Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.803
Filter
1.
Nat Commun ; 15(1): 8498, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39353946

ABSTRACT

Polycystic ovary syndrome (PCOS) is a heterogeneous condition, defined by oligo-/anovulation, hyper-androgenism and/or polycystic ovaries. Metabolic complications are common in patients suffering PCOS, including obesity, insulin resistance and type-2 diabetes, which severely compromise the clinical course of affected women. Yet, therapeutic options remain mostly symptomatic and of limited efficacy for the metabolic and reproductive alterations of PCOS. We report here the hormonal, metabolic and gonadal responses to the glucagon-like peptide-1 (GLP1)-based multi-agonists, GLP1/Estrogen (GLP1/E), GLP1/gastric inhibitory peptide (GLP1/GIP) and GLP1/GIP/Glucagon, in two mouse PCOS models, with variable penetrance of metabolic and reproductive traits, and their comparison with metformin. Our data illustrate the superior efficacy of GLP1/E vs. other multi-agonists and metformin in the management of metabolic complications of PCOS; GLP1/E ameliorates also ovarian cyclicity in an ovulatory model of PCOS, without direct estrogenic uterotrophic effects. In keeping with GLP1-mediated brain targeting, quantitative proteomics reveals changes in common and distinct hypothalamic pathways in response to GLP1/E between the two PCOS models, as basis for differential efficiency. Altogether, our data set the basis for the use of GLP1-based multi-agonists, and particularly GLP1/E, in the personalized management of PCOS.


Subject(s)
Disease Models, Animal , Glucagon-Like Peptide 1 , Metformin , Polycystic Ovary Syndrome , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/metabolism , Female , Animals , Glucagon-Like Peptide 1/metabolism , Metformin/therapeutic use , Metformin/pharmacology , Mice , Humans , Gastric Inhibitory Polypeptide/metabolism , Estrogens/metabolism , Ovary/drug effects , Ovary/metabolism , Insulin Resistance , Mice, Inbred C57BL
2.
Front Endocrinol (Lausanne) ; 15: 1410433, 2024.
Article in English | MEDLINE | ID: mdl-39359415

ABSTRACT

Background: Sex steroid hormones, primarily synthesized by gonadal somatic cells, are pivotal for sexual development and reproduction. Mice studies have shown that two transcription factors, steroidogenic factor 1 (SF-1) and Wilms' tumor 1 (WT1), are involved in gonadal development. However, their role in human gonadal somatic differentiation remains unclear. We therefore aimed to investigate the roles of SF-1 and WT1 in human gonadal steroidogenic cell differentiation. Methods: Using a transient lentivirus-mediated gene expression system, we assessed the effects of SF-1 and WT1 expression on the steroidogenic potential of human amniotic membrane-derived mesenchymal stem cells (hAmMSCs). Results: SF-1 and WT1-KTS, a splice variant of WT1, played distinct roles in human steroidogenic differentiation of hAmMSCs. SF-1 induced hAmMSC differentiation into progesterone- and androgen-producing cell lineages, whereas WT1-KTS promoted hAmMSC differentiation into estrogen-producing cell lineages. Conclusion: Our findings revealed that SF-1 and WT1-KTS play important roles in human gonadal steroidogenic cell differentiation, especially during ovarian development. These findings may pave the way for future studies on human ovarian differentiation and development.


Subject(s)
Amnion , Androgens , Cell Differentiation , Cell Lineage , Estrogens , Mesenchymal Stem Cells , Progesterone , Steroidogenic Factor 1 , WT1 Proteins , Humans , WT1 Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Steroidogenic Factor 1/metabolism , Steroidogenic Factor 1/genetics , Progesterone/metabolism , Progesterone/biosynthesis , Estrogens/metabolism , Androgens/metabolism , Amnion/cytology , Amnion/metabolism , Female , Cells, Cultured , RNA Splicing Factors
3.
J Biomed Sci ; 31(1): 95, 2024 Oct 11.
Article in English | MEDLINE | ID: mdl-39390588

ABSTRACT

BACKGROUND: Primary ovarian insufficiency (POI) is an early decline in ovarian function that leads to ovarian failure. Conventional treatments for POI are inadequate, and treatments based on mesenchymal stem cells (MSCs) have emerged as an option. However, the lack of consideration of the estrogen niche in ovarian tissue significantly reduces the therapeutic efficacy, with an unclear mechanism in the MSCs in POI treatment. Furthermore, the disruption of circadian rhythm associated with POI has not been previously addressed. METHODS: Conditioned medium (CM) and estradiol-conditioned medium (E2-CM) were generated from estrogen receptor positive MSCs (ER+pcMSCs). Chemotherapy-induced POI models were established using C57BL/6 mice (in vivo) and KGN cells (in vitro) treated with cyclophosphamide (CTX) or 4-hydroperoxycyclophosphamide (4-OOH-CP). Gene/protein expressions were detected using RT-qPCR, Western blotting, and immunohistochemistry assays. Locomotor activity was monitored for behavioral circadian rhythmicity. Cytokine arrays and miRNA analysis were conducted to analyze potential factors within CM/E2-CM. RESULTS: The secretome of ER+pcMSCs (CM and E2-CM) significantly reduced the CTX-induced defects in ovarian folliculogenesis and circadian rhythm. CM/E2-CM also reduced granulosa cell apoptosis and rescued angiogenesis in POI ovarian tissues. E2-CM had a more favorable effect than the CM. Notably, ER+pcMSC secretome restored CTX-induced circadian rhythm defects, including the gene expressions associated with the ovarian circadian clock (e.g., Rora, E4bp4, Rev-erbα, Per2 and Dbp) and locomotor activity. Additionally, the cytokine array analysis revealed a significant increase in cytokines and growth factors associated with immunomodulation and angiogenesis, including angiogenin. Neutralizing the angiogenin in CM/E2-CM significantly reduced its ability to promote HUVEC tube formation in vitro. Exosomal miRNA analysis revealed the miRNAs involved in targeting the genes associated with POI rescue (PTEN and PDCD4), apoptosis (caspase-3, BIM), estrogen synthesis (CYP19A1), ovarian clock regulation (E4BP4, REV-ERBα) and fibrosis (COL1A1). CONCLUSION: This study is the first to demonstrate that, in considering the estrogen niche in ovarian tissue, an estrogen-priming ER+pcMSC secretome achieved ovarian regeneration and restored the circadian rhythm in a CTX-induced POI mouse model. The potential factors involved include angiogenin and exosomal miRNAs in the ER+pcMSC secretome. These findings offer insights into potential stem cell therapies for chemotherapy-induced POI and circadian rhythm disruption.


Subject(s)
Circadian Rhythm , Cyclophosphamide , Mesenchymal Stem Cells , Mice, Inbred C57BL , Primary Ovarian Insufficiency , Female , Animals , Cyclophosphamide/adverse effects , Mice , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/therapy , Primary Ovarian Insufficiency/metabolism , Mesenchymal Stem Cells/metabolism , Circadian Rhythm/drug effects , Humans , Pregnancy , Secretome/metabolism , Placenta/metabolism , Placenta/drug effects , Estrogens/pharmacology , Estrogens/metabolism , Ovary/metabolism , Ovary/drug effects
4.
Sci Rep ; 14(1): 23030, 2024 10 03.
Article in English | MEDLINE | ID: mdl-39362931

ABSTRACT

Urinary tract infection (UTI) is one of the most common bacterial infections worldwide and the most common cause is uropathogenic Escherichia coli (UPEC). Current research is mostly focused on how UPEC affects host factors, whereas the effect of host factors on UPEC is less studied. Our previous studies have shown that estrogen alters UPEC virulence. However, the effect of this altered UPEC virulence on neutrophils is unknown. The aim of the present study was to investigate how the altered UPEC virulence mediated by estrogen modulates neutrophil responses. We found that estradiol-stimulated CFT073 increased neutrophil phagocytosis, NETs formation and intracellular ROS production. We observed that the total ROS production from neutrophils was reduced by estradiol-stimulated CFT073. We also found that estradiol-stimulated CFT073 induced less cytotoxicity in neutrophils. Additionally, we found that several cytokines and chemokines like IL-8, IL-1ß, CXCL6, MCP-1 and MCP-4 were increased upon estradiol-stimulated CFT073 infection. In conclusion, this study demonstrates that the estrogen-mediated alterations to UPEC virulence modulates neutrophil responses, most likely in a host-beneficial manner.


Subject(s)
Estrogens , Neutrophils , Phagocytosis , Reactive Oxygen Species , Urinary Tract Infections , Uropathogenic Escherichia coli , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/drug effects , Uropathogenic Escherichia coli/immunology , Uropathogenic Escherichia coli/pathogenicity , Humans , Estrogens/pharmacology , Estrogens/metabolism , Urinary Tract Infections/microbiology , Urinary Tract Infections/immunology , Phagocytosis/drug effects , Reactive Oxygen Species/metabolism , Estradiol/pharmacology , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Cytokines/metabolism , Extracellular Traps/metabolism , Virulence
5.
Nat Commun ; 15(1): 8447, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39349474

ABSTRACT

Younger premenopausal women are more prone to developing ovarian metastases (OM) of gastric cancer (GC) than metastases of other organs; however, the molecular mechanisms remain unclear. Here we perform single-cell RNA sequencing on 45 tumor samples from 18 GC patients with OM. Interestingly, fibroblasts in OM of GC express high levels of estrogen receptor (ER) and midkine (MDK), interacting with tumor cells through activating ER-MDK-LRP1 (low-density lipoprotein receptor-related protein 1) signaling axis. Functional experiments demonstrate that estrogen stimulation induces MDK secretion by ovarian fibroblasts, and binding of MDK to LRP1 increases GC cell migration and invasion. Furthermore, in vivo, estrogen stimulation remarkably augments ovarian engraftment and metastasis of LRP1+ GC cells. Collectively, our findings reveal that ER+ ovarian fibroblasts secrete MDK under estrogen influence, driving OM of GC via the MDK-LRP1 axis. Our study holds the potential to catalyze innovative therapeutic strategies aimed at intercepting and managing OM in GC.


Subject(s)
Estrogens , Fibroblasts , Low Density Lipoprotein Receptor-Related Protein-1 , Ovarian Neoplasms , Stomach Neoplasms , Humans , Female , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/genetics , Estrogens/metabolism , Animals , Fibroblasts/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Mice , Cell Line, Tumor , Cell Movement/drug effects , Signal Transduction , Receptors, Estrogen/metabolism , Gene Expression Regulation, Neoplastic , Middle Aged
6.
Int Immunopharmacol ; 142(Pt A): 113034, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39226826

ABSTRACT

Breast cancer remains one of the primary causes of cancer-related death. An imbalance of oestrogen homeostasis and an inflammatory tumor microenvironment (TME) are vital risk factors for the progression and metastasis of breast cancer. Here, we showed that oestrogen homeostasis was disrupted both in breast cancer patients and in a transgenic MMTV-PyMT mouse model of breast cancer, and significant levels of hydroxylated oestrogen accumulated in the mammary tissues of these patients and mice. We also observed that tumor-associated macrophages (TAMs) were the main population of immune cells present in the breast TME. TAM-dependent tumor metastasis could be triggered by hydroxylated oestrogen via NLRP3 inflammasome activation and IL-1ß production. Mechanistically, TAM-derived inflammatory cytokines induced the expression of matrix metalloproteinases (MMPs) in breast tumor cells, leading to breast tumor invasion and metastasis. Conceptually, our study reveals a previously unknown role of hydroxylated oestrogen in the reprogramming of the TME via NLRP3 inflammasome activation in TAMs, which ultimately facilitates breast cancer cells proliferation, migration, and invasion.


Subject(s)
Breast Neoplasms , Estrogens , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Tumor Microenvironment , Tumor-Associated Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Humans , Female , Breast Neoplasms/pathology , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Estrogens/metabolism , Inflammasomes/metabolism , Mice , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor Microenvironment/immunology , Disease Progression , Neoplasm Metastasis , Cell Line, Tumor , Hydroxylation , Mice, Transgenic , Interleukin-1beta/metabolism
7.
Genes (Basel) ; 15(9)2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39336780

ABSTRACT

Fish are exposed to increased water temperatures and aquatic pollutants, including endocrine-disrupting compounds (EDCs). Although each stressor can disturb fish liver metabolism independently, combined effects may exist. To unveil the molecular mechanisms behind the effects of EDCs and temperature, fish liver cell lines are potential models needing better characterisation. Accordingly, we exposed the rainbow trout RTL-W1 cells (72 h), at 18 °C and 21 °C, to ethynylestradiol (EE2), levonorgestrel (LNG), and a mixture of both hormones (MIX) at 10 µM. The gene expression of a selection of targets related to detoxification (CYP1A, CYP3A27, GST, UGT, CAT, and MRP2), estrogen exposure (ERα, VtgA), lipid metabolism (FAS, FABP1, FATP1), and temperature stress (HSP70b) was analysed by RT-qPCR. GST expression was higher after LNG exposure at 21 °C than at 18 °C. LNG further enhanced the expression of CAT, while both LNG and MIX increased the expressions of CYP3A27 and MRP2. In contrast, FAS expression only increased in MIX, compared to the control. ERα, VtgA, UGT, CYP1A, HSP70b, FABP1, and FATP1 expressions were not influenced by the temperature or the tested EDCs. The RTL-W1 model was unresponsive to EE2 alone, sensitive to LNG (in detoxification pathway genes), and mainly insensitive to the temperature range but had the potential to unveil specific interactions.


Subject(s)
Ethinyl Estradiol , Levonorgestrel , Oncorhynchus mykiss , Animals , Ethinyl Estradiol/toxicity , Levonorgestrel/pharmacology , Oncorhynchus mykiss/genetics , Oncorhynchus mykiss/metabolism , Estrogens/metabolism , Cell Line , Endocrine Disruptors/toxicity , Inactivation, Metabolic/genetics , Up-Regulation/drug effects , Progestins/pharmacology , Fish Proteins/genetics , Fish Proteins/metabolism , Liver/drug effects , Liver/metabolism , Water Pollutants, Chemical/toxicity , Temperature , Lipid Metabolism/drug effects , Lipid Metabolism/genetics
8.
Int J Mol Sci ; 25(18)2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39337631

ABSTRACT

Gender and biological sex have distinct impacts on the pathogenesis of type 2 diabetes (T2D). Estrogen deficiency is known to predispose female mice to T2D. In our previous study, we found that a high-fat, high-sucrose diet (HFHSD) induces T2D in male mice through the miR-10b-5p/KLF11/KIT pathway, but not in females, highlighting hormonal disparities in T2D susceptibility. However, the underlying molecular mechanisms of this hormonal protection in females remain elusive. To address this knowledge gap, we utilized ovariectomized, estrogen-deficient female mice, fed them a HFHSD to induce T2D, and investigated the molecular mechanisms involved in estrogen-deficient diabetic female mice, relevant cell lines, and female T2D patients. Initially, female mice fed a HFHSD exhibited a delayed onset of T2D, but ovariectomy-induced estrogen deficiency promptly precipitated T2D without delay. Intriguingly, insulin (INS) was upregulated, while insulin receptor (INSR) and protein kinase B (AKT) were downregulated in these estrogen-deficient diabetic female mice, indicating insulin-resistant T2D. These dysregulations of INS, INSR, and AKT were mediated by a miR-10a/b-5p-NCOR2 axis. Treatment with miR-10a/b-5p effectively alleviated hyperglycemia in estrogen-deficient T2D female mice, while ß-estradiol temporarily reduced hyperglycemia. Consistent with the murine findings, plasma samples from female T2D patients exhibited significant reductions in miR-10a/b-5p, estrogen, and INSR, but increased insulin levels. Our findings suggest that estrogen protects against insulin-resistant T2D in females through miR-10a/b-5p/NCOR2 pathway, indicating the potential therapeutic benefits of miR-10a/b-5p restoration in female T2D management.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , MicroRNAs , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Female , Mice , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/genetics , Insulin Resistance/genetics , Humans , Insulin/metabolism , Insulin/blood , Estrogens/metabolism , Estrogens/deficiency , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/genetics , Proto-Oncogene Proteins c-akt/metabolism , Male , Ovariectomy
9.
Biomolecules ; 14(9)2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39334811

ABSTRACT

Lower urinary tract symptoms (LUTS) are common in postmenopausal women. These symptoms are often linked to decreased estrogen levels following menopause. This study investigated the relationship between estrogen levels, alterations in bladder tissue structure, bladder function, and the incidence of urinary frequency. An age-appropriate bilateral ovariectomized mouse model (OVX) was developed to simulate conditions of estrogen deficiency. Mice were divided into three groups: a sham-operated control group, OVX, and an estradiol-treated group. The assessments included estrogen level measurement, urination frequency, cystometry, histological analysis, immunofluorescence staining, and real-time quantitative PCR. Additionally, we quantified the expression of the mechanosensitive channel proteins Piezo1 and TRPV4 in mouse bladder tissues. Lower estrogen levels were linked to increased voiding episodes and structural changes in mouse bladder tissues, notably a significant increase in Collagen III fiber deposition. There was a detectable negative relationship between estrogen levels and the expression of Piezo1 and TRPV4, mechanosensitive proteins in mouse bladder tissues, which may influence voiding frequency and nocturia. Estrogen treatment could improve bladder function, decrease urination frequency, and reduce collagen deposition in the bladder tissues. This study explored the connection between estrogen levels and urinary frequency, potentially setting the stage for novel methods to address frequent urination symptoms in postmenopausal women.


Subject(s)
Disease Models, Animal , Estrogens , Ion Channels , Lower Urinary Tract Symptoms , Menopause , TRPV Cation Channels , Urinary Bladder , Animals , Female , Mice , Menopause/metabolism , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urinary Bladder/drug effects , Estrogens/metabolism , Estrogens/pharmacology , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Ion Channels/metabolism , Ion Channels/genetics , Lower Urinary Tract Symptoms/metabolism , Lower Urinary Tract Symptoms/pathology , Mice, Inbred C57BL , Urination/drug effects , Ovariectomy
10.
Sci Adv ; 10(39): eadp2442, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39331714

ABSTRACT

Estrogens regulate eosinophilia in asthma and other inflammatory diseases. Further, peripheral eosinophilia and tumor-associated tissue eosinophilia (TATE) predicts a better response to immune checkpoint blockade (ICB) in breast cancer. However, how and if estrogens affect eosinophil biology in tumors and how this influences ICB efficacy has not been determined. Here, we report that estrogens decrease the number of peripheral eosinophils and TATE, and this contributes to increased tumor growth in validated murine models of breast cancer and melanoma. Moreover, estrogen signaling in healthy female mice also suppressed peripheral eosinophil prevalence by decreasing the proliferation and survival of maturing eosinophils. Inhibiting estrogen receptor (ER) signaling decreased tumor growth in an eosinophil-dependent manner. Further, the efficacy of ICBs was increased when administered in combination with anti-estrogens. These findings highlight the importance of ER signaling as a regulator of eosinophil biology and TATE and highlight the potential near-term clinical application of ER modulators to increase ICB efficacy in multiple tumor types.


Subject(s)
Breast Neoplasms , Eosinophilia , Eosinophils , Estrogens , Receptors, Estrogen , Signal Transduction , Animals , Female , Estrogens/metabolism , Estrogens/pharmacology , Mice , Signal Transduction/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Eosinophils/metabolism , Eosinophilia/metabolism , Eosinophilia/pathology , Humans , Receptors, Estrogen/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Immune Checkpoint Inhibitors/pharmacology , Disease Models, Animal
11.
Int J Mol Sci ; 25(18)2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39337355

ABSTRACT

The aim of this systematic review is to evaluate the impact of estrogen levels on the occurrence of temporomandibular disorders (TMDs) in humans. Searches were conducted in the same databases as follows: PubMed, the Cochrane Collaboration database, and the Scopus database. In accordance with the MeSH database and previous work, the following keywords were used: 'estrogens' and 'temporomandibular joint disorders'. Twelve studies were included in the review and were assessed for the quality of evidence. Estrogen levels are associated with pain modulation in the temporomandibular joint and the entire orofacial region. There is insufficient evidence to either confirm or refute the influence of estrogen on the occurrence of TMDs. The study was registered under the identifier: 10.17605/OSF.IO/BC7QF.


Subject(s)
Estrogens , Temporomandibular Joint Disorders , Humans , Temporomandibular Joint Disorders/metabolism , Temporomandibular Joint Disorders/blood , Temporomandibular Joint Disorders/etiology , Estrogens/metabolism , Estrogens/blood , Temporomandibular Joint/metabolism , Temporomandibular Joint/pathology , Female
12.
Int J Med Sci ; 21(11): 2149-2157, 2024.
Article in English | MEDLINE | ID: mdl-39239545

ABSTRACT

Estradiol (E2) deficiency arising from menopause is closely related to changes in body composition and declines of muscle mass and strength in elderly women. Whole-body vibration training (WBV) is an emerging approach expected to improve muscle mass and strength of older person, but the underlying mechanisms remain unclear. The balance between protein synthesis and degradation is a determining factor for muscle mass and strength, which is regulated by Akt-mTOR and FoxO1 signal pathway, respectively. In the present study, we firstly determined whether the effects of WBV on muscle mass and strength in ovariectomized female mice was affected by estrogen level, then investigated whether this was associated with Akt-mTOR and FoxO1 signal pathways. We found that (1) WBV, E2 supplementation (E) and WBV combined with E2 supplementation (WBV+E) significantly increased serum estradiol content, quadriceps muscle mass and grip strength in ovariectomized mice, accompanied with alterations of body composition (reducing fat content, increasing lean body mass and lean percent), furthermore, the altered degrees of these indicators by WBV+E were greater than WBV alone; (2) WBV, E and WBV+E remarkably increased the activities of Akt and mTOR and decreased FoxO1 activity, and the changed degrees by WBV+E were greater than WBV alone; (3) Pearson correlation coefficient revealed that serum estradiol content was positively correlated with Akt and mTOR activities, while inversely associated with FoxO1 activity. We concluded that WBV could significantly increase muscle mass and strength in ovariectomized mice, which might achieve through activating Akt-mTOR and suppressing FoxO1 signal pathways, and the improving effect of WBV on muscle mass and strength was better when in the presence of estrogen.


Subject(s)
Estradiol , Estrogens , Forkhead Box Protein O1 , Muscle Strength , Ovariectomy , TOR Serine-Threonine Kinases , Vibration , Animals , Female , Vibration/therapeutic use , Mice , Muscle Strength/physiology , TOR Serine-Threonine Kinases/metabolism , Estradiol/blood , Forkhead Box Protein O1/metabolism , Estrogens/blood , Estrogens/metabolism , Signal Transduction , Body Composition/physiology , Humans , Proto-Oncogene Proteins c-akt/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Physical Conditioning, Animal/physiology , Physical Conditioning, Animal/methods
13.
Horm Behav ; 165: 105632, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39244874

ABSTRACT

Female fertility signals are found across taxa, and the precision of such signals may be influenced by the relative strength of different sexual selection mechanisms. Among primates, more precise signals may be found in species with stronger direct male-male competition and indirect female mate choice, and less precise signals in species with stronger indirect male-male competition (e.g. sperm competition) and direct female mate choice. We tested this hypothesis in a wild population of Kinda baboons in Zambia, combining data on female signals with reproductive hormones (estrogen and progesterone metabolites) and intra- and inter-cycle fertility. We predicted that Kinda baboons will exhibit less precise fertility signals than other baboon species, as they experience weaker direct and stronger indirect male-male competition. The frequency of copulation calls and proceptive behavior did not vary with hormones or intra- or inter-cycle fertility in almost all models. Sexual swelling size was predicted by the ratio of estrogen to progesterone metabolites, and was largest in the fertile phase, but differences in size across days were small. Additionally, there was variability in the timing of ovulation relative to the day of sexual swelling detumescence across cycles and swelling size did not vary with inter-cycle fertility. Our results suggest that female Kinda baboon sexual swellings are less precise indicators of fertility compared to other baboon species, while signals in other modalities do not reflect variation in intra- and inter-cycle fertility. Female Kinda baboon sexual signals may have evolved as a strategy to reduce male monopolizability, allowing for more female control over reproduction by direct mate choice.


Subject(s)
Fertility , Progesterone , Sexual Behavior, Animal , Animals , Female , Fertility/physiology , Male , Sexual Behavior, Animal/physiology , Progesterone/metabolism , Progesterone/blood , Estrogens/metabolism , Copulation/physiology , Ovulation/physiology , Animal Communication
14.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39125736

ABSTRACT

Oestrogen plays a crucial physiological role in both women and men. It regulates reproductive functions and maintains various non-reproductive tissues through its receptors, such as oestrogen receptor 1/oestrogen receptor α (ESR1/Erα), oestrogen receptor 2/oestrogen receptor ß (ESR2/Erß), and G protein-coupled oestrogen receptor 1 (GPER). This hormone is essential for the proper functioning of women's ovaries and uterus. Oestrogen supports testicular function and spermatogenesis in men and contributes to bone density, cardiovascular health, and metabolic processes in both sexes. Nuclear receptors Er-α and Er-ß belong to the group of transcription activators that stimulate cell proliferation. In the environment, compounds similar in structure to the oestrogens compete with endogenous hormones for binding sites to receptors and to disrupt homeostasis. The lack of balance in oestrogen levels can lead to infertility, cancer, immunological disorders, and other conditions. Exogenous endocrine-active compounds, such as bisphenol A (BPA), phthalates, and organic phosphoric acid esters, can disrupt signalling pathways responsible for cell division and apoptosis processes. The metabolism of oestrogen and its structurally similar compounds can produce carcinogenic substances. It can also stimulate the growth of cancer cells by regulating genes crucial for cell proliferation and cell cycle progression, with long-term elevated levels linked to hormone-dependent cancers such as breast cancer. Oestrogens can also affect markers of immunological activation and contribute to the development of autoimmune diseases. Hormone replacement therapy, oral contraception, in vitro fertilisation stimulation, and hormonal stimulation of transgender people can increase the risk of breast cancer. Cortisol, similar in structure to oestrogen, can serve as a biomarker associated with the risk of developing breast cancer. The aim of this review is to analyse the sources of oestrogens and their effects on the endogenous and exogenous process of homeostasis.


Subject(s)
Estrogens , Humans , Estrogens/metabolism , Animals , Receptors, Estrogen/metabolism , Female , Male
15.
J Hazard Mater ; 479: 135637, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-39208633

ABSTRACT

While laccase humification has an efficient capacity to convert estrogenic pollutants, the roles of wheat (Triticum aestivum L.) root exudates (W-REs) in the enzymatic humification remain poorly understood. Herein, we presented the research into the effects of W-REs on 17ß-estradiol (E2) and bisphenol A (BPA) conversion in vitro laccase humification. W-REs inhibited E2 removal but promoted BPA conversion in the enzymatic humification, and the first-order kinetic constants for E2 and BPA were 0.27-0.69 and 0.28-0.55 h-1, respectively. Specialized small phenols and amino acids in W-REs were susceptible to laccase humification, resulting in increased copolymerization of estrogen and W-REs. In greenhouse hydroponics, the accumulated amounts of E2 (BPA) in the roots and shoots were estimated to be 0.87 (2.15) and 0.43 (0.51) nmol·plant-1 at day 3, respectively. By forming low- and eventually non-toxic copolymeric precipitates between estrogen and W-REs, laccase humification lowered the phytotoxicity and bioavailability of estrogen in the rhizosphere solution, consequently relieving its uptake, accumulation, and distribution in the wheat cells. This work sheds light on the roles of W-REs in regulating laccase-catalyzed estrogen humification, and gives an insight into the path of addressing organic contamination in the rhizosphere and ensuring food safety.


Subject(s)
Benzhydryl Compounds , Estradiol , Humic Substances , Laccase , Plant Roots , Triticum , Triticum/metabolism , Laccase/metabolism , Estradiol/metabolism , Estradiol/chemistry , Plant Roots/metabolism , Benzhydryl Compounds/metabolism , Benzhydryl Compounds/chemistry , Phenols/metabolism , Phenols/chemistry , Estrogens/metabolism , Estrogens/chemistry , Soil Pollutants/metabolism , Plant Exudates/metabolism , Plant Exudates/chemistry
16.
J Steroid Biochem Mol Biol ; 244: 106607, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39197539

ABSTRACT

Long-term administration of exogenous estrogen is known to cause urinary retention and marked, often fatal, bladder distention in both male and female mice. Estrogen-treated mice have increased bladder pressure and decreased urine flow, suggesting that urinary retention in estrogen-treated mice is due to infravesicular obstruction to urine outflow. Thus, the condition is commonly referred to as bladder outlet obstruction (BOO). Obesity can also lead to urinary retention. As the effects of estrogen are mediated by multiple receptors, including estrogen receptors ERα and ERß and the G protein-coupled estrogen receptor (GPER), we sought to determine whether GPER plays a role in estrogen-induced BOO, particularly in the context of obesity. Wild type and GPER knockout (KO) mice fed a high-fat diet were ovariectomized or left ovary-intact (sham surgery) and supplemented with slow-release estrogen or vehicle-only pellets. Supplementing both GPER KO and wild type obese mice with estrogen for 8 weeks resulted in weight loss, splenic enlargement, and thymic atrophy, as expected. However, estrogen-treated obese GPER KO mice developed abdominal distension, debilitation, and ulceration of the skin surrounding the urogenital opening. At necropsy, these mice had prominently distended bladders and hydronephrosis. In contrast, estrogen-treated obese wild type mice only rarely displayed these signs. Our results suggest that, under conditions of obesity, estrogen induces BOO as a result of ERα-driven pathways and that GPER expression is protective against BOO.


Subject(s)
Estrogens , Mice, Knockout , Obesity , Receptors, Estrogen , Receptors, G-Protein-Coupled , Urinary Retention , Animals , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Estrogen/metabolism , Receptors, Estrogen/genetics , Estrogens/metabolism , Female , Obesity/metabolism , Obesity/complications , Obesity/genetics , Mice , Urinary Retention/metabolism , Urinary Retention/genetics , Mice, Inbred C57BL , Mice, Obese , Diet, High-Fat/adverse effects , Ovariectomy , Male , Urinary Bladder Neck Obstruction/metabolism , Urinary Bladder Neck Obstruction/pathology , Urinary Bladder Neck Obstruction/genetics
17.
Int Immunopharmacol ; 141: 112906, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39173403

ABSTRACT

Hyperactive osteoclasts and hypoactive osteoblasts usually result in osteolytic conditions such as estrogen-deficiency bone loss. Few natural compounds that both attenuating bone resorption and enhancing bone formation could exert effects on this imbalance. 5-Deoxycajanin (5-D), an isoflavonoid extracted from Cajan leaf with estrogen-like properties, were found to have beneficial pharmacological effects on rebalancing the activities of osteoclasts and osteoblasts. This study revealed that 5-D at the same concentration could inhibit osteoclastogenesis of BMMs and promoted osteoblast differentiation of BMSCs. 5-D not only attenuated the fluorescent formation of RANKL-induced F-actin belts and NFATc1, but also activated ALP and RUNX2 expressions. As to downstream factor expressions, 5-D could block osteoclast-specific genes and proteins including NFATc1 and CTSK, while increased osteogenic genes and proteins including OPG and OCN, as confirmed by Real-time PCR and Western Blotting. Additionally, the network pharmacology and molecular docking identified the involvement of 5-D in the MIF and MAPK signaling pathways and the stable binding between 5-D and MAPK2K1. Further Western blot studies showed that 5-D decreased the phosphorylation of p38 and ERK in osteoclasts, but promoted these phosphorylations in osteoblasts. In a female C57BL/6J mouse model of estrogen deficiency-induced bone loss, 5-D demonstrated efficacy in enhancing BMD through attenuating osteoclast activities and promoting osteogenesis. These results underscore the potential application of 5-D on treating osteolysis resulting from hyperactive osteoclasts and hypoactive osteoblasts, shedding light on modulating osteoclast-osteoblast homeostasis.


Subject(s)
Bone Resorption , Estrogens , Osteoblasts , Osteoclasts , Animals , Osteoclasts/drug effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Mice , Female , Estrogens/deficiency , Estrogens/metabolism , Bone Resorption/drug therapy , Osteogenesis/drug effects , Homeostasis/drug effects , Mice, Inbred C57BL , Cells, Cultured , Cell Differentiation/drug effects , Ovariectomy , Isoflavones/pharmacology , Isoflavones/therapeutic use , RANK Ligand/metabolism , Humans , Molecular Docking Simulation , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism
19.
Int J Mol Sci ; 25(16)2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39201290

ABSTRACT

Breast cancer is the most diagnosed cancer in the world, and it is the primary cause of cancer death for women. The risk of breast cancer is increased by endogenous factors like hormones and exogenous factors like radiation exposure that causes damage to the mammary epithelial cells leading to an inflammatory response. Chronic inflammation creates a microenvironment composed of, among other factors, chemokines, and interleukins, which promote cancer. The gene expression of the interleukin 1 receptor type 1, the interleukin 1 receptor antagonist, the Interleukin 1 Receptor Accessory Protein, the interleukin 6 cytokine family signal transducer, the C-X-C motif chemokine ligand 3, the C-X-C motif chemokine ligand 5, and the C-X-C motif chemokine ligand 6 were analyzed in an estrogen and radiation experimental breast cancer model. Furthermore, the expression of these genes was correlated with immune cell infiltration, estrogen receptor expression, and their clinical relevance in breast cancer patients based on data provided by The Cancer Genome Atlas database online. Results given by the experimental breast cancer model showed that all genes related to inflammation respond to ionizing radiation alone or in combination with estrogen. On the other hand, the immune response depended on the breast cancer type and on the expression of the gene that encoded the estrogen receptor. Finally, the importance of the expression of these genes in breast cancer is such that high IL1R1 or IL1RAP is strongly related to patient survival. These findings may help to improve the understanding of the role of immune molecules in carcinogenesis and enhance therapeutic approaches.


Subject(s)
Breast Neoplasms , Estrogen Receptor alpha , Estrogens , Inflammation , Radiation, Ionizing , Female , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Estrogens/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Inflammation/metabolism , Humans , Mice , Gene Expression Regulation, Neoplastic , Tumor Microenvironment
20.
Int J Mol Sci ; 25(16)2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39201762

ABSTRACT

Alzheimer's disease (AD) is a long-term neurodegenerative condition that leads to the deterioration of neurons and synapses in the cerebral cortex, resulting in severe dementia. AD is significantly more prevalent in postmenopausal women, suggesting a neuroprotective role for estrogen. Estrogen is now known to regulate a wide array of physiological functions in the body by interacting with three known estrogen receptors (ERs) and with the ß-amyloid precursor protein, a key factor in AD pathogenesis. Recent experimental evidence indicates that new selective ER modulators and phytoestrogens may be promising treatments for AD for their neuroprotective and anti-apoptotic properties. These alternatives may offer fewer side effects compared to traditional hormone therapies, which are associated with risks such as cardiovascular diseases, cancer, and metabolic dysfunctions. This review sheds light on estrogen-based treatments that may help to partially prevent or control the neurodegenerative processes characteristic of AD, paving the way for further investigation in the development of estrogen-based treatments.


Subject(s)
Alzheimer Disease , Receptors, Estrogen , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Humans , Receptors, Estrogen/metabolism , Animals , Estrogens/metabolism , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Phytoestrogens/therapeutic use , Phytoestrogens/pharmacology , Phytoestrogens/metabolism , Amyloid beta-Protein Precursor/metabolism , Selective Estrogen Receptor Modulators/therapeutic use , Selective Estrogen Receptor Modulators/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL