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.
Adv Sci (Weinh) ; 11(23): e2310208, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38582508

RESUMO

The progestin regimen is one of the main therapeutic strategies for women with endometrial cancer who undergo conservative management. Although many patients respond well to initial therapy, progestin-refractory disease inevitably emerges, and the molecular basis underlying progestin resistance has not been comprehensively elucidated. Herein, they demonstrated progestin results in p38-dependent IDH1 Thr 77 phosphorylation (pT77-IDH1). pT77-IDH1 translocates into the nucleus and is recruited to chromatin through its interaction with OCT6. IDH1-produced α-ketoglutarate (αKG) then facilitates the activity of OCT6 to promote focal adhesion related target gene transcription to confer progestin resistance. Pharmacological inhibition of p38 or focal adhesion signaling sensitizes endometrial cancer cells to progestin in vivo. The study reveals p38-dependent pT77-IDH1 as a key mediator of progestin resistance and a promising target for improving the efficacy of progestin therapy.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Neoplasias do Endométrio , Isocitrato Desidrogenase , Progestinas , Feminino , Humanos , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/tratamento farmacológico , Progestinas/farmacologia , Progestinas/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Camundongos , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/metabolismo , Animais , Fosforilação , Linhagem Celular Tumoral , Modelos Animais de Doenças
4.
Steroids ; 207: 109427, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38663566

RESUMO

A variety of progestogens are widely used by women for contraception and menopausal hormone therapy. The progestogens undergo extensive metabolism by oral and parenteral routes of administration to form many metabolites. Although a small number of metabolites have been shown to be biologically active, most have not been tested for biologic activity. The present review shows that we know most about progesterone metabolism, followed by the metabolism of levonorgestrel and norethindrone. Very few studies have been carried out on metabolism of most of the progestogens. The clinical significance of this deficiency is that those progestogen metabolites that bind to the progesterone receptors may also bind to other steroid receptors and be responsible for some of the well-documented side effects of administered progestogens. We also discuss how obesity and genetic polymorphisms alter progestogen metabolism, and how development of oral progestogen formulations that are targeted to the colon, where the concentration of steroid-metabolizing enzymes is much lower than in the proximal gut, may have a beneficial effect on progestogen metabolism.


Assuntos
Progestinas , Humanos , Progestinas/metabolismo , Feminino , Menopausa/metabolismo , Menopausa/efeitos dos fármacos , Anticoncepção , Terapia de Reposição Hormonal , Animais
5.
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
6.
J Steroid Biochem Mol Biol ; 237: 106440, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38048919

RESUMO

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.


Assuntos
Neoplasias da Mama , Progestinas , Humanos , Feminino , Progestinas/farmacologia , Progestinas/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Sistema de Sinalização das MAP Quinases , Progesterona/metabolismo , Acetato de Medroxiprogesterona/farmacologia , Acetato de Medroxiprogesterona/metabolismo , Levanogestrel , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo
7.
Artigo em Alemão | MEDLINE | ID: mdl-37903595

RESUMO

The endocrine regulation of birth is based on an intensive exchange of signals between fetus, placenta and mother. Apart from sheep, our knowledge of the underlying processes is still very incomplete. However, current observations suggest substantial species differences. Of critical importance for the onset of the final steps of the signaling cascade leading to active labor is "prepartum progesterone withdrawal," which is based on luteolysis (e. g., cattle, goat, buffalo, camelids, pig) or a breakdown in placental progestogen production (sheep, horse), depending on the relevant progestogen source in late pregnancy. Knowledge of birth-associated regulatory processes allows species-specific regulatory mechanisms to be mimicked for drug-based induction of labor. Furthermore, species-independent mechanisms such as the inhibition of progesterone receptors are available. In addition to efficacy, other aspects such as tolerability for dams and offspring as well as drug regulations must be taken into account when selecting active ingredients under practical conditions.


Assuntos
Placenta , Progestinas , Bovinos , Gravidez , Feminino , Animais , Cavalos , Ovinos , Suínos , Progestinas/metabolismo , Especificidade da Espécie , Progesterona/metabolismo , Mamíferos/metabolismo , Parto/fisiologia
8.
Curr Protoc ; 3(10): e895, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37812178

RESUMO

Decidualization is a differentiation process involving shape reorganization from a fibroblast to an epithelioid-like appearance characteristic of endometrial stromal cells. For the study of in vitro decidualization, one needs to check that the cells have undergone this process effectively. Verification is usually done by analyzing the expression of decidual markers, but changes in morphology are a more comprehensive feature. However, morphological specificities (i.e., flatness) of endometrial cells prevent the use of existing automated tools. A simple and accurate methodology was developed to quantify the phenotypic changes that occur in an in vitro decidualization system. This approach analyzes cell circularity directly from light microscopy images to follow the effects of progesterone or progestin R5020 in combination with estradiol (E2) and cAMP in inducing the decidualization of human endometrial cells. A statistical model to detect the differences in the kinetics of decidualization of the two hormonal stimuli before all the cell population acquire the decidual phenotype was implemented. It was found that statistical differences in morphology between decidualized and control cells could be detected 2 days after the treatments. Here we detail the model applied, scripts, and input files in order to provide a useful, practical, and low-cost tool to evaluate morphological aspects of endometrial stromal differentiation. This method allows the verification of the effectiveness of the decidualization process of the stromal endometrial cells without having to use cell replicates, as other methods such as immunofluorescence and RT-qPCR assays require. Consequently, this approach can follow the kinetics of a living single replicate throughout the experiment. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Cell circularity quantification of human stromal endometrial cells using ImageJ Basic Protocol 2: Statistical analysis of cell circularity of human stromal endometrial cells.


Assuntos
Decídua , Endométrio , Feminino , Humanos , Decídua/metabolismo , Endométrio/metabolismo , Progesterona/farmacologia , Progesterona/metabolismo , Progestinas/metabolismo , Progestinas/farmacologia , Estradiol/farmacologia , Estradiol/metabolismo
9.
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
10.
Sci Rep ; 13(1): 5747, 2023 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-37029234

RESUMO

This study aimed to investigate the metabolite profile and inflammatory state of follicular fluid (FF) in women with stage III-IV ovarian endometriosis (OE) who underwent in vitro fertilization (IVF). A cohort of 20 consecutive patients with OE were recruited and received progestin-primed ovary stimulation (PPOS) protocol (study group), while another 20 OE patients received one-month ultra-long term protocol (control group) for IVF in this prospective, nonrandomized study. FF samples were obtained from dominant follicles during oocyte retrieval, and liquid chromatography-mass spectrometry (LC-MS) was used to investigate the metabolites profile of FF. Results showed that significant increases in the levels of proline, arginine, threonine, and glycine in patients who received PPOS protocol compared to the control group (P < 0.05). A panel of three metabolites (proline, arginine, and threonine) was identified as specific biomarkers of OE patients using PPOS protocol. Additionally, levels of interleukin-1ß, regulated on activation, normal T cell expressed and secreted, and tumor necrosis factor-α markedly decreased in women who received PPOS protocol compared to the control group (P < 0.05). In conclusion, PPOS protocol regulates the metabolism of several amino acids in the FF, which may play critical roles in the oocyte development and blastocyst formation, and their specific mechanism should be further elucidated.


Assuntos
Endometriose , Progestinas , Feminino , Humanos , Progestinas/metabolismo , Líquido Folicular/metabolismo , Endometriose/metabolismo , Ovário , Estudos Prospectivos , Fertilização in vitro/métodos , Esteroides/metabolismo , Arginina/metabolismo
11.
Mar Environ Res ; 186: 105938, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36870161

RESUMO

Progestins in aquatic environments are of increasing concern, as shown by the results of toxicological studies on adult invertebrates with external fertilization. However, their potential effects on the gametes and reproductive success of such animals remain largely unknown. Thus, the current study assessed the effect of in vitro exposure of environmentally relevant concentrations (10 ng/L and 1000 ng/L) of norgestrel (NGT) on the sperm of Pacific oyster Crassostrea gigas, analyzing sperm motility, ultrastructure, mitochondrial function, ATP status, characteristic enzyme activities, and DNA integrity underlying fertilization and hatching success. The results showed that NGT increased the percentage of motile sperm by elevating intracellular Ca2+ levels, Ca2+-ATPase activity, creatine kinase activity, and ATP content. Although superoxide dismutase activity was enhanced to eliminate reactive oxygen species generated by NGT, oxidative stress occurred, as indicated by the increase in malonaldehyde content and damage to plasma membranes and DNA. As a consequence, fertilization rates decreased. However, hatching rates did not alter significantly, possibly as a result of DNA repair processes. This study demonstrates oyster sperm as a useful, sensitive tool for toxicological research of progestins and provides ecologically relevant information on reproductive disturbance in oysters resulting from exposure to NGT.


Assuntos
Crassostrea , Animais , Masculino , Crassostrea/fisiologia , Norgestrel/metabolismo , Norgestrel/farmacologia , Progestinas/metabolismo , Progestinas/farmacologia , Motilidade dos Espermatozoides/fisiologia , Sêmen , Espermatozoides/fisiologia , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia
12.
Mol Cell Endocrinol ; 563: 111855, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36646303

RESUMO

The pro-inflammatory cytokine, chemokine (C-C motif) ligand 20 (CCL20), is emerging as a therapeutic target for immune-based therapies. Cooperative regulation of CCL20 by glucocorticoids and progestins used in endocrine therapy and pro-inflammatory mediators could modulate immune function and affect disease outcomes. We show that glucocorticoids as well as medroxyprogesterone acetate (MPA), the progestin widely used in injectable contraception in sub-Saharan Africa, cooperate with pro-inflammatory mediators to upregulate CCL20 protein and/or mRNA in human peripheral blood mononuclear cells (PBMCs) and human cervical cell lines. Changes in CCL20 mRNA levels were shown to be synergistic, as assessed by Chou analysis, cell- and gene-specific and to involve transcriptional regulation, with a requirement for a nuclear factor kappa B (NF-κB) site and glucocorticoid receptor (GR) involvement. The novel results suggest a mechanism whereby MPA, like glucocorticoids, may impact inflammation both systemically and in the genital tract in patients using MPA and/or glucocorticoid therapy.


Assuntos
Glucocorticoides , Acetato de Medroxiprogesterona , Humanos , Acetato de Medroxiprogesterona/farmacologia , Glucocorticoides/farmacologia , Glucocorticoides/metabolismo , Leucócitos Mononucleares/metabolismo , Progestinas/metabolismo , Receptores de Glucocorticoides/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Quimiocina CCL20/genética , Quimiocina CCL20/metabolismo
13.
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
14.
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
15.
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
16.
Endocrinology ; 163(11)2022 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-36041040

RESUMO

Gender differences in a wide variety of physiological parameters have implicated the ovarian hormones, estrogens and progesterone, in the regulation of numerous nonreproductive tissue functions. Rapid, nongenomic (nonclassical) progesterone actions mediated by membrane progesterone receptors (mPRs), which belong to the progestin and adipoQ receptor family, have been extensively investigated in reproductive and nonreproductive tissues since their discovery in fish ovaries 20 years ago. The 5 mPR subtypes (α, ß, γ, δ, ε) are widely distributed in vertebrate tissues and are often expressed in the same cells as the nuclear progesterone receptor (PR) and progesterone receptor membrane component 1, thereby complicating investigations of mPR-specific functions. Nevertheless, mPR-mediated progesterone actions have been identified in a wide range of reproductive and nonreproductive tissues and distinguished from nuclear PR-mediated ones by knockdown of these receptors with siRNA in combination with a pharmacological approach using mPR- and PR-specific agonists. There are several recent reviews on the roles of the mPRs in vertebrate reproduction and cancer, but there have been no comprehensive assessments of mPR functions in nonreproductive tissues. Therefore, this article briefly reviews mPR functions in a broad range of nonreproductive tissues. The evidence that mPRs mediate progesterone and progestogen effects on neuroprotection, lordosis behavior, respiratory control of apnea, olfactory responses to pheromones, peripheral nerve regeneration, regulation of prolactin secretion in prolactinoma, immune functions, and protective functions in vascular endothelial and smooth muscle cells is critically reviewed. The ubiquitous expression of mPRs in vertebrate tissues suggests mPRs regulate many additional nonreproductive functions that remain to be identified.


Assuntos
Neoplasias Hipofisárias , Receptores de Progesterona , Animais , Membrana Celular/metabolismo , Estrogênios/farmacologia , Feromônios/metabolismo , Feromônios/farmacologia , Neoplasias Hipofisárias/metabolismo , Progesterona/metabolismo , Progesterona/farmacologia , Progestinas/metabolismo , Prolactina/metabolismo , RNA Interferente Pequeno , Receptores de Progesterona/metabolismo
17.
Environ Sci Pollut Res Int ; 29(56): 85211-85228, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35794321

RESUMO

In this study, we investigated the effects of 1000 ng/l levonorgestrel (LNG) alone or combined with increased temperature of 20, 24, and 28 °C on the biochemical and physiological responses of the clam (Ruditapes decussatus) for 28 days. Our results revealed that female clams treated with levonorgestrel (LNG) alone showed enhancement of the antioxidant defense against oxidative stress related to the inductions of catalase (CAT), gluthatione -S -transferase (GST), and protein sulfhydryl (PSH), while the elevated temperatures of 20, 24, and 28 °C diminished most of the specific responses to LNG and was the main factor in the determining the responses to combine exposures. The responses of lysosomal membrane stability, alkaline phosphatase, and NADP+-dependent isocitrate dehydrogenase detected were the most common signs of an adverse effect in all exposures. Female clams' testosterone and estradiol responses to LNG were the most particular manifestations depending on the exposure. Overall, these findings showed clearly that chronic warming stress caused disruption in physiological, biochemical parameters of the female clam R. decussatus, and this may have implications for the whole organism and populations.


Assuntos
Bivalves , Poluentes Químicos da Água , Feminino , Animais , Progestinas/metabolismo , Progestinas/farmacologia , Levanogestrel/farmacologia , Levanogestrel/metabolismo , Poluentes Químicos da Água/análise , Estresse Oxidativo
18.
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
19.
Cancer Gene Ther ; 29(10): 1452-1462, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35388173

RESUMO

Progestin is one of the main hormone treatment regimens for early-stage estrogen receptor- and progesterone receptor (PR)-positive endometrial cancer (EC). However, the response rate of EC to progestins is unsatisfactory. Investigating the mechanisms related to progestin treatment could help improve treatment efficacy. Studies have demonstrated that normal endometrial stromal cells (ESCs) increase the inhibitory effect of progestin on EC cell proliferation via paracrine signaling, but the mechanisms involved remain unclear. In this study, we found that ESCs had different morphological features between progestin-sensitive and -insensitive EC tissues. ESCs presented typical decidualization changes in progestin-sensitive cases, while they remained slim in progestin-insensitive EC lesions, indicating no response. Furthermore, ESCs enhanced the inhibitory effect of medroxyprogesterone acetate (MPA) on EC cell proliferation by secreting neuron cell adhesion molecule (NrCAM). MPA treatment enhanced NrCAM secretion by ESCs. EC xenografts in BALB/C nude mice demonstrated that MPA combined with NrCAM had an increased tumor inhibitory effect compared with MPA or NrCAM alone. Mechanistically, MPA upregulated NrCAM expression in ESCs through PR. Specifically, NrCAM increased PR expression in EC cells through TET1-induced hydroxymethylation of the PRB gene promoter region. These findings indicate that NrCAM or NrCAM combined with progestins could be a new EC treatment.


Assuntos
Neoplasias do Endométrio , Progestinas , Animais , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Moléculas de Adesão Celular/farmacologia , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Endométrio , Epigênese Genética , Feminino , Humanos , Acetato de Medroxiprogesterona/metabolismo , Acetato de Medroxiprogesterona/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Oxigenases de Função Mista/genética , Progestinas/metabolismo , Progestinas/farmacologia , Proteínas Proto-Oncogênicas/genética , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Células Estromais/metabolismo , Células Estromais/patologia
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA