Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Exp Cell Res ; 434(1): 113866, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-38042247

RESUMEN

Endometrial carcinoma (EC) is a rising concern among gynecological malignancies. Iroquois Homeobox 2 (IRX2), a member of the Iroquois homeobox gene family, demonstrates variable effects in different cancer types, emphasizing the need for extensive exploration of its involvement in EC progression. Utilizing TCGA and GEO databases, as well as performing immunohistochemistry (IHC) analysis on clinical samples, we assessed the expression levels of IRX2 and its promoter methylation in EC. To understand the functional roles of IRX2, we conducted various assays including in vitro CCK-8 assays, colony formation assays, cell invasion assays, and cell apoptosis assays. Moreover, we utilized in vivo subcutaneous xenograft mouse models. Additionally, we performed KEGG pathway and gene set enrichment analyses to gain insights into the underlying mechanisms. To validate the regulatory relationship between IRX2 and RUVBL1, we employed chromatin immunoprecipitation and luciferase reporter assays. Our results indicate significantly reduced levels of IRX2 expression in EC, correlating with higher histological grades, advanced clinical stages, and diminished overall survival. We observed that DNA methylation of the IRX2 promoter suppresses its expression in EC, with cg26333652 and cg11793269 playing critical roles as methylated sites. In contrast, ectopic overexpression of IRX2 substantially inhibits cell proliferation and invasion, and promotes cell apoptosis. Additionally, we discovered that IRX2 exerts negative regulation on the expression of RUVBL1, which is upregulated in EC and associated with a poorer prognosis. In conclusion, our findings indicate that decreased expression of IRX2 facilitates EC cell growth through the regulation of RUVBL1 expression, thereby contributing to the development of EC. Hence, targeting the IRX2-RUVBL1 axis holds promise as a potential therapeutic strategy for EC treatment.


Asunto(s)
Neoplasias Endometriales , MicroARNs , Femenino , Humanos , Animales , Ratones , Transformación Celular Neoplásica/genética , Genes Homeobox , Apoptosis/genética , Neoplasias Endometriales/patología , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , MicroARNs/genética , ATPasas Asociadas con Actividades Celulares Diversas/genética , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Proteínas Portadoras/metabolismo , ADN Helicasas/metabolismo
2.
Funct Integr Genomics ; 23(2): 147, 2023 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-37145301

RESUMEN

Ovarian cancer (OC) has the worst prognosis among gynecological malignancies. Cisplatin (CDDP) is one of the most commonly used treatments for OC, but recurrence and metastasis are common due to endogenous or acquired resistance. High expression of ATP-binding cassette (ABC) transporters is an important mechanism of resistance to OC chemotherapy, but targeting ABC transporters in OC therapy remains a challenge. The expression of sortilin-related receptor 1 (SORL1; SorLA) in the response of OC to CDDP was determined by analysis of TCGA and GEO public datasets. Immunohistochemistry and western blotting were utilized to evaluate the expression levels of SORL1 in OC tissues and cells that were sensitive or resistant to CDDP treatment. The in vitro effect of SORL1 on OC cisplatin resistance was proven by CCK-8 and cell apoptosis assays. The subcutaneous xenotransplantation model verified the in vivo significance of SORL1 in OC. Finally, the molecular mechanism by which SORL1 regulates OC cisplatin resistance was revealed by coimmunoprecipitation, gene set enrichment analysis and immunofluorescence analysis. This study demonstrated that SORL1 is closely related to CDDP resistance and predicts a poor prognosis in OC. In vivo xenograft experiments showed that SORL1 knockdown significantly enhanced the effect of CDDP on CDDP-resistant OC cells. Mechanistically, silencing of SORL1 inhibits the early endosomal antigen 1 (EEA1) pathway, which impedes the stability of ATP-binding cassette B subfamily member 1 (ABCB1), sensitizing CDDP-resistant OC cells to CDDP. The findings of this study suggest that targeting SORL1 may represent a promising therapeutic approach for overcoming CDDP resistance in OC.


Asunto(s)
Cisplatino , Neoplasias Ováricas , Humanos , Femenino , Cisplatino/farmacología , Cisplatino/uso terapéutico , Cisplatino/metabolismo , Resistencia a Antineoplásicos/genética , Línea Celular Tumoral , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Transportadoras de Casetes de Unión a ATP/genética , Adenosina Trifosfato , Proteínas Relacionadas con Receptor de LDL/metabolismo , Proteínas Relacionadas con Receptor de LDL/farmacología , Proteínas Relacionadas con Receptor de LDL/uso terapéutico , Proteínas de Transporte de Membrana , Subfamilia B de Transportador de Casetes de Unión a ATP/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/uso terapéutico
3.
BMC Pregnancy Childbirth ; 21(1): 697, 2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34654394

RESUMEN

BACKGROUND: Cesarean scar pregnancy (CSP) remains a sporadic and special form of ectopic pregnancy in which the fertilized ovum is implanted on a previous cesarean scar within 12 weeks. This study aims to evaluate the optimal time interval between uterine artery embolization (UAE) and curettage modalities in order to provide the best clinical outcomes. METHODS: From January 2018 to December 2020, we recruited 61 patients with CSP. They were randomly divided into two groups depending on whether the time interval between UAE and dilatation and curettage (D&C) requires additional hospitalization: 31 patients received prophylactic UAE followed by D&C on the same day (0-12 h; group A) and 30 patients need hospitalization (12-72 h; group B). The clinical characteristics, diagnostic data, and outcomes of the two groups were compared and analyzed. RESULTS: A total of 59 (96.72%) cases had responded well to the first treatment. One patient in each arm undergone retreatment, but none of the 61 patients needed additional hysterectomy. There was no considerable relationship between the two groups with respect to the intraoperative hemorrhage during D&C, serum index (containing ß-hCG, hemoglobin, CRP, and D-dimer) on the first day after D&C, side effects (containing fever and abdominal pain), renal, hepatic, and coagulation function, time of CSP residual mass disappearance, and hospitalization cost. The time of serum ß-hCG resolution after surgery was 41.22 ± 14.97 days in group A and 66.67 ± 36.64 days in group B (P = 0.027), and group A treatment resulted in a shorten hospital stay as compared with group B (4.81 ± 2.74 days vs. 6.80 ± 2.14 days, P <  0.001). However, the average hourly serum ß-hCG decrease rate within 24 h and the leukocytes on the first day after D&C in group B were superior than in group A (P <  0.050). CONCLUSION: For patients with CSP, UAE followed by D&C on the same day (0-12 h) appears to have more advantages in hospitalization and recovery time, while the long time interval (12-72 h) may have a lower risk of inflammation and a more rapid decrease in serum ß-hCG level within 24 h after D&C surgery. The treatment of CSP should be individualized based on the conditions of patients.


Asunto(s)
Dilatación y Legrado Uterino/métodos , Embarazo Ectópico/terapia , Embolización de la Arteria Uterina/métodos , Adulto , Cesárea/efectos adversos , China , Gonadotropina Coriónica Humana de Subunidad beta/sangre , Cicatriz/etiología , Femenino , Humanos , Histerectomía , Tiempo de Internación , Embarazo , Estudios Prospectivos , Factores de Tiempo , Resultado del Tratamiento
4.
Cancer Sci ; 112(11): 4515-4525, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34490691

RESUMEN

We aimed to identify whether Rho GTPase activating proteins (RhoGAPs) were downregulated in cervical cancers and might be targeted to reduce the growth of cervical cancer using the GEO database and immunohistochemical analysis to identified changes in transcription and protein levels. We analyzed their proliferation, clone formation ability, and their growth as subcutaneous tumors in mice. To detect ARHGAP30 localization in cells, immunofluorescence assays were conducted. Mass spectrometry combined with immunoprecipitation experiments were used to identify binding proteins. Protein interactions were validated with co-immunoprecipitation assays. Western-blot and q-PCR were applied to analyze candidate binding proteins that were associated with ribosome biogenesis. Puromycin incorporation assay was used to detect the global protein synthesis rate. We identified that ARHGAP30 was the only downregulated RhoGAP and was related to the survival of cervical cancer patients. Overexpression of ARHGAP30 in cervical cancer cells inhibited cell proliferation and migration. ARHGAP30 immunoprecipitated proteins were enriched in the ribosome biogenesis process. ARHGAP30 was located in the nucleous and interacted with nucleolin (NCL). Overexpression of ARHGAP30 inhibited rRNA synthesis and global protein synthesis. ARHGAP30 overexpression induced the ubiquitination of NCL and decreased its protein level in Hela cells. The function of ARHGAP30 on cervical cancer cell ribosome biogenesis and proliferation was independent of its RhoGAP activity as assessed with a RhoGAP-deficient plasmid of ARHGAP30R55A . Overall, the findings revealed that ARHGAP30 was frequently downregulated and associated with shorter survival of cervical cancer patients. ARHGAP30 may suppress growth of cervical cancer by reducing ribosome biogenesis and protein synthesis through promoting ubiquitination of NCL.


Asunto(s)
Proliferación Celular , Proteínas Activadoras de GTPasa/metabolismo , Ribosomas/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Animales , Línea Celular Tumoral , Nucléolo Celular/metabolismo , Regulación hacia Abajo , Femenino , Células HeLa , Humanos , Inmunoprecipitación , Espectrometría de Masas , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de Neoplasias/metabolismo , Fosfoproteínas/metabolismo , Biosíntesis de Proteínas , ARN Ribosómico/biosíntesis , Proteínas de Unión al ARN/metabolismo , Ensayo de Tumor de Célula Madre , Ubiquitinación , Neoplasias del Cuello Uterino/mortalidad , Neoplasias del Cuello Uterino/patología , Nucleolina
5.
Cancer Lett ; 513: 36-49, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-33991616

RESUMEN

Endometrial cancer (EC) is becoming one of the most common gynecologic malignancies. Lipid metabolism is a hallmark feature of cancers. The molecular mechanisms underlying lipid metabolism in EC remain unclear. In this study, we revealed that many lipid metabolism-related genes were aberrantly expressed in endometrial cancer tissues, especially ACLY. Upregulated ACLY promoted EC cell proliferation and colony formation, and attenuated apoptosis. Mechanistically, cotreatment with obesity-related factors (estradiol, insulin and leptin) promoted nuclear translocation of ACLY through Akt-mediated phosphorylation of ACLY at Ser455. Nuclear-localized ACLY increased histone acetylation levels, thus resulting in upregulation of pyrimidine metabolism genes, such as DHODH. Moreover, STAT3 altered the ACLY expression at the transcriptional level via directly binding to its promoter region. In conclusion, our findings clarify the roles and mechanisms of ACLY in endometrial cancer and ACLY could link obesity risk factors to the regulation of histone acetylation. We believe that novel therapeutic strategies for EC can be designed by targeting the ACLY axis.


Asunto(s)
ATP Citrato (pro-S)-Liasa/metabolismo , Neoplasias Endometriales/metabolismo , Acetilación , Animales , Línea Celular Tumoral , Proliferación Celular/fisiología , Neoplasias Endometriales/patología , Femenino , Células HEK293 , Xenoinjertos , Humanos , Metabolismo de los Lípidos , Ratones , Persona de Mediana Edad , Pirimidinas/metabolismo , Factor de Transcripción STAT3/metabolismo
6.
Placenta ; 99: 141-151, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32798766

RESUMEN

INTRODUCTION: To gain insight into mechanisms of preeclampsia (PE)-dependent proteinuria, this study focused on whether preeclampsia serum (PES) could induce hyperpermeability in human renal glomerular endothelial cells (HRGECs) via the miRNAs-Caveolin-1 (CAV-1)-dependent pathway. METHODS: Bioinformatics approach was used to identify miRNAs targeting CAV1. Normal pregnancy serum (NPS) and severe PES were used to treat HRGECs monolayer to demonstrate if PES could induce the expression of identified miRNAs. A luciferase reporter assay was used to determine whether CAV1 was a direct target of miR-199a-5p, miR-199b-5p, and miR-204. The relationship between the expression of miR-199a-5p, miR-199b-5p, miR-204, and CAV1 in HRGECs was determined using quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blot. The gain-of-function and loss-of-function experiments were performed on HRGECs to investigate the effects of miR-199a-5p, miR-199b-5p, miR-204 on HRGECs permeability. RESULTS: We identified that CAV1 3'UTR has putative binding sites for miR-199a-5p, miR-199b-5p, and miR-204, whereas miR-199a-5p does not appear to be a direct regulator of CAV1. We detected that PE serum downregulated the expression of miR-199a-5p, miR-199b-5p and miR-204, increased expression of CAV1 and increased cell monolayer permeability in HRGECs. The level of CAV1 and permeability decreased when miR-199b-5p or miR-204, but not miR-199a-5p, were overexpressed. DISCUSSION: miR-199b-5p and miR-204 may play a role in PES-induced increasing permeability of HRGECs by regulating CAV1 expression.


Asunto(s)
Caveolina 1/metabolismo , Permeabilidad de la Membrana Celular/fisiología , Células Endoteliales/metabolismo , Glomérulos Renales/metabolismo , Preeclampsia/sangre , Caveolina 1/genética , Regulación hacia Abajo , Células Endoteliales/citología , Femenino , Humanos , Glomérulos Renales/citología , MicroARNs/genética , MicroARNs/metabolismo , Embarazo
7.
Oncogene ; 39(34): 5633-5648, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32661323

RESUMEN

Cervical cancer (CC) remains highest in the mortality of female reproductive system cancers, while cisplatin (CDDP) resistance is the one of main reasons for the lethality. Preceding evidence has supported that karyopherins are associated with chemoresistance. In this study, we simultaneously compared CDDP-incomplete responders with CDDP-complete responders of CC patients and CDDP-insensitive CC cell lines with CDDP-sensitive group. We finally identified that DNA-PKcs (PRKDC) was related to CDDP sensitivity after overlapping in CC sample tissues and CC cell lines. Further functional assay revealed that targeting PRKDC by shRNA and NU7026 (specific PRKDC inhibitor) could enhance CDDP sensitivity in vitro and in vivo, which was mediated by impairing DNA damage repair pathway in CC. Mechanistically, we found that PRKDC was transcriptionally upregulated by CCAAT/enhancer-binding protein delta (CEBPD), while intriguingly, CDDP treatment strengthened the transcriptional activity of CEBPD to PRKDC. We further disclosed that Importin 4 (IPO4) augmented the nuclear translocation of CEBPD through nuclear localization signals (NLS) to activate PRKDC-mediated DNA damage repair in response to CDDP. Moreover, we demonstrated that IPO4 and CEBPD knockdown improved CDDP-induced cytotoxicity in vitro and in vivo. Together, we shed the novel insight into the role of IPO4 in chemosensitivity and provide a clinical translational potential to enhance CC chemosensitivity since the IPO4-CEBPD-PRKDC axis is actionable via NU7026 (PRKDC inhibitor) or targeting IPO4 in combination with CDDP.


Asunto(s)
Proteína delta de Unión al Potenciador CCAAT/genética , Cisplatino/farmacología , Reparación del ADN/efectos de los fármacos , Proteína Quinasa Activada por ADN/genética , Proteínas de Transporte de Membrana/genética , Neoplasias del Cuello Uterino/genética , Transporte Activo de Núcleo Celular/genética , Animales , Antineoplásicos/farmacología , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Línea Celular Tumoral , Cromonas/farmacología , Daño del ADN , Reparación del ADN/genética , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Proteína Quinasa Activada por ADN/metabolismo , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Proteínas de Transporte de Membrana/metabolismo , Ratones Desnudos , Morfolinas/farmacología , Interferencia de ARN , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/terapia , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
8.
Appl Microbiol Biotechnol ; 104(6): 2481-2488, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32006050

RESUMEN

5-Hydroxytryptophan (5-HTP) is the precursor of the neurotransmitter serotonin and has been used for the treatment of various diseases such as depression, insomnia, chronic headaches, and binge eating associated obesity. The production of 5-HTP had been achieved in our previous report, by the development of a recombinant strain containing two plasmids for biosynthesis of L-tryptophan (L-trp) and subsequent hydroxylation. In this study, the L-trp biosynthetic pathway was further integrated into the E. coli genome, and the promoter strength of 3-deoxy-7-phosphoheptulonate synthase, which catalyzes the first step of L-trp biosynthesis, was engineered to increase the production of L-trp. Hence, the 5-HTP production could be manipulated by the regulation of copy number of L-trp hydroxylation plasmid. Finally, the 5-HTP production was increased to 1.61 g/L in the shaking flasks, which was 24% improvement comparing to the original producing strain, while the content of residual L-trp was successfully reduced from 1.66 to 0.2 g/L, which is beneficial for the downstream separation and purification. Our work shall promote feasible progresses for the industrial production of 5-HTP.


Asunto(s)
5-Hidroxitriptófano/biosíntesis , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Ingeniería Metabólica , Triptófano/biosíntesis , Técnicas de Cultivo Celular por Lotes , Vías Biosintéticas , Escherichia coli/metabolismo , Genoma Bacteriano , Microbiología Industrial , Serotonina/metabolismo
9.
EBioMedicine ; 51: 102609, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31915116

RESUMEN

BACKGROUND: Endometrial carcinoma (EC) is one of the most common gynecological malignancies among women. Maternal embryonic leucine Zipper Kinase (MELK) is upregulated in a variety of human tumors, where it contributes to malignant phenotype and correlates with a poor prognosis. However, the biological function of MELK in EC progression remains largely unknown. METHODS: We explored the MELK expression in EC using TCGA and GEO databases and verified it using clinical samples by IHC methods. CCK-8 assay, colony formation assay, cell cycle assay, wound healing assay and subcutaneous xenograft mouse model were generated to estimate the functions of MELK and its inhibitor OTSSP167. qRT-PCR, western blotting, co-immunoprecipitation, chromatin immunoprecipitation and luciferase reporter assay were performed to uncover the underlying mechanism concerning MELK during the progression of EC. FINDINGS: MELK was significantly elevated in patients with EC, and high expression of MELK was associated with serous EC, high histological grade, advanced clinical stage and reduced overall survival and disease-free survival. MELK knockdown decreased the ability of cell proliferation and migration in vitro and subcutaneous tumorigenesis in vivo. In addition, high expression of MELK could be regulated by transcription factor E2F1. Moreover, we found that MELK had a direct interaction with MLST8 and then activated mTORC1 and mTORC2 signaling pathway for EC progression. Furthermore, OTSSP167, an effective inhibitor, could inhibit cell proliferation driven by MELK in vivo and vitro assays. INTERPRETATION: We have explored the crucial role of the E2F1/MELK/mTORC1/2 axis in the progression of EC, which could be served as potential therapeutic targets for treatment of EC. FUNDING: This research was supported by National Natural Science Foundation of China (No:81672565), the Natural Science Foundation of Shanghai (Grant NO:17ZR1421400 to Dr. Zhihong Ai) and the fundamental research funds for central universities (No: 22120180595).


Asunto(s)
Progresión de la Enfermedad , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Estudios de Cohortes , Factor de Transcripción E2F1/metabolismo , Neoplasias Endometriales/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Modelos Biológicos , Análisis Multivariante , Naftiridinas/farmacología , Pronóstico , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética , Homóloga LST8 de la Proteína Asociada al mTOR/metabolismo
10.
Biochem Biophys Res Commun ; 522(4): 952-959, 2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-31810606

RESUMEN

Gallbladder carcinoma (GBC) is the most aggressive carcinoma of the biliary tract, effective chemotherapy was critical for the patients with unresectable GBC. However, chemotherapy resistance is still problematic for clinicians. Here, we identified a specific long non-coding RNA, SSTR5-AS1, in GBC patient that facilitates gemcitabine resistance. SSTR5-AS1 is significantly increased in GBC samples and cell lines, especially in gemcitabine-resistant cell lines, and higher SSTR5-AS1 expression was correlated with poorer overall survival rate in GBC patients. Our data revealed that upregulated SSTR5-AS1 facilitates gemcitabine resistance via inhibiting apoptosis. Knockdown of SSTR5-AS1 sensitized drug resistant GBC cells to gemcitabine in vitro and strongly inhibited xenografts formed by drug resistant GBC cells in vivo. Moreover, we found via streptavidin pull down assay that NONO specifically binds to sense sequence of SSTR5-AS1 and prevented proteasome mediated NONO degradation, which resulted in increased NONO protein level without affecting the transcription of NONO. NONO functions as the downstream effector of SSTR5-AS1 and is required for SSTR5-AS1 mediated gemcitabine resistance. Collectively, our data provided novel insights into lncRNA-mediated chemotherapy resistance and suggested a novel therapeutic target to improve chemotherapy strategies for unresectable GBC patients.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos/genética , Neoplasias de la Vesícula Biliar/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Desoxicitidina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Ratones Desnudos , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica/efectos de los fármacos , Proteolisis/efectos de los fármacos , ARN Largo no Codificante/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Gemcitabina
11.
J Cell Biochem ; 120(5): 8300-8310, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30548294

RESUMEN

PURPOSE: The dysregulation of long noncoding RNAs (lncRNAs) has been reported to be correlated to carcinogenesis and cancer progression. Endometrial cancer (EC), arising from the endometrium or the inner lining of the uterus, is one of the most common gynecological malignancies. We aim to explore the prognostic value of the lncRNAs in patients with endometrioid endometrial cancer (EEC) and to identify the potential lncRNA signature for predicting survival of patients with EEC. METHODS: We performed a genome-wide analysis of the lncRNA expression profiling in The Cancer Genome Atlas (TCGA)-Uterine Corpus Endometrial Carcinoma database (408 EEC) to identify the prognosis related lncRNAs for the EEC. The patients with EEC were randomly divided into a training set (204 for endometrioid) and a testing set (204 for endometrioid). The lncRNA signature was identified in the training set, and then independently validated in the testing set and the complete set (training set plus testing set). RESULTS: We developed a nine-lncRNA signature-based risk score in the patients with EEC. The risk score based on the novel lncRNAs signature was able to separate patients of training set into high-and low-risk groups with significantly different overall survival and progression-free survival. These can also be successfully confirmed in the testing set and complete set. CONCLUSION: A nine-lncRNA expression signature was identified and validated which can predict EEC patient's survival. These findings may have important implications in the understanding of the potential therapeutic methods for patients with EEC.

12.
J Reprod Immunol ; 129: 1-8, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30007203

RESUMEN

To explore new ideas about the pathogeny of preeclampsia (PE) proteinuria, this study focused on whether severe PE serum (PES) could induce high-molecular-weight protein (HMWP) hyperpermeability in glomerular endothelial cells (GEC) via the HMGB1-Caveolin-1 (CAV-1) pathway. Normal pregnancy serum (NPS) and severe PES were used to treat primary human GEC monolayer for 24 h. The CAV-1 inhibitor methyl-beta-cyclodextrin (MBCD), the HMGB1 inhibitor glycyrrhizicacid (GA), recombinant HMGB1 (rHMGB1) were also used to treat GEC monolayer that were stimulated by NPS or severe PES. The dynamic permeability of GEC to HMWP was detected by Evans blue-labeled BSA and CAV-1 expression in GEC was analyzed by immunofluorescence staining and Western blotting. We detected HMGB1 expression in placenta and serum in normal pregnancy and severe PE. The results showed that severe PES significantly promoted GEC hyperpermeability and CAV-1 expression. By inhibiting CAV-1 expression, MBCD reversed severe PES-induced GEC monolayer permeability. HMGB1 expression in PE placenta and serum was significantly increased. Compared with that in normal placenta, HMGB1expression was increased in the cytoplasm of syncytiotrophoblast cells in PE placenta. GA decreased the severe PES-induced hyperpermeability and CAV-1 expression in GEC. rHMGB1 induced high expression levels of CAV-1 and HMWP hyperpermeability in GEC. In conclusion, HMGB1 is increased in severe PE patients and induces the expression of CAV-1 in GEC. High expression of CAV-1 in GEC can promote HMWP hyperpermeability, which may contribute to the development of PE proteinuria.


Asunto(s)
Caveolina 1/metabolismo , Endotelio Vascular/fisiología , Proteína HMGB1/metabolismo , Placenta/metabolismo , Suero/metabolismo , Adulto , Caveolina 1/antagonistas & inhibidores , Caveolina 1/genética , Permeabilidad de la Membrana Celular , Células Cultivadas , Femenino , Ácido Glicirrínico/farmacología , Proteína HMGB1/antagonistas & inhibidores , Humanos , Preeclampsia , Embarazo , Proteinuria , ARN Interferente Pequeño/genética , Transducción de Señal , Regulación hacia Arriba , beta-Ciclodextrinas/farmacología
13.
Oncogene ; 37(18): 2394-2409, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29429992

RESUMEN

High-risk human papillomavirus oncoproteins E6 and E7 are the major etiological factors of cervical cancer but are insufficient for malignant transformation of cervical cancer. Dysregulated alternative splicing, mainly ascribed to aberrant splicing factor levels and activities, contributes to most cancer hallmarks. However, do E6 and E7 regulate the expression of splicing factors? Does alternative splicing acts as an "accomplice" of E6E7 to promote cervical cancer progression? Here, we identified that the splicing factor SRSF10, which promotes tumorigenesis of cervix, was upregulated by E6E7 via E2F1 transcriptional activation. SRSF10 modulates the alternate terminator of interleukin-1 receptor accessory protein exon 13 to increase production of the membrane form of interleukin-1 receptor accessory protein. SRSF10-mediated mIL1RAP upregulates the expression of the "don't eat me" signal CD47 to inhibit macrophage phagocytosis by promoting nuclear factor-κB activation, which is pivotal in inflammatory, immune, and tumorigenesis processes. Altogether, these data reveal a close relationship among HPV infection, alternative splicing and tumor immune evasion, and also suggests that the SRSF10-mIL1RAP-CD47 axis could be an attractive therapeutic target for the treatment of cervical cancer.


Asunto(s)
Empalme Alternativo/genética , Carcinogénesis/genética , Proteínas de Ciclo Celular/fisiología , Proteína Accesoria del Receptor de Interleucina-1/genética , Proteínas Represoras/fisiología , Factores de Empalme Serina-Arginina/fisiología , Neoplasias del Cuello Uterino/genética , Animales , Antígeno CD47/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Transformación Celular Viral/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Papillomavirus Humano 16/fisiología , Papillomavirus Humano 18/fisiología , Humanos , Ratones , Ratones Desnudos , FN-kappa B/metabolismo , Isoformas de Proteínas/genética , Transducción de Señal/genética , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/virología
14.
Int J Clin Exp Pathol ; 11(8): 3846-3857, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-31949772

RESUMEN

Endometrial cancer (EC) is one of the most important gynecological cancers, but its pathogenesis is not clearly understood, and it also lacks an effective treatment. The nuclear transcriptional protein forkhead box protein M1 (FOXM1) has crucial functions in the development and progression of cancer and is treated as a prognostic biomarker and therapeutic target in many types of cancers. However, the situation and underlying mechanisms of FOXM1's involvement in EC is largely underestimated. In our present study, we found FOXM1 was overexpressed in EC, including endometrioid (EEC) and serous (SEC). High expression of FOXM1 was meaningfully associated with a poor prognosis of EC patients as well as with EC pathological stages and clinical grades. Knocking down FOXM1 could significantly reduce the proliferation and migration capacity of AN3CA and ISHIKAWA cells. Furthermore, our RNA-seq results indicated that the knockdown of FOXM1 mainly affects downstream metabolic genes in EC cells. Finally, we also discovered one potential functional pathway, FOXM1-SLC27A2, which may contribute to EC progression. Taken together, the high expression of FOXM1 is closely associated with the prognosis, pathological stages, and clinical grades of EC patients. FOXM1 can promote the proliferation and migration of EC cells. Through SLC27A2, FOXM1 may influence the metabolic activity of EC cells, and FOXM1-SLC27A signaling could be treated as a potential cellular target for a therapeutic strategy of EC.

15.
Sci Rep ; 7: 41404, 2017 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-28112250

RESUMEN

3ß-Hydroxysteroid-Δ24 reductase (DHCR24), the final enzyme of the cholesterol biosynthetic pathway, has been associated with urogenital neoplasms. However, the function of DHCR24 in endometrial cancer (EC) remains largely elusive. Here, we analyzed the expression profile of DHCR24 and the progesterone receptor (PGR) in our tissue microarray of EC (n = 258), the existing EC database in GEO (Gene Expression Omnibus), and TCGA (The Cancer Genome Atlas). We found that DHCR24 was significantly elevated in patients with EC, and that the up-regulation of DHCR24 was associated with advanced clinical stage, histological grading, vascular invasion, lymphatic metastasis, and reduced overall survival. In addition, DHCR24 expression could be induced by insulin though STAT3, which directly binds to the promoter elements of DHCR24, as demonstrated by ChIP-PCR and luciferase assays. Furthermore, genetically silencing DHCR24 inhibited the metastatic ability of endometrial cancer cells and up-regulated PGR expression, which made cells more sensitive to progestin. Taken together, we have demonstrated for the first time the crucial role of the insulin/STAT3/DHCR24/PGR axis in the progression of EC by modulating the metastasis and progesterone response, which could serve as potential therapeutic targets for the treatment of EC with progesterone receptor loss.


Asunto(s)
Neoplasias Endometriales/enzimología , Neoplasias Endometriales/patología , Endometrio/anomalías , Insulina/efectos adversos , Proteínas del Tejido Nervioso/biosíntesis , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/biosíntesis , Regulación hacia Arriba/genética , Enfermedades Uterinas/enzimología , Anciano , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/genética , Endometrio/enzimología , Endometrio/patología , Inducción Enzimática/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Acetato de Medroxiprogesterona/farmacología , Acetato de Medroxiprogesterona/uso terapéutico , Persona de Mediana Edad , Invasividad Neoplásica , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Pronóstico , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Factor de Transcripción STAT3/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Enfermedades Uterinas/genética , Enfermedades Uterinas/patología
16.
Int J Clin Exp Med ; 8(4): 4954-67, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26131068

RESUMEN

Growth hormone (GH) is involved in the regulation of male and female infertility. Several clinical studies reveal that adjuvant GH treatment has a possible role in vitro fertilization (IVF) or intracytoplasmic sperm injection (ICSI), especially in poor ovarian responders (POR) undergoing IVF/ICSI. Recent studies suggest that GH addition in POR patients significantly improves the rate of clinical pregnancy and live birth. Databases including PubMed, Embase, the Cochrane Central China National Knowledge Infrastructure (CNKI) and Google Scholar were searched for randomized controlled trials (RCTs) or controlled clinical trials (CCTs) on the effectiveness of GH supplementation with gonadotrophins in IVF/ICSI for POR. Two reviewers independently screened literature according to the inclusion and exclusion criteria, extracted data, and assessed methodological quality. Meta Analyst Beta 3.13 software was used to meta-analysis. Eleven studies (six RCTs and five CCTs) and 3788 subjects (613 subjects in cases group and 3175 subjects in controls group) were included in our study. The results of meta-analysis showed that GH addition significantly increased serum E2 level on the day of HCG (OR = 0.55; 95% CI = 0.127-0.973) and MII oocyte number (OR = 0.827; 95% CI = 0.470-1.184). Furthermore, GH addition significantly improved the number of 2PN (OR = 0.934; 95% CI = 0.206-1.661) and obtained embryos (OR = 0.934; 95% CI = 0.206-1.661). However, no significant difference was found for the overall implantation rate was 8.8% (95% CI = -0.062-0.237) and clinical pregnancy rate was 5.1% (95% CI = -0.033-0.134). The present result revel that GH supplementation for IVF/ICSI in POR increases the probability of serum E2 level on the day of HCG, the number of MII oocyte, 2PN and obtained embryos. However, GH addition does not increase implantation rate and clinical pregnancy rates. Due to the limited quantity and quality of the included studies as well as the difference in methodology, we suggest this above could be taken as a reference for clinical analysis which needs to be further evaluated in its effects.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA