Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 791
Filtrar
1.
Cell ; 187(12): 2952-2968.e13, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38795705

RESUMO

Recent studies suggest that human-associated bacteria interact with host-produced steroids, but the mechanisms and physiological impact of such interactions remain unclear. Here, we show that the human gut bacteria Gordonibacter pamelaeae and Eggerthella lenta convert abundant biliary corticoids into progestins through 21-dehydroxylation, thereby transforming a class of immuno- and metabo-regulatory steroids into a class of sex hormones and neurosteroids. Using comparative genomics, homologous expression, and heterologous expression, we identify a bacterial gene cluster that performs 21-dehydroxylation. We also uncover an unexpected role for hydrogen gas production by gut commensals in promoting 21-dehydroxylation, suggesting that hydrogen modulates secondary metabolism in the gut. Levels of certain bacterial progestins, including allopregnanolone, better known as brexanolone, an FDA-approved drug for postpartum depression, are substantially increased in feces from pregnant humans. Thus, bacterial conversion of corticoids into progestins may affect host physiology, particularly in the context of pregnancy and women's health.


Assuntos
Microbioma Gastrointestinal , Glucocorticoides , Hidrogênio , Progestinas , Humanos , Progestinas/metabolismo , Hidrogênio/metabolismo , Feminino , Glucocorticoides/metabolismo , Gravidez , Animais , Família Multigênica , Fezes/microbiologia , Pregnanolona/metabolismo , Camundongos
2.
Proc Biol Sci ; 291(2022): 20240371, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38714210

RESUMO

Naked mole-rats (Heterocephalus glaber) live in large colonies with one breeding female (queen), one to three breeding males (BMs) and the remainder are non-reproductive subordinates. The animals have a linear dominance rank with the breeders at the top of the hierarchy. We investigated how dominance rank in naked mole-rats differs with exploration (the propensity to explore a novel environment) and related endocrine markers. Exploration behaviour, faecal progestagen metabolite (fPM), faecal glucocorticoid metabolite (fGCM), faecal androgen metabolite (fAM) and plasma prolactin concentrations were quantified in breeding, high-, middle- and low-ranked females and males from five naked mole-rat colonies. There were no significant differences between the dominance rank and exploration behaviour. Interestingly, the queens and high-ranking females had higher fGCM and fAM concentrations compared with middle- and low-ranked females. The queens had significantly higher fPM concentrations than all other ranked females, since they are responsible for procreation. In the males, the BMs had higher fGCM concentrations compared with high- and low-ranked males. In addition, BMs and middle-ranking males had overall higher prolactin levels than all other ranked males, which could be linked to cooperative care. Overall, the results suggest that physiological reproductive suppression is linked to high dominance rank.


Assuntos
Androgênios , Fezes , Ratos-Toupeira , Prolactina , Predomínio Social , Animais , Masculino , Feminino , Prolactina/metabolismo , Prolactina/sangue , Fezes/química , Ratos-Toupeira/fisiologia , Androgênios/metabolismo , Androgênios/sangue , Glucocorticoides/metabolismo , Comportamento Exploratório , Progestinas/metabolismo
3.
Reprod Biol Endocrinol ; 22(1): 20, 2024 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-38308254

RESUMO

BACKGROUND: Decidualization of endometrial cells is the prerequisite for embryo implantation and subsequent placenta formation and is induced by rising progesterone levels following ovulation. One of the hormone receptors contributing to endometrial homeostasis is Progesterone Receptor Membrane Component 1 (PGRMC1), a non-classical membrane-bound progesterone receptor with yet unclear function. In this study, we aimed to investigate how PGRMC1 contributes to human decidualization. METHODS: We first analyzed PGRMC1 expression profile during a regular menstrual cycle in RNA-sequencing datasets. To further explore the function of PGRMC1 in human decidualization, we implemented an inducible decidualization system, which is achieved by culturing two human endometrial stromal cell lines in decidualization-inducing medium containing medroxyprogesterone acetate and 8-Br-cAMP. In our system, we measured PGRMC1 expression during hormone induction as well as decidualization status upon PGRMC1 knockdown at different time points. We further conferred proximity ligation assay to identify PGRMC1 interaction partners. RESULTS: In a regular menstrual cycle, PGRMC1 mRNA expression is gradually decreased from the proliferative phase to the secretory phase. In in vitro experiments, we observed that PGRMC1 expression follows a rise-to-decline pattern, in which its expression level initially increased during the first 6 days after induction (PGRMC1 increasing phase) and decreased in the following days (PGRMC1 decreasing phase). Knockdown of PGRMC1 expression before the induction led to a failed decidualization, while its knockdown after induction did not inhibit decidualization, suggesting that the progestin-induced 'PGRMC1 increasing phase' is essential for normal decidualization. Furthermore, we found that the interactions of prohibitin 1 and prohibitin 2 with PGRMC1 were induced upon progestin treatment. Knocking down each of the prohibitins slowed down the decidualization process compared to the control, suggesting that PGRMC1 cooperates with prohibitins to regulate decidualization. CONCLUSIONS: According to our findings, PGRMC1 expression followed a progestin-induced rise-to-decline expression pattern during human endometrial decidualization process; and the correct execution of this expression program was crucial for successful decidualization. Thereby, the results of our in vitro model explained how PGRMC1 dysregulation during decidualization may present a new perspective on infertility-related diseases.


Assuntos
Progesterona , Proibitinas , Gravidez , Feminino , Humanos , Progesterona/farmacologia , Progesterona/metabolismo , Decídua/metabolismo , Receptores de Progesterona/genética , Progestinas/metabolismo , Endométrio/metabolismo , Células Estromais/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo
4.
Front Neuroendocrinol ; 67: 101032, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36029852

RESUMO

Progestins are an important component of hormonal contraceptives (HCs) and hormone replacement therapies (HRTs). Despite an increasing number of studies elucidating the effects of HCs and HRTs, little is known about the effects of different types of progestins included in these medications on the brain. Animal studies suggest that various progestins interact differently with sex steroid, mineralocorticoid and glucocorticoid receptors and have specific modulatory effects on neurotransmitter systems and on the expression of neuropeptides, suggesting differential impacts on cognition and behavior. This review focuses on the currently available knowledge from human behavioral and neuroimaging studies pooled with evidence from animal research regarding the effects of progestins on the brain. The reviewed information is highly relevant for improving women's mental health and making informed choices regarding specific types of contraception or treatment.


Assuntos
Neuropeptídeos , Progestinas , Animais , Humanos , Feminino , Progestinas/farmacologia , Progestinas/metabolismo , Encéfalo/metabolismo , Saúde da Mulher , Neuropeptídeos/metabolismo , Cognição
5.
Biochem Biophys Res Commun ; 639: 70-76, 2023 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-36470074

RESUMO

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.


Assuntos
Progestinas , Receptores de Progesterona , Feminino , Humanos , Anticoncepção , Menopausa , Progesterona/farmacologia , Progesterona/metabolismo , Congêneres da Progesterona/farmacologia , Progestinas/farmacologia , Progestinas/metabolismo , Promegestona , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Transcrição Gênica
6.
Neuroendocrinology ; 113(1): 14-35, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-35760047

RESUMO

Neuroactive steroids can be synthetic or endogenous molecules produced by neuronal and glial cells and peripheral glands. Examples include estrogens, testosterone, progesterone and its reduced metabolites such as 5α-dihydro-progesterone and allopregnanolone. Steroids produced by neurons and glia target the nervous system and are called neurosteroids. Progesterone and analog molecules, known as progestogens, have been shown to exhibit neurotrophic, neuroprotective, antioxidant, anti-inflammatory, glial modulatory, promyelinating, and remyelinating effects in several experimental models of neurodegenerative and injury conditions. Pleiotropic mechanisms of progestogens may act synergistically to prevent neuron degeneration, astrocyte and microglial reactivity, reducing morbidity and mortality. The aim of this review is to summarize the significant findings related to the actions of progesterone and other progestogens in experimental models and epidemiological and clinical trials of some of the most prevalent and debilitating chronic neurodegenerative disorders, namely, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and multiple sclerosis. We evaluated progestogen alterations under pathological conditions, how pathology modifies their levels, as well as the intracellular mechanisms and glial interactions underlying their neuroprotective effects. Furthermore, an analysis of the potential of natural progestogens and synthetic progestins as neuroprotective and regenerative agents, when administered as hormone replacement therapy in menopause, is also discussed.


Assuntos
Doença de Alzheimer , Progestinas , Feminino , Humanos , Progestinas/farmacologia , Progestinas/uso terapêutico , Progestinas/metabolismo , Progesterona/farmacologia , Progesterona/uso terapêutico , Progesterona/metabolismo , Neuroproteção , Doença de Alzheimer/metabolismo , Neurônios/metabolismo
7.
J Reprod Dev ; 69(4): 206-213, 2023 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-37344443

RESUMO

Progesterone (P4) and cortisol production increase in luteinized granulosa cells (LGCs) during the periovulatory period, but their interaction is not well established. Therefore, we investigated their interaction in cultured bovine LGCs. Granulosa cells were collected from follicles of 2-5 mm in diameter and cultured in DMEM/F-12 supplemented with 10% fetal calf serum for up to 14 days. P4 production and the expression of steroidogenic acute regulatory protein (STAR), cholesterol side-chain cleavage enzyme (CYP11A1), and 3ß-hydroxysteroid dehydrogenase type 1 (HSD3B1) rapidly increased until day 10 and remained high thereafter. No de novo production of cortisol from P4 was detected during the culture period. The expression of 11ß-hydroxysteroid dehydrogenase type 1 (HSD11B1), which converts cortisone to cortisol, increased dramatically on day two, decreased until day 8, and remained relatively constant. To investigate how P4 and cortisol influence each other's production, LGCs were treated with trilostane (a P4 synthesis inhibitor), nomegestrol acetate (NA, a synthetic progestogen), P4, and/or cortisol for 24 h on days 6 and 12 of culture. Trilostane suppressed P4 and STAR expression while elevating HSD11B1 and HSD3B1 expression and cortisol production. Concomitant treatment with NA or P4 dose-dependently decreased cortisol production and HSD11B1 and HSD3B1 expression but elevated STAR expression in both days 6 and 12. Conversely, cortisol treatment increased HSD11B1 and HSD3B1 expression and decreased STAR expression without influencing P4 production. These results indicate that progestogens suppress cortisol production by modulating HSD11B1 expression and that progestogens and cortisol differentially regulate STAR, HSD3B1, and HSD11B1 expression in bovine LGCs.


Assuntos
Hidrocortisona , Progesterona , Feminino , Animais , Bovinos , Progesterona/metabolismo , Hidrocortisona/metabolismo , Progestinas/metabolismo , Células da Granulosa/metabolismo , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Complexos Multienzimáticos , Células Cultivadas
8.
J Bacteriol ; 204(4): e0058321, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35285724

RESUMO

Membrane potential homeostasis is essential for cell survival. Defects in membrane potential lead to pleiotropic phenotypes, consistent with the central role of membrane energetics in cell physiology. Homologs of the progestin and AdipoQ receptors (PAQRs) are conserved in multiple phyla of Bacteria and Eukarya. In eukaryotes, PAQRs are proposed to modulate membrane fluidity and fatty acid (FA) metabolism. The role of bacterial homologs has not been elucidated. Here, we use Escherichia coli and Bacillus subtilis to show that bacterial PAQR homologs, which we name "TrhA," have a role in membrane energetics homeostasis. Using transcriptional fusions, we show that E. coli TrhA (encoded by yqfA) is part of the unsaturated fatty acid biosynthesis regulon. Fatty acid analyses and physiological assays show that a lack of TrhA in both E. coli and B. subtilis (encoded by yplQ) provokes subtle but consistent changes in membrane fatty acid profiles that do not translate to control of membrane fluidity. Instead, membrane proteomics in E. coli suggested a disrupted energy metabolism and dysregulated membrane energetics in the mutant, though it grew similarly to its parent. These changes translated into a disturbed membrane potential in the mutant relative to its parent under various growth conditions. Similar dysregulation of membrane energetics was observed in a different E. coli strain and in the distantly related B. subtilis. Together, our findings are consistent with a role for TrhA in membrane energetics homeostasis, through a mechanism that remains to be elucidated. IMPORTANCE Eukaryotic homologs of the progestin and AdipoQ receptor family (PAQR) have been shown to regulate membrane fluidity by affecting, through unknown mechanisms, unsaturated fatty acid (FA) metabolism. The bacterial homologs studied here mediate small and consistent changes in unsaturated FA metabolism that do not seem to impact membrane fluidity but, rather, alter membrane energetics homeostasis. Together, the findings here suggest that bacterial and eukaryotic PAQRs share functions in maintaining membrane homeostasis (fluidity in eukaryotes and energetics for bacteria with TrhA homologs).


Assuntos
Escherichia coli , Progestinas , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Membrana Celular/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Ácidos Graxos/metabolismo , Ácidos Graxos Insaturados , Homeostase , Progestinas/metabolismo
9.
Lab Invest ; 102(10): 1121-1131, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35710596

RESUMO

The pathogenesis of diabetic wounds is closely associated with the dysregulation of macrophage polarization. However, the underlying mechanism remains poorly understood. In this study, we aimed to investigate the potential effects of PAQR3 (progestin and adipoQ receptor 3) silencing in accelerating diabetic wound healing. We showed that PAQR3 silencing promoted skin wound healing and angiogenesis in diabetic mice, which was accompanied by enhanced M2 macrophage polarization and elevated expression of PPARγ (peroxisome proliferator-activated receptor γ). PAQR3 silencing also promoted M2 polarization and increased PPARγ protein level in PMA-treated THP-1 cells. Moreover, knockdown of PAQR3 in macrophages enhanced the migration of HaCaT cells and tube formation of HUVECs. The ubiquitination of PPARγ protein in macrophages was repressed by PAQR3 silencing. STUB1 (STIP1 homology and U-box-containing protein 1) binds with the PPARγ protein to mediate PPARγ ubiquitination and degradation in macrophages, which was impaired by PAQR3 silencing. The PPARγ inhibitor, GW9662, or STUB1 overexpression abrogated the enhanced M2 macrophage polarization induced by PAQR3 silencing. Therefore, these findings demonstrates that PAQR3 silencing accelerates diabetic wound healing by promoting M2 macrophage polarization and angiogenesis, which is mediated by the inhibition of STUB1-mediated PPARγ protein ubiquitination and degradation.


Assuntos
Diabetes Mellitus Experimental , PPAR gama , Animais , Diabetes Mellitus Experimental/metabolismo , Macrófagos/metabolismo , Camundongos , PPAR gama/metabolismo , Progestinas/metabolismo , Progestinas/farmacologia , Cicatrização
10.
Int J Cancer ; 150(9): 1481-1496, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-34935137

RESUMO

Progesterone receptors (PRs) ligands are being tested in luminal breast cancer. There are mainly two PR isoforms, PRA and PRB, and their ratio (PRA/PRB) may be predictive of antiprogestin response. Our aim was to investigate: the impact of the PR isoform ratio on metastatic behaviour, the PR isoform ratio in paired primary tumours and lymph node metastases (LNM) and, the effect of antiprogestin/progestins on metastatic growth. Using murine and human metastatic models, we demonstrated that tumours with PRB > PRA (PRB-H) have a higher proliferation index but less metastatic ability than those with PRA > PRB (PRA-H). Antiprogestins and progestins inhibited metastatic burden in PRA-H and PRB-H models, respectively. In breast cancer samples, LNM retained the same PRA/PRB ratio as their matched primary tumours. Moreover, PRA-H LNM expressed higher total PR levels than the primary tumours. The expression of NDRG1, a metastasis suppressor protein, was higher in PRB-H compared to PRA-H tumours and was inversely regulated by antiprogestins/progestins. The binding of the corepressor SMRT at the progesterone responsive elements of the NDRG1 regulatory sequences, together with PRA, impeded its expression in PRA-H cells. Antiprogestins modulate the interplay between SMRT and AIB1 recruitment in PRA-H or PRB-H contexts regulating NDRG1 expression and thus, metastasis. In conclusion, we provide a mechanistic interpretation to explain the differential role of PR isoforms in metastatic growth and highlight the therapeutic benefit of using antiprogestins in PRA-H tumours. The therapeutic effect of progestins in PRB-H tumours is suggested.


Assuntos
Neoplasias da Mama , Proteínas de Ciclo Celular , Peptídeos e Proteínas de Sinalização Intracelular , Receptores de Progesterona , Animais , Neoplasias da Mama/patologia , Proteínas de Ciclo Celular/metabolismo , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Metástase Neoplásica , Progesterona/farmacologia , Progestinas/metabolismo , Isoformas de Proteínas/metabolismo , Receptores de Progesterona/metabolismo
11.
Gen Comp Endocrinol ; 321-322: 114025, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35292264

RESUMO

Nuclear progestin receptor (PGR), which is induced in the follicles destined to undergo ovulation, is believed to be obligatory for rupture of the follicles during ovulation in vertebrates. Studies in some mammals and teleost medaka have revealed the outline of the central signaling pathway that leads to the PGR expression in the preovulatory follicles at ovulation. In this review, we summarize the current knowledge on what signaling mediators are involved in the LH-induced follicular expression of PGR at ovulation in these animals. LH-inducibility of follicular PGR expression is conserved. In both group of animals, activation of the LH receptor on the granulosa cell surface with LH commonly results in the increase of intracellular cAMP levels, while the downstream signaling cascades activated by high level of cAMP are totally different between mice and medaka. PGR is currently presumed to be induced via PKA/CREB-mediated transactivation and ERK1/2-dependent signaling in mice, but the receptor is induced via EPAC/RAP and AKT/CREB pathways in the teleost medaka. The differences and similarities in the signaling pathways for PGR expression between them is discussed from comparative and evolutionary aspects. We also discussed questions concerning PGR expression and its regulation needed to be investigated in future.


Assuntos
Oryzias , Receptores de Progesterona , Animais , Feminino , Células da Granulosa/metabolismo , Mamíferos/metabolismo , Camundongos , Oryzias/metabolismo , Ovulação/fisiologia , Congêneres da Progesterona , Progestinas/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Transdução de Sinais , Esteroides/metabolismo
12.
Reprod Biol Endocrinol ; 19(1): 47, 2021 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-33752672

RESUMO

BACKGROUND: Long-acting, reversible contraceptives (LARC; progestin only) are an increasingly common hormonal contraceptive choice in reproductive aged women looking to suppress ovarian function and menstrual cyclicity. The overall objective was to develop and validate a rodent model of implanted etonogestrel (ENG) LARC, at body size equivalent doses to the average dose received by women during each of the first 3 years of ENG subdermal rod LARC use. METHODS: Intact, virgin, female Sprague-Dawley rats (16-wk-old) were randomized to 1 of 4 groups (n = 8/group) of ENG LARC (high-0.30µg/d, medium-0.17µg/d, low-0.09µg/d, placebo-0.00µg/d) via a slow-release pellet implanted subcutaneously. Animals were monitored for 21 days before and 29 days following pellet implantation using vaginal smears, ultrasound biomicroscopy (UBM), saphenous blood draws, food consumption, and body weights. Data were analyzed by chi-square, non-parametric, univariate, and repeated measures 2-way ANOVA. RESULTS: Prior to pellet implantation there was no difference in time spent in estrus cycle phases among the treatment groups (p > 0.30). Following pellet implantation there was a dose-dependent impact on the time spent in diestrus and estrus (p < 0.05), with the high dose group spending more days in diestrus and fewer days in estrus. Prior to pellet insertion there was not an association between treatment group and estrus cycle classification (p = 0.57) but following pellet implantation there was a dose-dependent association with cycle classification (p < 0.02). Measurements from the UBM (ovarian volume, follicle count, corpora lutea count) indicate an alteration of ovarian function following pellet implantation. CONCLUSION: Assessment of estrus cyclicity indicated a dose-response relationship in the shift to a larger number of acyclic rats and longer in duration spent in the diestrus phase. Therefore, each dose in this model mimics some of the changes observed in the ovaries of women using ENG LARC and provides an opportunity for investigating the impacts on non-reproductive tissues in the future.


Assuntos
Anticoncepcionais Femininos/administração & dosagem , Desogestrel/administração & dosagem , Implantes de Medicamento/administração & dosagem , Estro/efeitos dos fármacos , Modelos Animais , Progestinas/administração & dosagem , Animais , Anticoncepcionais Femininos/metabolismo , Desogestrel/metabolismo , Relação Dose-Resposta a Droga , Implantes de Medicamento/metabolismo , Estro/metabolismo , Feminino , Humanos , Progestinas/metabolismo , Ratos , Ratos Sprague-Dawley , Roedores
13.
Front Neuroendocrinol ; 55: 100796, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31580837

RESUMO

Men and women differ in their vulnerability to a variety of stress-related illnesses, but the underlying neurobiological mechanisms are not well understood. This is likely due to a comparative dearth of neurobiological studies that assess male and female rodents at the same time, while human neuroimaging studies often don't model sex as a variable of interest. These sex differences are often attributed to the actions of sex hormones, i.e. estrogens, progestogens and androgens. In this review, we summarize the results on sex hormone actions in the hippocampus and seek to bridge the gap between animal models and findings in humans. However, while effects of sex hormones on the hippocampus are largely consistent in animals and humans, methodological differences challenge the comparability of animal and human studies on stress effects. We summarise our current understanding of the neurobiological mechanisms that underlie sex-related differences in behavior and discuss implications for stress-related illnesses.


Assuntos
Androgênios/metabolismo , Dendritos/fisiologia , Transtorno Depressivo , Estrogênios/metabolismo , Hipocampo , Neurogênese/fisiologia , Neuroesteroides/metabolismo , Progestinas/metabolismo , Caracteres Sexuais , Transtornos de Estresse Pós-Traumáticos , Estresse Psicológico , Animais , Dendritos/metabolismo , Transtorno Depressivo/metabolismo , Transtorno Depressivo/fisiopatologia , Feminino , Hipocampo/anatomia & histologia , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Humanos , Masculino , Transtornos de Estresse Pós-Traumáticos/metabolismo , Transtornos de Estresse Pós-Traumáticos/fisiopatologia , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia
14.
Biochem Biophys Res Commun ; 526(2): 466-471, 2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32234237

RESUMO

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.


Assuntos
Anticoncepção , Terapia de Reposição Hormonal , Progestinas/genética , Receptores de Mineralocorticoides/genética , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Feminino , Humanos , Progestinas/sangue , Progestinas/metabolismo , Receptores de Mineralocorticoides/metabolismo , Ativação Transcricional
15.
Acta Vet Hung ; 68(1): 79-84, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32384065

RESUMO

In women and females of different species of laboratory animals, oestrogens stimulate the renin-angiotensin-aldosterone system (RAAS) by increasing tissue and circulating levels of angiotensinogen and renin during the preovulatory period. Progesterone and cortisol compete with aldosterone for mineralocorticoid receptors, which results in increased Na+ reabsorption during the postovulatory period. The purpose of the current research was to analyse the relationship of oestradiol-17ß, progesterone and cortisol with RAAS in 23 mares during an oestrous cycle. During the preovulatory period, significant positive correlations of oestradiol-17ß with renin and aldosterone concentrations and negative correlations of progesterone with renin and aldosterone concentrations were found. In contrast, during the postovulatory period, oestradiol-17ß concentrations were positively correlated with angiotensin concentrations and progesterone was negatively correlated with this component of the RAAS. Cortisol concentrations were not correlated with the hormones of the RAAS, neither before nor after ovulation. This research demonstrates that, as occurs in other species, changes in the RAAS during the periovulatory period in mares may be modulated by variations in the concentrations of steroid hormones.


Assuntos
Estradiol/metabolismo , Estrogênios/metabolismo , Cavalos/fisiologia , Progesterona/metabolismo , Progestinas/metabolismo , Sistema Renina-Angiotensina/fisiologia , Animais , Feminino , Hidrocortisona/metabolismo , Espanha
16.
Zoo Biol ; 39(5): 315-324, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33464632

RESUMO

Aiming to improve our reproductive knowledge of large birds of prey, behavioral data and fecal steroids were assessed in captive pairs of Harpy eagles, a keystone species that is monogamous and resides in the Neotropics year-round. Adult individuals exhibited different reproductive outcomes and a breeding season extending beyond summer solstice (5-9 months) suggests that harpy eagles may not be absolutely photorefractory. Comparisons among breeding stages in males revealed that mean androgen levels in courtship were higher than in copulation and incubation, but no differences were detected in fecal progestagens or estrogens. Females had higher mean estrogen concentrations in courtship and copulation, whereas mean progestagen levels peaked during egg laying. Mean androgen concentrations were not significantly different among breeding stages in females. Assessment of six egg-lay cycles from three females demonstrated that fecal estrogens peaked predominantly between 31 and 18 days before oviposition (-31 to -18 days), and then remained low until 45 days after laying the first egg (+45 days). In contrast, fecal progestagens raised mostly between -20 and +1 day, lowering to baseline concentrations by +3 days. To our knowledge, this is the first study to describe in detail endocrine and behavioral data regarding reproduction in tropical eagles, which may serve in the future as a reference to developing breeding programs.


Assuntos
Animais de Zoológico , Águias/fisiologia , Estrogênios/química , Progestinas/química , Comportamento Sexual Animal/fisiologia , Animais , Estrogênios/metabolismo , Fezes/química , Feminino , Masculino , Oviposição/fisiologia , Fotoperíodo , Progestinas/metabolismo , Estações do Ano
17.
Artigo em Inglês | MEDLINE | ID: mdl-31658973

RESUMO

Millions of women are exposed simultaneously to antiretroviral drugs (ARVs) and progestin-based hormonal contraceptives. Yet the reciprocal modulation by ARVs and progestins of their intracellular functions is relatively unexplored. We investigated the effects of tenofovir disoproxil fumarate (TDF) and dapivirine (DPV), alone and in the presence of select steroids and progestins, on cell viability, steroid-regulated immunomodulatory gene expression, activation of steroid receptors, and anti-HIV-1 activity in vitro Both TDF and DPV modulated the transcriptional efficacy of a glucocorticoid agonist via the glucocorticoid receptor (GR) in the U2OS cell line. In TZM-bl cells, DPV induced the expression of the proinflammatory interleukin 8 (IL-8) gene while TDF significantly increased medroxyprogesterone acetate (MPA)-induced expression of the anti-inflammatory glucocorticoid-induced leucine zipper (GILZ) gene. However, peripheral blood mononuclear cell (PBMC) and ectocervical explant tissue viability and gene expression results, along with TZM-bl HIV-1 infection data, are reassuring and suggest that TDF and DPV, in combination with dexamethasone (DEX) or MPA, do not reciprocally modulate key biological effects in primary cells and tissue. We show for the first time that TDF induces progestogen-independent activation of the progesterone receptor (PR) in a cell line. The ability of TDF and DPV to influence GR and PR activity suggests that their use may be associated with steroid receptor-mediated off-target effects. This, together with cell line and individual donor gene expression responses in the primary models, raises concerns that reciprocal modulation may cause side effects in a cell- and donor-specific manner in vivo.


Assuntos
Antirretrovirais/efeitos adversos , Antirretrovirais/farmacologia , Receptores de Esteroides/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Inflamação/metabolismo , Células-Tronco de Sangue Periférico/efeitos dos fármacos , Células-Tronco de Sangue Periférico/metabolismo , Progestinas/metabolismo , Pirimidinas/efeitos adversos , Pirimidinas/farmacologia , Receptores de Glucocorticoides/metabolismo , Receptores de Progesterona/metabolismo , Tenofovir/efeitos adversos , Tenofovir/farmacologia , Fatores de Transcrição/metabolismo
18.
Biol Reprod ; 101(1): 162-176, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31107530

RESUMO

The current study aimed to elucidate the mechanisms underlying myometrial activation during equine placentitis related to progestogens and the progesterone receptor signaling pathways. Placentitis was induced via intracervical inoculation with Streptococcus equi ssp zooepidemicus in mares at approximately 290 days of gestation (placentitis group; n = 6) with uninoculated gestationally matched mares as controls (n = 4). Mares in the placentitis and control groups were euthanized, and myometrial samples were collected from two regions: region 1-parallel to active placentitis lesion with placental separation in placentitis group (P1) or caudal pole of the placenta in control group (C1); and region 2-parallel to apparently normal placenta without separation in placentitis group (P2) or uterine body in control group (C2). In the current study, SRD5A1 and AKR1C23, which encode for the key P4 metabolizing enzymes, were downregulated in P1 in comparison to C1, C2, and P2, and this was associated with a decline (P < 0.05) in 5αDHP, allopregnanolone (3αDHP), and 20αDHP in P1 in comparison to C1. Further, myometrial expression of PR was downregulated (P < 0.05) in P1 in comparison to C1 and P2, and this was associated with activation of the inflammatory cascade as reflected by significant upregulation of IL-1ß and IL-8 in P1 in comparison to C1, C2, and P2, and supported by increased tissue leukocytes in P1 in comparison to C1. In conclusion, equine placentitis is associated with a localized withdrawal of progestins and a downregulation of the PR in the myometrium concomitant with upregulation of inflammatory cytokines and subsequent myometrial activation.


Assuntos
Doenças dos Cavalos/metabolismo , Cavalos , Miométrio/metabolismo , Doenças Placentárias/metabolismo , Progestinas/metabolismo , Animais , Estudos de Casos e Controles , Corioamnionite/genética , Corioamnionite/metabolismo , Corioamnionite/patologia , Corioamnionite/veterinária , Citocinas/genética , Citocinas/metabolismo , Regulação para Baixo/genética , Feminino , Regulação da Expressão Gênica/genética , Doenças dos Cavalos/genética , Doenças dos Cavalos/patologia , Cavalos/genética , Cavalos/metabolismo , Mediadores da Inflamação/metabolismo , Miométrio/patologia , Doenças Placentárias/genética , Doenças Placentárias/patologia , Doenças Placentárias/veterinária , Gravidez , Complicações Infecciosas na Gravidez/genética , Complicações Infecciosas na Gravidez/metabolismo , Complicações Infecciosas na Gravidez/patologia , Complicações Infecciosas na Gravidez/veterinária , Progestinas/genética , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Transdução de Sinais/genética
19.
Curr Opin Obstet Gynecol ; 31(4): 267-278, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31276453

RESUMO

PURPOSE OF REVIEW: Postmenopausal endometriosis is a gynecologic disease, affecting 2-5% of postmenopausal woman. Current literature assessing the prevalence, pathogenesis, and treatment of this uncommon condition is limited, stressing the necessity for future research. This review examines the current literature on postmenopausal endometriosis to help inform clinical decision-making and point to novel approaches for treatment and management. RECENT FINDINGS: Although one unifying theory to explain the pathogenesis of endometriotic lesions has not been elucidated, estrogen dependence is central to the pathophysiological process. The total quantity of estrogen production is mediated by multiple enzymes in complex pathways. Recent studies have confirmed the presence of these necessary enzymes in endometriotic lesions thereby suggesting a local source of estrogen and a likely pathogenic contributor. More research is needed to fully elucidate the mechanism of local estrogen biosynthesis; however, the current data provide possible explanations for the presence of postmenopausal endometriosis in an otherwise systemically hypoestrogenic environment. SUMMARY: All suspected endometriosis lesions should be surgically excised for optimization of treatment and prevention of malignant transformation. If hormone replacement therapy is initiated, combined estrogen and progestin is recommended, even in the setting of previous hysterectomy, given the risk of disease reactivation and malignant transformation of endometriotic lesions. Further research is needed to understand the true prevalence, cause, and progression in this patient demographic. Histologic studies evaluating tissue lesions and peritoneal fluid for estrogen receptors, estrogen metabolizing enzymes, immune cells, and nerve fibers will aide in clinical management and treatment planning.


Assuntos
Endometriose/patologia , Terapia de Reposição Hormonal , Pós-Menopausa , Biópsia , Transformação Celular Neoplásica , Progressão da Doença , Estrogênios/metabolismo , Estrogênios/uso terapêutico , Feminino , Humanos , Histerectomia , Prevalência , Progestinas/metabolismo , Progestinas/uso terapêutico , Receptores de Estrogênio/metabolismo , Resultado do Tratamento
20.
Gen Comp Endocrinol ; 280: 24-34, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-30951726

RESUMO

Fecal hormone analysis shows high potential for noninvasive assessment of population-level patterns in stress and reproduction of marine mammals. However, the marine environment presents unique challenges for fecal sample collection. Data are still lacking on collection methodology and assay validations for most species, particularly for those mysticete whales that have variable diets. In this study we tested collection techniques for fecal samples of free-swimming humpback whales (Megaptera novaeangliae), and validated immunoassays for five steroid and thyroid hormones. Resulting data were used for preliminary physiological validations, i.e., comparisons to independently confirmed sex and reproductive state. Pregnant females had significantly higher fecal progestins and glucocorticoids than did other demographic categories of whales. Two possible cases of previously undetected pregnancies were noted. Males had significantly higher fecal testosterone metabolites than nonpregnant females. Fecal glucocorticoids were significantly elevated in pregnant females and mature males compared to nonpregnant females. Calf fecal samples had elevated concentrations of all fecal hormones. Fecal thyroid hormones showed a significant seasonal decline from spring to summer. Though sample sizes were small, and sampling was necessarily opportunistic, these patterns indicate that noninvasive fecal hormone analysis may facilitate studies of reproduction, stress and potentially energetics in humpback whales.


Assuntos
Fezes/química , Hormônios/metabolismo , Jubarte/fisiologia , Estresse Fisiológico , Animais , Feminino , Glucocorticoides/metabolismo , Masculino , Metaboloma , Gravidez , Progestinas/metabolismo , Reprodutibilidade dos Testes , Reprodução/fisiologia , Natação/fisiologia , Testosterona/metabolismo , Hormônios Tireóideos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA