HOME JOURNALS CONTACT

International Journal of Pharmacology

Year: 2022 | Volume: 18 | Issue: 8 | Page No.: 1537-1549
DOI: 10.3923/ijp.2022.1537.1549
Long Non-Coding RNAs: A Double-Edged Sword in Renal Cell Carcinoma Carcinogenesis
Tian Liu and Hui Zhao

Abstract: Renal cell carcinoma (RCC) is a malignant tumor that results in high mortality and a huge economic burden worldwide. Owing to the paucity of therapy options, it still has a poor prognosis. Therefore, it is of great importance to find an effective strategy for the treatment of RCC. Long Non-Coding RNAs (lncRNAs) emerge as a pivotal regulator of diverse cancers in many biological processes, including proliferation metastasis, apoptosis and metabolism by tethering with DNA, RNA or protein. Recently, a strong body of investigations revealed that lncRNAs play a critical role in carcinogenesis and cancer progression. Hence, this review provides a synopsis of the molecular mechanisms and clinical implications of lncRNAs in the progression of RCC and found that lncRNAs have the potential to be used clinically in RCC in the future.

Fulltext PDF Fulltext HTML

How to cite this article
Tian Liu and Hui Zhao, 2022. Long Non-Coding RNAs: A Double-Edged Sword in Renal Cell Carcinoma Carcinogenesis. International Journal of Pharmacology, 18: 1537-1549.

Keywords: diagnosis, prognosis, therapy, long non-coding RNA and Renal cell carcinoma

INTRODUCTION

Renal cell carcinoma (RCC), which comprises 90% of kidney cancers, was ranked sixth for mortality worldwide and accounts for 2% of human malignancies1-4. The RCC occurs more frequently in the elderly and in men, which resulted in more than 140000 deaths yearly5,6. Von hippel-lindau (VHL) gene, BRCA1-associated protein 1 (BAP1), polybromo-1 (PBRM1) and SET domain-containing 2 (SETD2) mutations are the key drivers in the development of RCC and many risk factors may contribute to RCC, including smoking, hypertension obesity and chronic kidney disease7,8. The RCC mainly contains 3 subtypes: Clear cell RCC (ccRCC), papillary RCC (pRCC) and chromophobe RCC (chRCC) and ccRCC is most frequent followed by pRCC and chRCC9,10. Currently, most patients with RCC are diagnosed at advanced stages due to the high ratio of relapse and metastasis. In addition, RCC exhibited high chemoresistance and radioresistance, which brings large obstacles to the therapy of RCC11,12. Thus, it is of urgent importance to explore the precise molecular mechanism of RCC for early diagnosis and treatment.

Only about 1-2% of the total RNA could be translated into protein, while more than 90% of RNA, namely Non-coding RNAs (ncRNAs), is thought to have no capacity to encode protein13,14. Owing to lacking sufficient knowledge, ncRNAs were initially viewed as transcriptional noise and attracted little attention. However, upon further study, ncRNAs were confirmed to play an essential role in multiple human diseases15-17. The ncRNAs longer than 200 nucleotides are called Long Non-coding RNAs (lncRNAs) that have been associated with various functions18. In the last decade, accumulating evidence has shown that several lncRNAs have emerged as pivotal regulators in multiple biological processes, such as cell growth and differentiation, tumorigenesis and inflammation19,20. Until now, lncRNAs found to be dysregulated in multiple cancers. A growing number of studies revealed that lncRNAs play an essential role in promoting the tumorigenesis and progression of human cancers, including RCC and served as regulators in many cellular behaviours, including proliferation, invasion, migration, as well as apoptosis21,22. Emerging evidence also indicated that aberrant expression lncRNAs are significantly associated with the diagnosis and prognosis of RCC23,24. Furthermore, they may have the potential to be therapeutic targets for RCC. In this review, we summarized the roles of lncRNAs in tumor initiation and progression of RCC and highlighted the possible utilization of lncRNAs for the diagnosis and therapy in RCC.

Molecular mechanisms of lncRNAs: In the past few decades, researchers have conducted considerable work and in-depth studies to clarify the molecular mechanisms of lncRNAs. A strong body of evidence indicated that lncRNAs regulate gene expression mainly at three levels namely epigenetic, transcriptional and post-transcriptional regulation. (a) Epigenetic level: Transcription process can be regulated by recruiting chromatin remodelling/modification complexes to specific sites and affect the chromosome structure, histone modification and DNA methylation (b) Transcriptional level: Affecting the process of gene transcription by binding with RNA polymerase II, pre-initiation complex and transcription factors and (c) Post-transcriptional level: Regulating mRNA processing in multiple aspects such as mRNA selective splicing, degradation or translation24,25.

Additionally, lncRNAs can sequester miRNA away from their target mRNA via interacting with miRNAs by acting as competing endogenous RNA (ceRNA) or RNA sponges. lncRNAs and mRNAs can interact by competing for miRNA binding, thus affecting their respective expression levels26. Relevant studies also showed that lncRNAs can bind to specific RNA or DNA sequences by base-pairing interactions and fold into advanced structures that regulate their interaction with proteins27. Moreover, lncRNAs could exert functions in both cytoplasm and nucleus via various mechanisms including the regulation of chromosome architecture and genomic regions, interaction with chromatin modification complexes, transcriptional enhancers activation, nuclear domains conformation, the transcriptional machinery interference and nuclear bodies’ structural formation and maintenance (Fig. 1)28. Collectively, increasing evidence demonstrated that lncRNAs exert an irreplaceable role in different diseases. The regulatory mechanisms of lncRNAs still need further investigation and conformation to enable them as novel targets for the diagnosis and treatment of diseases in the clinic.

lncRNAs in RCC: Recently, lncRNAs were confirmed as new players in human cancers. Accumulating evidence has demonstrated that lncRNAs are dysregulated and affect the progression of many malignant tumors, including RCC19. Many studies indicated that lncRNAs might be potential biomarkers of diagnosis and prognosis, targets for molecular targeted therapy and serve as new budding stars in RCC. Here, we illuminated the functions and mechanisms of some lncRNAs in the development of RCC to provide a good understanding of their roles in RCC.

Fig. 1: Molecular mechanisms of long non-coding RNAs
lncRNAs regulate gene expression in three ways, namely epigenetic regulation, transcriptional regulation and post-transcriptional regulation28

lncRNAs as oncogenes in RCC
MALAT: With more than 8000 nucleotides, the Metastasis-Associated Lung Adenocarcinoma Transcript 1 (MALAT1) is located on chromosome 11q13 and was confirmed to serve as a critical participant in multiple cancers29. Significantly, MALAT1 was upregulated in the RCC tissues and cells (A-498 and 786-O cells) compared to normal renal tissues30,31. The MALAT1 promoted the proliferation and migration of Caki-1 cells by targeting miR-22-3p via the phosphatidylinositide 3-kinase/protein kinase B (PI3K/Akt) signalling pathway. Similarly, overexpression of MALAT1 can promote the progression of RCC by the regulation of miR-203/baculoviral inhibitor of apoptosis repeat containing 5 (BIRC5) axis30,32. Other related studies indicated that MALAT1 deficiency inhibited the proliferation, invasion and migration of RCC cells by sponging miR-429 or targeting miR-200s/zinc finger E-box binding homeobox 2 (ZEB2) axis33,34. Moreover, MALAT1 was confirmed to interact with miR-205 and was activated by the c-Fos that interacted with the enhancer of zeste homolog 2 (EZH2), which is a PRC2 subunit. Knockdown of MALAT1 blocked epithelial-mesenchymal transition (EMT) in the progression of RCC, as indicated by the upregulation of E-cadherin and reduction of β-catenin via EZH231. Of note, knockdown of MALAT1 can also suppress sunitinib chemoresistance by downregulating miR-362-3p-mediated RasGAP SH3-domain-Binding Protein1 (G3BP1) expression, indicating that MALAT1 might serve as a therapeutic target for sunitinib-resistance RCC35. In conclusion, these findings indicated the in-depth role of MALAT1 in RCC carcinogenesis, suggesting a detailed mechanism in RCC progression (Fig. 2).

HOTAIR: The HOX Antisense intergenic RNA (HOTAIR), which is located on chromosome 12q13.13 with 2158 nucleotides and 6 exons, represents an important oncogene in various human cancers and is presented as a potential prognostic biomarker36,37. Notably, HOTAIR was upregulated in RCC cell lines (ACHN and Caki-1 cells) and tissues. Overexpression of miR-203 or HOTAIR inhibition repressed the proliferation, invasion and migration while promoting apoptosis of ACHN and Caki-1 cells. In contrast, HOTAIR deficiency inhibited the expression of E-cadherin, claudin, phosphatase and tensin homolog (PTEN), p21 and p27, indicating that HOTAIR suppression inhibited EMT and tumor metastasis38.

Fig. 2: lncRNA MALAT1 is an essential oncogene in the progression of renal cell carcinoma
lncRNA MALAT1 can affect the many processes of RCC, including cell proliferation, invasion, motility, viability, apoptosis and sunitinib resistance through targeting various pathways30,32-35

Moreover, HOTAIR promoted the progression of RCC by sponging miR-124 through α-2, 8-sialyltransferase 4. Knockdown of HOTAIR, a target of miR-217, inhibited tumorigenesis by downregulating the expression of Hypoxia-Inducible Factor-1α (HIF-1α) and anexelekto (AXL). Particularly, overexpression of HOTTIP had a significantly shorter overall survival (OS) and disease-free survival (DFS), suggesting that HOTTIP may serve as a prognostic predictor of RCC39,40. In addition, knockdown of HOTAIR by siRNA can also inhibit the proliferation, migration and invasion of A-498 and OS-RC-2 cells with a cell cycle arrest in the G0/G1 phase and with the reduced number of cells in the G2/M phase. Meanwhile, HOTAIR deficiency promoted the expression of p53, p21 and p16 at both mRNA and protein levels, whereas, downregulated the level of EZH2 protein with lower recruitment and binding abilities41. These studies indicated that HOTAIR was involved in the tumorigenesis of RCC and exhibited oncogenic properties, providing a novel therapeutic target for the treatment of RCC.

UCA1: The Urothelial carcinoma associated 1 (UCA1), which is located on chromosome 19, serves as an oncogene in many tumors and was firstly discovered in bladder cancer42,43. The expression of UCA1 was significantly upregulated in RCC tissues and cells (769-P, 786-O, ACHN, A498 and Caki2 cells). It has been shown to promote the proliferation and invasion of RCC cells, whereas, inhibited cell apoptosis by acting as a ceRNA of miR-129 to upregulate the level of SRY-box transcription factor 4 (SOX4) in RCC44. Furthermore, lncRNA UCA1 has been found to play a carcinogenic role in RCC. The expression of UCA1 promoted cell proliferation by inhibiting the expression of miR-495 and p21 in 786-O and ACHN cells. Overexpression of UCA1 was intimately associated with the advanced tumor-node-metastasis (TNM) stage and led to a shorter OS time45. These findings demonstrated that overexpression of UCA1 contributes to the progression and promotion of RCC and may serve as a potential molecular predictor for poor prognosis in RCC patients.

TUG1: With 4 exons, the Taurine Upregulated Gene 1 (TUG1) is located on chromosome 22q12 and exerts important regulatory functions in multiple types of human cancers46-48. In RCC tissues and cells (ACHN and OS-RC-2 cells), TUG1 was significantly increased. A new study implied that knockdown of TUG1 inhibited RCC formation by sponging miR-299-3P via downregulating Vascular Endothelial Growth Factor (VEGF) expression49. The expression of TUG1 can promote the proliferation, migration and invasion of RCC 786-O cells while inhibiting cell apoptosis by downregulating miR-196a. Meanwhile, TUG1 can promote the progression of RCC by regulating miR-9/yes-associated protein (YAP) axis50-52. In addition, TUG1 inhibited apoptosis and autophagy, whilst promoting the proliferation of RCC 786-0 and A498 cells by upregulating flotillin 1 (FLOT1) expression via sponging miR-31-5p53.

CYTOR: The Cytoskeleton Regulator (CYTOR), which consists of 828 bp, is located on chromosome 2p11.2. The CYTOR was firstly discovered in hepatic carcinoma and acts as a critical oncogene in various human cancers54. The CYTOR deficiency retarded the proliferation of RCC 786-O and Caki-1 cells while facilitating cell apoptosis by repressing miR-136-5p to regulate methionine adenosyltransferase (MAT) 2B. The MAT2B was demonstrated to affect RCC progression by interacting with Bcl2-associated athanogene 3 (BAG3), In addition, knockdown of CYTOR significantly inhibited the tumor growth in 786-O xenograft nude mice. Thus, CYTOR may act as a potential therapeutic target for RCC55.

AFAP1-AS1: Located on chromosome 4p16.1, the actin filament-associated protein 1-antisense RNA 1 (AFAP1-AS1) consists of 6810 nucleotides that are involved in many malignant tumors56. The upregulation of AFAP1-AS1 was observed in RCC tissues and cell lines (786-O, Caki-1, ACHN and A498 cells). In RCC patients, overexpression of AFAP1-AS1 had a poorer OS and was closely related to the TNM stage and lymph node metastasis of RCC patients. Knockdown of AFAP1-AS1 inhibited the proliferation, invasion, migration and EMT in Caki-1 and ACHN cells and inhibited tumor growth via PTEN/AKT signalling57. These results demonstrated that AFAP1-AS1 exhibits oncogenic properties and provides a potential therapeutic target for targeted interventional therapy of RCC.

HOTTIP: The HOXA Transcript at the Distal Tip (HOTTIP), which is located on the chromosomal locus 7p15.2, is transcribed from the HOXA cluster58. Upregulation of HOTTIP has been shown to play a critical role in various human cancers via epigenetic modifications, lncRNA-miRNA and lncRNA-protein interactions59. The HOTTIP deficiency inhibited the proliferation, migration and invasion by promoting autophagy through the PI3K/Akt/Atg13 signalling pathway and induced apoptosis by epigenetically downregulating large tumor suppressor 2 (LATS2) via interacting with EZH2 and Lysine-Specific Demethylase 1 (LSD1) in RCC ACHN and OSRC-2 cells. Moreover, HOTTIP overexpression led to larger tumor size and advanced TNM stage60,61. In addition, by acting as a ceRNA of miR-615, HOTTIP can increase the expression of IGF-2, leading to the promotion of RCC malignancy. Overexpression of HOTTIP had significant statistical relevance to the undesirable OS and DFS in RCC62.

H19: The lncRNA H19, which is expressed during embryonic progression and then is inhibited after birth, is transcribed from H19/Insulin-like Growth Factor 2 (IGF2) that is located on chromosome 11p15.563. The lncRNA H19 was over-expressed in RCC tissues and cells (768-O, ACHN and Caki-1 cells). Upregulation of lncRNA H19 inhibited the expression of miR-29a-3p by targeting E2F transcription factor 1 (E2F1) and led to a shorter OS64,65. In addition, knockdown of lncRNA H19 repressed the migration and invasion of RCC 786-O cells, whereas E2F1 overexpression reversed these effects65. Moreover, lncRNA H19 was significantly associated with the susceptibility and mortality of RCC66. These studies indicated that lncRNA H19 may be a genetic predictor for the prognosis of RCC and exert its function by regulating the miR-29a-3p/E2F1 axis.

LINC00641: Located on chromosome 14q11.2, the long intergenic non-coding RNA 00641 (LINC00641) is usually at a low level in a normal state and acts as both an oncogene and tumor suppressor in different types of cancers67,68. It was reported that LINC00641 was upregulated in RCC tissues and cell lines (GRC-1, 786-O, SN12-PM6, A498 and ACHN cells). The proliferation and invasion were inhibited, while apoptosis was promoted in RCC A498 and ACHN cells when LINC00641 was knocked down. Moreover, LINC00641 deficiency suppressed tumorigenesis in the xenograft tumor model. Mechanistically, LINC00641 affected the progression of RCC via sponging miR-340-5p68. In addition, overexpression of LINC00641 was closed associated with TNM stage and poor prognosis68.

PCED1B-AS1: The lncRNA PC-Esterase Domain Containing 1B Antisense RNA 1 (PCED1B-AS1) is implicated in multiple human malignant tumors, including hepatocellular carcinoma, glioblastoma and pancreatic cancer69-71. In a recent study, the significant upregulation of PCED1B-AS1 was observed in RCC tissues compared to adjacent normal renal tissue72. Knockdown of PCED1B-AS1 by siRNA promoted the migration, proliferation and EMT of A498 and Caki-1 cells by regulating the miR-484/zinc finger E-box-binding homeobox 1 (ZEB1) axis. In addition, PCED1B-AS1 might be a potential prognostic diagnostic marker and high expression of PCED1B-AS1 resulted in a shorter OS time, higher tumor stage and Fuhrman grade in RCC patients72. These findings demonstrated that overexpression of PCED1B-AS1 contributes to the promotion and progression of RCC.

SNHG1: Small nucleolar RNA host gene 1 (SNHG1) is a newly identified lncRNA, which is located at 11q12.3 with 11 exons73. The expression of SNHG1 was increased in RCC tissues and cell lines (ACHN, A498 and Caki 1)74. Knockdown of SNHG1 retarded proliferation and invasion of RCC Caki 1 cells by promoting the expression of miR 103a through targeting HMGA274. Furthermore, SNHG1 knockdown inhibited the immune escape ability of RCC A498 and 786-O cells by targeting miR 129 3p to downregulate STAT3 and PD L1. SNHG1 deficiency can also suppress EMT in RCC and RCC patients with high SNHG1 levels had a shorter OS than those with low SNHG1 levels75,76.

SNHG5: With six exons, the small nucleolar RNA host gene 5 (SNHG5), which is localized on chromosome 6q14.3 and consists of 524 bp, was firstly identified at a chromosomal translocation breakpoint77. The SNHG5 was recently confirmed as an aberrantly upregulated lncRNA in RCC tissues. High expression of SNHG5 was positively associated with TNM stage, tumor size, lymphatic invasion and distant metastasis78. On the one hand, knockdown of SNHG5 significantly inhibited the proliferation, invasion and migration of ACHN cells by acting as a ceRNA for miR-205-5p via targeting ZEB1 in RCC cells. On the other hand, overexpression of SNHG5 promoted tumorigenesis and metastasis in vivo78. Additionally, lncRNA SNHG5 deficiency inhibited the invasion and induced apoptosis in 786-O cells by promoting the expression of miR-363-3p. Inhibition of twist, a direct target of miR-363-3p, inhibited tumor metastasis and promoted apoptosis of 786-O cells by decreasing matrix metalloproteinase-9 (MMP-9) and MMP-2 at protein level79.

MIR4435-2HG: The lncRNA MIR4435-2 host gene (MIR4435-2HG) is located on chromosome 2q13 and was proved as carcinogenic lncRNA in a broad range of tumors80. Of note, MIR4435-2HG was found to be increased in RCC tissues and 769-P cells81. The MIR4435-2HG boosted the proliferation and invasion of 769-P cells by inhibiting the expression of miR-513a-5p that directly targeted Kruppel-like factor 6 (KLF6). Reciprocally, in vivo experiment further demonstrated that knockdown of MIR4435-2HG led to smaller tumor size and less tumor formation and these effects can be reversed by KLF6 overexpression81.

SNHG12: The Small nucleolar host gene 12 (SNHG12), which is located at chromosome 1p35.3, was discovered to be dysregulated in a series of cancers as a newly discovered lncRNA82,83. Recently, the level of SNHG12 was confirmed to be notably upregulated in the tissues of RCC patients and cell lines (A498 and 786-O cells)84. Overexpression of SNHG12 was associated with TNM stage, lymph node and distant metastases and resulted in a poor prognosis. In addition, SNHG12 prompted cell viability and invasion, whereas inhibited the apoptosis of RCC cells by decreasing the expression of miR-200c-5p by targeting collagen type XI α1 chain (COL11A1)84. These results indicated that SNHG12 exerts its oncogenic role through miR-200c-5p/COL11A1 axis.

CASC19: The long non-coding RNA cancer susceptibility 19 (CASC19) is located on the 8q24 region of the chromosome and is an important participant in the development of human cancers, such as gastric cancer, nasopharyngeal carcinoma and non-small cell lung cancer85,86. Recently, overexpression of CASC19 was observed in RCC tissues and cell lines (786-O and A498 cells)86. High expression of CASC19 resulted in larger tumor sizes, advanced TNM stage and a poor prognosis. Knockdown of CASC19 attenuated the proliferation, migration and invasion of RCC cells by sponging miR-532 via regulating ETS proto-oncogene 1 (ETS1) expression, indicating that CASC19 affecting the progression through miR-532/ETS1 Axis. In addition, further in vivo studies showed that interference of CASC19 inhibited tumor growth in A498 xenograft nude mice86. Collectively, these results indicated that CASC19/miR-532/ETS1 pathway might be used to monitor prognosis and provides potential therapeutic targets for the clinical management of RCC (Table 1).

lncRNAs as tumor suppressors in RCC
MEG3: The Maternally Expressed Gene 3 (MEG3) is located on chromosome 14q32.3 and is also known as Gene trap locus 2 (Gtl2)87. It was reported that the expression of MEG3 is downregulated in various cancers, including RCC and acts as a tumor suppressor88,89. The MEG3 promoted cell apoptosis via inhibiting miR-7 while inhibiting the proliferation, invasion and migration of A-498 cells by upregulating the expression of RAS like family 11 member B (RASL11B), which is the target of miR-7. Moreover, MEG3 induced the G0/G1 cell cycle arrest in RCC A-498 cells89. This study demonstrated that MEG3 exert an anti-cancer function in RCC by regulating miR-7/RASL11B signalling.

Table 1: Oncogenic lncRNAs in RCC
lncRNAs Changes Cell lines Location Targets/signaling Functions Mechanisms References
MALAT1 Up A-498 and 786-O Chromosome 11q13 miR-203, BIRC5, miR-205, miR-22-3p, PI3K/Akt signaling, miR-429, miR-200s ZEB2, miR-362-3p, G3BP1 Oncogene Proliferation, invasion, migration, EMT and sunitinib chemoresistance Li et al.30, Hirata et al.31, Zhang et al.32, Jiang et al.33 and Wang et al.35
HOTAIR Up ACHN and Caki-1 Chromosome 12q13.13 miR-203, PTEN, P21, P27 miR-124, HIF-1 α, AXL, p53, p21, p16 and EZH2 Oncogene Proliferation, invasion migration and apoptosis, EMT metastasis Dasgupta et al.38, Pan et al.39, Hong et al.40 and Wu et al.41
UCA1 Up 769P, 786O, ACHN, A498 and Caki2 Chromosome 19 miR-129, SOX4, miR-495, p21 Oncogene Proliferation, invasion and apoptosis Liu et al.44 and Lu et al.45
TUG1 Up ACHN and OS-RC-2 Chromosome 22q12 miR-299-3p, VEGF, miR-196a, miR-9, YAP, miR-31-5p, FLOT1 Oncogene Proliferation, migration, invasion, apoptosis and autophagy Li et al.49, Liu et al.50, Yang et al.51, Zhang et al.52 and Lv et al.53
CYTOR Up Caki-1, A498, ACHN and 7860 Chromosome 2p11.2 miR-136-5p, MAT2B, BAG3 Oncogene Proliferation and apoptosis Wang et al.55
AFAP1-AS1 Up 786-O and Caki-1 Chromosome 4p16.1 PTEN/AKT Oncogene Proliferation, invasion, migration and EMT Mu et al.57
HOTTIP Up 786-O, ACHN and OSRC-2 Chromosome 7p15.2 PI3K/Akt/Atg13, LATS2, EZH2, LSD1, miR-615, IGF-2 Oncogene Proliferation, invasion, migration, apoptosis and autophagy Su et al.60, Peng et al.61 and Wang et al.62
H19 Up 768-O, ACHN and Caki-1 Chromosome 11p15.5 miR-29a-3p, E2F1 Oncogene Migration and invasion Wang et al.64, He et al.65 and Cao et al.66
LINC00641 Up ACHN, 786-O, Caki-1 and Caki-2 Chromosome 2p11.2 miR-340-5p Oncogene Proliferation, invasion and apoptosis Zhang et al.68
PCED1B-AS1 Up A498 and Caki-1 Chromosome 12 miR-484/ZEB1 Oncogene Proliferation, migration and EMT Qin et al.72
SNHG1 Up ACHN, A498 and Caki 1 Chromosome 11q12.3 miR-103a, HMGA2, miR-129-3p, STAT3, PD-L1 Oncogene Proliferation, invasion, immune escape and EMT Ye et al.74 and Tian et al.75
SNHG5 Up ACHN Chromosome 6q14.3 miRNA-205-5p, ZEB1, miR-363-3p/twist, MMP-9, MMP-2 Oncogene Proliferation, invasion, migration, apoptosis and metastasis Xiang et al.78 and Li et al.79
MIR4435-2HG Up 769-P Chromosome 2q13 miR-513a-5p/KLF6 Oncogene Proliferation and invasion Zhu et al.81
SNHG12 Up A498, 768-O Chromosome 1p35.3 miR-200c-5p/COL 11A1 Oncogene Viability, invasion and apoptosis Xu et al.84
CASC19 Up 786-O and A498 Chromosome 8q24 miR-532/ETS1 Oncogene Proliferation, migration and invasion Luo et al.86


Table 2: Tumour suppressive lncRNAs in RCC
lncRNAs Changes Cell lines Location Targets/signaling Functions Mechanisms References
MEG3 Down A-498 and 786-O Chromosome 14q32.3 miR-7/RASL11B Tumour suppressor Proliferation, invasion and migration Ghafouri-Fard et al.88 and He et al.89
LINC01510 Down 786-O and Caki-2 Chromosome 7 β-catenin Tumour suppressor Proliferation and invasion Ma et al.90
LINC01939 Down ACHN and CAKI-1 Chromosome 2 miR-154, Wnt/β catenin Tumour suppressor Proliferation, migration and apoptosis Zhang et al.92
ZNF710-AS1-202 Down ACHN and 786-O - ZNF710 Tumour suppressor Proliferation, survival and apoptosis Li et al.93
ASB16-AS1 Down 786-O Chromosome 17q21 miR-185-5p/miR-214-3p-LARP1 Tumour suppressor Proliferation, migration, invasion, EMT and metastasis Li et al.94

LINC01510: In malignant RCC tissues, the expression of the LINC01510 was observed to be decreased90. Overexpression of LINC01510 declined the level of cyclin D1 (CCND1), cyclin E1 (CCNE1) and matrix metalloproteinases (MMPs) to affect the cell cycle and invasion of RCC 786-O and Caki-2 cells. In addition, ectopic expression of LINC01510 suppressed proliferation of RCC 786-O cells and attenuated the ability of invasion by boosting the expression of β-catenin, indicating that LINC01510 exerted tumor suppressive effect on RCC cell lines through Wnt/β-catenin signalling pathway90.

LINC01939: The long intergenic non-coding RNA 1939 (LINC01939) is a novel lncRNA and was first confirmed to be downregulated in gastric cancer and involved in cancer progression91. Recently, the latest study indicated that LINC01939 possessed decreased expression in RCC tissues92. Upregulation of LINC01939 inhibited the proliferation and migration, as well as induced the apoptosis of ACHN and CAKI-1 cells by decreasing the level of miR-154 with the bluntness of Wnt/β catenin and Notch92.

ZNF710-AS1-202: The ZNF710-AS1-202 is a newly-found lncRNA, which was first unveiled to be a tumor suppressor in RCC and was significantly declined in RCC tissues93. The expression of ZNF710-AS1-202 was associated with pathological grade, tumor size, local invasion and TNM stage. Moreover, RCC patients with high levels of ZNF710-AS1-202 had longer OS time and DFS than those with low ZNF710-AS1-202 levels, as measured by cancer genome atlas (TCGA) analysis93. The increase in ZNF710-AS1-202 promoted the proliferation and survival, as well as inhibited apoptosis of ACHN and 786-O cells by decreasing the expression of ZNF710 mRNA and increasing the expression of ZNF710 protein93.

ASB16-AS1: The lncRNA ASB16 antisense RNA 1 (ASB16-AS1) is a 2275-bp-long lncRNA encoded on chromosome 17q21 and was downregulated in RCC tissues compared to non-tumorous renal tissue94. The ASB16-AS1 deficiency promoted the proliferation, migration and invasion, inhibited EMT phenotype in 786-O cells and enhanced tumor growth and metastasis in 786-O xenograft mice by decreasing La-related protein 1 (LARP1) expression via sponging miR-185-5p and miR-214-3p. Moreover, low-level expression of ASB16-AS1 was associated with larger tumors and advanced tumor stage94. These findings suggested that ASB16-AS1 exerts its tumor-suppressive function in the progression of RCC through miR-185-5p/miR-214-3p-LARP1 pathway (Table 2).

lncRNAs act as diagnostic biomarkers in RCC: At the present stage, many RCC patients are diagnosed at the late stage when effective treatments cannot be applied. Approximately, one-third of RCC patients have been detected with metastatic RCC11. Therefore, potential novel biomarkers are needed to improve the early diagnosing rate of RCC. Evaluating the circulating level of lncRNAs in bodily fluids would be more practical and convenient than those in tissues for tumor diagnosis. Nowadays, more and more studies indicated that serum lncRNAs might be new diagnostic biomarkers in RCC. For example, the serum gradually increased during hepatocarcinogenesis (GIHCG) was significantly increased in RCC patients and can distinguish RCC patients from healthy controls with an AUC of 0.920 in the testing set95. Similarly, another study indicated that serum C-X-C motif chemokine ligand 2 (CXCL2) level was markedly higher than that in healthy subjects. Further ROC curve analysis showed that serum CXCL2 might be a potential diagnostic predictor for RCC and the AUC value was 0.86896. In addition, the level of LINC00887 was also increased in the serum of RCC patients compared with healthy volunteers with an AUC of 0.80397. These findings demonstrated that the signature of serum lncRNAs has the potential to be biomarkers for the early diagnosis of RCC patients. However, the low specificity or sensitivity of these serum diagnostic biomarkers and the small sample size for RCC screening have limited their clinical practicability. To date, there is no recognized lncRNA for the diagnosis of RCC. Consequently, further in-depth exploration of the clinical diagnostic value of lncRNA is necessary in the future study.

Promise and challenges of lncRNAs therapy in RCC: The lncRNAs have served as prospective candidates for RCC therapy and many studies performed in RCC cell lines and RCC cell xenograft mice model have revealed the potential targeting lncRNAs to affect the progression of RCC, which provides a strong basis for the treatment of RCC. However, lncRNAs in RCC are yet in their infancy and there is no sufficient evidence to confirm the effects of lncRNAs in RCC patients. For example, the level of lncRNA activated in RCC with sunitinib resistance (lncARSR) in plasma and tumor tissues was positively correlated with the drug resistance of RCC patients during sunitinib therapy98,99, but the sample size was small and still warrants further investigation. In addition, several critical issues need to be solved before lncRNAs can be used for RCC therapy. Firstly, whether the therapeutic capabilities of lncRNAs in animal models hold for humans remains obscure as lncRNAs are highly conserved among different species, thus limiting the exploration of the mechanism of lncRNAs in RCC100. Secondly, multiple studies have demonstrated that lncRNAs exert their regulatory functions in various biological processes by acting as ceRNAs. However, compared with highly expressed miRNAs, the expression level of most lncRNAs is low in both cytoplasm and nucleus. The sponge function is arduous to exert because miRNAs harbour only one putative binding site for each lncRNA.

CONCLUSION

Over the past few decades, an increasing number of lncRNAs have been confirmed to play essential roles in the pathogenesis of RCC. The lncRNAs can inhibit the proliferation, invasion and migration and induce the apoptosis of RCC cell lines by acting on various molecular targets. Moreover, the expressions of many lncRNAs are closely associated with the clinic’s pathological characteristics and prognosis. Emerging studies have defined lncRNAs as novel biomarkers of diagnosis and prognosis and potential therapeutic targets for RCC. Nevertheless, what we have done is far from enough. The clinical utilities of lncRNAs remain unclear and there is still a large obstacle before lncRNAs can be used clinically for the treatment of RCC. Therefore, the precise molecular mechanisms of lncRNAs in RCC are still worth further exploration. Although many challenges remain to be addressed, we anticipate that lncRNAs have a broader prospect as a viable candidate therapeutic targets in RCC.

SIGNIFICANCE STATEMENT

Renal cell carcinoma (RCC) is ranked sixth of all cancer deaths for morbidity and mortality worldwide. Owing to the paucity of effective diagnostic and treatment options, it is necessary to find and develop new strategies for the treatment of lung cancer. Long Non-Coding RNAs (lncRNAs) act as a critical regulatory factor of diverse cancers in many cellular regulatory processes, including proliferation metastasis, apoptosis and metabolism. In the past few decades, an increasing number of investigations indicated that lncRNAs play an important role in the pathogenesis and development of various cancers. The current review discusses the molecular mechanisms and clinical implications of lncRNAs in the progression of RCC and found that lncRNAs may be novel therapeutic targets for RCC.

ACKNOWLEDGMENT

This study was supported by the National Science Basic Research Program of Shaanxi, Basic Research Program of Xi'an Municipal Health Commission (No. 2022JQ-920, 2022yb41 and 2021JQ-938).

REFERENCES

  • Ochocki, J.D., S. Khare, M. Hess, D. Ackerman and B. Qiu et al., 2018. Arginase 2 suppresses renal carcinoma progression via biosynthetic cofactor pyridoxal phosphate depletion and increased polyamine toxicity. Cell Metab., 27: 1263-1280.
    CrossRef    Direct Link    


  • Choueiri, T.K. and R.J. Motzer, 2017. Systemic therapy for metastatic renal-cell carcinoma. N. Engl. J. Med., 376: 354-366.
    CrossRef    Direct Link    


  • Siegel, R.L., K.D. Miller, H.E. Fuchs and A. Jemal, 2021. Cancer statistics, 2021. CA: A Cancer J. Clinicians, 71: 7-33.
    CrossRef    Direct Link    


  • Sung, H., J. Ferlay, R.L. Siegel, M. Laversanne, I. Soerjomataram, A. Jemal and F. Bray, 2021. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: Cancer J. Clinicians, 71: 209-249.
    CrossRef    Direct Link    


  • Ruhle, A., N. Andratschke, S. Siva and M. Guckenberger, 2019. Is there a role for stereotactic radiotherapy in the treatment of renal cell carcinoma? Clin. Transl. Radiat. Oncol., 18: 104-112.
    CrossRef    Direct Link    


  • Zhao, H., L. Chen, T. Yang, Y.L. Feng and N.D. Vaziri et al., 2019. Aryl hydrocarbon receptor activation mediates kidney disease and renal cell carcinoma. J. Transl. Med., Vol. 17.
    CrossRef    


  • Chanza, N.M., A. Tripathi and L.C. Harshman, 2019. Adjuvant therapy options in renal cell carcinoma: Where do we stand? Curr. Treat. Options Oncol., Vol. 20.
    CrossRef    


  • Capitanio, U., K. Bensalah, A. Bex, S.A. Boorjian and F. Bray et al., 2019. Epidemiology of renal cell carcinoma. Eur. Urol., 75: 74-84.
    CrossRef    Direct Link    


  • Linehan, W.M., 2012. Genetic basis of kidney cancer: Role of genomics for the development of disease-based therapeutics. Genome Res., 22: 2089-2100.
    CrossRef    Direct Link    


  • Jiang, C., S. Zhao, B. Yang, T. Tang, R. Hou, X. Liu and H. Zhao, 2018. Rare ectopic metastasis from clear cell renal cell carcinoma to the chest wall after 10 years: A case report. Medicine, Vol. 97.
    CrossRef    


  • Ding, L., M. Jiang, R. Wang, D. Shen and H. Wang et al., 2021. The emerging role of small non-coding RNA in renal cell carcinoma. Transl. Oncol., Vol. 14.
    CrossRef    


  • Liu, T., L. Chen and G. Liu, 2021. A commentary on “Prevalence of benign pathology after partial nephrectomy for suspected renal tumor: A systematic review and meta-analysis” (Int J Surg 2020; 84: 161-170). Int. J. Surg., Vol. 91.
    CrossRef    


  • Ekine-Afolabi, B.A., A.A. Njan, S.O. Rotimi, R.I. Anu, A.M. Elbehi, E. Cash and A. Adeyeye, 2020. The impact of diet on the involvement of non-coding RNAs, extracellular vesicles, and gut microbiome-virome in colorectal cancer initiation and progression. Front. Oncol., Vol. 10.
    CrossRef    


  • Zhao, H., Y.L. Feng, T. Liu, J.J. Wang and J. Yu, 2021. MicroRNAs in organ fibrosis: From molecular mechanisms to potential therapeutic targets. Pathol. Res. Pract., Vol. 225.
    CrossRef    


  • Liu, J., J. Wu, L. Li, T. Li and J. Wang, 2020. The role of exosomal non-coding RNAs in coronary artery disease. Front. Pharmacol., Vol. 11.
    CrossRef    


  • Zhao, H., S.X. Ma, Y.Q. Shang, H.Q. Zhang and W. Su, 2019. microRNAs in chronic kidney disease. Clin. Chim. Acta, 491: 59-65.
    CrossRef    Direct Link    


  • Liu, T., Y. Pei, C. Li and M. Ye, 2019. Amount of escape estimation based on Bayesian and MCMC approaches for RNA interference. Mol. Ther. Nucleic Acids, 18: 893-902.
    CrossRef    Direct Link    


  • Zhang, X.Z., H. Liu and S.R. Chen, 2020. Mechanisms of long non-coding RNAs in cancers and their dynamic regulations. Cancers, Vol. 12.
    CrossRef    


  • Da, M., H. Jiang, Y. Xie, W. Jin and S. Han, 2021. The biological roles of exosomal long non-coding RNAs in cancers. OncoTargets Ther., 14: 271-287.
    CrossRef    Direct Link    


  • Zhao, H., Y.L. Feng, M. Wang, J.J. Wang, T. Liu and J. Yu, 2022. The Angelica dahurica: A review of traditional uses, phytochemistry and pharmacology. Front. Pharmacol., Vol. 13.
    CrossRef    


  • Zhang, H.D., L.H. Jiang, S.L. Zhong, J. Li and D.W. Sun et al., 2021. The role of long non-coding RNAs in drug resistance of cancer. Clin. Genet., 99: 84-92.
    CrossRef    Direct Link    


  • Arun, G., S.D. Diermeier and D.L. Spector, 2018. Therapeutic targeting of long non-coding RNAs in cancer. Trends Mol. Med., 24: 257-277.
    CrossRef    Direct Link    


  • Seles, M., G.C. Hutterer, T. Kiesslich, K. Pummer and I. Berindan-Neagoe et al., 2016. Current insights into long non-coding RNAs in renal cell carcinoma. Int. J. Mol. Sci., Vol. 17.
    CrossRef    


  • Sani, H.M., M. Hejazian, S.M.H. Khatibi, M. Ardalan and S.Z. Vahed, 2018. Long non-coding RNAs: An essential emerging field in kidney pathogenesis. Biomed. Pharmacother., 99: 755-765.
    CrossRef    Direct Link    


  • Gao, N., Y. Li, J. Li, Z. Gao and Z. Yang et al., 2020. Long non-coding RNAs: The regulatory mechanisms, research strategies, and future directions in cancers. Front. Oncol., Vol. 10.
    CrossRef    


  • Statello, L., C.J. Guo, L.L. Chen and M. Huarte, 2021. Gene regulation by long non-coding RNAs and its biological functions. Nat. Rev. Mol. Cell Biol., 22: 96-118.
    CrossRef    Direct Link    


  • Chen, C., Y. He, Y. Feng, W. Hong, G. Luo and Z. Ye, 2021. Long non-coding RNA review and implications in acute lung inflammation. Life Sci., Vol. 269.
    CrossRef    


  • Rusconi, F., E. Battaglioli and M. Venturin, 2020. Psychiatric disorders and lncRNAs: A synaptic match. Int. J. Mol. Sci., Vol. 21.
    CrossRef    


  • Amodio, N., L. Raimondi, G. Juli, M.A. Stamato, D. Caracciolo, P. Tagliaferri and P. Tassone, 2018. MALAT1: A druggable long non-coding RNA for targeted anti-cancer approaches. J. Hematol. Oncol., Vol. 11.
    CrossRef    


  • Li, Z., Z. Ma and X. Xu, 2018. Long non‑coding RNA MALAT1 correlates with cell viability and mobility by targeting miR‑22‑3p in renal cell carcinoma via the PI3K/Akt pathway. Oncol. Rep., 41: 1113-1121.
    CrossRef    Direct Link    


  • Hirata, H., Y. Hinoda, V. Shahryari, G. Deng and K. Nakajima et al., 2015. Long noncoding RNA MALAT1 promotes aggressive renal cell carcinoma through Ezh2 and interacts with miR-205. Cancer Res., 75: 1322-1331.
    CrossRef    Direct Link    


  • Zhang, H., W. Li, W. Gu, Y. Yan, X. Yao and J. Zheng, 2019. MALAT1 accelerates the development and progression of renal cell carcinoma by decreasing the expression of miR‐203 and promoting the expression of BIRC5. Cell Proliferation, Vol. 52.
    CrossRef    


  • Jiang, L.T., C.H. Wan, Q.H. Guo, S.J. Yang, J.D. Wu and J. Cai, 2018. Long noncoding RNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) promotes renal cell carcinoma progression via sponging miRNA-429. Med. Sci. Monit., 24: 1794-1801.
    CrossRef    Direct Link    


  • Xiao, H., K. Tang, P. Liu, K. Chen and J. Hu et al., 2015. LncRNA MALAT1 functions as a competing endogenous RNA to regulate ZEB2 expression by sponging miR-200s in clear cell kidney carcinoma. Oncotarget, 6: 38005-38015.
    CrossRef    Direct Link    


  • Wang, Z., X. Chang, G. Zhu, X. Gao and L. Chang, 2020. Depletion of lncRNA MALAT1 inhibited sunitinib resistance through regulating miR-362-3p-mediated G3BP1 in renal cell carcinoma. Cell Cycle, 19: 2054-2062.
    CrossRef    Direct Link    


  • Qu, X., S. Alsager, Y. Zhuo and B. Shan, 2019. HOX transcript antisense RNA (HOTAIR) in cancer. Cancer Lett., 454: 90-97.
    CrossRef    Direct Link    


  • Tang, Q. and S.S. Hann, 2018. HOTAIR: An oncogenic long non-coding RNA in human cancer. Cell. Physiol. Biochem., 47: 893-913.
    CrossRef    Direct Link    


  • Dasgupta, P., P. Kulkarni, S. Majid, V. Shahryari, Y. Hashimoto and N.S. Bhat et al., 2018. MicroRNA-203 inhibits long noncoding RNA HOTAIR and regulates tumorigenesis through epithelial-to-mesenchymal transition pathway in renal cell carcinoma. Mol. Cancer Ther., 17: 1061-1069.
    CrossRef    Direct Link    


  • Pan, Y., Y. Wu, J. Hu, Y. Shan and J. Ma et al., 2018. Long noncoding RNA HOTAIR promotes renal cell carcinoma malignancy through alpha-2, 8-sialyltransferase 4 by sponging microRNA-124. Cell Proliferation, Vol. 51.
    CrossRef    


  • Hong, Q., O. Li, W. Zheng, W.Z. Xiao and L. Zhang et al., 2017. LncRNA HOTAIR regulates HIF-1α/AXL signaling through inhibition of miR-217 in renal cell carcinoma. Cell Death Dis., Vol. 8.
    CrossRef    


  • Wu, Y., J. Liu, Y. Zheng, L. You, D. Kuang and T. Liu, 2014. Suppressed expression of long non-coding RNA HOTAIR inhibits proliferation and tumourigenicity of renal carcinoma cells. Tumor Biol., 35: 11887-11894.
    CrossRef    Direct Link    


  • Zhang, M., Y. Zhao, Y. Zhang, D. Wang and S. Gu et al., 2018. LncRNA UCA1 promotes migration and invasion in pancreatic cancer cells via the hippo pathway. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis., 1864: 1770-1782.
    CrossRef    Direct Link    


  • Neve, B., N. Jonckheere, A. Vincent and I. van Seuningen, 2018. Epigenetic regulation by lncRNAs: An overview focused on UCA1 in colorectal cancer. Cancers, Vol. 10.
    CrossRef    


  • Liu, Q., Y. Li, W. Lv, G. Zhang and X. Tian et al., 2018. UCA1 promotes cell proliferation and invasion and inhibits apoptosis through regulation of the miR129-SOX4 pathway in renal cell carcinoma. OncoTargets Ther., 11: 2475-2487.
    CrossRef    Direct Link    


  • Lu, Y., W.G. Liu, J.H. Lu, Z.J. Liu and H.B. Li et al., 2017. LncRNA UCA1 promotes renal cell carcinoma proliferation through epigenetically repressing p21 expression and negatively regulating miR-495. Tumor Biol., Vol. 39.
    CrossRef    


  • Li, Z., J. Shen, M.T.V. Chan and W.K.K. Wu, 2016. TUG1: A pivotal oncogenic long non-coding RNA of human cancers. Cell Proliferation, 49: 471-475.
    CrossRef    Direct Link    


  • Yu, C., L. Li, F. Xie, S. Guo, F. Liu, N. Dong and Y. Wang, 2018. LncRNA TUG1 sponges miR-204-5p to promote osteoblast differentiation through upregulating Runx2 in aortic valve calcification. Cardiovasc. Res., 114: 168-179.
    CrossRef    Direct Link    


  • Cui, Y.S., Y.P. Song and B.J. Fang, 2019. The role of long non‐coding RNAs in multiple myeloma. Eur. J. Haematol., 103: 3-9.
    CrossRef    Direct Link    


  • Li, Y., D. Zheng, L. Pan, Y. Dai, S. Cai, L. Zhao and H. Zhu, 2019. Knockdown of TUG1 by shRNA inhibited renal cell carcinoma formation by miR-299–3p/VEGF axis in vitro and in vivo. Eur. J. Pharmacol., Vol. 860.
    CrossRef    


  • Liu, S., Y. Yang, W. Wang and X. Pan, 2018. Long noncoding RNA TUG1 promotes cell proliferation and migration of renal cell carcinoma via regulation of YAP. J. Cell. Biochem., 119: 9694-9706.
    CrossRef    Direct Link    


  • Yang, Y., D.M. Sun, J.F. Yu, M. Zhang and C. Yi et al., 2018. Long noncoding RNA TUG1 promotes renal cell carcinoma cell proliferation, migration and invasion by downregulating microRNA‑196a. Mol. Med. Rep., 18: 5791-5798.
    CrossRef    Direct Link    


  • Zhang, M., W. Lu, Y. Huang, J. Shi and X. Wu et al., 2016. Downregulation of the long noncoding RNA TUG1 inhibits the proliferation, migration, invasion and promotes apoptosis of renal cell carcinoma. J. Mol. Histol., 47: 421-428.
    CrossRef    Direct Link    


  • Lv, D., Y. Xiang, Q. Yang, J. Yao and Q. Dong, 2020. Long non-coding RNA TUG1 promotes cell proliferation and inhibits cell apoptosis, autophagy in clear cell renal cell carcinoma via miR-31-5p/ FLOT1 axis. OncoTargets Ther., 13: 5857-5868.
    CrossRef    Direct Link    


  • Liang, J., X. Wei, Z. Liu, D. Cao and Y. Tang et al., 2018. Long noncoding RNA CYTOR in cancer: A TCGA data review. Clin. Chim. Acta, 483: 227-233.
    CrossRef    Direct Link    


  • Wang, D., X. Zhu, B. Siqin, C. Ren and F. Yi, 2022. Long non-coding RNA CYTOR modulates cancer progression through miR-136-5p/MAT2B axis in renal cell carcinoma. Toxicol. Appl. Pharmacol., Vol. 447.
    CrossRef    


  • Zhang, F., J. Li, H. Xiao, Y. Zou, Y. Liu and W. Huang, 2018. AFAP1‐AS1: A novel oncogenic long non‐coding RNA in human cancers. Cell Proliferation, Vol. 51.
    CrossRef    


  • Mu, Z., D. Dong, N. Wei, M. Sun and W. Wang et al., 2019. Silencing of lncRNA AFAP1-AS1 inhibits cell growth and metastasis in clear cell renal cell carcinoma. Oncol. Res. Featuring Preclinical Clin. Cancer Ther., 27: 653-661.
    CrossRef    Direct Link    


  • Fan, Y., T. Yan, Y. Chai, Y. Jiang and X. Zhu, 2018. Long noncoding RNA HOTTIP as an independent prognostic marker in cancer. Clin. Chim. Acta, 482: 224-230.
    CrossRef    Direct Link    


  • Lian, Y., Z. Cai, H. Gong, S. Xue, D. Wu and K. Wang, 2016. HOTTIP: A critical oncogenic long non-coding RNA in human cancers. Mol. BioSyst., 12: 3247-3253.
    CrossRef    Direct Link    


  • Su, Y., J. Lu, X. Chen, C. Liang, P. Luo, C. Qin and J. Zhang, 2019. Long non-coding RNA HOTTIP affects renal cell carcinoma progression by regulating autophagy via the PI3K/Akt/Atg13 signaling pathway. J. Cancer Res. Clin. Oncol., 145: 573-588.
    CrossRef    Direct Link    


  • Peng, F., X. Shi, Y. Meng, B. Dong and G. Xu et al., 2018. Long non-coding RNA HOTTIP is upregulated in renal cell carcinoma and regulates cell growth and apoptosis by epigenetically silencing of LATS2. Biomed. Pharmacother., 105: 1133-1140.
    CrossRef    Direct Link    


  • Wang, Q., G. Wu, Z. Zhang, Q. Tang and W. Zheng et al., 2018. Long non-coding RNA HOTTIP promotes renal cell carcinoma progression through the regulation of the miR-615/IGF-2 pathway. Int. J. Oncol., 53: 2278-2288.
    CrossRef    Direct Link    


  • Collette, J., X.L. Bourhis and E. Adriaenssens, 2017. Regulation of human breast cancer by the long non-coding RNA H19. Int. J. Mol. Sci., Vol. 18.
    CrossRef    


  • Wang, L., Y. Cai, X. Zhao, X. Jia and J. Zhang et al., 2015. Down-regulated long non-coding RNA H19 inhibits carcinogenesis of renal cell carcinoma. Neoplasma, 62: 412-418.
    CrossRef    Direct Link    


  • He, H., N. Wang, X. Yi, C. Tang and D. Wang, 2017. Long non-coding RNA H19 regulates E2F1 expression by competitively sponging endogenous miR-29a-3p in clear cell renal cell carcinoma. Cell Biosci., Vol. 7.
    CrossRef    


  • Cao, Q., P. Li, P. Cao, J. Qian and M. Du et al., 2020. Genetic variant in long non-coding RNA H19 modulates its expression and predicts renal cell carcinoma susceptibility and mortality. Front. Oncol., Vol. 10.
    CrossRef    


  • Hang, Q., J. Lu, L. Zuo and M. Liu, 2021. Linc00641 promotes the progression of gastric carcinoma by modulating the miR-429/Notch-1 axis. Aging, 13: 8497-8509.
    CrossRef    Direct Link    


  • Zhang, J., S. Jin, W. Xiao, X. Zhu, C. Jia and Z. Lin, 2021. Long noncoding RNA LINC00641 promotes renal cell carcinoma progression via sponging microRNA-340-5p. Cancer Cell Int., Vol. 21.
    CrossRef    


  • Fan, F., K. Chen, X. Lu, A. Li, C. Liu and B. Wu, 2021. Dual targeting of PD-L1 and PD-L2 by PCED1B-AS1 via sponging hsa-miR-194-5p induces immunosuppression in hepatocellular carcinoma. Hepatol. Int., 15: 444-458.
    CrossRef    Direct Link    


  • Yao, Z., Q. Zhang, F. Guo, S. Guo and B. Yang et al., 2020. Long noncoding RNA PCED1B-AS1 promotes the warburg effect and tumorigenesis by upregulating HIF-1α in glioblastoma. Cell Transplant., Vol. 29.
    CrossRef    


  • Zhang, Y., H. Ma and C. Chen, 2021. Long non‑coding RNA PCED1B‑AS1 promotes pancreatic ductal adenocarcinoma progression by regulating the miR‑411‑3p/HIF‑1α axis. Oncol. Rep., Vol. 46.
    CrossRef    


  • Qin, J., T. Zhu, W. Wu, H. Chen and Y. He, 2021. Long non-coding RNA PCED1B-AS1 promotes the progression of clear cell renal cell carcinoma through miR-484/ZEB1 axis. OncoTargets Ther., 14: 393-402.
    CrossRef    Direct Link    


  • Thin, K.Z., J.C. Tu and S. Raveendran, 2019. Long non-coding SNHG1 in cancer. Clin. Chim. Acta, 494: 38-47.
    CrossRef    Direct Link    


  • Ye, Z.H., D.W. Gui, X.Y. Wang, J. Wang and J.L. Fu, 2022. LncRNA SNHG1 promotes renal cell carcinoma progression through regulation of HMGA2 via sponging miR‐103a. J. Clin. Lab. Anal., Vol. 36.
    CrossRef    


  • Tian, P., J.X. Wei, J. Li, J.K. Ren and J.J. Yang, 2021. LncRNA SNHG1 regulates immune escape of renal cell carcinoma by targeting miR-129-3p to activate STAT3 and PD-L1. Cell Biol. Int., 45: 1546-1560.
    CrossRef    Direct Link    


  • Zhao, S., Y. Wang, M. Luo, W. Cui, X. Zhou and L. Miao, 2018. Long noncoding RNA small nucleolar RNA host gene 1 (SNHG1) promotes renal cell carcinoma progression and metastasis by negatively regulating miR-137. Med. Sci. Monit., 24: 3824-3831.
    CrossRef    Direct Link    


  • Li, Y.H., Y.Q. Hu, S.C. Wang, Y. Li and D.M. Chen, 2020. LncRNA SNHG5: A new budding star in human cancers. Gene, Vol. 749.
    CrossRef    


  • Xiang, W., L. Lv, G. Zhou, W. Wu, J. Yuan, C. Zhang and G. Jiang, 2020. The lncRNA SNHG5‐mediated miR‐205‐5p downregulation contributes to the progression of clear cell renal cell carcinoma by targeting ZEB1. Cancer Med., 9: 4251-4264.
    CrossRef    Direct Link    


  • Li, W.Z., Y. Zou, Z.Y. Song, Z.W. Wei, G. Chen, Q.L. Cai and Z. Wang, 2020. Long non-coding RNA SNHG5 affects the invasion and apoptosis of renal cell carcinoma by regulating the miR-363-3p-twist1 interaction. Am. J. Transl. Res., 12: 697-707.
    Direct Link    


  • Wang, S., X. Chen and T. Qiao, 2021. Long non-coding RNA MIR4435-2HG promotes the progression of head and neck squamous cell carcinoma by regulating the miR‑383‑5p/RBM3 axis. Oncol. Rep., Vol. 45.
    CrossRef    


  • Zhu, K., C. Miao, Y. Tian, Z. Qin and J. Xue et al., 2020. lncRNA MIR4435-2HG promoted clear cell renal cell carcinoma malignant progression via miR-513a-5p/KLF6 axis. J. Cell. Mol. Med., 24: 10013-10026.
    CrossRef    Direct Link    


  • Zhang, C., X. Ren, W. Zhang, L. He and L. Qi et al., 2020. Prognostic and clinical significance of long non-coding RNA SNHG12 expression in various cancers. Bioengineered, 11: 1112-1123.
    CrossRef    Direct Link    


  • Tamang, S., V. Acharya, D. Roy, R. Sharma and A. Aryaa et al., 2019. SNHG12: An LncRNA as a potential therapeutic target and biomarker for human cancer. Front. Oncol., Vol. 9.
    CrossRef    


  • Xu, C., H. Liang, J. Zhou, Y. Wang and S. Liu et al., 2020. lncRNA small nucleolar RNA host gene 12 promotes renal cell carcinoma progression by modulating the miR‑200c‑5p/collagen type XI α1 chain pathway. Mol. Med. Rep., 22: 3677-3686.
    CrossRef    Direct Link    


  • Liu, H., W. Zheng, Q. Chen, Y. Zhou and Y. Pan et al., 2021. lncRNA CASC19 contributes to radioresistance of nasopharyngeal carcinoma by promoting autophagy via AMPK-mTOR pathway. Int. J. Mol. Sci., Vol. 22.
    CrossRef    


  • Luo, Y., F. Liu, C. Yan, W. Qu and L. Zhu et al., 2020. Long non-coding RNA CASC19 sponges microRNA-532 and promotes oncogenicity of clear cell renal cell carcinoma by increasing ETS1 expression. Cancer Manag. Res., 12: 2195-2207.
    CrossRef    Direct Link    


  • Zhou, Y., X. Zhang and A. Klibanski, 2012. MEG3 noncoding RNA: A tumor suppressor. J. Mol. Endocrinol., 48: R45-R53.
    CrossRef    Direct Link    


  • Ghafouri-Fard, S. and M. Taheri, 2019. Maternally expressed gene 3 (MEG3): A tumor suppressor long non coding RNA. Biomed. Pharmacother., Vol. 118.
    CrossRef    


  • He, H., J. Dai, R. Zhuo, J. Zhao and H. Wang et al., 2018. Study on the mechanism behind lncRNA MEG3 affecting clear cell renal cell carcinoma by regulating miR‐7/RASL11B signaling. J. Cell. Physiol., 233: 9503-9515.
    CrossRef    Direct Link    


  • Ma, B., J. Zhang, W. Zhou, C. Chu, C. Zhao, Z. Zhang and T. Huang, 2018. LINC01510 suppresses cell proliferation and invasion by inhibiting Wnt/β-catenin signaling in renal cell carcinoma. Biochem. Biophys. Res. Commun., 505: 7-12.
    CrossRef    Direct Link    


  • Chen, C.L., Q. Ke, M. Luo, Z.Y. Gao, Z.J. Li, Z.G. Luo and D.B. Liu, 2018. Loss of LINC01939 expression predicts progression and poor survival in gastric cancer. Pathol. Res. Pract., 214: 1539-1543.
    CrossRef    Direct Link    


  • Zhang, R., W. Zhang, B. Xu, C. Lv, J. Hou and G. Zhang, 2020. Long intergenic non-coding RNA 1939 eliminates proliferation and migration of human renal cell carcinoma (RCC) cells by down-regulation of miR-154. Artif. Cells Nanomed. Biotechnol., 48: 695-702.
    CrossRef    Direct Link    


  • Li, G., M. Xie, Z. Huang, H. Li and P. Li et al., 2020. Overexpression of antisense long non-coding RNA ZNF710-AS1-202 promotes cell proliferation and inhibits apoptosis of clear cell renal cell carcinoma via regulation of ZNF710 expression. Mol. Med. Rep., 21: 2502-2512.
    CrossRef    Direct Link    


  • Li, M., B. Yin, M. Chen, J. Peng and X. Mu et al., 2021. Downregulation of the lncRNA ASB16-AS1 decreases LARP1 expression and promotes clear cell renal cell carcinoma progression via miR-185-5p/miR-214-3p. Front. Oncol., Vol. 10.
    CrossRef    


  • He, Z.H., X.H. Qin, X.L. Zhang, J.W. Yi and J.Y. Han, 2018. Long noncoding RNA GIHCG is a potential diagnostic and prognostic biomarker and therapeutic target for renal cell carcinoma. Eur. Rev. Med. Pharmacol. Sci., 22: 46-54.
    CrossRef    Direct Link    


  • Zheng, Z., F. Zhao, D. Zhu, J. Han and H. Chen et al., 2018. Long non-coding RNA LUCAT1 promotes proliferation and invasion in clear cell renal cell carcinoma through AKT/GSK-3β signaling pathway. Cell. Physiol. Biochem., 48: 891-904.
    CrossRef    Direct Link    


  • Xie, J., Y. Zhong, R. Chen, G. Li and Y. Luo et al., 2020. Serum long non‐coding RNA LINC00887 as a potential biomarker for diagnosis of renal cell carcinoma. FEBS Open Bio, 10: 1802-1809.
    CrossRef    Direct Link    


  • Qu, L., J. Ding, C. Chen, Z.J. Wu and B. Liu et al., 2016. Exosome-transmitted lncARSR promotes sunitinib resistance in renal cancer by acting as a competing endogenous RNA. Cancer Cell, 29: 653-668.
    CrossRef    Direct Link    


  • He, J., K. Zhou, B. Zhu, G. Zhang and X. Li et al., 2015. Dynamic contrast-enhanced CT characterization of Xp11.2 translocation/TFE3 gene fusions versus papillary renal cell carcinomas. BioMed Res. Int., Vol. 2015.
    CrossRef    


  • He, D., J. Zheng, J. Hu, J. Chen and X. Wei, 2020. Long non-coding RNAs and pyroptosis. Clin. Chim. Acta, 504: 201-208.
    CrossRef    Direct Link    

  • © Science Alert. All Rights Reserved