Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Steroid Biochem Mol Biol ; 237: 106440, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38048919

RESUMEN

Progestins used in hormonal contraceptives and menopausal hormone therapy (MHT) have been linked to increased breast cancer risk. Whether the association holds for all progestins is unclear and the underlying mechanisms remain poorly understood. We directly compared the effects of four progestins (medroxyprogesterone acetate (MPA), norethisterone acetate (NET-A), levonorgestrel (LNG) and drospirenone (DRSP)) to each other and the natural progestogen progesterone (P4) on selected cancer hallmarks. To provide mechanistic insight into these effects, we assessed the role of the progesterone receptor (PR), and the extracellular signal-related kinase (ERK1/2) and c-Jun N terminal (JNK) signaling pathways. We showed that the increased proliferation of the luminal T47D breast cancer cell line by P4 and all progestins, albeit to different extents, was inhibited by PR knockdown and inhibition of both the ERK1/2 and JNK pathways. While knockdown of the PR also blocked the upregulation of MKI67 and CCND1 mRNA expression by selected progestogens, only a role for the ERK1/2 pathway could be established in these effects. Similarly, only a role for the ERK1/2 pathway could be confirmed for progestogen-induced colony formation, whereas both the ERK1/2 and JNK pathways were required for cell migration in response to the three older progestins implicated in the etiology of breast cancer, MPA, NET-A and LNG. Together our results show that all the progestins elicit their effects on cell proliferation via a mechanism requiring the PR, ERK1/2 and JNK pathways. While the ERK1/2 and JNK pathways are also required for increased cell migration by the older progestins, only a role for the ERK1/2 pathway could be established in their effects on colony formation. Notably, the cytoplasmic PR was not needed for activation of the ERK1/2 pathway by the progestogens. Given that DRSP showed significantly lower proliferation than MPA and NET-A, and that it had no effect on breast cancer cell migration and colony formation, hormonal formulations containing the newer generation progestin DRSP may provide a better benefit/risk profile towards breast cancer than those containing the older generation progestins.


Asunto(s)
Neoplasias de la Mama , Progestinas , Humanos , Femenino , Progestinas/farmacología , Progestinas/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Sistema de Señalización de MAP Quinasas , Progesterona/metabolismo , Acetato de Medroxiprogesterona/farmacología , Acetato de Medroxiprogesterona/metabolismo , Levonorgestrel , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo
2.
PLoS One ; 18(12): e0295959, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38134043

RESUMEN

Progestin-only injectable contraceptives, mainly depo-medroxyprogesterone acetate intramuscular (DMPA-IM), are the most widely used contraceptive methods in sub-Saharan Africa. Insufficient robust data on their relative side-effects and serum concentrations limit understanding of reported outcomes in contraception trials. The WHICH clinical trial randomized HIV-negative women to DMPA-IM (n = 262) or norethisterone enanthate (NET-EN) (n = 259) at two South African sites between 2018-2019. We measured serum concentrations of study and non-study progestins at initiation (D0) and peak serum levels, one week after the 24-week injection [25 weeks (25W)], (n = 435) and investigated associations between study progestin levels, and BMI and weight of participants. Peak median serum concentrations were 6.59 (IQR 4.80; 8.70) nM for medroxyprogesterone (MPA) (n = 161) and 13.6 (IQR 9.01; 19.0) nM for norethisterone (NET) (n = 155). MPA was the most commonly quantifiable non-study progestin at D0 in both arms (54%) and at 25W in the NET-EN arm (27%), followed by NET at D0 in both arms (29%) and at 25W in the DMPA-IM arm (19%). Levonorgestrel was quantifiable in both arms [D0 (6.9%); 25W (3.4%)], while other progestins were quantifiable in ≤ 14 participants. Significant negative time-varying associations were detected between MPA and NET concentrations and weight and BMI in both contraceptive arms and a significant increase was detected for peak serum progestin concentrations for normal weight versus obese women. Contraceptive-related reported outcomes are likely confounded by MPA, more so than NET, with reported DMPA-IM effects likely underestimated, at sites where DMPA-IM is widely used, due to misreporting of contraceptive use before and during trials, and 'tail' effects of DMPA-IM use more than six months before trial enrolment. Peak serum levels of MPA and NET are negatively associated with BMI and weight, suggesting another source of variability between trial outcomes and a potential increase in side-effects for normal weight versus overweight and obese women. Trail registration: The clinical trial was registered with the Pan African Clinical Trials Registry (PACTR 202009758229976).


Asunto(s)
Acetato de Medroxiprogesterona , Progestinas , Femenino , Humanos , Acetato de Medroxiprogesterona/efectos adversos , Anticonceptivos , Índice de Masa Corporal , Noretindrona/farmacología , Obesidad
3.
Biochem Biophys Res Commun ; 671: 286-291, 2023 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-37315428

RESUMEN

The 3T3-L1 murine adipocyte cell line remains one of the most widely used models to study the mechanisms of obesity and related pathologies. Most studies investigate such mechanisms using mature adipocytes that have been chemically induced to differentiate for 7 days in media containing 25 mM glucose. However, the dysfunctional characteristics commonly observed in obesity including adipocyte hypertrophy, increased expression of inflammatory markers, enhanced production of reactive oxygen species (ROS), increased steroidogenic enzyme expression/activity and production of steroid hormones, are not necessarily mimicked in these cells. The aim of this study was to provide an inexpensive model which represents the well-known characteristics of obesity by manipulating the time of adipocyte differentiation and increasing the concentration of glucose in the cell media. Our results showed a glucose- and time-dependent increase in adipocyte hypertrophy, ROS production and gene expression of the pro-inflammatory cytokine interleukin-6 (IL-6), as well as a time-dependent increase in lipolysis and in the gene expression of the chemokine monocyte chemoattractant protein 1 (MCP1). We also showed that gene expression of the steroidogenic enzymes 11-beta-hydroxysteroid dehydrogenase type 1 (11ßHSD1), 17ßHSD type 7 and 12, as well as CYP19A1 (aromatase), were significantly higher in the hypertrophic model relative to the control adipocytes differentiated using the conventional method. The increase in 11ßHSD1 and 17ßHSD12 expression was consistent with the enhanced conversion of cortisone and androstenedione to cortisol and testosterone, respectively. As these characteristics reflect those commonly observed in obesity, hypertrophic 3T3-L1 adipocytes are an appropriate in vitro model to study mechanisms of adipocyte dysfunction in an era where the rise in obesity incidence is a global health concern, and where access to adipose tissue from obese patients are limited.


Asunto(s)
Adiposidad , Glucosa , Humanos , Ratones , Animales , Glucosa/metabolismo , Células 3T3-L1 , Especies Reactivas de Oxígeno/metabolismo , Adipocitos/metabolismo , Obesidad/metabolismo , Diferenciación Celular/genética , Hipertrofia/metabolismo
4.
J Steroid Biochem Mol Biol ; 232: 106348, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37315868

RESUMEN

Progestins (synthetic progestogens) are progesterone receptor (PR) ligands used globally by women in both hormonal contraception and menopausal hormone therapy. Although four generations of unique progestins have been developed, studies seldom distinguish between the activities of progestins via the two functionally distinct PR isoforms, PR-A and PR-B. Moreover, not much is known about the action of progestins in breast cancer tumors where PR-A is mostly overexpressed relative to PR-B. Understanding progestin action in breast cancer is crucial since the clinical use of some progestins has been associated with an increased risk of developing breast cancer. This study directly compared the agonist activities of selected progestins from all four generations for transactivation and transrepression via either PR-A or PR-B, and when PR-A and PR-B were co-expressed at ratios comparable to those detected in breast cancer tumors. Comparative dose-response analysis showed that earlier generation progestins mostly displayed similar efficacies for transactivation on a minimal progesterone response element via the PR isoforms, while most of the 4th generation progestins, similar to the natural progestogen, progesterone (P4), were more efficacious via PR-B. Most of the progestogens were however more potent via PR-A. We are the first to show that the efficacies of the selected progestogens via the individual PR isoforms were generally decreased when PR-A and PR-B were co-expressed, irrespective of the ratio of PR-A:PR-B. While the potencies of most progestogens via PR-B were enhanced when the ratio of PR-A relative to PR-B was increased, those via PR-A were minimally influenced. This study is also the first to report that all progestogens evaluated, except 1st generation medroxyprogesterone acetate and 4th generation drospirenone, displayed similar agonist activity for transrepression via PR-A and PR-B on a minimal nuclear factor kappa B containing promoter. Moreover, we showed that the progestogen activity for transrepression was significantly increased when PR-A and PR-B were co-expressed. Taken together, our results highlight that PR agonists (progestogens) do not always display the same activity via PR-A and PR-B, or when PR-A and PR-B are co-expressed at ratios mimicking those found in breast cancer tumors. These results suggest that biological responses are progestogen- and PR isoform-dependent and may differ in target tissues expressing varying PR-A:PR-B ratios.


Asunto(s)
Neoplasias de la Mama , Progestinas , Femenino , Humanos , Progestinas/farmacología , Progesterona/farmacología , Receptores de Progesterona/genética , Acetato de Medroxiprogesterona , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética
5.
Biochem Biophys Res Commun ; 639: 70-76, 2023 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-36470074

RESUMEN

Studies directly comparing the efficacies and potencies of multiple progestins used in contraception and menopausal hormone therapy (MHT) in parallel via human progesterone receptor isoform A (PR-A) in the same model system are limited, and how these parameters are influenced by the density of PR-A are unclear. This is surprising as it is known that the expression levels of PR-A vary in different tissues and diseases. We thus determined for the first time the relative efficacies and potencies for transactivation of the natural PR ligand, progesterone (P4), the PR-specific agonist promegestone (R5020), and selected progestins from all four generations in parallel via different densities of PR-A overexpressed in the MDA-MB-231 breast cancer cell line. Comparative dose-response analysis showed that P4, R5020, the 1st generation progestins medroxyprogesterone acetate and norethisterone, 2nd generation progestin levonorgestrel, 3rd generation progestin gestodene, as well as 4th generation progestins nesterone, nomegestrol acetate and drospirenone display differential agonist efficacies and potencies via PR-A. Moreover, we showed that the agonist efficacies and potencies of the progestins via PR-A were modulated in a density- and progestin-specific manner. Our finding that the potencies of the progestins via PR-A, at all densities, do not exceed reported progestin serum concentrations in women, suggest that these progestins are likely to elicit similar effects in vivo. We are the first to report that P4 and the selected progestins display similar agonist activity for transrepression via PR-A, and that the density of PR-A enhances the transrepression activity of some, but not all progestogens. Collectively, our findings provide proof of concept that the effects of the selected progestins via PR-A is progestin-specific and dependent on the density of the receptor, suggesting differential progestin responses in women using these progestins in contraception and MHT.


Asunto(s)
Progestinas , Receptores de Progesterona , Femenino , Humanos , Anticoncepción , Menopausia , Progesterona/farmacología , Progesterona/metabolismo , Congéneres de la Progesterona/farmacología , Progestinas/farmacología , Progestinas/metabolismo , Promegestona , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Transcripción Genética
6.
Front Endocrinol (Lausanne) ; 13: 959396, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36187129

RESUMEN

Progestins, synthetic compounds designed to mimic the activity of natural progesterone (P4), are used globally in menopausal hormone therapy. Although the older progestins medroxyprogesterone acetate (MPA) and norethisterone (NET) have been implicated in increased breast cancer risk, little is known regarding newer progestins, and no significant risk has been associated with P4. Considering that breast cancer is the leading cause of mortality in women, establishing which progestins increase breast cancer incidence and elucidating the underlying mechanisms is a global priority. We showed for the first time that the newer-generation progestin drospirenone (DRSP) is the least potent progestin in terms of proliferation of the estrogen-responsive MCF-7 BUS breast cancer cell line, while NET and P4 have similar potencies to estradiol (E2), the known driver of breast cancer cell proliferation. Notably, MPA, the progestin most frequently associated with increased breast cancer risk, was significantly more potent than E2. While all the progestogens enhanced the anchorage-independent growth of the MCF-7 BUS cell line, MPA promoted a greater number of colonies than P4, NET or DRSP. None of the progestogens inhibited E2-induced proliferation and anchorage-independent growth. We also showed that under non-estrogenic conditions, MPA and NET, unlike P4 and DRSP, increased the expression of the estrogen receptor (ER) target gene, cathepsin D, via a mechanism requiring the co-recruitment of ERα and the progesterone receptor (PR) to the promoter region. In contrast, all progestogens promoted the association of the PR and ERα on the promoter of the PR target gene, MYC, thereby increasing its expression under non-estrogenic and estrogenic conditions. These results suggest that progestins differentially regulate the way the PR and ER converge to modulate the expression of PR and ER-regulated genes. Our novel findings indicating similarities and differences between P4 and the progestins, emphasize the importance of comparatively investigating effects of individual progestins rather than grouping them as a class. Further studies are required to underpin the clinical relevance of PR/ERα crosstalk in response to different progestins in both normal and malignant breast tissue, to either confirm or refute their suitability in combination therapy for ER-positive breast cancer.


Asunto(s)
Neoplasias de la Mama , Receptores de Progesterona , Neoplasias de la Mama/patología , Catepsina D/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Acetato de Medroxiprogesterona/efectos adversos , Noretindrona/efectos adversos , Progesterona/farmacología , Congéneres de la Progesterona/efectos adversos , Progestinas/farmacología , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Regulación hacia Arriba
7.
Steroids ; 182: 108998, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35271867

RESUMEN

The glucocorticoid receptor (GR) regulates transcription of genes involved in multiple processes. Medroxyprogesterone acetate (MPA), widely used in the injectable contraceptive Depo-MPA (DMPA), has off-target effects via the GR, which may result in side-effects in endocrine therapy. However, very little is known about the GR activity of other progestins used in endocrine therapy. This study compared GR activities for several progestins, using whole cell binding, dose-response, and GR phosphorylation assays, in both a cell line model and peripheral blood mononuclear cells (PBMCs). MPA, etonogestrel (ETG) and nestorone (NES) exhibit greater relative binding affinities for the GR than levonorgestrel (LNG) and norethisterone/norethindrone (NET) and are partial GR agonists for transactivation but agonists for transrepression on synthetic promoters in COS-1 cells. MPA is a potent agonist for endogenous GR-regulated GILZ and IL6 genes in PBMCs. While ETG and NES also display agonist activity on IL6, they have little effect on GILZ. In contrast, LNG and NET exhibit little to no activity in transactivation models, while both exhibit some transrepressive activity but are generally less potent and/or efficacious than MPA. Antagonist and phosphorylation assays confirmed that MPA and NES act via the GR on endogenous genes in PBMCs. Our results suggest GR-mediated dose-dependent and gene-specific transcriptional side-effects are likely to occur at physiologically relevant concentrations in vivo for MPA, may possibly occur selectively for ETG and NES, but are unlikely to occur for LNG and NET. This suggests that these progestins will exhibit differential side-effects in endocrine therapy via the GR.


Asunto(s)
Acetato de Medroxiprogesterona , Progestinas , Animales , Células COS , Chlorocebus aethiops , Glucocorticoides/farmacología , Interleucina-6 , Leucocitos Mononucleares/metabolismo , Levonorgestrel , Acetato de Medroxiprogesterona/metabolismo , Noretindrona/metabolismo , Receptores de Glucocorticoides/metabolismo
8.
Endocrinology ; 162(3)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33340399

RESUMEN

Androgens are the obligatory precursors of estrogens. In humans, classic androgen biosynthesis yields testosterone, thought to represent the predominant circulating active androgen both in men and women. However, recent work has shown that 11-ketotestosterone, derived from the newly described 11-oxygenated androgen biosynthesis pathway, makes a substantial contribution to the active androgen pool in women. Considering that classic androgens are the obligatory substrates for estrogen biosynthesis catalyzed by cytochrome P450 aromatase, we hypothesized that 11-oxygenated androgens are aromatizable. Here we use steroid analysis by tandem mass spectrometry to demonstrate that human aromatase generates 11-oxygenated estrogens from 11-oxygenated androgens in 3 different cell-based aromatase expression systems and in human ex vivo placenta explant cultures. We also show that 11-oxygenated estrogens are generated as a byproduct of the aromatization of classic androgens. We show that 11ß-hydroxy-17ß-estradiol binds and activates estrogen receptors α and ß and that 11ß-hydroxy-17ß-estradiol and the classic androgen pathway-derived active estrogen, 17ß-estradiol, are equipotent in stimulating breast cancer cell line proliferation and expression of estrogen-responsive genes. 11-oxygenated estrogens were, however, not detectable in serum from individuals with high aromatase levels (pregnant women) and elevated 11-oxygenated androgen levels (patients with congenital adrenal hyperplasia or adrenocortical carcinoma). Our data show that while 11-oxygenated androgens are aromatizable in vitro and ex vivo, the resulting 11-oxygenated estrogens are not detectable in circulation, suggesting that 11-oxygenated androgens function primarily as androgens in vivo.


Asunto(s)
Estrógenos/análogos & derivados , Estrógenos/sangre , Oxígeno/química , Animales , Aromatasa/metabolismo , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Estradiol/análogos & derivados , Estradiol/química , Estradiol/metabolismo , Estrógenos/química , Femenino , Sangre Fetal/química , Sangre Fetal/metabolismo , Células HEK293 , Humanos , Recién Nacido , Células MCF-7 , Placenta/química , Placenta/metabolismo , Embarazo/sangre , Unión Proteica/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Testosterona/análogos & derivados , Testosterona/sangre , Testosterona/química
9.
Pharmacol Ther ; 222: 107789, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33316287

RESUMEN

Many different forms of hormonal contraception are used by millions of women worldwide. These contraceptives differ in the dose and type of synthetic progestogenic compound (progestin) used, as well as the route of administration and whether or not they contain estrogenic compounds. There is an increasing awareness that different forms of contraception and different progestins have different side-effect profiles, in particular their cardiovascular effects, effects on reproductive cancers and susceptibility to infectious diseases. There is a need to develop new methods to suit different needs and with minimal risks, especially in under-resourced areas. This requires a better understanding of the pharmacokinetics, metabolism, serum and tissue concentrations of progestins used in contraception as well as the biological activities of progestins and their metabolites via steroid receptors. Here we review the current knowledge on these topics and identify the research gaps. We show that there is a paucity of research on most of these topics for most progestins. We find that major impediments to clear conclusions on these topics include a lack of standardized methodologies, comparisons between non-parallel clinical studies and variability of data on serum concentrations between and within studies. The latter is most likely due, at least in part, to differences in intrinsic characteristics of participants. The review highlights the importance of insight on these topics in order to provide the best contraceptive options to women with minimal risks.


Asunto(s)
Anticoncepción , Anticonceptivos , Progestinas , Anticoncepción/efectos adversos , Anticoncepción/métodos , Anticonceptivos/sangre , Anticonceptivos/metabolismo , Anticonceptivos/farmacocinética , Femenino , Humanos , Progestinas/sangre , Progestinas/metabolismo , Progestinas/farmacocinética
10.
Biochem Biophys Res Commun ; 533(4): 879-885, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33008590

RESUMEN

Different progestogens are widely used in hormonal therapy and mediate their therapeutic actions via the progesterone receptor (PR). Little published data exist on their relative efficacies and potencies via the PR, while those available may be confounded by off-target receptors, different methodologies and model systems. We performed dose-response analysis to investigate the efficacies and potencies for transcription of progesterone and several progestins widely used in contraception via the B isoform of human PR (PR-B). We compared responses using three different cell lines and two different transient transfection conditions. Results show that in vitro biological responses via PR-B for the select progestogens can vary significantly in biocharacter, rank order and absolute values for efficacies and potencies, depending on the cell line and transfection condition. Progestogen rank orders for published relative binding affinities are mostly different to those for relative efficacies and potencies. These in vitro differences suggest that rank orders and absolute values of the efficacies and potencies of the progestogens are likely to vary in vivo in a cell-specific and progestogen-specific manner, and cannot easily be extrapolated from in vitro data, as is usually the practice. While obtaining such data in vivo is not possible, these in vitro data show proof of concept for likely significant cell- and progestogen-specific PR-B effects.


Asunto(s)
Agentes Anticonceptivos Hormonales/farmacología , Progestinas/farmacología , Receptores de Progesterona/metabolismo , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Agentes Anticonceptivos Hormonales/química , Humanos , Progesterona/química , Progesterona/farmacología , Progestinas/química , Transcripción Genética/efectos de los fármacos , Transfección
11.
Biochem Biophys Res Commun ; 526(2): 466-471, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32234237

RESUMEN

A variety of structurally and functionally distinct progestins is used in contraception and menopausal hormone therapy (MHT). Some progestins elicit off-target effects by binding to steroid receptors other than the progesterone receptor, which may impact their therapeutic and side-effect profiles. We directly compared the binding affinities, efficacies and potencies of selected progestins via the mineralocorticoid receptor (MR). We did not detect a significant difference in the affinities of medroxyprogesterone acetate (MPA), norethisterone acetate (NET-A), levonorgestrel (LNG), gestodene (GES), etonogestrel (ETG), nestorone (NES) and nomegestrel acetate (NoMAC) for the MR, while these were significantly lower compared to drospirenone (DRSP). While GES and NoMAC display affinities indistinguishable from progesterone (P4), the binding affinity of DRSP is significantly greater and all other progestins significantly lower than that of P4. Dose-response analyses showed that P4, GES and ETG display indistinguishable MR antagonist potencies for transactivation to the well-known MR antagonist spironolactone, while LNG, NoMAC and DRSP are significantly more potent than spironolactone and MPA, NET-A and NES are significantly less potent. Similar to our previous findings for NET-A, we show that LNG, GES, ETG and NES dissociate between transactivation and transrepression via the MR. Together our results provide strong evidence for progestin- and promoter-specific transcriptional effects via the MR, which are poorly predicted by relative binding affinities. A comparison of the binding affinities and potencies with reported free serum concentrations of progestins relative to the endogenous mineralocorticoid aldosterone, suggest that all progestins except MPA, NET-A and NES will likely compete with aldosterone for binding to the MR in vivo at doses used in hormonal therapy to elicit physiologically significant off-target effects.


Asunto(s)
Anticoncepción , Terapia de Reemplazo de Hormonas , Progestinas/genética , Receptores de Mineralocorticoides/genética , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Femenino , Humanos , Progestinas/sangre , Progestinas/metabolismo , Receptores de Mineralocorticoides/metabolismo , Activación Transcripcional
12.
J Steroid Biochem Mol Biol ; 189: 145-153, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30822501

RESUMEN

Steroid hormones regulate a variety of physiological processes, including reproductive function, and are widely used in hormonal therapy. Synthetic progestogens, or progestins, were designed to mimic progesterone (P4) for use in contraception and hormonal replacement therapy in women. Medroxyprogesterone acetate (MPA) and norethisterone (NET) are the most widely used injectable contraceptives in the developing world, while other progestins such as levonorgestrel (LNG), etonogestrel (ETG) and nestorone (NES) are used in or being developed for other forms of contraception. As concerns remain about the most appropriate choice of progestin and dosage, and the associated side-effects, the mechanisms and biological effects of progestins are frequently investigated in various in vitro mammalian cell line and tissue models. However, whether progestogens are differentially metabolised in different cell types in vivo or in vitro is unknown. For nine mammalian cell lines commonly used to investigate progestogen mechanisms of action, we developed and validated an ultra-high performance supercritical fluid chromatography-tandem mass spectrometry (UHPSFC-MS/MS) protocol for simultaneously quantifying the metabolism of the above-mentioned steroids. We show for the first time that, while 50-100% of P4 was metabolised within 24 h in all cell lines, the metabolism of the progestins is progestin- and cell line-specific. We also show that MPA and NET are significantly metabolised in human cervical tissue, but to a lesser extent than P4. Taken together, our findings suggest that differential progestogen metabolism may play a role in cell-specific therapeutic and side-effects. Relative affinities for binding to steroid receptors as well as potencies, efficacies and biocharacters for transcriptional activity of progestins, relative to P4, are most frequently determined using some of the cell lines investigated. Our results, however, suggest that differential metabolism of progestins and P4 may confound these results. In particular, metabolism may under-estimate the receptor-mediated intrinsic in vitro binding and dose-response values and predicted endogenous physiological effects of P4.


Asunto(s)
Anticonceptivos Femeninos/metabolismo , Progestinas/metabolismo , Animales , Línea Celular , Chlorocebus aethiops , Desogestrel/metabolismo , Humanos , Levonorgestrel/metabolismo , Acetato de Medroxiprogesterona/metabolismo , Noretindrona/metabolismo , Norprogesteronas/metabolismo , Progesterona/metabolismo , Espectrometría de Masas en Tándem
13.
J Mol Endocrinol ; 61(4): R133-R160, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-29899079

RESUMEN

Although hormone therapy is widely used by millions of women to relieve symptoms of menopause, it has been associated with several side-effects such as coronary heart disease, stroke and increased invasive breast cancer risk. These side-effects have caused many women to seek alternatives to conventional hormone therapy, including the controversial custom-compounded bioidentical hormone therapy suggested to not increase breast cancer risk. Historically estrogens and the estrogen receptor were considered the principal factors promoting breast cancer development and progression, however, a role for other members of the steroid receptor family in breast cancer pathogenesis is now evident, with emerging studies revealing an interplay between some steroid receptors. In this review, we discuss examples of hormone therapy used for the relief of menopausal symptoms, highlighting the distinction between conventional hormone therapy and custom-compounded bioidentical hormone therapy. Moreover, we highlight the fact that not all hormones have been evaluated for an association with increased breast cancer risk. We also summarize the current knowledge regarding the role of steroid receptors in mediating the carcinogenic effects of hormones used in menopausal hormone therapy, with special emphasis on the influence of the interplay or crosstalk between steroid receptors. Unraveling the intertwined nature of steroid hormone receptor signaling pathways in breast cancer biology is of utmost importance, considering that breast cancer is the most prevalent cancer among women worldwide. Moreover, understanding these mechanisms may reveal novel prevention or treatment options, and lead to the development of new hormone therapies that does not cause increased breast cancer risk.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptores de Esteroides/metabolismo , Animales , Neoplasias de la Mama/tratamiento farmacológico , Terapia de Reemplazo de Hormonas/métodos , Humanos , Receptores de Estrógenos/metabolismo
14.
Mol Cell Endocrinol ; 466: 86-97, 2018 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-28527781

RESUMEN

It is well-known that breast cancer is hormone-dependent and that steroid hormones exert their mitogenic effects by binding to estrogen, progesterone and androgen receptors. Vital to our understanding and treatment of this malignancy, is the local metabolism of steroid hormones in breast cancer tissue. This review summarises our current knowledge on steroid producing pathways in the adrenal, ovary and breast, while focussing on the availability of specific circulating hormone precursors and steroidogenic enzymes involved in the local synthesis and metabolism of steroid hormones in the breast. Consequently, we highlight alternate pathways that may be instrumental in the etiology of breast cancer.


Asunto(s)
Andrógenos/biosíntesis , Neoplasias de la Mama/metabolismo , Estrógenos/biosíntesis , Glucocorticoides/biosíntesis , Progesterona/biosíntesis , Glándulas Suprarrenales/metabolismo , Animales , Mama/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ovario/metabolismo
15.
J Steroid Biochem Mol Biol ; 174: 27-39, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28743541

RESUMEN

Conventional hormone therapy (HT) containing estrogens such as ethinylestradiol (EE) have been associated with an increased risk of breast cancer and cardiovascular disease resulting in women seeking safer alternatives that are claimed to have fewer health risks. One such alternative gaining popularity, is custom-compounded bioidentical (b)HT formulations containing bioidentical estradiol (bE2) and estriol (bE3). However, the preparation of these custom-compounded estrogens is not regulated, and depending on the route of synthesis, steroid mixtures with differing activities may be produced. Thus, an investigation into the activities of estrogens prepared by custom-compounded pharmacies is warranted. The aim of this study was therefore to directly compare the pharmacological properties of bE2 and bE3 of unknown purity relative to commercially available, pure E2, E3 and estrone (E1) standards as well as synthetic EE used in conventional HT via the human estrogen receptor (ER)-α and -ß. We determined precise equilibrium dissociation constants (Kd or Ki values) and showed that bE2 and bE3 display similar binding affinities to the E2 and E3 standards, while EE had a higher affinity for ERα, and E1 a lower affinity for ERß. Furthermore, all the estrogens display similar agonist efficacies, but not potencies, for transactivation on a minimal ERE-containing promoter via the individual ER subtypes. Although E2 and E3 were equally efficacious and potent on the endogenous ERE-containing pS2 promoter in the MCF-7 BUS breast cancer cell line co-expressing ERα and ERß, E1 was less efficacious and potent than E2. This study is the first to demonstrate that the bioidentical estrogens, commercially available estrogen standards and synthetic EE are full agonists for transrepression on both minimal and endogenous NFκB-containing promoters. Moreover, we showed that these estrogens all increase proliferation and anchorage-independent growth of MCF-7 BUS cells to a similar extent, suggesting that custom-compounded bHT may in fact not be a safer alternative to conventional HT. Furthermore, our results showing that E3 and E1 are not weak estrogens, and that E3 does not antagonize the activity of E2, suggest that the rationale behind the use of E3 and E1 in custom-compounded bHT formulations should be readdressed. Taken together, the results indicating that there is mostly no difference between the custom-compounded bioidentical estrogens, commercially available estrogen standards and synthetic EE, at concentrations reflecting serum levels in women using estrogen-containing HT, suggest that there is no clear advantage in choosing bHT above conventional HT.


Asunto(s)
Congéneres del Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Estrógenos/farmacología , Animales , Unión Competitiva , Células COS , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Chlorocebus aethiops , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Expresión Génica/efectos de los fármacos , Células HEK293 , Terapia de Reemplazo de Hormonas , Humanos
16.
Biochem Biophys Res Commun ; 491(1): 140-146, 2017 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-28711501

RESUMEN

Progestins used in endocrine therapies bind to multiple steroid receptors and are associated with several side-effects. It is thus important to understand the relationship between steroid receptor cross-reactivity and the side-effect profile of progestins. In cell lines that express negligible levels of steroid receptors, we report for the first time the binding affinities, potencies and efficacies of selected progestins from different generations determined in parallel. We show that the progestins bind to the androgen receptor (AR) with similar affinities to each other and progesterone, while none bind estrogen receptor (ER)-ß, and only norethisterone acetate, levonorgestrel and gestodene bind ERα. Comparative dose-response analysis revealed that progestins from the first three generations display similar androgenic activity to the natural androgen dihydrotestosterone for transactivation, while norethisterone acetate, levonorgestrel and gestodene are ERα agonists. We show for the first time that the anti-androgenic properties of progesterone and drospirenone are similar to the well-known AR antagonist hydroxyflutamide, while nomegestrol acetate is more potent and nestorone less potent than both hydroxyflutamide and progesterone. Moreover, we are the first to report that the older progestins, unlike progesterone and the fourth generation progestins, are efficacious ERα agonists for transrepression, while the selected progestins from the second and third generation are efficacious AR agonists for transrepression. Considering the progestin potencies and their reported free serum concentrations relative to dihydrotestosterone and estradiol, our results suggest that the progestins are likely to exert AR-, but not ERα- or ERß-mediated effects in vivo.


Asunto(s)
Andrógenos/metabolismo , Anticonceptivos Hormonales Orales/metabolismo , Estrógenos/metabolismo , Progestinas/metabolismo , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Células COS , Chlorocebus aethiops , Células HEK293 , Terapia de Reemplazo de Hormonas , Humanos , Unión Proteica , Receptor Cross-Talk/fisiología
17.
Mol Cell Endocrinol ; 441: 31-45, 2017 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-27616670

RESUMEN

Steroidogenesis refers to the de novo synthesis of steroid hormones from cholesterol by a number of sequential enzyme catalysed reactions in the adrenal and the gonads. In addition, circulating steroid hormone precursors are further metabolised in selected peripheral tissues. It has been suggested that the biosynthesis of endogenous steroid hormones can be modulated by progestins, used widely by women in female reproductive medicine. However, as a number of structurally diverse progestins with different pharmacological properties are available, it is possible that these synthetic compounds may vary in their effects on steroidogenesis. This review summarises the evidence indicating that progestins influence the biosynthesis of steroid hormones in the adrenal and gonads, as well as the metabolism of these endogenous hormones in the breast, highlighting the limitations to the current knowledge and directions for future research.


Asunto(s)
Terapia de Reemplazo de Hormonas , Progestinas/uso terapéutico , Esteroides/biosíntesis , Animales , Vías Biosintéticas , Humanos
18.
Mol Cell Endocrinol ; 441: 86-98, 2017 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-27664517

RESUMEN

16α-hydroxyprogesterone (16OHP4) is not well characterised in terms of metabolism and receptor interaction. We therefore investigated its metabolism by adrenal CYP11B and peripheral steroidogenic enzymes, SRD5A and AKR1C2. UHPLC-MS/MS analyses identified novel steroids: the biosynthesis of 4-pregnen-11ß,16α-diol-3,20-dione catalysed by CYP11B2; the 5α-reduction of the latter and 16OHP4 catalysed by SRD5A yielding 5α-pregnan-11ß,16α-diol-3,20-diovne and 5α-pregnan-16α-ol-3,20-dione (16OH-DHP4); and 16OH-DHP4 converted by AKR1C2 to 5α-pregnan-3α,16α-diol-20-one. Receptor studies showed 16OHP4, 16OH-DHP4, progesterone and dihydroprogesterone (DHP4) were weak partial AR agonists; 16OHP4, 16OH-DHP4 and DHP4 exhibited weak partial agonist activity towards PR-B with DHP4 also exhibiting partial agonist activity towards PR-A. Data showed that while the 5α-reduction of P4 decreased PR activation significantly, 16OHP4 and 16OH-DHP4 exhibited comparable receptor activation. Although the clinical relevance of 16OHP4 remains unclear the elevated 16OHP4 levels characteristic of 21OHD, CAH, PCOS, prostate cancer, testicular feminization syndrome and cryptorchidism likely contribute towards these clinical conditions, inducing receptor-activated target genes.


Asunto(s)
Hidroxiprogesteronas/metabolismo , Receptores Androgénicos/metabolismo , Receptores de Progesterona/metabolismo , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/metabolismo , Cromatografía Líquida de Alta Presión , Citocromo P-450 CYP11B2/metabolismo , Células HEK293 , Humanos , Hidroxilación , Hidroxiesteroide Deshidrogenasas/metabolismo , Proteínas de la Membrana/metabolismo , Peso Molecular , Esteroide 11-beta-Hidroxilasa/metabolismo , Espectrometría de Masas en Tándem
19.
PLoS One ; 11(10): e0164170, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27706226

RESUMEN

Progestins used in contraception and hormone replacement therapy are synthetic compounds designed to mimic the actions of the natural hormone progesterone and are classed into four consecutive generations. The biological actions of progestins are primarily determined by their interactions with steroid receptors, and factors such as metabolism, pharmacokinetics, bioavailability and the regulation of endogenous steroid hormone biosynthesis are often overlooked. Although some studies have investigated the effects of select progestins on a few steroidogenic enzymes, studies comparing the effects of progestins from different generations are lacking. This study therefore explored the putative modulatory effects of progestins on de novo steroid synthesis in the adrenal by comparing the effects of select progestins from the respective generations, on endogenous steroid hormone production by the H295R human adrenocortical carcinoma cell line. Ultra-performance liquid chromatography/tandem mass spectrometry analysis showed that the fourth-generation progestins, nestorone (NES), nomegestrol acetate (NoMAC) and drospirenone (DRSP), unlike the progestins selected from the first three generations, modulate the biosynthesis of several endogenous steroids. Subsequent assays performed in COS-1 cells expressing human 3ßHSD2, suggest that these progestins modulate the biosynthesis of steroid hormones by inhibiting the activity of 3ßHSD2. The Ki values determined for the inhibition of human 3ßHSD2 by NES (9.5 ± 0.96 nM), NoMAC (29 ± 7.1 nM) and DRSP (232 ± 38 nM) were within the reported concentration ranges for the contraceptive use of these progestins in vivo. Taken together, our results suggest that newer, fourth-generation progestins may exert both positive and negative physiological effects via the modulation of endogenous steroid hormone biosynthesis.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/metabolismo , Androstenos/farmacología , Megestrol/farmacología , Norpregnadienos/farmacología , Norprogesteronas/farmacología , Progestinas/farmacología , Esteroides/biosíntesis , 17-Hidroxiesteroide Deshidrogenasas/genética , Animales , Células COS , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Cromatografía Liquida , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Espectrometría de Masas en Tándem
20.
PLoS One ; 11(7): e0159867, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27442248

RESUMEN

Dihydrotestosterone (DHT) is regarded as the most potent natural androgen and is implicated in the development and progression of castration resistant prostate cancer (CRPC). Under castrate conditions, DHT is produced from the metabolism of the adrenal androgen precursors, DHEA and androstenedione. Recent studies have shown that the adrenal steroid 11ß-hydroxyandrostenedione (11OHA4) serves as the precursor to the androgens 11-ketotestosterone (11KT) and 11-ketodihydrotestosterone (11KDHT). In this study we comprehensively assess the androgenic activity of 11KT and 11KDHT. This is the first study, to our knowledge, to show that 11KT and 11KDHT, like T and DHT, are potent and efficacious agonists of the human androgen receptor (AR) and induced both the expression of representative AR-regulated genes as well as cellular proliferation in the androgen dependent prostate cancer cell lines, LNCaP and VCaP. Proteomic analysis revealed that 11KDHT regulated the expression of more AR-regulated proteins than DHT in VCaP cells, while in vitro conversion assays showed that 11KT and 11KDHT are metabolized at a significantly lower rate in both LNCaP and VCaP cells when compared to T and DHT, respectively. Our findings show that 11KT and 11KDHT are bona fide androgens capable of inducing androgen-dependant gene expression and cell growth, and that these steroids have the potential to remain active longer than T and DHT due to the decreased rate at which they are metabolised. Collectively, our data demonstrates that 11KT and 11KDHT likely play a vital, but overlooked, role in the development and progression of CRPC.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Testosterona/análogos & derivados , Andrógenos/metabolismo , Animales , Vías Biosintéticas , Línea Celular Tumoral , Proliferación Celular , Humanos , Masculino , Unión Proteica , Receptores Androgénicos/metabolismo , Elementos de Respuesta , Testosterona/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...