Open Access

Recent advances in unraveling the molecular mechanisms and functions of HOXA11‑AS in human cancers and other diseases (Review)

  • Authors:
    • Cheng Wei
    • Liangjuan Zhao
    • Hao Liang
    • Yingwei Zhen
    • Lei Han
  • View Affiliations

  • Published online on: March 19, 2020     https://doi.org/10.3892/or.2020.7552
  • Pages: 1737-1754
  • Copyright: © Wei et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

A large number of previously published research articles have demonstrated that the expression levels of long noncoding RNAs (lncRNAs) are generally dysregulated, either through overexpression or underexpression, in cancer and other types of disease. As a recently discovered lncRNA, HOXA11 antisense RNA (HOXA11‑AS) is able to serve as an oncogenic or tumor‑suppressor gene and serves a vital role in the processes of proliferation, invasion, and migration of cancer cells. HOXA11‑AS appears to be a major factor contributing to epigenetic modification, and exerts transcriptional, post‑transcriptional, translational and post‑translational regulatory effects on genes through a variety of mechanisms; for example, by competing endogenous RNA (ceRNA) and a molecular scaffold mechanism. A number of reports have demonstrated that HOXA11‑AS functions as a protein scaffold for polycomb repressive complex 2 (PRC2), lysine‑specific histone demethylase 1 (LSD1) and DNA methyltransferase 1 (DNMT1) to perform epigenetic modifications on chromosomes in the nucleus. Furthermore, HOXA11‑AS is also located in the cytoplasm and can act as a ceRNA, which sponges miRNAs. In addition, HOXA11‑AS may be useful as a biomarker for the diagnosis and prognosis of cancer. In the present review article, the clinical value, phenotype and mechanism of HOXA11‑AS in a variety of tumors types are briefly summarized, as well as its clinical value in certain additional diseases. The perspective of the authors is that HOXA11‑AS may represent an effective tumor marker and therapeutic target for cancer diagnosis and therapy.

Introduction

With the incidence of cancer and mortality rates rising rapidly worldwide, cancer is anticipated to become both the most significant obstacle to extending life expectancy, and the major cause of death worldwide. Cancer has already become the first or second main cause of mortality in 91 of 172 countries for persons aged 70 or less, and is ranked third or fourth in a further 22 countries, according to data published by the World Health Organization (WHO) in 2015 (1). During 2018, over 18.1 million new cases of cancer and 9.6 million cancer deaths were estimated to have occurred worldwide. For both the sexes, in 2018 Asia accounted for nearly one-half of all cancer cases, and more than one-half of cancer-associated deaths globally (1). Despite improvements in radiotherapy, chemotherapy and surgical treatment technologies, various types of cancer remain very difficult to treat, such as gastric cancer (GC), non-small cell lung cancer (NSCLC) and bladder cancer (BC) (2,3). Therefore, study of the pathogenesis and therapeutic targets of cancer is imperative.

The transcriptional background of all organisms has been shown to be much more complicated than was at first envisioned, since various protein-coding RNAs and noncoding RNAs (ncRNAs) are extensively transcribed from large portions of genomic sequences (4,5). Thus, to resolve these problems, a clear understanding of the intracellular transcriptional environment must be obtained. Benefiting from the development of new generation sequencing technologies, it has been shown that, in humans, over 70% of the genome sequence is transcribed. Among these transcripts, less than 2% of the transcripts contain protein-coding RNAs, whereas the remaining transcripts produce ncRNAs (69). Previously, it was considered that the products of most non-protein-coding genes were without any function, simply produced during the process of gene transcription. However, with the development of research in this area, there is a new understanding that the products of these non-protein-coding genes are involved in the regulation of a variety of biological processes in cells (69). At present, ncRNAs are a hot research topic in the life sciences.

NcRNAs comprise regulatory RNAs (for instance, miRNAs) and structural RNAs [such as small nuclear RNAs (snRNAs), ribosomal RNAs (rRNAs) and transfer RNAs (tRNAs)] (10). Thanks to recent advances in the acquisition of genome and transcriptome sequencing data, the catalogue of regulatory molecules now contains numerous long noncoding RNAs (lncRNAs), a loosely classified group of long RNA transcripts that lack protein-coding function (11,12). As early as 1991, it was found that X-inactive specific transcript (XIST) is involved in regulating X chromosome inactivation, providing an initial insight into lncRNA functionality (13,14). At present, 96,308 lncRNA genes as well as 172,216 lncRNA transcripts, have been identified in humans [data from NONCODE, the ncRNA database including sequence, documentation and analysis (http://www.bioinfo.org/noncode/)], which influence a great variety of cellular biological processes. lncRNAs were undervalued for a long time, being considered as merely transcriptional ‘garbage’ across the entire length of the genome (15). However, the expression of lncRNAs is now known to be dysregulated in a great majority of tumor types, including lung cancer (16), hepatocellular carcinoma (HCC) (17), glioma (18), colorectal cancer (CRC) (19), breast cancer (BC) (20), osteosarcoma (21), ovarian cancer (22), gastric cancer (GC) (23), and esophageal squamous cell carcinoma (ESCC) (24). Emerging studies have now shown that lncRNA has a vital role in a wide variety of cellular biological processes, including cellular differentiation, cell cycle, proliferation, migration, metabolism, as well as apoptosis, especially in tumor cells (25,26). These findings suggest that lncRNA functions as an oncogene or tumor suppressor through interaction with other biomolecules or via chromatin modifications (27).

lncRNAs are able to interfere in numerous cellular processes, including chromatin organization, transcription complex recruitment, post-transcriptional regulation, translation, and post-translational processes (12,28,29). Several of the main mechanisms of lncRNAs are discussed below.

Chromatin organization

The dosage compensation effect of the X chromosome in mammals provides a striking instance of lncRNA-mediated chromatin regulation. Xist, one of the first functionally annotated lncRNAs, regulates dosage compensation in female mammals by localizing to the X chromosome, and recruiting various factors directly or indirectly to accomplish X chromosome inactivation (XCI) (30). In brief, dosage compensation refers to the process by which the gene expression level of the two X chromosomes in female cells is made equivalent to the one X chromosome in male cells. The expression of lncRNA Xist in female cells is from one of the two X chromosomes, which subsequently changes the chromatin structure of the whole chromosome, whereby most of the genes in the inactive X chromosome are silenced during transcription (3133). Significantly, Xist can interact with polycomb repressive complex 2 (PRC2) via a structural domain called Repeat A, resulting in PRC2 and its cognate histone marker histone H3 lysine 27 trimethylation (H3K27me3) being located on the inactive X chromosome. lncRNAs can also recruit PRC2 to modulate distal genes throughout the genome (34). In addition, lncRNAs expressed only in embryonic stem cells are able to directly interact with chromatin to regulate gene expression and maintain pluripotency (35). The interaction between DNA and lncRNA is achieved either via sequence complementation or via combination in helical structures (28). Therefore, lncRNAs are associated with organization of the nuclear architecture and the general structuring of the genome, thereby affecting the expression of related genes (36).

Transcriptional regulation

At the transcriptional level, lncRNA is often used as a molecular scaffold to recruit two or more proteins to the promoter regions of their target genes, and modulates the transcription of the target genes (3739). These proteins include zinc-finger proteins, DNA methyltransferases, transcription factors (TFs), and other transcriptional regulators (40). Furthermore, lncRNA can act as an adaptor to recruit associated proteins into discrete complexes (41). For instance, the lncRNA HOTAIR synchronously binds both PRC2 and LSD1-CoREST complexes through specific structural domains of the RNA (42). This interaction effectively harmonizes the methylation of H3K27 and demethylation of H3K4me2, guaranteeing that gene silencing occurs. Functioning as a molecular scaffold is among the primary mechanisms by which HOXA11-AS functions.

Post-transcriptional regulation

At the post-transcriptional level, lncRNAs operate in diverse ways to affect miRNAs and mRNAs (43,44). First, lncRNAs are hypothesized to act as competing endogenous RNA (ceRNA), or as ‘RNA sponges’, which are able to interact with miRNAs, thereby blocking the interaction between miRNAs and mRNAs, thus decreasing their regulatory impact on target mRNAs (45,46). For example, HOXA11-AS serves as a ceRNA to modulate the expression level of the transcription factor Sp1 by sequestering miR-124. Findings demonstrated that improvements in cell invasion and proliferation mediated by HOXA11-AS were reversed by miR-124 (47). Secondly, lncRNAs can be precursors of miRNAs, which are able to modulate different processes in miRNA production and have microprocessor activity to complete primary transcripts via a mechanism independent of polyadenylation (48). For instance, lncRNA LOC554202 was shown to be the putative precursor of miR-31, exerting an important role in preventing metastasis of BC (49). In addition, the maturation of miR-145 can be hindered by lncRNA colon cancer-associated transcript 2 (CCAT2) via the repression of Dicer cleavage and cytoplasmic export (50). Moreover, lncRNAs also affect alternative splicing and the stability of mRNA. For example, first apoptosis signal (Fas)-antisense lncRNA-SAF complex is able to combine with the human splicing factor SPF45, resulting in removal of exon 6 during Fas splicing and the production of a soluble Fas protein that serves to inhibit Fas/Fas ligand (FASL)-mediated apoptosis in different human cell lines (51). In addition, lncRNA PDCD4-AS1 stabilizes programmed cell death 4 (PDCD4) RNA via formation of an RNA duplex that dictates the mutual effect between RNA decay promoting factors and PDCD4 RNA, in human BC (52).

Recently, numerous studies (19,53) have reported that exosomes can function as autocrine or paracrine factors to influence the significant biological functions that mediate intercellular interactions. An increasing amount of evidence has shown that cancer cells are able to release exosomes, and exosome-transmitted lncRNAs are able to promote tumor metastasis, angiogenesis and drug resistance. For example, lncRNA-APC1 is able to inhibit the production of exosomes, and reduce their stability by directly binding Rab5b mRNA, thereby inhibiting the growth, metastasis and tumor angiogenesis of CRC cells (19). Additionally, Qu et al (53) found that bioactive lncARSR [lncRNA activated in renal cell carcinoma (RCC) with sunitinib resistance] can be integrated into exosomes and delivered to sensitive cells in RCC. When exosomes containing lncARSR reached the sensitive RCC cells, lncARSR was released into the cytoplasm. The expression of the receptor tyrosine kinases AXL and c-MET in RCC cells was promoted by lncARSR competitively binding miR-34/miR-449, thus promoting sunitinib resistance. AXL and c-MET are responsible for lncARSR-mediated sunitinib resistance in RCC (53).

Translational process

In addition to the abovementioned effects, lncRNAs are also able to promote or inhibit the translational process. For example, dopaminergic neurons specifically express ubiquitin carboxy-terminal hydrolase L1 (Uchl1). Uchl1 can be modulated by its antisense transcript (AS Uchl1), which binds polysomes through its repetitive domain termed ‘SINEB2’ to facilitate cap-independent translation (54). Furthermore, lncRNA-p21, a post-transcriptional modulator, passively modulates translation of the transcripts of the transcription factor JUNB and β-catenin through incomplete complementary base pairing at diverse sites in the coding and non-coding regions [both 5′- and 3′-untranslated regions (UTRs)] of JUNB (8 sites) and β-catenin (15 sites) mRNA, leading to the formation of an lncRNA-p21-mRNA complex. The communication between mRNAs and Fragile X mental retardation protein (FMRP), as well as the translational repressor RCK, may be improved by the lncRNA-p21-mRNA complex, resulting in suppression of the translation target transcripts via the reduction of ribosome drop-off and polysome sizes (5456).

Post-translational modification

lncRNAs not only regulate the translational process, but also modify proteins produced after translation via mechanisms such as phosphorylation and ubiquitination. The activity and stability of a protein can be altered through these modifications (57). For instance, the lncRNA SLCO4A1-AS1 interacts with β-catenin to improve its stability through weakening the communication between glycogen synthase kinase β (GSKβ) and β-catenin, restricting its phosphorylation and leading to Wnt/β-catenin signaling activation in CRC (58). In addition, the lncRNA SNHG15 can sustain Slug stability by inhibiting the interaction between Slug and β-transducin repeat containing (BTRC) E3 ubiquitin protein ligase, blocking BTRC-mediated Slug ubiquitination in colon cancer (59).

It may be concluded that, from the chromatin level to transcription, post-transcription, translation and post-translational regulation, lncRNAs fulfill important roles in all aspects of cell physiology.

Discovery and description of HOXA11-AS

lncRNA HOXA11-AS, located in the HOXA gene cluster, has been reported to exert an impact on the occurrence of variety of human diseases and their subsequent development (60). HOXA11-AS is located on chromosome 7p15.2, and is referred to as HOXA11AS, HOXA11-AS1, HOXA11S, HOXA-AS5 or NCRNA00076. The chromosomal localization and secondary structure of HOXA11-AS are shown in Fig. 1. The length of the HOXA11-AS gene is 3,885 bp, whereas the HOXA11-AS transcript is 1628 bp in length (61). HOX genes are organized into four clusters (A, B, C and D) on four diverse chromosomes, and HOXA is a member of the homeobox (HOX) family in the human genome (62,63). The HOXA gene has a sense strand and an antisense strand that include protein-coding genes and ncRNA genes, respectively. The 5′-region of the HOXA gene refers to the direction of the sense strand relative to the direction of protein coding genes, and the most abundant protein-coding gene of the 5′-region is HOXA13. Furthermore, a further three protein-coding genes, HOXA9, HOXA10 and HOXA11, and 3 lncRNAs, HOXA10-AS, HOXA11-AS and HOTTIP, are located in the 5′-region (64,65). HOXA11-AS is a novel lncRNA that functions as an oncogene or tumor-suppressor gene in diverse types of tumor. For example, HOXA11-AS can serve as an oncogene in non-small cell lung cancer (NSCLC), HCC, glioma, BC, GC, renal cancer (RC), uveal melanoma (UM), laryngeal squamous cell carcinoma (LSCC), cervical cancer (CC), ESCC and osteosarcoma. By contrast, HOXA11-AS functions as a tumor suppressor in epithelial ovarian cancer (EOC) (7,66,67).

The subsequent sections of this review provide an overview of the clinical significance, biological functions, and molecular mechanisms of HOXA11-AS in tumors and several other diseases types, with the aim of intuitively understanding the role of HOXA11-AS in the occurrence and development of human disease.

HOXA11-AS in cancer
Nonsmall-cell lung cancer (NSCLC)

Lung cancer is the primary cause of cancer mortality and the most commonly occurring type of cancer worldwide (68,69). No fewer than 2.1 million people are diagnosed with lung cancer annually (1). Histologically, lung cancer can be divided into small cell lung cancer (SCLC) and NSCLC. Approximately 80–85% of newly diagnosed cases of lung cancer belong to the NSCLC type (70,71). Numerous lncRNAs, including HOXA11-AS, have been shown to have significant roles in NSCLC. Zhang et al (73,74) reported that the expression of HOXA11-AS is higher in both squamous cell carcinoma (SCC) and lung adenocarcinoma (LUAD) compared with that in normal lung tissues, and HOXA11-AS knockdown suppresses tumorigenesis, angiogenesis, proliferation, migration and invasion of NSCLC cells, inducing apoptosis by impeding the cell cycle at the G0/G1 or the G2/M phase. Moreover, Chen et al (75) identified that high levels of HOXA11-AS predict poor prognosis in patients with NSCLC. Furthermore, Yu et al (48) demonstrated that the expression level of HOXA11-AS is associated with lymph node metastasis and tumor size. Zhao et al (76) also reported that high levels of HOXA11-AS are associated with poor prognosis.

Mechanistically, Zhang et al (7274) revealed that HOXA11-AS expression is negatively correlated with dedicator of cytokinesis 8 (DOCK8) in SCC and LUAD. Those authors conjectured that HOXA11-AS could have an oncogenic role in the development and progression of NSCLC by modulating various pathways, such as the transforming growth factor (TGF)-β pathway, the phosphoinositide 3-kinase (PI3K)-Akt pathway and the Hippo signaling pathway, and genes, such as DOCK8 gene. In other studies, the same authors demonstrated that the expression of HOXA11-AS co-expressed genes in NSCLC may be partly regulated by the NSCLC pathway and that HOXA11-AS could affect various biological processes of NSCLC via regulation of the expression of miR-642b-3p by targeting the expression of phosphodiesterase 4D (PDE4D) or other target genes (7274). Additionally, Chen et al (75) found that HOXA11-AS interacts with DNA (cytosine-5)-methyltransferase 1 (DNMT1) and enhancer of zeste homolog 2 (EZH2), recruiting these proteins to the promoter regions of miR-200b and mediating methylation silencing of miR-200b in NSCLC cells, a process that promotes both NSCLC cell epithelial-mesenchymal transition (EMT) via regulation of the protein levels of E-cadherin, N-cadherin, Snail1/2 and ZEB1/2 and tumor progression. Yu et al (48) revealed that HOXA11-AS acts as a ceRNA to positively modulate the expression of transcription factor Sp1 by sequestering miR-124, a process that can inhibit cell proliferation and the invasion-promoting effects of HOXA11-AS. Futhermore, those authors found that HOXA11-AS can serve as an oncogene by promoting EMT via regulation of the protein levels of E-cadherin, β-catenin, vimentin and the EMT-mediating transcription factors Slug and Snail in NSCLC (48). Zhao et al (76) discovered that, in human LUAD cells, HOXA11-AS can function as a ceRNA to facilitate cisplatin tolerance through the miR-454-3p/Stat3 pathway (the abovementioned mechanisms are shown in Fig. 2A). Recent findings (77) have shown that membrane-bound extracellular vesicles, especially exosomes, serve significant roles as mediators for communication among different tissues and organs. Exosomes have a vital role in signal transduction among cells, and a wide range of biological functions. Wu et al (77) reported that high expression levels of HOXA11-AS in exosomes are closely associated with smoking and NSCLC in lung tissues. These findings indicate that HOXA11-AS has several functions associated with oncogenesis, regulating various physiological activities in NSCLC. These findings shed light upon the effects of HOXA11-AS on the progression of NSCLC and indicate that HOXA11-AS is a potential target for future treatment of NSCLC.

Hepatocellular carcinoma

Hepatocellular carcinoma (HCC), which accounts for 90% of cases of liver cancer, is the fifth most frequent cancer in men and the ninth in women worldwide (78,79). The mortality rate of HCC ranks second in the world (80,81). Recently, cancer-associated studies focused on lncRNAs have demonstrated that a considerable number of lncRNAs are involved in the progression of HCC (8284). Previous studies revealed that HOXA11-AS is overexpressed in HCC cells and tissues (8284). Liu et al (82) found that high expression levels of HOXA11-AS are significantly correlated with vascular invasion, cirrhosis, tumor size and Edmondson grade. The overall survival (OS) rate of patients with high levels of HOXA11-AS expression is markedly shorter compared with those with lower levels of HOXA11-AS expression. Overexpression of HOXA11-AS promotes cell cycle progression, proliferation, invasion, and EMT, as well as repressing apoptosis in HCC cells.

Yu et al (83) reported that HOXA11-AS recruits PRC2 to the promoter region of large tumor suppressor 1 (LATS1) to obstruct its transcription, thereby promoting HCC growth and inhibiting apoptosis and cell cycle progression at the G0/G1 phase. The results from flow cytometry experiments indicated that the cell cycle progression of HCC cells was stalled at the G0/G1 phase when HOXA11-AS RNAi was transfected into HCC cells (83). Zhan et al (84) showed that the overexpression of HOXA11-AS is able to facilitate HCC proliferation and invasion, and induce EMT by repressing the expression of miR-214-3p. In addition, these authors demonstrated that the expression of miR-214-3p in early clinical stages (I–II) was higher than that in advanced clinical stages (III–IV). Furthermore, Liu et al (82) reported that HOXA11-AS recruits EZH2 to the promoter region of dual specificity protein phosphatase 5 (DUSP5), thereby inhibiting the transcription of DUSP5, which is a downstream target of HOXA11-AS and can function as a tumor suppressor gene. HOXA11-AS performs an oncogenic role in HCC by interacting with PRC2 (the abovementioned mechanisms are shown in Fig. 2B). HOXA11-AS may function as an oncogene in HCC development. The interaction between HOXA11-AS and LATS1, DUSP5 or miR-214-3p may supply novel prognostic markers and therapeutic targets for HCC.

Glioma

Glioma, accounting for approximately 80% of primary malignant brain tumors, is the most aggressive primary tumor of the nervous system. Effective treatment of gliomas is very difficult, particularly glioblastomas (GBMs, also known as grade IV astrocytomas), with a median survival time for patients of less than 15 months using standard therapy (8588). To date, although many studies have been focused on seeking improvements in diagnosis and treatment, the trend of poor prognosis has not been reversed (89). Among the numerous biomolecules involved in the occurrence and development of glioma, lncRNAs have attracted sustained attention due to their abnormal expression during tumorigenesis. Wang et al (90) discovered that high levels of HOXA11-AS expression are closely correlated with OS in high-grade glioma, and HOXA11-AS may be an independent prognostic factor for GBM. HOXA11-AS overexpression might occur during initial gliomagenesis and increased levels might be maintained in higher grades of gliomas. Those authors reported that the expression levels of HOTTIP, HOXA9, HOXA10, and HOXA13 are prominently correlated with HOXA11-AS expression, and HOXA11-AS can alter the expression of cell cycle-associated proteins, thus regulating cell cycle progression. Moreover, Cui et al (91) and Xu et al (92) found that high expression levels of HOXA11-AS are correlated with decreased survival time and poorer prognosis compared with patients with lower HOXA11-AS expression. In addition, Xu et al (93) found that the overexpression of HOXA11-AS is associated with advanced stages of glioma and poor prognosis. Authors of that study reported that knocking down the expression of HOXA11-AS leads to suppression of the proliferation, migration, and invasion rates of glioma cells in vitro, with the consequent further enhancement of cell cycle arrest at the G0/G1 stage and improved apoptotic responses.

Cui et al (91) showed that miR-140-5p is able to directly target the 3′-UTR of HOXA11-AS, and the effects of HOXA11-AS knockdown are shown to be reversed by an miR-140-5p inhibitor, as determined by its effects on cell cycle arrest, proliferation and apoptosis. Xu et al (92) demonstrated that HOXA11-AS functions as a ceRNA by sponging miR-214-3p, which can directly target EZH2 and suppress its mRNA transcriptional level. It was thereby confirmed that HOXA11-AS may serve in an oncogenic role by regulating the miR-214-3p/EZH2 pathway. Yang et al (94) found that HOXA11-AS leads to a marked increase in proliferation, invasion and migration rates, while inhibiting apoptosis by absorbing miR-124-3p in glioma cells. Furthermore, Xu et al (93) found that HOXA11-AS can exert its oncogenic role by directly binding to miR-130a-5p as a ceRNA, which inhibits the inhibitory effect of miR-130a-5p on HMGB2 expression. HMGB2 has been demonstrated to be involved in several diseases, such as sepsis, arthritis and cancer (the abovementioned mechanisms are shown in Fig. 2C). The above results suggest that HOXA11-AS may be an oncogene participating in the prognosis and treatment response of specific GBM subtypes.

Colorectal cancer

Colorectal cancer (CRC) ranks third among the most common types of cancer in men, and second among women worldwide, with ~55% of CRC cases occurring in developed countries (95,96). The incidence of CRC has rapidly increased in China, and in 2011, it was shown to be the second most common cause of cancer-associated mortality (97). Although recent advances have been made in the diagnosis and treatment of CRC in recent years, the prognosis of patients diagnosed with CRC remains poor; in fact, the 5-year survival rate in patients with metastatic CRC has been shown to be less than 20% (98,99). Li et al (100) reported that HOXA11-AS is downregulated in CRC cell lines and tissues, and low expression levels of HOXA11-AS are associated with advanced tumor-lymph node-metastasis (TNM) stages, lymphatic metastasis, large tumor size and elevated levels of carcinoembryonic antigen (CEA). Authors of that study also showed that the lncRNA HOXA11-AS may distinguish CRC tissue from noncancerous tissue, and they further explored the correlation between HOXA11-AS and lymph node metastasis. Moreover, Chen et al (101) reported that the level of HOXA11-AS was markedly upregulated in CRC patients with liver metastasis, and the migration and invasion of CRC cells was facilitated by HOXA11-AS. In addition, Chen et al (101) found that HOXA11-AS can act as a ceRNA sequestering miR-125a-5p to modulate the expression of protein-arginine deiminase type-2 (PADI2), which was shown to facilitate metastasis and the invasion of CRC (the abovementioned mechanisms are shown in Fig. 2D). Consequently, a novel HOXA11-AS/miR-125a-5p/PADI2 pathway, involved in CRC liver metastasis, was identified. Taken together, these results demonstrated that HOXA11-AS is a potential biomarker and molecular therapy target in CRC.

Breast cancer

Breast cancer (BC) is one of the most frequent causes of cancer-associated mortality in women worldwide (102,103). Although great progress has been made in earlier diagnosis of this disease and in the effective of systemic therapies, the overall prognosis of BC remains unsatisfactory. In particular, distant metastasis is a barrier for successful treatment (104106). Su and Hu (107) discovered that lncRNA HOXA11-AS is overexpressed in human BC, and the expression of HOXA11-AS is markedly associated with metastasis, tumor size and TNM staging. Clinical statistics revealed that the expression of HOXA11-AS was correlated with Ki-67 protein and human epidermal growth factor receptor (HER2), but not with estrogen receptor (ER) or progesterone receptor (PR). Their results indicated that decreased expression levels of HOXA11-AS in human BC led to suppression in the rates of cell proliferation, migration and invasion, and cell cycle arrest at the G0/G1 phase. Li et al (108) also reported that low levels of HOXA11-AS expression inhibited cell proliferation and induced tumor cell apoptosis by inducing cell cycle arrest at the G0/G1 stage. By contrast, high levels of HOXA11-AS expression facilitated metastasis and invasion both in vitro and in vivo by exerting an influence on EMT in BC (the abovementioned mechanisms are shown in Fig. 2E). Note that the above articles (107,108) only determined the involvement of HOXA11-AS in these processes; the specific mechanisms have yet to be properly elucidated. These studies, however, have confirmed that HOXA11-AS exerts carcinogenic effects in BC, thereby providing some novel insights for even earlier diagnosis in the future, and for the therapy of BC.

Gastric cancer. Gastric cancer (GC) ranks the fifth among the most common types of cancer, and is the third leading cause of cancer mortality (109). GC is more common in men: The mortality rate in men is 2-fold higher compared with women (1). In men, GC is the most commonly occurring gastrointestinal malignancy in East Asia, the most common cancer in several Western Asian countries and the main cause of cancer deaths (110,111). Despite advances in surgical techniques and the successful development of targeted drugs, the 5-year OS rate, however, remains unsatisfactory, and the majority of patients are diagnosed with advanced cancer along with lymphatic metastasis (98). Sun et al (112) reported that HOXA11-AS is significantly upregulated in GC tissues. High levels of HOXA11-AS expression are closely related to poor differentiation, larger tumor size, lymph node metastasis and advanced pathological stage in GC. Researchers also found that progression-free survival (PFS) and OS rates in excess of 3 years in the group with high levels of HOXA11-AS expression were lower compared with the group with low levels of HOXA11-AS expression. HOXA11-AS overexpression leads to increased rates of cell growth, migration and invasion, and inhibition of apoptosis in GC. By contrast, Liu et al (113) reported that HOXA11-AS knockdown induces G0/G1 phase arrest in GC cells and decreases GC cell metastasis, migration and invasion, both in vitro and in vivo.

In terms of the underlying mechanism, Sun et al (112) demonstrated that HOXA11-AS acts as a protein scaffold to recruit EZH2, accompanied by DNMT1 or LSD1. In addition, HOXA11-AS functions as a miR-1297 sponge, thereby regulating the translation of EZHZ, a direct miR-1297 target. Knockdown of HOXA11-AS increases the expression of the tumor suppressors, serine protease 8 (PRSS8) and Krüppel-like Factor 2 (KLF2), leading to the promotion of cell proliferation and invasion and inhibiting apoptosis in GC cells. Those authors reported that EZH2 can directly bind to the promoter regions of PRSS8 and KLF2 and DNMT1 can directly bind to the promoter region of KLF2 to stimulate H3K27me3 modifications. LSD1 is also able to directly bind to the promoter region of PRSS8 to mediate H3K4 demethylation. Knockdown of HOXA11-AS suppresses the binding capability of DNA with several chromatin modification factors (namely, PRC2, LSD1 and DNMT1). Moreover, Sun et al (112) identified the potential regulator E2F1, which can directly bind to the promoter region of HOXA11-AS and regulate the transcription of HOXA11-AS. In addition, HOXA11-AS can function as an oncogene by regulating the miR-1297/EZH2 and HOXA11-AS-EZH2/DNMT1/LSD1-KLF2/PRSS8 pathways to promote GC cell proliferation, migration and invasion and inhibit apoptosis. Liu et al (113) identified that HOXA11-AS enhances β-catenin transcription by interacting with WD repeat-containing protein 5 (WDR5), and p21 transcription is subsequently inhibited via binding with EZH2. In addition, HOXA11-AS can interact with double-stranded RNA-binding protein staufen homolog 1 (STAU1) to promote KLF2 mRNA degradation. These findings revealed that HOXA11-AS may serve as an oncogene by modulating the HOXA11-AS-WDR5/EZH2/STAU1-β-catenin/p21/KLF2 pathway (Fig. 3A). Taken together, these results have revealed that HOXA11-AS can function as an oncogene in GC cells, further elucidating the role of HOXA11-AS in promoting better diagnosis of the disease, and developing further how lncRNA may be used in treatments of GC.

Renal cancer

Clear cell renal cell carcinoma (ccRCC) is a common malignant tumor of the urinary system, accounting for >80% of all types of RC (114). No fewer than 209,000 new cases of ccRCC are reported each year, and among these cases, approximately 25–30% of the patients present with distant metastasis and 40% of patients have local recurrence after the initial diagnosis (115,116). The biological behavior of ccRCC is extremely complex and the underlying molecular mechanisms have yet to be fully elucidated, factors which explain how prognosis is both poor and difficult to predict (117). Recently, Yang et al (118) found that HOXA11-AS was clearly upregulated in ccRCC cell lines and tissues, and high HOXA11-AS expression levels were very closely correlated with lymph node metastasis, tumor stage and advanced clinical stage. Downregulation of the HOXA11-AS transcript led to a marked decrease in the growth, proliferation, invasion, and EMT of the ccRCC cells. Regarding the mechanism, Yang et al (118) discovered that HOXA11-AS functions as a ceRNA to suppress the expression of miR-146b-5p, which subsequently modulates the expression of its downstream target, matrix metalloproteinase-16 (MMP16) in RC. Therefore, HOXA11-AS promotes RC cell invasion and proliferation by regulating the miR-146b-5p/MMP16 pathway (Fig. 3B). During this process, HOXA11-AS may function as an oncogene in ccRCC and may therefore represent an efficient therapeutic target for RCC.

Uveal melanoma. Uveal melanoma (UM) ranks first among primary intraocular malignant tumors in adults, and the uvea ranks second among the common sites of primary melanoma (119,120). The metastatic rate of UM is very high, and UM ultimately spreads to the liver in up to 50% of patients (121). Lu et al (122) discovered that HOXA11-AS is overexpressed in UM, and HOXA11-AS is shown to enhance the rates of UM cell proliferation and invasion, while repressing apoptosis. Authors of that study also reported that HOXA11-AS recruits EZH2 to the promoter region of p21 and mediates H3K27me3 to suppress its transcription. Furthermore, HOXA11-AS acts as a ceRNA for miR-124, which directly targets EZH2, and the cell proliferation and invasion-increasing effects of HOXA11-AS were attenuated upon miR-124 overexpression (Fig. 3C). Taken together, these studies indicate that HOXA11-AS has an oncogenic role through regulating the HOXA11-AS/EZH2/p21 and miR-124/EZH2 pathways in UM tumorigenesis, and HOXA11-AS may also represent a potential therapeutic target for treatment of UM.

Epithelial ovarian cancer

Ovarian malignancies are among the most commonly occurring malignancies in female reproductive organs. The most frequent ovarian malignancy type is cutaneous carcinoma, followed by malignant germ cell tumor. Among these malignant gynecological tumors, the mortality rate of epithelial ovarian cancer (EOC) is the highest, posing a serious threat to women's health (1). A large number of studies have shown that not only HOXA11-AS, but also many other lncRNAs act as oncogenes or tumor suppressor genes in different cancers. Ignarski et al (123) reported that the expression pattern of lncRNA genes is far more tissue- and cell-type specific than is the case for protein coding genes. HOXA11-AS exerts opposing effects in different types of cancer. Such expressional and functional discrepancies of HOXA11-AS in different types of cancer could be caused by distinct gene expression backgrounds in different tumors (124). For example, in EOC, HOXA11-AS can act as a tumor suppressor gene, which is different from the tumor mentioned above.

It has been reported that common germline genetic variants or single nucleotide polymorphisms (SNPs) affecting lncRNAs are conducive to the development of various types of cancer (125), such as EOC (126). Richards et al (126) reported that overexpression of HOXA11-AS results in a clear reduction in cell proliferation and survival, which are two main cellular processes associated with EOC development in both common and minor allele constructs. In particular, the existence of minor allele constructs markedly reduced the proliferation and survival of EOC cells compared with the common allele, and the expression of both alleles served to diminish cell migration and invasion. However, compared with the common allele, the minor allele exerted a more pronounced suppressive effect in both assays. Richards et al (126) found that the minor allele repressed the carcinogenic phenotypes to a greater extent compared with the common allele in EOC cells. The finding has identified a greatly decreased risk of EOC among women who have the HOXA11-AS rs17427875 T allele (126). In EOC tumor tissue, HOXA11-AS expression was reduced by over 60% on average compared with normal ovarian tissue. Richards et al (126) also found HOXA11-AS cannot exert any influence upon HOXA11 and HOXA13 (Fig. 3D). Thus, these studies suggest that HOXA11-AS may function as a tumor suppressor gene in EOC.

Laryngeal squamous cell carcinoma

Laryngeal squamous cell carcinoma (LSCC) is a commonly occurring malignant tumor of the upper respiratory tract. Laryngeal carcinoma is ranked second in terms of the most frequently occurring malignant tumors in the head and neck (127). Qu et al (128) found that the expression levels of HOXA11-AS are closely correlated with pathological grade, T (tumor) grade, clinical stage, neck nodal metastasis and advanced tumors of grade T3 to T4 in LSCC. Furthermore, advanced clinical stages, poor prognosis, poor differentiation and lymph node metastasis were identified in patients who expressed higher levels of HOXA11-AS. Authors of that study also found that HOXA11-AS was able to promote LSCC cell proliferation, invasion and migration (Fig. 3E). Notably, only the general effects attributable to HOXA11-AS were identified in the study of Qu et al (128), and the specific mechanism involved requires further investigation. However, their results have demonstrated that HOXA11-AS may function as an oncogene, suggesting that it is a potential novel biomarker and an efficient therapeutic target in LSCC.

Cervical cancer

It is estimated that CC is the fourth leading cause of cancer mortality, and ranks fourth out of the most commonly diagnosed cancers among women. In 2018, there were approximately 570,000 CC cases and 311,000 cancer-associated deaths worldwide (1). In fact, in terms of its incidence and the mortality rates, CC is ranked second behind BC (1). Kim et al (129) reported that the expression of HOXA11-AS is specifically upregulated in CC, and OS and 5-year survival rates are both reduced in CC patients with HOXA11-AS overexpression. Cox multivariate proportional hazards analysis revealed that nodal metastasis, tumor stage, and HOXA11-AS are independent prognosticators of OS. Those authors found that HOXA11-AS is associated with enhanced cell proliferation, also leading to increased rates of cell migration and invasion in CC. In terms of the underlying mechanism, HOXA11-AS promotes CC cell migration and invasion by upregulating the levels of MMP-9, MMP-2, and vascular endothelial growth factor (VEGF), and disordering of the EMT-associated genes suggested that HOXA11-AS may be involved in cell migration and invasion in CC (Fig. 3F). Kim et al (129) also showed that HOXA11-AS can promote activation of the genetic program that supports the cancer stem cell (CSC) phenotype and improves EMT, suggesting that HOXA11-AS may function as an oncogene in CC. Thus, HOXA11-AS may be a therapeutic target in the search for improved treatments for CC.

Esophageal squamous cell carcinoma

Among the different types of cancer, ESCC is ranked seventh in terms of its incidence and sixth in terms of overall mortality, which means that 1 out of every 20 cases of cancer-associated mortality in 2018 was estimated to have been caused by ESCC (1). Sun et al (130) found that high expression levels of HOXA11-AS are correlated with lymph node metastasis and histological grade in patients with ESCC. Compared with those patients with HOXA11-AS overexpression, patients with low HOXA11-AS expression exhibited both increased median disease-free survival (DFS) and median OS. Sun et al (130) found that lymph node metastasis and the expression of HOXA11-AS were independent poor prognosis factors in patients with ESCC (Fig. 3G). Taking all these findings into consideration, HOXA11-AS may have an oncogenic role in ESCC, and therefore HOXA11-AS may also represent a predictive marker in postoperative ESCC patients.

Osteosarcoma

Osteosarcoma is ranked the first among the most commonly occurring types of primary malignant bone cancer, and is placed second in terms of cancer-associated deaths in pediatrics (131). lncRNAs are well known to be involved in the development of most tumors. Cui et al (60) found that a high level of HOXA11-AS expression is correlated with distant metastasis, reduced OS and advanced clinical stage in patients with osteosarcoma. Their study suggested that HOXA11-AS knockdown in osteosarcoma induces cell cycle arrest at the G0/G1 phase, and HOXA11-AS overexpression led to a substantial improvement in cell invasion and growth rates in osteosarcoma cells via the competitive binding of miR-124-3p, which targets Rho-associated, coiled-coil-containing protein kinase 1 (ROCK1) (Fig. 3H). In this manner, HOXA11-AS may exert oncogenic functions by regulating the miR-124-3p/ROCK1 pathway. Investigating the underlying mechanistic roles of the HOXA11-AS/miR-124-3p/ROCK1 pathway may be an important step in developing novel osteosarcoma therapeutic strategies.

Other diseases
Fracture

Given the ever-increasing aging population, fractures have become a serious health problem, constituting the most common injuries sustained worldwide. Despite the body being capable of healing fractures, many risk factors have substantially delaying effects on the process of fracture healing, including advanced age, smoking, diabetes mellitus (DM) and anti-cancer drugs (132). Therefore, it is imperative to explore the mechanisms underlying fracture healing, notably in patients with these factors, is an urgent requirement. Numerous studies have been published that demonstrate how lncRNAs are able to participate in the occurrence and development of many diseases, including fracture healing (133). Wang et al (133) found that HOXA11-AS overexpression led to a suppression of OS-732 osteoblast proliferation and improved apoptosis. In addition, HOXA11-AS can act as a ceRNA by sequestering miR-124-3p to inhibit cell proliferation and enhance apoptosis (Fig. 3I). These results may provide a novel perspective for deciphering the mechanism of fracture healing.

Preeclampsia

Preeclampsia (PE) is the leading cause of pregnancy-associated death and fetal defects (134). PE is characterized as having a blood pressure exceeding 140/90 mmHg from the 20th week of pregnancy onwards. Between 3 and 5% of pregnant women experience PE, especially in developing countries (135). Xu et al (136) found that HOXA11-AS expression was markedly downregulated in pre-eclamptic placental tissues, and reducing HOXA11-AS expression led to a clear inhibition of trophoblast cell growth and migration. Mechanistically, those authors revealed that HOXA11-AS recruits LSD1 and EZH2 proteins to the RND3 gene promoter region in the nucleus to repress its expression in trophoblast cells. When the level of HOXA11-AS expression was reduced in trophoblast cells, its ability to bind to LSD1 and EZH2 was also reduced, leading to decreased LSD1 and EZH2 binding to the RND3 gene promoter region and decreasing the inhibitory effect on RND3, which suppresses cell growth and proliferation. Furthermore, HOXA11-AS facilitated the expression of HOXA7 in the cytoplasm via sequestration of miR-15b-5p, thus exerting an influence on trophoblast proliferation (Fig. 3J). These studies revealed that HOXA11-AS may have an oncogenic function via modulating the LSD1/EZH2-RND3 and HOXA11-AS/miR-15b-5p/HOXA7 pathways. Consequently, these results have verified that abnormal expression of HOXA11-AS is involved in the occurrence and development of PE, and that this lncRNA may function as a putative target for diagnosis and treatment in PE.

Diabetes mellitus (DM)

With rapid economic development and aging of the population, the number of patients with DM is increasing annually. Persistent hyperglycemia induces hyperglycemia-associated complications that pose severe medical risks, such as diabetic arteriosclerosis (DAA), atherosclerosis (AS) and cardiomyopathy (137). Studies have revealed that the incidence of cardio-cerebrovascular diseases in DM patients is markedly higher than in non-DM patients. In view of the role of lncRNAs in a variety of different tumor types and diseases, researchers have also examined the role of lncRNA in diabetes. Jin et al (138) found that expression levels of HOXA11-AS and pro-inflammatory genes were substantially increased in carotid endarterectomy specimens of DM patients, and HOXA11-AS expression was also significantly increased in the carotid arteries of DM mice. Mechanistically, HOXA11-AS knockdown reduces the expression of proliferation-associated gene (PCNA), the cell cycle-related genes p21 and p53, and platelet-derived growth factor (PDGF)-induced growth and migration of vascular smooth muscle cells (VSMCs) is repressed, significantly downregulating the expression of inflammation-associated genes in VSMCs induced by tumor necrosis factor-α (TNF-α). Moreover, in vascular endothelial cells (VECs), low expression levels of HOXA11-AS were shown to suppress the expression of TNF-α-induced pro-inflammatory genes and PDGF-induced vascular inflammation-related genes. Low expression levels of HOXA11-AS inhibited the PDGF-induced stimulation of the PI3K/AKT pathway by inhibiting the phosphorylation of PI3K and AKT in VSMCs and VECs (Fig. 3K). The biological functions of HOXA11-AS in DAA-induced inflammation should be further explored to identify potential new effective treatments for DAA.

Conclusion

In conclusion, an increasing number of studies have shown that lncRNAs are dysregulated in various types of cancer, and aberrant expression of lncRNAs is involved in the occurrence, development, and metastasis of cancer (139). lncRNAs function mainly by interacting with other DNA, RNA or protein molecules to exert their pre-transcriptional or post-transcriptional regulatory functions. At present, emerging in-depth studies are elucidating the regulatory effects of HOXA11-AS on a majority of different tumor types, including NSCLC, HCC and glioma, although its mechanisms and targets are generally found not to be similar when comparing among the different malignancies. Table I summarizes HOXA11-AS expression patterns and its functional and clinical value in different types of human cancer. Additionally, functional characteristics and molecular mechanisms of HOXA11-AS in diverse types of cancer and other diseases are summarized in Fig. 4. Although many studies have investigated HOXA11-AS, much remains to be determined before we are in a position to fully understand the mechanism of HOXA11-AS in diseases including BC, ESCC and LSCC. However, with further research, HOXA11-AS is likely to gain in importance as a novel target and guide for the prevention, diagnosis and treatment of tumors and additional diseases.

Table I.

HOXA11-AS expression pattern, function and clinical value in human.

Table I.

HOXA11-AS expression pattern, function and clinical value in human.

Cancer typesExpression levelGene typesNumber of casesCell lines involvedGenes, RNAs and proteins interact with HOXA11-ASSignaling pathwaysHOXA11-AS molecular mechanismsHOXA11-AS binding sites (5′→3′)Affected phenotypesAssociation with patients' outcome(Refs.)
Lung cancerUpOncogeneThree pairs NSCLC tissues (HOXA11-AS and HOXA11-AS RNAi)A549DOCK8TGF-beta pathway Zhang et al (2016) (72)
UpOncogene287 lung adenocarcinoma cases vs. 12 non-cancerous lung cases and 463 lung squamous cell carcinoma cases vs. 12 non-cancerous lung casesPC9, A549, H460 and H1299 ErbB, MAPK, Calcium, PI3K-Akt and P53 signaling pathways Proliferation, migration, invasion, apoptosis, tumorigenic angiogenic ability Zhang et al (2017) (73)
UpOncogene78 pairs NSCLC tissues and corresponding adjacent normal tissuesA549, H1299miR-124 HOXA11-AS/miR-124/Sp1 pathwayceRNA509–515 (NR_002795.2) (GTGCCTT)Proliferation, invasion, EMTTumor size, lymph node metastasisYu et al (2017) (47)
UpOncogene78 pairs NSCLC tissues and adjacent normal tissue samplesA549, H1299, 95D, 16HBEEZH2, DNMT1, miR-200b Scaffold Invasion, EMTPrognosis, lymph node metastasis, TNM stageChen et al (2017) (75)
UpOncogeneCell line studyA549, H157, A549-CR, H157-CR (cisplatin-Resistantcells), HEK-293TmiR-454-3p HOXA11-AS/miR-454-3p/Stat3 pathwayceRNA1057–1075 (ENST00000520395.2) (CTAXXXXXXTATT-GCACT)Proliferation, migration, apoptosis, EMTPrognosisZhao et al (2018) (76)
UpOncogeneCell line studyA549miR-642b-3p, PDE4D HOXA11-AS/miR-642b-3p/PDE4D axis ENST00000-520395.2 Zhang et al (2018) (74)
Liver cancerUpOncogene72 pairs HCC tissues and its adjacent tissue specimensHepG2, Hep3B, MHCC-97HLAST1, EZH2 Scaffold Proliferation, apoptosis, cell cycle progression, tumor formation ability Yu et al (2017) (83)
UpOncogene66 pairs HCC tissues and adjacent normal tissuesHepG2, Hep3BDUSP5, EZH2 Scaffold Proliferation, apoptosis, cell cycleVascular invasion, cirrhosis, tumor size and edmindson grade, prognosisLiu et al (2017) (82)
UpOncogene40 pairs of HCC samples and the adjacent noncancerous samplesHep3B, MHCC97-HmiR-214-3p HOXA11-AS/miR-214-3p axis Proliferation, invasion, EMT Zhan et al (2018) (84)
GliomaUpOncogene220 glioma cases and 5 normal brain samplesU87 (ATCC), LN229, U251, HA Proliferation, cell cycleGrade, prognosis, molecular, subtypesWang et al (2016) (90)
UpOncogene43 cases of glioma tissues and normal brain tissuesSHG44, U251, HAmiR-140-5p HOXA11-AS/miR-140-5p axisceRNA(HGNC 24957) (AAACCACT)Proliferation, apoptosis, cell cyclePrognosisCui et al (2017) (91)
UpOncogene45 pairs glioma tissues and the adjacent normal brain tissuesU251, U87 (ATCC), LN229, A172, NHA, HEK-293T, SHG-44miR-214-3p, EZH2 HOXA11-AS/miR-214-3p/EZH2 axisceRNA502–515 (NR_002795.2) (CXCXXXXGT-GCCTT)Proliferation, migration, invasion, tumor growthPrognosis, tumor size, gradeXu et al (2017) (92)
UpOncogene43 pairs of glioma tissues and noncancerous tissuesU251, U87 (ATCC)miR-130a-5p, HMGB2 pathway miR-130a-5p/HMGB2ceRNA1487–1494 (ENST00000520395.2) (ATTGCACT)Proliferation, migration, invasion, apoptosisPrognosis, advanced stageXu et al (2019) (93)
Colorectal cancerDownSuppressor gene84 CRC tissues and adjacent non-cancerous tissues, in addition to 3 CRC cell lines and 1 human normal colorectal cell line.CCD-18Co, HCT8, HCT116, RKO ENST0000-0520395.2Tumor size, TNM stage, lymph node metastasis, carcinoem-bryonic antigen levelLi et al (2016) (100)
UpOncogene30 primary CRC samples (15 patients with CRC) and liver metastasis and 15 patients with CRC without metastasis)Colo205, Lovo, HCT116, SW620, Caco-2, SW480miR-125a-5p, PADI2 HOXA11-AS/miR-125a-5p/PADI2 regulatory networkceRNA1482–1487 (NR_002795.2) (CCTGAG)Migration, invasionLiver metastasisChen et al (2017) (101)
Breast cancerUpOncogene100 pairs BC tissues and adjacent non-cancerous tissuesMCF10A, MDA-MB-231, MDA-MB-436, MCF7, T47D Proliferation, migration, invasion, colony formation, cell cycle progressionTumor size, metastasis, TNM stage, molecular subtypesSu and Hu (2017) (107)
UpOncogene68 pairs breast cancer tissues and adjacent normal tissue specimensMDA-MB-468, MDA-MB-231, SKBR3, MCF-10A Migration, invasion, apoptosis, cell cycle progression, EMTMetastasisLi et al (2017) (108)
Gastric cancerUpOncogene85 pairs gastric cancer and adjacent nontumor tissuesGES1, AGSmiR-1297, EZH2, LSD1, DNMT1Cell-cell adhesion pathways, HOXA11-AS/miR-1297/EZH2 cross-talkScaffold ceRNA457–469 (NR_002795.2) (CXXXXTXACTTGA) 5′ region bind with EZH2 3′ region bind with LSD1Growth, proliferation, migration, invasion, apoptosis, tumorigenesis, tumor progressionPrognosis, tumor size, differentiation, pathologic stage, lymph node metastasisSun et al (2016) (112)
UpOncogeneCell line studyAGSWDR5, STAU1, EZH2 Scaffold Proliferation, migration, invasion, cell cycle progression, EMTMetastasisLiu et al (2017) (113)
Renal cancerUpOncogene103 pairs ccRCC specimens and adjacent nontumor tissuesACHN, 786-O, A498, OSRC- 2, HK-2miR-146b-5pmiR-146b-5p/MMP16 axis.ceRNA15–42 (ENST00000522674.1) (AGCXXXXGAXXXTCAXXXXXXGTTCTCA)Growth, proliferation, invasion, EMTAdvanced clinical stage, tumor stage, lymphYang et al (2018) (118)
Uveal melanomaUpOncogeneFive primary UM samplesC918, MUM-2B and D78EZH2, miR-124 Scaffold ceRNA509–515 (NR_002795.2) (GTGCCTT)Growth, proliferation, invasion, apoptosis Lu et al (2017) (122)
Epithelial ovarian cancerDownSuppressor geneCase control study HOXA11-AS minor allele T and common allele A Proliferation, growth migration, invasionPrognosisRichards et al (2015) (126)
Laryngeal squamous cell carcinomaUpOncogene25 pairs cancerous and adjacent noncancerous tissuesAMC-HN-8 Proliferation, growth migration, invasionGrade, neck nodal metastasis, clinical stage, prognosisQu et al (2018) (128)
Cervical cancerUpOncogene92 cervical cancer tissues and 30 normal cervix samplesSiHa, HeLa, CaSki, ME-180, C33A and HOSEMMP-9, MMP-2, and VEGF Proliferation, growth, migration, invasion, sphere formation, EMT, stemness maintenancePrognosisKim et al (2016) (129)
Esophageal squamous cell carcinomaUpOncogene73 pairs ESCC tissues and adjacent tissue samplesHET-1A, EC109, EC9706 Histological grade, lymph node metastasis, prognosisSun et al (2018) (130)
OsteosarcomaUpOncogene51 pairs OS tissues and adjacent tissue samplesU2OS, MG-63, KHOS and NHostROCK1, miR-124-3p HOXA11-AS/miR-124-3p/ROCK1 signaling axisceRNA502–515 (NR_002795.2) (CXCXXXXGTGCCTT)Proliferation, invasion, cell cycle progressionClinical stage, distant metastasis, prognosisCui et al (2017) (60)

Acknowledgements

Not applicable.

Funding

This study was supported by grants (nos. 81773187 and 81572496) from the National Nature Science Foundation of China. Support was also received from the Tianjin High School Program for Young and Middle-aged Talents Backbone and the Tianjin Young Medical Talents Program.

Availability of data and materials

Not applicable.

Authors' contributions

LH and YZ conceived the review. CW and LZ drafted the manuscript and revised it before submission. CW and HL collected the references. All authors read and approved the final 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 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Li J, Li Z, Zheng W, Li X, Wang Z, Cui Y and Jiang X: PANDAR: A pivotal cancer-related long non-coding RNA in human cancers. Mol Biosyst. 13:2195–2201. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Hu Q, Tai S and Wang J: Oncogenicity of lncRNA FOXD2-AS1 and its molecular mechanisms in human cancers. Pathol Res Pract. 215:843–848. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Adams BD, Parsons C, Walker L, Zhang WC and Slack FJ: Targeting noncoding RNAs in disease. J Clin Invest. 127:761–771. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, et al: Landscape of transcription in human cells. Nature. 489:101–118. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Zhu S, Li W, Liu J, Chen CH, Liao Q, Xu P, Xu H, Xiao T, Cao Z, Peng J, et al: Genome-scale deletion screening of human long non-coding RNAs using a paired-guide RNA CRISPR-Cas9 library. Nat Biotechnol. 34:1279–1286. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Xue JY, Huang C, Wang W, Li HB, Sun M and Xie M: HOXA11-AS: A novel regulator in human cancer proliferation and metastasis. Onco Targets Ther. 11:4387–4393. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Zhao W, Ma X, Liu L, Chen Q, Liu Z, Zhang Z, Ma S, Wang Z, Li H, Wang Z and Wu J: SNHG20: A vital lncRNA in multiple human cancers. J Cell Physiol. Jan 15–2019.(Epub ahead of print). doi: 10.1002/jcp.28143.

9 

Schmitt AM and Chang HY: Long noncoding RNAs in cancer pathways. Cancer Cell. 29:452–463. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Anastasiadou E, Jacob LS and Slack FJ: Non-coding RNA networks in cancer. Nat Rev Cancer. 18:5–18. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Atianand MK, Caffrey DR and Fitzgerald KA: Immunobiology of long noncoding RNAs. Annu Rev Immunol. 35:177–198. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Khorkova O, Hsiao J and Wahlestedt C: Basic biology and therapeutic implications of lncRNA. Adv Drug Deliv Rev. 87:15–24. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Borsani G, Tonlorenzi R, Simmler MC, Dandolo L, Arnaud D, Capra V, Grompe M, Pizzuti A, Muzny D, Lawrence C, et al: Characterization of a murine gene expressed from the inactive X chromosome. Nature. 351:325–329. 1991. View Article : Google Scholar : PubMed/NCBI

14 

Gendrel AV and Heard E: Noncoding RNAs and epigenetic mechanisms during X-chromosome inactivation. Annu Rev Cell Dev Biol. 30:561–580. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Li ZX, Zhu QN, Zhang HB, Hu Y, Wang G and Zhu YS: MALAT1: A potential biomarker in cancer. Cancer Manag Res. 10:6757–6768. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Seiler J, Breinig M, Caudron-Herger M, Polycarpou-Schwarz M, Boutros M and Diederichs S: The lncRNA VELUCT strongly regulates viability of lung cancer cells despite its extremely low abundance. Nucleic Acids Res. 45:5458–5469. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Ma M, Xu H, Liu G, Wu J, Li C, Wang X, Zhang S, Xu H, Ju S, Cheng W, et al: Metabolism-induced tumor activator 1 (MITA1), an Energy Stress-inducible long noncoding RNA, promotes hepatocellular carcinoma metastasis. Hepatology. 70:215–230. 2019.PubMed/NCBI

18 

Lu YF, Cai XL, Li ZZ, Lv J, Xiang YA, Chen JJ, Chen WJ, Sun WY, Liu XM and Chen JB: lncRNA SNHG16 functions as an oncogene by sponging MiR-4518 and Up-regulating PRMT5 expression in Glioma. Cell Physiol Biochem. 45:1975–1985. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Wang FW, Cao CH, Han K, Zhao YX, Cai MY, Xiang ZC, Zhang JX, Chen JW, Zhong LP, Huang Y, et al: APC-activated long noncoding RNA inhibits colorectal carcinoma pathogenesis through reduction of exosome production. J Clin Invest. 129:727–743. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Kim J, Piao HL, Kim BJ, Yao F, Han Z, Wang Y, Xiao Z, Siverly AN, Lawhon SE, Ton BN, et al: Long noncoding RNA MALAT1 suppresses breast cancer metastasis. Nat Genet. 50:1705–1715. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Zhu C, Cheng D, Qiu X, Zhuang M and Liu Z: Long noncoding RNA SNHG16 promotes cell proliferation by sponging MicroRNA-205 and upregulating ZEB1 expression in osteosarcoma. Cell Physiol Biochem. 51:429–440. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Yan H, Li H, Li P, Li X, Lin J, Zhu L, Silva MA, Wang X, Wang P and Zhang Z: Long noncoding RNA MLK7-AS1 promotes ovarian cancer cells progression by modulating miR-375/YAP1 axis. J Exp Clin Cancer Res. 37:2372018. View Article : Google Scholar : PubMed/NCBI

23 

Zhuo W, Liu Y, Li S, Guo D, Sun Q, Jin J, Rao X, Li M, Sun M, Jiang M, et al: Long noncoding RNA GMAN, up-regulated in gastric cancer tissues, is associated with metastasis in patients and promotes translation of Ephrin A1 by competitively binding GMAN-AS. Gastroenterology. 156:676–691.e11. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Zhang E, Han L, Yin D, He X, Hong L, Si X, Qiu M, Xu T, De W, Xu L, et al: H3K27 acetylation activated-long non-coding RNA CCAT1 affects cell proliferation and migration by regulating SPRY4 and HOXB13 expression in esophageal squamous cell carcinoma. Nucleic Acids Res. 45:3086–3101. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Peng WX, Koirala P and Mo YY: lncRNA-mediated regulation of cell signaling in cancer. Oncogene. 36:5661–5667. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Wang Z, Yang B, Zhang M, Guo W, Wu Z, Wang Y, Jia L, Li S; Cancer Genome Atlas Research Network, ; Xie W and Yang D: lncRNA epigenetic landscape analysis identifies EPIC1 as an oncogenic lncRNA that interacts with MYC and promotes Cell-cycle progression in cancer. Cancer Cel. 33:706–720.e9. 2018. View Article : Google Scholar

27 

Bhan A, Soleimani M and Mandal SS: Long noncoding RNA and cancer: A new paradigm. Cancer Res. 77:3965–3981. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Chen YG, Satpathy AT and Chang HY: Gene regulation in the immune system by long noncoding RNAs. Nat Immunol. 18:962–972. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Kopp F and Mendell JT: Functional classification and experimental dissection of long noncoding RNAs. Cell. 172:393–407. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Chu C, Zhang QC, da Rocha ST, Flynn RA, Bharadwaj M, Calabrese JM, Magnuson T, Heard E and Chang HY: Systematic discovery of Xist RNA binding proteins. Cell. 161:404–416. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Samata M and Akhtar A: Dosage compensation of the X chromosome: A complex epigenetic assignment involving chromatin regulators and long noncoding RNAs. Annu Rev Biochem. 87:323–350. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Vallot C, Patrat C, Collier AJ, Huret C, Casanova M, Liyakat AT, Tosolini M, Frydman N, Heard E, Rugg-Gunn PJ and Rougeulle C: XACT noncoding RNA competes with XIST in the control of X Chromosome activity during human early development. Cell Stem Cell. 20:102–111. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Furlan G, Gutierrez Hernandez N, Huret C, Galupa R, van Bemmel JG, Romito A, Heard E, Morey C and Rougeulle C: The Ftx noncoding locus controls X chromosome inactivation independently of its RNA products. Mol Cell. 70:462–472.e8. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Sarma K, Cifuentes-Rojas C, Ergun A, Del Rosario A, Jeon Y, White F, Sadreyev R and Lee JT: ATRX directs binding of PRC2 to Xist RNA and Polycomb targets. Cell. 159:869–883. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Guttman M, Donaghey J, Carey BW, Garber M, Grenier JK, Munson G, Young G, Lucas AB, Ach R, Bruhn L, et al: lincRNAs act in the circuitry controlling pluripotency and differentiation. Nature. 477:295–300. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Khalil AM, Guttman M, Huarte M, Garber M, Raj A, Rivea Morales D, Thomas K, Presser A, Bernstein BE, van Oudenaarden A, et al: Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc Natl Acad Sci USA. 106:11667–11672. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Engreitz JM, Haines JE, Perez EM, Munson G, Chen J, Kane M, McDonel PE, Guttman M and Lander ES: Local regulation of gene expression by lncRNA promoters, transcription and splicing. Nature. 539:452–455. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Sun TT, He J, Liang Q, Ren LL, Yan TT, Yu TC, Tang JY, Bao YJ, Hu Y, Lin Y, et al: lncRNA GClnc1 promotes gastric carcinogenesis and may act as a modular scaffold of WDR5 and KAT2A complexes to specify the histone modification pattern. Cancer Discov. 6:784–801. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Li D, Liu X, Zhou J, Hu J, Zhang D, Liu J, Qiao Y and Zhan Q: Long noncoding RNA HULC modulates the phosphorylation of YB-1 through serving as a scaffold of extracellular signal-regulated kinase and YB-1 to enhance hepatocarcinogenesis. Hepatology. 65:1612–1627. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Long Y, Wang X, Youmans DT and Cech TR: How do lncRNAs regulate transcription? Sci Adv. 3:eaao21102017. View Article : Google Scholar : PubMed/NCBI

41 

Hu WL, Jin L, Xu A, Wang YF, Thorne RF, Zhang XD and Wu M: GUARDIN is a p53-responsive long non-coding RNA that is essential for genomic stability. Nat Cell Biol. 20:492–502. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Bhan A and Mandal SS: Estradiol-induced transcriptional regulation of long non-coding RNA, HOTAIR. Methods Mol Biol. 1366:395–412. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Grelet S, Link LA, Howley B, Obellianne C, Palanisamy V, Gangaraju VK, Diehl JA and Howe PH: Addendum: A regulated PNUTS mRNA to lncRNA splice switch mediates EMT and tumour progression. Nat Cell Biol. 19:14432017. View Article : Google Scholar : PubMed/NCBI

44 

Wu Q, Guo L, Jiang F, Li L, Li Z and Chen F: Analysis of the miRNA-mRNA-lncRNA networks in ER+ and ER-breast cancer cell lines. J Cell Mol Med. 19:2874–2887. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Song YX, Sun JX, Zhao JH, Yang YC, Shi JX, Wu ZH, Chen XW, Gao P, Miao ZF and Wang ZN: Non-coding RNAs participate in the regulatory network of CLDN4 via ceRNA mediated miRNA evasion. Nat Commun. 8:2892017. View Article : Google Scholar : PubMed/NCBI

46 

Shen L, Wang Q, Liu R, Chen Z, Zhang X, Zhou P and Wang Z: lncRNA lnc-RI regulates homologous recombination repair of DNA double-strand breaks by stabilizing RAD51 mRNA as a competitive endogenous RNA. Nucleic Acids Res. 46:717–729. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Yu W, Peng W, Jiang H, Sha H and Li J: lncRNA HOXA11-AS promotes proliferation and invasion by targeting miR-124 in human non-small cell lung cancer cells. Tumour Biol. 39:10104283177214402017. View Article : Google Scholar : PubMed/NCBI

48 

Dhir A, Dhir S, Proudfoot NJ and Jopling CL: Microprocessor mediates transcriptional termination of long noncoding RNA transcripts hosting microRNAs. Nat Struct Mol Biol. 22:319–327. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Augoff K, McCue B, Plow EF and Sossey-Alaoui K: miR-31 and its host gene lncRNA LOC554202 are regulated by promoter hypermethylation in triple-negative breast cancer. Mol Cancer. 11:52012. View Article : Google Scholar : PubMed/NCBI

50 

Yu Y, Nangia-Makker P, Farhana L and Majumdar A: A novel mechanism of lncRNA and miRNA interaction: CCAT2 regulates miR-145 expression by suppressing its maturation process in colon cancer cells. Mol Cancer. 16:1552017. View Article : Google Scholar : PubMed/NCBI

51 

Villamizar O, Chambers CB, Riberdy JM, Persons DA and Wilber A: Long noncoding RNA Saf and splicing factor 45 increase soluble Fas and resistance to apoptosis. Oncotarget. 7:13810–13826. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Jadaliha M, Gholamalamdari O, Tang W, Zhang Y, Petracovici A, Hao Q, Tariq A, Kim TG, Holton SE, Singh DK, et al: A natural antisense lncRNA controls breast cancer progression by promoting tumor suppressor gene mRNA stability. PLoS Genet. 14:e10078022018. View Article : Google Scholar : PubMed/NCBI

53 

Qu L, Ding J, Chen C, Wu ZJ, Liu B, Gao Y, Chen W, Liu F, Sun W, Li XF, et al: Exosome-Transmitted lncARSR promotes sunitinib resistance in renal cancer by acting as a competing endogenous RNA. Cancer Cell. 29:653–668. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Rashid F, Shah A and Shan G: Long non-coding RNAs in the cytoplasm. Genomics Proteomics Bioinformatics. 14:73–80. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Yoon JH, Abdelmohsen K, Srikantan S, Yang X, Martindale JL, De S, Huarte M, Zhan M, Becker KG and Gorospe M: LincRNA-p21 suppresses target mRNA translation. Mol Cell. 47:648–655. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Wang KC, Yang YW, Liu B, Sanyal A, Corces-Zimmerman R, Chen Y, Lajoie BR, Protacio A, Flynn RA, Gupta RA, et al: A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature. 472:120–124. 2011. View Article : Google Scholar : PubMed/NCBI

57 

He RZ, Luo DX and Mo YY: Emerging roles of lncRNAs in the post-transcriptional regulation in cancer. Genes Dis. 6:6–15. 2019. View Article : Google Scholar : PubMed/NCBI

58 

Yu J, Han Z, Sun Z, Wang Y, Zheng M and Song C: lncRNA SLCO4A1-AS1 facilitates growth and metastasis of colorectal cancer through β-catenin-dependent Wnt pathway. J Exp Clin Cancer Res. 37:2222018. View Article : Google Scholar : PubMed/NCBI

59 

Jiang H, Li T, Qu Y, Wang X, Li B, Song J, Sun X, Tang Y, Wan J, Yu Y, et al: Long non-coding RNA SNHG15 interacts with and stabilizes transcription factor Slug and promotes colon cancer progression. Cancer Lett. 425:78–87. 2018. View Article : Google Scholar : PubMed/NCBI

60 

Cui M, Wang J, Li Q, Zhang J, Jia J and Zhan X: Long non-coding RNA HOXA11-AS functions as a competing endogenous RNA to regulate ROCK1 expression by sponging miR-124-3p in osteosarcoma. Biomed Pharmacother. 92:437–444. 2017. View Article : Google Scholar : PubMed/NCBI

61 

Zheng LL, Li JH, Wu J, Sun WJ, Liu S, Wang ZL, Zhou H, Yang JH and Qu LH: deepBase v2.0: Identification, expression, evolution and function of small RNAs, lncRNAs and circular RNAs from deep-sequencing data. Nucleic Acids Res. 44:D196–D202. 2016. View Article : Google Scholar : PubMed/NCBI

62 

Spencer DH, Young MA, Lamprecht TL, Helton NM, Fulton R, O'Laughlin M, Fronick C, Magrini V, Demeter RT, Miller CA, et al: Epigenomic analysis of the HOX gene loci reveals mechanisms that may control canonical expression patterns in AML and normal hematopoietic cells. Leukemia. 29:1279–1289. 2015. View Article : Google Scholar : PubMed/NCBI

63 

Garcia-Fernández J: The genesis and evolution of homeobox gene clusters. Nat Rev Genet. 6:881–892. 2005. View Article : Google Scholar : PubMed/NCBI

64 

Kelly ZL, Michael A, Butler-Manuel S, Pandha HS and Morgan RG: HOX genes in ovarian cancer. J Ovarian Res. 4:162011. View Article : Google Scholar : PubMed/NCBI

65 

Cheng W, Liu J, Yoshida H, Rosen D and Naora H: Lineage infidelity of epithelial ovarian cancers is controlled by HOX genes that specify regional identity in the reproductive tract. Nat Med. 11:531–537. 2005. View Article : Google Scholar : PubMed/NCBI

66 

Lu S, Jiang X, Su Z, Cui Z, Fu W and Tai S: The role of the long non-coding RNA HOXA11-AS in promoting proliferation and metastasis of malignant tumors. Cell Biol Int. 42:1596–1601. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Lu CW, Zhou DD, Xie T, Hao JL, Pant OP, Lu CB and Liu XF: HOXA11 antisense long noncoding RNA (HOXA11-AS): A promising lncRNA in human cancers. Cancer Med. 7:3792–3799. 2018. View Article : Google Scholar : PubMed/NCBI

68 

Xu YJ, Du Y and Fan Y: Long noncoding RNAs in lung cancer: What we know in 2015. Clin Transl Oncol. 18:660–665. 2016. View Article : Google Scholar : PubMed/NCBI

69 

Kang CG, Lee HJ, Kim SH and Lee EO: Zerumbone suppresses Osteopontin-induced cell invasion through inhibiting the FAK/AKT/ROCK pathway in human Non-small cell lung cancer A549 cells. J Nat Prod. 79:156–160. 2016. View Article : Google Scholar : PubMed/NCBI

70 

Osmani L, Askin F, Gabrielson E and Li QK: Current WHO guidelines and the critical role of immunohistochemical markers in the subclassification of non-small cell lung carcinoma (NSCLC): Moving from targeted therapy to immunotherapy. Semin Cancer Biol. 52:103–109. 2018. View Article : Google Scholar : PubMed/NCBI

71 

Li Y, Xuan J, Song Y, Qi W, He B, Wang P and Qin L: Nanoporous Glass integrated in volumetric Bar-chart chip for Point-of-Care diagnostics of non-small cell lung cancer. Acs Nano. 10:1640–1647. 2016. View Article : Google Scholar : PubMed/NCBI

72 

Zhang Y, He RQ, Dang YW, Zhang XL, Wang X, Huang SN, Huang WT, Jiang MT, Gan XN, Xie Y, et al: Comprehensive analysis of the long noncoding RNA HOXA11-AS gene interaction regulatory network in NSCLC cells. Cancer Cell Int. 16:892016. View Article : Google Scholar : PubMed/NCBI

73 

Zhang Y, Chen WJ, Gan TQ, Zhang XL, Xie ZC, Ye ZH, Deng Y, Wang ZF, Cai KT, Li SK, et al: Clinical significance and effect of lncRNA HOXA11-AS in NSCLC: A study based on bioinformatics, in vitro and in vivo verification. Sci Rep. 7:5567–18. 2017. View Article : Google Scholar : PubMed/NCBI

74 

Zhang Y, Luo J, Wang X, Wang HL, Zhang XL, Gan TQ, Chen G and Luo DZ: A comprehensive analysis of the predicted targets of miR-642b-3p associated with the long non-coding RNA HOXA11-AS in NSCLC cells. Oncol Lett. 15:6147–6160. 2018.PubMed/NCBI

75 

Chen JH, Zhou LY, Xu S, Zheng YL, Wan YF and Hu CP: Overexpression of lncRNA HOXA11-AS promotes cell epithelial-mesenchymal transition by repressing miR-200b in non-small cell lung cancer. Cancer Cell Int. 17:642017. View Article : Google Scholar : PubMed/NCBI

76 

Zhao X, Li X, Zhou L, Ni J, Yan W, Ma R, Wu J, Feng J and Chen P: lncRNA HOXA11-AS drives cisplatin resistance of human LUAD cells via modulating miR-454-3p/Stat3. Cancer Sci. 109:3068–3079. 2018. View Article : Google Scholar : PubMed/NCBI

77 

Wu F, Yin Z, Yang L, Fan J, Xu J, Jin Y, Yu J, Zhang D and Yang G: Smoking induced extracellular vesicles release and their distinct properties in Non-small cell lung cancer. J Cancer. 10:3435–3443. 2019. View Article : Google Scholar : PubMed/NCBI

78 

Vogel A, Cervantes A, Chau I, Daniele B, Llovet J, Meyer T, Nault JC, Neumann U, Ricke J, Sangro B, et al: Hepatocellular carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 29 (Suppl 4):iv238–iv255. 2018. View Article : Google Scholar

79 

Koeberle D, Dufour JF, Demeter G, Li Q, Ribi K, Samaras P, Saletti P, Roth AD, Horber D, Buehlmann M, et al: Sorafenib with or without everolimus in patients with advanced hepatocellular carcinoma (HCC): A randomized multicenter, multinational phase II trial (SAKK 77/08 and SASL 29). Ann Oncol. 27:856–861. 2016. View Article : Google Scholar : PubMed/NCBI

80 

Tai WM, Yong WP, Lim C, Low LS, Tham CK, Koh TS, Ng QS, Wang WW, Wang LZ, Hartano S, et al: Ann A phase Ib study of selumetinib (AZD6244, ARRY-142886) in combination with sorafenib in advanced hepatocellular carcinoma (HCC). Ann Oncol. 27:2210–2215. 2016. View Article : Google Scholar : PubMed/NCBI

81 

Ciuleanu T, Bazin I, Lungulescu D, Miron L, Bondarenko I, Deptala A, Rodriguez-Torres M, Giantonio B, Fox NL, Wissel P, et al: A randomized, double-blind, placebo-controlled phase II study to assess the efficacy and safety of mapatumumab with sorafenib in patients with advanced hepatocellular carcinoma. Ann Oncol. 27:680–687. 2016. View Article : Google Scholar : PubMed/NCBI

82 

Liu B, Li J, Liu X, Zheng M, Yang Y, Lyu Q and Jin L: Long non-coding RNA HOXA11-AS promotes the proliferation HCC cells by epigenetically silencing DUSP5. Oncotarget. 8:109509–109521. 2017. View Article : Google Scholar : PubMed/NCBI

83 

Yu J, Hong JF, Kang J, Liao LH and Li CD: Promotion of lncRNA HOXA11-AS on the proliferation of hepatocellular carcinoma by regulating the expression of LATS1. Eur Rev Med Pharmacol Sci. 21:3402–3411. 2017.PubMed/NCBI

84 

Zhan M, He K, Xiao J, Liu F, Wang H, Xia Z, Duan X, Huang R, Li Y, He X, et al: lncRNA HOXA11-AS promotes hepatocellular carcinoma progression by repressing miR-214-3p. J Cell Mol Med. May 15–2018.(Epub ahead of print). doi: 10.1111/jcmm.13633. View Article : Google Scholar

85 

Gusyatiner O and Hegi ME: Glioma epigenetics: From subclassification to novel treatment options. Semin Cancer Biol. 51:50–58. 2018. View Article : Google Scholar : PubMed/NCBI

86 

Cohen ZR, Ramishetti S, Peshes-Yaloz N, Goldsmith M, Wohl A, Zibly Z and Peer D: Localized RNAi therapeutics of chemoresistant grade IV glioma using hyaluronan-grafted lipid-based nanoparticles. ACS Nano. 9:1581–1591. 2015. View Article : Google Scholar : PubMed/NCBI

87 

Zhang KL, Han L, Chen LY, Shi ZD, Yang M, Ren Y, Chen LC, Zhang JX, Pu PY and Kang CS: Blockage of a miR-21/EGFR regulatory feedback loop augments anti-EGFR therapy in glioblastomas. Cancer Lett. 342:139–149. 2014. View Article : Google Scholar : PubMed/NCBI

88 

Wang X, Wang K, Han L, Zhang A, Shi Z, Zhang K, Zhang H, Yang S, Pu P, Shen C, et al: PRDM1 is directly targeted by miR-30a-5p and modulates the Wnt/β-catenin pathway in a Dkk1-dependent manner during glioma growth. Cancer Lett. 331:211–219. 2013. View Article : Google Scholar : PubMed/NCBI

89 

Gonzalez-Quarante LH, Fernández Carballal C, Agarwal V, Vargas Lopez AJ, Gil de Sagredo Del Corral OL and Sola Vendrell E: Angiocentric Glioma in an elderly patient: Case report and review of the literature. World Neurosurg. 97:755.e5–755.e10. 2017. View Article : Google Scholar

90 

Wang Q, Zhang J, Liu Y, Zhang W, Zhou J, Duan R, Pu P, Kang C and Han L: A novel cell cycle-associated lncRNA, HOXA11-AS, is transcribed from the 5-prime end of the HOXA transcript and is a biomarker of progression in glioma. Cancer Lett. 373:251–259. 2016. View Article : Google Scholar : PubMed/NCBI

91 

Cui Y, Yi L, Zhao J and Jiang Y: Long Noncoding RNA HOXA11-AS functions as miRNA sponge to promote the glioma tumorigenesis through targeting miR-140-5p. DNA Cell Biol. 36:822–828. 2017. View Article : Google Scholar : PubMed/NCBI

92 

Xu C, He T, Li Z, Liu H and Ding B: Regulation of HOXA11-AS/miR-214-3p/EZH2 axis on the growth, migration and invasion of glioma cells. Biomed Pharmacother. 95:1504–1513. 2017. View Article : Google Scholar : PubMed/NCBI

93 

Xu CH, Xiao LM, Liu Y, Chen LK, Zheng SY, Zeng EM and Li DH: The lncRNA HOXA11-AS promotes glioma cell growth and metastasis by targeting miR-130a-5p/HMGB2. Eur Rev Med Pharmacol Sci. 23:241–252. 2019.PubMed/NCBI

94 

Yang JX, Liu B, Yang BY and Meng Q: Long non-coding RNA homeobox (HOX) A11-AS promotes malignant progression of glioma by targeting miR-124-3p. Neoplasma. 65:505–514. 2018. View Article : Google Scholar : PubMed/NCBI

95 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar : PubMed/NCBI

96 

Rychahou P, Haque F, Shu Y, Zaytseva Y, Weiss HL, Lee EY, Mustain W, Valentino J, Guo P and Evers BM: Delivery of RNA nanoparticles into colorectal cancer metastases following systemic administration. ACS Nano. 9:1108–1116. 2015. View Article : Google Scholar : PubMed/NCBI

97 

Zhang L, Cao F, Zhang G, Shi L, Chen S, Zhang Z, Zhi W and Ma T: Trends in and predictions of colorectal cancer incidence and mortality in china from 1990 to 2025. Front Oncol. 9:982019. View Article : Google Scholar : PubMed/NCBI

98 

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

99 

Lai Y, Wang C, Civan JM, Palazzo JP, Ye Z, Hyslop T, Lin J, Myers RE, Li B, Jiang B, et al: Effects of cancer stage and treatment differences on racial disparities in survival from colon cancer: A united states population-based study. Gastroenterology. 150:1135–1146. 2016. View Article : Google Scholar : PubMed/NCBI

100 

Li T, Xu C, Cai B, Zhang M, Gao F and Gan J: Expression and clinicopathological significance of the lncRNA HOXA11-AS in colorectal cancer. Oncol Lett. 12:4155–4160. 2016. View Article : Google Scholar : PubMed/NCBI

101 

Chen D, Sun Q, Zhang L, Zhou X, Cheng X, Zhou D, Ye F, Lin J and Wang W: The lncRNA HOXA11-AS functions as a competing endogenous RNA to regulate PADI2 expression by sponging miR-125a-5p in liver metastasis of colorectal cancer. Oncotarget. 8:70642–70652. 2017.PubMed/NCBI

102 

Anastasiadi Z, Lianos GD, Ignatiadou E, Harissis HV and Mitsis M: Breast cancer in young women: An overview. Updates Surg. 69:313–317. 2017. View Article : Google Scholar : PubMed/NCBI

103 

Zhang Y, Leonard M, Shu Y, Yang Y, Shu D, Guo P and Zhang X: Overcoming Tamoxifen resistance of human breast cancer by targeted gene silencing using multifunctional pRNA nanoparticles. Acs Nano. 11:335–346. 2017. View Article : Google Scholar : PubMed/NCBI

104 

Lemler DJ, Lynch ML, Tesfay L, Deng Z, Paul BT, Wang X, Hegde P, Manz DH, Torti SV and Torti FM: DCYTB is a predictor of outcome in breast cancer that functions via iron-independent mechanisms. Breast Cancer Res. 19:252017. View Article : Google Scholar : PubMed/NCBI

105 

Borin TF, Arbab AS, Gelaleti GB, Ferreira LC, Moschetta MG, Jardim-Perassi BV, Iskander AS, Varma NR, Shankar A, Coimbra VB, et al: Melatonin decreases breast cancer metastasis by modulating Rho-associated kinase protein-1 expression. J Pineal Res. 60:3–15. 2016. View Article : Google Scholar : PubMed/NCBI

106 

van Roosmalen W, Le Dévédec SE, Golani O, Smid M, Pulyakhina I, Timmermans AM, Look MP, Zi D, Pont C, de Graauw M, et al: Tumor cell migration screen identifies SRPK1 as breast cancer metastasis determinant. J Clin Invest. 125:1648–1664. 2015. View Article : Google Scholar : PubMed/NCBI

107 

Su J and Hu X: Long non-coding RNA HOXA11-AS promotes cell proliferation and metastasis in human breast cancer. Mol Med Rep. 16:4887–4894. 2017. View Article : Google Scholar : PubMed/NCBI

108 

Li W, Jia G, Qu Y, Du Q and Liu B and Liu B: Long Non-coding RNA (lncRNA) HOXA11-AS promotes breast cancer invasion and metastasis by regulating Epithelial-mesenchymal transition. Med Sci Monit. 23:3393–3403. 2017. View Article : Google Scholar : PubMed/NCBI

109 

Wang K, Liang Q, Li X, Tsoi H, Zhang J, Wang H, Go MY, Chiu PW, Ng EK, Sung JJ and Yu J: MDGA2 is a novel tumour suppressor cooperating with DMAP1 in gastric cancer and is associated with disease outcome. Gut. 65:1619–1631. 2016. View Article : Google Scholar : PubMed/NCBI

110 

Sugano K: Screening of gastric cancer in Asia. Best Pract Res Clin Gastroenterol. 29:895–905. 2015. View Article : Google Scholar : PubMed/NCBI

111 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2013. CA Cancer J Clin. 63:11–30. 2013. View Article : Google Scholar : PubMed/NCBI

112 

Sun M, Nie F, Wang Y, Zhang Z, Hou J, He D, Xie M, Xu L, De W, Wang Z and Wang J: lncRNA HOXA11-AS promotes proliferation and invasion of gastric cancer by scaffolding the chromatin modification factors PRC2, LSD1, and DNMT1. Cancer Res. 76:6299–6310. 2016. View Article : Google Scholar : PubMed/NCBI

113 

Liu Z, Chen Z, Fan R, Jiang B, Chen X, Chen Q, Nie F, Lu K and Sun M: Over-expressed long noncoding RNA HOXA11-AS promotes cell cycle progression and metastasis in gastric cancer. Mol Cancer. 16:822017. View Article : Google Scholar : PubMed/NCBI

114 

Grignon DJ and Che M: Clear cell renal cell carcinoma. Clin Lab Med. 25:305–316. 2005. View Article : Google Scholar : PubMed/NCBI

115 

Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, Negrier S, Chevreau C, Solska E, Desai AA, et al: Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med. 356:125–134. 2007. View Article : Google Scholar : PubMed/NCBI

116 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

117 

Ridge CA, Pua BB and Madoff DC: Epidemiology and staging of renal cell carcinoma. Semin Intervent Radiol. 31:3–8. 2014. View Article : Google Scholar : PubMed/NCBI

118 

Yang FQ, Zhang JQ, Jin JJ, Yang CY, Zhang WJ, Zhang HM, Zheng JH and Weng ZM: HOXA11-AS promotes the growth and invasion of renal cancer by sponging miR-146b-5p to upregulate MMP16 expression. J Cell Physiol. 233:9611–9619. 2018. View Article : Google Scholar : PubMed/NCBI

119 

Robertson AG, Shih J, Yau C, Gibb EA, Oba J, Mungall KL, Hess JM, Uzunangelov V, Walter V, Danilova L, et al: Integrative analysis identifies four molecular and clinical subsets in Uveal melanoma. Cancer Cell. 32:204–220.e15. 2017. View Article : Google Scholar : PubMed/NCBI

120 

Moore AR, Ceraudo E, Sher JJ, Guan Y, Shoushtari AN, Chang MT, Zhang JQ, Walczak EG, Kazmi MA, Taylor BS, et al: Recurrent activating mutations of G-protein-coupled receptor CYSLTR2 in uveal melanoma. Nat Genet. 48:675–680. 2016. View Article : Google Scholar : PubMed/NCBI

121 

Coupland SE, Lake SL, Zeschnigk M and Damato BE: Molecular pathology of uveal melanoma. Eye (Lond). 27:230–242. 2013. View Article : Google Scholar : PubMed/NCBI

122 

Lu Q, Zhao N, Zha G, Wang H, Tong Q and Xin S: lncRNA HOXA11-AS exerts oncogenic functions by repressing p21 and miR-124 in Uveal melanoma. DNA Cell Biol. 36:837–844. 2017. View Article : Google Scholar : PubMed/NCBI

123 

Ignarski M, Islam R and Müller RU: Long Non-coding RNAs in Kidney disease. Int J Mol Sci. 20:E32762019. View Article : Google Scholar : PubMed/NCBI

124 

Dong D, Mu Z, Zhao C and Sun M: ZFAS1: A novel tumor-related long non-coding RNA. Cancer Cell Int. 18:1252018. View Article : Google Scholar : PubMed/NCBI

125 

Cheetham SW, Gruhl F, Mattick JS and Dinger ME: Long noncoding RNAs and the genetics of cancer. Br J Cancer. 108:2419–2425. 2013. View Article : Google Scholar : PubMed/NCBI

126 

Richards EJ, Permuth-Wey J, Li Y, Chen YA, Coppola D, Reid BM, Lin H, Teer JK, Berchuck A, Birrer MJ, et al: A functional variant in HOXA11-AS, a novel long non-coding RNA, inhibits the oncogenic phenotype of epithelial ovarian cancer. Oncotarget. 6:34745–34757. 2015. View Article : Google Scholar : PubMed/NCBI

127 

Steuer CE, El-Deiry M, Parks JR, Higgins KA and Saba NF: An update on larynx cancer. CA Cancer J Clin. 67:31–50. 2017. View Article : Google Scholar : PubMed/NCBI

128 

Qu L, Jin M, Yang L, Sun C, Wang P, Li Y, Tian L, Liu M and Sun Y: Expression of long non-coding RNA HOXA11-AS is correlated with progression of laryngeal squamous cell carcinoma. Am J Transl Res. 10:573–580. 2018.PubMed/NCBI

129 

Kim HJ, Eoh KJ, Kim LK, Nam EJ, Yoon SO, Kim KH, Lee JK, Kim SW and Kim YT: The long noncoding RNA HOXA11 antisense induces tumor progression and stemness maintenance in cervical cancer. Oncotarget. 7:83001–83016. 2016. View Article : Google Scholar : PubMed/NCBI

130 

Sun XY, Wang XF, Cui YB, Cao XG, Zhao RH, Wei HY, Cao W and Wu W: Expression level and clinical significance of lncRNA HOXA11-AS in esophageal squamous cell carcinoma patients. Zhonghua Zhong Liu Za Zhi. 40:186–190. 2018.(In Chinese). PubMed/NCBI

131 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

132 

Wang C, Inzana JA, Mirando AJ, Ren Y, Liu Z, Shen J, O'Keefe RJ, Awad HA and Hilton MJ: NOTCH signaling in skeletal progenitors is critical for fracture repair. J Clin Invest. 126:1471–1481. 2016. View Article : Google Scholar : PubMed/NCBI

133 

Wang XN, Zhang LH, Cui XD, Wang MX, Zhang GY and Yu PL: lncRNA HOXA11-AS is involved in fracture healing through regulating mir-124-3p. Eur Rev Med Pharmacol Sci. 21:4771–4776. 2017.PubMed/NCBI

134 

Mirzakhani H, Litonjua AA, McElrath TF, O'Connor G, Lee-Parritz A, Iverson R, Macones G, Strunk RC, Bacharier LB, Zeiger R, et al: Early pregnancy vitamin D status and risk of preeclampsia. J Clin Invest. 126:4702–4715. 2016. View Article : Google Scholar : PubMed/NCBI

135 

Powe CE, Levine RJ and Karumanchi SA: Preeclampsia, a disease of the maternal endothelium: The role of antiangiogenic factors and implications for later cardiovascular disease. Circulation. 123:2856–2869. 2011. View Article : Google Scholar : PubMed/NCBI

136 

Xu Y, Wu D, Liu J, Huang S, Zuo Q, Xia X, Jiang Y, Wang S, Chen Y, Wang T and Sun L: Downregulated lncRNA HOXA11-AS affects trophoblast cell proliferation and migration by regulating RND3 and HOXA7 expression in PE. Mol Ther Nucleic Acids. 12:195–206. 2018. View Article : Google Scholar : PubMed/NCBI

137 

Hu L, Chang L, Zhang Y, Zhai L, Zhang S, Qi Z, Yan H, Yan Y, Luo X, Zhang S, et al: Platelets express activated P2Y12 receptor in patients with diabetes mellitus. Circulation. 136:817–833. 2017. View Article : Google Scholar : PubMed/NCBI

138 

Jin QS, Huang LJ, Zhao TT, Yao XY, Lin LY, Teng YQ, Kim SH, Nam MS, Zhang LY and Jin YJ: HOXA11-AS regulates diabetic arteriosclerosis-related inflammation via PI3K/AKT pathway. Eur Rev Med Pharmacol Sci. 22:6912–6921. 2018.PubMed/NCBI

139 

Grelet S, Link LA, Howley B, Obellianne C, Palanisamy V, Gangaraju VK, Diehl JA and Howe PH: A regulated PNUTS mRNA to lncRNA splice switch mediates EMT and tumour progression. Nat Cell Biol. 19:1105–1115. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2020
Volume 43 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wei C, Zhao L, Liang H, Zhen Y and Han L: Recent advances in unraveling the molecular mechanisms and functions of HOXA11‑AS in human cancers and other diseases (Review). Oncol Rep 43: 1737-1754, 2020
APA
Wei, C., Zhao, L., Liang, H., Zhen, Y., & Han, L. (2020). Recent advances in unraveling the molecular mechanisms and functions of HOXA11‑AS in human cancers and other diseases (Review). Oncology Reports, 43, 1737-1754. https://doi.org/10.3892/or.2020.7552
MLA
Wei, C., Zhao, L., Liang, H., Zhen, Y., Han, L."Recent advances in unraveling the molecular mechanisms and functions of HOXA11‑AS in human cancers and other diseases (Review)". Oncology Reports 43.6 (2020): 1737-1754.
Chicago
Wei, C., Zhao, L., Liang, H., Zhen, Y., Han, L."Recent advances in unraveling the molecular mechanisms and functions of HOXA11‑AS in human cancers and other diseases (Review)". Oncology Reports 43, no. 6 (2020): 1737-1754. https://doi.org/10.3892/or.2020.7552