drug effects , Receptors, Progesterone/antagonists & inhibitors , Animals , Cells, Cultured , Cholesterol/metabolism , Female , Granulosa Cells/cytology , Granulosa Cells/metabolism , Hormone Antagonists/pharmacology , Hypolipidemic Agents/pharmacology , Protein Prenylation/physiology , Rats , Rats, Sprague-Dawley , Simvastatin/pharmacology
5.
Mol Reprod Dev ; 73(10): 1277-83, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16868926

ABSTRACT

Progesterone receptor (PR) stimulation promotes survival in human and rat periovulatory granulosa cells. PR antagonists, Org 31710 and RU 486, both increase apoptosis and decrease cholesterol synthesis in these cells. The decrease in cholesterol synthesis also causes decreased synthesis of other products branching from the cholesterol synthesis pathway, including substrates for protein prenylation. In this study we focus on the link between apoptosis and prenylation in human periovulatory granulosa cells. A decreased cholesterol synthesis and increased apoptosis was verified in experiments with human periovulatory granulosa cells treated with the PR antagonists Org 31710 or RU 486 by measuring caspase-3/7 activity and incorporation of 14C-acetate into cholesterol and progesterone. Correspondingly, specific inhibition of cholesterol synthesis in periovulatory human granulosa cells using HMG-CoA reductase inhibitors (lovastatin or simvastatin) increased apoptosis, measured as caspase-3/7 activity. The increase in apoptosis caused by simvastatin or Org 31710 was partially reversed by addition of the protein prenylation precursors farnesol or geranylgeraniol. In addition, the prenylation inhibitors FTI R115777 and GGTI 2147 increased apoptosis in these cells. In conclusion our data suggest that PR antagonists increase apoptosis and reduce cholesterol synthesis in periovulatory granulosa cells and that the resulting depletion of substrates for protein prenylation may contribute to the increased apoptosis sensitivity.


Subject(s)
Apoptosis , Cholesterol/biosynthesis , Granulosa Cells/physiology , Protein Prenylation/drug effects , Apoptosis/drug effects , Cells, Cultured , Diterpenes/pharmacology , Estrenes/pharmacology , Farnesol/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Female , Furans/pharmacology , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Hormone Antagonists/pharmacology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Imidazoles/pharmacology , Leucine/analogs & derivatives , Leucine/pharmacology , Mifepristone/pharmacology , Ovulation , Quinolones/pharmacology , Receptors, Progesterone/antagonists & inhibitors
6.
Biol Reprod ; 72(3): 538-45, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15385411

ABSTRACT

Progesterone-receptor (PR) stimulation promotes survival in rat and human periovulatory granulosa cells. To investigate the mechanisms involved, periovulatory rat granulosa cells were incubated in vitro with or without the PR-antagonist Org 31710. Org 31710 caused the expected increase in apoptosis, and expression profiling using cDNA microarray analysis revealed regulation of several groups of genes with functional and/or metabolic connections. This regulation included decreased expression of genes involved in follicular rupture, increased stress responses, decreased angiogenesis, and decreased cholesterol synthesis. A decreased cholesterol synthesis was verified in experiments with both rat and human periovulatory granulosa cells treated with the PR-antagonists Org 31710 or RU 486 by measuring incorporation of [14C]acetate into cholesterol, cholesterol ester, and progesterone. Correspondingly, specific inhibition of cholesterol synthesis in periovulatory rat granulosa cells using 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors (lovastatin, mevastatin, or simvastatin) increased apoptosis, measured as DNA fragmentation and caspase-3/7 activity. The increase in apoptosis caused by simvastatin was reversed by addition of the cholesterol synthesis-intermediary mevalonic acid. These results show that PR antagonists reduce cholesterol synthesis in periovulatory granulosa cells and that cholesterol synthesis is important for granulosa cell survival.


Subject(s)
Apoptosis/physiology , Cholesterol/biosynthesis , Granulosa Cells/metabolism , Ovulation/physiology , Receptors, Progesterone/metabolism , Animals , Apoptosis/drug effects , Cell Survival/physiology , Estrenes/pharmacology , Female , Furans/pharmacology , Gene Expression Profiling , Gene Expression Regulation , Granulosa Cells/drug effects , Hormone Antagonists/pharmacology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , In Vitro Techniques , Mifepristone/pharmacology , Oligonucleotide Array Sequence Analysis , Rats , Rats, Sprague-Dawley , Receptors, Progesterone/antagonists & inhibitors , Signal Transduction/physiology
7.
Biol Reprod ; 68(3): 914-21, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12604642

ABSTRACT

The intracellular progesterone receptor (PR) in the mammalian ovary is a part of the physiological pathway that facilitates ovulation. Two PR isoforms (A and B) exist, with different molecular and biological functions. Previous studies have revealed that the cellular ratio of the PR isoforms is important for progesterone-responsive tissues and is under developmental control in different species. However, the relative expression of PR isoforms in the ovary is unknown. In this study we have demonstrated first that the expression of both PR isoforms in mouse granulosa cells was rapidly up-regulated by hCG treatment and dramatically down-regulated when the granulosa cells were undergoing luteinization. The relative level of protein expression of the A and B forms was 2:1 and the highest total PR protein expression was found after hCG stimulation. Second, we demonstrated that the expression of PR protein was specific to granulosa cells of periovulatory follicles and was absent in undifferentiated granulosa cells of growing follicles. It was not detected in other cell types (i.e., corpora lutea or any stage of follicles with features of apoptosis). Third, we demonstrated that treatment with the PR antagonist RU 486 in vivo resulted in down-regulation of both isoforms in parallel with increased activation of caspase-3, a decreased level of proliferating cell nuclear antigen, and a reduced rate of ovulation. Fourth, we demonstrated, in vitro, that the PR antagonists RU 486 and Org 31710 increased internucleosomal DNA fragmentation parallel with a decrease in DNA synthesis in granulosa cells, which express PR. These results indicate that PR and its isoforms participate in regulation of ovulation, along with suppression of granulosa cell apoptosis and promotion of cell survival in the mouse ovary.


Subject(s)
Apoptosis/physiology , Granulosa Cells/metabolism , Ovarian Follicle/metabolism , Receptors, Progesterone/physiology , Animals , Blotting, Western , Caspase 3 , Caspases/metabolism , Cell Division/physiology , Chorionic Gonadotropin/pharmacology , Estrenes/pharmacology , Female , Furans/pharmacology , Granulosa Cells/cytology , Hormone Antagonists/pharmacology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mifepristone/pharmacology , Ovarian Follicle/cytology , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/pharmacology , Protein Isoforms , Random Allocation , Receptors, Progesterone/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...