Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 13.485
Filter
1.
Pharmacol Res Perspect ; 12(4): e1234, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38961539

ABSTRACT

The association of hormonal contraception with increased risk of inflammatory bowel disease (IBD) observed in females suggests involvement of ovarian hormones, such as estradiol, and the estrogen receptors in the progression of intestinal inflammation. Here, we investigated the effects of prophylactic SERM2 and estradiol supplementation in dextran sulfate sodium-induced colitis using mice with intact ovaries and ovariectomized (OVX) female mice. We found that graded colitis score was threefold reduced in the OVX mice, compared to mice with intact ovaries. Estradiol supplementation, however, aggravated the colitis in OVX mice, increasing the colitis score to a similar level than what was observed in the intact mice. Further, we observed that immune infiltration and gene expression of inflammatory interleukins Il1b, Il6, and Il17a were up to 200-fold increased in estradiol supplemented OVX colitis mice, while a mild but consistent decrease was observed by SERM2 treatment in intact animals. Additionally, cyclo-oxygenase 2 induction was increased in the colon of colitis mice, in correlation with increased serum estradiol levels. Measured antagonist properties of SERM2, together with the other results presented here, indicates an exaggerating role of ERα signaling in colitis. Our results contribute to the knowledge of ovarian hormone effects in colitis and encourage further research on the potential use of ER antagonists in the colon, in order to alleviate inflammation.


Subject(s)
Colitis , Dextran Sulfate , Estradiol , Estrogen Receptor alpha , Ovariectomy , Animals , Female , Estrogen Receptor alpha/metabolism , Colitis/chemically induced , Colitis/metabolism , Colitis/drug therapy , Mice , Estradiol/pharmacology , Estradiol/blood , Mice, Inbred C57BL , Estrogens/pharmacology , Cyclooxygenase 2/metabolism , Disease Models, Animal , Interleukin-17/metabolism , Colon/pathology , Colon/drug effects , Colon/metabolism , Interleukin-6/metabolism , Interleukin-1beta/metabolism
3.
Endocrinology ; 165(8)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38980913

ABSTRACT

The resurgence of interest in psychedelics as treatments for psychiatric disorders necessitates a better understanding of potential sex differences in response to these substances. Sex as a biological variable (SABV) has been historically neglected in medical research, posing limits to our understanding of treatment efficacy. Human studies have provided insights into the efficacy of psychedelics across various diagnoses and aspects of cognition, yet sex-specific effects remain unclear, making it difficult to draw strong conclusions about sex-dependent differences in response to psychedelic treatments. Compounding this further, animal studies used to understand biological mechanisms of psychedelics predominantly use one sex and present mixed neurobiological and behavioral outcomes. Studies that do include both sexes often do not investigate sex differences further, which may hinder the translation of findings to the clinic. In reviewing sex differences in responses to psychedelics, we will highlight the direct interaction between estrogen (the most extensively studied steroid hormone) and the serotonin system (central to the mechanism of action of psychedelics), and the potential that estrogen-serotonin interactions may influence the efficacy of psychedelics in female participants. Estrogen influences serotonin neurotransmission by affecting its synthesis and release, as well as modulating the sensitivity and responsiveness of serotonin receptor subtypes in the brain. This could potentially influence the efficacy of psychedelics in females by modifying their therapeutic efficacy across menstrual cycles and developmental stages. Investigating this interaction in the context of psychedelic research could aid in the advancement of therapeutic outcomes, especially for conditions with sex-specific prevalence.


Subject(s)
Hallucinogens , Serotonin , Sex Characteristics , Hallucinogens/pharmacology , Humans , Female , Animals , Male , Serotonin/metabolism , Estrogens/pharmacology
4.
Int J Mol Sci ; 25(13)2024 Jun 30.
Article in English | MEDLINE | ID: mdl-39000355

ABSTRACT

Postmenopausal osteoporosis, characterized by an imbalance between osteoclast-mediated bone resorption and osteoblast-driven bone formation, presents substantial health implications. In this study, we investigated the role of black goat extract (BGE), derived from a domesticated native Korean goat, estrogen-like activity, and osteoprotective effects in vitro. BGE's mineral and fatty acid compositions were analyzed via the ICP-AES method and gas chromatography-mass spectrometry, respectively. In vitro experiments were conducted using MCF-7 breast cancer cells, MC3T3-E1 osteoblasts, and RAW264.7 osteoclasts. BGE exhibits a favorable amount of mineral and fatty acid content. It displayed antimenopausal activity by stimulating MCF-7 cell proliferation and augmenting estrogen-related gene expression (ERα, ERß, and pS2). Moreover, BGE positively impacted osteogenesis and mineralization in MC3T3-E1 cells through Wnt/ß-catenin pathway modulation, leading to heightened expression of Runt-related transcription factor 2, osteoprotegerin, and collagen type 1. Significantly, BGE effectively suppressed osteoclastogenesis by curtailing osteoclast formation and activity in RAW264.7 cells, concurrently downregulating pivotal signaling molecules, including receptor activator of nuclear factor κ B and tumor necrosis factor receptor-associated factor 6. This study offers a shred of preliminary evidence for the prospective use of BGE as an effective postmenopausal osteoporosis treatment.


Subject(s)
Cell Differentiation , Goats , Osteoblasts , Osteoclasts , Osteogenesis , Animals , Mice , RAW 264.7 Cells , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Cell Differentiation/drug effects , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/cytology , Humans , Estrogens/pharmacology , Cell Proliferation/drug effects , Wnt Signaling Pathway/drug effects , MCF-7 Cells , Tissue Extracts/pharmacology
5.
Anticancer Res ; 44(8): 3355-3364, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39060083

ABSTRACT

BACKGROUND/AIM: As an antagonist of bone morphogenetic protein (BMP), Noggin facilitates osteolytic bone metastases from breast cancer. The present study aimed to further dissect its role in oestrogen receptor (ER) positive breast cancer. MATERIALS AND METHODS: Noggin expression in ER positive breast cancer cell lines (MCF-7 and T-47D) was determined under conditions of oestrogen deprivation and treatment with 17-ß-oestradiol (E2). Activation of Smad1/5/8 in the oestrogen-regulated Noggin was examined using recombinant human BMP7 (rhBMP7) and a BMP receptor inhibitor (LDN-193189). The influence of Noggin on cellular functions was evaluated in MCF-7 and T-47D cell lines. Responses to tamoxifen and chemotherapy drugs were determined in MCF-7 and T-47D cells with Noggin over-expression using MTT assay. RESULTS: Noggin expression was negatively correlated with ERα in breast cancers. Noggin was up-regulated upon oestrogen deprivation, an effect that was eliminated by E2 Furthermore, increased levels of phosphorylated Smad1/5/8 were observed in the oestrogen-deprived MCF-7 and T-47D cells, which was prevented by E2 and LDN-193189, respectively. BMP7-induced Noggin expression and activation of Smad1/5/8 was also prevented by E2 and LDN-193189. Noggin over-expression resulted in an increase in the proliferation of both MCF-7 and T-47D cells. MCF-7 and T-47D cells over-expressing Noggin exhibited a good tolerance to tamoxifen (TAM), DTX, and 5-FU, but the percentage of viable cells was higher compared with the controls. CONCLUSION: Noggin expression can be repressed by oestrogen through inference with the BMP/Smad signalling. Over-expression of Noggin promotes the proliferation of MCF-7 and T-47D cells, contributing to drug resistance.


Subject(s)
Breast Neoplasms , Carrier Proteins , Estrogens , Signal Transduction , Smad Proteins , Tamoxifen , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Female , Signal Transduction/drug effects , Smad Proteins/metabolism , Estrogens/pharmacology , Estrogens/metabolism , MCF-7 Cells , Tamoxifen/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Protein 7/genetics , Gene Expression Regulation, Neoplastic/drug effects , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Estradiol/pharmacology
6.
Clin Epigenetics ; 16(1): 81, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890707

ABSTRACT

BACKGROUND: Papillary thyroid carcinoma (PTC) is a common endocrine malignancy. Studies have indicated that estrogen can regulate the expression of miRNAs in numerous malignancies. MiR-570-3p has been shown to have a regulatory function in various cancers. However, studies of the regulatory function of miR-570-3p and a direct link between estrogen (especially estradiol E2) and miR-570-3p in PTC have not been done. METHODS: Expression of miR-570-3p and its downstream target DPP4 in PTC tissues and cells was predicted using bioinformatics and validated by qRT-PCR and western blot assays. We then performed a series of gain-and-loss experiments to assess the functional significance of miR-570-3p/DPP4 axis in PTC progression in vitro and in vivo. Additionally, the methylation of the miR-570-3p promoter region was examined via bioinformatics analysis and MSP. Finally, the effects of E2 on PTC progression and the correlation between DNMT1/DNMT3A and EZH2 were predicted by bioinformatic tools and proved by luciferase reporter, ChIP, and co-IP assays. RESULTS: In PTC tumor tissues and cell lines, there was a lower expression level and a higher methylation level of miR-570-3p compared to normal tissues and cell lines. DPP4 was identified as the downstream target of miR-570-3p. Overexpression of miR-570-3p reduced the proliferative, migratory, and invasive capabilities, and promoted apoptosis, while overexpression of DPP4 reversed these effects in PTC cells. It was also discovered that DNMT1 and DNMT3A increased the CpG methylation level of the miR-570-3p promoter in an EZH2-dependent manner, which led to decreased expression of miR-570-3p. Furthermore, we observed that estrogen (E2) enhanced the methylation of miR-570-3p and suppressed its expression levels, resulting in augmented tumor growth in vivo in PTC. CONCLUSION: Estrogen regulates the EZH2/DNMTs/miR-570-3p/DPP4 signaling pathway to promote PTC progression.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methyltransferase 3A , Dipeptidyl Peptidase 4 , Enhancer of Zeste Homolog 2 Protein , Estrogens , Gene Expression Regulation, Neoplastic , MicroRNAs , Thyroid Cancer, Papillary , Thyroid Neoplasms , Humans , MicroRNAs/genetics , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/pathology , Dipeptidyl Peptidase 4/genetics , DNA Methyltransferase 3A/genetics , Cell Line, Tumor , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Estrogens/pharmacology , Estrogens/genetics , Gene Expression Regulation, Neoplastic/drug effects , Female , Mice , DNA Methylation/genetics , Animals , DNA (Cytosine-5-)-Methyltransferases/genetics , Cell Proliferation/genetics , Cell Proliferation/drug effects , Male , Promoter Regions, Genetic/genetics
7.
FASEB J ; 38(11): e23718, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38847487

ABSTRACT

Female carriers of a Duchenne muscular dystrophy (DMD) gene mutation manifest exercise intolerance and metabolic anomalies that may be exacerbated following menopause due to the loss of estrogen, a known regulator of skeletal muscle function and metabolism. Here, we studied the impact of estrogen depletion (via ovariectomy) on exercise tolerance and muscle mitochondrial metabolism in female mdx mice and the potential of estrogen replacement therapy (using estradiol) to protect against functional and metabolic perturbations. We also investigated the effect of estrogen depletion, and replacement, on the skeletal muscle proteome through an untargeted proteomic approach with TMT-labelling. Our study confirms that loss of estrogen in female mdx mice reduces exercise capacity, tricarboxylic acid cycle intermediates, and citrate synthase activity but that these deficits are offset through estrogen replacement therapy. Furthermore, ovariectomy downregulated protein expression of RNA-binding motif factor 20 (Rbm20), a critical regulator of sarcomeric and muscle homeostasis gene splicing, which impacted pathways involving ribosomal and mitochondrial translation. Estrogen replacement modulated Rbm20 protein expression and promoted metabolic processes and the upregulation of proteins involved in mitochondrial dynamics and metabolism. Our data suggest that estrogen mitigates dystrophinopathic features in female mdx mice and that estrogen replacement may be a potential therapy for post-menopausal DMD carriers.


Subject(s)
Estrogens , Mice, Inbred mdx , Muscle, Skeletal , RNA-Binding Proteins , Animals , Female , Mice , Estrogens/metabolism , Estrogens/pharmacology , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/genetics , Mice, Inbred C57BL , Ovariectomy , Mitochondria/metabolism , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/drug effects
8.
BMC Complement Med Ther ; 24(1): 227, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862934

ABSTRACT

OBJECTIVE: Endometrial cancer (EC) is an oestrogen-dependent tumour, the occurrence of which is closely related to an imbalance of oestrogen homeostasis. Our previous studies explored the effects of Resveratrol(Res) on oestrogen metabolism. However, systematic research on the exact mechanism of action of Res is still lacking. Based on network pharmacology, molecular docking and animal experiments, the effects and molecular mechanisms of Res on endometrial cancer were investigated. METHODS: The target of Res was obtained from the high-throughput experiment and reference-guided database of TCM (HERB) and the Encyclopedia of Traditional Chinese Medicine (ETCM) databases, and the target of endometrial cancer was obtained by using the Genecards database. Venny map was used to obtain the intersection target of Res in the treatment of endometrial cancer, and the protein interaction network of the intersection target was constructed by importing the data into the STRING database. Then, the drug-disease-target interaction network was constructed based on Cytoscape 3.9.1 software. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed for intersection targets using the OmicShare cloud platform. Res and core targets were analysed by molecular docking. EC model mice induced by MNNG were randomly divided into the control group, Res group, MNNG group, MNNG + Res group, and MNNG + Res + MAPK/ERKi group. The protein levels of ERK and p-ERK in the mouse uterus were detected by Western blot. The levels of E1, E2, E3, 16-epiE3, 17-epiE3, 2-MeOE1, 4-MeOE1, 2-MeOE2, 4-MeOE2, 3-MeOE1, 2-OHE1, 4-OHE1, 2-OHE2, 4-OHE2, and 16α-OHE1 in the serum and endometrial tissue of mice were measured by LC‒MS/MS. RESULTS: A total of 174 intersection targets of Res anti-endometrial cancer were obtained. The signalling pathways analysed by KEGG enrichment included the AGE-RAGE signalling pathway in diabetic complications, the PI3K-Akt signalling pathway and the MAPK signalling pathway. The top 10 core targets were MAPK3, JUN, TP53, CASP3, TNF, IL1B, AKT1, FOS, VEGFA and INS. Molecular docking showed that in addition to TNF, other targets had good affinity for Res, and the binding activity with MAPK3 was stable. Western blot results showed that Res increased the phosphorylation level of ERK and that MAPK/ERKi decreased ERK activation. In the LC-MS/MS analysis, the levels of 2-MeOE1, 2-MeOE2 and 4-MeOE1 in serum and uterine tissue showed a significantly decreasing trend in the MNNG group, while that of 4-OHE2 was increased (P < 0.05). The concentrations of 4-MeOE1 in serum and 2-MeOE1 and 2-MeOE2 in the endometrial tissue of mice were significantly increased after Res treatment, and those of 4-OHE2 in the serum and uterus of mice were significantly decreased (P < 0.05). Meanwhile, in the MAPK/ERKi intervention group, the effect of Res on the reversal of oestrogen homeostasis imbalance was obviously weakened. CONCLUSION: Res has multiple targets and multiple approaches in the treatment of endometrial cancer. In this study, it was found that Res regulates oestrogen metabolism by activating the MAPK/ERK pathway. This finding provides a new perspective for subsequent research on the treatment of endometrial cancer.


Subject(s)
Endometrial Neoplasms , Estrogens , MAP Kinase Signaling System , Molecular Docking Simulation , Resveratrol , Female , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/metabolism , Animals , Resveratrol/pharmacology , Mice , MAP Kinase Signaling System/drug effects , Estrogens/metabolism , Estrogens/pharmacology , Humans , Mice, Inbred BALB C , Network Pharmacology , Protein Interaction Maps
9.
Int J Mol Sci ; 25(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38892322

ABSTRACT

Estrogen (17ß-estradiol) deficiency post-menopause alters bone homeostasis whereby bone resorption by osteoclasts exceeds bone formation by osteoblasts, leading to osteoporosis in females. We established an in vitro model to examine the consequences of estrogen withdrawal (E2-WD) on osteoclasts derived from the mouse macrophage RAW 264.7 cell line and utilized it to investigate the mechanism behind the enhanced osteoclast activity post-menopause. We found that a greater population of osteoclasts that underwent E2-WD contained a podosome belt necessary for osteoclasts to adhere and resorb bone and possessed elevated resorptive activity compared to osteoclasts exposed to estrogen (E2) continuously. Our results show that compared to osteoclasts that received E2 continuously, those that underwent E2-WD had a faster rate of microtubule (MT) growth, reduced RhoA activation, and shorter podosome lifespan. Thus, altered podosome and MT dynamics induced by the withdrawal of estrogen supports podosome belt assembly/stability in osteoclasts, which may explain their enhanced bone resorption activity.


Subject(s)
Bone Resorption , Estrogens , Osteoclasts , Animals , Osteoclasts/metabolism , Osteoclasts/cytology , Mice , RAW 264.7 Cells , Estrogens/metabolism , Estrogens/pharmacology , Bone Resorption/metabolism , Podosomes/metabolism , Microtubules/metabolism , Female , rhoA GTP-Binding Protein/metabolism , Estradiol/pharmacology , Estradiol/metabolism , Cell Culture Techniques
10.
Aging (Albany NY) ; 16(11): 9876-9898, 2024 06 05.
Article in English | MEDLINE | ID: mdl-38843385

ABSTRACT

Estrogen is thought to have a role in slowing down aging and protecting cardiovascular and cognitive function. However, high doses of estrogen are still positively associated with autoimmune diseases and tumors with systemic inflammation. First, we administered exogenous estrogen to female mice for three consecutive months and found that the aorta of mice on estrogen develops inflammatory manifestations similar to Takayasu arteritis (TAK). Then, in vitro estrogen intervention was performed on mouse aortic vascular smooth muscle cells (MOVAS cells). Stimulated by high concentrations of estradiol, MOVAS cells showed decreased expression of contractile phenotypic markers and increased expression of macrophage-like phenotypic markers. This shift was blocked by tamoxifen and Krüppel-like factor 4 (KLF4) inhibitors and enhanced by Von Hippel-Lindau (VHL)/hypoxia-inducible factor-1α (HIF-1α) interaction inhibitors. It suggests that estrogen-targeted regulation of the VHL/HIF-1α/KLF4 axis induces phenotypic transformation of vascular smooth muscle cells (VSMC). In addition, estrogen-regulated phenotypic conversion of VSMC to macrophages is a key mechanism of estrogen-induced vascular inflammation, which justifies the risk of clinical use of estrogen replacement therapy.


Subject(s)
Estrogens , Hypoxia-Inducible Factor 1, alpha Subunit , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Macrophages , Muscle, Smooth, Vascular , Von Hippel-Lindau Tumor Suppressor Protein , Animals , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Macrophages/metabolism , Macrophages/drug effects , Mice , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/drug effects , Female , Estrogens/pharmacology , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Cell Transdifferentiation/drug effects , Phenotype , Aorta/pathology , Aorta/drug effects , Inflammation/metabolism
11.
Sci Rep ; 14(1): 14454, 2024 06 24.
Article in English | MEDLINE | ID: mdl-38914633

ABSTRACT

Hydrogen peroxide is considered deleterious molecule that cause cellular damage integrity and function. Its key redox signaling molecule in oxidative stress and exerts toxicity on a wide range of organisms. Thus, to understand whether oxidative stress alters visual development, zebrafish embryos were exposed to H2O2 at concentration of 0.02 to 62.5 mM for 7 days. Eye to body length ratio (EBR) and apoptosis in retina at 48 hpf, and optomotor response (OMR) at 7 dpf were all measured. To investigate whether hydrogen peroxide-induced effects were mediated by oxidative stress, embryos were co-incubated with the antioxidant, glutathione (GSH) at 50 µM. Results revealed that concentrations of H2O2 at or above 0.1 mM induced developmental toxicity, leading to increased mortality and hatching delay. Furthermore, exposure to 0.1 mM H2O2 decreased EBR at 48 hpf and impaired OMR visual behavior at 7 dpf. Additionally, exposure increased the area of apoptotic cells in the retina at 48 hpf. The addition of GSH reversed the effects of H2O2, suggesting the involvement of oxidative stress. H2O2 decreased the expression of eye development-related genes, pax6α and pax6ß. The expression of apoptosis-related genes, tp53, casp3 and bax, significantly increased, while bcl2α expression decreased. Antioxidant-related genes sod1, cat and gpx1a showed decreased expression. Expression levels of estrogen receptors (ERs) (esr1, esr2α, and esr2ß) and ovarian and brain aromatase genes (cyp19a1a and cyp19a1b, respectively) were also significantly reduced. Interestingly, co-incubation of GSH effectivity reversed the impact of H2O2 on most parameters. Overall, these results demonstrate that H2O2 induces adverse effects on visual development via oxidative stress, which leads to alter apoptosis, diminished antioxidant defenses and reduced estrogen production.


Subject(s)
Antioxidants , Apoptosis , Hydrogen Peroxide , Oxidative Stress , Zebrafish , Animals , Oxidative Stress/drug effects , Apoptosis/drug effects , Antioxidants/pharmacology , Antioxidants/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Glutathione/metabolism , Retina/drug effects , Retina/metabolism , Estrogens/pharmacology , Gene Expression Regulation, Developmental/drug effects , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Vision, Ocular/drug effects
12.
Int J Mol Sci ; 25(12)2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38928442

ABSTRACT

To enhance our understanding of teleost reproductive physiology, we identified six Sichuan bream (Sinibrama taeniatus) vitellogenin genes (vtg1-6) and characterized their sequence structures. We categorized them into type Ⅰ (vtg1,4,5 and 6), type Ⅱ (vtg2) and type Ⅲ (vtg3) based on differences in their subdomain structure. The promoter sequence of vtgs has multiple estrogen response elements, and their abundance appears to correlate with the responsiveness of vtg gene expression to estrogen. Gene expression analyses revealed that the vitellogenesis of Sichuan bream involves both heterosynthesis and autosynthesis pathways, with the dominant pathway originating from the liver. The drug treatment experiments revealed that 17ß-estradiol (E2) tightly regulated the level of vtg mRNA in the liver. Feeding fish with a diet containing 100 µg/g E2 for three weeks significantly induced vtg gene expression and ovarian development, leading to an earlier onset of vitellogenesis. Additionally, it was observed that the initiation of vtg transcription required E2 binding to its receptor, a process primarily mediated by estrogen receptor alpha in Sichuan bream. The findings of this study provide novel insights into the molecular information of the vitellogenin gene family in teleosts, thereby contributing to the regulation of gonadal development in farmed fish.


Subject(s)
Estrogens , Vitellogenins , Animals , Vitellogenins/genetics , Vitellogenins/metabolism , Estrogens/metabolism , Estrogens/pharmacology , Vitellogenesis/genetics , Estradiol/pharmacology , Estradiol/metabolism , Promoter Regions, Genetic , Female , Fish Proteins/genetics , Fish Proteins/metabolism , Phylogeny , Gene Expression Regulation/drug effects , Multigene Family , Liver/metabolism , Genome , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism
13.
Eur J Pharmacol ; 978: 176774, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38936452

ABSTRACT

AIM: Given estrogen's recognized regulatory influence on diverse metabolic and immune functions, this study sought to explore its potential impact on fibrosis and elucidate the underlying metabolic regulations. METHODS: Female mice underwent ovary removal surgery, followed by carbon tetrachloride (CCl4) administration to induce liver injury. Biochemical index analysis and histopathological examination were then conducted. The expression levels of alpha-smooth muscle actin (α-SMA), transforming growth factor-ß (TGF-ß), and collagen type 1 alpha 1 chain (COL1A1) were assessed using western blotting to further elucidate the extent of liver injury. Finally, metabolite extraction and metabolomic analysis were performed to evaluate metabolic changes. RESULTS: Ovary removal exacerbated CCl4-induced liver damage, while estrogen supplementation provided protection against hepatic changes resulting from OVX. Furthermore, estrogen mitigated liver injury induced by CCl4 treatment in vivo. Estrogen supplementation significantly restored liver damage induced by OVX and CCl4. Comparative analysis revealed significant alterations in pathways including aminoacyl-tRNA biosynthesis, glycine, serine, and threonine metabolism, lysine degradation, and taurine and hypotaurine metabolism in estrogen treatment. CONCLUSION: Estrogen supplementation alleviates liver injury induced by OVX and CCl4, highlighting its protective effects against fibrosis and associated metabolic alterations.


Subject(s)
Carbon Tetrachloride , Estrogens , Homeostasis , Liver Cirrhosis , Ovariectomy , Animals , Female , Carbon Tetrachloride/toxicity , Mice , Estrogens/pharmacology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/drug therapy , Homeostasis/drug effects , Liver/drug effects , Liver/pathology , Liver/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury/etiology , Mice, Inbred C57BL , Collagen Type I/metabolism , Transforming Growth Factor beta/metabolism
14.
Nat Aging ; 4(6): 839-853, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38858606

ABSTRACT

Thermogenic beige adipocytes are recognized as potential therapeutic targets for combating metabolic diseases. However, the metabolic advantages that they offer are compromised with aging. Here we show that treating mice with estrogen (E2), a hormone that decreases with age, can counteract the age-related decline in beige adipogenesis when exposed to cold temperature while concurrently enhancing energy expenditure and improving glucose tolerance in mice. Mechanistically, we found that nicotinamide phosphoribosyl transferase (NAMPT) plays a pivotal role in facilitating the formation of E2-induced beige adipocytes, which subsequently suppresses the onset of age-related endoplasmic reticulum (ER) stress. Furthermore, we found that targeting NAMPT signaling, either genetically or pharmacologically, can restore the formation of beige adipocytes by increasing the number of perivascular adipocyte progenitor cells. Conversely, the absence of NAMPT signaling prevents this process. Together, our findings shed light on the mechanisms regulating the age-dependent impairment of beige adipocyte formation and underscore the E2-NAMPT-controlled ER stress pathway as a key regulator of this process.


Subject(s)
Adipocytes, Beige , Adipogenesis , Aging , Endoplasmic Reticulum Stress , Estrogens , Nicotinamide Phosphoribosyltransferase , Nicotinamide Phosphoribosyltransferase/metabolism , Animals , Adipogenesis/drug effects , Endoplasmic Reticulum Stress/drug effects , Mice , Aging/drug effects , Aging/physiology , Estrogens/metabolism , Estrogens/pharmacology , Adipocytes, Beige/drug effects , Adipocytes, Beige/metabolism , Cytokines/metabolism , Signal Transduction/drug effects , Female , Mice, Inbred C57BL , Energy Metabolism/drug effects
15.
Genes (Basel) ; 15(6)2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38927745

ABSTRACT

Brain lipid homeostasis is an absolute requirement for proper functionality of nerve cells and neurological performance. Current evidence demonstrates that lipid alterations are linked to neurodegenerative diseases, especially Alzheimer's disease (AD). The complexity of the brain lipidome and its metabolic regulation has hampered the identification of critical processes associated with the onset and progression of AD. While most experimental studies have focused on the effects of known factors on the development of pathological hallmarks in AD, e.g., amyloid deposition, tau protein and neurofibrillary tangles, neuroinflammation, etc., studies addressing the causative effects of lipid alterations remain largely unexplored. In the present study, we have used a multifactor approach combining diets containing different amounts of polyunsaturated fatty acids (PUFAs), estrogen availabilities, and genetic backgrounds, i.e., wild type (WT) and APP/PS1 (FAD), to analyze the lipid phenotype of the frontal cortex in middle-aged female mice. First, we observed that severe n-3 PUFA deficiency impacts the brain n-3 long-chain PUFA (LCPUFA) composition, yet it was notably mitigated by hepatic de novo synthesis. n-6 LCPUFAs, ether-linked fatty acids, and saturates were also changed by the dietary condition, but the extent of changes was dependent on the genetic background and hormonal condition. Likewise, brain cortex phospholipids were mostly modified by the genotype (FAD>WT) with nuanced effects from dietary treatment. Cholesterol (but not sterol esters) was modified by the genotype (WT>FAD) and dietary condition (higher in DHA-free conditions, especially in WT mice). However, the effects of estrogen treatment were mostly observed in relation to phospholipid remodeling in a genotype-dependent manner. Analyses of lipid-derived variables indicate that nerve cell membrane biophysics were significantly affected by the three factors, with lower membrane microviscosity (higher fluidity) values obtained for FAD animals. In conclusion, our multifactor analyses revealed that the genotype, diet, and estrogen status modulate the lipid phenotype of the frontal cortex, both as independent factors and through their interactions. Altogether, the outcomes point to potential strategies based on dietary and hormonal interventions aimed at stabilizing the brain cortex lipid composition in Alzheimer's disease neuropathology.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Disease Models, Animal , Estrogens , Fatty Acids, Omega-3 , Frontal Lobe , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Alzheimer Disease/diet therapy , Animals , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-3/pharmacology , Mice , Frontal Lobe/metabolism , Frontal Lobe/drug effects , Frontal Lobe/pathology , Female , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Estrogens/metabolism , Estrogens/pharmacology , Mice, Transgenic , Presenilin-1/genetics , Presenilin-1/metabolism , Lipid Metabolism/drug effects , Humans
16.
Asian Pac J Cancer Prev ; 25(6): 2077-2087, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38918670

ABSTRACT

BACKGROUND: Breast cancer represents one of the leading causes of death worldwide. Apart from genetic factors, the sex hormone estrogen plays a pivotal role in breast cancer development. We are exposed to a plethora of estrogen mimics on a daily basis via various routes. Nevertheless, how xenoestrogens, the exogenous estrogen mimics, modulate cancer-associated signaling pathways and interact with specific genes is still underexplored. Hence, this study aims to explore the direct or indirect binding partners of xenoestrogens and their expression upon exposure to these estrogenic compounds. METHODS: The collection of genes linked to the xenoestrogens Octylphenol, Nonylphenol, Bisphenol-A, and 2,2-bis(4-hydroxyphenyl)-1,1,1-trichloroethane were gathered from the Comparative Toxicogenomics Database. Venny 2.1 was utilized to pinpoint the genes shared by these xenoestrogens. Subsequently, the shared genes underwent Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis using the Database for Annotation, Visualization, and Integrated Discovery bioinformatics resource. A xenoestrogen-protein interaction network was constructed using Search Tool for Interactions of Chemicals. The expressions of common genes were studied with the microarray dataset GSE5200 from the Gene Expression Omnibus database. Also, the expression of a common gene set within different breast cancer subtypes was identified using the University of California, Santa Cruz Xena. RESULTS: The genes linked to xenoestrogens were identified, and 13 genes were found to interact with all four xenoestrogens. Through DAVID analysis, the genes chosen are found to be enriched for various functions and pathways, including pathways in cancer, chemical carcinogenesis-receptor activation, and estrogen signaling pathways. The results of the Comparative Toxicogenomics Database and the chemical-protein interaction network derived from STITCH were similar. Microarray data analysis showed significantly high expression of all 13 genes in another study, with Bisphenol-A and Nonylphenol treated MCF-7 cells, most of the genes are expressed in luminal A or basal breast cancer subtype. CONCLUSION: In summary, the genes associated with the four xenoestrogens were mostly linked to pathways related to tumorigenesis, and the expression of these genes was found to be higher in breast cancer.


Subject(s)
Breast Neoplasms , Estrogens , Humans , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Estrogens/metabolism , Estrogens/pharmacology , Female , Computational Biology/methods , Computer Simulation , Protein Interaction Maps , Signal Transduction/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Benzhydryl Compounds
17.
Reproduction ; 168(2)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38833564

ABSTRACT

In brief: Atrazine, like oestrogen, disorganises laminin formation and reduces the number of germ cells and Sertoli cells in the developing testes of the tammar wallaby. This study suggests that interfering with the balance of androgen and oestrogen affects the integrity of laminin structure and testis differentiation. Abstract: The herbicide atrazine was banned in Europe in 2003 due to its endocrine disrupting activity but remains widely used. The integrity of the laminin structure in fetal testis cords requires oestrogen signalling but overexposure to xenoestrogens in the adult can cause testicular dysgenesis. However, whether xenoestrogens affect laminin formation in developing testes has not been investigated. Here we examined the effects of atrazine in the marsupial tammar wallaby during early development and compare it with the effects of the anti-androgen flutamide, oestrogen, and the oestrogen degrader fulvestrant. The tammar, like all marsupials, gives birth to altricial young, allowing direct treatment of the developing young during the male programming window (day 20-40 post partum (pp)). Male pouch young were treated orally with atrazine (5 mg/kg), flutamide (10 mg/kg), 17ß-oestradiol (2.5 mg/kg) and fulvestrant (1 mg/kg) daily from day 20 to 40 pp. Distribution of laminin, vimentin, SOX9 and DDX4, cell proliferation and mRNA expression of SRY, SOX9, AMH, and SF1 were examined in testes at day 50 post partum after the treatment. Direct exposure to atrazine, flutamide, 17ß-oestradiol, and fulvestrant all disorganised laminin but had no effect on vimentin distribution in testes. Atrazine reduced the number of germ cells and Sertoli cells when examined at day 40-50 pp and day 20 to 40 pp, respectively. Both flutamide and fulvestrant reduced the number of germ cells and Sertoli cells. Atrazine also downregulated SRY expression and impaired SOX9 nuclear translocation. Our results demonstrate that atrazine can compromise normal testicular differentiation during the critical male programming window.


Subject(s)
Atrazine , Cell Differentiation , Herbicides , Laminin , Testis , Male , Animals , Testis/drug effects , Testis/metabolism , Testis/cytology , Atrazine/pharmacology , Laminin/metabolism , Cell Differentiation/drug effects , Herbicides/pharmacology , Macropodidae/metabolism , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Sertoli Cells/cytology , Estrogens/pharmacology , Estrogens/metabolism , Endocrine Disruptors/pharmacology , Cell Count , Androgen Antagonists/pharmacology , Flutamide/pharmacology
18.
Mol Med Rep ; 30(1)2024 07.
Article in English | MEDLINE | ID: mdl-38785153

ABSTRACT

17ß­estradiol (E2) can inhibit cardiac fibrosis in female patients with heart failure (HF) and activate cell division cycle 42 (Cdc42), however it is unknown whether 17ß­estradiol (E2) can ameliorate differentiation and collagen synthesis in TGF­ß1­stimulated mouse cardiac fibroblasts (MCFs) by regulating cell division cycle 42 (Cdc42). The present study aimed to investigate the roles of estrogen and Cdc42 in preventing myocardial fibrosis and the underlying molecular mechanisms. An ELISA was used to measure the levels of E2 and Cdc42 in the serum of patients with heart failure (HF), and western blotting was used to measure the expression levels of Cdc42 in TGF­ß1­stimulated immortalized MCFs. MCFs were transfected with a Cdc42 overexpression (OE) lentivirus or small interfering RNA (siRNA), or treated with a Cdc42 inhibitor (MLS­573151), and the function of Cdc42 was assessed by western blotting, immunofluorescence staining, reverse transcription­quantitative PCR and dual­luciferase reporter assays. Western blotting and immunofluorescence staining were performed to verify the protective effect of E2 on TGF­ß1­stimulated MCFs, and the association between the protective effect and Cdc42. The results demonstrated that Cdc42 levels were increased in the serum of patients with HF and were positively correlated with the levels of E2; however, Cdc42 levels were decreased in TGF­ß1­stimulated MCFs. Cdc42 inhibited MCF differentiation and collagen synthesis, as indicated by the protein expression of α­smooth muscle actin, collagen I and collagen III. Mechanistically, Cdc42 inhibited the transcription of TGF­ß1 by promoting the expression of p21 (RAC1)­activated kinase 1 (Pak1)/JNK/c­Jun signaling pathway proteins and inhibiting the activity of the Tgfb1 gene promoter. In addition, E2 inhibited the differentiation and collagen synthesis of TGF­ß1­stimulated MCFs, and promoted the protein expression of Pak1, JNK and c­Jun, consistent with the effects of Cdc42, whereas the effects of E2 were abolished when Cdc42 was knocked down. The aforementioned findings suggested that E2 could inhibit differentiation and collagen synthesis in TGF­ß1­stimulated MCFs by regulating Cdc42 and the downstream Pak1/JNK/c­Jun signaling pathway.


Subject(s)
Cell Differentiation , Collagen , Estradiol , Estrogens , Fibroblasts , Transforming Growth Factor beta1 , cdc42 GTP-Binding Protein , cdc42 GTP-Binding Protein/metabolism , cdc42 GTP-Binding Protein/genetics , Animals , Cell Differentiation/drug effects , Mice , Transforming Growth Factor beta1/metabolism , Humans , Collagen/metabolism , Collagen/biosynthesis , Female , Fibroblasts/metabolism , Fibroblasts/drug effects , Estrogens/pharmacology , Estradiol/pharmacology , Middle Aged , Myocardium/metabolism , Heart Failure/metabolism , Male , Signal Transduction/drug effects
19.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731947

ABSTRACT

Estrogen plays an important role in osteoporosis prevention. We herein report the possible novel signaling pathway of 17ß-estradiol (E2) in the matrix mineralization of MC3T3-E1, an osteoblast-like cell line. In the culture media-containing stripped serum, in which small lipophilic molecules such as steroid hormones including E2 were depleted, matrix mineralization was significantly reduced. However, the E2 treatment induced this. The E2 effects were suppressed by ICI182,780, the estrogen receptor (ER)α, and the ERß antagonist, as well as their mRNA knockdown, whereas Raloxifene, an inhibitor of estrogen-induced transcription, and G15, a G-protein-coupled estrogen receptor (GPER) 1 inhibitor, had little or no effect. Furthermore, the E2-activated matrix mineralization was disrupted by PMA, a PKC activator, and SB202190, a p38 MAPK inhibitor, but not by wortmannin, a PI3K inhibitor. Matrix mineralization was also induced by the culture media from the E2-stimulated cell culture. This effect was hindered by PMA or heat treatment, but not by SB202190. These results indicate that E2 activates the p38 MAPK pathway via ERs independently from actions in the nucleus. Such activation may cause the secretion of certain signaling molecule(s), which inhibit the PKC pathway. Our study provides a novel pathway of E2 action that could be a therapeutic target to activate matrix mineralization under various diseases, including osteoporosis.


Subject(s)
Estradiol , Osteoblasts , Signal Transduction , Animals , Mice , Estradiol/pharmacology , Osteoblasts/metabolism , Osteoblasts/drug effects , Signal Transduction/drug effects , Calcification, Physiologic/drug effects , Cell Line , p38 Mitogen-Activated Protein Kinases/metabolism , Receptors, Estrogen/metabolism , Receptors, Estrogen/genetics , Estrogens/pharmacology , Estrogens/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics
20.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167227, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38733774

ABSTRACT

Olanzapine (OLA) is a highly obesogenic second-generation antipsychotic (SGA). Recently we demonstrated that, contrarily to OLA oral treatment, intraperitoneal (i.p.) administration resulted in weight loss and absence of hepatic steatosis in wild-type (WT) and protein tyrosine phosphatase 1B (PTP1B)-deficient (KO) male mice. This protection relied on two central-peripheral axes connecting hypothalamic AMPK with brown/inguinal white adipose tissue (BAT/iWAT) uncoupling protein-1 (UCP-1) and hypothalamic JNK with hepatic fatty acid synthase (FAS). Herein, we addressed OLA i.p. treatment effects in WT and PTP1B-KO female mice. Contrarily to our previous results in WT females receiving OLA orally, the i.p. treatment did not induce weight gain or hyperphagia. Molecularly, in females OLA failed to diminish hypothalamic phospho-AMPK or elevate BAT UCP-1 and energy expenditure (EE) despite the preservation of iWAT browning. Conversely, OLA i.p. treatment in ovariectomized mice reduced hypothalamic phospho-AMPK, increased BAT/iWAT UCP-1 and EE, and induced weight loss as occurred in males. Pretreatment of hypothalamic neurons with 17ß-estradiol (E2) abolished OLA effects on AMPK. Moreover, neither hypothalamic JNK activation nor hepatic FAS upregulation were found in WT and PTP1B-KO females receiving OLA via i.p. Importantly, this axis was reestablished upon ovariectomy. In this line, E2 prevented OLA-induced phospho-JNK in hypothalamic neurons. These results support the role of estrogens in sex-related dimorphism in OLA treatment. This study evidenced the benefit of OLA i.p. administration in preventing its obesogenic effects in female mice that could offer clinical value.


Subject(s)
Adipose Tissue, Brown , Estrogens , Hypothalamus , Liver , Mice, Knockout , Olanzapine , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Uncoupling Protein 1 , Animals , Female , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Hypothalamus/metabolism , Hypothalamus/drug effects , Mice , Liver/metabolism , Liver/drug effects , Estrogens/metabolism , Estrogens/pharmacology , Olanzapine/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Male , Energy Metabolism/drug effects , Injections, Intraperitoneal , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects , Mice, Inbred C57BL , Estradiol/pharmacology , Ovariectomy
SELECTION OF CITATIONS
SEARCH DETAIL