Open Access

Comparing tumor microRNA profiles of patients with long‑ and short‑term‑surviving glioblastoma

  • Authors:
    • Björn Schneider
    • Nora Lamp
    • Annette Zimpfer
    • Christian Henker
    • Andreas Erbersdobler
  • View Affiliations

  • Published online on: November 10, 2022     https://doi.org/10.3892/mmr.2022.12895
  • Article Number: 8
  • Copyright: © Schneider et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Glioblastoma is one of the most frequent primary brain tumors with a poor prognosis. Nevertheless, some patients show a prolonged survival. The aim of the present study was to compare the expression profiles of tumor derived microRNA (miR) of long‑term survivors with those of short‑term survivors in order to identify differentially expressed miRs as well as their target genes, which may elucidate mechanisms that play a role in varying tumor progression and, therefore, may influence survival. Formalin‑fixed paraffin‑embedded samples of 23 patients with glioblastoma were classified according to overall survival. Profiles of miR expression were determined using Nanostring technology. Expression levels of potential target genes of differentially expressed miRs were assessed using immunohistochemistry. MiR profiles of long‑term survivors differed from those of short‑term survivors. A total of three prominent differentially expressed miRs were highlighted: MiR‑130b‑3p, which is downregulated in long‑term survivors, and miR‑146b‑5p and miR‑148a‑3p, which are upregulated in long‑term survivors. Known tumor suppressor genes are among targets potentially affected by miR‑130b‑3p, whereas targets of miR‑146b‑5p and miR‑148a‑3p consist of several genes known to have a role in tumor invasion and aggressiveness. In conclusion, it was revealed that a type of miR‑signature was associated with short‑ and long‑term survival, potentially serving as biomarker for disease progression and providing a base for further functional studies.

Introduction

Glioblastoma (GBM) is the most frequent and fatal primary brain tumor. The common therapy regimen consisting of surgery, radiotherapy, and chemotherapy with Temozolomid (TMZ) (1,2) lack long lasting effects. The prognosis is dismal with a 5-year survival probability of 6.8% (varying with age at diagnosis) (3,4) which barely improved for decades (5), and a median overall survival (OS) of about 15–18 months after diagnosis which still remains at this low level despite several efforts (14).

Diverse therapeutic options are subject of actual or recent clinical studies, among them targeted approaches consisting of small-molecule kinase inhibitors (68) or antibodies (7), immunotherapy (9) or oncolytic viruses (10). But on the whole, the effects on OS remain rather low. Reasons for therapy failure include development of resistances caused by genetic heterogeneity or redundant signaling cascades, toxicity and other undesired side-effects, and low abundant drug availability on the tumor site due to inhibition by the blood-brain-barrier (6,7).

Nevertheless, some patients show a prolonged OS up to several years.

To date, several different factors, clinical and molecular, are discussed to cause or correlate with long-term survival (LTS). The possibility of gross tumor resection, a high Karnofsky performance status and a young age at diagnosis are considered as clinical factors for prolonged survival (11), also the location and whether the subventrical zone is affected or not might have an influence for progression and survival (12). Furthermore, for several patients, eligibility for a second surgery after tumor recurrence, is an option associated with prolonged OS (13).

On the molecular level, some expression signatures, mutations, chromosomal aberrations and MGMT methylation status are deemed to be factors influencing therapy response and survival (1416), but no common profile indicating LTS is found for this highly heterogeneous tumor entity (17). In contrary, some biomarkers like MGMT methylation are controversially discussed for outcome prediction ability (18).

Although several studies describe potential molecular features in connection with survival or prognosis (14,15), studies analyzing microRNA (miR) profiles are quite rare and mostly not concordant, describing different sets of miRs to be differentially expressed and therefore considered as potential biomarkers for survival (1923).

MiRs are short non-coding RNAs of ~22 nt length which derive from long primary transcripts which are matured in a multistep process. They influence the transcription and translation of targeted genes and are often deregulated in any kind of cancer (24), also in glioblastoma (25). Depending on the tissue, kind of dysregulation and affected targets, they have oncogenic and/or tumor suppressive character (26). They are considered as putative targets for directed therapies consisting of miR-mimics for upregulating repressed miRs, or anti-miRs or miR-sponges to suppress overexpressed miRs (2729).

The aim of this study is to compare miR expression profiles of short-time and long-time survivors to determine, whether there are survival-associated signatures. miRs showing significant differential expression between the survival groups are deemed as candidates for further analysis regarding potential target genes and influence on tumor behavior. These findings could contribute to more knowledge and deeper insights on molecular features of this highly complex and heterogenous disease.

Identifying differential expressed miRs and their potential target genes can give clues to factors affecting OS, which, in turn, could be interesting candidates for developing specific therapeutic approaches.

Materials and methods

Patient samples

Formalin-fixed paraffin-embedded (FFPE) samples of GBM patients were retrieved from the archive of the Institute of Pathology of the University Medicine Rostock. Tissue was fixed in 4% neutral buffered formalin (Grimm) and incubated overnight at room temperature. Afterwards, it was embedded in molten paraffin (Merck).

Inclusion criteria were diagnosed GBM WHO Grade IV, sufficient availability of material and tumor content. For this study, the number of patient samples for array-based microRNA profile screening was limited, so from all potential available samples, initially 24 were arbitrarily selected to obtain a balanced distribution of sex and survival. One sample had to be removed afterwards due to issues of data quality and completeness, so a total of 23 patients were suitable for analysis.

Patient data (sex, age at diagnosis, OS, kind of surgery, therapeutic treatments) were obtained from the department of neurosurgery and patients were assigned to two groups: LTS and short-term survivor (STS) using the median OS of the cohort as discriminator. Molecular characteristics (EGFR amplification, mutation status of IDH1 and IDH2) were obtained by earlier projects and/or routine diagnostic procedures.

Specimen collection was conducted in accordance with the ethics guidelines for the use of human material, approved by the Ethics Committee of the University of Rostock (Reference no. A 2009/34) and with informed written consent from all patients prior to surgery.

MicroRNA extraction

For microRNA extraction from 10 µm FFPE sections, the miRNeasy FFPE-Kit (Qiagen) was used following the manufacturer's protocol. Concentration of extracted RNA was determined with a Nanodrop spectrometer (Peqlab).

miR screening arrays

Analysis of microRNA expression was performed using the Nanostring nCounter System with the Human v3 miRNA assay (Nanostring). The analysis procedure was performed with 250 ng per sample by a service provider lab (Transcriptome and Genome Analysis Laboratory (TAL), Microarray and Deep-Sequencing Facility, University Medicine Göttingen) and analyzed by the authors with the nSolver 4.0 software (Nanostring) using standard settings for background subtraction and housekeeping genes-based normalization. Group-wise comparison (LTS vs. STS) was performed, delivering fold change and p-values. Results were visualized as boxplots generated by BoxPlotR (http://shiny.chemgrid.org/boxplotr/).

Determination of miR-targets

For determination of potential target genes of miR-130b-3p, miR-146b-5p and miR-148a-3p, targeted literature research was performed looking up PubMed (https://pubmed.ncbi.nlm.nih.gov) for corresponding papers using search strings including the miR-name and ‘glioblastoma’. Furthermore, the online microRNA-target interaction database miRTarBase (http://mirtarbase.cuhk.edu.cn) was consulted for predicted potential targets of these three miRs.

Immunohistochemistry

Protein expression analysis for PTEN and TRAF6 was performed by immunohistochemistry using 2 µm thick FFPE sections on coated glass slides (Dako). For deparaffinization, rehydration and antigen demasking, slides were incubated for 20 min at 97°C and pH 9 in EnVision FLEX Target Retrieval Solution, high pH (pH 9) (Dako).

For slide processing, an automatic IHC system, AutostainerLink48 (Dako) was used, according to the following (routine) protocol. All steps were performed at room temperature. Slides were rinsed with EnVision FLEX Wash Buffer (Dako). To reduce unspecific background staining, slides were incubated for 5 min with 100 µl of EnVision FLEX Peroxidase-Blocking Reagent, readty-to-use (Dako), a phosphate buffer containing H2O2, 15 mMol NaN3 and detergent, and rinsed afterwards with wash buffer. For PTEN, a monoclonal (clone 6H2.1) mouse-anti human PTEN antibody (Dako, cat. no. M3627) was used, dilution 1:100 in EnVision FLEX Antibody Diluent (Dako). For TRAF6, a monoclonal (clone EP592Y) rabbit-anti human TRAF6 antibody (Abcam; cat. no. ab40675), dilution 1:50, was used. Slides were incubated with 100 µl of first antibodies for 20 min. and rinsed afterwards with wash buffer. For TRAF6, a signal enhancement step was inserted incubating slides for 15 min with 100 µl of EnVision FLEX+ Rabbit (LINKER), ready-to-use (Dako, cat. no. SM805). As secondary reagent, EnVision FLEX/HRP, ready-to-use (Dako, cat. no. SM802), was used, containing dextran coupled with peroxidase molecules and goat secondary antibodies against rabbit and mouse immunoglobulins. Slides were incubated with 100 µl thereof for 20 min and rinsed afterwards with wash buffer. As substrate working solution 1 drop EnVision FLEX DAB+ Chromogen (Dako, cat. no. DM827) was mixed with 1 ml EnVision FLEX Substrate Buffer (Dako, cat. no. SM803). 200 µl thereof were applied to each slide. Slides were incubated for 10 min and rinsed with wash buffer afterwards. Finally, for counter staining, 100 µl of EnVision FLEX Hematoxylin (Dako, cat. no. SM806) were applied and slides were incubated for 5 min. Slides were rinsed with deionized water, washed for 5 min with wash buffer and rinsed once more with water. Slides were covered after dehydration with ethanol and xylol with coverslips (epredia) and CV mount (Leica) as mounting medium using the automated Coverslipper (Dako).

Stained slides were scanned using a Pannoramic Desk DWII slide scanner (3DHistech) with Pannoramic Scanner 2.2.0 software (3DHistech). Visualization and analysis were performed using CaseViewer 2.4 software (3DHistech). For presentation, representative regions the magnification was set to 10×, and the chosen sections were exported as JPEG.

Statistical analyses

Significance of differential miR expression was determined using the nSolver Software 4.0 (Nanostring) applying a paired, two-tailed Student's t-test. Statistical analyses concerning OS and clinical data were performed using SPSS Statistics version 28 (IBM) using the Kaplan-Meier method. Significance was assumed for P≤0.05.

Results

Patients' characteristics

Samples of 23 GBM patients were finally included in this study (Table I). Median age at diagnosis was 59.96 years (47.36–80.82 years). Median OS was 380 days (30–2041 days). Patients with OS >median OS were classified as LTS, the others as STS. 11 patients were male, 12 were female. All but one were wild type for IDH1 R132 and 12 carried an EGFR amplification. Sex and EGFR amplification status were evenly distributed over both survival groups. 19 of 23 patients underwent gross-total resection and 4 had a subtotal resection. 22 patients received radiotherapy, for one patient it is unknown. 15 patients were treated with TMZ, 7 received no TMZ, for one patient it is unknown. Of the patients without TMZ therapy, five were grouped into the STS group and 2 into LTS. Regarding the clinical data, only for the type of surgery, a significant influence on OS could be observed (P=0.001). No significant differences in OS were seen for age at diagnosis (P=0.566), sex (P=0.927), EGFR status (P=0.437) and TMZ treatment (P=0.158) (Fig. S1, Table II).

Table I.

Characteristics of analyzed patients.

Table I.

Characteristics of analyzed patients.

Patient no.Age at diagnosis (y)OS (d)SexIDH1 statusEGFR statusSurgeryRtxTMZSurvival status
167.36528MWtAmpGross-total resectionYesYesLTS
247.36143FWtAmpSubtotalYesYesSTS
347.72427MWtAmpGross-total resectionYesYesLTS
473.00297FWtAmpGross-total resectionYesNoSTS
559.96727MWtAmpGross-total resectionYesYesLTS
669.63380FWtAmpGross-total resectionYesYesSTS
750.052041MWtAmpGross-total resectionYesYesLTS
864.47148MWtAmpGross-total resectionYesYesSTS
976.46883FWtAmpGross-total resectionYesNoLTS
1069.35148FWtAmpGross-total resectionYesYesSTS
1153.22236MWtAmpSubtotalN/AN/ASTS
1257.18687FWtNormGross-total resectionYesYesLTS
1359.75357FWtNormGross-total resectionYesYesSTS
1457.46586FWtNormGross-total resectionYesYesLTS
1549.17253MWtNormGross-total resectionYesYesSTS
1677.23777FWtNormGross-total resectionYesYesLTS
1753.63129MWtNormGross-total resectionYesNoSTS
1880.82489FWtNormGross-total resectionYesYesLTS
1949.62120FWtNormGross-total resectionYesNoSTS
2066.2955MWtNormSubtotalYesNoSTS
2171.0930MR132HNormSubtotalYesNoSTS
2270.50397FWtAmpGross-total resectionYesNoLTS
2356.74871MWtNormGross-total resectionYesYesLTS

[i] y, years; OS, overall survival; d, days; wt, wild type; R132H, IDH1 R132H mutation; norm, normal copy number; amp, amplification; STS, short-term survivor; LTS, long-term survivor; Rtx, radiotherapy.

Table II.

Overview of clinical parameters and their influence on overall survival.

Table II.

Overview of clinical parameters and their influence on overall survival.

Clinical parameterVariables, n (%)Influence on OS (P-value)
Age at diagnosis, years 0.566
  <6011 (47.83%)
  >6012 (52.17%)
Median age, years59.96
Age range, years47.36-80.82
Sex 0.927
  Female12 (52.17%)
  Male11 (47.83%)
EGFR status 0.437
  EGRF normal11 (52.17%)
  EGFR amplified12 (47.83%)
IDH1/2 status ND
  Wild-type22 (95.65%)
  Mutated1 (4.35%)
Tmz 0.158
  Yes15 (65.2%)
  No7 (30.4 %)
  Unknown:1 (4.35%)
Radiotherapy ND
  Yes22 (95.65%)
  Unknown1 (4.35%)
Type of surgery 0.001
  Gross-total19 (82.6%)
  Subtotal4 (17.4%)

[i] OS, overall survival; ND, not determined; TMZ, temozolomid; IDH1/2 status, mutation status of IDH1 and IDH2 genes.

miR expression profiling

The miR-expression profiling by the Nanostring nCounter software revealed on the whole just moderate changes in miR expression comparing profiles of LTS and STS (Table SI) for most of the miRs analyzed. But some miRs outperformed the moderate fold change values, rendering them as potential candidates as significant targets. Finally, three miRs were choosen for further analysis: miR-130b-3p, downregulated in LTS, miR-146b-5p and miR-148a-5p, both upregulated in LTS (Table III, Fig. 1). All three of them fulfilled the requirements for being further considered: a top fold change value with high significance, and robust absolute expression values (normalized counts) in both groups. MiRs showing high, significant fold change values, but only very low counts, weren't considered further.

Table III.

Top 10 significantly differentially expressed miRs showing fold-change values of the LTS group (‘true’) compared with the STS group (‘false’).

Table III.

Top 10 significantly differentially expressed miRs showing fold-change values of the LTS group (‘true’) compared with the STS group (‘false’).

Probe nameAccession no.Fold-change LTS vs. STSP-value
hsa-miR-146b-5pMIMAT00028092.530.002
hsa-miR-130b-3pMIMAT0000691−2.720.007
hsa-miR-148a-3pMIMAT00002433.200.007
hsa-miR-302b-3pMIMAT00007151.780.011
hsa-miR-3065-5pMIMAT00150663.900.012
hsa-miR-301b-3pMIMAT0004958−3.170.012
hsa-miR-887-3pMIMAT00049512.240.013
hsa-miR-23a-3pMIMAT00000781.940.017
hsa-miR-21-5pMIMAT00000761.790.019
hsa-miR-142-3pMIMAT00004342.020.019

[i] Fold-change data of all miRs analyzed are presented in Table SI. STS, short-term survivor; LTS, long-term survivor; miR, microRNA.

Although there are overlaps of expression values between the two survival groups (Fig. 1), a kind of signature can be seen: low miR-130b-3p expression and high expression of miR-146b-5p and miR-148a-3p for LTS; and vice versa for STS. High and low expression states are separated by the median expression values. Correlating survival and expression status for each case shows that 11/23 cases (5 LTS and 6 STS) completely correspond to this signature and another 6 cases (4 LTS and 2 STS) in 2 of 3 miRs. Another 6 cases (2 LTS and 4 STS) show just one miR but no case shows none (Fig. 2).

Determination of potential target genes

Targeted PubMed search and specific investigation of miRTarBase revealed several genes proposed to be potential targets of miRs 130b-3p, 146b-5p and 148a-3p (Table IV). These two following were chosen to start further analyses with: PTEN as target of miR-130b-3p and important TSG in GBM, and TRAF6 as target of miR-146b-5b, an invasion promoting factor. For miR-148a-3p the situation is more complex, as it is described to function either as oncogene or as TSG in GBM, so due to its ambivalent role, target studies were delayed.

Table IV.

Literature and database predicted target genes for miR-130b-3p, miR-146b-5p and miR 148a-3p.

Table IV.

Literature and database predicted target genes for miR-130b-3p, miR-146b-5p and miR 148a-3p.

miRLiteraturemiRTarBase
miR-130b-3pPTEN (31); PPARγ (30); MST, SAV1 (32)RUNX3, ZEB1, IGF1, TP53INP1
miR-146b-5pTRAF6 (40,41); MMP16 (37); EGFR (34)TRAF6, IRAK1, MMP16, KIT, EGFR
miR-148a-3pITGA9 (49); DLGAP1 (46); MIG, BIM (52)DNMT3B, DNMT1, IKBKB, MMP7

[i] miR, microRNA.

IHC analysis of PTEN and TRAF6

Immunohistochemistry of PTEN and TRAF6 was successful in 22 of 23 cases. Although IHC is not considered as a quantitative method, it is observable, that a partial loss or decrease of PTEN expression occurs in some cases, mostly in samples with a higher miR-130b expression (Fig. 3), especially samples #11, #10 and #1.

Similarly, for TRAF6, a reduction or partial loss in expression is observable predominantly in samples with higher miR-146b expression (Fig. 4), especially samples #4, #1 and #5.

In summary, PTEN and TRAF6 could be considered as putative targets of miR-130b and miR-146b, respectively, at least in cases with higher miR expression levels.

Discussion

This study shows a significant differential miR expression profile comparing LTS and STS, highlighting miR-130b-3p, miR-146b-5p and miR-148a-3p as most significantly differentially expressed miRs. As the definition of LTS is non-uniform throughout the literature, in our study, the median OS of the cohort (380 days) was used as delimiter to split the cohort in two more or less even groups. These data give the indication, that miR-130b-3p acts as oncomir, while miR-146b-5p and miR-148a-3p have tumor suppressor properties.

miR-130b is shown to be overexpressed in glioblastoma in contrast to non-neoplastic brain tissue, as well as in other tumor entities, and promotes cell proliferation by inhibiting PPARγ, leading to decreased E-cadherin levels (30). Furthermore, it was shown, that suppression of miR-130b inhibits GBM cell proliferation and invasion and induces apoptosis via the PTEN/AKT signaling pathway. In this context, PTEN was shown to be a direct target of miR-130b (31). Another study shows more oncogenic properties of miR-130b, demonstrating that its upregulation enhances a stem cell-like phenotype by inactivating the Hippo signaling pathway with MST1/2 and SAV1 as direct targets (32). Earlier, miR-130b expression was associated with progression from lower to high grade glioma (33). Together with these, our data, showing a higher expression in STS samples, underpin the oncogenic character of miR-130b.

miR-146b-5p is linked with tumor suppressive properties. It is shown to target EGFR and hence, at least in vitro, reduces invasion and migration of glioma cells (34). In the cohort analyzed in our study, immunohistochemistry showed, that the EGFR expression was only dependent on the EGFR amplification status, but not on the survival group (data not shown). But for in vitro models it is shown that EGFR amplification is often lost during cultivation (35), except for special cell culture conditions (36). Another target deemed to be regulated by miR-146b-5p is MMP16, whose inhibition leads to decreased migration and invasion (37,38). MMP16 is a member of matrix metalloproteases, important factors for migration and metastasis as they contribute to proteolysis of the extracellular matrix, enabling tumor cell migration (39). TRAF6 is another direct target of miR-146b-5p whose repression inhibits proliferation and progression, while promoting apoptosis. It is associated with a better prognosis (40). Furthermore, miR-146b-5p mediated TRAF6 repression is considered to suppress TMZ resistance (41). TRAF6 is correlated with a worse prognosis and oncogenic properties promoting invasion by upregulating another matrix-metalloprotease, MMP9 (42). In context with our data these studies underpin the correlation of miR-146-5p, TRAF6 expression and survival. Furthermore, miR-146b-5p inhibits GSCs and radioresistance by targeting SMARCA5 (43) and the β-catenin pathway (44). Although most studies attest miR-146b-5p tumor suppressive properties, some oncogenic potential cannot be totally excluded, as its upregulation is also associated with GBM recurrence (45). But the data acquired in our own study are rather concordant with the described tumor suppressive properties.

For miR-148a, its role in GBM seems to be ambivalent, according to the current literature. Several studies nominate different targets whose repression depict the oncogenic character of miR-148a, where others describe tumor suppressive properties. Repression of DLGAP1 leads to loss of cell polarity, therefore promoting growth, migration, invasion and EMT (46). Overexpression of miR-148a deemed to be induced by NFκB and targeting QKI and SKP1 activates TGFβ signaling and is associated with progression and augmented tumor aggressiveness (47). In a TCGA based study, it is shown that miR-148a targets FIH1, an HIF1 inhibitor and therefore promotes HIF1a and NOTCH1 signaling, enhancing vascularization, growth and survival (48). But also the opposite effect on vascularization is described, as miR-148a has shown to have tumor suppressive properties targeting ITGA9 (49), a cell adhesion factor involved in NOTCH1 controlled vascularization, leading to decreased angiogenesis (50). More oncogenic properties are demonstrated with targets like GADD45A, whose repression stimulates β-catenin and MMP9, promoting invasion, migration and stemness (51). Also positively influencing EGFR activity by targeting MIG2 and inhibiting apoptosis by targeting BIM is described (52). Even exosomal delivery of miR-148a seems to have a positive effect to proliferation and progression by targeting CADM1, leading to STAT3 activation (53). On the other side, more tumor suppressive properties are known. miR-148a targets ROCK-1, a factor promoting migration in several tumors (54). Even therapeutic effects are described, as in vivo experiments in mice show a prolonged survival when miR-148a is co-delivered with miR-296-5p by nanoparticles, targeting OCT4 and SOX2, reducing stemness (29). Although studies describing oncogenic properties of miR-148a in GBM predominate, our own data show, that it is higher expressed in LTS, rendering it more to the tumor suppressive side.

Considering the exemplarily selected targets PTEN and TRAF6, the impression is not that clear. The expression of these targets was assessed by IHC. This method is not considered as quantitative and not sensitive enough to identify minor changes in expression which is characteristic for miR-mediated regulation. Also, there are much more factors influencing genes' translational activity. But the data show, that at least at the upper ranges of miR expression, a decrease in target gene expression is observable, but there's no linearity.

To elucidate the downstream effects of differential miR expression, identify the real target genes in these tumors, and observe the effects on tumor cell behavior, functional approaches with GBM in vitro models using transfection of miR mimics or antagomiRs, followed by analyses on different levels are needed.

This study is limited by a quite low number of patients analyzed and a small selection of potential target genes. To improve the value of robustness of the data, a consecutive study based on the present data should include a higher number of patients. Furthermore, next to functional studies, the selection of potential target genes should be extended to elucidate more details of the molecular mechanisms of tumor behavior underlying the outcome. Also, the study is carried out on FFPE material, which offers lesser quality and opportunities for deeper analysis. Fresh material would serve as a source better quality nucleic acids and proteins for deeper molecular analysis, and would even offer the opportunity to be taken in culture to provide cell lines or xenografts for in vitro or in vivo studies.

In summary, this study shows a significant correlation of differential miR-expression and survival status. It emphasizes the role of regulatory components/epigenetic factors, in this case microRNAs, within the complex interactions determining tumor behavior and outcome. These results hopefully help to clarify the so far widely unconcordant approaches to determine outcome predicting biomarkers. Although the regulatory impact to the few selected, potential miR target genes is rather low, the connection between miR expression and survival is significant. These data provide a good basis for further, functional studies.

Supplementary Material

Supporting Data
Supporting Data

Acknowledgements

The authors would like to thank the technicians of the Institute of Pathology (University Medicine Rostock, Rostock, Germany), Mrs. Kerstin Westphal, Mrs. Heike Clasen and Mrs. Beate Krause for performing immunohistochemistry, and Mrs. Susanne Höffer for the sections.

Funding

Funding: No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

BS designed the study, acquired, analyzed and interpreted data and wrote the manuscript. NL analyzed and interpreted immunohistochemistry data. AZ analyzed and interpreted immunohistochemistry data, contributed to the statistical analyses and critically edited the manuscript. CH acquired the patient data and substantially contributed to their analyses and interpretation, and critically edited the manuscript. AE provided patient material, substantially contributed to the concept of the study, and critically contributed to the manuscript. BS and AZ confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Ethics Committee of the University of Rostock (approval no. A2009/34). Informed consent was obtained from all subjects involved in the study.

Patient consent for publication

Not applicable

Competing interests

The authors declare that they have no competing interests.

Authors' information

Dr Björn Schneider, ORCID-ID: 0000-0002-0282-7330.

References

1 

Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJB, Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, et al: Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 10:459–466. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJB, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 352:987–996. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Wen PY, Weller M, Lee EQ, Alexander BM, Barnholtz-Sloan JS, Barthel FP, Batchelor TT, Bindra RS, Chang SM, Chiocca EA, et al: Glioblastoma in adults: A society for neuro-oncology (SNO) and european society of neuro-oncology (EANO) consensus review on current management and future directions. Neuro Oncol. 22:1073–1113. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Ostrom QT, Cioffi G, Gittleman H, Patil N, Waite K, Kruchko C and Barnholtz-Sloan JS: CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2012–2016. Neuro Oncol. 21 (Suppl 5):v1–v100. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Miller KD, Ostrom QT, Kruchko C, Patil N, Tihan T, Cioffi G, Fuchs HE, Waite KA, Jemal A, Siegel RL and Barnholtz-Sloan JS: Brain and other central nervous system tumor statistics, 2021. CA Cancer J Clin. 71:381–406. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Heffron TP: Challenges of developing small-molecule kinase inhibitors for brain tumors and the need for emphasis on free drug levels. Neuro Oncol. 20:307–312. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Le Rhun E, Preusser M, Roth P, Reardon DA, van den Bent M, Wen P, Reifenberger G and Weller M: Molecular targeted therapy of glioblastoma. Cancer Treat Rev. 80:1018962019. View Article : Google Scholar : PubMed/NCBI

8 

Perryman R, Renziehausen A, Shaye H, Kostagianni AD, Tsiailanis AD, Thorne T, Chatziathanasiadou MV, Sivolapenko GB, El Mubarak MA, Han GW, et al: Inhibition of the angiotensin II type 2 receptor AT2R is a novel therapeutic strategy for glioblastoma. Proc Natl Acad Sci USA. 119:e21162891192022. View Article : Google Scholar : PubMed/NCBI

9 

Cao TQ, Wainwright DA, Lee-Chang C, Miska J, Sonabend AM, Heimberger AB and Lukas RV: Next steps for immunotherapy in glioblastoma. Cancers (Basel). 14:40232022. View Article : Google Scholar : PubMed/NCBI

10 

Todo T, Ito H, Ino Y, Ohtsu H, Ota Y, Shibahara J and Tanaka M: Intratumoral oncolytic herpes virus G47∆ for residual or recurrent glioblastoma: A phase 2 trial. Nat Med. 28:1630–1639. 2022. View Article : Google Scholar : PubMed/NCBI

11 

Anselmo P, Maranzano E, Selimi A, Lupattelli M, Palumbo I, Bini V, Casale M, Trippa F, Bufi A, Arcidiacono F and Aristei C: Clinical characterization of glioblastoma patients living longer than 2 years: A retrospective analysis of two Italian institutions. Asia Pac J Clin Oncol. 17:273–279. 2020. View Article : Google Scholar : PubMed/NCBI

12 

Jiang H, Yu K, Li M, Cui Y, Ren X, Yang C, Zhao X and Lin S: Classification of progression patterns in glioblastoma: Analysis of predictive factors and clinical implications. Front Oncol. 10:5906482020. View Article : Google Scholar : PubMed/NCBI

13 

Pasqualetti F, Montemurro N, Desideri I, Loi M, Giannini N, Gadducci G, Malfatti G, Cantarella M, Gonnelli A, Montrone S, et al: Impact of recurrence pattern in patients undergoing a second surgery for recurrent glioblastoma. Acta Neurol Belg. 122:441–446. 2022. View Article : Google Scholar : PubMed/NCBI

14 

Jovčevska I: Genetic secrets of long-term glioblastoma survivors. Bosn J Basic Med Sci. 19:116–124. 2019.PubMed/NCBI

15 

Gately L, McLachlan SA, Philip J, Rathi V and Dowling A: Molecular profile of long-term survivors of glioblastoma: A scoping review of the literature. J Clin Neurosci. 68:1–8. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Lei CG, Jia XY and Sun WJ: Establish six-gene prognostic model for glioblastoma based on multi-omics data of TCGA database. Yi Chuan. 43:665–679. 2021.PubMed/NCBI

17 

Richardson TE, Kumar A, Xing C, Hatanpaa KJ and Walker JM: Overcoming the odds: Toward a molecular profile of long-term survival in glioblastoma. J Neuropathol Exp Neurol. 79:1031–1037. 2020. View Article : Google Scholar : PubMed/NCBI

18 

Butler M, Pongor L, Su YT, Xi L, Raffeld M, Quezado M, Trepel J, Aldape K, Pommier Y and Wu J: MGMT status as a clinical biomarker in glioblastoma. Trends Cancer. 6:380–391. 2020. View Article : Google Scholar : PubMed/NCBI

19 

Henriksen M, Johnsen KB, Andersen HH, Pilgaard L and Duroux M: MicroRNA expression signatures determine prognosis and survival in glioblastoma multiforme-a systematic overview. Mol Neurobiol. 50:896–913. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Zhang W, Zhang J, Yan W, You G, Bao Z, Li S, Kang C, Jiang C, You Y, Zhang Y, et al: Whole-genome microRNA expression profiling identifies a 5-microRNA signature as a prognostic biomarker in Chinese patients with primary glioblastoma multiforme. Cancer. 119:814–824. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Srinivasan S, Patric IRP and Somasundaram K: A ten-microRNA expression signature predicts survival in glioblastoma. PLoS One. 6:e174382011. View Article : Google Scholar : PubMed/NCBI

22 

Niyazi M, Zehentmayr F, Niemöller OM, Eigenbrod S, Kretzschmar H, Schulze-Osthoff K, Tonn JC, Atkinson M, Mörtl S and Belka C: MiRNA expression patterns predict survival in glioblastoma. Radiat Oncol. 6:1532011. View Article : Google Scholar : PubMed/NCBI

23 

Areeb Z, Stylli SS, Koldej R, Ritchie DS, Siegal T, Morokoff AP, Kaye AH and Luwor RB: MicroRNA as potential biomarkers in Glioblastoma. J Neurooncol. 125:237–248. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Di Leva G, Garofalo M and Croce CM: MicroRNAs in cancer. Annu Rev Pathol. 9:287–314. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Rolle K: miRNA Multiplayers in glioma. From bench to bedside. Acta Biochim Pol. 62:353–365. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Svoronos AA, Engelman DM and Slack FJ: OncomiR or tumor suppressor? The duplicity of microRNAs in cancer. Cancer Res. 76:3666–3670. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Chen L and Kang C: miRNA interventions serve as ‘magic bullets’ in the reversal of glioblastoma hallmarks. Oncotarget. 6:38628–38642. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Ananta JS, Paulmurugan R and Massoud TF: Tailored nanoparticle codelivery of antimiR-21 and antimiR-10b augments Glioblastoma Cell Kill by Temozolomide: Toward a ‘personalized’ anti-microRNA therapy. Mol Pharm. 13:3164–3175. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Lopez-Bertoni H, Kozielski KL, Rui Y, Lal B, Vaughan H, Wilson DR, Mihelson N, Eberhart CG, Laterra J and Green JJ: Bioreducible polymeric nanoparticles containing multiplexed cancer stem cell regulating miRNAs inhibit Glioblastoma growth and prolong survival. Nano Lett. 18:4086–4094. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Gu JJ, Zhang JH, Chen HJ and Wang SS: MicroRNA-130b promotes cell proliferation and invasion by inhibiting peroxisome proliferator-activated receptor-γ in human glioma cells. Int J Mol Med. 37:1587–1593. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Gu JJ, Fan KC, Zhang JH, Chen HJ and Wang SS: Suppression of microRNA-130b inhibits glioma cell proliferation and invasion, and induces apoptosis by PTEN/AKT signaling. Int J Mol Med. 41:284–292. 2018.PubMed/NCBI

32 

Zhu G, Wang Y, Mijiti M, Wang Z, Wu PF and Jiafu D: Upregulation of miR-130b enhances stem cell-like phenotype in glioblastoma by inactivating the Hippo signaling pathway. Biochem Biophys Res Commun. 465:194–199. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Malzkorn B, Wolter M, Liesenberg F, Grzendowski M, Stühler K, Meyer HE and Reifenberger G: Identification and functional characterization of microRNAs involved in the malignant progression of gliomas. Brain Pathol. 20:539–550. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Katakowski M, Zheng X, Jiang F, Rogers T, Szalad A and Chopp M: MiR-146b-5p suppresses EGFR expression and reduces in vitro migration and invasion of glioma. Cancer Invest. 28:1024–1030. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Pandita A, Aldape KD, Zadeh G, Guha A and James CD: Contrasting in vivo and in vitro fates of glioblastoma cell subpopulations with amplified EGFR. Genes Chromosomes Cancer. 39:29–36. 2004. View Article : Google Scholar : PubMed/NCBI

36 

William D, Mokri P, Lamp N, Linnebacher M, Classen CF, Erbersdobler A and Schneider B: Amplification of the EGFR gene can be maintained and modulated by variation of EGF concentrations in in vitro models of glioblastoma multiforme. PLoS One. 12:e01852082017. View Article : Google Scholar : PubMed/NCBI

37 

Li Y, Wang Y, Yu L, Sun C, Cheng D, Yu S, Wang Q, Yan Y, Kang C, Jin S, et al: miR-146b-5p inhibits glioma migration and invasion by targeting MMP16. Cancer Lett. 339:260–269. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Le Zhang, Wang J, Fu Z, Ai Y, Li Y and Wang Y and Wang Y: Sevoflurane suppresses migration and invasion of glioma cells by regulating miR-146b-5p and MMP16. Artif Cells Nanomed Biotechnol. 47:3306–3314. 2019. View Article : Google Scholar : PubMed/NCBI

39 

Stetler-Stevenson WG, Aznavoorian S and Liotta LA: Tumor cell interactions with the extracellular matrix during invasion and metastasis. Annu Rev Cell Biol. 9:541–573. 1993. View Article : Google Scholar : PubMed/NCBI

40 

Liu J, Xu J, Li H, Sun C, Yu L, Li Y, Shi C, Zhou X, Bian X, Ping Y, et al: miR-146b-5p functions as a tumor suppressor by targeting TRAF6 and predicts the prognosis of human gliomas. Oncotarget. 6:29129–29142. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Qian Z, Zhou S, Zhou Z, Yang X, Que S, Lan J, Qiu Y and Lin Y: miR-146b-5p suppresses glioblastoma cell resistance to temozolomide through targeting TRAF6. Oncol Rep. 38:2941–2950. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Sun J, Zhao B, Du K and Liu P: TRAF6 correlated to invasion and poor prognosis of glioblastoma via elevating MMP9 expression. Neuroreport. 30:127–133. 2019. View Article : Google Scholar : PubMed/NCBI

43 

Wang H, Tan L, Dong X, Liu L, Jiang Q, Li H, Shi J, Yang X, Dai X, Qian Z and Dong J: MiR-146b-5p suppresses the malignancy of GSC/MSC fusion cells by targeting SMARCA5. Aging. 12:13647–13667. 2020. View Article : Google Scholar : PubMed/NCBI

44 

Yang W, Yu H, Shen Y, Liu Y, Yang Z and Sun T: MiR-146b-5p overexpression attenuates stemness and radioresistance of glioma stem cells by targeting HuR/lincRNA-p21/β-catenin pathway. Oncotarget. 7:41505–41526. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Khwaja SS, Cai C, Badiyan SN, Wang X and Huang J: The immune-related microRNA miR-146b is upregulated in glioblastoma recurrence. Oncotarget. 9:29036–29046. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Li Y, Li W, Zeng X, Tang X, Zhang S, Zhong F, Peng X, Zhong Y, Rosol TJ, Deng X, et al: The role of microRNA-148a and downstream DLGAP1 on the molecular regulation and tumor progression on human glioblastoma. Oncogene. 38:7234–7248. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Wang H, Pan JQ, Luo L, Ning XJ, Ye ZP, Yu Z and Li WS: NF-κB induces miR-148a to sustain TGF-β/Smad signaling activation in glioblastoma. Mol Cancer. 14:22015. View Article : Google Scholar : PubMed/NCBI

48 

Wong HA, Fatimy RE, Onodera C, Wei Z, Yi M, Mohan A, Gowrisankaran S, Karmali P, Marcusson E, Wakimoto H, et al: The cancer genome atlas analysis predicts microRNA for targeting cancer growth and vascularization in Glioblastoma. Mol Ther. 23:1234–1247. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Xu TJ, Qiu P, Zhang YB, Yu SY, Xu GM and Yang W: MiR-148a inhibits the proliferation and migration of glioblastoma by targeting ITGA9. Hum Cell. 32:548–556. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Guichet P-O, Guelfi S, Teigell M, Hoppe L, Bakalara N, Bauchet L, Duffau H, Lamszus K, Rothhut B and Hugnot JP: Notch1 stimulation induces a vascularization switch with pericyte-like cell differentiation of glioblastoma stem cells. Stem Cells. 33:21–34. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Cui D, Sajan P, Shi J, Shen Y, Wang K, Deng X, Zhou L, Hu P and Gao L: MiR-148a increases glioma cell migration and invasion by downregulating GADD45A in human gliomas with IDH1 R132H mutations. Oncotarget. 8:25345–25361. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Kim J, Zhang Y, Skalski M, Hayes J, Kefas B, Schiff D, Purow B, Parsons S, Lawler S and Abounader R: microRNA-148a is a prognostic oncomiR that targets MIG6 and BIM to regulate EGFR and apoptosis in glioblastoma. Cancer Res. 74:1541–1553. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Cai Q, Zhu A and Gong L: Exosomes of glioma cells deliver miR-148a to promote proliferation and metastasis of glioblastoma via targeting CADM1. Bull Cancer. 105:643–651. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Fang Z, Weng Y, Xiao F and Yu J: LncRNA RP11-390F4.3 inhibits invasion and migration of glioblastoma cells by downregulating ROCK1. Neuroreport. 32:888–893. 2021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2023
Volume 27 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Schneider B, Lamp N, Zimpfer A, Henker C and Erbersdobler A: Comparing tumor microRNA profiles of patients with long‑ and short‑term‑surviving glioblastoma. Mol Med Rep 27: 8, 2023
APA
Schneider, B., Lamp, N., Zimpfer, A., Henker, C., & Erbersdobler, A. (2023). Comparing tumor microRNA profiles of patients with long‑ and short‑term‑surviving glioblastoma. Molecular Medicine Reports, 27, 8. https://doi.org/10.3892/mmr.2022.12895
MLA
Schneider, B., Lamp, N., Zimpfer, A., Henker, C., Erbersdobler, A."Comparing tumor microRNA profiles of patients with long‑ and short‑term‑surviving glioblastoma". Molecular Medicine Reports 27.1 (2023): 8.
Chicago
Schneider, B., Lamp, N., Zimpfer, A., Henker, C., Erbersdobler, A."Comparing tumor microRNA profiles of patients with long‑ and short‑term‑surviving glioblastoma". Molecular Medicine Reports 27, no. 1 (2023): 8. https://doi.org/10.3892/mmr.2022.12895