Taurine‑upregulated gene 1: A vital long non‑coding RNA associated with cancer in humans (Review)

  • Authors:
    • Wen‑Yu Wang
    • Yan‑Fen Wang
    • Pei Ma
    • Tong‑Peng Xu
    • Yong‑Qian Shu
  • View Affiliations

  • Published online on: September 12, 2017     https://doi.org/10.3892/mmr.2017.7472
  • Pages: 6467-6471
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

It is widely reported that long non‑coding RNAs (lncRNAs) are involved in regulating cell differentiation, proliferation, apoptosis and other biological processes. Certain lncRNAs have been found to be crucial in various types of tumor. Taurine‑upregulated gene 1 (TUG1) has been shown to be expressed in a tissue‑specific pattern and exert oncogenic or tumor suppressive functions in different types of cancer in humans. According to previous studies, TUG1 is predominantly located in the nucleus and may regulate gene expression at the transcriptional level. It mediates chromosomal remodeling and coordinates with polycomb repressive complex 2 (PRC2) to regulate gene expression. Although the mechanisms of how TUG1 affects the tumor genesis process remain to be fully elucidated, increasing studies have suggested that TUG1 offers potential as a diagnostic and prognostic biomarker, and as a therapeutic target in certain types of tumor. This review aims to summarize current evidence concerning the characteristics, mechanisms and associations with cancer of TUG1.

Introduction

Long non-coding RNAs (lncRNAs) are characterized as a subgroup of RNAs>200 nucleotides in length without, or with limited, protein-coding potential (13). Previous studies have demonstrated that certain lncRNAs have regulatory roles in diverse biological processes at the epigenetic, transcriptional and post-transcriptional levels due to their various structural and biochemical characteristics (49). The aberrant expression of lncRNAs has been shown in various human diseases, including cancer (Table I).

Table I.

Effects of taurine-upregulated gene 1in various types of cancer.

Table I.

Effects of taurine-upregulated gene 1in various types of cancer.

Author, yearCancerExpressionClinical significanceFunctionAssociated factors(Refs.)
Zhang et al, 2014Non-small cell lung cancerDownregulatedPrognosisProliferation; cell cycleP53; PRC2; HOXB7(15)
Li et al, 2016GliomaDownregulatedPathological stage; tumor sizeNot reportedNot reported(16)
Zhang et al, 2013OsteosarcomaUpregulatedNot reportedNot reportedNot reported(17)
Han et al, 2013; Tan et al, 2015Bladder cancerUpregulatedTNM stage; overall survivalInvasion; radioresistancemiR145; ZEB2(18,19)
Sun et al, 2016Colorectal cancerUpregulatedPrognosisProliferation; invasion; metastasisHDAC1(20)
Xu et al, 2015Esophageal squamous cell carcinomaUpregulatedFamily history; tumor locationProliferation; migrationNot reported(21)
Zhang et al, 2016GastricUpregulatedInvasion depth; TNM stage; overall survivalProliferation; apoptosis; cell cycleEZH2; p57(22)
Huang et al, 2015Hepatocellular carcinomaUpregulatedTumor size; BCLC stageProliferation; apoptosis; cell cycleSP1; KLF2(23)
Isin et al, 2014Malignant melanomaUpregulatedStageNot reportedNot reported(35)

[i] PRC2, polycomb repressive complex 2; HOXB7, homeobox B7; miR, microRNA; ZEB2, Zinc finger E-box binding homeobox 2; HDAC1, histone deacetylase 1; EZH2, enhancer of zeste homolog 2; SP1, stimulatory protein 1; KLF2, Kruppel-like factor 2; BCLC, Barcelona Clinic Liver Cancer.

Taurine-upregulated gene 1 (TUG1), a 7.1-kb lncRNA, was originally detected in a genomic screen for genes upregulated in response to taurine treatment in developing mouse retinal cells (10). It is located on chromosome 22q12.2 in the human genome, and has been reported to be expressed in a tissue-specific pattern and to exert oncogenic or tumor suppressive functions in different types of cancer in humans (1114). The downregulation of TUG1 has been detected in glioma and non-small-cell lung cancer (NSCLC), with TUG1 shown to induce apoptosis as a tumor suppressor (15,16). By contrast, the overexpression of TUG1 has been reported in osteosarcoma (17), bladder cancer (18,19), colorectal cancer (CRC) (20), esophageal squamous cell carcinoma (ESCC) (21), gastric cancer (22) and hepatocellular cancer (HCC) (23). TUG1 was shown to function as an oncogene by promoting cell proliferation and was correlated with a poor prognosis (1723).

Polycomb repressive complex 2 (PRC2) is a methyltransferase, which is composed of enhancer of zeste homolog 2 (EZH2), suppressor of zeste 12 (SUZ12) and embryonic ectoderm development, and is capable of catalyzing the di- and trimethylation of lysine residue 27 of histone 3 (H3K27me3), which regulates gene expression. Various lncRNAs, including TUG1, modulate specific genetic loci by recruiting and binding to PRC2 protein complexes, and PRC2-mediated epigenetic regulation is vital in tumorigenesis and development (2427). The knockdown of TUG1 results in wide changes in gene expression, particularly the upregulation of cell-cycle genes, indicating that TUG1 is important in cell proliferation and apoptosis through effects on the cell cycle (22). However, the comprehensive mechanisms remain to be fully elucidated.

TUG1 in human cancer

Colorectal cancer (CRC)

It was previously reported that the expression of TUG1 was significantly enhanced in CRC tumor tissues, compared with that in paratumor tissues. Further analysis showed that the expression of TUG1 was negatively correlated with overall survival rates in patients (20). The stable knockdown of histone deacetylase 1 (HDAC1) induced the expression of TUG1, indicating that the expression of TUG1 is regulated by histone modification (20,28). In vitro experiments have confirmed that the knockdown of TUG1 suppresses the colony formation, migration and invasion of CRC cells in vitro. In addition, an in vivo liver metastasis model revealed that the overexpression of TUG1 increased the number of metastatic tumor nodules in the liver, indicating that TUG1 promoted CRC metastasis (20). The molecular mechanism by which TUG1 promotes the invasion and metastasis of CRC has also been investigated. It was demonstrated that the overexpression of TUG1 reduced the expression of E-cadherin, and upregulated the expression levels of N-cadherin, vimentin and fibronectin, whereas knockdown of the expression of TUG1 showed the opposite effects. This suggested that TUG1 may affect CRC metastasis and invasion through mediating epithelial-mesenchymal transition (EMT)-associated gene expression (20,29). However, the mechanisms by which HDAC1 affects TUG1 and regulates EMT require further investigation.

Bladder cancer

The expression of TUG1 was also found to be upregulated in bladder cancer tissues and cell lines. A higher expression of TUG1 was found to be associated with poorer tumor-necrosis-metastasis (TNM) staging and shorter overall survival rates (18,19). Subsequent investigations revealed that TUG1 promoted bladder cancer cell invasion and radioresistance. The expression level of epithelial markers increased whereas those of mesenchymal markers decreased following the overexpression of TUG1, indicating that TUG1 was involved in bladder cancer through EMT (19,29). TUG1 acted as a microRNA (miRNA) sponge, as miRNA (miR)-145 was able to bind to TUG1 and exhibit reciprocal regulatory effects. In addition, Zinc finger E-box binding homeobox 2 (ZEB2), a transcription factor regulating the EMT marker E-cadherin (30), has been identified as a direct target of miR-145 (31). The evidence above indicates a possible mechanism by which TUG1 is involved in EMT and the radioresistance in bladder cancer through the miR-145/ZEB2 axis.

Hepatocellular cancer (HCC)

A previous study detected an upregulation in the expression of TUG1 in HCC tissues, which was confirmed to be associated with tumor size and Barcelona Clinic Liver Cancer stage (23). The transcription factor, stimulatory protein 1 (SP1) was later confirmed to directly bind to TUG1 promoter regions and positively regulate the expression of TUG1. In previous in vitro and xenograft model experiments, the functions of TUG1 in inhibiting cell proliferation and inducing cell apoptosis in HCC were demonstrated. Kruppel-like factor 2 (KLF2) was identified as a novel downstream gene of TUG1, which was found to be involved in HCC cell G0/G1 arrest, suppression of cell proliferation and the induction of apoptosis. TUG1 inhibited the transcription of KLF2 through binding to EZH2/SUZ12, the core subunits of PRC2, to the KLF2 gene promoter locus in HCC cells, thus acting as an oncogenic factor (32).

Gastric cancer (GC)

A previous study observed that TUG1 was overexpressed in GC cells, and was positively correlated with invasion depth and TNM stage, but negatively correlated with overall survival rates (22). In vitro and in vivo experiments confirmed that TUG1 suppressed GC cell proliferation through its effects on cell cycle progression in a pattern of G0/G1 arrest. Subsequent investigation of the mechanism demonstrated that TUG1 specifically targeted EZH2 and epigenetically regulated the expression of cyclin-dependent kinase inhibitor (CKI) family members, including p15, p16, p21, p27 and p57 (22,33,34). Analysis of the mechanism showed that TUG1 epigenetically repressed CKIs by binding to PRC2, and thereby regulated the cell cycle to promote GC cell proliferation.

ESCC

It was previously found that TUG1 was overexpressed in ESCC. Patients with a family history of esophageal cancer and upper segment ESCC were shown to express higher levels of TUG1. TUG1 also promoted the proliferation and migration of ESCC cell lines in vitro (21).

Osteosarcoma

TUG1 is upregulated in osteosarcoma tissues and cells (17). Experiments have demonstrated that TUG1 acts as an oncogenic gene via increasing osteosarcoma cell proliferation and affecting apoptosis. However, the detailed mechanisms require further investigation.

Multiple myeloma (MM)

It has been reported that, in the plasma of patients with MM, the expression of TUG1 was upregulated and showed marked association with clinical stages (35).

Glioma

Unlike the overexpression of TUG1 found in the types of cancer described above, TUG1 was downregulated in human glioma tissues and cells, and negatively associated with advanced pathological progression, serving as an indicator of poor prognosis (16). TUG1 exerts its antitumor function in glioma through promoting cell apoptosis via intrinsic pathways mediated by caspase-3 together with caspase-9, and simultaneously suppressing anti-apoptotic pathways mediated by B-cell lymphoma 2 (3639).

Non-small-cell lung cancer (NSCLC)

The expression of TUG1 was also shown to be downregulated in human NSCLC tissues and correlated with poor prognosis of patients (15). TUG1 has been shown to modulate NSCLC cell proliferation in vitro and in vivo through alterations in cell cycle progression. Further analysis demonstrated that p53 was able to directly bind to the promoter region of TUG1 and regulate the expression of TUG1. In addition, investigations have suggested that homeobox B7 (HOXB7), a known oncogene, is a downstream gene of TUG1, and it was suggested that TUG1 targets PRC2 to regulate HOXB7 at the transcriptional level (40,41). The role of HOXB7 has also been investigated, which showed that HOXB7 promoted NSCLC cell proliferation via activating the AKT and mitogen-activated protein kinase pathways (15,38,39,42,43). Therefore, the p53/TUG1/PRC2/HOXB7 axis was found to be vital in the tumorigenesis and progression of NSCLC, which may be a target for future therapy.

Conclusions and perspectives

In previous years, widespread investigations have been performed on the biological roles and clinical significance of lncRNAs. The deregulation of lncRNAs is capable of affecting tumor development, functioning as tumor inducers or suppressors. One of the most comprehensively investigated lncRNAs is the hox transcript antisense intergenic RNA (HOTAIR). HOTAIR has been identified as an oncogenic gene, recruiting PRC2 and interacting with lysine-specific demethylase1 (LSD1), regulating the expression of its downstream targets (8,11,42).

This review discusses TUG1 and its association with cancer in humans. TUG1 is expressed in a tissue-specific pattern, showing oncogenic or tumor inhibiting capacities in different types of cancer in humans (Table I). The downregulation of TUG1 is observed in glioma and NSCLC, showing that TUG1 serves as a tumor suppressor. By contrast, the overexpression of TUG1 has been reported in ESCC, CRC, HCC, bladder cancer and osteosarcoma, indicating that TUG1 functions as an oncogene. The mechanisms underlying the biological functions of TUG1 are shown in Figs. 1 and 2. These findings indicate that the expression of TUG1 can be enhanced or weakened by upstream or downstream interference to slow down or alter tumor progression. The level of TUG1 in tumor tissues correlates with tumor stage and prognosis, which may be utilized as a diagnostic and prognostic biomarker clinically. In a previous study, TUG1 was moderately elevated in secreted exosomes (44), and this may enable the isolation of exosomes from blood plasma or serum to assist in monitoring the state of disease dynamically (45). However, further investigations are required in order to fully elucidate the mechanisms underlying TUG1 and cancer development. TUG1 may offer potential as a novel diagnostic biomarker and therapeutic approach for clinical utilization.

Acknowledgements

This review was supported by grants from National Natural Science Foundation of China (grant no. 81272532).

References

1 

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. View Article : Google Scholar : PubMed/NCBI

2 

Mercer TR, Dinger ME and Mattick JS: Long non-coding RNAs: Insights into functions. Nat Rev Genet. 10:155–159. 2009. View Article : Google Scholar : PubMed/NCBI

3 

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

4 

Amaral PP and Mattick JS: Noncoding RNA in development. Mamm Genome. 19:454–492. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Blackshaw S, Harpavat S, Trimarchi J, Cai L, Huang H, Kuo WP, Weber G, Lee K, Fraioli RE, Cho SH, et al: Genomic analysis of mouse retinal development. PLoS Biol. 2:e2472004. View Article : Google Scholar : PubMed/NCBI

6 

Dinger ME, Amaral PP, Mercer TR, Pang KC, Bruce SJ, Gardiner BB, Askarian-Amiri ME, Ru K, Soldà G, Simons C, et al: Long noncoding RNAs in mouse embryonic stem cell pluripotency and differentiation. Genome Res. 18:1433–1445. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Cesana M, Cacchiarelli D, Legnini I, Santini T, Sthandier O, Chinappi M, Tramontano A and Bozzoni I: A long noncoding RNA controls muscle differentiation by functioning as a competing endogenous RNA. Cell. 147:358–369. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, Goodnough LH, Helms JA, Farnham PJ, Segal E and Chang HY: Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell. 129:1311–1323. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Ginger MR, Shore AN, Contreras A, Rijnkels M, Miller J, Gonzalez-Rimbau MF and Rosen JM: A noncoding RNA is a potential marker of cell fate during mammary gland development. Proc Natl Acad Sci USA. 103:5781–5786. 2006; View Article : Google Scholar : PubMed/NCBI

10 

Young TL, Matsuda T and Cepko CL: The noncoding RNA taurine upregulated gene 1 is required for differentiation of the murine retina. Curr Biol. 15:501–512. 2005. View Article : Google Scholar : PubMed/NCBI

11 

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. View Article : Google Scholar : PubMed/NCBI

12 

Khaitan D, Dinger ME, Mazar J, Crawford J, Smith MA, Mattick JS and Perera RJ: The melanoma-upregulated long noncoding RNA SPRY4-IT1 modulates apoptosis and invasion. Cancer Res. 71:3852–3862. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Yuan SX, Yang F, Yang Y, Tao QF, Zhang J, Huang G, Yang Y, Wang RY, Yang S, Huo XS, et al: Long noncoding RNA associated with microvascular invasion in hepatocellular carcinoma promotes angiogenesis and serves as a predictor for hepatocellular carcinoma patients' poor recurrence-free survival after hepatectomy. Hepatology. 56:2231–2241. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Ji P, Diederichs S, Wang W, Böing S, Metzger R, Schneider PM, Tidow N, Brandt B, Buerger H, Bulk E, et al: MALAT-1, a novel noncoding RNA and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene. 22:8031–8041. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Zhang EB, Yin DD, Sun M, Kong R, Liu XH, You LH, Han L, Xia R, Wang KM, Yang JS, et al: P53-regulated long non-coding RNA TUG1 affects cell proliferation in human non-small cell lung cancer, partly through epigenetically regulating HOXB7 expression. Cell Death Dis. 5:e12432014. View Article : Google Scholar : PubMed/NCBI

16 

Li J, Zhang M, An G and Ma Q: LncRNA TUG1 acts as a tumor suppressor in human glioma by promoting cell apoptosis. Exp Biol Med (Maywood). 241:644–649. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Zhang Q, Geng PL, Yin P, Wang XL, Jia JP and Yao J: Down-regulation of long non-coding RNA TUG1 inhibits osteosarcoma cell proliferation and promotes apoptosis. Asian Pac J Cancer Prev. 14:2311–2315. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Han Y, Liu Y, Gui Y and Cai Z: Long intergenic non-coding RNA TUG1 is overexpressed in urothelial carcinoma of the bladder. J Surg Oncol. 107:555–559. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Tan J, Qiu K, Li M and Liang Y: Double-negative feedback loop between long non-coding RNA TUG1 and miR-145 promotes epithelial to mesenchymal transition and radioresistance in human bladder cancer cells. FEBS Lett. 589:3175–3181. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Sun J, Ding C, Yang Z, Liu T, Zhang X, Zhao C and Wang J: The long non-coding RNA TUG1 indicates a poor prognosis for colorectal cancer and promotes metastasis by affecting epithelial-mesenchymal transition. J Transl Med. 14:422016. View Article : Google Scholar : PubMed/NCBI

21 

Xu Y, Wang J, Qiu M and Xu L, Li M, Jiang F, Yin R and Xu L: Upregulation of the long noncoding RNA TUG1 promotes proliferation and migration of esophageal squamous cell carcinoma. Tumour Biol. 36:1643–1651. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Zhang E, He X, Yin D, Han L, Qiu M, Xu T, Xia R, Xu L, Yin R and De W: Increased expression of long noncoding RNA TUG1 predicts a poor prognosis of gastric cancer and regulates cell proliferation by epigenetically silencing of p57. Cell Death Dis. 7:e21092016. View Article : Google Scholar : PubMed/NCBI

23 

Huang MD, Chen WM, Qi FZ, Sun M, Xu TP, Ma P and Shu YQ: Long non-coding RNA TUG1 is up-regulated in hepatocellular carcinoma and promotes cell growth and apoptosis by epigenetically silencing of KLF2. Mol Cancer. 14:1652015. View Article : Google Scholar : PubMed/NCBI

24 

Paul TA, Bies J, Small D and Wolff L: Signatures of polycomb repression and reduced H3K4 trimethylation are associated with p15INK4b DNA methylation in AML. Blood. 115:3098–3108. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Aoki R, Chiba T, Miyagi S, Negishi M, Konuma T, Taniguchi H, Ogawa M, Yokosuka O and Iwama A: The polycomb group gene product Ezh2 regulates proliferation and differentiation of murine hepatic stem/progenitor cells. J Hepatol. 52:854–863. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Chen H, Gu X, Su IH, Bottino R, Contreras JL, Tarakhovsky A and Kim SK: Polycomb protein Ezh2 regulates pancreatic beta-cell Ink4a/Arf expression and regeneration in diabetes mellitus. Genes Dev. 23:975–985. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Fan T, Jiang S, Chung N, Alikhan A, Ni C, Lee CC and Hornyak TJ: EZH2-dependent suppression of a cellular senescence phenotype in melanoma cells by inhibition of p21/CDKN1A expression. Mol Cancer Res. 9:418–429. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Zilio N, Codlin S, Vashisht AA, Bitton DA, Head SR, Wohlschlegel JA, Bähler J and Boddy MN: A novel histone deacetylase complex in the control of transcription and genome stability. Mol Cell Biol. 34:3500–3514. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Thiery JP, Acloque H, Huang RY and Nieto MA: Epithelial-mesenchymal transitions in development and disease. Cell. 139:871–890. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Vandewalle C, Comijn J, De Craene B, Vermassen P, Bruyneel E, Andersen H, Tulchinsky E, Van Roy F and Berx G: SIP1/ZEB2 induces EMT by repressing genes of different epithelial cell-cell junctions. Nucleic Acids Res. 33:6566–6578. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Fernandez-Zapico ME, Lomberk GA, Tsuji S, DeMars CJ, Bardsley MR, Lin YH, Almada LL, Han JJ, Mukhopadhyay D, Ordog T, et al: A functional family-wide screening of SP/KLF proteins identifies a subset of suppressors of KRAS-mediated cell growth. Biochem J. 435:529–537. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Kotake Y, Nakagawa T, Kitagawa K, Suzuki S, Liu N, Kitagawa M and Xiong Y: Long non-coding RNA ANRIL is required for the PRC2 recruitment to and silencing of p15(INK4B) tumor suppressor gene. Oncogene. 30:1956–1962. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Prensner JR, Iyer MK, Balbin OA, Dhanasekaran SM, Cao Q, Brenner JC, Laxman B, Asangani IA, Grasso CS, Kominsky HD, et al: Transcriptome sequencing across a prostate cancer cohort identifies PCAT-1, an unannotated lincRNA implicated in disease progression. Nat Biotechnol. 29:742–749. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Isin M, Ozgur E, Cetin G, Erten N, Aktan M, Gezer U and Dalay N: Investigation of circulating lncRNAs in B-cell neoplasms. Clin Chim Acta. 431:255–259. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Ghavami S, Hashemi M, Ande SR, Yeganeh B, Xiao W, Eshraghi M, Bus CJ, Kadkhoda K, Wiechec E, Halayko AJ and Los M: Apoptosis and cancer: Mutations within caspase genes. J Med Genet. 46:497–510. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Brentnall M, Rodriguez-Menocal L, De Guevara RL, Cepero E and Boise LH: Caspase-9, caspase-3 and caspase-7 have distinct roles during intrinsic apoptosis. BMC Cell Biol. 14:322013. View Article : Google Scholar : PubMed/NCBI

38 

Liao WT, Jiang D, Yuan J, Cui YM, Shi XW, Chen CM, Bian XW, Deng YJ and Ding YQ: HOXB7 as a prognostic factor and mediator of colorectal cancer progression. Clin Cancer Res. 17:3569–3578. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Jin K, Kong X, Shah T, Penet MF, Wildes F, Sgroi DC, Ma XJ, Huang Y, Kallioniemi A, Landberg G, et al: The HOXB7 protein renders breast cancer cells resistant to tamoxifen through activation of the EGFR pathway. Proc Natl AcadSci USA. 109:2736–2741. 2012; View Article : Google Scholar

40 

Storti P, Donofrio G, Colla S, Airoldi I, Bolzoni M, Agnelli L, Abeltino M, Todoerti K, Lazzaretti M, Mancini C, et al: HOXB7 expression by myeloma cells regulates their pro-angiogenic properties in multiple myeloma patients. Leukemia. 25:527–537. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Yuan W, Zhang X, Xu Y, Li S, Hu Y and Wu S: Role of HOXB7 in regulation of progression and metastasis of human lung adenocarcinoma. Mol Carcinog. 53:49–57. 2014. View Article : Google Scholar : PubMed/NCBI

42 

di Pietro M, Lao-Sirieix P, Boyle S, Cassidy A, Castillo D, Saadi A, Eskeland R and Fitzgerald RC: Evidence for a functional role of epigenetically regulated midcluster HOXB genes in the development of Barrett esophagus. Proc Natl Acad Sci USA. 109:9077–9082. 2012; View Article : Google Scholar : PubMed/NCBI

43 

Wu X, Chen H, Parker B, Rubin E, Zhu T, Lee JS, Argani P and Sukumar S: HOXB7, a homeodomain protein, is overexpressed in breast cancer and confers epithelial-mesenchymal transition. Cancer Res. 66:9527–9534. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Gezer U, Özgür E, Cetinkaya M, Isin M and Dalay N: Long non-coding RNAs with low expression levels in cells are enriched in secreted exosomes. Cell Biol Int. 38:1076–1079. 2014.PubMed/NCBI

45 

Bang C and Thum T: Exosomes: New players in cell-cell communication. Int J Biochem Cell Biol. 44:2060–2064. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 16 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang WY, Wang YF, Ma P, Xu TP and Shu YQ: Taurine‑upregulated gene 1: A vital long non‑coding RNA associated with cancer in humans (Review). Mol Med Rep 16: 6467-6471, 2017
APA
Wang, W., Wang, Y., Ma, P., Xu, T., & Shu, Y. (2017). Taurine‑upregulated gene 1: A vital long non‑coding RNA associated with cancer in humans (Review). Molecular Medicine Reports, 16, 6467-6471. https://doi.org/10.3892/mmr.2017.7472
MLA
Wang, W., Wang, Y., Ma, P., Xu, T., Shu, Y."Taurine‑upregulated gene 1: A vital long non‑coding RNA associated with cancer in humans (Review)". Molecular Medicine Reports 16.5 (2017): 6467-6471.
Chicago
Wang, W., Wang, Y., Ma, P., Xu, T., Shu, Y."Taurine‑upregulated gene 1: A vital long non‑coding RNA associated with cancer in humans (Review)". Molecular Medicine Reports 16, no. 5 (2017): 6467-6471. https://doi.org/10.3892/mmr.2017.7472