Abstract
Long noncoding RNAs (lncRNAs) are pervasively transcribed from the mammalian genome as transcripts that are usually >200 nucleotides long. LncRNAs generally do not encode proteins but are involved in a variety of physiological processes, principally as epigenetic regulators. HOX transcript antisense intergenic RNA (HOTAIR) is a well-characterized lncRNA that has been implicated in several cancers and in various other diseases. HOTAIR is a repressor lncRNA and regulates various repressive chromatin modifications. However, recent studies have revealed additional functions of HOTAIR in regulation of protein degradation, microRNA (miRNA) sponging, NF-κB activation, inflammation, immune signaling, and DNA damage response. Herein, we have summarized the diverse functions and modes of action of HOTAIR in protein degradation, inflammation, DNA repair, and diseases, beyond its established functions in gene silencing.
Keywords: lncRNA, HOTAIR, Inflammation, DNA repair, gene regulation, chromatin modification
Introduction
The human genome is huge with 3 billion base pairs, and the vast majority of this DNA does not code for protein but rather for transcripts that are mostly non-translated, called non-coding RNA (ncRNAs).1 Long ncRNAs (lncRNAs) are pervasively transcribed from the genome as transcription products usually >200 nucleotides long and, like protein-coding transcripts, lncRNAs may be capped and polyadenylated.2 To date, approximately 60,000 lncRNAs have been identified, many of which affect the transcription of various proteins and non-coding genes, such as microRNAs (miRNAs).3 LncRNAs play post-transcriptional roles in the cell to regulate dosage compensation, imprinting, and developmental gene expression.4 LncRNAs modulate various aspects of the epigenetic machinery, and their dysregulation impacts several aspects of cellular homeostasis, including proliferation, mobility, chromatin remodeling, gene expression, post-synthetic processing, and differentiation.5
HOX transcript antisense intergenic RNA (HOTAIR) is a noncoding transcript encoded from chromosome 12 within the HOXC gene cluster and acts in trans to silence the HOXD locus of chromosome 2, as well as genes on other chromosomes.6–7 HOTAIR is transcribed from the antisense strand by RNA polymerase II and is 2.2 kb long, spliced, and polyadenylated.8 It is overexpressed in a wide variety of cancers and is associated with the promotion of oncogenesis and metastasis,9 and is one of the most well-characterized lncRNAs implicated in gene silencing. HOTAIR interacts with chromatin-modifying enzymes such as polycomb repressive complex 2 (PRC2) and lysine specific demethylase 1 (LSD1), multi-protein complexes that are major players in gene silencing.10 HOTAIR recruits these chromatin-modifying enzymes to gene promoters and suppresses the expression of tumor suppressors, thereby facilitating tumor growth. However, several recent studies revealed that HOTAIR may have additional roles beyond its well-known functions in gene repression, including modulatory roles in certain neurological,11 orthopedic,12 and metabolic13 conditions through several mechanisms, including protein degradation,14–15 NF-κB activation,16–17 microRNA sponging,18–19 and immune signaling.20–21 This review will focus on the functions of HOTAIR beyond repression and will describe its diverse roles in protein degradation, inflammation, DNA damage response, cell signaling, mechanisms of action, and roles in various diseases.
HOTAIR overexpression in cancer
HOTAIR is an oncogenic lncRNA that is overexpressed in a variety of solid tumors, including breast cancer,22 esophageal cancer,23 hepatocellular carcinoma,19 lung cancer,24 colorectal cancer,25 and others.26–28 HOTAIR overexpression is positively correlated with tumorigenesis, angiogenesis, cancer stem cell differentiation, progression, invasion, drug resistance, metastasis, and poor prognosis in a wide array of human malignancies via various signaling pathways.28–32 For example, in lung cancers, an increased HOTAIR level is associated with cell motility and invasion, and contributes to chemoresistance in small-cell lung cancer and lung adenocarcinoma, at least partly by regulating the expression of the tumor-suppressor p21.24, 33–34 HOTAIR overexpression is associated with distant metastasis and poor prognosis in colon cancer25 and with malignant proliferation, invasion, and metastasis in breast cancer.35 HOTAIR was found to be highly expressed in lymph node metastases from primary melanoma, and its knockdown reduced cell motility and invasion in human melanoma.36 By shortening the cell cycle and promoting malignant cell proliferation, HOTAIR has been found to play a role in the development and progression of adrenocortical carcinoma.37 Independent of other prognostic variables, HOTAIR levels in glioma patients are associated with higher grade and significantly shortened survival.38 HOTAIR deregulation has been identified in various gynecologic cancers, including endometrial and cervical cancers.39 It has been shown to act as an oncogene in gastric cancer, possibly by de-repressing HER2, and a functional single nucleotide polymorphism (SNP) in the HOTAIR gene may affect the risk of this malignancy.40–41 HOTAIR is significantly upregulated in cancerous gastric tissues and there is evidence for an inverse relationship between HOTAIR and poly r(C)-binding protein 1 (PCBP-1), which is an inhibitor of gastric cancer pathogenesis.42 Elevated HOTAIR levels in esophageal squamous cell carcinoma (ESCC) tissues are positively associated with ESCC metastases, higher stage, and significantly reduced overall survival in human patients, and its knockdown diminishes tumor cell invasiveness and migration potential.43 Specific HOTAIR SNPs have also been strongly associated with susceptibility to pancreatic cancer and with malignant cell proliferation.44–45
HOTAIR in non-malignant disorders
Besides its well-known associations with a multitude of cancers, HOTAIR overexpression has been associated with various neurological,11 musculoskeletal,46 and metabolic13 disorders. Increased activity of leucine-rich repeat kinase 2 (LRRK2) is strongly implicated in the pathogenesis of Parkinson’s disease (PD); in a mouse model of PD, HOTAIR upregulated this enzyme and specifically increased the stability of its mRNA transcript.47 PD mice-model-based studies demonstrated that disease progression could be significantly suppressed by inhibition of HOTAIR, which binds miR-126–5p to indirectly upregulate its target RAB3IP, a guanine nucleotide exchange factor that may suppress neuronal autophagy, and to promote apoptosis in PD cells.48 Deregulation of the p35/CDK5 (cyclin-dependent kinase 5) complex is closely associated with the onset and progression of Alzheimer’s disease, and HOTAIR negatively regulates mRNA levels of the gene that encodes for p35, cyclin-dependent kinase 5 regulatory subunit 1 (CDK5R1).49 HOTAIR was found to be upregulated in vitamin D-deficient patients with multiple sclerosis, suggesting a possible role in the pathogenesis of this disease specifically through a vitamin D-mediated immune process.50
HOTAIR appears to play roles in the most common types of arthritis. Its overexpression was identified in the blood mononuclear cells and serum exosomes of patients with rheumatoid arthritis, possibly indicating its involvement in delivering inflammatory components to nearby cells or through the circulatory system, and the same study showed that HOTAIR contributed to rheumatoid arthritis pathogenesis by upregulating matrix metalloproteinase (MMP)-2 and MMP-13 in osteoclasts and synoviocytes, promoting joint destruction.51 The pathogenesis of osteoarthritis is associated with HOTAIR overexpression, possibly through a HOTAIR/miR-17–5p/FUT2/β-catenin axis,46 with high HOTAIR levels being shown to induce apoptosis and to diminish chondrocyte viability.52
HOTAIR is also involved in disorders of energy metabolism. It is significantly upregulated in patients with type II diabetes13 and was found to participate in hepatic insulin resistance by inhibiting SIRT1, a protein deacetylase necessary for maintaining metabolic homeostasis in hepatocytes.53 Likely via promoter methylation, HOTAIR suppresses SIRT1 expression in liver cells exposed to hepatitis C virus core protein, contributing to dysregulation of several genes related to glucose and lipid metabolism.54 Interestingly, a study on nasopharyngeal carcinoma showed that knockdown of HOTAIR led to downregulation of fatty acid synthase and inhibition of de novo synthesis of cellular free fatty acids in cancer cells.55 A non-comprehensive list of diseases with demonstrated HOTAIR activity and some of its functions and modes of action is presented in Table 1.
Table 1.
Disease type | Factors linked to HOTAIR overexpression | Proposed mechanism of action | REFs |
---|---|---|---|
Lung cancer | Invasion, metastasis, 5FU chemoresistance, poor prognosis | Epigenetic downregulation of tumor suppressors; promotion of EMT via derepression of Snail by sponging miR-34a-5p; promotion of cell cycle progression |
18–20, 45,46 |
Colorectal cancer | Distant metastasis, poor prognosis, 5FU resistance | Suppression of miR-34a, miR-214, and miR-218; EMT induction | 21,47–49 |
Breast cancer | Proliferation, invasion, metastasis, resistance to radiotherapy | Sponging miR-218; EMT promotion |
22, 50, 51 |
Liver cancer | Tumorigenesis, proliferation, metastasis | Suppression of miR-217; upregulation of CCL2; EMT promotion via sponging miR-23b-3p; repression of miR-1 |
52–57 |
Esophageal cancer | Metastasis, progression, poor prognosis | Derepression of CCND1 by sponging miR-1 | 30, 58 |
Nasopharyngeal carcinoma | Oncogenesis, angiogenesis, tumor progression, poor prognosis | Activation of VEGFA |
44, 59, 60 |
Glioma | Higher tumor grade, progression, shortened survival | Sponging of miR-141 and miR-148b-3p |
25, 61, 62 |
Head and neck squamous cell carcinoma | Proliferation, invasion, migration | Derepression of PI3K/AKT pathway by sponging miR-206 | 63 |
Renal cell carcinoma | Tumorigenesis, progression, poor prognosis | HIF-1α/AXL signaling by sponging miR-217; upregulation of IGFBP2 | 64, 65 |
Melanoma | Lymph node metastasis, invasion | Unknown | 23 |
Gastric carcinoma | Pathogenesis, larger tumor size, extensive metastasis, poor prognosis | Derepression of HER2 by sponging miR-331–3p; activation of EMT via Snail derepression by sponging of miR-34a and epigenetic suppression of E-cadherin |
27–29, 66, 67 |
Ovarian cancer | Proliferation, invasion, tumor size, metastasis, platinum chemoresistance | Inhibition of cell cycle arrest via upregulation of CCND1 and 2 by miR-206 sponging; miR-373 sponging; NF-κB pathway activation | 68 |
Cervical cancer | Advanced stage, lymph node invasion, metastasis, tumor progression, poor prognosis | Repression of tumor-suppressor miR-206; derepression of HLA-G by sponging miR-148a |
26, 69, 70 |
Pancreatic cancer | Increased risk, proliferation, poor prognosis | Inhibition of cell cycle arrest via sponging miR-613 |
31, 32, 71 |
Prostate cancer | Malignant cell growth and invasion | Epigenetic AR suppression in non-CRPC; AR stabilization via blocking ubiquitination in CRPC; repression of tumor suppressor miR-193a | 72–74 |
Adrenocortical carcinoma | Tumorigenesis, progression | Promotion of cell cycle progression by upregulation of CCND1 | 24 |
Chromic myeloid leukemia | Acquired multidrug resistance | Activation of PI3K/AKT signaling | 75, 76 |
Acute myeloid leukemia | Poor prognosis | Upregulation of proto-oncogene c-KIT via sponging miR-193a | 77 |
Chondrosarcoma | Higher tumor grade, poor prognosis | Silencing of anti-oncogene miR-454–3p | 78 |
Type 2 diabetes | Hepatic insulin resistance | Inhibition of SIRT1 |
35, 42, 43 |
Parkinson’s disease | Pathogenesis | Upregulation of LRRK2, sponging of MiR-126–5p | 36, 37 |
Alzheimer’s disease | Onset and progression | Downregulation of p35 (activator of CDK5) |
38 |
Multiple sclerosis with vitamin D deficiency | Pathogenesis | Unknown | 39 |
Rheumatoid arthritis | Pathogenesis | Upregulation of MMP-2 and −13 | 40 |
Osteoarthritis | Increased apoptosis and reduced chondrocyte viability | Sponging miR-130a-3p | 41 |
Sepsis | Activation of NF-κB pathway and upregulation of inflammatory cytokines | Recruitment of coactivators and transcription factors to NF-κB binding sites on IL-6 and iNOS promoters; induced expression of IL-6R by sponging miR-211 |
70, 79, 80 |
Abbreviations: AR, androgen receptor; CCL2, chemokine (C-C motif) ligand 2; CCND1 & 2, cyclin D1 and D2; CDK5, cyclin-dependent kinase 5; CRPC, castration-resistant prostate cancer; EMT, epithelial-to-mesenchymal transition; HER2, human epidermal growth factor receptor 2; HLA-G, human leukocyte antigen-G; IGFBP2, insulin growth factor-binding protein 2; IL-6R, interleukin-6R; iNOS, inducible nitric oxide synthase; LRRK2, leucine-rich repeat kinase 2; MMP, matrix metalloproteinase; NF-κB, nuclear factor κB; SIRT1, sirtuin 1; VEGFA, vascular endothelial growth factor A.
HOTAIR in chromatin modification and transcriptional control
HOTAIR is classically known as repressor lncRNA.8 It can function as a scaffold by interacting via its 5’ end with EZH2, a subunit of polycomb repressive complex 2 (PRC2), an epigenetic silencing complex that binds to approximately 20% of all lncRNAs and exerts H3K27 tri-methylase activity via its catalytic EZH2 subunit to act as a transcription repressor.8 In addition, HOTAIR interacts with lysine-specific demethylase 1A (LSD1), a histone modifier that suppresses transcription via H3K4 demethylation via its 3’ end (Figure 1).4 HOTAIR and PRC2 were shown to be functionally interdependent in cancer invasiveness, and the genome-wide silencing of metastasis-suppressor genes via HOTAIR’s targeted recruitment of PRC2 to those genes’ promoters has been demonstrated.9 Portoso et al. reported that HOTAIR overexpression was able to repress a limited number of target genes independently of PRC2, suggesting that there are multiple mechanisms through which HOTAIR induces transcriptional repression of other genes.56
HOTAIR has also been demonstrated to exert transcriptional control via mechanisms that do not involve the PRC2/LSD1 complex. For example, in a study on liver cancer stem cells, HOTAIR blocked the recruitment of CREB, P300, and RNA polymerase II to the promoter region of SETD2, a histone methyltransferase specific for H3K36me3, which is associated with transcription activation.57 In contrast, HOTAIR promoted angiogenesis in nasopharyngeal carcinoma by directly activating the transcription of angiogenic factor VEGFA via binding to HOTAIR-responsive elements in the VEGFA promoter.58
Regulation of miRNA by HOTAIR: epigenetic silencing and competitive binding
In addition to epigenetically regulating various protein-coding genes via epigenomic modulations, HOTAIR silences many microRNA (miRNAs). MiRNAs are highly conserved small non-coding RNAs of 17–25 nt in length that play a major regulatory role in cells by binding directly to the 3’-UTR of target messenger RNAs (mRNAs) and causing mRNA cleavage and/or translational repression.59 Some miRNAs are considered proto-oncogenes while others act as tumor suppressors by inhibiting the translation of oncogenic proteins. Indeed, perturbations in the expression levels of specific miRNAs have been found to be associated with the vast majority of human malignancies.60–61 Over half of the 140 lncRNAs analyzed in one study affected their targets in a miRNA-dependent manner.62 HOTAIR is no exception, silencing its target miRNAs in various ways. For example, in gastric cancer, miR-34a modulates targets that accelerate metastasis by contributing to epithelial-to-mesenchymal transition (EMT); HOTAIR contributes to EMT in this system by recruiting PRC2 to the miR-34a promoter, where the EZH2 subunit directly binds and mediates repression via H3K27 trimethylation.63 In pancreatic cancer, HOTAIR silences miR-663b by disrupting the balance between H3K27me3 and H3K4me3 at its promoter; this miRNA targets insulin-like growth factor 2, the overexpression of which is closely associated with worse prognosis.45 Studies on chondrosarcoma cell lines showed that the HOTAIR/EZH2 complex epigenetically silenced miR-454–3p via CpG-island methylation; this miRNA acts as an anti-oncogene by targeting STAT3 and ATG12 (involved in cell growth and autophagosome formation, respectively).64
Besides being transcriptionally repressed via histone modification, miRNAs can be sponged by HOTAIR acting as a competitive endogenous RNA (ceRNA), indirectly causing the de-repression of miRNA targets.65 An example of this is shown in Figure 2. Several studies have demonstrated that HOTAIR acts as ceRNA to upregulate specific tumorigenic genes. For example, HOTAIR acts as a sink for miR-331–3p in gastric cancer cells, leading to de-repression of its direct target, human epithelial growth factor receptor (HER2), which oncogenically activates cellular networks involved in proliferation, motility, and angiogenesis.40 By competitively binding miR-193a, HOTAIR modulates the expression of the proto-oncogene c-KIT in acute myeloid leukemia cells,66 and in ovarian cancer, it acts as a sink for miR-373 to de-repress Rab22a, a small GTPase that modulates cancer cell proliferation.67 Overexpression of the transcription factor ZEB1 is involved in malignant transformation and is targeted by miR-23b-3p for suppression; HOTAIR sponges miR-23b-3p in hepatocellular carcinoma (HCC) cells, thus promoting invasion and migration.19, 68 MiR-218 is sponged by HOTAIR in breast cancer, inhibiting cell apoptosis and DNA damage from radiotherapy.69 SKA2, overexpressed in glioma tissues, is a direct target of miR-141 but is de-repressed by HOTAIR sponging of this miRNA.70 Also in glioma, an in vitro model showed that HOTAIR knockdown increased the permeability of the blood-tumor barrier (BTB) by binding to miR-148b-3p and thereby reducing the expression of several proteins related to tight junctions.71 HOTAIR competitively binds to miR-206, which directly targets STC2 to inhibit activation of the PI3K/AKT signaling pathway in head and neck squamous cell carcinoma (HNSCC), and knockdown of HOTAIR reduced tumor growth in an HNSCC mouse model.72
Renal cell carcinoma tumorigenesis was promoted by HOTAIR functioning as a ceRNA for miR-217, thus de-repressing HIF-1α,73 and researchers have identified an aberrantly activated autoregulatory feedback loop involving HOTAIR’s interference with tumor-suppressing miR-193a in a prostate cancer study.74 Knockdown of HOTAIR in a HeLa cervical cancer cell line enhanced the expression of the tumor suppressor miR-206, which targets MKL1, a transcription factor involved in inducing cancer cell migration and invasion.75 Antagonistic expression of miR-217 and HOTAIR in HCC has been observed, suggesting that overexpression of HOTAIR leads to tumorigenesis and proliferation via downregulation of miR-217,76 a miRNA that directly targets E2F3 protein, which is found at high levels in HCC metastatic tissues.77 HOTAIR’s ability to promote malignant progression in colon cancer may be partly due to interactions with miR-34a25 and miR-214.78 The latter mechanism may involve a direct target of miR-214 known as ST6GAL1, which restructures sialylated glycoproteins on the surface of colorectal cancer cells to enhance malignancy.78 It has been suggested that HOTAIR may be capable of sequestering several miRNAs at once, and since a single miRNA can control multiple genes, this may help explain why HOTAIR has multiple properties in malignances.40
It should be noted that HOTAIR/miRNA interactions are not limited to malignant processes, and the effects of HOTAIR overexpression are not always deleterious (see Table 2). HOTAIR protectively influences the proliferative and apoptotic properties of vascular smooth muscle cells by targeting the nuclear transcription factor PPARα via sponging miR-130b-3p.79 In intervertebral disc degeneration, HOTAIR expression is decreased; this is undesirable because HOTAIR is able to suppress the TNF-α-induced apoptosis of nucleus pulposus cells by modulating the miR-34a/Bcl-2 axis and sponging miR-34a-5p with the result of downregulating genes involved in apoptosis via the Notch signaling pathway.80–81 HOTAIR also has a protective function in acute myocardial infarction (AMI) partly via negative modulation of miR-1, which is markedly increased post-AMI.82 HOTAIR overexpression also protected against cell injury due to oxidative stress in a cardiomyocyte cell line (H9C2) by sponging miR-125, de-repressing its protein target MMP2, a matrix metalloproteinase important in extracellular matrix homeostasis.83 Interestingly, HOTAIR can also modulate miRNA levels at the transcriptional level. Cheng et al. showed that HOTAIR transcriptionally inhibits miR-122 via CpG island methylation, mediated by DNMTs.84
Table 2.
Condition or cell type | Role of HOTAIR | Proposed mechanism of action | REFs |
---|---|---|---|
Vascular smooth muscle cells | Protection of apoptotic and proliferative properties | Upregulation of PPARα via sponging miR-130b-3p | 90 |
Intervertebral disc degeneration | Prevents apoptosis of nucleus pulposus cells | Downregulation of apoptosis-related genes (Notch signaling pathway) via sponging miR-34a-5p |
91, 92 |
Acute myocardial infarction | Protective factor for cardiomyocytes and biomarker for human AMI | Suppression of miR-1 | 93 |
Oxidatively stressed cardiomyocytes | Protection against cell injury | Sponging miR-125 | 94 |
Rheumatoid arthritis | Anti-inflammatory effect on LPS-treated chondrocytes | Inhibition of NF-κB | 104 |
HOTAIR modulation of the NF-κB inflammatory pathway and immune signaling
The pro-inflammatory transcription factor NF-κB is a master regulator of hundreds of genes involved in the proliferation and survival of cells, and its prompt activation and termination are crucial for appropriate inflammatory response. Dysregulation of NF-κB is found in many diseases, including almost all malignancies, where constitutively activated NF-κB promotes tumor cell proliferation, angiogenesis, and EMT while suppressing apoptosis.85–86 Recent studies from our laboratory demonstrate that HOTAIR promotes NF-κB activation by downregulating its inhibitor, IκBα, while HOTAIR itself appears to be transcriptionally regulated by NF-κB in response to lipopolysaccharide (LPS) treatment in macrophages.87 SiRNA-mediated knockdown of HOTAIR suppressed the LPS-induced activation of cytokines and pro-inflammatory genes including IL-6 and iNOS, and this was mediated via downregulation of NF-κB activation.87 Though the exact mechanism is not clear, we predict that HOTAIR functionally modulates the ubiquitin ligases and/or kinases for the NF-κB inhibitor, IκBα, and thus controls IκBα degradation and hence NF-κB activation (Figure 3). Additionally, metabolic reprogramming plays a central role in macrophage activation, inflammation, and immune response. In a recent study, our laboratory demonstrated that HOTAIR regulates expression of glucose transporter Glut1 and metabolism in macrophages during inflammation.88 LPS induces NF-κB binding and enrichment at the Glut1 promoter but this was reduced upon HOTAIR knockdown, which also caused a significant reduction in the otherwise 10-fold increase in glucose uptake under LPS stimulation.88 We also found that HOTAIR suppresses gene expression of PTEN (a tumor suppressor that blocks PI3K/AKT activation) during LPS-induced inflammation in macrophages.88 These findings suggest a critical regulatory role for HOTAIR in in glucose metabolism via modulation of Glut1 expression and glucose uptake during inflammation. Importantly, in addition to HOTAIR, we have discovered a series of long-noncoding RNAs (we termed them as LinfRNAs – long noncoding inflammation-associated RNAs) that are associated with inflammation and immune response in higher eukaryotes, including humans (manuscripts in preparation). An independent study also demonstrated that HOTAIR regulates NF-κB activation and cytokine expression during DNA damage such as that induced by platinum chemotherapy, creating a positive feedback loop cascade (Figure 3) that sustains an activated DNA damage response, thus reducing chemotherapeutic efficacy and contributing to cellular senescence.89 HOTAIR was upregulated via activation of NF-κB in gluteofemoral fat in a mouse model of obesity and sedentary lifestyle, pointing to a possible mechanism by which those co-morbidities are correlated to risk of colorectal cancer.90
In LPS-induced sepsis, HOTAIR was found to regulate TNF-α production via activation of the NF-κB pathway in cardiomyocytes.91 In UVB-treated keratinocytes, overexpressed HOTAIR promotes the expression of PKR (double-stranded RNA-dependent protein kinase), which is involved in activating the NF-κB pathway.92 HOTAIR appears to be linked to the significant upregulation of the inflammatory cytokines IFN-γ, IL-6, IL-17, TNF-α, IL-1β, and IL-6R in monocytes via sponging of miR-211.93 HOTAIR was found to promote colorectal cancer cell viability and 5FU chemotherapy resistance by epigenetically silencing miR-218, which targets VOPP1, an activator of NF-κB signaling.94 In a rat model of acute myocardial infarction, HOTAIR upregulated the receptor of advanced glycation end-products (RAGE) in cardiomyocytes, activating NF-κB signaling.95 In contrast, in certain cells or conditions, HOTAIR can inactivate the NF-κB pathway and act in an anti-inflammatory manner. For example, it was found to significantly inhibit NF-κB expression in LPS-treated chondrocytes, leading to decreased IL-1β and TNF-α and thereby conferring a protective effect in rheumatoid arthritis.96
Recent studies have also revealed novel roles of HOTAIR in immune signaling, both as a target and as a regulator of various immunity factors. HCC cells with HOTAIR overexpression secrete higher levels of chemokine (C-C motif) ligand 2 (CCL2), a humoral immunity factor that is required for recruitment of tumor-associated macrophages and accumulation of myeloid-derived suppressor cells, both of which are critical components of the tumor microenvironment and promote tumor growth and metastasis.97 The cause of this CCL2 upregulation may be due to upregulation of its transcriptional factor SP1; it has been shown that miR-326 targets SP1, and that HOTAIR suppresses miR-326.98 Human leukocyte antigen-G (HLA-G) is expressed by tumor cells to help them evade the host’s immune surveillance; this antigen is targeted for downregulation by miR-148a, which is downregulated by HOTAIR via competitive binding in cervical cancer cells.99 A study using cell cultures with overexpressed HOTAIR in rheumatoid arthritis exosomes showed that HOTAIR may attract active macrophages.51 HOTAIR may also indirectly promote the progression of sepsis by sponging miR-211, which induces the expression of IL-6R and contributes to initiation of the host immune response.93 HOTAIR itself may also be a target of certain inflammatory immune cells. Data from a study on prostate cancer showed that infiltrating mast cells suppress androgen receptor (AR) expression by upregulating HOTAIR, which then promotes binding of the PRC2 complex to the AR promoter.100 In addition, HOTAIR is induced in primary macrophages upon stimulation with lipopolysaccharide (LPS) and it is required for the LPS-induced proteasomal degradation of IκBα and subsequent NF-κB activation, causing the downregulation of IL-6 and iNOS.87
HOTAIR in DNA repair
Systematic in vivo and in vitro studies from various laboratories, including ours, have established that HOTAIR is an oncogenic lncRNA, playing diverse roles in promoting carcinogenesis.101–102 In addition to the conventional role of HOTAIR in altering the epigenomic landscape, recent studies have reported that HOTAIR expression significantly modulates the DNA damage response (DDR), which in turn aids in oncogenesis, malignant transformation, and chemotherapeutic resistance.17,103–104 However, the underlying molecular mechanisms through which HOTAIR modulates the DDR remain elusive. HOTAIR has been shown to induce apoptosis and inflammation in UVB-exposed keratinocytes.92 DNA damage was shown to induce the expression of HOTAIR in a p53-dependent manner, which indicates that HOTAIR plays a role in p53-regulated DDR.105 HOTAIR was also shown to modulate the DNA damage response pathway through suppression of p21 and p5310. Recently, Özeş et. al. not only observed overexpression of HOTAIR in recurrent platinum-resistant ovarian tumors compared to primary ovarian tumors but also demonstrated that HOTAIR activates the NF-κB signaling cascade during platinum-induced DDR, and the DDR-mediated activation of NF-κB induces HOTAIR.89 It thus establishes a positive feedback loop with sustained NF-κB activation and persistent DNA-damage signaling, resulting in cellular senescence and chemotherapy resistance in ovarian and other cancers (see Figure 4).89 In vitro RNAi-based HOTAIR depletion studies using breast cancer lines that highly overexpress HOTAIR showed that HOTAIR-depleted breast adenocarcinoma cells became significantly sensitized to ionizing radiation, leading to augmented DNA damage through avoiding sponging of miR-218.69 This suggests that a HOTAIR-miR-218 axis plays a role in preventing DNA damage, but the mechanism through which this axis might regulate the DDR remains elusive. A recent report showed that HOTAIR promotes malignant growth of human liver cancer stem cells via inhibiting the recruitment of CREB, P300, and RNA polymerase II onto the SETD2 promoter region, in turn inhibiting SETD2 expression and subsequently its phosphorylation (Figure 4).57 SETD2 binding capacity to its cognate substrate histone H3 is reduced, leading to consequent reduction of trimethyl histone H3 lysine-36 marks, which prevents mismatch DNA repair.57
Dysregulated HOTAIR expression has thus been demonstrated to unequivocally alter the epigenomic landscape and to affect gene expression and contribute to tumorigenesis. However, the mere modulation of HOTAIR expression can also modulate signaling pathways that play a critical role in regulating the DDR, enabling a signaling cascade that aids in the malignant transformation of various tumors. Therefore, knockdown of HOTAIR in combination with blocking DNA-damaging agents might represent a promising treatment strategy for targeting tumors that show HOTAIR overexpression.
It is interesting to note that HOTAIR is not alone among the lncRNAs as master regulators of cellular DNA damage and repair processes (see Table 3).103 This role is reflected in the names given to some of these lncRNAs, such as NORAD (noncoding RNA activated by DNA damage).106 Some are also classified as damage-induced lncRNAs (dilncRNAs) and for example may be synthesized by RNA polymerase II (RNAPII) after binding to the MRE11–RAD50–NBS1 complex and being recruited to double-strand breaks (DSBs).107 LncRNAs employ an array of mechanisms to mediate DSB repair.108 DilncRNAs are transcribed from broken DNA ends and help to recruit the homologous recombination (HR) proteins BRCA1, BRCA2, and RAD51,109 and the lncRNA BGL3 regulates the HR pathway via recruitment by PARP1 to DSBs, where it enhances the accumulation of the BRCA1/BARD1 complex and controls DNA end resection.110 The recombinase RAD51 is crucial for the high-fidelity repair of DSBs, and its formation is increased by overexpression of the lncRNA TODRA (transcribed in the opposite direction of RAD51).111 LncRNA- JADE induces histone H4 acetylation in the DDR by transcriptionally activating Jade1, a key component in the HBO1 (human acetylase binding to ORC1) histone acetylation complex.112 CCND1-upstream intergenic DNA repair lncRNAs 1 and 2 (CUPID1 and CUPID2), which are induced by estrogen stimulation, modulate DDR by affecting the choice of DDR pathway.113 The lncRNA DDSR1 (DNA damage- sensitive RNA1) is induced by various DSB agents and interferes with DDR signaling.114 The transcription factor E2F1 upregulates ANRIL in response to DNA damage, and this lncRNA in turn inhibits certain CDK inhibitors at the late stage of the DDR, which promotes the cell’s ability to return to normal after necessary DNA repair has been accomplished.115
Table 3.
LncRNA | Role in DDR | REFS |
---|---|---|
ANRIL | CDK inhibition in late-stage DDR | 117 |
APTR | PRC2 recruitment for p21 repression | 125 |
BGL3 | Recruitment of PARP1 to DSBs; accumulation of BRCA1/BARD1 | 112 |
CUPID1 and CUPID2 | Influence choice of DDR pathway | 115 |
DilncRNA | Recruitment of HR proteins (BRCA1, BRCA2, and RAD51) |
109, 110 |
DDSR1 | Interference with DDR signaling | 116 |
DINO | Required for p53-dependent gene expression; triggers cell cycle arrest and amplifies damage signaling | 118 |
DNM3OS | Inhibition of DNA-repair proteins following irradiation in esophageal cancer cells | 132 |
GUARDIN | Sequestration of mRNA-23a to prevent end-to-end chromosomal fusion by maintaining TRF2 | 127 |
JADE | Activation of HBO1 to promote H4 acetylation | 114 |
lincRNA-p21 | Activation of tumor suppressor p21 | 121–123 |
Meg3 | Interaction with PTBP3 to interfere with p53 activation in epithelium | 131 |
NEAT1 | Promotion of replication-stress-induced ATR signaling | 129 |
NORAD | Sequestration of proteins (e.g., PUMILIO) that drive chromosomal instability | 108 |
PANDA | Pro-apoptotic via interaction with NF-YA |
124 |
PR-lncRNA-1 and PR-lncRNA-10 | Required for efficient binding of p53, transcriptional activation of p53 targets | 119 |
RP11–624C23.1, RP11–203E8, and RP11–446E9 | Increased apoptosis after DNA-damaging chemotherapy in childhood leukemia cells | 133 |
TERRA | Protection against damage to telomeres and other chromosomal sites |
134 |
TODRA | Enhanced synthesis of RAD51 for DSB repair | 113 |
TP53TG1 | Suppression of tumorigenic protein YBX1 |
128 |
WRAP53 | p53 induction | 130 |
Unsurprisingly, many of the lncRNAs that play roles in DDR mechanisms do so by either inducing or being induced by p53. For example, DINO (damage-induced noncoding) lncRNA is transcriptionally activated by p53 in the context of DNA damage and is necessary for p53-dependent gene expression, and its expression is sufficient to activate cell cycle arrest and damage signaling even without DNA damage being present.116 Sánchez et al. defined a set of 18 lncRNAs that are bona fide p53 transcriptional targets, two of which (PR-lncRNA-1 and PR-lncRNA-10) were observed to be required for p53’s efficient binding to, and transcriptional activation of, some of its other direct transcriptional targets.117 Another study found 39 lncRNAs associated with p53, and approximately half of these were specifically associated with the induction of DNA damage.118 Several lncRNAs that participate in, or interfere with, p53 pathways do so via links to p21, which is induced by p53 in response to DNA damage. For example, lincRNA-p21 has been found to affect global gene expression in a variety of ways by serving to activate p21.119 P53 transcriptionally activates lincRNA-p21 promoters in response to DNA damage, and lincRNA-p21 inhibition allows the expression of hundreds of genes that p53 represses under normal circumstances.120 DNA damage and overexpression of the tumor suppressor ING1b also induces lincRNA-p21.121 PANDA (p21-associated ncRNA DNA-damage-activated) interacts with NF-YA, a transcription factor involved in pro-apoptotic gene expression.122 The lncRNA APTR (Alu-mediated p21 transcriptional regulator) recruits PRC2 to repress the transcription of p21.123
LncRNAs have been shown to be induced by UVC-induced DDR, with the suggestion of involvement in regulation of p53 signaling pathways.124 The lncRNA GUARDIN responds to p53 by sequestering mRNA-23a to prevent end-to-end chromosomal fusion by maintaining TRF2 (telomeric repeat-binding factor 2) expression.125 Epigenetic loss of lncRNA TP53TG1 by methylation-mediated silencing enriches the nuclear content of RNA binding protein YBX1; this tumorigenic protein activates PI3K and its oncogenic target, AKT, a kinase that promotes the degradation of p53.126 NEAT1 helps to attenuate p53 activation as part of a negative feedback loop in which it promotes replication-stress-induced ATR signaling.127 WRAP53 is required for DNA-damage-related p53 induction,128 and p53 activation induces Meg3 (maternally expressed gene 3) in epithelial cells, where this lncRNAs interacts with RNA binding protein polypyrimidine tract binding protein 3, thus contributing to regulation of gene expression related to DDR.129
Various lncRNAs have demonstrated DDR roles in specific cancers. DNM3OS is a fibroblast-promoted lncRNA that confers radioresistance in esophageal squamous cell carcinoma by promoting the expression of DNA damage markers but also inhibiting the expression of several DNA-repair proteins following irradiation.130 Three lncRNAs (RP11–624C23.1, RP11–203E8, and RP11–446E9) were found to significantly increase apoptosis after exposure to a DNA-damaging chemotherapeutic agent (camptothecin) in childhood leukemia cells.131 A single-telomere, long noncoding member of a group of telomeric repeat-containing RNAs (TERRA) was shown to be involved in maintaining genomic integrity in a stomach adenocarcinoma cell line, as its depletion led to increased damage in telomeres and other chromosomal sites.132
HOTAIR promotion of epithelial-to-mesenchymal transition
HOTAIR has been shown to promote epithelial-to-mesenchymal transition (EMT), a process associated with tumor progression, invasion, and metastasis. EMT is regulated via many signaling pathways and transcription factors as well as by many miRNAs.133 Loss of the protein E-cadherin induces EMT, and E-cadherin was found to be lost in 37.1% of invasive ductal breast malignancies.134 In human lung cancer cells, HOTAIR sponges miR-34a-5p, leading to de-repression of the transcription factor Snail that in turn is a repressor of E-cadherin.135 Snail is also de-repressed by HOTAIR in gastric cancer via repression of miR-34a by recruiting PRC2 to its promoter region, to which the EZH2 subunit directly binds for mediation of H3K27me3.63 Snail was also demonstrated, in TGFβ-treated human hepatoma cells, to form an epithelial-gene-repressing complex with EZH2 by using HOTAIR as a bridge, and this complex promoted EMT.136 Carcinoma-associated fibroblasts transactivated HOTAIR in breast cancer cells via TGF-β1 paracrine secretion, promoting EMT and consequential metastatic activity.137 HOTAIR knockdown significantly reversed EMT in gastric cancer by increasing E-cadherin expression in a PRC2-dependent manner,138 and ablation of HOTAIR expression prevented the induction of EMT and the arising of cancer stem cells in colon and breast cancer cell lines, demonstrating a requirement of HOTAIR for EMT.139
HOTAIR involvement in ubiquitination and proteasomal degradation
In addition to its epigenomic, transcriptional, and translational modulation functions, various studies have also elucidated novel post-translational functions of cytoplasmic HOTAIR. Yoon et al. demonstrated that HOTAIR can induce ubiquitin-mediated proteolysis by acting as a scaffold transcript that enhances E3-mediated ubiquitination of substrate proteins and targets them for proteasomal degradation.140 In brief, HOTAIR interacts with E3 ubiquitin ligases Dzip3 and Mex3b to facilitate the degradation of their respective substrates, Ataxin-1 and Snurportin-1 (Figure 5). Therefore, in the context of senescent cells, human antigen R (HuR) associates with HOTAIR and lowers its stability by recruiting the let7-Ago2 complex (during the normal cell cycle); however, in the cellular senescence state, when HuR levels are alleviated, let7-Ago2-complex-mediated degradation of HOTAIR decreases and HOTAIR is stabilized, leading to rapid degradation of Ataxin-1 and Snurportin-1, thus preventing premature senescence.140 This HOTAIR-mediated mechanism is also oncogenic because Ataxin-1 activates the promoter of E-cadherin, which is a key tumor suppressor.141
HOTAIR acts as a ubiquitination scaffold between PLK1 and its substrates, such as the transcription repression factors SUZ12 and ZNF198, in hepatitis B-virus-induced liver carcinogenesis. Mex3b has been identified as the E3 ligase that binds to HOTAIR before ubiquitinating SUZ12 for degradation.142 HOTAIR was recently shown to also interact with RUNX3, inducing its proteasome-mediated degradation via ubiquitination dependent on MEX3B (an E3 ubiquitin ligase), thus providing a novel regulatory mechanism responsible for silencing the expression of RUNX3 in gastric cancer.143 This HOTAIR–RUNX3 interaction might work as a new therapeutic target for metastatic gastric cancer. Other studies have determined that HOTAIR can also block ubiquitination. For example, Zhang et al. demonstrated that HOTAIR promoted castration-resistant prostate cancer by binding to the androgen receptor (AR) protein, stabilizing AR and blocking its degradation by preventing it from interacting with E3 ligase MDM2 (Figure 5).144
HOTAIR inhibition of cell cycle arrest
One of the myriad ways through which HOTAIR overexpression promotes malignancy is by inhibiting cell cycle arrest of tumor cells. For example, two major positive regulators of the cell cycle that are associated with metastasis, cyclin D1 and D2 (CCND1 and CCND2), are targeted for downregulation by miR-206, which is negatively modulated by HOTAIR in ovarian cancer.67 CCND1 is also de-repressed by HOTAIR binding to miR-1 in esophageal squamous cell carcinoma.145 HOTAIR overexpression in adrenocortical carcinoma cells causes upregulation of cyclin D1 and other genes related to the cell cycle, with the effect of promoting cell cycle progression and cell proliferation.37 In lung cancer, HOTAIR was found to regulate the Rb-E2F pathway, promoting cell cycle passage through the G1-S phase restriction point.146 In yet another example of miRNA sponging, HOTAIR suppresses miR-613 in pancreatic cancer cells, contributing to malignant growth since miR-613 normally induces cell cycle arrest at the G0/G1 phase.147
Regulation of HOTAIR
As an oncogenic lncRNA, transcription regulation of HOTAIR is critical for its cellular functions and disease implications. Indeed, multiple studies, including ours, demonstrate that HOTAIR expression is influenced in a variety of ways; for example, studies from our laboratory demonstrated that HOTAIR expression is elevated in breast cancer cells and that estradiol activates HOTAIR transcription in estrogen-receptor (ER)-positive breast cancer cells in an estradiol-dependent manner.148 ERs and ER co-regulators such as those belonging to the mixed lineage leukemia (MLL) family of histone methyltransferases play critical roles in regulation of HOTAIR expression in an estradiol environment. Additionally, studies from our laboratory also demonstrate that the endocrine disruptors bisphenol-A and diethylstilbestrol regulate HOTAIR expression by inducing estrogen-response elements (EREs) in its promoter; these EREs are bound by ERs and ER co-regulators including MLL-histone methyltransferases and CBP/p300 histone acetylases.149 Since HOTAIR is an oncogenic lncRNA, its upregulation by estrogen-mimicking endocrine-disrupting chemicals is indicative of the potential risk of such EDCs in cancer and other endocrine regulated diseases. Additionally, HOTAIR expression is elevated in hypoxia,150 which is a major driving force for tumor cell proliferation151 and angiogenesis.152 Our recent studies demonstrate that MLL1 histone methylase, which is a crucial player in hypoxia signaling and angiogenesis, coordinates with hypoxia inducible factor, HIF-1α, to regulate hypoxia-induced HOTAIR expression.150 These observations further suggest a pro-oncogenic functionality of HOTAIR and its potential interplay with other tumorigenic factors, such as HIF and MLL-histone methylases. Cancer-associated fibroblasts secrete the cytokine TGF-β1, which activates SMADs in a paracrine manner; the activated SMADs then directly bind to HOTAIR’s promoter site to induce its transcription.137 Other transcription factors that directly bind the HOTAIR promoter region include c-Myc in gallbladder cancer,153 FOXC1 in HCC,154 and HOXA9 in glioma.155 HOTAIR is also upregulated by overexpression of the hepatitis C virus core protein.54
In addition to the transcriptional induction of HOTAIR via epigenetic alteration of its promoter, HOTAIR expression is post-transcriptionally altered by various miRNAs. Genistein, a soy isoflavone studied for its cancer-inhibition effects in prostate cancer cells, was shown to upregulate miR-34a and miR-141, both of which directly target HOTAIR for repression.156–157 Interestingly, there is evidence that HOTAIR may also be mechanoresponsive. For example, its levels are decreased in certain heart valvular cells exposed to cyclic stretch, with data suggesting that the repression occurs via β-catenin in a stretch-responsive signaling pathway.158
CONCLUSION
A sizeable body of literature presently exists on the multitude of roles played by HOTAIR in human physiology through varied modes of action. Beyond its well-known role as a regulator of chromatin modification and gene repression, HOTAIR exhibits much more dynamic roles in the regulation of various signaling pathways, microRNA sponging, NF-κB activation, inflammation, immune signaling, modulation of proteasomal degradation via regulation of ubiquitin ligase activity, and DNA damage response. Most reports have focused on its roles in malignancy, but some studies have also demonstrated its involvement in neurodegenerative disorders such as Alzheimer’s and Parkinson’s, inflammatory and immune diseases, arthritis, and diabetes. In general, HOTAIR appears to exhibit two major modes of action: a) Gene silencing via recruitment of chromatin-modifying enzymes (histone methylases and demethylases and DNA methylases) and epigenetic alterations; and b) modulation of inflammatory response via regulation of NF-kB pathway. We predict that there may be unifying mechanisms of action of HOTAIR in most inflammatory and metabolic diseases, where it is found to be up-regulated. In addition to gene repression, HOTAIR-mediated modulation of the NF-kB pathway-mediated inflammatory process might be the central and one of the common mechanisms in cancer, neurodegenerative diseases, diabetes, arthritis, and other inflammatory diseases.
Along with critical roles of HOTAIR in gene regulation and cell signaling, HOTAIR has been implicated as a diagnostic and therapeutic target. Similar to proteins, most lncRNAs are expressed in a cell-and-tissue-type-specific manner and in far greater amounts,159–160 and many cell-free lncRNAs that were originally only detected in tissues are detectable and stable in various bodily fluids.161 For these reasons, lncRNAs, including HOTAIR, show great promise as novel, non-invasive, and specific biomarkers for diagnosis and prognosis of cancer and other diseases. In NSCLC patients, HOTAIR is detectable in plasma, where its levels are not only elevated but also associated with histology subtype and tumor-node-metastasis (TNM) stage, with greater diagnostic accuracy than CEA levels alone and even greater accuracy when used in combination with that traditional serum biomarker.162 Clinical data show that serum HOTAIR levels also correlate to TNM stage in esophageal squamous cell carcinoma.163 Computational analysis revealed that plasma HOTAIR levels could diagnose gastric cancer with 88% sensitivity and 84% specificity, and higher levels were associated with higher tumor grade, advanced stage, and metastasis.164 Serum HOTAIR is significantly higher in patients with glioblastoma multiforme compared to healthy controls, with 86.1% sensitivity and 87.5% specificity, and is significantly correlated with high-grade brain tumors.165 Plasma HOTAIR has also been suggested as a viable biomarker for diagnosis of acute myocardial infarction.82 Beyond detection in the blood, lncRNAs can also be found in the urine; for example, HOTAIR is enriched in urinary exosomes in bladder cancer patients and correlates with disease progression.166 Remarkably, HOTAIR and other lncRNAs can even be detected in the saliva; for example, salivary levels of HOTAIR are significantly higher in pancreatic cancer patients (with sensitivity and specificity of 60–97%) compared to patients with benign pancreatic tumors and a healthy cohort, and are significantly decreased after curative pancreatectomy.167 Thus, HOTAIR indeed holds potential as a therapeutic target and biomarker for various human diseases, including cancer. Overall, HOTAIR is one of the most well-studied lncRNAs, and is a critical regulator of gene expression, epigenetics, and cell signaling. A vast number of functional studies on HOTAIR indeed suggest that lncRNAs are functional, not mere transcriptional noise. Although further research is necessary before a full understanding of HOTAIR’s functions, mechanisms, and regulation will be achieved, studies so far have highlighted potential uses for HOTAIR as a therapeutic target and biomarker for various human diseases, including cancer.
Research highlights.
HOTAIR is a well-characterized lncRNA that has been implicated in several cancers and other diseases. HOTAIR is a well-known as a repressor lncRNA and regulates various repressive chromatin modifications. However, recent studies have revealed additional functions of HOTAIR in regulation of protein degradation, microRNA (miRNA) sponging, NF-κB activation, inflammation, immune signaling, and DNA damage response. Herein, we have summarized diverse functions and modes of action of HOTAIR, with an emphasis on the following.
Roles in cancer
Roles in chromatin modification and gene silencing
Implications in protein degradation, inflammation and immune signaling.
Implications in DNA repair
Potential implications in diagnosis and therapeutics
Acknowledgements
We thank all the Mandal lab members for helpful discussions. Research in the Mandal laboratory is supported by grants from the National Institute of Health 1R15HL142032 and The Schwartzberg Companies.
Footnotes
Author statement.
Authors declares that there is no conflict of interest. The manuscript is not submitted elsewhere for publication. RP as review the literature and written the manuscript. AB has guided and helped writing the reviews. SSM has supervised the overall process, reviewed the content, and finalized the whole manuscript.
COI statement
There is no conflict of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
REFERENCES
- 1.Rinn JL; Chang HY, Genome regulation by long noncoding RNAs. Annu Rev Biochem 2012, 81, 145–166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Kung JTY; Colognori D; Lee JT, Long Noncoding RNAs: Past, Present, and Future. Genetics 2013, 193 (3), 651–669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Tan JY; Sirey T; Honti F; Graham B; Piovesan A; Merkenschlager M; Webber C; Ponting CP; Marques AC, Extensive microRNA-mediated crosstalk between lncRNAs and mRNAs in mouse embryonic stem cells. Genome Research 2015, 25 (5), 655–666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Tsai MC; Manor O; Wan Y; Mosammaparast N; Wang JK; Lan F; Shi Y; Segal E; Chang HY, Long Noncoding RNA as Modular Scaffold of Histone Modification Complexes. Science 2010, 329 (5992), 689–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Di Gesualdo F; Capaccioli S; Lulli M, A pathophysiological view of the long non-coding RNA world. Oncotarget 2014, 5 (22), 10976–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.He S; Liu S; Zhu H, The sequence, structure and evolutionary features of HOTAIR in mammals. BMC Evolutionary Biology 2011, 11, 102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Johnsson P; Lipovich L; Grandér D; Morris KV, Evolutionary conservation of long noncoding RNAs; sequence, structure, function. Biochimica et biophysica acta 2014, 1840 (3), 1063–1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Rinn JL; Kertesz M; Wang JK; Squazzo SL; Xu X; Brugmann SA; Goodnough LH; Helms JA; Farnham PJ; Segal E; Chang HY, Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell 2007, 129 (7), 1311–1323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Gupta RA; Shah N; Wang KC; Kim J; Horlings HM; Wong DJ; Tsai MC; Hung T; Argani P; Rinn JL; Wang Y; Brzoska P; Kong B; Li R; West RB; van de Vijver MJ; Sukumar S; Chang HY, Long noncoding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 2010, 464 (7291), 1071–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Rinn JL; Kertesz M; Wang JK; Squazzo SL; Xu X; Brugmann SA; Goodnough H; Helms JA; Farnham PJ; Segal E; Chang HY, Functional Demarcation of Active and Silent Chromatin Domains in Human HOX Loci by Non-Coding RNAs. Cell 2007, 129 (7), 1311–1323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Zhou F; Xie S; Li J; Duan S, Long noncoding RNA HOTAIR promotes cell apoptosis by sponging miR-221 in Parkinson’s disease. RSC Advances 2019, 9 (51), 29502–29510. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 12.Hu J; Wang Z; Shan Y; Pan Y; Ma J; Jia L, Long non-coding RNA HOTAIR promotes osteoarthritis progression via miR-17–5p/FUT2/β-catenin axis. Cell Death & Disease 2018, 9 (7), 711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Sathishkumar C; Prabu P; Mohan V; Balasubramanyam M, Linking a role of lncRNAs (long non-coding RNAs) with insulin resistance, accelerated senescence, and inflammation in patients with type 2 diabetes. Human Genomics 2018, 12 (1), 41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Yoon J-H; Abdelmohsen K; Kim J; Yang X; Martindale JL; Tominaga-Yamanaka K; White EJ; Orjalo AV; Rinn JL; Kreft SG; Wilson GM; Gorospe M, Scaffold function of long non-coding RNA HOTAIR in protein ubiquitination. Nature Communications 2013, 4 (1), 2939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Zhang H; Diab A; Fan H; Mani SKK; Hullinger R; Merle P; Andrisani O, PLK1 and HOTAIR Accelerate Proteasomal Degradation of SUZ12 and ZNF198 during Hepatitis B Virus-Induced Liver Carcinogenesis. Cancer Research 2015, 75 (11), 2363–2374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Obaid M; Udden SMN; Deb P; Shihabeddin N; Zaki MH; Mandal SS, LncRNA HOTAIR regulates lipopolysaccharide-induced cytokine expression and inflammatory response in macrophages. Scientific Reports 2018, 8 (1), 1–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Özeş AR; Miller DF; Özeş ON; Fang F; Liu Y; Matei D; Huang T; Nephew KP, NF-κB-HOTAIR axis links DNA damage response, chemoresistance and cellular senescence in ovarian cancer. Oncogene 2016, 35 (41), 5350–5361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Zhang Z; Cheng J; Wu Y; Qiu J; Sun Y; Tong X, LncRNA HOTAIR controls the expression of Rab22a by sponging miR-373 in ovarian cancer. Molecular Medicine Reports 2016, 14 (3), 2465–2472. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 19.Yang T; He X; Chen A; Tan K; Du X, LncRNA HOTAIR contributes to the malignancy of hepatocellular carcinoma by enhancing epithelial-mesenchymal transition via sponging miR-23b-3p from ZEB1. Gene 2018, 670, 114–122. [DOI] [PubMed] [Google Scholar]
- 20.Fujisaka Y; Iwata T; Tamai K; Nakamura M; Mochizuki M; Shibuya R; Yamaguchi K; Shimosegawa T; Satoh K, Long non-coding RNA HOTAIR up-regulates chemokine (C-C motif) ligand 2 and promotes proliferation of macrophages and myeloid-derived suppressor cells in hepatocellular carcinoma cell lines. Oncology Letters 2018, 15 (1), 509–514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Sun JCJJGHQSXZ, Long non-coding RNA HOTAIR modulates HLA-G expression by absorbing miR-148a in human cervical cancer. International Journal of Oncology 2020, 49 (3), 943–952. [DOI] [PubMed] [Google Scholar]
- 22.Bhan A; Hussain I; Ansari KI; Kasiri S; Bashyal A; Mandal SS, Antisense-transcript long noncoding RNA (lncRNA) HOTAIR is transcriptionally induced by estradiol. J Mol Biol 2013, 425 (19), 3707–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.al C. e., Upregulation of the long non-coding rna hotair promotes esophageal squamous cell carcinoma metastasis and poor prognosis - Chen - 2013 - Molecular Carcinogenesis - Wiley Online Library. 2020. [DOI] [PubMed] [Google Scholar]
- 24.W Z; Y A; Y L; XW X, Role of HOTAIR Long Noncoding RNA in Metastatic Progression of Lung Cancer. European review for medical and pharmacological sciences 2014, 18 (13). [PubMed] [Google Scholar]
- 25.Peng, LncRNA HOTAIR promotes colon cancer development by down-regulating miRNA-34a. 2019. [DOI] [PubMed] [Google Scholar]
- 26.Hajjari M; Salavaty A, HOTAIR: an oncogenic long non-coding RNA in different cancers. Cancer Biology & Medicine 2015, 12 (1), 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bhan A; Soleimani M; Mandal SS, Long Noncoding RNA and Cancer: A New Paradigm. Cancer Res 2017, 77 (15), 3965–3981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Bhan A; Mandal SS, LncRNA HOTAIR: A master regulator of chromatin dynamics and cancer. Biochimica et Biophysica Acta (BBA) - Reviews on Cancer 2015, 1856 (1), 151–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Zhang S; Chen S; Yang G; Gu F; Li M; Zhong B; Hu J; Hoffman A; Chen M, Long Noncoding RNA HOTAIR as an Independent Prognostic Marker in Cancer: A Meta-Analysis. PLoS ONE 2014, 9 (8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Wu Z-H; Wang X-L; Tang H-M; Jiang T; Chen J; Lu S; Qiu G-Q; Peng Z-H; Yan D-W, Long non-coding RNA HOTAIR is a powerful predictor of metastasis and poor prognosis and is associated with epithelial-mesenchymal transition in colon cancer. Oncology Reports 2014, 32 (1), 395–402. [DOI] [PubMed] [Google Scholar]
- 31.Zhou X; Chen J; Tang W, The molecular mechanism of HOTAIR in tumorigenesis, metastasis, and drug resistance. Acta biochimica et biophysica Sinica 2014. [DOI] [PubMed] [Google Scholar]
- 32.Yuan C; Ning Y; Pan Y, Emerging roles of HOTAIR in human cancer. Journal of Cellular Biochemistry 2020, 121 (5–6), 3235–3247. [DOI] [PubMed] [Google Scholar]
- 33.Fang S; Shen Y; Chen B; Wu Y; Jia L; Li Y; Zhu Y; Yan Y; Li M; Chen R; Guo L; Chen X; Chen Q, H3K27me3 induces multidrug resistance in small cell lung cancer by affecting HOXA1 DNA methylation via regulation of the lncRNA HOTAIR. In Ann Transl Med, 2018; Vol. 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Liu Z; Sun M; Lu K; Liu J; Zhang M; Wu W; De W; Wang Z; Wang R, The long noncoding RNA HOTAIR contributes to cisplatin resistance of human lung adenocarcinoma cells via downregualtion of p21(WAF1/CIP1) expression. PloS One 2013, 8 (10), e77293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Mozdarani H; Ezzatizadeh V; Rahbar Parvaneh R, The emerging role of the long non-coding RNA HOTAIR in breast cancer development and treatment. Journal of Translational Medicine 2020, 18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Tang L; Zhang W; Su B; Yu B, Long Noncoding RNA HOTAIR Is Associated with Motility, Invasion, and Metastatic Potential of Metastatic Melanoma. Biomed Res Int 2013, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.al Y. e., LncRNA HOTAIR participates in the development and progression of adrenocortical carcinoma via regulating cell cycle. 2018. [DOI] [PubMed] [Google Scholar]
- 38.Xavier-Magalhães A; Gonçalves CS; Fogli A; Lourenço T; Pojo M; Pereira B; Rocha M; Lopes MC; Crespo I; Rebelo O; Tão H; Lima J; Moreira R; Pinto AA; Jones C; Reis RM; Costello JF; Arnaud P; Sousa N; Costa BM, The long non-coding RNA HOTAIR is transcriptionally activated by HOXA9 and is an independent prognostic marker in patients with malignant glioma. In Oncotarget, 2018; Vol. 9, pp 15740–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Li J; Wang J; Zhong Y; Guo R; Chu D; Qiu H; Yuan Z, HOTAIR: a key regulator in gynecologic cancers. Cancer Cell Int 2017, 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Liu X-H; Sun M; Nie F-Q; Ge Y-B; Zhang E-B; Yin D-D; Kong R; Xia R; Lu K-H; Li J-H; De W; Wang K-M; Wang Z-X, Lnc RNA HOTAIR functions as a competing endogenous RNA to regulate HER2 expression by sponging miR-331–3p in gastric cancer. Molecular Cancer 2014, 13, 92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Pan W; Liu L; Wei J; Ge Y; Zhang J; Chen H; Zhou L; Yuan Q; Zhou C; Yang M, A functional lncRNA HOTAIR genetic variant contributes to gastric cancer susceptibility. Molecular Carcinogenesis 2016, 55 (1), 90–96. [DOI] [PubMed] [Google Scholar]
- 42.Zhang Z-Z; Shen Z-Y; Shen Y-Y; Zhao E-H; Wang M; Wang C-J; Cao H; Xu J, HOTAIR Long Noncoding RNA Promotes Gastric Cancer Metastasis through Suppression of Poly r(C)-Binding Protein (PCBP) 1. Molecular Cancer Therapeutics 2015, 14 (5), 1162–1170. [DOI] [PubMed] [Google Scholar]
- 43.FJ C; M S; SQ L; QQ W; L J; ZL L; GZ Z; G C; L J; HW X; CM W; J L; W D; M W; XF C, Upregulation of the Long Non-Coding RNA HOTAIR Promotes Esophageal Squamous Cell Carcinoma Metastasis and Poor Prognosis. Molecular carcinogenesis 2013, 52 (11). [DOI] [PubMed] [Google Scholar]
- 44.Jiang D; Xu L; Ni J; Zhang J; Cai M; Shen L, Functional polymorphisms in LncRNA HOTAIR contribute to susceptibility of pancreatic cancer. In Cancer Cell Int, 2019; Vol. 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Cai H; An Y; Chen X; Sun D; Chen T; Peng Y; Zhu F; Jiang Y; He X, Epigenetic inhibition of miR-663b by long non-coding RNA HOTAIR promotes pancreatic cancer cell proliferation via up-regulation of insulin-like growth factor 2. Oncotarget 2016, 7 (52), 86857–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Hu J; Wang Z; Shan Y; Pan Y; Ma J; Jia L, Long non-coding RNA HOTAIR promotes osteoarthritis progression via miR-17–5p/FUT2/β-catenin axis. Cell Death Dis 2018, 9 (7). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wang S; Zhang X; Guo Y; Rong H; Liu T, The long noncoding RNA HOTAIR promotes Parkinson’s disease by upregulating LRRK2 expression. Oncotarget 2017, 8 (15), 24449–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Q L; S H; Y D; N J; Y L, LncRNA HOTAIR Targets miR-126–5p to Promote the Progression of Parkinson’s Disease Through RAB3IP. Biological chemistry 2019, 400 (9). [DOI] [PubMed] [Google Scholar]
- 49.Spreafico M; Grillo B; Rusconi F; Battaglioli E; Venturin M, Multiple Layers of CDK5R1 Regulation in Alzheimer’s Disease Implicate Long Non-Coding RNAs. In Int J Mol Sci, 2018; Vol. 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kakhki MP; Nikravesh A; Farsani ZS; Sahraian MA; Behmanesh M, HOTAIR but not ANRIL long non-coding RNA contributes to the pathogenesis of multiple sclerosis. Immunology 2018, 153 (4), 479–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Song J; Kim D; Han J; Kim Y; Lee M; Jin E-J, PBMC and exosome-derived Hotair is a critical regulator and potent marker for rheumatoid arthritis. Clinical and Experimental Medicine 2014, 15 (1), 121–126. [DOI] [PubMed] [Google Scholar]
- 52.He B; Jiang D, HOTAIR-induced apoptosis is mediated by sponging miR-130a-3p to repress chondrocyte autophagy in knee osteoarthritis. Cell Biology International 2020, 44 (2), 524–535. [DOI] [PubMed] [Google Scholar]
- 53.Li M; Guo Y; Wang XJ; Duan BH; Li L, HOTAIR Participates in Hepatic Insulin Resistance via Regulating SIRT1. European review for medical and pharmacological sciences 2018. [DOI] [PubMed] [Google Scholar]
- 54.Li Z-Q; Gu X-Y; Hu J-X; Ping Y; Li H; Yan J-Y; Li J; Sun R; Yu Z-J; Zhang Y, Hepatitis C virus core protein impairs metabolic disorder of liver cell via HOTAIR-Sirt1 signalling. Bioscience Reports 2016, 36 (3). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Ma DD; Yuan LL; Lin LQ, LncRNA HOTAIR contributes to the tumorigenesis of nasopharyngeal carcinoma via up-regulating FASN. European Review for Medical and Pharmacological Sciences 2017, 21 (22), 5143–5152. [DOI] [PubMed] [Google Scholar]
- 56.Portoso M; Ragazzini R; Brenčič Ž; Moiani A; Michaud A; Vassilev I; Wassef M; Servant N; Sargueil B; Margueron R, PRC2 is dispensable for HOTAIR-mediated transcriptional repression. The EMBO journal 2017, 36 (8), 981–994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Li H; An J; Wu M; Zheng Q; Gui X; Li T; Pu H; Lu D, LncRNA HOTAIR promotes human liver cancer stem cell malignant growth through downregulation of SETD2. Oncotarget 2015, 6 (29), 27847–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Fu W-M; Lu Y-F; Hu B-G; Liang W-C; Zhu X; Yang H -d.; Li, G.; Zhang, J.-F., Long noncoding RNA Hotair mediated angiogenesis in nasopharyngeal carcinoma by direct and indirect signaling pathways. Oncotarget 2016, 7 (4), 4712–4723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Bartel DP, MicroRNA Target Recognition and Regulatory Functions. Cell 2009, 136 (2), 215–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Gozuacik D; Akkoc Y; Ozturk DG; Kocak M, Autophagy-Regulating microRNAs and Cancer. Front Oncol 2017, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Ling H; Girnita L; Buda O; Calin GA, Non-coding RNAs: the cancer genome dark matter that matters! Clinical Chemistry and Laboratory Medicine 2017, 55 (5), 705–714. [DOI] [PubMed] [Google Scholar]
- 62.Tan JY; Sirey T; Honti F; Graham B; Piovesan A; Merkenschlager M; Webber C; Ponting CP; Marques AC, Extensive microRNA-mediated crosstalk between lncRNAs and mRNAs in mouse embryonic stem cells. Genome Res 2015, 25 (5), 655–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Liu Y.w.; Sun M; Xia R; Zhang E. b.; Liu X. h.; Zhang Z. h.; Xu T. p.; De W; Liu B. r.; Wang Z. x., LincHOTAIR epigenetically silences miR34a by binding to PRC2 to promote the epithelial-to-mesenchymal transition in human gastric cancer. Cell Death & Disease 2015, 6, e1802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Bao X; Ren T; Huang Y; Sun K; Wang S; Liu K; Zheng B; Guo W, Knockdown of long non-coding RNA HOTAIR increases miR-454–3p by targeting Stat3 and Atg12 to inhibit chondrosarcoma growth. Cell Death & Disease 2017, 8 (2), e2605–e2605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Salmena L; Poliseno L; Tay Y; Kats L; Pandolfi PP, A ceRNA hypothesis: the Rosetta stone of a hidden RNA language? Cell 2011, 146 (3), 353–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Gao C.-y. X.-q. X. . F.-y. H Z.-j. X. H.-y. Y. B.-Z. L. -b. J. L.-y. W. S.-m. T, Long non-coding RNA HOTAIR modulates c-KIT expression through sponging miR-193a in acute myeloid leukemia - Xing - 2015 - FEBS Letters - Wiley Online Library. FEBS Letters 2015, 589 (15), 1981–1987. [DOI] [PubMed] [Google Scholar]
- 67.Zhang Z; Cheng J; Wu Y; Qiu J; Sun Y; Tong X, LncRNA HOTAIR controls the expression of Rab22a by sponging miR-373 in ovarian cancer. Mol Med Rep 2016, 14 (3), 2465–72. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 68.Caramel J; Ligier M; Puisieux A, Pleiotropic Roles for ZEB1 in Cancer. Cancer Research 2018, 78 (1), 30–35. [DOI] [PubMed] [Google Scholar]
- 69.Hu X; Ding D; Zhang J; Cui J, Knockdown of lncRNA HOTAIR sensitizes breast cancer cells to ionizing radiation through activating miR-218. Bioscience Reports 2019, 39 (4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Bian EB; Ma CC; He XJ; Wang C; Zong G; Wang HL; Zhao B, Epigenetic modification of miR-141 regulates SKA2 by an endogenous ‘sponge’ HOTAIR in glioma. Oncotarget 2016, 7 (21), 30610–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Sa L; Li Y; Zhao L; Liu Y; Wang P; Liu L; Li Z; Ma J; Cai H; Xue Y, The Role of HOTAIR/miR-148b-3p/USF1 on Regulating the Permeability of BTB. Front Mol Neurosci 2017, 10. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 72.Li T; Qin Y; Zhen Z; Shen H; Cong T; Schiferle E; Xiao S, Long non-coding RNA HOTAIR/microRNA-206 sponge regulates STC2 and further influences cell biological functions in head and neck squamous cell carcinoma. Cell Proliferation 2019, 52 (5), e12651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Hong Q; Li O; Zheng W; Xiao W; Zhang L; Wu D; Cai G; He JC; Chen X, LncRNA HOTAIR regulates HIF-1α/AXL signaling through inhibition of miR-217 in renal cell carcinoma. Cell Death Dis 2017, 8 (5), e2772-. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 74.Ling Z; Wang X; Tao T; Zhang L; Guan H; You Z; Lu K; Zhang G; Chen S; Wu J; Qian J; Liu H; Xu B; Chen M, Involvement of aberrantly activated HOTAIR/EZH2/miR-193a feedback loop in progression of prostate cancer. J Exp Clin Cancer Res 2017, 36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Zheng P; Yin Z; Wu Y; Xu Y; Luo Y; Zhang TC, LncRNA HOTAIR promotes cell migration and invasion by regulating MKL1 via inhibition miR206 expression in HeLa cells. Cell Commun Signal 2018, 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Wang LP; Wang JP; Wang XP, HOTAIR contributes to the growth of liver cancer via targeting miR-217. Oncol Lett 2018, 15 (5), 7963–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Su J; Wang Q; Liu Y; Zhong M, miR-217 inhibits invasion of hepatocellular carcinoma cells through direct suppression of E2F3. Molecular and Cellular Biochemistry 2014, 392 (1), 289–296. [DOI] [PubMed] [Google Scholar]
- 78.Liu B; Liu Q; Pan S; Huang Y; Qi Y; Li S; Xiao Y; Jia L, The HOTAIR/miR-214/ST6GAL1 crosstalk modulates colorectal cancer procession through mediating sialylated c-Met via JAK2/STAT3 cascade. J Exp Clin Cancer Res 2019, 38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.H X; B W; YS X, LncRNA HOTAIR Regulates the Proliferation and Apoptosis of Vascular Smooth Muscle Cells Through Targeting miRNA-130b-3p/PPARα Axis. European review for medical and pharmacological sciences 2019, 23 (24). [DOI] [PubMed] [Google Scholar]
- 80.T S; Y H; W T; H S; Z Y; J G, The Long Noncoding RNA HOTAIR Serves as a microRNA-34a-5p Sponge to Reduce Nucleus Pulposus Cell Apoptosis via a NOTCH1-mediated Mechanism. Gene 2019, 715. [DOI] [PubMed] [Google Scholar]
- 81.Y Y; X Z; Z L; L K; Y H, LncRNA HOTAIR Suppresses TNF-α Induced Apoptosis of Nucleus Pulposus Cells by Regulating miR-34a/Bcl-2 Axis. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie 2018, 107. [DOI] [PubMed] [Google Scholar]
- 82.Gao L; Liu Y; Guo S; Yao R; Wu L; Xiao L; Wang Z; Liu Y; Zhang Y, Circulating Long Noncoding RNA HOTAIR is an Essential Mediator of Acute Myocardial Infarction. Cellular Physiology and Biochemistry: International Journal of Experimental Cellular Physiology, Biochemistry, and Pharmacology 2017, 44 (4), 1497–1508. [DOI] [PubMed] [Google Scholar]
- 83.Li L; Zhang M; Chen W; Wang R; Ye Z; Wang Y; Li X; Cai C, LncRNA-HOTAIR inhibition aggravates oxidative stress-induced H9c2 cells injury through suppression of MMP2 by miR-125. Acta Biochimica Et Biophysica Sinica 2018, 50 (10), 996–1006. [DOI] [PubMed] [Google Scholar]
- 84.Cheng D; Deng J; Zhang B; He X; Meng Z; Li G; Ye H; Zheng S; Wei L; Deng X; Chen R; Zhou J, LncRNA HOTAIR epigenetically suppresses miR-122 expression in hepatocellular carcinoma via DNA methylation. EBioMedicine 2018, 36, 159–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.BB A, Nuclear factor-kappaB: The Enemy Within. Cancer cell 2004, 6 (3). [DOI] [PubMed] [Google Scholar]
- 86.Xia Y; Shen S; Verma IM, NF-κB, an active player in human cancers. Cancer Immunol Res 2014, 2 (9), 823–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Obaid M; Udden SMN; Deb P; Shihabeddin N; Zaki MH; Mandal SS, LncRNA HOTAIR regulates lipopolysaccharide-induced cytokine expression and inflammatory response in macrophages. Sci Rep 2018, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Obaid M; Udden SMN; Alluri P; Mandal SS, LncRNA HOTAIR regulates glucose transporter Glut1 expression and glucose uptake in macrophages during inflammation. Sci Rep 2021, 11 (1), 232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Özeş AR; Miller DF; Özeş ON; Fang F; Liu Y; Matei D; Huang T; Nephew KP, NF-κB-HOTAIR axis links DNA damage response, chemoresistance and cellular senescence in ovarian cancer. Oncogene 2016, 35 (41), 5350–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Lu X; Bai D; Liu X; Zhou C; Yang G, Sedentary lifestyle related exosomal release of Hotair from gluteal-femoral fat promotes intestinal cell proliferation. Scientific Reports 2017, 7 (1), 45648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.H W; J L; W L; G L; Z L, LncRNA-HOTAIR Promotes TNF-α Production in Cardiomyocytes of LPS-induced Sepsis Mice by Activating NF-κB Pathway. Biochemical and biophysical research communications 2016, 471 (1). [DOI] [PubMed] [Google Scholar]
- 92.Liu G; Zhang W, Long non-coding RNA HOTAIR promotes UVB-induced apoptosis and inflammatory injury by up-regulation of PKR in keratinocytes. Braz J Med Biol Res 2018, 51 (8). [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 93.Chen J; Gu X; Zhou L; Wang S; Zhu L; Huang Y; Cao F, Long non-coding RNA-HOTAIR promotes the progression of sepsis by acting as a sponge of miR-211 to induce IL-6R expression. Experimental and Therapeutic Medicine 2019, 18 (5), 3959–3967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Li P; Zhang X; Wang L; Du L; Yang Y; Liu T; Li C; Wang C, lncRNA HOTAIR Contributes to 5FU Resistance through Suppressing miR-218 and Activating NF-κB/TS Signaling in Colorectal Cancer. Mol Ther Nucleic Acids 2017, 8, 356–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.W L; L Z; ZB R; MX W; Y R; W L, HOTAIR Promotes Inflammatory Response After Acute Myocardium Infarction by Upregulating RAGE. European review for medical and pharmacological sciences 2018, 22 (21). [DOI] [PubMed] [Google Scholar]
- 96.HJ Z; QF W; SJ W; HJ Z; XY Z; Q, G.; YH, C.; XH, W., LncRNA HOTAIR Alleviates Rheumatoid Arthritis by Targeting miR-138 and Inactivating NF-κB Pathway. International immunopharmacology 2017, 50. [DOI] [PubMed] [Google Scholar]
- 97.Y F; T I; K T; M N; M M; R S; K Y; T S; K S, Long Non-Coding RNA HOTAIR Up-Regulates Chemokine (C-C Motif) Ligand 2 and Promotes Proliferation of Macrophages and Myeloid-Derived Suppressor Cells in Hepatocellular Carcinoma Cell Lines. Oncology letters 2018, 15 (1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.J L; S L; Z C; J W; Y C; Z X; M J; W Y, miR-326 Reverses Chemoresistance in Human Lung Adenocarcinoma Cells by Targeting Specificity Protein 1. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine 2016, 37 (10). [DOI] [PubMed] [Google Scholar]
- 99.Sun J, Chu H, Ji J, Huo G, Song Q, Zhang X, Long non-coding RNA HOTAIR modulates HLA-G expression by absorbing miR-148a in human cervical cancer. International Journal of Oncology 2016, 49 (3), 943–952. [DOI] [PubMed] [Google Scholar]
- 100.L L; Q D; H X; Z Y; D H; L L; W S; S Y; C C, Infiltrating Mast Cells Enhance Prostate Cancer Invasion via Altering LncRNA-HOTAIR/PRC2-androgen Receptor (AR)-MMP9 Signals and Increased Stem/Progenitor Cell Population. Oncotarget 2015, 6 (16). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Ma M; Li C; Zhang Y; Weng M; Zhang M; Qin Y; Gong W; Quan Z, Long non-coding RNA HOTAIR, a c-Myc activated driver of malignancy, negatively regulates miRNA-130a in gallbladder cancer. Mol Cancer 2014, 13, 156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.C PA; AS F; BR M; R d. B E. L. B.; MC B; d. S. JE; V V; MA Z; WA S, Brief Report: The lincRNA Hotair Is Required for Epithelial-To-Mesenchymal Transition and Stemness Maintenance of Cancer Cell Lines. Stem cells (Dayton, Ohio) 2013, 31 (12). [DOI] [PubMed] [Google Scholar]
- 103.Su M; Wang H; Wang W; Wang Y; Ouyang L; Pan C; Xia L; Cao D; Liao Q, LncRNAs in DNA damage response and repair in cancer cells. Acta Biochimica et Biophysica Sinica 2018, 50 (5), 433–439. [DOI] [PubMed] [Google Scholar]
- 104.Tehrani SS; Karimian A; Parsian H; Majidinia M; Yousefi B, Multiple Functions of Long Non-Coding RNAs in Oxidative Stress, DNA Damage Response and Cancer Progression. J Cell Biochem 2018, 119 (1), 223–236. [DOI] [PubMed] [Google Scholar]
- 105.Liu H; Li Z; Wang C; Feng L; Huang H; Liu C; Li F, Expression of long non-coding RNA-HOTAIR in oral squamous cell carcinoma Tca8113 cells and its associated biological behavior. American Journal of Translational Research 2016, 8 (11), 4726–4734. [PMC free article] [PubMed] [Google Scholar]
- 106.Lee S; Kopp F; Chang T-C; Sataluri A; Chen B; Sivakumar S; Yu H; Xie Y; Mendell, Joshua T, Noncoding RNA NORAD Regulates Genomic Stability by Sequestering PUMILIO Proteins. Cell 2016, 164 (1), 69–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Michelini F; Pitchiaya S; Vitelli V; Sharma S; Gioia U; Pessina F; Cabrini M; Wang Y; Capozzo I; Iannelli F; Matti V; Francia S; Shivashankar GV; Walter NG; d’Adda di Fagagna F, Damage-induced lncRNAs control the DNA damage response through interaction with DDRNAs at individual double-strand breaks. Nature Cell Biology 2017, 19 (12), 1400–1411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Thapar R, Regulation of DNA Double-Strand Break Repair by Non-Coding RNAs. Molecules 2018, 23 (11), 2789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.D’Alessandro G; Whelan DR; Howard SM; Vitelli V; Renaudin X; Adamowicz M; Iannelli F; Jones-Weinert CW; Lee M; Matti V; Lee WTC; Morten MJ; Venkitaraman AR; Cejka P; Rothenberg E; d’Adda di Fagagna F, BRCA2 controls DNA:RNA hybrid level at DSBs by mediating RNase H2 recruitment. Nature Communications 2018, 9 (1), 5376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Hu Z; Mi S; Zhao T; Peng C; Peng Y; Chen L; Zhu W; Yao Y; Song Q; Li X; Li X; Jia C; Pei H, BGL3 lncRNA mediates retention of the BRCA1/BARD1 complex at DNA damage sites. The EMBO Journal 2020, 39 (12), e104133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Gazy I; Zeevi DA; Renbaum P; Zeligson S; Eini L; Bashari D; Smith Y; Lahad A; Goldberg M; Ginsberg D; Levy-Lahad E, TODRA, a lncRNA at the RAD51 Locus, Is Oppositely Regulated to RAD51, and Enhances RAD51-Dependent DSB (Double Strand Break) Repair. PLOS ONE 2015, 10 (7), e0134120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Wan G; Hu X; Liu Y; Han C; Sood AK; Calin GA; Zhang X; Lu X, A novel non-coding RNA lncRNA-JADE connects DNA damage signalling to histone H4 acetylation. The EMBO Journal 2013, 32 (21), 2833–2847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Betts JA; Moradi Marjaneh M; Al-Ejeh F; Lim YC; Shi W; Sivakumaran H; Tropée R; Patch AM; Clark MB; Bartonicek N; Wiegmans AP; Hillman KM; Kaufmann S; Bain AL; Gloss BS; Crawford J; Kazakoff S; Wani S; Wen SW; Day B; Möller A; Cloonan N; Pearson J; Brown MA; Mercer TR; Waddell N; Khanna KK; Dray E; Dinger ME; Edwards SL; French JD, Long Noncoding RNAs CUPID1 and CUPID2 Mediate Breast Cancer Risk at 11q13 by Modulating the Response to DNA Damage. The American Journal of Human Genetics 2017, 101 (2), 255–266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Sharma V; Khurana S; Kubben N; Abdelmohsen K; Oberdoerffer P; Gorospe M; Misteli T, A BRCA1-interacting lncRNA regulates homologous recombination. EMBO reports 2015, 16 (11), 1520–1534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Wan G; Mathur R; Hu X; Liu Y; Zhang X; Peng G; Lu X, Long non-coding RNA ANRIL (CDKN2B-AS) is induced by the ATM-E2F1 signaling pathway. Cellular Signalling 2013, 25 (5), 1086–1095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Schmitt AM; Garcia JT; Hung T; Flynn RA; Shen Y; Qu K; Payumo AY; Peres-da-Silva A; Broz DK; Baum R; Guo S; Chen JK; Attardi LD; Chang HY, An inducible long noncoding RNA amplifies DNA damage signaling. Nature Genetics 2016, 48 (11), 1370–1376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Sánchez Y; Segura V; Marín-Béjar O; Athie A; Marchese FP; González J; Bujanda L; Guo S; Matheu A; Huarte M, Genome-wide analysis of the human p53 transcriptional network unveils a lncRNA tumour suppressor signature. Nature Communications 2014, 5 (1), 5812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Guttman M; Amit I; Garber M; French C; Lin MF; Feldser D; Huarte M; Zuk O; Carey BW; Cassady JP; Cabili MN; Jaenisch R; Mikkelsen TS; Jacks T; Hacohen N; Bernstein BE; Kellis M; Regev A; Rinn JL; Lander ES, Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature 2009, 458 (7235), 223–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Dimitrova N; Zamudio, Jesse R; Jong Robyn M.; Soukup D; Resnick R; Sarma K; Ward Amanda J.; Raj A; Lee Jeannie T.; Sharp Phillip A.; Jacks T, LincRNA-p21 Activates p21 In cis to Promote Polycomb Target Gene Expression and to Enforce the G1/S Checkpoint. Molecular Cell 2014, 54 (5), 777–790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Huarte M; Guttman M; Feldser D; Garber M; Koziol MJ; Kenzelmann-Broz D; Khalil AM; Zuk O; Amit I; Rabani M; Attardi LD; Regev A; Lander ES; Jacks T; Rinn JL, A Large Intergenic Noncoding RNA Induced by p53 Mediates Global Gene Repression in the p53 Response. Cell 2010, 142 (3), 409–419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Tran UM; Rajarajacholan U; Soh J; Kim T. s.; Thalappilly, S.; Sensen, C. W.; Riabowol, K., LincRNA-p21 acts as a mediator of ING1b-induced apoptosis. Cell Death & Disease 2015, 6 (3), e1668-e1668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Hung T; Wang Y; Lin MF; Koegel AK; Kotake Y; Grant GD; Horlings HM; Shah N; Umbricht C; Wang P; Wang Y; Kong B; Langerød A; Børresen-Dale A-L; Kim SK; van de Vijver M; Sukumar S; Whitfield ML; Kellis M; Xiong Y; Wong DJ; Chang HY, Extensive and coordinated transcription of noncoding RNAs within cell-cycle promoters. Nature Genetics 2011, 43 (7), 621–629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Negishi M; Wongpalee SP; Sarkar S; Park J; Lee KY; Shibata Y; Reon BJ; Abounader R; Suzuki Y; Sugano S; Dutta A, A New lncRNA APTR, Associates with and Represses the CDKN1A/p21 Promoter by Recruiting Polycomb Proteins. PLOS ONE 2014, 9 (4), e95216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Xu D; Wang Y; Wang J; Qi F; Sun Y, The Potential Regulatory Roles of lncRNAs in DNA Damage Response in Human Lymphocytes Exposed to UVC Irradiation. BioMed Research International 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Hu WL; Jin L; Xu A; Wang YF; Thorne RF; Zhang XD; Wu M, GUARDIN is a p53-responsive long non-coding RNA that is essential for genomic stability. Nature Cell Biology 2018, 20 (4), 492–502. [DOI] [PubMed] [Google Scholar]
- 126.Diaz-Lagares A; Crujeiras AB; Lopez-Serra P; Soler M; Setien F; Goyal A; Sandoval J; Hashimoto Y; Martinez-Cardús A; Gomez A; Heyn H; Moutinho C; Espada J; Vidal A; Paúles M; Galán M; Sala N; Akiyama Y; Martínez-Iniesta M; Farré L; Villanueva A; Gross M; Diederichs S; Guil S; Esteller M, Epigenetic inactivation of the p53-induced long noncoding RNA TP53 target 1 in human cancer. Proceedings of the National Academy of Sciences 2016, 113 (47), E7535–E7544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Adriaens C; Standaert L; Barra J; Latil M; Verfaillie A; Kalev P; Boeckx B; Wijnhoven PWG; Radaelli E; Vermi W; Leucci E; Lapouge G; Beck B; van den Oord J; Nakagawa S; Hirose T; Sablina AA; Lambrechts D; Aerts S; Blanpain C; Marine J-C, p53 induces formation of NEAT1 lncRNA-containing paraspeckles that modulate replication stress response and chemosensitivity. Nature Medicine 2016, 22 (8), 861–868. [DOI] [PubMed] [Google Scholar]
- 128.Mahmoudi S; Henriksson S; Corcoran M; Méndez-Vidal C; Wiman KG; Farnebo M, Wrap53, a Natural p53 Antisense Transcript Required for p53 Induction upon DNA Damage. Molecular Cell 2016, 64 (5), 1009. [DOI] [PubMed] [Google Scholar]
- 129.Shihabudeen Haider Ali MS; Cheng X; Moran M; Haemmig S; Naldrett MJ; Alvarez S; Feinberg MW; Sun X, LncRNA Meg3 protects endothelial function by regulating the DNA damage response. Nucleic Acids Research 2019, 47 (3), 1505–1522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Zhang H; Hua Y; Jiang Z; Yue J; Shi M; Zhen X; Zhang X; Yang L; Zhou R; Wu S, Cancer-associated Fibroblast–promoted LncRNA DNM3OS Confers Radioresistance by Regulating DNA Damage Response in Esophageal Squamous Cell Carcinoma. Clinical Cancer Research 2019, 25 (6), 1989–2000. [DOI] [PubMed] [Google Scholar]
- 131.Gioia R; Drouin S; Ouimet M; Caron M; St-Onge P; Richer C; Sinnett D, LncRNAs downregulated in childhood acute lymphoblastic leukemia modulate apoptosis, cell migration, and DNA damage response. Oncotarget 2017, 8 (46), 80645–80650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Avogaro L; Querido E; Dalachi M; Jantsch MF; Chartrand P; Cusanelli E, Live-cell imaging reveals the dynamics and function of single-telomere TERRA molecules in cancer cells. RNA Biology 2018, 15 (6), 787–796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Prieto-García E; Díaz-García CV; García-Ruiz I; Agulló-Ortuño MT, Epithelial-to-mesenchymal transition in tumor progression. Medical Oncology 2017, 34 (7), 122. [DOI] [PubMed] [Google Scholar]
- 134.ElMoneim HMA; Zaghloul NM, Expression of e-cadherin, n-cadherin and snail and their correlation with clinicopathological variants: an immunohistochemical study of 132 invasive ductal breast carcinomas in Egypt. Clinics (Sao Paulo) 2011, 66 (10), 1765–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.F Z; J L; C M; X T; Q T; J W; X C; J X; XB Y; SS H, Novel Regulation of miR-34a-5p and HOTAIR by the Combination of Berberine and Gefitinib Leading to Inhibition of EMT in Human Lung Cancer. Journal of cellular and molecular medicine 2020, 24 (10). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Battistelli C; Cicchini C; Santangelo L; Tramontano A; Grassi L; Gonzalez FJ; de Nonno V; Grassi G; Amicone L; Tripodi M, The Snail repressor recruits EZH2 to specific genomic sites through the enrollment of the lncRNA HOTAIR in epithelial-to-mesenchymal transition. Oncogene 2017, 36 (7), 942–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Y R; HH J; YQ X; X Z; XH Z; YF W; X S; ZY Z; T S; Y D; WP T; XL Z; CS K; M M, Paracrine and Epigenetic Control of CAF-induced Metastasis: The Role of HOTAIR Stimulated by TGF-ß1 Secretion. Molecular cancer 2018, 17 (1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Song Y; Wang R; Li L-W; Liu X; Wang Y-F; Wang Q-X; Zhang Q, Long non-coding RNA HOTAIR mediates the switching of histone H3 lysine 27 acetylation to methylation to promote epithelial-to-mesenchymal transition in gastric cancer. International Journal of Oncology 2018, 54 (1), 77–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Pádua Alves C; Fonseca AS; Muys BR; de Barros E Lima Bueno R; Bürger MC; de Souza JES; Valente V; Zago MA; Silva WA, Brief report: The lincRNA Hotair is required for epithelial-to-mesenchymal transition and stemness maintenance of cancer cell lines. Stem Cells (Dayton, Ohio) 2013, 31 (12), 2827–2832. [DOI] [PubMed] [Google Scholar]
- 140.Yoon JH; Abdelmohsen K; Kim J; Yang X; Martindale JL; Tominaga-Yamanaka K; White EJ; Orjalo AV; Rinn JL; Kreft SG; Wilson GM; Gorospe M, Scaffold function of long non-coding RNA HOTAIR in protein ubiquitination. Nat Commun 2013, 4, 2939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Lee S; Hong S; Kim S; Kang S, Ataxin-1 occupies the promoter region of E-cadherin in vivo and activates CtBP2-repressed promoter. Biochim Biophys Acta 2011, 1813 (5), 713–22. [DOI] [PubMed] [Google Scholar]
- 142.Zhang H; Xing Z; Mani SK; Bancel B; Durantel D; Zoulim F; Tran EJ; Merle P; Andrisani O, RNA helicase DEAD box protein 5 regulates Polycomb repressive complex 2/Hox transcript antisense intergenic RNA function in hepatitis B virus infection and hepatocarcinogenesis. Hepatology 2016, 64 (4), 1033–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.HOTAIR induces the ubiquitination of Runx3 by interacting with Mex3b and enhances the invasion of gastric cancer cells | SpringerLink. [DOI] [PubMed] [Google Scholar]
- 144.A Z; JC Z; J K; KW F; YA Y; D C; YY M; J Y, LncRNA HOTAIR Enhances the Androgen-Receptor-Mediated Transcriptional Program and Drives Castration-Resistant Prostate Cancer. Cell reports 2015, 13 (1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Ren K; Li Y; Lu H; Li Z; Li Z; Wu K; Li Z; Han X, Long Noncoding RNA HOTAIR Controls Cell Cycle by Functioning as a Competing Endogenous RNA in Esophageal Squamous Cell Carcinoma. Translational Oncology 2016, 9 (6), 489–497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Liu M; Zhang H; Li Y; Wang R; Ren D; Liu H; Kang C; Chen J, HOTAIR, a long noncoding RNA, is a marker of abnormal cell cycle regulation in lung cancer. Cancer Sci 2018, 109 (9), 2717–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Cai H; Yao J; An Y; Chen X; Chen W; Wu D; Luo B; Yang Y; Jiang Y; Sun D; He X, LncRNA HOTAIR acts a competing endogenous RNA to control the expression of notch3 via sponging miR-613 in pancreatic cancer. Oncotarget 2017, 8 (20), 32905–32917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Bhan A; Hussain I; Ansari KI; Kasiri S; Bashyal A; Mandal SS, Antisense Transcript Long Noncoding RNA (lncRNA) HOTAIR is Transcriptionally Induced by Estradiol. Journal of Molecular Biology 2013, 425 (19), 3707–3722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Bhan A; Hussain I; Ansari KI; Bobzean SAM; Perrotti LI; Mandal SS, Bisphenol-A and diethylstilbestrol exposure induces the expression of breast cancer associated long noncoding RNA HOTAIR in vitro and in vivo. J Steroid Biochem Mol Biol 2014, 141, 160–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.A B; P D; N; KI A; M B; SS M, Histone Methylase MLL1 Coordinates With HIF and Regulate lncRNA HOTAIR Expression Under Hypoxia. Gene 2017, 629. [DOI] [PubMed] [Google Scholar]
- 151.Zeng W; Liu P; Pan W; Singh SR; Wei Y, Hypoxia and hypoxia inducible factors in tumor metabolism. Cancer Lett 2015, 356 (2 Pt A), 263–7. [DOI] [PubMed] [Google Scholar]
- 152.Acker T; Plate KH, Role of hypoxia in tumor angiogenesis-molecular and cellular angiogenic crosstalk. Cell Tissue Res 2003, 314 (1), 145–55. [DOI] [PubMed] [Google Scholar]
- 153.Ma M-Z; Li C-X; Zhang Y; Weng M-Z; Zhang M.-d.; Qin Y-Y; Gong W; Quan Z-W, Long non-coding RNA HOTAIR, a c-Myc activated driver of malignancy, negatively regulates miRNA-130a in gallbladder cancer. Molecular Cancer 2014, 13, 156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Su DN; Wu SP; Chen HT; He JH, HOTAIR, a long non-coding RNA driver of malignancy whose expression is activated by FOXC1, negatively regulates miRNA-1 in hepatocellular carcinoma. Oncol Lett 2016, 12 (5), 4061–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Xavier-Magalhães A; Gonçalves CS; Fogli A; Lourenço T; Pojo M; Pereira B; Rocha M; Lopes MC; Crespo I; Rebelo O; Tão H; Lima J; Moreira R; Pinto AA; Jones C; Reis RM; Costello JF; Arnaud P; Sousa N; Costa BM, The long non-coding RNA HOTAIR is transcriptionally activated by HOXA9 and is an independent prognostic marker in patients with malignant glioma. Oncotarget 2018, 9 (21), 15740–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Chiyomaru T; Yamamura S; Fukuhara S; Yoshino H; Kinoshita T; Majid S; Saini S; Chang I; Tanaka Y; Enokida H; Seki N; Nakagawa M; Dahiya R, Genistein Inhibits Prostate Cancer Cell Growth by Targeting miR-34a and Oncogenic HOTAIR. PLoS One 2013, 8 (8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Chiyomaru T; Fukuhara S; Saini S; Majid S; Deng G; Shahryari V; Chang I; Tanaka Y; Enokida H; Nakagawa M; Dahiya R; Yamamura S, Long non-coding RNA HOTAIR is targeted and regulated by miR-141 in human cancer cells. The Journal of Biological Chemistry 2014, 289 (18), 12550–12565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Carrion K; Dyo J; Patel V; Sasik R; Mohamed SA; Hardiman G; Nigam V, The Long Non-Coding HOTAIR Is Modulated by Cyclic Stretch and WNT/β-CATENIN in Human Aortic Valve Cells and Is a Novel Repressor of Calcification Genes. PLoS One 2014, 9 (5). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Mercer TR; Dinger ME; Mattick JS, Long non-coding RNAs: insights into functions. Nat Rev Genet 2009, 10 (3), 155–9. [DOI] [PubMed] [Google Scholar]
- 160.Cabili MN; Trapnell C; Goff L; Koziol M; Tazon-Vega B; Regev A; Rinn JL, Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev 2011, 25 (18), 1915–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Shi T; Gao G; Cao Y, Long Noncoding RNAs as Novel Biomarkers Have a Promising Future in Cancer Diagnostics. Dis Markers 2016, 2016, 9085195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Li N; Wang Y; Liu X; Luo P; Jing W; Zhu M; Tu J, Identification of Circulating Long Noncoding RNA HOTAIR as a Novel Biomarker for Diagnosis and Monitoring of Non-Small Cell Lung Cancer. Technol Cancer Res Treat 2017, 16 (6), 1060–1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Wang W; He X; Zheng Z; Ma X; Hu X; Wu D; Wang M, Serum HOTAIR as a novel diagnostic biomarker for esophageal squamous cell carcinoma. Mol Cancer 2017, 16 (1), 75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Elsayed ET; Salem PE; Darwish AM; Fayed HM, Plasma long non-coding RNA HOTAIR as a potential biomarker for gastric cancer. Int J Biol Markers 2018, 1724600818760244. [DOI] [PubMed] [Google Scholar]
- 165.Tan SK; Pastori C; Penas C; Komotar RJ; Ivan ME; Wahlestedt C; Ayad NG, Serum long noncoding RNA HOTAIR as a novel diagnostic and prognostic biomarker in glioblastoma multiforme. Mol Cancer 2018, 17 (1), 74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Berrondo C; Flax J; Kucherov V; Siebert A; Osinski T; Rosenberg A; Fucile C; Richheimer S; Beckham CJ, Expression of the Long Non-Coding RNA HOTAIR Correlates with Disease Progression in Bladder Cancer and Is Contained in Bladder Cancer Patient Urinary Exosomes. PLoS One 2016, 11 (1), e0147236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Xie Z; Chen X; Li J; Guo Y; Li H; Pan X; Jiang J; Liu H; Wu B, Salivary HOTAIR and PVT1 as novel biomarkers for early pancreatic cancer. Oncotarget 2016, 7 (18), 25408–19. [DOI] [PMC free article] [PubMed] [Google Scholar]