Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Int J Oncol ; 64(2)2024 Feb.
Article in English | MEDLINE | ID: mdl-38063241

ABSTRACT

Following the publication of the above article, a concerned reader drew to the Editor's attention that, regarding the western blots featured in Fig. 3B on p. 670, the bands featured in the U251 and U251­MC lanes for the miR­21 and U6 experiments appeared to be duplicates of each other. Moreover, certain of these data were strikingly similar to data that appeared in another article published at around the same time featuring some of the same authors (again, with apparent duplications of bands within the same gel slices, as they were presented). After having conducted an internal investigation of this matter, the Editor of International Journal of Oncology has judged that the apparently anomalous grouping of the data could not have been attributed to pure coincidence. Therefore, the Editor has decided that this article should be retracted from the publication on the grounds of an overall lack of confidence in the data. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a reply. The Editor sincerely apologizes to the readership for any incovenience caused, and we thank the reader for bringing this matter to our attention. [International Journal of Oncology 36: 665­672, 2010; DOI: 10.3892/ijo_00000542].

2.
Mol Cancer ; 13: 63, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24650032

ABSTRACT

BACKGROUND: Epidermal growth factor receptor (EGFR) is amplified in 40% of human glioblastomas. However, most glioblastoma patients respond poorly to anti-EGFR therapy. MicroRNAs can function as either oncogenes or tumor suppressor genes, and have been shown to play an important role in cancer cell proliferation, invasion and apoptosis. Whether microRNAs can impact the therapeutic effects of EGFR inhibitors in glioblastoma is unknown. METHODS: miR-566 expression levels were detected in glioma cell lines, using real-time quantitative RT-PCR (qRT-PCR). Luciferase reporter assays and Western blots were used to validate VHL as a direct target gene of miR-566. Cell proliferation, invasion, cell cycle distribution and apoptosis were also examined to confirm whether miR-566 inhibition could sensitize anti-EGFR therapy. RESULTS: In this study, we demonstrated that miR-566 is up-regulated in human glioma cell lines and inhibition of miR-566 decreased the activity of the EGFR pathway. Lentiviral mediated inhibition of miR-566 in glioblastoma cell lines significantly inhibited cell proliferation and invasion and led to cell cycle arrest in the G0/G1 phase. In addition, we identified von Hippel-Lindau (VHL) as a novel functional target of miR-566. VHL regulates the formation of the ß-catenin/hypoxia-inducible factors-1α complex under miR-566 regulation. CONCLUSIONS: miR-566 activated EGFR signaling and its inhibition sensitized glioblastoma cells to anti-EGFR therapy.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , ErbB Receptors/genetics , Glioblastoma/genetics , MicroRNAs/genetics , Signal Transduction , Animals , Blotting, Western , Cell Line, Tumor , ErbB Receptors/metabolism , Fluorescent Antibody Technique , Glioblastoma/metabolism , Heterografts , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Nude , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Transfection , Von Hippel-Lindau Tumor Suppressor Protein/genetics
3.
Cancer Lett ; 342(1): 139-49, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24012640

ABSTRACT

Epidermal growth factor receptors (EGFR) expression is frequently amplified in human glioblastoma cells. Nimotuzumab, a monoclonal antibody (mAb) against EGFR, has been used globally in clinics as an anti-cancer agent. It is largely unknown whether the blockade of miR-21, a microRNA that is upregulated in glioma cells, could amplify the effects of nimotuzumab. Herein, we have demonstrated that miR-21 directly targets von Hippel-Lindau (VHL) and peroxisome-proliferator-activated receptor α (PPARα) and that miR-21 regulates EGFR/AKT signaling through VHL/ß-catenin and the PPARα/AP-1 axis. Further, the expression of miR-21 is regulated by EGFR via the activation of ß-catenin and AP-1. These data indicate that a feedback loop exists between miR-21 and EGFR. We also show that the combination of nimotuzumab and an inhibitor of miR-21 is superior to single-agent therapy. These results clarify a novel association between miR-21 and EGFR in the regulation of cancer cell progression.


Subject(s)
Brain Neoplasms/metabolism , ErbB Receptors/metabolism , Glioblastoma/metabolism , MicroRNAs/genetics , 3' Untranslated Regions , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Base Sequence , Binding Sites , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , PPAR alpha/genetics , PPAR alpha/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Transcription Factor AP-1/metabolism , Tumor Burden , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Wnt Signaling Pathway , Xenograft Model Antitumor Assays , beta Catenin/metabolism
4.
Neuro Oncol ; 15(12): 1595-603, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24203894

ABSTRACT

BACKGROUND: Long noncoding RNA Hox transcript antisense intergenic RNA (HOTAIR) has been characterized as a negative prognostic factor in breast and colon cancer patients. The clinical significance and function of HOTAIR in glioma remains unclear. METHODS: We analyzed the clinical significance of HOTAIR in 3 different glioma cohorts with gene expression data, including correlation with tumor grade, prognosis, and molecular subtype. The function of HOTAIR in glioma was explored by performing gene set enrichment analysis and in vitro and in vivo experiments. RESULTS: HOTAIR expression was closely associated with glioma grade and poor prognosis. Multivariate Cox regression analysis revealed that HOTAIR was an independent prognostic factor in glioblastoma multiforme patients. HOTAIR expression correlated with glioma molecular subtype, including those of The Cancer Genome Atlas. HOTAIR was preferentially expressed in the classical and mesenchymal subtypes compared with the neural and proneural subtypes. A gene set enrichment analysis designed to show gene set differences between patients with high and low HOTAIR expression indicated that HOTAIR expression was associated with gene sets involved in cell cycle progression. HOTAIR reduction induced colony formation suppression, cell cycle G0/G1 arrest, and orthotopic tumor growth inhibition. CONCLUSION: Our data establish that HOTAIR is an important long noncoding RNA that primarily serves as a prognostic factor for glioma patient survival, as well as a biomarker for identifying glioma molecular subtypes, a critical regulator of cell cycle progression.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Glioma/genetics , Mesoderm/pathology , RNA, Long Noncoding/genetics , Adult , Animals , Blotting, Western , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Case-Control Studies , Cell Cycle , Cell Proliferation , Female , Follow-Up Studies , Gene Expression Profiling , Glioma/mortality , Glioma/pathology , Humans , Male , Mesoderm/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Grading , Oligonucleotide Array Sequence Analysis , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Neuro Oncol ; 15(3): 279-89, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23295773

ABSTRACT

BACKGROUND: Aberrant activation of beta-catenin/TCF4 and STAT3 signaling in glioblastoma multiforme (GBM) has been reported. However, the molecular mechanisms related to this process are still poorly understood. METHODS: Genome-wide screening of the binding characteristics of the transcription factors TCF4 and STAT3 in GBM cells was performed by chromatin immunoprecipitation sequencing (ChIP-seq) assay. Hierarchical clustering was used to analyze the association of TCF4 and STAT3 coregulated genes with The Cancer Genome Atlas (TCGA) GBM subtypes (classical, mesenchymal, neural, and proneural). New molecular classification of GBM was proposed and validated in Western and Asian populations. RESULTS: We identified 1250 overlapping putative target genes that were coregulated by TCF4 and STAT3. Further, the coregulated genes had the potential to guide TCGA GBM subtypes. Finally, we proposed a new molecular classification of GBM into 2 subtypes (proneural-like and mesenchymal-like) and showed that the new classification could be applied to both Western and Asian populations. In addition, the GBM response to temozolomide therapy differed depending on its subtype; mesenchymal-like GBM benefited, while there was no benefit for proneural-like GBM. CONCLUSIONS: This is the first comprehensive study to combine a ChIP-seq assay of TCF4 and STAT3 and data mining of patient cohorts to derive molecular subtypes of GBM.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Biomarkers, Tumor/genetics , Brain Neoplasms/classification , Genome, Human , Glioblastoma/classification , STAT3 Transcription Factor/metabolism , Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Binding Sites , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Chromatin Immunoprecipitation , Cohort Studies , DNA Methylation , Female , Follow-Up Studies , Gene Expression Profiling , Glioblastoma/genetics , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Male , Mesoderm/metabolism , Mesoderm/pathology , Middle Aged , Neurons/metabolism , Neurons/pathology , Oligonucleotide Array Sequence Analysis , Prognosis , Promoter Regions, Genetic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , Survival Rate , Transcription Factor 4 , Transcription Factors/genetics
6.
CNS Neurosci Ther ; 19(2): 98-108, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23230963

ABSTRACT

BACKGROUND AND AIMS: Currently temozolomide (TMZ) as a potent agent is widely used to treat the glioblastoma multiforme (GBM), whereas recurrence due to intrinsic or acquired therapeutic resistance often occurs. Combination chemotherapy with TMZ may be a promising therapeutic strategy to improve treatment efficacy. METHODS: Aspirin, TMZ, and aspirin-/TMZ-coloaded poly (L-lactide-co-glycolide) (PLGA) microspheres were prepared by spray drying, and cytotoxicities of glioblastoma cells were measured. RESULTS: Aspirin microsphere treatment induced slight apoptosis and modestly inhibited proliferation of LN229 and U87 cells in vitro and in vivo through inhibition of ß-catenin transactivation. However, aspirin-/TMZ-coloaded microspheres presented synergistic antitumor efficacy compared with single TMZ-loaded microspheres. Aspirin/TMZ microspheres induced more apoptosis and repressed proliferation of LN229 and U87 cells. Corresponding to inhibition of ß-catenin signaling, ß-catenin/TCF4 transcriptional activity and STAT3 luciferase activity were strongly suppressed, and downstream targets expression was decreased. Furthermore, aspirin/TMZ microsphere intratumoral injection downregulated the expression of ß-catenin, TCF4, pAKT, pSTAT3, and PCNA and delayed tumor growth in nude mice harboring subcutaneous LN229 xenografts. CONCLUSIONS: Aspirin sensitized TMZ chemotherapy efficacy through inhibition of ß-catenin transactivation; furthermore, the coloaded microspheres achieved a sustained release action to reduce the TMZ dosage, offering the potential for improved treatment of glioblastomas.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Aspirin/administration & dosage , Dacarbazine/analogs & derivatives , Glioma/drug therapy , Transcriptional Activation/drug effects , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism , Animals , Cell Line, Tumor , Dacarbazine/administration & dosage , Drug Carriers/administration & dosage , Drug Synergism , Glioma/genetics , Glioma/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Microspheres , Random Allocation , Temozolomide , Transcriptional Activation/genetics , Treatment Outcome , Xenograft Model Antitumor Assays/methods , beta Catenin/genetics
7.
CNS Neurosci Ther ; 18(9): 722-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22709411

ABSTRACT

BACKGROUND AND PURPOSE: As an important oncogenic miRNA, miR-21 has been reported to play crucial roles in glioblastoma (GBM) carcinogenesis. However, the precise biological function and molecular mechanism of miR-21 in GBM remain elusive. This study is designed to explore the mechanism of miR-21 involved in the control of GBM cell growth. METHODS AND RESULTS: MTT assay, cell cycle analysis, and apoptosis analysis showed that reduction of miR-21 inhibited cell growth in U87 and LN229 GBM cells. Further, reduction of miR-21 decreased the expression of human telomerase reverse transcriptase (hTERT) and repressed STAT3 expression and STAT3 phosphorylation. STAT3 inhibition led to a remarkable depletion of hTERT at both mRNA and protein levels by binding to the hTERT gene promoter by performing luciferase reporter assay and chromatin Immunoprecipitation PCR. Finally, knockdown of miR-21 considerably inhibited tumor growth and diminished the expression of STAT3 and hTERT in xenograft model. CONCLUSION: Our findings indicate that miR-21 regulates hTERT expression mediated by STAT3, therefore controlling GBM cell growth.


Subject(s)
Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/physiology , Glioblastoma/metabolism , MicroRNAs/metabolism , STAT3 Transcription Factor/metabolism , Telomerase/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Glioblastoma/genetics , Glioblastoma/pathology , Humans
8.
CNS Neurosci Ther ; 18(7): 573-83, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22630347

ABSTRACT

AIMS: MicroRNA-21 (miR-21) expression is increased in many types of human malignancy, including glioma. Recent studies report that miR-21 regulates cell invasion by targeting RECK, however, the underlying transcriptional regulation of miR-21 in glioma cells remains elusive. RESULTS: Here, we identify a positive correlation between miR-21 expression and pathological grade in glioma tissues. We demonstrate that ß-catenin pathway regulates miR-21 expression in human umbilical vein endothelial cell and glioma cells, and that this regulation is signal transducer and activator of transcription 3 (STAT3)-dependent. Further, chromatin immunoprecipitation and luciferase reporter analysis demonstrate that miR-21 is controlled by an upstream promoter containing a conserved STAT3 binding site. Notably, knockdown of miR-21-inhibited cell invasion by increasing RECK expression and decreased tumor growth in a xenograft model. CONCLUSION: These data provide compelling evidence that ß-catenin regulation of miR-21 via STAT3 plays a role in glioma cell invasion and proliferation and indicate that STAT3 is a potential therapeutic target for glioma intervention.


Subject(s)
GPI-Linked Proteins/metabolism , Gene Expression Regulation, Neoplastic , Glioma/metabolism , MicroRNAs/metabolism , STAT3 Transcription Factor/physiology , beta Catenin/physiology , Animals , Cell Line, Tumor , Down-Regulation/genetics , GPI-Linked Proteins/genetics , Gene Knockdown Techniques/methods , Gene Targeting/methods , Glioma/genetics , Glioma/pathology , Humans , Mice , Mice, Nude , MicroRNAs/biosynthesis , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Umbilical Veins/cytology , Umbilical Veins/metabolism , Up-Regulation/genetics , Xenograft Model Antitumor Assays/methods
9.
Zhonghua Wai Ke Za Zhi ; 50(11): 1015-20, 2012 Nov.
Article in Chinese | MEDLINE | ID: mdl-23302488

ABSTRACT

OBJECTIVES: To study the different expression of miRNA between pediatric and adult types of brainstem gliomas, and to provide the target miRNAs for explore the mechanism and miRNA interference of the malignant progression of pediatric BSG. METHODS: miRNA expression profiles in orthotopic models which could simulate the BSG heterogeneity were examined by microarray and analyzed to obtain the aberrantly expressed miRNAs. The two types of human BSG tissue were utilized to verify the microarray data by qRT-PCR and in situ hybridization for the putative causative miRNAs. RESULTS: There were 216 miRNAs detected in both the pediatric BSG group and the adult BSG group, 39 miRNAs to be differential expressed in the pediatric BSG group versus adult group, including 10 up-regulated and 29 down-regulated. qRT-PCR and in situ hybridization indicated good consistency with that of the microarray method. CONCLUSIONS: Aberrantly expressed miRNA may serve as putative causative involvement of malignant progression of pediatric BSG, thereby might be potentially novel targets for therapy.


Subject(s)
Brain Stem Neoplasms/metabolism , Glioma/metabolism , MicroRNAs/metabolism , Adult , Age Factors , Animals , Brain Stem , Child , Disease Models, Animal , Female , Gene Expression Profiling , Humans , In Situ Hybridization , Oligonucleotide Array Sequence Analysis , Rats
10.
Mol Cancer ; 9: 229, 2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20813046

ABSTRACT

BACKGROUND: MiR-221 and miR-222 (miR-221/222) are frequently up-regulated in various types of human malignancy including glioblastoma. Recent studies have reported that miR-221/222 regulate cell growth and cell cycle progression by targeting p27 and p57. However the underlying mechanism involved in cell survival modulation of miR-221/222 remains elusive. RESULTS: Here we showed that miR-221/222 inhibited cell apoptosis by targeting pro-apoptotic gene PUMA in human glioma cells. Enforced expression of miR-22/222 induced cell survival whereas knockdown of miR-221/222 rendered cells to apoptosis. Further, miR-221/222 reduced PUMA protein levels by targeting PUMA-3'UTR. Introducing PUMA cDNA without 3'UTR abrogated miR-221/222-induced cell survival. Notably, knockdown of miR-221/222 induces PUMA expression and cell apoptosis and considerably decreases tumor growth in xenograft model. Finally, there was an inverse relationship between PUMA and miR-221/222 expression in glioma tissues. CONCLUSION: To our knowledge, these data indicate for the first time that miR-221/222 directly regulate apoptosis by targeting PUMA in glioblastoma and that miR-221/222 could be potential therapeutic targets for glioblastoma intervention.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Glioblastoma/metabolism , Glioblastoma/therapy , MicroRNAs/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Blotting, Northern , Blotting, Western , Cell Line, Tumor , Cell Survival/physiology , Female , Glioblastoma/genetics , Humans , Immunohistochemistry , In Situ Hybridization , In Vitro Techniques , Mice , Mice, Nude , MicroRNAs/genetics , NIH 3T3 Cells , Proto-Oncogene Proteins/genetics , Xenograft Model Antitumor Assays
11.
Mol Cancer ; 9: 252, 2010 Sep 21.
Article in English | MEDLINE | ID: mdl-20858269

ABSTRACT

ß-catenin, a key factor in the Wnt signaling pathway, has essential functions in the regulation of cell growth and differentiation. Aberrant ß-catenin signaling has been linked to various disease pathologies, including an important role in tumorigenesis. Here, we review the regulation of the Wnt signaling pathway as it relates to ß-catenin signaling in tumorigenesis, with particular focus on the role of microRNAs. Finally, we discuss the potential of ß-catenin targeted therapeutics for cancer treatment.


Subject(s)
MicroRNAs/metabolism , Signal Transduction/physiology , beta Catenin/metabolism , Animals , Humans , MicroRNAs/genetics , Models, Biological , Neoplasms/genetics , Neoplasms/metabolism , Signal Transduction/genetics , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics
12.
Int J Oncol ; 37(2): 299-305, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20596657

ABSTRACT

miRNAs are non-coding, single-stranded RNAs that regulate target gene expression by repressing translation or promoting RNA cleavage. Dicer is an essential component of the miRNA processing machinery. To identify a role for miRNAs in tumorigenesis, we designed an adenovirus expressing small hairpin RNA (shRNA) to silence Dicer and globally suppress the maturation of miRNAs. We identified that the impairment of miRNA processing conferred an enhanced proliferative activity and invasive ability on each of three tumor cell lines in vitro. Inhibition of Dicer was associated with activation of p-Akt and enhanced expression of the cell cycle associating molecules, cyclin A and PCNA, as well as MMP-2 and MMP-9, proteins involved in tumor cell invasion. Adenoviral gene silencing of Dicer in subcutaneous MCF-7 xenografts significantly increased tumor growth in vivo compared to tumors infected with non-loading adenovirus. Increased tumor growth was associated with p-Akt activation and upregulation of cyclin A, PCNA MMP-2 and MMP-9. These findings demonstrate that global reduction of miRNA processing by silencing Dicer enhances tumor proliferation and invasion, and the p-Akt pathway may contribute to this phenotype via the downstream molecules, cyclin A, PCNA, MMP-2 and MMP-9.


Subject(s)
Cell Proliferation , Neoplasms/pathology , Ribonuclease III/genetics , Animals , Cell Proliferation/drug effects , Down-Regulation/drug effects , Down-Regulation/physiology , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Genetic Therapy , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Invasiveness , Neoplasms/genetics , Neoplasms/metabolism , RNA, Small Interfering/pharmacology , Ribonuclease III/antagonists & inhibitors , Transfection , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
J Mater Sci Mater Med ; 21(9): 2673-81, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20535631

ABSTRACT

The aim of present study is to conceive a biodegradable poly(ethylene glycol)-polylactide (PEG-PLA) copolymer nanoparticle which can be surface biofunctionalized with ligands via biotin-avidin interactions and used as a potential drug delivery carrier targeting to brain glioma in vivo. For this aim, a new method was employed to synthesize biotinylated PEG-PLA copolymers, i.e., esterification of PEG with biotinyl chloride followed by copolymerization of hetero-biotinylated PEG with lactide. PEG-PLA nanoparticles bearing biotin groups on surface were prepared by nanoprecipitation technique and the functional protein transferrin (Tf) were coupled to the nanoparticles by taking advantage of the strong biotin-avidin complex formation. The flow cytometer measurement demonstrated the targeting ability of the nanoparticles to tumor cells in vitro, and the fluorescence microscopy observation of brain sections from C6 glioma tumor-bearing rat model gave the intuitive proof that Tf functionalized PEG-PLA nanoparticles could penetrate into tumor in vivo.


Subject(s)
Brain Neoplasms/drug therapy , Glioma/drug therapy , Lactic Acid/chemistry , Micelles , Polyethylene Glycols/chemistry , Transferrin/chemistry , Humans
14.
Int J Oncol ; 36(3): 665-72, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20126987

ABSTRACT

Activation of the AKT (serine-threonine kinase) pathway is a common feature in glioblastoma cells. Downstream factors of the AKT pathway are involved in cell proliferation, apoptosis, cellular migration and angiogenesis. Micro-RNAs (miRNAs) are highly conserved small non-coding RNAs that block targeted mRNA expression at the post-transcriptional level. The aim of this study was to investigate the role of the AKT pathway in regulating miRNA. The changes of miRNA expression profile in human glioblastome U251 cells after AKT small interfering RNA transfection were examined by a microarray, and confirmed by Northern blotting. Down-regulation of AKT expression by siRNA decreased the activity of AKT pathway in U251 cells. Interruption of AKT pathway suppressed the expression of NF-kappaB and c-Myc, furthermore, the expression of a set of miRNAs was also changed after AKT siRNA transfection. There are putative binding sites of NF-kappaB and c-Myc in the promoters of several up-regulated miRNAs, indicating these transcription factors may also be involved in the regulation of miRNA expression, thus affecting the activity of AKT pathway in tumorigenesis. We provide new components of the regulatory function of AKT pathway to better understand the regulatory network mediated by downstream transcription factors. The understanding of the regulatory function of AKT pathway is crucial in tailored therapy of gliomas.


Subject(s)
Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , MicroRNAs/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Binding Sites , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Line, Tumor , Gene Expression Profiling , Glioma/drug therapy , Glioma/metabolism , Humans , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/metabolism , RNA, Messenger/metabolism , Signal Transduction , Transcription, Genetic
15.
J Biomater Sci Polym Ed ; 21(3): 303-14, 2010.
Article in English | MEDLINE | ID: mdl-20178687

ABSTRACT

MicroRNAs have been demonstrated to be deregulated in different types of cancer. miR-21 is a key player in the majority of cancers. Down-regulation of miR-21 in glioblastoma cells leads to repression of cell growth, increased cellular apoptosis and cell-cycle arrest, which can theoretically enhance the chemotherapeutic effect in cancer therapy. In this study, the poly(amidoamine) (PAMAM) dendrimer was employed as a carrier to co-deliver antisense-miR-21 oligonucleotide (as-miR-21) and 5-fluorouracil (5-FU) to achieve delivery of as-miR-21 to human glioblastoma cells and enhance the cytotoxicity of 5-FU antisense therapy. The inhibitory effect toward brain tumors was evaluated by MTT assay, and measurements of cell apoptosis and invasion using the human brain glioma cell line U251. PAMAM could be simultaneously loaded with 5-FU and as-miR-21, forming a complex smaller than 100 nm in diameter. Both the chemotherapeutant and as-miR-21 could be efficiently introduced into tumor cells. The co-delivery of as-miR-21 significantly improved the cytotoxicity of 5-FU and dramatically increased the apoptosis of U251 cells, while the migration ability of the tumor cells was decreased. These results suggest that our co-delivery system may have important clinical applications in the treatment of miR-21-overexpressing glioblastoma.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Cell Proliferation/drug effects , Dendrimers/chemistry , Fluorouracil/administration & dosage , Glioma/drug therapy , MicroRNAs/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic , Glioma/genetics , Humans , MicroRNAs/genetics , Transfection
16.
BMC Cancer ; 10: 27, 2010 Jan 31.
Article in English | MEDLINE | ID: mdl-20113523

ABSTRACT

BACKGROUND: Substantial data indicate that the oncogene microRNA 21 (miR-21) is significantly elevated in glioblastoma multiforme (GBM) and regulates multiple genes associated with cancer cell proliferation, apoptosis, and invasiveness. Thus, miR-21 can theoretically become a target to enhance the chemotherapeutic effect in cancer therapy. So far, the effect of downregulating miR-21 to enhance the chemotherapeutic effect to taxol has not been studied in human GBM. METHODS: Human glioblastoma U251 (PTEN-mutant) and LN229 (PTEN wild-type) cells were treated with taxol and the miR-21 inhibitor (in a poly (amidoamine) (PAMAM) dendrimer), alone or in combination. The 50% inhibitory concentration and cell viability were determined by the MTT assay. The mechanism between the miR-21 inhibitor and the anticancer drug taxol was analyzed using the Zheng-Jun Jin method. Annexin V/PI staining was performed, and apoptosis and the cell cycle were evaluated by flow cytometry analysis. Expression of miR-21 was investigated by RT-PCR, and western blotting was performed to evaluate malignancy related protein alteration. RESULTS: IC(50) values were dramatically decreased in cells treated with miR-21 inhibitor combine with taxol, to a greater extent than those treated with taxol alone. Furthermore, the miR-21 inhibitor significantly enhanced apoptosis in both U251 cells and LN229 cells, and cell invasiveness was obviously weakened. Interestingly, the above data suggested that in both the PTEN mutant and the wild-type GBM cells, miR-21 blockage increased the chemosensitivity to taxol. It is worth noting that the miR-21 inhibitor additively interacted with taxol on U251cells and synergistically on LN229 cells. Thus, the miR-21 inhibitor might interrupt the activity of EGFR pathways, independently of PTEN status. Meanwhile, the expression of STAT3 and p-STAT3 decreased to relatively low levels after miR-21 inhibitor and taxol treatment. The data strongly suggested that a regulatory loop between miR-21 and STAT3 might provide an insight into the mechanism of modulating EGFR/STAT3 signaling. CONCLUSIONS: Taken together, the miR-21 inhibitor could enhance the chemo-sensitivity of human glioblastoma cells to taxol. A combination of miR-21 inhibitor and taxol could be an effective therapeutic strategy for controlling the growth of GBM by inhibiting STAT3 expression and phosphorylation.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , Paclitaxel/pharmacology , Cell Line, Tumor , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Inhibitory Concentration 50 , Models, Biological , Phosphorylation , STAT3 Transcription Factor/metabolism , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology
17.
Technol Cancer Res Treat ; 9(1): 77-86, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20082533

ABSTRACT

The successful of anti-cancer treatment are often limited by the development of drug resistance. Recent work has highlighted the involvement of non-coding RNAs, microRNAs(miRNAs) in cancer development, and their possible involvement in the evolution of drug resistance has been proposed. In this study, we combine taxol chemotherapy and miR-21 inhibitor treatment via polyamidoamine (PAMAM) dendrimers vector to evaluate the effects of combination therapy on suppression of breast cancer cells. The 50% inhibitory concentration (IC50) values for taxol were significantly decreased to a greater extent in the cells transfected with miR-21 inhibitor compared with cells treated with taxol alone. Taxol treatment also increased the percentage of apoptotic breast cancer cells in miR-21 inhibitor transfected cells compared with control cells. Furthermore, treatment of the miR-21 inhibitor-transfected cells with the anti-cancer drugs taxol resulted in significantly reduced cell viability and invasiveness compared with control cells. These results indicated that the miR-21 plays an important role in the resistance of breast carcinoma cells to chemotherapeutic drugs. Therefore, miR-21 inhibitor gene therapy combined with taxol chemotherapy might represent a promising novel therapeutic approach for the treatment of breast malignancies.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/therapy , Dendrimers/pharmacology , Genetic Therapy , MicroRNAs/antagonists & inhibitors , Paclitaxel/pharmacology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Movement , Cell Proliferation/drug effects , Combined Modality Therapy , Down-Regulation , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/metabolism , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Transfection
18.
Med Oncol ; 27(3): 843-52, 2010 Sep.
Article in English | MEDLINE | ID: mdl-19728186

ABSTRACT

The over-expression/amplification of the epidermal growth factor receptor (EGFR) gene and mutation/deletion of tumor suppressor PTEN gene are main genetic changes identified in glioblastomas. These two genetic changes play a critical role in the formation of many malignant tumors and have been shown to be the important therapeutic targets. In this study, we used an expression plasmid that expresses small hairpin RNA-targeting sequences of human EGFR and wild-type PTEN cDNA to examine the growth inhibitive effects in U251 glioma cells. It was found that down-regulation of EGFR expression and up-regulation of PTEN expression resulted in the suppression of cell proliferation, arrest of cell cycle, reduction in cell invasion and promotion of cell apoptosis in vitro. In addition, the growth of the subcutaneous U251 glioma in the nude mice treated with expression plasmid was significantly inhibited. Our results demonstrated that the expression plasmid could exert proliferation and invasion inhibition effects on U251 cells in vitro and in vivo. It suggested that combinatory gene therapy targeting EGFR and PTEN would be a new strategy in gene therapy of glioblastoma.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Genetic Therapy , Glioblastoma/therapy , Neoplasm Proteins/antagonists & inhibitors , PTEN Phosphohydrolase/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/therapeutic use , Animals , Apoptosis , Base Sequence , Cell Line, Tumor/drug effects , DNA, Complementary/genetics , ErbB Receptors/biosynthesis , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Targeting , Genes, erbB-1 , Glioblastoma/pathology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Neoplasm Invasiveness , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , PTEN Phosphohydrolase/biosynthesis , PTEN Phosphohydrolase/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacology , Xenograft Model Antitumor Assays
19.
J Neurooncol ; 98(3): 329-40, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20035367

ABSTRACT

Our previous study demonstrated that SEPT7 was downregulated at mRNA level in human gliomas. This study is to further examine the expression of SEPT7 in glioma samples and characterizes its role on cell cycle progression and growth of glioma cells. mRNA and protein expression of SEPT7 were detected by RT-PCR, immunohistochemical staining, and western blot analysis in human glioma specimens and normal brain tissues. A pcDNA3-SEPT7 expression plasmid was constructed and transfected into human glioblastoma cell line U251, and cell proliferation and apoptosis were examined. The growth of established U251 and TJ905 subcutaneous xenograft gliomas was measured in nude mice treated with pcDNA3-SEPT7 and U251 xenograft tumors treated with SEPT7 siRNA. SEPT7 expression is negatively correlated with the increase of glioma grade. Overexpression of SEPT7 is able to inhibit cell proliferation and arrest cell cycle progression in the G0/G1 phase both in vitro and in vivo. Knocking down further the already low endogenous expression of SEPT7 in U251 xenograft tumors with siRNA leads to faster tumor growth compared with control tumors. This study demonstrates that SEPT7 is involved in gliomagenesis and suppresses glioma cell growth.


Subject(s)
Brain Neoplasms/metabolism , Cytoskeletal Proteins/metabolism , GTP-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/physiology , Glioma/metabolism , Analysis of Variance , Animals , Apoptosis/physiology , Brain/metabolism , Brain Neoplasms/pathology , Caspase 3/metabolism , Cell Cycle/physiology , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Cytoskeletal Proteins/genetics , Disease Models, Animal , Flow Cytometry/methods , GTP-Binding Proteins/genetics , Glial Fibrillary Acidic Protein/metabolism , Glioma/pathology , Humans , In Situ Nick-End Labeling/methods , Mice , Mice, Nude , Neoplasm Transplantation/mortality , Neoplasm Transplantation/pathology , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Septins , Time Factors , Tissue Array Analysis/methods , Transfection/methods
20.
Zhonghua Zhong Liu Za Zhi ; 31(10): 721-6, 2009 Oct.
Article in Chinese | MEDLINE | ID: mdl-20021821

ABSTRACT

OBJECTIVE: To study the inhibitory effect of knocking down microRNA(miR)-221 and miR-222 on human glioma cell growth and its possible mechanism. METHODS: miRNA-221/222 antisense oligonucleotides (antisense miR221/222) were transfected into human glioma U251 cells by lipofectamine. Northern blot analysis was conducted to detect the mRNA expression of miR-221/222 in the control and transfected cell groups. The proliferation activity of cells was determined by MTT assay. Cell invasion ability was examined by transwell assay, and cell cycle kinetics and apoptosis were detected with flow cytometry. The expression of relevant proteins was analyzed by Western blotting. The therapeutic efficacy of antisense miR221/222 on the growth of xenograft tumors in nude mice were also observed. RESULTS: In the antisense miR-221/222-transfected cells, the expression of miR-221/222 was significantly reduced; the cell invasion ability was suppressed, cell cycle was blocked at G(0)/G(1) phase, and apoptotic cells were increased. The growth of xenograft tumors treated with antisense miR-221/222 was also inhibited. In antisense miR-221/222 treated tumor cells, the expression of bcl-2 was down-regulated while connexin43, p27, PUMA, caspase-3, PTEN, TIMP3 and Bax up-regulated, and p53 expression not changed. CONCLUSION: There is a significant inhibitory effect of antisense miR-221/222 on the growth of human glioma U251 cells. miR-221/222 may be considered as a candidate target for gene therapy of human gliomas.


Subject(s)
Cell Proliferation , Glioma/pathology , MicroRNAs/biosynthesis , Animals , Apoptosis , Base Sequence , Caspase 3/metabolism , Cell Cycle , Cell Line, Tumor , Down-Regulation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Genetic Therapy , Glioma/metabolism , Humans , Ki-67 Antigen/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Molecular Sequence Data , Neoplasm Transplantation , Oligonucleotides, Antisense/pharmacology , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...