Next Article in Journal
Spontaneous Tumor Regression and Reversion: Insights and Associations with Reduced Dietary Phosphate
Previous Article in Journal
Past Happiness and Broken Future Horizon of Oncological Patients during Chemotherapy—A Quantitative Exploration of a Phenomenological Hypothesis
Previous Article in Special Issue
Implementation of HER2 Testing in Endometrial Cancer, a Summary of Real-World Initial Experience in a Large Tertiary Cancer Center
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Promotive and Inhibitory Role of Long Non-Coding RNAs in Endometrial Cancer Course—A Review

1
Department of Gynecology, Gynecology Oncology and Obstetrics, Fryderyk Chopin University Hospital, 35-055 Rzeszow, Poland
2
Laboratory of Molecular Biology, Centre for Innovative Research in Medical and Natural Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
3
Institute of Medical Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
*
Authors to whom correspondence should be addressed.
Cancers 2024, 16(11), 2125; https://doi.org/10.3390/cancers16112125
Submission received: 29 April 2024 / Revised: 28 May 2024 / Accepted: 31 May 2024 / Published: 3 June 2024
(This article belongs to the Special Issue Endometrial Cancer: Old Questions and New Perspectives (Volume II))

Abstract

:

Simple Summary

Endometrial cancer has emerged as the main gynecological malignant tumour in developed countries. The development of molecular techniques allows the precise profiling of each case. This affects the selection of a therapy and the patient’s prognosis. Long non-coding RNAs, which influence tumour development, are a novelty in endometrial cancer. They can both stimulate and inhibit the progression of the disease. Their number is constantly growing, and the mechanisms of action are not fully discovered. Some of them may become targets for new molecular therapies in the future.

Abstract

Endometrial cancer is one of the most common malignant tumours in women. The development of this tumour is associated with several genetic disorders, many of which are still unknown. One type of RNA molecules currently being intensively studied in many types of cancer are long non-coding RNAs (lncRNAs). LncRNA-coding genes occupy a large fraction of the human genome. LncRNAs regulate many aspects of cell development, metabolism, and other physiological processes. Diverse types of lncRNA can function as a tumour suppressor or an oncogene that can alter migration, invasion, cell proliferation, apoptosis, and immune system response. Recent studies suggest that selected lncRNAs are important in an endometrial cancer course. Our article describes over 70 lncRNAs involved in the development of endometrial cancer, which were studied via in vivo and in vitro research. It was proved that lncRNAs could both promote and inhibit the development of endometrial cancer. In the future, lncRNAs may become an important therapeutic target. The aim of this study is to review the role of lncRNAs in the development of carcinoma of uterine body.

1. Introduction

1.1. Endometrial Cancer

Endometrial cancer (EC) is considered one of the most common malignant tumours of the female reproductive system. In 2020, 417,367 new cases of this cancer were diagnosed worldwide. A total of 97,370 women died, and still the incidence of EC continues increasing, especially in North America and Western Europe [1]. This is related to the ageing of the population, but also to the increasing rate of obesity, which is the main risk factor for this cancer as it is responsible for over 50% of cases [2]. Other risk factors include smoking, type 2 diabetes, polycystic ovarian syndrome, or estrogen therapy after menopause [3]. EC can be divided into morphological and molecular classifications. Historically, Bokhman (1983) suggested the first subtype classification of endometrial carcinoma, which distinguished between type I and type II endometrial cancer. The classification was based on clinical and hormonal traits. Type I, called endometrioid endometrial cancer, includes grade I-II endometroid adenocarcinoma. It accounts for 80% of all EC and is correlated with an excessive estrogen stimulation. Type II, called non-endometrioid endometrial cancer, is less common, and has a worse prognosis than type I. Type II EC is divided into grade III endometrioid adenocarcinoma, serous clear cell, undifferentiated, and carcinosarcomas [4,5]. The Cancer Genome Atlas presented the molecular classification of EC. It consists of four subgroups: POLE (ultra-mutated), microsatellite instability hypermutated or mismatch repair deficiency microsatellite instable (MSI), copy-number low (microsatellite stable), and copy-number high or p53 abnormal (serous-like) [6,7]. Each subgroup is correlated with different genes mutations and prognosis. EC is treated by hysterectomy with pelvic lymph nodes excision. Depending on the stage of cancer, chemotherapy, radiotherapy, and chemoradiotherapy can also be deployed [3].

1.2. Long Non-Coding RNAs (lncRNAs)

Non-coding RNAs are molecules which are not translated into proteins. However, this definition is not fully correct. Recent research has shown that some long non-coding RNAs (lncRNAs) encode the formation of small peptide molecules. LncRNAs can influence cell metabolism and tissue functioning in various ways, not only in connection with proteins synthesis. According to their length, they can be divided into two groups: short non-coding RNA and long non-coding RNA. It was established that lncRNA is a transcripted particle of RNA that is more than 200 nucleotides in length and is not translated into proteins. According to the origin (genomic origin), there are differentiated subtypes of lncRNA: intronic lncRNA, intergenic lncRNA, antisense lncRNA, bidirectional lncRNA, enhancer RNA, and circular lncRNA (Table 1) [8,9,10].
LncRNA is a product of polymerase RNA. Hence, the biogenesis of this molecule is like messenger RNA (mRNA). Due to polyadenylation, alternative splicing, and cleavage different isoforms of lncRNA can be created. Additionally, LncRNAs can be modified by some transcription factors like p53, Sox2, or nuclear factor-kappa B (NF-κB) [11,12]. LncRNA can be found in the nucleus, nucleolus, cytoplasm, and mitochondria. Currently, many lncRNA molecules are being discovered in the human body, and their number is constantly growing. Even though they are not responsible for the formation of proteins, they perform several crucial functions in the body. LncRNA takes part in the regulation of the genome, cell structure, and gene expression. It regulates the chromatin structure, transcription, translation, and processing of RNA. LncRNAs are also involved in the development of cells, the regulation of the immune system, the expression of cytokines, and the maintenance of homeostasis [13,14,15]. On the other hand, lncRNAs have an impact on pathological processes. They influence the development of the cardiovascular system, as well as cardiovascular diseases such as hypertension, heart failure, and coronary artery disease [16]. Moreover, it has been proved that lncRNAs participate in pathogenesis of certain neurological conditions such as Alzheimer’s disease, Parkinson’s disease, and lateral spinal cord sclerosis. It is correlated with the role of lncRNAs in synapse formation and neural cell functioning [17]. LncRNAs are also crucial factors in cancer development, the course of the disease and efficiency of treatment [18].
Due to the growing number of endometrial cancer cases, the search for individualized therapy that is less burdensome for patients is required. LncRNAs have broad and incompletely described effects on a number of metabolic processes. Due to the lack of a similar article, the topic of our review is the comprehensive description of the stimulatory and inhibitory effects of lncRNAs on the course of endometrial cancer. We took into account the effect of lncRNA both promoting and inhibiting the progression of endometrial cancer because in the future there is a chance to use drugs with different mechanisms of action.

2. Materials and Methods

A search for articles about the lncRNA in endometrial cancer was performed. The database of Pubmed was analyzed. The search was undertaken in April 2024. To find articles, the following keywords were used: “endometrial cancer”, “lncRNA”, ”long non-coding RNA”, “cancer of the uterine body”. A three-step analysis of found articles—title, abstract, and entire text—was undertaken. The inclusion criteria were as follows: (1) articles from the last 10 years; (2) manuscripts written in English; (3) original articles; (4) articles with access to the entire text; (5) studies on cells, animals, and humans.

3. LncRNA Mechanisms of Action

The action of lncRNAs is related to their influence on gene expression. The mechanisms of this action are complicated and still not fully understood. LncRNAs can regulate gene expression at multiple levels by interacting with RNA, DNA, and proteins. LncRNA mechanisms of action can be divided into three main groups: (1) regulation of gene expression at the transcription level by binding to DNA or transcription factors; (2) post-transcriptional action by interfering in the activities of mRNAs, miRNAs, or proteins; (3) epigenetic influence—activation or repression of gene expression by impact on chromatin complexes. LncRNAs have an ability to create hybrid structures with DNA (triple helices). It can change accessibility to chromatin and mediate gene silencing or activation [19,20,21]. The activation of proto-oncogene sphingosine kinase 1 (SPHK1) by lncRNA KHPS1 is an example of such a mechanism This molecule creates a triple helix with the SPHK1 enhancer to activate the transcription of SPHK1. Experimentally, KHPS1 was replaced by other lncRNA-MEG3. The correlation between SPHK1 and MEG3 did not activate this proto-oncogene [22].
LncRNAs are important gene expression regulators. They are located between genes, where they can have an impact on transcription. Antisense and bidirectional lncRNAs are particularly involved in these processes. The lncRNA transcript can regulate adjacent gene expression. Moreover, the transcription or splicing of the lncRNA can influence chromatin state and therethrough the expression of adjacent genes. LncRNAs, on the other hand, can suppress or activate polymerase II promotors, as well as change histone structure. Additionally, they can modify the enhancers of genes to activate or block expression [23,24,25,26]. LncRNAs can also function as post-transcriptional, translational, and post-translational regulators. Some lncRNAs are translated into peptides [27]. Long non-coding RNA can regulate protein functions by binding to specific RNA sequences and creating lncRNA–protein complexes. Another mechanism is to modify the splicing of pre-mRNA. It happens through alteration of splicing factors and splicing blocking by establishing connections with pre-mRNA. Moreover, LncRNAs can also make connections with structures that interact with certain proteins. This may cause the blocking or enhancement of the action [28,29,30]. LncRNAs can bind with other RNAs and activate proteins engaged in mRNA degeneration [31].
Finally, lncRNA has an impact on proteins located in the neighbourhood of chromatin where it can regulate DNA expression. These molecules are also connected with nuclear condensates—compartments engaged in cell functions [32]. On the other hand, lncRNA is a scaffolding particle. This means that this RNA creates architectural scaffolds for RNA and protein interactions in the nucleus and in the cytoplasm [33].
Some lncRNAs have special complementary sites to microRNAs (miRNAs) which, in turn, participate in the regulation of mRNA expression. Thanks to complementary fragments, lncRNA molecules combine with miRNA, which reduces the pool of free molecules necessary for the functioning of mRNA. This affects the regulation of all cellular processes [34].
In summary, the above description provides only a brief explanation of the mechanisms of lncRNAs’ action. The issue itself is broad and the mechanisms of action of lncRNAs are not yet fully understood. The functions of lncRNAs in the body are summarized in Table 2.

4. The Role of lncRNA in Endometrial Cancer

As mentioned earlier, lncRNAs influence the occurrence and the course of carcinogenesis. Individual long non-coding RNA molecules are associated with various cancers. Below there are described general characteristics of certain lncRNA molecules and their impact on the development, course, and treatment of endometrial cancer.

4.1. Long Non-Coding Antisense RNAs (-AS) Group

Antisense RNAs are the largest group of lncRNAs. It is a natural antisense transcript which contains single-stranded RNA that is complementary to messenger RNA (mRNA)—a protein-coding molecule. Antisense lncRNAs can have an impact on gene expression by blocking the translation of mRNA into proteins. Depending on their mechanism of action, antisense lncRNAs can stimulate or inhibit the development of endometrial cancer. The first example of an antisense lncRNA is NR2F1-AS1. It promotes the progression of endometrial cancer. This antisense lncRNA stimulates the migration and invasion of EC cells due to a decrease in microRNA-363 (miR-363) expression through the phosphoinositide 3-kinases/serine/threonine protein kinase/glycogen synthase kinase-3β (PI3K/AKT/GSK-3β) pathway. MiR-363 inhibits the expression of the SRY-related HMG-box 4 (SOX4) oncogene, which promotes tumour growth and metastasis [35]. In another study, LOXL1 antisense lncRNA has a similar impact on EC as it promotes the invasion and progression of cancer by downregulation expression of miR-28-5p. This microRNA decreases the expression of Ras-related protein Rap-1b gene (RAP1B), which is a part of the oncogene family. RAP1B stimulates the development of endometrial cancer [36]. MCM3AP-AS1 is another antisense lncRNA promoting EC development. This molecule stimulates the expression of the vascular endothelial growth factor (VEGF) gene. It engages in the formation of new blood vessels, which increases the blood supply to the tumour and promotes its development and metastasis. MCM3AP-AS1 works by blocking miR-126, which in turn blocks VEGF expression [37]. It has been shown that AFAP1-AS1 contributes to the progression of EC by promoting vascular endothelial growth factor A (VEGFA) expression. This gene increases the angiogenesis of tumour vessels. AFAP1-AS1 blocks miR-545-3p from acting [38]. NNT-AS1 is another antisense lncRNA that boosts the development of endometrial cancer. This lncRNA inhibits the expression of miR-30c-microRNA, which blocks EC progression. NNT-AS promotes further growth of estrogen-mediated uterine corpus cancer [39]. Interestingly, DLX6 antisense-1 stimulates the proliferation and invasion of endometrial cancer cells, simultaneously impeding apoptosis. It was proved that silencing of this lncRNA repressed the proliferation and invasion of EC cells. It is regulated by the interaction with p300/E2F1 acetyltransferase [40]. TTN-AS1 promotes uterine corpus endometrial cancer development. This molecule promotes the expression of pumilio homolog 2 (PUM2), which is a translational regulator. TTN-AS1 blocks miR-376a-3p expression, which is the factor that suppresses EC progression [41]. Moreover, GATA3-AS1 stimulates the invasion and migration of EC cells by blocking miR-361, and promotes the activation of Arrestin beta 2 gene (ARRB2), which, among others, is involved in cecum lymphoma occurrence. The GATA3-AS1/miR-361/ARRB2 axis was shown to regulate EC cell proliferation, invasion, and migration [42]. In another study, it was demonstrated that HOXB-AS3 also stimulates uterine corpus endometrial cancer by sponging miR-498-5p. This microRNA downregulates the expression of disintegrin and metalloproteinase domain-9 (ADAM-9). This enzyme level is increased in different cancers including EC [43]. Furthermore, HOXB-AS3 has an impact on lipid metabolism in EC by binding to polypyrimidine tract-binding protein 1 (PTBP1). It promotes supply of energy to growing tumour [44]. Another antisense lncRNA HOXA-AS2 promotes the invasion and proliferation of endometrial cancer type 1 by silencing miRNA-302c-3p and promoting the expression of zinc finger X-chromosomal protein (ZFX). This protein stimulates a growth and migration of cancer cells [45]. The next lncRNA HOXC-AS2 advertises EC development by blocking miR-876-5p. It decreases the expression of hexokinase domain-containing 1 (HKDC1). This enzyme provides a supply of glucose for tumour development, and promotes the production of reactive oxygen species. These forms of oxygen have an impact on pyroptosis, which is a form of programmed cell death and which plays a crucial role in a wide range of diseases [46]. The FIGO stage, histological grade, and lymph node metastasis are correlated with another long non-coding RNA-RHPN1-AS1. It reacts with mitogen-activated protein kinases (MAPKs) and extracellular signal-regulated kinases (ERKs)—the MAPK/ERK pathway [47]. Another mechanism of antisense lncRNA action is up-regulating cyclin D1, a protein that alters the cell cycle and promotes tumorigenesis. The regulation of cyclin D1 is associated with the presence of lncRNA ABHD11-AS1 [48]. LncRNA DCST1-AS1 enhances tumour growth, migration, and invasion. This lncRNA induces the expression of cell adhesion molecule 1 (CADM1) and homeobox B5 (HOXB5) by miR-873-5p and miR-665. Increased expression of HOXB5 and CADM1 is correlated with tumour development [49]. The overexpression of another lncRNA-DSCAM-AS1 is associated with higher expression levels of the prolactin gene (PRL) and estrogen receptor α (Erα), a more aggressive course of EC, and a shorter survival period of patients. The oncogenic role of DSCAM-AS1 is correlated with the blocking of miR-136-5p actions [50,51]. In other research, it has been shown that VPS9D1-AS1 can contribute to EC progression by binding to miR-520a-5p and promoting the expression of Baculoviral IAP Repeat-Containing 5 (BIRC5). The protein coded by this gene inhibits the apoptosis of endometrial cancer cells. VPS9D1-AS1 regulates the expression by sponging miR-377-3p and stimulates the glucocorticoid-regulated kinase 1 (SGK1) activity. It correlates with lymph node metastasis and the FIGO stage. SGK1 is engaged in cellular stress response and cell survival [52,53]. Other examples of antisense lncRNAs that promote EC development are BMPR1B-AS1, which sponges miR-7-2-3p and modulates the DCLK1/Akt/NF-κB pathway, and ZFAS. This lncRNA promotes higher expression of cyclin D1, ZNRD1-AS1 and NIFK-AS1 [54,55,56]. It has been shown that antisense lncRNAs can promote EC development by reducing the effectiveness of chemotherapy., whereas LncRNA TMPO-AS1 promotes glucose transporter-1 (GLUT1)-dependent glycolysis and resistance to paclitaxel therapies. Paclitaxel is one of the cytostatics that is used in the treatment of different cancers including endometrial cancer. GLUT1 stimulates the proliferation and invasion of endometrial cancer cells. TMPO-AS1 elevated GLUT1 expression by directly binding to two tumour suppressors, microRNAs-miR-140 and miR-143. The downregulation of TMPO-AS1 significantly inhibited EC proliferation and reduced paclitaxel resistance [57]. Moreover, CDKN2B-AS1 has a similar effect and it increases resistance to paclitaxel by blocking the expression of miR-125a-5p [58]. The promotion of endometrial cancer cell proliferation and its migration and paclitaxel resistance is correlated with the higher expression of another antisense lncRNA, FGD5-AS1. Moreover, FGD5-AS1 impedes immune escape via the PD-1/PD-L1 checkpoint [59].
Some antisense lncRNAs have inhibitory effects on the development of endometrial cancer. This is related to various molecular mechanisms. The first example is FRMD6-AS2. This lncRNA suppresses tumour growth, migration, and invasion by the activation of Hippo signalling. The Hippo pathway regulates cell proliferation, and its activation suppresses this process. FRMD6-AS2 has also been shown to have an impact on the function of contraction and actin-mediated cell movement [60]. It has been shown that MTCP1-AS1 is another lncRNA that has a negative impact on EC cell proliferation, migration, invasion, and epithelial to mesenchymal transition. This lncRNA inhibits miR-650 and promotes the expression of mothers against decapentaplegic homolog 7 (SMAD-7). It is a member of the group which is engaged in leukocytes signals transfer to cells. SMADs are correlated with tumour growth suppression [61]. LncRNA can also inhibit EC progression by stimulating the apoptosis of endometrial cancer cells. The example is EIF1AX-AS1. The mechanism of this action is that EIF1AX-AS1 mediates the eukaryotic translation initiation factor 1A X-linked (EIF1AX) mRNA instability by binding to poly C binding protein 1 (PCBP1). The instability of this mRNA results in the dysfunction of EC cells [62]. Moreover, lncRNA can increase the degradation of Aurora kinase A (AURKA), which is involved in cell proliferation. This mechanism is related to lncRNA SOCS2-AS1. This molecule suppresses EC cell proliferation and promotes cell-cycle arrest, as well as apoptosis [63,64]. In another study, it was proved that GATA6-AS inhibited EC progression by downregulation of metalloproteinase 9 (MMP-9) expression. This protein promotes metastasis and angiogenesis due to the decomposition of the extracellular matrix [65]. In the study by Liu et al., it was proved that OIP5-AS1 inhibited EC progression by blocking miR-200c-3p and controlled the phosphatase and tensin homolog (PTEN/AKT) pathway. PTEN is a tumour suppressor gene and participates in cell cycle, angiogenesis, migration, and invasion [66]. LncRNA inhibits the migration and invasion of EC cells by decreasing expression of neuromedin U (NMU). It is proved that this neuropeptide promotes endometrial cancer development. This is how another lncRNA HAND2-AS1 works [67]. Apart from these all, antisense lncRNAs can also mitigate EC progression by the stimulation of the immune system activity. They may exert such an effect through various mechanisms, including stimulating the activity of macrophages and other leukocytes, signalling pathways, or cytokine secretion [68,69]. The main characteristics of long non-coding antisense RNAs are summarized in Table 3.

4.2. Long Intergenic Non-Coding RNA (LINC)

Long intergenic non-coding RNA is one kind of lncRNA involved in various cellular functions, including the regulation of gene expression and chromatin remodelling. The abnormal expression of lincRNAs can induce or suppress EC development.
One of the lincs that is important in the development of EC is linc02936. It promotes EC progression by inhibiting ferroptosis—the process of programmed cell death dependent on iron and mediated by enzyme ceruloplasmin. Linc02936 upregulates the expression of ceruloplasmin by binding to SIX Homeobox 1 (SIX1) [70]. Another lncRNA is linc01016. It stimulates EC progression by inhibiting miR-302a-3p and miR-3130-3p. These microRNAs block the expression of nuclear factor YA (NFYA), which promotes endometrial cancer development [71]. Furthermore, another molecule, linc01410, stimulates the expression of chromodomain helicase DNA-binding protein 7 (CHD7) and inhibits miR-23c. This protein engages in promoting the angiogenesis of the tumour. CHD7 is impeded by miR-23c. Hence, linc01410 promotes EC progression [72]. In another study, it was proved that linc01857 was engaged in the EC course as well. The decreased expression of this lncRNA promotes EC cell apoptosis and blocks proliferation and migration. It stabilizes proto-oncogene MYCN, as well as inhibits miR-19b-3p expression, which has a negative impact on MYCN expression [73]. Moreover, lincs can promote EC progression by binding to MYC and deactivation of PTEN. This is a potential mechanism of action of inc00470 [74]. Apart from all this, linc01194 stimulates EC progression by connecting to insulin-like growth factor 2 binding protein 1 (IGF2BP1). This factor promotes the expression of sex-determining region Y-box 2 (SOX2). This gene increases tumour development and promotes metastasis occurrence [75]. Linc can either promote endometroid cancer proliferation by stimulating overexpression of VEGFA, and thus increases the angiogenesis of tumour vessels. It is correlated with linc01541 [76]. Linc00958 has a comprehensive impact on EC development. This lncRNA promotes insulin-like growth factor mRNA-binding protein 3 (IGF2BP3). This protein, in turn, upregulates the expression of E2F3—a gene that increases EC cell proliferation and tumour growth. Moreover, linc00958 can upregulate PHD Finger Protein 6 (PHF6) by impeding miR-3174 activity. PHF6 has an oncogenic function in EC. Linc00958 may also inhibit miR-145-3p expression, and as a result, it leads to an increase in transcription factor 4 (TCF-4) activity, which causes EC progression [77,78,79]. Furthermore, linc01224 and linc01170 bind to and downregulate miR-485-5p. This results in an increase in threonine-protein kinase (AKT3) expression, which leads to EC progression and metastasis [80,81]. In another study, linc01220 generates the overexpression of mitogen-activated protein kinase 11 (MAPK11). This kinase increases the proliferation of EC cells and stimulates the progression of the disease [82]. Another oncogenic long intergenic non-coding RNA is linc00461. This lincRNA upregulates the expression of cyclooxygenase-2 (COX-2) by decreasing the expression of miR-219-5p, for which COX-2 is a target. COX-2 promotes tumour growth and EC progression, as well as decreases its immune system activity [83]. The last lincRNA to mention that promotes EC progression is linc01106. This molecule inhibits the activity of miR-449a and upregulates the expression of proto-oncogene MET, which has a negative impact on EC course [84].
On the other hand, there is a group of lincRNAs that inhibit the development of endometrial cancer. Linc00672 suppresses the progression of EC by two mechanisms. Firstly, this lncRNA promotes the p53-induced downregulation of LIM and SH3 Protein 1 (LASP1). This protein induces cytoskeleton alteration, the progression of cancer, and metastasis. Secondly, ling00672 increases chemosensitivity to paclitaxel [85]. Forkhead box protein O1 (FOXO1) inhibits endometrial cancer growth by suppressing angiogenesis and promoting apoptosis. FOXO1 is upregulated by linc00261 and downregulated by miR-182, whose expression is impeded by linc00261 [86]. Polypyrimidine tract binding protein 1 (PTBP1) has a negative impact on prognosis of EC by promoting the formation of pseudopodia, which are engaged in the invasion and occurrence of metastasis. Linc00478 inhibits EC progression by downregulating PTBP1 expression [87]. The last linc to mention here is linc01589. It suppresses EC development by the stimulation of immune system activation due to the increasing expression of B cells, T cells, and NK cells [88]. The main characteristics of long intergenic non-coding RNA are summarized in Table 4.

4.3. Maternally Expressed Gene 3 (MEG3)

There is also a group of specific lncRNAs which are of particular interest to scientists. They have an impact on the course of different cancers. The first example, maternally expressed gene 3 (MEG3), is an intergenic lncRNA that participates in the course of endometrial cancer. It is expressed in a wide range of tissues and regulates gene activity. MEG3 is considered as a tumour suppressor in cancers including EC. MEG3 is downregulated in uterine corpus endometrial cancer cells. Its effect in blocking EC development may be related to several mechanisms. MEG3 increases the expression of programmed death-ligand-1 (PD-L1) which acts as a tumour suppressor, and its levels are downregulated in EC cells. PDL-L1 downregulation is correlated with an aggressive course of cancer and poor prognosis. PD-L1 has an anti-tumour activity through inhibiting myeloid cell leukemia-1 (MCL1) expression. MCL1 promotes cell survival and the epithelial–mesenchymal transition. This process induces invasion and promotes metastasis occurrence. PD-L1 expression is downregulated by miR-216a. MEG3 stimulates PD-L1 expression by inhibiting activation of miR-216a. Hence, MEG3 suppresses cells migration and invasion [89]. Another potential mechanism of MEG3 action is the downregulating activity of phosphoinositide 3-kinases/mammalian target of rapamycin (PI3K/m-TOR) signalling. PI3K/m-TOR dysfunction is correlated with carcinogenesis—it promotes progression and invasion, and inhibits apoptosis [90]. Moreover, MEG3 can downregulate the activity of neurogenic locus notch homolog protein 1 (Notch 1) and hairy and enhancer of split-1 (HES-1). Both proteins are engaged in cancer progression by promoting EC cell proliferation and metastasis, and inhibiting apoptosis [91]. Interestingly, lncRNAs can influence each other’s expression. An example is the downregulation of MEG3 expression by another lncRNA, PSMG3-AS1. MEG3 inhibits EC progression, while PSMG3-AS1 promotes the invasion of this cancer [92].

4.4. Homeobox (HOX) Transcript Antisense Intergenic RNA (HOTAIR)

Another well-known lncRNA molecule, the role of which was investigated in several types of cancers including breast, lung, and liver, is homeobox (HOX) transcript antisense intergenic RNA (HOTAIR). It can regulate the expression of homeobox cluster genes. HOTAIR is upregulated in a wide range of malignancies, and it is correlated with overall survival and progression-free survival. In research conducted by He et al., HOTAIR expression was higher among patients with EC than in the control group. The expression of HOTAIR was correlated with EC grade and lymph node metastasis. It was also correlated with a depth of myometrial invasion and lymphovascular space invasion. Moreover, the higher expression of HOTAIR indicated a poorer overall survival ratio [93]. HOTAIR expression can be upregulated by estrogen, which promotes EC cell migration. Thus, the estrogen–HOTAIR axis is a potential target in EC therapy [94]. In other research, it was proved that HOTAIR expression was higher in G3 EC than in G1/G2 [95]. Similar actions exhibit long non-coding antisense RNA myeloid-specific 1 (HOTAIRM1). In EC, HOTAIRM1 expression is correlated with the FIGO stage and lymph node metastasis. The downregulation of this lncRNA expression leads to the inhibition of cell proliferation, migration, and invasion and to the epithelial–mesenchymal transition [96]. The HOTAIR mechanism of action is still unclear. However, in EC, it acts as an oncogene by the activation of PI3K/Akt pathway and suppression of PTEN expression [97]. Estrogen-induced HOTAIR expression can be regulated by a variety of factors. One example is miR-646. This microRNA inhibits the expression of HOTAIR in EC [98]. In the research by Łuczak et al., it was proved that there were no differences in HOTAIR expression between different subtypes of EC. A higher expression of HOTAIR was correlated with a shorter overall survival [99]. The overexpression of HOTAIR in EC may also affect a conservative treatment with progesterone. In a study by Chi et al., HOTAIR expression has been shown to be inversely correlated with progesterone receptor B (PRB) expression. The knockdown of HOTAIR can promote medroxyprogesterone sensitivity by upregulating PRB. Hence, the downregulation of HOTAIR can be a potential therapeutic target to overcoming progesterone resistance [100]. The downregulation of HOTAIR by lentivirus-mediated RNA interference in EC cells was correlated with significant suppression of migration, invasion, and cell proliferation. Moreover, it caused cell cycle arrest at the G0/G1 phase. A decline in HOTAIR expression resulted with a decrease in tumorigenesis [101].

4.5. H19

Another lncRNA is H19. It is an lncRNA located in 11 chromosomes in the neighbourhood with the insulin-like growth factor II (IGF-2) gene. It is one of the major oncogenic genes and is engaged in the development of a wide range of tumours, including endometrial cancer. H19 participates in carcinogenesis at various stages of disease development and through various mechanisms. According to Lottin et al., H19 is expressed in stromal and myometrial cells of uterus. A high expression is observed in the vicinity of malignant epithelial cells. The expression is correlated with the invasion of cancer cells into the myometrium and disease progression in general [102]. It may be a result of promoting the epithelial–mesenchymal transition by H19 [103]. Moreover, it can promote homeobox A10 (HOXA10) expression by inhibiting miR-612. HOXA10 is engaged in tumour growth and metastasis occurrence, as well as mediating the epithelial–mesenchymal transition. MiR-612 impedes EC progression by blocking HOXA10 expression [104]. Interestingly, H19 levels in EC can be reduced using metformin [105].

4.6. Nuclear-Enriched Abundant Transcript 1 (NEAT1)

Another lncRNA that is upregulated in EC and accelerates malignant growth and metastasis is nuclear-enriched abundant transcript 1 (NEAT1). According to existing research, it is proved that NEAT1 is highly expressed in EC cells. A decrease in NEAT1 expression results in the suppression of proliferation, migration, and invasion. NEAT1 downregulates the expression of miR-144-3p, and that leads to the upregulation of Enhancer of zester homolog 2 (EZH2). EZH2 inhibits the transcription of tumour suppressor genes [106]. High NEAT1 expression is correlated with poor prognosis. It can also downregulate miR-361, the particle that is engaged in the suppression of prometastatic gene expression. NEAT1 also promotes the expression of oncogene signal transducer and activator of transcription 3 (STAT3) by blocking miR-26a activity [107,108]. NEAT1 activity can be positively correlated with the FIGO stage and lymph node metastasis. It increases the migration and invasion ability. NEAT1 increases expression of MMP-2, MMP-7, and the insulin-like growth factor 1 (IGF1) [109]. NEAT1 can also suppress miR-214-3p and promote the expression of high-mobility group protein (HMGA1) to regulate the Wnt/β-catenin pathway and stimulate EC progression [110]. Finally, NEAT1 can impede the expression of miR-202-3p and promote T cell immunoglobulin and mucin domain 4 (TIMD4). TIMD4 promotes the progression of EC and reduces the time free from progression [111].

4.7. Small Nucleolar Host Genes (SNHG)

Another group of lncRNAs that take part in course of endometrial cancer are small nucleolar host genes (SNHGs). They are a group of lncRNAs that are overexpressed in many cancers. In general, they are involved in tumour development—SNHGs increase the proliferation, cell cycle progression, invasion, and metastasis of cancer cells. SNHGs are created from both introns and exons. They can exert impact on a wide range of cells functions at the DNA/RNA/protein level. The first example of molecule from this group is SNHG9. It promotes EC cell proliferation and glycolysis, processes that generate glucose—a source of energy for tumour development [112]. They can also stimulate the development of EC by upregulating the levels of matrix metalloproteinase MMP2 and MMP9. These enzymes promote invasion and metastasis. This is correlated with SNHG12. SNHG12 expression can be upregulated by the activity of zinc family member 2 (ZIC2). This leads to the promotion of EC cell proliferation and migration by activating the Notch signalling pathway. On the other hand, SNGH12 expression can be suppressed by miR-4429 [113,114]. On the other hand, the next small nucleolar host gene—SNHG14—acts as an obstructive factor in EC. In previous research, it was proved that the overexpression of SNGH14 hampered the migration, invasion, and viability of endometrial cancer cells. This lncRNA inhibits miR-93-5p and upregulates zinc finger and BTB domain-containing protein 7A (ZBTB7A)—a factor that acts a tumour suppressor [115]. However, the role of SNHG14 in endometrial cancer is not entirely clear. According to other research, SNHG14 can promote the proliferation of EC cells. High expression of SNHG14 was correlated with a larger tumour size, shorter overall survival, and a more advanced pathological stage. The expression of SNHG14 was downregulated by miR-655-3P [116]. Other SNHGs—16 and 25—promote the proliferation and malignancy of EC. SNHG16 stimulates hexokinase 2 (HX2) expression by blocking miR-490-3p. HX2 increases glycolysis and endometrial carcinoma proliferation [117]. Higher expression of SNHG25 is correlated with shorter overall survival rates among patients with EC. SNHG25 upregulates the level of fatty acid synthase (FASN), which plays the role of an oncogene and impedes the expression of microRNA-497-5p [118]. The main characteristics of small nucleolar host genes are summarized in Table 5.

4.8. LncRNA-Associated Transcripts (ATs)

LncRNA-associated transcripts are a group of lncRNAs that are correlated with several types of tumours. The example of this kind of lncRNA is colon cancer-associated transcript 1 (CCAT1). CCAT1 acts as a promotor of EC progression, and it stimulates proliferation and migration. This activity is correlated with the downregulation of miR-181a-5p [119]. In other research, CCAT1 expression was 9.3-fold higher in endometrial cancer compared to normal endometrium, and the inhibition of this lncRNA expression was correlated with a significant proliferation reduction [120]. Another lncRNA AT that is engaged with the EC course is prostate cancer-associated transcript 1 (PCAT1). This lncRNA expression is positively correlated with myometrial invasion, the FIGO stage, lymph node metastasis, and a shorter overall survival. Overexpression of PCAT1 is relevant to changes in the molecular level of different molecules [121]. PCAT1 promotes the downregulation of E-cadherin in correlation with EZH2. E-cadherin downregulation is associated with an increase in the epithelial–mesenchymal transition (EMT). It stimulates the progression and metastasis of EC [122]. Another lncRNA that promotes EC development is retinoblastoma-associated transcript-1 (RBAT1). This molecule increases cell viability and suppresses cell apoptosis. Moreover, RBAT1 reduces the chemosensitivity of EC cells to carboplatin/paclitaxel. A potential mechanism is correlated with promoting the expression of multidrug-resistant-related protein and the inhibition of miR-27b [123]. The last LncRNA in this group is bladder cancer-associated transcript 2 (BLACAT2). High expression of this molecule is correlated with migration and invasion of EC cells. It also promotes proliferation. BLACAT2 suppresses miR-378a-3p activity, and as a result, it induces mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK/ERK) pathway. It is correlated with tumour development [124]. The main characteristics of lncRNA-associated transcripts are summarized in Table 6.

4.9. Other Long Non-Coding RNAs Associated with Endometrial Cancer

In addition to the above, there are also other lncRNAs that influence the course of endometrial cancer, and that cannot be assigned to a specific group. Some of them stimulate tumour development, while others may inhibit it. The number of lncRNAs among which associations with EC are being discovered is constantly growing.

4.10. LncRNAs That Promote Endometrial Cancer Progression

Another group of lncRNAs includes a variety of molecules. Most of them stimulate the development of endometrial cancer. LncRNA deleted in lymphocytic leukaemia 1 (DLEU1) is an example of a factor which promotes EC development. DLEU1 increases viability, migration, and invasion, and reduces the proportion of apoptosis. It is correlated with an increase in PI3K/AKT/mTOR pathway expression. DLEU1 also decreases the activity of miR-490 [125,126]. The DLEU1 level can be considered as an independent prognostic factor in EC [127]. Another example of a molecule from this group is DLEU2. It can promote EC progression by upregulating hexokinase 2 (HK2) expression. It leads to intensified glycolysis and epithelial–mesenchymal transition [128]. Another lncRNA whose high expression is correlated with a high FIGO stage and poor tumour differentiation is LncRNA-ATB. It is inhibited by miR-126 [129]. Furthermore, lncRNA that promotes EC development by the upregulation of the proliferation of cells is BANCR. The expression of this molecule is positively correlated with ERK/MAPK signalling pathway activation and the production of MMP2 and MMP1. These molecules stimulate invasion and metastasis [130]. Moreover, lncRNA PVT1 stimulates cell proliferation, migration, and invasion, and blocks apoptosis. PVT1 inhibits the miR-195-5p activity. It causes the upregulation of acidic fibroblast growth factor receptor (FGFR1) and basic fibroblast growth factor (FGF2). Both factors activate the PI3K/AKT and MAPK/Erk pathways, which promote EC progression. PVT1 can also stimulate the expression of Centromere Protein H (CENP-H) by impeding miR-612 [131,132]. LncRNA XIST participates in EC growth by stimulating the expression of Centromere coiled-coil protein 110 (CCP110), a protein that stimulates migration, invasion, and proliferation. XIST blocks the activity of miR-129-2-3p, a factor that impedes CCP110 expression [133]. Another lncRNA is NDRG1. It promotes viability, migration, and invasion. NDRG1 stimulates neovascularization by upregulating the expression of VEGFA [134]. The next lncRNA that upregulates the epithelial–mesenchymal transition is MIR210HG. It is correlated with tumorigenesis, metastasis, and drug resistance. This lncRNA inhibits miR-337-3p and promotes the Wnt/β-catenin and TGF-β/Smad3 signalling pathways. These pathways engage in the epithelial–mesenchymal transition [135]. LSINCT5 also activates the Wnt/β-catenin pathway by stabilization of High-mobility group AT-hook 2 (HMGA2) [136]. Another lncRNA is ROR, which promotes the expression of the Notch1 protein, and causes the upregulation of proliferation and downregulation of apoptosis in EC [137]. LncRNA FIRRE promotes endometrial cancer development and reduces radiotherapy sensitivity. FIRRE downregulates the expression of miR-199b-5p. This microRNA blocks the activity of sirtuin 1 (SIRT1), which takes part in the FIRRE-mediated stimulation of the autophagy process. Moreover, FIRRE increases the viability and proliferation of EC cells [138]. Another lncRNA molecule which promotes cell proliferation, migration, and invasion in endometrial cancer is LINP1. It can activate PI3K/AKT signalling and promotes tumour invasion [139]. In another study, it was proved that the high expression of lncRNA RNA (THOR) was correlated with a poor overall survival rate. THOR promotes the proliferation, invasion, and migration of EC cells. It is in relation to the upregulation of the ERK/AKT pathway [140]. Furthermore, PCGEM1 is upregulated in endometrial cancer. This lncRNA acts as a tumour development promotor. PCGEM1 inhibits the expression of miR-129-5p, as well as stimulates STAT3 activity [141]. Another lncRNA that participates in EC growth via the MAPK pathway activation is HEIH. It leads to the promotion of chemo-resistance in endometrial cancer cells and enhances cell proliferation and viability [142]. Another lncRNA that promotes EC growth via increasing the epithelial–mesenchymal transition is SLERT. It elevates vimentin and N-cadherin expression and downregulates E-cadherin [143]. TUG1 demonstrates a similar mechanism of action, and it also favours the epithelial–mesenchymal transition due to increasing the levels of E-cadherin and a decrease in N-cadherin. The overexpression of this molecule is correlated with lower survival rates among patients with EC and with lymph node metastasis [144]. TUG1 acts as a suppressor of miR-299 and miR-34a-5p expression [145]. The last one lncRNA to mention here has a diverse impact on EC development. It is UCA1. It promotes cell proliferation, colony formation, invasion, and lymph node metastasis, and inhibits apoptosis. UCA1 overexpression is correlated with a higher malignancy of EC. A decrease in UCA1 expression leads to the suppression of tumour growth and cancer progression. This lncRNA inhibits miR-204-5p and promotes Kruppel-like factor 5 (KLF5) and relaxin-like family peptide receptor 1 (RXFP1). These factors participate in metastasis formation [146,147,148].

4.11. lncRNAs That Suppress Endometrial Cancer Progression

In addition to lncRNAs that stimulate the development of endometrial cancer, there are also those that block its growth. They act through various molecular mechanisms and influence the proliferation, migration, and invasion of cancer cells. The first lncRNA FER1L4 inhibits EC progression by the upregulation of PTEN and inhibition of Akt activation. It leads to a decline in proliferation, as well as an increase in apoptosis [149]. Lnc-NA stimulates the apoptosis of EC by activation of the nuclear receptor subfamily 4 group A member 1 (NR4A1) gene. It leads to the activation of the apoptosis signalling pathway to inhibit tumour progression [150]. Another LncRNA, MONC, impedes endometrial cancer tumour development, as well as migration and invasion. MONC decreases the expression of miR-636 and impedes the epithelial-to-mesenchymal transition process. The downregulation of miR-636 is correlated with the upregulation of Glucuronic Acid Epimerase (GLCE), which inhibits the progression of EC [151]. In another study, LncRNA TUSC7 inhibits cell proliferation, cycle progression, and metastasis. TUSC7 acts as a suppressor of miR-616, which is a factor that blocks the expression of suppressor of cytokine signalling 4 (SOCS4). SOCS4 inhibits the angiogenesis and development of tumour. Moreover, TUSC7 can block an activity of miR-23b, and as a result, it increases the sensitivity to chemotherapy based on cisplatin and paclitaxel [64,152,153]. Another lncRNA that impedes EC progression is NORAD. The downregulation of this molecule is correlated with EC progression (FIGO stage) and poor outcome. NORAD stimulates the activity of pro-apoptotic Far Upstream Element-Binding Protein 1 (FUBP1). It leads to the apoptosis of EC cells [154]. The next lncRNA that inhibits EC development is ZXF1. It impedes the migration and invasion of EC, as well as regulates cell cycle and proliferation. ZXF1 can stimulate the expression of cyclin-dependent kinase inhibitor 1A (P21) via inhibiting the miR-378a-3p- and CDC20-mediated degradation of P21. P21 participates in cell cycle arrest and the stimulation of cell apoptosis [155]. Another lncRNA, FENDDR, suppresses cell proliferation and stimulates apoptosis by the downregulation of SOX4 expression [156]. LncRNA can also block EC cells in the G1 phase. An example of this is XLEC1. It inhibits the proliferation and migration of EC cells. XLEC1 blocks cells in the G1 phase via Myc Promoter-Binding Protein-1 (MBP-1) activation and the suppression of c-Myc expression. C-myc acts as an oncogene that participates in cellular growth [157]. Furthermore, lncRNA GAS5 functions as a tumour development suppressor by the upregulation of cancer cell apoptosis. GAS5 enhances the expression of PTEN via the suppression of miR-103 activity. Moreover, GAS5 inhibits EC progression by regulating the immune system. GAS5 promotes phagocytosis, antigen presentation, and the activation of cytotoxic T cells. It changes the phenotype of tumour-associated macrophages from a pro-tumour to an anti-tumour [158,159]. The last lncRNA to mention that suppresses EC growth is lncRNA-LA16C-313D11.11. It also acts as a promotor of PTEN expression. LA16C-313D11.11 inhibits the activity of microRNA-205-5p—a factor that blocks PTEN [160]. The main characteristics of other lncRNAs are summarized in Table 7.

5. Conclusions

In summary, various lncRNAs may influence the course of endometrial cancer. Most of them stimulate tumour growth by increasing proliferation, migration, and invasion while reducing apoptosis. However, some of them inhibit the progression of endometrial cancer by blocking the cell cycle, proliferation, migration, and invasion due to blocking the epithelial–mesenchymal transition. They can also induce the apoptosis of cancer cells. LncRNAs exert their effects through a variety of molecular mechanisms. They stimulate or inhibit the activity of molecular pathways, proteins, and enzymes. They also influence the expression of several microRNA molecules. In the future, selected lncRNAs may be used as prognostic factors determining the clinical course of the disease, the degree of cancer malignancy, and the prognosis for patients. They may also be considered when selecting the optimal therapy. Some of them can become direct therapeutic targets. The influence of drugs on the inhibition or stimulation of lncRNAs may affect the course of the disease and improve the prognosis and overall survival rate. LncRNAs are a part of the modern approach to the development and a course of cancer diseases as a set of genetic and molecular disorders encompassing a number of factors. Further research should focus on isolating panels of lncRNAs or single molecules that would constitute prognostic factors for the course of endometrial cancer. We should also look for drugs that, by acting on selected long non-coding RNAs, would inhibit the progression or recurrence of the cancer. The literature data indicate that changing the expression of selected lncRNAs may be important for tumour development, invasion, and metastasis. Potential therapeutic targets among lncRNAs may include, in particular, molecules such as MEG3 or HOTAIR. This is related to their comprehensive impact on oncogenesis, studied in many cancers. The first research on the use of these molecules is currently being conducted.

Author Contributions

Conceptualization, P.J., T.K. and I.Z.; methodology, P.J., T.K., S.P. and N.P.; software, A.K. (Adrianna Korzystka), A.W. and M.M.; validation, P.J., A.B., N.P., S.P. and I.Z.; formal analysis, I.Z. and T.K.; investigation, P.J., M.M., A.K. (Aleksandra Kmieć) and A.W.; resources, P.J., A.B., N.P. and S.P.; data curation, P.J., A.K. (Adrianna Korzystka), A.K. (Aleksandra Kmieć), A.W. and M.M.; writing—original draft preparation, P.J., I.Z., N.P., S.P. and A.B.; writing—review and editing, T.K., I.Z., M.M., A.K. (Adrianna Korzystka), A.K. (Aleksandra Kmieć) and A.W.; visualization, P.J., I.Z., N.P. and S.P.; supervision, I.Z. and T.K.; project administration, P.J., I.Z. and T.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Onstad, M.A.; Schmandt, R.E.; Lu, K.H. Addressing the Role of Obesity in Endometrial Cancer Risk, Prevention, and Treatment. J. Clin. Oncol. 2016, 34, 4225–4230. [Google Scholar] [CrossRef] [PubMed]
  3. Crosbie, E.J.; Kitson, S.J.; McAlpine, J.N.; Mukhopadhyay, A.; Powell, M.E.; Singh, N. Endometrial cancer. Lancet 2022, 399, 1412–1428. [Google Scholar] [CrossRef] [PubMed]
  4. Bokhman, J.V. Two pathogenetic types of endometrial carcinoma. Gynecol. Oncol. 1983, 15, 10–17. [Google Scholar] [CrossRef] [PubMed]
  5. Makker, V.; MacKay, H.; Ray-Coquard, I.; Levine, D.A.; Westin, S.N.; Aoki, D.; Oaknin, A. Endometrial cancer. Nat. Rev. Dis. Primers 2021, 7, 88. [Google Scholar] [CrossRef] [PubMed]
  6. Yen, T.T.; Wang, T.L.; Fader, A.N.; Shih, I.M.; Gaillard, S. Molecular Classification and Emerging Targeted Therapy in Endometrial Cancer. Int. J. Gynecol. Pathol. 2020, 39, 26–35. [Google Scholar] [CrossRef] [PubMed]
  7. Cancer Genome Atlas Research, N.; Kandoth, C.; Schultz, N.; Cherniack, A.D.; Akbani, R.; Liu, Y.; Shen, H.; Robertson, A.G.; Pashtan, I.; Shen, R.; et al. Integrated genomic characterization of endometrial carcinoma. Nature 2013, 497, 67–73. [Google Scholar] [CrossRef] [PubMed]
  8. Choi, S.W.; Kim, H.W.; Nam, J.W. The small peptide world in long noncoding RNAs. Brief. Bioinform. 2019, 20, 1853–1864. [Google Scholar] [CrossRef]
  9. Li, K.; Ramchandran, R. Natural antisense transcript: A concomitant engagement with protein-coding transcript. Oncotarget 2010, 1, 447–452. [Google Scholar] [CrossRef]
  10. Fernandes, J.C.R.; Acuna, S.M.; Aoki, J.I.; Floeter-Winter, L.M.; Muxel, S.M. Long Non-Coding RNAs in the Regulation of Gene Expression: Physiology and Disease. Noncoding RNA 2019, 5, 17. [Google Scholar] [CrossRef]
  11. Guttman, M.; Amit, I.; Garber, M.; French, C.; Lin, M.F.; Feldser, D.; Huarte, M.; Zuk, O.; Carey, B.W.; Cassady, J.P.; et al. Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature 2009, 458, 223–227. [Google Scholar] [CrossRef]
  12. Quinn, J.J.; Chang, H.Y. Unique features of long non-coding RNA biogenesis and function. Nat. Rev. Genet. 2016, 17, 47–62. [Google Scholar] [CrossRef] [PubMed]
  13. Statello, L.; Guo, C.J.; Chen, L.L.; Huarte, M. Author Correction: Gene regulation by long non-coding RNAs and its biological functions. Nat. Rev. Mol. Cell Biol. 2021, 22, 159. [Google Scholar] [CrossRef]
  14. Mattick, J.S.; Amaral, P.P.; Carninci, P.; Carpenter, S.; Chang, H.Y.; Chen, L.L.; Chen, R.; Dean, C.; Dinger, M.E.; Fitzgerald, K.A.; et al. Long non-coding RNAs: Definitions, functions, challenges and recommendations. Nat. Rev. Mol. Cell Biol. 2023, 24, 430–447. [Google Scholar] [CrossRef] [PubMed]
  15. Pisignano, G.; Ladomery, M. Epigenetic Regulation of Alternative Splicing: How LncRNAs Tailor the Message. Noncoding RNA 2021, 7, 21. [Google Scholar] [CrossRef] [PubMed]
  16. Cai, B.; Zhang, Y.; Zhao, Y.; Wang, J.; Li, T.; Zhang, Y.; Jiang, Y.; Jin, X.; Xue, G.; Li, P.; et al. Long Noncoding RNA-DACH1 (Dachshund Homolog 1) Regulates Cardiac Function by Inhibiting SERCA2a (Sarcoplasmic Reticulum Calcium ATPase 2a). Hypertension 2019, 74, 833–842. [Google Scholar] [CrossRef]
  17. Li, L.; Zhuang, Y.; Zhao, X.; Li, X. Long Non-coding RNA in Neuronal Development and Neurological Disorders. Front. Genet. 2018, 9, 744. [Google Scholar] [CrossRef]
  18. Qian, Y.; Shi, L.; Luo, Z. Long Non-coding RNAs in Cancer: Implications for Diagnosis, Prognosis, and Therapy. Front. Med. 2020, 7, 612393. [Google Scholar] [CrossRef]
  19. Chen, Y.; Li, Z.; Chen, X.; Zhang, S. Long non-coding RNAs: From disease code to drug role. Acta Pharm. Sin. B 2021, 11, 340–354. [Google Scholar] [CrossRef]
  20. Schmitz, K.M.; Mayer, C.; Postepska, A.; Grummt, I. Interaction of noncoding RNA with the rDNA promoter mediates recruitment of DNMT3b and silencing of rRNA genes. Genes. Dev. 2010, 24, 2264–2269. [Google Scholar] [CrossRef]
  21. O’Leary, V.B.; Ovsepian, S.V.; Carrascosa, L.G.; Buske, F.A.; Radulovic, V.; Niyazi, M.; Moertl, S.; Trau, M.; Atkinson, M.J.; Anastasov, N. PARTICLE, a Triplex-Forming Long ncRNA, Regulates Locus-Specific Methylation in Response to Low-Dose Irradiation. Cell Rep. 2015, 11, 474–485. [Google Scholar] [CrossRef] [PubMed]
  22. Blank-Giwojna, A.; Postepska-Igielska, A.; Grummt, I. lncRNA KHPS1 Activates a Poised Enhancer by Triplex-Dependent Recruitment of Epigenomic Regulators. Cell Rep. 2019, 26, 2904–2915.e4. [Google Scholar] [CrossRef] [PubMed]
  23. Latos, P.A.; Pauler, F.M.; Koerner, M.V.; Senergin, H.B.; Hudson, Q.J.; Stocsits, R.R.; Allhoff, W.; Stricker, S.H.; Klement, R.M.; Warczok, K.E.; et al. Airn transcriptional overlap, but not its lncRNA products, induces imprinted Igf2r silencing. Science 2012, 338, 1469–1472. [Google Scholar] [CrossRef] [PubMed]
  24. Rosa, S.; Duncan, S.; Dean, C. Mutually exclusive sense-antisense transcription at FLC facilitates environmentally induced gene repression. Nat. Commun. 2016, 7, 13031. [Google Scholar] [CrossRef] [PubMed]
  25. Pandya-Jones, A.; Markaki, Y.; Serizay, J.; Chitiashvili, T.; Leon, W.R.M.; Damianov, A.; Chronis, C.; Papp, B.; Chen, C.K.; McKee, R.; et al. Publisher Correction: A protein assembly mediates Xist localization and gene silencing. Nature 2020, 586, E30. [Google Scholar] [CrossRef] [PubMed]
  26. Engreitz, J.M.; Haines, J.E.; Perez, E.M.; Munson, G.; Chen, J.; Kane, M.; McDonel, P.E.; Guttman, M.; Lander, E.S. Local regulation of gene expression by lncRNA promoters, transcription and splicing. Nature 2016, 539, 452–455. [Google Scholar] [CrossRef] [PubMed]
  27. Hartford, C.C.R.; Lal, A. When Long Noncoding Becomes Protein Coding. Mol. Cell Biol. 2020, 40, e00528-19. [Google Scholar] [CrossRef]
  28. Yin, Q.F.; Yang, L.; Zhang, Y.; Xiang, J.F.; Wu, Y.W.; Carmichael, G.G.; Chen, L.L. Long noncoding RNAs with snoRNA ends. Mol. Cell 2012, 48, 219–230. [Google Scholar] [CrossRef] [PubMed]
  29. Wu, H.; Yin, Q.F.; Luo, Z.; Yao, R.W.; Zheng, C.C.; Zhang, J.; Xiang, J.F.; Yang, L.; Chen, L.L. Unusual Processing Generates SPA LncRNAs that Sequester Multiple RNA Binding Proteins. Mol. Cell 2016, 64, 534–548. [Google Scholar] [CrossRef]
  30. Yap, K.; Mukhina, S.; Zhang, G.; Tan, J.S.C.; Ong, H.S.; Makeyev, E.V. A Short Tandem Repeat-Enriched RNA Assembles a Nuclear Compartment to Control Alternative Splicing and Promote Cell Survival. Mol. Cell 2018, 72, 525–540.e13. [Google Scholar] [CrossRef]
  31. Karakas, D.; Ozpolat, B. The Role of LncRNAs in Translation. Noncoding RNA 2021, 7, 16. [Google Scholar] [CrossRef] [PubMed]
  32. Banani, S.F.; Lee, H.O.; Hyman, A.A.; Rosen, M.K. Biomolecular condensates: Organizers of cellular biochemistry. Nat. Rev. Mol. Cell Biol. 2017, 18, 285–298. [Google Scholar] [CrossRef] [PubMed]
  33. Luo, J.; Qu, L.; Gao, F.; Lin, J.; Liu, J.; Lin, A. LncRNAs: Architectural Scaffolds or More Potential Roles in Phase Separation. Front. Genet. 2021, 12, 626234. [Google Scholar] [CrossRef] [PubMed]
  34. Bosson, A.D.; Zamudio, J.R.; Sharp, P.A. Endogenous miRNA and target concentrations determine susceptibility to potential ceRNA competition. Mol. Cell 2014, 56, 347–359. [Google Scholar] [CrossRef]
  35. Wang, L.; Zhao, S.; Mingxin, Y.U. LncRNA NR2F1-AS1 is involved in the progression of endometrial cancer by sponging miR-363 to target SOX4. Pharmazie 2019, 74, 295–300. [Google Scholar] [CrossRef] [PubMed]
  36. Yang, X.; Xing, G.; Liu, S.; Li, B.; He, Y.; Wang, F. LncRNA LOXL1-AS1 promotes endometrial cancer progression by sponging miR-28-5p to upregulate RAP1B expression. Biomed. Pharmacother. 2020, 125, 109839. [Google Scholar] [CrossRef] [PubMed]
  37. Yu, J.; Fan, Q.; Li, L. The MCM3AP-AS1/miR-126/VEGF axis regulates cancer cell invasion and migration in endometrioid carcinoma. World J. Surg. Oncol. 2021, 19, 213. [Google Scholar] [CrossRef]
  38. Zhong, Y.; Wang, Y.; Dang, H.; Wu, X. LncRNA AFAP1-AS1 contributes to the progression of endometrial carcinoma by regulating miR-545-3p/VEGFA pathway. Mol. Cell Probes 2020, 53, 101606. [Google Scholar] [CrossRef]
  39. Shen, J.; Yuan, Z.; Sheng, J.; Feng, X.; Wang, H.; Wang, Y.; Zhou, Y. Long non-coding RNA NNT-AS1 positively regulates NPM1 expression to affect the proliferation of estrogen-mediated endometrial carcinoma by interacting. J. Cancer 2022, 13, 112–123. [Google Scholar] [CrossRef]
  40. Zhao, H.; Xu, Q. Long non-coding RNA DLX6-AS1 mediates proliferation, invasion and apoptosis of endometrial cancer cells by recruiting p300/E2F1 in DLX6 promoter region. J. Cell Mol. Med. 2020, 24, 12572–12584. [Google Scholar] [CrossRef]
  41. Shen, L.; Wu, Y.; Li, A.; Li, L.; Shen, L.; Jiang, Q.; Li, Q.; Wu, Z.; Yu, L.; Zhang, X. LncRNA TTN-AS1 promotes endometrial cancer by sponging miR-376a-3p. Oncol. Rep. 2020, 44, 1343–1354. [Google Scholar] [CrossRef] [PubMed]
  42. Liu, Y.X.; Yuan, S.; Liu, X.J.; Huang, Y.X.; Qiu, P.; Gao, J.; Deng, G.P. LncRNA GATA3-AS1 promoted invasion and migration in human endometrial carcinoma by regulating the miR-361/ARRB2 axis. J. Mol. Med. 2022, 100, 1271–1286. [Google Scholar] [CrossRef] [PubMed]
  43. Xing, Y.; Sun, X.; Li, F.; Jiang, X.; Jiang, A.; Li, X.; Lv, R.; Shao, L. Long non-coding RNA (lncRNA) HOXB-AS3 promotes cell proliferation and inhibits apoptosis by regulating ADAM9 expression through targeting miR-498-5p in endometrial carcinoma. J. Int. Med. Res. 2021, 49, 3000605211013548. [Google Scholar] [CrossRef] [PubMed]
  44. Zhou, Q.; Kong, D.; Li, W.; Shi, Z.; Liu, Y.; Sun, R.; Ma, X.; Qiu, C.; Liu, Z.; Hou, Y.; et al. LncRNA HOXB-AS3 binding to PTBP1 protein regulates lipid metabolism by targeting SREBP1 in endometrioid carcinoma. Life Sci. 2023, 320, 121512. [Google Scholar] [CrossRef] [PubMed]
  45. Song, N.; Zhang, Y.; Kong, F.; Yang, H.; Ma, X. HOXA-AS2 promotes type I endometrial carcinoma via miRNA-302c-3p-mediated regulation of ZFX. Cancer Cell Int. 2020, 20, 359. [Google Scholar] [CrossRef] [PubMed]
  46. Guo, J.; Ye, F.; Xie, W.; Zhang, X.; Zeng, R.; Sheng, W.; Mi, Y.; Sheng, X. The HOXC-AS2/miR-876-5p/HKDC1 axis regulates endometrial cancer progression in a high glucose-related tumor microenvironment. Cancer Sci. 2022, 113, 2297–2310. [Google Scholar] [CrossRef] [PubMed]
  47. Zhang, X.J.; Qi, G.T.; Zhang, X.M.; Wang, L.; Li, F.F. lncRNA RHPN1-AS1 promotes the progression of endometrial cancer through the activation of ERK/MAPK pathway. J. Obstet. Gynaecol. Res. 2021, 47, 533–543. [Google Scholar] [CrossRef] [PubMed]
  48. Liu, Y.; Wang, L.L.; Chen, S.; Zong, Z.H.; Guan, X.; Zhao, Y. LncRNA ABHD11-AS1 promotes the development of endometrial carcinoma by targeting cyclin D1. J. Cell Mol. Med. 2018, 22, 3955–3964. [Google Scholar] [CrossRef] [PubMed]
  49. Wang, J.; Lei, C.; Shi, P.; Teng, H.; Lu, L.; Guo, H.; Wang, X. LncRNA DCST1-AS1 Promotes Endometrial Cancer Progression by Modulating the MiR-665/HOXB5 and MiR-873-5p/CADM1 Pathways. Front. Oncol. 2021, 11, 714652. [Google Scholar] [CrossRef]
  50. Treeck, O.; Weber, F.; Fritsch, J.; Skrzypczak, M.; Schuler-Toprak, S.; Buechler, C.; Ortmann, O. DSCAM-AS1 Long Non-Coding RNA Exerts Oncogenic Functions in Endometrial Adenocarcinoma via Activation of a Tumor-Promoting Transcriptome Profile. Biomedicines 2022, 10, 1727. [Google Scholar] [CrossRef]
  51. Li, L.; Chen, P.; Huang, B.; Cai, P. lncRNA DSCAM-AS1 facilitates the progression of endometrial cancer via miR-136-5p. Oncol. Lett. 2021, 22, 825. [Google Scholar] [CrossRef] [PubMed]
  52. Chen, L.; Shen, M. LncRNA VPS9D1-AS1 Sponging miR-520a-5p Contributes to the Development of Uterine Corpus Endometrial Carcinoma by Enhancing BIRC5 Expression. Mol. Biotechnol. 2022, 64, 1328–1339. [Google Scholar] [CrossRef] [PubMed]
  53. Peng, T.F.; Zhou, Y.J.; Zhou, J.; Zhou, Y.; Li, X.C.; Ouyang, Q. Long non-coding RNA VPS9D1-AS1 enhances proliferation, invasion, and epithelial-mesenchymal transition in endometrial cancer via miR-377-3p/SGK1. Kaohsiung J. Med. Sci. 2022, 38, 1048–1059. [Google Scholar] [CrossRef] [PubMed]
  54. Lai, T.; Qiu, H.; Si, L.; Zhen, Y.; Chu, D.; Guo, R. Long noncoding RNA BMPR1B-AS1 facilitates endometrial cancer cell proliferation and metastasis by sponging miR-7-2-3p to modulate the DCLK1/Akt/NF-kappaB pathway. Cell Cycle 2022, 21, 1599–1618. [Google Scholar] [CrossRef] [PubMed]
  55. Sun, Y.; Gao, X.; Li, P.; Song, L.; Shi, L. LncRNA ZFAS1, as a poor prognostic indicator, promotes cell proliferation and epithelial-mesenchymal transition in endometrial carcinoma. Per Med. 2021, 18, 43–53. [Google Scholar] [CrossRef] [PubMed]
  56. Peng, J.T.; Li, M.C. A functional Cis-eQTL locus in lncRNA ZNRD1-AS1 contributes to the susceptibility of endometrial cancer. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 7802–7807. [Google Scholar] [CrossRef] [PubMed]
  57. Dong, P.; Wang, F.; Taheri, M.; Xiong, Y.; Ihira, K.; Kobayashi, N.; Konno, Y.; Yue, J.; Watari, H. Long Non-Coding RNA TMPO-AS1 Promotes GLUT1-Mediated Glycolysis and Paclitaxel Resistance in Endometrial Cancer Cells by Interacting with miR-140 and miR-143. Front. Oncol. 2022, 12, 912935. [Google Scholar] [CrossRef] [PubMed]
  58. Shang, C.; Ao, C.N.; Cheong, C.C.; Meng, L. Long Non-coding RNA CDKN2B Antisense RNA 1 Gene Contributes to Paclitaxel Resistance in Endometrial Carcinoma. Front. Oncol. 2019, 9, 27. [Google Scholar] [CrossRef] [PubMed]
  59. Hao, M.; Li, T.; Xiao, L.; Liu, Y. METTL3-induced FGD5-AS1 contributes to the tumorigenesis and PD-1/PD-L1 checkpoint to enhance the resistance to paclitaxel of endometrial carcinoma. J. Cell Mol. Med. 2023, 28, e17971. [Google Scholar] [CrossRef]
  60. Wang, J.; Li, Z.; Wang, X.; Ding, Y.; Li, N. The tumor suppressive effect of long non-coding RNA FRMD6-AS2 in uteri corpus endometrial carcinoma. Life Sci. 2020, 243, 117254. [Google Scholar] [CrossRef]
  61. Gao, Q.; Huang, Q.; Li, F.; Luo, F. LncRNA MCTP1-AS1 Regulates EMT Process in Endometrial Cancer by Targeting the miR-650/SMAD7 Axis. Onco Targets Ther. 2021, 14, 751–761. [Google Scholar] [CrossRef]
  62. Lv, C.; Sun, J.; Ye, Y.; Lin, Z.; Li, H.; Liu, Y.; Mo, K.; Xu, W.; Hu, W.; Draz, E.; et al. Long noncoding RNA EIF1AX-AS1 promotes endometrial cancer cell apoptosis by affecting EIF1AX mRNA stabilization. Cancer Sci. 2022, 113, 1277–1291. [Google Scholar] [CrossRef] [PubMed]
  63. Jian, F.; Che, X.; Zhang, J.; Liu, C.; Liu, G.; Tang, Y.; Feng, W. The long-noncoding RNA SOCS2-AS1 suppresses endometrial cancer progression by regulating AURKA degradation. Cell Death Dis. 2021, 12, 351. [Google Scholar] [CrossRef]
  64. Gadd, M.S.; Bulatov, E.; Ciulli, A. Serendipitous SAD Solution for DMSO-Soaked SOCS2-ElonginC-ElonginB Crystals Using Covalently Incorporated Dimethylarsenic: Insights into Substrate Receptor Conformational Flexibility in Cullin RING Ligases. PLoS ONE 2015, 10, e0131218. [Google Scholar] [CrossRef] [PubMed]
  65. Zhao, Y.; Zou, X.; Wang, G.; Liu, Y.; Zhang, C.; Lu, W.; Li, Q. Effects of GATA6-AS/MMP9 on malignant progression of endometrial carcinoma. J. BUON 2021, 26, 1789–1795. [Google Scholar]
  66. Liu, Y.; Cai, X.; Cai, Y.; Chang, Y. lncRNA OIP5-AS1 Suppresses Cell Proliferation and Invasion of Endometrial Cancer by Regulating PTEN/AKT via Sponging miR-200c-3p. J. Immunol. Res. 2021, 2021, 4861749. [Google Scholar] [CrossRef]
  67. Yang, X.; Wang, C.C.; Lee, W.Y.W.; Trovik, J.; Chung, T.K.H.; Kwong, J. Long non-coding RNA HAND2-AS1 inhibits invasion and metastasis in endometrioid endometrial carcinoma through inactivating neuromedin U. Cancer Lett. 2018, 413, 23–34. [Google Scholar] [CrossRef] [PubMed]
  68. Liu, J.; Yin, J.; Wang, Y.; Cai, L.; Geng, R.; Du, M.; Zhong, Z.; Ni, S.; Huang, X.; Yu, H.; et al. A comprehensive prognostic and immune analysis of enhancer RNA identifies IGFBP7-AS1 as a novel prognostic biomarker in Uterine Corpus Endometrial Carcinoma. Biol. Proced. Online 2022, 24, 9. [Google Scholar] [CrossRef]
  69. Zhou, Y.X.; Zhao, W.; Mao, L.W.; Wang, Y.L.; Xia, L.Q.; Cao, M.; Shen, J.; Chen, J. Long non-coding RNA NIFK-AS1 inhibits M2 polarization of macrophages in endometrial cancer through targeting miR-146a. Int. J. Biochem. Cell Biol. 2018, 104, 25–33. [Google Scholar] [CrossRef]
  70. Zhang, Z.; Li, B.; Wang, Z.; Yang, L.; Peng, J.; Wang, H.; Wang, Y.; Hong, L. Novel LncRNA LINC02936 Suppresses Ferroptosis and Promotes Tumor Progression by Interacting with SIX1/CP Axis in Endometrial Cancer. Int. J. Biol. Sci. 2024, 20, 1356–1374. [Google Scholar] [CrossRef]
  71. Pan, X.; Li, D.; Huo, J.; Kong, F.; Yang, H.; Ma, X. LINC01016 promotes the malignant phenotype of endometrial cancer cells by regulating the miR-302a-3p/miR-3130-3p/NFYA/SATB1 axis. Cell Death Dis. 2018, 9, 303. [Google Scholar] [CrossRef] [PubMed]
  72. Lu, M.; Ding, N.; Zhuang, S.; Li, Y. LINC01410/miR-23c/CHD7 functions as a ceRNA network to affect the prognosis of patients with endometrial cancer and strengthen the malignant properties of endometrial cancer cells. Mol. Cell Biochem. 2020, 469, 9–19. [Google Scholar] [CrossRef] [PubMed]
  73. Cao, R.; Zhao, J.; Zhang, J. LINC01857 Exacerbates the Malignant Behaviors of Endometrial Carcinoma Cells by Sponging miR-19b-3p and Recruiting ELAVL1 to Upregulate MYCN. Gynecol. Obstet. Investig. 2023, 88, 16–29. [Google Scholar] [CrossRef] [PubMed]
  74. Yi, T.; Song, Y.; Zuo, L.; Wang, S.; Miao, J. LINC00470 Stimulates Methylation of PTEN to Facilitate the Progression of Endometrial Cancer by Recruiting DNMT3a Through MYC. Front. Oncol. 2021, 11, 646217. [Google Scholar] [CrossRef] [PubMed]
  75. Huang, Z.; Shen, F.; Chen, J.; Xie, B.; Chen, X.; Zhao, Y.; Chen, S. LncRNA linc01194 promotes the progress of endometrial carcinoma by up-regulating SOX2 through binding to IGF2BP1. J. Gynecol. Oncol. 2024, 35, e21. [Google Scholar] [CrossRef] [PubMed]
  76. Qiao, D.; Qin, X.; Yang, H.; Liu, X.; Liu, L.; Liu, S.; Jia, Z. Estradiol mediates the interaction of LINC01541 and miR-429 to promote angiogenesis of G1/G2 endometrioid adenocarcinoma in-vitro: A pilot study. Front. Oncol. 2022, 12, 951573. [Google Scholar] [CrossRef] [PubMed]
  77. Wang, C.; Kong, F.; Ma, J.; Miao, J.; Su, P.; Yang, H.; Li, Q.; Ma, X. IGF2BP3 enhances the mRNA stability of E2F3 by interacting with LINC00958 to promote endometrial carcinoma progression. Cell Death Discov. 2022, 8, 279. [Google Scholar] [CrossRef]
  78. Li, Q.Y.; Shen, J.Q.; Li, J.H.; Dai, D.F.; Saeed, M.; Li, C.X. LINC00958/miR-3174/PHF6 axis is responsible for triggering proliferation, migration and invasion of endometrial cancer. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 6853–6861. [Google Scholar] [CrossRef]
  79. Jiang, Y.; Qiao, Z.; Jiang, J.; Zhang, J. LINC00958 promotes endometrial cancer cell proliferation and metastasis by regulating the miR-145-3p/TCF4 axis. J. Gene Med. 2021, 23, e3345. [Google Scholar] [CrossRef]
  80. Zuo, X.; Li, W.; Yan, X.; Ma, T.; Ren, Y.; Hua, M.; Yang, H.; Wu, H.; Zhu, H. Long non-coding RNA LINC01224 promotes cell proliferation and inhibits apoptosis by regulating AKT3 expression via targeting miR-485-5p in endometrial carcinoma. Oncol. Rep. 2021, 46, 186. [Google Scholar] [CrossRef]
  81. Zhang, Z.; Liu, X.; Xu, H.; Li, P.; Zeng, T.; Du, W.; Lu, X. LINC01170 promotes the progression of endometrial carcinoma by activating the AKT pathway. J. BUON 2018, 23, 1745–1752. [Google Scholar] [PubMed]
  82. Li, Y.; Kong, C.; Wu, C.; Wang, Y.; Xu, B.; Liang, S.; Ying, X. Knocking down of LINC01220 inhibits proliferation and induces apoptosis of endometrial carcinoma through silencing MAPK11. Biosci. Rep. 2019, 39, BSR20181794. [Google Scholar] [CrossRef] [PubMed]
  83. Wang, Y.; Yin, L. LINC00461 Promoted Endometrial Carcinoma Growth and Migration by Targeting MicroRNA-219-5p/Cyclooxygenase-2 Signaling Axis. Cell Transplant. 2021, 30, 963689721989616. [Google Scholar] [CrossRef] [PubMed]
  84. Gao, X.; Yu, L.; Zhang, J.; Xue, P. Silencing of Long Non-Coding RNA LINC01106 Suppresses the Proliferation, Migration and Invasion of Endometrial Cancer Cells Through Regulating the miR-449a/MET Axis. Onco Targets Ther. 2020, 13, 9643–9655. [Google Scholar] [CrossRef] [PubMed]
  85. Li, W.; Li, H.; Zhang, L.; Hu, M.; Li, F.; Deng, J.; An, M.; Wu, S.; Ma, R.; Lu, J.; et al. Long non-coding RNA LINC00672 contributes to p53 protein-mediated gene suppression and promotes endometrial cancer chemosensitivity. J. Biol. Chem. 2017, 292, 5801–5813. [Google Scholar] [CrossRef]
  86. Fang, Q.; Sang, L.; Du, S. Long noncoding RNA LINC00261 regulates endometrial carcinoma progression by modulating miRNA/FOXO1 expression. Cell Biochem. Funct. 2018, 36, 323–330. [Google Scholar] [CrossRef] [PubMed]
  87. Feng, Y.; Dong, Y.; Rao, B.; Yu, Y.; Su, W.; Zeng, J.; Zhao, E.; Chen, Y.; Fang, S.; Zhou, Y.; et al. A novel LINC00478 serves as a tumor suppressor in endometrial carcinoma progression. J. Cancer Res. Clin. Oncol. 2023, 149, 14927–14940. [Google Scholar] [CrossRef]
  88. Chen, R.; An, J.; Wang, Y.; Yang, L.; Lin, Q.; Wang, Y. LINC01589 serves as a potential tumor-suppressor and immune-related biomarker in endometrial cancer: A review. Medicine 2023, 102, e33536. [Google Scholar] [CrossRef] [PubMed]
  89. Xu, D.; Dong, P.; Xiong, Y.; Chen, R.; Konno, Y.; Ihira, K.; Yue, J.; Watari, H. PD-L1 Is a Tumor Suppressor in Aggressive Endometrial Cancer Cells and Its Expression Is Regulated by miR-216a and lncRNA MEG3. Front. Cell Dev. Biol. 2020, 8, 598205. [Google Scholar] [CrossRef]
  90. Sun, K.X.; Wu, D.D.; Chen, S.; Zhao, Y.; Zong, Z.H. LncRNA MEG3 inhibit endometrial carcinoma tumorigenesis and progression through PI3K pathway. Apoptosis 2017, 22, 1543–1552. [Google Scholar] [CrossRef]
  91. Guo, Q.; Qian, Z.; Yan, D.; Li, L.; Huang, L. LncRNA-MEG3 inhibits cell proliferation of endometrial carcinoma by repressing Notch signaling. Biomed. Pharmacother. 2016, 82, 589–594. [Google Scholar] [CrossRef] [PubMed]
  92. Huang, S.; Chen, J.; Gao, X.; Shang, Z.; Ma, X.; Zhang, X.; Li, J.; Yin, R.; Meng, X. LncRNAs PSMG3-AS1 and MEG3 negatively regulate each other to participate in endometrial carcinoma cell proliferation. Mamm. Genome 2022, 33, 502–507. [Google Scholar] [CrossRef] [PubMed]
  93. He, X.; Bao, W.; Li, X.; Chen, Z.; Che, Q.; Wang, H.; Wan, X.P. The long non-coding RNA HOTAIR is upregulated in endometrial carcinoma and correlates with poor prognosis. Int. J. Mol. Med. 2014, 33, 325–332. [Google Scholar] [CrossRef] [PubMed]
  94. Wang, H.; Ma, X.; Jiang, Z.; Xia, D.; Sui, F.; Fu, F.; Dai, Y. Estrogen promotes the proliferation and migration of endometrial cancer cells by upregulating the expression of lncRNA HOTAIR. Gynecol. Endocrinol. 2023, 39, 2269248. [Google Scholar] [CrossRef] [PubMed]
  95. Chen, L.; Zheng, X.; Liu, W.; Sun, Y.; Zhao, S.; Tian, L.; Tian, W.; Xue, F.; Kang, C.; Wang, Y. Compound AC1Q3QWB upregulates CDKN1A and SOX17 by interrupting the HOTAIR-EZH2 interaction and enhances the efficacy of tazemetostat in endometrial cancer. Cancer Lett. 2023, 578, 216445. [Google Scholar] [CrossRef] [PubMed]
  96. Li, X.; Pang, L.; Yang, Z.; Liu, J.; Li, W.; Wang, D. LncRNA HOTAIRM1/HOXA1 Axis Promotes Cell Proliferation, Migration And Invasion In Endometrial Cancer. Onco Targets Ther. 2019, 12, 10997–11015. [Google Scholar] [CrossRef] [PubMed]
  97. Zhang, X.H.; Hu, P.; Xie, Y.Q.; Kang, Y.J.; Li, M. Long Noncoding RNA HOTAIR Promotes Endometrial Carcinoma Cell Proliferation by Binding to PTEN via the Activating Phosphatidylinositol 3-Kinase/Akt Signaling Pathway. Mol. Cell Biol. 2019, 39, e00251-19. [Google Scholar] [CrossRef] [PubMed]
  98. Zhou, Y.X.; Wang, C.; Mao, L.W.; Wang, Y.L.; Xia, L.Q.; Zhao, W.; Shen, J.; Chen, J. Long noncoding RNA HOTAIR mediates the estrogen-induced metastasis of endometrial cancer cells via the miR-646/NPM1 axis. Am. J. Physiol. Cell Physiol. 2018, 314, C690–C701. [Google Scholar] [CrossRef] [PubMed]
  99. Luczak, A.; Supernat, A.; Lapinska-Szumczyk, S.; Jachimowicz, D.; Majewska, H.; Gulczynski, J.; Zaczek, A.J. HOTAIR in relation to epithelial-mesenchymal transition and cancer stem cells in molecular subtypes of endometrial cancer. Int. J. Biol. Markers 2016, 31, e245–e251. [Google Scholar] [CrossRef]
  100. Chi, S.; Liu, Y.; Zhou, X.; Feng, D.; Xiao, X.; Li, W.; Zhao, Y.; Wang, H. Knockdown of long non-coding HOTAIR enhances the sensitivity to progesterone in endometrial cancer by epigenetic regulation of progesterone receptor isoform B. Cancer Chemother. Pharmacol. 2019, 83, 277–287. [Google Scholar] [CrossRef]
  101. Huang, J.; Ke, P.; Guo, L.; Wang, W.; Tan, H.; Liang, Y.; Yao, S. Lentivirus-mediated RNA interference targeting the long noncoding RNA HOTAIR inhibits proliferation and invasion of endometrial carcinoma cells in vitro and in vivo. Int. J. Gynecol. Cancer 2014, 24, 635–642. [Google Scholar] [CrossRef]
  102. Lottin, S.; Adriaenssens, E.; Berteaux, N.; Lepretre, A.; Vilain, M.O.; Denhez, E.; Coll, J.; Dugimont, T.; Curgy, J.J. The human H19 gene is frequently overexpressed in myometrium and stroma during pathological endometrial proliferative events. Eur. J. Cancer 2005, 41, 168–177. [Google Scholar] [CrossRef] [PubMed]
  103. Zhao, L.; Li, Z.; Chen, W.; Zhai, W.; Pan, J.; Pang, H.; Li, X. H19 promotes endometrial cancer progression by modulating epithelial-mesenchymal transition. Oncol. Lett. 2017, 13, 363–369. [Google Scholar] [CrossRef] [PubMed]
  104. Zhang, L.; Wang, D.L.; Yu, P. LncRNA H19 regulates the expression of its target gene HOXA10 in endometrial carcinoma through competing with miR-612. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 4820–4827. [Google Scholar] [CrossRef]
  105. Aminimoghaddam, S.; Fooladi, B.; Noori, M.; Nickhah Klashami, Z.; Kakavand Hamidi, A.; Amoli, M.M. The Effect of Metformin on Expression of Long Non-coding RNA H19 in Endometrial Cancer. Med. J. Islam. Repub. Iran. 2021, 35, 155. [Google Scholar] [CrossRef]
  106. Wang, W.; Ge, L.; Xu, X.J.; Yang, T.; Yuan, Y.; Ma, X.L.; Zhang, X.H. LncRNA NEAT1 promotes endometrial cancer cell proliferation, migration and invasion by regulating the miR-144-3p/EZH2 axis. Radiol. Oncol. 2019, 53, 434–442. [Google Scholar] [CrossRef] [PubMed]
  107. Dong, P.; Xiong, Y.; Yue, J.; Xu, D.; Ihira, K.; Konno, Y.; Kobayashi, N.; Todo, Y.; Watari, H. Long noncoding RNA NEAT1 drives aggressive endometrial cancer progression via miR-361-regulated networks involving STAT3 and tumor microenvironment-related genes. J. Exp. Clin. Cancer Res. 2019, 38, 295. [Google Scholar] [CrossRef] [PubMed]
  108. Fan, J.T.; Zhou, Z.Y.; Luo, Y.L.; Luo, Q.; Chen, S.B.; Zhao, J.C.; Chen, Q.R. Exosomal lncRNA NEAT1 from cancer-associated fibroblasts facilitates endometrial cancer progression via miR-26a/b-5p-mediated STAT3/YKL-40 signaling pathway. Neoplasia 2021, 23, 692–703. [Google Scholar] [CrossRef]
  109. Li, Z.; Wei, D.; Yang, C.; Sun, H.; Lu, T.; Kuang, D. Overexpression of long noncoding RNA, NEAT1 promotes cell proliferation, invasion and migration in endometrial endometrioid adenocarcinoma. Biomed. Pharmacother. 2016, 84, 244–251. [Google Scholar] [CrossRef]
  110. Wang, J.; Zhao, X.; Guo, Z.; Ma, X.; Song, Y.; Guo, Y. Regulation of NEAT1/miR-214-3p on the growth, migration and invasion of endometrial carcinoma cells. Arch. Gynecol. Obstet. 2017, 295, 1469–1475. [Google Scholar] [CrossRef]
  111. Xu, C.; Zhai, J.; Fu, Y. Overexpression of Nuclear Enriched Autosomal Transcript 1 Facilitates Cell Proliferation, Migration Invasion, and Suppresses Apoptosis in Endometrial Cancer by Targeting MicroRNA-202-3p/T Cell Immunoglobulin and Mucin Domain 4 Axis. Cancer Biother. Radiopharm. 2022, 37, 815–823. [Google Scholar] [CrossRef] [PubMed]
  112. Wang, L.; Huang, Q.; Lin, Q.; Chen, L.; Shi, Q. Knockdown of long non-coding RNA small nucleolar RNA host gene 9 or hexokinase 2 both suppress endometrial cancer cell proliferation and glycolysis. J. Obstet. Gynaecol. Res. 2021, 47, 2196–2203. [Google Scholar] [CrossRef] [PubMed]
  113. Cai, P.; Wu, M.; Zhang, B.; Wu, S.; Wei, H.; Wei, L. Long non-coding RNA SNHG12 regulates cell proliferation, invasion and migration in endometrial cancer by targeting miR-4429. Mol. Med. Rep. 2020, 22, 2842–2850. [Google Scholar] [CrossRef] [PubMed]
  114. Cai, P.; Li, G.; Wu, M.; Zhang, B.; Bai, H. ZIC2 upregulates lncRNA SNHG12 expression to promote endometrial cancer cell proliferation and migration by activating the Notch signaling pathway. Mol. Med. Rep. 2021, 24, 632. [Google Scholar] [CrossRef] [PubMed]
  115. Zhang, K.; Cai, Y.; Zhou, Q.; Sun, H.; Wei, J. Long Non-Coding RNA SNHG14 Impedes Viability, Migration and Invasion of Endometrial Carcinoma Cells Through Modulating miR-93-5p/ZBTB7A Axis. Cancer Manag. Res. 2020, 12, 9515–9525. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, G.F.; Wen, L.N. LncRNA SNHG14 promotes proliferation of endometrial cancer through regulating microRNA-655-3p. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 10410–10418. [Google Scholar] [CrossRef] [PubMed]
  117. Zhang, G.; Ma, A.; Jin, Y.; Pan, G.; Wang, C. LncRNA SNHG16 induced by TFAP2A modulates glycolysis and proliferation of endometrial carcinoma through miR-490-3p/HK2 axis. Am. J. Transl. Res. 2019, 11, 7137–7145. [Google Scholar] [PubMed]
  118. He, Y.; Xu, S.; Qi, Y.; Tian, J.; Xu, F. Long noncoding RNA SNHG25 promotes the malignancy of endometrial cancer by sponging microRNA-497-5p and increasing FASN expression. J. Ovarian Res. 2021, 14, 163. [Google Scholar] [CrossRef] [PubMed]
  119. Yu, J.; Jiang, L.; Gao, Y.; Sun, Q.; Liu, B.; Hu, Y.; Han, X. LncRNA CCAT1 negatively regulates miR-181a-5p to promote endometrial carcinoma cell proliferation and migration. Exp. Ther. Med. 2019, 17, 4259–4266. [Google Scholar] [CrossRef]
  120. Treeck, O.; Skrzypczak, M.; Schuler-Toprak, S.; Weber, F.; Ortmann, O. Long non-coding RNA CCAT1 is overexpressed in endometrial cancer and regulates growth and transcriptome of endometrial adenocarcinoma cells. Int. J. Biochem. Cell Biol. 2020, 122, 105740. [Google Scholar] [CrossRef]
  121. Zhao, X.; Fan, Y.; Lu, C.; Li, H.; Zhou, N.; Sun, G.; Fan, H. PCAT1 is a poor prognostic factor in endometrial carcinoma and associated with cancer cell proliferation, migration and invasion. Bosn. J. Basic. Med. Sci. 2019, 19, 274–281. [Google Scholar] [CrossRef] [PubMed]
  122. Zhang, C.; Shao, S.; Zhang, Y.; Wang, L.; Liu, J.; Fang, F.; Li, P.; Wang, B. LncRNA PCAT1 promotes metastasis of endometrial carcinoma through epigenetical downregulation of E-cadherin associated with methyltransferase EZH2. Life Sci. 2020, 243, 117295. [Google Scholar] [CrossRef] [PubMed]
  123. Fan, L.; Wang, C.; Zhan, P.; Liu, Y. LncRNA RBAT1 reduces chemosensitivity of cancer cells to carboplatin/paclitaxel by sponging miR-27b in endometrial carcinoma. J. Ovarian Res. 2023, 16, 147. [Google Scholar] [CrossRef] [PubMed]
  124. Zhang, C.; Wang, R.; Li, M.; Yang, Q. Long non-coding RNA BLACAT2/miR-378a-3p/YY1 feedback loop promotes the proliferation, migration and invasion of uterine corpus endometrial carcinoma. Oncol. Rep. 2023, 49, 108. [Google Scholar] [CrossRef] [PubMed]
  125. Du, Y.; Wang, L.; Chen, S.; Liu, Y.; Zhao, Y. lncRNA DLEU1 contributes to tumorigenesis and development of endometrial carcinoma by targeting mTOR. Mol. Carcinog. 2018, 57, 1191–1200. [Google Scholar] [CrossRef] [PubMed]
  126. Shao, W.; Li, Y.; Chen, F.; Jia, H.; Jia, J.; Fu, Y. Long non-coding RNA DLEU1 contributes to the development of endometrial cancer by sponging miR-490 to regulate SP1 expression. Pharmazie 2018, 73, 379–385. [Google Scholar] [CrossRef] [PubMed]
  127. Shan, L.; Zhao, T.; Wang, Y. Upregulation of Serum lncRNA DLEU1 Predicts Progression of Premalignant Endometrial Lesion and Unfavorable Clinical Outcome of Endometrial Cancer. Technol. Cancer Res. Treat. 2020, 19, 1533033820965589. [Google Scholar] [CrossRef] [PubMed]
  128. Dong, P.; Xiong, Y.; Konno, Y.; Ihira, K.; Kobayashi, N.; Yue, J.; Watari, H. Long non-coding RNA DLEU2 drives EMT and glycolysis in endometrial cancer through HK2 by competitively binding with miR-455 and by modulating the EZH2/miR-181a pathway. J. Exp. Clin. Cancer Res. 2021, 40, 216. [Google Scholar] [CrossRef]
  129. Zheng, X.; Liu, M.; Song, Y.; Feng, C. Long Noncoding RNA-ATB Impairs the Function of Tumor Suppressor miR-126-Mediated Signals in Endometrial Cancer for Tumor Growth and Metastasis. Cancer Biother. Radiopharm. 2019, 34, 47–55. [Google Scholar] [CrossRef]
  130. Wang, D.; Wang, D.; Wang, N.; Long, Z.; Ren, X. Long Non-Coding RNA BANCR Promotes Endometrial Cancer Cell Proliferation and Invasion by Regulating MMP2 and MMP1 via ERK/MAPK Signaling Pathway. Cell Physiol. Biochem. 2016, 40, 644–656. [Google Scholar] [CrossRef]
  131. Kong, F.; Ma, J.; Yang, H.; Yang, D.; Wang, C.; Ma, X. Long non-coding RNA PVT1 promotes malignancy in human endometrial carcinoma cells through negative regulation of miR-195-5p. Biochim. Biophys. Acta Mol. Cell Res. 2018, 1865, 1479–1490. [Google Scholar] [CrossRef] [PubMed]
  132. Cong, R.; Kong, F.; Ma, J.; Li, Q.; Yang, H.; Ma, X. The PVT1/miR-612/CENP-H/CDK1 axis promotes malignant progression of advanced endometrial cancer. Am. J. Cancer Res. 2021, 11, 1480–1502. [Google Scholar] [PubMed]
  133. Chen, S.; Liang, Y.; Shen, Y.; Wang, X. lncRNA XIST/miR-129-2-3p axis targets CCP110 to regulate the proliferation, invasion and migration of endometrial cancer cells. Exp. Ther. Med. 2023, 25, 159. [Google Scholar] [CrossRef] [PubMed]
  134. Chen, S.; Wang, L.L.; Sun, K.X.; Liu, Y.; Guan, X.; Zong, Z.H.; Zhao, Y. LncRNA TDRG1 enhances tumorigenicity in endometrial carcinoma by binding and targeting VEGF-A protein. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864 Pt B, 3013–3021. [Google Scholar] [CrossRef]
  135. Ma, J.; Kong, F.F.; Yang, D.; Yang, H.; Wang, C.; Cong, R.; Ma, X.X. lncRNA MIR210HG promotes the progression of endometrial cancer by sponging miR-337-3p/137 via the HMGA2-TGF-beta/Wnt pathway. Mol. Ther. Nucleic Acids 2021, 24, 905–922. [Google Scholar] [CrossRef] [PubMed]
  136. Jiang, H.; Li, Y.; Li, J.; Zhang, X.; Niu, G.; Chen, S.; Yao, S. Long noncoding RNA LSINCT5 promotes endometrial carcinoma cell proliferation, cycle, and invasion by promoting the Wnt/beta-catenin signaling pathway via HMGA2. Ther. Adv. Med. Oncol. 2019, 11, 1758835919874649. [Google Scholar] [CrossRef] [PubMed]
  137. Zeng, S.Y.; Liu, C.Q.; Zhuang, Y.; Chen, Y.; Gu, L.L.; Shi, S.Q. LncRNA ROR promotes proliferation of endometrial cancer cells via regulating Notch1 pathway. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 5970–5978. [Google Scholar] [CrossRef] [PubMed]
  138. Cai, J.; Wang, R.; Chen, Y.; Zhang, C.; Fu, L.; Fan, C. LncRNA FIRRE regulated endometrial cancer radiotherapy sensitivity via the miR-199b-5p/SIRT1/BECN1 axis-mediated autophagy. Genomics 2024, 116, 110750. [Google Scholar] [CrossRef] [PubMed]
  139. Zhang, X.H.; Li, M.; Kang, Y.J.; Xie, Y.Q.; Cao, Y.X. Long non-coding RNA LINP1 functions as an oncogene in endometrial cancer progression by regulating the PI3K/AKT signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 6830–6838. [Google Scholar] [CrossRef]
  140. Zhang, H.Q.; Li, T.; Li, C.; Hu, H.T.; Zhu, S.M.; Lu, J.Q.; Chen, X.J.; Huang, H.F.; Wu, Y.T. LncRNA THOR promotes endometrial cancer progression through the AKT and ERK signaling pathways. Med. Oncol. 2022, 39, 207. [Google Scholar] [CrossRef]
  141. Li, Q.; Shen, F.; Zhao, L. The relationship between lncRNA PCGEM1 and STAT3 during the occurrence and development of endometrial carcinoma. Biomed. Pharmacother. 2018, 107, 918–928. [Google Scholar] [CrossRef] [PubMed]
  142. Guo, J.L.; Tang, T.; Li, J.H.; Yang, Y.H.; Zhang, L.; Quan, Y. LncRNA HEIH Enhances Paclitaxel-Tolerance of Endometrial Cancer Cells via Activation of MAPK Signaling Pathway. Pathol. Oncol. Res. 2020, 26, 1757–1766. [Google Scholar] [CrossRef] [PubMed]
  143. Tian, J.; Cheng, H.; Wang, N.; Wang, C. SLERT, as a novel biomarker, orchestrates endometrial cancer metastasis via regulation of BDNF/TRKB signaling. World J. Surg. Oncol. 2023, 21, 27. [Google Scholar] [CrossRef]
  144. Chen, Q.; Schatz, C.; Cen, Y.; Chen, X.; Haybaeck, J.; Li, B. LncRNA TUG1 promotes the migration and invasion in type I endometrial carcinoma cells by regulating E-N cadherin switch. Taiwan. J. Obstet. Gynecol. 2022, 61, 780–787. [Google Scholar] [CrossRef]
  145. Liu, L.; Chen, X.; Zhang, Y.; Hu, Y.; Shen, X.; Zhu, W. Long non-coding RNA TUG1 promotes endometrial cancer development via inhibiting miR-299 and miR-34a-5p. Oncotarget 2017, 8, 31386–31394. [Google Scholar] [CrossRef]
  146. Jing, S.; Feng, Y.; He, X.L.; Wang, Y. Effects of lncRNA-UCA1 targeting miR-204-5p on the proliferation, migration, apoptosis and immune escape of endometrial carcinoma cells. Zhonghua Zhong Liu Za Zhi 2023, 45, 56–63. [Google Scholar] [CrossRef] [PubMed]
  147. Liu, T.; Wang, X.; Zhai, J.; Wang, Q.; Zhang, B. Long Noncoding RNA UCA1 Facilitates Endometrial Cancer Development by Regulating KLF5 and RXFP1 Gene Expressions. Cancer Biother. Radiopharm. 2021, 36, 521–533. [Google Scholar] [CrossRef] [PubMed]
  148. Lu, L.; Shen, Y.; Tseng, K.F.; Liu, W.; Duan, H.; Meng, W. Silencing of UCA1, a poor prognostic factor, inhibited the migration of endometrial cancer cell. Cancer Biomark. 2016, 17, 171–177. [Google Scholar] [CrossRef] [PubMed]
  149. Qiao, Q.; Li, H. LncRNA FER1L4 suppresses cancer cell proliferation and cycle by regulating PTEN expression in endometrial carcinoma. Biochem. Biophys. Res. Commun. 2016, 478, 507–512. [Google Scholar] [CrossRef]
  150. Sun, L.; Zhou, R.; Dong, J.; Liu, S.; Jiao, Y.; Wang, L.; Hu, S.; He, P.; Liu, X.; Zhao, X.; et al. Lnc-NA inhibits proliferation and metastasis in endometrioid endometrial carcinoma through regulation of NR4A1. J. Cell Mol. Med. 2019, 23, 4699–4710. [Google Scholar] [CrossRef]
  151. Li, Y.; Huo, J.; He, J.; Ma, X. LncRNA MONC suppresses the malignant phenotype of Endometrial Cancer Stem Cells and Endometrial Carcinoma Cells by regulating the MiR-636/GLCE axis. Cancer Cell Int. 2021, 21, 331. [Google Scholar] [CrossRef]
  152. Wu, X.; Cai, D.; Zhang, F.; Li, M.; Wan, Q. Long noncoding RNA TUSC7 inhibits cell proliferation, migration and invasion by regulating SOCS4 (SOCS5) expression through targeting miR-616 in endometrial carcinoma. Life Sci. 2019, 231, 116549. [Google Scholar] [CrossRef] [PubMed]
  153. Shang, C.; Lang, B.; Ao, C.N.; Meng, L. Long non-coding RNA tumor suppressor candidate 7 advances chemotherapy sensitivity of endometrial carcinoma through targeted silencing of miR-23b. Tumour Biol. 2017, 39, 1010428317707883. [Google Scholar] [CrossRef]
  154. Han, T.; Wu, Y.; Hu, X.; Chen, Y.; Jia, W.; He, Q.; Bian, Y.; Wang, M.; Guo, X.; Kang, J.; et al. NORAD orchestrates endometrial cancer progression by sequestering FUBP1 nuclear localization to promote cell apoptosis. Cell Death Dis. 2020, 11, 473. [Google Scholar] [CrossRef]
  155. Kong, D.; Hou, Y.; Li, W.; Ma, X.; Jiang, J. LncRNA-ZXF1 stabilizes P21 expression in endometrioid endometrial carcinoma by inhibiting ubiquitination-mediated degradation and regulating the miR-378a-3p/PCDHA3 axis. Mol. Oncol. 2022, 16, 813–829. [Google Scholar] [CrossRef]
  156. Shen, J.; Feng, X.P.; Hu, R.B.; Wang, H.; Wang, Y.L.; Qian, J.H.; Zhou, Y.X. N-methyladenosine reader YTHDF2-mediated long noncoding RNA FENDRR degradation promotes cell proliferation in endometrioid endometrial carcinoma. Lab. Investig. 2021, 101, 775–784. [Google Scholar] [CrossRef]
  157. Li, F.; Li, H.; Zhang, L.; Li, W.; Deng, J.; An, M.; Wu, S.; Lu, X.; Ma, R.; Wang, Y.; et al. X chromosome-linked long noncoding RNA lnc-XLEC1 regulates c-Myc-dependent cell growth by collaborating with MBP-1 in endometrial cancer. Int. J. Cancer 2019, 145, 927–940. [Google Scholar] [CrossRef] [PubMed]
  158. Guo, C.; Song, W.Q.; Sun, P.; Jin, L.; Dai, H.Y. LncRNA-GAS5 induces PTEN expression through inhibiting miR-103 in endometrial cancer cells. J. Biomed. Sci. 2015, 22, 100. [Google Scholar] [CrossRef] [PubMed]
  159. Tu, J.; Tan, X.; Chen, Y.; Chen, Y.; Li, Z.; Zhang, Y.; Chen, X.; Yang, H.; Chen, H.; Yu, Z. Growth arrest-specific transcript 5 represses endometrial cancer development by promoting antitumor function of tumor-associated macrophages. Cancer Sci. 2022, 113, 2496–2512. [Google Scholar] [CrossRef]
  160. Xin, W.; Zhao, S.; Han, X.; Zhao, P.; Yu, H.; Gao, X.; Li, P.; Wu, Q.; Ding, J.; Hua, K. lncRNA LA16c-313D11.11 modulates the development of endometrial cancer by binding to and inhibiting microRNA-205-5p function and indirectly increasing PTEN activity. Int. J. Oncol. 2020, 57, 355–363. [Google Scholar] [CrossRef]
Table 1. Main characteristics of subtypes of lncRNA.
Table 1. Main characteristics of subtypes of lncRNA.
Type of lncRNACharacteristic Features
Intronic lncRNALocated in intronic region of protein-coding gene
Intergenic lncRNASituated between two protein-coding genes
Antisense lncRNATranscription from complementary DNA strands
Bidirectional lncRNAArise from bidirectional transcription of protein-coding genes
Enhancer lncRNAOriginate from enhancer regions of protein-coding gene promoters; involved in the mediation of transcription factor positioning
Circular lncRNAlncRNA that undergo back splicing
Table 2. Summary of biological functions of lncRNAs.
Table 2. Summary of biological functions of lncRNAs.
Biological Functions of lncRNAs
Regulation of chromatin structure
Alteration of DNA methylation
Influence transcription process
Post-transcriptional regulation
Stimulation of DNA/RNA decay
Ensure stability of mRNA
Encoding peptides and protein relocalization
Regulation of translation and post-translation modification
Table 3. Summary of long non-coding antisense RNA actions.
Table 3. Summary of long non-coding antisense RNA actions.
Long Non-Coding Antisense RNA
Name of lncRNAImpact on Endometrial Cancer CourseMolecular Target of lncRNA Action
NR2F1-AS1Promoting progressionSOX4
LOXL1RAP1B
MCM3AP-AS1VEGF
AFAP1-AS1VEGFA
NNT-AS1Estrogen
DLX6-AS1p300/E2F1 acetyltransferase
TTN-AS1PUM2
GATA3-AS1ARRB2
HOXB-AS3ADAM9; lipid metabolism through PTBP1
HOXA-AS2ZFX
HOXC-AS2HKDC1
RHPN1-AS1MAPK/ERK pathway
ABHD11-AS1Cyclin D1
DCST1-AS1CADM1 and HOXB5
DSCAM-AS1PRL and Erα
VPS9D1-AS1BIRC5 and SGK1
BMPR1B-AS1DCLK1/Akt/NF-κB pathway and ZFAS
TMPO-AS1GLUT1 and paclitaxel resistance
CDKN2B-AS1Paclitaxel resistance
FGD5-AS1PD-1/PD-L1 checkpoint and paclitaxel resistance
FRMD6-AS2Inhibiting progressionHippo pathway
MTCP1-AS1SMAD-7
EIF1AX-AS1EIF1AX
SOCS2-AS1AURKA
GATA6-ASMMP-9
OIP5-AS1PTEN/AKT pathway
HAND2-AS1NMU
Table 4. Summary of long intergenic non-coding (LINC) RNA actions.
Table 4. Summary of long intergenic non-coding (LINC) RNA actions.
Long Intergenic Non-Coding RNA (LINC)
Name of lncRNAImpact on Endometrial Cancer CourseMolecular Target of lncRNA Action
Linc02936Promoting progressionFerroptosis
Linc01016NFYA
Linc01410CHD7
Linc01857MYCN
Linc00470MYC
Linc01194SOX2
Linc01541VEGFA
Linc00958E2F3; PHF6; TCF-4
Linc01224 and LINC01170AKT3
Linc01220MAPK11
Linc00461COX-2
Linc01106.MET
Linc00672Inhibiting progressionp-53-induced LASP1 downregulation; increasing chemosensitivity to paclitaxel
Linc00261FOXO1
Linc00478PTBP1
Linc01589Immune system activation
Table 5. Summary of small nucleolar host gene (SNHG) actions.
Table 5. Summary of small nucleolar host gene (SNHG) actions.
Small Nucleolar Host Genes (SNHGs)
Name of lncRNAImpact on Endometrial Cancer CourseMolecular Target of lncRNA Action
SNHG9Promoting progressionGlycolysis
SNHG12Promoting progression MMP2 and MMP9
SNHG14 Inhibiting progression ZBTB7A
SNHG16Promoting progression HX2
SNHG25 Promoting progressionFASN
Table 6. Summary of long non-coding RNA-associated transcript actions.
Table 6. Summary of long non-coding RNA-associated transcript actions.
LncRNA-Associated Transcripts (ATs)
Name of lncRNAImpact on Endometrial Cancer CourseMolecular Target of lncRNA Action
CCAT1Promoting progressionmiR-181a-5p
PCAT1epithelial–mesenchymal transition
RBAT1carboplatin/paclitaxel chemosensitivity downregulation
BLACAT2 MEK/ERK pathway
Table 7. Summary of other long non-coding RNA actions.
Table 7. Summary of other long non-coding RNA actions.
Other Long Non-Coding RNAs Associated with Endometrial Cancer
Name of lncRNAImpact on Endometrial Cancer CourseMolecular Target of lncRNA Action
HOTAIRPromoting progression PI3K/Akt pathway; PTEN suppression
H19HOXA10
NEAT1EZH2; STAT3; HMGA1; TIMD4; MMP-2, MMP-7 and IGF1
DLEU1PI3K/AKT/mTOR pathway
DLEU2HK2
LncRNA-ATBATB
BANCRERK/MAPK pathway; MMP2 and MMP1
PVT1FGFR1 and FGF2; CENP-H
XISTCCP110
NDRG1VEGFA
MIR210HGWnt/β-catenin and TGF-β/Smad3 signalling pathways
LSINCT5HMGA2
RORNotch1
FIRRESIRT1
LINP1PI3K/AKT signalling
THORERK/AKT pathway
PCGEM1STAT3
HEIHMAPK pathway
SLERTVimentin and E-cadherin upregulation; N-cadherin downregulation
TUG1Epithelial–mesenchymal transition
UCA1KLF5 and RXFP1
MEG3Inhibiting progressionPD-L1; PI3K/m-TOR signalling; Notch 1 and HES-1 expression
FER1L4PTEN
NANR4A1
MONCGLCE
TUSC7SOCS4
NORADFUBP1
ZXF1P21
FENDDR SOX4
XLEC1 MBP-1 activation and c-Myc downregulation
GAS5PTEN
lncRNA-LA16C-313D11.11PTEN
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jasielski, P.; Zawlik, I.; Bogaczyk, A.; Potocka, N.; Paszek, S.; Maźniak, M.; Witkoś, A.; Korzystka, A.; Kmieć, A.; Kluz, T. The Promotive and Inhibitory Role of Long Non-Coding RNAs in Endometrial Cancer Course—A Review. Cancers 2024, 16, 2125. https://doi.org/10.3390/cancers16112125

AMA Style

Jasielski P, Zawlik I, Bogaczyk A, Potocka N, Paszek S, Maźniak M, Witkoś A, Korzystka A, Kmieć A, Kluz T. The Promotive and Inhibitory Role of Long Non-Coding RNAs in Endometrial Cancer Course—A Review. Cancers. 2024; 16(11):2125. https://doi.org/10.3390/cancers16112125

Chicago/Turabian Style

Jasielski, Patryk, Izabela Zawlik, Anna Bogaczyk, Natalia Potocka, Sylwia Paszek, Michał Maźniak, Aleksandra Witkoś, Adrianna Korzystka, Aleksandra Kmieć, and Tomasz Kluz. 2024. "The Promotive and Inhibitory Role of Long Non-Coding RNAs in Endometrial Cancer Course—A Review" Cancers 16, no. 11: 2125. https://doi.org/10.3390/cancers16112125

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop