MicroRNA-142-3p is involved in regulation of MGMT expression in glioblastoma cells

Background Glioblastoma multiforme (GBM) is the most malignant brain tumor, and there is no effective treatment strategy. Patients with GBM have a median overall survival of only 14.6 months. Current treatment consists of safe and maximal surgical excision, followed by concurrent chemoradiotherapy and maintenance chemotherapy. There are several obstacles that hinder the effectiveness of this aggressive treatment. Temozolomide (TMZ) is an oral alkylating drug that acts through alkylating the O6 position of guanine in DNA that leads to cell death. However, the expression and enzymatic activity of the DNA repair protein MGMT limits the therapeutic benefit from treatment with TMZ. MGMT reduces the efficacy of alkylating drugs by removing the methyl or alkyl group from damaged O6-methylguanine. Expression levels of MGMT play an important role in the outcome of GBM patients. miRNAs are a group of small regulatory RNAs that control target gene expression by binding to mRNAs. miR-142-3p has been found to be an important factor in the development and maintenance of the oncogenic state. Results In this study, we sought to investigate whether miR-142-3p can regulate MGMT gene expression in GBM cells. Here, we show that miR-142-3p downregulates MGMT expression through binding to the 3′-UTR of MGMT mRNA, thus affecting protein translation. Responsiveness to TMZ was significantly enhanced after transfection with miR-142-3p. Overexpression of miR-142-3p also sensitized GBM cells to alkylating drugs. Conclusion Above all, our findings demonstrate that miR-142-3p plays a critical role in regulating MGMT expression, has great potential for future clinical applications, and acts as a new diagnostic marker for this intractable disease.

[1]  Sagar Agarwal,et al.  Delivery of molecularly targeted therapy to malignant glioma, a disease of the whole brain , 2011, Expert Reviews in Molecular Medicine.

[2]  C. Burge,et al.  Conserved Seed Pairing, Often Flanked by Adenosines, Indicates that Thousands of Human Genes are MicroRNA Targets , 2005, Cell.

[3]  C. Croce,et al.  MicroRNA gene expression deregulation in human breast cancer. , 2005, Cancer research.

[4]  Jun Zhao,et al.  MiR‐142‐3p represses TGF‐β‐induced growth inhibition through repression of TGFβR1 in non‐small cell lung cancer , 2014, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[5]  I. Ferrer,et al.  Prognostic Significance of O6-Methylguanine-DNA Methyltransferase Determined by Promoter Hypermethylation and Immunohistochemical Expression in Anaplastic Gliomas , 2005, Clinical Cancer Research.

[6]  S. Bellusci,et al.  MicroRNA‐142 is a multifaceted regulator in organogenesis, homeostasis, and disease , 2017, Developmental dynamics : an official publication of the American Association of Anatomists.

[7]  J. Sarkaria,et al.  A genome-wide miRNA screen revealed miR-603 as a MGMT-regulating miRNA in glioblastomas , 2014, Oncotarget.

[8]  Guang-Yuh Chiou,et al.  Epigenetic regulation of the miR142-3p/interleukin-6 circuit in glioblastoma. , 2013, Molecular cell.

[9]  F. Kisseljov MicroRNAs and cancer , 2014, Molecular Biology.

[10]  A. Straube,et al.  Glioblastoma-dependent differentiation and angiogenic potential of human mesenchymal stem cells in vitro , 2011, Journal of Neuro-Oncology.

[11]  K. Kosik,et al.  MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. , 2005, Cancer research.

[12]  F. Slack,et al.  Oncomirs — microRNAs with a role in cancer , 2006, Nature Reviews Cancer.

[13]  Jennie W. Taylor,et al.  Treatment Considerations for MGMT-Unmethylated Glioblastoma , 2014, Current Neurology and Neuroscience Reports.

[14]  G. Reifenberger,et al.  MGMT Promoter Methylation Is a Strong Prognostic Biomarker for Benefit from Dose-Intensified Temozolomide Rechallenge in Progressive Glioblastoma: The DIRECTOR Trial , 2015, Clinical Cancer Research.

[15]  J. Menten,et al.  MGMT promoter hypermethylation correlates with a survival benefit from temozolomide in patients with recurrent anaplastic astrocytoma but not glioblastoma. , 2009, European journal of cancer.

[16]  Y. Katayama,et al.  Gene expression profiling predicts response to temozolomide in malignant gliomas. , 2010, International journal of oncology.

[17]  R. Stupp,et al.  For Personal Use. Only Reproduce with Permission from the Lancet Publishing Group. Review Temozolomide for Brain Tumours Current and Future Developments in the Use of Temozolomide for the Treatment of Brain Tumours , 2022 .

[18]  K. Gunsalus,et al.  A non-canonical site reveals the cooperative mechanisms of microRNA-mediated silencing , 2017, Nucleic acids research.

[19]  Keiji Suzuki,et al.  miR-195, miR-455-3p and miR-10a( *) are implicated in acquired temozolomide resistance in glioblastoma multiforme cells. , 2010, Cancer letters.

[20]  Takayuki Murata,et al.  MicroRNA Inhibition of Translation Initiation in Vitro by Targeting the Cap-Binding Complex eIF4F , 2007, Science.

[21]  Deye Song,et al.  MALAT1 promotes osteosarcoma development by regulation of HMGB1 via miR-142–3p and miR-129–5p , 2017, Cell cycle.

[22]  H. Too,et al.  MicroRNA as potential modulators in chemoresistant high-grade gliomas , 2014, Journal of Clinical Neuroscience.

[23]  H. Friedman,et al.  Temozolomide and treatment of malignant glioma. , 2000, Clinical cancer research : an official journal of the American Association for Cancer Research.

[24]  Y. Akao,et al.  MicroRNAs 143 and 145 are possible common onco-microRNAs in human cancers. , 2006, Oncology reports.

[25]  M. Hegi,et al.  Glioma epigenetics: From subclassification to novel treatment options. , 2017, Seminars in cancer biology.

[26]  Juyoung Ham,et al.  MicroRNA Target Recognition: Insights from Transcriptome-Wide Non-Canonical Interactions , 2016, Molecules and cells.

[27]  M. Kiebler,et al.  Faculty Opinions recommendation of Argonaute HITS-CLIP decodes microRNA-mRNA interaction maps. , 2009 .

[28]  K. Srivenugopal,et al.  Ubiquitination-dependent proteolysis of O6-methylguanine-DNA methyltransferase in human and murine tumor cells following inactivation with O6-benzylguanine or 1,3-bis(2-chloroethyl)-1-nitrosourea. , 1996, Biochemistry.

[29]  P. Xiao,et al.  MiR-142-3p functions as a potential tumor suppressor directly targeting HMGB1 in non-small-cell lung carcinoma. , 2015, International journal of clinical and experimental pathology.

[30]  B. Tannous,et al.  miR-101 is down-regulated in glioblastoma resulting in EZH2-induced proliferation, migration, and angiogenesis , 2010, Oncotarget.

[31]  Yi-tao Jia,et al.  MiR-142-3p Functions as a Potential Tumor Suppressor in Human Osteosarcoma by Targeting HMGA1 , 2014, Cellular Physiology and Biochemistry.

[32]  Scar,et al.  Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. , 2000, The New England journal of medicine.

[33]  M. Ferracin,et al.  miRNA array screening reveals cooperative MGMT-regulation between miR-181d-5p and miR-409-3p in glioblastoma , 2016, Oncotarget.

[34]  S. Shibata,et al.  Relationship between expression of O6-methylguanine-DNA methyltransferase, glutathione-S-transferase π in glioblastoma and the survival of the patients treated with nimustine hydrochloride: An immunohistochemical analysis , 2003, Neurological research.

[35]  Tracy T Batchelor,et al.  Glioblastoma recurrence after cediranib therapy in patients: lack of "rebound" revascularization as mode of escape. , 2011, Cancer research.

[36]  B. Bataille,et al.  Correlation of Clinical Features and Methylation Status of MGMT Gene Promoter in Glioblastomas , 2004, Journal of Neuro-Oncology.

[37]  Yiheng Zhou,et al.  MGMT promoter methylation and glioblastoma prognosis: a systematic review and meta-analysis. , 2013, Archives of medical research.

[38]  Y. Miao,et al.  MicroRNA-142-3p inhibits cell proliferation and invasion of cervical cancer cells by targeting FZD7 , 2015, Tumor Biology.

[39]  J. Qiao,et al.  MiR-142-3p Overexpression Increases Chemo-Sensitivity of NSCLC by Inhibiting HMGB1-Mediated Autophagy , 2017, Cellular Physiology and Biochemistry.

[40]  D. Rao,et al.  miRNA dysregulation in cancer: towards a mechanistic understanding , 2014, Front. Genet..

[41]  R. Mirimanoff,et al.  MGMT gene silencing and benefit from temozolomide in glioblastoma. , 2005, The New England journal of medicine.

[42]  L. Lim,et al.  Widespread siRNA "off-target" transcript silencing mediated by seed region sequence complementarity. , 2006, RNA.

[43]  R. Mirimanoff,et al.  Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. , 2005, The New England journal of medicine.

[44]  Min Liu,et al.  MicroRNA‐142‐3p, a new regulator of RAC1, suppresses the migration and invasion of hepatocellular carcinoma cells , 2011, FEBS letters.

[45]  L. Lim,et al.  MicroRNA targeting specificity in mammals: determinants beyond seed pairing. , 2007, Molecular cell.