Next Article in Journal
Clinical Implications of Circulating Tumor Cells in Patients with Esophageal Squamous Cell Carcinoma: Cancer-Draining Blood Versus Peripheral Blood
Previous Article in Journal
Overcoming Resistance to Checkpoint Inhibitors with Combination Strategies in the Treatment of Non-Small Cell Lung Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Long Non-Coding RNAs, Nuclear Receptors and Their Cross-Talks in Cancer—Implications and Perspectives

by
Prabha Tiwari
1,* and
Lokesh P. Tripathi
2,3,*
1
Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
2
Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Kanagawa, Japan
3
AI Center for Health and Biomedical Research (ArCHER), National Institutes of Biomedical Innovation, Health and Nutrition, Kento Innovation Park NK Building, 3-17 Senrioka Shinmachi, Settsu 566-0002, Osaka, Japan
*
Authors to whom correspondence should be addressed.
Cancers 2024, 16(16), 2920; https://doi.org/10.3390/cancers16162920
Submission received: 5 June 2024 / Revised: 30 July 2024 / Accepted: 14 August 2024 / Published: 22 August 2024

Abstract

:

Simple Summary

Long non-coding RNAs (lncRNAs) exert their influence on transcriptional regulation and genome organization by associating with transcription factors, chromatin, RNAs and various proteins. In particular, studies have established a widespread involvement of cross-talks between lncRNAs and nuclear receptors (NRs) in pathology of diseases such as cancer. A deeper understanding of these cross-talks will not only help gain greater insights into cancer physiology, but also guide the development for newer and improved therapeutic strategies aimed at cancer.

Abstract

Long non-coding RNAs (lncRNAs) play key roles in various epigenetic and post-transcriptional events in the cell, thereby significantly influencing cellular processes including gene expression, development and diseases such as cancer. Nuclear receptors (NRs) are a family of ligand-regulated transcription factors that typically regulate transcription of genes involved in a broad spectrum of cellular processes, immune responses and in many diseases including cancer. Owing to their many overlapping roles as modulators of gene expression, the paths traversed by lncRNA and NR-mediated signaling often cross each other; these lncRNA-NR cross-talks are being increasingly recognized as important players in many cellular processes and diseases such as cancer. Here, we review the individual roles of lncRNAs and NRs, especially growth factor modulated receptors such as androgen receptors (ARs), in various types of cancers and how the cross-talks between lncRNAs and NRs are involved in cancer progression and metastasis. We discuss the challenges involved in characterizing lncRNA-NR associations and how to overcome them. Furthering our understanding of the mechanisms of lncRNA-NR associations is crucial to realizing their potential as prognostic features, diagnostic biomarkers and therapeutic targets in cancer biology.

1. Introduction

Non-coding RNAs (ncRNAs) are defined as transcripts with little or no protein-coding potential [1]. Genes encoding ncRNAs comprise a significant portion of the genome in higher eukaryotes. Until recently, ncRNAs were viewed as “junk” or “transcriptional noise”, but their biological functions have now been increasingly recognized. These functions include regulating gene expression, modulating the activities of transcription factors, chromatin remodeling, nuclear and cytoplasmic protein scaffolding and coregulation with other RNAs [1,2]. NcRNAs are broadly categorized as long non-coding RNAs (lncRNAs), which are typically defined as transcripts that are >200 nucleotides long [1], small ncRNAs such as microRNAs (miRNAs) and circular RNAs (circRNAs) [3]. Functions, protein binding properties and subcellular localization of lncRNAs typically correlate with short motifs (k-mers) rather than linear sequence homology [4]. Besides nuclear localization, lncRNAs can also associate with cytoplasm and cytoplasmic organelles such as mitochondria and ribosomes; they tend to acquire secondary and tertiary structures, which strongly correlate with their functions [5,6]. In general, lncRNAs can bind to DNA, RNA or proteins, are less conserved compared to other classes of RNAs [7,8], and their interactions may often be context-dependent or cell-type dependent [9]. These interactions enable lncRNAs to participate in a wide array of cellular processes in normal as well as in disease conditions [10,11,12]. In particular, lncRNA interactions with different transcription factors (TFs) play key roles in determining gene expression and are also implicated in pathophysiology of several human diseases [1,13,14]. Among the TFs, the nuclear receptors (NRs) frequently associate with lncRNAs; NRs are one of the largest groups of TFs that regulate the expression of functional genes involved in various biological processes such as development, immune responses and metabolism [15]. NRs are intracellular receptors; some NRs, including the androgen receptor (AR) and the glucocorticoid receptor (GR), are located in the cytoplasm and translocated to the nucleus after stimulation by agonists; other receptors such as retinoic acid receptors (RAR) are located in the nucleus and are bound to specific sequences even in the absence of ligands [16,17]. NRs are modulated by various endogenous ligands such as hormones, growth factors, fatty acids and their metabolites; although NRs with no known endogenous ligands, hence classified as orphan receptors, have also been reported [18]. Functional roles of NRs include transactivation or transrepression of target genes and coregulation of other transcriptional regulators [19]. NcRNAs can regulate NRs directly, or coregulate via direct interactions with NR-associated coactivators and corepressors, and also function as downstream targets of NRs in feedback loops [20,21]. Given the extensive and often overlapping involvement of lncRNAs and NRs in cellular physiology and diseases, investigating lncRNA-NR cross-talks is crucial to a deeper understanding of the pathogenesis of various diseases as well as in the identification of novel therapeutic targets [22]. In this review, we have specifically focused on how lncRNA-NR cross-talks drive cell proliferation, progression, tumorigenesis and metastasis in various types of cancer. We discuss the challenges in characterizing lncRNA-NR cross-talks in cancer systems and the probable approaches to generate more in-depth coverage of these associations. We finally deliberate on how a deeper understanding of lncRNA-NR cross-talks may assist in the development of newer and more effective diagnostics and anticancer therapies.

2. LncRNA Classification, Functional Characterization and Their Roles in Cancer

2.1. Functional Repertoire of lncRNAs

The complexity and diversity of lncRNA functions make their classification a challenging task [1]. Nevertheless, lncRNAs can be classified by genomic location (i.e., intergenic, intronic, sense, antisense), transcriptional regulation of nearby genes (i.e., cis, trans) and their effects on proteins (i.e., signal, decoy, guide, scaffold) [23]. LncRNAs can also be classified based on association with repeat elements or by their stability [24]. LncRNAs exert their functions primarily via biomolecular interactions, but also by undergoing structural modifications [1,25]. Therefore, a systematic characterization of lncRNA interactions with other biomolecules in the form of lncRNA interactome maps will help to better understand their biological functions [26]. Accordingly, genome-wide technologies have been developed to map system-wide RNA-chromatin interactions [27]. While secondary structure determination of lncRNAs is difficult due to variation in sizes and the heterogeneous nature of the transcripts, various methods such as enzymatic footprinting, chemical probing, nuclear magnetic resonance (NMR) spectroscopy and comparative sequence analysis have been employed to determine lncRNA secondary structure [6] and to correlate them with lncRNA function [28]. Functional roles played by lncRNAs include regulation of local gene expression in cis, at distant genomic locations in trans, epigenetic and enhancer regulation [10,29,30,31].
Regulation of gene expression by lncRNAs is a multilayered process and involves many different mechanisms. Broadly, lncRNAs modulate gene expression by (a) associating with chromatin and (b) by modulating RNA splicing, transcript stability and translation. Chromatin regulation by lncRNAs is facilitated by direct or indirect interactions (via proteins) between lncRNAs and specific-DNA regions, which facilitate the modification of the local chromatin environment and govern the changes in gene expression [32,33]. A key feature of direct interactions between lncRNAs and DNA is the formation of hybrid structures, namely RNA-DNA-DNA triplexes that are formed between RNAs and double-stranded DNA [34] and R-loops that are formed between RNA and single-stranded DNA [35]. Transcriptional and post-transcriptional regulation by lncRNAs variously involves gene silencing as in the case of XIST lncRNA [36], lncRNAs functioning as micro RNA (miRNA) sponges, thereby restricting the ability of miRNAs to target expressed mRNAs [37] or functioning as decoys of chromatin modifiers, and thus preventing them from associating with the target gene promoters [38]. A subset of lncRNAs, namely natural antisense transcripts that are transcribed from the opposite strands of the genes, are also deeply involved in gene regulatory processes [39]. Multiple studies have suggested key roles for lncRNAs in alternate splicing of genes that can have diverse implications for the development, differentiation and pathogenesis of various diseases such as cancer [40]. For a detailed discussion on the mechanisms of gene regulation by lnRNAs, readers are also encouraged to see the review on the subject from Statello and colleagues [41].
There is a widespread interest in the characterization of the lncRNA repertoire and their functions at the whole genome level. The FANTOM6 (Functional Annotation of Mammalian Genomes) initiative is aimed at a system-wide characterization of lncRNA functions in different cellular contexts and has provided novel insights into functional roles of many different lncRNAs [42,43]. There is also a growing cognizance of networks of extensive cross-talks between lncRNAs and other cellular factors, especially TFs such as NRs to determine gene expression, modulate signaling pathways and regulate cellular physiology [1,42,43].

2.2. LncRNAs in Cancer

There is an increasing acknowledgment of the involvement of lncRNAs in virtually all tumorigenesis and cancer progression-related pathways such as Wnt/β-catenin, Hippo, Notch, NF-κB and TGF-β that are involved in proliferation, differentiation, metastasis and apoptosis [44,45]. For instance, metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) and nuclear-enriched abundant transcript 1 (NEAT1) are strongly implicated in cancer progression and metastasis in various cancer types [46,47,48]. MALAT1 is overexpressed and displays increased mutations in many cancer types [49,50,51]. Moreover, various cell-cycle regulatory proteins involved in cancer progression such as cyclins/cyclin-dependent kinases and transcription factors (E2F, p53) are known to be regulated by lncRNAs [52]. Many cellular processes in cancer stem cells (CSCs) that are known to initiate tumor formation can be regulated by lncRNAs such as MALAT1, ROR, HOTAIR, H19, UCA1 and ARSR [53,54]. Autophagy, which can play dual roles in both the promotion and inhibition of tumorigenesis, can be regulated by lncRNAs such as NBR2, MEG3, PTENP1, GAS5, HULC and HOTAIR [55,56]. Indeed, several autophagy-related lncRNAs having prognostic value were identified in bladder cancer [57]. Tumor microenvironment and peripheral immune landscape perturbations play important roles in cancer progression and therapy [58]. Hence, there have been efforts aimed at mapping the single-cell lncRNA landscape of T cells towards a better understanding of T cell regulation in cancer immunity and immunotherapies [59]. Both cancer and immune cells undergo metabolic reprogramming during tumorigenesis and cancer progression [60,61]. It is reported that lncRNAs can regulate both metabolism and the immune microenvironment through generation and utilization of metabolites [62]. Roles of lncRNAs in cancer glucose metabolism via regulation of various glucose transporters, kinases and metabolic pathways have also been noted [63]. Moreover, multiple studies have suggested key roles for lncRNAs MALAT1, LINC-PINT and NEAT1 in cancer [64], and in therapeutic resistance in breast cancer [65]. Recently, a CRISPR-based pipeline using non-small cell lung cancer (NSCLC) suggested that cancer vulnerabilities can be targeted by potent antisense oligonucleotide inhibitors [66]. Studies have also suggested potential roles of lncRNAs as biomarkers in cancer diagnosis such as prostate cancer antigen 3 (PCA3) for prostate cancer and EGFR-AS1 and MIR205HG as predictors of antiepidermal growth-factor receptor drug response [67,68]. Tumor drug resistance, which is a formidable obstacle in successful chemotherapy, can be modulated by lncRNAs in several ways, such as regulation of gene expression, alteration of drug metabolism, drug efflux, drug targets, epithelial–mesenchymal transition, and inhibition of drug-induced apoptosis [69]. These studies have indicated extensive roles of lncRNAs in cancer therapy [70] and there is an ever-growing discourse on the potential for lncRNAs being targeted for precision therapy in human cancer [71]. Concurrently, there has been an emergence of curated repositories that catalogue lncRNA-cancer associations. For instance, Lnc2RNA is a database of experimentally characterized associations between lncRNAs and various human cancers [72]. Another repository, Cancer LncRNA Census (CLC) leverages direct functional or genetic evidence to assign causal lncRNA-cancer roles for 122 GENCODE lncRNAs with established roles in cancer processes [49]. The well-characterized roles of lncRNAs in various cancer types are summarized in Table S1.

3. Molecular Functions of NRs and Their Roles in Cancer

3.1. Functional Repertoire of NRs

NRs are typically composed of an N-terminal domain (NTD), a DNA binding domain (DBD), a hinge region, a ligand-binding domain (LBD) and a C-terminal domain [73]. A large number of NRs have been discovered as a part of multimember families in different species (for instance, 48, 49, 49 and 47 NRs have been reported in human, mouse, rat and dolphin, respectively), and they are broadly divided into four subgroups [74,75]. Type I receptors such as androgen receptors (ARs) and progesterone receptors (PRs) bind to direct repeats as homodimers upon ligand activation and regulate the expression of target genes involved in cellular physiology and pathophysiology [21,76,77]. Type II NRs such as peroxisome-proliferation-activated receptor (PPAR) and retinoid acid receptor (RAR) form heterodimers with retinoid X receptor (RXR) upon activation and translocate to specific DNA motifs (AGGTCA) termed PPAR response elements (PPRE). PPARs bind to the 5′ end of the PPRE, while RXR binds to the 3′ end [78]. These complexes regulate diverse functions including lipid metabolism, energy homeostasis, gene expression, differentiation, proliferation and apoptosis and inflammatory conditions [79,80,81,82,83,84]. PPAR superfamily comprises three subtypes, namely, PPARα, PPARβ/δ and PPARγ, which are encoded by separate genes and display differential tissue distribution [85]. Apart from fatty acids and their metabolites, PPARs are also activated by several hypolipidemic and antidiabetic drugs via binding with the ligand-binding domain (LBD), which causes changes in their conformation and allows them to regulate the expression of their target genes [83,84,86]. The number of carbon atoms, degree of saturation and configuration of fatty acids play important roles in the activation of PPARs [87]. Type III NRs bind to direct repeat and palindromic sequences as homodimers and can regulate constitutive transactivation and transrepression functions without ligand activation [73]. Type IV NRs bind to DNA at a single consensus half-site preceded by a 5′-A/T-rich flanking sequence as monomers [88].
Coregulators are acknowledged to play critical roles in the facilitating NR-regulated transcriptional control [89]. It has also been established that chromatin accessibility, epigenetic modifications and lncRNAs play important roles in the activation of NRs [90]. Further, NR dimerization can play significant roles in gene regulation through their binding to the promoters and/or enhancers of the target genes [91].

3.2. NRs in Cancer

NRs such as AR, estrogen receptor (ER), farnesoid X receptor (FXR), glucocorticoid receptor (GR), hepatocyte nuclear factor 4 alpha (HNF4α), liver X receptors (LXR), PPARs and pregnane X receptor (PXR) function as regulators of both inflammation and cancer [92,93,94]. Cross-talks between TLRs and PPARs are known to play important roles in various inflammatory diseases including cancer via modulation of various common signaling pathways such as NF-κB and AP-1 [95]. The roles of steroid receptors AR and ER in both prostate and breast cancers are well established [96]. Indeed, AR and ER coexpression can determine the efficacy of hormone receptor-mediated radiosensitization in breast cancer [97]. Apart from AR and ER, GR-mediated signaling pathways are emerging as key players in breast and prostate cancers [98]. The GR-related signaling can lead to either cancer progression or cancer suppression in different cancer types [99]. It has been shown that GR activation can also trigger anticancer drug tolerance via modulation of the CDKN1C gene in lung cancer [100]. In particular, the interplay among hormones, growth factors such as EGF (epidermal growth factor)- and IGF (insulin-like growth factor)-mediated signaling and even lncRNAs, plays an important role in the pathogenesis of many cancer types such as breast and prostate cancer [101,102,103,104,105] (Figure 1). Other widely studied NRs, PPARα, PPARβ/δ and PPARγ, play important roles in both cancer progression and inhibition via modulation of metabolism, inflammation, cell proliferation and apoptosis in various cancer types in a context-dependent manner [106,107,108]. Many studies have suggested potential roles of PPAR modulators in cancer therapeutics [109,110]. NR HNF4α is involved in the regulation of various biological processes, for instance, inflammation, cell proliferation, and drug and lipid metabolism [111]. Also, the tumor-promoting role of HNF4α through induction of cell proliferation, invasion, metastasis and angiogenesis is well studied [112]. The tumor-suppressor role of HNF4α through the regulation of p21-mediated cellular senescence [113] in prostate cancer is also reported. HNF4α is a promising therapeutic target in a variety of cancers via the regulation of various signaling pathways (i.e., Wnt/β-catenin, NF-κB, STAT3, TGFβ) [94]. LXRs are expressed in cancer, stromal and immune cells, and can regulate various signaling pathways involved in lipogenesis, inflammation, cell cycle, and metabolism [114]. Therapeutic roles of LXR ligands via modulation of cholesterol homeostasis in various cancer types such as breast, glioblastoma and melanoma are emerging [115,116,117]. The FXR nuclear receptor is activated by bile acids and plays important roles in the regulation of metabolism [118]. Expression and functional roles of FXR in various types of cancers such as breast, pancreatic, gastric, cervical, colon and hepatocellular carcinoma have been reported [119]. In addition to the regulation of metabolism, FXR regulates proliferation and apoptosis in cancer cells [119,120]. It was demonstrated that the overexpression of FXR can inhibit migration, adhesion and angiogenesis of bladder cancers through proteasome degradation and VEGF reduction [121]. Furthermore, various studies have suggested a therapeutic potential for FXR-regulated targets such as matrix metalloproteinase 7 in colon, and Wnt/β-catenin in colorectal cancer, respectively [122,123]. PXR is an orphan NR that is activated by both endobiotics and xenobiotics, which can be expressed in several cancer types, such as prostate, breast, cervical and ovarian [124]. PXR can regulate various important cellular events such as proliferation, cell cycle, apoptosis, metabolism and chemotherapeutic drug resistance [125]. Roles of PXR polymorphisms in the chemotherapy of cancer patients have also been suggested [126]. A list of NRs with well-characterized roles in various cancer types is summarized in Table S1.

4. Cross-Talks between lncRNAs and NRs in Cancer

Many roles of lncRNAs and NRs in cancer networks are driven by cross-talks between them; these cross-talks determine gene expression, regulate metabolism and signaling pathway outcomes and modulate immune resistance in various types of cancer. The cross-talks between lncRNAs and NRs may happen in many different ways, such as transcriptional regulation, coactivation, chromatin reorganization, interplay between the shared constituents of signaling pathways, etc., and they regulate many different biological processes, including those associated with human disorders such as atherosclerosis, Alzheimer’s disease, various cancers [52] and liver diseases [127] (Figure 2 and Table 1).
LncRNA-NR cross-talks may be viewed as broadly falling into two mechanistic classes. The first category includes instances where lncRNAs and NRs associate with each other via direct physical contacts (Figure 2A,B); it is well established that apart from direct activation by ligands, NRs can also be regulated by direct interactions with lncRNAs and in turn NRs can also regulate lncRNAs via mutual regulatory feedback loops [147]. The second category involves lncRNAs and NRs functionally regulating each other by modulating gene expression, transcript stabilization, regulation of splicing and cross-talks between lncRNA-mediated and NR-mediated signaling pathways, via overlapping cellular factors (Figure 2C–G). Recently, Cantile et al. have discussed at length the reciprocal interactions between endocrine NRs and lncRNAs in different tumor diseases [148]. Below we discuss well-characterized examples of regulatory associations between individual lncRNAs and specific NRs and their functional outcomes on gene expression and other physiological processes in cancer progression and tumorigenesis.

4.1. LncRNA Cross-Talks with ARs

AR, which is activated by binding with circulating androgenic hormones is increasingly implicated with essential roles in various cancer types including prostate cancer pathogenesis and therapy [132] and breast cancer [149]. Several AR-signaling-associated lncRNAs such as PCAL7, PRKAG2-AS1 and HOXC-AS1 can act as diagnostic biomarkers for prostate cancer [132,150]. Additionally, many lncRNAs such as PCGEM1, CTBP1-AS, PCAL7, HOTAIR, CRPC-Lnc#6, SOCS2-AS1, NXTAR and ARNILA associate with ARs in different cancers and modulate various aspects of tumorigenesis and carcinogenesis via diverse mechanisms (Figure 2 and Table 1). For instance, Hung et al. demonstrated that prostate cancer gene expression marker 1 (PCGEM1), which is an AR-regulated, prostate-specific lncRNA strongly elevated in prostate cancer, regulates metabolic programming in tumor cells by functioning as a coactivator for both AR and c-Myc (Figure 2A) [128]. Takayama et al. showed that positive regulation of AR transcription by CTBP1-AS played a significant role in prostate cancer progression [130]. Multiple studies have also established important roles for lncRNA-AR cross-talks in castration-resistant prostate cancer (CRPC), a more aggressive form of prostate cancer; these cross-talks are diverse and involve varied underlying mechanisms. For instance, Li et al. showed that PCAL7, an AR-induced lncRNA, binds with and stabilizes Huntingtin-interacting protein 1 (HIP1), which in turn activates AR signaling; this positive feedback loop plays an essential role in CRPC progression (Figure 2E) [132]. Takayama et al. characterized a set of AR-regulated lncRNAs that were upregulated in CRPC tissues and directly modulate the spliceosome complex; since the dysregulation of splicing machinery plays a key role in CRPC progression, lncRNA-mediated perturbations in the spliceosome complex may facilitate CRPC progression (Figure 2D) [133]. Misawa et al. demonstrated that androgen-regulated lncRNA, suppressor of cytokine signaling 2-antisense transcript 1 (SOCS2-AS1), which is highly expressed in CRPC, inhibited apoptosis and promoted cancer cell survival. SOCS2-AS1 also modulated AR signaling by interacting with AR, promoting cofactor recruitment and epigenetic modification of AR-target genes in CRPC [134]. Additionally, Ghildiyal et al. showed that lncRNA NXSTAR functions as a tumor suppressor gene, which is negatively regulated by AR and which itself negatively regulates AR and AR-V7 expression in an epigenetic fashion (Figure 2F) [135]. It was also demonstrated that impeding AR binding with lncRNA SLNCR blocked melanoma invasion, indicating key oncogenic roles of lncRNA-AR complexes and their implied therapeutic potential [147]. Additionally, in a study from Zhang et al., lncRNA HOTAIR, by binding to the AR N-terminal domain (NTD), demonstrably stabilized the AR protein by preventing its ubiquitination and subsequent degradation and contributed to CRPC proliferation [131].
Among other cancer types, AR is strongly associated with different forms of breast cancer and the roles of lncRNAs in AR signaling pathways in progression of breast cancer are being increasingly recognized [136,149,151]. Huang et al. built a regression analysis-based risk model to highlight a cluster of androgen-receptor signaling-pathway-related long non-coding RNAs (ARSP-related lncRNAs) that were strongly linked with the incidence and progression of breast cancer and were demonstrated to effectively predict the prognoses of breast cancer patients [151]. Yang et al. identified AR negatively induced lncRNA (ARNILA), which is transcriptionally repressed by AR and which itself functions as a sponge RNA for miR-204 to facilitate expression of its target gene Sox4, thereby promoting cancer progression, invasion and metastasis in triple negative breast cancer (TNBC) [136].

4.2. LncRNA Cross-Talks with ERs

ERs are a group of nuclear-localized or membrane-localized TFs that upon activation by steroid estrogens, bind to estrogen response elements (EREs) and modulate gene expression in various cellular processes and diseases including cancer. ERs and lncRNAs can mutually regulate each other to modulate various processes such as antiestrogen resistance in breast cancer [152]. Estrogen-regulated lncRNAs such as HOTAIR, MALAT1, MIAT, DSCAM-AS1, LINC00472, LINC01016, EGOT, LINC-ROR and LINP1 can modulate ER-dependent transcriptional changes, histone modifications and enhancer–promoter looping interactions [153] (Table 1). Among specific examples, Liang et al. showed that lncRNA DLGAP1 antisense RNA 2 (DLGAP1-AS2) was significantly upregulated in ER-positive breast cancers. DLGAP1-AS2 upregulation facilitated increased ER-signaling via the inhibition of AFF3 degradation, which in turn increased cell viability, inhibited apoptosis and induced resistance to the drug tamoxifen; DLGAP1-AS2 therefore offered a promising target for anti-breast cancer therapy aimed at overcoming tamoxifen resistance [137]. Miano et al. showed that ER-regulated lncRNA Down syndrome cell adhesion molecule antisense 1 (DSCAM-AS1) was highly expressed in Erα+ breast carcinoma and its expression correlated with EMT and invasiveness, suggesting that ER-DSCAM-AS1 interplay may play an important role in breast cancer [138].
ER may also modulate lncRNA transcription via attaching to genomic binding sites; Fang et al., for instance, highlighted an oncogenic role for estrogen-inducible lncRNA (ERINA) in ER-positive breast cancer. ERINA was transactivated by ER via an intronic ER-binding region and it promoted tumor proliferation and cell-cycle progression chiefly via binding to and preventing the interaction of TF E2F1 with tumor suppressor retinoblastoma protein (RB1). Since ERINA knockdown impeded cancer progression and tumorigenesis, it is a promising candidate as a breast cancer biomarker and target for anti-breast cancer therapy [139].
In contrast, the roles of lncRNAs in directly regulating ER-activity have also been reported. Yuan et al. performed a computational analysis of NGS data for ER-regulated and breast cancer-related lncRNAs and observed that Erα-regulated lncRNA 1 (ERLC1), a breast tissue specific lncRNA, was transcriptionally activated by Erα and in turn ERLC1 stabilized estrogen receptor 1 (ESR1) transcript to modulate Erα signaling, therefore emerging as a promising therapeutic target in breast cancer (Figure 2G) [140]. Among other examples, functional roles of lncRNA TMPO-AS1 in breast cancer progression via the stabilization of ESR1 mRNA expression have been reported [154]. It was also observed that ER-regulated lncRNA LINC00263 is differentially expressed in male and female patients [155]; these examples have indicated that ER-lncRNA-ESR1 interplay offers a promising avenue for targeted anti-breast cancer therapies.

4.3. LncRNA Cross-Talks with GRs

GR is a cytosol-localized factor that upon binding by glucocorticoids, either activates gene expression in the nucleus or prevents the translocation of other TFs to the nucleus, thereby repressing gene expression. As a quintessential example of a direct physical interaction-mediated lncRNA-NR cross-talk (Figure 2B), Kino et al. reported that lncRNA Gas5 can function as a GRE decoy by binding to the GR DNA-binding domain (DBD) and subsequently limiting GR binding to glucocorticoid response elements (GREs) in the target genes; this Gas5-GR interplay was demonstrated to modulate GR activity in HeLa cancer cells [156].

4.4. LncRNA Cross-Talks with PPARs

PPARs play important roles in regulating carbohydrate and fatty acid metabolism and are also implicated in many cancer types. LncRNA-PPAR cross-talks have been implicated in the progression of Hepatocellular carcinoma (HCC) (Table 1). Specifically, PPARα directly binds to the lncRNA Gm15441 promoter to upregulate its expression and attenuate hepatic inflammation by decreasing the NLR family pyrin domain containing 3 (NLRP3) inflammasome activation [157], which is known to play important roles in cancer [158]. Furthermore, the inhibition of PPARα enrichment on the lncRNA-APC promoter enforced its expression and inhibited colorectal carcinoma pathogenesis via the reduction in exosome production [159]. It was recently reported that lncRNA small nucleolar RNA host gene 1 (SNHG1) is expressed in several cancer types such as breast, colorectal, pancreatic and neuroblastoma [160,161,162] as a competitive endogenous RNA and can inhibit PPARγ expression to promote bladder cancer pathogenesis [163]. The role of lncRNA MALAT1 in cancer-associated cachexia through inhibition of adipogenesis via regulation of PPARγ has also been reported [164]. HULC (highly upregulated in liver cancer) is an lncRNA specifically overexpressed in HCC; HULC upregulates PPARα to activate acyl-CoA synthetase subunit ACSL1, thereby aberrantly modulating lipid metabolism in hepatoma cells and in the development of HCC [141].

4.5. LncRNA Cross-Talks with HNF4α

Hepatocyte nuclear factor 4 alpha (HNF4α) is the most abundant DNA-binding protein in liver and binds to DNA as a homodimer to regulate expression of genes in lipid metabolism; aberrant expression of HNF4α is a prominent feature in many diseases including cancer. Various lncRNAs can associate with HNF4α to modulate tumor progression in different cancer types (Table 1). Specifically, lncRNAs, such as LINC00858 in colon cancer, LOC100996425 in prostate cancer and LINC00511 in colorectal cancer, were shown to induce tumorigenesis and cancer progression via regulation of HNF4α and associated signaling pathways such as AMPK/mTOR [165,166,167]. Deng et al. showed that lncRNA SNHG16, which is involved in the pathogenesis of neuroblastoma, regulated miR-542-3p/HNF4 axis via the RAS/RAF/MEK/ERK signaling pathway [168]. Further, Jin et al. demonstrated that lncRNA HOTAIR can recruit SNAIL to HNF4α promoter to inhibit its expression and promote epithelial–mesenchymal transition of colorectal cancer [169]. Studies have also indicated that lncRNA-HNF4α cross-talks could be a therapeutic target for anticancer strategies. Chen et al. demonstrated that in invasive mucinous lung adenocarcinoma (IMA), HNF4α transactivated lncRNA BC200 (brain cytoplasmic 200 lncRNA), which in turn functioned as a scaffold for the fragile X messenger ribonucleoprotein 1 (FMR1), an mRNA-binding protein that stabilizes cancer-related mRNAs and HNF4α mRNA in a positive feedback loop (Figure 2D). Furthermore, mycophenolic acid (MPA), a component of the FDA-approved drug MMF (mycophenolate mofetil), functioned as an HNF4α antagonist to impede the progression of IMA, possibly by disrupting the HNF4α-BC200-FMR1 circuit and thereby providing a promising avenue for anti-IMA therapy [143].

4.6. LncRNA Cross-Talks with RAR

RARs form heterodimers with RXRs upon retinoid binding and bind to retinoid acid response elements (RARE) in the promoters of their target genes [170]. Some lncRNAs are known to associate with RAR signaling in cancer. Among the earliest examples, Zhao et al., using NB4, a promyelotic cell line, showed that the expression of HOXA cluster antisense RNA 2 (HOXA-AS2) was induced by RAR signaling in acute promyelocytic leukemia (APL), which in turn suppresses pro-apoptotic pathways [144]. Likewise, Haji et al. demonstrated that lncRNA H19 expression was upregulated in APL by RAR signaling, which in turn disrupted telomerase activity by interfering with the interaction between telomerase reverse transcriptase (hTERT) enzyme and telomerase RNA component (hTR), thereby contributing to tumor suppression [145]. Therefore, H19 offers an attractive target for therapeutic intervention in AML and related cancer types. Further examples of RAR/RXR-lncRNA cross-talks are summarized in Table 1. For a more detailed perspective of lncRNA-RAR/RXR cross-talks in cancer, readers are directed to refer to the review by Cantile et al. [148].

4.7. LncRNA Cross-Talks with LXR

Liver X receptor (LXR) belongs to a family of cholesterol ligand-activated TFs that are significant modulators of fatty acid and glucose metabolism. Wan et al. reported that an LXR agonist, LXR-623-induced lncRNA LINC01125, can suppress proliferation of breast cancer cells through the PTEN/AKT/p53 signaling pathway. This observation suggested that targeting NRs using receptor agonists such as LXR-623 may be a useful strategy for antitumor therapies [171].

4.8. LncRNA Cross-Talks as Therapeutic Targets

Therapeutic modulation of lncRNA-NR contacts to control the downstream processes and impede or even reverse cancer proliferation is an attractive, yet challenging task. There are multiple studies and applications towards targeting lncRNAs and NRs in diseases; in one such study, He at al. demonstrated the therapeutic potential for the antiangiogenic drug Sunitinib, which is widely used for renal cell carcinoma (RCC) therapy. The study showed that Sumatinib enhanced the expression of ECVSR lncRNA, thereby stabilizing ERβ mRNA. This stabilizing effect resulted in an increased ECVSR-ERβ-Hif2α signaling, leading to increased cancer stem cell (CSC) phenotype and vascular mimicry formation of RCC cells [172].

5. Multiomics Approach to Characterizing the Regulatory Networks Underlying lncRNA-NR Cross-Talks

While many lncRNAs-NR cross-talks in cancer have been individually characterized, a clear understanding of the general mechanisms underlying these associations remains elusive. LncRNAs are implicated in the regulation of gene expression typically via biomolecular interactions [173], and lncRNA-NR cross-talks, like all cellular networks, will often encompass different organizational levels involving interactions between RNAs, DNA/chromatin, proteins and small molecule metabolites, among others. Therefore, properly characterizing lncRNA-NR cross-talks is a non-trivial task. To actively advance our understanding of the lncRNA-NR cross-regulatory networks in cancer, these associations should preferably be visualized through multiple lenses. To achieve this, it is necessary to employ a holistic multiomics approach combining genome-wide RNA-chromatin interactions, chromatin–chromatin (DNA-DNA) interactions, protein–protein interactions (PPIs), RNA–protein interactions (RPIs), RNA-RNA interactions and small molecule (chemical) interactions with global gene expression (transcriptome) data to assemble lncRNA-NR gene regulatory networks (GRNs) to characterize lncRNA-NR cross-talks in different cellular contexts, pinpoint key modulators of these processes and prioritize candidates for experimental validation, biomarker discovery and therapeutic intervention in diseases such as cancer (Figure 3).

5.1. High-Throughput Omics Methodologies

Different omics platforms provide distinct high-throughput data on different organizational levels of a cell or an organism. Therefore, an integrative multiomics approach that leverages multiple omics data-types together with bioinformatics tools for data analysis and pattern discovery is well suited to characterize different layers of lncRNA-NR cross-regulatory networks.

5.2. Transcriptome

The entirety of the RNA population within a cell, tissue or a living system is referred to as the transcriptome and the study of the transcriptome is known as transcriptomics. The two most widely used techniques to profile the transcriptome are microarrays and RNA-seq [174]. Both lncRNAs and TFs such as NRs display cell-specific expression; hence, global transcriptome profiling can help identify sets of coexpressed lncRNAs and NR-coding genes in specific cancer types (Figure 3). However, it is also well established that lncRNAs, in general, have more restricted expression than protein-coding messenger RNAs (mRNAs); this is in part attributed to high cellular specificity of lncRNA expression and disproportionately high expression of lncRNAs in cellular subpopulations [1]. Therefore, global transcriptome profiling coupled with single-cell RNA-seq analysis is well suited to profile lncRNA and TF (including NRs) coexpression in specific cancer types. One such initiative is the RNA Atlas, which combines multiple RNA sequencing technologies together with lncRNA-target prediction to expand the catalog of heterogeneous transcripts in the human transcriptome and analyze correlations with lncRNAs and TFs such as NRs [175].

5.3. Biomolecular Interactions Underlying lncRNA-NR Cross-Talks

Various high-throughput experiments can offer detailed insights into the different types of biomolecular associations that typically underlie lncRNA-NR cross-talks. Technologies such as cross-linking and immunoprecipitation (IP) followed by sequencing (CLIP-seq) and associated methods can potentially capture direct binding between RNAs such as lncRNAs and proteins such as NRs [176]. Gene regulatory interactions between lncRNAs and NRs can be profiled on a genome-wide scale by using technologies that capture protein–DNA interactions [177] and those that profile RNA–chromatin interactions [27]; these include one-to-many methods such as CHIRP-seq [178] and CHART-seq [179] and many-to-many methods such as RADICL-seq [180] and GRID-seq [181], among others. PPIs between NRs and other cellular proteins can be mapped using different experimental techniques such as the yeast two-hybrid system (Y2H) [182], affinity purification–mass spectrometry (AP-MS) for one-to-many PPI mapping [183] and cofractionation and mass spectrometry [184] and PROPER-seq, for the mapping of many-to-many PPIs [185]. Chromatin–chromatin interactions involving lncRNA and NR loci can be mapped using chromosome conformation capture techniques such as Hi-C [186], while lncRNA-NR mRNA interactions can be characterized with methods such as RIC-seq [187].

5.4. Computational Tools

Computational and statistical methods to assess huge volumes of genomics and transcriptomics data have been pivotal to new discoveries in RNA biology. For instance, computational methods such as catRAPID [188] can be used to rapidly predict RNA-protein (NR) interactions on a global scale, which can then be prioritized for experimental validation. Likewise, in silico methods for PPI prediction [189,190] can complement and qualitatively assess PPIs likely to play important roles in lncRNA-NR cross-talks. However, individual analytical methods, though useful in their own right, will provide only specific insights into the many layers that make up the lncRNA-NR cross-regulatory networks. A multiomics data analysis approach, which seeks to combine different omics data types and different tools for their analysis into a unified analytical framework, is better suited to understand the depth of lncRNA-NR cross-talks. These methods for data classification and categorization can take many forms and differ in scope and content but can broadly be viewed as falling into two categories—unsupervised (such as clustering [191] and matrix factorization [192]) and supervised (such as network modeling [193] and most machine-learning algorithms [194]). For a detailed overview of integrative omics methods for building lncRNA gene regulatory networks and knowledge discovery, readers are directed to reviews by Tripathi et al. [195] and Zhao et al. [2].

6. Conclusions and Perspectives

LncRNAs can regulate NR signaling either individually or in conjunction with other coregulators in a context-dependent manner and in feedback loops. Multiple studies have increasingly highlighted the cross-talks between lncRNAs and NRs in the regulation of chromatin modifications and cellular processes involved in tumor progression, resistance and metastasis. Emerging studies have also highlighted the roles of lncRNA-NR cross-talks in metabolic reprogramming of cells, which can lead to cancer progression. Dysregulation of NR-mediated signaling is observed in a variety of hormone-dependent cancers. Studies have also suggested roles of NR-associated lncRNAs in cancer as prognostic biomarkers and in the development of novel treatment options. Since most of the genome is composed of lncRNAs, exploring lncRNAs and NR-interactions can provide a wider range of prognostic and therapeutic opportunities, especially in the treatment of hormone-dependent cancers such as breast and prostate cancers. A deeper understanding of lncRNA-NR cross-talks can best be obtained from a combined approach that integrates different data types in a multiomics framework. Consequently, computational tools that can facilitate integrative analysis of multidimensional omics data to infer representative lncRNA-NR regulatory networks will help gain more knowledge about these associations and help prioritize targets for experimental and clinical validation.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers16162920/s1, Table S1: Selected lncRNAs and NRs associated with cancers.

Author Contributions

P.T. and L.P.T. conceived and designed the review. P.T. and L.P.T. were responsible for the data collection and summary. Both P.T. and L.P.T. contributed to drafting the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors acknowledge institutional support from the RIKEN Center for Integrative Medical Sciences. No explicit funding was sought or provided for this study.

Conflicts of Interest

The authors declare no competing financial interests.

References

  1. 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]
  2. Zhao, X.; Lan, Y.; Chen, D. Exploring long non-coding RNA networks from single cell omics data. Comput. Struct. Biotechnol. J. 2022, 20, 4381–4389. [Google Scholar] [CrossRef]
  3. Ghafouri-Fard, S.; Shoorei, H.; Taheri, M.; Sanak, M. Emerging role of non-coding RNAs in allergic disorders. Biomed. Pharmacother. 2020, 130, 110615. [Google Scholar] [CrossRef] [PubMed]
  4. Kirk, J.M.; Kim, S.O.; Inoue, K.; Smola, M.J.; Lee, D.M.; Schertzer, M.D.; Wooten, J.S.; Baker, A.R.; Sprague, D.; Collins, D.W.; et al. Functional classification of long non-coding RNAs by k-mer content. Nat. Genet. 2018, 50, 1474–1482. [Google Scholar] [CrossRef]
  5. Carlevaro-Fita, J.; Johnson, R. Global Positioning System: Understanding Long Noncoding RNAs through Subcellular Localization. Mol. Cell 2019, 73, 869–883. [Google Scholar] [CrossRef] [PubMed]
  6. Graf, J.; Kretz, M. From structure to function: Route to understanding lncRNA mechanism. BioEssays 2020, 42, 2000027. [Google Scholar] [CrossRef] [PubMed]
  7. Xia, W.; He, Y.; Gan, Y.; Zhang, B.; Dai, G.; Ru, F.; Jiang, Z.; Chen, Z.; Chen, X. Long Non-coding RNA: An Emerging Contributor and Potential Therapeutic Target in Renal Fibrosis. Front. Genet. 2021, 12, 682904. [Google Scholar] [CrossRef] [PubMed]
  8. Peng, Z.; Liu, C.; Wu, M. New insights into long noncoding RNAs and their roles in glioma. Mol. Cancer 2018, 17, 61. [Google Scholar] [CrossRef]
  9. Anastasiadou, E.; Jacob, L.S.; Slack, F.J. Non-coding RNA networks in cancer. Nat. Rev. Cancer 2018, 18, 5–18. [Google Scholar] [CrossRef]
  10. Slack, F.J.; Chinnaiyan, A.M. The Role of Non-coding RNAs in Oncology. Cell 2019, 179, 1033–1055. [Google Scholar] [CrossRef]
  11. Rey, F.; Zuccotti, G.V.; Carelli, S. Long non-coding RNAs in metabolic diseases: From bench to bedside. Trends Endocrinol. Metab. 2021, 32, 747–749. [Google Scholar] [CrossRef]
  12. Geng, H.; Tan, X.-D. Functional diversity of long non-coding RNAs in immune regulation. Genes Dis. 2016, 3, 72–81. [Google Scholar] [CrossRef]
  13. Ghafouri-Fard, S.; Askari, A.; Hussen, B.M.; Taheri, M.; Mokhtari, M. A long non-coding RNA with important roles in the carcinogenesis. Front. Cell Dev. Biol. 2022, 10, 1037149. [Google Scholar] [CrossRef] [PubMed]
  14. Long, Y.; Wang, X.; Youmans, D.T.; Cech, T.R. How do lncRNAs regulate transcription? Sci. Adv. 2017, 3, eaao2110. [Google Scholar] [CrossRef] [PubMed]
  15. Noguchi, M.; Shimizu, M.; Lu, P.; Takahashi, Y.; Yamauchi, Y.; Sato, S.; Kiyono, H.; Kishino, S.; Ogawa, J.; Nagata, K.; et al. Lactic acid bacteria-derived γ-linolenic acid metabolites are PPARδ ligands that reduce lipid accumulation in human intestinal organoids. J. Biol. Chem. 2022, 298, 102534. [Google Scholar] [CrossRef]
  16. Umemoto, T.; Fujiki, Y. Ligand-dependent nucleo-cytoplasmic shuttling of peroxisome proliferator-activated receptors, PPARα and PPARγ. Genes Cells 2012, 17, 576–596. [Google Scholar] [CrossRef]
  17. Levin, E.R.; Hammes, S.R. Nuclear receptors outside the nucleus: Extranuclear signalling by steroid receptors. Nat. Rev. Mol. Cell Biol. 2016, 17, 783–797. [Google Scholar] [CrossRef] [PubMed]
  18. de Vera, I.M.S. Advances in Orphan Nuclear Receptor Pharmacology: A New Era in Drug Discovery. ACS Pharmacol. Transl. Sci. 2018, 1, 134–137. [Google Scholar] [CrossRef]
  19. Pawlak, M.; Lefebvre, P.; Staels, B. General molecular biology and architecture of nuclear receptors. Curr. Top. Med. Chem. 2012, 12, 486–504. [Google Scholar] [CrossRef]
  20. Sun, M.; Kraus, W.L. From Discovery to Function: The Expanding Roles of Long NonCoding RNAs in Physiology and Disease. Endocr. Rev. 2015, 36, 25–64. [Google Scholar] [CrossRef]
  21. Jafari, H.; Hussain, S.; Campbell, M.J. Nuclear Receptor Coregulators in Hormone-Dependent Cancers. Cancers 2022, 14, 2402. [Google Scholar] [CrossRef] [PubMed]
  22. Sheng, L.; Ye, L.; Zhang, D.; Cawthorn, W.P.; Xu, B. New Insights into the Long Non-coding RNA SRA: Physiological Functions and Mechanisms of Action. Front. Med. 2018, 5, 244. [Google Scholar] [CrossRef] [PubMed]
  23. Ma, L.; Bajic, V.B.; Zhang, Z. On the classification of long non-coding RNAs. RNA Biol. 2013, 10, 924–933. [Google Scholar] [CrossRef] [PubMed]
  24. Laurent, G., St.; Wahlestedt, C.; Kapranov, P. The Landscape of long noncoding RNA classification. Trends Genet. 2015, 31, 239–251. [Google Scholar] [CrossRef] [PubMed]
  25. Zampetaki, A.; Albrecht, A.; Steinhofel, K. Corrigendum: Long Non-coding RNA Structure and Function: Is There a Link? Front. Physiol. 2019, 10, 1127. [Google Scholar] [CrossRef]
  26. Kazimierczyk, M.; Kasprowicz, M.K.; Kasprzyk, M.E.; Wrzesinski, J. Human Long Noncoding RNA Interactome: Detection, Characterization and Function. Int. J. Mol. Sci. 2020, 21, 1027. [Google Scholar] [CrossRef]
  27. Kato, M.; Carninci, P. Genome-Wide Technologies to Study RNA-Chromatin Interactions. Non-Coding RNA 2020, 6, 20. [Google Scholar] [CrossRef]
  28. Chillón, I.; Marcia, M. The molecular structure of long non-coding RNAs: Emerging patterns and functional implications. Crit. Rev. Biochem. Mol. Biol. 2020, 55, 662–690. [Google Scholar] [CrossRef]
  29. Núñez-Martínez, H.N.; Recillas-Targa, F. Emerging Functions of lncRNA Loci beyond the Transcript Itself. Int. J. Mol. Sci. 2022, 23, 6258. [Google Scholar] [CrossRef]
  30. Chen, J.; Wang, Y.; Wang, C.; Hu, J.F.; Li, W. LncRNA Functions as a New Emerging Epigenetic Factor in Determining the Fate of Stem Cells. Front. Genet. 2020, 11, 277. [Google Scholar] [CrossRef]
  31. Chen, H.; Du, G.; Song, X.; Li, L. Non-coding Transcripts from Enhancers: New Insights into Enhancer Activity and Gene Expression Regulation. Genom. Proteom. Bioinform. 2017, 15, 201–207. [Google Scholar] [CrossRef] [PubMed]
  32. Isoda, T.; Moore, A.J.; He, Z.; Chandra, V.; Aida, M.; Denholtz, M.; van Hamburg, P.J.; Fisch, K.M.; Chang, A.N.; Fahl, S.P.; et al. Non-coding Transcription Instructs Chromatin Folding and Compartmentalization to Dictate Enhancer-Promoter Communication and T Cell Fate. Cell 2017, 171, 103–119.e118. [Google Scholar] [CrossRef] [PubMed]
  33. Yang, F.; Deng, X.; Ma, W.; Berletch, J.B.; Rabaia, N.; Wei, G.; Moore, J.M.; Filippova, G.N.; Xu, J.; Liu, Y.; et al. The lncRNA Firre anchors the inactive X chromosome to the nucleolus by binding CTCF and maintains H3K27me3 methylation. Genome Biol. 2015, 16, 52. [Google Scholar] [CrossRef] [PubMed]
  34. Leisegang, M.S.; Warwick, T.; Stötzel, J.; Brandes, R.P. RNA-DNA triplexes: Molecular mechanisms and functional relevance. Trends Biochem. Sci. 2024, 49, 532–544. [Google Scholar] [CrossRef]
  35. Tan-Wong, S.M.; Dhir, S.; Proudfoot, N.J. R-Loops Promote Antisense Transcription across the Mammalian Genome. Mol. Cell 2019, 76, 600–616.e606. [Google Scholar] [CrossRef]
  36. Engreitz, J.M.; Pandya-Jones, A.; McDonel, P.; Shishkin, A.; Sirokman, K.; Surka, C.; Kadri, S.; Xing, J.; Goren, A.; Lander, E.S.; et al. The Xist lncRNA exploits three-dimensional genome architecture to spread across the X chromosome. Science 2013, 341, 1237973. [Google Scholar] [CrossRef]
  37. Cesana, M.; Cacchiarelli, D.; Legnini, I.; Santini, T.; Sthandier, O.; Chinappi, M.; Tramontano, A.; Bozzoni, I. A long noncoding RNA controls muscle differentiation by functioning as a competing endogenous RNA. Cell 2011, 147, 358–369. [Google Scholar] [CrossRef]
  38. Jain, A.K.; Xi, Y.; McCarthy, R.; Allton, K.; Akdemir, K.C.; Patel, L.R.; Aronow, B.; Lin, C.; Li, W.; Yang, L.; et al. LncPRESS1 Is a p53-Regulated LncRNA that Safeguards Pluripotency by Disrupting SIRT6-Mediated De-acetylation of Histone H3K56. Mol. Cell 2016, 64, 967–981. [Google Scholar] [CrossRef]
  39. Werner, A.; Kanhere, A.; Wahlestedt, C.; Mattick, J.S. Natural antisense transcripts as versatile regulators of gene expression. Nat. Rev. Genet. 2024, 1–15. [Google Scholar] [CrossRef]
  40. Ouyang, J.; Zhong, Y.; Zhang, Y.; Yang, L.; Wu, P.; Hou, X.; Xiong, F.; Li, X.; Zhang, S.; Gong, Z.; et al. Long non-coding RNAs are involved in alternative splicing and promote cancer progression. Br. J. Cancer 2022, 126, 1113–1124. [Google Scholar] [CrossRef]
  41. Statello, L.; Guo, C.J.; Chen, L.L.; Huarte, M. Gene regulation by long non-coding RNAs and its biological functions. Nat. Rev. Mol. Cell Biol. 2021, 22, 96–118. [Google Scholar] [CrossRef] [PubMed]
  42. Yip, C.W.; Hon, C.-C.; Yasuzawa, K.; Sivaraman, D.M.; Ramilowski, J.A.; Shibayama, Y.; Agrawal, S.; Prabhu, A.V.; Parr, C.; Severin, J.; et al. Antisense-oligonucleotide-mediated perturbation of long non-coding RNA reveals functional features in stem cells and across cell types. Cell Rep. 2022, 41, 111893. [Google Scholar] [CrossRef] [PubMed]
  43. Ramilowski, J.A.; Yip, C.W.; Agrawal, S.; Chang, J.-C.; Ciani, Y.; Kulakovskiy, I.V.; Mendez, M.; Ooi, J.L.C.; Ouyang, J.F.; Parkinson, N.; et al. Corrigendum: Functional annotation of human long noncoding RNAs via molecular phenotyping. Genome Res. 2020, 30, 1060–1072. [Google Scholar] [CrossRef] [PubMed]
  44. Zhao, S.; Zhang, X.; Chen, S.; Zhang, S. Long noncoding RNAs: Fine-tuners hidden in the cancer signaling network. Cell Death Discov. 2021, 7, 283. [Google Scholar] [CrossRef]
  45. Ahmad, S.; Abbas, M.; Ullah, M.F.; Aziz, M.H.; Beylerli, O.; Alam, M.A.; Syed, M.A.; Uddin, S.; Ahmad, A. Long non-coding RNAs regulated NF-κB signaling in cancer metastasis: Micromanaging by not so small non-coding RNAs. Semin. Cancer Biol. 2022, 85, 155–163. [Google Scholar] [CrossRef]
  46. Sun, Y.; Ma, L. New Insights into Long Non-Coding RNA MALAT1 in Cancer and Metastasis. Cancers 2019, 11, 216. [Google Scholar] [CrossRef]
  47. Park, M.K.; Zhang, L.; Min, K.W.; Cho, J.H.; Yeh, C.C.; Moon, H.; Hormaechea-Agulla, D.; Mun, H.; Ko, S.; Lee, J.W.; et al. NEAT1 is essential for metabolic changes that promote breast cancer growth and metastasis. Cell Metab. 2021, 33, 2380–2397.e9. [Google Scholar] [CrossRef]
  48. Wen, S.; Wei, Y.; Zen, C.; Xiong, W.; Niu, Y.; Zhao, Y. Long non-coding RNA NEAT1 promotes bone metastasis of prostate cancer through N6-methyladenosine. Mol. Cancer 2020, 19, 171. [Google Scholar] [CrossRef]
  49. Carlevaro-Fita, J.; Lanzós, A.; Feuerbach, L.; Hong, C.; Mas-Ponte, D.; Pedersen, J.S.; Abascal, F.; Amin, S.B.; Bader, G.D.; Barenboim, J.; et al. Cancer LncRNA Census reveals evidence for deep functional conservation of long noncoding RNAs in tumorigenesis. Commun. Biol. 2020, 3, 56. [Google Scholar] [CrossRef]
  50. Lanzós, A.; Carlevaro-Fita, J.; Mularoni, L.; Reverter, F.; Palumbo, E.; Guigó, R.; Johnson, R. Discovery of Cancer Driver Long Noncoding RNAs across 1112 Tumour Genomes: New Candidates and Distinguishing Features. Sci. Rep. 2017, 7, 41544. [Google Scholar] [CrossRef]
  51. Rheinbay, E.; Nielsen, M.M.; Abascal, F.; Wala, J.A.; Shapira, O.; Tiao, G.; Hornshøj, H.; Hess, J.M.; Juul, R.I.; Lin, Z.; et al. Analyses of non-coding somatic drivers in 2658 cancer whole genomes. Nature 2020, 578, 102–111. [Google Scholar] [CrossRef] [PubMed]
  52. Ghafouri-Fard, S.; Shoorei, H.; Anamag, F.T.; Taheri, M. The Role of Non-Coding RNAs in Controlling Cell Cycle Related Proteins in Cancer Cells. Front. Oncol. 2020, 10, 608975. [Google Scholar] [CrossRef] [PubMed]
  53. Chen, S.; Zhu, J.; Wang, F.; Guan, Z.; Ge, Y.; Yang, X.; Cai, J. LncRNAs and their role in cancer stem cells. Oncotarget 2017, 8, 110685. [Google Scholar] [CrossRef]
  54. Schwerdtfeger, M.; Desiderio, V.; Kobold, S.; Regad, T.; Zappavigna, S.; Caraglia, M. Long non-coding RNAs in cancer stem cells. Transl. Oncol. 2021, 14, 101134. [Google Scholar] [CrossRef] [PubMed]
  55. Lim, S.M.; Mohamad Hanif, E.A.; Chin, S.-F. Is targeting autophagy mechanism in cancer a good approach? The possible double-edge sword effect. Cell Biosci. 2021, 11, 56. [Google Scholar] [CrossRef]
  56. Yang, L.; Wang, H.; Shen, Q.; Feng, L.; Jin, H. Long non-coding RNAs involved in autophagy regulation. Cell Death Dis. 2017, 8, e3073. [Google Scholar] [CrossRef]
  57. Wan, J.; Guo, C.; Fang, H.; Xu, Z.; Hu, Y.; Luo, Y. Autophagy-Related Long Non-coding RNA Is a Prognostic Indicator for Bladder Cancer. Front. Oncol. 2021, 11, 647236. [Google Scholar] [CrossRef]
  58. Hiam-Galvez, K.J.; Allen, B.M.; Spitzer, M.H. Systemic immunity in cancer. Nat. Rev. Cancer 2021, 21, 345–359. [Google Scholar] [CrossRef]
  59. Luo, H.; Bu, D.; Shao, L.; Li, Y.; Sun, L.; Wang, C.; Wang, J.; Yang, W.; Yang, X.; Dong, J.; et al. Single-cell Long Non-coding RNA Landscape of T Cells in Human Cancer Immunity. Genom. Proteom. Bioinform. 2021, 19, 377–393. [Google Scholar] [CrossRef]
  60. Xia, L.; Oyang, L.; Lin, J.; Tan, S.; Han, Y.; Wu, N.; Yi, P.; Tang, L.; Pan, Q.; Rao, S.; et al. The cancer metabolic reprogramming and immune response. Mol. Cancer 2021, 20, 28. [Google Scholar] [CrossRef]
  61. Biswas, S.K. Metabolic Reprogramming of Immune Cells in Cancer Progression. Immunity 2015, 43, 435–449. [Google Scholar] [CrossRef] [PubMed]
  62. Zhang, Y.; Mao, Q.; Xia, Q.; Cheng, J.; Huang, Z.; Li, Y.; Chen, P.; Yang, J.; Fan, X.; Liang, Y.; et al. Noncoding RNAs link metabolic reprogramming to immune microenvironment in cancers. J. Hematol. Oncol. 2021, 14, 169. [Google Scholar] [CrossRef]
  63. Fan, C.; Tang, Y.; Wang, J.; Xiong, F.; Guo, C.; Wang, Y.; Zhang, S.; Gong, Z.; Wei, F.; Yang, L.; et al. Role of long non-coding RNAs in glucose metabolism in cancer. Mol. Cancer 2017, 16, 130. [Google Scholar] [CrossRef] [PubMed]
  64. Bhan, A.; Soleimani, M.; Mandal, S.S. Long Noncoding RNA and Cancer: A New Paradigm. Cancer Res. 2017, 77, 3965–3981. [Google Scholar] [CrossRef] [PubMed]
  65. Shaath, H.; Vishnubalaji, R.; Elango, R.; Khattak, S.; Alajez, N.M. Single-cell long noncoding RNA (lncRNA) transcriptome implicates MALAT1 in triple-negative breast cancer (TNBC) resistance to neoadjuvant chemotherapy. Cell Death Discov. 2021, 7, 23. [Google Scholar] [CrossRef]
  66. Esposito, R.; Polidori, T.; Meise, D.F.; Pulido-Quetglas, C.; Chouvardas, P.; Forster, S.; Schaerer, P.; Kobel, A.; Schlatter, J.; Kerkhof, E.; et al. Multi-hallmark long noncoding RNA maps reveal non-small cell lung cancer vulnerabilities. Cell Genom. 2022, 2, 100171. [Google Scholar] [CrossRef]
  67. Nath, A.; Lau, E.Y.T.; Lee, A.M.; Geeleher, P.; Cho, W.C.S.; Huang, R.S. Discovering long noncoding RNA predictors of anticancer drug sensitivity beyond protein-coding genes. Proc. Natl. Acad. Sci. USA 2019, 116, 22020–22029. [Google Scholar] [CrossRef]
  68. 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]
  69. Liu, K.; Gao, L.; Ma, X.; Huang, J.-J.; Chen, J.; Zeng, L.; Ashby, C.R.; Zou, C.; Chen, Z.-S. Long non-coding RNAs regulate drug resistance in cancer. Mol. Cancer 2020, 19, 54. [Google Scholar] [CrossRef]
  70. Winkle, M.; El-Daly, S.M.; Fabbri, M.; Calin, G.A. Noncoding RNA therapeutics—Challenges and potential solutions. Nat. Rev. Drug Discov. 2021, 20, 629–651. [Google Scholar] [CrossRef]
  71. Wu, M.; Zhang, X.; Han, X.; Pandey, V.; Lobie, P.E.; Zhu, T. The potential of long noncoding RNAs for precision medicine in human cancer. Cancer Lett. 2021, 501, 12–19. [Google Scholar] [CrossRef] [PubMed]
  72. Gao, Y.; Shang, S.; Guo, S.; Li, X.; Zhou, H.; Liu, H.; Sun, Y.; Wang, J.; Wang, P.; Zhi, H.; et al. Lnc2Cancer 3.0: An updated resource for experimentally supported lncRNA/circRNA cancer associations and web tools based on RNA-seq and scRNA-seq data. Nucleic Acids Res. 2021, 49, D1251–D1258. [Google Scholar] [CrossRef] [PubMed]
  73. Porter, B.A.; Ortiz, M.A.; Bratslavsky, G.; Kotula, L. Structure and Function of the Nuclear Receptor Superfamily and Current Targeted Therapies of Prostate Cancer. Cancers 2019, 11, 1852. [Google Scholar] [CrossRef] [PubMed]
  74. Luan, Z.L.; Zhang, C.; Ming, W.H.; Huang, Y.Z.; Guan, Y.F.; Zhang, X.Y. Nuclear receptors in renal health and disease. EBioMedicine 2022, 76, 103855. [Google Scholar] [CrossRef]
  75. Zhao, Y.; Zhang, K.; Giesy, J.P.; Hu, J. Families of Nuclear Receptors in Vertebrate Models: Characteristic and Comparative Toxicological Perspective. Sci. Rep. 2015, 5, 8554. [Google Scholar] [CrossRef]
  76. Sever, R.; Glass, C.K. Signaling by Nuclear Receptors. Cold Spring Harb. Perspect. Biol. 2013, 5, a016709. [Google Scholar] [CrossRef]
  77. Arnal, J.-F.; Lenfant, F.; Metivier, R.; Flouriot, G.; Henrion, D.; Adlanmerini, M.; Fontaine, C.; Gourdy, P.; Chambon, P.; Katzenellenbogen, B.; et al. Membrane and Nuclear Estrogen Receptor Alpha Actions: From Tissue Specificity to Medical Implications. Physiol. Rev. 2017, 97, 1045–1087. [Google Scholar] [CrossRef]
  78. Rigano, D.; Sirignano, C.; Taglialatela-Scafati, O. The potential of natural products for targeting PPARα. Acta Pharm. Sin. B 2017, 7, 427–438. [Google Scholar] [CrossRef]
  79. Penvose, A.; Keenan, J.L.; Bray, D.; Ramlall, V.; Siggers, T. Comprehensive study of nuclear receptor DNA binding provides a revised framework for understanding receptor specificity. Nat. Commun. 2019, 10, 2514. [Google Scholar] [CrossRef]
  80. Li, B.; Cai, S.-Y.; Boyer, J.L. The role of the retinoid receptor, RAR/RXR heterodimer, in liver physiology. Biochim. Biophys. Acta Mol. Basis Dis. 2021, 1867, 166085. [Google Scholar] [CrossRef]
  81. Decara, J.; Rivera, P.; López-Gambero, A.J.; Serrano, A.; Pavón, F.J.; Baixeras, E.; Rodríguez de Fonseca, F.; Suárez, J. Peroxisome Proliferator-Activated Receptors: Experimental Targeting for the Treatment of Inflammatory Bowel Diseases. Front. Pharmacol. 2020, 11, 730. [Google Scholar] [CrossRef]
  82. Saika, A.; Nagatake, T.; Hirata, S.-i.; Sawane, K.; Adachi, J.; Abe, Y.; Isoyama, J.; Morimoto, S.; Node, E.; Tiwari, P.; et al. ω3 fatty acid metabolite, 12-hydroxyeicosapentaenoic acid, alleviates contact hypersensitivity by downregulation of CXCL1 and CXCL2 gene expression in keratinocytes via retinoid X receptor α. FASEB J. 2021, 35, e21354. [Google Scholar] [CrossRef]
  83. Saika, A.; Tiwari, P.; Nagatake, T.; Node, E.; Hosomi, K.; Honda, T.; Kabashima, K.; Kunisawa, J. Mead acid inhibits retinol-induced irritant contact dermatitis via peroxisome proliferator-activated receptor alpha. Front. Mol. Biosci. 2023, 10, 1097955. [Google Scholar] [CrossRef] [PubMed]
  84. Nagatake, T.; Kishino, S.; Urano, E.; Murakami, H.; Kitamura, N.; Konishi, K.; Ohno, H.; Tiwari, P.; Morimoto, S.; Node, E.; et al. Intestinal microbe-dependent ω3 lipid metabolite αKetoA prevents inflammatory diseases in mice and cynomolgus macaques. Mucosal Immunol. 2022, 15, 289–300. [Google Scholar] [CrossRef] [PubMed]
  85. Dubois, V.; Eeckhoute, J.; Lefebvre, P.; Staels, B. Distinct but complementary contributions of PPAR isotypes to energy homeostasis. J. Clin. Investig. 2017, 127, 1202–1214. [Google Scholar] [CrossRef]
  86. Hong, F.; Xu, P.; Zhai, Y. The Opportunities and Challenges of Peroxisome Proliferator-Activated Receptors Ligands in Clinical Drug Discovery and Development. Int. J. Mol. Sci. 2018, 19, 2189. [Google Scholar] [CrossRef] [PubMed]
  87. Korbecki, J.; Bobiński, R.; Dutka, M. Self-regulation of the inflammatory response by peroxisome proliferator-activated receptors. Inflamm. Res. 2019, 68, 443–458. [Google Scholar] [CrossRef]
  88. Moraitis, A.N.; Giguère, V. Transition from Monomeric to Homodimeric DNA Binding by Nuclear Receptors: Identification of RevErbAα Determinants Required for RORα Homodimer Complex Formation. Mol. Endocrinol. 1999, 13, 431–439. [Google Scholar] [CrossRef]
  89. Millard, C.J.; Watson, P.J.; Fairall, L.; Schwabe, J.W.R. An evolving understanding of nuclear receptor coregulator proteins. J. Mol. Endocrinol. 2013, 51, T23–T36. [Google Scholar] [CrossRef]
  90. Gadaleta, R.M.; Magnani, L. Nuclear receptors and chromatin: An inducible couple. J. Mol. Endocrinol. 2014, 52, R137–R149. [Google Scholar] [CrossRef]
  91. Beinsteiner, B.A.-O.; Markov, G.A.-O.; Erb, S.A.-O.X.; Chebaro, Y.A.-O.; McEwen, A.A.-O.; Cianférani, S.; Laudet, V.A.-O.; Moras, D.A.-O.; Billas, I.A.-O. A structural signature motif enlightens the origin and diversification of nuclear receptors. PLoS Genet. 2021, 17, e1009492. [Google Scholar] [CrossRef]
  92. Alatshan, A.; Benkő, S. Nuclear Receptors as Multiple Regulators of NLRP3 Inflammasome Function. Front. Immunol. 2021, 12, 630569. [Google Scholar] [CrossRef] [PubMed]
  93. Zhao, L.; Zhou, S.; Gustafsson, J.-Å. Nuclear Receptors: Recent Drug Discovery for Cancer Therapies. Endocr. Rev. 2019, 40, 1207–1249. [Google Scholar] [CrossRef] [PubMed]
  94. Lv, D.-D.; Zhou, L.-Y.; Tang, H. Hepatocyte nuclear factor 4α and cancer-related cell signaling pathways: A promising insight into cancer treatment. Exp. Mol. Med. 2021, 53, 8–18. [Google Scholar] [CrossRef]
  95. Dana, N.A.-O.; Vaseghi, G.A.-O.; Haghjooy Javanmard, S.A.-O. Crosstalk between Peroxisome Proliferator-Activated Receptors and Toll-Like Receptors: A Systematic Review. Adv. Pharm. Bull. 2019, 9, 12. [Google Scholar] [CrossRef]
  96. Karamouzis, M.V.; Papavassiliou, K.A.; Adamopoulos, C.; Papavassiliou, A.G. Targeting Androgen/Estrogen Receptors Crosstalk in Cancer. Trends Cancer 2016, 2, 35–48. [Google Scholar] [CrossRef]
  97. Michmerhuizen, A.R.; Lerner, L.M.; Ward, C.; Pesch, A.M.; Zhang, A.; Schwartz, R.; Wilder-Romans, K.; Eisner, J.R.; Rae, J.M.; Pierce, L.J.; et al. Androgen and oestrogen receptor co-expression determines the efficacy of hormone receptor-mediated radiosensitisation in breast cancer. Br. J. Cancer 2022, 127, 927–936. [Google Scholar] [CrossRef]
  98. Kach, J.; Conzen, S.D.; Szmulewitz, R.Z. Targeting the glucocorticoid receptor in breast and prostate cancers. Sci. Transl. Med. 2015, 7, 305ps19. [Google Scholar] [CrossRef] [PubMed]
  99. Mayayo-Peralta, I.; Zwart, W.; Prekovic, S. Duality of glucocorticoid action in cancer: Tumor-suppressor or oncogene? Endocr.-Relat. Cancer 2021, 28, R157–R171. [Google Scholar] [CrossRef]
  100. Prekovic, S.; Schuurman, K.; Mayayo-Peralta, I.; Manjón, A.G.; Buijs, M.; Yavuz, S.; Wellenstein, M.D.; Barrera, A.; Monkhorst, K.; Huber, A.; et al. Glucocorticoid receptor triggers a reversible drug-tolerant dormancy state with acquired therapeutic vulnerabilities in lung cancer. Nat. Commun. 2021, 12, 4360. [Google Scholar] [CrossRef]
  101. Zhu, M.L.; Kyprianou, N. Androgen receptor and growth factor signaling cross-talk in prostate cancer cells. Endocr. Relat. Cancer 2008, 15, 841–849. [Google Scholar] [CrossRef]
  102. Ghafouri-Fard, S.; Abak, A.; Mohaqiq, M.; Shoorei, H.; Taheri, M. The Interplay Between Non-coding RNAs and Insulin-Like Growth Factor Signaling in the Pathogenesis of Neoplasia. Front. Cell Dev. Biol. 2021, 9, 634512. [Google Scholar] [CrossRef]
  103. Kerr, A.; Baxter, R.C. Noncoding RNA actions through IGFs and IGF binding proteins in cancer. Oncogene 2022, 41, 3385–3393. [Google Scholar] [CrossRef] [PubMed]
  104. Matsushita, M.; Fujita, K.; Hatano, K.; De Velasco, M.A.; Uemura, H.; Nonomura, N. Connecting the Dots Between the Gut–IGF-1–Prostate Axis: A Role of IGF-1 in Prostate Carcinogenesis. Front. Endocrinol. 2022, 13, 852382. [Google Scholar] [CrossRef] [PubMed]
  105. Levin, E.R. Bidirectional Signaling between the Estrogen Receptor and the Epidermal Growth Factor Receptor. Mol. Endocrinol. 2003, 17, 309–317. [Google Scholar] [CrossRef] [PubMed]
  106. Wagner, N.A.-O.; Wagner, K.A.-O. PPAR Beta/Delta and the Hallmarks of Cancer. Cells 2020, 9, 1133. [Google Scholar] [CrossRef]
  107. Tan, Y.; Wang, M.; Yang, K.; Chi, T.; Liao, Z.; Wei, P. PPAR-α Modulators as Current and Potential Cancer Treatments. Front. Oncol. 2021, 11, 599995. [Google Scholar] [CrossRef]
  108. Hernandez-Quiles, M.; Broekema, M.F.; Kalkhoven, E. PPARgamma in Metabolism, Immunity, and Cancer: Unified and Diverse Mechanisms of Action. Front. Endocrinol. 2021, 12, 624112. [Google Scholar] [CrossRef]
  109. Chi, T.; Wang, M.; Wang, X.; Yang, K.; Xie, F.; Liao, Z.; Wei, P. PPAR-γ Modulators as Current and Potential Cancer Treatments. Front. Oncol. 2021, 11, 737776. [Google Scholar] [CrossRef]
  110. Cheng, H.A.-O.; Yip, Y.S.; Lim, E.K.Y.; Wahli, W.A.-O.; Tan, N.A.-O. PPARs and Tumor Microenvironment: The Emerging Roles of the Metabolic Master Regulators in Tumor Stromal-Epithelial Crosstalk and Carcinogenesis. Cancers 2021, 13, 2153. [Google Scholar] [CrossRef]
  111. Lu, H. Crosstalk of HNF4α with extracellular and intracellular signaling pathways in the regulation of hepatic metabolism of drugs and lipids. Acta Pharm. Sin. B 2016, 6, 393–408. [Google Scholar] [CrossRef] [PubMed]
  112. Wang, Z.; Zhang, Y.; Zhang, J.; Deng, Q.; Liang, H. Controversial roles of hepatocyte nuclear receptor 4α on tumorigenesis (Review). Oncol. Lett. 2021, 21, 356. [Google Scholar] [CrossRef] [PubMed]
  113. Wang, Z.; Li, Y.; Wu, D.; Yu, S.; Wang, Y.; Leung Chan, F. Nuclear receptor HNF4α performs a tumor suppressor function in prostate cancer via its induction of p21-driven cellular senescence. Oncogene 2020, 39, 1572–1589. [Google Scholar] [CrossRef] [PubMed]
  114. Lin, C.-Y.; Gustafsson, J.-Å. Targeting liver X receptors in cancer therapeutics. Nat. Rev. Cancer 2015, 15, 216–224. [Google Scholar] [CrossRef]
  115. Komati, R.; Spadoni, D.; Zheng, S.; Sridhar, J.; Riley, K.E.; Wang, G. Ligands of Therapeutic Utility for the Liver X Receptors. Molecules 2017, 22, 88. [Google Scholar] [CrossRef]
  116. Nguyen, T.T.T.; Ishida, C.T.; Shang, E.; Shu, C.; Torrini, C.; Zhang, Y.; Bianchetti, E.; Sanchez-Quintero, M.J.; Kleiner, G.; Quinzii, C.M.; et al. Activation of LXRβ inhibits tumor respiration and is synthetically lethal with Bcl-xL inhibition. EMBO Mol. Med. 2019, 11, e10769. [Google Scholar] [CrossRef]
  117. Ma, L.; Vidana Gamage, H.E.; Tiwari, S.; Han, C.; Henn, M.A.; Krawczynska, N.; Dibaeinia, P.; Koelwyn, G.J.; Das Gupta, A.; Bautista Rivas, R.O.; et al. The Liver X Receptor Is Selectively Modulated to Differentially Alter Female Mammary Metastasis-associated Myeloid Cells. Endocrinology 2022, 163, bqac072. [Google Scholar] [CrossRef]
  118. Zhou, M.; Wang, D.; Li, X.; Cao, Y.; Yi, C.; Wiredu Ocansey, D.K.; Zhou, Y.; Mao, F. Farnesoid-X receptor as a therapeutic target for inflammatory bowel disease and colorectal cancer. Front. Pharmacol. 2022, 13, 1016836. [Google Scholar] [CrossRef]
  119. Huang, X.; Wang, B.; Chen, R.; Zhong, S.; Gao, F.; Zhang, Y.; Niu, Y.; Li, C.; Shi, G. The Nuclear Farnesoid X Receptor Reduces p53 Ubiquitination and Inhibits Cervical Cancer Cell Proliferation. Front. Cell Dev. Biol. 2021, 9, 583146. [Google Scholar] [CrossRef]
  120. Girisa, S.; Rana, V.; Parama, D.; Dutta, U.; Kunnumakkara, A.B.; Donev, R. Chapter Three—Differential roles of farnesoid X receptor (FXR) in modulating apoptosis in cancer cells. In Advances in Protein Chemistry and Structural Biology; Academic Press: Cambridge, MA, USA, 2021; Volume 126, pp. 63–90. [Google Scholar]
  121. Lai, C.-R.; Wang, H.-H.; Chang, H.-H.; Tsai, Y.-L.; Tsai, W.-C.; Lee, C.-R.; Changchien, C.-Y.; Cheng, Y.-C.; Wu, S.-T.; Chen, Y. Enhancement of Farnesoid X Receptor Inhibits Migration, Adhesion and Angiogenesis through Proteasome Degradation and VEGF Reduction in Bladder Cancers. Int. J. Mol. Sci. 2022, 23, 5259. [Google Scholar] [CrossRef]
  122. Peng, Z.; Chen, J.; Drachenberg, C.B.; Raufman, J.P.; Xie, G. Farnesoid X receptor represses matrix metalloproteinase 7 expression, revealing this regulatory axis as a promising therapeutic target in colon cancer. J. Biol. Chem. 2019, 294, 8529–8542. [Google Scholar] [CrossRef]
  123. Yu, J.; Li, S.; Guo, J.; Xu, Z.; Zheng, J.; Sun, X. Farnesoid X receptor antagonizes Wnt/β-catenin signaling in colorectal tumorigenesis. Cell Death Dis. 2020, 11, 640. [Google Scholar] [CrossRef] [PubMed]
  124. Pondugula, S.R.; Pavek, P.; Mani, S. Pregnane X Receptor and Cancer: Context-Specificity is Key. Nucl. Recept. Res. 2016, 3, 101198. [Google Scholar] [CrossRef] [PubMed]
  125. Creamer, B.A.; Sloan, S.N.B.; Dennis, J.A.-O.; Rogers, R.; Spencer, S.; McCuen, A.; Persaud, P.; Staudinger, J.L. Associations between Pregnane X Receptor and Breast Cancer Growth and Progression. Cells 2020, 9, 2295. [Google Scholar] [CrossRef] [PubMed]
  126. Skandalaki, A.; Sarantis, P.; Theocharis, S. Pregnane X Receptor (PXR) Polymorphisms and Cancer Treatment. Biomolecules 2021, 11, 1142. [Google Scholar] [CrossRef]
  127. Wu, J.; Nagy, L.E.; Liangpunsakul, S.; Wang, L. Non-coding RNA crosstalk with nuclear receptors in liver disease. Biochim. Biophys. Acta Mol. Basis Dis. 2021, 1867, 166083. [Google Scholar] [CrossRef]
  128. Hung, C.L.; Wang, L.Y.; Yu, Y.L.; Chen, H.W.; Srivastava, S.; Petrovics, G.; Kung, H.J. A long noncoding RNA connects c-Myc to tumor metabolism. Proc. Natl. Acad. Sci. USA 2014, 111, 18697–18702. [Google Scholar] [CrossRef]
  129. Parolia, A.; Crea, F.; Xue, H.; Wang, Y.; Mo, F.; Ramnarine, V.R.; Liu, H.H.; Lin, D.; Saidy, N.R.N.; Clermont, P.-L.; et al. The long non-coding RNA PCGEM1 is regulated by androgen receptor activity in vivo. Mol. Cancer 2015, 14, 46. [Google Scholar] [CrossRef]
  130. Takayama, K.-i.; Horie-Inoue, K.; Katayama, S.; Suzuki, T.; Tsutsumi, S.; Ikeda, K.; Urano, T.; Fujimura, T.; Takagi, K.; Takahashi, S.; et al. Androgen-responsive long noncoding RNA CTBP1-AS promotes prostate cancer. EMBO J. 2013, 32, 1665–1680. [Google Scholar] [CrossRef]
  131. Zhang, A.; Zhao, J.C.; Kim, J.; Fong, K.-w.; Yang, Y.A.; Chakravarti, D.; Mo, Y.-Y.; Yu, J. LncRNA HOTAIR Enhances the Androgen-Receptor-Mediated Transcriptional Program and Drives Castration-Resistant Prostate Cancer. Cell Rep. 2015, 13, 209–221. [Google Scholar] [CrossRef]
  132. Li, Z.; Teng, J.; Jia, Z.; Zhang, G.; Ai, X. The long non-coding RNA PCAL7 promotes prostate cancer by strengthening androgen receptor signaling. J. Clin. Lab. Anal. 2021, 35, e23645. [Google Scholar] [CrossRef]
  133. Takayama, K.-i.; Fujimura, T.; Suzuki, Y.; Inoue, S. Identification of long non-coding RNAs in advanced prostate cancer associated with androgen receptor splicing factors. Commun. Biol. 2020, 3, 393. [Google Scholar] [CrossRef] [PubMed]
  134. Misawa, A.; Takayama, K.-i.; Urano, T.; Inoue, S. Androgen-induced Long Noncoding RNA (lncRNA) SOCS2-AS1 Promotes Cell Growth and Inhibits Apoptosis in Prostate Cancer Cells. J. Biol. Chem. 2016, 291, 17861–17880. [Google Scholar] [CrossRef] [PubMed]
  135. Ghildiyal, R.; Sawant, M.; Renganathan, A.; Mahajan, K.; Kim, E.H.; Luo, J.; Dang, H.X.; Maher, C.A.; Feng, F.Y.; Mahajan, N.P. Loss of Long Noncoding RNA NXTAR in Prostate Cancer Augments Androgen Receptor Expression and Enzalutamide Resistance. Cancer Res. 2022, 82, 155–168. [Google Scholar] [CrossRef] [PubMed]
  136. Yang, F.; Shen, Y.; Zhang, W.; Jin, J.; Huang, D.; Fang, H.; Ji, W.; Shi, Y.; Tang, L.; Chen, W.; et al. An androgen receptor negatively induced long non-coding RNA ARNILA binding to miR-204 promotes the invasion and metastasis of triple-negative breast cancer. Cell Death Differ. 2018, 25, 2209–2220. [Google Scholar] [CrossRef] [PubMed]
  137. Liang, X.; Zhao, Y.; Fang, Z.; Shao, N.; Zhai, D.; Zhang, M.; Yu, L.; Shi, Y. DLGAP1-AS2 promotes estrogen receptor signalling and confers tamoxifen resistance in breast cancer. Mol. Biol. Rep. 2022, 49, 3939–3947. [Google Scholar] [CrossRef]
  138. Miano, V.; Ferrero, G.; Reineri, S.; Caizzi, L.; Annaratone, L.; Ricci, L.; Cutrupi, S.; Castellano, I.; Cordero, F.; De Bortoli, M. Luminal long non-coding RNAs regulated by estrogen receptor alpha in a ligand-independent manner show functional roles in breast cancer. Oncotarget 2016, 7, 3201. [Google Scholar] [CrossRef]
  139. Fang, Z.; Wang, Y.; Wang, Z.; Xu, M.; Ren, S.; Yang, D.; Hong, M.; Xie, W. ERINA Is an Estrogen-Responsive LncRNA That Drives Breast Cancer through the E2F1/RB1 Pathway. Cancer Res. 2020, 80, 4399–4413. [Google Scholar] [CrossRef]
  140. Yuan, H.; Yan, L.; Wu, M.; Shang, Y.; Guo, Q.; Ma, X.; Zhang, X.; Zhu, Y.; Wu, Z.; Lobie, P.E.; et al. Analysis of the Estrogen Receptor-Associated LncRNA Landscape Identifies a Role for ERLC1 in Breast Cancer Progression. Cancer Res. 2022, 82, 391–405. [Google Scholar] [CrossRef]
  141. Cui, M.; Xiao, Z.; Wang, Y.; Zheng, M.; Song, T.; Cai, X.; Sun, B.; Ye, L.; Zhang, X. Long Noncoding RNA HULC Modulates Abnormal Lipid Metabolism in Hepatoma Cells through an miR-9–Mediated RXRA Signaling Pathway. Cancer Res. 2015, 75, 846–857. [Google Scholar] [CrossRef]
  142. Li, X.; Zhao, Q.; Qi, J.; Wang, W.; Zhang, D.; Li, Z.; Qin, C. lncRNA Ftx promotes aerobic glycolysis and tumor progression through the PPARγ pathway in hepatocellular carcinoma. Int. J. Oncol. 2018, 53, 551–566. [Google Scholar] [CrossRef] [PubMed]
  143. Chen, X.; Zhao, Y.; Wang, D.; Lin, Y.; Hou, J.; Xu, X.; Wu, J.; Zhong, L.; Zhou, Y.; Shen, J.; et al. The HNF4α-BC200-FMR1–Positive Feedback Loop Promotes Growth and Metastasis in Invasive Mucinous Lung Adenocarcinoma. Cancer Res. 2021, 81, 5904–5918. [Google Scholar] [CrossRef] [PubMed]
  144. Zhao, H.; Zhang, X.; Frazão, J.B.; Condino-Neto, A.; Newburger, P.E. HOX antisense lincRNA HOXA-AS2 is an apoptosis repressor in all Trans retinoic acid treated NB4 promyelocytic leukemia cells. J. Cell. Biochem. 2013, 114, 2375–2383. [Google Scholar] [CrossRef]
  145. El Hajj, J.; Nguyen, E.; Liu, Q.; Bouyer, C.; Adriaenssens, E.; Hilal, G.; Ségal-Bendirdjian, E. Telomerase regulation by the long non-coding RNA H19 in human acute promyelocytic leukemia cells. Mol. Cancer 2018, 17, 85. [Google Scholar] [CrossRef] [PubMed]
  146. Zheng, C.; Li, X.; Ren, Y.; Yin, Z.; Zhou, B. Long Noncoding RNA RAET1K Enhances CCNE1 Expression and Cell Cycle Arrest of Lung Adenocarcinoma Cell by Sponging miRNA-135a-5p. Front. Genet. 2020, 10, 1348. [Google Scholar] [CrossRef] [PubMed]
  147. Schmidt, K.; Weidmann, C.A.; Hilimire, T.A.; Yee, E.; Hatfield, B.M.; Schneekloth, J.S., Jr.; Weeks, K.M.; Novina, C.D. Targeting the Oncogenic Long Non-coding RNA SLNCR1 by Blocking Its Sequence-Specific Binding to the Androgen Receptor. Cell Rep. 2020, 30, 541–554.e5. [Google Scholar] [CrossRef]
  148. Cantile, M.; Cerrone, M.; Di Bonito, M.; Moccia, P.; Tracey, M.; Ferrara, G.; Budillon, A. Endocrine nuclear receptors and long non-coding RNAs reciprocal regulation in cancer (Review). Int. J. Oncol. 2024, 64, 7. [Google Scholar] [CrossRef]
  149. Kumar, S.; Prajapati, K.S.; Singh, A.K.; Kushwaha, P.P.; Shuaib, M.; Gupta, S. Long non-coding RNA regulating androgen receptor signaling in breast and prostate cancer. Cancer Lett. 2021, 504, 15–22. [Google Scholar] [CrossRef]
  150. Morgan, R.; da Silveira, W.A.; Kelly, R.C.; Overton, I.; Allott, E.H.; Hardiman, G. Long non-coding RNAs and their potential impact on diagnosis, prognosis, and therapy in prostate cancer: Racial, ethnic, and geographical considerations. Expert Rev. Mol. Diagn. 2021, 21, 1257–1271. [Google Scholar] [CrossRef]
  151. Huang, G.; Cao, H.; Liu, G.; Chen, J. Role of androgen receptor signaling pathway-related lncRNAs in the prognosis and immune infiltration of breast cancer. Sci. Rep. 2022, 12, 20631. [Google Scholar] [CrossRef]
  152. Huang, L.A.-O.; Liang, G.A.-O.; Zhang, Q.A.-O.; Zhao, W.A.-O. The Role of Long Noncoding RNAs in Antiestrogen Resistance in Breast Cancer: An Overview and Update. J. Breast Cancer 2020, 23, 129. [Google Scholar] [CrossRef]
  153. Sedano, M.J.; Harrison, A.L.; Zilaie, M.; Das, C.; Choudhari, R.A.-O.; Ramos, E.; Gadad, S.A.-O. Emerging Roles of Estrogen-Regulated Enhancer and Long Non-Coding RNAs. Int. J. Mol. Sci. 2020, 21, 3711. [Google Scholar] [CrossRef]
  154. Mitobe, Y.; Ikeda, K.; Suzuki, T.; Takagi, K.; Kawabata, H.; Horie-Inoue, K.; Inoue, S. ESR1-Stabilizing Long Noncoding RNA TMPO-AS1 Promotes Hormone-Refractory Breast Cancer Progression. Mol. Cell. Biol. 2019, 39, e00261-00219. [Google Scholar] [CrossRef] [PubMed]
  155. Liu, S.; Lai, W.; Shi, Y.; Liu, N.; Ouyang, L.; Zhang, Z.; Chen, L.; Wang, X.; Qian, B.; Xiao, D.; et al. Annotation and cluster analysis of long noncoding RNA linked to male sex and estrogen in cancers. NPJ Precis. Oncol. 2020, 4, 5. [Google Scholar] [CrossRef]
  156. Kino, T.; Hurt, D.E.; Ichijo, T.; Nader, N.; Chrousos, G.P. Noncoding RNA gas5 is a growth arrest-and starvation-associated repressor of the glucocorticoid receptor. Sci. Signal. 2010, 3, ra8. [Google Scholar] [CrossRef] [PubMed]
  157. Brocker, C.N.; Kim, D.; Melia, T.; Karri, K.; Velenosi, T.J.; Takahashi, S.; Aibara, D.; Bonzo, J.A.; Levi, M.; Waxman, D.J.; et al. Long non-coding RNA Gm15441 attenuates hepatic inflammasome activation in response to PPARA agonism and fasting. Nat. Commun. 2020, 11, 5847. [Google Scholar] [CrossRef] [PubMed]
  158. Sharma, B.R.; Kanneganti, T.-D. NLRP3 inflammasome in cancer and metabolic diseases. Nat. Immunol. 2021, 22, 550–559. [Google Scholar] [CrossRef] [PubMed]
  159. Wang, F.-W.; Cao, C.-H.; Han, K.; Zhao, Y.-X.; Cai, M.-Y.; Xiang, Z.-C.; Zhang, J.-X.; Chen, J.-W.; Zhong, L.-P.; Huang, Y.; et al. APC-activated long noncoding RNA inhibits colorectal carcinoma pathogenesis through reduction of exosome production. J. Clin. Investig. 2021, 131, e149666. [Google Scholar] [CrossRef]
  160. Zhao, Y.; Qin, Z.S.; Feng, Y.; Tang, X.J.; Zhang, T.; Yang, L. Long non-coding RNA (lncRNA) small nucleolar RNA host gene 1 (SNHG1) promote cell proliferation in colorectal cancer by affecting P53. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 976–984. [Google Scholar]
  161. Chen, S.; Guo, W.; Meng, M.; Wu, D.; Zhou, T.; Wang, L.; Xu, J. LncRNA SNHG1 Promotes the Progression of Pancreatic Cancer by Regulating FGFR1 Expression via Competitively Binding to miR-497. Front. Oncol. 2022, 12, 813850. [Google Scholar] [CrossRef]
  162. Hsu, C.-L.; Yin, C.-F.; Chang, Y.-W.; Fan, Y.-C.; Lin, S.-H.; Wu, Y.-C.; Huang, H.-C.; Juan, H.-F. LncRNA SNHG1 regulates neuroblastoma cell fate via interactions with HDAC1/2. Cell Death Dis. 2022, 13, 809. [Google Scholar] [CrossRef] [PubMed]
  163. Cai, H.; Xu, H.; Lu, H.; Xu, W.; Liu, H.; Wang, X.; Zhou, G.; Yang, X. LncRNA SNHG1 Facilitates Tumor Proliferation and Represses Apoptosis by Regulating PPARγ Ubiquitination in Bladder Cancer. Cancers 2022, 14, 4740. [Google Scholar] [CrossRef] [PubMed]
  164. Han, J.; Shen, L.; Zhan, Z.; Liu, Y.; Zhang, C.; Guo, R.; Luo, Y.; Xie, Z.; Feng, Y.; Wu, G. The long noncoding RNA MALAT1 modulates adipose loss in cancer-associated cachexia by suppressing adipogenesis through PPAR-γ. Nutr. Metab. 2021, 18, 27. [Google Scholar] [CrossRef] [PubMed]
  165. Xu, T.; Wu, K.; Zhang, L.; Zheng, S.; Wang, X.; Zuo, H.; Wu, X.; Tao, G.; Jiang, B.; Zhang, L. Long non-coding RNA LINC00858 exerts a tumor-promoting role in colon cancer via HNF4α and WNK2 regulation. Cell. Oncol. 2020, 43, 297–310. [Google Scholar] [CrossRef]
  166. Wang, X.; Song, B.; Zang, M.; Ji, H.; Yang, H.; Jiang, S.; Yang, X. LOC100996425 acts as a promoter in prostate cancer by mediating hepatocyte nuclear factor 4A and the AMPK/mTOR pathway. J. Cell. Mol. Med. 2021, 25, 8174–8186. [Google Scholar] [CrossRef]
  167. Lu, Y.; Yu, Y.; Liu, F.; Han, Y.; Xue, H.; Sun, X.; Jiang, Y.; Tian, Z. LINC00511-dependent inhibition of IL-24 contributes to the oncogenic role of HNF4α in colorectal cancer. Am. J. Physiol. Gastrointest. Liver Physiol. 2021, 320, G338–G350. [Google Scholar] [CrossRef]
  168. Deng, D.; Yang, S.; Wang, X. Long non-coding RNA SNHG16 regulates cell behaviors through miR-542-3p/HNF4α axis via RAS/RAF/MEK/ERK signaling pathway in pediatric neuroblastoma cells. Biosci. Rep. 2020, 40, BSR20200723. [Google Scholar] [CrossRef]
  169. Jin, L.; Pan, Y.-L.; Zhang, J.; Cao, P.-G. LncRNA HOTAIR recruits SNAIL to inhibit the transcription of HNF4α and promote the viability, migration, invasion and EMT of colorectal cancer. Transl. Oncol. 2021, 14, 101036. [Google Scholar] [CrossRef]
  170. Dawson, M.I.; Xia, Z. The retinoid X receptors and their ligands. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2012, 1821, 21–56. [Google Scholar] [CrossRef]
  171. Wan, W.; Hou, Y.; Wang, K.; Cheng, Y.; Pu, X.; Ye, X. The LXR-623-induced long non-coding RNA LINC01125 suppresses the proliferation of breast cancer cells via PTEN/AKT/p53 signaling pathway. Cell Death Dis. 2019, 10, 248. [Google Scholar] [CrossRef]
  172. He, M.; Yang, H.; Shi, H.; Hu, Y.; Chang, C.; Liu, S.; Yeh, S. Sunitinib increases the cancer stem cells and vasculogenic mimicry formation via modulating the lncRNA-ECVSR/ERβ/Hif2-α signaling. Cancer Lett. 2022, 524, 15–28. [Google Scholar] [CrossRef] [PubMed]
  173. Ferrer, J.; Dimitrova, N. Transcription regulation by long non-coding RNAs: Mechanisms and disease relevance. Nat. Rev. Mol. Cell Biol. 2024, 25, 396–415. [Google Scholar] [CrossRef]
  174. Lowe, R.A.-O.; Shirley, N.; Bleackley, M.; Dolan, S.A.-O.; Shafee, T.A.-O. Transcriptomics technologies. PLoS Comput. Biol. 2017, 13, e1005457. [Google Scholar] [CrossRef] [PubMed]
  175. Lorenzi, L.; Chiu, H.-S.; Avila Cobos, F.; Gross, S.; Volders, P.-J.; Cannoodt, R.; Nuytens, J.; Vanderheyden, K.; Anckaert, J.; Lefever, S.; et al. The RNA Atlas expands the catalog of human non-coding RNAs. Nat. Biotechnol. 2021, 39, 1453–1465. [Google Scholar] [CrossRef] [PubMed]
  176. Hafner, M.; Katsantoni, M.; Köster, T.; Marks, J.; Mukherjee, J.; Staiger, D.; Ule, J.; Zavolan, M. CLIP and complementary methods. Nat. Rev. Methods Primers 2021, 1, 20. [Google Scholar] [CrossRef]
  177. Cozzolino, F.; Iacobucci, I.; Monaco, V.; Monti, M. Protein–DNA/RNA Interactions: An Overview of Investigation Methods in the -Omics Era. J. Proteome Res. 2021, 20, 3018–3030. [Google Scholar] [CrossRef]
  178. Chu, C.; Qu, K.; Zhong, F.L.; Artandi, S.E.; Chang, H.Y. Genomic maps of long noncoding RNA occupancy reveal principles of RNA-chromatin interactions. Mol. Cell 2011, 44, 667–678. [Google Scholar] [CrossRef]
  179. Simon, M.D.; Wang, C.I.; Kharchenko, P.V.; West, J.A.; Chapman, B.A.; Alekseyenko, A.A.; Borowsky, M.L.; Kuroda, M.I.; Kingston, R.E. The genomic binding sites of a noncoding RNA. Proc. Natl. Acad. Sci. USA 2011, 108, 20497–20502. [Google Scholar] [CrossRef]
  180. Bonetti, A.; Agostini, F.; Suzuki, A.M.; Hashimoto, K.; Pascarella, G.; Gimenez, J.; Roos, L.; Nash, A.J.; Ghilotti, M.; Cameron, C.J.F.; et al. RADICL-seq identifies general and cell type–specific principles of genome-wide RNA-chromatin interactions. Nat. Commun. 2020, 11, 1018. [Google Scholar] [CrossRef]
  181. Li, X.; Zhou, B.; Chen, L.; Gou, L.-T.; Li, H.; Fu, X.-D. GRID-seq reveals the global RNA–chromatin interactome. Nat. Biotechnol. 2017, 35, 940–950. [Google Scholar] [CrossRef]
  182. Fields, S.; Song, O.-K. A novel genetic system to detect protein–protein interactions. Nature 1989, 340, 245–246. [Google Scholar] [CrossRef]
  183. Dunham, W.H.; Mullin, M.; Gingras, A.C. Affinity-purification coupled to mass spectrometry: Basic principles and strategies. Proteomics 2012, 12, 1576–1590. [Google Scholar] [CrossRef]
  184. Yang, J.; Wagner, S.A.; Beli, P. Illuminating Spatial and Temporal Organization of Protein Interaction Networks by Mass Spectrometry-Based Proteomics. Front. Genet. 2015, 6, 344. [Google Scholar] [CrossRef]
  185. Johnson, K.L.; Qi, Z.; Yan, Z.; Wen, X.; Nguyen, T.C.; Zaleta-Rivera, K.; Chen, C.J.; Fan, X.; Sriram, K.; Wan, X.; et al. Revealing protein-protein interactions at the transcriptome scale by sequencing. Mol. Cell 2021, 81, 4091–4103.e4099. [Google Scholar] [CrossRef] [PubMed]
  186. Lieberman-Aiden, E.; van Berkum, N.L.; Williams, L.; Imakaev, M.; Ragoczy, T.; Telling, A.; Amit, I.; Lajoie, B.R.; Sabo, P.J.; Dorschner, M.O.; et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 2009, 326, 289–293. [Google Scholar] [CrossRef] [PubMed]
  187. Cai, Z.; Cao, C.; Ji, L.; Ye, R.; Wang, D.; Xia, C.; Wang, S.; Du, Z.; Hu, N.; Yu, X.; et al. RIC-seq for global in situ profiling of RNA–RNA spatial interactions. Nature 2020, 582, 432–437. [Google Scholar] [CrossRef]
  188. Armaos, A.; Colantoni, A.; Proietti, G.; Rupert, J.; Tartaglia, G.G. catRAPID omics v2.0: Going deeper and wider in the prediction of protein–RNA interactions. Nucleic Acids Res. 2021, 49, W72–W79. [Google Scholar] [CrossRef] [PubMed]
  189. Murakami, Y.; Tripathi, L.P.; Prathipati, P.; Mizuguchi, K. Network analysis and in silico prediction of protein–protein interactions with applications in drug discovery. Curr. Opin. Struct. Biol. 2017, 44, 134–142. [Google Scholar] [CrossRef] [PubMed]
  190. Tripathi, L.P.; Chen, Y.-a.; Mizuguchi, K.; Murakami, Y. Network-Based Analysis for Biological Discovery. Encycl. Bioinform. Comput. Biol. 2019, 283–291. [Google Scholar]
  191. Ronan, T.; Qi, Z.; Naegle, K.M. Avoiding common pitfalls when clustering biological data. Sci. Signal. 2016, 9, re6. [Google Scholar] [CrossRef]
  192. Li, W.; Zhang, S.; Liu, C.C.; Zhou, X.J. Identifying multi-layer gene regulatory modules from multi-dimensional genomic data. Bioinformatics 2012, 28, 2458–2466. [Google Scholar] [CrossRef] [PubMed]
  193. Robinson, J.L.; Nielsen, J. Integrative analysis of human omics data using biomolecular networks. Mol. BioSystems 2016, 12, 2953–2964. [Google Scholar] [CrossRef] [PubMed]
  194. Lin, E.; Lane, H.Y. Machine learning and systems genomics approaches for multi-omics data. Biomark. Res. 2017, 5, 2. [Google Scholar] [CrossRef] [PubMed]
  195. Tripathi, L.P.; Esaki, T.; Itoh, M.N.; Chen, Y.-A.; Mizuguchi, K. Integrative Analysis of Multi-Omics Data. In Encyclopedia of Bioinformatics and Computational Biology; Ranganathan, S., Gribskov, M., Nakai, K., Schönbach, C., Eds.; Academic Press: Oxford, UK, 2019; pp. 194–199. [Google Scholar]
Figure 1. A representative interplay between growth factors, hormones and lncRNAs in cancer proliferation. The binding of the IGF-1 to IGF-1 receptor (IGF-1R) triggers the PI3K/AKT-mTOR and Jak pathways that contribute to increased expression of Erα-induced genes, thereby leading to accelerated breast cancer growth and proliferation. EGFR signaling can also trigger PI3k-AKT signaling, thereby contributing to the activation of Erα-responsive genes. LncRNA NR2F1-AS1 can also modulate IGF-1R activation by sponging miR-338-3p, a negative regulator of IGF-1. In prostate cancer, IGF-1R signaling activates AR signaling via the IGF-1R-FOXO1 (forkhead box protein O1) signaling axis. AR activation stimulates the expression of HIF1α-induced genes, which contribute to cancer proliferation. HIF1α-responsive genes may also be triggered by EGFR via the PI3K-AKT signaling axis. HOTAIR is an androgen-repressed lncRNA that is upregulated in castration-resistant prostate cancer (CRPC); HOTAIR associates with AR and drives its androgen-independent activation that subsequently leads to induction of AR-target genes in CRPC proliferation.
Figure 1. A representative interplay between growth factors, hormones and lncRNAs in cancer proliferation. The binding of the IGF-1 to IGF-1 receptor (IGF-1R) triggers the PI3K/AKT-mTOR and Jak pathways that contribute to increased expression of Erα-induced genes, thereby leading to accelerated breast cancer growth and proliferation. EGFR signaling can also trigger PI3k-AKT signaling, thereby contributing to the activation of Erα-responsive genes. LncRNA NR2F1-AS1 can also modulate IGF-1R activation by sponging miR-338-3p, a negative regulator of IGF-1. In prostate cancer, IGF-1R signaling activates AR signaling via the IGF-1R-FOXO1 (forkhead box protein O1) signaling axis. AR activation stimulates the expression of HIF1α-induced genes, which contribute to cancer proliferation. HIF1α-responsive genes may also be triggered by EGFR via the PI3K-AKT signaling axis. HOTAIR is an androgen-repressed lncRNA that is upregulated in castration-resistant prostate cancer (CRPC); HOTAIR associates with AR and drives its androgen-independent activation that subsequently leads to induction of AR-target genes in CRPC proliferation.
Cancers 16 02920 g001
Figure 2. Mechanisms underlying cross-talks between lncRNAs and NRs. lncRNAs and NRs can mutually regulate each other through direct physical associations: (A) Transcriptional coactivation—PCGEM1 functions as a coactivator for AR and cMyc and promotes proliferation. (B) Decoy binding—GAS5 binds to the GR DBD as a GRE decoy and limits its binding to GREs in the target genes. LncRNAs and NRs may also mutually regulate or interact indirectly with each other in different ways. (C) Negative feed-forward circuitry—NXSTAR and AR mutually negatively impact transcription. (D) Feedback regulation—positive feedback loop BC200 stabilizes HNF4α mRNA. (E) Activation of signaling—PCAL7 stabilizes HIP1 and activates AR signaling. (F) Regulation of splicing—CRPC-linked lncRNAs associate with splicing factors such U2 to positively regulate AR expression. (G) Transcriptional activation and miRNA sequestration—Erα transcriptionally activates ERLC1; ERLC1 stabilizes ESR1 transcript by sequestrating miR-129. Red × means that because of the preceding event, the downstream process is stopped from proceeding further.
Figure 2. Mechanisms underlying cross-talks between lncRNAs and NRs. lncRNAs and NRs can mutually regulate each other through direct physical associations: (A) Transcriptional coactivation—PCGEM1 functions as a coactivator for AR and cMyc and promotes proliferation. (B) Decoy binding—GAS5 binds to the GR DBD as a GRE decoy and limits its binding to GREs in the target genes. LncRNAs and NRs may also mutually regulate or interact indirectly with each other in different ways. (C) Negative feed-forward circuitry—NXSTAR and AR mutually negatively impact transcription. (D) Feedback regulation—positive feedback loop BC200 stabilizes HNF4α mRNA. (E) Activation of signaling—PCAL7 stabilizes HIP1 and activates AR signaling. (F) Regulation of splicing—CRPC-linked lncRNAs associate with splicing factors such U2 to positively regulate AR expression. (G) Transcriptional activation and miRNA sequestration—Erα transcriptionally activates ERLC1; ERLC1 stabilizes ESR1 transcript by sequestrating miR-129. Red × means that because of the preceding event, the downstream process is stopped from proceeding further.
Cancers 16 02920 g002
Figure 3. High-throughput omics technologies can provide heterogeneous lncRNA and biomolecular interaction data; these data can be assembled into an lncRNA-centric interactome that in conjunction with gene expression data may then be used to generate lncRNA-NR gene regulatory networks (GRNs), to hypothesize the mechanisms underlying lncRNA-NR interactions and their phenotypic outcomes. These outcomes can then be leveraged to rank and prioritize biologically and therapeutically relevant candidates for biomedical and drug-target discovery.
Figure 3. High-throughput omics technologies can provide heterogeneous lncRNA and biomolecular interaction data; these data can be assembled into an lncRNA-centric interactome that in conjunction with gene expression data may then be used to generate lncRNA-NR gene regulatory networks (GRNs), to hypothesize the mechanisms underlying lncRNA-NR interactions and their phenotypic outcomes. These outcomes can then be leveraged to rank and prioritize biologically and therapeutically relevant candidates for biomedical and drug-target discovery.
Cancers 16 02920 g003
Table 1. Selected NR and lncRNA cross-talks and their functional roles in cancers.
Table 1. Selected NR and lncRNA cross-talks and their functional roles in cancers.
NRslncRNAsCancer TypeMechanismsImpact on Cancer
Pathogenesis
Refs.
ARPCGEM1ProstateCoactivator for AR and c-Myc, tumor metabolism and carcinogenesis Promotes cancer progression.[128,129]
CTBP1-ASProstatePromotes AR transcription and cell cycle.Promotes cancer progression.[130]
HOTAIRCRPCBinds to N-terminal domain of AR, preventing its ubiquitination and degradation. Enhanced cell migration and proliferation.[131]
PCAL-7CRPCInteracts with and stabilizes HIP1 protein to activate AR signaling.Promotes cell growth and invasion and cancer proliferation.[132]
CRPC-Lnc#6CRPCModulates AR splicing.Abnormalities in target gene expression.[133]
SOCS2-AS1CRPCApoptosis-inhibition.Enhanced cell growth and migration.[134]
NXTARCRPCInhibits AR/AR-V7; AR negatively regulates NXTAR in a negative feed-forward NXTAR-AR circuitry.Loss of NXTAR leads to enhanced tumorigenesis.[135]
ARNILATriple Negative Breast Cancer (TNBC)AR suppresses ARNILA transcription by promoter bindingSequesters miR-204 leading to upregulation of Sox4, promotes EMT, invasion and metastasis.[136]
ERαDLGAP1-AS2BreastBinds to and inhibits AFF3 degradation, promoting ER signaling.Confers tamoxifen resistance.[137]
DSCAM-AS1Erα positively regulates DSCAM-AS1 expression.Correlates inversely with EMT markers; cancer progression.[138]
ERINAER transactivates ERINA via intronic binding site; ERINA binds to E2F1 and prevents interaction with RB1.Promotes cell-cycle progression and tumor proliferation; correlated with poor patient survival.[139]
ERLC1Erα transcriptionally activates ERLC1; ERLC1 stabilizes ESR1 transcript by sequestrating miR-129.Promotes resistance to antiestrogen therapies in breast cancer.[140]
PPARαHULCHepatocellular carcinomaDeregulates lipid metabolism via miR-9, PPARA, and ACSL1 pathway.Malignant development.[141]
PPARγFtxAerobic glycolysisTumor progression.[142]
HNF4αBC200Invasive mucinous lung adenocarcinoma (IMA)HNF4α-BC200-FMR1 positive feedback loop stabilizes cancer-related and HNF4α mRNAs. Tumor growth and metastasis.[143]
RAR/RXRHOXA-AS2Acute promyelocytic leukemia (APL)RAR signaling upregulates HOXA-AS2, which in turn reduces caspase expression.Tumor growth and metastasis.[144]
H19APLRAR signaling upregulates H19, which perturbs telomerase activity.Tumor growth and metastasis.[145]
RAET1KLungRAET1K counteracts RA activity by sponging miR-135a-5p, thereby enhancing Cyclin E1 expression.Cell-cycle progression in tumorigenesis.[146]
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

Tiwari, P.; Tripathi, L.P. Long Non-Coding RNAs, Nuclear Receptors and Their Cross-Talks in Cancer—Implications and Perspectives. Cancers 2024, 16, 2920. https://doi.org/10.3390/cancers16162920

AMA Style

Tiwari P, Tripathi LP. Long Non-Coding RNAs, Nuclear Receptors and Their Cross-Talks in Cancer—Implications and Perspectives. Cancers. 2024; 16(16):2920. https://doi.org/10.3390/cancers16162920

Chicago/Turabian Style

Tiwari, Prabha, and Lokesh P. Tripathi. 2024. "Long Non-Coding RNAs, Nuclear Receptors and Their Cross-Talks in Cancer—Implications and Perspectives" Cancers 16, no. 16: 2920. https://doi.org/10.3390/cancers16162920

APA Style

Tiwari, P., & Tripathi, L. P. (2024). Long Non-Coding RNAs, Nuclear Receptors and Their Cross-Talks in Cancer—Implications and Perspectives. Cancers, 16(16), 2920. https://doi.org/10.3390/cancers16162920

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