Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(17)2024 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-39273151

RESUMEN

Gastric cancer is one of the most common cancers worldwide, and new therapeutic strategies are urgently needed. Ferroptosis is an intracellular iron-dependent cell death induced by the accumulation of lipid peroxidation, a mechanism different from conventional apoptosis and necrosis. Therefore, induction of ferroptosis is expected to be a new therapeutic strategy. Glutathione peroxidase 4 (GPX4) and ferroptosis suppressor protein 1 (FSP1) have been identified as the major inhibitors of ferroptosis. Herein, we performed immunohistochemistry for GPX4, FSP1, and 4-HNE using tissues from patients with gastric cancer and investigated the relationship between these factors and prognosis. Patients with high GPX4 expression or high GPX4 expression and low 4-HNE accumulation tended to have a poor prognosis (p = 0.036, 0.023), whereas those with low FSP1 expression and high 4-HNE accumulation had a good prognosis (p = 0.033). The synergistic induction of cell death by inhibiting GPX4 and FSP1 in vitro was also observed, indicating that the cell death was non-apoptotic. Our results indicate that the expression and accumulation of lipid peroxidation-related factors play an important role in the clinicopathological significance of gastric cancer and that novel therapeutic strategies targeting GPX4 and FSP1 may be effective in treating patients with gastric cancer who have poor prognosis.


Asunto(s)
Biomarcadores de Tumor , Ferroptosis , Peroxidación de Lípido , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Neoplasias Gástricas , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Humanos , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Pronóstico , Femenino , Masculino , Biomarcadores de Tumor/metabolismo , Anciano , Persona de Mediana Edad , Ferroptosis/efectos de los fármacos , Línea Celular Tumoral , Aldehídos/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/genética
2.
Mol Biol Evol ; 39(9)2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35959649

RESUMEN

The emergence of the placenta is a revolutionary event in the evolution of therian mammals, to which some LTR retroelement-derived genes, such as PEG10, RTL1, and syncytin, are known to contribute. However, therian genomes contain many more LTR retroelement-derived genes that may also have contributed to placental evolution. We conducted large-scale evolutionary genomic and transcriptomic analyses to comprehensively search for LTR retroelement-derived genes whose origination coincided with therian placental emergence and that became consistently expressed in therian placentae. We identified NYNRIN as another Ty3/Gypsy LTR retroelement-derived gene likely to contribute to placental emergence in the therian stem lineage. NYNRIN knockdown inhibited the invasion of HTR8/SVneo invasive-type trophoblasts, whereas the knockdown of its nonretroelement-derived homolog KHNYN did not. Functional enrichment analyses suggested that NYNRIN modulates trophoblast invasion by regulating epithelial-mesenchymal transition and extracellular matrix remodeling and that the ubiquitin-proteasome system is responsible for the functional differences between NYNRIN and KHNYN. These findings extend our knowledge of the roles of LTR retroelement-derived genes in the evolution of therian mammals.


Asunto(s)
Placenta , Retroelementos , Animales , Femenino , Genoma , Mamíferos/genética , Embarazo , Retroelementos/genética , Trofoblastos
3.
J Pharmacol Sci ; 153(4): 188-196, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37973216

RESUMEN

Human endometrial stromal cells (ESCs) undergo differentiation, known as decidualization, and endometrial epithelial cells mature around the embryo implantation stage. In the uterus, cyclooxygenase 2 (COX2), the rate-limiting enzyme that produces prostaglandin E2, is expressed in endometrial stromal and epithelial cells, and promotes decidualization of the former cells. Our recent study demonstrated that progesterone receptor membrane component 1 (PGRMC1) is downregulated during decidualization and may be involved in cellular senescence associated with decidualization via the transcription factor forkhead box protein O1 (FOXO1). Therefore, we investigated the role of PGRMC1 in COX2 expression during differentiation and maturation of endometrial stromal and epithelial cells. Inhibition or knockdown of PGRMC1 significantly enhanced differentiation stimuli-induced COX2 expression in both cell types. However, this COX2 expression was suppressed by FOXO1 knockdown or nuclear factor-kappa B (NF-κB) inhibition. Silencing of COX2 expression inhibited PGRMC1 knockdown-induced expression of decidual markers in ESCs. Thus, PGRMC1 may be linked to FOXO1- and NF-κB-mediated COX2 expression in endometrial cells. Taken together, our data suggest that downregulation of PGRMC1 expression facilitates differentiation of endometrial cells, i.e., decidualization and glandular maturation, via upregulation of COX2 expression.


Asunto(s)
Decidua , FN-kappa B , Femenino , Humanos , AMP Cíclico/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Decidua/metabolismo , Endometrio , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , FN-kappa B/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
4.
Reprod Med Biol ; 22(1): e12537, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37614815

RESUMEN

Purpose: Extravillous trophoblasts (EVTs) invade the endometrium to establish a fetomaternal interaction during pregnancy. Epidermal growth factor (EGF) and heparin-binding EGF-like growth factor (HB-EGF) stimulate EVT invasion by binding to the EGF receptor (EGFR). We examined the role of the small GTP-binding protein Rap1 in EGF- and HB-EGF-stimulated EVT invasion. Methods: Expression of Rap1 in the first-trimester placenta was examined by immunohistochemistry. Effect of EGF or HB-EGF on Rap1 activation (GTP-Rap1) and Rap1 knockdown on invasion was assessed in EVT cell line (HTR-8/SVneo). In addition, effect of Rap1 knockdown and Rap1GAP (a Rap1 inactivator) overexpression on the activation of EGF signaling and EGFR expression were examined. Results: Rap1 was expressed by EVTs, villous cytotrophoblasts, and syncytiotrophoblasts in the placenta. EGF and HB-EGF activated Rap1 and promoted invasion of HTR-8/SVneo, and these effects were inhibited by Rap1 knockdown. The EGF- and HB-EGF-induced phosphorylation of AKT, ERK1/2, p38MAPK, and Src was inhibited by Rap1 knockdown. Furthermore, the knockdown of Rap1 reduced the EGFR protein level. Overexpression of Rap1GAP repressed EGF- and HB-EGF-induced Rap1 activation and reduced EGFR expression. Conclusion: Rap1 may function as a mediator of EGF and HB-EGF signaling pathways and can modulate EGFR expression in EVTs during placental development.

5.
J Pharmacol Sci ; 150(4): 259-266, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36344048

RESUMEN

Uterine leiomyosarcoma is an aggressive soft tissue tumor. Stathmin, a phosphoprotein that modulates microtubule dynamics, is highly expressed in many malignancies including leiomyosarcoma. The microtubule-depolymerizing agent eribulin has been recently approved for treating malignant soft tissue tumors. Although eribulin inhibits microtubule polymerization, little is known about the relationship between eribulin treatment and stathmin dynamics. In this study, we explored the role of stathmin expression in the action of eribulin in leiomyosarcoma cells. Eribulin induced phosphorylation of stathmin and reduced expression of subunits A and C of protein phosphatase 2A (PP2A) in a leiomyosarcoma cell line. The PP2A activator FTY720 reduced levels of phosphorylated stathmin. Eribulin decreased stathmin protein levels without affecting stathmin mRNA expression. Furthermore, stathmin knockdown attenuated the inhibitory effects of eribulin on cell viability, whereas stathmin overexpression enhanced the anti-proliferative effect of eribulin. Eribulin-resistant leiomyosarcoma cell lines had enhanced expression of the class Ⅰ ß-tubulin TUBB1, multi-drug resistance 1 protein MDR1 and breast cancer-resistance protein BCRP, and decreased expression of stathmin. Taken together, these results suggest that stathmin expression modulates the pharmacological efficacy of eribulin in uterine leiomyosarcoma cells.


Asunto(s)
Leiomiosarcoma , Estatmina , Humanos , Estatmina/genética , Estatmina/metabolismo , Estatmina/farmacología , Leiomiosarcoma/tratamiento farmacológico , Leiomiosarcoma/genética , Leiomiosarcoma/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Proteínas de Neoplasias/metabolismo , Microtúbulos/metabolismo , Microtúbulos/patología
6.
J Pharmacol Sci ; 150(4): 267-274, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36344049

RESUMEN

The pathogenesis of hypertensive disorder of pregnancy (HDP), which affects about 10% of pregnant women, is still incompletely understood. Our previous study showed that endoplasmic reticulum (ER) stress influences high-temperature requirement A serine peptidase 1 (HTRA1) expression and trophoblast invasion. However, the involvement of ER stress in the regulation of HTRA subtype expression and pathophysiology of HDP has not been characterized in extravillous trophoblasts (EVTs). To investigate this, HTR8/SVneo EVTs cell line was treated with the ER stress inducers Thapsigargin (Thap) or Tunicamycin (Tuni). Treatment with either Thap or Tuni inhibited trophoblast invasion, reduced HTRA1 and HTRA3 expression, but did not alter HTRA2 or HTRA4 expression. Knockdown of HTRA1 or HTRA3 also inhibited trophoblast invasion. Furthermore, treatment with either ER stress inducer or HTRA1 silencing increased the ratio of soluble fms-like tyrosine kinase-1/placental growth factor (sFLT1/PlGF), which is a marker of HDP. Immunohistochemical analysis revealed that HTRA1 is localized to EVTs and the endometrial decidua in the placenta of patients with HDP. These results suggest that factors that cause ER stress could result in the inhibition of EVTs invasion via HTRA1.


Asunto(s)
Trofoblastos , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Humanos , Femenino , Embarazo , Trofoblastos/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Estrés del Retículo Endoplásmico/genética , Temperatura , Factor de Crecimiento Placentario , Placenta/química , Placenta/metabolismo , Movimiento Celular/genética , Serina Peptidasa A1 que Requiere Temperaturas Altas/genética , Serina Peptidasa A1 que Requiere Temperaturas Altas/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/análisis , Serina Endopeptidasas/metabolismo , Serina Proteasas/genética , Serina Proteasas/metabolismo
7.
Biol Pharm Bull ; 45(11): 1627-1635, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36328498

RESUMEN

Eribulin, an inhibitor of microtubule dynamics, is used for treating breast cancers and sarcomas. The microtubule-destabilizing protein stathmin may modulate the antiproliferative activity of eribulin on breast cancer cells and leiomyosarcoma cells. The antitumor activity of eribulin in ovarian cancers has not been fully explored, so the present study aimed to determine the antitumor efficacy of eribulin and the involvement of stathmin in ovarian cancers. In a xenograft model of ovarian cancer, eribulin treatment reduced the tumor weight, which was accompanied by an increased level of phosphorylated stathmin. Eribulin stimulated the phosphorylation of stathmin in cultured cancer cell lines. The eribulin-induced phosphorylation of stathmin was inhibited by treatment with FTY720, an activator of protein phosphatase 2A (PP2A), and eribulin downregulated the expression of PP2A subunits. Furthermore, stathmin knockdown abrogated the inhibitory effects of eribulin on cell viability. Eribulin enhanced the antiproliferative effects of paclitaxel and concomitantly decreased stathmin expression. These results suggest that eribulin-induced phosphorylation of stathmin, mediated in part by PP2A downregulation, reduces stathmin activity and enhances the antiproliferative effects of paclitaxel in ovarian cancer. Collectively, the results of this study indicate that eribulin may suppress the proliferation of ovarian cancer cells partly by regulating the activity of stathmin.


Asunto(s)
Neoplasias Ováricas , Paclitaxel , Humanos , Femenino , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Estatmina/metabolismo , Estatmina/farmacología , Línea Celular Tumoral , Neoplasias Ováricas/metabolismo , Microtúbulos
8.
Int J Mol Sci ; 23(4)2022 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-35216073

RESUMEN

The serine protease inhibitor alpha1-antitrypsin (A1AT) may possess protective functions of impaired organs in a manner independent of its protease inhibitor activity. A1AT expression has been shown to fluctuate in patients with pregnancy-induced hypertension, which suggests that A1AT may play a role in the syncytialization of villous trophoblasts. A1AT expression was knocked down in primary trophoblasts. RNA was extracted from these cells and subjected to RNA-sequencing analysis to determine the levels of expression of markers of syncytialization and inflammation. In addition, A1AT protein was localized in trophoblastic cells in placental tissues. Knockdown of A1AT upregulated the expression of FOSL1 and markers of syncytialization, as well as cell fusion, whereas overexpression of A1AT had the opposite effects. FOSL1 overexpression stimulated syncytialization, similar to the effects of A1AT knock down. Inhibitors of p38MAPK and JNK reduce the expression of inflammatory factors, whereas a p38MAPK inhibitor suppressed FOSL1 expression. Collectively, these findings indicated A1AT may negatively regulate inflammatory responses by controlling the activation of p38MAPK and JNK, and that p38MAPK mediates trophoblast syncytialization by altering FOSL1 expression. Therefore, a dysfunction in A1AT could be responsible for abnormal placental formation and pregnancy-associated disorders.


Asunto(s)
Inflamación/metabolismo , Trofoblastos/metabolismo , alfa 1-Antitripsina/metabolismo , Biomarcadores/metabolismo , Línea Celular , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Placenta/metabolismo , Embarazo
9.
Biochem Biophys Res Commun ; 571: 174-180, 2021 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-34330061

RESUMEN

Decidualization - the differentiation of endometrial stromal cells (ESCs) into decidual cells - is a crucial step for successful embryo implantation and placentation that is initiated in the secretory phase of the menstrual cycle. During decidualization, ESCs undergo proliferation arrest and secrete inflammatory mediators, including senescence-associated secretory phenotype (SASP). Although several senolytic agents improve age-related diseases, their effects on cellular senescence in decidualizing ESCs has not been explored. To do this, we treated decidualized ESCs with the senolytic agents Quercetin (Que), Dasatinib (Das), and BPTES. Que decreased the number of senescence-associated ß-galactosidase (SA-ß-Gal) positive cells and expression of senescence markers in ESCs treated with the decidual stimulus (dibutyryl-cAMP plus progesterone: DP). Concomitantly, Que markedly increased the expression of the decidualization markers IGFBP1, PRL, and FOXO1, in decidualizing ESCs. Similar to Que, Das also stimulated decidualization. Treatment with a combination of Que and Das synergistically increased the expression of decidualization markers and senescence markers compared with treatment with Que or Das alone. However, BPTES did not enhance the expression of decidualization markers. These results imply that treatment with Que and/or Das can remove senescent decidual cells and enhance the decidualization of the rest of ESCs. Thus, senolytic modulation of abnormal ESC decidualization could alleviate infertility caused by dysfunctions of endometrial receptivity and embryo implantation.


Asunto(s)
Dasatinib/farmacología , Endometrio/efectos de los fármacos , Quercetina/farmacología , Células del Estroma/efectos de los fármacos , Sulfuros/farmacología , Tiadiazoles/farmacología , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Femenino , Humanos
10.
Int J Mol Sci ; 22(7)2021 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-33916165

RESUMEN

Alpha-1 antitrypsin (A1AT) is a glycoprotein that has been shown to protect tissues from proteolytic damage under various inflammatory conditions. Several studies show that A1AT may be associated with pre-eclampsia. However, the role of A1AT expression in placental physiology is not fully understood. In the present study, we aim to characterize the expression and function of placental A1AT. A1AT knockdown is found to reduce the expression of the serine protease HTRA1 in a trophoblast cell line. In addition, A1AT overexpression (A1AT-OE) increases the expression of HTRA1, IL6, CXCL8, and several markers of endoplasmic reticulum (ER) stress. Treatment with tunicamycin or thapsigargin, which induces ER stress, increases HTRA1 expression. Furthermore, immunohistochemistry reveals that HTRA1 is expressed in trophoblasts and the endometrial decidual cells of human placentas. An invasion assay shows that A1AT and HTRA1 stimulate cell invasion, but treatment with the ER stress inhibitors reduces the expression of HTRA1 and ER stress markers and prevents cell invasion in A1AT-OE trophoblasts. These results suggest that endogenous A1AT regulates inflammatory cytokine expression and HTRA1-induced trophoblast invasion via the induction of ER stress. It is concluded that an imbalance in the functional link between A1AT and ER stress at the maternal-fetal interface might cause abnormal placental development.


Asunto(s)
Estrés del Retículo Endoplásmico , Inflamación/metabolismo , Trofoblastos/metabolismo , Respuesta de Proteína Desplegada , alfa 1-Antitripsina/metabolismo , Biomarcadores/metabolismo , Línea Celular , Femenino , Serina Peptidasa A1 que Requiere Temperaturas Altas/metabolismo , Humanos , Preeclampsia/etiología , Embarazo
11.
Int J Mol Sci ; 22(19)2021 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-34638846

RESUMEN

Previous in vitro studies have suggested that calreticulin (CALR), which is responsible for the folding and quality control of glycoproteins, may be associated with decidualization. However, its precise role in regulating decidualization has not been explored in vivo. Here, we used pregnant rat models to examine endometrial CALR expression during the peri-implantation period. We also examined whether polypectomy, a procedure that could ameliorate infertility, alters the endometrial expression levels of CALR and several implantation factors in women diagnosed as infertile. In rats, uterine CALR was expressed at a high level at the implantation site, and a marked increase in CALR expression was observed in decidual cells of normal pregnancy. In addition, endometrial CALR expression was enhanced by either administration of estradiol-17ß in the delayed implantation rat model or the artificial induction of decidualization in the pseudopregnant rat. In cultured stromal cells, siRNA-mediated silencing of CALR inhibited the decidual stimulus-induced expression of prolactin, decidual/trophoblast prolactin-related protein, and connexin 43. In humans, the endometrial expression levels of the mRNAs encoding CALR and the implantation-related factor insulin-like growth factor binding protein (IGFBP)-7 tended to increase after polypectomy. The strongest positive correlation between expression levels before polypectomy was observed for IGFBP-7 and CALR, and the strength of this correlation increased after the surgery. Thus, endometrial CALR may play a role in the formation of decidua, and the polypectomy of infertile patients may result in the co-operative expression of endometrial factors, including CALR, that could enhance endometrial receptivity.


Asunto(s)
Blastocisto/metabolismo , Calreticulina/genética , Decidua/metabolismo , Implantación del Embrión , Endometrio/metabolismo , Animales , Blastocisto/citología , Calreticulina/metabolismo , Endometrio/citología , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Infertilidad Femenina/genética , Masculino , Embarazo , Interferencia de ARN , Ratas , Células del Estroma/citología , Células del Estroma/metabolismo , Factores de Tiempo , Útero/metabolismo
12.
Molecules ; 26(2)2021 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-33477249

RESUMEN

Solubility of several anthraquinone derivatives in supercritical carbon dioxide was readily available in the literature, but correcting ability of the existing models was poor. Therefore, in this work, two new models have been developed for better correlation based on solid-liquid phase equilibria. The new model has five adjustable parameters correlating the solubility isotherms as a function of temperature. The accuracy of the proposed models was evaluated by correlating 25 binary systems. The proposed models observed provide the best overall correlations. The overall deviation between the experimental and the correlated results was less than 11.46% in averaged absolute relative deviation (AARD). Moreover, exiting solubility models were also evaluated for all the compounds for the comparison purpose.


Asunto(s)
Antraquinonas/química , Dióxido de Carbono/química , Solubilidad
13.
Reprod Fertil Dev ; 30(11): 1454-1461, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29735004

RESUMEN

Protein kinase A (PKA) signalling accompanies elevated intracellular cAMP levels during endometrial stromal cell (ESC) decidualisation. Exchange protein directly activated by cAMP (EPAC), an alternate mediator of cAMP signalling, promotes PKA analogue-induced decidualisation; however, the precise mechanism by which EPAC and PKA co-operatively stimulate decidualisation has not been characterised. To examine the role of CCAAT/enhancer-binding protein (C/EBP) in EPAC- and PKA-mediated decidualisation of primary human ESCs, a reporter plasmid containing the 332bp region upstream from the transcription initiation site of the decidual prolactin (dPRL) gene was generated and the promoter activity was evaluated using a luciferase assay. The dPRL promoter activity was increased by treatment of transfected ESCs with the PKA-selective cAMP analogue N6-phenyl-cAMP (Phe) and enhanced further by co-treatment with the EPAC-selective cAMP analogue 8-(4-chlorophenyltio)-2'-O-methyl cAMP (CPT). Treatment with forskolin, an adenylyl cyclase activator, had a similar effect on reporter activity. Site-directed mutagenesis of the C/EBPß- and/or C/EBPδ-binding site in the dPRL promoter abolished Phe/CPT-mediated elevation of the reporter activity. EPAC2 knockdown markedly reduced Phe-stimulated C/EBPß and C/EBPδ mRNA levels, as well as forkhead box O1 (FOXO1) protein levels. These results suggest that EPAC signalling enhances PKA-mediated dPRL expression in ESCs by acting on C/EBP response elements in the promoter region of the gene.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Decidua/metabolismo , Endometrio/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Prolactina/metabolismo , Células del Estroma/metabolismo , Activación Transcripcional/genética , Proteínas Potenciadoras de Unión a CCAAT/genética , Colforsina/farmacología , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Decidua/efectos de los fármacos , Endometrio/citología , Endometrio/efectos de los fármacos , Femenino , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Prolactina/genética , Regiones Promotoras Genéticas , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos
14.
J Pharmacol Sci ; 135(2): 89-95, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29056256

RESUMEN

Infection-associated pregnancy complications cause premature delivery. Caspase-1 is involved in the maturation of interleukin (IL)-1ß, which is activated by the NLRP3 inflammasome. To characterize the significance of the NLRP3 inflammasome pathway in the placenta, the effects of activators and inhibitors on NLRP3-related molecules were examined using isolated primary trophoblasts. Caspase-1 and IL-1ß mRNA expression was markedly increased in response to lipopolysaccharide (LPS), a toll-like receptor (TLR)4 ligand. Treatment with the potassium ionophore nigericin significantly increased the level of activated caspase-1. Treatment with either LPS or nigericin stimulated IL-1ß secretion, whereas pretreatment with the ATP-sensitive K+ channel inhibitor glibenclamide, the Rho-associated coiled-coil kinase inhibitor Y-27632, or a caspase-1 inhibitor significantly decreased nigericin-induced IL-1ß secretion. In addition, dibutyryl-cAMP, which induces trophoblast differentiation, decreased expression of NLRP3, caspase-1, and IL-1ß. These findings suggest that trophoblasts can secrete IL-1ß through the NLRP3/caspase-1 pathway, which is suppressed by glibenclamide, and that the TLR4-mediated NLRP3 inflammasome pathway is more likely to be stimulated in undifferentiated than differentiated trophoblasts. Our data support the hypothesis that inhibition of the NLRP3 inflammasome can suppress placental inflammation-associated disorders.


Asunto(s)
Gliburida/farmacología , Inflamasomas/fisiología , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/fisiología , Bloqueadores de los Canales de Potasio/farmacología , Trofoblastos/metabolismo , Amidas/farmacología , Bucladesina/farmacología , Caspasa 1/metabolismo , Caspasa 1/fisiología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Humanos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ionóforos de Potasio/farmacología , Embarazo , Piridinas/farmacología , Trofoblastos/citología , Células U937
15.
Biol Pharm Bull ; 40(5): 638-644, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28458349

RESUMEN

Pioglitazone improves sepsis-induced organ injury accompanied with anti-inflammatory effects on visceral adipose tissue. However, its action in adipose immune cells remains to be ascertained. We investigated the effects of pioglitazone on visceral adipose macrophage population and polarisation in cecal ligation and puncture (CLP)-induced sepsis mice. Eight-week-old male mice were assigned to 3 groups: 1) sham-operated group, 2) CLP group, or 3) pioglitazone-treated CLP group. Pioglitazone (10 mg/kg) was injected intraperitonally for 7 d and CLP surgery was performed. Visceral adipose tissues were collected 24 h after the surgery. mRNA expression of several macrophage markers (inducible nitric oxide synthase (iNOS) for M1, arginase1 (Arg1) and interleukin (IL)-10 for M2, CD163 and F4/80 for mature macrophages) and inflammatory adipokines (IL-6, monocyte chemoattractant protein-1: MCP-1) was quantified by real-time RT-PCR. Tissue sections were subjected to the immunohistochemical analysis and the terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) assay. CLP significantly enhanced Arg1, IL-10 and iNOS mRNA expressions as compared with the sham group, and pioglitazone significantly increased the mRNA level of CD163 and F4/80 in CLP mice. Expression of IL-6 and MCP-1 stimulated by CLP was reduced by pioglitazone treatment. Increased CD11b/c- and CD163-positive cells as well as apoptotic cells were observed in the CLP group and the pioglitazone-treated group. The data indicate that M1/M2 macrophage activation of visceral adipose tissues is induced in CLP-induced mice, and the function of macrophages recruited from surrounding organs may be modulated by pioglitazone treatment.


Asunto(s)
Hipoglucemiantes/farmacología , Grasa Intraabdominal/patología , Macrófagos/efectos de los fármacos , Sepsis/patología , Tiazolidinedionas/farmacología , Adipoquinas/metabolismo , Animales , Arginasa/análisis , Biomarcadores/análisis , Ciego , Quimiocina CCL2/metabolismo , Hipoglucemiantes/administración & dosificación , Inyecciones Intraperitoneales , Interleucina-10/sangre , Ligadura , Masculino , Ratones , Pioglitazona , Punciones , Tiazolidinedionas/administración & dosificación
16.
Artículo en Inglés | MEDLINE | ID: mdl-27174800

RESUMEN

Microsomal prostaglandin E synthase-1 (mPGES-1) is primarily expressed in granulosa cells (GCs) in the preovulatory follicle. Both prostaglandin E2 (PGE2) and progesterone (P4) are implicated in various reproductive functions. Here, we demonstrate that mPges-1 may be a direct downstream target gene of the P4 receptor and P4-stimulated PGE2 secretion can stimulate P4 production in a newly generated mouse GC line (GtsT). Treatment of GtsT cells with a P4 receptor agonist, norgestrel, markedly increased mPGES-1 expression detected by RT-PCR analysis. PGE2 secretion measured by an enzyme-linked immunosorbent assay was enhanced by P4 treatment. Luciferase assays revealed that the proximal promoter region of the mPges-1 gene was responsible for the effects of P4 treatment. Conversely, PGE2 treatment stimulated P4 secretion, which coordinated with mRNA expression of steroidogenic acute regulatory protein. Taken together, P4 may regulate mPGES-1 expression to increase PGE2 secretion and in turn P4 production. An autocrine loop between P4 and PGE2 might function to maintain the increased levels of both in GCs.


Asunto(s)
Dinoprostona/metabolismo , Retroalimentación Fisiológica , Células de la Granulosa/metabolismo , Progesterona/metabolismo , Prostaglandina-E Sintasas/genética , Receptores de Progesterona/genética , Animales , Comunicación Autocrina , Dinoprostona/farmacología , Femenino , Regulación de la Expresión Génica , Genes Reporteros , Células de la Granulosa/citología , Células de la Granulosa/efectos de los fármacos , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Microsomas/metabolismo , Norgestrel/farmacología , Cultivo Primario de Células , Progesterona/farmacología , Regiones Promotoras Genéticas , Prostaglandina-E Sintasas/metabolismo , Receptores de Progesterona/agonistas , Receptores de Progesterona/metabolismo , Transducción de Señal , Acetato de Tetradecanoilforbol/farmacología
17.
Reproduction ; 147(6): 897-906, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24586073

RESUMEN

The optimal decidualization of endometrial stromal cells (ESCs) following embryo implantation is one of the critical steps to establish pregnancy in rodents and humans. This step is intricately regulated by ovarian hormones. Using in vitro human ESCs model, we previously showed that activation of a cAMP mediator, exchange protein directly activated by cAMP (EPAC), promotes ovarian steroid- or cAMP analog-induced decidualization. However, expressions and functions of EPAC and RAP1 in the uterus during pregnancy have not yet been examined. In this study, we found that the expression of EPAC2 and RAP1 was markedly upregulated in the decidual cells at the implantation sites on days 7 and 9 of pregnancy in rats. Furthermore, both delayed-implantation and artificial decidualization models showed that EPAC2 and RAP1 expression was enhanced in decidual cells. Significant activation of cAMP-responsive element-binding protein (CREB), a central transcriptional factor of cAMP signaling, was observed in decidual cells. These spatiotemporal expressions of protein related EPAC pathway are overlapped by sites with activated cAMP signaling, indicating the association of EPAC signaling with decidualization. Strikingly, further studies in in vitro rat decidualization model showed that the cAMP analog and medroxyprogesterone stimulated the expression of decidual markers, while knockdown of EPAC1/2 and RAP1 attenuated the expressions of these markers. Together, these findings suggest that EPAC and RAP1 are the crucial factors for endometrial decidualization in rat pregnancy.


Asunto(s)
Decidua/metabolismo , Implantación del Embrión , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células del Estroma/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Animales , Células Cultivadas , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Deciduoma/metabolismo , Implantación Tardía del Embrión , Femenino , Edad Gestacional , Factores de Intercambio de Guanina Nucleótido/genética , Embarazo , Ratas , Ratas Wistar , Transducción de Señal , Factores de Tiempo , Regulación hacia Arriba , Proteínas de Unión al GTP rap1/genética
18.
J Surg Res ; 189(2): 295-303, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24713471

RESUMEN

BACKGROUND: Pioglitazone modulates adipocyte differentiation and enhances adiponectin promoter activity to increase plasma adiponectin levels. We investigated the effects of pioglitazone on cecal ligation and puncture (CLP)-induced visceral-adipose-tissue inflammation and lung injury in mice. MATERIALS AND METHODS: Eight-wk-old male mice were assigned to three groups: (1) a sham-operated control group, (2) a CLP group, and (3) a pioglitazone-treated CLP group. Pioglitazone (10 mg/kg) was injected intraperitoneally for 7 d. Serum, lung, and visceral adipose tissue were collected 24 h after surgery. Tumor necrosis factor α (TNF-α) levels in peritoneal lavage fluid were measured by an enzyme-linked immunosorbent assay, and TNF-α and interleukin 6 messenger RNA (mRNA) expression levels in visceral adipose tissue were quantified by real-time polymerase chain reaction. Lung tissue specimens were stained with hematoxylin-eosin, and the terminal deoxynucleotidyl transferase dUTP-mediated nick-end labeling method was used to evaluate tissue damage. RESULTS: TNF-α levels in peritoneal lavage fluid were significantly higher in the CLP group than in the sham group. TNF-α levels in the pioglitazone-treated CLP group were significantly lower than those in the CLP group. TNF-α and interleukin 6 mRNA expression levels of visceral adipose tissue were significantly higher in the CLP group than in the sham group. Pioglitazone treatment decreased the mRNA expression levels of these cytokines compared with the respective values in the CLP group. Histopathologic analysis of lung tissue revealed significantly increased numbers of terminal deoxynucleotidyl transferase dUTP-mediated nick-end labeling-positive cells in the CLP group compared with the sham group. CONCLUSIONS: Pioglitazone effectively prevents lung injury caused by CLP-induced sepsis by maintaining the anti-inflammatory status of visceral adipose tissue.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Tejido Adiposo/patología , Hipoglucemiantes/uso terapéutico , Mediadores de Inflamación/fisiología , Sepsis/complicaciones , Tiazolidinedionas/uso terapéutico , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/patología , Tejido Adiposo/metabolismo , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Endotoxinas/análisis , Hipoglucemiantes/farmacología , Interleucina-6/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Pioglitazona , Tiazolidinedionas/farmacología , Factor de Necrosis Tumoral alfa/sangre
19.
J Reprod Dev ; 60(6): 447-53, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25212428

RESUMEN

Angiogenesis in the developing corpus luteum (CL) is a prerequisite for establishment and maintenance of an early pregnancy. To explore the physiological significance of insulin-like growth factor-binding protein-7 (IGFBP7) in the developing CL, the effects of IGFBP7 on vascular endothelial growth factor (VEGFA)- and luteinizing hormone (LH)-induced in vitro tube formation were tested using isolated luteal microvascular endothelial cells (LECs). Capillary-like tube formation of LECs and their proliferation were stimulated by both VEGFA and LH. IGFBP7 treatment suppressed VEGFA- or LH-induced tube formation. The proliferation and migration of LECs, and phosphorylation of mitogen-activated protein kinase kinase and extracellular signal-regulated kinase 1/2 were inhibited by IGFBP7. Furthermore, IGFBP7 attenuated VEGFA-enhanced cyclooxygenase (COX)-2 mRNA expression and prostaglandin E2 secretion. These findings suggest the possibility that luteal IGFBP7 secretion may suppress the stimulatory effect of VEGFA on angiogenesis in the early CL.


Asunto(s)
Cuerpo Lúteo/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Animales , Western Blotting , Movimiento Celular , Proliferación Celular , Factores de Crecimiento Endotelial/farmacología , Femenino , Técnicas In Vitro , Hormona Luteinizante/farmacología , Embarazo , Ratas , Factor A de Crecimiento Endotelial Vascular/farmacología
20.
Sci Rep ; 14(1): 287, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38168580

RESUMEN

The fusion of mononuclear trophoblasts into multinucleate syncytiotrophoblasts is the critical event in the process of syncytialization, and its dysregulation can lead to pregnancy complications, notably hypertensive disorders of pregnancy (HDP). Oxidative stress may disrupt trophoblast syncytialization in HDP. Specifically, placentas with HDP exhibit impaired mitochondria, giving rise to the generation of reactive oxygen species (ROS) and subsequent oxidative stress. Quercetin, a bioflavonoid known for its antioxidant and anti-aging properties, has the potential to mitigate oxidative stress during trophoblast syncytialization. However, the precise mechanism underlying the action of quercetin in these processes remains to be elucidated. To explore the impact of quercetin on syncytialization, mitochondrial function, and ROS generation, cyclic AMP-stimulated BeWo cells were treated with quercetin. The expression of markers associated with cell fusion, mitochondrial function, and oxidative stress was determined using qPCR and western blotting. Additionally, morphological syncytialization and mitophagy (mitochondrial degradation) were assessed by immunofluorescence analysis. Our results revealed that quercetin increased the expression of syncytialization markers and promoted cell fusion. Furthermore, this compound also upregulated markers associated with mitophagy and mitochondrial fusion, which are corroborated by visual evidence of mitophagy through the fluorescence microscope. Cell fusion naturally stimulated ROS generation, which was attenuated by quercetin. Quercetin downregulated the expression of NRF2 and HO-1 during syncytialization, while increasing the expression of sirtuin1/3/6, which are known to play essential roles in antioxidant responses. In conclusion, quercetin effectively regulates mitochondrial function through its antioxidant properties and the suppression of ROS generation, ultimately promoting trophoblast fusion, suggesting that the flavonoid has the potential to ameliorate pregnancy-related disorder stemming from placental dysplasia.


Asunto(s)
Placenta , Quercetina , Embarazo , Humanos , Femenino , Placenta/metabolismo , Quercetina/farmacología , Quercetina/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Antioxidantes/farmacología , Antioxidantes/metabolismo , Trofoblastos/metabolismo , Mitocondrias/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA