Open Access

lncRNA HIF1A‑AS2: A potential oncogene in human cancers (Review)

  • Authors:
    • Yang Liu
    • Yunyan Zhang
    • Cha Chen
    • Youqiang Li
  • View Affiliations

  • Published online on: August 13, 2021     https://doi.org/10.3892/br.2021.1461
  • Article Number: 85
  • Copyright: © Liu 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

Long non‑coding RNAs (lncRNAs) are transcripts that are >200 nucleotides, but with no open reading frame. An increasing number of lncRNAs have been identified following the development of second‑generation sequencing technologies, and they have since become a research hotspot. Functionally, they play a vital role in tumor progression, including in tumor proliferation, migration, invasion, apoptosis and acquisition of drug resistance. They regulate gene expression primarily through interaction with DNA, RNA and proteins at the epigenetic, transcriptional and post‑transcriptional levels. Endogenous hypoxia‑inducible factor 1α antisense RNA 2 (lncRNA HIF1A‑AS2) is aberrantly expressed and involved the development/progression of various types of tumors, such as bladder cancer, glioblastoma, breast cancer and osteosarcoma. It plays a vital role in the proliferation, apoptosis, migration, invasion and epithelial‑mesenchymal transformation of various tumor cells. This review summarizes the current body of knowledge on the biological functions and related molecular mechanisms of lncRNA HIF1A‑AS2 in the development/progression of human tumors and other diseases.

1. Introduction

Technological advances have driven an improved understanding of protein-coding genes; however, the functional roles of non-coding (nc)RNAs are relatively less well understood. ncRNAs account for >90% of the human genome, whereas protein-coding genes account for only 1.5% (1,2). Based on transcript size, ncRNAs are divided into two groups: Small ncRNAs with transcripts <200 nucleotides and long ncRNAs (lncRNAs) with transcripts >200 nucleotides in length (3). lncRNAs, first discovered in the sequencing of cDNA libraries in mouse cells (4), are mRNA-like transcripts that are likely transcribed by RNA polymerase II (RNA pol II), but which lack a stable open reading frame (5). Initially, these non-coding RNAs were viewed as by-products and noise of the transcription process (4). However, with the continuous development of gene technologies, a large number of studies have found that lncRNAs are involved in various physiological and pathological processes.

lncRNA HIF1A-AS2, also known as HIF1A-AS2, is the endogenous antisense transcript of hypoxia-inducible factor 1α (HIF1α), and 3'aHIF, termed HIF1α antisense RNA 2 (HIF1A-AS2), is localized at chromosome 14q23.2, and is 2,052 nucleotides in length. In 1999, it was first discovered to be abnormally expressed in clear cell renal carcinoma by Thrash-Bingham and Tartof (6), and was identified as the endogenous antisense transcript, which could bind to the 3' untranslated region (3'UTR) of HIF1α mRNA in a complementary manner (Fig. 1), and this bound form is referred to as aHIF. In 2002, Rossignol et al (7) reported that HIF1A-AS2 was expressed in several human tissues, both physiologically, and when the tissues had become cancerous. These findings attracted increased focus on HIF1A-AS2. Further studies demonstrated that HIF1A-AS2 was aberrantly expressed in various human diseases, including preeclampsia (PE), epithelial ovarian cancer (EOC), colorectal cancer (CRC), gastric cancer (GC), breast cancer (BC), bladder cancer, osteosarcoma (OS), renal cell carcinoma, non-small cell lung cancer (NSCLC) and glioblastoma (GBM). Chen et al (8) reported that the expression levels of HIF1A-AS2 were upregulated in GC tissues and cells, and this upregulated expression was correlated with Tumor-Node-Metastasis stage, tumor invasion, lymph node metastasis and a poor prognosis. Lin et al (9) also demonstrated upregulated expression of HIF1A-AS2 in 60 OS tissues compared with the adjacent healthy tissues. Thus, HIF1A-AS2 may serve as a promising target for treatment of several types of cancer.

However, several studies demonstrated that the expression levels of HIF1A-AS2 in tumor tissues was abnormal, indicating the potential correlation between HIF1A-AS2 and cancer. Therefore, this review summarizes the current body of knowledge regarding the aberrant expression of this lncRNA (Table I), its function and the regulatory mechanisms of HIF1A-AS2 in several types of cancer.

Table I

Expression and function of long non-coding RNA HIF1α antisense RNA in PE and various types of cancer.

Table I

Expression and function of long non-coding RNA HIF1α antisense RNA in PE and various types of cancer.

DiseaseChange in expressionRoleBiological functionRelated genesRefs.
PEDownPathogenicProliferation, migration, invasion, pro-apoptosis, cell cycle arrestLSD1, PHLDA1(14)
Epithelial ovarian cancerUpOncogenicProliferation, migration and invasionBax, caspase-7, caspase-9, BCL-2, caspase-3(18)
Colorectal cancerUpOncogenicProliferation, migration and invasionmiR-129-5p, miR-33b-5p DNMT3A(23)
Gastric cancerUpOncogenicProliferation, migration and invasion-(8)
Breast cancerUpOncogenicProliferation, migration and invasionmiR-548c-3p, HIF1α, VEGF(30)
Bladder cancerUpOncogenicProliferation, migration, invasion and anti-apoptosis-(35)
OsteosarcomaUpOncogenicProliferation, migration, invasion and anti-apoptosismiR-33b-5p, SIRT6, miR-129-5p(9,40)
GlioblastomaUpOncogenicNeurosphere formationIGF2BP2, DHX9, HMGA1(43)
Renal cancerUpOncogenicProliferation, migration, invasion and anti-apoptosisHIF1α, miR-130-5p(6,50)
Non-small cell lung cancerUpOncogenicProliferation, migration, invasion and anti-apoptosismiR-153b-5p, S100A14(54)

[i] HIF1α, hypoxia-inducible factor 1α; LSD1, lysine-specific demethylase 1; PHLDA1, pleckstrin homology like domain, family A, member 1; IGF2BP2, insulin-like growth factor 2; DHX9, ATP-dependent RNA helicase A; HMGA1, high mobility group AT-hook 1; VEGF, vascular endothelial growth factor; BCL-2, B-cell lymphoma 2; BAX, Bcl-2-associated X protein; miRNA, microRNA; PE, preeclampsia.

2. Expression and function of lncRNA HIF1A-AS2 in several types of cancer

PE

PE is one of the leading causes of maternal death and a pregnancy-specific disease, affecting 3-14% of parturients worldwide (10). Although PE has been extensively studied (11), the underlying pathogenesis of PE remains elusive. However, it is hypothesized that inadequate trophoblastic invasion may cause PE (12,13). Wu et al (14) reported that HIF1A-AS2 expression was significantly downregulated in the tissues of 52 patients with PE compared with the adjacent normal samples. Knockdown of HIF1A-AS2 expression significantly inhibited proliferation, migration and invasion, as well as inducing G0/G1 cell cycle arrest and increased cell apoptosis in two trophoblast cell lines (HTR/SVneo and JAR). In contrast, overexpression of HIF1A-AS2 exerted the opposite effect. Mechanistically, a subcellular localization assay indicated that HIF1A-AS2 was primarily localized in the cell nucleus; thus, it may play a role in regulation of transcription. Further experiments showed that HIF1A-AS2 inhibited the transcription of pleckstrin homology like domain, family A, member 1 (PHLDA1), which plays a significant role in the activation-induced apoptosis following binding to lysine-specific demethylase (LSD1) at the epigenetic level. Furthermore, chromatin immunoprecipitation assays showed LSD1 and H3K4 me2 enrichment in the promoter region of the PHLDA1 gene (Fig. 2A) after transfection with small-interfering (si)-HIF1A-AS2. Thus, HIF1A-AS2 may be a useful diagnostic biomarker for PE.

EOC

Ovarian cancer (OC) is one of the most common types of malignant tumors in females, with EOC being the most common, accounting for 80-90% of OC cases (15,16) . Although EOC treatments have improved notably, even in developed countries, such as the United States and Canada, the overall survival remains at only 47% 5 years after diagnosis (17). Therefore, investigating the molecular mechanism and finding effective therapeutic targets for management of EOC is of great importance. Qiu et al (18) reported that the expression of HIF1A-AS2 in EOC tissues was significantly higher compared with the normal controls, and HIF1A-AS2 was a lncRNA that was upregulated under hypoxic conditions. Thus, the following assays were performed under hypoxic conditions. Functional assays revealed that knockdown of HIF1A-AS2 promoted cell apoptosis and weakened tumorigenesis in nude mice. In contrast, overexpression of HIF1A-AS2 inhibited EOC cell apoptosis and enhanced cell proliferation.

Further mechanistic experiments showed that HIF1A-AS2 functions by regulating the mitochondrial apoptosis pathway-related genes (Fig. 2B). Briefly, HIF1A-AS2 knockdown resulted in increased expression of Bax, Bcl-2, caspase-7, and caspase-9 at the mRNA level under hypoxic conditions. Thus, overexpression of HIF1A-AS2 may serve as a diagnostic biomarker for EOC.

CRC

CRC is the third most common type of cancer and the fourth leading cause of cancer-associated death globally (19,20). At present, chemotherapy is an essential treatment for CRC; however, both the incidence and death rate of CRC is increasing rapidly (21,22). Thus, it is crucial to identify novel critical genes involved in the pathogenesis of CRC to develop effective treatments. Lin et al (23) observed upregulated expression of HIF1A-AS2 in CRC tissues and cells compared with the healthy controls. Moreover, high expression of HIF1A-AS2 was strongly associated with a poor prognosis and advanced TNM stages in patients with CRC. Functionally, knockdown of HIF1A-AS2 inhibited the proliferation, invasion and epithelial-mesenchymal transformation (EMT) of CRC cells in-vitro. HIF1A-AS2 mechanistically functioned as a competing endogenous (ce)RNA binding to microRNA (miR)-129-5p (Fig. 2C), a tumor suppressor. Consistent with this, DNMT3A was identified to be a target of miR-129–5p. The critical role of the HIF1A-AS2/miR-129-5p/DNMT3A axis in the proliferation, invasion and EMT of CRC cells was further confirmed by reverse transcription-quantitative PCR and dual- luciferase reporter assays. Thus, due to its oncogenic role and clinical significance in colorectal cancer, HIF1A-AS2 may be considered a diagnostic biomarker and prognostic indicator for CRC.

GC

GC is the third leading cause of cancer-associated death worldwide, with ~1,000,000 newly diagnosed cases each year, and a higher rate of occurrence in East Asia (24,25). The majority of patients are diagnosed with advanced stage GC, and thus, GC has a high mortality rate (26). Therefore, it has been a central issue to study the pathogenic mechanisms of GC and identify effective tumor markers to improve early diagnosis. Chen et al (8) reported that HIF1A-AS2 was upregulated in 38 GC samples and four human GC cell lines compared with the matched paracarcinoma tissues or a normal GC cell line (GES-1), respectively. The high expression of HIF1A-AS2 was significantly associated with a more advanced TNM stage, tumor invasion, lymph node metastasis and a poor prognosis. Functionally, knockdown of HIF1A-AS2 suppressed the proliferative ability of GC cells in-vitro and restrained tumor weight and volume in nude mice. In addition, it was found that HIF1A-AS2 had value in the early diagnosis of GC and could be used as a potential diagnostic marker for detection of GC. Therefore, HIF1A-AS2 is a potential tumorigenic gene in GC, but its molecular mechanisms have not been studied, to the best of our knowledge.

BC

BC is the most common malignancy and the leading cause of cancer-related death in women (27). Breast cancer tumors usually express a combination of the following receptors: Estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor (HER2). Cases that lack expression of these three receptors are termed triple-negative breast cancer (TNBC). TNBC accounts for ~20% of all breast cancer cases, ad is most common in women >40(28). TNBC is highly invasive, with high mortality and recurrence rates. Current treatments for TNBC include surgery, chemotherapy, radiotherapy and targeted therapy. However, the median overall survival rarely extends beyond 18 months in patients with advanced BC (29). Therefore, it is essential to study the molecular mechanism and identify novel biomarkers for management of TNBC. Guo et al (30) showed that HIF1A-AS2 was significantly overexpressed in four BC cell lines compared with a normal mammary epithelial cell line. Knockdown of HIF1A-AS2 levels effectively suppress proliferation, invasion, EMT and senescence of MCF-7 cell lines in-vitro. In vivo studies also showed that tumor growth was reduced after the knockdown of HIF1A-AS2 by short hairpin (sh)RNA targeting HIF1A-AS2 in-vivo, thus indicating that HIF1A-AS2 functions as an oncogene. Mechanistically, a HIF1A-AS2/miR-548c-3p/HIF1a/VEGF axis was confirmed to regulate the proliferation, invasion, migration and EMT of BC cells. Jiang et al (31) also reported that expression of HIF1A-AS2 was increased in 33 TNBC tissues compared with the adjacent normal breast tissues. Knockdown of HIF1A-AS2 functionally suppressed TNBC cell proliferation. These results indicated that HIF1A-AS2 was involved in the pathogenesis of TNBC, suggesting that it could be a prognostic indicator or therapeutic target for TNBC.

Bladder cancer

Bladder cancer is one of the most common malignancies of the urinary system worldwide, posing a severe threat to human health (32). Surgery, radiotherapy and chemotherapy are the primary modes of treatment for bladder cancer; however, the 5-year overall survival rate is only 50-60% (33). Although several studies have demonstrated novel biomarkers for the early detection and diagnosis of bladder cancer, the survival rate of patients with bladder cancer remains very low (34). Therefore, it is necessary to identify novel biomarkers to improve the early diagnosis and prognosis of bladder cancer. Chen et al (35) revealed that the expression of HIF1A-AS2 was significantly upregulated in 44 bladder cancer samples and cancer cell lines (5637 and T24) compared with the matched normal peritumoral tissues or the SVHUC-1 normal bladder cell line. In addition, the upregulated HIF1A-AS2 expression was closely related to histological grade, tumor invasion depth and TNM stage. These results indicated that lncRNA HIF1A-AS2 may function as an oncogene in bladder cancer. Functionally, knockdown of HIF1A-AS2 significantly inhibited bladder cancer cell proliferation and migration, and increased apoptosis. Conversely, overexpression of HIF1A-AS2 had the opposite effect.

Furthermore, a tetracycline-induced shRNA using medical synthetic biology techniques was designed, which could effectively inhibit the expression of HIF1A-AS2 in a dose-dependent manner, and in turn inhibited cell growth and migration, and induced apoptosis in bladder cancer cells. It also indicated that tetracycline-induced shRNA may be a novel approach for quantitatively controlling specific targets in human cancers, and may be an effective treatment method for bladder cancer. Thus, HIF1A-AS2 may serve as a target for the treatment of bladder cancer; however, the exact molecular regulatory mechanisms in bladder cancer require further study.

OS

OS is a skeletal system primary malignant tumor, common amongst the younger population, particularly children and adolescents (36,37). OS accounts for 60% of all sarcoma cases, characterized by early metastasis, high aggressiveness, a high rate of disability and a high recurrence rate (38). Despite advances in OS treatment, the overall survival of patients has not substantially increased, the 5-year overall survival still remains only 20% over the past 30 years (39). Thus, understanding the molecular mechanism of OS and identifying novel therapeutic targets is of great clinical significance to improve early diagnosis and survival rates of patients with OS. Lin et al (9) observed increased HIF1A-AS2 expression in 60 OS samples and four OS cell lines when compared with the 60 adjacent normal samples or the hFOB 1.19 cells, respectively. In addition, high expression of HIF1A-AS2 was significantly associated with a larger tumor size, higher tumor grade, advanced stage disease and distance of metastasis.

Furthermore, Kaplan-Meier survival analysis showed that the 5-year survival rate of the high HIF1A-AS2 expression group was lower than the low HIF1A-AS2 expression group. Knockdown of HIF1A-AS2 resulted in decreased cell proliferation, migration and invasion, increased cell cycle arrest in the G0/G1-phase and an increased percentage of apoptotic cells. In in-vivo experiments, knockdown of HIF1A-AS2 resulted in reduced tumor size in nude mice. Mechanistically, HIF1A-AS2/miR-33b-5p/SIRT6 was confirmed to regulate OS cell proliferation, migration and apoptosis. Wang et al (40) also confirmed increased expression of HIF1A-AS2 in 30 OS samples and four OS cell lines compared with the adjacent normal tissues and osteoblast cell lines, respectively. Moreover, high HIF1A-AS2 expression was associated with poor survival rates. Functional assays revealed that HIF1A-AS2 overexpression promoted osteosarcoma cell proliferation, cell cycle progression and invasion. HIF1A-AS2 mechanistically served as a ceRNA to negatively regulate miR-129–5p (Fig. 2C). Thus, HIF1A-AS2 may be an effective diagnostic and prognostic indicator of OS.

GBM

GBM is the most common and aggressive primary malignant brain tumor, with a median patient survival time of 14-16 months (41). GBM is a highly proliferative and invasive tumor with a poor prognosis. Despite advances in GBM treatment, patients are still likely to face a poor prognosis (42). Therefore, new therapeutic methods and targets are required. lncRNAs are involved in the development of GBM. Mineo et al (43) reported that HIF1A-AS2 contributes to the formation of stem-like glioma cells (GSCs) in the tumor microenvironment and their adaptation to hypoxia. Based on characterization of the GBM genome and transcriptome, GBM can be divided into several cellular subtypes, including mesenchymal (M), proneural (P), neural (N), and classical (C) (44). Patients with the aggressive and predominant M subtype exhibit a particularly high degree of tumor necrosis (45). It was observed that HIF1A-AS2 expression was significantly increased in the GSCs of patients with the M subtype. Moreover, knockdown of HIF1A-AS2 led to reduced growth, decreased cellular activity and decreased neurosphere-forming capacity of M GSC cells (43).

Furthermore, the HIF1A-AS2 expression is increased under hypoxic conditions. In order to clarify the pro-oncogenic function of HIF1A-AS2, researchers revealed that knockdown of HIF1A-AS2 by shRNA resulted in smaller tumor sizes in nude mice. Mechanistic experiments showed that HIF1A-AS2 could bind to IGF2BP2 and DHX9 to directly modulate the expression of HMGA1 (Fig. 2D) and maintain the growth of M GSCs under hypoxic conditions (43). In addition, Liao et al (46) showed that the upregulated HIF1A-AS2 expression could mediate radiation resistance of the glioma, leading to tumor recurrence following radiotherapy by regulating expression of apoptotic proteins. Knockdown of HIF1A-AS2 increased the expression of the pro-apoptotic protein caspase 7 and the number of apoptotic cells. Thus, HIF1A-AS2 may be a novel diagnostic indicator and potential therapeutic target for the management of GBM.

RCC

RCC is one of the most common malignancies of the urinary system, and accounts for 2-3% of all malignancies (47,48). The estimated number of new cases and deaths worldwide in 2018 were 403,262 and 175,098, respectively (49). Relatively fewer biomarkers for RCC have been identified when compared with other types of cancer. Thus, it is essential to identify novel and sensitive biomarkers to predict the progress and prognosis of the disease. In 1999, Thrash-Bingham and Tartof (6) first discovered a natural antisense transcript that could bind to the 3'UTR of HIF1α mRNA in non-papillary kidney cancer and termed it aHIF, for which the official gene symbol is now HIF1A-AS2. Expression of HIF1A-AS2 is increased in non-papillary renal carcinoma cells compared with the control cells, but not in papillary renal carcinoma cells. It is hypothesized that decreased HIF1a mRNA expression through HIF1A-AS2 may serve an important role in regulating P53 to regulate progression of cancer, but this mechanism requires further investigation (6). Zhu et al (50) also reported increased expression of HIF1A-AS2 in kidney cancer tissues and RCC cells compared with the non-cancerous tissues. In addition, knockdown of HIF1A-AS2 inhibited renal cancer cell proliferation, invasion and migration, whilst accelerating cell apoptosis. Overexpression of HIF1A-AS2 resulted in the opposite effect. HIF1A-AS2 mechanistically functions as a ceRNA, binding to miR-130a-5p (Fig. 2C) to modulate renal carcinoma progression. Thus, HIF1A-AS2 may be a promising diagnostic biomarker and a potential therapeutic target for management of renal cancer.

NSCLC

Lung cancer is the most common type of cancer and the leading cause of cancer-associated death worldwide. The majority of patients are diagnosed with advanced stage disease in the first instance, and NSCLC accounts for nearly 85% of patients with lung cancer (51). Despite advances in cancer treatment, lung cancer has a high mortality rate, accounting for 18.4% of all cancer deaths (52,53). Thus, understanding the molecular mechanism of NSCLC and identifying novel therapeutic targets is of great clinical significance. Zhang et al (54) reported elevated expression levels of HIF1A-AS2 in NSCLC tissues and cell lines, and this increased expression was associated with a poor prognosis. However, knockdown of HIF1A-AS2 resulted in decreased cell proliferation, migration and invasion, and an increased percentage of apoptotic cells. Mechanistically, a HIF1A-AS2/miR-153–5p/S100A14 axis was confirmed to regulate NSCLC cell proliferation, migration and apoptosis (Fig. 2C). Thus, HIF1A-AS2 may be an effective diagnostic and prognostic indicator for NSCLC.

3. Conclusions and future perspective

A wealth of studies have shown that lncRNAs exert their functions through various mechanisms, such as associating with transcription factors, chromatin modifiers, signaling adapters, enzymes and miRNAs, to influence gene expression, post-translational modifications and protein activities (55).

HIF1A-AS2 has been reported to regulate cellular pathological processes, but is primarily focused on tumors. HIF1A-AS2 is primarily functions as a protein scaffold, protein decoy and a ceRNA. Mineo et al (43) reported that HIF1A-AS2 acts as a protein scaffold to bind both IGF2BP2 and DHX9 to modulate the expression of HMGA1. Wu et al (14) reported that HIF1A-AS2 functions as a protein decoy to inhibit the transcription of PHLDA1 by binding to LSD1, a histone demethylase. Additionally, HIF1A-AS2 acts as a molecular sponge to bind miRNAs to further affect expression of other genes (23,40,50,54,56). Although significant achievements have been obtained with regard to understanding the role of HIF1A-AS2 in various types of cancer, further studies are still required with regard to its regulatory function, as lncRNAs often exhibit several complex regulatory functions/mechanisms.

Studies have shown that HIF1A-AS2 may serve as a novel biomarker for the clinical diagnosis of several types of cancer. These data demonstrate that upregulated expression of HIF1A-AS2 is associated with poor overall survival and an unfavorable prognosis, such as in TNBC, OS and CRC. Nevertheless, the clinical diagnostic value of HIF1A-AS2 in these types of cancer needs to be validated using large-scale multicenter cohorts.

Acknowledgements

Not applicable.

Funding

This study was supported by grants from The National Natural Science Fund of China (grant no. 81601736); Science and Technology Planning Project of Panyu District, China (grant no. 2019-Z04-85); Medical Scientific Research Foundation of Guangdong Province, China (grant no. A2020560); Basic and Applied Research Project of Guangzhou Research Program, China (grant no. 202102080539).

Availability of data and materials

Not applicable.

Authors' contributions

YLi conceived and designed the study. YLiu, YZ and CC participated in drafting and revising the article. All authors have read and approved the final manuscript. Data authentication is not applicable.

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 

Slack FJ and Chinnaiyan AM: The role of non-coding RNAs in oncology. Cell. 179:1033–1055. 2019.PubMed/NCBI View Article : Google Scholar

2 

Esteller M: Non-coding RNAs in human disease. Nat Rev Genet. 12:861–874. 2011.PubMed/NCBI View Article : Google Scholar

3 

Khandelwal A, Bacolla A, Vasquez KM and Jain A: Long non-coding RNA: A new paradigm for lung cancer. Mol Carcinog. 54:1235–1251. 2015.PubMed/NCBI View Article : Google Scholar

4 

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

5 

Bhat SA, Ahmad SM, Mumtaz PT, Malik AA, Dar MA, Urwat U, Shah RA and Ganai NA: Long non-coding RNAs: Mechanism of action and functional utility. Noncoding RNA Res. 1:43–50. 2016.PubMed/NCBI View Article : Google Scholar

6 

Thrash-Bingham CA and Tartof KD: aHIF: A natural antisense transcript overexpressed in human renal cancer and during hypoxia. J Natl Cancer Inst. 91:143–151. 1999.PubMed/NCBI View Article : Google Scholar

7 

Rossignol F, Vaché C and Clottes E: Natural antisense transcripts of hypoxia-inducible factor 1alpha are detected in different normal and tumour human tissues. Gene. 299:135–140. 2002.PubMed/NCBI View Article : Google Scholar

8 

Chen WM, Huang MD, Kong R, Xu TP, Zhang EB, Xia R, Sun M, De W and Shu YQ: Antisense long noncoding RNA HIF1A-AS2 is upregulated in gastric cancer and associated with poor prognosis. Dig Dis Sci. 60:1655–1662. 2015.PubMed/NCBI View Article : Google Scholar

9 

Lin H, Zhao Z, Hao Y and He J and He J: Long noncoding RNA HIF1A-AS2 facilitates cell survival and migration by sponging miR-33b-5p to modulate SIRT6 expression in osteosarcoma. Biochem Cell Biol. 98:284–292. 2020.PubMed/NCBI View Article : Google Scholar

10 

Wu P, Haththotuwa R, Kwok CS, Babu A, Kotronias RA, Rushton C, Zaman A, Fryer AA, Kadam U, Chew-Graham CA, et al: Preeclampsia and future cardiovascular health: A systematic review and meta-analysis. Circ Cardiovasc Qual Outcomes. 10(e003497)2017.PubMed/NCBI View Article : Google Scholar

11 

Phipps EA, Thadhani R, Benzing T and Karumanchi SA: Pre-eclampsia: Pathogenesis, novel diagnostics and therapies (vol 15, pg 275, 2019). Nat Rev Nephrol. 15:386. 2019.PubMed/NCBI View Article : Google Scholar

12 

Irminger-Finger I, Jastrow N and Irion O: Preeclampsia: A danger growing in disguise. Int J Biochem Cell Biol. 40:1979–1983. 2008.PubMed/NCBI View Article : Google Scholar

13 

Ma Y, Liang X, Wu H, Zhang C and Ma Y: Long non coding RNA NR_002794 is upregulated in pre eclampsia and regulates the proliferation, apoptosis and invasion of trophoblast cells. Mol Med Rep. 20:4567–4575. 2019.PubMed/NCBI View Article : Google Scholar

14 

Wu D, Yang N, Xu Y, Wang S, Zhang Y, Sagnelli M, Hui B, Huang Z and Sun L: lncRNA HIF1A antisense RNA 2 modulates trophoblast cell invasion and proliferation through upregulating PHLDA1 expression. Mol Ther Nucleic Acids. 16:605–615. 2019.PubMed/NCBI View Article : Google Scholar

15 

Tang X, Liu S, Liu Y, Lin X, Zheng T, Liu X, Qiu J and Hua K: Circulating serum exosomal aHIF is a novel prognostic predictor for epithelial ovarian cancer. Onco Targets Ther. 12:7699–7711. 2019.PubMed/NCBI View Article : Google Scholar

16 

Guler E, Smith DA, Somarouthu B, Gujrathi R, Ramaiya NH and Tirumani SH: Overview of imaging findings associated with systemic therapies in advanced epithelial ovarian cancer. Abdom Radiol (NY). 45:828–841. 2020.PubMed/NCBI View Article : Google Scholar

17 

Lheureux S, Braunstein M and Oza AM: Epithelial ovarian cancer: Evolution of management in the era of precision medicine. CA Cancer J Clin. 69:280–304. 2019.PubMed/NCBI View Article : Google Scholar

18 

Qiu JJ, Lin XJ, Zheng TT, Tang XY and Hua KQ: Natural antisense transcript of hypoxia-inducible factor 1 regulates hypoxic cell apoptosis in epithelial ovarian cancer. Onco Targets Ther. 11:9101–9110. 2018.PubMed/NCBI View Article : Google Scholar

19 

Yang J, McDowell A, Kim EK, Seo H, Lee WH, Moon CM, Kym SM, Lee DH, Park YS, Jee YK, et al: Development of a colorectal cancer diagnostic model and dietary risk assessment through gut microbiome analysis. Exp Mol Med. 51:1–15. 2019.PubMed/NCBI View Article : Google Scholar

20 

Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global patterns and trends in colorectal cancer incidence and mortality. Gut. 66:683–691. 2017.PubMed/NCBI View Article : Google Scholar

21 

Yuan W, Peng S, Wang J, Wei C, Ye Z, Wang Y, Wang M, Xu H, Jiang S, Sun D, et al: Identification and characterization of circRNAs as competing endogenous RNAs for miRNA-mRNA in colorectal cancer. PeerJ. 7(e7602)2019.PubMed/NCBI View Article : Google Scholar

22 

Dong Y, Zhang Y, Kang W, Wang G, Chen H, Higashimori A, Nakatsu G, Go M, Tong JH, Zheng S, et al: VSTM2A suppresses colorectal cancer and antagonizes Wnt signaling receptor LRP6. Theranostics. 9:6517–6531. 2019.PubMed/NCBI View Article : Google Scholar

23 

Lin J, Shi Z, Yu Z and He Z: lncRNA HIF1A-AS2 positively affects the progression and EMT formation of colorectal cancer through regulating miR-129-5p and DNMT3A. Biomed Pharmacother. 98:433–439. 2018.PubMed/NCBI View Article : Google Scholar

24 

Fan H, Jin X, Liao C, Qiao L and Zhao W: MicroRNA-301b-3p accelerates the growth of gastric cancer cells by targeting zinc finger and BTB domain containing 4. Pathol Res Pract. 215(152667)2019.PubMed/NCBI View Article : Google Scholar

25 

Wan P, Bai X, Yang C, He T, Luo L, Wang Y, Fan M, Wang Z, Lu L, Yin Y, et al: miR-129-5p inhibits proliferation, migration, and invasion in rectal adenocarcinoma cells through targeting E2F7. J Cell Physiol. 235:5689–5701. 2020.PubMed/NCBI View Article : Google Scholar

26 

Wang LL, Zhang L and Cui XF: Downregulation of long noncoding RNA LINC01419 inhibits cell migration, invasion, and tumor growth and promotes autophagy via inactivation of the PI3K/Akt1/mTOR pathway in gastric cancer. Ther Adv Med Oncol. 11(1758835919874651)2019.PubMed/NCBI View Article : Google Scholar

27 

Lee KL, Kuo YC, Ho YS and Huang YH: Triple-negative breast cancer: Current understanding and future therapeutic breakthrough targeting cancer Stemness. Cancers (Basel). 11(1334)2019.PubMed/NCBI View Article : Google Scholar

28 

Khan MA, Jain VK, Rizwanullah M, Ahmad J and Jain K: PI3K/AKT/mTOR pathway inhibitors in triple-negative breast cancer: A review on drug discovery and future challenges. Drug Discov Today. 24:2181–2191. 2019.PubMed/NCBI View Article : Google Scholar

29 

Song N, Zhao L, Zhu M and Zhao J: 99mTc-Labeled LyP-1 for SPECT Imaging of Triple Negative Breast Cancer. Contrast Media Mol Imaging. 2019(9502712)2019.PubMed/NCBI View Article : Google Scholar

30 

Guo X, Lee S and Cao P: The inhibitive effect of sh-HIF1A-AS2 on the proliferation, invasion, and pathological damage of breast cancer via targeting miR-548c-3p through regulating HIF-1α/VEGF pathway in vitro and vivo. Onco Targets Ther. 12:825–834. 2019.PubMed/NCBI View Article : Google Scholar

31 

Jiang YZ, Liu YR, Xu XE, Jin X, Hu X, Yu KD and Shao ZM: Transcriptome Analysis of triple-negative breast cancer reveals an integrated mRNA-lncRNA signature with predictive and prognostic value. Cancer Res. 76:2105–2114. 2016.PubMed/NCBI View Article : Google Scholar

32 

Yu G, Zhou H, Yao W, Meng L and Lang B: lncRNA TUG1 promotes cisplatin resistance by regulating CCND2 via epigenetically silencing miR-194-5p in bladder cancer. Mol Ther Nucleic Acids. 16:257–271. 2019.PubMed/NCBI View Article : Google Scholar

33 

Miao L, Liu HY, Zhou C and He X: LINC00612 enhances the proliferation and invasion ability of bladder cancer cells as ceRNA by sponging miR-590 to elevate expression of PHF14. J Exp Clin Cancer Res. 38(143)2019.PubMed/NCBI View Article : Google Scholar

34 

Zhuang C, Ma Q, Zhuang C, Ye J, Zhang F and Gui Y: lncRNA GClnc1 promotes proliferation and invasion of bladder cancer through activation of MYC. FASEB J. 33:11045–11059. 2019.PubMed/NCBI View Article : Google Scholar

35 

Chen M, Zhuang C, Liu Y, Li J, Dai F, Xia M, Zhan Y, Lin J, Chen Z, He A, et al: Tetracycline-inducible shRNA targeting antisense long non-coding RNA HIF1A-AS2 represses the malignant phenotypes of bladder cancer. Cancer Lett. 376:155–164. 2016.PubMed/NCBI View Article : Google Scholar

36 

Qi X, Yu XJ, Wang XM, Song TN, Zhang J, Guo XZ, Li GJ and Shao M: Knockdown of KCNQ1OT1 suppresses cell invasion and sensitizes osteosarcoma cells to CDDP by upregulating DNMT1-mediated Kcnq1 expression. Mol Ther Nucleic Acids. 17:804–818. 2019.PubMed/NCBI View Article : Google Scholar

37 

Jiang N, Wang X, Xie X, Liao Y, Liu N, Liu J, Miao N, Shen J and Peng T: lncRNA DANCR promotes tumor progression and cancer stemness features in osteosarcoma by upregulating AXL via miR-33a-5p inhibition. Cancer Lett. 405:46–55. 2017.PubMed/NCBI View Article : Google Scholar

38 

Zhang CL, Zhu KP and Ma XL: Antisense lncRNA FOXC2-AS1 promotes doxorubicin resistance in osteosarcoma by increasing the expression of FOXC2. Cancer Lett. 396:66–75. 2017.PubMed/NCBI View Article : Google Scholar

39 

Zhong L, Liao D, Li JJ, Liu WQ, Wang JX, Zeng CL, Wang X, Cao ZL, Zhang RH, Li M, et al: Rab22a-NeoF1 fusion protein promotes osteosarcoma lung metastasis through its secretion into exosomes. Signal Transduct Target Ther. 6(59)2021.PubMed/NCBI View Article : Google Scholar

40 

Wang X, Peng L, Gong X, Zhang X and Sun R: lncRNA HIF1A-AS2 promotes osteosarcoma progression by acting as a sponge of miR-129-5p. Aging (Albany NY). 11:11803–11813. 2019.PubMed/NCBI View Article : Google Scholar

41 

Tang C, Wang Y, Zhang L, Wang J, Wang W, Han X, Mu C and Gao D: Identification of novel lncRNA targeting Smad2/PKCα signal pathway to negatively regulate malignant progression of glioblastoma. J Cell Physiol. 235:3835–3848. 2020.PubMed/NCBI View Article : Google Scholar

42 

Ji J, Xu R, Ding K, Bao G, Zhang X, Huang B, Wang X, Martinez A, Wang X, Li G, et al: Long Noncoding RNA SChLAP1 forms a growth-promoting complex with HNRNPL in human glioblastoma through stabilization of ACTN4 and activation of NF-κB signaling. Clin Cancer Res. 25:6868–6881. 2019.PubMed/NCBI View Article : Google Scholar

43 

Mineo M, Ricklefs F, Rooj AK, Lyons SM, Ivanov P, Ansari KI, Nakano I, Chiocca EA, Godlewski J and Bronisz A: The Long Non-coding RNA HIF1A-AS2 facilitates the maintenance of mesenchymal glioblastoma stem-like cells in hypoxic niches. Cell Rep. 15:2500–2509. 2016.PubMed/NCBI View Article : Google Scholar

44 

Wang L, Babikir H, Müller S, Yagnik G, Shamardani K, Catalan F, Kohanbash G, Alvarado B, Di Lullo E, Kriegstein A, et al: The phenotypes of proliferating glioblastoma cells reside on a single axis of variation. Cancer Discov. 9:1708–1719. 2019.PubMed/NCBI View Article : Google Scholar

45 

Bhat KPL, Balasubramaniyan V, Vaillant B, Ezhilarasan R, Hummelink K, Hollingsworth F, Wani K, Heathcock L, James JD, Goodman LD, et al: Mesenchymal differentiation mediated by NF-κB promotes radiation resistance in glioblastoma. Cancer Cell. 24:331–346. 2013.PubMed/NCBI View Article : Google Scholar

46 

Liao K, Ma X, Chen B, Lu X, Hu Y, Lin Y, Huang R and Qiu Y: Upregulated AHIF-mediated radioresistance in glioblastoma. Biochem Biophys Res Commun. 509:617–623. 2019.PubMed/NCBI View Article : Google Scholar

47 

Kovacs G, Wilkens L, Papp T and de Riese W: Differentiation between papillary and nonpapillary renal cell carcinomas by DNA analysis. J Natl Cancer Inst. 81:527–530. 1989.PubMed/NCBI View Article : Google Scholar

48 

Korenaga Y, Naito K, Okayama N, Hirata H, Suehiro Y, Hamanaka Y, Matsuyama H and Hinoda Y: Association of the BCRP C421A polymorphism with nonpapillary renal cell carcinoma. Int J Cancer. 117:431–434. 2005.PubMed/NCBI View Article : Google Scholar

49 

Zhang C, Huang D, Liu A, Xu Y, Na R and Xu D: Genome-wide screening and cohorts validation identifying novel lncRNAs as prognostic biomarkers for clear cell renal cell carcinoma. J Cell Biochem. 121:2559–2570. 2020.PubMed/NCBI View Article : Google Scholar

50 

Zhu Y, Yang Z, Chen H, Pan Y, Gong L, Chen F, Jin X, Wen S, Li Y and Chen G: lncRNAHIF1A-AS2 promotes renal carcinoma cell proliferation and migration via miR-130a-5p/ERBB2 pathway. Onco Targets Ther. 13:9807–9820. 2020.PubMed/NCBI View Article : Google Scholar

51 

The Lancet: Lung cancer: Some progress, but still a lot more to do. The Lancet. 394(1880)2019.PubMed/NCBI View Article : Google Scholar

52 

Ramalingam SS, Owonikoko TK and Khuri FR: Lung cancer: New biological insights and recent therapeutic advances. CA Cancer J Clin. 61:91–112. 2011.PubMed/NCBI View Article : Google Scholar

53 

Dawson Q: NELSON trial: Reduced lung-cancer mortality with volume CT screening. Lancet Respir Med. 8:236. 2020.PubMed/NCBI View Article : Google Scholar

54 

Zhang W, Liu K, Pei Y, Tan J, Ma J and Zhao J: Long noncoding RNA HIF1A-AS2 promotes non-small cell lung cancer progression by the miR-153-5p/S100A14 axis. Onco Targets Ther. 13:8715–8722. 2020.PubMed/NCBI View Article : Google Scholar

55 

Wang P: The Opening of Pandora's Box: An emerging role of long noncoding RNA in viral infections. Front Immunol. 9(3138)2019.PubMed/NCBI View Article : Google Scholar

56 

Mu L, Wang Y, Su H, Lin Y, Sui W, Yu X and Lv Z: HIF1A-AS2 promotes the proliferation and metastasis of gastric cancer cells through miR-429/PD-L1 axis. Dig Dis Sci Feb 8, 2021 (Epub ahead of print). doi: 10.1007/s10620-020-06819-w.

Related Articles

Journal Cover

October-2021
Volume 15 Issue 4

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu Y, Zhang Y, Chen C and Li Y: lncRNA HIF1A‑AS2: A potential oncogene in human cancers (Review). Biomed Rep 15: 85, 2021
APA
Liu, Y., Zhang, Y., Chen, C., & Li, Y. (2021). lncRNA HIF1A‑AS2: A potential oncogene in human cancers (Review). Biomedical Reports, 15, 85. https://doi.org/10.3892/br.2021.1461
MLA
Liu, Y., Zhang, Y., Chen, C., Li, Y."lncRNA HIF1A‑AS2: A potential oncogene in human cancers (Review)". Biomedical Reports 15.4 (2021): 85.
Chicago
Liu, Y., Zhang, Y., Chen, C., Li, Y."lncRNA HIF1A‑AS2: A potential oncogene in human cancers (Review)". Biomedical Reports 15, no. 4 (2021): 85. https://doi.org/10.3892/br.2021.1461