Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
J Cell Biol ; 223(2)2024 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-38241019

RESUMEN

Exportin receptors are concentrated in the nucleus to transport essential cargoes out of it. A mislocalization of exportins to the cytoplasm is linked to disease. Hence, it is important to understand how their containment within the nucleus is regulated. Here, we have studied the nuclear efflux of exportin2 (cellular apoptosis susceptibility protein or CAS) that delivers karyopherinα (Kapα or importinα), the cargo adaptor for karyopherinß1 (Kapß1 or importinß1), to the cytoplasm in a Ran guanosine triphosphate (RanGTP)-mediated manner. We show that the N-terminus of CAS attenuates the interaction of RanGTPase activating protein 1 (RanGAP1) with RanGTP to slow GTP hydrolysis, which suppresses CAS nuclear exit at nuclear pore complexes (NPCs). Strikingly, a single phosphomimetic mutation (T18D) at the CAS N-terminus is sufficient to abolish its nuclear retention and coincides with metastatic cellular behavior. Furthermore, downregulating Kapß1 disrupts CAS nuclear retention, which highlights the balance between their respective functions that is essential for maintaining the Kapα transport cycle. Therefore, NPCs play a functional role in selectively partitioning exportins in the cell nucleus.


Asunto(s)
Núcleo Celular , Proteína de Susceptibilidad a Apoptosis Celular , Carioferinas , Proteína de Unión al GTP ran , Transporte Activo de Núcleo Celular/fisiología , Transporte Biológico , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Carioferinas/metabolismo , Poro Nuclear/metabolismo , Proteína de Unión al GTP ran/metabolismo , Humanos , Proteína de Susceptibilidad a Apoptosis Celular/genética , Proteína de Susceptibilidad a Apoptosis Celular/metabolismo
2.
Cell Prolif ; 55(5): e13226, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35403306

RESUMEN

Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer. However, the treatment regimens for TNBC are limited. Chromosome segregation 1-like (CSE1L), also called cellular apoptosis susceptibility protein (CAS), is highly expressed in breast cancer and plays a crucial role in the progression of various tumours. However, the involvement of CAS in TNBC remains elusive. In this study, we showed that the expression of CAS was higher in TNBC samples than in non-TNBC samples in the Gene Expression Omnibus database. Knockdown of CAS inhibited MDA-MB-231 cell growth, migration and invasion. Further RNA-seq analysis revealed that complement pathway activity was significantly elevated. Of note, complement component 3 (C3), the key molecule in the complement pathway, was significantly upregulated, and the expression of C3 was negatively correlated with that of CAS in breast cancer. Lower C3 expression was related to poor prognosis. Interestingly, the expression level of C3 was positively correlated with the infiltration of multiple immune cells. Taken together, our findings suggest that CAS participates in the development of TNBC through C3-mediated immune cell suppression and might constitute a potential therapeutic target for TNBC.


Asunto(s)
Complemento C3/metabolismo , Neoplasias de la Mama Triple Negativas , Mama/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Proteína de Susceptibilidad a Apoptosis Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias de la Mama Triple Negativas/patología
3.
Int J Oncol ; 60(6)2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35417036

RESUMEN

The chromosome segregation 1­like (CSE1L) protein, which regulates cellular mitosis and apoptosis, was previously found to be overexpressed in colorectal cancer (CRC) cells harboring mutations. Therefore, regulating CSE1L expression may confer chemotherapeutic effects against CRC. The gut microflora can regulate gene expression in colonic cells. In particular, metabolites produced by the gut microflora, including the short­chain fatty acid butyrate, have been shown to reduce CRC risk. Butyrates may exert antioncogenic potential in CRC cells by modulating p53 expression. The present study evaluated the association between CSE1L expression and butyrate treatment from two non­transformed colon cell lines (CCD­18Co and FHC) and six CRC cell lines (LS 174T, HCT116 p53+/+, HCT116 p53­/­, Caco­2, SW480 and SW620). Lentiviral knockdown of CSE1L and p53, reverse transcription­quantitative PCR (CSE1L, c­Myc and p53), western blotting [CSE1L, p53, cyclin (CCN) A2, CCNB2 and CCND1], wound healing assay (cell migration), flow cytometry (cell cycle analysis) and immunofluorescence staining (CSE1L and tubulin) were adopted to verify the effects of butyrate on CSE1L­expressing CRC cells. The butyrate­producing gut bacteria Butyricicoccus pullicaecorum was administered to mice with 1,2­dimethylhydrazine­induced colon tumors before the measurement of CSE1L expression. The effects of B. pullicaecorum on CSE1L expression were then assessed by immunohistochemical staining for CSE1L and p53 in tissues from CRC­bearing mice. Non­cancerous colon cells with the R273H p53 mutation or CRC cells haboring p53 mutations were found to exhibit significantly higher CSE1L expression levels. CSE1L knockdown in HCT116 p53­/­ cells resulted in G1­and G2/M­phase cell cycle arrest. Furthermore, in HCT116 p53­/­ cells, CSE1L expression was already high at interphase, increased at prophase, peaked during metaphase before declining at cytokinesis but remained relatively high compared with that in HCT116 expressing wild­type p53. Significantly decreased expression levels of CSE1L were also observed in HCT116 p53­/­ cells that were treated with butyrate for 24 h. In addition, the migration of HCT116 p53­/­ cells was significantly decreased after CSE1L knockdown or butyrate treatment. Tumors with more intense nuclear p53 staining and weaker CSE1L staining were found in mice bearing DMH/DSS­induced CRC that were administered with B. pullicaecorum. Taken together, the results indicated that butyrate can impair CSE1L­induced tumorigenic potential. In conclusion, butyrate­producing microbes, such as B. pullicaecorum, may reverse the genetic distortion caused by p53 mutations in CRC by regulating CSE1L expression levels.


Asunto(s)
Butiratos , Proteína de Susceptibilidad a Apoptosis Celular , Neoplasias Colorrectales , Proteína p53 Supresora de Tumor , Animales , Apoptosis , Butiratos/farmacología , Células CACO-2 , Proliferación Celular , Proteína de Susceptibilidad a Apoptosis Celular/genética , Segregación Cromosómica , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Suplementos Dietéticos , Células HCT116 , Humanos , Ratones , Mutación , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
4.
Cell Death Dis ; 13(1): 45, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013112

RESUMEN

PHY34 is a synthetic small molecule, inspired by a compound naturally occurring in tropical plants of the Phyllanthus genus. PHY34 was developed to have potent in vitro and in vivo anticancer activity against high grade serous ovarian cancer (HGSOC) cells. Mechanistically, PHY34 induced apoptosis in ovarian cancer cells by late-stage autophagy inhibition. Furthermore, PHY34 significantly reduced tumor burden in a xenograft model of ovarian cancer. In order to identify its molecular target/s, we undertook an unbiased approach utilizing mass spectrometry-based chemoproteomics. Protein targets from the nucleocytoplasmic transport pathway were identified from the pulldown assay with the cellular apoptosis susceptibility (CAS) protein, also known as CSE1L, representing a likely candidate protein. A tumor microarray confirmed data from mRNA expression data in public databases that CAS expression was elevated in HGSOC and correlated with worse clinical outcomes. Overexpression of CAS reduced PHY34 induced apoptosis in ovarian cancer cells based on PARP cleavage and Annexin V staining. Compounds with a diphyllin structure similar to PHY34 have been shown to inhibit the ATP6V0A2 subunit of V(vacuolar)-ATPase. Therefore, ATP6V0A2 wild-type and ATP6V0A2 V823 mutant cell lines were tested with PHY34, and it was able to induce cell death in the wild-type at 246 pM while the mutant cells were resistant up to 55.46 nM. Overall, our data demonstrate that PHY34 is a promising small molecule for cancer therapy that targets the ATP6V0A2 subunit to induce autophagy inhibition while interacting with CAS and altering nuclear localization of proteins.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Núcleo Celular/metabolismo , Proteína de Susceptibilidad a Apoptosis Celular/metabolismo , Cistadenocarcinoma Seroso/metabolismo , Neoplasias Ováricas/metabolismo , ATPasas de Translocación de Protón/antagonistas & inhibidores , Transporte Activo de Núcleo Celular/efectos de los fármacos , Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proteína de Susceptibilidad a Apoptosis Celular/genética , Cistadenocarcinoma Seroso/tratamiento farmacológico , Cistadenocarcinoma Seroso/patología , Femenino , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Phyllanthus/química , Pronóstico
5.
Bioengineered ; 12(1): 6967-6980, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34516344

RESUMEN

MicroRNA-451a (miR-451a) has been implicated in the initiation and progression of multiple cancers. However, the regulatory mechanisms underlying its function in nasopharyngeal carcinoma (NPC) are poorly understood. Thus, we investigated in detail the role of the microRNA-451a/chromosome segregation 1-like (miR-45a/CSE1L) axis and its regulatory mechanism in NPC. We examined the levels of miR-451a and CSE1L in NPC, and assessed the effects of miR-451a and CSE1L on NPC by cell functional experiments. Furthermore, we elucidated the direct regulatory effect of miR-451a on CSE1L by the luciferase reporter assay, RNA pull-down assay, and RNA immunoprecipitation and validated our observations by calculating the Pearson's correlation coefficient. We found that miR-451a was down-regulated in NPC cells, and its over-expression attenuated cell proliferation, migration, and invasion, and tumor growth in 5-8 F and SUNE-1 cells and promoted apoptosis. Moreover, CSE1L was the direct gene target of miR-451a, and its over-expression abrogated miR-451a-dependent inhibition of malignancy in 5-8 F and SUNE-1 cells. The Pearson's correlation coefficient indicated a negative correlation between CSE1L and miR-451a. miR-451a serves as a tumor suppressor and targets CSE1L. miR-451a suppresses CSE1L expression, thereby reducing proliferation, invasion, and migration and increasing apoptosis of NPC cells.


Asunto(s)
Proteína de Susceptibilidad a Apoptosis Celular/genética , MicroARNs/genética , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas , Adulto , Animales , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Proteína de Susceptibilidad a Apoptosis Celular/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , MicroARNs/metabolismo , Persona de Mediana Edad , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/metabolismo , Carcinoma Nasofaríngeo/patología , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Transcriptoma/genética
6.
BMC Cancer ; 21(1): 175, 2021 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-33602168

RESUMEN

BACKGROUND: Apoptosis-related genes(Args)play an essential role in the occurrence and progression of hepatocellular carcinoma(HCC). However, few studies have focused on the prognostic significance of Args in HCC. In the study, we aim to explore an efficient prognostic model of Asian HCC patients based on the Args. METHODS: We downloaded mRNA expression profiles and corresponding clinical data of Asian HCC patients from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) databases. The Args were collected from Deathbase, a database related to cell death, combined with the research results of GeneCards、National Center for Biotechnology Information (NCBI) databases and a lot of literature. We used Wilcoxon-test and univariate Cox analysis to screen the differential expressed genes (DEGs) and the prognostic related genes (PRGs) of HCC. The intersection genes of DEGs and PGGs were seen as crucial Args of HCC. The prognostic model of Asian HCC patients was constructed by least absolute shrinkage and selection operator (lasso)- proportional hazards model (Cox) regression analysis. Kaplan-Meier curve, Principal Component Analysis (PCA) analysis, t-distributed Stochastic Neighbor Embedding (t-SNE) analysis, risk score curve, receiver operating characteristic (ROC) curve, and the HCC data of ICGC database and the data of Asian HCC patients of Kaplan-Meier plotter database were used to verify the model. RESULTS: A total of 20 of 56 Args were differentially expressed between HCC and adjacent normal tissues (p < 0.05). Univariate Cox regression analysis showed that 10 of 56 Args were associated with survival time and survival status of HCC patients (p < 0.05). There are seven overlapping genes of these 20 and 10 genes, including BAK1, BAX, BNIP3, CRADD, CSE1L, FAS, and SH3GLB1. Through Lasso-Cox analysis, an HCC prognostic model composed of BAK1, BNIP3, CSE1L, and FAS was constructed. Kaplan-Meier curve, PCA, t-SNE analysis, risk score curve, ROC curve, and secondary verification of ICGC database and Kaplan-Meier plotter database all support the reliability of the model. CONCLUSIONS: Lasso-Cox regression analysis identified a 4-gene prognostic model, which integrates clinical and gene expression and has a good effect. The expression of Args is related to the prognosis of HCC patients, but the specific mechanism remains to be further verified.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/genética , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Pueblo Asiatico/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/metabolismo , Proteína de Susceptibilidad a Apoptosis Celular/genética , Proteína de Susceptibilidad a Apoptosis Celular/metabolismo , Biología Computacional/métodos , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Pronóstico , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Receptor fas/genética , Receptor fas/metabolismo
7.
Eur Rev Med Pharmacol Sci ; 24(10): 5429-5435, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32495878

RESUMEN

OBJECTIVE: CSE1L (human chromosomal segregation 1-like) is reported to be able to affect cell apoptosis, invasiveness, and migration. The purpose of this study was to uncover the regulatory effects of CSE1L on cell phenotypes of oral cancer and the underlying mechanism. MATERIALS AND METHODS: CSE1L levels in oral cancer cells were determined by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR) and Western blot. CSE1L overexpression and knockdown models were constructed in CAL-27 and HN6 cells, respectively. Changes in proliferative and migratory abilities in oral cancer cells affected by CSE1L and microphthalmia-associated transcription factor (MITF) were assessed by cell counting kit-8 (CCK-8), 5-Ethynyl-2'-deoxyuridine (EdU) and wound healing assay. Meanwhile, potential influences of CSE1L and MITF on relative levels of E-cadherin and Vimentin in oral cancer cells were detected. Finally, regulatory effects of CSE1L and MITF on the Akt/mTOR pathway were evaluated by detecting expression levels of p-Akt, Akt, p-mTOR, and mTOR. RESULTS: CSE1L was upregulated in oral cancer cells. Knockdown of CSE1L in HN6 cells attenuated proliferative and migratory abilities, as well as downregulated Vimentin and upregulated E-cadherin. Overexpression of CSE1L in CAL-27 cells yielded the opposite results. MIFT level was positively regulated by CSE1L. Overexpression of MITF partially reversed regulatory effects of CSE1L on proliferative ability of oral cancer cells. Moreover, silence of CSE1L suppressed the Akt/mTOR pathway, which was reversed by overexpression of MITF. CONCLUSIONS: CSE1L promotes the proliferative and migratory abilities in oral cancer cells by positively regulating MITF, thus activating the Akt/mTOR pathway.


Asunto(s)
Movimiento Celular , Proteína de Susceptibilidad a Apoptosis Celular/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , Neoplasias de la Boca/metabolismo , Proliferación Celular , Proteína de Susceptibilidad a Apoptosis Celular/genética , Humanos , Factor de Transcripción Asociado a Microftalmía/genética , Neoplasias de la Boca/patología , Células Tumorales Cultivadas
8.
Med Oncol ; 37(5): 43, 2020 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-32270348

RESUMEN

Breast cancer is the most common cancer in women. Although several studies demonstrated cellular apoptosis susceptibility protein (CAS) involved in the development of breast cancer, the underlying mechanisms of CAS regulating cell processes in the breast cancer remain elusive. In the present study, we explored the possible mechanism of CAS in contributing to the cell proliferation in the breast cancer cell line MCF-7. Knockdown of CAS led to the reduction of cell viability and proliferation. Furthermore, cell cycle was arrested in G0/G1 phase after knocking down CAS with the decrease of cyclinD1. In addition, RNA-seq analysis for the CAS knockdown cells demonstrated that total eleven genes were significantly altered (Fold changes > 2). Of note, the expression of cyp24a1 was dramatically increased in the shCAS cells compared to that of shNC cells as well as confirmed by quantitative real-time polymerase chain reaction (qPCR). These observations clarified the previous conflicting results on the cell fates of the breast cells regulated by CAS and provide new insight into the role of CAS in the development of breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , Proliferación Celular/genética , Proteína de Susceptibilidad a Apoptosis Celular/metabolismo , Vitamina D3 24-Hidroxilasa/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Ciclo Celular/genética , Supervivencia Celular/genética , Proteína de Susceptibilidad a Apoptosis Celular/genética , Bases de Datos Genéticas , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Pronóstico
9.
Cell Prolif ; 52(2): e12549, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30485574

RESUMEN

OBJECTIVES: CSE1L has been reported to be highly expressed in various tumours. Testicular germ cell tumours are common among young males, and seminoma is the major type. However, whether CSE1L has functions in the seminoma is unclear. MATERIALS AND METHODS: The expression of CSE1L was detected by immunohistochemistry in seminoma tissues and non-tumour normal testis tissues from patients. CSE1L distribution during cell mitosis was determined by immunofluorescent staining with CSE1L, α-tubulin and γ-tubulin antibodies. The effects of Cse1L knockdown on cell proliferation and cell cycle progression were determined by Cell Counting Kit-8 assay, flow cytometry, PH3 staining and bromodeoxyuridine incorporation assay. RESULTS: CSE1L was significantly enriched in the seminoma tissue compared with the non-tumour normal testis tissue. CSE1L also co-localized with α-tubulin in the cells with a potential to divide. In the seminoma cell line TCam-2, CSE1L was associated with the spindles and the centrosomes during cell division. The knockdown of CSE1L in TCam-2 cells attenuated the cells' proliferative capacity. Cell cycle assay revealed that the CSE1L-deficient cells were mainly arrested in the G0/G1 phase and moderately delayed in the G2/M phase. The proportion of cells with multipolar spindle and abnormal spindle geometry was obviously increased by CSE1L expression silencing in the TCam-2 cells. CONCLUSIONS: Overall, these findings showed that CSE1L plays a pivotal role in maintaining cell proliferation and cell division in seminomas.


Asunto(s)
Proteína de Susceptibilidad a Apoptosis Celular/metabolismo , Mitosis , Seminoma/metabolismo , Neoplasias Testiculares/metabolismo , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Proteína de Susceptibilidad a Apoptosis Celular/análisis , Proteína de Susceptibilidad a Apoptosis Celular/genética , Eliminación de Gen , Humanos , Masculino , Seminoma/genética , Seminoma/patología , Neoplasias Testiculares/genética , Neoplasias Testiculares/patología , Testículo/metabolismo , Testículo/patología
10.
Aging (Albany NY) ; 10(8): 2062-2078, 2018 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-30144787

RESUMEN

Colorectal cancer (CRC) is the third most common malignancy in the United States. Chemotherapeutic resistance is a massive obstacle for cancer treatment. The roles and molecular basis of long non-coding RNA BRAF-activated noncoding RNA (BANCR) in CRC progression and adriamycin (ADR) resistance have not been extensively identified. In this study, we found that BANCR and CSE1L expressions were upregulated in CRC tumor tissues. Meanwhile, CSE1L expression was correlated with depth of CRC. BANCR silencing suppressed cell proliferation and invasion capacity, increased apoptotic rate and potentiated cell sensitivity to ADR. CSE1L downregulation triggered a reduction of cell proliferation and invasion ability, and an increase of apoptosis rate and cell sensitivity to ADR. CSE1L overexpression attenuated si-BANCR-mediated anti-proliferation, anti-invasion and pro-apoptosis effects in CRC cells. BANCR acted as a molecular sponge of miR-203 to sequester miR-203 away from CSE1L in CRC cells, resulting in the upregulation of CSE1L expression. CSE1L knockdown inhibited expressions of DNA-repair-related proteins (53BP1 and FEN1) in HCT116 cells. BANCR knockdown also inhibited tumor growth and enhanced ADR sensitivity in CRC mice model. In conclusion, BANCR knockdown suppressed CRC progression and strengthened chemosensitization of CRC cells to ADR possibly by regulating miR-203/CSE1L axis, indicating that BANCR might be a promising target for CRC treatment.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , ARN Largo no Codificante/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Proteína de Susceptibilidad a Apoptosis Celular/genética , Proteína de Susceptibilidad a Apoptosis Celular/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , MicroARNs , Neoplasias Experimentales , ARN Largo no Codificante/genética , Distribución Aleatoria
11.
Am J Pathol ; 188(7): 1597-1607, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29630856

RESUMEN

Epithelial-to-mesenchymal transition (EMT) is postulated to be a prerequisite for the establishment of endometriosis (EMS), a common reproductive disorder in women. Our previous studies have demonstrated the elevated expression of transmembrane glycoprotein CD147 and its prosurvival effect on abnormal cells in endometriosis. Intriguingly, CD147 is known to promote EMT in cancers. However, the involvement of CD147 in EMT during the establishment of endometriosis remains incompletely understood. We found that CD147 promotes EMT in human endometrial adenocarcinoma cell line Ishikawa. We identified a novel CD147-interacting partner, cellular apoptosis susceptibility protein (CAS), which stabilized the interaction between E-cadherin (E-cad) and ß-catenin (ß-cat) by forming the CAS/E-cad/ß-cat complex. Down-regulation of CAS led to the release and nuclear translocation of ß-cat from E-cad, resulting in the overexpression of the EMT-promoting gene SNAIL. Interestingly, overexpression of CD147 impaired the interaction between CAS and E-cad and triggered the release of ß-cat from the CAS/E-cad/ß-cat complex, which in turn led to EMT. Furthermore, CAS was down-regulated in EMS, with elevated levels of CD147 and nuclear ß-cat. These findings suggest a previously undefined role of CAS in regulating EMT and reveal the involvement of a CD147-induced EMT signaling pathway in pathogenic progression of EMS.


Asunto(s)
Antígenos CD/metabolismo , Basigina/metabolismo , Cadherinas/metabolismo , Proteína de Susceptibilidad a Apoptosis Celular/metabolismo , Neoplasias Endometriales/patología , Endometriosis/patología , Transición Epitelial-Mesenquimal , beta Catenina/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adulto , Antígenos CD/genética , Basigina/genética , Cadherinas/genética , Estudios de Casos y Controles , Membrana Celular , Proteína de Susceptibilidad a Apoptosis Celular/genética , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Endometriosis/genética , Endometriosis/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Persona de Mediana Edad , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal , Células Tumorales Cultivadas , beta Catenina/genética
12.
Proc Natl Acad Sci U S A ; 115(17): E4013-E4022, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29636421

RESUMEN

Epigenetic silencing can be mediated by various mechanisms, and many regulators remain to be identified. Here, we report a genome-wide siRNA screening to identify regulators essential for maintaining gene repression of a CMV promoter silenced by DNA methylation. We identified CSE1L (chromosome segregation 1 like) as an essential factor for the silencing of the reporter gene and many endogenous methylated genes. CSE1L depletion did not cause DNA demethylation. On the other hand, the methylated genes derepressed by CSE1L depletion largely overlapped with methylated genes that were also reactivated by treatment with histone deacetylase inhibitors (HDACi). Gene silencing defects observed upon CSE1L depletion were linked to its nuclear import function for certain protein cargos because depletion of other factors involved in the same nuclear import pathway, including KPNAs and KPNB1 proteins, displayed similar derepression profiles at the genome-wide level. Therefore, CSE1L appears to be critical for the nuclear import of certain key repressive proteins. Indeed, NOVA1, HDAC1, HDAC2, and HDAC8, genes known as silencing factors, became delocalized into cytosol upon CSE1L depletion. This study suggests that the cargo specificity of the protein nuclear import system may impact the selectivity of gene silencing.


Asunto(s)
Núcleo Celular/metabolismo , Proteína de Susceptibilidad a Apoptosis Celular/metabolismo , Silenciador del Gen/fisiología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/genética , Línea Celular , Núcleo Celular/genética , Proteína de Susceptibilidad a Apoptosis Celular/genética , Metilación de ADN/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Estudio de Asociación del Genoma Completo , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Antígeno Ventral Neuro-Oncológico , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , beta Carioferinas/genética , beta Carioferinas/metabolismo
13.
Clin. transl. oncol. (Print) ; 19(5): 562-570, mayo 2017. graf
Artículo en Inglés | IBECS | ID: ibc-162189

RESUMEN

Objective. Recent studies have identified Engrailed-2 (EN-2), a homeobox-containing transcription factor, as a candidate oncogene in prostate cancer (PC). Therapeutic targeting on EN-2, however, is limited because the mechanism underlying EN-2 overexpression in prostatic cancer cells is unknown. This study was to investigate the potential regulatory role of miR-33a on EN-2 expression and explore this signaling axis in ability of prostate cancer survival and metastasis. Methods. The relative expression of miR-33a and EN-2 in paired prostate cancer tissue and adjacent normal tissue as well as in prostate cancer cell lines, PC3 and DU145, was determined using quantitative real-time PCR or western blot, respectively. Cells survival, migration and invasion were evaluated by assays of MTT, TUNEL and Boyden chamber assays, respectively. Direct regulation of EN-2 by miR-33a was examined by luciferase reporter assay. Results. The data showed that miR-33a was upregulated and EN-2 was downregulated in both prostate cancer tissue and prostate cancer cells. miR-33a overexpression suppresses prostate cancer cell survival and metastasis. miR-33a can directly act on EN-2 expression by binding to 3′UTR of its mRNA. Also, miR-33a negatively regulated EN-2 mRNA and protein expression. In pcDNA-EN-2 and miR-33a mimic co-transfected PC3 and DU145 cells, EN-2 overexpression reverses the anti-cell survival and metastasis actions of miR-33a overexpression. The pivotal role of miR-33a in inhibiting prostate tumor growth was confirmed in xenograft models of prostate cancer. Conclusion. Our data suggest that the functional interaction of miR-33a and EN-2 is involved in tumorigenesis of prostate cancer. Also in this process EN-2 serves as a negative responder for miR-33a (AU)


No disponible


Asunto(s)
Humanos , Masculino , Neoplasias de la Próstata/complicaciones , Neoplasias de la Próstata/patología , Metástasis de la Neoplasia/fisiopatología , ARN Mensajero/genética , Proteína de Susceptibilidad a Apoptosis Celular/genética , Movimiento Celular/genética , Biomarcadores de Tumor/genética , Neoplasias de la Próstata/genética , Reacción en Cadena de la Polimerasa/tendencias , ARN Mensajero/metabolismo , Western Blotting , Luciferasas/análisis , Análisis de Varianza , Represión Epigenética/genética , Proto-Oncogenes/genética , Proteínas Oncogénicas/genética
14.
Sci Rep ; 7: 43893, 2017 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-28272419

RESUMEN

Influenza viruses exploit host factors to successfully replicate in infected cells. Using small interfering RNA (siRNA) technology, we identified six human genes required for influenza A virus (IAV) replication. Here we focused on the role of acid phosphatase 2 (ACP2), as its knockdown showed the greatest inhibition of IAV replication. In IAV-infected cells, depletion of ACP2 resulted in a significant reduction in the expression of viral proteins and mRNA, and led to the attenuation of virus multi-cycle growth. ACP2 knockdown also decreased replication of seasonal influenza A and B viruses and avian IAVs of the H7 subtype. Interestingly, ACP2 depletion had no effect on the replication of Ebola or hepatitis C virus. Because ACP2 is known to be a lysosomal acid phosphatase, we assessed the role of ACP2 in influenza virus entry. While neither binding of the viral particle to the cell surface nor endosomal acidification was affected in ACP2-depleted cells, fusion of the endosomal and viral membranes was impaired. As a result, downstream steps in viral entry were blocked, including nucleocapsid uncoating and nuclear import of viral ribonucleoproteins. Our results established ACP2 as a necessary host factor for regulating the fusion step of influenza virus entry.


Asunto(s)
Fosfatasa Ácida/metabolismo , Virus de la Influenza A/fisiología , Virus de la Influenza B/fisiología , Células A549 , Fosfatasa Ácida/antagonistas & inhibidores , Fosfatasa Ácida/genética , Animales , Proteína de Susceptibilidad a Apoptosis Celular/antagonistas & inhibidores , Proteína de Susceptibilidad a Apoptosis Celular/genética , Proteína de Susceptibilidad a Apoptosis Celular/metabolismo , Perros , Endosomas/metabolismo , Humanos , Subtipo H7N7 del Virus de la Influenza A/fisiología , Células de Riñón Canino Madin Darby , Microscopía Fluorescente , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Internalización del Virus , Replicación Viral
15.
Am J Pathol ; 186(10): 2761-8, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27521996

RESUMEN

Human cellular apoptosis susceptibility (chromosomal segregation 1-like, CSE1L) gene plays a role in nuclear-to-cytoplasm transport and chromosome segregation during mitosis, cellular proliferation, and apoptosis. CSE1L is involved in colon carcinogenesis. CSE1L gene expression was assessed with three data sets using Affymetrix U133 + gene chips on normal human colonic mucosa (NR), adenomas (ADs), and colorectal carcinoma (CRC). CSE1L protein expression in CRC, AD, and NR from the same patients was measured by immunohistochemistry using a tissue microarray. We evaluated CSE1L expression in CRC cells (HCT116, SW480, and HT29) and its biological functions. CSE1L mRNA was significantly increased in all AD and CRC compared with NR (P < 0.001 and P = 0.02, respectivly). We observed a change in CSE1L staining intensity and cellular localization by immunohistochemistry. CSE1L was significantly increased during the transition from AD to CRC when compared with NR in a CRC tissue microarray (P = 0.01 and P < 0.001). HCT116, SW480, and HT29 cells also expressed CSE1L protein. CSE1L knockdown by shRNA inhibited protein, resulting in decreased cell proliferation, reduced colony formation in soft agar, and induction of apoptosis. CSE1L protein is expressed early and across all stages of CRC development. shRNA knockdown of CSE1L was associated with inhibition of tumorigenesis in CRC cells. CSE1L may represent a potential target for treatment of CRC.


Asunto(s)
Adenoma/patología , Carcinogénesis/genética , Proteína de Susceptibilidad a Apoptosis Celular/genética , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Adenoma/genética , Adenoma/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis/genética , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular , Proteína de Susceptibilidad a Apoptosis Celular/metabolismo , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Citoplasma/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Transporte de Proteínas , Análisis de Matrices Tisulares , Adulto Joven
16.
Exp Cell Res ; 319(17): 2627-36, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23948303

RESUMEN

The human homolog of the yeast cse1 gene (CSE1L) is over-expressed in ovarian cancer. CSE1L forms complex with Ran and importin-α and has roles in nucleocytoplasmic traffic and gene expression. CSE1L accumulated in the nucleus of ovarian cancer cell lines, while it was localized also in the cytoplasm of other cancer cell lines. Nuclear localization depended on AKT, which was constitutively active in ovarian cancer cells, as the CSE1L protein translocated to the cytoplasm when AKT was inactivated. Moreover, the expression of a constitutively active AKT forced the translocation of CSE1L from the cytoplasm to the nucleus in other cancer cells. Nuclear accrual of CSE1L was associated to the nuclear accumulation of the phosphorylated Ran Binding protein 3 (RanBP3), which depended on AKT as well. Also in samples of human ovarian cancer, AKT activation was associated to nuclear accumulation of CSE1L and phosphorylation of RanBP3. Expression profiling of ovarian cancer cells after CSE1L silencing showed that CSE1L was required for the expression of genes promoting invasion and metastasis. In agreement, CSE1L silencing impaired motility and invasiveness of ovarian cancer cells. Altogether these data show that in ovarian cancer cells activated AKT by affecting RanBP3 phosphorylation determines the nuclear accumulation of CSE1L and likely the nuclear concentration of transcription factors conveying pro-oncogenic signals.


Asunto(s)
Núcleo Celular/metabolismo , Proteína de Susceptibilidad a Apoptosis Celular/metabolismo , Neoplasias Ováricas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Activación Transcripcional , Transporte Activo de Núcleo Celular , Línea Celular Tumoral , Movimiento Celular , Proteína de Susceptibilidad a Apoptosis Celular/genética , Citoplasma/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Nucleares/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Fosforilación , Transcripción Genética
17.
Am J Surg ; 206(3): 418-27, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23806821

RESUMEN

BACKGROUND: Ras plays an important role in colorectal cancer progression. CSE1L (chromosome segregation 1-like) gene maps to 20q13, a chromosomal region that correlates with colorectal cancer development. We investigated the association of CSE1L with Ras in colorectal cancer progression. METHODS: The effect of CSE1L on metastasis-stimulating activity of Ras was studied in an animal model with tumor cells expressing CSE1L-specific shRNA and v-H-Ras. CSE1L expression was evaluated by the immunohistochemical analysis of 127 surgically resected colorectal tumors. K-Ras mutations were analyzed by direct sequencing. RESULTS: CSE1L knockdown reduced Ras-induced metastasis of B16F10 melanoma cells in C57BL/6 mice. v-H-Ras expression altered the cellular trafficking of CSE1L and increased CSE1L secretion. Most colorectal tumors were positive for CSE1L staining (98.4%, 125 of 127). Colorectal tumors with K-Ras mutation or high cytoplasmic CSE1L expression were correlated with T status (depth of tumor penetration; P = .004), stage (P = .004), and lymph node metastasis (P = .019). CONCLUSIONS: CSE1L may be a target for treating Ras-associated tumors. Analysis of K-Ras mutation and CSE1L expression may provide valuable clinical and pathological information to aid in the determination of treatment options for colorectal cancer.


Asunto(s)
Proteína de Susceptibilidad a Apoptosis Celular/genética , Neoplasias Colorrectales/genética , Genes ras/genética , Mutación , Metástasis de la Neoplasia/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Biomarcadores de Tumor/genética , Biopsia , Codón , Progresión de la Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente , Predisposición Genética a la Enfermedad , Humanos , Immunoblotting , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad
18.
Asian Pac J Cancer Prev ; 14(2): 1017-21, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23621178

RESUMEN

The cellular apoptosis susceptibility (CSE1L) gene has been demonstrated to regulate multiple cellular mechanisms including the mitotic spindle check point as well as proliferation and apoptosis. However, the importance of CSE1L in human colon cancer is largely unknown. In the present study, we examined expression levels of CSE1L mRNA by semiquantitative RT-PCR. A lentivirus-mediated small interfering RNA (siRNA) was used to knock down CSE1L expression in the human colon cancer cell line RKO. Changes in CSE1L target gene expression were determined by RT-PCR. Cell proliferation was examined by a high content screening assay. In vitro tumorigenesis was measured by colony-formation assay. Cell cycle distribution and apoptosis were detected by flow cytometric analysis. We found CSE1L mRNA to be expressed in human colon cancer cells. Using a lentivirus based RNAi approach, CSE1L expression was significantly inhibited in RKO cells, causing cell cycle arrest in the G2/M and S phases and a delay in cell proliferation, as well as induction of apoptosis and an inhibition of colony growth capacity. Collectively, the results suggest that silencing of CSE1L may be a potential therapeutic approach for colon cancer.


Asunto(s)
Apoptosis/genética , Proteína de Susceptibilidad a Apoptosis Celular/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/genética , Humanos , Puntos de Control de la Fase M del Ciclo Celular/genética , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Interferente Pequeño , Puntos de Control de la Fase S del Ciclo Celular/genética
19.
Ann Oncol ; 24(1): 193-201, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22967991

RESUMEN

BACKGROUND: Aggressive mature B-cell non-Hodgkin's lymphomas (BCL) sharing features of Burkitt's lymphoma (BL) and diffuse large B-cell lymphoma (DLBCL) (intermediate BL/DLBCL) but deviating with respect to one or more characteristics are increasingly recognized. The limited knowledge about these biologically heterogeneous lymphomas hampers their assignment to a known entity, raising incertitude about optimal treatment approaches. We therefore searched for discriminative, prognostic, and predictive factors for their better characterization. PATIENTS AND METHODS: We analyzed 242 cytogenetically defined aggressive mature BCL for differential protein expression. Marker selection was based on recent gene-expression profile studies. Predictive models for diagnosis were established and validated by a different set of lymphomas. RESULTS: CSE1L- and inhibitor of DNA binding-3 (ID3)-overexpression was associated with the diagnosis of BL and signal transduction and transcription-3 (STAT3) with DLBCL (P<0.001 for all markers). All three markers were associated with patient outcome in DLBCL. A new algorithm discriminating BL from DLBCL emerged, including the expression of CSE1L, STAT3, and MYC translocation. This 'new classifier' enabled the identification of patients with intermediate BL/DLBCL who benefited from intensive chemotherapy regimens. CONCLUSION: The proposed algorithm, which is based on markers with reliable staining properties for routine diagnostics, represents a novel valid tool in separating BL from DLBCL. Most interestingly, it allows segregating intermediate BL/DLBCL into groups with different treatment requirements.


Asunto(s)
Algoritmos , Biomarcadores de Tumor/genética , Linfoma de Burkitt/diagnóstico , Proteína de Susceptibilidad a Apoptosis Celular/genética , Genes myc , Linfoma de Células B Grandes Difuso/diagnóstico , Factor de Transcripción STAT3/genética , Western Blotting , Linfoma de Burkitt/genética , Femenino , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Linfoma de Células B Grandes Difuso/genética , Masculino , Persona de Mediana Edad , Pronóstico
20.
Brain Pathol ; 23(4): 426-39, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23252729

RESUMEN

MicroRNA-137 (miR-137) expression has been reported to be decreased in astrocytic tumors in two expression profiling studies but its role in the pathogenesis of oligodendroglial tumors is still limited. In this study, we demonstrate that miR-137 expression is significantly downregulated in a cohort of 35 oligodendroglial tumors and nine glioma cell lines compared with normal brains. Lower miR-137 expression is associated with shorter progressive-free survival and overall survival. Restoration of miR-137 expression in an oligodendroglial cells TC620, and also glioblastoma cells of U87 and U373 significantly suppressed cell growth, anchorage-independent growth, as well as invasion. Demethylation and deacetylation treatments resulted in upregulation of miR-137 expression in TC620 cells. In silico analysis showed that CSE1 chromosome segregation 1-like (yeast) (CSE1L) is a potential target gene of miR-137. Luciferase reporter assay demonstrated that miR-137 negatively regulates CSE1L by interaction between miR-137 and complementary sequences in the 3' UTR of CSE1L. Immunohistochemistry revealed that CSE1L is upregulated in oligodendroglial tumors. Knockdown of CSE1L resulted in similar outcomes as overexpressing miR-137 in oligodendroglioma cells and glioblastoma cells. Overall, our data suggest that miR-137 regulates growth of glioma cells and targets CSE1L, providing further understanding in the tumorigenesis of gliomas.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proteína de Susceptibilidad a Apoptosis Celular/metabolismo , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica/fisiología , MicroARNs/metabolismo , Oligodendroglioma/metabolismo , Adulto , Anciano , Azacitidina/análogos & derivados , Azacitidina/farmacología , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Bromodesoxiuridina/metabolismo , Línea Celular Tumoral , Proliferación Celular , Proteína de Susceptibilidad a Apoptosis Celular/genética , Niño , Estudios de Cohortes , Colágeno , Decitabina , Regulación hacia Abajo/efectos de los fármacos , Combinación de Medicamentos , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Ácidos Hidroxámicos/farmacología , Estimación de Kaplan-Meier , Laminina , Masculino , MicroARNs/genética , Persona de Mediana Edad , Oligodendroglioma/mortalidad , Oligodendroglioma/patología , Proteoglicanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...