Next Article in Journal
Real-Time SPR Biosensing to Detect and Characterize Fast Dissociation Rate Binding Interactions Missed by Endpoint Detection and Implications for Off-Target Toxicity Screening
Previous Article in Journal
Not Just PA28γ: What We Know About the Role of PA28αβ in Carcinogenesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

LncRNA–Protein Interactions: A Key to Deciphering LncRNA Mechanisms

1
Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
2
State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou 450000, China
3
Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
4
China-US (Henan) Hormel Cancer Institute, Zhengzhou 450008, China
*
Author to whom correspondence should be addressed.
Biomolecules 2025, 15(6), 881; https://doi.org/10.3390/biom15060881
Submission received: 24 April 2025 / Revised: 8 June 2025 / Accepted: 13 June 2025 / Published: 17 June 2025
(This article belongs to the Section Molecular Biophysics: Structure, Dynamics, and Function)

Abstract

Long non-coding RNAs (lncRNAs) have emerged as pivotal regulators in a multitude of biological processes. However, their functional basis, particularly structure-based functional characteristics, remains elusive. lncRNAs exert their influence primarily through intricate interactions with various cellular components. Among these, interactions with proteins have garnered increasing attention. Recent research highlights the significance of the interactions with proteins as a plausible mechanism underlying lncRNA functions. Here, we delve into the interactions between lncRNAs and RNA-binding proteins (RBPs), explore their implications in cellular processes, and examine bioinformatic and experimental approaches for characterizing these interactions. We introduce an innovative ISD strategy to decipher the mysterious mechanism of lncRNAs. Through reviewing the recent advances in the study of proteins and their complexes, we incorporate the ISD strategy into our integrated structural analysis pipeline for comprehensively understanding the structure-function relationship of lncRNAs. Advances in the development of innovative therapeutic approaches based on lncRNA-protein interactions (LPIs) are reviewed accordingly.

1. Introduction

LncRNAs (Long non-coding RNAs) are non-coding RNAs that harbor sequences over 200 base pairs [1]. Although present at a low abundance, an increasing number of studies indicates that lncRNAs play vital roles in various cellular processes, such as differentiation and signal transduction [2]. Due to their lack of protein-coding capability, lncRNA sequences typically provide limited insights into their functional roles. Predicting lncRNA function solely from primary sequence similarity is also challenging, as their molecules exhibit low conservation across species [3,4]. A significant portion of lncRNAs adopts two-dimensional (2D) or three-dimensional (3D) structures when interacting with partner molecules to carry out cellular functions [5]. Even a combination of multiple modules, formed by conserved patches within the sequence, can be defined as functional motifs. However, determining the structures of such motifs, as well as the entire lncRNA molecule, remains a significant challenge [6]. Non-conserved primary sequence fragments are often active and tend to be flexible, which contributes to the high heterogeneity of RNA molecules in structural study pipelines. Additionally, the low abundance of lncRNAs and the presence of multiple distinct modules necessitate efficient approaches to obtain properly folded RNAs with stable structures [7,8].
The functionalization of most lncRNAs involves direct interaction with various partners. Notably, the protein partner has been drawing more and more attention due to its significant roles in LncRNA function [9]. Therefore, proteins are confirmed to be the first and principal partner of lncRNAs [10,11]. The multiple modular scaffolds within lncRNA sequences provide suitable binding interfaces or docking pockets, assembling diverse combinations of proteins [12]. For instance, lncRNAs can function as components of ribonucleoprotein complexes (RNPs) to support molecular functions. LncRNA-protein interactions (LPIs) are accepted as major functional units in metabolic processes, many of which have been shown to be closely related to human diseases [13,14,15,16,17]. Accordingly, the development of recently emerging and progressively improving methods to explore LPIs has greatly advanced our understanding of lncRNA mechanisms.
To identify LPIs, the mainstream strategy is to isolate their interactome based on the pulldown principle, followed by characterization of these interactions. For instance, workflows can be designed around a specific lncRNA to systematically explore its binding proteins [18]. An alternative strategy involves separately centering investigations on the protein partners, with the two strategies ultimately combined for complementary validation of their interactions. Recent molecular docking techniques, combined with various algorithms, allow for the investigation of LPIs through computer simulations. Commonly used web servers for such studies include, but are not limited to, HADDOCK and P3DOCK [19,20]. The accuracy of docking predictions can be greatly enhanced when structural information about the protein or lncRNA is available, and these structures can subsequently be experimentally validated. In alignment, various databases such as RBP2GO, RNA Bricks, and NPIDB cataloging LPI networks are emerging and undergoing continuous refinement [21,22,23]. Some of these now not only provide interaction details but also include information on specific binding motifs of lncRNA. To optimize the use of these resources, guide matrices and advanced search functionalities have been developed, allowing for more precise retrieval of relevant data [24]. In recent years, machine learning-based algorithms for predicting LPIs have been gradually maturing and showing great potential. Particularly deep learning methods, also known as artificial neural networks, have been shown to achieve high accuracy in predicting lncRNA-binding proteins. For instance, the DRPScore approach has been reported to evaluate native-like structures with a success rate of 91.67% on a testing set of RNA-protein complexes [25].
Overall, identifying or predicting the proteins that interact with lncRNAs cannot fully capture the molecular mechanisms underlying the structure and function of lncRNA. particularly the 3D structural mechanisms that are crucial for understanding the functional basis of lncRNAs and discovering disease prevention strategies. Recent research has provided us with new insights: Once lncRNA-binding proteins are identified, these proteins can, to some extent, be considered stabilizers of the lncRNA [26]. By applying a broader range of existing protein- or RNA-based approaches, it is becoming feasible to elucidate structural mechanisms of lncRNAs from the perspective of their protein partners or to consider lncRNA-protein complexes as independent operational units in cellular processes when investigating lncRNA’s roles in human diseases.
Therefore, in the first part of this review, we outline recent advances in uncovering the cellular mechanisms of lncRNAs, primarily through their protein-binding partners, including the biological consequences of lncRNA–protein interactions in cellular regulation and human disease. In the following core sections, we focus on experimental approaches for studying LPIs, from current methods for identification of LPIs to the structural–functional analysis of lncRNAs in their protein-bound state. We provide an in-depth discussion on the determination of secondary and tertiary structures and the study of structure–function relationships of lncRNAs within these complexes. We carefully examine emerging approaches in this area, attempt to formulate a systematic approach for overcoming the limitations mentioned above, and propose strategies to advance future research. We highlight our ISD strategy as a productive approach for uncovering the multiple roles of lncRNAs, particularly when integrated into our structural analysis framework. Naturally, the rising recognition of LPIs in diseases, along with ongoing efforts to develop therapies targeting these interactions, underscores the significance of lncRNAs in the body, which we further discuss in the final section.

2. Mechanisms of LncRNA–Protein Interactions

LncRNAs exhibit remarkable functional diversity through their ability to interact with one or more protein partners via a range of molecular mechanisms. Although lncRNAs are generally more abundant in the nucleus, they are also more prevalent in the cytoplasm than previously thought. This difference in subcellular localization has important implications for the functional roles of lncRNAs in various biological processes [27].
Early studies categorized lncRNAs into five archetypal functional classes: guides, scaffolds, decoys, enhancers, and signals, each representing a distinct mode of action across various biological processes [28,29]. These archetypes remain highly relevant when examining lncRNA–protein interactions, as each type tends to involve specific modes of protein binding and regulatory mechanisms. For instance, guide lncRNAs recruit chromatin-modifying or transcriptional regulatory proteins to specific genomic loci, thereby directing site-specific gene regulation. Scaffold lncRNAs serve as structural platforms that bring together multiple proteins into functional complexes, often facilitating enzymatic cascades or chromatin remodeling. Decoy lncRNAs bind and sequester transcription factors or RNA-binding proteins (RBPs), preventing them from acting on their usual targets and modulating downstream signaling. Enhancer-like lncRNAs interact with transcriptional co-activators or mediators, helping to stabilize or spatially organize protein assemblies that activate gene expression. lncRNAs can also function as signals by integrating spatial and temporal expression patterns to mediate physiologically specific recruitment of protein partners [30], as Figure 1 shows.
Importantly, these categories are not mutually exclusive. A single lncRNA may contain multiple structural or sequence domains that mediate interactions with different proteins either simultaneously or under different conditions. This modular and functionally dynamic architecture allows lncRNAs to act pleiotropically, playing distinct regulatory roles depending on the biological setting. The availability of protein partners, subcellular localization, and the lncRNA’s structural configuration collectively determine the functional output of these interactions [31]. This classification enhances our understanding of how lncRNAs interact with protein partners and contribute to a broad spectrum of cellular functions.

3. Biological Consequences of LPIs

3.1. Subcellular Localization and Functional Compartmentalization

Sequence-encoded signals are believed to guide lncRNA localization, yet their low sequence conservation and extensive alternative splicing complicate the prediction of subcellular distribution [32]. lncRNAs also exhibit pronounced tissue-specific expression, and their localization is often influenced by the availability of local protein partners, highlighting a cooperative interplay between lncRNAs and proteins in defining both spatial positioning and compartment-specific activity [33,34]. Interactions with RBPs help determine whether lncRNAs are retained in the nucleus or exported to the cytoplasm, especially when functional data is limited [35]. Chromatin-associated lncRNAs are typically found in the nucleus, participating in transcriptional regulation through chromatin remodeling [36]. However, recent evidence shows that some of these lncRNAs also localize to the cytoplasm, where they scaffold signaling protein complexes such as TLR–TRIF [37]. LncRNAs like LINC00473 and SAMMON are found at specific subcellular sites, including mitochondria and lipid droplet interfaces, via direct protein interactions. Some others exhibit dynamic localization patterns influenced by physiological conditions [38]. Given that classification of lncRNAs is often based on subcellular localization, understanding protein-mediated compartmentalization provides important clues to their biological roles. For example, in lung adenocarcinoma, reduced LINC00472 expression is closely linked to clinical outcomes, with its function dependent on interactions with proteins such as YBX1 [39].
Owing to their versatile modes of protein interaction, lncRNAs frequently act as structural and regulatory hubs that assemble protein complexes into distinct functional compartments within the cell. For example, GlycoLINC orchestrates glycolytic metabolism by bringing together key enzymes into a multi-component complex [40], while SLERT regulates RNA polymerase I activity via stoichiometric interaction with DDX21 [41]. In certain conditions, multiple lncRNAs co-localize with shared protein partners to form higher-order regulatory structures, such as the nuclear bodies in Prader–Willi syndrome, where sno-lncRNAs and SPA-lncRNAs recruit splicing regulators, including RBFOX2 and TDP43, through sequence-specific motifs [42,43,44]. lncRNAs also contribute to the formation of membraneless and phase-separated condensates. For example, NORAD sequesters PUM proteins within such condensates, modulating RBPs’ availability and function.

3.2. Regulation of Gene Expression

LncRNAs regulate gene expression through interactions with protein partners, ranging from influencing DNA repair, chromatin remodeling, and transcription [45,46].
For example, GUARDIN, a p53-inducible lncRNA, binds to BRCA1 and BARD1, facilitating the recruitment of DNA double-strand break (DSB) repair complexes [13]. Similarly, NORAD interacts with RBMX and TOP1 to form a ribonucleoprotein complex that maintains mitotic integrity and prevents chromosomal instability [47]. Multiple other lncRNAs, such as LINP1, NIHCOLE, and LRIK, enhance non-homologous end joining (NHEJ) by scaffolding Ku70/Ku80 and recruiting DNA-PKcs to DSB sites [48]. lncRNAs also regulate chromatin states and gene transcription. Some act by modifying chromatin architecture, such as L1 elements, which interact with EZH2 and SUZ12 to deposit H3K27me3 marks and regulate cortical development [49]. Others exert dual regulatory effects: HOTAIR activates GLI2 transcription via interaction with the androgen receptor (AR), while ZFAT-AS1 suppresses CDX2 by recruiting PRC2 to its promoter, leading to gene silencing through H3K27me3 [50,51].
Epigenetic gene regulation by lncRNAs frequently involves DNA methylation machinery and chromatin modifiers. For instance, CDKN2B-AS1 can form RNA–DNA triplexes to recruit proteins like EZH2 and CTCF, influencing local methylation status. The nuclear pore protein NUP153 represents another layer of lncRNA-mediated chromatin regulation. NUP153 modulates gene activation by coordinating CTCF and cohesin occupancy at topologically associating domain (TAD) boundaries, influencing bivalent gene expression in embryonic stem cells [52]. Similarly, Meg3 facilitates TAD formation at the Dlk1-Dio3 locus by binding CTCF, thus enabling cis-silencing through 3D chromatin architecture remodeling [53]. During epidermal differentiation, NEAT1 exemplifies lncRNA-mediated transcriptional control. Initially repressed by ΔNp63-HDAC1, NEAT1 levels rise upon differentiation, forming paraspeckles and associating with promoters of epithelial transcription factors to sustain their expression [54].

3.3. Modulation of Cellular Processes

LPIs influence a wide array of cellular functions by shaping the expression and activity of key regulatory molecules. These complexes contribute to essential processes such as cell proliferation, immune regulation, and metabolism in various signaling pathways.
For instance, SDNOR promotes granulosa cell proliferation and resistance to oxidative stress by inhibiting SOX9, a key factor in steroidogenesis and cell cycle regulation [55], while CHASERR enhances glioma progression by upregulating TRIM14 through miR-6893-3p sequestration and activating the PTEN/p-Akt/mTOR and Wnt/β-catenin pathways [56].
In muscle development, LncMREF cooperates with the chromatin remodeler Smarca5 to enhance chromatin accessibility and activate MyoD expression via recruitment of p300/CBP and H3K27ac deposition [57], while Has2os promotes myocyte fusion and differentiation by inhibiting the JNK/MAPK pathway [58]. Similarly, SNHG1 enhances STAT3 phosphorylation, promoting chondrogenic differentiation and angiogenesis in jaw bone marrow mesenchymal stem cells [59]. In tumor settings, lncRNAs reshape the immune landscape by modulating macrophage plasticity, T cell activity, and MDSC function, contributing to immune evasion and therapy resistance [60]. In hepatocellular carcinoma, MIAT promotes immune escape by stabilizing STAT3 through miR-411-5p inhibition, resulting in increased PD-L1 expression [61]. Integrative studies have also revealed immune-related lncRNA–protein networks across diseases like pulpitis, where distinct molecular subtypes have been characterized [62]. Moreover, LPIs contribute to metabolic regulation by organizing enzymatic complexes, as exemplified by glycoLINC, which scaffolds glycolytic enzymes to coordinate metabolic flux [6,63].

3.4. Implications in Human Diseases

The biological consequences of LPIs are increasingly recognized in diverse diseases. For instance, LINC00472 is significantly downregulated in lung adenocarcinoma, correlating with poor prognosis and TNM stage [38]. MALAT1 aggravates renal tubular injury in diabetic nephropathy by enhancing AMPK/mTOR signaling through stabilization of the LIN28A–Nox4 interaction [64]. In Prader–Willi syndrome, the loss of sno-lncRNAs and SPA-lncRNAs from chromosome 15q11–13 disrupts nuclear body formation by impairing interactions with splicing factors such as RBFOX2 and TDP43 [42,43]. A substantial number of LPIs are implicated in cellular signaling pathways, and many have been experimentally validated as contributors to disease progression. These data are documented in comprehensive resources like RAID, NONCODE, and NPInter [65,66,67], while specialized databases such as Lnc2Cancer and LncPCD provide focused insights into LPIs involved in cancer and programmed cell death [68,69]. Figure 2 summarizes recent findings on how LPIs are implicated in disease processes.

4. Experimental Approaches for Studying LPIs

4.1. Identification of LPIs

A variety of technologies that focus on either proteins or lncRNAs have been developed to identify LPIs nowadays [150]. Experimental methods generally rely on the pull-down principle and fall into categories of protein-centric and RNA-centric approaches, as Figure 3 shows.
In protein-centric techniques, the target protein is usually isolated at an early stage, followed by sequencing of associated lncRNA to map the interactions along the transcriptome. In the case of in vitro performances, the protein of interest is immobilized on a matrix, and a synthetic lncRNA library flows through the bound protein. High-affinity lncRNAs (Hits) are pulled down and subjected to sequencing. Advanced representatives are RNA Bind-n-Seq (RBNS) methods, which allow statistical modeling and the determination of dissociation constants between RNA and its binding proteins by deep sequencing [151,152]. A limitation of this method is that it is difficult to distinguish false positives. LncRNA hits that do not bind a protein inherently may show certain affinity and get selected, particularly when selection cycles are not conducted under stringent conditions. To mitigate this, affinity thresholds should be adjusted to reflect natural ligand interactions, and RNA libraries should be generated from cellular transcriptomes or by utilizing genomic fragments [153].
Alternatively, in vivo approaches have gained popularity in recent years. In vivo protein-centric methods usually utilize immunoprecipitation (IP) to isolate RNA-protein complexes, followed by co-purified RNA detection using microarrays (RIP-chip, RNA Immunoprecipitation-Chip) or RNA immunoprecipitation sequencing (RIPSeq) [154,155]. While these methods identify bound transcripts, they do not provide direct information about the binding site. Advanced methods were developed to recognize the binding sites and screen bound complexes in parallel with high throughput. For example, crosslinking immunoprecipitation RNA-sequencing (CLIP-Seq) and some of its variants, including HITS-CLIP, PAR-CLIP, and iCLIP [156,157]. These methods take advantage of covalent bonds induced by ultraviolet (UV) light between RNA and protein at the binding site, enabling the isolation of protein–RNA complexes by immunoprecipitation at high accuracy. The bound RNA is subsequently reverse-transcribed for next-generation sequencing. Crosslinking protein to modified lncRNA facilitates precise identification of protein types and their binding sites within the lncRNA sequence [158,159].
To enhance specificity and reproducibility, enhanced CLIP (eCLIP) incorporates an input control and has become a standardized technique within the ENCODE [160]. In addition, formaldehyde crosslinking immunoprecipitation (fCLIP) uses formaldehyde instead of UV to capture interactions that are otherwise difficult to crosslink, expanding the detectable spectrum of RNA–protein complexes [161]. Given the sequencing-based nature of these techniques, appropriate computational tools such as HTSeq-CLIP are essential for accurate data interpretation and downstream analysis [162].
To detect novel RBPs that interact with specific lncRNAs, an RNA-centric approach is commonly used instead (Figure 3). In this technique, the RNA of interest is purified and used as bait to capture interacting proteins. The associated protein complexes are then identified by enzymatically digesting the RNA and analyzing the proteins using mass spectrometry (MS), protein arrays, or other techniques [163,164]. In vitro-transcribed (IVT) RNAs tagged with biotin can be captured using streptavidin-conjugated beads, facilitating the isolation of the interacting protein network [165].
Due to the fact that synthetic RNA may misfold, leading to nonspecific binding with proteins in cell lysis solution. An in vivo RNA-centric method was developed to avoid this problem and preserve the context of natural RNA-protein interactions [166,167,168]. For example, RNA purification-based chromatin isolation (ChIRP), capture hybridization analysis of RNA targets (CHART), and RNA antisense purification (RAP) all isolate the relevant endogenous lncRNA using antisense oligonucleotides [169,170,171,172]. Ultimately, proteins that are eluted undergo analysis via mass spectrometry (MS). Certain modifications were also employed to explore protein with a specific RNA. For example, Pant et al. reported a method called MORPH-MS (Multiple Oligo-assisted RNA Pulldown via Hybridization) that uses a biotinylated oligo to collect all the antisense oligos rather than several separate biotinylated oligos. This method effectively extracts the relevant RNA along with its associated proteins [173]. One limitation of these methods is that their performance often depends on the efficiency of oligo hybridization and the abundance of target transcripts, making optimization crucial for low-expression lncRNAs. More recently, proximity labeling-based RNA-centric approaches such as RPL and CARPID have expanded the landscape of detectable LPIs in live cells. These techniques rely on catalytically inactive Cas13 (dCas13) fused to enzymes like APEX2 or biotin ligase, enabling biotinylation of proteins in close proximity to a target lncRNA. This allows comprehensive proteomic profiling of lncRNA-associated proteins under native cellular conditions [174,175].
In addition to experimental methods, in silico methods such as artificial neural networks have been developed to predict LPIs. These include integrated machine learning, combined graph auto-encoders (LPICGAE), deep autoencoder & marginal fisher analysis (DFRPI), and DeepLPI [176,177,178,179]. Further examples of similar techniques can be found in articles [180,181,182,183]. Moreover, online tools have been developed and are available for inputting predictions directly, for example, RNAincoder, SoCube, Nucpred, DeepBtoD, DeepA-RBPBS, and PST-PRNA [184,185,186,187,188,189]. In addition, databases such as POSTAR3 integrate large-scale CLIP-seq datasets with functional annotations to systematically analyze RBP binding sites, RNA structural features, disease-associated mutations, and miRNA-mediated decay events, thereby offering a multi-dimensional reference for CLIP data interpretation [190].
Compared with experimental approaches, in silico tools offer high-throughput screening capabilities and require no physical material, making them suitable for prioritizing candidate LPIs. However, prediction quality heavily depends on the training data and algorithmic features. Thus, experimental validation remains essential, particularly when moving toward functional interpretation. Table 1 illustrates the characteristics of common methods for detecting LPIs.

4.2. Structural Insights into LncRNA–Protein Binding

Most lncRNAs possess higher-order structures that are often critical for their function. Various methods have been developed to study the structure of lncRNAs. Notably, some studies not only address the structural prediction challenge but also explore its application in RNA-based therapeutic development. In the case of HOTAIR, which promotes EMT by enabling Snail-mediated EZH2 recruitment, a deletion mutant (HOTAIR-sbid) retaining the Snail-binding domain but lacking the EZH2-binding region acted as a dominant negative. HOTAIR-sbid impaired tumor cell invasiveness, motility, and TGFβ-induced EMT, highlighting how domain-based strategies can circumvent structural challenges in lncRNA-targeted therapy [204]. From functional analysis, the roles of lncRNAs can be inferred even without full structural resolution. For example, chemical probing of lncTCF7, a conserved lncRNA implicated in cancer, revealed modular structural domains and conserved regions, one of which overlaps with a known protein-binding site. As the first structural model of lncTCF7, this work highlights how domain-level insights can guide functional understanding in the absence of complete structural information [205]. We will further discuss the methods used to study lncRNA structure in the following sections.

4.2.1. Probing LncRNA Secondary Structure

In lncRNA molecules, complementary base pairs typically form structures such as bulges, junctions, loops, helices, subdomains, and pseudoknots, which are defined as secondary structures [206]. In many cases, the function of lncRNA is affected, and to some extent, determined by their highly conserved secondary structures. Especially when they provide recognition sites for unique protein components. Therefore, the knowledge of secondary structure is a primary and essential information source for the identity of protein interplay [207,208]. For instance, NORAD interacts with PUMILIO proteins to preserve genomic stability. An analysis of its sequence identified multiple repeated secondary domains (NRUs). Each domain features PUMILIO response elements (PRE) that exhibit strong and specific binding to PUMILIO proteins (PUM1 and PUM2) [209,210].
Conventional probe methods primarily rely on thermodynamic calculations and dynamic programming. Such as in vitro chemical probing, which has produced extremely accurate secondary structures that can also be confirmed by X-ray crystallography [211]. It’s worth noting that a single sequence of RNA (e.g., riboswitch RNA) may form multiple and mutually competitive secondary structures to determine certain functions. In this context, the binding ligand concentration causes a change in the secondary structure’s equilibrium, which either creates or destroys an ON/OFF transcriptional terminator state [212]. Meanwhile, certain RNA can dynamically adjust secondary structures in response to temperature changes [213]. The development of high-throughput sequencing (HTS) technologies has revolutionized RNA structure analysis, enabling the simultaneous examination of thousands of RNA molecules or entire transcriptomes. Additionally, the RNA-protein interaction interface can be indirectly probed by observing changes when comparing free lncRNA with RNA-bound states. These advancements have greatly enhanced our understanding of RNA structure [214,215]. Recently, a method called LLPS-CLIR-MS (liquid-liquid phase separation combined with cross-linking of isotope-labeled RNA and tandem mass spectrometry) was reported to investigate protein-RNA interactions inside biomolecular condensates. This technique enables the characterization of RNA-protein interactions within condensates at residue-specific resolution [216].
Once homogenous lncRNA is purified, advanced RNA structure probing techniques such as SHAPE-MaP and DMS-MaP can be applied in combination with next-generation sequencing to enable high-throughput structural analysis and accurate determination of secondary structures. These approaches provide a powerful framework for exploring lncRNA structure-function relationships [217]. SHAPE-MaP and DMS-MaP techniques can probe lncRNA flexibility and, when combined with other methods, provide insights into RNA structure within the cellular context by characterizing in vivo nucleotide dynamics [218]. Recently, a method was reported that enables accurate RNA structure probing of all four nucleotides in living cells. By leveraging the unique mutational signature of N1-methylguanine, it improves DMS profiling to capture guanine reactivity, providing richer structural data and supporting more precise RNA structure modeling [219].
As an alternative, a variety of computational techniques, including machine learning, multiple sequence alignment, free-energy estimates, and direct coupling analysis, can be used to predict the secondary structure of RNA [220,221,222,223]. For example, DiffScan, a method for high-resolution analysis of structure probing data, was recently reported to have an improved accuracy in detecting SVRs (structurally variable regions). It can work across multiple platforms like icSHAPE and DMS-seq, showing robust performance in identifying SVRs at nucleotide resolution [224]. Other web server programs such as RNAstructure, Vfold2D, linear-time heuristic, REDfold, and bpRNA-align have also emerged [225,226,227,228,229].

4.2.2. Analysis of LncRNA Tertiary Structure

3D of lncRNAs is very important for understanding their function. LPIs enable the characterization of high-resolution 3D structures of lncRNA. Recent chemical probing-based next-generation sequencing (M2-seq) coupled with mutational profiles has the potential to provide tertiary structural information [230]. Probes like dimethyl sulfate (DMS) and SHAPE (Selective 2′-hydroxyl acylation analyzed by primer extension) react with RNA to reflect structural dynamics. To detect modifications, reverse transcription (RT) or third-generation sequencing methods (e.g., Oxford Nanopore) are commonly used. Techniques like SHAPE-MaP and DMS-MaP-Seq are commonly employed [219,231]. The BASH MaP method has been reported for studying RNA tertiary structure by measuring N7G (N7 position of guanosine) accessibility. When combined with the DAGGER (Deconvolution Assisted by N7G Gaps Enabled by Reduction and Depurination) pipeline, this method reveals alternative RNA conformations, especially for RNAs with G-quadruplexes. This approach has been shown to improve RNA structure prediction, overcoming limitations of traditional methods in analyzing complex RNA structures [232].
In addition, multiple computer algorithms were developed to identify RNA structural ensembles from large data generated by sequencing-based chemical probing, which show increasing potential; examples include DREEM (Detection of RNA Folding Ensembles using Expectation-Maximization), DaVinci (Determination of the Variation in the RNA Structure Conformation through Stochastic Context-Free Grammar), and DANCE-MaP (Deconvolution and Annotation of RiboNucleic Conformational Ensembles Measured by Mutational Profiling) [231,233,234]. Overall, all methods are missing a direct 3D map at the atomic range of the target lncRNA [8].
The tremendous advancement of structural biology technology, particularly cryoEM, has resulted in unparalleled successes in the understanding of proteins and protein complexes that have revolutionized our knowledge of ligand binding, complex formation, and self-assembly—all critical processes in biology [235,236]. Nevertheless, the number of individual lncRNA structural models currently available is limited. This constrains efforts to elucidate their enigmatic structure-function relationships. RNA structural biology has significantly lagged behind protein structural biology [1,237].
To some extent, techniques used for protein 3D characterization can also be applied to lncRNA. X-ray crystallography has succeeded in solving the 3D structures of some RNA [238,239]. Small-angle X-ray scattering experiments can be used to characterize the RNA systems that are too flexible or come in a variety of conformations with low resolution [240]. LncRNAs have also been studied using atomic force microscopy (AFM, Multimode 8, Nanoscope V, Bruker, Santa Barbara, CA, USA), which allows for the analysis of 3D structures in a solution-free environment [241]. In order to differentiate between extended and compact conformations of lncRNA systems, parameters such as 3D size have been evaluated using fluorescence correlation spectroscopy (FCS). For instance, HOTAIR was shown to be less compact than ribosomes but to display a more compact structure when compared to mRNA transcripts [242]. In summary, studies that solely focus on RNA systems have generated a large amount of structural data, but the scope of structure-function roles is still limited by the multiple faces of lncRNA characteristics (e.g., heterogeneity) [12]. Not to mention that characterizing the majority of lncRNA is still challenging due to their intrinsic properties, particularly those containing flexible segments.
Single LncRNA–Protein Complex Structural Analysis
Obtaining stable lncRNA samples is the key step for structural observation. Many lncRNAs adopt certain 3D shapes only when induced by binding partners, which are different from linear lncRNAs [240,243]. The protein binding partners help to stabilize lncRNAs for structure determination in these cases. Benefiting from advances in protein structure resolution, structure investigation of lncRNAs can be operationalized by indirectly visualizing the complexes formed by lncRNAs and proteins. Then, in principle, all available approaches used for proteins can be adapted to study lncRNA (Figure 4).
Accordingly, we propose a novel strategy to further explore the structural intricacies of lncRNA from the perspective of protein binding, which is ISD (Inducing-Screening-Decomposing). In this strategy, we can firstly induce a protein partner for stabilizing lncRNA. Aditionally, for lncRNA with multiple binding partners, screening for stable partners or parallel analysis is beneficial for comprehensive understanding. Since the lncRNAs harbor a huge range of heterogeneity in size. A significant number of IncRNAs interact with more than one protein; the structure study can be optimized according to functional sub-motifs that regulate the proteins they bind to (decomposing). We have successfully incorporated ISD strategy into our integrated analysis pipeline (refer to Figure 5). This is especially important when it comes to the functional characteristics of lncRNA, because low abundance and multiple functional roles in cells require lncRNA to interact stoichiometrically with target molecules so as to have detectable regulatory effects [30].
With regard to available techniques, recent advances in artificial intelligence (AI) demonstrate great potential in structural prediction [244,245]. Tools like AlphaFold and ProteinBERT (RFAA) have demonstrated remarkable accuracy in predicting protein structures [246,247]. Their accuracy decreases when predicting the binding mechanisms of complexes, especially at the interface [248,249]. An effective compensatory approach is to use proteins with known structures to predict unknown lncRNA partners. Nevertheless, AI-driven deep learning algorithms are rapidly evolving and continuously improving their accuracy on protein complexes. For example, RoseTTAFoldNA has been shown to efficiently generate 3D structural models for protein-RNA complexes with higher accuracy [250]. Other deep-learning-based methods include DeepDISOBind and AI-integrated networks, while MSAs (multiple sequence alignments) provide essential input data for these tools [251,252,253].
Overall, lncRNA structure prediction can be approached in multiple steps, and integrating protein-binding information can significantly enhance prediction accuracy. For example, first identifying protein interaction partners (e.g., via CLIP-seq data), then predicting secondary structure with RNA-binding constraints using tools like SPOT-RNA, followed by protein-guided tertiary modeling through integrative methods such as RoseTTAFoldNA or constrained docking, and finally validating models with geometric checks and simulations. For instance, AlphaFold3 has been released with the aim of extending accurate predictions to RNA structures and RNA–protein complexes. However, recent evaluations suggest that AlphaFold3 currently struggles with non-canonical RNA interactions, and its predictions in this domain remain unreliable, especially where training data are scarce. It is therefore recommended to combine AI-based predictions with docking or structure-informed approaches for now [254]. Table 2 summarizes common AI-based methods for modeling lncRNA structures and their complexes with proteins.
In addition, databases such as MARS (Master database of All possible RNA Sequences) and ViRBase v3.0 greatly expand the scope of RNA homology search by integrating multiple large-scale RNA and genome resources, thereby improving alignment quality and structure prediction accuracy [255,256].
Table 2. Common AI-based methods for modeling lncRNA structures and their complexes.
Table 2. Common AI-based methods for modeling lncRNA structures and their complexes.
Tool NameRNA
Struct.
lPI
Struct.
Key FeaturesSuitabilityRef.
RNA-ComposerYesNoFast conversion from 2D to 3D structureSuitable for short lncRNA fragments with known secondary structure, web-based and fast[257]
3dRNA v2.0YesLimitedAssembles 3D structure from motifs and 2D inputSuitable for moderate-length lncRNAs, accuracy depends on quality of 2D input[258]
P-FARFAR2YesLimitedParallelized enhancement of FARFAR2, multithreaded greedy sampling for low-energy structure assemblySuitable for high-precision RNA 3D modeling, better efficiency and accuracy over FARFAR2, especially for complex/larger RNAs[259]
FARNA/FARFAR2YesLimitedHigh-precision folding, but computationally intensiveBest for small lncRNA segments, high-quality modeling with heavy compute requirements[260]
RNA-MSMIndirectlyNoMSA-based RNA language model, captures structural information via attention maps and embeddingsOutperforms BERT-like RNA models in predicting base-pairing and solvent accessibility, suitable for structure-function tasks[261]
MoEFold2DNo (2D)NoMixture-of-experts model combining DL and physics-based predictions, auto-detects in/out-of-distribution sequencesOffers high accuracy for in-distribution and robust predictions, useful when training/test domains differ[262]
RNA-LLM FoldingNoNoEvaluates multiple pretrained RNA language models for RNA secondary structure predictionProvides benchmark datasets and unified experimental framework, shows LLMs can improve prediction in high-homology regions, with limitations in generalization[263]
RPI-SENoYesEnsemble model using PWM, Legendre moments (proteins), and k-mer features (ncRNAs), high accuracy and robustnessEffective computational predictor for ncRNA-protein interactions, suitable for accelerating interaction studies with sequence data only[264]
SPOT-RNA2YesNoHigh-accuracy secondary structure prediction, supports pseudoknotsGreat for generating input structures for 3D modeling of lncRNA[261]
MXfold2NoNoCombines neural networks and thermodynamics for better generalizationFast and accurate, ideal for batch secondary structure prediction[265]
RNAstructureNoNoSupports SHAPE data and pseudoknots, energy minimizationStable and widely used, excellent for combining with experimental data[266]
IntaRNANoYesPredicts RNA–protein interaction regionsUseful for predicting lncRNA binding regions with proteins[267]
catRAPIDNoYesSequence-based scoring of RNA–protein interaction strengthUseful for large-scale screening of LPIs[196]
RPI-EDLCNNoYesEnsemble deep learning framework based on CapsuleNet, integrates sequence, secondary structure, motif, and physicochemical features, used for feature extractionHigh accuracy in predicting ncRNA-protein interactions across multiple datasets, effective across species[268]
LPI-MFFNo YesMulti-source information fusion (PPI, sequence, structure, physico-chemical), feature selection by random forestHigh accuracy and robustness across multiple datasets, good generalization for RPI prediction[269]
DRPScoreYesYesDeep learning model for identifying native-like RNA-protein complex structures across docking types (bound/unbound)Suitable for RNA-protein complex modeling, high accuracy in selecting native-like structures even under flexible docking scenarios[25]
AlphaFold3YesYesAccurate protein 3D structure prediction, limited in RNA/RNA-protein complexProvides protein structure for docking; essential if structure is unknown[254]
RoseTTAFoldYesYesDeep learning-based 3D structure prediction for RNA-protein complexes, with confidence scoresSuitable for predicting structures of complexes without known homology [250]
Experimental methods play a crucial role in validating these predicted structural models. X-ray crystallography has long provided high-resolution insights into protein-RNA complexes, providing high-resolution structures that reveal detailed molecular interactions once the target lncRNA is stabilized. For instance, TRIM71 interacts with the lncRNA Trincr1 to repress the FGF/ERK pathway in mouse ESCs (embryonic stem cells). Recently an X-ray crystallography study determined an RNA motif in Trincr1 that is specifically recognized by TRIM71 [270]. Nuclear magnetic resonance (NMR) imaging has been extensively used to study small-scale systems. It provides a precise way to record details about RNA dynamics, its various configurations, and the speeds at which these configurations change [271]. Fragmented (decomposing) regions or motifs of lncRNA with a stabilized protein can be employed in an NMR pipeline [240,241].
With recent advances in cryo-electron microscopy (cryoEM), the number of RNA and protein-RNA structure models in the PDB has grown significantly. For instance, the non-coding 7S RNA inhibits transcription via mitochondrial RNA polymerase dimerization. The dimerization of POLRMT is induced by interactions with 7S RNA. CryoEM analysis, focusing on 2D classification and 3D reconstruction, obtained a low-resolution structure of POLRMT-POLRMT complexes. By using decomposition and negative control methods, researchers were able to map the density of 7S RNA, which appears to bind and regulate the wrapping of POLRMT during dimerization [272]. In the study of lncRNA ADIPINT interacting with pyruvate carboxylase in adipocyte metabolism, the RNA was sequentially truncated to explore its binding patterns with proteins, using methods analogous to the decomposition techniques proposed in our ISD proposal [273].
Integrated Structural Approaches
It is essential to select and optimize approaches tailored to each target’s characteristics. While a single method may not fully capture lncRNA dynamics, integrated structural analysis can overcome these limitations and enhance the accuracy of structural determination [274,275]. As Figure 5 shows, we propose an overall diagram to reveal the structure-function relationship of lncRNA. ISD strategies are expected to be used during implementation.
A typical example is the ribosome, which has been examined using various technologies over time. Generally, small-angle X-ray scattering (SAXS) is generally used as an initial approach for higher-resolution structure determination, paving the way for techniques like X-ray crystallography or cryo-electron microscopy. The sample preparation requirements for SAXS are relatively less stringent. A low-resolution structure obtained from SAXS is a valuable resource for further high-resolution modeling and complex studies in cryo-EM. For example, SAXS was utilized to determine the low-resolution structures of the Braveheart lncRNA and the Braveheart-CNBP ribonucleoprotein complex. These results were supported by computational 3D modeling and phylogenetic analysis [240].

5. Development of Novel Biomedical Interventions Targeting LPIs

Emerging technologies are being developed to target lncRNA-protein for disease treatment, given its essential roles in biological processes. Traditionally, methods such as small interfering RNAs (siRNAs) and antisense oligonucleotides (ASOs) have been used to degrade lncRNA by directly targeting through base pairing. A notable concern for these methods is the off-target effects [276,277,278]. Advanced approaches, such as CRISPR/Cas9, can be applied for lncRNA knockout regardless of cellular location but are limited by a relatively narrow spectrum of targetable lncRNAs [279,280].
Since LPIs play a vital role in diverse diseases, as discussed above, specifically focusing on the interactions provides creative solutions for broadening the range of targetable LncRNA in drug development. Two main approaches are currently available: The first approach involves the development of small molecules that target lncRNA-bound protein partners to intervene in lncRNA-associated physiological processes. In fact, protein-based drug design occupies a very important position in pharmaceutical areas [281] due to factors such as the close connection between protein structure and function, the abundance of atomic models obtained by state-of-art technologies, and the potential for compound optimization [282,283,284]. For example, Lin28 is overexpressed in many cancer cells and disrupts the maturation of RNA let-7 by directly binding to let-7 microRNA precursors. Small-molecule Lin28 inhibitors have been discovered to prevent Lin28 from binding to let-7, presenting a promising therapeutic strategy [285]. Numerous Lin28 inhibitors have been identified through various high-throughput screening (HTS) techniques [286,287].
The second approach focuses on targeting the interaction interface by designing molecules that disrupt the spatial structure of lncRNAs or interfere with LPIs. For instance, even though EZH2 (Enhancer of zeste homolog 2) is an attractive drug target for cancers, the design of EZH2 inhibitors is constrained by diverse undesigned effects on gene expression. The lncRNA BDNF-AS (brain-derived neurotrophic factor antisense) binds EZH2 in a concentration-dependent manner. AlphaScreen was employed to quantify lncRNA-EZH2 interaction, leading to the identification of a small molecule, ellipticine, which effectively inhibits the interaction between BDNF-AS and EZH2 [288,289]. This study highlights the effectiveness of high-throughput screening in identifying regulators of LPIs. Furthermore, it has been reported that the ENE (element for nuclear expression) triplexes in lncRNA MALAT1 and KSHV PAN serve as unique platforms for protein binding, such as MALAT1 and KSHV PAN, and serve as unique platforms for protein binding, including interactions with methyltransferase-like protein 16 (METTL16). Several small ligands have been discovered that selectively bind to the ENE triplexes and disrupt these protein-lncRNA interactions [290,291].

6. Conclusions

As more lncRNAs have been discovered in recent years, their roles in cellular activities are receiving growing attention. Understanding the cellular mechanism of lncRNA, particularly for those with advanced structures, is crucial for studying the significant correlation between structure and function. However, due to the distinguishable properties compared to proteins, it is difficult to explore the structure-function relationship of lncRNA solely from resolving certain configurations formed by their sequences. Moreover, the availability of high-resolution structures for individual lncRNAs remains extremely limited. Additionally, predicting the structure or function of lncRNAs through sequence alignment is largely constrained by the low conservation of their sequences.
We emphasize the importance of focusing on the LPI in lncRNA research, as proteins have significantly contributed to a more comprehensive understanding of lncRNA in various contexts. By expanding attention from lncRNA itself to its interaction with the primary protein ligand, LPIs can be treated as functional units. This approach, on the one hand, enables deeper insights into the cellular roles of lncRNAs from a broader biological process perspective. On the other hand, utilizing proteins as stabilizing ligands for lncRNAs during structural investigations can effectively address many of the challenges faced in lncRNA research, as discussed in the main section.
Protein binding is a critical step for lncRNAs to exert their functions. Structural information obtained from solving lncRNA-protein complexes provides a robust foundation for interpreting the structure-function relationships of lncRNAs, which could subsequently facilitate the development of novel therapeutic strategies. Except for structure-based small compound screening from bond protein or lPIs. Strategies such as antisense oligonucleotides and siRNAs can all be designed to target functional domains or disrupt essential structural regions of lncRNAs, thereby selectively interfering with their pathological roles. While notable progress has been made, the clinical application of lncRNA-targeted treatments still faces challenges. These include poor sequence conservation across species, extensive alternative splicing, and tissue-specific localization, all of which complicate the development of reliable disease models and functional studies. Off-target effects and delivery specificity also present ongoing concerns, particularly for systemically administered therapeutics. Integrative strategies such as transcriptome-wide association studies (TWAS), along with improved disease models and structure-based screening, will be essential to overcome these barriers and facilitate the development of safe, effective, and precise lncRNA-based interventions. In this review, we have examined the physiological roles of lncRNAs and provided a comprehensive overview of recent advances in LPIs. We further summarized progress in lncRNA-associated diseases and structure-based drug design centered on LPIs, aiming to inspire further developments of innovative treatments in the future.

Author Contributions

Z.W. and M.A. designed and wrote the initial draft, K.L. edited and corrected the manuscript, Z.D., Y.J. and K.L. revised the manuscript and the figures. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundations of China (No, 82472998), the Postdoctoral Fellowship Program of CPSF (No, GZC20241560).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The 3D model of lncRNA-protein complex illustrated in this study was made by PyMOL (Version 3.1, Schrödinger, LLC) from an available structure in PDB database (ID: 3ho8), all the figures were created with the assistance of BioRender.

Conflicts of Interest

The authors declare no conflicts of interest.

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]
  2. Wang, Z.; Xie, C.; Chen, X. Diagnostic and Therapeutic Role of Non-Coding RNAs Regulating Programmed Cell Death in Melanoma. Front. Oncol. 2024, 14, 1476684. [Google Scholar] [CrossRef]
  3. Ponting, C.P.; Haerty, W. Genome-Wide Analysis of Human Long Noncoding RNAs: A Provocative Review. Annu. Rev. Genom. Hum. Genet. 2022, 23, 153–172. [Google Scholar] [CrossRef]
  4. Chen, L.-L.; Kim, V.N. Small and Long Non-Coding RNAs: Past, Present, and Future. Cell 2024, 187, 6451–6485. [Google Scholar] [CrossRef]
  5. Rocca, R.; Grillone, K.; Citriniti, E.L.; Gualtieri, G.; Artese, A.; Tagliaferri, P.; Tassone, P.; Alcaro, S. Targeting Non-Coding RNAs: Perspectives and Challenges of in-Silico Approaches. Eur. J. Med. Chem. 2023, 261, 115850. [Google Scholar] [CrossRef]
  6. Constanty, F.; Shkumatava, A. lncRNAs in Development and Differentiation: From Sequence Motifs to Functional Characterization. Development 2021, 148, dev182741. [Google Scholar] [CrossRef]
  7. Ma, H.; Jia, X.; Zhang, K.; Su, Z. Cryo-EM Advances in RNA Structure Determination. Sig. Transduct. Target Ther. 2022, 7, 58. [Google Scholar] [CrossRef]
  8. Deng, J.; Fang, X.; Huang, L.; Li, S.; Xu, L.; Ye, K.; Zhang, J.; Zhang, K.; Zhang, Q.C. RNA Structure Determination: From 2D to 3D. Fundam. Res. 2023, 3, 727–737. [Google Scholar] [CrossRef]
  9. Alvarez-Dominguez, J.R.; Lodish, H.F. Emerging Mechanisms of Long Noncoding RNA Function during Normal and Malignant Hematopoiesis. Blood 2017, 130, 1965–1975. [Google Scholar] [CrossRef]
  10. Ribeiro, D.M.; Zanzoni, A.; Cipriano, A.; Delli Ponti, R.; Spinelli, L.; Ballarino, M.; Bozzoni, I.; Tartaglia, G.G.; Brun, C. Protein Complex Scaffolding Predicted as a Prevalent Function of Long Non-Coding RNAs. Nucleic Acids Res. 2018, 46, 917–928. [Google Scholar] [CrossRef]
  11. Chu, C.; Spitale, R.C.; Chang, H.Y. Technologies to Probe Functions and Mechanisms of Long Noncoding RNAs. Nat. Struct. Mol. Biol. 2015, 22, 29–35. [Google Scholar] [CrossRef]
  12. Graf, J.; Kretz, M. From Structure to Function: Route to Understanding lncRNA Mechanism. BioEssays 2020, 42, 2000027. [Google Scholar] [CrossRef]
  13. Statello, L.; Guo, C.-J.; Chen, L.-L.; Huarte, M. Author Correction: Gene Regulation by Long Non-Coding RNAs and Its Biological Functions. Nat. Rev. Mol. Cell Biol. 2021, 22, 159. [Google Scholar] [CrossRef]
  14. Shaath, H.; Vishnubalaji, R.; Elango, R.; Kardousha, A.; Islam, Z.; Qureshi, R.; Alam, T.; Kolatkar, P.R.; Alajez, N.M. Long Non-Coding RNA and RNA-Binding Protein Interactions in Cancer: Experimental and Machine Learning Approaches. Semin. Cancer Biol. 2022, 86, 325–345. [Google Scholar] [CrossRef]
  15. Nadhan, R.; Isidoro, C.; Song, Y.S.; Dhanasekaran, D.N. Signaling by LncRNAs: Structure, Cellular Homeostasis, and Disease Pathology. Cells 2022, 11, 2517. [Google Scholar] [CrossRef]
  16. Yadav, V.K.; Kumar, A.; Tripathi, P.P.; Gupta, J. Long Noncoding RNAs in Intestinal Homeostasis, Regeneration, and Cancer. J. Cell. Physiol. 2021, 236, 7801–7813. [Google Scholar] [CrossRef]
  17. He, D.; Zheng, J.; Hu, J.; Chen, J.; Wei, X. Long Non-Coding RNAs and Pyroptosis. Clin. Chim. Acta 2020, 504, 201–208. [Google Scholar] [CrossRef]
  18. Gerber, A.P. RNA-Centric Approaches to Profile the RNA–Protein Interaction Landscape on Selected RNAs. ncRNA 2021, 7, 11. [Google Scholar] [CrossRef]
  19. Honorato, R.V.; Trellet, M.E.; Jiménez-García, B.; Schaarschmidt, J.J.; Giulini, M.; Reys, V.; Koukos, P.I.; Rodrigues, J.P.G.L.M.; Karaca, E.; van Zundert, G.C.P.; et al. The HADDOCK2.4 Web Server for Integrative Modeling of Biomolecular Complexes. Nat. Protoc. 2024, 19, 3219–3241. [Google Scholar] [CrossRef]
  20. Zheng, J.; Hong, X.; Xie, J.; Tong, X.; Liu, S. P3DOCK: A Protein–RNA Docking Webserver Based on Template-Based and Template-Free Docking. Bioinformatics 2020, 36, 96–103. [Google Scholar] [CrossRef]
  21. Caudron-Herger, M.; Jansen, R.E.; Wassmer, E.; Diederichs, S. RBP2GO: A Comprehensive Pan-Species Database on RNA-Binding Proteins, Their Interactions and Functions. Nucleic Acids Res. 2021, 49, D425–D436. [Google Scholar] [CrossRef]
  22. Chojnowski, G.; Waleń, T.; Bujnicki, J.M. RNA Bricks—A Database of RNA 3D Motifs and Their Interactions. Nucl. Acids Res. 2014, 42, D123–D131. [Google Scholar] [CrossRef]
  23. Kirsanov, D.D.; Zanegina, O.N.; Aksianov, E.A.; Spirin, S.A.; Karyagina, A.S.; Alexeevski, A.V. NPIDB: Nucleic Acid—Protein Interaction Database. Nucleic Acids Res. 2012, 41, D517–D523. [Google Scholar] [CrossRef]
  24. Philip, M.; Chen, T.; Tyagi, S. A Survey of Current Resources to Study lncRNA-Protein Interactions. ncRNA 2021, 7, 33. [Google Scholar] [CrossRef]
  25. Zeng, C.; Jian, Y.; Vosoughi, S.; Zeng, C.; Zhao, Y. Evaluating Native-like Structures of RNA-Protein Complexes through the Deep Learning Method. Nat. Commun. 2023, 14, 1060. [Google Scholar] [CrossRef]
  26. Yang, Y.; Wen, L.; Zhu, H. Unveiling the Hidden Function of Long Non-Coding RNA by Identifying Its Major Partner-Protein. Cell Biosci. 2015, 5, 59. [Google Scholar] [CrossRef]
  27. Kaczynski, T.J.; Au, E.D.; Farkas, M.H. Exploring the lncRNA Localization Landscape within the Retinal Pigment Epithelium under Normal and Stress Conditions. BMC Genom. 2022, 23, 539. [Google Scholar] [CrossRef]
  28. Wang, K.C.; Chang, H.Y. Molecular Mechanisms of Long Noncoding RNAs. Mol. Cell 2011, 43, 904–914. [Google Scholar] [CrossRef]
  29. Ahmad, P.; Bensaoud, C.; Mekki, I.; Rehman, M.; Kotsyfakis, M. Long Non-Coding RNAs and Their Potential Roles in the Vector–Host–Pathogen Triad. Life 2021, 11, 56. [Google Scholar] [CrossRef]
  30. Kopp, F.; Mendell, J.T. Functional Classification and Experimental Dissection of Long Noncoding RNAs. Cell 2018, 172, 393–407. [Google Scholar] [CrossRef]
  31. Palihati, M.; Saitoh, N. RNA in Chromatin Organization and Nuclear Architecture. Curr. Opin. Genet. Dev. 2024, 86, 102176. [Google Scholar] [CrossRef]
  32. Bridges, M.C.; Daulagala, A.C.; Kourtidis, A. LNCcation: lncRNA Localization and Function. J. Cell Biol. 2021, 220, e202009045. [Google Scholar] [CrossRef]
  33. Poloni, J.d.F.; Oliveira, F.H.S.d.; Feltes, B.C. Localization Is the Key to Action: Regulatory Peculiarities of lncRNAs. Front. Genet. 2024, 15, 1478352. [Google Scholar] [CrossRef]
  34. Li, Z.; Tian, J.-M.; Chu, Y.; Zhu, H.-Y.; Wang, J.-J.; Huang, J. Long Non-Coding RNA PVT1 (PVT1) Affects the Expression of CCND1 and Promotes Doxorubicin Resistance in Osteosarcoma Cells. J. Bone Oncol. 2023, 43, 100512. [Google Scholar] [CrossRef]
  35. Zuckerman, B.; Ulitsky, I. Predictive Models of Subcellular Localization of Long RNAs. RNA 2019, 25, 557–572. [Google Scholar] [CrossRef]
  36. Leucci, E.; Vendramin, R.; Spinazzi, M.; Laurette, P.; Fiers, M.; Wouters, J.; Radaelli, E.; Eyckerman, S.; Leonelli, C.; Vanderheyden, K.; et al. Melanoma Addiction to the Long Non-Coding RNA SAMMSON. Nature 2016, 531, 518–522. [Google Scholar] [CrossRef]
  37. Aznaourova, M.; Janga, H.; Sefried, S.; Kaufmann, A.; Dorna, J.; Volkers, S.M.; Georg, P.; Lechner, M.; Hoppe, J.; Dökel, S.; et al. Noncoding RNA MaIL1 Is an Integral Component of the TLR4–TRIF Pathway. Proc. Natl. Acad. Sci. USA 2020, 117, 9042–9053. [Google Scholar] [CrossRef]
  38. Tran, K.-V.; Brown, E.L.; DeSouza, T.; Jespersen, N.Z.; Nandrup-Bus, C.; Yang, Q.; Yang, Z.; Desai, A.; Min, S.Y.; Rojas-Rodriguez, R.; et al. Human Thermogenic Adipocyte Regulation by the Long Noncoding RNA LINC00473. Nat. Metab. 2020, 2, 397–412. [Google Scholar] [CrossRef]
  39. Deng, X.; Xiong, W.; Jiang, X.; Zhang, S.; Li, Z.; Zhou, Y.; Xiang, B.; Zhou, M.; Li, X.; Li, G.; et al. LncRNA LINC00472 Regulates Cell Stiffness and Inhibits the Migration and Invasion of Lung Adenocarcinoma by Binding to YBX1. Cell Death Dis. 2020, 11, 945. [Google Scholar] [CrossRef]
  40. Zhu, Y.; Jin, L.; Shi, R.; Li, J.; Wang, Y.; Zhang, L.; Liang, C.-Z.; Narayana, V.K.; De Souza, D.P.; Thorne, R.F.; et al. The Long Noncoding RNA glycoLINC Assembles a Lower Glycolytic Metabolon to Promote Glycolysis. Mol. Cell 2022, 82, 542–554.e6. [Google Scholar] [CrossRef]
  41. Wu, M.; Xu, G.; Han, C.; Luan, P.-F.; Xing, Y.-H.; Nan, F.; Yang, L.-Z.; Huang, Y.; Yang, Z.-H.; Shan, L.; et al. lncRNA SLERT Controls Phase Separation of FC/DFCs to Facilitate Pol I Transcription. Science 2021, 373, 547–555. [Google Scholar] [CrossRef]
  42. Sledziowska, M.; Winczura, K.; Jones, M.; Almaghrabi, R.; Mischo, H.; Hebenstreit, D.; Garcia, P.; Grzechnik, P. Non-Coding RNAs Associated with Prader–Willi Syndrome Regulate Transcription of Neurodevelopmental Genes in Human Induced Pluripotent Stem Cells. Hum. Mol. Genet. 2023, 32, 608–620. [Google Scholar] [CrossRef]
  43. Li, R.; Fox, A.H. SPArking Interest in the Long Noncoding RNA World: A New Class of 5′ SnoRNA-Stabilized LncRNA That Influences Alternative Splicing. Mol. Cell 2016, 64, 435–437. [Google Scholar] [CrossRef]
  44. Wu, H.; Yin, Q.-F.; Luo, Z.; Yao, R.-W.; Zheng, C.-C.; Zhang, J.; Xiang, J.-F.; Yang, L.; Chen, L.-L. Unusual Processing Generates SPA LncRNAs That Sequester Multiple RNA Binding Proteins. Mol. Cell 2016, 64, 534–548. [Google Scholar] [CrossRef]
  45. Crespi, M. Long Non-Coding RNAs Reveal New Regulatory Mechanisms Controlling Gene Expression. Comptes Rendus Biol. 2023, 345, 15–39. [Google Scholar] [CrossRef]
  46. Zhao, C.; Xie, W.; Zhu, H.; Zhao, M.; Liu, W.; Wu, Z.; Wang, L.; Zhu, B.; Li, S.; Zhou, Y.; et al. LncRNAs and Their RBPs: How to Influence the Fate of Stem Cells? Stem. Cell Res. Ther. 2022, 13, 175. [Google Scholar] [CrossRef]
  47. Dodel, M.; Guiducci, G.; Dermit, M.; Krishnamurthy, S.; Alard, E.L.; Capraro, F.; Rekad, Z.; Stojic, L.; Mardakheh, F.K. TREX Reveals Proteins That Bind to Specific RNA Regions in Living Cells. Nat. Methods 2024, 21, 423–434. [Google Scholar] [CrossRef]
  48. Shaw, A.; Gullerova, M. Home and Away: The Role of Non-Coding RNA in Intracellular and Intercellular DNA Damage Response. Genes 2021, 12, 1475. [Google Scholar] [CrossRef]
  49. Mangoni, D.; Simi, A.; Lau, P.; Armaos, A.; Ansaloni, F.; Codino, A.; Damiani, D.; Floreani, L.; Di Carlo, V.; Vozzi, D.; et al. LINE-1 Regulates Cortical Development by Acting as Long Non-Coding RNAs. Nat. Commun. 2023, 14, 4974. [Google Scholar] [CrossRef]
  50. Poltronieri, P. Regulatory RNAs: Role as Scaffolds Assembling Protein Complexes and Their Epigenetic Deregulation. Explor. Target. Anti-Tumor Ther. 2024, 5, 841–876. [Google Scholar] [CrossRef]
  51. Bai, J.-Y.; Jin, B.; Ma, J.-B.; Liu, T.-J.; Yang, C.; Chong, Y.; Wang, X.; He, D.; Guo, P. HOTAIR and Androgen Receptor Synergistically Increase GLI2 Transcription to Promote Tumor Angiogenesis and Cancer Stemness in Renal Cell Carcinoma. Cancer Lett. 2021, 498, 70–79. [Google Scholar] [CrossRef]
  52. Kadota, S.; Ou, J.; Shi, Y.; Lee, J.T.; Sun, J.; Yildirim, E. Nucleoporin 153 Links Nuclear Pore Complex to Chromatin Architecture by Mediating CTCF and Cohesin Binding. Nat. Commun. 2020, 11, 2606. [Google Scholar] [CrossRef]
  53. Farhadova, S.; Ghousein, A.; Charon, F.; Surcis, C.; Gomez-Velazques, M.; Roidor, C.; Di Michele, F.; Borensztein, M.; De Sario, A.; Esnault, C.; et al. The Long Non-Coding RNA Meg3 Mediates Imprinted Gene Expression during Stem Cell Differentiation. Nucleic Acids Res. 2024, 52, 6183–6200. [Google Scholar] [CrossRef]
  54. Fierro, C.; Gatti, V.; La Banca, V.; De Domenico, S.; Scalera, S.; Corleone, G.; Fanciulli, M.; De Nicola, F.; Mauriello, A.; Montanaro, M.; et al. The Long Non-Coding RNA NEAT1 Is a ΔNp63 Target Gene Modulating Epidermal Differentiation. Nat. Commun. 2023, 14, 3795. [Google Scholar] [CrossRef]
  55. Huo, Y.; Li, Q.; Yang, L.; Li, X.; Sun, C.; Liu, Y.; Liu, H.; Pan, Z.; Li, Q.; Du, X. SDNOR, a Novel Antioxidative lncRNA, Is Essential for Maintaining the Normal State and Function of Porcine Follicular Granulosa Cells. Antioxidants 2023, 12, 799. [Google Scholar] [CrossRef]
  56. Wu, X.; Fu, M.; Ge, C.; Zhou, H.; Huang, H.; Zhong, M.; Zhang, M.; Xu, H.; Zhu, G.; Hua, W.; et al. m6A-Mediated Upregulation of lncRNA CHASERR Promotes the Progression of Glioma by Modulating the miR-6893-3p/TRIM14 Axis. Mol. Neurobiol. 2024, 61, 5418–5440. [Google Scholar] [CrossRef]
  57. Lv, W.; Jiang, W.; Luo, H.; Tong, Q.; Niu, X.; Liu, X.; Miao, Y.; Wang, J.; Guo, Y.; Li, J.; et al. Long Noncoding RNA lncMREF Promotes Myogenic Differentiation and Muscle Regeneration by Interacting with the Smarca5/P300 Complex. Nucleic Acids Res. 2022, 50, 10733–10755. [Google Scholar] [CrossRef]
  58. Chen, W.; Chen, W.; Liu, P.; Qian, S.; Tao, S.; Huang, M.; Xu, W.; Li, C.; Chen, X.; Lin, H.; et al. Role of lncRNA Has2os in Skeletal Muscle Differentiation and Regeneration. Cells 2022, 11, 3497. [Google Scholar] [CrossRef]
  59. Liu, H.; Liu, H.; Yang, Q.; Fan, Z. LncRNA SNHG1 Enhances Cartilage Regeneration by Modulating Chondrogenic Differentiation and Angiogenesis Potentials of JBMMSCs via Mitochondrial Function Regulation. Stem. Cell Res. Ther. 2024, 15, 177. [Google Scholar] [CrossRef]
  60. Lu, J.; Ma, H.; Wang, Q.; Song, Z.; Wang, J. Chemotherapy-Mediated lncRNA-Induced Immune Cell Plasticity in Cancer Immunopathogenesis. Int. Immunopharmacol. 2024, 141, 112967. [Google Scholar] [CrossRef]
  61. Zhang, X.; Pan, B.; Qiu, J.; Ke, X.; Shen, S.; Wang, X.; Tang, N. lncRNA MIAT Targets miR-411-5p/STAT3/PD-L1 Axis Mediating Hepatocellular Carcinoma Immune Response. Int. J. Exp. Pathol. 2022, 103, 102–111. [Google Scholar] [CrossRef]
  62. Du, J.; Liu, L.; Yang, F.; Leng, S.; Zhang, L.; Huang, D. Identification of Immune-Related lncRNA Regulatory Network in Pulpitis. Dis. Mark. 2022, 2022, 7222092. [Google Scholar] [CrossRef]
  63. Srinivas, T.; Mathias, C.; Oliveira-Mateos, C.; Guil, S. Roles of lncRNAs in Brain Development and Pathogenesis: Emerging Therapeutic Opportunities. Mol. Ther. 2023, 31, 1550–1561. [Google Scholar] [CrossRef]
  64. Song, P.; Chen, Y.; Liu, Z.; Liu, H.; Xiao, L.; Sun, L.; Wei, J.; He, L. LncRNA MALAT1 Aggravates Renal Tubular Injury via Activating LIN28A and the Nox4/AMPK/mTOR Signaling Axis in Diabetic Nephropathy. Front. Endocrinol. 2022, 13, 895360. [Google Scholar] [CrossRef]
  65. Yi, Y.; Zhao, Y.; Li, C.; Zhang, L.; Huang, H.; Li, Y.; Liu, L.; Hou, P.; Cui, T.; Tan, P.; et al. RAID v2.0: An Updated Resource of RNA-Associated Interactions across Organisms. Nucleic Acids Res. 2017, 45, D115–D118. [Google Scholar] [CrossRef]
  66. Frankish, A.; Diekhans, M.; Jungreis, I.; Lagarde, J.; Loveland, J.E.; Mudge, J.M.; Sisu, C.; Wright, J.C.; Armstrong, J.; Barnes, I.; et al. GENCODE 2021. Nucleic Acids Research 2021, 49, D916–D923. [Google Scholar] [CrossRef]
  67. Kang, J.; Tang, Q.; He, J.; Li, L.; Yang, N.; Yu, S.; Wang, M.; Zhang, Y.; Lin, J.; Cui, T.; et al. RNAInter v4.0: RNA Interactome Repository with Redefined Confidence Scoring System and Improved Accessibility. Nucleic Acids Res. 2022, 50, D326–D332. [Google Scholar] [CrossRef]
  68. He, N.; Li, D.; Xu, F.; Jin, J.; Li, L.; Tian, L.; Chen, B.; Li, X.; Ning, S.; Wang, L.; et al. LncPCD: A Manually Curated Database of Experimentally Supported Associations between lncRNA-Mediated Programmed Cell Death and Diseases. Database 2023, 2023, baad087. [Google Scholar] [CrossRef]
  69. 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]
  70. Zhao, Y.; Ning, J.; Teng, H.; Deng, Y.; Sheldon, M.; Shi, L.; Martinez, C.; Zhang, J.; Tian, A.; Sun, Y.; et al. Long Noncoding RNA Malat1 Protects against Osteoporosis and Bone Metastasis. Nat. Commun. 2024, 15, 2384. [Google Scholar] [CrossRef]
  71. Zhao, X.; Chen, Q.; Cai, Y.; Chen, D.; Bei, M.; Dong, H.; Xu, J. TRA2A Binds With LncRNA MALAT1 To Promote Esophageal Cancer Progression By Regulating EZH2/β-Catenin Pathway. J. Cancer 2021, 12, 4883–4890. [Google Scholar] [CrossRef]
  72. Wu, C.; Chen, W.; Yu, F.; Yuan, Y.; Chen, Y.; Hurst, D.R.; Li, Y.; Li, L.; Liu, Z. Long Noncoding RNA HITTERS Protects Oral Squamous Cell Carcinoma Cells from Endoplasmic Reticulum Stress-Induced Apoptosis via Promoting MRE11-RAD50-NBS1 Complex Formation. Adv. Sci. 2020, 7, 2002747. [Google Scholar] [CrossRef]
  73. Duan, Y.; Jia, Y.; Wang, J.; Liu, T.; Cheng, Z.; Sang, M.; Lv, W.; Qin, J.; Liu, L. Long Noncoding RNA DGCR5 Involves in Tumorigenesis of Esophageal Squamous Cell Carcinoma via SRSF1-Mediated Alternative Splicing of Mcl-1. Cell Death Dis. 2021, 12, 587. [Google Scholar] [CrossRef]
  74. Yu, C.-H.; Fang, C.-Y.; Yu, C.-C.; Hsieh, P.-L.; Liao, Y.-W.; Tsai, L.-L.; Chu, P.-M. LINC00312/YBX1 Axis Regulates Myofibroblast Activities in Oral Submucous Fibrosis. Int. J. Mol. Sci. 2020, 21, 2979. [Google Scholar] [CrossRef]
  75. Wu, Y.; Zhao, Y.; Huan, L.; Zhao, J.; Zhou, Y.; Xu, L.; Hu, Z.; Liu, Y.; Chen, Z.; Wang, L.; et al. An LTR Retrotransposon-Derived Long Noncoding RNA lncMER52A Promotes Hepatocellular Carcinoma Progression by Binding P120-Catenin. Cancer Res. 2020, 80, 976–987. [Google Scholar] [CrossRef]
  76. Lin, Y.-H.; Wu, M.-H.; Liu, Y.-C.; Lyu, P.-C.; Yeh, C.-T.; Lin, K.-H. LINC01348 Suppresses Hepatocellular Carcinoma Metastasis through Inhibition of SF3B3-Mediated EZH2 Pre-mRNA Splicing. Oncogene 2021, 40, 4675–4685. [Google Scholar] [CrossRef]
  77. Liao, M.; Yao, D.; Wu, L.; Luo, C.; Wang, Z.; Zhang, J.; Liu, B. Targeting the Warburg Effect: A Revisited Perspective from Molecular Mechanisms to Traditional and Innovative Therapeutic Strategies in Cancer. Acta Pharm. Sin. B 2024, 14, 953–1008. [Google Scholar] [CrossRef]
  78. Wang, C.; Li, Y.; Yan, S.; Wang, H.; Shao, X.; Xiao, M.; Yang, B.; Qin, G.; Kong, R.; Chen, R.; et al. Interactome Analysis Reveals That lncRNA HULC Promotes Aerobic Glycolysis through LDHA and PKM2. Nat. Commun. 2020, 11, 3162. [Google Scholar] [CrossRef]
  79. Haga, Y.; Bandyopadhyay, D.; Khatun, M.; Tran, E.; Steele, R.; Banerjee, S.; Ray, R.; Nazzal, M.; Ray, R.B. Increased Expression of Long Non-Coding RNA FIRRE Promotes Hepatocellular Carcinoma by HuR-CyclinD1 Axis Signaling. J. Biol. Chem. 2024, 300, 107247. [Google Scholar] [CrossRef]
  80. Zhou, J.; Liu, T.; Xu, H.; Wang, Y.; Yang, T.; Liu, L. LncRNA FIRRE Promotes the Proliferation and Metastasis of Hepatocellular Carcinoma by Regulating the Expression of PXN through Interacting with MBNL3. Biochem. Biophys. Res. Commun. 2022, 625, 188–195. [Google Scholar] [CrossRef]
  81. Malakoti, F.; Targhazeh, N.; Karimzadeh, H.; Mohammadi, E.; Asadi, M.; Asemi, Z.; Alemi, F. Multiple Function of lncRNA MALAT1 in Cancer Occurrence and Progression. Chem. Biol. Drug Des. 2023, 101, 1113–1137. [Google Scholar] [CrossRef]
  82. Jen, J.; Tang, Y.-A.; Lu, Y.-H.; Lin, C.-C.; Lai, W.-W.; Wang, Y.-C. Oct4 Transcriptionally Regulates the Expression of Long Non-Coding RNAs NEAT1 and MALAT1 to Promote Lung Cancer Progression. Mol. Cancer 2017, 16, 104. [Google Scholar] [CrossRef]
  83. Huang, J.; Lin, C.; Dong, H.; Piao, Z.; Jin, C.; Han, H.; Jin, D. Targeting MALAT1 Induces DNA Damage and Sensitize Non-Small Cell Lung Cancer Cells to Cisplatin by Repressing BRCA1. Cancer Chemother. Pharmacol. 2020, 86, 663–672. [Google Scholar] [CrossRef]
  84. Mao, G.; Mu, Z.; Wu, D.A. Exosomal lncRNA FOXD3-AS1 Upregulates ELAVL1 Expression and Activates PI3K/Akt Pathway to Enhance Lung Cancer Cell Proliferation, Invasion, and 5-Fluorouracil Resistance. Acta Biochim. Biophys. Sin. 2021, 53, 1484–1494. [Google Scholar] [CrossRef]
  85. Qian, X.; Yang, J.; Qiu, Q.; Li, X.; Jiang, C.; Li, J.; Dong, L.; Ying, K.; Lu, B.; Chen, E.; et al. LCAT3, a Novel m6A-Regulated Long Non-Coding RNA, Plays an Oncogenic Role in Lung Cancer via Binding with FUBP1 to Activate c-MYC. J. Hematol. Oncol. 2021, 14, 112. [Google Scholar] [CrossRef]
  86. Hussain, S.A.; Venkatesh, T. YBX1/lncRNA SBF2-AS1 Interaction Regulates Proliferation and Tamoxifen Sensitivity via PI3K/AKT/MTOR Signaling in Breast Cancer Cells. Mol. Biol. Rep. 2023, 50, 3413–3428. [Google Scholar] [CrossRef]
  87. Sun, X.; Wang, R.; Tan, M.; Tian, X.; Meng, J. LncRNA LINC00680 Promotes Lung Adenocarcinoma Growth via Binding to GATA6 and Canceling GATA6-Mediated Suppression of SOX12 Expression. Exp. Cell Res. 2021, 405, 112653. [Google Scholar] [CrossRef]
  88. Cong, Z.; Diao, Y.; Li, X.; Jiang, Z.; Xu, Y.; Zhou, H.; Qiang, Y.; Wu, H.; Shen, Y. Long Non-Coding RNA Linc00665 Interacts with YB-1 and Promotes Angiogenesis in Lung Adenocarcinoma. Biochem. Biophys. Res. Commun. 2020, 527, 545–552. [Google Scholar] [CrossRef]
  89. Wu, A.; Tang, J.; Guo, Z.; Dai, Y.; Nie, J.; Hu, W.; Liu, N.; Ye, C.; Li, S.; Pei, H.; et al. Long Non-Coding RNA CRYBG3 Promotes Lung Cancer Metastasis via Activating the eEF1A1/MDM2/MTBP Axis. Int. J. Mol. Sci. 2021, 22, 3211. [Google Scholar] [CrossRef]
  90. Bao, T.; Liu, X.; Hu, J.; Ma, M.; Li, J.; Cao, L.; Yu, B.; Cheng, H.; Zhao, S.; Tian, Z. Recruitment of PVT1 Enhances YTHDC1-Mediated m6A Modification of IL-33 in Hyperoxia-Induced Lung Injury During Bronchopulmonary Dysplasia. Inflammation 2023, 47, 469–482. [Google Scholar] [CrossRef]
  91. Sun, J.; Jin, T.; Niu, Z.; Guo, J.; Guo, Y.; Yang, R.; Wang, Q.; Gao, H.; Zhang, Y.; Li, T.; et al. LncRNA DACH1 Protects against Pulmonary Fibrosis by Binding to SRSF1 to Suppress CTNNB1 Accumulation. Acta Pharm. Sin. B 2022, 12, 3602–3617. [Google Scholar] [CrossRef]
  92. Zhou, L.; Xu, D.; Sha, W.; Shen, L.; Lu, G. Long Non-Coding RNA MALAT1 Interacts with Transcription Factor Foxo1 to Regulate SIRT1 Transcription in High Glucose-Induced HK-2 Cells Injury. Biochem. Biophys. Res. Commun. 2018, 503, 849–855. [Google Scholar] [CrossRef]
  93. Liu, T.; Wang, H.; Fu, Z.; Wang, Z.; Wang, J.; Gan, X.; Wang, A.; Wang, L. Methyltransferase-like 14 Suppresses Growth and Metastasis of Renal Cell Carcinoma by Decreasing Long Noncoding RNA NEAT1. Cancer Sci. 2022, 113, 446–458. [Google Scholar] [CrossRef]
  94. Xie, X.; Lin, J.; Fan, X.; Zhong, Y.; Chen, Y.; Liu, K.; Ren, Y.; Chen, X.; Lai, D.; Li, X.; et al. LncRNA CDKN2B-AS1 Stabilized by IGF2BP3 Drives the Malignancy of Renal Clear Cell Carcinoma through Epigenetically Activating NUF2 Transcription. Cell Death Dis. 2021, 12, 201. [Google Scholar] [CrossRef]
  95. Hjazi, A.; Ghaffar, E.; Asghar, W.; Alauldeen Khalaf, H.; Ikram Ullah, M.; Mireya Romero-Parra, R.; Hussien, B.M.; Abdulally Abdulhussien Alazbjee, A.; Singh Bisht, Y.; Fakri Mustafa, Y.; et al. CDKN2B-AS1 as a Novel Therapeutic Target in Cancer: Mechanism and Clinical Perspective. Biochem. Pharmacol. 2023, 213, 115627. [Google Scholar] [CrossRef]
  96. Ageeli Hakami, M. Diabetes and Diabetic Associative Diseases: An Overview of Epigenetic Regulations of TUG1. Saudi J. Biol. Sci. 2024, 31, 103976. [Google Scholar] [CrossRef]
  97. Wang, S.; Yi, P.; Wang, N.; Song, M.; Li, W.; Zheng, Y. LncRNA TUG1/miR-29c-3p/SIRT1 Axis Regulates Endoplasmic Reticulum Stress-Mediated Renal Epithelial Cells Injury in Diabetic Nephropathy Model in Vitro. PLoS ONE 2021, 16, e0252761. [Google Scholar] [CrossRef]
  98. Song, X.; Gao, F.; Li, H.; Qin, W.; Chai, C.; Shi, G.; Yang, H. Long Noncoding RNA THRIL Promotes Foam Cell Formation and Inflammation in Macrophages. Cell Biol. Int. 2023, 47, 156–166. [Google Scholar] [CrossRef]
  99. Xiao, J.; Lu, Y.; Yang, X. THRIL Mediates Endothelial Progenitor Cells Autophagy via AKT Pathway and FUS. Mol. Med. 2020, 26, 86. [Google Scholar] [CrossRef]
  100. Zhang, Y.-Y.; Tan, R.-Z.; Yu, Y.; Niu, Y.-Y.; Yu, C. LncRNA GAS5 Protects against TGF-β-Induced Renal Fibrosis via the Smad3/miRNA-142-5p Axis. Am. J. Physiol. Ren. Physiol. 2021, 321, F517–F526. [Google Scholar] [CrossRef]
  101. Tang, R.; Wang, Y.-C.; Mei, X.; Shi, N.; Sun, C.; Ran, R.; Zhang, G.; Li, W.; Staveley-O’Carroll, K.F.; Li, G.; et al. LncRNA GAS5 Attenuates Fibroblast Activation through Inhibiting Smad3 Signaling. Am. J. Physiol. Cell Physiol. 2020, 319, C105–C115. [Google Scholar] [CrossRef]
  102. Wang, Y.; Chen, D.; Xie, H.; Zhou, S.; Jia, M.; He, X.; Guo, F.; Lai, Y.; Tang, X.X. LncRNA GAS5 Suppresses TGF-Β1-Induced Transformation of Pulmonary Pericytes into Myofibroblasts by Recruiting KDM5B and Promoting H3K4me2/3 Demethylation of the PDGFRα/β Promoter. Mol. Med. 2023, 29, 32. [Google Scholar] [CrossRef]
  103. Grossi, E.; Raimondi, I.; Goñi, E.; González, J.; Marchese, F.P.; Chapaprieta, V.; Martín-Subero, J.I.; Guo, S.; Huarte, M. A lncRNA-SWI/SNF Complex Crosstalk Controls Transcriptional Activation at Specific Promoter Regions. Nat. Commun. 2020, 11, 936. [Google Scholar] [CrossRef]
  104. Jiang, M.; Zhang, S.; Yang, Z.; Lin, H.; Zhu, J.; Liu, L.; Wang, W.; Liu, S.; Liu, W.; Ma, Y.; et al. Self-Recognition of an Inducible Host lncRNA by RIG-I Feedback Restricts Innate Immune Response. Cell 2018, 173, 906–919.e13. [Google Scholar] [CrossRef]
  105. Lin, H.; Jiang, M.; Liu, L.; Yang, Z.; Ma, Z.; Liu, S.; Ma, Y.; Zhang, L.; Cao, X. The Long Noncoding RNA Lnczc3h7a Promotes a TRIM25-Mediated RIG-I Antiviral Innate Immune Response. Nat. Immunol. 2019, 20, 812–823. [Google Scholar] [CrossRef]
  106. Zou, S.; Gou, X.; Wen, K. Advances in the Role of Long Non-coding RNAs and RNA-binding Proteins in Regulating DNA Damage Repair in Cancer Cells. Int. J. Mol. Med. 2023, 52, 93. [Google Scholar] [CrossRef]
  107. Wang, D.; Zhou, Z.; Wu, E.; Ouyang, C.; Wei, G.; Wang, Y.; He, D.; Cui, Y.; Zhang, D.; Chen, X.; et al. LRIK Interacts with the Ku70–Ku80 Heterodimer Enhancing the Efficiency of NHEJ Repair. Cell Death Differ. 2020, 27, 3337–3353. [Google Scholar] [CrossRef]
  108. Deng, B.; Xu, W.; Wang, Z.; Liu, C.; Lin, P.; Li, B.; Huang, Q.; Yang, J.; Zhou, H.; Qu, L. An LTR Retrotransposon-Derived lncRNA Interacts with RNF169 to Promote Homologous Recombination. EMBO Rep. 2019, 20, e47650. [Google Scholar] [CrossRef]
  109. Typas, D.; Mailand, N. An Unorthodox Partnership in DNA Repair Pathway Choice. EMBO Rep. 2019, 20, e49105. [Google Scholar] [CrossRef]
  110. Wang, J.; Chen, H.; Deng, Q.; Chen, Y.; Wang, Z.; Yan, Z.; Wang, Y.; Tang, H.; Liang, H.; Jiang, Y. High Expression of RNF169 Is Associated with Poor Prognosis in Pancreatic Adenocarcinoma by Regulating Tumour Immune Infiltration. Front. Genet. 2023, 13, 1022626. [Google Scholar] [CrossRef]
  111. Shuai, Y.; Ma, Z.; Liu, W.; Yu, T.; Yan, C.; Jiang, H.; Tian, S.; Xu, T.; Shu, Y. TEAD4 Modulated LncRNA MNX1-AS1 Contributes to Gastric Cancer Progression Partly through Suppressing BTG2 and Activating BCL2. Mol. Cancer 2020, 19, 6. [Google Scholar] [CrossRef]
  112. Han, T.; Jing, X.; Bao, J.; Zhao, L.; Zhang, A.; Miao, R.; Guo, H.; Zhou, B.; Zhang, S.; Sun, J.; et al. H. Pylori Infection Alters Repair of DNA Double-Strand Breaks via SNHG17. J. Clin. Investig. 2020, 130, 3901–3918. [Google Scholar] [CrossRef]
  113. Pirlog, R.; Drula, R.; Nutu, A.; Calin, G.A.; Berindan-Neagoe, I. The Roles of the Colon Cancer Associated Transcript 2 (CCAT2) Long Non-Coding RNA in Cancer: A Comprehensive Characterization of the Tumorigenic and Molecular Functions. Int. J. Mol. Sci. 2021, 22, 12491. [Google Scholar] [CrossRef]
  114. Chen, B.; Dragomir, M.P.; Fabris, L.; Bayraktar, R.; Knutsen, E.; Liu, X.; Tang, C.; Li, Y.; Shimura, T.; Ivkovic, T.C.; et al. The Long Noncoding RNA CCAT2 Induces Chromosomal Instability Through BOP1-AURKB Signaling. Gastroenterology 2020, 159, 2146–2162.e33. [Google Scholar] [CrossRef]
  115. Zhao, C.; Ling, X.; Xia, Y.; Yan, B.; Guan, Q. The m6A Methyltransferase METTL3 Controls Epithelial-Mesenchymal Transition, Migration and Invasion of Breast Cancer through the MALAT1/miR-26b/HMGA2 Axis. Cancer Cell Int. 2021, 21, 441. [Google Scholar] [CrossRef]
  116. Lu, D.; Di, S.; Zhuo, S.; Zhou, L.; Bai, R.; Ma, T.; Zou, Z.; Chen, C.; Sun, M.; Tang, J.; et al. The Long Noncoding RNA TINCR Promotes Breast Cancer Cell Proliferation and Migration by Regulating OAS1. Cell Death Discov. 2021, 7, 41. [Google Scholar] [CrossRef]
  117. Zhang, Y.; Huang, Y.-X.; Wang, D.-L.; Yang, B.; Yan, H.-Y.; Lin, L.-H.; Li, Y.; Chen, J.; Xie, L.-M.; Huang, Y.-S.; et al. LncRNA DSCAM-AS1 Interacts with YBX1 to Promote Cancer Progression by Forming a Positive Feedback Loop That Activates FOXA1 Transcription Network. Theranostics 2020, 10, 10823–10837. [Google Scholar] [CrossRef]
  118. Abo Elwafa, R.; Abd Elrahman, A.; Ghallab, O. Long Intergenic Non-Coding RNA-P21 Is Associated with Poor Prognosis in Chronic Lymphocytic Leukemia. Clin. Transl. Oncol. 2021, 23, 92–99. [Google Scholar] [CrossRef]
  119. Amirinejad, R.; Rezaei, M.; Shirvani-Farsani, Z. An Update on Long Intergenic Noncoding RNA P21: A Regulatory Molecule with Various Significant Functions in Cancer. Cell Biosci. 2020, 10, 82. [Google Scholar] [CrossRef]
  120. Huang, Y.; Yi, Q.; Feng, J.; Xie, W.; Sun, W.; Sun, W. The Role of lincRNA-P21 in Regulating the Biology of Cancer Cells. Hum. Cell 2022, 35, 1640–1649. [Google Scholar] [CrossRef]
  121. Xiong, J.; Lian, W.; Zhao, R.; Gao, K. METTL3/MALAT1/ELAVL1 Axis Promotes Tumor Growth in Ovarian Cancer. OncoTargets Ther. 2024, 17, 85–97. [Google Scholar] [CrossRef]
  122. Wang, Z.; Wang, X.; Zhang, T.; Su, L.; Liu, B.; Zhu, Z.; Li, C. LncRNA MALAT1 Promotes Gastric Cancer Progression via Inhibiting Autophagic Flux and Inducing Fibroblast Activation. Cell Death Dis. 2021, 12, 368. [Google Scholar] [CrossRef]
  123. Farooqi, A.; Zahid, R.; Naureen, H.; Attar, R.; Gazouli, M.; Berardi, R.; Szelachowska, J.; Matkowski, R.; Pawlak, E. Regulation of ROCK1/2 by Long Non-coding RNAs and Circular RNAs in Different Cancer Types (Review). Oncol. Lett. 2022, 23, 159. [Google Scholar] [CrossRef]
  124. Dai, R.; Zhou, Y.; Chen, Z.; Zou, Z.; Pan, Z.; Liu, P.; Gao, X. Lnc-MUC20-9 Binds to ROCK1 and Functions as a Tumor Suppressor in Bladder Cancer. J. Cell Biochem. 2020, 121, 4214–4225. [Google Scholar] [CrossRef]
  125. Mehmandar-Oskuie, A.; Jahankhani, K.; Rostamlou, A.; Arabi, S.; Sadat Razavi, Z.; Mardi, A. Molecular Landscape of LncRNAs in Bladder Cancer: From Drug Resistance to Novel LncRNA-Based Therapeutic Strategies. Biomed. Pharmacother. 2023, 165, 115242. [Google Scholar] [CrossRef]
  126. Chen, C.; Luo, Y.; He, W.; Zhao, Y.; Kong, Y.; Liu, H.; Zhong, G.; Li, Y.; Li, J.; Huang, J.; et al. Exosomal Long Noncoding RNA LNMAT2 Promotes Lymphatic Metastasis in Bladder Cancer. J. Clin. Investig. 2019, 130, 404–421. [Google Scholar] [CrossRef]
  127. Wu, R.; Li, L.; Bai, Y.; Yu, B.; Xie, C.; Wu, H.; Zhang, Y.; Huang, L.; Yan, Y.; Li, X.; et al. The Long Noncoding RNA LUCAT1 Promotes Colorectal Cancer Cell Proliferation by Antagonizing Nucleolin to Regulate MYC Expression. Cell Death Dis. 2020, 11, 908. [Google Scholar] [CrossRef]
  128. Zhou, Q.; Hou, Z.; Zuo, S.; Zhou, X.; Feng, Y.; Sun, Y.; Yuan, X. LUCAT1 Promotes Colorectal Cancer Tumorigenesis by Targeting the Ribosomal Protein L40-MDM2-P53 Pathway through Binding with UBA52. Cancer Sci. 2019, 110, 1194–1207. [Google Scholar] [CrossRef]
  129. Deng, X.; Kong, F.; Li, S.; Jiang, H.; Dong, L.; Xu, X.; Zhang, X.; Yuan, H.; Xu, Y.; Chu, Y.; et al. A KLF4/PiHL/EZH2/HMGA2 Regulatory Axis and Its Function in Promoting Oxaliplatin-Resistance of Colorectal Cancer. Cell Death Dis. 2021, 12, 485. [Google Scholar] [CrossRef]
  130. Deng, X.; Li, S.; Kong, F.; Ruan, H.; Xu, X.; Zhang, X.; Wu, Z.; Zhang, L.; Xu, Y.; Yuan, H.; et al. Long Noncoding RNA PiHL Regulates P53 Protein Stability through GRWD1/RPL11/MDM2 Axis in Colorectal Cancer. Theranostics 2020, 10, 265–280. [Google Scholar] [CrossRef]
  131. Yue, M.; Liu, T.; Yan, G.; Luo, X.; Wang, L. LINC01605, Regulated by the EP300-SMYD2 Complex, Potentiates the Binding between METTL3 and SPTBN2 in Colorectal Cancer. Cancer Cell Int. 2021, 21, 504. [Google Scholar] [CrossRef]
  132. Jiang, T.; Qi, J.; Xue, Z.; Liu, B.; Liu, J.; Hu, Q.; Li, Y.; Ren, J.; Song, H.; Xu, Y.; et al. The m6A Modification Mediated-lncRNA POU6F2-AS1 Reprograms Fatty Acid Metabolism and Facilitates the Growth of Colorectal Cancer via Upregulation of FASN. Mol. Cancer 2024, 23, 55. [Google Scholar] [CrossRef]
  133. Zhou, Y.; Shao, Y.; Hu, W.; Zhang, J.; Shi, Y.; Kong, X.; Jiang, J. A Novel Long Noncoding RNA SP100-AS1 Induces Radioresistance of Colorectal Cancer via Sponging miR-622 and Stabilizing ATG3. Cell Death Differ. 2023, 30, 111–124. [Google Scholar] [CrossRef]
  134. Chen, Q.; Zhou, L.; Ma, D.; Hou, J.; Lin, Y.; Wu, J.; Tao, M. LncRNA GAS6-AS1 Facilitates Tumorigenesis and Metastasis of Colorectal Cancer by Regulating TRIM14 through miR-370-3p/miR-1296-5p and FUS. J. Transl. Med. 2022, 20, 356. [Google Scholar] [CrossRef]
  135. Chen, X.; Song, J.; Wang, X.; Sun, D.; Liu, Y.; Jiang, Y. LncRNA LINC00460: Function and Mechanism in Human Cancer. Thorac. Cancer 2022, 13, 3–14. [Google Scholar] [CrossRef]
  136. Hou, P.; Meng, S.; Li, M.; Lin, T.; Chu, S.; Li, Z.; Zheng, J.; Gu, Y.; Bai, J. LINC00460/DHX9/IGF2BP2 Complex Promotes Colorectal Cancer Proliferation and Metastasis by Mediating HMGA1 mRNA Stability Depending on m6A Modification. J. Exp. Clin. Cancer Res. 2021, 40, 52. [Google Scholar] [CrossRef]
  137. Zhao, K.; Wang, X.; Xue, X.; Li, L.; Hu, Y. A Long Noncoding RNA Sensitizes Genotoxic Treatment by Attenuating ATM Activation and Homologous Recombination Repair in Cancers. PLoS Biol. 2020, 18, e3000666. [Google Scholar] [CrossRef]
  138. Cai, J.; Wang, R.; Chen, Y.; Zhang, C.; Fu, L.; Fan, C. LncRNA FIRRE Regulated Endometrial Cancer Radiotherapy Sensitivity via the miR-199b-5p/SIRT1/BECN1 Axis-Mediated Autophagy. Genomics 2024, 116, 110750. [Google Scholar] [CrossRef]
  139. Wang, Y.; Li, Z.; Xu, S.; Li, W.; Chen, M.; Jiang, M.; Fan, X. LncRNA FIRRE Functions as a Tumor Promoter by Interaction with PTBP1 to Stabilize BECN1 mRNA and Facilitate Autophagy. Cell Death Dis. 2022, 13, 98. [Google Scholar] [CrossRef]
  140. Liu, Q.; Dai, G.; Wu, Y.; Wang, X.; Song, M.; Li, X.; Wu, Z.; Xia, R. LncRNA FIRRE Stimulates PTBP1-induced Smurf2 Decay, Stabilizes B-cell Receptor, and Promotes the Development of Diffuse Large B-cell Lymphoma. Hematol. Oncol. 2022, 40, 554–566. [Google Scholar] [CrossRef]
  141. Gu, N.; Wang, Y.; Li, L.; Sui, X.; Liu, Z. The Mechanism of lncRNA MALAT1 Targeting the miR-124-3p/IGF2BP1 Axis to Regulate Osteogenic Differentiation of Periodontal Ligament Stem Cells. Clin. Oral Invest. 2024, 28, 219. [Google Scholar] [CrossRef]
  142. Ding, A.; Li, C.-H.; Yu, C.-Y.; Zhou, H.-T.; Zhang, Z.-H. Long Non-Coding RNA MALAT1 Enhances Angiogenesis during Bone Regeneration by Regulating the miR-494/SP1 Axis. Lab. Investig. 2021, 101, 1458–1466. [Google Scholar] [CrossRef]
  143. Ghafouri-Fard, S.; Azimi, T.; Taheri, M. Cervical Carcinoma High Expressed 1 (CCHE1): An Oncogenic lncRNA in Diverse Neoplasms. Biomed. Pharmacother. 2021, 142, 112003. [Google Scholar] [CrossRef]
  144. Zhang, Z.; Yu, T.; Geng, W. Long Non-Coding RNA CCHE1 Participates in Postoperative Distant Recurrence but Not Local Recurrence of Osteosarcoma Possibly by Interacting with ROCK1. BMC Musculoskelet. Disord. 2020, 21, 462. [Google Scholar] [CrossRef]
  145. Yao, X.-Y.; Liu, J.-F.; Luo, Y.; Xu, X.-Z.; Bu, J. LncRNA HOTTIP Facilitates Cell Proliferation, Invasion, and Migration in Osteosarcoma by Interaction with PTBP1 to Promote KHSRP Level. Cell Cycle 2021, 20, 283–297. [Google Scholar] [CrossRef]
  146. Fu, C.; Xin, J.; Zhang, W.; Lai, J.; Huang, Z. LINC00992 Exerts Oncogenic Activities in Prostate Cancer via Regulation of SOX4. Exp. Cell Res. 2021, 408, 112855. [Google Scholar] [CrossRef]
  147. Tan, X.; Chen, W.; Lv, D.; Yang, T.; Wu, K.; Zou, L.; Luo, J.; Zhou, X.; Liu, G.; Shu, F.; et al. LncRNA SNHG1 and RNA Binding Protein hnRNPL Form a Complex and Coregulate CDH1 to Boost the Growth and Metastasis of Prostate Cancer. Cell Death Dis. 2021, 12, 138. [Google Scholar] [CrossRef]
  148. Muller, C.S.M.; Giner, I.S.; Zambalde, É.P.; Carvalho, T.M.; Ribeiro, E.M.D.S.F.; Carvalho De Oliveira, J.; Mathias, C.; Gradia, D.F. The Potential of NORAD–PUMILIO–RALGAPB Regulatory Axis as a Biomarker in Breast Cancer. ncRNA 2022, 8, 76. [Google Scholar] [CrossRef]
  149. Elguindy, M.M.; Mendell, J.T. NORAD-Induced Pumilio Phase Separation Is Required for Genome Stability. Nature 2021, 595, 303–308. [Google Scholar] [CrossRef]
  150. Zhao, D.; Wang, C.; Yan, S.; Chen, R. Advances in the Identification of Long Non-Coding RNA Binding Proteins. Anal. Biochem. 2022, 639, 114520. [Google Scholar] [CrossRef]
  151. Lambert, N.J.; Robertson, A.D.; Burge, C.B. RNA Bind-n-Seq: Measuring the Binding Affinity Landscape of RNA-Binding Proteins. In Methods in Enzymology; Elsevier: Amsterdam, The Netherlands, 2015; Volume 558, pp. 465–493. ISBN 978-0-12-801934-4. [Google Scholar]
  152. Hanelt, T.N.; Treiber, N.; Treiber, T.; Lehmann, G.; Eichner, N.; Rothmeier, T.; Schmid, G.; Reichelt, R.; Zambelli, F.; Pavesi, G.; et al. Endo-Bind-n-Seq: Identifying RNA Motifs of RNA Binding Proteins Isolated from Endogenous Sources. Life Sci. Alliance 2025, 8, e202402782. [Google Scholar] [CrossRef]
  153. Ye, X.; Jankowsky, E. High Throughput Approaches to Study RNA-Protein Interactions in Vitro. Methods 2020, 178, 3–10. [Google Scholar] [CrossRef]
  154. Baldini, L.; Labialle, S. Using Native RIP, UV-CLIP or fCLIP to Address Protein–RNA Interactions In Vivo. In Small Non-Coding RNAs; Rederstorff, M., Ed.; Methods in Molecular Biology; Springer: New York, NY, USA, 2021; Volume 2300, pp. 89–98. ISBN 978-1-07-161385-6. [Google Scholar]
  155. Martindale, J.; Gorospe, M.; Idda, M. Ribonucleoprotein Immunoprecipitation (RIP) Analysis. BIO-Protocol 2020, 10, e3488. [Google Scholar] [CrossRef]
  156. Kapral, T.H.; Farnhammer, F.; Zhao, W.; Lu, Z.J.; Zagrovic, B. Widespread Autogenous mRNA–Protein Interactions Detected by CLIP-Seq. Nucleic Acids Res. 2022, 50, 9984–9999. [Google Scholar] [CrossRef]
  157. Chen, X.; Castro, S.A.; Liu, Q.; Hu, W.; Zhang, S. Practical Considerations on Performing and Analyzing CLIP-Seq Experiments to Identify Transcriptomic-Wide RNA-Protein Interactions. Methods 2019, 155, 49–57. [Google Scholar] [CrossRef]
  158. Zheng, M.; Lin, Y.; Wang, W.; Zhao, Y.; Bao, X. Application of Nucleoside or Nucleotide Analogues in RNA Dynamics and RNA-Binding Protein Analysis. WIREs RNA 2022, 13, e1722. [Google Scholar] [CrossRef]
  159. Kiefer, L.; Schofield, J.A.; Simon, M.D. Expanding the Nucleoside Recoding Toolkit: Revealing RNA Population Dynamics with 6-Thioguanosine. J. Am. Chem. Soc. 2018, 140, 14567–14570. [Google Scholar] [CrossRef]
  160. Van Nostrand, E.L.; Pratt, G.A.; Shishkin, A.A.; Gelboin-Burkhart, C.; Fang, M.Y.; Sundararaman, B.; Blue, S.M.; Nguyen, T.B.; Surka, C.; Elkins, K.; et al. Robust Transcriptome-Wide Discovery of RNA-Binding Protein Binding Sites with Enhanced CLIP (eCLIP). Nat. Methods 2016, 13, 508–514. [Google Scholar] [CrossRef]
  161. Kim, B.; Kim, V.N. fCLIP-Seq for Transcriptomic Footprinting of dsRNA-Binding Proteins: Lessons from DROSHA. Methods 2019, 152, 3–11. [Google Scholar] [CrossRef]
  162. Sahadevan, S.; Sekaran, T.; Ashaf, N.; Fritz, M.; Hentze, M.W.; Huber, W.; Schwarzl, T. Htseq-Clip: A Toolset for the Preprocessing of eCLIP/iCLIP Datasets. Bioinformatics 2023, 39, btac747. [Google Scholar] [CrossRef]
  163. Castello, A.; Fischer, B.; Frese, C.K.; Horos, R.; Alleaume, A.-M.; Foehr, S.; Curk, T.; Krijgsveld, J.; Hentze, M.W. Comprehensive Identification of RNA-Binding Domains in Human Cells. Mol. Cell 2016, 63, 696–710. [Google Scholar] [CrossRef]
  164. Shan, M.; Gregory, B.D. Using RNA Affinity Purification Followed by Mass Spectrometry to Identify RNA-Binding Proteins (RBPs). In RNA Tagging; Heinlein, M., Ed.; Methods in Molecular Biology; Springer: New York, NY, USA, 2020; Volume 2166, pp. 241–253. ISBN 978-1-07-160711-4. [Google Scholar]
  165. Fernandez-Chamorro, J.; Francisco-Velilla, R.; Embarc-Buh, A.; Martinez-Salas, E. Identification of RNA-Binding Proteins Associated to RNA Structural Elements. In RNA Scaffolds; Ponchon, L., Ed.; Methods in Molecular Biology; Springer: New York, NY, USA, 2021; Volume 2323, pp. 109–119. ISBN 978-1-07-161498-3. [Google Scholar]
  166. Azizi, H.; Papadopoulou, B. In Vivo Tethering System to Isolate RNA-Binding Proteins Regulating mRNA Decay in Leishmania. In Trypanosomatids; Michels, P.A.M., Ginger, M.L., Zilberstein, D., Eds.; Methods in Molecular Biology; Springer: New York, NY, USA, 2020; Volume 2116, pp. 325–338. ISBN 978-1-07-160293-5. [Google Scholar]
  167. Janvier, A.; Hayek, H.; Alghoul, F.; Gross, L.; Allmang, C.; Martin, F.; Eriani, G. Purification of In Vivo or In Vitro-Assembled RNA-Protein Complexes by RNA Centric Methods. In Advanced Technologies for Protein Complex Production and Characterization; Vega, M.C., Fernández, F.J., Eds.; Advances in Experimental Medicine and Biology; Springer Nature Switzerland: Cham, Switzerland, 2024; Volume 3234, pp. 17–29. ISBN 978-3-031-52192-8. [Google Scholar]
  168. Dvir, S.; Argoetti, A.; Lesnik, C.; Roytblat, M.; Shriki, K.; Amit, M.; Hashimshony, T.; Mandel-Gutfreund, Y. Uncovering the RNA-Binding Protein Landscape in the Pluripotency Network of Human Embryonic Stem Cells. Cell Rep. 2021, 35, 109198. [Google Scholar] [CrossRef]
  169. Zheng, J.; Dou, R.; Zhang, X.; Zhong, B.; Fang, C.; Xu, Q.; Di, Z.; Huang, S.; Lin, Z.; Song, J.; et al. LINC00543 Promotes Colorectal Cancer Metastasis by Driving EMT and Inducing the M2 Polarization of Tumor Associated Macrophages. J. Transl. Med. 2023, 21, 153. [Google Scholar] [CrossRef]
  170. Alfeghaly, C.; Behm-Ansmant, I.; Maenner, S. Study of Genome-Wide Occupancy of Long Non-Coding RNAs Using Chromatin Isolation by RNA Purification (ChIRP). In Small Non-Coding RNAs; Rederstorff, M., Ed.; Methods in Molecular Biology; Springer: New York, NY, USA, 2021; Volume 2300, pp. 107–117. ISBN 978-1-07-161385-6. [Google Scholar]
  171. Kim, J.; Ma, L. Capturing Endogenous Long Noncoding RNAs and Their Binding Proteins Using Chromatin Isolation by RNA Purification. In Long Non-Coding RNAs; Zhang, L., Hu, X., Eds.; Methods in Molecular Biology; Springer: New York, NY, USA, 2021; Volume 2372, pp. 85–92. ISBN 978-1-07-161696-3. [Google Scholar]
  172. Xing, S.; Wang, J.; Wu, R.; Hefti, M.M.; Crary, J.F.; Lu, Y. Identification of HnRNPC as a Novel Tau Exon 10 Splicing Factor Using RNA Antisense Purification Mass Spectrometry. RNA Biol. 2022, 19, 104–116. [Google Scholar] [CrossRef]
  173. Pant, P.; Kumarswamy, R. Multiple Oligo Assisted RNA Pulldown via Hybridization Followed by Mass Spectrometry (MORPH-MS) for Exploring the RNA-Protein Interactions. RNA Biol. 2024, 21, 56–64. [Google Scholar] [CrossRef]
  174. Weissinger, R.; Heinold, L.; Akram, S.; Jansen, R.-P.; Hermesh, O. RNA Proximity Labeling: A New Detection Tool for RNA-Protein Interactions. Molecules 2021, 26, 2270. [Google Scholar] [CrossRef]
  175. Yi, W.; Li, J.; Zhu, X.; Wang, X.; Fan, L.; Sun, W.; Liao, L.; Zhang, J.; Li, X.; Ye, J.; et al. CRISPR-Assisted Detection of RNA-Protein Interactions in Living Cells. Nat. Methods 2020, 17, 685–688. [Google Scholar] [CrossRef]
  176. Teng, Z.; Zhang, Y.; Dai, Q.; Wu, C.; Li, D. Constructing Discriminative Feature Space for LncRNA–Protein Interaction Based on Deep Autoencoder and Marginal Fisher Analysis. Comput. Biol. Med. 2023, 157, 106711. [Google Scholar] [CrossRef]
  177. Shaw, D.; Chen, H.; Xie, M.; Jiang, T. DeepLPI: A Multimodal Deep Learning Method for Predicting the Interactions between lncRNAs and Protein Isoforms. BMC Bioinform. 2021, 22, 24. [Google Scholar] [CrossRef]
  178. Chen, Y.; Fu, X.; Li, Z.; Peng, L.; Zhuo, L. Prediction of lncRNA–Protein Interactions via the Multiple Information Integration. Front. Bioeng. Biotechnol. 2021, 9, 647113. [Google Scholar] [CrossRef]
  179. Zhao, J.; Sun, J.; Shuai, S.C.; Zhao, Q.; Shuai, J. Predicting Potential Interactions between lncRNAs and Proteins via Combined Graph Auto-Encoder Methods. Brief. Bioinform. 2023, 24, bbac527. [Google Scholar] [CrossRef]
  180. Zhao, X.; Zhang, Y.; Du, X. DFpin: Deep Learning–Based Protein-Binding Site Prediction with Feature-Based Non-Redundancy from RNA Level. Comput. Biol. Med. 2022, 142, 105216. [Google Scholar] [CrossRef]
  181. Wang, Y.; Pan, Z.; Mou, M.; Xia, W.; Zhang, H.; Zhang, H.; Liu, J.; Zheng, L.; Luo, Y.; Zheng, H.; et al. A Task-Specific Encoding Algorithm for RNAs and RNA-Associated Interactions Based on Convolutional Autoencoder. Nucleic Acids Res. 2023, 51, e110. [Google Scholar] [CrossRef]
  182. Wei, J.; Chen, S.; Zong, L.; Gao, X.; Li, Y. Protein–RNA Interaction Prediction with Deep Learning: Structure Matters. Brief. Bioinform. 2022, 23, bbab540. [Google Scholar] [CrossRef]
  183. Agarwal, A.; Kant, S.; Bahadur, R.P. Efficient Mapping of RNA-binding Residues in RNA-binding Proteins Using Local Sequence Features of Binding Site Residues in protein-RNA Complexes. Proteins 2023, 91, 1361–1379. [Google Scholar] [CrossRef]
  184. Li, P.; Liu, Z.-P. PST-PRNA: Prediction of RNA-Binding Sites Using Protein Surface Topography and Deep Learning. Bioinformatics 2022, 38, 2162–2168. [Google Scholar] [CrossRef]
  185. Wang, Y.; Chen, Z.; Pan, Z.; Huang, S.; Liu, J.; Xia, W.; Zhang, H.; Zheng, M.; Li, H.; Hou, T.; et al. RNAincoder: A Deep Learning-Based Encoder for RNA and RNA-Associated Interaction. Nucleic Acids Res. 2023, 51, W509–W519. [Google Scholar] [CrossRef]
  186. Zhang, H.; Lu, M.; Lin, G.; Zheng, L.; Zhang, W.; Xu, Z.; Zhu, F. SoCube: An Innovative End-to-End Doublet Detection Algorithm for Analyzing scRNA-Seq Data. Brief. Bioinform. 2023, 24, bbad104. [Google Scholar] [CrossRef]
  187. Agarwal, A.; Singh, K.; Kant, S.; Bahadur, R.P. A Comparative Analysis of Machine Learning Classifiers for Predicting Protein-Binding Nucleotides in RNA Sequences. Comput. Struct. Biotechnol. J. 2022, 20, 3195–3207. [Google Scholar] [CrossRef]
  188. Du, X.; Zhao, X.; Zhang, Y. DeepBtoD: Improved RNA-Binding Proteins Prediction via Integrated Deep Learning. J. Bioinform. Comput. Biol. 2022, 20, 2250006. [Google Scholar] [CrossRef]
  189. Du, Z.; Xiao, X.; Uversky, V.N. DeepA-RBPBS: A Hybrid Convolution and Recurrent Neural Network Combined with Attention Mechanism for Predicting RBP Binding Site. J. Biomol. Struct. Dyn. 2022, 40, 4250–4258. [Google Scholar] [CrossRef]
  190. Zhao, W.; Zhang, S.; Zhu, Y.; Xi, X.; Bao, P.; Ma, Z.; Kapral, T.H.; Chen, S.; Zagrovic, B.; Yang, Y.T.; et al. POSTAR3: An Updated Platform for Exploring Post-Transcriptional Regulation Coordinated by RNA-Binding Proteins. Nucleic Acids Res. 2022, 50, D287–D294. [Google Scholar] [CrossRef]
  191. 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]
  192. Blue, S.M.; Yee, B.A.; Pratt, G.A.; Mueller, J.R.; Park, S.S.; Shishkin, A.A.; Starner, A.C.; Van Nostrand, E.L.; Yeo, G.W. Transcriptome-Wide Identification of RNA-Binding Protein Binding Sites Using seCLIP-Seq. Nat. Protoc. 2022, 17, 1223–1265. [Google Scholar] [CrossRef]
  193. Ramanathan, M.; Porter, D.F.; Khavari, P.A. Methods to Study RNA-Protein Interactions. Nat. Methods 2019, 16, 225–234. [Google Scholar] [CrossRef]
  194. Wei, B.; Zhang, Y.; Gong, X. DeepLPI: A Novel Deep Learning-Based Model for Protein–Ligand Interaction Prediction for Drug Repurposing. Sci. Rep. 2022, 12, 18200. [Google Scholar] [CrossRef]
  195. Ding, Y.; Sun, Y.; Wang, H.; Zhao, H.; Yin, R.; Zhang, M.; Pan, X.; Zhu, X. Atherosis-Associated Lnc_000048 Activates PKR to Enhance STAT1-Mediated Polarization of THP-1 Macrophages to M1 Phenotype. Neural Regen. Res. 2024, 19, 2488–2498. [Google Scholar] [CrossRef]
  196. 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]
  197. Laverty, K.U.; Jolma, A.; Pour, S.E.; Zheng, H.; Ray, D.; Morris, Q.; Hughes, T.R. PRIESSTESS: Interpretable, High-Performing Models of the Sequence and Structure Preferences of RNA-Binding Proteins. Nucleic Acids Res. 2022, 50, e111. [Google Scholar] [CrossRef]
  198. Horlacher, M.; Wagner, N.; Moyon, L.; Kuret, K.; Goedert, N.; Salvatore, M.; Ule, J.; Gagneur, J.; Winther, O.; Marsico, A. Towards in Silico CLIP-Seq: Predicting Protein-RNA Interaction via Sequence-to-Signal Learning. Genome Biol. 2023, 24, 180. [Google Scholar] [CrossRef]
  199. Qiao, Y.; Yang, R.; Liu, Y.; Chen, J.; Zhao, L.; Huo, P.; Wang, Z.; Bu, D.; Wu, Y.; Zhao, Y. DeepFusion: A Deep Bimodal Information Fusion Network for Unraveling Protein-RNA Interactions Using in Vivo RNA Structures. Comput. Struct. Biotechnol. J. 2024, 23, 617–625. [Google Scholar] [CrossRef]
  200. Zhu, H.; Yang, Y.; Wang, Y.; Wang, F.; Huang, Y.; Chang, Y.; Wong, K.-C.; Li, X. Dynamic Characterization and Interpretation for Protein-RNA Interactions across Diverse Cellular Conditions Using HDRNet. Nat. Commun. 2023, 14, 6824. [Google Scholar] [CrossRef]
  201. Wu, Z.; Basu, S.; Wu, X.; Kurgan, L. qNABpredict: Quick, Accurate, and Taxonomy-Aware Sequence-Based Prediction of Content of Nucleic Acid Binding Amino Acids. Protein Sci. 2023, 32, e4544. [Google Scholar] [CrossRef]
  202. Zhang, F.; Li, M.; Zhang, J.; Kurgan, L. HybridRNAbind: Prediction of RNA Interacting Residues across Structure-Annotated and Disorder-Annotated Proteins. Nucleic Acids Res. 2023, 51, e25. [Google Scholar] [CrossRef]
  203. Chu, L.-C.; Christopoulou, N.; McCaughan, H.; Winterbourne, S.; Cazzola, D.; Wang, S.; Litvin, U.; Brunon, S.; Harker, P.J.; McNae, I.; et al. pyRBDome: A Comprehensive Computational Platform for Enhancing RNA-Binding Proteome Data. Life Sci. Alliance 2024, 7, e202402787. [Google Scholar] [CrossRef]
  204. Battistelli, C.; Garbo, S.; Riccioni, V.; Montaldo, C.; Santangelo, L.; Vandelli, A.; Strippoli, R.; Tartaglia, G.G.; Tripodi, M.; Cicchini, C. Design and Functional Validation of a Mutant Variant of the LncRNA HOTAIR to Counteract Snail Function in Epithelial-to-Mesenchymal Transition. Cancer Res. 2021, 81, 103–113. [Google Scholar] [CrossRef]
  205. Owens, M.C.; Clark, S.C.; Yankey, A.; Somarowthu, S. Identifying Structural Domains and Conserved Regions in the Long Non-Coding RNA lncTCF7. Int. J. Mol. Sci. 2019, 20, 4770. [Google Scholar] [CrossRef]
  206. Fu, J.; Yu, L.; Yan, H.; Tang, S.; Wang, Z.; Dai, T.; Chen, H.; Zhang, S.; Hu, H.; Liu, T.; et al. LncRNAs in Non-Small Cell Lung Cancer: Novel Diagnostic and Prognostic Biomarkers. Front. Mol. Biosci. 2023, 10, 1297198. [Google Scholar] [CrossRef]
  207. Delli Ponti, R.; Armaos, A.; Marti, S.; Tartaglia, G.G. A Method for RNA Structure Prediction Shows Evidence for Structure in lncRNAs. Front. Mol. Biosci. 2018, 5, 111. [Google Scholar] [CrossRef]
  208. Spetale, F.E.; Murillo, J.; Villanova, G.V.; Bulacio, P.; Tapia, E. FGGA-Lnc: Automatic Gene Ontology Annotation of lncRNA Sequences Based on Secondary Structures. Interface Focus. 2021, 11, 20200064. [Google Scholar] [CrossRef]
  209. Chorostecki, U.; Saus, E.; Gabaldón, T. Structural Characterization of NORAD Reveals a Stabilizing Role of Spacers and Two New Repeat Units. Comput. Struct. Biotechnol. J. 2021, 19, 3245–3254. [Google Scholar] [CrossRef]
  210. Lee, S.; Kopp, F.; Chang, T.-C.; Sataluri, A.; Chen, B.; Sivakumar, S.; Yu, H.; Xie, Y.; Mendell, J.T. Noncoding RNA NORAD Regulates Genomic Stability by Sequestering PUMILIO Proteins. Cell 2016, 164, 69–80. [Google Scholar] [CrossRef]
  211. Solayman, M.; Litfin, T.; Singh, J.; Paliwal, K.; Zhou, Y.; Zhan, J. Probing RNA Structures and Functions by Solvent Accessibility: An Overview from Experimental and Computational Perspectives. Brief. Bioinform. 2022, 23, bbac112. [Google Scholar] [CrossRef]
  212. Sanbonmatsu, K. Getting to the Bottom of lncRNA Mechanism: Structure–Function Relationships. Mamm. Genome 2022, 33, 343–353. [Google Scholar] [CrossRef]
  213. Qi, Y.; Zhang, Y.; Mu, Q.; Zheng, G.; Zhang, M.; Chen, B.; Huang, J.; Ma, C.; Wang, X. RNA Secondary Structurome Revealed Distinct Thermoregulation in Plasmodium Falciparum. Front. Cell Dev. Biol. 2022, 9, 766532. [Google Scholar] [CrossRef]
  214. Chan, D.; Feng, C.; England, W.E.; Wyman, D.; Flynn, R.A.; Wang, X.; Shi, Y.; Mortazavi, A.; Spitale, R.C. Diverse Functional Elements in RNA Predicted Transcriptome-Wide by Orthogonal RNA Structure Probing. Nucleic Acids Res. 2021, 49, 11868–11882. [Google Scholar] [CrossRef]
  215. Spitale, R.C.; Incarnato, D. Probing the Dynamic RNA Structurome and Its Functions. Nat. Rev. Genet. 2023, 24, 178–196. [Google Scholar] [CrossRef]
  216. De Vries, T.; Novakovic, M.; Ni, Y.; Smok, I.; Inghelram, C.; Bikaki, M.; Sarnowski, C.P.; Han, Y.; Emmanouilidis, L.; Padroni, G.; et al. Specific Protein-RNA Interactions Are Mostly Preserved in Biomolecular Condensates. Sci. Adv. 2024, 10, eadm7435. [Google Scholar] [CrossRef]
  217. Yankey, A.; Clark, S.C.; Owens, M.C.; Somarowthu, S. Purification and Structural Characterization of the Long Noncoding RNAs. Methods Mol. Biol. 2021, 2372, 93–110. [Google Scholar] [CrossRef]
  218. Saha, K.; Ghosh, G. Chemical Probing of RNA Structure In Vivo Using SHAPE-MaP and DMS-MaP. Methods Mol. Biol. 2023, 2666, 81–93. [Google Scholar] [CrossRef]
  219. Mitchell, D.; Cotter, J.; Saleem, I.; Mustoe, A.M. Mutation Signature Filtering Enables High-Fidelity RNA Structure Probing at All Four Nucleobases with DMS. Nucleic Acids Res. 2023, 51, 8744–8757. [Google Scholar] [CrossRef]
  220. Feltes, B.C.; Pinto, É.S.M.; Mangini, A.T.; Dorn, M. NIAS-Server 2.0: A Versatile Complementary Tool for Structural Biology Studies. J. Comput. Chem. 2023, 44, 1610–1623. [Google Scholar] [CrossRef]
  221. Bugnon, L.A.; Edera, A.A.; Prochetto, S.; Gerard, M.; Raad, J.; Fenoy, E.; Rubiolo, M.; Chorostecki, U.; Gabaldón, T.; Ariel, F.; et al. Secondary Structure Prediction of Long Noncoding RNA: Review and Experimental Comparison of Existing Approaches. Brief. Bioinform. 2022, 23, bbac205. [Google Scholar] [CrossRef]
  222. Singh, J.; Paliwal, K.; Zhang, T.; Singh, J.; Litfin, T.; Zhou, Y. Improved RNA Secondary Structure and Tertiary Base-Pairing Prediction Using Evolutionary Profile, Mutational Coupling and Two-Dimensional Transfer Learning. Bioinformatics 2021, 37, 2589–2600. [Google Scholar] [CrossRef]
  223. Matarrese, M.A.G.; Loppini, A.; Nicoletti, M.; Filippi, S.; Chiodo, L. Assessment of Tools for RNA Secondary Structure Prediction and Extraction: A Final-User Perspective. J. Biomol. Struct. Dyn. 2023, 41, 6917–6936. [Google Scholar] [CrossRef]
  224. Yu, B.; Li, P.; Zhang, Q.C.; Hou, L. Differential Analysis of RNA Structure Probing Experiments at Nucleotide Resolution: Uncovering Regulatory Functions of RNA Structure. Nat. Commun. 2022, 13, 4227. [Google Scholar] [CrossRef]
  225. Lasher, B.; Hendrix, D.A. bpRNA-Align: Improved RNA Secondary Structure Global Alignment for Comparing and Clustering RNA Structures. RNA 2023, 29, 584–595. [Google Scholar] [CrossRef]
  226. Ali, S.E.; Mittal, A.; Mathews, D.H. RNA Secondary Structure Analysis Using RNAstructure. Curr. Protoc. 2023, 3, e846. [Google Scholar] [CrossRef]
  227. Cheng, Y.; Zhang, S.; Xu, X.; Chen, S.-J. Vfold2D-MC: A Physics-Based Hybrid Model for Predicting RNA Secondary Structure Folding. J. Phys. Chem. B 2021, 125, 10108–10118. [Google Scholar] [CrossRef]
  228. Zhang, H.; Zhang, L.; Liu, K.; Li, S.; Mathews, D.H.; Huang, L. Linear-Time Algorithms for RNA Structure Prediction. In RNA Structure Prediction; Kawaguchi, R.K., Iwakiri, J., Eds.; Methods in Molecular Biology; Springer: New York, NY, USA, 2023; Volume 2586, pp. 15–34. ISBN 978-1-07-162767-9. [Google Scholar]
  229. Chen, C.-C.; Chan, Y.-M. REDfold: Accurate RNA Secondary Structure Prediction Using Residual Encoder-Decoder Network. BMC Bioinform. 2023, 24, 122. [Google Scholar] [CrossRef]
  230. Bose, E.; Xiong, S.; Jones, A.N. Probing RNA Structure and Dynamics Using Nanopore and next Generation Sequencing. J. Biol. Chem. 2024, 300, 107317. [Google Scholar] [CrossRef]
  231. Yang, M.; Zhu, P.; Cheema, J.; Bloomer, R.; Mikulski, P.; Liu, Q.; Zhang, Y.; Dean, C.; Ding, Y. In Vivo Single-Molecule Analysis Reveals COOLAIR RNA Structural Diversity. Nature 2022, 609, 394–399. [Google Scholar] [CrossRef]
  232. Oleynikov, M.; Jaffrey, S.R. RNA Tertiary Structure and Conformational Dynamics Revealed by BASH MaP. Elife 2024, 13, RP98540. [Google Scholar]
  233. Olson, S.W.; Turner, A.-M.W.; Arney, J.W.; Saleem, I.; Weidmann, C.A.; Margolis, D.M.; Weeks, K.M.; Mustoe, A.M. Discovery of a Large-Scale, Cell-State-Responsive Allosteric Switch in the 7SK RNA Using DANCE-MaP. Mol. Cell 2022, 82, 1708–1723.e10. [Google Scholar] [CrossRef]
  234. Luo, L.; Chiu, L.-Y.; Sugarman, A.; Gupta, P.; Rouskin, S.; Tolbert, B.S. HnRNP A1/A2 Proteins Assemble onto 7SK snRNA via Context Dependent Interactions. J. Mol. Biol. 2021, 433, 166885. [Google Scholar] [CrossRef]
  235. Callaway, E. Revolutionary Cryo-EM Is Taking over Structural Biology. Nature 2020, 578, 201. [Google Scholar] [CrossRef]
  236. Shepherd, D.C.; Dalvi, S.; Ghosal, D. From Cells to Atoms: Cryo-EM as an Essential Tool to Investigate Pathogen Biology, Host–Pathogen Interaction, and Drug Discovery. Mol. Microbiol. 2022, 117, 610–617. [Google Scholar] [CrossRef]
  237. Townshend, R.J.L.; Eismann, S.; Watkins, A.M.; Rangan, R.; Karelina, M.; Das, R.; Dror, R.O. Geometric Deep Learning of RNA Structure. Science 2021, 373, 1047–1051. [Google Scholar] [CrossRef]
  238. El Omari, K.; Duman, R.; Mykhaylyk, V.; Orr, C.M.; Latimer-Smith, M.; Winter, G.; Grama, V.; Qu, F.; Bountra, K.; Kwong, H.S.; et al. Experimental Phasing Opportunities for Macromolecular Crystallography at Very Long Wavelengths. Commun. Chem. 2023, 6, 219. [Google Scholar] [CrossRef]
  239. Skeparnias, I.; Zhang, J. Structural Basis of NEAT1 lncRNA Maturation and menRNA Instability. Nat. Struct. Mol. Biol. 2024, 31, 1650–1654. [Google Scholar] [CrossRef]
  240. Kim, D.N.; Thiel, B.C.; Mrozowich, T.; Hennelly, S.P.; Hofacker, I.L.; Patel, T.R.; Sanbonmatsu, K.Y. Zinc-Finger Protein CNBP Alters the 3-D Structure of lncRNA Braveheart in Solution. Nat. Commun. 2020, 11, 148. [Google Scholar] [CrossRef]
  241. Uroda, T.; Anastasakou, E.; Rossi, A.; Teulon, J.M.; Pellequer, J.L.; Annibale, P.; Pessey, O.; Inga, A.; Chillón, I.; Marcia, M. Conserved Pseudoknots in lncRNA MEG3 Are Essential for Stimulation of the p53 Pathway. Mol. Cell. 2019, 75, 982–995.e9. [Google Scholar] [CrossRef]
  242. Borodavka, A.; Singaram, S.W.; Stockley, P.G.; Gelbart, W.M.; Ben-Shaul, A.; Tuma, R. Sizes of Long RNA Molecules Are Determined by the Branching Patterns of Their Secondary Structures. Biophys. J. 2016, 111, 2077–2085. [Google Scholar] [CrossRef]
  243. Jones, A.N.; Sattler, M. Challenges and Perspectives for Structural Biology of lncRNAs—The Example of the Xist lncRNA A-Repeats. J. Mol. Cell Biol. 2019, 11, 845–859. [Google Scholar] [CrossRef]
  244. Qiu, X.; Li, H.; Ver Steeg, G.; Godzik, A. Advances in AI for Protein Structure Prediction: Implications for Cancer Drug Discovery and Development. Biomolecules 2024, 14, 339. [Google Scholar] [CrossRef]
  245. Qin, Y.; Chen, Z.; Peng, Y.; Xiao, Y.; Zhong, T.; Yu, X. Deep Learning Methods for Protein Structure Prediction. MedComm–Future Med. 2024, 3, e96. [Google Scholar] [CrossRef]
  246. Chen, L.; Li, Q.; Nasif, K.F.A.; Xie, Y.; Deng, B.; Niu, S.; Pouriyeh, S.; Dai, Z.; Chen, J.; Xie, C.Y. AI-Driven Deep Learning Techniques in Protein Structure Prediction. Int. J. Mol. Sci. 2024, 25, 8426. [Google Scholar] [CrossRef]
  247. Brandes, N.; Ofer, D.; Peleg, Y.; Rappoport, N.; Linial, M. ProteinBERT: A Universal Deep-Learning Model of Protein Sequence and Function. Bioinformatics 2022, 38, 2102–2110. [Google Scholar] [CrossRef]
  248. Krapp, L.F.; Abriata, L.A.; Cortés Rodriguez, F.; Dal Peraro, M. PeSTo: Parameter-Free Geometric Deep Learning for Accurate Prediction of Protein Binding Interfaces. Nat. Commun. 2023, 14, 2175. [Google Scholar] [CrossRef]
  249. Bryant, P. Deep Learning for Protein Complex Structure Prediction. Curr. Opin. Struct. Biol. 2023, 79, 102529. [Google Scholar] [CrossRef]
  250. Baek, M.; McHugh, R.; Anishchenko, I.; Jiang, H.; Baker, D.; DiMaio, F. Accurate Prediction of Protein–Nucleic Acid Complexes Using RoseTTAFoldNA. Nat. Methods 2024, 21, 117–121. [Google Scholar] [CrossRef]
  251. Green, A.G.; Elhabashy, H.; Brock, K.P.; Maddamsetti, R.; Kohlbacher, O.; Marks, D.S. Large-Scale Discovery of Protein Interactions at Residue Resolution Using Co-Evolution Calculated from Genomic Sequences. Nat. Commun. 2021, 12, 1396. [Google Scholar] [CrossRef]
  252. Liu, H.; Zhuo, C.; Gao, J.; Zeng, C.; Zhao, Y. AI-Integrated Network for RNA Complex Structure and Dynamic Prediction. Biophys. Rev. (Melville) 2024, 5, 041304. [Google Scholar] [CrossRef]
  253. Zhang, F.; Zhao, B.; Shi, W.; Li, M.; Kurgan, L. DeepDISOBind: Accurate Prediction of RNA-, DNA- and Protein-Binding Intrinsically Disordered Residues with Deep Multi-Task Learning. Brief. Bioinform. 2022, 23, bbab521. [Google Scholar] [CrossRef]
  254. Hennig, J. Structural Biology of RNA and Protein-RNA Complexes after AlphaFold3. Chembiochem 2025, 26, e202401047. [Google Scholar] [CrossRef]
  255. Chen, K.; Litfin, T.; Singh, J.; Zhan, J.; Zhou, Y. MARS and RNAcmap3: The Master Database of All Possible RNA Sequences Integrated with RNAcmap for RNA Homology Search. Genom. Proteom. Bioinform. 2024, 22, qzae018. [Google Scholar] [CrossRef]
  256. Cheng, J.; Lin, Y.; Xu, L.; Chen, K.; Li, Q.; Xu, K.; Ning, L.; Kang, J.; Cui, T.; Huang, Y.; et al. ViRBase v3.0: A Virus and Host ncRNA-Associated Interaction Repository with Increased Coverage and Annotation. Nucleic Acids Res. 2022, 50, D928–D933. [Google Scholar] [CrossRef]
  257. Sarzynska, J.; Popenda, M.; Antczak, M.; Szachniuk, M. RNA Tertiary Structure Prediction Using RNAComposer in CASP15. Proteins 2023, 91, 1790–1799. [Google Scholar] [CrossRef]
  258. Zhang, Y.; Xiong, Y.; Yang, C.; Xiao, Y. 3dRNA/DNA: 3D Structure Prediction from RNA to DNA. J. Mol. Biol. 2024, 436, 168742. [Google Scholar] [CrossRef]
  259. Kamga Youmbi, F.I.; Kengne Tchendji, V.; Tayou Djamegni, C. P-FARFAR2: A Multithreaded Greedy Approach to Sampling Low-Energy RNA Structures in Rosetta FARFAR2. Comput. Biol. Chem. 2023, 104, 107878. [Google Scholar] [CrossRef]
  260. Watkins, A.M.; Rangan, R.; Das, R. FARFAR2: Improved De Novo Rosetta Prediction of Complex Global RNA Folds. Structure 2020, 28, 963–976.e6. [Google Scholar] [CrossRef]
  261. Zhang, Y.; Lang, M.; Jiang, J.; Gao, Z.; Xu, F.; Litfin, T.; Chen, K.; Singh, J.; Huang, X.; Song, G.; et al. Multiple Sequence Alignment-Based RNA Language Model and Its Application to Structural Inference. Nucleic Acids Res. 2024, 52, e3. [Google Scholar] [CrossRef]
  262. Qiu, X. Robust RNA Secondary Structure Prediction with a Mixture of Deep Learning and Physics-Based Experts. Biol. Methods Protoc. 2025, 10, bpae097. [Google Scholar] [CrossRef]
  263. Zablocki, L.I.; Bugnon, L.A.; Gerard, M.; Di Persia, L.; Stegmayer, G.; Milone, D.H. Comprehensive Benchmarking of Large Language Models for RNA Secondary Structure Prediction. Brief Bioinform. 2025, 26, bbaf137. [Google Scholar] [CrossRef]
  264. Yi, H.-C.; You, Z.-H.; Wang, M.-N.; Guo, Z.-H.; Wang, Y.-B.; Zhou, J.-R. RPI-SE: A Stacking Ensemble Learning Framework for ncRNA-Protein Interactions Prediction Using Sequence Information. BMC Bioinform. 2020, 21, 60. [Google Scholar] [CrossRef]
  265. Sato, K.; Akiyama, M.; Sakakibara, Y. RNA Secondary Structure Prediction Using Deep Learning with Thermodynamic Integration. Nat. Commun. 2021, 12, 941. [Google Scholar] [CrossRef]
  266. Zhu, M.; Mathews, D.H. Sequence Design Using RNAstructure. Methods Mol. Biol. 2025, 2847, 17–31. [Google Scholar] [CrossRef]
  267. Raden, M.; Miladi, M. How to Do RNA-RNA Interaction Prediction? A Use-Case Driven Handbook Using IntaRNA. Methods Mol. Biol. 2024, 2726, 209–234. [Google Scholar] [CrossRef]
  268. Li, X.; Qu, W.; Yan, J.; Tan, J. RPI-EDLCN: An Ensemble Deep Learning Framework Based on Capsule Network for ncRNA-Protein Interaction Prediction. J. Chem. Inf. Model. 2024, 64, 2221–2235. [Google Scholar] [CrossRef]
  269. Liang, Y.; Yin, X.; Zhang, Y.; Guo, Y.; Wang, Y. Predicting lncRNA-Protein Interactions through Deep Learning Framework Employing Multiple Features and Random Forest Algorithm. BMC Bioinform. 2024, 25, 108. [Google Scholar] [CrossRef]
  270. Shi, F.; Zhang, K.; Cheng, Q.; Che, S.; Zhi, S.; Yu, Z.; Liu, F.; Duan, F.; Wang, Y.; Yang, N. Molecular Mechanism Governing RNA-Binding Property of Mammalian TRIM71 Protein. Sci. Bull. 2024, 69, 72–81. [Google Scholar] [CrossRef]
  271. Liu, B.; Shi, H.; Al-Hashimi, H.M. Developments in Solution-State NMR Yield Broader and Deeper Views of the Dynamic Ensembles of Nucleic Acids. Curr. Opin. Struct. Biol. 2021, 70, 16–25. [Google Scholar] [CrossRef]
  272. Zhu, X.; Xie, X.; Das, H.; Tan, B.G.; Shi, Y.; Al-Behadili, A.; Peter, B.; Motori, E.; Valenzuela, S.; Posse, V.; et al. Non-Coding 7S RNA Inhibits Transcription via Mitochondrial RNA Polymerase Dimerization. Cell 2022, 185, 2309–2323.e24. [Google Scholar] [CrossRef]
  273. Kerr, A.G.; Wang, Z.; Wang, N.; Kwok, K.H.M.; Jalkanen, J.; Ludzki, A.; Lecoutre, S.; Langin, D.; Bergo, M.O.; Dahlman, I.; et al. The Long Noncoding RNA ADIPINT Regulates Human Adipocyte Metabolism via Pyruvate Carboxylase. Nat. Commun. 2022, 13, 2958. [Google Scholar] [CrossRef]
  274. Schwalbe, H.; Audergon, P.; Haley, N.; Amaro, C.A.; Agirre, J.; Baldus, M.; Banci, L.; Baumeister, W.; Blackledge, M.; Carazo, J.M.; et al. The Future of Integrated Structural Biology. Structure 2024, 32, 1563–1580. [Google Scholar] [CrossRef]
  275. Ziegler, S.J.; Mallinson, S.J.B.; St. John, P.C.; Bomble, Y.J. Advances in Integrative Structural Biology: Towards Understanding Protein Complexes in Their Cellular Context. Comput. Struct. Biotechnol. J. 2021, 19, 214–225. [Google Scholar] [CrossRef]
  276. Huang, S.; Hao, X.-Y.; Li, Y.-J.; Wu, J.; Xiang, D.-X.; Luo, S. Nonviral Delivery Systems for Antisense Oligonucleotide Therapeutics. Biomater. Res. 2022, 26, 49. [Google Scholar] [CrossRef]
  277. Hammond, S.M.; Aartsma-Rus, A.; Alves, S.; Borgos, S.E.; Buijsen, R.A.M.; Collin, R.W.J.; Covello, G.; Denti, M.A.; Desviat, L.R.; Echevarría, L.; et al. Delivery of Oligonucleotide-based Therapeutics: Challenges and Opportunities. EMBO Mol. Med. 2021, 13, e13243. [Google Scholar] [CrossRef]
  278. Collotta, D.; Bertocchi, I.; Chiapello, E.; Collino, M. Antisense Oligonucleotides: A Novel Frontier in Pharmacological Strategy. Front. Pharmacol. 2023, 14, 1304342. [Google Scholar] [CrossRef]
  279. Esposito, R.; Bosch, N.; Lanzós, A.; Polidori, T.; Pulido-Quetglas, C.; Johnson, R. Hacking the Cancer Genome: Profiling Therapeutically Actionable Long Non-Coding RNAs Using CRISPR-Cas9 Screening. Cancer Cell 2019, 35, 545–557. [Google Scholar] [CrossRef]
  280. Jia, Z.; Wang, Y.; Sun, X.; Zhao, X.; Zhang, Y.; Xu, S.; Wang, Y.; Li, Y. Effect of lncRNA XLOC_005950 Knockout by CRISPR/Cas9 Gene Editing on Energy Metabolism and Proliferation in Osteosarcoma MG63 Cells Mediated by hsa-miR-542-3p. Oncol. Lett. 2021, 22, 669. [Google Scholar] [CrossRef]
  281. Aplin, C.; Milano, S.K.; Zielinski, K.A.; Pollack, L.; Cerione, R.A. Evolving Experimental Techniques for Structure-Based Drug Design. J. Phys. Chem. B 2022, 126, 6599–6607. [Google Scholar] [CrossRef]
  282. Saur, M.; Hartshorn, M.J.; Dong, J.; Reeks, J.; Bunkoczi, G.; Jhoti, H.; Williams, P.A. Fragment-Based Drug Discovery Using Cryo-EM. Drug Discov. Today 2020, 25, 485–490. [Google Scholar] [CrossRef]
  283. Stanzione, F.; Giangreco, I.; Cole, J.C. Use of Molecular Docking Computational Tools in Drug Discovery. In Progress in Medicinal Chemistry; Elsevier: Amsterdam, The Netherlands, 2021; Volume 60, pp. 273–343. ISBN 978-0-323-85056-8. [Google Scholar]
  284. Velmurugan, D.; Pachaiappan, R.; Ramakrishnan, C. Recent Trends in Drug Design and Discovery. Curr. Top. Med. Chem. 2020, 20, 1761–1770. [Google Scholar] [CrossRef]
  285. Radaeva, M.; Ho, C.-H.; Xie, N.; Zhang, S.; Lee, J.; Liu, L.; Lallous, N.; Cherkasov, A.; Dong, X. Discovery of Novel Lin28 Inhibitors to Suppress Cancer Cell Stemness. Cancers 2022, 14, 5687. [Google Scholar] [CrossRef]
  286. Byun, W.G.; Lim, D.; Park, S.B. Discovery of Small-Molecule Modulators of Protein–RNA Interactions by Fluorescence Intensity-Based Binding Assay. ChemBioChem 2020, 21, 818–824. [Google Scholar] [CrossRef]
  287. Borgelt, L.; Li, F.; Hommen, P.; Lampe, P.; Hwang, J.; Goebel, G.L.; Sievers, S.; Wu, P. Trisubstituted Pyrrolinones as Small-Molecule Inhibitors Disrupting the Protein–RNA Interaction of LIN28 and Let-7. ACS Med. Chem. Lett. 2021, 12, 893–898. [Google Scholar] [CrossRef]
  288. Pedram Fatemi, R.; Salah-Uddin, S.; Modarresi, F.; Khoury, N.; Wahlestedt, C.; Faghihi, M.A. Screening for Small-Molecule Modulators of Long Noncoding RNA-Protein Interactions Using AlphaScreen. SLAS Discov. 2015, 20, 1132–1141. [Google Scholar] [CrossRef]
  289. Li, A.; Bouhss, A.; Clément, M.-J.; Bauvais, C.; Taylor, J.P.; Bollot, G.; Pastré, D. Using the Structural Diversity of RNA: Protein Interfaces to Selectively Target RNA with Small Molecules in Cells: Methods and Perspectives. Front. Mol. Biosci. 2023, 10, 1298441. [Google Scholar] [CrossRef]
  290. Mwangi, M.N.; Yonkunas, M.J.; Ageeli, A.A.; McGovern-Gooch, K.R.; Yilmaz, S.; Baird, N.J. A Newly Identified Peripheral Duplex Anchors and Stabilizes the MALAT1 Triplex. Biochemistry 2024, 63, 2280–2292. [Google Scholar] [CrossRef]
  291. Swain, M.; Ageeli, A.A.; Kasprzak, W.K.; Li, M.; Miller, J.T.; Sztuba-Solinska, J.; Schneekloth, J.S.; Koirala, D.; Piccirili, J.; Fraboni, A.J.; et al. Dynamic Bulge Nucleotides in the KSHV PAN ENE Triple Helix Provide a Unique Binding Platform for Small Molecule Ligands. Nucleic Acids Res. 2021, 49, 13179–13193. [Google Scholar] [CrossRef]
Figure 1. Schematic illustration of major archetypes of LPIs (dashed boxes).
Figure 1. Schematic illustration of major archetypes of LPIs (dashed boxes).
Biomolecules 15 00881 g001
Figure 2. Recent uncovered lncRNA-protein partners and their associated diseases [39,64,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,,137,138,139,140,141,142,143,144,145,146,147,148,149].
Figure 2. Recent uncovered lncRNA-protein partners and their associated diseases [39,64,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,,137,138,139,140,141,142,143,144,145,146,147,148,149].
Biomolecules 15 00881 g002
Figure 3. Schematic description of current approaches for detecting LPI.
Figure 3. Schematic description of current approaches for detecting LPI.
Biomolecules 15 00881 g003
Figure 4. Illustration of current high-resolution 3D structural analysis methods for lncRNA-protein complexes.
Figure 4. Illustration of current high-resolution 3D structural analysis methods for lncRNA-protein complexes.
Biomolecules 15 00881 g004
Figure 5. Schematic diagram of proposed integrated structural analysis of lncRNA from the perspective of protein partners; the ISD strategy is incorporated into the pipeline.
Figure 5. Schematic diagram of proposed integrated structural analysis of lncRNA from the perspective of protein partners; the ISD strategy is incorporated into the pipeline.
Biomolecules 15 00881 g005
Table 1. Illustrates commonly used methods for detecting LPIs.
Table 1. Illustrates commonly used methods for detecting LPIs.
MethodPrincipleAdvantagesLimitationsRef.
Protein-CentricRIP-Seq/RIP-chipImmunoprecipitation of RBP, followed by RNA identificationSimple, applicable in vivoCannot map precise binding sites[155]
CLIP-Seq (HITS-CLIP, iCLIP, PAR-CLIP)UV crosslinking, IP of RBP, and sequencingSingle-nucleotide resolution; transcriptome-wideLabor-intensive,RNA loss during purification[156,168,191]
eCLIPEnhanced CLIP with size-matched input controlImproved specificity and reproducibilityRequires large sample amounts[160]
fCLIPFormaldehyde-based crosslinking CLIP variantCaptures transient or weak interactionsLower resolution compared to UV-based CLIP[161]
RBNSIn vitro pull-down of lncRNAs with immobilized RBPQuantifies binding affinity, statistical modelingIn vitro only, prone to false positives[152]
seCLIPSize-matched input and simplified protocol to purify and sequence LPIsHigh resolution, efficiency, and scalabilityStill relies on UV crosslinking and may require optimization [192]
RNA-CentricIVT RNABiotin-labeled RNA baits used to isolate RBPsStraightforward, customizableIn vitro only, RNA misfolding risks[165]
[193]
CHIRP/CHART/RAPHybridization of biotinylated probes to native RNA, IP of LPIsIn vivo relevance, genome-scale coverageRequires high expression levels and probe optimization[170]
MORPH-MSMultiplex probe hybridization followed by MSEfficient RNA retrieval with fewer oligosStill dependent on transcript abundance[173]
RAP-MSBiotinylated antisense probes capture target RNA and associated proteins, proteins identified by MSSpecific, enables in vivo RBP identificationRequires effective probe design, low resolution for binding sites[172]
RPL (RNA Proximity Labeling)Labeling enzymes are directed to specific RNAs to biotinylate nearby proteinsIn vivo compatible, identifies weak/transient interactionsRequires expression optimization[174]
CARPIDCRISPR assisted RNA–protein interaction detectionSpecific to endogenous RNAs, works without crosslinkingLimited by gRNA design and accessibility[175]
In SilicoLPICGAE, DFRPI, DeepLPIMachine learning-based prediction from sequence and structural dataFast screening, captures nonlinear patternsDependent on training data quality[176,179,194]
RNAInter, RPISeq, catRAPIDRule-based or probabilistic sequence feature modelsWeb-accessible, user-friendlyLimited performance on unseen RNA–protein pairs[67,195,196]
RNAincoder, SoCube, DeepBtoDAdvanced neural nets trained on interaction databasesImproved performance for large-scale predictionsLimited for low-abundance RNAs[185,186,188]
DeepA-RBPBSDeep learning model using CNN, BiGRU, and attention to predict RBP binding sites from RNA sequence and structureCaptures both sequence and structural features, effective across multiple RBPsRelies on well-annotated training data, may be sensitive to feature encoding quality[189]
PRIESSTESSUsing regression on binding data, integrating both sequence and secondary structure featuresCaptures both sequence and structure specificity, applicable to diverse RBPsMay oversimplify complex binding rules, depends on quality of input data[197]
RBPNetPredicts CLIP-seq crosslink count distribution from RNA sequence at single-nucleotide resolution, using bias correction and attribution methodsHigh-resolution predictions, supports eCLIP, iCLIP, and miCLIP, captures known and novel motifs, enables variant impact analysis via in silico mutagenesisRequires large-scale training data, computationally intensive, performance depends on data quality and assay consistency[198]
DeepFusionIntegrating RNA sequence and in vivo structural features from DMS-seq to predict RBP bindingCombines local motifs and global context, improved accuracy over sequence-only models, applicable to RNA degradation studiesDepends on high-quality DMS-seq data, computationally intensive[199]
HDRNetDeep learning-based framework to predict dynamic RBP binding events under diverse cellular conditionsHigh accuracy in cross cell type prediction, interpretable motifs, applicable to disease association studiesRequires large, diverse annotated datasets, black-box model may limit mechanistic insight[200]
qNABpredictPredicts nucleic acid-binding residue content using support vector regression and computed features from protein sequenceMuch faster than residue-level methods, provides informative summary, suitable for large scale analysisLess detailed than residue-level tool, focuses on content rather than precise binding residues[201]
HybridRNAbindMeta-model integrating structure- and disorder-trained predictors for RNA-binding residuesReduces cross-predictions, works well across structured and disordered regionsPerformance depends on quality/diversity of input models, modestly effective on non-matching annotations[202]
pyRBDomePipeline integrating ML-based RBS predictions with structural data to refine RBPs datasetsEnhances confidence in RNA-binding proteome data, improves RBS detection via ensemble learningRelies on existing structure/ML data quality, structural benchmarks still have known limitations[203]
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

Wang, Z.; Aftab, M.; Dong, Z.; Jiang, Y.; Liu, K. LncRNA–Protein Interactions: A Key to Deciphering LncRNA Mechanisms. Biomolecules 2025, 15, 881. https://doi.org/10.3390/biom15060881

AMA Style

Wang Z, Aftab M, Dong Z, Jiang Y, Liu K. LncRNA–Protein Interactions: A Key to Deciphering LncRNA Mechanisms. Biomolecules. 2025; 15(6):881. https://doi.org/10.3390/biom15060881

Chicago/Turabian Style

Wang, Zuoneng, Muhammad Aftab, Zigang Dong, Yanan Jiang, and Kangdong Liu. 2025. "LncRNA–Protein Interactions: A Key to Deciphering LncRNA Mechanisms" Biomolecules 15, no. 6: 881. https://doi.org/10.3390/biom15060881

APA Style

Wang, Z., Aftab, M., Dong, Z., Jiang, Y., & Liu, K. (2025). LncRNA–Protein Interactions: A Key to Deciphering LncRNA Mechanisms. Biomolecules, 15(6), 881. https://doi.org/10.3390/biom15060881

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