Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Br J Cancer ; 116(10): 1350-1357, 2017 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-28419078

RESUMEN

BACKGROUND: MicroRNAs (miRs) are involved in the regulation of many processes that contribute to malignancy, including cell proliferation, radiation resistance, invasion and metastasis. The role of miR-330-3p, an miR upregulated in breast cancer, remains unclear. METHODS: We examine the association of miR-330-3p with distant relapse-free survival in the Oxford cohort of breast cancer patients. We also study miR-330-3p function using in vitro invasion and ex ovo metastasis assays. Using in vitro luciferase assays, we validate a novel target gene for miR-330-3p, Collagen And Calcium Binding EGF Domains 1 (CCBE1). We assess functional consequences of CCBE1 loss by using siRNA-mediated knockdown followed by in vitro invasion assays. Lastly, we examine the expression profile of CCBE1 in breast carcinomas in the Curtis and TCGA Breast Cancer data sets using Oncomine Platform as well as distant relapse-free and overall survival of patients in the Helsinki University breast cancer data set according to CCBE1 expression status. RESULTS: miR-330-3p is enriched in breast cancer, and higher levels of miR-330-3p expression are associated with lower distant relapse-free survival in a cohort of breast cancer patients. Consistent with these observations, overexpression of miR-330-3p in breast cancer cell lines results in greater invasiveness in vitro, and miR-330-3p-overexpressing cells also metastasise more aggressively ex ovo. We identify CCBE1 as a direct target of miR-330-3p, and show that knockdown of CCBE1 results in a greater invasive capacity. Accordingly, in breast cancer patients CCBE1 is frequently downregulated, and its loss is associated with reduced distant relapse-free and overall survival. CONCLUSIONS: We show for the first time that miR-330-3p targets CCBE1 to promote invasion and metastasis. miR-330-3p and CCBE1 may represent promising biomarkers in breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas de Unión al Calcio/genética , Carcinoma Ductal de Mama/genética , Carcinoma Intraductal no Infiltrante/genética , MicroARNs/genética , Proteínas Supresoras de Tumor/genética , Carcinoma Ductal de Mama/patología , Carcinoma Ductal de Mama/secundario , Carcinoma Intraductal no Infiltrante/patología , Carcinoma Intraductal no Infiltrante/secundario , Línea Celular Tumoral , Supervivencia sin Enfermedad , Regulación hacia Abajo , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Invasividad Neoplásica/genética , Metástasis de la Neoplasia , Tasa de Supervivencia
2.
Radiol Imaging Cancer ; 6(2): e230020, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38334470

RESUMEN

Purpose To investigate the feasibility of low-dose positron emission mammography (PEM) concurrently to MRI to identify breast cancer and determine its local extent. Materials and Methods In this research ethics board-approved prospective study, participants newly diagnosed with breast cancer with concurrent breast MRI acquisitions were assigned independently of breast density, tumor size, and histopathologic cancer subtype to undergo low-dose PEM with up to 185 MBq of fluorine 18-labeled fluorodeoxyglucose (18F-FDG). Two breast radiologists, unaware of the cancer location, reviewed PEM images taken 1 and 4 hours following 18F-FDG injection. Findings were correlated with histopathologic results. Detection accuracy and participant details were examined using logistic regression and summary statistics, and a comparative analysis assessed the efficacy of PEM and MRI additional lesions detection (ClinicalTrials.gov: NCT03520218). Results Twenty-five female participants (median age, 52 years; range, 32-85 years) comprised the cohort. Twenty-four of 25 (96%) cancers (19 invasive cancers and five in situ diseases) were identified with PEM from 100 sets of bilateral images, showcasing comparable performance even after 3 hours of radiotracer uptake. The median invasive cancer size was 31 mm (range, 10-120). Three additional in situ grade 2 lesions were missed at PEM. While not significant, PEM detected fewer false-positive additional lesions compared with MRI (one of six [16%] vs eight of 13 [62%]; P = .14). Conclusion This study suggests the feasibility of a low-dose PEM system in helping to detect invasive breast cancer. Though large-scale clinical trials are essential to confirm these preliminary results, this study underscores the potential of this low-dose PEM system as a promising imaging tool in breast cancer diagnosis. ClinicalTrials.gov registration no. NCT03520218 Keywords: Positron Emission Digital Mammography, Invasive Breast Cancer, Oncology, MRI Supplemental material is available for this article. © RSNA, 2024 See also commentary by Barreto and Rapelyea in this issue.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Persona de Mediana Edad , Neoplasias de la Mama/diagnóstico por imagen , Fluorodesoxiglucosa F18 , Tomografía de Emisión de Positrones/métodos , Estudios Prospectivos , Electrones , Radiofármacos , Tomografía Computarizada por Rayos X , Mamografía
3.
Biochem Biophys Res Commun ; 441(4): 726-31, 2013 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-24184480

RESUMEN

ATG4B belongs to the autophagin family of cysteine proteases required for autophagy, an emerging target of cancer therapy. Developing pharmacological ATG4B inhibitors is a very active area of research. However, detailed studies on the role of ATG4B during anticancer therapy are lacking. By analyzing PC-3 and C4-2 prostate cancer cells overexpressing dominant negative ATG4B(C74A)in vitro and in vivo, we show that the effects of ATG4B(C74A) are cell type, treatment, and context-dependent. ATG4B(C74A) expression can either amplify the effects of cytotoxic therapies or contribute to treatment resistance. Thus, the successful clinical application of ATG4B inhibitors will depend on finding predictive markers of response.


Asunto(s)
Autofagia/efectos de los fármacos , Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/uso terapéutico , Terapia Molecular Dirigida , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Proteínas Relacionadas con la Autofagia , Línea Celular Tumoral , Cisteína Endopeptidasas/genética , Inhibidores de Cisteína Proteinasa/farmacología , Docetaxel , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos , Humanos , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Tolerancia a Radiación , Taxoides/farmacología , Taxoides/uso terapéutico , Topotecan/farmacología , Topotecan/uso terapéutico
4.
Oncol Rep ; 42(3): 1047-1056, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31322262

RESUMEN

MicroRNAs are small non­coding RNA molecules which act as modulators of gene function, and have been identified as playing important roles in cancer as both tumor suppressors and oncogenes. The present study aimed to examine the role of miR­198 in prostate cancer aggression by analyzing how it influences several hallmarks of cancer. Abundance of miR­198 in prostate cancer and association with clinical characteristics was analyzed using a CPC­Gene prostate cancer dataset. Overexpression of miR­198 was performed using transient transfection of miR­198 mimic prior to assaying proliferation, cell cycle, and colony formation in LNCaP and DU145 cell lines using standard protocols. In vivo tumor formation in athymic nude mice was examined using LNCaP xenografts with stable overexpression conferred using lentiviral miR­198 transduction. Protein and mRNA abundance of MIB1 was determined using western blotting and RT­qPCR respectively, while miR­198 binding to MIB1 was validated using a luciferase reporter assay. miR­198 abundance was lower in high Gleason grade prostate cancer relative to intermediate and low­grade cancer. Overexpression of miR­198 diminished proliferation of prostate cancer cell lines, increased G0/G1 cell cycle arrest, and significantly impaired colony formation. Elevated miR­198 abundance was also demonstrated to impair tumor formation in vivo using LNCaP xenografts. Mindbomb E3 ubiquitin protein ligase 1 (MIB1) was demonstrated to be directly targeted by miR­198, and knockdown of MIB1 recapitulated the effects of miR­198 on proliferation and colony formation. The present evidence supports miR­198 as an important tumor suppressor in prostate cancer, and demonstrates for the first time that it acts by targeting MIB1. The present study reinforces the importance and complexity of miRNA in regulating prostate cancer aggression.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Neoplasias de la Próstata/patología , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Humanos , Masculino , Ratones , Ratones Desnudos , Pronóstico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Transcriptoma , Células Tumorales Cultivadas , Ubiquitina-Proteína Ligasas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Mol Oncol ; 12(8): 1324-1341, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29845714

RESUMEN

Recurrence of high-grade prostate cancer after radiotherapy is a significant clinical problem, resulting in increased morbidity and reduced patient survival. The molecular mechanisms of radiation resistance are being elucidated through the study of microRNA (miR) that negatively regulate gene expression. We performed bioinformatics analyses of The Cancer Genome Atlas (TCGA) dataset to evaluate the association between miR-106a and its putative target lipopolysaccharide-induced TNF-α factor (LITAF) in prostate cancer. We characterized the function of miR-106a through in vitro and in vivo experiments and employed transcriptomic analysis, western blotting, and 3'UTR luciferase assays to establish LITAF as a bona fide target of miR-106a. Using our well-characterized radiation-resistant cell lines, we identified that miR-106a was overexpressed in radiation-resistant cells compared to parental cells. In the TCGA, miR-106a was significantly elevated in high-grade human prostate tumors relative to intermediate- and low-grade specimens. An inverse correlation was seen with its target, LITAF. Furthermore, high miR-106a and low LITAF expression predict for biochemical recurrence at 5 years after radical prostatectomy. miR-106a overexpression conferred radioresistance by increasing proliferation and reducing senescence, and this was phenocopied by knockdown of LITAF. For the first time, we describe a role for miRNA in upregulating ATM expression. LITAF, not previously attributed to radiation response, mediates this interaction. This route of cancer radioresistance can be overcome using the specific ATM kinase inhibitor, KU-55933. Our research provides the first report of miR-106a and LITAF in prostate cancer radiation resistance and high-grade disease, and presents a viable therapeutic strategy that may ultimately improve patient outcomes.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Recurrencia Local de Neoplasia/genética , Proteínas Nucleares/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/radioterapia , Factores de Transcripción/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Masculino , Regulación hacia Arriba/efectos de la radiación
6.
Oncotarget ; 8(3): 4668-4689, 2017 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-27902466

RESUMEN

Radioresistance remains a significant obstacle in the treatment of Prostate Cancer (PCa). To simulate the clinical scenario of irradiation resistance (IRR), we created DU145-IRR PCa cell lines by treatment with 2 Gy daily IR for 59 fractions. DU145-IRR cells acquired an aggressive phenotype as evidenced by increased clonogenic survival, tumorigenic potential and invasiveness. We performed transcriptome profiling to discover dysregulated genes in DU145-IRR cells and identified the long non-coding RNA (lncRNA), Urothelial carcinoma-associated 1 (UCA1). We first investigated the role of UCA1 in radiation response and found that UCA1 abundance was significantly higher in DU145-IRR cells compared to control cells. UCA1 siRNA-knockdown reversed the aggressive phenotype and significantly increased sensitivity to IR. UCA1 depletion inhibited growth, induced cell cycle arrest at the G2/M transition and decreased activation of the pro-survival Akt pathway. We then studied the clinical significance of UCA1 expression in two independent cohorts of PCa patients: MSKCC (130 patients) and CPC-GENE (209 patients). UCA1 over-expression was associated with decreased 5-year disease-free survival in MSKCC patients (HR = 2.9; p = 0.007) and a trend toward lower biochemical recurrence-free survival in CPC-GENE patients (HR = 2.7; p = 0.05). We showed for the first time that UCA1 depletion induces radiosensitivity, decreases proliferative capacity and disrupts cell cycle progression, which may occur through altered Akt signaling and induced cell cycle arrest at the G2/M transition. Our results indicate that UCA1 might have prognostic value in PCa and be a potential therapeutic target.


Asunto(s)
Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , ARN Largo no Codificante/genética , Tolerancia a Radiación , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Clasificación del Tumor , Neoplasias de la Próstata/radioterapia , Análisis de Supervivencia , Regulación hacia Arriba
7.
Oncotarget ; 6(26): 22439-51, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26068950

RESUMEN

MicroRNA contribute to tumor radiation resistance, which is an important clinical problem, and thus we are interested in identifying and characterizing their function. We demonstrate that miR-620 contributes to radiation resistance in cancer cells by increasing proliferation, and decreasing the G2/M block. We identify the hydroxyprostaglandin dehydrogenase 15-(nicotinamide adenine dinucleotide) (HPGD/15-PGDH) tumor suppressor gene as a direct miR-620 target, which results in increased prostaglandin E2 (PGE2) levels. Furthermore, we show that siRNA targeting of HPGD or administration of exogenous PGE2 recapitulates radioresistance. Targeting of the EP2 receptor that responds to PGE2 using pharmacological or genetic approaches, abrogates radioresistance. Tumor xenograft experiments confirm that miR-620 increases proliferation and tumor radioresistance in vivo. Regulation of PGE2 levels via targeting of HPGD by miR-620 is an innovative manner by which a microRNA can induce radiation resistance.


Asunto(s)
Hidroxiprostaglandina Deshidrogenasas/metabolismo , MicroARNs/administración & dosificación , Neoplasias/terapia , Animales , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/radioterapia , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Dinoprostona/metabolismo , Dinoprostona/farmacología , Femenino , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Masculino , Ratones , Ratones Desnudos , MicroARNs/genética , Neoplasias/enzimología , Neoplasias/genética , Neoplasias/radioterapia , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/radioterapia , Neoplasias Pancreáticas/terapia , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/radioterapia , Neoplasias de la Próstata/terapia , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/genética , Radioterapia Adyuvante , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
8.
World J Gastroenterol ; 20(46): 17376-87, 2014 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-25516649

RESUMEN

AIM: To investigate the function of Pea3 in colorectal carcinoma (CRC) invasion and metastatic potential. METHODS: The expression of Pea3 during clinical progression of human CRC was investigated using Oncomine Research Edition. To assay Pea3 expression in established CRC cell lines, we performed western blotting of cell lysates. We employed shRNA-mediated knockdown of Pea3 in HCT116 (HCT) and LS174T CRC cells which was confirmed by real-time quantitative PCR (qPCR) and western blotting. Transwell invasion assays, MTS proliferation assays, anoikis assays, and fluorometric matrix metalloprotease (MMP) assays were performed to determine the effects of Pea3 knockdown on invasion, proliferation, anoikis and MMP activity in CRC cells in vitro. Alterations in epithelial-mesenchymal transition (EMT) and matrix metalloprotease (MMP) mRNA levels were determined by qPCR. CRC cells were injected into the flanks of nude mice to generate xenografts and tumor growth monitored with serial calliper measurements. To assay metastatic potential, CRC cells were injected into the spleen of nude mice, and histological analysis performed on the livers 21 d later. RESULTS: We demonstrated that reduction of Pea3 expression in CRC cells significantly impaired their invasive capacity (HCT.shPea3, 0.28 ± 0.04 fold, P < 0.01; LS.shPea3, 0.15 ± 0.04 fold; SW.shPea3, 0.23 ± 0.03, P < 0.01), reduced anoikis resistance (HCT.shPea3 75.4% ± 1.9% viable cells vs HCT.shCtrl 88.6% ± 0.6% viable cells, P < 0.01; LS.shPea3 71.7% ± 0.5% viable cells vs LS.Ctrl 89.6% ± 0.3% viable cells, P < 0.005, but had no effect on proliferation (HCT.shCtrl AUC 5098 ± 123 vs HCT.shPea3 5689 ± 151, P < 0.05; LS.shCtrl AUC 5600 ± 324.1 vs LS.shPea3 6423 ± 400, P < 0.05). In vivo, HCT.shPea3 and HCT.shCtrl tumour xenografts grew at a similar rate (HCT.shPea3 2.64 ± 0.82 fold vs HCT.shCtrl 2.88 ± 0.80 fold, P > 0.05). In keeping with a pro-metastatic function for Pea3 in CRC, several EMT markers and MMPs were downregulated in shPea3-expressing cells, suggesting that Pea3 may exert its effects through these processes. A reduction in overall MMP activity was observed in HCT.shPea3 cells compared to their control counterparts (HCT.shPea3 0.61 ± 0.04 fold, P < 0.005). This translated in vivo to the complete absence of metastases in the livers of mice that were grafted with CRC cells lacking Pea3. Conversely, CRC cells expressing Pea3 formed liver metastases in all mice. CONCLUSION: Our study implicates Pea3 as a mediator of metastases, and provides a biological rationale for the adverse prognosis associated with elevated Pea3 expression in human CRC.


Asunto(s)
Proteínas E1A de Adenovirus/metabolismo , Carcinoma/metabolismo , Movimiento Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas E1A de Adenovirus/genética , Animales , Anoicis , Carcinoma/genética , Carcinoma/secundario , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HEK293 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/prevención & control , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-ets , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Transfección , Carga Tumoral
9.
Cancer Res ; 73(23): 6972-86, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24145350

RESUMEN

Radiation resistance poses a major clinical challenge in cancer treatment, but little is known about how microRNA (miR) may regulate this phenomenon. In this study, we used next-generation sequencing to perform an unbiased comparison of miR expression in PC3 prostate cancer cells rendered resistant to fractionated radiation treatment. One miR candidate found to be upregulated by ionizing radiation was miR-95, the enforced expression of which promoted radiation resistance in a variety of cancer cells. miR-95 overexpression recapitulated an aggressive phenotype including increased cellular proliferation, deregulated G2-M checkpoint following ionizing radiation, and increased invasive potential. Using combined in silico prediction and microarray expression analyses, we identified and validated the sphingolipid phosphatase SGPP1, an antagonist of sphingosine-1-phosphate signaling, as a target of miR-95 that promotes radiation resistance. Consistent with this finding, cell treatment with FTY720, a clinically approved small molecule inhibitor of S1P signaling, sensitized miR-95 overexpressing cells to radiation treatment. In vivo assays extended the significance of these results, showing that miR-95 overexpression increased tumor growth and resistance to radiation treatment in tumor xenografts. Furthermore, reduced tumor necrosis and increased cellular proliferation were seen after radiation treatment of miR-95 overexpressing tumors compared with control tumors. Finally, miR-95 expression was increased in human prostate and breast cancer specimens compared with normal tissue. Together, our work reveals miR-95 expression as a critical determinant of radiation resistance in cancer cells.


Asunto(s)
Proteínas de la Membrana/genética , MicroARNs/fisiología , Neoplasias/genética , Monoéster Fosfórico Hidrolasas/genética , Tolerancia a Radiación/genética , Animales , Línea Celular Tumoral , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Desnudos , Invasividad Neoplásica , Neoplasias/patología , Interferencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA