Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
2.
Int J Oncol ; 63(2)2023 Aug.
Article in English | MEDLINE | ID: mdl-37417373

ABSTRACT

Subsequently to the publication of the above article, an interested reader drew to the authors' attention that two pairs of the culture plate images in Fig. 4A-C on p. 60 appeared to be the same, although the images were shown in different orientations; moreover, the 'NC/0 and DEX+miR132' and 'DEX and miR132' pairings of images in the scratch-wound assay experiments shown in Fig. 4B also appeared to be overlapping, such that these were apparently derived from the same original source where the results of differently performed experiments were intended to have been portrayed. After re­examining their original data, the authors have realized that some of the data in Fig. 4A and B were inadvertently assembled incorrectly. The revised version of Fig. 4, showing all the correct data for the culture plate images in Fig. 4A-C (specifically, the images fifth along on the right for Fig. 4B and C have been revised) and the correct images for 'NC/0' and 'DEX/0' in Fig. 4D is shown on on the next page. The authors are grateful to the Editor of International Journal of Oncology for allowing them this opportunity to publish a Corrigendum, and all the authors agree with its publication. Furthermore, the authors apologize to the readership for any inconvenience caused. [International Journal of Oncology 54: 53­64, 2019; DOI: 10.3892/ijo.2018.4616].

3.
Int J Oncol ; 54(1): 53-64, 2019 01.
Article in English | MEDLINE | ID: mdl-30387838

ABSTRACT

Glucocorticoids (GCs) such as dexamethasone (DEX) are administered as cancer co­treatment for palliative purposes due to their pro­apoptotic effects in lymphoid cancer and limited side effects associated with cancer growth and chemotherapy. However, there is emerging evidence that GCs induce therapy resistance in most epithelial tumors. Our recent data reveal that DEX promotes the progression of pancreatic ductal adenocarcinoma (PDA). In the present study, we examined 1 primary and 2 established PDA cell lines, and 35 PDA tissues from patients who had received (n=14) or not received (n=21) GCs prior to surgery. Through microRNA microarray analysis, in silico, and RT­qPCR analyses, we identified 268 microRNAs differentially expressed between DEX­treated and untreated cells. With a focus on cancer progression, we selected miR­132 and its target gene, transforming growth factor-ß2 (TGF­ß2), as top candidates. miR­132 mimics directly bound to the 3' untranslated region (3'UTR) of a TGF­ß2 luciferase construct and enhanced expression, as shown by increased luciferase activity. By contrast, DEX inhibited miR­132 expression via promoter methylation. miR­132 mimics also reduced DEX­induced clonogenicity, migration and expression of vimentin and E­cadherin in vitro and in tumor xenografts. In patients, GC intake prior to surgery enhanced global hypermethylation and expression of TGF­ß2 in tissues; expression of miR­132 was detected but could not be quantified. Our results demonstrate that DEX­mediated inhibition of miR­132 is a key mediator in the progression of pancreatic cancer, and the findings provide a foundation for miRNA­based therapies.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , DNA Methylation , Dexamethasone/adverse effects , Down-Regulation , MicroRNAs/genetics , Pancreatic Neoplasms/pathology , Transforming Growth Factor beta2/genetics , 3' Untranslated Regions , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Chick Embryo , Dexamethasone/pharmacology , Disease Progression , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Promoter Regions, Genetic/drug effects , Signal Transduction , Transforming Growth Factor beta2/metabolism
4.
Sci China Life Sci ; 61(5): 541-549, 2018 05.
Article in English | MEDLINE | ID: mdl-29307109

ABSTRACT

Late-stage melanoma is refractory to current therapies. MicroRNAs (miRNAs) can modulate many physiological and pathological processes of melanoma. Studies have demonstrated that miR-137 acts as a tumor suppressor by inhibiting the proliferation of melanoma cells through targeting multiple mRNAs. The glyoxalase system member glyoxalase 1 (GLO1) is the principal scavenging enzyme of methylglyoxal (MG), a toxic byproduct of glycolysis. Using 35S in vivo/vitro labelling analysis for dynamic proteomics (SiLAD), we found that miR-137 downregulated the expression of GLO1 in melanoma cells. Bioinformatics analysis predicted that GLO1 is a direct target of miR-137. This was validated by dual luciferase reporter assay. Quantitative RT-PCR (qRT-PCR) and western blot analysis indicated that miR-137 could decrease endogenous GLO1 expression. Furthermore, siRNA targeting of GLO1 mimicked inhibition of melanoma cell proliferation caused by miR-137 overexpression. Re-expression of GLO1 was able to restore miR-137-mediated suppression of melanoma cell proliferation. Therefore, these results suggest that miR-137 inhibits the proliferation of melanoma cells by targeting GLO1.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Lactoylglutathione Lyase/genetics , Melanoma/metabolism , Melanoma/pathology , MicroRNAs/metabolism , 3' Untranslated Regions , Cell Line, Tumor , Cell Proliferation/physiology , Gene Knockdown Techniques , Humans
5.
Mol Cancer ; 16(1): 23, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28137273

ABSTRACT

BACKGROUND: Cancer stem cells are suggested to contribute to the extremely poor prognosis of pancreatic ductal adenocarcinoma and dysregulation of symmetric and asymmetric stem cell division may be involved. Anticancer benefits of phytochemicals like the polyphenol quercetin, present in many fruits, nuts and vegetables, could be expedited by microRNAs, which orchestrate cell-fate decisions and tissue homeostasis. The mechanisms regulating the division mode of cancer stem cells in relation to phytochemical-induced microRNAs are poorly understood. METHODS: Patient-derived pancreas tissue and 3 established pancreatic cancer cell lines were examined by immunofluorescence and time-lapse microscopy, microRNA microarray analysis, bioinformatics and computational analysis, qRT-PCR, Western blot analysis, self-renewal and differentiation assays. RESULTS: We show that symmetric and asymmetric division occurred in patient tissues and in vitro, whereas symmetric divisions were more extensive. By microarray analysis, bioinformatics prediction and qRT-PCR, we identified and validated quercetin-induced microRNAs involved in Notch signaling/cell-fate determination. Further computational analysis distinguished miR-200b-3p as strong candidate for cell-fate determinant. Mechanistically, miR-200b-3p switched symmetric to asymmetric cell division by reversing the Notch/Numb ratio, inhibition of the self-renewal and activation of the potential to differentiate to adipocytes, osteocytes and chondrocytes. Low miR-200b-3p levels fostered Notch signaling and promoted daughter cells to become symmetric while high miR-200b-3p levels lessened Notch signaling and promoted daughter cells to become asymmetric. CONCLUSIONS: Our findings provide a better understanding of the cross talk between phytochemicals, microRNAs and Notch signaling in the regulation of self-renewing cancer stem cell divisions.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , MicroRNAs/genetics , Neoplastic Stem Cells/drug effects , Pancreatic Neoplasms/genetics , Quercetin/pharmacology , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Self Renewal/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplastic Stem Cells/cytology , Pancreatic Neoplasms/drug therapy , Up-Regulation
6.
Oncotarget ; 7(36): 58367-58380, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27521217

ABSTRACT

Pancreatic Ductal Adenocarcinoma (PDA) is a highly malignant tumor with poor prognosis. MicroRNAs (miRs) may offer novel therapeutic approaches to treatment. The polyphenol quercetin, present in many fruits and vegetables, possesses anti-carcinogenic properties. To unravel the effect of quercetin to miR signaling we performed miR profiling in PDA cells before and after quercetin treatment, followed by biostatistical analysis. miR let-7c was among the top up-regulated candidates after quercetin treatment, as measured by qRT-PCR and confirmed in two established and one primary PDA cell lines. By computational analysis we identified the Notch-inhibitor Numbl as let-7c target gene. This was strengthened by luciferase assays, where lipofected let-7c mimics induced a Numbl 3-UTR wild type construct, but not the mutated counterpart. Let-7c induced Numbl mRNA and protein expression but inhibited Notch just like quercetin. It also inhibited colony formation, wound healing, and protein expression of progression markers. In vivo xenotransplantation of PDA cells and subsequent intravenous injection of let-7c resulted in a significant decrease in tumor mass without obvious toxic effects in the fertilized chick egg model. The delivery rate of the miR mimics to the tumor mass was 80%, whereas minor amounts were present in host tissue. By immunohistochemistry we demonstrated that let-7c inhibited Notch and progression markers but up-regulated Numbl. These findings show that quercetin-induced let-7c decreases tumor growth by posttranscriptional activation of Numbl and indirect inhibition of Notch.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , MicroRNAs/metabolism , Pancreatic Neoplasms/metabolism , Quercetin/pharmacology , 3' Untranslated Regions , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Chick Embryo , Computational Biology , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Neoplasm Transplantation , Neoplastic Stem Cells , Osteoblasts/metabolism , Pancreatic Neoplasms/genetics , Receptor, Notch1/metabolism , Signal Transduction/drug effects , Up-Regulation , Pancreatic Neoplasms
7.
Cancer Lett ; 373(1): 130-137, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26828016

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is among the most lethal malignancies and resistance to chemotherapy prevents the therapeutic outcome. MicroRNAs provide a novel therapeutic strategy. Here, the established and primary human PDA cell lines PANC-1, AsPC-1, MIA-PaCa2, AsanPaCa, BxPC-3 and three gemcitabine-resistant subclones were examined. A gene expression profiling revealed that the ribonucleotide reductase M1 (RRM1) was upregulated in gemcitabine-resistant cells, which was confirmed by qRT-PCR, Western blot analysis and immunostaining. Inhibition of RRM1 by lipotransfection of siRNA reduced its expression and reversed gemcitabine resistance. The expression of RRM1 correlated to gemcitabine resistance in vitro and was higher in malignant patient pancreas tissue compared to non-malignant pancreas tissue. By microRNA expression profiling, we identified microRNA-101-3p as top-downregulated candidate. Lipotransfection of microRNA-101-3p mimics inhibited the expression of RRM1, reduced the luciferase activity of its 3'UTR and sensitized for gemcitabine-induced cytotoxicity. These results underline the relevance of microRNA-101-3p-driven regulation of RRM1 in drug resistance and suggest the co-delivery of microRNA-101-3p and gemcitabine for more effective therapy outcome.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , MicroRNAs/metabolism , Pancreatic Neoplasms/drug therapy , Tumor Suppressor Proteins/metabolism , 3' Untranslated Regions , Binding Sites , Carcinoma, Pancreatic Ductal/enzymology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , RNA Interference , Ribonucleoside Diphosphate Reductase , Time Factors , Transfection , Tumor Suppressor Proteins/genetics , Up-Regulation , Gemcitabine
8.
Exp Dermatol ; 24(12): 947-52, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26186482

ABSTRACT

MicroRNAs (miRNA) are key players in a variety of cancers including malignant melanoma. miR-137 has been reported to be a tumor suppressor in melanoma and several targets have been identified for this miRNA. We previously developed a novel proteomics technology, (35) S in vivo/vitro labelling analysis for dynamic proteomics (SiLAD). Because of its high sensitivity in analysing protein expression rates, SiLAD has the potential to unravel miRNA effects on mRNAs coding for proteins with long half-lives or high abundance. Using SiLAD, we discovered that miR-137 significantly downregulated the expression rate of p21-activated kinase 2 (PAK2) in melanoma cells. Bioinformatics analysis predicted PAK2 as a direct target of miR-137, which was confirmed by luciferase reporter assay and Western blot analysis. We found that overexpression of miR-137 inhibited the proliferation of melanoma cells, which could be phenocopied by knockdown of PAK2 using siRNAs. Furthermore, overexpression of PAK2 restored miR-137-mediated suppression of cell proliferation. These findings indicate that miR-137 could inhibit proliferation through targeting PAK2 in melanoma cells.


Subject(s)
Melanoma/genetics , Melanoma/pathology , MicroRNAs/genetics , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/genetics , Apoptosis/genetics , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Down-Regulation , Gene Knockdown Techniques , Humans , Melanoma/metabolism , MicroRNAs/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA, Small Interfering/genetics , Transfection , p21-Activated Kinases/metabolism
9.
J Invest Dermatol ; 133(3): 768-775, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23151846

ABSTRACT

MicroRNAs are small noncoding RNAs that regulate gene expression and have important roles in various types of cancer. Previously, miR-137 was reported to act as a tumor suppressor in different cancers, including malignant melanoma. In this study, we show that low miR-137 expression is correlated with poor survival in stage IV melanoma patients. We identified and validated two genes (c-Met and YB1) as direct targets of miR-137 and confirmed two previously known targets, namely enhancer of zeste homolog 2 (EZH2) and microphthalmia-associated transcription factor (MITF). Functional studies showed that miR-137 suppressed melanoma cell invasion through the downregulation of multiple target genes. The decreased invasion caused by miR-137 overexpression could be phenocopied by small interfering RNA knockdown of EZH2, c-Met, or Y box-binding protein 1 (YB1). Furthermore, miR-137 inhibited melanoma cell migration and proliferation. Finally, miR-137 induced apoptosis in melanoma cell lines and decreased BCL2 levels. In summary, our study confirms that miR-137 acts as a tumor suppressor in malignant melanoma and reveals that miR-137 regulates multiple targets including c-Met, YB1, EZH2, and MITF.


Subject(s)
Down-Regulation/physiology , Melanoma/pathology , MicroRNAs/metabolism , Neoplasm Invasiveness/pathology , Oncogenes/physiology , Skin Neoplasms/pathology , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/genetics , Enhancer of Zeste Homolog 2 Protein , Humans , Melanoma/metabolism , Melanoma/mortality , MicroRNAs/genetics , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Neoplasm Staging , Oncogenes/genetics , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , RNA, Small Interfering/pharmacology , Skin Neoplasms/metabolism , Skin Neoplasms/mortality , Survival Rate , Y-Box-Binding Protein 1/genetics , Y-Box-Binding Protein 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...