Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.461
Filtrer
1.
Transl Psychiatry ; 14(1): 283, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997258

RÉSUMÉ

Return to use, or relapse, is a major challenge in the treatment of opioid use disorder (OUD). Relapse can be precipitated by several factors, including exposure to drug-conditioned cues. Identifying successful treatments to mitigate cue-induced relapse has been challenging, perhaps due to extinction memory recall (EMR) deficits. Previously, inhibition of estradiol (E2) signaling in the basolateral amygdala (BLA) impaired heroin-cue EMR. This effect was recapitulated by antagonism of BLA estrogen receptors (ER) in a sex-specific manner such that blocking ERα in males, but ERß in females, impaired EMR. However, it is unclear whether increased E2 signaling, in the BLA or systemically, enhances heroin-cue EMR. We hypothesized that ERß agonism would enhance heroin-cue EMR in a sex- and region-specific manner. To determine the capacity of E2 signaling to improve EMR, we pharmacologically manipulated ERß across several translationally designed experiments. First, male and female rats acquired heroin or sucrose self-administration. Next, during a cued extinction session, we administered diarylpropionitrile (DPN, an ERß agonist) and tested anxiety-like behavior on an open field. Subsequently, we assessed EMR in a cue-induced reinstatement test and, finally, measured ERß expression in several brain regions. Across all experiments, females took more heroin and sucrose than males and had greater responses during heroin-cued extinction. Administration of DPN in the BLA enhanced EMR in females only, driven by ERß's impacts on memory consolidation. Interestingly, however, systemic DPN administration improved EMR for heroin cues in both sexes across several different tests, but did not impact sucrose-cue EMR. Immunohistochemical analysis of ERß expression across several different brain regions showed that females only had greater expression of ERß in the basal nucleus of the BLA. Here, in several preclinical experiments, we demonstrated that ERß agonism enhances heroin-cue EMR and has potential utility in combatting cue-induced relapse.


Sujet(s)
Signaux , Récepteur bêta des oestrogènes , Extinction (psychologie) , Héroïne , Rappel mnésique , Animaux , Mâle , Femelle , Récepteur bêta des oestrogènes/agonistes , Récepteur bêta des oestrogènes/métabolisme , Héroïne/pharmacologie , Rats , Extinction (psychologie)/effets des médicaments et des substances chimiques , Extinction (psychologie)/physiologie , Rappel mnésique/effets des médicaments et des substances chimiques , Rappel mnésique/physiologie , Nitriles/pharmacologie , Groupe nucléaire basolatéral/métabolisme , Groupe nucléaire basolatéral/effets des médicaments et des substances chimiques , Propionates/pharmacologie , Facteurs sexuels , Autoadministration , Rat Sprague-Dawley , Dépendance à l'héroïne/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
2.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-39000147

RÉSUMÉ

Glyphosate, the active ingredient of several broad-spectrum herbicides, is widely used throughout the world, although many adverse effects are known. Among these, it has been recognized as an endocrine disruptor. This work aimed to test the effects and potential endocrine disrupting action of glyphosate on PNT1A human prostate cells, an immortalized non-tumor epithelial cell line, possessing both ERα and ERß estrogen receptors. The results showed that glyphosate induces cytotoxicity, mitochondrial dysfunction, and rapid activation of ERα and ERß via nuclear translocation. Molecular analysis indicated a possible involvement of apoptosis in glyphosate-induced cytotoxicology. The apoptotic process could be attributed to alterations in mitochondrial metabolism; therefore, the main parameters of mitochondrial functionality were investigated using the Seahorse analyzer. Impaired mitochondrial function was observed in glyphosate-treated cells, with reductions in ATP production, spare respiratory capacity, and proton leakage, along with increased efficiency of mitochondrial coupling. Finally, the results of immunofluorescence analysis demonstrated that glyphosate acts as an estrogen disruptor determining the nuclear translocation of both ERs. Nuclear translocation occurred independent of dose, faster than the specific hormone, and persisted throughout treatment. In conclusion, the results collected show that in non-tumor prostate cells glyphosate can cause cell death and acts as a xenoestrogen, activating estrogen receptors. The consequent alteration of hormonal functions can have negative effects on the reproductive health of exposed animals, compromising their fertility.


Sujet(s)
Apoptose , Récepteur alpha des oestrogènes , Récepteur bêta des oestrogènes , Glycine , , Mitochondries , Prostate , Glycine/analogues et dérivés , Glycine/pharmacologie , Glycine/toxicité , Humains , Mâle , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Récepteur bêta des oestrogènes/métabolisme , Récepteur alpha des oestrogènes/métabolisme , Prostate/effets des médicaments et des substances chimiques , Prostate/métabolisme , Prostate/anatomopathologie , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire , Herbicides/toxicité , Perturbateurs endocriniens/toxicité , Perturbateurs endocriniens/pharmacologie , Survie cellulaire/effets des médicaments et des substances chimiques
3.
Front Endocrinol (Lausanne) ; 15: 1408677, 2024.
Article de Anglais | MEDLINE | ID: mdl-38978624

RÉSUMÉ

Infertility is becoming a major public health problem, with increasing frequency due to medical, environmental and societal causes. The increasingly late age of childbearing, growing exposure to endocrine disruptors and other reprotoxic products, and increasing number of medical reproductive dysfunctions (endometriosis, polycystic ovary syndrome, etc.) are among the most common causes. Fertility relies on fine-tuned control of both neuroendocrine function and reproductive behaviors, those are critically regulated by sex steroid hormones. Testosterone and estradiol exert organizational and activational effects throughout life to establish and activate the neural circuits underlying reproductive function. This regulation is mediated through estrogen receptors (ERs) and androgen receptor (AR). Estradiol acts mainly via nuclear estrogen receptors ERα and ERß. The aim of this review is to summarize the genetic studies that have been undertaken to comprehend the specific contribution of ERα and ERß in the neural circuits underlying the regulation of the hypothalamic-pituitary-gonadal axis and the expression of reproductive behaviors, including sexual and parental behavior. Particular emphasis will be placed on the neural role of these receptors and the underlying sex differences.


Sujet(s)
Récepteur alpha des oestrogènes , Récepteur bêta des oestrogènes , Reproduction , Humains , Récepteur alpha des oestrogènes/métabolisme , Récepteur bêta des oestrogènes/métabolisme , Animaux , Reproduction/physiologie , Femelle , Comportement procréatif/physiologie , Mâle
4.
Cell Commun Signal ; 22(1): 367, 2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39030619

RÉSUMÉ

Colorectal cancer (CRC) is a common gastrointestinal malignancy with higher incidence and mortality rates in men compared to women, potentially due to the effects of estrogen signaling. There is substantial evidence supporting the significant role of 17ß-Estradiol (E2) in reducing CRC risk in females, although this perspective remains debated. E2 has been demonstrated to inhibit CRC cell proliferation and migration at the cellular level by enhancing DNA mismatch repair, modulating key gene expression, triggering cell cycle arrest, and reducing activity of migration factors. Furthermore, E2 contributes to promote a tumor microenvironment unfavorable for CRC growth by stimulating ERß expression, reducing inflammatory responses, reversing immunosuppression, and altering the gut microbiome composition. Conversely, under conditions of high oxidative stress, hypoxia, and nutritional deficiencies, E2 may facilitate CRC development through GPER-mediated non-genomic signaling. E2's influence on CRC involves the genomic and non-genomic signals mediated by ERß and GPER, respectively, leading to its dual roles in anticancer activity and carcinogenesis. This review aims to summarize the potential mechanisms by which E2 directly or indirectly impacts CRC development, providing insights into the phenomenon of sexual dimorphism in CRC and suggesting potential strategies for prevention and treatment.


Sujet(s)
Tumeurs colorectales , Oestradiol , Humains , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Oestradiol/métabolisme , Animaux , Récepteur bêta des oestrogènes/métabolisme , Récepteur bêta des oestrogènes/génétique , Microenvironnement tumoral , Transduction du signal
5.
FASEB J ; 38(11): e23719, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38837828

RÉSUMÉ

Chronic disruption of circadian rhythms by night shift work is associated with an increased breast cancer risk. However, little is known about the impact of night shift on peripheral circadian genes (CGs) and circadian-controlled genes (CCGs) associated with breast cancer. Hence, we assessed central clock markers (melatonin and cortisol) in plasma, and peripheral CGs (PER1, PER2, PER3, and BMAL1) and CCGs (ESR1 and ESR2) in peripheral blood mononuclear cells (PBMCs). In day shift nurses (n = 12), 24-h rhythms of cortisol and melatonin were aligned with day shift-oriented light/dark schedules. The mRNA expression of PER2, PER3, BMAL1, and ESR2 showed 24-h rhythms with peak values in the morning. In contrast, night shift nurses (n = 10) lost 24-h rhythmicity of cortisol with a suppressed morning surge but retained normal rhythmic patterns of melatonin, leading to misalignment between cortisol and melatonin. Moreover, night shift nurses showed disruption of rhythmic expressions of PER2, PER3, BMAL1, and ESR2 genes, resulting in an impaired inverse correlation between PER2 and BMAL1 compared to day shift nurses. The observed trends of disrupted circadian markers were recapitulated in additional day (n = 20) and night (n = 19) shift nurses by measurement at early night and midnight time points. Taken together, this study demonstrated the misalignment of cortisol and melatonin, associated disruption of PER2 and ESR2 circadian expressions, and internal misalignment in peripheral circadian network in night shift nurses. Morning plasma cortisol and PER2, BMAL1, and ESR2 expressions in PBMCs may therefore be useful biomarkers of circadian disruption in shift workers.


Sujet(s)
Horloges circadiennes , Rythme circadien , Hydrocortisone , Mélatonine , Horaire de travail posté , Humains , Femelle , Mélatonine/métabolisme , Mélatonine/sang , Adulte , Horaire de travail posté/effets indésirables , Horloges circadiennes/génétique , Hydrocortisone/sang , Hydrocortisone/métabolisme , Rythme circadien/physiologie , Protéines circadiennes Period/génétique , Protéines circadiennes Period/métabolisme , Infirmières et infirmiers , Agranulocytes/métabolisme , Récepteur alpha des oestrogènes/métabolisme , Récepteur alpha des oestrogènes/génétique , Récepteur bêta des oestrogènes/métabolisme , Récepteur bêta des oestrogènes/génétique , Facteurs de transcription ARNTL/génétique , Facteurs de transcription ARNTL/métabolisme , Tolérance à l'horaire de travail/physiologie , Conditions de Travail
6.
Ecotoxicol Environ Saf ; 280: 116521, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38850708

RÉSUMÉ

The aim of this study is to investigate the role of estrogen receptor ß (ERß) in nonylphenol (NP) - induced depression - like behavior in rats and its impact on the regulation of the TPH2/5-HT pathway. In the in vitro experiment, rat basophilic leukaemia cells (RBL-2H3) cells were divided into the four groups: blank group, NP group (20 µM), ERß agonist group (0.01 µM), and NP+ERß agonist group (20 µM+0.01 µM). For the in vivo experiment, 72 adult male Sprague-Dawley rats were randomly divided into following six groups: the Control, NP (40 mg/kg) group, ERß agonist (2 mg/kg, Diarylpropionitrile (DPN)) group, ERß inhibitor (0.1 mg/kg, 4-(2-phenyl-5,7-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-3-yl) phenol (PHTPP)) group, NP+ERß agonist (40 mg/kg NP + 2 mg/kg DPN) group, and NP+ERß inhibitor (40 mg/kg NP + 0.1 mg/kg PHTPP) group, with 12 rats in each group. Each rat in drug group were given NP by gavage and/or received a single intraperitoneal injection of DPN 2 mg/kg or PHTPP 0.1 mg/kg. Both in vivo and in vitro, NP group showed a decrease in the expression levels of ERß, tryptophan hydroxylase (TPH1), and tryptophan hydroxylase-2 (TPH2) genes and proteins, and reduced levels of DA, NE, and 5-hydroxytryptophan (5-HT) neurotransmitters. RBL-2H3 cells showed signs of cell shrinkage, with rounded cells, increased suspension and more loosely arranged cells. The effectiveness of the ERß agonist stimulation exhibited an increase exceeding 60% in RBL-2H3 cells. The application of ERß agonist resulted in an alleviation the aforementioned alterations. ERß agonist activated the TPH2/5-HT signaling pathways. Compared to the control group, the NP content in the brain tissue of the NP group was significantly increased. The latency to eat for the rats was longer and the amount of food consumed was lower, and the rats had prolonged immobility time in the behavioral experiment of rats. The expression levels of ERß, TPH1, TPH2, 5-HT and 5-HITT proteins were decreased in the NP group, suggesting NP-induced depression-like behaviours as well as disturbances in the secretion of serum hormones and monoamine neurotransmitters. In the NP group, the midline raphe nucleus showed an elongated nucleus with a dark purplish-blue colour, nuclear atrophy, displacement and pale cytoplasm. ERß might ameliorate NP-induced depression-like behaviors, and secretion disorders of serum hormones and monoamine neurotransmitters via activating TPH2/5-HT signaling pathways.


Sujet(s)
Dépression , Récepteur bêta des oestrogènes , Phénols , Rat Sprague-Dawley , Sérotonine , Tryptophane 5-monooxygenase , Animaux , Tryptophane 5-monooxygenase/métabolisme , Récepteur bêta des oestrogènes/métabolisme , Phénols/toxicité , Mâle , Rats , Sérotonine/métabolisme , Dépression/induit chimiquement , Dépression/traitement médicamenteux , Dépression/métabolisme , Agents neuromédiateurs/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Nitriles/toxicité , Nitriles/pharmacologie , Propionates/toxicité , Propionates/pharmacologie , Pyrazoles , Pyrimidines
7.
Wei Sheng Yan Jiu ; 53(3): 441-454, 2024 May.
Article de Chinois | MEDLINE | ID: mdl-38839586

RÉSUMÉ

OBJECTIVE: To investigate the effects of long-term(7 days and 14 days) bisphenol S(BPS) exposure on the ERß-MAPK signaling pathway, hormone secretion phenotype and cell cycle in human normal ovarian epithelial cells IOSE 80 at actual human exposure level. METHODS: Physiologically based pharmacokinetic model combined with BPS levels in the serum of women along the Yangtze River in China was used to determine the dosing concentrations of BPS, and vehicle control and 17 ß-estradiol(E_2) control were used. Complete medium with corresponding concentrations(0, 6.79×10~(-6), 6.79×10~(-4), 6.79×10~(-2), 6.79 µmol/L BPS and 10 nmol/L E_2) was replaced every 2 days. mRNA expressions of estrogen receptor(ERß and GPR30), key genes in MAPK signaling pathway(P38/JNK/ERK signaling pathway) and gonadotropin-releasing hormone-related genes(GnRH-I, GnRH-II and GnRH-R) were measured by qPCR. The ERß-MAPK signaling pathway inhibitors were employed to detect the effect of long-term exposure to BPS on the cell cycle by flow cytometry. Dose-response relationship analysis was performed to calculate the benchmark does lower confidence limits. RESULTS: Compared to the vehicle control, after 7 days exposure to BPS, the ratio of G_2/M phase was significantly increased(P<0.05), and the mRNA expressions of GnRH-I, GnRH-II and GnRH-R were significantly decreased(P<0.05); after 14 days exposure to BPS, the mRNA expressions of ESR2, MAPK3, and MAPK9 were significantly increased(P<0.05), and the mRNA expressions of GnRH-II and GnRH-R were significantly decreased(P<0.05). The GnRH-II mRNA expression level of BPS treatment for 7 days; the G_0/G_1 phase ratio, MAPK3 and MAPK8 mRNA expression level of BPS exposure for 14 days; and the GnRH-I mRNA expression level after BPS treatment for 7 days and 14 days showed a good dose-response relationship but with poor fit. CONCLUSION: Long-term low-dose exposure to BPS may cause cell cycle arrest by activating the ERß-MAPK signaling pathway, and may lead to changes in the hormone secretion of IOSE 80 cells.


Sujet(s)
Cellules épithéliales , Récepteur bêta des oestrogènes , Système de signalisation des MAP kinases , Ovaire , Phénols , Sulfones , Humains , Phénols/toxicité , Femelle , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Récepteur bêta des oestrogènes/métabolisme , Récepteur bêta des oestrogènes/génétique , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Ovaire/effets des médicaments et des substances chimiques , Ovaire/métabolisme , Sulfones/toxicité , Lignée cellulaire
8.
Gene ; 927: 148678, 2024 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-38906392

RÉSUMÉ

Estrogen and estrogen receptors (ERα and ERß) regulate a multitude of complicated physiological and pathological processes. Jan-Ake Gustafsson's group discovered ERß in 1996, this crucial finding gives us new insights into the understanding of estrogen signaling. ERß is highly expressed in the ovary and particularly exists in granulosa cells (GCs). ERß is a key transcription factor in the maintenance of ovarian granulosa cell growth, differentiation, and homeostasis, and the ovulation function of ovarian follicles and oocytes. Additionally, ERß can modulate the steroidogenic transcriptional program through phosphorylation and regulate both gonadotropin response and FOXL2 expression within the ovary. In this review, we focus on the role of ERß in regulating ovarian granulosa cell development and homeostasis, particularly its significance in ovarian cancer (OC), premature ovarian failure (POF), and polycystic ovary syndrome (PCOS). It also highlights the prospects of small molecule compounds targeting ERß, providing a new strategy for the treatment of ovarian-related diseases.


Sujet(s)
Récepteur bêta des oestrogènes , Tumeurs de l'ovaire , Ovaire , Syndrome des ovaires polykystiques , Insuffisance ovarienne primitive , Humains , Femelle , Récepteur bêta des oestrogènes/métabolisme , Récepteur bêta des oestrogènes/génétique , Syndrome des ovaires polykystiques/métabolisme , Syndrome des ovaires polykystiques/génétique , Ovaire/métabolisme , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/génétique , Insuffisance ovarienne primitive/métabolisme , Insuffisance ovarienne primitive/génétique , Cellules de la granulosa/métabolisme , Animaux , Protéine L2 à motif en tête de fourche/métabolisme , Protéine L2 à motif en tête de fourche/génétique , Transduction du signal , Maladies ovariennes/métabolisme , Maladies ovariennes/génétique , Maladies ovariennes/anatomopathologie
9.
Int J Mol Sci ; 25(11)2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38892441

RÉSUMÉ

In this narrative review, we attempt to provide an overview of the evidence regarding the role of estrogen (receptors) in cutaneous melanoma (CM). We reviewed 68 studies and 4 systematic reviews and meta-analyses published from 2002 up to and including 2022. The prevailing presence of estrogen receptor ß (ERß) instead of estrogen receptor α (ERα) in CM is notable, with ERß potentially playing a protective role and being less frequently detected in progressive cases. While men with CM generally experience a less favorable prognosis, this distinction may become negligible with advancing age. The role of oral contraceptives (OC) and hormone replacement therapy (HRT) in CM remains controversial. However, recent studies tend to associate the use of these exogenous hormones with a heightened risk of CM, mostly only when using estrogen therapy and not in combination with progesterone. On the contrary, the majority of studies find no substantial influence of in vitro fertilization (IVF) treatment on CM risk. Reproductive factors, including younger age at first childbirth, higher parity, and shorter reproductive life, show conflicting evidence, with some studies suggesting a lower CM risk. We suggest an important role for estrogens in CM. More research is needed, but the integration of estrogens and targeting the estrogen receptors in melanoma therapy holds promise for future developments in the field.


Sujet(s)
Oestrogènes , Mélanome , Humains , Mélanome/métabolisme , Oestrogènes/métabolisme , Récepteurs des oestrogènes/métabolisme , Tumeurs cutanées/métabolisme , Femelle , Récepteur bêta des oestrogènes/métabolisme , Récepteur alpha des oestrogènes/métabolisme
11.
Scand J Med Sci Sports ; 34(6): e14668, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38802727

RÉSUMÉ

Multiple intramuscular variables have been proposed to explain the high variability in resistance training induced muscle hypertrophy across humans. This study investigated if muscular androgen receptor (AR), estrogen receptor α (ERα) and ß (ERß) content and fiber capillarization are associated with fiber and whole-muscle hypertrophy after chronic resistance training. Male (n = 11) and female (n = 10) resistance training novices (22.1 ± 2.2 years) trained their knee extensors 3×/week for 10 weeks. Vastus lateralis biopsies were taken at baseline and post the training period to determine changes in fiber type specific cross-sectional area (CSA) and fiber capillarization by immunohistochemistry and, intramuscular AR, ERα and ERß content by Western blotting. Vastus lateralis volume was quantified by MRI-based 3D segmentation. Vastus lateralis muscle volume significantly increased over the training period (+7.22%; range: -1.82 to +18.8%, p < 0.0001) but no changes occurred in all fiber (+1.64%; range: -21 to +34%, p = 0.869), type I fiber (+1.33%; range: -24 to +41%, p = 0.952) and type II fiber CSA (+2.19%; range: -23 to +29%, p = 0.838). However, wide inter-individual ranges were found. Resistance training increased the protein expression of ERα but not ERß and AR, and the increase in ERα content was positively related to changes in fiber CSA. Only for the type II fibers, the baseline capillary-to-fiber-perimeter index was positively related to type II fiber hypertrophy but not to whole muscle responsiveness. In conclusion, an upregulation of ERα content and an adequate initial fiber capillarization may be contributing factors implicated in muscle fiber hypertrophy responsiveness after chronic resistance training.


Sujet(s)
Récepteur alpha des oestrogènes , Récepteur bêta des oestrogènes , Fibres musculaires squelettiques , Muscle quadriceps fémoral , Récepteurs aux androgènes , Entraînement en résistance , Humains , Mâle , Entraînement en résistance/méthodes , Femelle , Récepteur bêta des oestrogènes/métabolisme , Récepteur alpha des oestrogènes/métabolisme , Jeune adulte , Récepteurs aux androgènes/métabolisme , Muscle quadriceps fémoral/métabolisme , Muscle quadriceps fémoral/vascularisation , Muscle quadriceps fémoral/imagerie diagnostique , Fibres musculaires squelettiques/métabolisme , Fibres musculaires squelettiques/physiologie , Adulte , Hypertrophie , Vaisseaux capillaires , Imagerie par résonance magnétique
12.
Genes (Basel) ; 15(5)2024 05 17.
Article de Anglais | MEDLINE | ID: mdl-38790265

RÉSUMÉ

The estrogen receptor signaling pathway plays an important role in vertebrate embryonic development and sexual differentiation. There are four major estrogen receptors in zebrafish: esr1, esr2a, esr2b and gper. However, the specific role of different estrogen receptors in zebrafish is not clear. To investigate the role of esr2b in zebrafish development and reproduction, this study utilized TALENs technology to generate an esr2b knockout homozygous zebrafish line. The number of eggs laid by esr2b knockout female zebrafish did not differ significantly from that of wild zebrafish. The embryonic development process of wild-type and esr2b knockout zebrafish was observed, revealing a significant developmental delay in the esr2b knockout zebrafish. Additionally, mortality rates were significantly higher in esr2b knockout zebrafish than in their wild-type counterparts at 24 hpf. The reciprocal cross experiment between esr2b knockout zebrafish and wild-type zebrafish revealed that the absence of esr2b resulted in a decline in the quality of zebrafish oocytes, while having no impact on sperm cells. The knockout of esr2b also led to an abnormal sex ratio in the adult zebrafish population, with a female-to-male ratio of approximately 1:7. The quantitative PCR (qPCR) and in situ hybridization results demonstrated a significant downregulation of cyp19ab1b expression in esr2b knockout embryos compared to wild-type embryos throughout development (at 2 dpf, 3 dpf and 4 dpf). Additionally, the estrogen-mediated induction expression of cyp19ab1b was attenuated, while the estradiol-induced upregulated expression of vtg1 was disrupted. These results suggest that esr2b is involved in regulating zebrafish oocyte development and sex differentiation.


Sujet(s)
Récepteur bêta des oestrogènes , Protéines de poisson-zèbre , Danio zébré , Animaux , Femelle , Mâle , Aromatase/génétique , Aromatase/métabolisme , Développement embryonnaire , Récepteur bêta des oestrogènes/génétique , Récepteur bêta des oestrogènes/métabolisme , Régulation de l'expression des gènes au cours du développement , Techniques de knock-out de gènes , Ovocytes/métabolisme , Ovocytes/croissance et développement , Différenciation sexuelle , Sexe-ratio , Danio zébré/génétique , Protéines de poisson-zèbre/génétique , Protéines de poisson-zèbre/métabolisme
13.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 132-138, 2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38814223

RÉSUMÉ

We investigated the influence of 17ß-estradiol (17ß-E2) on cartilage extracellular matrix (ECM) homeostasis in postmenopausal women. We focused on the roles of estrogen receptors (ESR) and SOX6 in 17ß-E2-mediated stimulation of ECM metabolism during chondrocyte (CH) degeneration. We compared the expression of anabolic genes (collagen II and aggrecan) and catabolic genes (MMPs and TIMPs) in IL-1ß-induced CH degeneration in vitro, with and without 17ß-E2 supplementation. We separately silenced the SOX6, ESR1, and ESR2 genes in CHs to determine their impact on 17ß-E2 treatment. Additionally, we used Chromatin immunoprecipitation followed by DNA sequencing (ChIP-seq) and luciferase assays to investigate protein-DNA interactions within ESR2 and SOX6-promoter complexes. After three days of IL-1ß treatment, ESR1/2, SOX6, collagen II, aggrecan, and TIMP1/3 were decreased, while MMP3/9/13 were increased. The addition of 17ß-E2 partially reversed these effects, but silencing SOX6, ESR1, or ESR2 weakened the protective effects of 17ß-E2. Silencing ESR2, but not ESR1, abolished the upregulation of SOX6 induced by 17ß-E2. ESR2 was found to bind the SOX6 promoter and regulate SOX6 expression. 17ß-E2 upregulates SOX6 through ESR2 mediation, and the synergistic effect of 17ß-E2 and ESR2 on SOX6 balances ECM metabolism in CHs.


Sujet(s)
Chondrocytes , Oestradiol , Récepteur bêta des oestrogènes , Matrice extracellulaire , Interleukine-1 bêta , Facteurs de transcription SOX-D , Chondrocytes/métabolisme , Chondrocytes/effets des médicaments et des substances chimiques , Oestradiol/pharmacologie , Humains , Récepteur bêta des oestrogènes/métabolisme , Récepteur bêta des oestrogènes/génétique , Femelle , Matrice extracellulaire/métabolisme , Matrice extracellulaire/effets des médicaments et des substances chimiques , Facteurs de transcription SOX-D/métabolisme , Facteurs de transcription SOX-D/génétique , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/pharmacologie , Récepteur alpha des oestrogènes/métabolisme , Récepteur alpha des oestrogènes/génétique , Régions promotrices (génétique)/génétique , Cellules cultivées
14.
J Steroid Biochem Mol Biol ; 242: 106526, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38657699

RÉSUMÉ

Estrogen drives the growth of some cancers, such as breast cancer, via estrogen receptor alpha (ERα). Estrogen also activates ERß, but whether ERß is expressed and has a role in different cancers is debated. The use of nonspecific antibodies has contributed to the confusion, and this review delves into ERß's controversial role in cancer and focuses on tumor expression that can be supported by non-antibody-dependent assays. We discuss its expression at the transcript level and focus on its potential role in lymphoma, granulosa cell tumors, testicular, and adrenal cancers, emphasizing recent findings and the complexities that necessitate further research.


Sujet(s)
Récepteur bêta des oestrogènes , Tumeurs , Humains , Récepteur bêta des oestrogènes/métabolisme , Récepteur bêta des oestrogènes/génétique , Tumeurs/métabolisme , Tumeurs/génétique , Femelle , Animaux , Mâle , Régulation de l'expression des gènes tumoraux , Tumeurs du testicule/métabolisme , Tumeurs du testicule/génétique , Tumeurs du testicule/anatomopathologie , Tumeur de la granulosa/métabolisme , Tumeur de la granulosa/génétique , Tumeur de la granulosa/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Tumeurs de la surrénale/génétique , Tumeurs de la surrénale/métabolisme , Tumeurs de la surrénale/anatomopathologie , Lymphomes/métabolisme , Lymphomes/génétique , Lymphomes/anatomopathologie
15.
Poult Sci ; 103(6): 103734, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38636201

RÉSUMÉ

Dietary supplementation with bioactive substances that can regulate lipid metabolism is an effective approach for reducing excessive fat deposition in chickens. Genistein (GEN) has the potential to alleviate fat deposition; however, the underlying mechanism of GEN's fat-reduction action in chickens remains unclear. Therefore, the present study aimed to explore the underlying mechanism of GEN on the reduction of fat deposition from a novel perspective: intercellular transmission of adipokine between adipocytes and hepatocytes. The findings showed that GEN enhanced the secretion of adiponectin (APN) in chicken adipocytes, and the enhancement effect of GEN was completely blocked when the cells were pretreated with inhibitors targeting estrogen receptor ß (ERß) or proliferator-activated receptor γ (PPARγ) signals, respectively. Furthermore, the results demonstrated that both co-treatment with GEN and APN or treatment with the medium supernatant (Med SUP) derived from chicken adipocytes treated with GEN significantly decreased the content of triglyceride and increased the protein levels of ERß, Sirtuin 1 (SIRT1) and phosphor-AMP-activated protein kinase (p-AMPK) in chicken hepatocytes compared to the cells treated with GEN or APN alone. Moreover, the increase in the protein levels of SIRT1 and p-AMPK induced by GEN and APN co-treatment or Med SUP treatment were blocked in chicken hepatocytes pretreated with the inhibitor of ERß signals. Importantly, the up-regulatory effect of GEN and APN co-treatment or Med SUP treatment on the protein level of p-AMPK was also blocked in chicken hepatocytes pretreated with a SIRT1 inhibitor; however, the increase in the protein level of SIRT1 induced by GEN and APN co-treatment or Med SUP treatment was not reversed when the hepatocytes were pretreated with an AMPK inhibitor. In conclusion, the present study demonstrated that GEN enhanced APN secretion by activating the ERß-Erk-PPARγ signaling pathway in chicken adipocytes. Subsequently, adipocyte-derived APN synergized with GEN to activate the ERß-mediated SIRT1-AMPK signaling pathway in chicken hepatocytes, ultimately reducing fat deposition. These findings provide substantial evidence from a novel perspective, supporting the potential use of GEN as a dietary supplement to prevent excessive fat deposition in poultry.


Sujet(s)
Adiponectine , Poulets , Récepteur bêta des oestrogènes , Génistéine , Hépatocytes , Transduction du signal , Sirtuine-1 , Animaux , Génistéine/pharmacologie , Génistéine/administration et posologie , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Sirtuine-1/métabolisme , Récepteur bêta des oestrogènes/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Adiponectine/métabolisme , AMP-Activated Protein Kinases/métabolisme , Protéines aviaires/métabolisme , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Tissu adipeux/effets des médicaments et des substances chimiques
16.
J Biol Inorg Chem ; 29(2): 177-186, 2024 03.
Article de Anglais | MEDLINE | ID: mdl-38581541

RÉSUMÉ

The anti-proliferative activity of the known metalloantibiotic {[Ag(CIPH)2]NO3∙0.75MeOH∙1.2H2O} (CIPAG) (CIPH = ciprofloxacin) against the human breast adenocarcinoma cancer cells MCF-7 (hormone dependent (HD)) and MDA-MB-231 (hormone independent (HI)) is evaluated. The in vitro toxicity and genotoxicity of the metalloantibiotic were estimated toward fetal lung fibroblast (MRC-5) cells. The molecular mechanism of the CIPAG activity against MCF-7 cells was clarified by the (i) cell morphology, (ii) cell cycle arrest, (iii) mitochondrial membrane permeabilization, and (iv) by the assessment of the possible differential effect of CIPAG on estrogen receptor alpha (ERα) and estrogen receptor beta (ERß) transcriptional activation, applying luciferase reporter gene assay. Moreover, the ex vivo mechanism of CIPAG was clarified by its binding affinity toward calf thymus (CT-DNA).


Sujet(s)
Antinéoplasiques , Tumeurs du sein , Ciprofloxacine , Humains , Ciprofloxacine/pharmacologie , Ciprofloxacine/composition chimique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Femelle , Récepteur bêta des oestrogènes/métabolisme , Récepteur bêta des oestrogènes/génétique , Argent/composition chimique , Argent/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Récepteur alpha des oestrogènes/métabolisme , ADN/métabolisme , ADN/composition chimique , Tests de criblage d'agents antitumoraux , Animaux , Cellules MCF-7 , Lignée cellulaire tumorale
17.
Anat Histol Embryol ; 53(3): e13042, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38634511

RÉSUMÉ

To observe developmental changes in the ovarian tissue structure and distribution characteristics of oestrogen receptors (ERs) in the ovaries of Huanghuai goats at different ages, we selected healthy Huanghuai goats ewes and divided them into five groups (i.e. 3-, 30-, 60-, 90- and 120-day-old groups), with 10 animals in each group. The serum was separated after blood collection through the jugular vein, and the contents of oestrogen (E) and progesterone (P) in the serum of Huanghuai goats at each age were determined. Three goats were randomly selected from each group and sacrificed after anaesthesia, and the ovarian tissue was quickly obtained and placed in 4% paraformaldehyde fixative to prepare the tissue sections. Using HE, oestrogen receptors were immunohistochemically stained and observed. These results showed many primordial follicles and occasional secondary follicles in the ovaries of 3-day-old Huanghuai goats. Ovarian reticular structures were observed in 30-day-old ovarian medulla, with occasional near-mature growing follicles. Mature follicles and corpus luteum were occasionally detected in 60-day-old ovarian cortex. The 90-120-day-old ovarian cortices contained growing and mature follicles, and the number of mature follicles and corpora lutea increased, implying a significant luteal involution period. The E and P contents in the 120-day-old group were significantly higher than those in the 3-, 30-, 60- and 90-day-old groups. The levels of ERα and ERß in the 3- and 30-day-old groups were mainly distributed in the granulosa cells of ovarian reproductive epithelial cells, primordial follicles, atretic follicles, and primary and secondary follicles. The ERα and ERß levels of the 60-, 90- and 120-day-old groups were also distributed in the granulosa cells and luteal cells of mature follicles, especially in the 120-day-old endometrial cells of mature follicles, where ERß was distributed significantly. The overall expression of ERß in the ovary was higher than that of ERα. The results of this study provide basic data on the ovarian development and the specific expression of ERs and PRs in the ovaries of Huanghuai white goats, which play an important role in ovarian development and precocity.


Sujet(s)
Ovaire , Récepteurs des oestrogènes , Femelle , Animaux , Ovis , Ovaire/métabolisme , Récepteurs des oestrogènes/analyse , Récepteurs des oestrogènes/métabolisme , Récepteur alpha des oestrogènes/analyse , Récepteur bêta des oestrogènes , Capra/métabolisme
18.
BMC Cancer ; 24(1): 505, 2024 Apr 22.
Article de Anglais | MEDLINE | ID: mdl-38649856

RÉSUMÉ

Lung adenocarcinoma is the main type of lung cancer in women. Our previous findings have evidenced that 25-hydroxycholesterol (25-HC) promotes migration and invasion of lung adenocarcinoma cells (LAC), during which LXR as a 25-HC receptor plays an important role. Estrogen receptor beta (ERß) is a receptor of 27-hydroxycholesterol that is structurally analogous to 25-HC, but its role in the functional actions of 25-HC remained largely unknown. In this study, we demonstrated that 25-HC treatment triggered ERß expression in LAC. Knockdown of ERß inhibited 25-HC-mediated proliferation, migration and invasion, and reduced 25-HC-induced LAC metastasis in vivo. Further investigation revealed that ERß knockdown restrained the expression of TNFRSF17 (BCMA). In vivo experiments also confirmed that ERß knockdown blocked 25-HC-induced TNFRSF17 expression. TNFRSF17 knockdown also restrained 25-HC-induced proliferation, migration and invasion. Bioinformatic analysis showed that the levels of ERß and TNFRSF17 were elevated in lung adenocarcinoma, and were closely related to tumor stages and nodal metastasis status. These results suggested that 25-HC promoted the proliferation and metastasis of LAC by regulating ERß/TNFRSF17 axis.


Sujet(s)
Adénocarcinome pulmonaire , Mouvement cellulaire , Prolifération cellulaire , Récepteur bêta des oestrogènes , Hydroxycholestérols , Tumeurs du poumon , Animaux , Femelle , Humains , Mâle , Souris , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/secondaire , Lignée cellulaire tumorale , Récepteur bêta des oestrogènes/métabolisme , Récepteur bêta des oestrogènes/génétique , Régulation de l'expression des gènes tumoraux , Hydroxycholestérols/pharmacologie , Hydroxycholestérols/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/secondaire , Tumeurs du poumon/génétique , Souris de lignée BALB C , Souris nude , Métastase tumorale , Transduction du signal
19.
Cells ; 13(7)2024 Apr 06.
Article de Anglais | MEDLINE | ID: mdl-38607081

RÉSUMÉ

Increased activation of ovarian primordial follicles in Erß knockout (ErßKO) rats becomes evident as early as postnatal day 8.5. To identify the ERß-regulated genes that may control ovarian primordial follicle activation, we analyzed the transcriptome profiles of ErßKO rat ovaries collected on postnatal days 4.5, 6.5, and 8.5. Compared to wildtype ovaries, ErßKO ovaries displayed dramatic downregulation of Indian hedgehog (Ihh) expression. IHH-regulated genes, including Hhip, Gli1, and Ptch1, were also downregulated in ErßKO ovaries. This was associated with a downregulation of steroidogenic enzymes Cyp11a1, Cyp19a1, and Hsd17b1. The expression of Ihh remained very low in ErßKO ovaries despite the high levels of Gdf9 and Bmp15, which are known upregulators of Ihh expression in the granulosa cells of activated ovarian follicles. Strikingly, the downregulation of the Ihh gene in ErßKO ovaries began to disappear on postnatal day 16.5 and recovered on postnatal day 21.5. In rat ovaries, the first wave of primordial follicles is rapidly activated after their formation, whereas the second wave of primordial follicles remains dormant in the ovarian cortex and slowly starts activating after postnatal day 12.5. We localized the expression of Ihh mRNA in postnatal day 8.5 wildtype rat ovaries but not in the age-matched ErßKO ovaries. In postnatal day 21.5 ErßKO rat ovaries, we detected Ihh mRNA mainly in the activated follicles in the ovaries' peripheral regions. Our findings indicate that the expression of Ihh in the granulosa cells of the activated first wave of ovarian follicles depends on ERß.


Sujet(s)
Récepteur bêta des oestrogènes , Protéines Hedgehog , Animaux , Femelle , Rats , Récepteur bêta des oestrogènes/génétique , Récepteur bêta des oestrogènes/métabolisme , Protéines Hedgehog/génétique , Protéines Hedgehog/métabolisme , Follicule ovarique/métabolisme , Ovaire/métabolisme , ARN messager/métabolisme
20.
Int J Mol Sci ; 25(7)2024 Apr 08.
Article de Anglais | MEDLINE | ID: mdl-38612938

RÉSUMÉ

Glioblastoma multiforme (GBM) is a malignant tumor with a higher prevalence in men and a higher survival rate in transmenopausal women. It exhibits distinct areas influenced by changing environmental conditions. This study examines how these areas differ in the levels of estrogen receptors (ERs) which play an important role in the development and progression of many cancers, and whose expression levels are often correlated with patient survival. This study utilized two research models: an in vitro model employing the U87 cell line and a second model involving tumors resected from patients (including tumor core, enhancing tumor region, and peritumoral area). ER expression was assessed at both gene and protein levels, with the results validated using confocal microscopy and immunohistochemistry. Under hypoxic conditions, the U87 line displayed a decrease in ERß mRNA expression and an increase in ERα mRNA expression. In patient samples, ERß mRNA expression was lower in the tumor core compared to the enhancing tumor region (only in males when the study group was divided by sex). In addition, ERß protein expression was lower in the tumor core than in the peritumoral area (only in women when the study group was divided by sex). Immunohistochemical analysis indicated the highest ERß protein expression in the enhancing tumor area, followed by the peritumoral area, and the lowest in the tumor core. The findings suggest that ER expression may significantly influence the development of GBM, exhibiting variability under the influence of conditions present in different tumor areas.


Sujet(s)
Glioblastome , Mâle , Humains , Femelle , Glioblastome/génétique , Récepteur bêta des oestrogènes/génétique , Expression des gènes , Oestrogènes , ARN messager/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE