Skip to main content
Erschienen in: Cancer Cell International 1/2017

Open Access 01.12.2017 | Primary Research

Restoration of miR-29b exerts anti-cancer effects on glioblastoma

verfasst von: Jaekyung Shin, Hyun Geun Shim, Taeyoung Hwang, Hyungsin Kim, Shin-Hyuk Kang, Yun-Sik Dho, Sung-Hye Park, Sang Jeong Kim, Chul-Kee Park

Erschienen in: Cancer Cell International | Ausgabe 1/2017

Abstract

Background

Glioblastoma multiforme (GBM) is known as one of the most fatal forms of cancer. MicroRNAs have been widely implicated in the regulation of mammalian development and pathogenesis. The brain-enriched miR-29 subfamilies are known to be exclusively expressed in the developing brain, and they are aberrantly down-regulated in GBM. This study aims to elucidate the role of miR-29b in GBM development and the feasibility of therapeutic targeting using conjugated nanoparticles.

Methods

After confirmation of miR-29b expression levels in GBM tissues by analysis of open source data, the anticancer effect of miR-29b was tested by the introduction of syn-hsa-miR-29b-3p in the A172 GBM cell line. In vitro studies of cell viability and apoptosis and ex vivo study using GBM tissue slice cultures from 3 patients and nanoparticle delivery of miR-29b were performed.

Results

We discovered an increase in apoptotic cell populations with the introduction of miR-29b in the GBM cell line. An established human-derived GBM tissue slice culture system confirmed the anticancer effect of miR-29b-conjugated nanoparticles. Using PCR array, we found that exogenous miR-29b inhibits the expression of COL1A2, COL3A1, COL4A1, ELN, ITGA11, MMP24, and SPARC, which mediates an anticancer effect.

Conclusions

miR-29b may serve as a putative therapeutic molecule when its expression is restored using a nanoparticle delivery system in GBM.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12935-017-0476-9) contains supplementary material, which is available to authorized users.

Background

Glioblastoma multiforme (GBM) is known as one of the most fatal forms of brain cancer in humans, with an average survival duration of approximately 14 months [1]. However, because of pacesetting and intensive research on genomic profiles in GBM, a substantial amount of knowledge regarding its oncogenesis is rapidly accumulating [2]. The current understanding of the genomic characteristics of GBM extend from the genetic and epigenetic characteristics of the disease to their regulatory mechanisms encompassing posttranslational modifications, chromosome remodelling, and small and long non-coding RNAs (ncRNAs).
MicroRNAs (miRNAs) are small, ncRNAs of approximately 22 nucleotides in length that can regulate the expression of messenger RNAs (mRNAs) [3]. The targets of a single miRNA can be multiple and multiple miRNAs can be used against a single target [4]. Accumulating evidence has demonstrated that miRNAs act as crucial regulators of cancer [5]. Since the initial studies that demonstrated the aberrant expression of miRNAs in GBM samples, hundreds of papers have been published regarding the role of miRNAs in GBM [68]. An extensive review of all those studies concluded that 253 miRNAs are significantly up-regulated and 95 miRNAs are down-regulated in GBM tissues compared with those in normal brain tissues [8]. However, most of the aberrantly expressed miRNAs are yet to be functionally characterized [8].
Based on our previous study, we investigated miR-29b for its function in inhibiting the self-renewal and proliferation of neural stem cells, which can significantly affect neurogenesis [9]. Therefore, we postulated that there might be a certain role of miR-29b in brain tumour development considering the similarity of tumour progenitor cells with neural stem cells. In humans, the genetic loci encoding miR-29 consist of two gene clusters, the miR-29a/b-1 locus on chromosome 7q32 and the miR-29b-2/c locus on 1q23. While miR-29a localizes to the cytoplasm, miR-29b and miR-29c are found in the nucleus [10, 11]. There is evidence of the role of miR-29s in a variety of cancer types [12]. Here, we demonstrate that miR-29b is a tumour suppressor that is deregulated in GBM, and its restoration can exert an anti-cancer effect.

Methods

Cell line and culture

The human GBM cell line A172 was purchased from American Type Culture Collection (ATCC® CRL-1620™, Manassas, VA, USA) and cultured in Dulbecco’s Modified Eagle’s Medium (DMEM, WelGene, Dae-Ku, Korea) containing 10% foetal bovine serum (FBS, Gibco, Grand Island, NY, USA) and 5% antibiotics (streptomycin) in a humidified atmosphere of 5% CO2 and 95% air at 37 °C.

RNA interference and reagents

For RNA interference, non-targeting control siRNA (#D-001610-01-05, Dharmacon, Lafayette, CO, USA) and syn-hsa-miR-29b-3p (#MSY0000100, Qiagen, Hilden, Germany) were purchased, and Lipofectamine® RNAiMAX Reagent (#13776-150, Invitrogen, Carlsbad, CA, USA) was used to transfect cells.

Flow cytometry

A total of 1 × 105 A172 cells were plated in 6-well dishes and incubated with 30–50 nM control siRNA or syn-hsa-miR-29b-3p with Lipofectamine® RNAiMAX Reagent according to the manufacturer’s protocol for 3 days at 37 °C. The cells were fixed with ice-cold 70% ethanol after harvesting, washed once with phosphate-buffered saline (PBS) and resuspended in PBS, and then ribonuclease A (0.1%) and propidium iodide (PI, 10 µg/ml) were added to the cells. The cells were incubated for 30 min in the dark at room temperature. For apoptosis assays, cells were harvested and suspended in Annexin V binding buffer (#556570, BD Biosciences Pharmingen, San Diego, CA, USA) at a density of 1 × 105 cells/100 μl. Next, 5 μl of Annexin V-FITC and 5 μl of PI were added to each sample, and the samples were incubated in the dark at room temperature for 15 min.
Fluorescence events were quantified after laser excitation of the fluorescent dye with a fluorescence-activated cell sorter (FACSCalibur, Becton–Dickinson, Heidelberg, Germany) with a cell count of 1 × 105–106 cells per 0.2–0.3 ml. The data were processed using Cell Quest Pro software (version 5.2.1, BD Biosciences).

Cell viability

Cell viability was tested using the LIVE/DEAD® Viability/Cytotoxicity Kit (#L3224, Molecular Probes, Inc., Eugene, OR, USA) according to the manufacturer’s instructions. In brief, 5 × 104 A172 cells were seeded onto a 0.1% gelatine-coated slides, and 20 μM syn-hsa-miR-29b-3p was added to a final concentration of 30–50 nM. Calcein-AM and ethidium bromide were added for 15 min at room temperature. The slides were mounted with 1× PBS and immediately photographed.

Glioblastoma-derived tissue slice culture

Glioblastoma samples were obtained from 3 patients during surgery after the patients provided informed consent and the samples were immersed in ice-cold sterile 1× HBSS, orientated, mounted, and immobilized using cyanoacrylate glue immediately after resection. Using a Vibratome VT1200 (Leica Microsystems Inc., Buffalo Grove, IL, USA), the most viable regions of the tissue were selected, and 400 μm-thick slices were cut. The vibration amplitude was set at 2.0 mm, and the slicing speed was 0.03–0.05 mm/s. The tissue slices were placed in 6-well plates on membrane inserts with 0.4 μm pores (#PICM03050, Millipore Corp., Bedford, MA, USA). The tissue slices were cultured at 37 °C in a humidified incubator with 5% CO2 for 10 days using 1 ml of MEM supplemented with 10% FBS. The genetic characteristics of the samples used for the tissue slice culture are summarized in Additional file 1: Table S1.

Nanoparticle complex

We used InViVojection™ RNAi-nano Red reagent (#DHMSN-vivoRFRNA25, Lemonex Inc., Seoul, Korea) for the delivery of control siRNA and miR-29b into tissue slice cultures according to the manufacturer’s instructions. In brief, 80 μl of InViVojection™ reagent was mixed with 10 μl of 10× PBS in the dark, and 10 μl of miR-29b (100 μM stock) or control siRNA was added. After incubation at room temperature for 1 h, the reagent mixture was carefully dropped on top of the slices to soak in. The incorporation of nanoparticle complex into tissues was confirmed by transmission electron microscopy (TEM).

Immunohistochemistry

The cultured tissue slices at day 0 and at day 7 were formalin-fixed and paraffin-embedded (FFPE) and cut into 4 μm sections for immunohistochemistry (IHC).
To assay proliferation, primary antibodies against human Ki-67 (1:1000, #M724029, DAKO, Glostrup, Denmark) were used. A nuclear algorithm of the Aperio ScanScope image analysis program (Aperio Technologies, Vista, CA, USA) was used to calculate the nuclear positivity in hot spots for the Ki-67 labelling index.
To assay apoptosis, terminal transferase was used to visualize fragmented DNA (terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling, TUNEL labelling, In Situ Cell Death Detection Kit, POD, #1,684,817, Roche Applied Science, Mannheim, Germany). The apoptotic index (%) was calculated by dividing the number of apoptotic nuclei by the total number of nuclei counted. The cells were counterstained with methyl green.

Polymerase chain reaction array

To identify the targets of human miR-29b in GBM cells, we performed polymerase chain reaction (PCR) using the RT2 Profiler™ PCR Array kit (#PAHS-6012Z, SA Biosciences, Frederick, MD, USA). Ct values were converted to fold change, using web-accessible analysis programs (http://​pcrdataanalysis.​sabiosciences.​com/​pcr/​arrayanalysis.​php). Fold change (\(2^{{ - \Delta \Delta {\text{C}}_{\text{T}} }}\)) was calculated from the normalized gene expression (\(2^{{ - \Delta {\text{C}}_{\text{T}} }}\)) in the test sample divided by the normalized gene expression (\(2^{{ - \Delta {\text{C}}_{\text{T}} }}\)) in the control sample. We present the target molecules with their log2 fold change (x-axis) and the corresponding log2 p values (y-axis) on a volcano plot.

Public database usage and statistical analysis

To screen the miR-29b expression levels in GBM tissues, we curated and combined the open source data from two previous studies comparing GBM tissues (n = 9) and non-tumour brain tissues (n = 6) using miRNA-seq [13, 14]. Specifically, raw counts of the sequencing reads for commonly identified miRNAs in both studies were obtained for every sample using the supplementary data from the studies. The differential expression of miRNAs between glioblastoma and non-tumour brain tissues was statistically evaluated using the R package DESeq2 that uses a negative binomial model [15]. In particular, we controlled for batch effect arising from the integration of the two separate studies by considering a variable that identifies the study providing the samples being analysed.
An integrated network analysis of the gene of interest was conducted and visualized using PCViz in Pathway Commons (http://​www.​pathwaycommons.​org) [16].
Quantitative data were expressed as the mean ± the standard error of the mean (SEM). The statistical significance between two groups was tested using the two-tailed Student’s t test. A p value of < 0.05 was considered significant.

Results

Expression of miR-29b is suppressed in GBM

We used open source miRNA profiling data from 2 previously published studies to compare the levels of expression of miR-29b between GBM and normal brain tissues [13, 14]. After correction for the batch effect of each dataset, a total of 9 GBM and 6 normal brain tissues could be analysed for 423 miRNAs that were commonly expressed. Among those miRNAs, a total of 170 miRNAs were significantly differentially expressed between the GBM and normal brain tissues (Table 1; Additional file 1: Tables S2, FDR < 0.1). Based on our previous study, we focused on miR-29b for its function in inhibiting the self-renewal and proliferation of neural stem cells, which can significantly affect neurogenesis [9]. miR-29b was found to be significantly down-regulated in GBM (Fig. 1, log2 (Normal/GBM) = 1.34, p = 0.02, FDR = 0.06).
Table 1
A total of 76 differentially expressed microRNAs with significantly lower expression in glioblastoma (GBM) compared to non-tumour brain tissues
No.
MicroRNA
log2(FC)
log2(FC-standard error)
z-statistic
p value
FDR
1
hsa-miR-124
7.5068
1.0387
7.2270
0.0000
0.0000
2
hsa-miR-218
6.6782
0.9775
6.8322
0.0000
0.0000
3
hsa-miR-873
5.4139
1.0405
5.2031
0.0000
0.0000
4
hsa-miR-7
4.7696
0.7774
6.1350
0.0000
0.0000
5
hsa-miR-1912
4.5089
1.8598
2.4243
0.0153
0.0429
6
hsa-miR-129star
4.5065
0.7294
6.1782
0.0000
0.0000
7
hsa-miR-139-3p
4.3718
0.8802
4.9666
0.0000
0.0000
8
hsa-miR-383
4.1483
1.0614
3.9083
0.0001
0.0007
9
hsa-miR-495
4.0638
0.9937
4.0894
0.0000
0.0004
10
hsa-miR-139-5p
3.8873
0.9888
3.9314
0.0001
0.0007
11
hsa-miR-138
3.7940
0.8260
4.5930
0.0000
0.0001
12
hsa-miR-485-5p
3.6911
0.7200
5.1262
0.0000
0.0000
13
hsa-miR-323-3p
3.5294
0.6566
5.3754
0.0000
0.0000
14
hsa-miR-487b
3.5207
0.7118
4.9464
0.0000
0.0000
15
hsa-miR-485-3p
3.4738
0.7005
4.9591
0.0000
0.0000
16
hsa-miR-1286
3.4257
0.9717
3.5256
0.0004
0.0025
17
hsa-miR-1224-5p
3.3436
0.7768
4.3045
0.0000
0.0002
18
hsa-miR-132
3.3423
0.6392
5.2287
0.0000
0.0000
19
hsa-miR-543
3.2264
0.6293
5.1272
0.0000
0.0000
20
hsa-miR-490-3p
3.1761
0.7582
4.1889
0.0000
0.0003
21
hsa-miR-490-5p
3.1672
0.8236
3.8454
0.0001
0.0009
22
hsa-miR-410
3.0772
0.8013
3.8405
0.0001
0.0009
23
hsa-miR-668
3.0100
0.7603
3.9588
0.0001
0.0006
24
hsa-miR-323b-3p
2.9812
0.8378
3.5582
0.0004
0.0023
25
hsa-miR-212
2.8464
0.6536
4.3552
0.0000
0.0002
26
hsa-miR-628-5p
2.8262
0.5173
5.4638
0.0000
0.0000
27
hsa-miR-124star
2.8005
0.8573
3.2666
0.0011
0.0055
28
hsa-miR-136
2.7895
0.7544
3.6977
0.0002
0.0014
29
hsa-miR-935
2.7497
0.7178
3.8304
0.0001
0.0009
30
hsa-miR-539
2.7319
0.9807
2.7857
0.0053
0.0194
31
hsa-miR-504
2.7238
0.8304
3.2800
0.0010
0.0053
32
hsa-miR-487a
2.7167
0.7557
3.5948
0.0003
0.0021
33
hsa-miR-138-1star
2.6958
0.5442
4.9533
0.0000
0.0000
34
hsa-miR-132star
2.6106
0.8890
2.9365
0.0033
0.0145
35
hsa-miR-770-5p
2.5856
0.9006
2.8711
0.0041
0.0165
36
hsa-miR-433
2.5657
1.0518
2.4394
0.0147
0.0415
37
hsa-miR-4446-3p
2.5107
0.9984
2.5147
0.0119
0.0351
38
hsa-miR-154star
2.5037
0.6443
3.8859
0.0001
0.0008
39
hsa-miR-375
2.4955
0.7418
3.3642
0.0008
0.0042
40
hsa-miR-431star
2.4278
0.8736
2.7791
0.0055
0.0196
41
hsa-miR-411star
2.3678
0.5828
4.0629
0.0000
0.0004
42
hsa-miR-342-3p
2.3513
0.6636
3.5430
0.0004
0.0024
43
hsa-miR-376astar
2.3385
0.8987
2.6022
0.0093
0.0293
44
hsa-miR-431
2.3223
1.1063
2.0991
0.0358
0.0851
45
hsa-miR-496
2.2728
0.8521
2.6672
0.0076
0.0254
46
hsa-miR-7-1star
2.1583
0.8207
2.6299
0.0085
0.0274
47
hsa-miR-769-3p
2.1347
0.5331
4.0047
0.0001
0.0005
48
hsa-miR-655
2.0954
0.9282
2.2574
0.0240
0.0606
49
hsa-miR-432
2.0845
0.7328
2.8443
0.0045
0.0175
50
hsa-miR-203
2.0536
0.7244
2.8349
0.0046
0.0175
51
hsa-miR-382
2.0529
0.7358
2.7901
0.0053
0.0193
52
hsa-miR-329
2.0327
0.8035
2.5298
0.0114
0.0344
53
hsa-miR-491-5p
2.0173
0.7588
2.6586
0.0078
0.0257
54
hsa-miR-221
1.9337
0.6314
3.0623
0.0022
0.0102
55
hsa-miR-379
1.9324
0.7444
2.5961
0.0094
0.0296
56
hsa-miR-369-5p
1.8083
0.7021
2.5755
0.0100
0.0307
57
hsa-miR-330-3p
1.8050
0.6517
2.7699
0.0056
0.0199
58
hsa-miR-379star
1.7531
0.7448
2.3540
0.0186
0.0502
59
hsa-miR-149
1.7529
0.6027
2.9083
0.0036
0.0153
60
hsa-miR-1
1.7498
0.4676
3.7418
0.0002
0.0012
61
hsa-miR-889
1.7203
0.7117
2.4174
0.0156
0.0435
62
hsa-miR-136star
1.6970
0.6581
2.5789
0.0099
0.0306
63
hsa-miR-153
1.5730
0.6368
2.4703
0.0135
0.0392
64
hsa-miR-766
1.5411
0.6396
2.4093
0.0160
0.0438
65
hsa-miR-377star
1.4851
0.6731
2.2063
0.0274
0.0678
66
hsa-miR-411
1.4768
0.6314
2.3389
0.0193
0.0513
67
hsa-miR-377
1.4701
0.6668
2.2046
0.0275
0.0678
68
hsa-miR-95
1.4609
0.6567
2.2247
0.0261
0.0656
69
hsa-miR-376a
1.4324
0.7042
2.0342
0.0419
0.0972
70
hsa-miR-127-5p
1.3517
0.6337
2.1329
0.0329
0.0787
71
hsa-miR-29b
1.3389
0.5925
2.2597
0.0238
0.0606
72
hsa-miR-425
1.3189
0.6458
2.0421
0.0411
0.0960
73
hsa-let-7d
1.2599
0.5884
2.1411
0.0323
0.0776
74
hsa-miR-191
1.1826
0.4351
2.7181
0.0066
0.0222
75
hsa-miR-598
1.1699
0.5339
2.1911
0.0284
0.0697
76
hsa-miR-769-5p
1.1113
0.4806
2.3121
0.0208
0.0543
Whole set of differentially expressed microRNAs between GBM and non-tumor brain tissues are listed in Additional file 1: Table S2. A total of 170 microRNAs were found to be significant after combining open source data from two previous studies comparing GBM (n = 9) and non-tumour brain tissues (n = 6) with miRNA-seq [13, 14]. Log2(FC) indicates log2(fold change of non-tumour brain tissues to GBM)

Restoration of miR-29b induces cell death and growth arrest in a GBM cell line

Introduction of miR-29b into A172, a PTEN-deficient GBM cell line, showed successful overexpression (Fig. 2a). FACS analysis with Annexin V staining revealed a marked increase in the apoptotic cell fraction after miR-29b treatment (Fig. 2b). The LIVE/DEAD cell staining confirmed a decrease in the viability of the miR-29b-treated cells (Fig. 2c). Cell cycle analysis showed a significant S-phase arrest after miR-29b treatment (Fig. 2d). Overall, miR-29b acts as a tumour suppressor, which can inhibit cell growth and induce apoptosis in vitro. To confirm the growth arrest effect of miR-29b, we performed flow cytometry experiments using the BrdU/7-AAD assay. Expression of miR-29b was followed by a significant induction of apoptosis and S-phase arrest in A172 cells, while the fraction of G1 and G2/M phase remained unaltered (Fig. 2e).

Introduction of nanoparticle loaded with miR-29b exerts anti-cancer effects on GBM-derived tissues in culture

We cultured GBM tissue slices (n = 3) for testing the anticancer effects of miR-29b ex vivo. The delivery of miR-29b into cancer cells in cultured tissue could be effectively achieved using nanoparticles (Fig. 3a). The anticancer effects were evaluated 7 days after treatment. There was a marked decrease in proliferation and significant increase in apoptosis after miR-29b treatment (Fig. 3b, c).

Targets of miR-29b in GBM cells

To elucidate miR-29b target genes in GBM, we analysed A172 cells using the Human miR-29 Targets RT2 Profiler PCR Array (QIAGEN #PAHS-6012Z) (Additional file 1: Table S3). By comparing the expression levels of 96 candidate target genes before and after miR-29b treatment, we found 75 genes that showed significant changes (p < 0.05) of either increased expression (10 genes) or decreased expression (65 genes). The noticeably down-regulated genes were COL1A2, COL4A1, COL2A1, ITGA11, MMP24, and SPARC (Fig. 4a). A network was constructed based on the regulatory relationships of the genes of interest using Pathway Commons (Fig. 4b). Among them, SPARC was found to be of particular interest, as its expression was remarkably reduced (21.14-fold decreased) after miR-29b treatment from being highly expressed in the GBM cell line.

Discussion

Similar to other cancer types, it has been theorized that GBM arises from neural stem cells (NSCs) that undergo genetic mutations in tumour suppressor genes and oncogenes [17]. Non-neoplastic cells that transform and produce gliomas are likely the direct descendants of common NSCs [18]. Therefore, it is plausible that oncogenesis of GBM shares a common genetic developmental mechanism with NSCs. We have previously shown that miR-29b plays a pivotal role in neurogenesis by regulating the inhibitor of β-catenin and T cell factor (ICAT)-mediated Wnt/β-catenin signalling in NSCs, which impacts their self-renewal and proliferation [9]. MicroRNAs regulate a wide range of gene expression in a post-transcriptional manner, and their aberrant function affects a variety of steps in tumour development. Among the microRNAs that are known to be deregulated in GBMs, miR-29b attenuates stemness in NSCs during brain development [9]. In the present study, we showed that the restoration of down-regulated miR-29b in GBM cells exerts anticancer effects by inhibiting cell cycle arrest. Furthermore, we demonstrated that the loss of ECM molecules in A172 cells accelerates cell apoptosis in GBMs. These results can be mediated via the epithelial-mesenchymal transition (EMT)-related processes that are correlated with ECM molecules including COL1A2, COL3A1, COL4A1, ELN, ITGA11 and MMP24. Accumulating evidence has shown that the miR-29 family is involved in multiple cancer types [5]. Also, miR-29b is generally recognized as a fundamental regulator of EMT [5].
Among the possible functional mediators of miR-29b, SPARC is of particular interest, as its expression was remarkably reduced after miR-29b treatment in GBM. SPARC induces changes in the migration [19], invasion [20] and angiogenesis of GBM cells in vitro. Additionally, SPARC affects tumour cell behaviour to activate the PI3K (phosphoinositide-3-kinase)-AKT pathway, a key mediator of cell survival [21]. SPARC can act as a potent oncogene and therapeutic RNAi target in GBM. This is in line with a previous study on breast cancer cells showing that C1QTNF6, SPARC, and COL4A2 were targeted by miR-29b [22]. Another study showed that miR-29b exerts a critical suppressive role on colorectal cancer mediated by the inhibition of Tiam1 and EMT [23].

Conclusions

miR-29b may be used as a therapeutic molecule when its expression is restored, which indicates its potential role in the molecular therapy of patients with GBM. Although the induction of apoptosis is the predominant anti-cancer role of miR-29b, analysis of the mediators of the miR-29b effect suggests that diverse mechanisms can contribute to tumour suppression, which need to be evaluated in the future.

Authors’ contributions

JS and CKP designed this study. JS, HGS, and HK carried out the study. TH and YSD performed the genomic analysis. SHP performed histological evaluation. JS, TH, and CKP drafted the manuscript. SHK, and SJK supervised the experimental work and reviewed the manuscript. All authors read and approved the final manuscript.

Acknowledgements

We thank Dr. Hyun Cheol Chung for his constant support and helpful advice.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

The datasets analysed during the current available from the corresponding authors on reasonable request.
All authors are responsible for the submission of this article and accept the conditions of submission.
This study was performed under the approval of the Institutional Review Board of Seoul National University Hospital, and all experiments were performed in accordance with relevant guidelines and regulations. Written informed consent was obtained from all patients for the usage of samples.

Funding

This study was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (NRF-2015R1D1A1A09057171) in Korea and the Seoul National University Hospital Research Fund (03-2015-0160).

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–96.CrossRefPubMed Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–96.CrossRefPubMed
2.
Zurück zum Zitat Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, Zheng S, Chakravarty D, Sanborn JZ, Berman SH, et al. The somatic genomic landscape of glioblastoma. Cell. 2013;155(2):462–77.CrossRefPubMedPubMedCentral Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, Zheng S, Chakravarty D, Sanborn JZ, Berman SH, et al. The somatic genomic landscape of glioblastoma. Cell. 2013;155(2):462–77.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–97.CrossRefPubMed Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–97.CrossRefPubMed
5.
Zurück zum Zitat Yan B, Guo Q, Fu FJ, Wang Z, Yin Z, Wei YB, Yang JR. The role of miR-29b in cancer: regulation, function, and signaling. OncoTargets Ther. 2015;8:539–48. Yan B, Guo Q, Fu FJ, Wang Z, Yin Z, Wei YB, Yang JR. The role of miR-29b in cancer: regulation, function, and signaling. OncoTargets Ther. 2015;8:539–48.
6.
Zurück zum Zitat Ciafre SA, Galardi S, Mangiola A, Ferracin M, Liu CG, Sabatino G, Negrini M, Maira G, Croce CM, Farace MG. Extensive modulation of a set of microRNAs in primary glioblastoma. Biochem Biophys Res Commun. 2005;334(4):1351–8.CrossRefPubMed Ciafre SA, Galardi S, Mangiola A, Ferracin M, Liu CG, Sabatino G, Negrini M, Maira G, Croce CM, Farace MG. Extensive modulation of a set of microRNAs in primary glioblastoma. Biochem Biophys Res Commun. 2005;334(4):1351–8.CrossRefPubMed
7.
Zurück zum Zitat Chan JA, Krichevsky AM, Kosik KS. MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Can Res. 2005;65(14):6029–33.CrossRef Chan JA, Krichevsky AM, Kosik KS. MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Can Res. 2005;65(14):6029–33.CrossRef
8.
Zurück zum Zitat Moller HG, Rasmussen AP, Andersen HH, Johnsen KB, Henriksen M, Duroux M. A systematic review of microRNA in glioblastoma multiforme: micro-modulators in the mesenchymal mode of migration and invasion. Mol Neurobiol. 2013;47(1):131–44.CrossRefPubMed Moller HG, Rasmussen AP, Andersen HH, Johnsen KB, Henriksen M, Duroux M. A systematic review of microRNA in glioblastoma multiforme: micro-modulators in the mesenchymal mode of migration and invasion. Mol Neurobiol. 2013;47(1):131–44.CrossRefPubMed
9.
Zurück zum Zitat Shin J, Shin Y, Oh SM, Yang H, Yu WJ, Lee JP, Huh SO, Lee SH, Suh YH, Chung S, et al. MiR-29b controls fetal mouse neurogenesis by regulating ICAT-mediated Wnt/beta-catenin signaling. Cell Death Dis. 2014;5:e1473.CrossRefPubMedPubMedCentral Shin J, Shin Y, Oh SM, Yang H, Yu WJ, Lee JP, Huh SO, Lee SH, Suh YH, Chung S, et al. MiR-29b controls fetal mouse neurogenesis by regulating ICAT-mediated Wnt/beta-catenin signaling. Cell Death Dis. 2014;5:e1473.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Amodio N, Rossi M, Raimondi L, Pitari MR, Botta C, Tagliaferri P, Tassone P. miR-29s: a family of epi-miRNAs with therapeutic implications in hematologic malignancies. Oncotarget. 2015;6(15):12837–61.CrossRefPubMedPubMedCentral Amodio N, Rossi M, Raimondi L, Pitari MR, Botta C, Tagliaferri P, Tassone P. miR-29s: a family of epi-miRNAs with therapeutic implications in hematologic malignancies. Oncotarget. 2015;6(15):12837–61.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Hwang HW, Wentzel EA, Mendell JT. A hexanucleotide element directs microRNA nuclear import. Science. 2007;315(5808):97–100.CrossRefPubMed Hwang HW, Wentzel EA, Mendell JT. A hexanucleotide element directs microRNA nuclear import. Science. 2007;315(5808):97–100.CrossRefPubMed
12.
Zurück zum Zitat Park SY, Lee JH, Ha M, Nam JW, Kim VN. miR-29 miRNAs activate p53 by targeting p85 alpha and CDC42. Nat Struct Mol Biol. 2009;16(1):23–9.CrossRefPubMed Park SY, Lee JH, Ha M, Nam JW, Kim VN. miR-29 miRNAs activate p53 by targeting p85 alpha and CDC42. Nat Struct Mol Biol. 2009;16(1):23–9.CrossRefPubMed
13.
Zurück zum Zitat Skalsky RL, Cullen BR. Reduced expression of brain-enriched microRNAs in glioblastomas permits targeted regulation of a cell death gene. PLoS ONE. 2011;6(9):e24248.CrossRefPubMedPubMedCentral Skalsky RL, Cullen BR. Reduced expression of brain-enriched microRNAs in glioblastomas permits targeted regulation of a cell death gene. PLoS ONE. 2011;6(9):e24248.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Hua D, Mo F, Ding D, Li L, Han X, Zhao N, Foltz G, Lin B, Lan Q, Huang Q. A catalogue of glioblastoma and brain MicroRNAs identified by deep sequencing. OMICS. 2012;16(12):690–9.CrossRefPubMedPubMedCentral Hua D, Mo F, Ding D, Li L, Han X, Zhao N, Foltz G, Lin B, Lan Q, Huang Q. A catalogue of glioblastoma and brain MicroRNAs identified by deep sequencing. OMICS. 2012;16(12):690–9.CrossRefPubMedPubMedCentral
15.
16.
Zurück zum Zitat Cerami EG, Gross BE, Demir E, Rodchenkov I, Babur O, Anwar N, Schultz N, Bader GD, Sander C. Pathway Commons, a web resource for biological pathway data. Nucleic Acids Res. 2011;39(Database issue):685–90.CrossRef Cerami EG, Gross BE, Demir E, Rodchenkov I, Babur O, Anwar N, Schultz N, Bader GD, Sander C. Pathway Commons, a web resource for biological pathway data. Nucleic Acids Res. 2011;39(Database issue):685–90.CrossRef
18.
Zurück zum Zitat de Almeida Sassi F, Lunardi Brunetto A, Schwartsmann G, Roesler R, Abujamra AL. Glioma revisited: from neurogenesis and cancer stem cells to the epigenetic regulation of the niche. J Oncol. 2012;2012:537861. de Almeida Sassi F, Lunardi Brunetto A, Schwartsmann G, Roesler R, Abujamra AL. Glioma revisited: from neurogenesis and cancer stem cells to the epigenetic regulation of the niche. J Oncol. 2012;2012:537861.
19.
Zurück zum Zitat Kunigal S, Gondi CS, Gujrati M, Lakka SS, Dinh DH, Olivero WC, Rao JS. SPARC-induced migration of glioblastoma cell lines via uPA-uPAR signaling and activation of small GTPase RhoA. Int J Oncol. 2006;29(6):1349–57.PubMedPubMedCentral Kunigal S, Gondi CS, Gujrati M, Lakka SS, Dinh DH, Olivero WC, Rao JS. SPARC-induced migration of glioblastoma cell lines via uPA-uPAR signaling and activation of small GTPase RhoA. Int J Oncol. 2006;29(6):1349–57.PubMedPubMedCentral
20.
Zurück zum Zitat Golembieski WA, Thomas SL, Schultz CR, Yunker CK, McClung HM, Lemke N, Cazacu S, Barker T, Sage EH, Brodie C, et al. HSP27 mediates SPARC-induced changes in glioma morphology, migration, and invasion. Glia. 2008;56(10):1061–75.CrossRefPubMed Golembieski WA, Thomas SL, Schultz CR, Yunker CK, McClung HM, Lemke N, Cazacu S, Barker T, Sage EH, Brodie C, et al. HSP27 mediates SPARC-induced changes in glioma morphology, migration, and invasion. Glia. 2008;56(10):1061–75.CrossRefPubMed
21.
Zurück zum Zitat Shi Q, Bao S, Maxwell JA, Reese ED, Friedman HS, Bigner DD, Wang XF, Rich JN. Secreted protein acidic, rich in cysteine (SPARC), mediates cellular survival of gliomas through AKT activation. J Biol Chem. 2004;279(50):52200–9.CrossRefPubMed Shi Q, Bao S, Maxwell JA, Reese ED, Friedman HS, Bigner DD, Wang XF, Rich JN. Secreted protein acidic, rich in cysteine (SPARC), mediates cellular survival of gliomas through AKT activation. J Biol Chem. 2004;279(50):52200–9.CrossRefPubMed
22.
Zurück zum Zitat Wang C, Gao C, Zhuang JL, Ding C, Wang Y. A combined approach identifies three mRNAs that are down-regulated by microRNA-29b and promote invasion ability in the breast cancer cell line MCF-7. J Cancer Res Clin Oncol. 2012;138(12):2127–36.CrossRefPubMed Wang C, Gao C, Zhuang JL, Ding C, Wang Y. A combined approach identifies three mRNAs that are down-regulated by microRNA-29b and promote invasion ability in the breast cancer cell line MCF-7. J Cancer Res Clin Oncol. 2012;138(12):2127–36.CrossRefPubMed
23.
Zurück zum Zitat Wang B, Li W, Liu H, Yang L, Liao Q, Cui S, Wang H, Zhao L. miR-29b suppresses tumor growth and metastasis in colorectal cancer via downregulating Tiam1 expression and inhibiting epithelial-mesenchymal transition. Cell Death Dis. 2014;5:e1335.CrossRefPubMedPubMedCentral Wang B, Li W, Liu H, Yang L, Liao Q, Cui S, Wang H, Zhao L. miR-29b suppresses tumor growth and metastasis in colorectal cancer via downregulating Tiam1 expression and inhibiting epithelial-mesenchymal transition. Cell Death Dis. 2014;5:e1335.CrossRefPubMedPubMedCentral
Metadaten
Titel
Restoration of miR-29b exerts anti-cancer effects on glioblastoma
verfasst von
Jaekyung Shin
Hyun Geun Shim
Taeyoung Hwang
Hyungsin Kim
Shin-Hyuk Kang
Yun-Sik Dho
Sung-Hye Park
Sang Jeong Kim
Chul-Kee Park
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Cancer Cell International / Ausgabe 1/2017
Elektronische ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-017-0476-9

Weitere Artikel der Ausgabe 1/2017

Cancer Cell International 1/2017 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.