Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Diabetes Metab Syndr Obes ; 17: 1727-1738, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38645653

RESUMEN

Purpose: Visceral fat accumulation can negatively affect uric acid metabolism in healthy adults. The hypertriglyceridemic-waist (HTGW) phenotype is a predictor of diabetes and cardiometabolic risk. This study aimed to investigate the association between the HTGW phenotype and asymptomatic hyperuricemia in Korean adults. Patients and Methods: The study included 23,240 adults, aged 20-80 years who underwent comprehensive health examinations at a general hospital in Gyeonggi Province, Korea, from January 2020 to December 2022. The HTGW phenotype was defined as the simultaneous presence of elevated serum triglyceride (TG) levels and increased waist circumference (WC). The diagnostic capability of the HTGW phenotype for hyperuricemia and its association with the condition were assessed using the receiver operating characteristic (ROC) curve and logistic regression analysis. Results: The prevalence of hyperuricemia in the HTGW phenotype was 3.44 times higher than that in the normal TG normal waist (NTNW) phenotype. Compared with those in the NTNW group, the hazard ratios for developing hyperuricemia in the HTGW group were 2.887 (2.566-3.249, P <0.001) for men and 7.341 (5.139-10.487, P <0.001) for women, and these values remained significant after adjusting for potential confounders. The stratified analysis revealed that the HTGW phenotype, coupled with diabetes, had the highest probability of developing asymptomatic hyperuricemia (2.55 times). ROC curve analysis revealed that the area under the curve values of the WC*TG index for hyperuricemia diagnosis were 0.702, 0.627, and 0.685 for all participants, men, and women, respectively. Conclusion: Among Korean adults, the HTGW phenotype was closely related to hyperuricemia in both men and women and showed a particularly strong association in patients with diabetes. It may be used in combination with an indicator that can complement its accuracy for identifying individuals at high risk of hyperuricemia.

2.
Reprod Sci ; 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38637476

RESUMEN

Endometrial cancer is the most common gynecologic malignancy. PTEN is a negative regulator of PI3K signaling and is deficient in > 50% of primary human endometrial cancer. Amplification of ERBB2 promotes tumorigenesis and pathogenesis of several human cancers. However, the effect of ERBB2 targeting has not been studied in endometrial cancer with PTEN mutations. The murine model Pgrcre/+Erbb2f/fPtenf/f (Erbb2d/d Ptend/d) was developed to evaluate the effect of ERBB2 targeted therapy in endometrial cancer with PTEN deficiency. Histopathological and molecular analysis was performed for Ptend/d and Erbb2d/dPtend/d mice. Histopathological analysis revealed that Erbb2d/dPtend/d mice significantly reduced development and progression of endometrial cancer compared to Ptend/d mice. Furthermore, percentage of proliferative cells in Erbb2d/dPtend/d mice revealed anti-tumorigenic effect of Erbb2 ablation compared to Ptend/d mice. Our results demonstrate that Erbb2 ablation reveals a significant suppression of tumorigenesis on endometrial cancer of Ptend/d mice. Our results suggest that Erbb2 functions as an oncogene in endometrial cancer of Ptend/d mice implying that Erbb2 targeting can be used as an effective therapeutic approach for treatment of endometrial cancer with PTEN deficiency to hinder cancer development.

3.
Reprod Sci ; 31(6): 1632-1641, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38388922

RESUMEN

Endometrial cancer (EC) is the most common gynecologic malignancy. While the majority of patients present with early-stage and low-grade EC and have an excellent prognosis, a subset has metastatic disease at presentation or develops distant recurrence after initial treatment of the primary. However, the lack of prognostic biomarkers for metastatic EC is a critical barrier. Arginase 1 (ARG1) regulates the last step of the urea cycle, and an increase in ARG1 has been correlated as a poor prognostic factor in a variety of cancers. In the present study, ARG1 expression was evaluated as a potential prognostic marker for metastatic EC in endometrial hyperplasia and cancer of mice with Pten mutation as well as Pten and Mig-6 double mutations. While Pten mutation in the uterus is not sufficient for distant metastasis, mice with concurrent ablation of Mig-6 and Pten develop distant metastasis. Our immunostaining and RT-qPCR analysis revealed that the expression of ARG1 in early stage of EC as well as endometrial hyperplasia from mice deficient in Mig-6 and Pten mutations significantly increased compared to Pten mutation in the uterus. The results suggest that a high level of ARG1 is associated with poor prognosis in association with EC of mouse.


Asunto(s)
Arginasa , Biomarcadores de Tumor , Neoplasias Endometriales , Fosfohidrolasa PTEN , Femenino , Neoplasias Endometriales/patología , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Animales , Arginasa/genética , Arginasa/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Pronóstico , Ratones , Humanos , Mutación , Hiperplasia Endometrial/genética , Hiperplasia Endometrial/metabolismo , Hiperplasia Endometrial/patología , Metástasis de la Neoplasia
4.
Exp Mol Med ; 55(8): 1795-1805, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37524875

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a chronic, fatal, fibrotic, interstitial lung disease of unknown cause. Despite extensive studies, the underlying mechanisms of IPF development remain unknown. Here, we found that p300 was upregulated in multiple epithelial cells in lung samples from patients with IPF and mouse models of lung fibrosis. Lung fibrosis was significantly diminished by the alveolar type II (ATII) cell-specific deletion of the p300 gene. Moreover, we found that ubiquitin C-terminal hydrolase L3 (UCHL3)-mediated deubiquitination of p300 led to the transcriptional activation of the chemokines Ccl2, Ccl7, and Ccl12 through the cooperative action of p300 and C/EBPß, which consequently promoted M2 macrophage polarization. Selective blockade of p300 activity in ATII cells resulted in the reprogramming of M2 macrophages into antifibrotic macrophages. These findings demonstrate a pivotal role for p300 in the development of lung fibrosis and suggest that p300 could serve as a promising target for IPF treatment.


Asunto(s)
Células Epiteliales Alveolares , Fibrosis Pulmonar Idiopática , Ubiquitina Tiolesterasa , Animales , Ratones , Quimiocina CCL2/genética , Enzimas Desubicuitinizantes , Fibrosis Pulmonar Idiopática/genética , Pulmón , Humanos , Ubiquitina Tiolesterasa/metabolismo , Proteína p300 Asociada a E1A
5.
Stem Cell Res ; 67: 103048, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36801602

RESUMEN

Cardiac muscle troponin T protein binds to tropomyosin and regulates the calcium-dependent actin-myosin interaction on thin filaments in cardiomyocytes. Recent genetic studies have revealed that TNNT2 mutations are strongly linked to dilated cardiomyopathy (DCM). In this study, we generated YCMi007-A, a human induced pluripotent stem cell (hiPSC) line from a DCM patient with a p. Arg205Trp mutation in the TNNT2 gene. The YCMi007-A cells show high expression of pluripotent markers, normal karyotype, and differentiation into three germ layers. Thus, YCMi007-A-an established iPSC-could be useful for the investigation of DCM.


Asunto(s)
Cardiomiopatía Dilatada , Células Madre Pluripotentes Inducidas , Humanos , Cardiomiopatía Dilatada/genética , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Troponina T/genética , Troponina T/metabolismo , Heterocigoto , Mutación
6.
BMB Rep ; 56(2): 114-119, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36593107

RESUMEN

Liver fibrosis is caused by chronic liver damage and results in the aberrant accumulation of extracellular matrix during disease progression. Despite the identification of the HAT enzyme p300 as a major factor for liver fibrosis, the development of therapeutic agents targeting the regulation of p300 has not been reported. We validated a novel p300 inhibitor (A6) on the improvement of liver fibrosis using two mouse models, mice on a choline-deficient high-fat diet and thioacetamide-treated mice. We demonstrated that pathological hall-marks of liver fibrosis were significantly diminished by A6 treatment through Masson's trichrome and Sirius red staining on liver tissue and found that A6 treatment reduced the expression of matricellular protein genes. We further showed that A6 treatment improved liver fibrosis by reducing the stability of p300 protein via disruption of p300 binding to AKT. Our findings suggest that targeting p300 through the specific inhibitor A6 has potential as a major therapeutic avenue for treating liver fibrosis. [BMB Reports 2023; 56(2): 114-119].


Asunto(s)
Histonas , Cirrosis Hepática , Ratones , Animales , Histonas/metabolismo , Hígado/metabolismo , Modelos Animales de Enfermedad , Dieta Alta en Grasa
7.
Int J Mol Sci ; 25(1)2023 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-38203534

RESUMEN

Enterotoxigenic Bacteroides fragilis (ETBF) causes colitis and is implicated in inflammatory bowel diseases and colorectal cancer. The ETBF-secreted B. fragilis toxin (BFT) causes cleavage of the adherence junction, the E-cadherin, resulting in the large intestine showing IL-17A inflammation in wild-type (WT) mice. However, intestinal pathology by ETBF infection is not fully understood in B-cell-deficient mice. In this study, ETBF-mediated inflammation was characterized in B-cell-deficient mice (muMT). WT or muMT C57BL/6J mice were orally inoculated with ETBF and examined for intestinal inflammation. The indirect indicators for colitis (loss of body weight and cecum weight, as well as mortality) were increased in muMT mice compared to WT mice. Histopathology and inflammatory genes (Nos2, Il-1ß, Tnf-α, and Cxcl1) were elevated and persisted in the large intestine of muMT mice compared with WT mice during chronic ETBF infection. However, intestinal IL-17A expression was comparable between WT and muMT mice during infection. Consistently, flow cytometry analysis applied to the mesenteric lymph nodes showed a similar Th17 immune response in both WT and muMT mice. Despite elevated ETBF colonization, the ETBF-infected muMT mice showed no histopathology or inflammation in the small intestine. In conclusion, B cells play a protective role in ETBF-induced colitis, and IL-17A inflammation is not attributed to prompted colitis in B-cell-deficient mice. Our data support the fact that B cells are required to ameliorate ETBF infection-induced colitis in the host.


Asunto(s)
Infecciones Bacterianas , Colitis , Animales , Ratones , Ratones Endogámicos C57BL , Bacteroides fragilis , Interleucina-17/genética , Inflamación
8.
Nat Commun ; 13(1): 1101, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35232969

RESUMEN

Female subfertility is highly associated with endometriosis. Endometrial progesterone resistance is suggested as a crucial element in the development of endometrial diseases. We report that MIG-6 is downregulated in the endometrium of infertile women with endometriosis and in a non-human primate model of endometriosis. We find ERBB2 overexpression in the endometrium of uterine-specific Mig-6 knockout mice (Pgrcre/+Mig-6f/f; Mig-6d/d). To investigate the effect of ERBB2 targeting on endometrial progesterone resistance, fertility, and endometriosis, we introduce Erbb2 ablation in Mig-6d/d mice (Mig-6d/dErbb2d/d mice). The additional knockout of Erbb2 rescues all phenotypes seen in Mig-6d/d mice. Transcriptomic analysis shows that genes differentially expressed in Mig-6d/d mice revert to their normal expression in Mig-6d/dErbb2d/d mice. Together, our results demonstrate that ERBB2 overexpression in endometrium with MIG-6 deficiency causes endometrial progesterone resistance and a nonreceptive endometrium in endometriosis-related infertility, and ERBB2 targeting reverses these effects.


Asunto(s)
Endometriosis , Infertilidad Femenina , Péptidos y Proteínas de Señalización Intracelular , Receptor ErbB-2 , Enfermedades Uterinas , Animales , Endometriosis/genética , Endometriosis/metabolismo , Endometrio/anomalías , Endometrio/metabolismo , Femenino , Infertilidad Femenina/genética , Infertilidad Femenina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Progesterona/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Enfermedades Uterinas/genética , Enfermedades Uterinas/metabolismo
9.
Biochem Biophys Res Commun ; 550: 151-157, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33706098

RESUMEN

Endometriosis is a disorder in which endometrial cells normally limited to the lining of the uterus proliferate outside the uterine cavity and can cause pelvic pain and infertility. ARID1A levels are significantly reduced in the eutopic endometrium from women with endometriosis. Uterine specific Arid1a knock-out mice were infertile due to loss of epithelial progesterone receptor (PGR) signaling. However, the functional association of ARID1A and PGR in endometriosis has not been studied. We examined the expression patterns and co-localization of ARID1A and PGR in eutopic endometrium from women with and without endometriosis using immunostaining and Western blot analysis. ARID1A and PGR proteins co-localized in the epithelium during the proliferative and the early secretory phases. Our immunoprecipitation analysis and proximity ligation assay (PLA) revealed physical interaction between ARID1A and PGR-A but not PGR-B in the mouse and human endometrium. ARID1A levels positively correlated with PGR levels in the eutopic endometrium of women with endometriosis. Our results bring new perspectives on the molecular mechanisms involved in endometrial receptivity and progesterone resistance in endometriosis. The interrelationship between ARID1A and PGR may contribute to explaining the non-receptive endometrium in endometriosis-related infertility.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Endometriosis/metabolismo , Endometrio/metabolismo , Receptores de Progesterona/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Unión al ADN/deficiencia , Endometriosis/patología , Endometrio/patología , Femenino , Células HEK293 , Humanos , Inmunoprecipitación , Ratones , Unión Proteica , Receptores de Progesterona/deficiencia , Factores de Transcripción/deficiencia
10.
Mol Hum Reprod ; 27(6)2021 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-33693877

RESUMEN

About 40% of women with infertility and 70% of women with pelvic pain suffer from endometriosis. The pregnancy rate in women undergoing IVF with low endometrial integrin αvß3 (LEI) expression is significantly lower compared to the women with high endometrial integrin αvß3 (HEI). Mid-secretory eutopic endometrial biopsies were obtained from healthy controls (C; n=3), and women with HEI (n=4) and LEI (n=4) and endometriosis. Changes in gene expression were assessed using human gene arrays and DNA methylation data were derived using 385 K Two-Array Promoter Arrays. Transcriptional analysis revealed that LEI and C groups clustered separately with 396 differentially expressed genes (DEGs) (P<0.01: 275 up and 121 down) demonstrating that transcriptional and epigenetic changes are distinct in the LEI eutopic endometrium compared to the C and HEI group. In contrast, HEI vs C and HEI vs LEI comparisons only identified 83 and 45 DEGs, respectively. The methylation promoter array identified 1304 differentially methylated regions in the LEI vs C comparison. The overlap of gene and methylation array data identified 14 epigenetically dysregulated genes and quantitative RT-PCR analysis validated the transcriptomic findings. The analysis also revealed that aryl hydrocarbon receptor (AHR) was hypomethylated and significantly overexpressed in LEI samples compared to C. Further analysis validated that AHR transcript and protein expression are significantly (P<0.05) increased in LEI women compared to C. The increase in AHR, together with the altered methylation status of the 14 additional genes, may provide a diagnostic tool to identify the subset of women who have endometriosis-associated infertility.


Asunto(s)
Metilación de ADN , Endometriosis/genética , Endometrio/metabolismo , Infertilidad Femenina/etiología , Integrina alfaVbeta3/biosíntesis , Transcriptoma , Adolescente , Adulto , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Biopsia , Regulación hacia Abajo , Endometriosis/complicaciones , Endometriosis/metabolismo , Endometrio/patología , Femenino , Humanos , Infertilidad Femenina/genética , Integrina alfaVbeta3/genética , Persona de Mediana Edad , Análisis de Componente Principal , Receptores de Hidrocarburo de Aril/biosíntesis , Receptores de Hidrocarburo de Aril/genética , Adulto Joven
11.
FASEB J ; 35(2): e21209, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33222288

RESUMEN

Though endometriosis and infertility are clearly associated, the pathophysiological mechanism remains unclear. Previous work has linked endometrial ARID1A loss to endometriosis-related endometrial non-receptivity. Here, we show in mice that ARID1A binds and regulates transcription of the Foxa2 gene required for endometrial gland function. Uterine-specific deletion of Arid1a compromises gland development and diminishes Foxa2 and Lif expression. Deletion of Arid1a with Ltf-iCre in the adult mouse endometrial epithelium preserves the gland development while still compromising the gland function. Mice lacking endometrial epithelial Arid1a are severely sub-fertile due to defects in implantation, decidualization, and endometrial receptivity from disruption of the LIF-STAT3-EGR1 pathway. FOXA2 is also reduced in the endometrium of women with endometriosis in correlation with diminished ARID1A, and both ARID1A and FOXA2 are reduced in nonhuman primates induced with endometriosis. Our findings describe a role for ARID1A in the endometrial epithelium supporting early pregnancy establishment through the maintenance of gland function.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Implantación del Embrión , Endometrio/metabolismo , Factores de Transcripción/metabolismo , Adulto , Animales , Proteínas de Unión al ADN/genética , Femenino , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/metabolismo , Humanos , Factor Inhibidor de Leucemia/genética , Factor Inhibidor de Leucemia/metabolismo , Ratones , Ratones Endogámicos C57BL , Embarazo , Factores de Transcripción/genética
12.
Exp Mol Med ; 52(10): 1754-1765, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33060769

RESUMEN

Adenomyosis is defined as the presence of ectopic nests of endometrial glands and stroma within the myometrium. Adenomyosis is a common cause of dysmenorrhea, menorrhagia, and chronic pelvic pain but is often underdiagnosed. Despite its prevalence and severity of symptoms, its pathogenesis and etiology are poorly understood. Our previous study showed that aberrant activation of ß-catenin results in adenomyosis through epithelial-mesenchymal transition. Using transcriptomic and ChIP-seq analysis, we identified activation of TGF-ß signaling in the uteri of mutant mice that expressed dominant stabilized ß-catenin in the uterus. There was a strong positive correlation between ß-catenin and TGF-ß2 proteins in women with adenomyosis. Furthermore, treatment with pirfenidone, a TGF-ß inhibitor, increased E-cadherin expression and reduced cell invasiveness in Ishikawa cells with nuclear ß-catenin. Our results suggest that ß-catenin activates TGF-ß-induced epithelial-mesenchymal transition in adenomyosis. This finding describes the molecular pathogenesis of adenomyosis and the use of TGF-ß as a potential therapeutic target for adenomyosis.


Asunto(s)
Adenomiosis/metabolismo , Susceptibilidad a Enfermedades , Transición Epitelial-Mesenquimal , Factor de Crecimiento Transformador beta/metabolismo , beta Catenina/metabolismo , Adenomiosis/etiología , Adenomiosis/patología , Animales , Sitios de Unión , Cadherinas/metabolismo , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , Unión Proteica , Factor de Crecimiento Transformador beta/farmacología
13.
BMC Cancer ; 19(1): 810, 2019 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-31412816

RESUMEN

BACKGROUND: Endometrial cancer is the most common gynecological cancer. G-protein coupled receptor 64 (GPR64) belongs to a family of adhesion GPCRs and plays an important role in male fertility. However, the function of GPR64 has not been studied in endometrial cancer. Our objective is to investigate the role of GPR64 in endometrial cancer. METHODS: We examined the levels of GPR64 in human endometrioid endometrial carcinoma by immunohistochemistry analysis. To determine a tumor suppressor role of GPR64 in endometrial cancer, we used a siRNA loss of function approach in human endometrial adenocarcinoma cell lines. RESULTS: GPR64 levels were remarkably lower in 10 of 21 (47.62%) of endometrial carcinoma samples compared to control. Depletion of GPR64 by siRNA transfection revealed an increase of colony formation ability, cell proliferation, cell migration, and invasion activity in Ishikawa and HEC1A cells. The expression of Connexin 43 (Cx43), a member of the large family of gap junction proteins, was reduced through activation of AMP-activated protein kinase (AMPK) in Ishikawa cells with GPR64-deficicy. CONCLUSIONS: These results suggest that GPR64 plays an important tumor suppressor role in endometrial cancer.


Asunto(s)
Carcinoma Endometrioide/patología , Neoplasias Endometriales/patología , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Carcinoma Endometrioide/genética , Carcinoma Endometrioide/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Conexina 43/metabolismo , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Invasividad Neoplásica , Fosforilación , ARN Interferente Pequeño , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética
14.
Clin Cancer Res ; 25(12): 3617-3629, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-30814110

RESUMEN

PURPOSE: Because of disease heterogeneity, limited studies on effective chemotherapies and therapeutic agents for advanced gastric cancer are available. Erythrocyte membrane protein band 4.1-like 5 (EPB41L5) has critical roles in renal and breast cancer metastasis. However, its role in metastatic gastric cancer remains unknown. EXPERIMENTAL DESIGN: The specimens of 78 gastric cancer patients were analyzed by oligonucleotide microarray and survival analysis. In vitro experiments and metastatic mice models were used to assess the effects of EPB41L5 on gastric cancer metastasis. RESULTS: Gastric cancer patients with high EPB41L5 levels had poor prognosis and low survival rate. Further, TGFß1-induced EPB41L5 expression promoted gastric cancer cell migration and invasion by Smad-dependent TGFß signaling. Phospho-Smad3 recruitment to the EPB41L5 promoter was significantly inhibited by a TGFß inhibitor. EPB41L5 overexpression increased lung metastasis of gastric cancer cells in nude mice, which was completely reversed by anti-EPB41L5 monoclonal antibody treatment. Importantly, p120-catenin knockdown abolished EPB41L5-enhanced gastric cancer cell metastasis. Anti-EPB41L5 monoclonal antibody treatment blocked the association of EPB41L5 with p120-catenin. CONCLUSIONS: TGFß/EPB41L5/p120-catenin axis regulates gastric cancer cell metastasis, and EPB41L5 is a promising therapeutic target for advanced gastric cancer.


Asunto(s)
Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Proteínas de la Membrana/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Embrión de Pollo , Femenino , Xenoinjertos , Humanos , Neoplasias Pulmonares/genética , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Pronóstico , Regiones Promotoras Genéticas , Transducción de Señal , Neoplasias Gástricas/genética , Tasa de Supervivencia
15.
Sci Transl Med ; 11(474)2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30626716

RESUMEN

Endometriosis is a disease in which tissue that normally grows inside the uterus grows outside the uterus and causes chronic pelvic pain and infertility. However, the exact mechanisms of the pathogenesis of endometriosis-associated infertility are unknown. Epigenetic dysregulation has recently been implicated in infertility. Here, we report a reduction of histone deacetylase 3 (HDAC3) protein amounts in eutopic endometrium of infertile women with endometriosis compared to a control group. To investigate the effect of HDAC3 loss in the uterus, we generated mice with conditional ablation of Hdac3 in progesterone receptor (PGR)-positive cells (Pgrcre/+Hdac3f/f ; Hdac3d/d ). Loss of Hdac3 in the uterus of mice results in infertility due to implantation failure and decidualization defect. Expression microarray and ChIP-seq analyses identified COL1A1 and COL1A2 as direct targets of HDAC3 in both mice and humans. Reduction of HDAC3 abrogated decidualization in a primary culture of human endometrial stromal cells (hESCs) similar to that observed in infertile patients with endometriosis. Whereas attenuation of HDAC3 resulted in p300 recruitment to Col1a1 and Col1a2 genes in the uterus of mice as well as hESCs, inhibition of p300 permitted hESCs to undergo decidualization. Collectively, we found attenuation of HDAC3 and overexpression of collagen type I in the eutopic endometrium of infertile patients with endometriosis. HDAC3 loss caused a defect of decidualization through the aberrant transcriptional activation of Col1a1 and Col1a2 genes in mice and COL1A1 and COL1A2 genes in humans. Our results suggest that HDAC3 is critical for endometrial receptivity and decidualization.


Asunto(s)
Endometrio/enzimología , Endometrio/patología , Histona Desacetilasas/deficiencia , Infertilidad Femenina/enzimología , Infertilidad Femenina/patología , Adolescente , Adulto , Animales , Colágeno/genética , Colágeno/metabolismo , Decidua/patología , Implantación del Embrión , Endometriosis/enzimología , Endometriosis/patología , Femenino , Histona Desacetilasas/metabolismo , Humanos , Ratones Endogámicos C57BL , Persona de Mediana Edad , Papio , Progesterona/metabolismo , Transducción de Señal , Células Madre/metabolismo , Adulto Joven
16.
J Cell Physiol ; 234(3): 2649-2658, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30144069

RESUMEN

Although programed cell death 5 (PDCD5) is an important protein in p53-mediated proapoptotic signaling, very little is known about PDCD5-related cell death. In this study, we report that serine/threonine kinase 31 (STK31) interacts with PDCD5, which maintains the stability of PDCD5. STK31 overexpression significantly activated PDCD5 stabilization and p53-mediated apoptosis in response to etoposide (ET). However, STK31 knockdown did not enhance apoptosis by ET treatment. Moreover, when STK31 was depleted, PDCD5 inhibited the activation of the p53 signaling pathway with ET, indicating that the PDCD5-STK31 network has an essential role in p53 activation. Importantly, STK31 activated the p53 signaling pathway by genotoxic stress through positive regulation of PDCD5-mediated apoptosis. We thus demonstrated that overexpression of STK31 greatly inhibited tumorigenic growth and increased the chemosensitivity of HCT116 human colorectal carcinoma cells. Taken together, these findings demonstrate that the STK31-PDCD5 complex network regulates apoptosis of cancer cells, and STK31 is a positive apoptosis regulator that inhibits tumorigenesis of colon cancer cells by inducing PDCD5-mediated apoptosis in response to genotoxic stress.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Etopósido/farmacología , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteína p53 Supresora de Tumor/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/patología , Daño del ADN/efectos de los fármacos , Humanos , Proteínas de Neoplasias/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
17.
BMC Cancer ; 18(1): 605, 2018 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-29843645

RESUMEN

BACKGROUND: Aberrant hyperactivation of epithelial proliferation, AKT signaling, and association with unopposed estrogen (E2) exposure is the most common endometrial cancer dysfunction. In the normal uterus, progesterone (P4) inhibits proliferation by coordinating stromal-epithelial cross-talk, which we previously showed is mediated by the function of Mitogen-inducible gene 6 (Mig-6). Despite their attractive characteristics, non-surgical conservative therapies based on progesterone alone have not been universally successful. One barrier to this success has been the lack of understanding of the P4 effect on endometrial cells. METHOD: To further understand the role of Mig-6 and P4 in controlling uterine proliferation, we developed a Sprr2f-cre driven mouse model where Mig-6 is specifically ablated only in the epithelial cells of the uterus (Sprr2f cre+ Mig-6 f/f ). We examined P4 effect and regulation of AKT signaling in the endometrium of mutant mice. RESULTS: Sprr2f cre+ Mig-6 f/f mice developed endometrial hyperplasia. P4 treatment abated the development of endometrial hyperplasia and restored morphological and histological characteristics of the uterus. P4 treatment reduced cell proliferation which was accompanied by decreased AKT signaling and the restoration of stromal PGR and ESR1 expression. Furthermore, our in vitro studies revealed an inhibitory effect of MIG-6 on AKT phosphorylation as well as MIG-6 and AKT protein interactions. CONCLUSIONS: These data suggest that endometrial epithelial cell proliferation is regulated by P4 mediated Mig-6 inhibition of AKT phosphorylation, uncovering new mechanisms of P4 action. This information may help guide more effective non-surgical interventions in the future.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias Endometriales/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Progesterona/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Proliferación Celular , Proteínas Ricas en Prolina del Estrato Córneo/genética , Endometrio/citología , Endometrio/metabolismo , Endometrio/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Noqueados , Modelos Animales , Fosforilación , Receptores de Progesterona/metabolismo , Transducción de Señal
18.
Asian-Australas J Anim Sci ; 31(8): 1176-1182, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29642667

RESUMEN

OBJECTIVE: Progesterone receptor (PGR) and AT-rich interactive domain 1A (ARID1A) have important roles in the establishment and maintenance of pregnancy in the uterus. In present studies, we examined the expression of mitochondrial tumor suppressor 1 (MTUS1) in the murine uterus during early pregnancy as well as in response to ovarian steroid hormone treatment. METHODS: We performed quantitative reverse transcription polymerase chain reaction and immunohistochemistry analysis to investigate the regulation of MTUS1 by ARID1A and determined expression patterns of MTUS1 in the uterus during early pregnancy. RESULTS: The expression of MTUS1 was detected on day 0.5 of gestation (GD 0.5) and then gradually increased until GD 3.5 in the luminal and glandular epithelium. However, the expression of MTUS1 was significantly reduced in the uterine epithelial cells of Pgrcre/+Arid1af/f and Pgr knockout (PRKO) mice at GD 3.5. Furthermore, MTUS1 expression was remarkably induced after P4 treatment in the luminal and glandular epithelium of the wild-type mice. However, the induction of MTUS1 expression was not detected in uteri of Pgrcre/+Arid1af/f or PRKO mice treated with P4. CONCLUSION: These results suggest that MTUS1 is a novel target gene by ARID1A and PGR in the uterine epithelial cells.

19.
PLoS One ; 13(1): e0191433, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29346447

RESUMEN

The PI3K/AKT signaling pathway plays a critical role in the maintenance of equilibrium between cell survival and apoptosis. The Pik3ca gene is mutated in a range of human cancers. It has been found to be oncogenic, and mutations lead to constitutive activation of the PI3K/AKT pathway. The expression patterns of PIK3CA proteins in the uterus of mice during early pregnancy indicate that it may play a role in the regulation of glandular epithelial cells, which is required to support uterine receptivity. To further investigate the role of Pik3ca in uterine function, Pik3ca was conditionally ablated only in the PGR-positive cells (Pgrcre/+Pik3caf/f; Pik3cad/d). A defect of uterine gland development and decidualization led to subfertility observed in Pik3cad/d mice. Pik3cad/d mice showed significantly decreased uterine weight compared to Pik3caf/f mice. Interestingly, a significant decrease of gland numbers were detected in Pik3cad/d mice compared to control mice. In addition, we found a decrease of Foxa2 expression, which is a known uterine gland marker in Pik3cad/d mice. Furthermore, the excessive proliferation of endometrial epithelial cells was observed in Pik3cad/d mice. Our studies suggest that Pik3ca has a critical role in uterine gland development and female fertility.


Asunto(s)
Fosfatidilinositol 3-Quinasas/metabolismo , Útero/crecimiento & desarrollo , Animales , Western Blotting , Proliferación Celular , Fosfatidilinositol 3-Quinasa Clase I , Decidua/citología , Decidua/crecimiento & desarrollo , Implantación del Embrión , Femenino , Fertilidad , Ratones , Ratones Endogámicos C57BL , Mutación , Fosfatidilinositol 3-Quinasas/genética , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa , Útero/citología , Útero/metabolismo
20.
Reprod Sci ; 25(2): 207-213, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28673206

RESUMEN

The four jointed box 1 (FJX1) is a regulator of angiogenesis, and the levels of FJX1 are increased in several types of cancer. Angiogenesis plays a critical role in endometrial growth as well as in several gynecologic disorders including endometriosis. However, the function of FJX1 has not been studied in endometriosis. Therefore, we examined the levels of FJX1 in eutopic endometrium from women with or without endometriosis. The levels of FJX1 protein did not change in endometrial cells during the menstrual cycle in endometrium from women without endometriosis. However, its levels were significantly higher in the secretory phase of the eutopic endometrium from women with endometriosis when compared to women without endometriosis. Hypoxia-inducible factor-1α (HIF1α) is known as a key mediator of endometriosis by regulating genes essential to estrogen production, angiogenesis, proliferation, inflammation, and extracellular invasion. It has been reported that FJX1 induces an increase in HIF1α through posttranslational stabilization. The results of our Western blot analysis reveal a significant positive correlation between FJX1 and HIF1α proteins in endometrium of women with and without endometriosis. This overexpression of FJX1 was confirmed by sequential analysis of the eutopic endometrium during endometriosis progression, using an induced model of endometriosis in the baboon. Therefore, our results suggest that high levels of FJX1 proteins may play an important role in the pathogenesis of endometriosis.


Asunto(s)
Endometriosis/metabolismo , Endometrio/metabolismo , Proteínas de la Membrana/metabolismo , Neovascularización Patológica/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Papio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...