Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.678
Filtrer
1.
Sci Rep ; 14(1): 18400, 2024 08 08.
Article de Anglais | MEDLINE | ID: mdl-39117675

RÉSUMÉ

Estrogens regulate numerous physiological and pathological processes, including wide-ranging effects in wound healing. The effects of estrogens are mediated through multiple estrogen receptors (ERs), including the classical nuclear ERs (ERα and ER ß ), that typically regulate gene expression, and the 7-transmembrane G protein-coupled estrogen receptor (GPER), that predominantly mediates rapid "non-genomic" signaling. Estrogen modulates the expression of various genes involved in epidermal function and regeneration, inflammation, matrix production, and protease inhibition, all critical to wound healing. Our previous work demonstrated improved myocutaneous wound healing in female mice compared to male mice. In the current study, we employed male and female GPER knockout mice to investigate the role of this estrogen receptor in wound revascularization and tissue viability. Using a murine myocutaneous flap model of graded ischemia, we measured real-time flap perfusion via laser speckle perfusion imaging. We conducted histologic and immunohistochemical analyses to assess skin and muscle viability, microvascular density and vessel morphology. Our results demonstrate that GPER is crucial in wound healing, mediating effects that are both dependent and independent of sex. Lack of GPER expression is associated with increased skin necrosis, reduced flap perfusion and altered vessel morphology. These findings contribute to understanding GPER signaling in wound healing and suggest possible therapeutic opportunities by targeting GPER.


Sujet(s)
Souris knockout , Néovascularisation physiologique , Récepteurs des oestrogènes , Récepteurs couplés aux protéines G , Cicatrisation de plaie , Animaux , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Mâle , Souris , Femelle , Peau/métabolisme , Peau/vascularisation , Ischémie/métabolisme , Lambeaux chirurgicaux
2.
Technol Cancer Res Treat ; 23: 15330338241277699, 2024.
Article de Anglais | MEDLINE | ID: mdl-39161322

RÉSUMÉ

Introduction: A strong association was previously established between body mass index (BMI) and female reproductive system tumors; however, the causal relationship is unclear. We conducted a Mendelian randomization (MR) study to further explore this association. Methods: Genetic information for BMI was retrieved from a published genome-wide association study involving 339,224 participants. Genetic associations with five common female reproductive system tumors were obtained from the FinnGen, UK Biobank studies, and other large consortia. Results: Genetic predisposition towards BMI exhibits a significant association with multiple tumors of the female reproductive system. Specifically, for every 1-unit increase in BMI log-transformed odds ratio (OR). The OR fluctuations overall for patients with breast cancer ranged from 0.661 to 0.996 (95% confidence interval [CI],0.544-1.000, P < 0.05). When stratified by estrogen receptor (ER) status, the OR for patients with ER (+) breast cancer ranged from 0.782 to 0.844 (95% CI, 0.616-0.994, P < 0.05) and that for those with ER (-) breast cancer ranged from 0.663 to 0.789 (95% CI, 0.498-0.991, P < 0.05). Additionally, ORs were as follows for cancer types: 1.577-1.908 (95% CI, 1.049-2.371, P < 0.05) for endometrial carcinoma; 1.216-1.303 (95% CI, 1.021-1.591, P < 0.05) for high-grade serous ovarian cancer; 1.217 (95% CI, 1.034-1.432, P < 0.05) for low-grade malignant serous ovarian cancer; and 1.502 (95% CI, 1.112-2.029, P < 0.05) for endometrioid ovarian carcinoma. Furthermore, our findings indicated that genetic predisposition towards BMI did not exhibit a causal association with uterine fibroids, cervical precancerous lesions, or cervical cancer itself. Conclusion: A genetic association was established between a high BMI and high risk of developing multiple tumors of the female reproductive system and their associated subtypes. This underscores the significance of taking measures to prevent reproductive system tumors in women who have a high BMI.


Sujet(s)
Indice de masse corporelle , Prédisposition génétique à une maladie , Étude d'association pangénomique , Analyse de randomisation mendélienne , Humains , Femelle , Odds ratio , Polymorphisme de nucléotide simple , Tumeurs de l'appareil génital féminin/génétique , Tumeurs de l'appareil génital féminin/étiologie , Tumeurs de l'appareil génital féminin/épidémiologie , Facteurs de risque , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique
3.
J Hazard Mater ; 477: 135379, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-39096633

RÉSUMÉ

Tris (2,6-dimethylphenyl) phosphate (TDMPP), a novel organic phosphorus flame retardant (OPFR), has been found to have estrogenic activity. Estrogens are critical in regulating various biological responses during liver development. However, the effects of TDMPP on zebrafish liver development remain largely unexplored. Here, we utilized a chemical genetic screening approach to assess the estrogenic effects of TDMPP on liver development and to elucidate the underlying molecular mechanism. Our findings revealed that zebrafish larvae exposed to environmentally relevant concentrations of TDMPP (0.05 and 0.5 µM) exhibited concentration-dependent liver impairments, including reduced liver size, histopathological changes, and hepatocyte apoptosis. In addition, E2 caused similar adverse effects to TDMPP, but the pharmacological blockade of estrogen synthesis alleviated the effects on liver development. Chemical inhibitors and morpholino knockdown assays indicated that the reduction of esr2a blocked TDMPP-induced liver impairments, which was further confirmed in the esr2a-/- mutant line. Subsequently, transcriptomic analysis showed that the estrogen receptor activated by TDMPP inhibited the expression of smc2, which was linked to the suppression of liver development through p53 activation. Consistently, overexpression of smc2 and inhibition of p53 evidently rescued hepatic damages induced by TDMPP. Taken together, the above findings identified esr2a, downstream smc2, and p53 as important regulators for the estrogenic effects of TDMPP on liver development. Our work fills crucial gaps in the current knowledge of TDMPP's hepatotoxicity, providing new insights into the adverse effects of TDMPP and the molecular mechanisms of action. These findings underscore the need for further ecological risk assessment and regulatory considerations.


Sujet(s)
Foie , Transduction du signal , Protéine p53 suppresseur de tumeur , Protéines de poisson-zèbre , Danio zébré , Animaux , Apoptose/effets des médicaments et des substances chimiques , Ignifuges/toxicité , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Organophosphates/toxicité , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Protéines de poisson-zèbre/génétique , Protéines de poisson-zèbre/métabolisme
4.
Methods Mol Biol ; 2846: 133-150, 2024.
Article de Anglais | MEDLINE | ID: mdl-39141234

RÉSUMÉ

Gonadal steroid hormones, namely, testosterone, progesterone, and estrogens, influence the physiological state of an organism through the regulation of gene transcription. Steroid hormones activate nuclear hormone receptor (HR), transcription factors (TFs), which bind DNA in a tissue- and cell type-specific manner to influence cellular function. Identifying the genomic binding sites of HRs is essential to understanding mechanisms of hormone signaling across tissues and disease contexts. Traditionally, chromatin immunoprecipitation followed by sequencing (ChIP-seq) has been used to map the genomic binding of HRs in cancer cell lines and large tissues. However, ChIP-seq lacks the sensitivity to detect TF binding in small numbers of cells, such as genetically defined neuronal subtypes in the brain. Cleavage Under Targets & Release Under Nuclease (CUT&RUN) resolves most of the technical limitations of ChIP-seq, enabling the detection of protein-DNA interactions with as few as 100-1000 cells. In this chapter, we provide a stepwise CUT&RUN protocol for detecting and analyzing the genome-wide binding of estrogen receptor α (ERα) in mouse brain tissue. The steps described here can be used to identify the genomic binding sites of most TFs in the brain.


Sujet(s)
Séquençage après immunoprécipitation de la chromatine , Chromatine , Animaux , Chromatine/métabolisme , Chromatine/génétique , Souris , Séquençage après immunoprécipitation de la chromatine/méthodes , Sites de fixation , Immunoprécipitation de la chromatine/méthodes , Encéphale/métabolisme , Récepteur alpha des oestrogènes/métabolisme , Récepteur alpha des oestrogènes/génétique , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Liaison aux protéines , Humains , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique
5.
Sheng Wu Gong Cheng Xue Bao ; 40(7): 2308-2321, 2024 Jul 25.
Article de Chinois | MEDLINE | ID: mdl-39044593

RÉSUMÉ

This study aims to explore the roles of three estrogen receptors (Esr1, Esr2, and Gper1) in early differentiation of embryonic gonads of Trachemys scripta. The expression characteristics of the receptor genes were studied first. The Esr1, Esr2, and Gper1 agonists PPT, WAY 200070, and G-1 were respectively injected into the embryos at the male-producing temperature (MPT) before initiation of gonadal differentiation. The sex reversal of the treated embryonic gonads was analyzed in terms of morphological structure of gonads, distribution pattern of germ cells, and expression of key genes and proteins involved in sex differentiation. The expression level of esr1 during the critical stage of sex differentiation was higher than those of esr2 and gper1 (very low expression) and was particularly high in the gonads at the female-producing temperature (FPT). After treatment with PPT, the MPT gonads presented obviously feminized morphology and structure, with the germ cells exhibiting a female distribution pattern. Furthermore, the mRNA expression levels of the key genes (dmrt1, amh, and sox9) for male differentiation were down-regulated significantly, while those of the key genes (foxl2 and cyp19a1) for female differentiation were up-regulated observably. The fluorescent signals of Amh and Sox9 expression almost disappeared, while Foxl2 and Arom were activated to express abundantly, which fully demonstrated the sex reversal of the gonads from male to female (sex reversal rate: 70.27%). However, the MPT gonads treated with WAY 200070 and G-1 still differentiated into testes, and the expression patterns of the key genes and proteins were similar to those in male gonads. The above results demonstrate that activation of Esr1 alone can fully initiate the early female differentiation process of gonads, suggesting that estrogen may induce early ovarian differentiation via Esr1 in Trachemys scripta. The findings provide a basis for further revealing the mechanisms of estrogen regulation in sex determination and differentiation of turtles.


Sujet(s)
Récepteur alpha des oestrogènes , Ovaire , Différenciation sexuelle , Tortues , Animaux , Femelle , Différenciation sexuelle/génétique , Ovaire/métabolisme , Ovaire/croissance et développement , Récepteur alpha des oestrogènes/génétique , Récepteur alpha des oestrogènes/métabolisme , Mâle , Tortues/génétique , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Régulation de l'expression des gènes au cours du développement/effets des médicaments et des substances chimiques
6.
Int J Mol Sci ; 25(14)2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39063107

RÉSUMÉ

It is not entirely clear how the interaction between joint inflammation and the central nervous system (CNS) response in rheumatoid arthritis (RA) works, and what pathophysiology underlies the sex differences in coexisting neuropsychiatric comorbidities. It is known that estrogen hormones reduce inflammation in RA and that this occurs mainly via the stimulation of G protein-coupled receptor-30 (GPR30), also known as G protein-coupled estrogen receptor (GPER) 1. However, changes in GPR30 expression and sex differences induced by local and systemic inflammation in RA are not yet known. Our aim was to reveal sex differences in the expression and association of joint GPR30 with local and systemic inflammation, clinical course and furthermore with hippocampal GPR30 expression during pristane-induced arthritis (PIA) in Dark Agouti (DA) rats, an animal model of RA. Furthermore, we demonstrated sex-specific differences in the association between joint and systemic inflammation and hippocampal microglia during PIA. Our results suggest sex-specific differences not only in the clinical course and serum levels of pro-inflammatory cytokines but also in the expression of GPR30. Female rats show greater synovial inflammation and greater damage to the articular cartilage compared to males during PIA attack. Male rats express higher levels of synovial and cartilaginous GPR30 than females during PIA, which correlates with a less severe clinical course. The correlation between synovial and cartilaginous GPR30 and joint inflammation scores (Krenn and Mankin) in male rats suggests that the more severe the joint inflammation, the higher the GPR30 expression. At the same time, there is no particular upregulation of hippocampal GPR30 in males. On the other hand, female rats express higher levels of neuroprotective GPR30 in the hippocampus than male rats at the basic level and during PIA attack. In addition, females have a higher number of Iba-1+ cells in the hippocampus during PIA attack that strongly correlates with the clinical score, serum levels of IL-17A, and Krenn and Mankin scores. These results suggest that male rats are better protected from inflammation in the joints and female rats are better protected from the inflammation in the hippocampus during a PIA attack, independently of microglia proliferation. However, in the remission phase, synovial GPR30 expression suddenly increases in female rats, as does hippocampal GPR30 expression in males. Further experiments with a longer remission period are needed to investigate the molecular background of these sex differences, as well as microglia phenotype profiling.


Sujet(s)
Polyarthrite rhumatoïde , Modèles animaux de maladie humaine , Hippocampe , Récepteurs couplés aux protéines G , Animaux , Femelle , Mâle , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/génétique , Hippocampe/métabolisme , Rats , Inflammation/métabolisme , Arthrite expérimentale/métabolisme , Arthrite expérimentale/anatomopathologie , Caractères sexuels , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Microglie/métabolisme , Facteurs sexuels , Terpènes
7.
Clin Lab ; 70(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38965945

RÉSUMÉ

BACKGROUND: The aim of this study was to explore the causal relationship between different serum iron statuses (ferritin, transferrin, transferrin saturation, and serum iron) and the occurrence of estrogen receptor (ER)-positive or ER-negative breast cancer. METHODS: The summary data on serum iron status exposure were gathered from the IEU OpenGWAS Project, the UK Biobank, and other databases. Concurrently, the summary data for ER+ and ER- breast cancer are sourced from the Breast Cancer Association Consortium (BCAC). By examining the causal link between iron status and breast cancer, we deployed five distinct Mendelian randomization (MR) algorithms, namely MR-Egger, inverse variance weighted (IVW), weighted median, simple mode, and MR-PRESSO. To assess heterogeneity and horizontal pleiotropy, Cochran's Q and MR-Egger algorithms were applied, respectively. RESULTS: Elevated ferritin levels are associated with an increased risk of ER-negative breast cancer (OR(IVW) = 1.042, 95% CI (1.005, 1.081), p = 0.025; OR (weighted median) = 1.050, 95% CI (1.001, 1.102), p = 0.046; and OR (MR-PRESSO) = 1.042, 95% CI (1.005, 1.081), p = 0.039). Conversely, an increase in the serum iron level is linked to a reduced risk of ER-negative breast cancer (OR (IVW) = 0.791, 95% CI (0.649, 0.962), p = 0.019; and OR (MR-PRESSO) = 0.791, 95% CI (0.649, 0.962), p = 0.028). However, there is no evidence of a causal relationship between transferrin, transferrin saturation, and ER-negative breast cancer. For ER-positive breast cancer, none of the four different iron statuses demonstrated a causal relationship. CONCLUSIONS: Ferritin is positively correlated with ER-negative breast cancer, while serum iron is negatively associated with ER-negative breast cancer. However, there is no causal relationship between the four iron statuses and ER-positive breast cancer.


Sujet(s)
Tumeurs du sein , Ferritines , Fer , Analyse de randomisation mendélienne , Récepteurs des oestrogènes , Humains , Tumeurs du sein/sang , Tumeurs du sein/génétique , Ferritines/sang , Femelle , Fer/sang , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Transferrine/analyse , Transferrine/métabolisme , Facteurs de risque
8.
Int J Mol Sci ; 25(13)2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-39000600

RÉSUMÉ

Women with type 2 diabetes (T2D) have a higher risk of being diagnosed with breast cancer and have worse survival than non-diabetic women if they do develop breast cancer. However, more research is needed to elucidate the biological underpinnings of these relationships. Here, we found that forkhead box A1 (FOXA1), a forkhead family transcription factor, and metformin (1,1-dimethylbiguanide hydrochloride), a medication used to treat T2D, may impact hormone-receptor-positive (HR+) breast cancer (BC) tumor cell growth and metastasis. Indeed, fourteen diabetes-associated genes are highly expressed in only three HR+ breast cancer cell lines but not the other subtypes utilizing a 53,805 gene database obtained from NCBI GEO. Among the diabetes-related genes, FOXA1, MTA3, PAK4, FGFR3, and KIF22 were highly expressed in HR+ breast cancer from 4032 breast cancer patient tissue samples using the Breast Cancer Gene Expression Omnibus. Notably, elevated FOXA1 expression correlated with poorer overall survival in patients with estrogen-receptor-positive/progesterone-receptor-positive (ER+/PR+) breast cancer. Furthermore, experiments demonstrated that loss of the FOXA1 gene inhibited tumor proliferation and invasion in vitro using MCF-7 and T47D HR+ breast cancer cell lines. Metformin, an anti-diabetic medication, significantly suppressed tumor cell growth in MCF-7 cells. Additionally, either metformin treatment or FOXA1 gene deletion enhanced tamoxifen-induced tumor growth inhibition in HR+ breast cancer cell lines within an ex vivo three-dimensional (3D) organoid model. Therefore, the diabetes-related medicine metformin and FOXA1 gene inhibition might be a new treatment for patients with HR+ breast cancer when combined with tamoxifen, an endocrine therapy.


Sujet(s)
Tumeurs du sein , Prolifération cellulaire , Facteur nucléaire hépatocytaire HNF-3 alpha , Metformine , Facteur nucléaire hépatocytaire HNF-3 alpha/métabolisme , Facteur nucléaire hépatocytaire HNF-3 alpha/génétique , Humains , Metformine/pharmacologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Femelle , Prolifération cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Invasion tumorale , Cellules MCF-7 , Récepteurs à la progestérone/métabolisme , Récepteurs à la progestérone/génétique
9.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-39000231

RÉSUMÉ

Following metastatic spread, many hormone receptor positive (HR+) patients develop a more aggressive phenotype with an observed loss of the HRs estrogen receptor (ER) and progesterone receptor (PR). During metastasis, breast cancer cells are exposed to high magnitudes of fluid shear stress (FSS). Unfortunately, the role for FSS on the regulation of HR expression and function during metastasis is not fully understood. This study was designed to elucidate the impact of FSS on HR+ breast cancer. Utilizing a microfluidic platform capable of exposing breast cancer cells to FSS that mimics in situ conditions, we demonstrate the impact of FSS exposure on representative HR+ breast cancer cell lines through protein and gene expression analysis. Proteomics results demonstrated that 540 total proteins and 1473 phospho-proteins significantly changed due to FSS exposure and pathways of interest included early and late estrogen response. The impact of FSS on response to 17ß-estradiol (E2) was next evaluated and gene expression analysis revealed repression of ER and E2-mediated genes (PR and SDF1) following exposure to FSS. Western blot demonstrated enhanced phosphorylation of mTOR following exposure to FSS. Taken together, these studies provide initial insight into the effects of FSS on HR signaling in metastatic breast cancer.


Sujet(s)
Tumeurs du sein , Régulation de l'expression des gènes tumoraux , Récepteurs des oestrogènes , Récepteurs à la progestérone , Contrainte mécanique , Humains , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Femelle , Récepteurs à la progestérone/métabolisme , Récepteurs à la progestérone/génétique , Lignée cellulaire tumorale , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Oestradiol/pharmacologie , Phosphorylation , Transduction du signal , Sérine-thréonine kinases TOR/métabolisme , Protéomique/méthodes , Cellules MCF-7 , Chimiokine CXCL12/métabolisme , Chimiokine CXCL12/génétique
10.
ASN Neuro ; 16(1): 2368382, 2024.
Article de Anglais | MEDLINE | ID: mdl-39024550

RÉSUMÉ

Ventromedial hypothalamic nucleus (VMN) growth hormone-releasing hormone (Ghrh) neurotransmission shapes counterregulatory hormone secretion. Dorsomedial VMN Ghrh neurons express the metabolic-sensitive transcription factor steroidogenic factor-1/NR5A1 (SF-1). In vivo SF-1 gene knockdown tools were used here to address the premise that in male rats, SF-1 may regulate basal and/or hypoglycemic patterns of Ghrh, co-transmitter biosynthetic enzyme, and estrogen receptor (ER) gene expression in these neurons. Single-cell multiplex qPCR analyses showed that SF-1 regulates basal profiles of mRNAs that encode Ghrh and protein markers for neurochemicals that suppress (γ-aminobutyric acid) or enhance (nitric oxide; glutamate) counterregulation. SF-1 siRNA pretreatment respectively exacerbated or blunted hypoglycemia-associated inhibition of glutamate decarboxylase67 (GAD67/GAD1) and -65 (GAD65/GAD2) transcripts. Hypoglycemia augmented or reduced nitric oxide synthase and glutaminase mRNAs, responses that were attenuated by SF-1 gene silencing. Ghrh and Ghrh receptor transcripts were correspondingly refractory to or increased by hypoglycemia, yet SF-1 knockdown decreased both gene profiles. Hypoglycemic inhibition of ER-alpha and G protein-coupled-ER gene expression was amplified by SF-1 siRNA pretreatment, whereas as ER-beta mRNA was amplified. SF-1 knockdown decreased (corticosterone) or elevated [glucagon, growth hormone (GH)] basal counterregulatory hormone profiles, but amplified hypoglycemic hypercorticosteronemia and -glucagonemia or prevented elevated GH release. Outcomes document SF-1 control of VMN Ghrh neuron counterregulatory neurotransmitter and ER gene transcription. SF-1 likely regulates Ghrh nerve cell receptivity to estradiol and release of distinctive neurochemicals during glucose homeostasis and systemic imbalance. VMN Ghrh neurons emerge as a likely substrate for SF-1 control of glucose counterregulation in the male rat.


Sujet(s)
Hormone de libération de l'hormone de croissance , Neurones , Rat Sprague-Dawley , Facteur stéroïdogène-1 , Noyau ventromédial de l'hypothalamus , Animaux , Mâle , Hormone de libération de l'hormone de croissance/métabolisme , Hormone de libération de l'hormone de croissance/génétique , Noyau ventromédial de l'hypothalamus/métabolisme , Facteur stéroïdogène-1/métabolisme , Facteur stéroïdogène-1/génétique , Neurones/métabolisme , Rats , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Glutamate decarboxylase/métabolisme , Glutamate decarboxylase/génétique , Régulation de l'expression des gènes , Hypoglycémie/métabolisme , Petit ARN interférent/pharmacologie
11.
Gene ; 928: 148769, 2024 Nov 30.
Article de Anglais | MEDLINE | ID: mdl-39025340

RÉSUMÉ

There is increasing evidence that gender impacts the onset and progression of cardiovascular pathology. However, it is vastly unclear how this variable determines the ultimate outcomes, particularly in the setting of pressure overload-induced left ventricular hypertrophy (LVH). This study was carried out to fill this gap, at least in part, by assessing myocardial expression of G protein-coupled estrogen receptor (GPER) in female and male rats afflicted with LVH. Both female and male rats underwent abdominal aorta banding to induce LVH or were kept intact as control groups. At the end of the experiment, carotid artery catheterization was performed to measure systolic (SBP) and diastolic (DBP) blood pressure. Fibrosis and cardiomyocyte cross-sectional area were assessed by conventional histological analyses. Protein and mRNA expression were evaluated by Western blot/immunofluorescence staining and real-time RT-PCR technique, respectively. In LVH groups, male rats exhibited higher SBP and DBP, heart weight to body weight ratio, and fibrosis compared with female rats. However, both sexes showed a similar increase in cardiomyocyte size after LVH induction. In female, but not in male rats, LVH instigated the GPER mRNA and protein expression in the heart. These results, confirm a significant interaction between gender and myocardial remodeling in terms of GPER expression. Thus, it can be argued that sex differences in the cardiac GPER expression may be responsible for sex differences in the pressure overload-induced LVH. In other words, the female heart seems to unleash stronger protection against pressure overload than that of males in light of a higher GPER expression.


Sujet(s)
Hypertrophie ventriculaire gauche , Récepteurs des oestrogènes , Récepteurs couplés aux protéines G , Animaux , Mâle , Femelle , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Rats , Hypertrophie ventriculaire gauche/métabolisme , Hypertrophie ventriculaire gauche/génétique , Hypertrophie ventriculaire gauche/anatomopathologie , Hypertrophie ventriculaire gauche/étiologie , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Pression sanguine , Myocarde/métabolisme , Myocarde/anatomopathologie , Caractères sexuels , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Fibrose , Rat Sprague-Dawley , Facteurs sexuels
12.
Pharmacogenet Genomics ; 34(7): 246-251, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38842463

RÉSUMÉ

Studies have reported overexpression of NAT1 gene for xenobiotic metabolizing arylamine N -acetyltransferase type 1 in estrogen receptor positive breast tumors, and this association has been linked to patient chemoresistance and response to tamoxifen. We probed the expression of NAT1 , using quantitative reverse transcription PCR to screen clinically characterized breast cancer tissue cDNA arrays. Primers detecting all NAT1 alternative transcripts were used, and the protocol and results are reported according to consensus guidelines. The clinical information about 166 tumor samples screened is provided, including tumor stage, estrogen and progesterone receptor status and HER2 expression. NAT1 was found to be significantly ( P  < 0.001) upregulated in hormone receptor positive vs. negative tumors. No correlation was apparent between NAT1 and tumor stage or HER2 expression. Our findings demonstrate a strong correlation between the expression of NAT1 and steroid hormone receptors in breast tumors, supporting its possible utility as a pharmacogenetic biomarker or drug target. Of the two polymorphic NAT genes, NAT1 is the one primarily expressed in breast tissue, and is subjected to regulation by two differential promoters and more than one polyadenylation signal. Hormonal factors may enhance NAT1 gene expression at the transcriptional or epigenetic level, and tamoxifen has additionally been shown to inhibit NAT1 enzymatic activity. The outcome of tamoxifen treatment is also more favorable in patients with NAT1 overexpressing tumors. The study adds to the growing body of evidence implicating NAT1 in breast cancer and its pharmacological treatment.


Sujet(s)
Arylamine N-acetyltransferase , Tumeurs du sein , Isoenzymes , Récepteurs des oestrogènes , Humains , Arylamine N-acetyltransferase/génétique , Tumeurs du sein/génétique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Femelle , Récepteurs des oestrogènes/génétique , Récepteurs des oestrogènes/métabolisme , Isoenzymes/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Récepteurs à la progestérone/génétique , Récepteurs à la progestérone/métabolisme , Tamoxifène/usage thérapeutique , Tamoxifène/pharmacologie , Adulte d'âge moyen , Récepteur ErbB-2/génétique , Récepteur ErbB-2/métabolisme
13.
Int J Mol Sci ; 25(11)2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38892447

RÉSUMÉ

Bisphenol-A (BPA), a synthetic compound ubiquitously present in the environment, can act as an endocrine disruptor by binding to both canonical and non-canonical estrogen receptors (ERs). Exposure to BPA has been linked to various cancers, in particular, those arising in hormone-targeted tissues such as the breast. In this study, we evaluated the effect of BPA intake through drinking water on ErbB2/neu-driven cancerogenesis in BALB-neuT mice, transgenic for a mutated ErbB2/neu receptor gene, which reproducibly develop carcinomas in all mammary glands. In this model, BPA accelerated mammary cancerogenesis with an increase in the number of tumors per mouse and a concurrent decrease in tumor-free and overall survival. As assessed by immunohistochemistry, BALB-neuT tumors were ER-negative but expressed high levels of the alternative estrogen receptor GPR30, regardless of BPA exposure. On the other hand, BPA exposure resulted in a marked upregulation of progesterone receptors in preinvasive tumors and of Ki67, CD31, and phosphorylated Akt in invasive tumors. Moreover, based on several infiltration markers of immune cells, BPA favored an immunosuppressive tumor microenvironment. Finally, in vitro cell survival studies performed on a cell line established from a BALB-neuT breast carcinoma confirmed that BPA's impact on cancer progression can be particularly relevant after chronic, low-dose exposure.


Sujet(s)
Composés benzhydryliques , Souris de lignée BALB C , Phénols , Récepteurs des oestrogènes , Microenvironnement tumoral , Animaux , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Femelle , Souris , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Eau de boisson , Tumeurs expérimentales de la mamelle/induit chimiquement , Tumeurs expérimentales de la mamelle/anatomopathologie , Tumeurs expérimentales de la mamelle/métabolisme , Souris transgéniques , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Récepteurs à la progestérone/métabolisme , Récepteurs à la progestérone/génétique , Carcinogenèse/induit chimiquement , Carcinogenèse/effets des médicaments et des substances chimiques , Perturbateurs endocriniens/toxicité
14.
Genes (Basel) ; 15(6)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38927728

RÉSUMÉ

Background: Breast cancer (BC) has the highest morbidity rate and the second-highest mortality rate of all cancers among women. Recently, multi-cancer genome profiling (multi-CGP) tests have become clinically available. In this study, we aimed to clarify the significance of multi-CGP testing of BC by using the large clinical dataset from The Center for Cancer Genomics and Advanced Therapeutics (C-CAT) profiling database in Japan. Materials and Methods: A total of 3744 BC cases were extracted from the C-CAT database, which enrolled 60,250 patients between June 2019 and October 2023. Of the 3744 BC cases, a total of 3326 cases for which the C-CAT included information on ER, PR, and HER2 status were classified into four subtypes, including TNBC, HR+/HER2-, HR+/HER2+, and HR-/HER2+. Comparisons between groups were performed by the χ2 test or Fisher's exact test using EZR. Kaplan-Meier curves were created using the log-rank test. Results: Of all 3326 cases analyzed, 1114 (33.5%) were TNBC cases, HR+/HER2- accounted for 1787 cases (53.7%), HR+/HER2+ for 260 cases (7.8%), and HR-/HER2+ for 165 cases (5.0%). Genetic abnormalities were most frequently detected in TP53 (58.0%), PIK3CA (35.5%), MYC (18.7%), FGF19 (15.5%), and GATA3 (15.1%) across all BCs. The rate of TMB-High was 12.3%, and the rate of MSI-High was 0.3%, in all BC cases. Therapeutic drugs were proposed for patients with mutations in six genes: PIK3CA, ERBB2, PTEN, FGFR1, ESR1, and AKT1. The prognoses of HR+/HER2- cases were significantly (p = 0.044) better in the treated group than in the untreated group. Conclusions: These findings suggest that cancer gene panel testing is useful for HR+/HER2- cases.


Sujet(s)
Tumeurs du sein , Récepteur ErbB-2 , Humains , Femelle , Japon/épidémiologie , Adulte d'âge moyen , Tumeurs du sein/génétique , Récepteur ErbB-2/génétique , Sujet âgé , Adulte , Études rétrospectives , Marqueurs biologiques tumoraux/génétique , Récepteurs des oestrogènes/génétique , Récepteurs des oestrogènes/métabolisme , Récepteurs à la progestérone/génétique , Récepteurs à la progestérone/métabolisme , Sujet âgé de 80 ans ou plus , Pronostic , Mutation , Analyse de profil d'expression de gènes/méthodes , Phosphatidylinositol 3-kinases de classe I/génétique
15.
Hum Genomics ; 18(1): 67, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38886847

RÉSUMÉ

BACKGROUND: Sex-related differences in colorectal (CRC) incidence and mortality are well-documented. However, the impact of sex on metabolic pathways that drive cancer growth is not well understood. High expression of asparagine synthetase (ASNS) is associated with inferior survival for female CRC patients only. Here, we used a CRISPR/Cas9 technology to generate HCT116 ASNS-/- and HCT 116 ASNS+/+ cancer cell lines. We examine the effects of ASNS deletion on tumor growth and the subsequent rewiring of metabolic pathways in male and female Rag2/IL2RG mice. RESULTS: ASNS loss reduces cancer burden in male and female tumor-bearing mice (40% reduction, q < 0.05), triggers metabolic reprogramming including gluconeogenesis, but confers a survival improvement (30 days median survival, q < 0.05) in female tumor-bearing mice alone. Transcriptomic analyses revealed upregulation of G-protein coupled estrogen receptor (GPER1) in tumors from male and female mice with HCT116 ASNS-/- xenograft. Estradiol activates GPER1 in vitro in the presence of ASNS and suppresses tumor growth. CONCLUSIONS: Our study indicates that inferior survival for female CRC patients with high ASNS may be due to metabolic reprogramming that sustains tumor growth. These findings have translational relevance as ASNS/GPER1 signaling could be a future therapeutic target to improve the survival of female CRC patients.


Sujet(s)
Aspartate-ammonia ligase , Animaux , Humains , Femelle , Mâle , Souris , Aspartate-ammonia ligase/génétique , Aspartate-ammonia ligase/métabolisme , Cellules HCT116 , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Prolifération cellulaire/génétique , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/métabolisme , Régulation de l'expression des gènes tumoraux/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Hétérogreffes , Facteurs sexuels , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor
16.
Int J Mol Sci ; 25(11)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38892065

RÉSUMÉ

Hormone receptor-positive and HER2-negative breast cancer (HR+/HER2-BC) is the most common type with a favorable prognosis under endocrine therapy. However, it still demonstrates unpredictable progression and recurrences influenced by high tumoral diversity and microenvironmental status. To address these heterogeneous molecular characteristics of HR+/HER2-BC, we aimed to simultaneously characterize its transcriptomic landscape and genetic architecture at the same resolution. Using advanced single-cell RNA and DNA sequencing techniques together, we defined four distinct tumor subtypes. Notably, the migratory tumor subtype was closely linked to genomic alterations of EGFR, related to the tumor-promoting behavior of IL6-positive inflammatory tumor-associated fibroblast, and contributing to poor prognosis. Our study comprehensively utilizes integrated analysis to uncover the complex dynamics of this breast cancer subtype, highlighting the pivotal role of the migratory tumor subtype in influencing surrounding cells. This sheds light on potential therapeutic targets by offering enhanced insights for HR+/HER2-BC treatment.


Sujet(s)
Tumeurs du sein , Fibroblastes associés au cancer , Mouvement cellulaire , Récepteur ErbB-2 , Humains , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Femelle , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Régulation de l'expression des gènes tumoraux , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Microenvironnement tumoral , Lignée cellulaire tumorale , Récepteurs à la progestérone/métabolisme , Récepteurs à la progestérone/génétique , Pronostic , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Analyse sur cellule unique
17.
Cell Mol Biol Lett ; 29(1): 87, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38867189

RÉSUMÉ

BACKGROUND: Alzheimer's disease (AD) is a progressive neurodegenerative disease and apolipoprotein E (APOE) genotypes (APOE2, APOE3, and APOE4) show different AD susceptibility. Previous studies indicated that individuals carrying the APOE2 allele reduce the risk of developing AD, which may be attributed to the potential neuroprotective role of APOE2. However, the mechanisms underlying the protective effects of APOE2 is still unclear. METHODS: We analyzed single-nucleus RNA sequencing and bulk RNA sequencing data of APOE2 and APOE3 carriers from the Religious Orders Study and Memory and Aging Project (ROSMAP) cohort. We validated the findings in SH-SY5Y cells and AD model mice by evaluating mitochondrial functions and cognitive behaviors respectively. RESULTS: The pathway analysis of six major cell types revealed a strong association between APOE2 and cellular stress and energy metabolism, particularly in excitatory and inhibitory neurons, which was found to be more pronounced in the presence of beta-amyloid (Aß). Moreover, APOE2 overexpression alleviates Aß1-42-induced mitochondrial dysfunction and reduces the generation of reactive oxygen species in SH-SY5Y cells. These protective effects may be due to ApoE2 interacting with estrogen-related receptor alpha (ERRα). ERRα overexpression by plasmids or activation by agonist was also found to show similar mitochondrial protective effects in Aß1-42-stimulated SH-SY5Y cells. Additionally, ERRα agonist treatment improve the cognitive performance of Aß injected mice in both Y maze and novel object recognition tests. ERRα agonist treatment increased PSD95 expression in the cortex of agonist-treated-AD mice. CONCLUSIONS: APOE2 appears to enhance neural mitochondrial function via the activation of ERRα signaling, which may be the protective effect of APOE2 to treat AD.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Apolipoprotéine E2 , , Mitochondries , Neurones , Récepteurs des oestrogènes , Transduction du signal , Animaux , Femelle , Humains , Mâle , Souris , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Peptides bêta-amyloïdes/métabolisme , Apolipoprotéine E2/génétique , Apolipoprotéine E2/métabolisme , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Mitochondries/métabolisme , Neurones/métabolisme , Espèces réactives de l'oxygène/métabolisme , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique
18.
Pestic Biochem Physiol ; 202: 105947, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38879334

RÉSUMÉ

Until recently, chemical pesticides were one of the most effective means of controlling agricultural pests; therefore, the search for insecticide targets for agricultural pests has been an ongoing problem. Estrogen-related receptors (ERRs) are transcription factors that regulate cellular metabolism and energy homeostasis in animals. Silkworms are highly sensitive to chemical pesticides, making them ideal models for pesticide screening and evaluation. In this study, we detected ERR expression in key organs involved in pesticide metabolism in silkworms (Bombyx mori), including the fat body and midgut. Using ChIP-seq technology, many estrogen- related response elements were identified in the 2000-bp promoter region upstream of metabolism-related genes, almost all of which were potential ERR target genes. The ERR inhibitor, XCT-790, and the endocrine disruptor, bisphenol A, significantly inhibited expression of the ERR target genes, BmTreh-1, BmTret-1, BmPK, BmPFK, and BmHK, in the fat bodies of silkworms, resulting in pupation difficulties in silkworm larvae that ultimately lead to death. In addition, based on the clarification that the ERR can bind to XCT-790, as observed through biofilm interferometry, its three-dimensional spatial structure was predicted, and using molecular docking techniques, small-molecule compounds with a stronger affinity for the ERR were identified. In summary, utilizing the powerful metabolic regulatory function of ERR in Lepidoptera pests, the developed small molecule inhibitors of ERR can be used for future control of Lepidoptera pests.


Sujet(s)
Bombyx , Simulation de docking moléculaire , Phénols , Récepteurs des oestrogènes , Animaux , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Bombyx/métabolisme , Bombyx/génétique , Bombyx/effets des médicaments et des substances chimiques , Phénols/pharmacologie , Composés benzhydryliques/pharmacologie , Larve/métabolisme , Larve/effets des médicaments et des substances chimiques , Larve/génétique , Insecticides/pharmacologie , Protéines d'insecte/métabolisme , Protéines d'insecte/génétique , Corps gras/métabolisme , Corps gras/effets des médicaments et des substances chimiques , Perturbateurs endocriniens/pharmacologie , Perturbateurs endocriniens/métabolisme , Nitriles , Thiazoles
19.
Oncogene ; 43(31): 2431-2446, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38937602

RÉSUMÉ

Mortalin (encoded by HSPA9) is a mitochondrial chaperone often overexpressed in cancer through as-yet-unknown mechanisms. By searching different RNA-sequencing datasets, we found that ESRRA is a transcription factor highly correlated with HSPA9 in thyroid cancer, especially in follicular, but not C cell-originated, tumors. Consistent with this correlation, ESRRA depletion decreased mortalin expression only in follicular thyroid tumor cells. Further, ESRRA expression and activity were relatively high in thyroid tumors with oncocytic characteristics, wherein ESRRA and mortalin exhibited relatively high functional overlap. Mechanistically, ESRRA directly regulated HSPA9 transcription through a novel ESRRA-responsive element located upstream of the HSPA9 promoter. Physiologically, ESRRA depletion suppressed thyroid tumor cell survival via caspase-dependent apoptosis, which ectopic mortalin expression substantially abrogated. ESRRA depletion also effectively suppressed tumor growth and mortalin expression in the xenografts of oncocytic or ESRRA-overexpressing human thyroid tumor cells in mice. Notably, our Bioinformatics analyses of patient data revealed two ESRRA target gene clusters that contrast oncocytic-like and anaplastic features of follicular thyroid tumors. These findings suggest that ESRRA is a tumor-specific regulator of mortalin expression, the ESRRA-mortalin axis has higher significance in tumors with oncocytic characteristics, and ESRRA target gene networks can refine molecular classification of thyroid cancer.


Sujet(s)
Survie cellulaire , , Régulation de l'expression des gènes tumoraux , Réseaux de régulation génique , Protéines du choc thermique HSP70 , Récepteurs des oestrogènes , Tumeurs de la thyroïde , Humains , Tumeurs de la thyroïde/génétique , Tumeurs de la thyroïde/anatomopathologie , Tumeurs de la thyroïde/métabolisme , Animaux , Souris , Protéines du choc thermique HSP70/génétique , Protéines du choc thermique HSP70/métabolisme , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Lignée cellulaire tumorale , Survie cellulaire/génétique , Apoptose/génétique , Régions promotrices (génétique)/génétique , Protéines mitochondriales
20.
Genes Genomics ; 46(8): 899-907, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38847971

RÉSUMÉ

BACKGROUND: RNA-binding proteins (RBPs) perform various biological functions in humans and are associated with several diseases, including cancer. Therefore, RBPs have emerged as novel therapeutic targets. Although recent investigations have shown that RBPs have crucial functions in breast cancer (BC), detailed research is underway to determine the RBPs that are closely related to cancers. OBJECTIVE: To provide an insight into estrogen receptor (ER) regulation by cold-inducible RNA binding protein (CIRBP) as a novel therapeutic target. RESULTS: By analyzing the genomic data, we identified a potential RBP in BC. We found that CIRBP is highly correlated with ER function and influences clinical outcomes, such as patient survival and endocrine therapy responsiveness. In addition, CIRBP influences the proliferation of BC cells by directly binding to ER-RNA. CONCLUSION: Our results suggest that CIRBP is a novel upstream regulator of ER and that the interplay between CIRBP and ER may be associated with the clinical relevance of BC.


Sujet(s)
Tumeurs du sein , Protéines de liaison à l'ARN , Récepteurs des oestrogènes , Humains , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Femelle , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Prolifération cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Lignée cellulaire tumorale , Cellules MCF-7 , Génomique/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE