Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.770
Filtrar
1.
Front Endocrinol (Lausanne) ; 14: 1152634, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37265700

RESUMEN

Aim: Sepsis is a life-threatening disease with high mortality worldwide. Septic females have lower severity and mortality than the males, suggesting estrogen exerts a protective action, but nothing is known about the role of vascular endothelial estrogen receptor subtypes in this process. In the present study, we aimed to study the estrogen receptors on mesenteric arterioles in normal and sepsis mice and to elucidate the underlying mechanisms. Methods: Sepsis was induced in mice by intraperitoneal injection of LPS. The changes in the expression and release of the serum and cell supernatant proinflammatory cytokines, including TNF-α, IL-1ß and IL-6, were measured by qPCR and ELISA, and the functions of multiple organs were analyzed. The functional activities of mouse mesenteric arterioles were determined by a Mulvany-style wire myograph. The expression of phospholipase C (PLC) and inositol 1,4,5-trisphosphate receptor (IP3R) in endothelial cells were examined by Western blot and their functions were characterized by cell Ca2+ imaging. Results: Septic female mice had higher survival rate than the male mice, and pretreatment with E2 for 5 days significantly improved the survival rate and inhibited proinflammatory cytokines in septic male mice. E2 ameliorated pulmonary, intestinal, hepatic and renal multiple organ injuries in septic male mice; and ER subtypes inhibited proinflammatory cytokines in endothelial cells via PLC/IP3R/Ca2+ pathway. E2/ER subtypes immediately induced endothelial-derived hyperpolarization (EDH)-mediated vasorelaxation via PLC/IP3R/Ca2+ pathway, which was more impaired in septic male mice. E2/ER subtypes could rescue the impaired acetylcholine (ACh)-induced EDH-mediated vasorelaxation in septic male mice. Conclusions: E2 through ER subtypes mediates anti-inflammation and vasorelaxation via genomic and nongenomic actions in sepsis. Mechanistically, activation of endothelial ER subtypes reduces proinflammatory cytokines and induces EDH-mediated vasorelaxation via PLC/IP3R/Ca2+ pathway, leading to amelioration of sepsis-induced organ injury and survival rate.


Asunto(s)
Receptores de Estrógenos , Sepsis , Ratones , Masculino , Femenino , Animales , Receptores de Estrógenos/fisiología , Vasodilatación/fisiología , Células Endoteliales/metabolismo , Citocinas , Genómica , Sepsis/complicaciones , Sepsis/metabolismo
2.
Artículo en Inglés | MEDLINE | ID: mdl-35362389

RESUMEN

Breast cancer, even today, can cause death. Therefore, prevention and early detection are fundamental factors. The mechanisms that favour it are genetic and epigenetic, and seem to play a significant role; also, the microbiota can change estrogen levels and can induce chronic inflammation in the neoplastic site, alternating the balance between proliferation and cell death. Activated steroid hormone receptors induce transcription of genes that encode for proteins involved in cell proliferation and activate another transduction pathway, inducing cell cycle progression and cell migration. These important studies have allowed to develop therapies with selective modulators of estrogen receptors (SERMs), able to block their proliferative and pro-tumorigenic action. Of fundamental importance is also the role played by the microbiota in regulating the metabolism of estrogens and their levels in the blood. There are microbial populations that are able to promote the development of breast cancer, through the production of enzymes responsible for the deconjugation of estrogens, the increase of these in the intestine, subsequent circulation and migration to other locations, such as the udder. Other microbial populations are, instead, able to synthesize estrogen compounds or mimic estrogenic action, and interfere with the metabolism of drugs, affecting the outcome of therapies. The microbial composition of the intestine and hormonal metabolism depend largely on eating habits; the consumption of fats and proteins favours the increase of estrogen in the blood, unlike a diet rich in fiber. Therefore, in-depth knowledge of the microbiota present in the intestine-breast axis could, in the future, encourage the development of new diagnostic and therapeutic approaches to breast cancers.


Asunto(s)
Neoplasias de la Mama , Microbioma Gastrointestinal , Animales , Neoplasias de la Mama/metabolismo , Estrógenos/uso terapéutico , Femenino , Humanos , Receptores de Estrógenos/fisiología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Esteroides/uso terapéutico
3.
PLoS One ; 17(1): e0262389, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35061800

RESUMEN

G protein-coupled estrogen receptor (GPER) was reported to be a potential target in the breast cancer therapy. This study aimed to illuminate the function of GPER and its mediated PI3K/AKT pathway in cryptotanshinone (CPT) inducing cell apoptosis and antiproliferation effect on GPER positive breast cancer MCF-7 cells. Cell proliferation was tested by MTT assay. Apoptosis rates were tested by Annexin V-FITC/PI double staining and the cell cycle was researched by flow cytometry. Autodock vina was applied to make molecular docking between CPT or estradiol and GPER. siRNA technique and GPER specific agonist G-1 or antagonist G-15 were applied to verify the mediated function of GPER. Apoptosis and cell cycle related proteins, as well as the key proteins on PI3K/AKT signaling pathway were detected by western blot. The results indicated that CPT could exert antiproliferation effects by arresting cell cycle in G2/M phase and downregulating the expression of cyclin D, cyclin B and cyclin A. Besides, apoptosis induced by CPT was observed. CPT might be a novel GPER binding compounds. Significantly, suppression of PI3K/AKT signal transduction by CPT was further increased by G-1 and decreased by G-15. The study revealed that the effect of antiproliferation and apoptosis treating with CPT on MCF-7 cells might be through the downregulation of PI3K/AKT pathway mediated by activated GPER.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Fenantrenos/farmacología , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , China , Estrógenos/farmacología , Femenino , Humanos , Células MCF-7 , Fenantrenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Estrógenos/fisiología , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal/efectos de los fármacos
4.
J Gynecol Obstet Hum Reprod ; 51(1): 102242, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34715402

RESUMEN

BACKGROUND: The aim of this study was to evaluate the impact of Lymphovascular Space Invasion (LVSI) on Overall Survival (OS) and Recurrence-Free Survival (RFS) in patients managed for high-grade serous epithelial ovarian cancer (HGSOC). MATERIALS AND METHODS: Retrospective multicenter study by the FRANCOGYN research group between January 2001 and December 2018. All patients managed for HGSOC and for whom histological slides for the review of LVSI were available, were included. The characteristics of patients with LVSI (LVSI group) were compared to those without LVSI (No LVSI group). A Cox analysis for OS and RFS analysis was performed in all populations. RESULTS: Over the study period, 410 patients were included in the thirteen institutions. Among them, 289 patients had LVSI (33.9%). LVSI was an independent predictive factor for poorer Overall and Recurrence-Free Survival. LVSI affected OS (p<0.001) and RFS (p<0.001), Association of LVSI status and estrogen receptor status (ER) also affected OS and RFS (p = 0.04; p = 0.04 respectively). CONCLUSION: The presence of LVSI in HGSOC has an impact on OS and RFS and should be routinely included in the pathology examination along with ER status.


Asunto(s)
Neoplasias Ováricas/fisiopatología , Receptores de Estrógenos/clasificación , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Francia , Humanos , Estimación de Kaplan-Meier , Metástasis Linfática/patología , Persona de Mediana Edad , Neoplasias Ováricas/complicaciones , Neoplasias Ováricas/patología , Pronóstico , Receptores de Estrógenos/fisiología , Estudios Retrospectivos
5.
Cancer Lett ; 523: 82-99, 2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-34610415

RESUMEN

Many breast cancer patients harbor high estrogen receptor (ER) expression in tumors that can be treated with endocrine therapy, which includes aromatase inhibitors (AI); unfortunately, resistance often occurs. Mitochondrial dysfunction has been thought to contribute to progression and to be related to hormone receptor expression in breast tumors. Mitochondrial alterations in AI-resistant breast cancer have not yet been defined. In this study, we characterized mitochondrial alterations and their roles in AI resistance. MCF-7aro AI-resistant breast cancer cells were shown to have significant changes in mitochondria. Low expressions of mitochondrial genes and proteins could be poor prognostic factors for breast cancer patients. Long-term mitochondrial inhibitor treatments-mediated mitochondrial stress adaptation could induce letrozole resistance. ERα-amphiregulin (AREG) loop signaling was activated and contributed to mitochondrial stress adaptation-mediated letrozole resistance. The up-regulation of AREG-epidermal growth factor receptor (EGFR) crosstalk activated the PI3K/Akt/mTOR and ERK pathways and was responsible for ERα activation. Moreover, mitochondrial stress adaptation-increased intracellular levels of reactive oxygen species (ROS) and calcium were shown to induce AREG expression and secretion. In conclusion, our results support the claim that mitochondrial stress adaptation contributes to AI resistance via ROS/calcium-mediated AREG-ERα loop signaling and provide possible treatment targets for overcoming AI resistance.


Asunto(s)
Anfirregulina/fisiología , Inhibidores de la Aromatasa/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Calcio/metabolismo , Mitocondrias/fisiología , Especies Reactivas de Oxígeno/metabolismo , Receptores de Estrógenos/fisiología , Inhibidores de la Aromatasa/uso terapéutico , Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos , Femenino , Humanos , Letrozol/farmacología , Sistema de Señalización de MAP Quinasas , Células MCF-7 , Mitocondrias/efectos de los fármacos , Transducción de Señal/fisiología
6.
Life Sci ; 286: 120029, 2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-34634322

RESUMEN

Cisplatin is a platinum-based chemotherapeutic drug used in the standard treatment of various solid cancers including testicular, bladder, head and neck, cervical and ovarian cancer. Although successful clinical responses are observed in patients following initial cisplatin treatment, resistance to cisplatin ultimately develops in most patients, leading to therapeutic failure. Multiple molecular mechanisms contributing to cisplatin resistance in cancer cells have been identified to date. In this review, we discuss the effect of estrogen, estrogen receptors (ERs) and estrogen-related receptors (ERRs) on cisplatin resistance in various cancer types. We highlight that estrogen treatment or increased expression of ERs or ERRs are generally associated with higher cisplatin resistance in cancer in vitro, mostly due to decreased caspase activity, increased anti-apoptotic protein levels such as BCL-2, higher drug efflux and higher levels of antioxidant enzymes. Targeted inhibition of ERs or estrogen production in combination with cisplatin treatment thus can be a useful strategy to overcome chemoresistance in certain cancer types. Estrogen levels and ER status can also be considered to identify cancer patients with a high potential of therapy response against cisplatin. A better mechanistic understanding of the involvement of estrogen, ERs and ERRs in the development of cisplatin resistance is needed to improve the management of cancer treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos/fisiología , Neoplasias/tratamiento farmacológico , Receptores de Estrógenos/fisiología , Femenino , Humanos
7.
Reprod Fertil Dev ; 33(12): 736-745, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34602123

RESUMEN

The long-term storage of spermatozoa in the female reproductive tract is limited by the innate immune system. Oestrogen plays a role in regulating the innate immune system. Thus, exploring the expression of genes in the Toll-like receptor (TLR) 2/4 pathway and oestrogen receptors in the oviduct of Mauremys reevesii could contribute to our understanding of the mechanism of sperm storage. In this study, three parts of the oviduct (isthmus, uterus and vagina) in three mated and unmated female turtles were used to perform immunohistochemistry and real-time quantitative polymerase chain reaction (qPCR). Immunohistochemistry revealed that the TLR2/4 protein was mainly distributed in epithelial tissues and glandular cell membranes, and that TLR2/4 levels in the oviduct were significantly decreased in mated compared with unmated turtles. Real-time qPCR indicated that TLR2/4, myeloid differentiation factor 88 (MyD88), interleukin 1 receptor associated kinase 4 (IRAK4), TNF receptor associated factor 6 (TRAF6), interferon regulatory factor 3 (IRF3) and interleukin 6 (IL6) mRNA expression was significantly higher in the oviduct of unmated than mated turtles, whereas the opposite was true for the expression of oestrogen receptor 1 (ESR1) and progesterone receptor (PGR). These results indicate that when spermatozoa are stored in the oviduct, an increase in oestrogen suppresses the immune response induced by the TLR2/4 pathway so that spermatozoa are not removed as a foreign substance, but stored until fertilisation. The findings of this study are relevant to our understanding of the relationship between sperm storage and the innate immune system in the oviduct of reptiles.


Asunto(s)
Inmunidad Innata/fisiología , Oviductos/metabolismo , Receptores de Estrógenos/fisiología , Espermatozoides/fisiología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Femenino , Masculino , Tortugas
8.
APMIS ; 129(12): 694-699, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34582047

RESUMEN

Urinary bladder cancer is worldwide one of the most diagnosed and costly types of cancer. One puzzle in the bladder cancer diagnosis is the disproportional relationship between genders. Males are more likely to be diagnosed with bladder cancer whereas females typically are diagnosed with more adverse disease and worse prognosis, which has led to speculation of the potential role of sex hormones and their receptors in this disease. Estrogen receptors are present in the human bladder, and their role in bladder cancer oncogenesis is increasingly becoming a focus for researchers around the world. This mini-review aims to give a brief overview of the status of female bladder cancer, and to which extend the sex hormones receptors play a role in this. A literature search was performed and included all female original studies on bladder cancer and hormone receptors. Estrogen-receptor alpha seems to be anti-oncogenic whereas estrogen-receptor beta is exhibiting its function pro-oncogenic. The receptor functions may be exercised through mRNA transcriptions and enzymes. Epidemiological studies indicate a potential increase in incidence of bladder cancer for females with earlier age at menopause, and clinical trials are investigating Tamoxifen as a potential treatment in bladder cancer. Increasing evidence supports the theory of bladder cancer development and progression as being partly hormone-dependent. This can lead to a change in conceptual background of bladder cancer etiology and development in the future. Further studies are required to more precise map the use of anti-hormonal drugs in the treatment of this cancer.


Asunto(s)
Neoplasias de la Vejiga Urinaria/etiología , Antagonistas de Estrógenos/uso terapéutico , Receptor alfa de Estrógeno/fisiología , Glucuronosiltransferasa/fisiología , Hormonas Esteroides Gonadales/fisiología , Disparidades en el Estado de Salud , Humanos , Receptores de Estrógenos/fisiología , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Receptor Relacionado con Estrógeno ERRalfa
9.
Int J Oncol ; 59(4)2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34476497

RESUMEN

Pyroptosis is mediated by gasdermins and serves a critical role in ionizing radiation (IR)­induced damage in normal tissues, but its role in cancer radiotherapy and underlying mechanisms remains unclear. Long non­coding (lnc) RNAs serve important roles in regulating the radiosensitivity of cancer cells. The present study aimed to investigate the mechanistic involvement of lncRNAs in IR­induced pyroptosis in human colorectal cancer HCT116 cells. LncRNA, microRNA (miR)­448 and gasdermin E (GSDME) levels were evaluated using reverse transcription­quantitative polymerase chain reaction. Protein expression and activation of gasdermins were measured using western blotting. The binding association between miR­448 and GSDME was assessed using the dual­luciferase reporter assay. Pyroptosis was examined using phase­contrast microscopy, flow cytometry, Cell Counting Kit­8 assay and lactate dehydrogenase release assay. IR dose­dependently induced GSDME­mediated pyroptosis in HCT116 cells. GSDME was identified as a downstream target of miR­448. LncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) was upregulated in response to IR and enhanced GSDME expression by negatively regulating miR­448 expression. Notably, NEAT1 knockdown suppressed IR­induced pyroptosis, full­length GSDME expression and GSDME cleavage compared with that in irradiated cells. In addition, NEAT1 knockdown rescued the IR­induced decrease in cell viability in HCT116 cells. The findings of the present study indicated that lncRNA NEAT1 modulates IR­induced pyroptosis and viability in HCT116 cells via miR­448 by regulating the expression, but not activation of GSDME. The present study provides crucial mechanistic insight into the potential role of lncRNA NEAT1 in IR­induced pyroptosis.


Asunto(s)
Neoplasias Colorrectales/patología , MicroARNs/fisiología , Piroptosis/efectos de la radiación , ARN Largo no Codificante/fisiología , Receptores de Estrógenos/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/radioterapia , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Humanos , Tolerancia a Radiación , Receptores de Estrógenos/fisiología
10.
Int J Mol Sci ; 22(15)2021 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-34360813

RESUMEN

Proper cardiac function depends on the coordinated expression of multiple gene networks related to fuel utilization and mitochondrial ATP production, heart contraction, and ion transport. Key transcriptional regulators that regulate these gene networks have been identified. Among them, estrogen-related receptors (ERRs) have emerged as crucial modulators of cardiac function by regulating cellular metabolism and contraction machinery. Consistent with this role, lack of ERRα or ERRγ results in cardiac derangements that lead to functional maladaptation in response to increased workload. Interestingly, metabolic inflexibility associated with diabetic cardiomyopathy has been recently associated with increased mitochondrial fatty acid oxidation and expression of ERRγ, suggesting that sustained expression of this nuclear receptor could result in a cardiac pathogenic outcome. Here, we describe the generation of mice with cardiac-specific overexpression of ERRγ, which die at young ages due to heart failure. ERRγ transgenic mice show signs of dilated cardiomyopathy associated with cardiomyocyte hypertrophy, increased cell death, and fibrosis. Our results suggest that ERRγ could play a role in mediating cardiac pathogenic responses.


Asunto(s)
Cardiomiopatía Dilatada/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Receptores de Estrógenos/fisiología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocardio/patología , Miocitos Cardíacos/patología
11.
FASEB J ; 35(8): e21757, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34233045

RESUMEN

Pyroptosis and intrinsic apoptosis are two forms of regulated cell death driven by active caspases where plasma membrane permeabilization is induced by gasdermin pores. Caspase-1 induces gasdermin D pore formation during pyroptosis, whereas caspase-3 promotes gasdermin E pore formation during apoptosis. These two types of cell death are accompanied by mitochondrial outer membrane permeabilization due to BAK/BAX pore formation in the external membrane of mitochondria, and to some extent, this complex also affects the inner mitochondrial membrane facilitating mitochondrial DNA relocalization from the matrix to the cytosol. However, the detailed mechanism responsible for this process has not been investigated. Herein, we reported that gasdermin processing is required to induce mitochondrial DNA release from cells during pyroptosis and apoptosis. Gasdermin targeted at the plasma membrane promotes a fast mitochondrial collapse along with the initial accumulation of mitochondrial DNA in the cytosol and then facilitates the DNA's release from the cell when the plasma membrane ruptures. These findings demonstrate that gasdermin action has a critical effect on the plasma membrane and facilitates the release of mitochondrial DNA as a damage-associated molecular pattern.


Asunto(s)
Apoptosis/fisiología , ADN Mitocondrial/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de Unión a Fosfato/fisiología , Piroptosis/fisiología , Animales , Caspasas/metabolismo , Membrana Celular/metabolismo , Células HEK293 , Humanos , Técnicas In Vitro , Inflamasomas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas de Unión a Fosfato/deficiencia , Proteínas de Unión a Fosfato/genética , Pirina/metabolismo , Receptores de Estrógenos/fisiología
12.
Int J Mol Sci ; 22(14)2021 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-34299224

RESUMEN

Inflammation is important for the initiation and progression of breast cancer. We have previously reported that in monocytes, estrogen regulates TLR4/NFκB-mediated inflammation via the interaction of the Erα isoform ERα36 with GPER1. We therefore investigated whether a similar mechanism is present in breast cancer epithelial cells, and the effect of ERα36 expression on the classic 66 kD ERα isoform (ERα66) functions. We report that estrogen inhibits LPS-induced NFκB activity and the expression of downstream molecules TNFα and IL-6. In the absence of ERα66, ERα36 and GPER1 are both indispensable for this effect. In the presence of ERα66, ERα36 or GPER1 knock-down partially inhibits NFκB-mediated inflammation. In both cases, ERα36 overexpression enhances the inhibitory effect of estrogen on inflammation. We also verify that ERα36 and GPER1 physically interact, especially after LPS treatment, and that GPER1 interacts directly with NFκB. When both ERα66 and ERα36 are expressed, the latter acts as an inhibitor of ERα66 via its binding to estrogen response elements. We also report that the activation of ERα36 leads to the inhibition of breast cancer cell proliferation. Our data support that ERα36 is an inhibitory estrogen receptor that, in collaboration with GPER1, inhibits NFκB-mediated inflammation and ERα66 actions in breast cancer cells.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Neoplasias de la Mama , Línea Celular Tumoral , Células Epiteliales/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/fisiología , Estrógenos/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/metabolismo , Interleucina-6/metabolismo , Células MCF-7 , Monocitos/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Receptores de Estrógenos/fisiología , Receptores Acoplados a Proteínas G/fisiología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
13.
Physiol Rep ; 9(14): e14948, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34288542

RESUMEN

Estradiol (E2) inhibits fluid intake in several species, which may help to defend fluid homeostasis by preventing excessive extracellular fluid volume. Although this phenomenon is well established using the rat model, it has only been studied directly in young adults. Because aging influences the neuronal sensitivity to E2 and the fluid intake effects of E2 are mediated in the brain, we tested the hypothesis that aging influences the fluid intake effects of E2 in female rats. To do so, we examined water and NaCl intake in addition to the pressor effect after central angiotensin II treatment in young (3-4 months), middle-aged (10-12 months), and old (16-18 months) ovariectomized rats treated with estradiol benzoate (EB). As expected, EB treatment reduced water and NaCl intake in young rats. EB treatment, however, did not reduce water intake in old rats, nor did it reduce NaCl intake in middle-aged or old rats. The ability of EB to reduce blood pressure was, in contrast, observed in all three age groups. Next, we also measured the gene expression of estrogen receptors (ERs) and the angiotensin type 1 receptor (AT1R) in the areas of the brain that control fluid balance. ERß, G protein estrogen receptor (GPER), and AT1R were reduced in the paraventricular nucleus of the hypothalamus in middle-aged and old rats, compared to young rats. These results suggest the estrogenic control of fluid intake is modified by age. Older animals lost the fluid intake effects of E2, which correlated with decreased ER and AT1R expression in the hypothalamus.


Asunto(s)
Envejecimiento/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Ingestión de Líquidos/efectos de los fármacos , Estradiol/análogos & derivados , Frecuencia Cardíaca/efectos de los fármacos , Envejecimiento/fisiología , Animales , Presión Sanguínea/fisiología , Ingestión de Líquidos/fisiología , Estradiol/administración & dosificación , Femenino , Frecuencia Cardíaca/fisiología , Ovariectomía/efectos adversos , Ovariectomía/tendencias , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/fisiología , Receptores de Estrógenos/fisiología
14.
Nat Commun ; 12(1): 3596, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34155205

RESUMEN

One of the earliest maturation steps in cardiomyocytes (CMs) is the sarcomere protein isoform switch between TNNI1 and TNNI3 (fetal and neonatal/adult troponin I). Here, we generate human induced pluripotent stem cells (hiPSCs) carrying a TNNI1EmGFP and TNNI3mCherry double reporter to monitor and isolate mature sub-populations during cardiac differentiation. Extensive drug screening identifies two compounds, an estrogen-related receptor gamma (ERRγ) agonist and an S-phase kinase-associated protein 2 inhibitor, that enhances cardiac maturation and a significant change to TNNI3 expression. Expression, morphological, functional, and molecular analyses indicate that hiPSC-CMs treated with the ERRγ agonist show a larger cell size, longer sarcomere length, the presence of transverse tubules, and enhanced metabolic function and contractile and electrical properties. Here, we show that ERRγ-treated hiPSC-CMs have a mature cellular property consistent with neonatal CMs and are useful for disease modeling and regenerative medicine.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Receptores de Estrógenos/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Modelos Biológicos , Miocitos Cardíacos/metabolismo , Receptores de Estrógenos/química , Proteínas Quinasas Asociadas a Fase-S/antagonistas & inhibidores , Sarcolema/efectos de los fármacos , Sarcolema/metabolismo , Sarcómeros/efectos de los fármacos , Sarcómeros/metabolismo , Transcriptoma/efectos de los fármacos , Troponina I/genética , Troponina I/metabolismo
15.
J Clin Endocrinol Metab ; 106(9): 2547-2565, 2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-34058008

RESUMEN

CONTEXT: Suboptimal endometrial thickening is associated with lower pregnancy rates and occurs in some infertile women treated with clomiphene. OBJECTIVE: To examine cellular and molecular differences in the endometrium of women with suboptimal vs optimal endometrial thickening following clomiphene. METHODS: Translational prospective cohort study from 2018 to 2020 at a university-affiliated clinic. Reproductive age women with unexplained infertility treated with 100 mg of clomiphene on cycle days 3 to 7 who developed optimal (≥8mm; n = 6, controls) or suboptimal (<6mm; n = 7, subjects) endometrial thickness underwent preovulatory blood and endometrial sampling. The main outcome measures were endometrial tissue architecture, abundance and location of specific proteins, RNA expression, and estrogen receptor (ER) α binding. RESULTS: The endometrium of suboptimal subjects compared with optimal controls was characterized by a reduced volume of glandular epithelium (16% vs 24%, P = .01), decreased immunostaining of markers of proliferation (PCNA, ki67) and angiogenesis (PECAM-1), increased immunostaining of pan-leukocyte marker CD45 and ERß, but decreased ERα immunostaining (all P < .05). RNA-seq identified 398 differentially expressed genes between groups. Pathway analysis of differentially expressed genes indicated reduced proliferation (Z-score = -2.2, P < .01), decreased angiogenesis (Z-score = -2.87, P < .001), increased inflammation (Z-score = +2.2, P < .01), and ERß activation (Z-score = +1.6, P < .001) in suboptimal subjects. ChIP-seq identified 6 genes bound by ERα that were differentially expressed between groups (P < .01), some of which may play a role in implantation. CONCLUSION: Women with suboptimal endometrial thickness after clomiphene exhibit aberrant ER expression patterns, architectural changes, and altered gene and protein expression suggesting reduced proliferation and angiogenesis in the setting of increased inflammation.


Asunto(s)
Clomifeno/efectos adversos , Endometrio/efectos de los fármacos , Receptores de Estrógenos/fisiología , Adulto , Proliferación Celular/efectos de los fármacos , Endometrio/patología , Estrógenos/fisiología , Femenino , Hormonas Esteroides Gonadales/sangre , Humanos , Receptores de Estrógenos/análisis
16.
Front Endocrinol (Lausanne) ; 12: 643870, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33796076

RESUMEN

Epidemiological data have indicated that there are some sex-related differences in bladder cancer. Indeed, the incidence of bladder cancer in men has been substantially higher than that in women throughout the world, while women tend to have higher stage disease and poorer prognosis. These gender disparities have prompted to investigate sex hormones and their cognitive receptors in bladder cancer. Specifically, estrogen receptors, including estrogen receptor-α and estrogen receptor-ß, have been shown to contribute to urothelial carcinogenesis and cancer progression, as well as to modulating chemosensitivity in bladder cancer, although conflicting findings exist. Meanwhile, immunohistochemical studies in surgical specimens have assessed the expression of estrogen receptors and related proteins as well as its associations with clinicopathologic features of bladder cancer and patient outcomes. This review article summarizes and discusses available data indicating that estrogen receptor signaling plays an important role in urothelial cancer.


Asunto(s)
Receptores de Estrógenos/fisiología , Neoplasias de la Vejiga Urinaria/metabolismo , Urotelio/patología , Animales , Cognición , Progresión de la Enfermedad , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Estrógenos/metabolismo , Femenino , Humanos , Inmunohistoquímica , Masculino , Ratones , Pronóstico , Ratas , Factores Sexuales , Resultado del Tratamiento
17.
Front Endocrinol (Lausanne) ; 12: 578250, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815268

RESUMEN

Persistent inflammation and persistent pain are major medical, social and economic burdens. As such, related pharmacotherapy needs to be continuously improved. The peptide ERα17p, which originates from a part of the hinge region/AF2 domain of the human estrogen receptor α (ERα), exerts anti-proliferative effects in breast cancer cells through a mechanism involving the hepta-transmembrane G protein-coupled estrogen receptor (GPER). It is able to decrease the size of xenografted human breast tumors, in mice. As GPER has been reported to participate in pain and inflammation, we were interested in exploring the potential of ERα17p in this respect. We observed that the peptide promoted anti-hyperalgesic effects from 2.5 mg/kg in a chronic mice model of paw inflammation induced by the pro-inflammatory complete Freund's adjuvant (CFA). This action was abrogated by the specific GPER antagonist G-15, leading to the conclusion that a GPER-dependent mechanism was involved. A systemic administration of a Cy5-labeled version of the peptide allowed its detection in both, the spinal cord and brain. However, ERα17p-induced anti-hyperalgesia was detected at the supraspinal level, exclusively. In the second part of the study, we have assessed the anti-inflammatory action of ERα17p in mice using a carrageenan-evoked hind-paw inflammation model. A systemic administration of ERα17p at a dose of 2.5 mg/kg was responsible for reduced paw swelling. Overall, our work strongly suggests that GPER inverse agonists, including ERα17p, could be used to control hyperalgesia and inflammation.


Asunto(s)
Receptor alfa de Estrógeno/química , Fragmentos de Péptidos/farmacología , Receptores de Estrógenos/fisiología , Receptores Acoplados a Proteínas G/fisiología , Anestésicos/farmacología , Animales , Antiinflamatorios/farmacología , Antineoplásicos/farmacología , Artritis Experimental/inducido químicamente , Artritis Experimental/genética , Artritis Experimental/metabolismo , Artritis Experimental/patología , Agonismo Inverso de Drogas , Femenino , Adyuvante de Freund , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/genética , Hiperalgesia/metabolismo , Hiperalgesia/patología , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/metabolismo , Masculino , Ratones , Receptores Acoplados a Proteínas G/agonistas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
18.
Transl Res ; 234: 31-42, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33567346

RESUMEN

Nociception and opioid antinociception in females are pliable processes, varying qualitatively and quantitatively over the reproductive cycle. Spinal estrogenic signaling via membrane estrogen receptors (mERs), in combination with multiple other signaling molecules [spinal dynorphin, kappa-opioid receptors (KOR), glutamate and metabotropic glutamate receptor 1 (mGluR1)], appears to function as a master coordinator, parsing functionality between pronociception and antinociception. This provides a window into pharmacologically accessing intrinsic opioid analgesic/anti-allodynic systems. In diestrus, membrane estrogen receptor alpha (mERα) signals via mGluR1 to suppress spinal endomorphin 2 (EM2) analgesia. Strikingly, in the absence of exogenous opioids, interfering with this suppression in a chronic pain model elicits opioid anti-allodynia, revealing contributions of endogenous opioid(s). In proestrus, robust spinal EM2 analgesia is manifest but this requires spinal dynorphin/KOR and glutamate-activated mGluR1. Furthermore, spinal mGluR1 blockade in a proestrus chronic pain animal (eliminating spinal EM2 analgesia) exacerbates mechanical allodynia, revealing tempering by endogenous opioid(s). A complex containing mu-opioid receptor, KOR, aromatase, mGluRs, and mERα are foundational to eliciting endogenous opioid anti-allodynia. Aromatase-mERα oligomers are also plentiful, in a central nervous system region-specific fashion. These can be independently regulated and allow estrogens to act intracellularly within the same signaling complex in which they are synthesized, explaining asynchronous relationships between circulating estrogens and central nervous system estrogen functionalities. Observations with EM2 highlight the translational relevance of extensively characterizing exogenous responsiveness to endogenous opioids and the neuronal circuits that mediate them along with the multiplicity of estrogenic systems that concomitantly function in phase and out-of-phase with the reproductive cycle.


Asunto(s)
Analgesia , Estrógenos/fisiología , Glutamatos/fisiología , Nocicepción/fisiología , Péptidos Opioides/fisiología , Analgesia/métodos , Analgésicos/farmacología , Animales , Sistema Nervioso Central/fisiología , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/fisiopatología , Femenino , Humanos , Masculino , Modelos Neurológicos , Receptores de Estrógenos/fisiología , Receptores de Glutamato/fisiología , Receptores Opioides/fisiología , Investigación Biomédica Traslacional
19.
Domest Anim Endocrinol ; 74: 106527, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32799038

RESUMEN

The steroid hormones 17-ß estradiol (E2) and progesterone (P4) can regulate capacitation, hyperactive motility, and the acrosome reaction (AR) during the sperm transit through the female tract. Moreover, exogenous P4 and E2 can induce the AR in ovine spermatozoa, and progesterone receptor (PR) and estrogen receptors (ERα and ERß) are present in these cells. Thus, to investigate whether the effects both steroid hormones in ram sperm capacitation and AR are receptor-mediated, we incubated them with receptor agonists (tanaproget 1 µM and 5 µM for PR or resveratrol 5 µM and 10 µM for ER) or antagonists (mifepristone 4 µM and 40 µM for PR or tamoxifen 5 µM and 10 µM for ER) in capacitating conditions. The addition of receptor modulators did not affect sperm viability or total motility, although changes in progressive motility were detected. The incubation with both receptor agonists increased the percentage of acrosome-reacted spermatozoa, evaluated by chlortetracycline staining, when compared with the capacitated nontreated sample (Cap-C, P < 0.001). Moreover, the ER agonist resveratrol 10 µM provoked a greater AR than E2 (P < 0.01). Furthermore, the incubation with the receptor antagonists prevented the induction of the AR by P4 or E2, as the antagonists-treated spermatozoa presented a similar CTC pattern to that of Cap-C. In conclusion, these results confirm that P4 and E2 can induce the AR in ram spermatozoa and that this effect is receptor-mediated.


Asunto(s)
Reacción Acrosómica/efectos de los fármacos , Reacción Acrosómica/fisiología , Receptores de Estrógenos/fisiología , Receptores de Progesterona/fisiología , Espermatozoides/fisiología , Animales , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Supervivencia Celular , Células Cultivadas , Estradiol/farmacología , Estrógenos/farmacología , Masculino , Progesterona/farmacología , Receptores de Estrógenos/antagonistas & inhibidores , Resveratrol/farmacología , Ovinos , Capacitación Espermática/efectos de los fármacos , Capacitación Espermática/fisiología , Motilidad Espermática , Tamoxifeno/farmacología
20.
J Biochem Mol Toxicol ; 35(2): e22641, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32996202

RESUMEN

Retinal vascularization is arrested at the early (hyperoxia) stage in retinopathy of prematurity (ROP), a leading cause of blindness in children. Estrogen was reported to alleviate ROP by inhibiting reactive oxygen species, the upstream signaling molecules of endoplasmic reticulum stress (ERS). Astrocytes have long been proposed to guide angiogenesis, because they form a reticular network that provides a substrate for migrating endothelial cells. However, the factors that control the vascularization of the immature retina and the therapeutic mechanism of estrogen in early ROP remain poorly understood. This study aimed to investigate the role of G-protein-coupled estrogen receptor (GPER), an estrogen receptor distributed in the endoplasmic reticulum (ER), in protecting retinal astrocytes under hyperoxia and the association with ERS. The results showed that GPER was widely expressed in retinal astrocytes. GPER activation increases cell viability, decreases apoptosis, and autophagy of retinal astrocytes, decreases inositol-1,4,5-triphosphate receptor activity, and increases Ca2+ concentration in ER of astrocytes under hyperoxia. GPER blockade reversed all of these changes. Together, our findings indicate that GPER can protect the survival of retinal astrocytes by inhibiting ERS under hyperoxia.


Asunto(s)
Astrocitos/metabolismo , Estrés del Retículo Endoplásmico , Estrógenos/fisiología , Hiperoxia/metabolismo , Receptores de Estrógenos/fisiología , Receptores Acoplados a Proteínas G/fisiología , Retina/metabolismo , Retinopatía de la Prematuridad/metabolismo , Animales , Apoptosis , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Retina/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA