Loading

Journal of Cancer Biology and Research

RAS Oncogene in Brain Tumors, Let-7 MicroRNA Involvement

Mini Review | Open Access | Volume 5 | Issue 1

  • 1. Department of Social, Human and Health Sciences, University of Cassino and Southern Lazio, Italy
  • 2. IRCCS Neuromed, Pozzilli, Italy
+ Show More - Show Less
Corresponding Authors
Samantha Messina, Department of Human Sciences, Society and Health, University of Cassino and Southern Lazio, Italy -Via S. Angelo (Campus Folcara) - 03043 Cassino (FR) – Italy, Tel: 39 0776 2993936
ABSTRACT

RAS oncogenes are master regulators of cancers. Somatic mutation in KRAS, HRAS and  NRAS genes account for approximately 30% of human cancers. KRAS is the most frequently  mutated isoform in RAS-driven cancers. Brain cancers are RAS-driven cancers despite have  no RAS mutation or amplification and its pivotal role in brain tumorigenesis has been well  documented. Indeed, it’s generally accepted that glioblastoma shows aberrant activation of  RAS/MAPK cascade due to mutations in upstream and downstream regulators. Since pioneering  study reporting that let-7 miRNA acted as tumor suppressor by repressing RAS oncogene,  growing evidence has suggested the importance of miRNA targeting the RAS-MAPK in brain  oncogenesis. Let-7 family members are direct and strong regulator of the RAS family. KRAS,  NRAS and HRAS mRNAs contain let-7 binding sites in 3’UTR sequences with clinical outcomes  in some cancer. Although the expression levels of let-7 miRNA family are not reduced in brain  tumors (with few exceptions), growing evidences show that let-7 miRNA inhibits the malignant   behavior - proliferation, migration and invasion - of glioma cells and glioma stem-like cells  as well as the tumor size in nude mice xenograft transplanted glioblastoma (GBM) via KRAS inhibition. More recently, genetic loss of let-7 is involved in neuroblastoma oncogenesis placing  let-7 disruption at the center of neuroblastoma pathogenesis. In addition, loss of let-7 increases  resistance to certain chemotherapeutic drugs and to radiation therapy in GBM. We aim this  review at summarizing and updating current knowledge on the contribution of let-7 miRNA  interplay with KRAS to oncogenesis of brain tumors

KEYWORDS

Glioblastoma ;RAS oncogenes ; let-7 microRNA ; Oncogenesi.

CITATION

Messina S, Frati L (2017) RAS Oncogene in Brain Tumors, Let-7 MicroRNA Involvement. J Cancer Biol Res 5(1): 1096

ABBREVIATIONS

miRNA: microRNA; let-7: lethal-7; GBM: Glioblastoma; 3’UTR: 3’ Untranslated Region; NSCL: Non-small Lung Cancer; ERK: Extracellular Signal-regulated Kinase; MAPK: Mitogenactivated Protein Kinase; HMGA2: High-mobility Group AThook 2; CSCs: Cancer Stem-like Cells; EGFR: Epidermal Growth Factor Receptor; NF1: Neurofibromin 1; PDGFR: Platelet-Derived Growth Factor; TMZ: Temozolomide; BBB: Blood Brain Barrier; ICV: Intracerebro-ventricular

INTRODUCTION

The observation that RAS is directly regulated by microRNAs (miRNAs) added a new facet to the regulation of RAS [1]. MicroRNAs (miRNAs) are a class of non-coding RNAs that function as endogenous triggers of the RNA interference pathway. Aberrant miRNA expression can contribute to tumorigenesis, but which of the many miRNA-target relationships are relevant to this process has been unclear. Lethal-7 (let-7) is a bona fide tumor suppressor gene, at least in lung and colon cancers.

Let-7 Targets Multiple Oncogenes (RAS, c-MYC, HMGA2, and so on) and the prominent (but not exclusive) mechanisms by which let-7 exerts a tumor suppressive role is by repressing the translation of the three RAS proteins (HRAS, NRAS, and KRAS) and c-MYC, a downstream effector of RAS-ERK[1-6]. There are also reports that suggest an oncogenic function for let-7 [7,8]. Thus, it is conceivable that let-7 may have pleiotropic cell-type specific biological effects depending on the milieu of expressed genes inside the cells.

The past decade has provided considerable insight into the critical regulatory roles that miRNAs exert over key cancer relevant signaling networks such as the RAS-ERK pathway. Our knowledge of how miRNAs can modulate RAS-ERK pathway activation continues to grow as potential miRNA-mRNA regulatory networks are identified using a variety of strategies. Three major paradigms of miRNA-mediated RAS-ERK regulation have emerged from these studies. miRNAs can impact the translation of (i) core RAS-ERK pathway components (e.g., let-7 targets HRAS, NRAS, and KRAS) [1], (ii) critical pathway regulatory proteins that are required for the proper spatio-temporal control of RASERK signaling [8-10] and (iii) upstream drivers and downstream effector/regulatory molecules [11,12]. Exist many examples of miRNAs that regulate RAS-ERK pathway activity in a variety of cancer [6]. Indeed, these miRNAs represent potential therapeutic substrates and targets that can be modulated in the treatment of cancer. In general, there is a very clear link between loss of let-7 expression and the development of poorly differentiated, aggressive cancers [13,14]. Studies analyzing in vitro and in vivo models of non-small cell lung cancer (NSCLC) show that let-7 expression is inversely correlated with the expression of KRAS, a critical promoter of NSCLC tumorigenesis. Let-7 abrogates tumor development and RAS-ERK signaling in an autochthonous model of NSCLC driven by activated KRAS (KRASG12D) [15,16]. Consistent with this previous result, a tumor suppressive role for let-7 was observed in a study analyzing a xenograft model of NSCLC [2] and increased expression of let-7a substantially reduces tumor burden in a KRAS murine lung cancer model [17]. Additionally, in a breast cancer context, let-7 antagonizes the maintenance, survival, and self-renewal of cancer stem-like cells (CSCs), and this suppressive activity was correlated with the reduced expression of RAS and HMGA2 [18]. Thus, by suppressing RAS expression, let-7 can attenuate RAF-MEK-ERK signaling and dependent oncogenic phenotypes regardless of the RAS-mutation status of cancers. These studies suggest that let-7 can act as both a cancer-preventative and cancer-therapeutic agent, and point to let-7 supplementation as a promising strategy to target RAS-ERK signaling in the treatment of cancers. In this review, we evaluate miRNA let-7 that hold great promise as potential therapeutic targets in the treatment of brain cancers by impacting RAS-ERK signaling.

Let-7 in brain tumors

Brain tumors encompass a wide spectrum of over 120 histologically, demographically, clinically and molecularly distinct diseases [19] (Table 1).

Table 1: WHO classification of tumors of the central nervous system [19] and summary of main studies reporting let-7 miRNA expression in brain tumors.

Grade

Examples

Criteria

Reference

WHO I

Pilocytic astrocytoma

Low proliferating, discrete non-invasive tumor

 

WHO II

Diffuse astrocytoma

Modest proliferating, partly invasive tumor

 

WHO III

Anaplastic astrocytoma

Anaplastic ependymoma

Fast proliferating invasive tumor

 

WHO IV

Embryonal tumors

Medulloblastoma

Neuroblastoma

 

 

Glioblastoma multiforme

 

Rapidly proliferating highly invasive tumor

Wang et al 2012; Turner et al 2010; Tannoet al 2016; Powers et al 2016; Song et al 2016; Wang et al 2013; Lee et al 2011; Wang et al 2016[20-27]

Glioblastoma is the most common malignant primary brain tumor in adults. Despite advances in understanding the molecular mechanisms underlying these tumors, current treatments are ineffective [28-32]. Therapeutic trials against driving genetic abnormalities amenable to therapeutic targeting such as the epidermal growth factor receptor EGFR, have been largely negative in most brain cancers [33]. The standard of care for most brain tumors remains focused on maximal surgical resection, radiotherapy and chemotherapy. Indirect targeting of the tumor through anti-angiogenics (for example, bevacizumab) and immunotherapies (vaccines, adoptive therapies, immune checkpoint inhibitors and oncolytic viruses) have demonstrated preclinical activity but mixed efficacy in clinical trials [34,35].

Let-7 family members are direct and strong regulator of the RAS family. K-RAS, N-RAS and H-RAS. mRNAs contain let-7 binding sites in 3’UTR sequences [36,37].There is a small but growing body of literature regarding the predictive utility of a Let7 microRNA-binding-site polymorphism in the 3′-untranslated region 3’UTR of KRAS (KRAS-LCS6) formany cancer outcome, although the results are conflicting [38-40]. MicroRNA detection has rapidly emerged as potential biomarkers, in patients with glioblastoma [41]. Based on the complete sequencing of the human genome as well as several high-throughput genomic technologies, the Cancer Genome Atlas (TCGA) has defined RAS/ MAPK as one of the main pathways involved in GBM [42] and the expression levels of let-7 miRNA family are not reduced in GBM [43]. Regardless its expression levels, let-7 miRNA is able to impair glioblastoma growth and cellular migration via RAS inhibition [25,44]. Specifically, the authors showed that forced expression of let-7 miRNA reduced expression of pan-RAS, N-RAS, and K-RAS reducing proliferation and migration as well as the tumor size in nude mice xenograft transplanted GBM [45]. Particularly, over expressed let-7a inhibited glioma cell malignancy by directly targeting KRAS [23] and, more recently, let-7b inhibits the malignant behavior (proliferation, migration and invasion) of glioma cells and glioma stem-like cells [24]. In other brain tumors, indeed, tumor suppressor microRNA let-7 is able to inhibit cell proliferation in human neuroblastoma by targeting MYC [45] and its repression by lin28 in an in vivo mouse model induces neuroblastoma development [46]. Indeed, focal deletion of let-7 family members was found in medulloblastoma (let-7a-2 and let-7e) [18] and the let-7 family was validated in spontaneous and radiation-induced medulloblastoma (MB) [22]. More recently, genetic loss of let-7 is involved in neuroblastoma oncogenesis placing let-7 disruption at the center of neuroblastoma pathogenesis [23]. Conversely, miR-let7g found up-regulated in anaplastic medulloblastoma (MB) and differentially expressed in desmoplastic medulloblastoma (MB) [21] although the functional consequences of their dysregulation were not still investigated. Moreover, human glioblastomas often develop resistance to radiation therapy and recently has been suggested that let-7 could exert a role in mediating such resistance in down- or up-regulation the relative expression level of let-7 family in irradiated human glioblastoma cells [47]. It is perplexing how let?7 affects oncogenesis, as the large influx of new miRNAs and other kinds of non-coding RNAs are continuously defined. Numerous oncogenes and signaling pathways were demonstrated to be targets of let-7 miRNAs besides RAS (as such MYC) and KRAS is target of many other miRNA in gliomagenesis [48,49].

RAS oncogenes in brain tumors

The biological functions of the RAS family (Harvey rat sarcoma viral oncogene homolog (HRAS), Kirsten rat sarcoma viral oncogene homolog (KRAS) and neuroblastoma RAS viral oncogene homolog (NRAS) have been extensively studied for decades. Though only 1% of the GBM tumors have a RAS mutation or amplification, 10% of GBM tumors contain neurofibromin 1 (NF1) inactivating genetic alterations that lead to hyperactive RAS activity by enhancing the intrinsic GTPase activity [42,50]. Three recent reports focused on miRNAs targeting RAS in GBM and showed that miR-143-3p directly targets NRAS [51], let-7a5p directly targets KRAS [25], and both NRAS and RRAS (related RAS viral oncogene homolog, HRAS homolog) are direct targets of miR-124-3p [52].

Deregulated RAS signaling is an important step in cancer initiation, with activating RAS mutations implicated in 30% of all cancers [53]. Unlike many human tumors, RAS mutation is not common in human glioma with some exceptions such as cerebellar GBM [54]. Hyperactive RAS signaling alone is sufficient to produce gliomas that closely resemble human tumors in glioma mouse models. Thus the RAS pathway is central in human gliomagenesis. Primary GBMs, and more recently relapsed neuroblastoma, are associated with disturbed RAS signaling and expression of oncogenic HRAS results in a malignant phenotype in glioma cell lines [55-58]. Particularly K-RAS oncogene is strongly involved in glioblastoma tumorigenesis [56,59-61] although KRAS mutations are nearly absent in malignant gliomas [62]. Therefore, the observed deregulation of the Ras-RAFERK signaling pathway in gliomas is generally attributed to its upstream positive regulators, including, EGFR and PlateletDerived Growth Factor (PDGFR) known to be highly active in the majority of malignant gliomas [50]. It is likely that mechanisms other than mutations contribute to RAS-MAPK pathway activation in wild-type cancers. Recently, epigenetic alterations were described to potentiate this activation in human tumors [6] and dysregulation of physiologic microRNA (miR) activity has been shown to play an important role in tumor gliomagenesis but the functional relevance of this regulatory layer is currently unknown [63-68].

mi-RNA in treatment of brain tumors

Current postoperative standard treatment for GBM patients is mainly based on unselective induction of DNA damage via radiotherapy and alkylating agents such as Temozolimide (TMZ) [28-32]. The majority of drugs specifically targeting key signalling pathways and mechanisms of gliomagenesis, such as RTK signaling (erlotinib, gefitinib, cetuximab and imatinib) or angiogenesis (bevacizumab and cediranib) do not provide a significant survival benefit when tested alone or in combination with other therapies [31,69]. Though development and optimization of improved miR delivery methods (the principal problem) is necessary [70], targeting RAS-ERK signaling by miR-based therapeutics holds great promise in the treatment of cancers that are reliant on this signaling pathway. Therapeutic anti-miRs are currently being developed for cancer therapy [71,72], however, this is still at an early stage and has not yet been introduced in a clinical setting. Thus far, no clinical trial on miRNA intervention has been conducted in GBM [73]. Interestingly, some authors reported the functional efficiency of anti-miR Let-7a delivery in vivo evaluating its practicality for preclinical study in glioblastoma [74]. In this report authors demonstrate that anti-miRs are able to penetrate the Blood Brain Barrier (BBB) (which makes systemic treatment more difficult) without the use of any viral or lipid carriers, and intracerebro-ventricular administration of anti-miRs could lead to whole-brain distribution, including the tumor region.

CONCLUSION

Control of KRAS expression by the let-7 family of miRNAs has been well documented. The significance of this control mechanism has been highlighted in a number of studies that report a correlation between let-7 expression and cancer. Low levels of let-7 expression in human tumors correlate with high levels of KRAS (at least in lung and colorectal cancers). The control over KRAS expression and activity in glioblastoma (wild-type KRAS tumors) indicates that let-7, even without oncogenic mutation, can regulate RAS activity and that this might be a general phenomenon related to the interactions between tumor suppressor genes (let-7) and proto-oncogenes (KRAS) or oncogenes (KRASG12V or G12D). The possibility of upregulation of tumor suppressor along epigenetic downregulation of protooncogenes is an interesting phenomenon and should be studied in future research.

REFERENCES

1. Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng A, et al. RAS is regulated by the let-7 microRNA family. Cell. 2005; 120: 635- 647.

2. He XY, Chen JX, Zhang Z, Li CL, Peng QL, Peng HM. The let-7a microRNA protects from growth of lung carcinoma by suppression of k-Ras and c-Myc in nude mice. J Cancer Res Clin Oncol. 2010; 136: 1023-1028.

3. Gunzburg MJ, Sivakumaran A, Pendini NR, Yoon JH, Gorospe M, Wilce MC, et al. Cooperative interplay of let-7 mimic and HuR with MYC RNA. Cell Cycle. 2015; 14: 2729-2733.

4. Sampson VB, Rong NH, Han J, Yang Q, Aris V, Soteropoulos P, et al. MicroRNA let-7a down-regulates MYC and reverts MYC-induced growth in Burkitt lymphoma cells. Cancer Res. 2007; 67: 9762-9770.

5. Wong TS, Man OY, Tsang CM, Tsao SW, Tsang RK, Chan JY, et al. MicroRNA let-7 suppresses nasopharyngeal carcinoma cells proliferation through downregulating c-Myc expression. J Cancer Res Clin Oncol. 2011; 137: 415-422.

6. Masliah-Planchon J, Garinet S, Pasmant E. RAS-MAPK pathway epigenetic activation in cancer: miRNAs in action. Oncotarget. 2016; 7: 38892-38907.

7. Brueckner B, Stresemann C, Kuner R, Mund C, Musch T, Meister M, et al. The human let-7a-3 locus contains an epigenetically regulated microRNA gene with oncogenic function. Cancer Res. 2007; 67: 1419- 1423.

8. Meng F, Henson R, Wehbe-Janek H, Smith H, Ueno Y, Patel T. The MicroRNA let-7a modulates interleukin-6-dependent STAT-3 survival signaling in malignant human cholangiocytes. J Biol Chem. 2007; 282: 8256-8264.

9. Hatley ME, Patrick DM, Garcia MR, Richardson JA, Bassel-Duby R, van Rooij E, et al. Modulation of K-Ras-dependent lung tumorigenesis by MicroRNA-21. Cancer Cell. 2010; 18: 282-293.

10. Sharma V, Dixit D, Koul N, Mehta VS, Sen E. Ras regulates interleukin1β-induced HIF-1α transcriptional activity in glioblastoma. J Mol Med (Berl). 2011; 123-136.

11. Zawistowski JS, Nakamura K, Parker JS, Granger DA, Golitz BT, Johnson GL. MicroRNA 9-3p targets β1 integrin to sensitize claudin-low breast cancer cells to MEK inhibition. Mol Cell Biol. 2013; 33: 2260-2274.

12. Lin X, Chen L, Yao Y, Zhao R, Cui X, Chen J, et al. CCL18-mediated downregulation of miR98 and miR27b promotes breast cancer metastasis. Oncotarget. 2015; 6: 20485-20499.

13. Petrillo M, Zannoni GF, Beltrame L, Martinelli E, DiFeo A, Paracchini L, et al. Identification of high-grade serous ovarian cancer miRNA species associated with survival and drug response in patients receiving neoadjuvant chemo. Ann Oncol. 2016; 27: 625-634.

14. Ma L, Li GZ, Wu ZS, Meng G. Prognostic significance of let-7b expression in breast cancer and correlation to its target gene of BSG expression. Med Oncol. 2014; 31: 773.

15. Esquela-Kerscher A, Trang P, Wiggins JF, Patrawala L, Cheng A, Ford L, et al. The let-7 microRNA reduces tumor growth in mouse models of lung cancer. Cell Cycle. 2008; 7: 759-764.

16. Kumar MS, Erkeland SJ, Pester RE, Chen CY, Ebert MS, Sharp PA, et al. Suppression of non-small cell lung tumor development by the let-7 microRNA family. Proc Natl Acad Sci USA. 2008; 105: 3903-3908.

17. Trang P, Medina PP, Wiggins JF, Ruffino L, Kelnar K, Omotola M, et al. Regression of murine lung tumors by the let-7 microRNA. Oncogene. 2010; 29: 1580-1587.

18. Yu F, Yao H, Zhu P, Zhang X, Pan Q, Gong C, et al. let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell. 2007; 131: 1109-1123.

19. Louis DN, Perry A, Reifenberger G, von Deimling A, FigarellaBranger D, Cavenee WK, et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol. 2016; 131: 803-820.

20. Wang Y, Hu X, Greshock J, Shen L, Yang X, Shao Z, et al. Genomic DNA copy-number alterations of the let-7 family in human cancers. PLoS One. 2012; 7: e44399.

21. Turner JD, Williamson R, Almefty KK, Nakaji P, Porter R, Tse V, et al. The many roles of microRNAs in brain tumor biology. Neurosurg Focus. 2010; 28: E3.

22. Tanno B, Babini G, Leonardi S, Giardullo P, De Stefano I, Pasquali E, et al. Ex vivo miRNome analysis in Ptch1+/- cerebellum granule cells reveals a subset of miRNAs involved in radiation-induced medulloblastoma.Oncotarget. 2016; 7: 68253-68269.

23. Powers JT, Tsanov KM, Pearson DS, Roels F, Spina CS, Ebright R, et al. Multiple mechanisms disrupt the let-7 microRNA family in neuroblastoma. Nature. 2016; 535: 246-251.

24. Song H, Zhang Y, Liu N, Zhang D, Wan C, Zhao S, et al. Let-7b inhibits the malignant behavior of glioma cells and glioma stem-like cells via downregulation of E2F2. J Physiol Biochem. 2016; 72:733-744.

25. Wang XR, Luo H, Li HL, Cao L, Wang XF, Yan W, et al. Overexpressed let-7a inhibits glioma cell malignancy by directly targeting K-ras, independently of PTEN. Neuro Oncol. 2013; 15: 1491-1501.

26. Lee ST, Chu K, Oh HJ, Im WS, Lim JY, Kim SK, et al. Let-7 microRNA inhibits the proliferation of human glioblastoma cells. J Neurooncol. 2011; 102: 19-24.

27. Wang X, Xin Z, Xu Y, Ma J. Upregulated miRNA-622 inhibited cell proliferation, motility, and invasion via repressing Kirsten rat sarcoma in glioblastoma. Tumour Biol. 2016; 37: 5963-5970.

28. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005; 352: 987-996.

29. Stupp R, Hegi ME. Brain cancer in 2012: Molecular characterization leads the way. Nat Rev Clin Oncol. 2013; 10: 69-70.

30. Stupp R, Taillibert S, Kanner AA, Kesari S, Steinberg DM, Toms SA, et al. Maintenance Therapy With Tumor-Treating Fields Plus Temozolomide vs Temozolomide Alone for Glioblastoma: A Randomized Clinical Trial. JAMA. 2015; 314: 2535-2543.

31. Gilbert MR, Sulman EP, Mehta MP. Bevacizumab for newly diagnosed glioblastoma. N Engl J Med. 2014; 370: 2048-2049.

32. Gilbert MR. Antiangiogenic Therapy for Glioblastoma: Complex Biology and Complicated Results. J Clin Oncol. 2016; 34: 1567-1569.

33. Khosla D. Concurrent therapy to enhance radiotherapeutic outcomes in glioblastoma. Ann Transl Med. 2016; 4: 54.

34. Jason T. Huse & Eric CH. Targeting brain cancer: advances in the molecular pathology of malignant glioma and medulloblastoma. Nature Reviews Cancer. 2010; 319-331.

35. Omuro A, DeAngelis LM. Glioblastoma and other malignant gliomas: a clinical review. JAMA. 2013; 310: 1842-1850.

36. Büssing I, Slack FJ, Grosshans H. let-7 microRNAs in development, stem cells and cancer. Trends Mol Med. 2008; 14: 400-409.

37. Yu ML, Wang JF, Wang GK, You XH, Zhao XX, Jing Q, et al. Vascular smooth muscle cell proliferation is influenced by let-7d microRNA and its interaction with KRAS. Circ J. 2011; 75: 703-709.

38. Graziano F, Canestrari E, Loupakis F, Ruzzo A, Galluccio N, Santini D, et al. Genetic modulation of the Let-7 microRNA binding to KRAS 3’-untranslated region and survival of metastatic colorectal cancer patients treated with salvage cetuximab-irinotecan. Pharmacogenomics J. 2010; 10: 458-464.

39. Christensen BC, Moyer BJ, Avissar M, Ouellet LG, Plaza SL, McClean MD, et al. A let-7 microRNA-binding site polymorphism in the KRAS 3’ UTR is associated with reduced survival in oral cancers. Carcinogenesis. 2009; 30: 1003-1007.

40. Langevin SM, Christensen BC. Let-7 microRNA-binding-site polymorphism in the 3’UTR of KRAS and colorectal cancer outcome: a systematic review and meta-analysis. Cancer Med. 2014; 3: 1385- 1395.

41. Areeb Z, Stylli SS, Koldej R, et al. MicroRNA as potential biomarkers in Glioblastoma. J Neurooncol. 2015; 125: 237-248.

42. Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, et al. The somatic genomic landscape of glioblastoma. Cell. 2013; 155: 462-477.

43. Wang Z, Lin S, Zhang J, Xu Z, Xiang Y, Yao H, et al. Loss of MYC and E-box3 binding contributes to defective MYC-mediated transcriptional suppression of human MC-let-7a-1~let-7d in glioblastoma. Oncotarget. 2016; 7: 56266-56278.

44. Lee ST, Chu K, Oh HJ, Im WS, Lim JY, Kim SK, et al. Let-7 microRNA inhibits the proliferation of human glioblastoma cells. J Neurooncol. 2011; 102: 19-24.

45. Buechner J, T mte E, Haug BH, Henriksen JR, L kke C, Fl gstad T, et al. Tumour-suppressor microRNAs let-7 and mir-101 target the protooncogene MYCN and inhibit cell proliferation in MYCN-amplified neuroblastoma. Br J Cancer. 2014; 105: 296-303.

46. Molenaar JJ, Domingo-Fernández R, Ebus ME, Lindner S, Koster J, Drabek K, et al. LIN28B induces neuroblastoma and enhances MYCN levels via let-7 suppression. Nat Genet. 2012; 44: 1199-1206.

47. Chaudhry MA, Sachdeva H, Omaruddin RA. Radiation-induced micro-RNA modulation in glioblastoma cells differing in DNA-repair pathways. DNA Cell Biol. 2010; 29: 553-561.

48. Zhao Y, Pang D, Wang C, Zhong S, Wang S. MicroRNA-134 modulates glioma cell U251 proliferation and invasion by targeting KRAS and suppressing the ERK pathway. Tumour Biol. 2016; 37: 11485-11493.

49. Li Y, Li Y, Ge P, Ma C. MiR-126 Regulates the ERK Pathway via Targeting KRAS to Inhibit the Glioma Cell Proliferation and Invasion. Mol Neurobiol. 2017; 54: 137-145.

50. Lo HW. Targeting Ras-RAF-ERK and its interactive pathways as a novel therapy for malignant gliomas. Curr Cancer Drug Targets. 2010; 10: 840-848.

51. Wang L, Yamaguchi S, Burstein MD, Terashima K, Chang K, Ng HK, et al. Novel somatic and germline mutations in intracranial germ cell tumours. Nature. 2014; 511: 241-245.

52. Shi Z, Chen Q, Li C, Wang L, Qian X, Jiang C, et al. MiR-124 governs glioma growth and angiogenesis and enhances chemosensitivity by targeting R-Ras and N-Ras. Neuro Oncol. 2014; 16: 1341-1353.

53. Prior IA, Lewis PD, Mattos C. A comprehensive survey of Ras mutations in cancer. Cancer Res. 2012; 72: 2457-2467.

54. Milinkovic VP, Skender Gazibara MK, Manojlovic Gacic EM, Gazibara TM, Tanic NT. The impact of TP53 and RAS mutations on cerebellar glioblastomas. Exp Mol Pathol. 2014; 97: 202-207.

55. Knobbe CB, Reifenberger J, Reifenberger G. Mutation analysis of the Ras pathway genes NRAS, HRAS, KRAS and BRAF in glioblastomas. Acta Neuropathol. 2004; 108: 467-470.

56. Ding H, Roncari L, Shannon P, Wu X, Lau N, Karaskova J, et al. Astrocyte-specific expression of activated p21-ras results in malignant astrocytoma formation in a transgenic mouse model of human gliomas. Cancer Res. 2001; 61: 3826-3836.

57. Vitucci M, Karpinich NO, Bash RE, Werneke AM, Schmid RS, White KK. Cooperativity between MAPK and PI3K signaling activation is required for glioblastoma pathogenesis. Neuro Oncol. 2013; 15:1317- 1329.

58. Eleveld TF, Oldridge DA, Bernard V, Koster J, Daage LC, Diskin SJ, et al. Relapsed neuroblastomas show frequent RAS-MAPK pathway mutations. Nat Genet. 2015; 47: 864-871.

59. Dasgupta B, Li W, Perry A, Gutmann DH. Glioma formation in neurofibromatosis 1 reflects preferential activation of K-RAS in astrocytes. Cancer Res. 2005; 65: 236-245.

60. Holmen SL, Williams BO. Essential role for Ras signaling in glioblastoma maintenance. Cancer Res. 2005; 65: 8250-8255.

61. Bunda S, Burrell K, Heir P, Zeng L, Alamsahebpour A, Kano Y, et al. Inhibition of SHP2-mediated dephosphorylation of Ras suppresses oncogenesis. Nat Commun. 2015; 6: 8859. 62.Gömöri E, Dóczi T, Pajor L, Matolcsy A. Sporadic p53 mutations and absence of ras mutations in glioblastomas. Acta Neurochir (Wien). 1999; 593-599.

63. Gabriely G, Yi M, Narayan RS, Niers JM, Wurdinger T, Imitola J, et al. Human glioma growth is controlled by microRNA-10b. Cancer Res. 2011; 71: 3563-3572.

64. Godlewski J, Nowicki MO, Bronisz A, Williams S, Otsuki A, Nuovo G, et al. Targeting of the Bmi-1 oncogene/stem cell renewal factor by microRNA-128 inhibits glioma proliferation and self-renewal. Cancer Res. 2008; 68: 9125-9130.

65. Kim H, Huang W, Jiang X, Pennicooke B, Park PJ, Johnson MD. Integrative genome analysis reveals an oncomir/oncogene cluster regulating glioblastoma survivorship. Proc Natl Acad Sci U S A. 2010; 107: 2183-2188.

66. Kim TM, Huang W, Park R, Park PJ, Johnson MD. A developmental taxonomy of glioblastoma defined and maintained by MicroRNAs. Cancer Res. 2011; 71: 3387-3399.

67. Kwak HJ, Kim YJ, Chun KR, Woo YM, Park SJ, Jeong JA, et al. Downregulation of Spry2 by miR-21 triggers malignancy in human gliomas. Oncogene. 2011; 30: 2433-2442.

68. Wang X, Xin Z, Xu Y, Ma J. Upregulated miRNA-622 inhibited cell proliferation, motility, and invasion via repressing Kirsten rat sarcoma in glioblastoma. Tumour Biol. 2016; 37: 5963-5970.

69. Tanaka S, Louis DN, Curry WT, Batchelor TT, Dietrich J. Diagnostic and therapeutic avenues for glioblastoma: no longer a dead end? Nat Rev Clin Oncol. 2013; 10: 14-26.

70. Kasinski AL, Kelnar K, Stahlhut C, Orellana E, Zhao J, Shimer E, et al. A combinatorial microRNA therapeutics approach to suppressing nonsmall cell lung cancer. Oncogene. 2015; 34: 3547-3555.

71. Garofalo M, Leva GD, Croce CM. MicroRNAs as anti-cancer therapy. Curr Pharm Des. 2014; 20: 5328-5335.

72. Ji W, Sun B, Su C. Targeting MicroRNAs in Cancer Gene Therapy. Genes (Basel). 2017; 8: 21.

73. Chen L, Kang C. miRNA interventions serve as ‘magic bullets’ in the reversal of glioblastoma hallmarks. Oncotarget. 2015; 6: 38628- 38642.

74. Kim DG, Kim KH, Seo YJ, Yang H, et al. Anti-miR delivery strategies to bypass the blood-brain barrier in glioblastoma therapy. Oncotarget. 2016; 7: 29400-29411.

 

Messina S, Frati L (2017) RAS Oncogene in Brain Tumors, Let-7 MicroRNA Involvement. J Cancer Biol Res 5(1): 1096.

Received : 15 Dec 2017
Accepted : 21 Jan 2017
Published : 24 Jan 2017
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X