Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 286
Filtrar
1.
Rev. chil. obstet. ginecol. (En línea) ; 87(6): 404-411, dic. 2022. ilus, tab
Artigo em Espanhol | LILACS | ID: biblio-1423742

RESUMO

En algunos estudios se ha asociado a la terapia de reemplazo hormonal (TRH) con estrógenos y progestinas a un mayor riesgo de cáncer de mama que la terapia con estrógenos solos. Sin embargo, dependiendo de su naturaleza algunas progestinas serían más seguras que otras. Se buscaron y analizaron artículos atingentes al tema en las bases de datos Google Scholar, PubMed, Science, SciELO y Cochrane, introduciendo los siguientes términos: terapia de reemplazo hormonal y cáncer de mama, progestinas y cáncer de mama, receptor de progesterona. Específicamente se ha asociado a las progestinas sintéticas acetato de medroxiprogesterona, noretisterona y levonorgestrel con un mayor riesgo de cáncer de mama, no así a la progesterona natural, a la progesterona oral micronizada ni a la didrogesterona. La progesterona natural, progesterona micronizada y didrogesterona serían más seguras en TRH para evitar el desarrollo de cáncer de mama, lo que estaría dado por la mayor especificidad en su acción.


In some studies, hormone replacement therapy (HRT) with estrogens and progestins has been associated with a higher risk of breast cancer than therapy with estrogens alone. However, depending on their nature, some progestins may be safer than others. This article analyzes the mode of action of progesterone in breast tissue and also the role of some progestins in the development of this pathology. Articles related to the subject were searched for and analyzed in Google Scholar, PubMed, Science, SciELO and Cochrane databases, introducing the following terms: hormone replacement therapy and breast cancer, progestins and breast cancer, progesterone receptor. Specifically, synthetic progestins medroxyprogesterone acetate, norethisterone, and levonorgestrel have been associated with an increased risk of breast cancer, but not natural progesterone, micronized oral progesterone, or dydrogesterone. Natural progesterone, micronized progesterone and dydrogesterone would be safer in HRT to prevent the development of breast cancer, which would be due to the greater specificity of their action.


Assuntos
Humanos , Feminino , Progestinas/efeitos adversos , Neoplasias da Mama/induzido quimicamente , Progestinas/classificação , Progestinas/fisiologia , Receptores de Progesterona , Medição de Risco , Terapia de Reposição Hormonal/efeitos adversos , Estrogênios/efeitos adversos
2.
Cells ; 11(11)2022 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-35681480

RESUMO

The role of membrane progesterone receptors (mPRs), which belong to the progestin and adipoQ receptor (PAQR) family, in mediating rapid, nongenomic (non-classical) progestogen actions has been extensively studied since their identification 20 years ago. Although the mPRs have been implicated in progestogen regulation of numerous reproductive and non-reproductive functions in vertebrates, several critical aspects of their structure and signaling functions have been unresolved until recently and remain the subject of considerable debate. This paper briefly reviews recent developments in our understanding of the structure and functional characteristics of mPRs. The proposed membrane topology of mPRα, the structure of its ligand-binding site, and the binding affinities of steroids were predicted from homology modeling based on the structures of other PAQRs, adiponectin receptors, and confirmed by mutational analysis and ligand-binding assays. Extensive data demonstrating that mPR-dependent progestogen regulation of intracellular signaling through mPRs is mediated by activation of G proteins are reviewed. Close association of mPRα with progesterone membrane receptor component 1 (PGRMC1), its role as an adaptor protein to mediate cell-surface expression of mPRα and mPRα-dependent progestogen signaling has been demonstrated in several vertebrate models. In addition, evidence is presented that mPRs can regulate the activity of other hormone receptors.


Assuntos
Progesterona , Receptores de Progesterona , Animais , Ligantes , Progesterona/metabolismo , Progestinas/fisiologia , Receptores de Progesterona/metabolismo , Transdução de Sinais/fisiologia
3.
Nucleic Acids Res ; 49(22): 12716-12731, 2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34850111

RESUMO

Here, we report that in T47D breast cancer cells 50 pM progestin is sufficient to activate cell cycle entry and the progesterone gene expression program. At this concentration, equivalent to the progesterone blood levels found around the menopause, progesterone receptor (PR) binds only to 2800 genomic sites, which are accessible to ATAC cleavage prior to hormone exposure. These highly accessible sites (HAs) are surrounded by well-organized nucleosomes and exhibit breast enhancer features, including estrogen receptor alpha (ERα), higher FOXA1 and BRD4 (bromodomain containing 4) occupancy. Although HAs are enriched in RAD21 and CTCF, PR binding is the driving force for the most robust interactions with hormone-regulated genes. HAs show higher frequency of 3D contacts among themselves than with other PR binding sites, indicating colocalization in similar compartments. Gene regulation via HAs is independent of classical coregulators and ATP-activated remodelers, relying mainly on MAP kinase activation that enables PR nuclear engagement. HAs are also preferentially occupied by PR and ERα in breast cancer xenografts derived from MCF-7 cells as well as from patients, indicating their potential usefulness as targets for therapeutic intervention.


Assuntos
Neoplasias da Mama/genética , Elementos Facilitadores Genéticos , Regulação Neoplásica da Expressão Gênica , Progestinas/fisiologia , Animais , Neoplasias da Mama/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células , Cromatina , Receptor alfa de Estrogênio/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases , Células MCF-7 , Camundongos , Promegestona/farmacologia , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
4.
Addict Biol ; 26(6): e13079, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34374475

RESUMO

Stimulant use disorder is associated with significant global health burden. Despite evidence for sex differences in the development and maintenance of stimulant use disorder, few studies have focused on mechanisms underpinning distinct trajectories in females versus males, including the effect of the ovarian sex hormones estrogen and progesterone. This review aimed to identify and synthesise the existing preclinical and clinical literature on the effect of ovarian sex hormones on stimulant consumption in females. A systematic search of peer-reviewed literature identified 1593 articles, screened using the following inclusion criteria: (1) adult female humans or animals, (2) using stimulant drugs, (3) ovarian sex hormones were administered exogenously OR were measured in a validated manner and (4) with stimulant consumption as an outcome measure. A total of 50 studies (3 clinical and 47 preclinical) met inclusion criteria. High-estrogen (low progesterone) phases of the menstrual/estrus cycle were associated with increased stimulant use in preclinical studies, while there were no clinical studies examining estrogen and stimulant consumption. Consistent preclinical evidence supported progesterone use reducing stimulant consumption, which was also identified in one clinical study. The review was limited by inconsistent data reporting across studies and different protocols across preclinical laboratory paradigms. Importantly, almost all studies examined cocaine use, with impact on methamphetamine use a significant gap in the existing evidence. Given the safety and tolerability profile of progesterone, further research is urgently needed to address this gap, to explore the potential therapeutic utility of progesterone as a treatment for stimulant use disorder.


Assuntos
Estimulantes do Sistema Nervoso Central/administração & dosagem , Estrogênios/fisiologia , Hormônios Esteroides Gonadais/fisiologia , Progestinas/fisiologia , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Animais , Relação Dose-Resposta a Droga , Estrogênios/farmacologia , Feminino , Humanos , Ciclo Menstrual/fisiologia , Progestinas/farmacologia , Transtornos Relacionados ao Uso de Substâncias/psicologia
5.
Addict Biol ; 24(2): 157-169, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-29280252

RESUMO

Sex hormones play an important role in establishing sex-distinctive brain structural and functional variations that could contribute to the sex differences in alcohol consumption behavior. Here, we systematically reviewed articles that studied sex hormone impacts on alcohol consumption and alcohol use disorder (AUD). An extensive literature search conducted in MEDLINE, PubMed, Scopus and CINAHL databases identified 776 articles, which were then evaluated for pre-specified criteria for relevance and quality assurance. A total of 50 articles, including 19 human studies and 31 animal studies, were selected for this review. Existing evidence supports the association of increased testosterone level and increased risk for alcohol use and AUD in males but results are inconclusive in females. In contrast, the evidence supports the association of increased estrogen level and increased alcohol use in females, with mixed findings reported in males. Much less is known about the impact of progestins on alcohol use and misuse in human subjects. Future observational and experimental studies conducted in both sexes with a comprehensive hormone panel are needed to elucidate the impact of the interplay between various sex hormone levels during various developmental stages on alcohol use-related phenotypes and AUD.


Assuntos
Consumo de Bebidas Alcoólicas/fisiopatologia , Alcoolismo/etiologia , Hormônios Esteroides Gonadais/fisiologia , Animais , Modelos Animais de Doenças , Estrogênios/fisiologia , Feminino , Humanos , Masculino , Progestinas/fisiologia
6.
Climacteric ; 21(4): 321-325, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29583028

RESUMO

It is well established that unopposed estrogen for hormone therapy in postmenopausal women (MHT) induces a dose-related stimulation of the endometrium associated with an increased risk of hyperplasia and endometrial cancer. Progesterone acts physiologically to counteract the proliferative effects of estradiol during the menstrual cycle. In MHT, progestogens protect the endometrium against the proliferative effects of estrogens in women with a uterus. Recent data suggest that, whereas micronized progesterone is apparently safer for the breast, it could be less efficient than synthetic progestin on the endometrium. An update on progestogen and endometrial safety in MHT is the subject of this review.


Assuntos
Mama/efeitos dos fármacos , Endométrio/efeitos dos fármacos , Terapia de Reposição de Estrogênios/métodos , Progesterona/fisiologia , Progestinas/fisiologia , Neoplasias da Mama/induzido quimicamente , Neoplasias do Endométrio/induzido quimicamente , Terapia de Reposição de Estrogênios/efeitos adversos , Feminino , Humanos , Perimenopausa , Progesterona/efeitos adversos , Progestinas/efeitos adversos , Ensaios Clínicos Controlados Aleatórios como Assunto
7.
Chemosphere ; 190: 17-24, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28972919

RESUMO

The aim of this study was to assess the adverse effects of synthetic progestin norgestrel (NGT) on the reproduction of zebrafish by measuring the egg production, histology and transcriptional expression profiles along the hypothalamic-pituitary-gonadal (HPG) axis in adult zebrafish. After a pre-exposure period of 7 days, adult zebrafish were exposed to 6, 29 and 69 ng L-1 NGT for 21 days. The results showed that exposure to 69 ng L-1 NGT led to a significant up-regulation of follicle stimulating hormone, beta polypeptide (fshb), luteinizing hormone, beta polypeptide (lhb), progesterone receptor (pgr), estrogen receptor 1 (esr1) and androgen receptor (ar) genes in the brains, as well as significant up-regulation of hydroxysteroid 20-beta dehydrogenase (hsd20b) and hydroxysteroid 11-beta dehydrogenase 2 (hsd11b2) genes and down-regulation of 11-beta-hydroxylase (cyp11b) gene in the ovaries of females. In the testes of males, an overall down-regulation of steroidogenic acute regulatory protein (star), cytochrome P450-mediated side-chain cleavage enzyme (cyp11a1), cyp11b, hsd20b, hydroxysteroid 17-beta dehydrogenase type 3 (hsd17b3), hsd11b2 and ar genes were observed following exposure to different treatments of NGT. These transcriptional alterations imply that NGT could exhibit the potent progestogenic and androgenic activities in zebrafish. Egg production as well as histology in the ovaries and testes was not affected by NGT. Taken together, the overall results demonstrated that NGT could significantly affect transcriptional expression levels of genes related to HPG axis in zebrafish, and whether that change translates to additional physiological effects is needed further research.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Norgestrel/farmacologia , Reprodução/efeitos dos fármacos , Peixe-Zebra/fisiologia , Animais , Anticoncepcionais Orais Sintéticos/farmacologia , Feminino , Gonadotropinas Hipofisárias/genética , Hormônios Hipotalâmicos/genética , Masculino , Norgestrel/metabolismo , Progestinas/fisiologia , Receptores de Progesterona/genética , Peixe-Zebra/metabolismo
8.
Sci Rep ; 7(1): 17697, 2017 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-29255206

RESUMO

HIV prevention research is focused on combining antiretrovirals (ARV) and progestin contraceptives to prevent HIV infection and pregnancy. The possibility that progestins compromise ARV anti-HIV activity prompted us to evaluate the effects of progestins on tenofovir (TFV) and TFV-alafenamide (TAF) on HIV infection and intracellular TFV-diphosphate (TFV-DP) concentrations in blood and genital CD4+ T cells. Following incubation of blood CD4+ T cells with TFV or TAF, Medroxyprogesterone acetate (MPA), but not Levonorgestrel, Norethisterone or progesterone, suppressed the anti-HIV effect of TFV by reducing intracellular TFV-DP, but had no effect on TAF inhibition of infection or TFV-DP. In contrast, with genital CD4+ T cells, MPA suppressed TAF inhibition of HIV infection and lowered of TFV-DP concentrations without affecting TFV protection. These findings demonstrate that MPA selectively compromises TFV and TAF protection in blood and genital CD4+ T cells and suggests that MPA may decrease ARV protection in individuals who use ARV intermittently for prevention.


Assuntos
Linfócitos T CD4-Positivos/efeitos dos fármacos , Anticoncepcionais Orais Hormonais/farmacologia , Infecções por HIV/prevenção & controle , Adenina/análogos & derivados , Adenina/farmacologia , Adulto , Alanina , Fármacos Anti-HIV/farmacologia , Antirretrovirais/uso terapêutico , Células Cultivadas , Anticoncepcionais/farmacologia , Anticoncepcionais Orais Hormonais/efeitos adversos , Feminino , Genitália Feminina/efeitos dos fármacos , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Humanos , Pessoa de Meia-Idade , Organofosfatos/farmacologia , Progestinas/antagonistas & inibidores , Progestinas/fisiologia , Tenofovir/farmacologia , Tenofovir/uso terapêutico
9.
Theriogenology ; 99: 119-123, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28708492

RESUMO

In this study it was determined the progression of uterine gland development from late gestation to puberty in domestic felids. Cell proliferation patterns for luminal (LE), glandular epithelium (GE) as well as stroma (S) were also described. Twenty-four uteri from female kittens: 45 and 65 days of gestation and 1 to 5, 8, 12, 16, 20 and 24 weeks postnatally were obtained. Uterine cross-sections were submitted for routine histological and immunohistochemical quantification of proliferating cell nuclear antigen (PCNA) techniques. Although prenatal uteri presented no indication of adenogenesis, 1 week old uteri revealed an incipient budding of the LE. During the second week budding increased and a mild degree of tubulogenesis of the GE into the stroma was detected. From the third to fifth weeks coiling, branching and cross-sections of glands appeared. These latter findings were more evident in week 8 when GE began to penetrate through much of the S to week 24. PCNA immunostaining revealed that DNA synthesis decreased throughout the study in the 3 cell compartments; (P < 0.01). Luminal proliferation began prenatally, it maintained up to postnatal week 8 to markedly decrease to puberty (P < 0.01). From postnatal week 3 up to week 8, GE mitotic activity was elevated becoming low thereafter (P < 0.01). Stroma actively proliferated prenatally (P < 0.01), diminishing up to week 8 (P < 0.01) and again during the last weeks (P < 0.01) of the study. It was concluded that, in domestic felids, proliferation of LE begins prenatally, histological uterine adenogenesis commenced during the first postnatal week and both events concluded by postnatal weeks 5-8.


Assuntos
Gatos/crescimento & desenvolvimento , Endométrio/anatomia & histologia , Endométrio/crescimento & desenvolvimento , Ciclo Menstrual/fisiologia , Maturidade Sexual/fisiologia , Animais , Feminino , Progestinas/fisiologia
10.
Menopause ; 24(2): 203-209, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27749742

RESUMO

OBJECTIVE: Expression of progesterone receptor membrane component 1 (PGRMC1) has been shown to be higher in breast cancer than normal tissue. We have previously shown that certain progestogens strongly stimulate proliferation of breast cancer cells overexpressing PGRMC1, and therefore hypothesize that PGRMC1 may play a critical role in breast cancer progression. Because little information is available if expression of PGRMC1 is also associated with worse prognosis for breast cancer patients, in this study we investigated the clinicopathologic significance of PGRMC1 expression in breast cancer tissue. METHODS: Expression of PGRMC1 was analyzed by immunohistochemical staining of primary tumor tissues obtained from 69 breast cancer patients. A labeling score was developed, and results were correlated with tumor size, lymph node metastasis, and clinical outcome. RESULTS: Overexpression of PGRMC1 is correlating with larger tumor size and lymph node metastasis. Kaplan-Meier survival curves indicate that patients with PGRMC1 tumors have poorer disease-free and overall survival independent from the estrogen receptor status than breast cancer patients with PGRMC1 tumors. CONCLUSIONS: Our findings suggest that the expression of PGRMC1 might be useful for predicting prognosis in patients with breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Membrana/metabolismo , Fenótipo , Receptores de Estrogênio/análise , Receptores de Progesterona/metabolismo , Mama/patologia , Neoplasias da Mama/química , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Proliferação de Células , Progressão da Doença , Feminino , Humanos , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Progestinas/fisiologia , Prognóstico
11.
Compr Physiol ; 6(3): 1295-337, 2016 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-27347894

RESUMO

Sex differences in neurological disease exist in incidence, severity, progression, and symptoms and may ultimately influence treatment. Cognitive disturbances are frequent in neuropsychiatric disease with men showing greater cognitive impairment in schizophrenia, but women showing more severe dementia and cognitive decline with Alzheimer's disease. Although there are no overall differences in intelligence between the sexes, men, and women demonstrate slight but consistent differences in a number of cognitive domains. These include a male advantage, on average, in some types of spatial abilities and a female advantage on some measures of verbal fluency and memory. Sex differences in traits or behaviors generally indicate the involvement of sex hormones, such as androgens and estrogens. We review the literature on whether adult levels of testosterone and estradiol influence spatial ability in both males and females from rodent models to humans. We also include information on estrogens and their ability to modulate verbal memory in men and women. Estrone and progestins are common components of hormone therapies, and we also review the existing literature concerning their effects on cognition. We also review the sex differences in the hippocampus and prefrontal cortex as they relate to cognitive performance in both rodents and humans. There has been greater recognition in the scientific literature that it is important to study both sexes and also to analyze study findings with sex as a variable. Only by examining these sex differences can we progress to finding treatments that will improve the cognitive health of both men and women. © 2016 American Physiological Society. Compr Physiol 6:1295-1337, 2016.


Assuntos
Cognição/fisiologia , Hormônios Esteroides Gonadais/fisiologia , Aprendizagem/fisiologia , Memória/fisiologia , Sistemas Neurossecretores/fisiologia , Sistema Nervoso Central/fisiologia , Estrogênios/fisiologia , Hipocampo/anatomia & histologia , Hipocampo/fisiologia , Terapia de Reposição Hormonal/métodos , Humanos , Neurogênese/fisiologia , Córtex Pré-Frontal/fisiologia , Progestinas/fisiologia , Caracteres Sexuais
12.
J Neuroendocrinol ; 28(7)2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27306650

RESUMO

When steroids, such as pregnenolone, progesterone and oestrogen, are synthesised de novo in neural tissues, they are more specifically referred to as neurosteroids. These neurosteroids bind specific receptors to promote essential brain functions. Pregnenolone supports cognition and protects mouse hippocampal cells against glutamate and amyloid peptide-induced cell death. Progesterone promotes myelination, spinogenesis, synaptogenesis, neuronal survival and dendritic growth. Allopregnanolone increases hippocampal neurogenesis, neuronal survival and cognitive functions. Oestrogens, such as oestradiol, regulate synaptic plasticity, reproductive behaviour, aggressive behaviour and learning. In addition, neurosteroids are neuroprotective in animal models of Alzheimer's disease, Parkinson's disease, brain injury and ageing. Using in situ hybridisation and/or immunohistochemistry, steroidogenic enzymes, including cytochrome P450 side-chain cleavage, 3ß-hydroxysteroid dehydrogenase/Δ5-Δ4 isomerase, cytochrome P450arom, steroid 5α-reductase and 3α-hydroxysteroid dehydrogenase, have been detected in numerous brain regions, including the hippocampus, hypothalamus and cerebral cortex. In the present review, we summarise some of the studies related to the synthesis and function of oestrogens and progestagens in the central nervous system.


Assuntos
Encéfalo/fisiologia , Encéfalo/fisiopatologia , Estrogênios/fisiologia , Progestinas/fisiologia , Animais , Encéfalo/enzimologia , Encéfalo/metabolismo , Estrogênios/biossíntese , Humanos , Fármacos Neuroprotetores , Progestinas/biossíntese , Receptores de Neurotransmissores/fisiologia
13.
Biochem Biophys Res Commun ; 473(1): 29-34, 2016 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-26993165

RESUMO

Previous studies indicated that maturation inducing hormone, 17α, 20ß-Dihydroxy-4-pregnen-3-one (DHP), probably through nuclear progestin receptor (Pgr), might be involved in spermatogenesis and oogenesis in fish. To further elucidate DHP actions in teleostean ovarian differentiation, we analyzed the expression of pgr in the ovary of Nile tilapia (Oreochromis niloticus), and performed RU486 (a synthetic Pgr antagonist) treatment in XX fish from 5 days after hatching (dah) to 120 dah. Tilapia Pgr was abundantly expressed in the follicular cells surrounding oocytes at 30 and 90 dah. Continuous RU486 treatment led to the blockage of oogenesis and masculinization of somatic cells in XX fish. Termination of RU486 treatment and maintenance in normal condition resulted in testicular differentiation, and estrogen compensation in RU486-treated XX fish successfully restored oogenesis. In RU486-treated XX fish, transcript levels of female dominant genes were significantly reduced, while male-biased genes were evidently augmented. Meanwhile, both germ cell mitotic and meiotic markers were substantially reduced. Consistently, estrogen production levels were significantly declined in RU486-treated XX fish. Taken together, our data further proved that DHP, possibly through Pgr, might be essential in the ovarian differentiation and estrogen production in fish.


Assuntos
Ciclídeos/fisiologia , Ovário/citologia , Progestinas/fisiologia , Receptores de Progesterona/antagonistas & inibidores , Animais , Diferenciação Celular , Estrogênios/química , Feminino , Perfilação da Expressão Gênica , Hidroxiprogesteronas/química , Técnicas Imunoenzimáticas , Imuno-Histoquímica , Masculino , Meiose , Mifepristona/química , Mitose , Reação em Cadeia da Polimerase , Diferenciação Sexual , Espermatogênese
14.
Hum Reprod Update ; 22(4): 497-515, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26912000

RESUMO

BACKGROUND: Human pregnancy requires robust hemostasis to prevent hemorrhage during extravillous trophoblast (EVT) invasion of the decidualized endometrium, modification of spiral arteries and post-partum processes. However, decidual hemorrhage (abruption) can occur throughout pregnancy from poorly transformed spiral arteries, causing fetal death or spontaneous preterm birth (PTB), or it can promote the aberrant placentation observed in intrauterine growth restriction (IUGR) and pre-eclampsia; all leading causes of perinatal or maternal morbidity and mortality. In non-fertile cycles, the decidua undergoes controlled menstrual bleeding. Abnormal uterine bleeding (AUB) accompanying progestin-only, long-acting, reversible contraception (pLARC) accounts for most discontinuations of these safe and highly effective agents, thereby contributing to unwanted pregnancies and abortion. The aim of this study was to investigate the role of decidual cells in uterine hemostasis, menstruation, inflammation, adverse pregnancy outcomes and abnormal uterine bleeding. METHODS: We conducted a critical review of the literature arising from PubMed searches up to December 2015, regarding in situ and in vitro expression and regulation of several specific proteins involved in uterine hemostasis in decidua and cycling endometrium. In addition, we discussed clinical and molecular mechanisms associated with pLARC-induced AUB and pregnancy complications with abruptions, chorioamnionitis or pre-eclampsia. RESULTS: Progestin-induced decidualization of estradiol-primed human endometrial stromal cells (HESCs) increases in vivo and in vitro expression of tissue factor (TF) and type-1 plasminogen activator inhibitor (PAI-1) while inhibiting plasminogen activators (PAs), matrix metalloproteinases (MMPs), and the vasoconstrictor, endothelin-1 (ET-1). These changes in decidual cell-derived regulators of hemostasis, fibrinolysis, extracellular matrix (ECM) turnover, and vascular tone prevent hemorrhage during EVT invasion and vascular remodeling. In non-fertile cycles, progesterone withdrawal reduces TF and PAI-1 while increasing PA, MMPs and ET-1, causing menstrual-associated bleeding, fibrinolysis, ECM degradation and ischemia. First trimester decidual hemorrhage elicits later adverse outcomes including pregnancy loss, pre-eclampsia, abruption, IUGR and PTB. Decidual hemorrhage generates excess thrombin that binds to decidual cell-expressed protease-activated receptors (PARs) to induce chemokines promoting shallow placentation; such bleeding later in pregnancy generates thrombin to down-regulate decidual cell progesterone receptors and up-regulate cytokines and MMPs linked to PTB. Endometria of pLARC users display ischemia-induced excess vasculogenesis and progestin inhibition of spiral artery vascular smooth muscle cell proliferation and migration leading to dilated fragile vessels prone to bleeding. Moreover, aberrant TF-derived thrombin signaling also contributes to the pathogenesis of endometriosis via induction of angiogenesis, inflammation and cell survival. CONCLUSION: Perivascular decidualized HESCs promote endometrial hemostasis during placentation yet facilitate menstruation through progestational regulation of hemostatic, proteolytic, and vasoactive proteins. Pathological endometrial hemorrhage elicits excess local thrombin generation, which contributes to pLARC associated AUB, endometriosis and adverse pregnancy outcomes through several biochemical mechanisms.


Assuntos
Decídua/citologia , Hemostasia , Ciclo Menstrual/fisiologia , Hemorragia Uterina/etiologia , Decídua/irrigação sanguínea , Decídua/metabolismo , Implantação do Embrião/fisiologia , Endométrio/fisiologia , Feminino , Humanos , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Pré-Eclâmpsia/etiologia , Gravidez , Resultado da Gravidez , Progestinas/fisiologia , Tromboplastina/metabolismo
15.
Anim Reprod Sci ; 165: 11-16, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26709013

RESUMO

Efforts to increase mink reproductive success (live births and litter sizes) can be partly assessed by measurement of blood progesterone levels. However, the stress of blood sampling increases the incidence of failed matings, aborted fetuses and death of the dam. We have therefore non-invasively measured fecal progesterone metabolite (progestin) concentrations during the reproductive cycle of mink. We tested the hypothesis that fecal progestin concentrations during the window of implantation (late March-early April) will, (1): be higher for whelping than non-whelping mink, and (2): be higher for mink mated multiple times, compared to single matings. Mink were mated once (March 3), twice (March 3 and 10) or three times (March 3, 10 and 11) and fecal progestin concentrations determined from March 1 to April 30. The percent mink in each group giving birth to live offspring was 42.8%, 80.8% and 92.3% for mink mated once, twice or three times, respectively (P<0.05). Litter sizes did not differ among mink mated once (5.22±0.55), twice (6.29±0.35) or three times (6.08±0.32; P>0.05). Mean fecal progestin concentrations from mating to diapause (March 19) did not differ between mink that whelped or not, nor in response to the number of times mated. However, mean fecal progestin concentrations for mink that whelped were higher on March 25 (peri-implantation) than March 19 after being mated once (51.96±2.96 vs 23.53±1.89nM/g dry wt; P<0.05), twice (66.00±1.60 vs 25.57±1.28nM/g dry wt; P<0.05) or three times (66.48±1.42/g vs 19.16±1.09nM/g dry wt; P<0.05). During implantation (April 5), mean fecal progestin concentrations for mink that whelped after being mated once (146.60±10.02nM/g dry wt), twice (162.10±5.64nM/g dry wt) or three times (188.50±3.92nM/g dry wt) were significantly higher than for those that failed to whelp; 119.30±8.87nM/g dry wt, 77.84±5.86nM/g dry wt. and 118.9±6.55nM/g dry wt., respectively (P<0.05). Our findings suggest that measurement of fecal progestin concentrations during blastocyst reactivation and implantation may be a useful indicator of successful pregnancies in mink.


Assuntos
Fezes/química , Vison/fisiologia , Progestinas/fisiologia , Animais , Feminino , Gravidez , Taxa de Gravidez , Progestinas/química
16.
Acta Obstet Gynecol Scand ; 94 Suppl 161: 3-7, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26443945

RESUMO

The physiologic and clinical value of progesterone is undisputed and a cornerstone of human reproduction. Better understanding of the exact dynamics and effects of endogenous progesterone secretion, as well as its therapeutic actions, is critical to ensure optimal clinical results in artificial reproduction technology, and to enhance chances of successfully completing pregnancy. Novel progesterone-based drugs and administration regimens will provide clinicians with greater options to make the management and treatment of infertile couples less burdensome and more successful.


Assuntos
Progesterona/uso terapêutico , Progestinas/uso terapêutico , Técnicas de Reprodução Assistida , Endométrio/metabolismo , Feminino , Humanos , Progesterona/fisiologia , Progestinas/fisiologia , Hemorragia Uterina/tratamento farmacológico
17.
Pharmacol Biochem Behav ; 137: 1-6, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26190222

RESUMO

The following experiment was designed to test two specific questions: (1) Does the antiprogestin, RU486, reduce emergence of lordosis behavior and/or proceptivity in rats given repeated treatment with 10µg estradiol benzoate (EB) and/or a single high dose (40µg) of EB? (2) Does RU486 accentuate the effects of a 5min restraint experience on sexual behaviors in rats given repeated treatment with estradiol benzoate (EB) and/or a high dose of EB? RU486 was used to determine if a high dose and/or repeated treatment with EB enhanced proceptivity and reduced the response to mild stress through an intracellular progesterone receptor-mediated process. Ovariectomized Fischer rats were injected with a single dose of 10 or 40µg estradiol benzoate (EB) or received 4consecutiveweeks of treatment with 10µg EB. Forty-eight hours after the last treatment with EB, rats were injected with 5mg/kg of the antiprogestin, RU486, or the RU486 vehicle. That afternoon, rats were monitored for sexual behaviors. Sexually-receptive rats were then restrained for 5min and again tested for sexual behaviors. A separate set of rats received 4consecutiveweeks of 10µg EB treatment before treatment with a higher (5mg/rat) dose of RU486. Lordosis to mount ratios, lordosis quality, proceptivity, and resistance were monitored. RU486 had no effect on the emergence of sexual behaviors but did accentuate the lordosis-inhibitory effect of restraint in rats given a single treatment with EB. Rats treated for 4consecutiveweeks with EB showed no effect of restraint and were unaffected by RU486. These findings lead to the suggestion that repeated EB initiates select behavioral effects that are not mimicked by acute EB treatment and that the intracellular progesterone receptor may not be involved.


Assuntos
Estradiol/análogos & derivados , Antagonistas de Hormônios/farmacologia , Mifepristona/farmacologia , Progestinas/antagonistas & inibidores , Comportamento Sexual Animal/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Estradiol/administração & dosagem , Feminino , Masculino , Ovariectomia , Progestinas/fisiologia , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Restrição Física , Comportamento Sexual Animal/fisiologia
18.
Steroids ; 100: 21-6, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25911435

RESUMO

Previously, we established cell lines stably producing goldfish membrane progestin receptor α (goldfish mPRα) proteins, which mediate steroidal nongenomic actions. In this study, we transfected these cell lines (MDA-MD-231) with cDNAs encoding a recombinant luciferase gene (GloSensor). These cells can be used for monitoring the effects of ligands that bind to mPR by means of luminescence, the intensity of which reflects intracellular cyclic adenosine monophosphate (cAMP) levels. Luminescence intensity of the cells increased significantly when cells were treated with forskolin, strong activator of adenylyl cyclase. Then, we established a strategy to measure changes in luminescence that correlated with the actions of the ligands. The actions of ligands were measurable by the prevention of stimulation caused by forskolin after ligand stimulation. The studies using these cell lines indicated that cAMP concentrations were decreased specifically by the mPR ligands 17α,20ß-dihydroxy-4-pregnen-3-one, diethylstilbestrol and progesterone. Furthermore, pertussis toxin inhibited the decrease in cAMP levels caused by mPR ligands. These results support evidence from previous results that mPRα is coupled to an inhibitory G protein.


Assuntos
Proteínas de Peixes/fisiologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Progestinas/fisiologia , Receptores de Progesterona/fisiologia , Linhagem Celular Tumoral , AMP Cíclico/metabolismo , Humanos , Sistemas do Segundo Mensageiro
19.
J Steroid Biochem Mol Biol ; 139: 201-22, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23583279

RESUMO

In this review we focused on steroid metabolomics in human fetuses and newborns and its role in the physiology and pathophysiology of human pregnancy and subsequent stages of human life, and on the physiological relevance of steroids influencing the nervous systems with regards to their concentrations in the fetus. Steroid profiling provides valuable data for the diagnostics of diseases related to altered steroidogenesis in the fetal and maternal compartments and placenta. We outlined a potential use of steroid metabolomics for the prediction of reproductive disorders, misbalance of hypothalamic-pituitary-adrenal axis, and impaired insulin sensitivity in subsequent stages of human life. A possible role of steroids exhibiting a non-genomic effect in the development of gestational diabetes and in the neuroprotection via negative modulation of AMPA/kainate receptors was also indicated. Increasing progesterone synthesis and catabolism, declining production of tocolytic 5ß-pregnane steroids, and rising activities of steroid sulfotransferases with the approaching term may be of importance in sustaining pregnancy. An increasing trend was demonstrated with advancing gestation toward the production of ketones (and 3ß-hydroxyl groups in the case of 3α-hydroxy-steroids) was demonstrated in the fetus on the expense of 3α-hydroxy-, 17ß-hydroxy-, and 20α-hydroxy-groups weakening in the sequence C17, C3, and C20. There was higher production of active progestogen but lower production of active estrogen and GABAergic steroids with the approaching term. Rising activities of placental CYP19A1 and oxidative isoforms of HSD17B, and of fetal CYP3A7 with advancing gestation may protect the fetus from hyperestrogenization. This article is part of a Special Issue entitled 'Pregnancy and Steroids'.


Assuntos
Encéfalo/metabolismo , Feto/metabolismo , Hormônios Esteroides Gonadais/fisiologia , Corticosteroides/fisiologia , Animais , Estrogênios/fisiologia , Feminino , Desenvolvimento Fetal , Humanos , Gravidez , Progestinas/fisiologia
20.
Mol Endocrinol ; 27(11): 1808-24, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24014651

RESUMO

Progesterone (P4) stimulates proliferation of the mammary epithelium by a mechanism that involves paracrine signaling mediated from progesterone receptor (PR)-positive to neighboring PR-negative cells. Here we used a primary mouse mammary epithelial cell (MEC) culture system to define the molecular mechanism by which P4 regulates the expression of target gene effectors of proliferation including the paracrine factor receptor and activator of nuclear factor κB ligand (RANKL). MECs from adult virgin mice grown and embedded in three-dimensional basement-membrane medium resemble mammary ducts in vivo structurally and with respect to other properties including a heterogeneous pattern of PR expression, P4 induction of RANKL and other target genes in a PR-dependent manner, and a proliferative response to progestin. RANKL was demonstrated to have multiple functional P4-responsive enhancers that bind PR in a hormone-dependent manner as detected by chromatin immunoprecipitation assay. P4 also stimulated recruitment of signal transducer and activator of transcription (Stat)5a to RANKL enhancers through an apparent tethering with PR. Analysis of primary MECs from Stat5a knockout mice revealed that P4 induction of RANKL and a broad range of other PR target genes required Stat5a, as did P4-stimulated cell proliferation. In the absence of Stat5a, PR binding was lost at selective RANKL enhancers but was retained with others, suggesting that Stat5a acts to facilitate PR DNA binding at selective sites and to function as a coactivator with DNA-bound PR at others. These results show that RANKL is a direct PR target gene and that Stat5a has a novel role as a cofactor in PR-mediated transcriptional signaling in the mammary gland.


Assuntos
Células Epiteliais/metabolismo , Ligante RANK/metabolismo , Receptores de Progesterona/metabolismo , Fator de Transcrição STAT5/metabolismo , Animais , Células Cultivadas , Elementos Facilitadores Genéticos , Feminino , Expressão Gênica , Regulação da Expressão Gênica , Glândulas Mamárias Animais/citologia , Camundongos , Camundongos Endogâmicos BALB C , Progesterona/fisiologia , Progestinas/fisiologia , Prolactina/fisiologia , Ligação Proteica , Ligante RANK/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...