Long non‑coding RNAs in prostate tumorigenesis and therapy (Review)

  • Authors:
    • Ganggang Jiang
    • Zhengming Su
    • Xue Liang
    • Yiqiao Huang
    • Ziquan Lan
    • Xianhan Jiang
  • View Affiliations

  • Published online on: September 24, 2020     https://doi.org/10.3892/mco.2020.2146
  • Article Number: 76
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Prostate cancer (PCa) is one of the most frequently diagnosed malignancy. Although there have been many advances in PCa diagnosis and therapy, the concrete mechanism remains unknown. Long non‑coding RNAs (lncRNAs) are novel biomarkers associated with PCa, and their dysregulated expression is closely associated with risk stratification, diagnosis and carcinogenesis. Accumulating evidence has suggested that lncRNAs play important roles in prostate tumorigenesis through relevant pathways, such as androgen receptor interaction and PI3K/Akt. The present review systematically summarized the potential clinical utility of lncRNAs and provided a novel guide for their function in PCa.

1. Introduction

Prostate cancer (PCa) is the second most common malignancy in Western countries and accounts for 10% of cancer-related deaths (1). Approximately 26,730 deaths occurred in 2017 due to PCa in the United States. The causes of this disease include age, functional testicles, and family heredity. Most cases of PCa undergo a series of processes from androgen sensitivity neoplasia to metastatic castration-resistant PCa (mCRPC), which is presently incurable. Currently available means for diagnosis depend on prostate specific antigen (PSA) and pathological biopsy. Since PSA testing was introduced, the incidence of localized PCa has increased significantly; PSA testing can predict cancer risk and treatment outcome (2). According to the literature, almost 240,000 individuals developed PCa in the United States yearly; however, <15% of these patients eventually died; mortality is largely dependent upon PSA testing and reasonable treatment of PCa at an early stage (3-6). However, serum PSA is not particular to PCa, and levels can be enhanced in benign prostatic hyperplasia (BPH) (7) and prostatitis (8) even after a digital rectal examination. Therefore, the lack of specificity limits its further development. Low specificity has caused unnecessary biopsies, thereby leading to the overtreatment of indolent cancers. Pathological biopsy is the standard in diagnosing PCa, but it is an invasive examination, which will cause hemorrhage, infection, and even blood poisoning. Hence, a novel biomarker for PCa diagnosis and therapy should is urgently needed.

RNA plays a crucial part in the regulation of gene expression and genome organization (9,10). RNA serves as a template for protein synthesis and exerts many functions (11). The current research has concluded that only 10% of the genome is made up of protein-coding genes. A large part of the genome (~70%) is actively transcribed, which indicates that noncoding RNAs (ncRNAs) account for an overwhelming percentage of the human transcriptome (Fig. 1). At the start, NcRNAs, which is viewed as ‘noisy RNAs,’ have no transcriptional function and account for almost 90% RNAs in humans (12). ncRNAs can be divided into two major groups according to their sizes, as follows: Long (lncRNA, >200 bp) and small ncRNAs (<200 bp) (13). Small ncRNAs include termini-associated short, transcription initiation, splice site, and antisense termini-associated short RNAs (14). Most lncRNAs are generated similar to other mRNAs, as emphasized by RNA polymerase II activity and histone modifications associated with transcription initiation and elongation (15). lncRNAs can be divided into intragenic and intergenic lncRNAs according to their locations in the genome relative to protein coding genes (16). lncRNAs also play a role as decoys for transcription factors (17) and regulate protein activity (18,19). lncRNAs are aberrantly expressed in a several human diseases, including kidney cancer (20), colorectal cancer (21), endometrial cancer (22), testicular cancer (23,24), breast cancer (25), and hematological cancers (26,27). Bussemakers (28) first found DD3PCA3, which is a lncRNA and is a potential diagnostic biomarker for PCa in 1999. This discovery started the research on the involvement of lncRNAs in PCa. We summarized the viewpoints, as follows (Fig. 1).

2. Key pathways dysregulated in prostate cancer by lncRNAs

Androgen receptor (AR) interaction

The AR, as a protein coding gene, is situated on the X chromosome, is approximately 110 kD and made up of four functional regions, namely, (1) the hinge region, (2) ligand-binding domain, (3) N-terminal transactivation domain, and (4) DNA-binding domain (29-31). The AR is a nuclear transcription factor required for normal prostate development and PCa; thus, it plays key roles in PCa initiation and progression (32,33). PCa undergoes progression from androgen-sensitive to resistance to castration. Androgen deprivation therapy (ADT) is the frontline treatment for PCa at the late stages. However, after 12 to 24 months of androgen deprivation, PCa will eventually progress to the lethal form of the disease known as castration-resistant PCa (CRPC), which is eventually fatal for patients with PCa. The reactivation of the AR is central to the progression of CRPC, and treatment mechanisms may also be mediated by the AR signaling axis. AR-dependent resistance mechanisms include AR enhancement, AR single-base substitution, changed intratumoral androgen biosynthesis, and the expression of constitutively active AR splice variants (34-37). lncRNAs can function as oncogenic and tumor suppressor in PCa through AR signaling axis (Table I).

Table I

lncRNAs in prostate cancer.

Table I

lncRNAs in prostate cancer.

lncRNALocationExpressionPathway(Refs.)
PCGEM12q32AR interaction(39,40)
HOTAIR12q13.13AR interaction(49)
CTBP1-AS4p16.3AR interaction(51)
PCA39q21-22AR interaction(56)
UCA119p13.12PI3K/Akt pathway(67)
LINC0129614q11.2PI3K/Akt pathway(68)
PCAT-18q24.21by C-MYC protein(72)
SChLAP12q31.1Interfering with SWI/SNF tumor-inhibiting complex(75)
H1911p15.5Down-regulation of TGFBI(86)
TODRA15q15.1Causing DSB repair by HR(88)

[i] lncRNA, long non-coding RNA.

PCGEM1

PCGEM1, as the earliest prostate-specific lncRNA, is located on chromosome 2q32 and overexpressed in nearly 84% of patients with PCa. Recent studies revealed the PRNCR1 binding site to AR 549-623 location and the PCGEM1 combining site to the N-terminal location of AR. PCGEM1 and PRNCR1 interact with AR (38) in a recently published report. In various PCa cells, lncRNA cannot be detected in AR-null cell lines, such as DU145 and PC3 (39,40). The coalition of PRNCR1 and AR leads to enrollment of DOT1L methyltransferase methylating AR and allows the subsequent interaction of PCGEM1 with the methylated AR. In turn, PCGEM1 recruits PYGO2 (Pygopus 2), thereby allowing the binding of AR to H3K4me3 chromatin marks to the promoter regions of AR-regulated genes and leading to their activation (38).

A positive correlation of PCGEM1 with AR3, which is one of the most important splice variants that play a key role in castration resistance, was observed (41-43). This AD-PCGEM1-AR3 axis can explain several reasons why the effectiveness of ADT can only be sustained for a short time. Heterogeneous nuclear ribonucleoprotein A1 (HnRNP A1) and U2AF65, as splicing factors, play key roles in AR3 expression. When hnRNP A1 combines with PCGEM1, the coalition activity of hnRNP A1 to AR pre-mRNA is weakened. By contrast, the binding activity of U2AF65 to AR pre-mRNA is enhanced. A specific molecular mechanism does not explain why interaction between U2AF65 and PCGEM1 is dominant. The binding of PCGEM1 to U2AF65 is more competitive than that of hnRNP A1 to AR pre-mRNA (44).

HOX transcript antisense RNA (HOTAIR)

HOTAIR lncRNA is a 2.2 kb-long transcript and overexpressed in a variety of cancer types, such as breast cancer, colorectal cancer, lung cancer, and pancreatic cancer (45-48). HOTAIR is sensitive to androgen and inhibited by androgen severely, and its expression inhibits AR ubiquitination and avoids AR protein degradation (49). HOTAIR lncRNA is entirely abolished after AR target gene is knocked down via RNA interference (49). Zhang et al (49) concluded that HOTAIR overexpression enhances aggressivity in PCa and upregulates in enzalutamide-resistant PCa cells. Therefore, attention should be paid to HOTAIR as a potential therapy target in enzalutamide-resistant patients with PCa in the future.

C-Terminal binding protein 1 antisense (CTBP1-AS) CTBP1-AS, which is situated in the AS region of C-terminal binding protein 1 (CTBP1), is related to AR signaling pathway and is overexpressed in both local PCa patients and metastatic PCa patients, but not in Benign Prostatic Hyperplasia (BPH). It is recruited to AR-binding sites. The CTBP1-AS lncRNA directly inhibited the expression of CTBP1(50), which acted as the corepressor of AR by recruiting the RNA binding transcriptional repressor PTB-associated splicing factor and histone deacetylases. Thus, CTBP1-AS can enhance AR transcriptional activity. Takayama et al (51) have reported that upregulation of CTBP1-AS and downregulation of CTBP1 in PCa. CTBP1-AS knockdown inhibited cell proliferation in hormone-depleted condition in both cell lines; in contrast, CTBP1-AS overexpression induced tumor growth after castration (51).

PCa gene 3 (PCA3)

PCA3 is a lncRNA that was initially named as DD3 and is located on chromosome 9q21-22 in antisense direction within the intron 6 of the Prune homolog 2 gene (PRUNE2 or BMCC1) (52). PCA3 is overexpressed in PCa cell lines (53,54) and modulates PCa cell survival partly according to the AR pathway, which is involved in the oncogenesis of PCa. Meanwhile, the positive rates of its sensitivity and specificity are 82.3 and 89.0%, respectively, compared with PSA, which showed only 57.4 and 53.8% (55). Lemos et al (56) reported that PCA3 may regulate AR signal pathway through AR cofactors (57), such as ARA 54, ARA 70, CBP, and P300, when PCA3 and ERK are silenced, and Akt protein phosphorylation levels stayed the same. Thus, the preferred method should be activate AR. Both AR cofactors, including coactivators and corepressors, were upregulated, which indicated that PCA3 may be a negative modulator to AR and aberrant cofactor activity because altered or changed expression levels may be factors to the progression to mCRPC. A future potential therapy for PCa patients, especially mCRPC, is the application of PCA3.

Phosphatidylinositol 3-kinase (PI3K)/Akt pathway

PI3K/Akt pathway is one of the key signal transduction pathways regulating cell proliferation. PI3K/AKT/mTOR signaling, PTEN/PI3K/AKT pathway, and PI3K/AKT/NF-kappaB/BMP-2-Smad axis play important roles in cancer progression and development (58). PI3K enzymes regulate cellular signal transduction. The PI3K/Akt pathway mainly includes PI3K activation, recruiting pleckstrin homology (PH) domain-containing proteins, phosphorylation, activating AKT, and activating necessary downstream targets (59). PI3K/AKT/mTOR is overexpressed in 30-50% of all prostate cancers (60), and its signal is regulated in PCa cellular proliferation (61), apoptosis (62), invasion, and migration (63).

lncRNA-ATB

lncRNA-ATB is first identified in hepatocellular carcinoma (64). Xu et al (65) reported that lncRNA-ATB is overexpressed in PCa tissues compared with normal tissues, and it is related to high PSA level, high Gleason score, and biochemical recurrence when the knockdown of lncRNA-ATB and PI3K/Akt signaling pathways is inhibited. Meanwhile, the roles of lncRNA-ATB in the invasion, migration, and tumor growth remain unclear.

Urothelial carcinoma associated 1 (UCA1)

UCA1 is a lncRNA that is related to various cancer types (66). Ghiam et al (67) revealed that UCA1 expression is high in PCa cells when UCA1 knockdown enhances radiosensitivity in classic PCa cell lines and irradiation-resistant PCa cells due to PI3K/Akt pathway downregulation.

LINC01296

LINC01296 is located at chromosome 14q11.2. Wu et al (68) found that LINC01296 is upregulated in LNCaP cell lines but not in normal cell lines. Meanwhile, when silencing LINC01296, the protein expression level of PI3K-Akt-mTOR signaling pathway significantly decreased compared with that of normal cell. Therefore, the lncRNA LOC400891 regulates cell proliferation through the PI3K–Akt-mTOR signaling pathway (69).

Act as a tumorigenesis or a tumor-inhibiting gene

Genome instability is the main factor in the promotion of cancer. The aberrant expression of lncRNAs is related to the development and progression of PCa and plays an important role in tumorigenesis or tumor-inhibiting in patients with PCa. Several lncRNAs are upregulated as oncogenes, whereas others are downregulated in cancer.

PCa-associated lncRNA transcripts 1 (PCAT-1)

PCAT-1, which is located in the 8q24.21 gene desert with nearly 725 kb upstream of the c-MYC oncogene (70), is overexpressed in patients with PCa (71). C-MYC protein is upregulated by PCAT-1, thereby resulting in specific gene expression programs and cell proliferation (72). When PCAT-1 is knocked down in LNCaP cells, cellular proliferation is diminished, thereby indicating that it is a potential novel biomarker for colorectal cancer metastasis.

Second chromosome locus associated with prostate-1 (SChLAP1)

SChLAP1 is a novel biomarker that is highly upregulated in PCa (73-75) and associated with a high risk of CRPC, thereby leading to tumor cell invasion and metastasis. SChLAP1 can interfere with the SWI/SNF tumor-inhibiting complex (76) to promote tumor metastasis. SChLAP1 damages genomic binding and SNF5-mediated gene expression regulation. Similarly, Mehra et al (75) found that knocking down SChLAP1 can inhibit cell proliferation and migration in bladder cancer cell lines.

Noncoding nuclear-enriched abundant transcript2 (NEAT2)

The lncRNA NEAT2 is 7 kb long and is also known as MALAT1. NEAT2 is highly overexpressed in a series of cancers, including prostate cancer (77), osteosarcoma (78), pancreatic cancer (79), breast cancer (80), bladder cancer (81), and esophageal cancer (82,83). When NEAT2 is knocked down, cell hyperproliferation and metastasis are inhibited in a PCa cell, thereby leading to cell cycle block in the G0/G1 phases (77). NEAT2 promotes the activation of PRC2 by connecting to the polycomb protein enhancer of zeste homolog 2 (EZH2) and enhances the EZH2-mediated inhibition of polycomb-dependent target gene E-cadherin in clear renal cancer (84). Meanwhile, NEAT2 controls cell cycle progression by regulating the oncogenic transcription factor B-MYB (Mybl2) (85).

H19

The H19 gene, which is transcribed from H19/Igf2 gene cluster, is located on human chromosome 11p15.5. H19 can acts as a tumor suppressor gene. Zhu et al (86) found that the decreased H19 expression is significant in metastatic prostate cell compared with local prostate epithelial cell. Hence, H19/miR-675 axis inhibits PCa metastasis according to TGFBI downregulation.

Other genes

Double-strand DNA breaks (DSBs) are potentially lethal DNA lesions. Homologous recombination (HR) is an effective pathway for eliminating DSBs and repairing injured DNA replication forks. RAD51 is the core recombinase involved in HR, and increased RAD51 levels may cause tumorigenesis. Prensner et al (87) found that lncRNA PCAT1 is involved in the DSB repair process in PCa. TODRA is a novel lncRNA that is also known as RAD51 antisense RNA 1 and is located on 15q15.1. TODRA plays a role in RAD51 regulation. Gazy et al (88) reported that the overexpression of TODRA causes DSB repair by HR and also enhances the fraction of RAD51 foci formed after DNA damage.

Recent studies indicated that lncRNAs can recognize miRNA elements that can be targeted by miRNAs (89). A Zebrafish model where miR-125b regulates 7sl lncRNA expression is a typical example of the miRNA-lncRNA interaction (90). Meanwhile, HOTAIR downregulation is targeted by the tumor suppressor miR-34a, thereby inhibiting CRPC cell growth (91).

3. Therapeutic potential of lncRNAs in patients with cancer

lncRNAs has multiple functions and high cell-type specificity. Thus, lncRNAs can provide an avenue for PCa diagnosis, prognosis, and therapy. Currently, the use of lncRNA for PCa patients is being explored.

lncRNA targeting strategy

The RNAi technology, which interferes with RNA expression through antisense technologies, can be widely used for the weak expression levels of lncRNAs with oncology potential (92). The key cancer-associated genes with therapeutic siRNAs have been suppressed in clinical trials.

Another method for inhibiting cancer-associated RNA is by using catalytic nucleic acids, including antisense oligonucleotides (ASOs) or by using small molecule inhibitors that can also be used to modulate lncRNAs. The small molecule inhibitors prevent the interaction of HOTAIR with LSD1 or PRC2 complexes, thereby restricting the metastatic potential of breast cancer (93).

Targeting lncRNAs through the CRISPER/Cas system

Currently, CRISPER/Cas (clustered regularly interspaced short palindromicrepeats/CRISPER-associated system) take advantage of knocking out targeting gene for treating PCa patients. Meanwhile, Shechner et al (94) have ever reported of using CRISPER/Cas system successfully.

4. Conclusions

lncRNAs are potential novel biomarkers as therapeutic targets for patients with PCa. Considering the multiple and varied processes of PCa, its treatment should be planned precisely for each patient. At the same time, when modern technology is used to kill tumors, the safety of normal tissues should be ensured. However, in spite of its advantage over other therapeutic options, many questions need to be addressed.

The biggest challenge is that further research and large-scale validation studies are imperative before the successful application to clinical trials, because the molecular mechanisms of lncRNAs and pathogenesis of PCa have not been thoroughly understood. Nevertheless, the clearer the lncRNA functions are, the better their field of therapeutic usage will be. The next challenge is that the current lncRNA marker candidates are mostly based on a small sample, and the lack of validity limits further development. Thus, the effectiveness of lncRNA markers have to be prospectively verified in large and varied datasets. Research in animal models and clinical trials is needed to evaluate the potential side effects, including toxicity, body distribution, pharmacokinetics, and pharmacodynamics data.

Hence, lncRNAs are intriguing targets in treating patients with PCa, and their potential in therapy can be remarkable.

Acknowledgements

The authors would like to thank Miss Chen Gao (Department of Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China) for assistance.

Funding

The present study was supported by funding from Science and Technology Plan Project of Guangzhou of China (grant no. 201510010177), Medical Science and Technology Program of Guangzhou of China (grant nos. 20171A011329 and 20171A010325), Youth Scientific Research Project of Guangzhou Medical University of China (grant no. 2015A14) and Medical Research Foundation of Guangdong Province of China (grant no. A2018093).

Availability of data and materials

All data generated or analyzed during the present study are included in this article.

Authors' contributions

GJ, ZS, XL, YH, ZL and XJ conceived and designed the study. GJ and ZS wrote the manuscript. GJ, ZS, XL, YH, ZL and XJ collected the data. ZS and XJ reviewed and edited the manuscript. All authors read and approved the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel R, Ma J, Zou Z and Jemal A: Cancer statistics, 2014. CA Cancer J Clin. 64:9–29. 2014.PubMed/NCBI View Article : Google Scholar

2 

Carter HB and Pearson JD: PSA velocity for the diagnosis of early prostate cancer. A new concept. Urol Clin North Am. 20:665–670. 1993.PubMed/NCBI

3 

Gandellini P, Folini M, Longoni N, Pennati M, Binda M, Colecchia M, Salvioni R, Supino R, Moretti R, Limonta P, et al: miR-205 Exerts tumor-suppressive functions in human prostate through down-regulation of protein kinase Cepsilon. Cancer Res. 69:2287–2295. 2009.PubMed/NCBI View Article : Google Scholar

4 

Tucci P, Agostini M, Grespi F, Markert EK, Terrinoni A, Vousden KH, Muller PA, Dotsch V, Kehrloesser S, Sayan BS, et al: Loss of p63 and its microRNA-205 target results in enhanced cell migration and metastasis in prostate cancer. Proc Natl Acad Sci USA. 109:15312–15317. 2012.PubMed/NCBI View Article : Google Scholar

5 

Gandellini P, Profumo V, Casamichele A, Fenderico N, Borrelli S, Petrovich G, Santilli G, Callari M, Colecchia M, Pozzi S, et al: miR-205 regulates basement membrane deposition in human prostate: Implications for cancer development. Cell Death Differ. 19:1750–1760. 2012.PubMed/NCBI View Article : Google Scholar

6 

Bonci D, Coppola V, Musumeci M, Addario A, Giuffrida R, Memeo L, D'Urso L, Pagliuca A, Biffoni M, Labbaye C, et al: The miR-15a-miR-16-1 cluster controls prostate cancer by targeting multiple oncogenic activities. Nat Med. 14:1271–1277. 2008.PubMed/NCBI View Article : Google Scholar

7 

Park TY, Chae JY, Kim JW, Kim JW, Oh MM, Yoon CY and Moon du G: Prostate-specific antigen mass and free prostate-specific antigen mass for predicting the prostate volume of korean men with biopsy-proven benign prostatic hyperplasia. Korean J Urol. 54:609–614. 2013.PubMed/NCBI View Article : Google Scholar

8 

Wang Y, Hu HL, Liu ZF, Sun WZ, Chen XX and Wu CL: Diagnosis and treatment of xanthogranulomatous prostatitis: A case report and review of the literature. Zhonghua Nan Ke Xue. 19:149–152. 2013.PubMed/NCBI(In Chinese).

9 

ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature. 489:57–74. 2012.PubMed/NCBI View Article : Google Scholar

10 

Ma W, Ay F, Lee C, Gulsoy G, Deng X, Cook S, Hesson J, Cavanaugh C, Ware CB, Krumm A, et al: Fine-scale chromatin interaction maps reveal the cis-regulatory landscape of human lincRNA genes. Nat Methods. 12:71–78. 2015.PubMed/NCBI View Article : Google Scholar

11 

Ling H, Vincent K, Pichler M, Fodde R, Berindan-Neagoe I, Slack FJ and Calin GA: Junk DNA and the long non-coding RNA twist in cancer genetics. Oncogene. 34:5003–5011. 2015.PubMed/NCBI View Article : Google Scholar

12 

Kapranov P, Cheng J, Dike S, Nix DA, Duttagupta R, Willingham AT, Stadler PF, Hertel J, Hackermuller J, Hofacker IL, et al: RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science. 316:1484–1488. 2007.PubMed/NCBI View Article : Google Scholar

13 

Brosnan CA and Voinnet O: The long and the short of noncoding RNAs. Curr Opin Cell Biol. 21:416–425. 2009.PubMed/NCBI View Article : Google Scholar

14 

Gibb EA, Brown CJ and Lam WL: The functional role of long non-coding RNA in human carcinomas. Mol Cancer. 10(38)2011.PubMed/NCBI View Article : Google Scholar

15 

Mercer TR and Mattick JS: Structure and function of long noncoding RNAs in epigenetic regulation. Nat Struct Mol Biol. 20:300–307. 2013.PubMed/NCBI View Article : Google Scholar

16 

Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, Guernec G, Martin D, Merkel A, Knowles DG, et al: The GENCODE v7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression. Genome Res. 22:1775–1789. 2012.PubMed/NCBI View Article : Google Scholar

17 

Kino T, Hurt DE, Ichijo T, Nader N and Chrousos GP: Noncoding RNA gas5 is a growth arrest- and starvation-associated repressor of the glucocorticoid receptor. Sci Signal. 3(ra8)2010.PubMed/NCBI View Article : Google Scholar

18 

Mallory AC and Shkumatava A: lncRNAs in vertebrates: Advances and challenges. Biochimie. 117:3–14. 2015.PubMed/NCBI View Article : Google Scholar

19 

Yeh E, Cunningham M, Arnold H, Chasse D, Monteith T, Ivaldi G, Hahn WC, Stukenberg PT, Shenolikar S, Uchida T, et al: A signalling pathway controlling c-Myc degradation that impacts oncogenic transformation of human cells. Nat Cell Biol. 6:308–318. 2004.PubMed/NCBI View Article : Google Scholar

20 

Seles M, Hutterer GC, Kiesslich T, Pummer K, Berindan-Neagoe I, Perakis S, Schwarzenbacher D, Stotz M, Gerger A and Pichler M: Current insights into long non-coding RNAs in renal cell carcinoma. Int J Mol Sci. 17(573)2016.PubMed/NCBI View Article : Google Scholar

21 

Smolle M, Uranitsch S, Gerger A, Pichler M and Haybaeck J: Current status of long non-coding RNAs in human cancer with specific focus on colorectal cancer. Int J Mol Sci. 15:13993–14013. 2014.PubMed/NCBI View Article : Google Scholar

22 

Smolle MA, Bullock MD, Ling H, Pichler M and Haybaeck J: Long non-coding RNAs in endometrial carcinoma. Int J Mol Sci. 16:26463–26472. 2015.PubMed/NCBI View Article : Google Scholar

23 

Bezan A, Gerger A and Pichler M: MicroRNAs in testicular cancer: Implications for pathogenesis, diagnosis, prognosis and therapy. Anticancer Res. 34:2709–2713. 2014.PubMed/NCBI

24 

Ling H, Krassnig L, Bullock MD and Pichler M: MicroRNAs in testicular cancer diagnosis and prognosis. Urol Clin North Am. 43:127–134. 2016.PubMed/NCBI View Article : Google Scholar

25 

Cerk S, Schwarzenbacher D, Adiprasito JB, Stotz M, Hutterer GC, Gerger A, Ling H, Calin GA and Pichler M: Current status of long Non-coding RNAs in human breast cancer. Int J Mol Sci. 17(1485)2016.PubMed/NCBI View Article : Google Scholar

26 

Zebisch A, Hatzl S, Pichler M, Wolfler A and Sill H: Therapeutic resistance in acute myeloid leukemia: The role of non-coding RNAs. Int J Mol Sci. 17(2080)2016.PubMed/NCBI View Article : Google Scholar

27 

Troppan K, Wenzl K, Deutsch A, Ling H, Neumeister P and Pichler M: MicroRNAs in diffuse large B-cell lymphoma: Implications for pathogenesis, diagnosis, prognosis and therapy. Anticancer Res. 34:557–564. 2014.PubMed/NCBI

28 

Bussemakers MJ, van Bokhoven A, Verhaegh GW, Smit FP, Karthaus HF, Schalken JA, Debruyne FM, Ru N and Isaacs WB: DD3: A new prostate-specific gene, highly overexpressed in prostate cancer. Cancer Res. 59:5975–5979. 1999.PubMed/NCBI

29 

Gelmann EP: Molecular biology of the androgen receptor. J Clin Oncol. 20:3001–3015. 2002.PubMed/NCBI View Article : Google Scholar

30 

Claessens F, Denayer S, Van Tilborgh N, Kerkhofs S, Helsen C and Haelens A: Diverse roles of androgen receptor (AR) domains in AR-mediated signaling. Nucl Recept Signal. 6(e008)2008.PubMed/NCBI View Article : Google Scholar

31 

Heemers HV and Tindall DJ: Androgen receptor (AR) coregulators: A diversity of functions converging on and regulating the AR transcriptional complex. Endocr Rev. 28:778–808. 2007.PubMed/NCBI View Article : Google Scholar

32 

Chang CS, Kokontis J and Liao ST: Molecular cloning of human and rat complementary DNA encoding androgen receptors. Science. 240:324–326. 1988.PubMed/NCBI View Article : Google Scholar

33 

Heinlein CA and Chang C: Androgen receptor in prostate cancer. Endocr Rev. 25:276–308. 2004.PubMed/NCBI View Article : Google Scholar

34 

Jentzmik F, Azoitei A, Zengerling F, Damjanoski I and Cronauer MV: Androgen receptor aberrations in the era of abiraterone and enzalutamide. World J Urol. 34:297–303. 2016.PubMed/NCBI View Article : Google Scholar

35 

Mitsiades N: A road map to comprehensive androgen receptor axis targeting for castration-resistant prostate cancer. Cancer Res. 73:4599–4605. 2013.PubMed/NCBI View Article : Google Scholar

36 

Scher HI and Sawyers CL: Biology of progressive, castration-resistant prostate cancer: Directed therapies targeting the androgen-receptor signaling axis. J Clin Oncol. 23:8253–8261. 2005.PubMed/NCBI View Article : Google Scholar

37 

Karantanos T, Corn PG and Thompson TC: Prostate cancer progression after androgen deprivation therapy: Mechanisms of castrate resistance and novel therapeutic approaches. Oncogene. 32:5501–5511. 2013.PubMed/NCBI View Article : Google Scholar

38 

Yang L, Lin C, Jin C, Yang JC, Tanasa B, Li W, Merkurjev D, Ohgi KA, Meng D, Zhang J, et al: lncRNA-dependent mechanisms of androgen-receptor-regulated gene activation programs. Nature. 500:598–602. 2013.PubMed/NCBI View Article : Google Scholar

39 

Srikantan V, Zou Z, Petrovics G, Xu L, Augustus M, Davis L, Livezey JR, Connell T, Sesterhenn IA, Yoshino K, et al: PCGEM1, a prostate-specific gene, is overexpressed in prostate cancer. Proc Natl Acad Sci USA. 97:12216–12221. 2000.PubMed/NCBI View Article : Google Scholar

40 

Parolia A, Crea F, Xue H, Wang Y, Mo F, Ramnarine VR, Liu HH, Lin D, Saidy NR, Clermont PL, et al: The long non-coding RNA PCGEM1 is regulated by androgen receptor activity in vivo. Mol Cancer. 14(46)2015.PubMed/NCBI View Article : Google Scholar

41 

Hu R, Dunn TA, Wei S, Isharwal S, Veltri RW, Humphreys E, Han M, Partin AW, Vessella RL, Isaacs WB, et al: Ligand-independent androgen receptor variants derived from splicing of cryptic exons signify hormone-refractory prostate cancer. Cancer Res. 69:16–22. 2009.PubMed/NCBI View Article : Google Scholar

42 

Hu R, Lu C, Mostaghel EA, Yegnasubramanian S, Gurel M, Tannahill C, Edwards J, Isaacs WB, Nelson PS, Bluemn E, et al: Distinct transcriptional programs mediated by the ligand-dependent full-length androgen receptor and its splice variants in castration-resistant prostate cancer. Cancer Res. 72:3457–3462. 2012.PubMed/NCBI View Article : Google Scholar

43 

Khurana N, Kim H, Chandra PK, Talwar S, Sharma P, Abdel-Mageed AB, Sikka SC and Mondal D: Multimodal actions of the phytochemical sulforaphane suppress both AR and AR-V7 in 22Rv1 cells: Advocating a potent pharmaceutical combination against castration-resistant prostate cancer. Oncol Rep. 38:2774–2786. 2017.PubMed/NCBI View Article : Google Scholar

44 

Zhang Z, Zhou N, Huang J, Ho TT, Zhu Z, Qiu Z, Zhou X, Bai C, Wu F, Xu M and Mo YY: Regulation of androgen receptor splice variant AR3 by PCGEM1. Oncotarget. 7:15481–15491. 2016.PubMed/NCBI View Article : Google Scholar

45 

Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL, et al: Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 464:1071–1076. 2010.PubMed/NCBI View Article : Google Scholar

46 

Kogo R, Shimamura T, Mimori K, Kawahara K, Imoto S, Sudo T, Tanaka F, Shibata K, Suzuki A, Komune S, et al: Long noncoding RNA HOTAIR regulates polycomb-dependent chromatin modification and is associated with poor prognosis in colorectal cancers. Cancer Res. 71:6320–6326. 2011.PubMed/NCBI View Article : Google Scholar

47 

Nakagawa T, Endo H, Yokoyama M, Abe J, Tamai K, Tanaka N, Sato I, Takahashi S, Kondo T and Satoh K: Large noncoding RNA HOTAIR enhances aggressive biological behavior and is associated with short disease-free survival in human non-small cell lung cancer. Biochem Biophys Res Commun. 436:319–324. 2013.PubMed/NCBI View Article : Google Scholar

48 

Kim K, Jutooru I, Chadalapaka G, Johnson G, Frank J, Burghardt R, Kim S and Safe S: HOTAIR is a negative prognostic factor and exhibits pro-oncogenic activity in pancreatic cancer. Oncogene. 32:1616–1625. 2013.PubMed/NCBI View Article : Google Scholar

49 

Zhang A, Zhao JC, Kim J, Fong KW, Yang YA, Chakravarti D, Mo YY and Yu J: lncRNA HOTAIR Enhances the Androgen-receptor-mediated transcriptional program and drives castration-resistant prostate cancer. Cell Rep. 13:209–221. 2015.PubMed/NCBI View Article : Google Scholar

50 

Takayama K, Tsutsumi S, Katayama S, Okayama T, Horie-Inoue K, Ikeda K, Urano T, Kawazu C, Hasegawa A, Ikeo K, et al: Integration of cap analysis of gene expression and chromatin immunoprecipitation analysis on array reveals genome-wide androgen receptor signaling in prostate cancer cells. Oncogene. 30:619–630. 2011.PubMed/NCBI View Article : Google Scholar

51 

Takayama K, Horie-Inoue K, Katayama S, Suzuki T, Tsutsumi S, Ikeda K, Urano T, Fujimura T, Takagi K, Takahashi S, et al: Androgen-responsive long noncoding RNA CTBP1-AS promotes prostate cancer. EMBO J. 32:1665–1680. 2013.PubMed/NCBI View Article : Google Scholar

52 

Clarke RA, Zhao Z, Guo AY, Roper K, Teng L, Fang ZM, Samaratunga H, Lavin MF and Gardiner RA: New genomic structure for prostate cancer specific gene PCA3 within BMCC1: Implications for prostate cancer detection and progression. PLoS One. 4(e4995)2009.PubMed/NCBI View Article : Google Scholar

53 

de Kok JB, Verhaegh GW, Roelofs RW, Hessels D, Kiemeney LA, Aalders TW, Swinkels DW and Schalken JA: DD3(PCA3), a very sensitive and specific marker to detect prostate tumors. Cancer Res. 62:2695–2698. 2002.PubMed/NCBI

54 

van Bokhoven A, Varella-Garcia M, Korch C, Johannes WU, Smith EE, Miller HL, Nordeen SK, Miller GJ and Lucia MS: Molecular characterization of human prostate carcinoma cell lines. Prostate. 57:205–225. 2003.PubMed/NCBI View Article : Google Scholar

55 

Yang Z, Yu L and Wang Z: PCA3 and TMPRSS2-ERG gene fusions as diagnostic biomarkers for prostate cancer. Chin J Cancer Res. 28:65–71. 2016.PubMed/NCBI View Article : Google Scholar

56 

Lemos AE, Ferreira LB, Batoreu NM, de Freitas PP, Bonamino MH and Gimba ER: PCA3 long noncoding RNA modulates the expression of key cancer-related genes in LNCaP prostate cancer cells. Tumour Biol. 37:11339–11348. 2016.PubMed/NCBI View Article : Google Scholar

57 

Sung YY and Cheung E: Androgen receptor co-regulatory networks in castration-resistant prostate cancer. Endocr Relat Cancer. 21:R1–R11. 2013.PubMed/NCBI View Article : Google Scholar

58 

Lui GY, Kovacevic Z, Richardson V, Merlot AM, Kalinowski DS and Richardson DR: Targeting cancer by binding iron: Dissecting cellular signaling pathways. Oncotarget. 6:18748–18779. 2015.PubMed/NCBI View Article : Google Scholar

59 

Chen H, Zhou L, Wu X, Li R, Wen J, Sha J and Wen X: The PI3K/AKT pathway in the pathogenesis of prostate cancer. Front Biosci (Landmark Ed). 21:1084–1091. 2016.PubMed/NCBI View Article : Google Scholar

60 

Morgan TM, Koreckij TD and Corey E: Targeted therapy for advanced prostate cancer: Inhibition of the PI3K/Akt/mTOR pathway. Curr Cancer Drug Targets. 9:237–249. 2009.PubMed/NCBI View Article : Google Scholar

61 

Gao N, Zhang Z, Jiang BH and Shi X: Role of PI3K/AKT/mTOR signaling in the cell cycle progression of human prostate cancer. Biochem Biophys Res Commun. 310:1124–1132. 2003.PubMed/NCBI View Article : Google Scholar

62 

Kim SM, Park JH, Kim KD, Nam D, Shim BS, Kim SH and Ahn KS, Choi SH and Ahn KS: Brassinin induces apoptosis in PC-3 human prostate cancer cells through the suppression of PI3K/Akt/mTOR/S6K1 signaling cascades. Phytother Res. 28:423–431. 2014.PubMed/NCBI View Article : Google Scholar

63 

Vo BT, Morton D Jr, Komaragiri S, Millena AC, Leath C and Khan SA: TGF-β effects on prostate cancer cell migration and invasion are mediated by PGE2 through activation of PI3K/AKT/mTOR pathway. Endocrinology. 154:1768–1779. 2013.PubMed/NCBI View Article : Google Scholar

64 

Yuan JH, Yang F, Wang F, Ma JZ, Guo YJ, Tao QF, Liu F, Pan W, Wang TT, Zhou CC, et al: A long noncoding RNA activated by TGF-β promotes the invasion-metastasis cascade in hepatocellular carcinoma. Cancer Cell. 25:666–681. 2014.PubMed/NCBI View Article : Google Scholar

65 

Xu S, Yi XM, Tang CP, Ge JP, Zhang ZY and Zhou WQ: Long non-coding RNA ATB promotes growth and epithelial-mesenchymal transition and predicts poor prognosis in human prostate carcinoma. Oncol Rep. 36:10–22. 2016.PubMed/NCBI View Article : Google Scholar

66 

Xue M, Chen W and Li X: Urothelial cancer associated 1: A long noncoding RNA with a crucial role in cancer. J Cancer Res Clin Oncol. 142:1407–1419. 2016.PubMed/NCBI View Article : Google Scholar

67 

Ghiam AF, Taeb S, Huang X, Huang V, Ray J, Scarcello S, Hoey C, Jahangiri S, Fokas E, Loblaw A, et al: Long non-coding RNA urothelial carcinoma associated 1 (UCA1) mediates radiation response in prostate cancer. Oncotarget. 8:4668–4689. 2017.PubMed/NCBI View Article : Google Scholar

68 

Wu J, Cheng G, Zhang C, Zheng Y, Xu H, Yang H and Hua L: Long noncoding RNA LINC01296 is associated with poor prognosis in prostate cancer and promotes cancer-cell proliferation and metastasis. Onco Targets Ther. 10:1843–1852. 2017.PubMed/NCBI View Article : Google Scholar

69 

Wang J, Cheng G, Li X, Pan Y, Qin C, Yang H, Hua L and Wang Z: Overexpression of long non-coding RNA LOC400891 promotes tumor progression and poor prognosis in prostate cancer. Tumour Biol. 37:9603–9613. 2016.PubMed/NCBI View Article : Google Scholar

70 

Thorne H, Mitchell G and Fox S: kConFab: A familial breast cancer consortium facilitating research and translational oncology. J Natl Cancer Inst Monogr. 2011:79–81. 2011.PubMed/NCBI View Article : Google Scholar

71 

Du Z, Fei T, Verhaak RG, Su Z, Zhang Y, Brown M, Chen Y and Liu XS: Integrative genomic analyses reveal clinically relevant long noncoding RNAs in human cancer. Nat Struct Mol Biol. 20:908–913. 2013.PubMed/NCBI View Article : Google Scholar

72 

Prensner JR, Chen W, Han S, Iyer MK, Cao Q, Kothari V, Evans JR, Knudsen KE, Paulsen MT, Ljungman M, et al: The long non-coding RNA PCAT-1 promotes prostate cancer cell proliferation through cMyc. Neoplasia. 16:900–908. 2014.PubMed/NCBI View Article : Google Scholar

73 

Prensner JR, Zhao S, Erho N, Schipper M, Iyer MK, Dhanasekaran SM, Magi-Galluzzi C, Mehra R, Sahu A, Siddiqui J, et al: RNA biomarkers associated with metastatic progression in prostate cancer: A multi-institutional high-throughput analysis of SChLAP1. Lancet Oncol. 15:1469–1480. 2014.PubMed/NCBI View Article : Google Scholar

74 

Mehra R, Udager AM, Ahearn TU, Cao X, Feng FY, Loda M, Petimar JS, Kantoff P, Mucci LA and Chinnaiyan AM: Overexpression of the long Non-coding RNA SChLAP1 independently predicts lethal prostate cancer. Eur Urol. 70:549–552. 2016.PubMed/NCBI View Article : Google Scholar

75 

Mehra R, Shi Y, Udager AM, Prensner JR, Sahu A, Iyer MK, Siddiqui J, Cao X, Wei J, Jiang H, et al: A novel RNA in situ hybridization assay for the long noncoding RNA SChLAP1 predicts poor clinical outcome after radical prostatectomy in clinically localized prostate cancer. Neoplasia. 16:1121–1127. 2014.PubMed/NCBI View Article : Google Scholar

76 

Prensner JR, Iyer MK, Sahu A, Asangani IA, Cao Q, Patel L, Vergara IA, Davicioni E, Erho N, Ghadessi M, et al: The long noncoding RNA SChLAP1 promotes aggressive prostate cancer and antagonizes the SWI/SNF complex. Nat Genet. 45:1392–1398. 2013.PubMed/NCBI View Article : Google Scholar

77 

Ren S, Liu Y, Xu W, Sun Y, Lu J, Wang F, Wei M, Shen J, Hou J, Gao X, et al: Long noncoding RNA MALAT-1 is a new potential therapeutic target for castration resistant prostate cancer. J Urol. 190:2278–2287. 2013.PubMed/NCBI View Article : Google Scholar

78 

Viazzi F, Bonino B, Ratto E, Desideri G and Pontremoli R: Hyperuricemia, diabetes and hypertension. G Ital Nefrol. 32 (Suppl 62)(gin/32.S62.10)2015.PubMed/NCBI(In Italian).

79 

Mei YH, Yu JP and Li G: An extramedullary plasmacytoma in the kidney of a 14-year-old girl: Case report and review of the literature. Medicine (Baltimore). 96(e6092)2017.PubMed/NCBI View Article : Google Scholar

80 

Zhao Z, Chen C, Liu Y and Wu C: 17β-Estradiol treatment inhibits breast cell proliferation, migration and invasion by decreasing MALAT-1 RNA level. Biochem Biophys Res Commun. 445:388–393. 2014.PubMed/NCBI View Article : Google Scholar

81 

Ying L, Chen Q, Wang Y, Zhou Z, Huang Y and Qiu F: Upregulated MALAT-1 contributes to bladder cancer cell migration by inducing epithelial-to-mesenchymal transition. Mol Biosyst. 8:2289–2294. 2012.PubMed/NCBI View Article : Google Scholar

82 

Hu L, Wu Y, Tan D, Meng H, Wang K, Bai Y and Yang K: Up-regulation of long noncoding RNA MALAT1 contributes to proliferation and metastasis in esophageal squamous cell carcinoma. J Exp Clin Cancer Res. 34(7)2015.PubMed/NCBI View Article : Google Scholar

83 

Wang X, Li M, Wang Z, Han S, Tang X, Ge Y, Zhou L, Zhou C, Yuan Q and Yang M: Silencing of long noncoding RNA MALAT1 by miR-101 and miR-217 inhibits proliferation, migration, and invasion of esophageal squamous cell carcinoma cells. J Biol Chem. 290:3925–3935. 2015.PubMed/NCBI View Article : Google Scholar

84 

Hirata H, Hinoda Y, Shahryari V, Deng G, Nakajima K, Tabatabai ZL, Ishii N and Dahiya R: Long noncoding RNA MALAT1 promotes aggressive renal cell carcinoma through Ezh2 and interacts with miR-205. Cancer Res. 75:1322–1331. 2015.PubMed/NCBI View Article : Google Scholar

85 

Joaquin M and Watson RJ: Cell cycle regulation by the B-Myb transcription factor. Cell Mol Life Sci. 60:2389–2401. 2003.PubMed/NCBI View Article : Google Scholar

86 

Zhu M, Chen Q, Liu X, Sun Q, Zhao X, Deng R, Wang Y, Huang J, Xu M, Yan J and Yu J: lncRNA H19/miR-675 axis represses prostate cancer metastasis by targeting TGFBI. FEBS J. 281:3766–3775. 2014.PubMed/NCBI View Article : Google Scholar

87 

Prensner JR, Chen W, Iyer MK, Cao Q, Ma T, Han S, Sahu A, Malik R, Wilder-Romans K, Navone N, et al: PCAT-1, a long noncoding RNA, regulates BRCA2 and controls homologous recombination in cancer. Cancer Res. 74:1651–1660. 2014.PubMed/NCBI View Article : Google Scholar

88 

Gazy I, Zeevi DA, Renbaum P, Zeligson S, Eini L, Bashari D, Smith Y, Lahad A, Goldberg M, Ginsberg D and Levy-Lahad E: TODRA, a lncRNA at the RAD51 locus, is oppositely regulated to RAD51, and enhances RAD51-dependent DSB (Double Strand Break) repair. PLoS One. 10(e0134120)2015.PubMed/NCBI View Article : Google Scholar

89 

Tay Y, Rinn J and Pandolfi PP: The multilayered complexity of ceRNA crosstalk and competition. Nature. 505:344–352. 2014.PubMed/NCBI View Article : Google Scholar

90 

Jalali S, Bhartiya D, Lalwani MK, Sivasubbu S and Scaria V: Systematic transcriptome wide analysis of lncRNA-miRNA interactions. PLoS One. 8(e53823)2013.PubMed/NCBI View Article : Google Scholar

91 

Chiyomaru T, Yamamura S, Fukuhara S, Yoshino H, Kinoshita T, Majid S, Saini S, Chang I, Tanaka Y, Enokida H, et al: Genistein inhibits prostate cancer cell growth by targeting miR-34a and oncogenic HOTAIR. PLoS One. 8(e70372)2013.PubMed/NCBI View Article : Google Scholar

92 

Salehi S, Taheri MN, Azarpira N, Zare A and Behzad-Behbahani A: State of the art technologies to explore long non-coding RNAs in cancer. J Cell Mol Med. 21:3120–3140. 2017.PubMed/NCBI View Article : Google Scholar

93 

Tsai MC, Spitale RC and Chang HY: Long intergenic noncoding RNAs: New links in cancer progression. Cancer Res. 71:3–7. 2011.PubMed/NCBI View Article : Google Scholar

94 

Shechner DM, Hacisuleyman E, Younger ST and Rinn JL: Multiplexable, locus-specific targeting of long RNAs with CRISPR-display. Nat Methods. 12:664–670. 2015.PubMed/NCBI View Article : Google Scholar

95 

St Laurent G, Wahlestedt C and Kapranov P: The Landscape of long noncoding RNA classification. Trends Genet. 31:239–251. 2015.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

December-2020
Volume 13 Issue 6

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jiang G, Su Z, Liang X, Huang Y, Lan Z and Jiang X: Long non‑coding RNAs in prostate tumorigenesis and therapy (Review). Mol Clin Oncol 13: 76, 2020
APA
Jiang, G., Su, Z., Liang, X., Huang, Y., Lan, Z., & Jiang, X. (2020). Long non‑coding RNAs in prostate tumorigenesis and therapy (Review). Molecular and Clinical Oncology, 13, 76. https://doi.org/10.3892/mco.2020.2146
MLA
Jiang, G., Su, Z., Liang, X., Huang, Y., Lan, Z., Jiang, X."Long non‑coding RNAs in prostate tumorigenesis and therapy (Review)". Molecular and Clinical Oncology 13.6 (2020): 76.
Chicago
Jiang, G., Su, Z., Liang, X., Huang, Y., Lan, Z., Jiang, X."Long non‑coding RNAs in prostate tumorigenesis and therapy (Review)". Molecular and Clinical Oncology 13, no. 6 (2020): 76. https://doi.org/10.3892/mco.2020.2146