#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Long Non-coding RNAs as Regulators of the Mitogen-activated Protein Kinase (MAPK) Pathway in Cancer


Dlouhé nekódující molekuly RNA jako regulátory mitogenem aktivované proteinkinázové dráhy (MAPK) v nádorech

Úvod:
Mitogenem aktivovaná proteinkinázová dráha (MAPK) přispívá k regulaci mnoha buněčných funkcí, jako je proliferace a diferenciace buněk, mobilita a apoptóza. Extracelulární signálně regulovaná kináza 1/2 (ERK1/2), c-Jun N-terminální kináza (JNK) /p38 a ERK5 jsou tři hlavní moduly v této dráze. Kaskády Raf-ERK1/2 a JNK přispívají k proliferaci buněk, migraci a přežití a jsou hlavními regulátory maligního fenotypu. Tato dráha je sama regulována několika vnějšími signály, stejně jako bočními signály z jiných signálních drah, které vytvářejí komplexní síť. Dlouhé nekódující RNA (lncRNA) jako hlavní modulátory genové exprese na transkripční a posttranskripční úrovni a také regulují tuto dráhu. Kromě toho můžou lncRNA sloužit jako biomarker a cíl nových léčebných strategií u pacientů s nádory.

Cíl:
Prozkoumat roli lncRNA v regulaci dráhy MAPK.

Závěr:
Vzhledem k úloze této dráhy v patogenezi několika typů nádorů dochází ke změnám exprese lncRNA, které vedou ke změnám v dráze MAPK, což vede k inhibici apoptózy a indukci buněčné proliferace a migrace. Některé lncRNA se navíc podílejí na spojení mezi MAPK a jinými drahami souvisejícími s nádory, jako je dráha PI3K/Akt prostřednictvím regulace určitých sdílených proteinů mezi těmito drahami. Na základě dostupnosti některých protinádorových léčiv, modulujících tuto dráhu, by identifikace lncRNA, ovlivňující tuto dráhu pomohla při vytváření účinných terapií.

Klíčová slova:
RNA – dlouhé nekódování – mitogenem aktivované proteinové kinázy – signální transdukce

Autoři deklarují, že v souvislosti s předmětem studie nemají žádné komerční zájmy.

Redakční rada potvrzuje, že rukopis práce splnil ICMJE kritéria pro publikace zasílané do biomedicínských časopisů.

Obdrženo:
5. 12. 2017

Přijato:
5. 2. 2018


Authors: Tasharrofi Behnoosh;  Ghafouri-Fard Soudeh
Authors place of work: Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
Published in the journal: Klin Onkol 2018; 31(2): 95-102
Category: Přehled
doi: https://doi.org/10.14735/amko201895

Summary

Background:
The mitogen-activated protein kinase (MAPK) pathway contributes to regulation of many cellular functions, such as cell proliferation and differentiation, mobility and apoptosis. Extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun N-terminal kinase (JNK) /p38 and ERK5 construct the three main modules in this pathway. The Raf-ERK1/2 and JNK cascades contribute in cell proliferation, migration, and survival and are principal regulators of malignant phenotype. This pathway is itself regulated by several outside signals as well as lateral signals from other pathways, which construct a complex network. Long non-coding RNAs (lncRNAs) as principal modulators of gene expression at transcriptional and post-transcriptional levels also regulate this pathway. In addition, lncRNA signature can be used as biomarker and target of novel treatment strategies in cancer patients.

Aim:
To explore the role of lncRNAs in regulation of MAPK pathway.

Conclusion:
Considering the role of this pathway in the pathogenesis of several cancers, alterations in lncRNA expression lead to changes in MAPK pathway resulting in inhibition of apoptosis and induction of cell proliferation and migration. Moreover, some lncRNAs participate in cross-talk between MAPK and other cancer-related pathways, such as PI3K/Akt pathway through regulation of certain shared proteins between these pathways. Based on the availability of certain anticancer drugs that modulate this pathway, identification of lncRNAs that affect this pathway would help in establishment of effective therapies.

Key words:
RNA – long noncoding – mitogen-activated protein kinases – signal transduction

Introduction

The mitogen-activated protein kinase (MAPK) pathway contributes to a wide range of cellular functions, such as cell proliferation and differentiation, mobility and apoptosis. Several lines of evidence suggest that this pathway does not work as a linear signaling pathway that directly conveys certain signals to a definite cellular response. Instead, several outside signals as well as lateral signals from other pathways contribute to construction of a complex network through which MAPK pathway operates [1]. Abnormal regulation of this pathway participates in the pathogenesis of several malignancies [2]. Extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun N-terminal kinase (JNK) / p38 and ERK5 construct the three main modules in this pathway [2]. The Raf-ERK1/2 and JNK cascades contribute to cell proliferation, migration, and survival and are principal regulators of malignant phenotype [3].

Among putative regulators of this pathway are long non-coding RNAs (lncRNAs). LncRNAs make a high proportion of human transcripts, which are not translated to proteins [4,5]. With sizes more than 200 base pairs, they regulate expression of protein coding genes a well as microRNAs (miRNAs) [6]. Subsequently, they affect telomere biology, chromatin dynamics, genome structure organization [7], DNA repair [8] and tissue homeostasis [9]. They also participate in tumorigenesis process via different mechanisms, such as transcription repression [10], induction of epithelial-mesenchymal transition (EMT) [11], inhibition of tumor suppressors expression [12,13], over-expression of certain oncogenes [14,15], regulation of tumor-associated signaling pathways [16] as well as modulation of tumor microenvironment [17]. Recently, a handful of lncRNAs have been shown to regulate the MAPK pathway through modulation of the expression of different proteins within this pathway. We discuss specific roles of these lncRNAs in the following sections. Special site of their action in the MAPK pathway has been demonstrated in Fig. 1. In addition, their characteristics, such as their genomic location and their expression pattern in cancers have been summarized in Tab. 1.

Schema of mitogen-activated protein kinase (MAPK) pathway with several proteins involved in it. Long non-coding RNAs (demonstrated in red terapeziuses) regulate this pathway at several points.
Fig. 1. Schema of mitogen-activated protein kinase (MAPK) pathway with several proteins involved in it. Long non-coding RNAs (demonstrated in red terapeziuses) regulate this pathway at several points.

Tab. 1. Detailed characteristics of long non-coding RNAs that regulate MAPK pathway.
Detailed characteristics of long non-coding RNAs that regulate MAPK pathway.

Antisense RNA in the INK4 locus (ANRIL)

Aberrant expression of ANRIL has been detected in a variety of cancers including glioma in which its expression has been associated with tumor grade and patients survival. In vitro studies have shown that ANRIL silencing results in suppression of cell proliferation as well as induction of cell cycle arrest and apoptosis in glioma cells by modulation of expression of ERK, JNK and p38 from MAPK signaling pathway [18].

BMP/OP-Responsive Gene (BORG)

The expression levels of this lncRNA have been associated with invasive and metastatic potential of breast tumors. Its role in breast cancer pathogenesis is exerted through binding with Tripartite Motif Containing 28 (TRIM28) protein and subsequent enhancement of its localization and transcriptional repressive activity. BORG-expressing breast cancer cells attain metastatic characteristics through inhibition of p38 MAPK activity and enhancement of ERK1/2 function. This BORG-mediated alteration in MAPK function was TRIM28 dependent as demonstrated by high p38 MAPK: ERK1/2 activity ratios in TRIM28 silenced cells [19].

BRAF-activated non-coding RNA (BANCR)

Elevated expression of BANCR has been detected in human malignant melanoma cell lines and tissues. BANCR silencing has suppressed proliferation of tumor cells and inhibited MAPK pathway through down-regulation of ERK1/2 and JNK [3]. In addition, BANCR silencing in endometrial cancer cells inhibited their proliferation and arrested the cell cycle at G0/G1. BANCR anti-apoptotic role is possibly exerted through up-regulation of Cyclin D1 and Bcl-2. Besides, BANCR activates ERK/MAPK pathway and then induces expression of matrix metalloproteinases 1 and 2 (MMP1 and MMP2) in endometrial cancer cells leading to enhancement in proliferation, migration, and invasion of these cells [20]. In colorectal cancer (CRC) and lung cancer, BANCR activates mitogen-activated protein kinase (MEK) /ERK and JNK/MAPK pathways, resp. [21,22].

Cancer susceptibility candidate 2 (CASC2)

Its role as a tumor suppressor gene was first demonstrated in endometrial cancer in which its down-regulation has been detected [23,24]. Subsequently, its down-regulation has been confirmed in glioma and renal cell carcinoma tissues and cell lines where CASC2 exerts its tumor suppressive role through down-regulation of miR-21 [25,26]. In lung cancer, its down-regulation was significantly correlated with advanced tumor, node and metastasis (TNM) stage and tumor size indicating its role as an independent predictor for overall survival (OS) of these patients [27]. Furthermore, lower expression of this lncRNA in CRC patients was associated with advanced TNM stage. In vitro studies have shown that it functions as a competing endogenous RNA (ceRNA) for miR-18 leading to up-regulation of expression of PIAS3 and STAT3 downstream genes and subsequent suppression of CRC cell proliferation [28]. More recently, its aberrant expression has been demonstrated in hepatocellular carcinoma (HCC) and gastric cancer where it has been shown to exert its role via inhibition of MAPK pathway [29,30]. In CASC2 over-expressed CRC cells, phosphorylation of ERK and JNK was decreased showing the role of CASC2 in inhibition of the MAPK pathway [29].

Colon cancer-associated transcript-1 (CCAT1)

The expression levels of CCAT1 increase in a variety of tumors including medulloblastoma. Its silencing has remarkably suppressed cell proliferation, induced cell cycle arrest in medulloblastoma cells and decreased phosphorylated levels of MAPK, ERK and MEK. So the oncogenic roles of CCAT1 in medulloblastoma is possibly exerted via regulating the MAPK pathway [31].

Colorectal neoplasia differentially expressed (CRNDE)

The elevated expression of this lncRNA has been detected in colorectal adenomas and cancers compared with normal colon epithelia [32]. In addition, high plasma levels of a certain splice variant of CRNDE is a putative plasma biomarker for CRC with a sensitivity of 87% and specificity of 93% [32]. Up-regulation of this lncRNA has been demonstrated in lymphocytic leukemias as well as various solid tumors [33]. Notably, in addition to anti-apoptotic effects, CRNDE has a role in enhancement of proliferation, migration and invasion of glioma stem cells [34]. Its over-expression has been associated with poor patients‘ outcome in ovarian cancer [35] and CRC patients [36]. More recently, over-expression of this lncRNA has been demonstrated in a large cohort of CRC patients with remarkable associations between its high expression and advanced cancer stages as well as poor prognosis [37]. Notably, the mRNA expression profile of CRNDE-knocked out CRC cells shows its regulatory function on the Ras and MAPK pathways. In addition, many of CRNDE-induced Ras/MAPK signaling genes participate in CRC development [37].

Cytoskeleton regulator RNA (CYTOR)

This intergenic lncRNA has a crucial role in cell proliferation, cell migration, and cytoskeletal organization. Its expression has been increased in all subtypes of breast cancer as well as a variety of other malignancies including thyroid, stomach, lung, renal, and liver cancers. Using an innovative method based on the recognition of correlations between the expression of lncRNA and protein-coding genes which participate in certain functions, CYTOR has been shown to be associated with the epidermal growth factor receptor (EGFR), mammalian target of rapamycin (mTOR), and MAPK pathways [38].

Insulin-like growth factor 2 antisense 1 (Igf2as)

The expression of this lncRNA has been increased in HCC cells and tissues. Its silencing inhibited cell proliferation and migration and induced apoptosis through inactivation of ERK/MAPK signaling pathway [39].

LincRNA regulator of reprogramming (linc-RoR)

This lncRNA was initially recognized to alter reprogramming of induced pluripotent stem cells [40]. Subsequently, it has been demonstrated to act as a miRNA sponge which changes the expression of the principal transcription factors in human embryonic stem cells [41]. Besides, it both inhibits p53 translation [42] and enhances c-Myc expression through interaction with heterogeneous nuclear ribonucleoprotein I (hnRNP I) [43]. While in triple negative breast cancer, linc-RoR increases cell invasion via miR-145/ARF6 pathway [44], in estrogen receptor positive breast tumors it activates MAPK/ERK pathway through diminishing the stability of dual specificity phosphatase 7 (DUSP7) which down-regulates ERK [45].

LncRNA-AWPPH

This lncRNA has been firstly identified as an over-expressed lncRNA in HCC whose expression was correlated with invasion, advanced TNM stage as well as poor recurrence-free and OS. In vitro studies have shown its interaction with Y-Box Binding Protein 1 (YBX1) which leads to up-regulation of SNAIL1 and PIK3CA expressions and activation of the PI3K/AKT pathway [46].

Lnc00462717

In glioma tissues, its low expression was associated with the malignant status. In vitro studies have shown its suppressive effect on glioma cell proliferation, survival and migration, which is possibly due to its negative effect on MDM2 expression. In addition, it has a regulatory role on MAPK pathway through decreasing the phosphorylation of p38, ERK1/2, and JNK [47].

LncRNA-low expression in tumor (LET)

It is a tumor suppressor lncRNA, which is down-regulated in a variety of malignancies including nasopharyngeal carcinoma (NPC). In vitro studies have demonstrated its role in regulation of cell proliferation, adhesion and invasion of NPC through modulation of MAPK/ERK pathway genes. p-ERK1/2 and p-MEK were considerably down-regulated in LET over-expressed cells with no change in total-ERK1/2 and total-MEK levels. So LET inhibits the cell growth and invasion of NPC cells via suppression of ERK signaling pathway [48].

Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1)

This lncRNA has been known as an oncogene associated with metastasis potent-ial of several cancers. MALAT1 enhances cancer cell migration and the EMT by modulation of the expression of several genes [49]. MALAT1 silencing in gall bladder cancer (GBC) cell lines has led to suppression of the proliferation and metastasis of these cell in addition to down-regulation of the expression of phosphorylated MEK1/2, ERK 1/2, MAPK, and JNK 1/2/3 leading to inhibition of MAPK pathway [50]. Moreover, knockout of MALAT1 in lung cancer cells has resulted in down-regulation of numerous metastasis related genes including Glypican 6 (GPC6) and C-X-C motif chemokine 5 (CXCL5) [51] both of which participate in regulation of MAPK pathway [52,53]. Notably, MALAT1 has been one of the most considerably up-regulated lncRNAs during neuroblastoma-derived Neuro-2a (N2a) cell differentiation whose silencing has impaired neurite outgrowth and induced cell death possibly via inactivation of MAPK signaling pathway as well as aberrant activation of Peroxisome proliferator-activated receptor (PPAR) and P53 signaling pathway [54].

Metastatic renal cell carcinoma-associated transcript 1 (MRCCAT1)

This lncRNA is over-expressed in metastatic clear cell renal cell carcinoma (ccRCC) tissues. Its expression is associated with the metastatic phenotype of these cells as well as patients‘ outcome. MRCCAT1 inhibits Natriuretic Peptide Receptor 3 (NPR3) transcription by recruiting polycomb repressive complex 2 (PRC2) to NPR3 promoter leading to activation of p38-MAPK signaling pathway. Forced over-expression of this lncRNA has resulted in elevation of phosphorylated p38 levels while the phosphorylation levels of ERK and JNK have not been altered [55].

Maternally expressed gene 3 (MEG3)

This imprinted gene is expressed in a variety of normal tissues [56]. On the other hand, its expression has been decreased or lost in several tumors including glioma [57], meningiomas [58], hepatoma [59], gastric cancer [60] and breast cancer [61]. Its tumor suppressive effects have been demonstrated by several in vitro studies [57,60]. In breast cancer cells, it has a fundamental role in regulation of cell proliferation and invasion as well as expression of angiogenesis-related factors which is at least partially exerted through AKT signaling pathway [61].

Melanoma highly expressed noncoding RNA (MHENCR)

Its expression has been increased in melanoma tissues and metastatic melanoma in association with poor survival of melanoma patients. Its silencing has resulted in inhibition of cells proliferation and migration as well as enhancement of cell cycle arrest and apoptosis. It acts as a ceRNA for miR-425 and miR-489 leading to over-expression of their target genes IGF1 and SPIN1 and subsequent activation of PI3K-Akt pathway [62]. Considering the role of miR-489 in regulation of MAPK signaling [63], MHENCR might be involved in regulation of this pathway as well.

PTEN pseudogene-1 (PTENP1)

Its over-expression in MCF7 breast can-cer cells has resulted in inhibition of cell proliferation and migration as well as down-regulation of cyclin A2, CDK2, p-AKT, p-p44/42 MAPK and p-p38 MAPK, so this lncRNA can impede the proliferation and migration of breast cancer cells through modulation of AKT and MAPK signaling pathways [64].

RMEL3 (ENSG00000250961)

Elevated expression of RMEL3 in melanoma is significantly linked with the existence of BRAFV600E mutation. Its knock-down has considerably decreased colony formation in melanoma cell lines that harbor this mutation. Its expression levels have been correlated with the expression of several MAPK and PI3K pathways genes. Besides, its silencing has diminished expression of pAKT and BRAF in addition to several activators or effectors of MAPK and PI3K pathways such as FGF2, FGF3, DUSP6, ITGB3 and GNG2. Consequently, both in silico analysis and experimental data confirmed its role in MAPK and PI3K signaling [65].

Taurine-upregulated gene 1 (TUG1)

This lncRNA has a role in regulation of gene expression through engagement of PRC2. Its down-regulation has been demonstrated in a variety of cancers including non-small cell lung carcinoma (NSCLC) tissues in which its down-expression has been associated with higher TNM stage and tumor size, as well as poor patients‘ outcome. It has been shown to be a direct transcriptional target of p53 whose silencing leads to enhancement of cell proliferation and up-regulation of homeobox B7 (HOXB7) expression. As the levels of p-ERK, p-AKT and p-GSK3b were increased following TUG1 silencing, it has been hypothesized that TUG1 contributes in AKT/MAPK pathway through modulation of HOXB7 expression [66].

Urothelial carcinoma associated 1 (UCA1)

This lncRNA has an oncogenic role in human bladder cancer pathogenesis. Its silencing has resulted in down-regulation of p300 (encoded by EP300) and its coactivator cAMP response element-binding protein (CREB). In addition, UCA1 modulates AKT activity and cell cycle progression through CREB [67]. CREB is also been phosphorylated through activation of MAPK-interacting kinase (MNK) in MAPK cascade [68].

Up-regulated in hepatocellular carcinoma (URHC)

Up-regulation of this lncRNA in HCC cell lines has been demonstrated through using lncRNA microarray. In addition, its over-expression in HCC tissues has been correlated with poor OS. In vitro studies have shown its role in modulation of cell proliferation and apoptosis through down-regulation of leucine zipper containing kinase AZK (ZAK). ERK, JNK and p38 kinase phosphorylation has been altered in URHC knocked-down cells, which implies that the role of URHC in modulation of cell growth and apoptosis might be exerted through inactivation of the MAPK signaling pathway. Consequently, URHC regulates cell proliferation and apoptosis via ERK/MAPK inhibition by targeting ZAK [69].

Discussion

Recent studies have demonstrated that lncRNA signature can be used as biomarker and target of novel treatment strategies in cancer patients [70,71]. LncRNAs participate in principal cancer-related processes, such as cell proliferation, cell migration, and EMT through the modulation of important signaling pathways including mTOR and MAPK pathways [38]. A comprehensive genome-wide transcriptional investigation of lncRNA signature in breast cancer samples has led to identification of lncRNA specific profile in breast cancer subtypes. Of note, among the nine lncRNAs that constitute the luminal B signature, one chief lncRNA category was linked to the MAPK pathway [38]. Considering the prominent role of MAPK pathway in the pathogenesis of melanoma and existence of MAPK targeting therapeutic modalities in melanoma [72], expression analysis of MAPK-related lncRNAs in this kind of cancer is of special significance.

LncRNAs regulatory roles in fundamental cellular pathways are exerted through modulation of gene expression at the transcriptional or posttranscriptional level. Some of their functions have been elaborated, while others need to be clarified. For instance, the regulatory role of CRNDE in regulation of MAPK pathway in CRC [37] and in the maintenance of glioma stem cells [34] have been demonstrated. Considering the role of MAPK in glioma stem cells [73], future studies are needed to elucidate the functional link between the MAPK pathway, CRNDE and stem cell maintenance in glioma. Besides, consistent with the existence of a complex interaction network between lncRNAs, miRNAs and protein coding genes especially in the context of cancer, CASC2 has been shown to exert its tumor suppressive role via modulation of both miRNAs and protein coding genes including those located in MAPK pathway [25,26,29]. Additionally, as revealed by independent studies, a certain lncRNA might regulate expression of different pathways in distinct types of cancers. For instance, while MALAT1 activates MAPK pathway in GBC [50], it induces the phosphoinositide 3-kinase (PI3K/Akt) pathway in osteosarcoma cells without any significant alteration in MAPK pathway in these cells [74].

Notably, lncRNA-AWPPH and UCA1 possibly participate in cross-talk between MAPK and PI3K/Akt pathways through regulation of certain shared proteins between these pathways. Previous reports have demonstrated that these two signaling pathways interact with each other at multiple levels. However, as revealed by both in silico and experimental studies, such interactions are context-dependent [75] which is in line with the proposed background-reliant role of lncRNAs in regulation of these pathways.

Considering the advent of pathway-based therapeutic modalities for cancer especially in the context of precision medicine [76], expression analysis of lncRNAs that modulate certain cancer related pathways is of clinical importance. Future studies are needed to elaborate function of lncRNAs in these pathways in each cancer type.

The authors declare they have no potential conflicts of interest concerning drugs, products, or services used in the study.

The Editorial Board declares that the manuscript met the ICMJE recommendation for biomedical papers.

Submitted: 5. 12. 2017

Accepted: 5. 2. 2018

Soudeh Ghafouri-Fard, MD, PhD

Department of Medical Genetics

Shahid Beheshti University of Medical Sciences

Bldg No.2 SBUMS

Arabi Ave, Daneshjoo Blvd, Velenjak Tehran, Iran

e-mail: s.ghafourifard@sbmu.ac.ir 


Zdroje

1. Schaeffer HJ, Weber MJ. Mitogen-activated protein kinases: Specific messages from ubiquitous messengers. Mol Cell Biol 1999; 19 (4): 2435–2444.

2. Sebolt-Leopold JS, Herrera R. Targeting the mitogen-activated protein kinase cascade to treat cancer. Nat Rev Cancer 2004; 4 (12): 937–947. doi: 10.1038/nrc1503.

3. Li R, Zhang L, Jia L et al. Long non-coding RNA BANCR promotes proliferation in malignant melanoma by regulating MAPK pathway activation. Plos One 2014; 9 (6): e100893. doi: 10.1371/journal.pone.0100893.

4. Soudyab M, Iranpour M, Ghafouri-Fard S. The role of long non-coding RNAs in breast cancer. Arch Iran Med 2016; 19 (7): 508–517. doi: 0161907/AIM.0011.

5. Iranpour M, Soudyab M, Geranpayeh L et al. Expression analysis of four long noncoding RNAs in breast cancer. Tumour Biol 2016; 37 (3): 2933–2940. doi: 10.1007/s13277-015-4135-2.

6. Nikpayam E, Tasharrofi B, Sarrafzadeh S et al. The role of long non-coding RNAs in ovarian cancer. Iran Biomed J 2017; 21 (1): 3–15. doi: 10.6091/.21.1.24.

7. Dianatpour A, Ghafouri-Fard S. Long non coding RNA expression intersecting cancer and spermatogenesis: A systematic review. Asian Pac J Cancer Prev 2017; 18 (10): 2601–2610. doi: 10.22034/APJCP.2017.18.10. 2601.

8. Dianatpour A, Ghafouri-Fard S. The role of long non coding RNAs in the repair of dna double strand breaks. Int J Mol Cell Med 2017; 6 (1): 1–12.

9. Melissari MT, Grote P. Roles for long non-coding RNAs in physiology and disease. Pflugers Archiv 2016; 468 (6): 945–958. doi: 10.1007/s00424-016-1804-y.

10. Sarrafzadeh S, Geranpayeh L, Ghafouri-Fard S. Expression analysis of long non-coding PCAT-1in breast cancer. Int J Hematol Oncol Stem Cell Res 2017; 11 (3): 185–191.

11. Nikpayam E, Soudyab M, Tasharrofi B et al. Expression analysis of long non-coding ATB and its putative target in breast cancer. Breast Dis 2017; 37 (1): 11–20. doi: 10.3233/BD-160264.

12. Khorshidi HR, Taheri M, Noroozi R et al. ANRIL genetic variants in iranian breast cancer patients. Cell J 2017; 19 (Suppl 1): 72–78. doi: 10.22074/cellj.2017.4496.

13. Taheri M, Pouresmaeili F, Omrani MD et al. Association of ANRIL gene polymorphisms with prostate cancer and benign prostatic hyperplasia in an Iranian population. Biomark Med 2017; 11 (5): 413–422. doi: 10.2217/bmm-2016-0378.

14. Taheri M, Habibi M, Noroozi R et al. HOTAIR genetic variants are associated with prostate cancer and benign prostate hyperplasia in an Iranian population. Gene 2017; 613: 20–24. doi: 10.1016/j.gene.2017.02.031.

15. Khorshidi HR, Taheri M, Noroozi R et al. Investigation of the association of HOTAIR single nucleotide polymorphisms and risk of breast cancer in an iranian population. Int J Cancer Manag 2017; 10 (5): e7498. doi: 10.5812/ijcm.7498.

16. Sarrafzadeh S, Geranpayeh L, Tasharrofi B et al. Expression study and clinical correlations of MYC and CCAT2 in breast cancer patients. Iran Biomed J 2017; 21 (5): 303–311.

17. Tasharrofi B, Soudyab M, Nikpayam E et al. Comparative expression analysis of hypoxia-inducible factor-alpha and its natural occurring antisense in breast cancer tissues and adjacent noncancerous tissues. Cell Biochem Funct 2016; 34 (8): 572–578. doi: 10.1002/cbf.3230.

18. Xu XC, Wan XF, Zhang ZR. Long non-coding RNA ANRIL promotes tumorigenesis in glioma via MAPK signaling pathways. Int J Clin Exp Patho 2016; 9 (10): 10803–10809.

19. Gooding AJ, Zhang B, Jahanbani FK et al. The lncRNA BORG drives breast cancer metastasis and disease recurrence. Sci Rep 2017; 7: 12698. doi: 10.1038/s41598-017-12716-6.

20. Wang DN, Wang DB, Wang N et al. Long non-coding RNA BANCR promotes endometrial cancer cell proliferation and invasion by regulating MMP2 and MMP1 via ERK/MAPK signaling pathway. Cell Physiol Biochem 2016; 40 (3–4): 644–656. doi: 10.1159/000452577.

21. Quo QH, Zhao Y, Chen JJ et al. BRAF-activated long non-coding RNA contributes to colorectal cancer migration by inducing epithelial-mesenchymal transition. Oncol Lett 2014; 8 (2): 869–875. doi: 10.3892/ol.2014.2154.

22. Jiang WJ, Zhang DD, Xu BN et al. Long non-coding RNA BANCR promotes proliferation and migration of lung carcinoma via MAPK pathways. Biomed Pharmacother 2015; 69: 90–95. doi: 10.1016/j.biopha.2014.11.027.

23. Baldinu P, Cossu A, Manca A et al. Identification of a novel candidate gene, CASC2, in a region of common allelic loss at chromosome 10q26 in human endometrial cancer. Hum Mutat 2004; 23 (4): 318–326. doi: 10.1002/humu.20015.

24. Baldinu P, Cossu A, Manca A et al. CASC2a gene is down-regulated in endometrial cancer. Anticancer Res 2007; 27 (1A): 235–243.

25. Wang P, Liu YH, Yao YL et al. Long non-coding RNA CASC2 suppresses malignancy in human gliomas by miR-21. Cell Signal 2015; 27 (2): 275–282. doi: 10.1016/j.cellsig.2014.11.011.

26. Cao Y, Xu R, Xu X et al. Downregulation of lncRNA CASC2 by microRNA-21 increases the proliferation and migration of renal cell carcinoma cells. Mol Med Rep 2016; 14 (1): 1019–1025. doi: 10.3892/mmr.2016.5337.

27. He X, Liu Z, Su J et al. Low expression of long noncoding RNA CASC2 indicates a poor prognosis and regulates cell proliferation in non-small cell lung cancer. Tumour Biol 2016; 37 (7): 9503–9510. doi: 10.1007/s13277-016-4787-6.

28. Huang G, Wu X, Li S et al. The long noncoding RNA CASC2 functions as a competing endogenous RNA by sponging miR-18a in colorectal cancer. Sci Rep 2016 20; 6: 26524. doi: 10.1038/srep26524.

29. Gan Y, Han N, He X et al. Long non-coding RNA CASC2 regulates cell biological behaviour through the MAPK signalling pathway in hepatocellular carcinoma. Tumor Biol 2017; 39 (6): 1010428317706229. doi: 10.1177/1010428317706229.

30. Li P, Xue WJ, Feng Y et al. Long non-coding RNA CASC2 suppresses the proliferation of gastric cancer cells by regulating the MAPK signaling pathway. Am J Transl Res 2016; 8 (8): 3522–3529.

31. Gao R, Zhang R, Zhang C et al. Long noncoding RNA CCAT1 promotes cell proliferation and metastasis in human medulloblastoma via MAPK pathway. Tumori 2017: 0. doi: 10.5301/tj.5000662.

32. Graham LD, Pedersen SK, Brown GS et al. Colorectal neoplasia differentially expressed (CRNDE), a novel gene with elevated expression in colorectal adenomas and adenocarcinomas. Genes Cancer 2011; 2 (8): 829–840. doi: 10.1177/1947601911431081.

33. Ellis BC, Molloy PL, Graham LD. CRNDE: a long non-coding RNA involved in cancer, neurobiology, and development. Front Genet 2012; 3: 270. doi: 10.3389/fgene.2012.00270.

34. Zheng J, Li X, Wang P et al. CRNDE affects the malignant biological characteristics of human glioma stem cells by negatively regulating miR-186. Oncotarget 2015; 6 (28): 25339–25355. doi: 10.18632/oncotarget.4509.

35. Szafron LM, Balcerak A, Grzybowska EA et al. The putative oncogene, CRNDE, is a negative prognostic factor in ovarian cancer patients. Oncotarget 2015; 6 (41): 43897–43910. doi: 10.18632/oncotarget.6016.

36. Liu T, Zhang X, Yang Y et al. Increased expression of the long noncoding RNA CRNDE-h indicates a poor prognosis in colorectal cancer, and is positively correlated with IRX5 mRNA expression. Onco Targets Ther 2016; 9: 1437–1448. doi: 10.2147/OTT.S98268.

37. Jiang H, Wang Y, Ai M et al. Long noncoding RNA CRNDE stabilized by hnRNPUL2 accelerates cell proliferation and migration in colorectal carcinoma via activating Ras/MAPK signaling pathways. Cell Death Dis 2017; 8 (6): e2862. doi: 10.1038/cddis.2017.258.

38. Van Grembergen O, Bizet M, de Bony EJ et al. Portraying breast cancers with long noncoding RNAs. Sci Adv 2016; 2 (9): e 1600220. doi: 10.1126/sciadv.1600220.

39. Bao H, Guo CG, Qiu PC et al. Long non-coding RNA Igf2as controls hepatocellular carcinoma progression through the ERK/MAPK signaling pathway. Oncol Lett 2017; 14 (3): 2831–2837. doi: 10.3892/ol.2017. 6492.

40. Loewer S, Cabili MN, Guttman M et al. Large intergenic non-coding RNA-RoR modulates reprogramming of human induced pluripotent stem cells. Nat Genet 2010; 42 (12): 1113–1117. doi: 10.1038/ng.710.

41. Wang Y, Xu Z, Jiang J et al. Endogenous miRNA sponge lincRNA-RoR regulates Oct4, Nanog, and Sox2 in human embryonic stem cell self-renewal. Dev Cell 2013; 25 (1): 69–80. doi: 10.1016/j.devcel.2013.03.002.

42. Zhang A, Zhou N, Huang J et al. The human long non-coding RNA-RoR is a p53 repressor in response to DNA damage. Cell Res 2013; 23 (3): 340–350. doi: 10.1038/cr.2012.164.

43. Huang J, Zhang A, Ho TT et al. Linc-RoR promotes c-Myc expression through hnRNP I and AUF1. Nucleic Acids Res 2016; 44 (7): 3059–3069. doi: 10.1093/nar/gkv1353.

44. Eades G, Wolfson B, Zhang Y et al. LincRNA-RoR and miR-145 regulate invasion in triple-negative breast cancer via targeting ARF6. Mol Cancer Res 2015; 13 (2): 330–338. doi: 10.1158/1541-7786.MCR-14-0251.

45. Peng WX, Huang JG, Yang L et al. Linc-RoR promotes MAPK/ERK signaling and confers estrogen-independent growth of breast cancer. Mol Cancer 2017; 16 (1): 161. doi: 10.1186/s12943-017-0727-3.

46. Zhao XD, Liu YB, Yu S. Long noncoding RNA AWPPH promotes hepatocellular carcinoma progression through YBX1 and serves as a prognostic biomarker. Biochim Biophys Acta 2017; 1863 (7): 1805–1816. doi: 10.1016/j.bbadis.2017.04.014.

47. Wang AQ, Meng MZ, Zhao XH et al. Long non-coding RNA ENST00462717 suppresses the proliferation, survival, and migration by inhibiting MDM2/MAPK pathway in glioma. Biochem Biophys Res Commun 2017; 485 (2): 513–521. doi: 10.1016/j.bbrc.2017.02.015.

48. Chen L, Sun L, Dong L et al. The role of long noncoding RNA-LET in cell proliferation and invasion of nasopharyngeal carcinoma and its mechanism. Onco Targets Ther 2017; 10: 2769–2778. doi: 10.2147/OTT.S126907.

49. Ying L, Chen Q, Wang Y et al. Upregulated MALAT-1 contributes to bladder cancer cell migration by inducing epithelial-to-mesenchymal transition. Mol Biosyst 2012; 8 (9): 2289–2294. doi: 10.1039/c2mb25070e.

50. Wu XS, Wang XA, Wu WG et al. MALAT1 promotes the proliferation and metastasis of gallbladder cancer cells by activating the ERK/MAPK pathway. Cancer Biol Ther 2014; 15 (6): 806–814. doi: 10.4161/cbt.28584.

51. Gutschner T, Hämmerle M, Eißmann M et al. The noncoding RNA MALAT1 is a critical regulator of the metastasis phenotype of lung cancer cells. Cancer Res 2013; 73 (3): 1180–1189. doi: 10.1158/0008-5472.CAN-12-2850.

52. Yiu GK, Kaunisto A, Chin YR et al. NFAT promotes carcinoma invasive migration through glypican-6. Biochemical J 2011; 440 (1): 157–166. doi: 10.1042/BJ20110 530.

53. Merabova N, Kaminski R, Krynska B et al. JCV agnoprotein-induced reduction in CXCL5/LIX secretion by oligodendrocytes is associated with activation of apoptotic signaling in neurons. J Cell Physiol 2012; 227 (8): 3119–3127. doi: 10.1002/jcp.23065.

54. Chen L, Feng PM, Zhu X et al. Long non-coding RNA Malat1 promotes neurite outgrowth through activation of ERK/MAPK signalling pathway in N2a cells. J Cell Mol Med 2016; 20 (11): 2102–2110. doi: 10.1111/jcmm.12904.

55. Li JK, Chen C, Liu JY et al. Long noncoding RNA MRCCAT1 promotes metastasis of clear cell renal cell carcinoma via inhibiting NPR3 and activating p38-MAPK signaling. Mol Cancer 2017; 16 (1): 111. doi: 10.1186/s12943-017-0681-0.

56. Zhang X, Zhou YL, Mehta KR et al. A pituitary-derived MEG3 isoform functions as a growth suppressor in tumor cells. J Clin Endocrinol Metab 2003; 88 (11): 5119–5126. doi: 10.1210/jc.2003-030222.

57. Wang PJ, Ren ZQ, Sun PY. Overexpression of the long non-coding RNA MEG3 impairs in vitro glioma cell proliferation. J Cell Biochem 2012; 113 (6): 1868–1874. doi: 10.1002/jcb.24055.

58. Zhang X, Gejman R, Mahta A et al. Maternally expressed gene 3, an imprinted noncoding RNA gene, is associated with meningioma pathogenesis and progression. Cancer Res 2010; 70 (6): 2350–2358. doi: 10.1158/0008-5472.CAN-09-3885.

59. Zhu J, Liu S, Ye F et al. Long Noncoding RNA MEG3 Interacts with p53 protein and regulates partial p53 target genes in hepatoma cells. Plos One 2015; 10 (10): e0139790. doi: 10.1371/journal.pone.0139790.

60. Sun M, Xia R, Jin F et al. Downregulated long noncoding RNA MEG3 is associated with poor prognosis and promotes cell proliferation in gastric cancer. Tumour biology 2014; 35 (2): 1065–1073. doi: 10.1007/s13277-013-1142-z.

61. Zhang CY, Yu MS, Li X et al. Overexpression of long non-coding RNA MEG3 suppresses breast cancer cell proliferation, invasion, and angiogenesis through AKT pathway. Tumour Biol 2017; 39 (6): 1010428317701311. doi: 10.1177/1010428317701311.

62. Chen XJ, Dong H, Liu S et al. Long noncoding RNA MHENCR promotes melanoma progression via regulating miR-425/489-mediated PI3K-Akt pathway. Am J Transl Res 2017; 9 (1): 90–102.

63. Patel Y, Shah N, Lee JS et al. A novel double-negative feedback loop between miR-489 and the HER2-SHP2-MAPK signaling axis regulates breast cancer cell proliferation and tumor growth. Oncotarget 2016; 7 (14): 18295–18308. doi: 10.18632/oncotarget.7577.

64. Chen S, Wang Y, Zhang JH et al. Long non-coding RNA PTENP1 inhibits proliferation and migration of breast cancer cells via AKT and MAPK signaling pathways. Oncol Lett 2017; 14 (4): 4659–4662. doi: 10.3892/ol.2017.6823.

65. Goedert L, Pereira CG, Roszik J et al. RMEL3, a novel BRAFV600E-associated long noncoding RNA, is required for MAPK and PI3K signaling in melanoma. Oncotarget 2016; 7 (24): 36711–36718. doi: 10.18632/oncotarget. 9164.

66. Zhang EB, Yin DD, Sun M et al. P53-regulated long non-coding RNA TUG1 affects cell proliferation in human non-small cell lung cancer, partly through epigenetically regulating HOXB7 expression. Cell Death Dis 2014; 5: e1243. doi: 10.1038/cddis.2014.201.

67. Yang C, Li X, Wang Y et al. Long non-coding RNA UCA1 regulated cell cycle distribution via CREB through PI3-K dependent pathway in bladder carcinoma cells. Gene 2012; 496 (1): 8–16. doi: 10.1016/j.gene.2012.01.012.

68. Cargnello M, Roux PP. Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases (vol 75, pg 50, 2011). Microbiol Mol Biol Rev 2012; 76 (2): 496. doi: 10.1128/MMBR.00013-12.

69. Xu WH, Zhang JB, Dang Z et al. Long Non-coding RNA URHC regulates cell proliferation and apoptosis via ZAK through the ERK/MAPK signaling pathway in hepatocellular carcinoma. Int J Biol Sci 2014; 10 (7): 664–676. doi: 10.7150/ijbs.8232.

70. Taheri M, Omrani MD, Ghafouri-Fard S. Long non-coding RNAs expression in renal cell carcinoma. Journal of Biology and Today‘s World 2017; 6 (12): 240–247.

71. Faramarzi S, Dianatpour A, Ghafouri-Fard S. Discovering the role of long non-coding RNAs in regulation of steroid receptors signaling in cancer. Journal of Biology and Today‘s World 2017; 6 (12): 248–258.

72. Inamdar GS, Madhunapantula SV, Robertson GP. Targeting the MAPK pathway in melanoma: Why some approaches succeed and other fail. Biochem Pharmacol 2010; 80 (5): 624–637. doi: 10.1016/j.bcp.2010.04. 029.

73. Riddick G, Kotliarova S, Rodriguez V et al. A core regulatory circuit in glioblastoma stem cells links MAPK activation to a transcriptional program of neural stem cell identity. Sci Rep 2017; 7: 43605. doi: 10.1038/srep43 605.

74. Dong YQ, Liang GJ, Yuan B et al. MALAT1 promotes the proliferation and metastasis of osteosarcoma cells by activating the PI3K/Akt pathway. Tumour Biol 2015; 36 (3): 1477–1486. doi: 10.1007/s13277-014-2631-4.

75. Aksamitiene E, Kiyatkin A, Kholodenko BN. Cross-talk between mitogenic Ras/MAPK and survival PI3K/Akt pathways: a fine balance. Biochemical Soc Trans 2012; 40 (1): 139–146. doi: 10.1042/BST20110609.

76. Taylor AD, Micheel CM, Anderson IA et al. The path (way) less traveled: a pathway-oriented approach to providing information about precision cancer medicine on my cancer genome. Transl Oncol 2016; 9 (2): 163–165. doi: 10.1016/j.tranon.2016.03.001.

Štítky
Dětská onkologie Chirurgie všeobecná Onkologie

Článek vyšel v časopise

Klinická onkologie

Číslo 2

2018 Číslo 2
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

Svět praktické medicíny 1/2024 (znalostní test z časopisu)
nový kurz

Koncepce osteologické péče pro gynekology a praktické lékaře
Autoři: MUDr. František Šenk

Sekvenční léčba schizofrenie
Autoři: MUDr. Jana Hořínková

Hypertenze a hypercholesterolémie – synergický efekt léčby
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Význam metforminu pro „udržitelnou“ terapii diabetu
Autoři: prof. MUDr. Milan Kvapil, CSc., MBA

Všechny kurzy
Kurzy Podcasty Doporučená témata Časopisy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#