Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
International Journal of Epigenetics
Join Editorial Board Propose a Special Issue
Print ISSN: 2752-5406 Online ISSN: 2752-5414
Journal Cover
January-December 2025 Volume 5 Issue 1

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
January-December 2025 Volume 5 Issue 1

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML

  • Supplementary Files
    • Supplementary_Data1.pdf
    • Supplementary_Data2.pdf
Article Open Access

Role of lncRNAs related to NRs in the regulation of gene expression

  • Authors:
    • Katerina Pierouli
    • Louis Papageorgiou
    • George P. Chrousos
    • Elias Eliopoulos
    • Dimitrios Vlachakis
  • View Affiliations / Copyright

    Affiliations: Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens 11855, Greece
    Copyright: © Pierouli et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].
  • Article Number: 3
    |
    Published online on: April 22, 2025
       https://doi.org/10.3892/ije.2025.26
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Long non‑coding RNAs (lncRNAs) play a key role in regulating gene expression, influencing various cellular pathways by interacting with transcription factors, other RNA molecules such as microRNAs (miRNAs) and DNA. The present focused on the role of lncRNAs related to nuclear receptors (NRs), which are a family of transcription factors activated by ligands. NRs are involved in vital biological processes, such as metabolism, immune response, reproduction and development. Herein, six novel sequence motifs were discovered within lncRNAs that respond to multiple NRs, suggesting they are not specific to a single receptor. Of note, one of these motifs, was complementary to miRNA hsa‑miR‑1908‑3p, suggesting that lncRNAs containing this motif may function as miRNA sponges, regulating the expression of ~487 target mRNAs. The motifs were also found in key regulatory regions of the human genome, particularly on chromosome 19, including in adeno‑associated virus integration site 1, a region with a high regulatory role in gene expression. Additionally, an evolutionary analysis was conducted, revealing that these motifs are highly conserved across species, including Mus musculus, Euglena gracilis and Saccharomyces cerevisiae, indicating their ancient origins. Based on these results, it is suggested that these motifs may represent ancestral binding sites for NR precursors, and may function as backup mechanisms in modern organisms, ensuring the functional versatility and evolutionary conservation of NR‑mediated gene regulation.

Introduction

As per the central dogma of biology, RNA functions as an intermediate molecule, assisting transfer genetic information from the DNA level to the protein synthesis level. For a number of years, this process was considered the primary pathway for gene expression. However, over time, the functional importance of RNA transcripts and other non-coding regions, that are not directly encoded to proteins, has been recognized. Various of these non-coding RNAs (ncRNAs) are involved in several cellular functions, including the regulation of gene expression, alternative splicing, RNA processing, the inhibition of translation and mRNA degradation. In fact, RNA plays a crucial role in almost every level of genome regulation (1-3).

ncRNAs are typically divided into two main classes: Structural and regulatory. Structural ncRNAs include ribosomal RNAs, transfer RNAs and small nuclear RNAs. On the other hand, regulatory ncRNAs are further classified into several subcategories, such as microRNAs (miRNAs/miRs), Piwi-interacting RNAs, small interfering RNAs and long ncRNAs (lncRNAs). In addition, a subclass of promoter-associated RNAs that interacts with RNA promoters and is produced by genomic enhancer regions (enhancer RNAs) has previously been described (4).

lncRNAs are a highly diverse and heterogeneous class of ncRNAs, varying in their characteristics, locations in the genome and functional roles. They are the most abundant class of non-coding transcripts, and they typically have low expression levels and are >200 nucleotides in length (5). These RNAs can be categorized based on their location in the genome, into intronic, intergenic (lincRNAs), divergent, sense and antisense lncRNAs (6,7). Unlike small ncRNAs, lncRNAs are less conserved across species and their roles in the regulation of gene expression are not yet fully understood (8).

Even though the limited number of functional lncRNAs have been well characterized, they play essential roles in the regulation of gene expression at various stages. lncRNAs are involved in post-transcriptional regulation by controlling protein synthesis, RNA maturation and RNA transport, as well as in gene silencing through modulation of chromatin structure. Due to the vast variety of lncRNAs, categorizing them is challenging. However, several studies have classified them based on their molecular functions, as signaling molecules that relay temporal, spatial and developmental information; as decoys that isolate various RNA molecules and proteins to inhibit their activity; as guides that direct epigenetic regulators and transcriptional factors to specific genomic loci; and as scaffolds that facilitate the formation of macromolecular complexes with a various functions (8). Notably, individual lncRNAs may function through more than one mechanism, potentially leading to the combination of functions that create more complex functions in cellular procedures. Thus, understanding the shared features of these mechanisms is crucial for elucidating the functions of lncRNAs and their crucial roles in cellular regulation (8).

In addition to the aforementioned functions, several lncRNAs regulate gene expression by binding to miRNAs, functioning as ‘miRNA sponges’. These lncRNAs contain multiple binding sites for one or more miRNAs and prevent their interaction with target mRNAs. As a result, the target mRNAs of the miRNAs are upregulated, increasing their expression (8,9).

The direct and indirect interactions between lncRNAs and transcription factors are well-documented. For instance, lncRNA Gas5, which is activated in the absence of growth factors, contains a hairpin structure sequence pattern that resembles the DNA-binding site of the glucocorticoid receptor (GR), a member of the family of nuclear receptors (NRs). By acting as a decoy, Gas5 blocks the access of GR to DNA, thus inhibiting the transcription of GR metabolic target genes (10). NRs are transcription factors that bind to DNA and regulate the expression of genes involved in processes, such as homeostasis, stress response and metabolism (11). The dysregulation of NRs has been reported in various diseases, including neurodegenerative diseases, cancer, metabolic syndromes, diabetes, obesity and autism (12). Thus, understanding the mechanisms of interaction between lncRNAs and NRs is critical for determining the complex molecular pathways involved and could lead to the development of more effective and specialized therapies for plenty of diseases.

To further explore the regulatory roles of lncRNAs in gene expression, the present study examined lncRNAs whose expression levels were altered following the activation of an NR. These lncRNAs were classified into two groups as follows: Those that are upregulated and those that are downregulated. The aim was to explore whether any lncRNAs fail to exhibit a regulatory function. Considering the evolutionary conservation of lncRNAs, it was hypothesized that the presence of sequence motifs within lncRNA sequences could provide insight into their regulatory roles. These motifs, if present with non-random frequency, may serve as signatures that contribute to the regulatory functions of lncRNAs.

Using the MEME algorithm, the lncRNA sequences from the aforementioned groups were analyzed and six specific sequence motifs (three in each group) were identified. The potential roles of these motifs were investigated, providing insight into how lncRNAs containing these motifs may regulate a wide range of biological processes. According to the results obtained, one of the discovered motifs was found to be complementary to the miRNA hsa-miR-1908-3p, suggesting that lncRNAs containing this motif may act as sponges for this miRNA, regulating the expression of its target mRNAs. Additionally, all six motifs were detected in specific genomic regions of human genome, particularly on chromosome 19, with a high percentage of occurrence. Among these regions on chromosome 19, the motifs were aligned with the adeno-associated virus (AAV) integration site 1 (AAVS1), a known hotspot for the AAV integration. Notably, a number of lncRNAs containing at least one of these motifs, participate in various biological pathways such as development, immune response, inflammation, cell cycle, regulation of gene expression and homeostasis, highlighting the diverse roles of lncRNAs in cellular pathways. To explore the evolutionary significance of these motifs, the present study investigated their presence across various species, including Mus musculus, Squamata (lizard), Danio rerio (zebrafish), Drosophila melanogaster, Caenorhabditis elegans, Escherichia coli (K12 MG1655), Saccharomyces cerevisiae, Bacillus subtilis (PY79) and Euglena gracilis, which differ both evolutionarily and genetically. This cross-species analysis revealed that these motifs are evolutionary conserved, suggesting their potential importance in fundamental biological processes. The conservation of these motifs across a wide range of species highlights their potential functional relevance, warranting further investigation into their roles in gene regulation and interaction with NRs.

Data and methods

The present study developed an in-house pipeline for the analysis of the collected dataset of lncRNAs with known expression levels, as documented in the literature. These lncRNAs are related to the activation and expression levels of NRs. This pipeline includes the following key steps: i) Literature review and data retrieval: A comprehensive search in current literature was conducted to identify lncRNAs whose expression levels are known to be associated with the activation of NRs. In this stage, the corresponding RNA sequences were also retrieved for further analysis. ii) Classification of lncRNAs: The identified lncRNAs were classified into two groups. The first group included lncRNAs whose expression was upregulated following the activation of an NR, while the second group included lncRNAs whose expression was downregulated in response to the activation of an NR. Subsequently, conserved sequence motifs were identified within these two lncRNA sub-groups. iii) Statistical evaluation of motif significance: The identified motifs were statistically evaluated to determine their significance. This involved estimating the probability of these motifs occurring randomly within the entire human DNA. iv) Exploration of motif functionality: In this step, we explored the potential functions of the identified motifs, focusing on their role in gene regulation. This included investigating their potential to function as binding sites for miRNAs, examining their presence within genome regulatory regions and exploring their conservation through evolution. These analyses helped broaden the understanding of the roles of these lncRNAs in cellular processes. A detailed description of the in-house pipeline is provided below.

Data collection and filtering and annotation. The IDs of lncRNAs related to the NRs were collected from the literature according to the study by Foulds et al (13). The IDs of lncRNAs had the following formats: i) Name of lncRNA (e.g., MALAT-1); ii) ID such as AC145110.1; iii) ID such as NR_026765; iv) ID such as RP1-148H17.1.

The first three ID formats correspond to different types of IDs detected at GenBank, a comprehensive genetic sequence database available at NCBI (ncbi.nlm.nih.gov). The fourth ID format derived from LNCipedia database (14), a novel database for human lncRNA transcripts and genes.

Based on these ID formats, the dataset of lncRNAs was classified into four subcategories. To retrieve the corresponding sequences for each subcategory, an in-house algorithm was developed using the MATLAB bioinformatics toolbox. The details of the four algorithms used for sequence retrieval are presented in Data S1.

Following the creation of the four individual algorithms for each type of lncRNA ID, we developed a unified algorithm designed to retrieve the sequences of the entire dataset, containing all ID formats simultaneously. The aggregate algorithm used for retrieving all lncRNA sequences is presented in Data S2.

Dataset classification and motif exploration. In order to further classify the lncRNAs within the two sub-datasets, the present study investigated the presence of joint motifs across the lncRNA sequences in each subgroup. Following the collection, filtering and annotation of the data, the Multiple EM for Motif Elicitation (MEME) software, version 4.10.0(15), was employed to identify joint motifs within the lncRNA sequences of each subgroup. MEME is a part of the ΜEME Suite (https://meme-suite.org/meme/), which is a comprehensive set of tools for motif-based sequence analysis. This tool is designed to detect motifs or sequence patterns that occur repeatedly in a group of related nucleotide or protein sequences, finding common biological functions and structural features across different sequences. The term ‘pattern’ or ‘motif’ refers to a short, sequence that repeats within a group of similar sequences. The motifs have no gaps, while the MEME algorithm calculates how often each motif appears across sequences, allowing the detection of multiple motifs (15). For the purposes of the present study, the MEME algorithm was run on a local server using the Linux operating system.

Statistical evaluation of the randomness of the occurrence of the discovered motifs. To calculate the probability of finding specific nucleotide bases of each motif at a random position within the 3.2 billion bases of the human DNA, it was assumed that the occurrence of a nucleotide at one position in the motif's sequence is independent of the occurrence of nucleotides at other positions. This assumption represents the simplest hypothesis, involving the fewest parameters. Thus, the rule of probabilities for independent events was applied, which defines the probability of multiple events occurring independently. Specifically, two events are considered independent if the occurrence or non-occurrence of one does not influence the probability of the other event's occurrence or non-occurrence (16,17). To calculate the probability of both events A and B occurring together, the formula for independent events was used: P (A and B)=P (A) x P (B). This approach was applied to assess the randomness of each motif existence within human DNA.

Exploration of the potential role of the identified motifs. A total of six motifs were discovered, three in each subgroup of lncRNAs. To explore the potential role of the collected lncRNAs in the regulation of gene expression, the present study focused on the lncRNAs that contained at least one of the discovered motifs. Since motifs are usually conserved through evolution and contribute crucial functions to the regions they occupy, the authors proceeded to explore the potential roles of these motifs in genome regulation, which could reflect the functions of the lncRNAs that contain them. For this purpose, two different approaches were pursued. The first included querying the miRBase database (https://www.mirbase.org/) to identify mature miRNAs with complementary sequences to these motifs, suggesting the possibility of miRNA-lncRNA interaction. The second approach included the exploration of the presence of these motif sequences across whole human DNA in order to determine their existence in other genomic regions, assess whether they were associated with regulatory regions, and explore potential roles in genomic regulation. In addition, the evolutionary conservation of the motifs was explored to determine the maintenance of their functions through evolution.

Exploration of the interactions of miRNA target genes. The main purpose of this approach was to identify the potential role of lncRNAs containing the identified motifs in functioning as ‘sponges’ for miRNAs, where the motifs serve as interaction/binding regions between lncRNAs and miRNAs. For this purpose, the blastn algorithm from miRbase was used, with the complement sequence of each motif as input. This allowed this algorithm to search for similarities between the motif sequences and known miRNA sequences in the miRBase. Subsequently, the authors further investigated the predicted target genes of the identified miRNA using the bioinformatic tool miRTarget from the miRDB database (16), and the TargetScanS database (Release 8.0) (17-19).

Finally, the location of the binding sites in the target mRNAs was examined by using the multiple alignment algorithm ClustalW, using as input the mRNA sequence of the targets and the complementary sequence of miR-1908-3p.

Exploration of motif sequences in whole human DNA. To investigate whether these motif sequences also appear in other regions of the genome, the online tool Find Individual Motif Occurrences (FIMO) from the MEME Suite was used. This tool is an algorithm designed to identify specific matches of a given motif within a set of sequences, specialized for analyzing shorter sequences such as motifs. Similar to a BLAST algorithm, FIMO searches for matches to each motif. When employed locally, motifs must be in MEME Motif Format; however, the online version of FIMO supports additional motif formats.

Subsequently, an algorithm was developed using the MATLAB Bioinformatics Toolbox to create a chromosome plot for each identified motif. This plot displayed the locations of the motif sequences by marking them with a red dot next to the corresponding regions on the chromosome where they were detected. The input for this algorithm was the output from the FIMO tool in tsv format. The specific algorithm we developed is presented in Data S3.

The FIMO tool was also used to examine the presence of the identified sequence motifs in the genomes of various organisms, including Mus musculus, Squamata (lizard), Danio rerio (zebrafish), Drosophila melanogaster, Caenorhabditis elegans, Escherichia coli (K12 MG1655), Saccharomyces cerevisiae and Bacillus subtilis (PY79), to assess their evolutionary conservation. Additionally, the present study investigated the presence of these motifs in the genome of Euglena gracilis, a protist believed to possess an ancient form of NRs. Due to the unavailability of Euglena's genome in the database of the FIMO tool, the ‘Matcher’ tool available on the Galaxy platform (usegalaxy.org) was employed to check for potential sequence matches (20).

Galaxy is an open-source, web-based platform that aids computational biology research by providing a comprehensive suite of tools for data analysis. The ‘Matcher’ tool within this platform compares two different sequences to identify regions of similarity, supporting both local and global alignments. This tool enables the identification of conserved genomic regions across species and helps in detecting functionally important motifs that have been preserved through evolution. This tool is widely used in genomics and bioinformatics for tasks such as gene annotation, mutation analysis, and the discovery of conserved regulatory regions (21).

Results and Discussion

Data collection, filtering and annotation

The initial dataset, collected from the study by Foulds et al (13), contained 1,823 lncRNA IDs, with sequences for 1,788 lncRNAs successfully identified and retrieved. Following the removal of the duplicate sequences, the final dataset consisted of 1,753 lncRNAs. These were then classified into two subgroups as follows: The first subgroup contained 778 upregulated lncRNAs and the second subgroup contained 975 downregulated lncRNAs (Table SI).

Dataset classification and motif identification

The MEME algorithm was employed to identify potential motifs within the lncRNA sequences of each subgroup. The results of this analysis led to the identification of six motifs: Three motifs in the upregulated lncRNA group and three in the downregulated lncRNA group.

In the upregulated lncRNAs group following NR activation, the first identified motif was GATCCGCCCGCCTCGGCCTCCCAAAGTGCTGGGATTACAGG, found in 140 lncRNAs from the total 778 (Fig. S1). The second motif discovered in this group was TGGCCAGGCTGGTCTCGAACTCCTGACCT, present in 127 lncRNAs (Fig. S2), while the third motif was TAATCCCAGCACTTTGGGAGGCCGAGG, which occurs in 115 lncRNAs (Fig. S3).

For the downregulated lncRNA group following NR activation, the first motif discovered was GTGGCTCACGCCTGTAATCCCAGCACTTTGGGAGGCCGAGG, found in 145 lncRNA out of the total of 975 (Fig. S4). The second motif discovered was AGGTCAGGAGTTCGAGACCAGCCTGGCCAACATGGTGAAAC, which is present in 128 lncRNAs (Fig. S5), and the third motif was CCTCAGCCTCCCGAGTAGCTGGGACTACAGGCGC, found in 129 lncRNAs from this group (Fig. S6).

Statistical evaluation

To calculate the probability of each motif appearing randomly at a position within the 3.2 billion base pairs of human DNA, the following steps were taken:

The first motif from the upregulated lncRNAs subgroup consists of 41 nucleotide bases. Since each base (A, C, G, or T) has a ¼ probability of being in a specific position in the sequence, the probability of finding the full sequence of these 41 nucleotide bases is: (¼)41=2.23517418x10-25.

Taking into consideration the total number of human DNA base pairs (3.2 billion or 3.2x109), the probability of this sequence occurring randomly at any position in the genome is: 3.2x109x2.23517418x10-25=7.15255974x10-16.

Accordingly, for the second motif in the upregulated lncRNA subgroup, consisting of 29 nucleotides, the probability calculation is as follows: (¼)29=1.8626451x10-18. The probability of finding this specific sequence randomly in human genome is: 3.2x109x1.8626451x10-18=5.96174432x10-10. For the third motif in the upregulated lncRNAs subgroup, which consists of 27 nucleotides, the results are as follows: (¼)27=7.8886x10-17. Thus, the probability of its random occurrence is: 3.2x109x7.8886x10-17=2.52435x10-7.

In the downregulated lncRNA subgroup, both first and second motifs consist of 41 nucleotides; thus, the same probability calculations apply: (¼)41=2.23517418x10-25. Thus, the probability of their random occurrence is: 3.2x109x2.23517418x10-25=7.15255974x10-16. The third motif in the downregulated lncRNA subgroup, consists of 34 nucleotides: (¼)34=1.421085x10-21. Thus, the probability of its random occurrence is: 3.2x109x1.421085x10-21=4.5474755x10-13.

The calculated probabilities are extraordinarily low compared to common biological standards and are consistent with the hypothesis that these motifs are not randomly occurring in the genome (22).

The above-calculated probabilities suggest that the motifs discovered are non-random occurrences in the human genome, even when assuming independent events. However, the nucleotide sequence of DNA is linked to biological functions. The order of the bases is strictly determined by the way nucleotides bind each other in the double helix structure. The complementary pairing of nucleotides restricts the possible sequences that DNA can form, meaning that the probability of these motifs occurring randomly in the genome would likely be even lower than the values calculated above.

Exploration of the potential role of the identified motifs. Exploration of the miRNA-target gene interactions

As previously mentioned, lncRNAs can function as miRNA sponges, regulating the expression of mRNA targets (8,9). Considering this regulatory role, we searched for potential miRNAs complementary to the six identified motifs in miRBase. In the present study, the search revealed that the motif GATCCGCCCGCCTCGGCCTCCCAAAGTGCTGGGATTACAGG was complementary to hsa-miR-1908-3p.

miRNAs typically bind to complementary sequences in mRNAs, mainly targeting the 3' untranslated regions (UTRs) (23), while there is evidence to indicate that they can also target the 5' UTRs and coding regions, thus inhibiting mRNA expression (24). To predict the potential targets of miRNAs, several computational approaches have been developed, such as miRanda (25), mirSVR (26), PicTar (27), TargetScan (28), TargetScanS (29), RNA22(30), PITA (31), RNAhybird (32) and DIANA-microT (33). In addition, there are miRNA target prediction databases such as TarBase (34), PicTar (27) and TargetScan (28).

Thus, to identify mRNAs targeted by hsa-miR-1908-3p, whose expression may be affected by the interaction between lncRNAs and this miRNA, the miRDB database was used. This database, through its miRTarget bioinformatics tool, predicts the mRNA based on high-throughput sequencing experiments. By this analysis, 11 targets were identified, including METTL21A, CHFR, MTA1, CAVIN4, IGFBP3, GRB2, DYNLL2, H2AFX, TNRC6C, C1orf229 and ELOVL2 (Table I). In addition, the TargetScanS (Release 8.0) database (17-19,35) was also searched, and 487 predicted targets were found (Table SII), including the aforementioned 11 targets.

Table I

The target genes of mir-1908-3p were predicted using the miRTarget tool of miRDB.

Table I

The target genes of mir-1908-3p were predicted using the miRTarget tool of miRDB.

Gene symbolGene descriptionmiRNA nameTarget score
METTL21AMethyltransferase like 21A hsa-miR-1908-3p80
CHFRCheckpoint with forkhead and ring finger domains hsa-miR-1908-3p62
MTA1Metastasis associated 1 hsa-miR-1908-3p62
CAVIN4Caveolae associated protein 4 hsa-miR-1908-3p61
IGFBP3Insulin like growth factor binding protein 3 hsa-miR-1908-3p61
GRB2Growth factor receptor bound protein 2 hsa-miR-1908-3p56
DYNLL2Dynein light chain LC8-type 2 hsa-miR-1908-3p55
H2AFXH2A histone family member X hsa-miR-1908-3p53
TNRC6CTrinucleotide repeat containing 6C hsa-miR-1908-3p53
C1orf229Chromosome 1 open reading frame 229 hsa-miR-1908-3p51
ELOVL2ELOVL fatty acid elongase 2 hsa-miR-1908-3p50

miR-1908 is encoded by the first intron of the FADS1 gene and is highly expressed in mature human adipocytes, where it is likely involved in regulating the preadipocyte differentiation (36). The promoter region of miR-1908 contains two binding sites of NF-κB, suggesting its transcription is regulated by this transcription factor (37). miR-1908 was first discovered in human embryonic stem cells in 2008(38), and its functional role in melanoma metastasis and angiogenesis was explored in 2012(39). More specifically, miR-1908, in conjunction with miR-199a-3p and miR-199a-5p, inhibits apolipoprotein E and DNAJ heat shock protein family 23 member A4, which reduces the interaction of Apo-E with low-density lipoprotein receptor-related protein (LRP)-1 and LRP-8. This interaction promotes melanoma cell invasion and endothelial cell recruitment, resulting in poor prognosis (39). Moreover, miR-1908 dysregulation has been shown to be associated with various types of cancer, where its dysregulation tends to enhance cell proliferation, invasion, migration and angiogenesis, thus reducing the survival rate of affected patients (40,41). Finally, free fatty acids and adipokines, such as resistin and leptin, are considered to downregulate the expression of miR-1908, suggesting a role of this miRNA in regulating obesity-related insulin resistance (42).

Exploration of motif sequences in whole human DNA

For the analysis of motif sequences across the whole human genome using the FIMO tool, the following results were obtained for each of the six motifs. In the upregulated lncRNA subgroup, the first motif appeared 53,524 times, the second motif was detected 51,673 times and the third motif was found 53,936 times. In the downregulated lncRNA subgroup, the first motif (fourth in total) was found 63,126 times, the second motif (fifth in total) was detected 58,442 times and the third motif (sixth in total) was found 57,943 times. Using these results, karyotypes were created for each motif, and the percentage of the occurrences of each motif across all chromosomes was calculated. The karyotypes are presented in Fig. S7, Fig. S8, Fig. S9, Fig. S10, Fig. S11 and Fig. S12.

Subsequently, the present study focused on the 1,000 highest-scoring loci which occurred using the FIMO tool for each motif across the human genome. Using these 1,000 loci, karyotypes for each motif were also created, and the percentage of occurrences of each motif was also calculated for each chromosome (Fig. 1, Fig. 2, Fig. 3, Fig. 4, Fig. 5 and Fig. 6).

Figure 1

Human karyotype for the 1,000 higher-scored loci that present the percentage of the occurrence of motif 1 in each chromosome.

Figure 2

Human karyotype for the 1,000 higher-scored loci that presents the percentage of the occurrence of motif 2 in each chromosome.

Figure 3

Human karyotype for the 1,000 higher-scored loci that presents the percentage of the occurrence of motif 3 in each chromosome.

Figure 4

Human karyotype for the 1000 higher-scored loci that presents the percentage of the occurrence of motif 4 in each chromosome.

Figure 5

Human karyotype for the 1,000 higher-scored loci that presents the percentage of the occurrence of motif 5 in each chromosome.

Figure 6

Human karyotype for the 1,000 higher-scored loci that presents the percentage of the occurrence of motif 6 in each chromosome.

In addition, the chromosome plots for each motif were created, where the 1,000 loci are represented as red dots beside the corresponding region. These chromosome plots are presented in Fig. S13, Fig. S14, Fig. S15, Fig. S16, Fig. S17 and Fig. S18.

Based on these karyotypes and chromosome plots, it was observed that chromosome 19 exhibited a fairly high percentage of motif occurrence relative to its size. In fact, for all six motifs, the percentage of occurrence in chromosome 19 was higher than that of the longer chromosomes.

According to the literature, human chromosome 19 is recognized for its unusual nature, which was noted even before the initial publication of its DNA sequence (43). One of its unusual features is the considerably high gene density, which exceeds the genome-wide average by >2-fold and includes 20 large, tandemly clustered gene families (43). In addition, chromosome 19 contains a considerable number of segmental duplications, with 6.2% of its sequence lying within intrachromosomal segmental duplications (43). The sequence divergence within these duplications implies that these events occurred between 30 and 40 million years ago (44). These duplications likely played a pivotal role in the evolution of phenotypic characteristics that occurred by genes on chromosome 19 across primates, including humans. Moreover, chromosome 19 presents an unusually high repeat content, comprising 55%, with Alu repeats comprising 26% of the sequence of the chromosome (44). Another notable feature is the GC content (48%) of chromosome 19, the highest of any human chromosome, compared to the GC content of the whole human genome of 41%. This elevated GC content enhances the potential for wide gene regulation via DNA methylation, particularly at CpG islands, as well as in CpG regions located within promoters and enhancers (45).

According to the chromosome plots, the six identified motifs were detected in various regions of chromosome 19. Between those regions, all six motifs aligned with a specific locus, AAVS1, located on chromosome 19q13.42. This site was first mapped in 1991 Kotin et al (46) and Samulski et al (47), who were investigating the integration mechanism of the AAV into the host genome. According to these studies, the AAV preferentially intergrades into the AAVS1 region of human chromosome 19q13.42, a unique phenomenon among eukaryotic DNA viruses (48).

Several features make the AAVS1 region particularly noteworthy. Notably, the locus for myotonic dystrophy (49), as well as a common breakpoint related to chronic B-cell leukemia (BCL-3) (47,48), are located at this region. In addition, AAVS1 undergoes frequent sister chromatid exchanges (50). The alignments of motifs with the AAVS1 region were developed using the Clustal Omega program (51) and visualized with Jalview version 2.11.2.6 program (52) (Fig. S19, Fig. S20, Fig. S21, Fig. S22, Fig. S23 and Fig. S24).

The AAVS1 region is located within the first intron of the phosphatase 1 regulatory subunit 12C (PPP1R12C) gene, which encodes a protein with a poorly defined function. This region exhibits several unique features that contribute to its unique nature. First, it includes a tetrad GACT repeat, which functions as an AAV Rep-binding element, also detected in the AAV2 inverted terminal repeats, facilitating the exclusive integration of the AAV genome in the presence of AAV replication protein. Second, chromosome 19 is GC-rich, with a higher number of CpG islands relative to GpC islands, a characteristic linked to regions of transcriptional activity in vertebrates, which may be relevant to gene expression (53-55). Third, several putative transcription factor-binding sites are present in AAVS1, including CREB, AP-1, AP-2 and Sp1 (48,56). Fourth, topoisomerase I is predicted to cleave at least two sites within AAVS1(48). Fifth, Lamartina et al (56) demonstrated an open chromatin conformation of AAVS1, accompanied by promoter activity on chromosome 19 in cultured cells. All these features position AAVS1 as an approachable region, subject to regulation by both host and viral regulatory elements.

The potential of the AAVS1 site as a target for gene therapy applications has been investigated and well-characterized. This site is known for its ability to integrate exogenous DNA with limited disruption to endogenous genes, providing a preferred site for stable transgene expression (57). A key feature that facilitates the usage of the AAVS1 site in gene therapy is its open chromatin structure, which is accompanied by endogenous insulator elements that protect the integrated DNA sequences from trans-activation or repression (58).

The insertion of exogenous genes into the human genome is conducted mainly using lentiviral and gamma-retroviral vectors. However, these vectors tend to integrate randomly within the genome (59-61), which can lead to unpredictable interactions between the transgene and the host genome. Such random integration may result in the attenuation or complete silencing of the transgene (62-64) or, more critically, in the dysregulation of host gene expression (64,65), which poses significant risks in gene therapy application. One promising strategy to mitigate these risks involves the use of gene-editing tools that enable site-specific insertion of transgenes into genomic safe harbors (GSHs) (64,66,67). GSHs are genomic locus that support a stable environment for transgene expression without interfering the integrity of endogenous genes (64,68,69). The AAVS1 site is considered a GSH that provides a stable transgene expression (70) due to the presence of flanking insulator regions (58). According to the study by Lombardo et al (66), the GSH AAVS1 can support transgene expression across different human cell types, facilitated by the active and open chromatin structure of the PPP1R12C gene located in this region (66).

The regulatory role of lncRNAs has been well-characterized. Numerous studies have detected that lncRNAs function as crucial regulators of gene expression, participating in various pathways by acting as decoys, guides and scaffolds for molecular interactions. In the present study, it was hypothesized that the regulatory roles of lncRNAs are indisputable, and to investigate this hypothesis, an in-house pipeline was developed to analyze a collection of lncRNAs. This pipeline enabled the identification of conserved motifs within their sequences that may contribute to their regulatory functions. The analysis revealed that one of the motifs may function as a sponge for a specific miRNA, thereby suppressing the function of the miRNA and leading to the upregulation of its mRNA targets. The function of miRNAs in the suppression of the translation of mRNA targets is well known, with implications for numerous pathological conditions, such as neurodegeneration (71-73), cancer (72,74,75) and cardiovascular diseases (74,76,77). Consequently, the interaction between lncRNAs and miRNAs could have significant consequences, either promoting disease prevention and treatment or exacerbating disease progression and severity.

Furthermore, all six motifs identified in the present study were detected across numerous regions of the human genome, with chromosome 19 presenting a markedly high frequency of motif occurrence relative to other chromosomes. Focusing on chromosome 19, it was found that these motifs were aligned with the AAVS1 regulatory region, a genomic locus known for its unique features, including the specific integration of the AAV, the presence of transcription factor binding sites, an AAV Rep-binding element, high GC-content, open chromatin, and two cleavage sites for topoisomerase I. It is noteworthy that the identified motifs demonstrate a pronounced GC content, a feature commonly associated with key regulatory regions, including transcription factor binding sites, polymerase binding sites, and loci involved in chromatin modification and DNA methylation. GC-rich sequences are often enriched at genomic sites that regulate transcription and chromatin remodeling due to their structural properties, such as enhanced stability and the presence of CpG sites that are key for epigenetic regulation (78-81). The existence of these motifs in regions integral to the regulation of the human genome, combined with their GC-rich nature, suggests their role in regulating gene expression through DNA methylation processes. Consequently, these motifs and the lncRNAs containing them, likely function as pivotal regulatory elements in gene expression.

In addition to the potential functions of lncRNAs that contain the identified motifs-based on the presence of these motif sequences in well-characterized regulatory regions of DNA and their interactions with other molecules, including miRNAs, the present study further investigated the documented roles of the lncRNAs containing at least one of these motifs in the literature (82-124) and relevant databases such as NCBI (ncbi.nlm.nih.gov) and GeneCards (125). Despite the incomplete annotation and characterization of these lncRNAs, the majority of the examined lncRNAs are located either within intergenic space, introns of coding genes, or are antisense to coding genes. These lncRNAs are believed to influence the functions of their associated genes, mainly by suppressing gene functions. The lncRNAs examined are involved in various biological processes, such as development, cellular proliferation, immune responses, inflammation, transmembrane transport, gene expression regulation, chromatin and chromosome structure modulation, differentiation, cell cycle control, spermatogenesis, metabolic regulation, and several functions related to the nervous and cardiovascular systems. The results from the present study, as depicted in the diagrams shown in Fig. S25, Fig. S26, Fig. S27, Fig. S28, Fig. S29 and Fig. S30, illustrate the involvement of lncRNAs with each specific motif in these diverse biological pathways.

The emergence of NRs dates back >1.5 billion years, originating in early eukaryotic organisms as ligand-dependent transcription factors. These receptors were responsive to environmental cues such as hormones, light, and metabolic shifts. The earliest NRs likely played a critical role in regulating gene expression, helping primitive eukaryotes adapt to dynamic external conditions, thus aiding their survival and proliferation. Of note, unicellular organisms such as Euglena gracilis, a photosynthetic protist, contain an ancestral version of the NR, providing evidence that these mechanisms were present well before the evolution of multicellular organisms. This discovery enhances the understanding of the origins of NRs and their evolutionary significance (126).

As evolution progressed, NRs diversified into a complex protein family with distinct functions. Different subfamilies evolved to regulate a wide variety of biological processes across animals, plants, fungi and even bacteria. In modern organisms, NRs govern critical processes such as metabolism, immune response, reproduction, and development. The diversity of these receptors is reflected in the large number of subfamilies, including steroid hormone receptors, thyroid hormone receptors, peroxisome proliferator-activated receptors and orphan NRs. Despite their varied roles, these receptors retain a conserved ligand-binding domain that allows them to interact with specific signaling molecules to regulate gene expression (126).

The present study discovered six novel sequence motifs within the lncRNA sequences under investigation. These lncRNAs exhibited altered expression levels upon the activation of NRs, either increasing or decreasing in abundance. Notably, several of these lncRNAs, which contain at least one of the identified motifs, are regulated by multiple NRs. This suggests that these motifs may be associated with NR function but do not show specificity to any single receptor, underscoring the complexity of the regulatory network between NRs and ncRNAs. This finding highlights the importance of further research to explore their broader biological implications.

To investigate the evolutionary conservation of these motifs, the FIMO tool was applied to examine their presence in various organisms. The present study analyzed species such as Mus musculus, Squamata (lizard), Danio rerio (zebrafish), Drosophila melanogaster, Caenorhabditis elegans, Escherichia coli (K12 MG1655), Saccharomyces cerevisiae and Bacillus subtilis (PY79). Additionally, the present study explored the presence of these motifs in the genome of Euglena gracilis using the ‘Matcher’ tool on the Galaxy platform. A phylogenetic tree was subsequently constructed using the iTOL tool (Interactive Tree Of Life), which enabled the visualization and annotation of the evolutionary associations among the organisms and the conservation of the motifs across species (127). The resulting phylogenetic tree (Fig. 7) clearly illustrates the evolutionary conservation of these motifs, which were identified repeatedly in the studied organisms (Table II).

Figure 7

Phylogenetic tree representing the species: Homo sapiens, Mus musculus, Squamata (lizard), Danio rerio (zebrafish), Drosophila melanogaster, Caenorhabditis elegans, Escherichia coli (K12 MG1655), Saccharomyces cerevisiae, Bacillus subtilis (PY79) and Euglena gracilis.

Table II

Occurrence frequency of the motifs in the genome sequences of the species Mus musculus, Squamata (lizard), Danio rerio (zebrafish), Drosophila melanogaster, Caenorhabditis elegans, Escherichia coli, Saccharomyces cerevisiae, Bacillus subtilis (PY79) and Euglena gracilis.

Table II

Occurrence frequency of the motifs in the genome sequences of the species Mus musculus, Squamata (lizard), Danio rerio (zebrafish), Drosophila melanogaster, Caenorhabditis elegans, Escherichia coli, Saccharomyces cerevisiae, Bacillus subtilis (PY79) and Euglena gracilis.

SpeciesMotif 1Motif 2Motif 3Motif 4Motif 5Motif 6
Mus musculus61,75583,48456,87064,32581,20781,748
Squamata (lizard)50,26753,38298,62652,82251,30859,874
Danio rerio66,23891,87054,89691,91097,17142,731
Drosophila melanogaster41,65835,39729,32033,96044,61060,662
Caenorhabditis elegans42,98220,44621,76829,07627,32140,169
Escherichia coli9036826446791269760
Saccharomyces cerevisiae3,0602,3863,4163,1103,7705,483
Bacillus subtilis912663662638865830
Euglena gracilis3641,6571,2785415221,711

The genetic similarities between humans and the other organisms examined in the present study exhibit significant variation. For instance, humans and Mus musculus (mouse) share ~85% genetic similarity, reflecting a divergence of ~70 million years ago, making the mouse an essential model for human genetic research, particularly in disease mechanisms and therapeutic development (128). Squamata, a diverse group of reptiles, includes species that serve as critical models for studying thermoregulation, behavior and reproductive strategies. Research on lizards and snakes has provided important insights into immunity, skin regeneration, and other physiological processes crucial for survival in varied environments (129). The genetic similarity between humans and different Squamata species varies considerably, with lizards sharing ~70% genetic similarity with humans, diverging ~250 million years ago. The six motifs identified in the present study were also present in Squamata, suggesting their ancient evolutionary origins. Danio rerio (zebrafish), with a genetic similarity to humans of ~70%, diverged ~400 million years ago and is an critical model organism for developmental biology and gene function studies (130). Drosophila melanogaster (fruit fly), sharing ~60% of its genes with humans and diverging ~600 million years ago, is extensively used in genetic regulation, neurobiology, and cellular research (131). Caenorhabditis elegans (nematode), with ~40% genetic similarity to humans, has been instrumental in research on aging, developmental biology and cellular processes (132). Escherichia coli K12 MG1655, although lacking direct gene homologs in humans, plays a crucial role in understanding fundamental gene regulation and bacterial genetics (133). Saccharomyces cerevisiae (yeast) shares ~23% of its genes with humans and diverged from humans >1 billion years ago, rendering it a key organism for cell biology, genomics and metabolism research (134). Bacillus subtilis PY79, a soil bacterium, has markedly contributed to the understanding of bacterial gene regulation and stress responses (135). Finally, Euglena gracilis, a single-celled eukaryote, is considered to share ancient NR mechanisms with humans, offering insights into the early evolution of eukaryotic gene regulation (136).

The presence of the discovered motifs in species that diverged long before humans, including organisms with distinct genomes, such as Euglena gracilis and Saccharomyces cerevisiae, combined with the lack of specificity in these motifs, suggests they may represent ancestral binding sites for the precursors of NRs. The conservation of these motifs across a broad evolutionary spectrum emphasizes their potential role in regulating gene expression via NR binding. Given that these motifs do not exhibit specificity for any particular NR, it was hypothesized that they may have functioned as general binding sites for ancestral NR precursors (137). Moreover, these motifs play a crucial role in the evolution of NR function by enabling NRs to interact with a broader range of binding sites, facilitating gene regulation in response to environmental and internal cues (126,138).

The widespread presence of these motifs across different evolutionary lineages points to their ancient origins, likely predating the highly specific NRs found in modern organisms. In early eukaryotes, these motifs could have functioned as genomic binding sites for precursor forms of NRs, which were less specific in their binding preferences compared to the more specialized NRs currently observed (139). This generalization would have been essential in the early stages of NR evolution, allowing for flexible and robust gene regulation. These early, versatile genomic binding sites could have allowed primordial NR-like proteins to modulate gene expression effectively, even in the absence of the highly refined receptor-genome interactions seen in contemporary systems.

The conservation of these motifs across such diverse species also suggests that they may have functioned as backup mechanisms, ensuring the continued regulation of essential genes when canonical NR binding sites, such as the glucocorticoid response element for GR, were unavailable or dysfunctional (139). In this sense, these motifs may have served as a safety net, allowing the NR system to maintain gene expression and cellular functions even in the absence of optimal binding site availability. This backup role would have been particularly important in the early evolutionary stages, when the molecular machinery for precise gene regulation was still evolving (140).

As NRs became more specialized through evolution, the ancestral motifs likely persisted due to their role in maintaining regulatory flexibility. In modern organisms, these motifs may still contribute to the robustness of gene regulation, particularly in cases where the canonical NR-binding sites are compromised or unable to function. Their ability to interact with multiple NRs enhances the adaptability of the organism to different signaling environments, providing a more versatile and resilient regulatory system (140).

The preservation of these motifs across such a broad evolutionary spectrum underscores their importance in the evolutionary trajectory of gene regulation. These motifs may represent a foundational aspect of NR functionality, offering a mechanism that allowed early organisms to adapt to changing environments by ensuring continued gene regulation, even when specific receptor-binding sites were not fully evolved. The fact that these motifs can still be found in modern organisms suggests that they continue to play a role in gene regulation, particularly in situations where canonical NR interactions are suboptimal or unavailable (126).

This broader, more flexible role in gene regulation may have allowed organisms to thrive in variable environments and adapt to new challenges. The conservation of motifs across both ancient and modern species highlights their functional importance and their potential as key elements in maintaining the adaptability of the NR signaling system throughout evolutionary history.

To date, numerous studies have examined and described the potential function of lncRNAs in the regulation of gene expression and their abilities to act as biomarkers or as pharmaceutical targets, or even therapeutic molecules (141-144). In the present study, six conserved motifs were detected for the first time in the sequences of the lncRNAs studied which are suggested to participate in the regulatory function of lncRNAs that contain them. However, to fully understand the role of these motifs, and the lncRNAs containing them, in gene regulation, future research is required to explore their functional significance in greater detail. Experimental studies could confirm whether these motifs act as alternative binding sites for NRs or other transcription factors. Additionally, further investigations into the mechanisms through which these motifs contribute to the regulation of gene expression in both ancient and modern species are warranted to provide valuable insight into the evolution of NR-mediated gene regulation and its ongoing role in cellular adaptability.

Supplementary Material

The first discovered motif in the upregulated lncRNA subgroup.
The second discovered motif in the upregulated lncRNA subgroup.
The third discovered motif in the upregulated lncRNA subgroup.
The first discovered motif in the downregulated lncRNA subgroup.
The second discovered motif in the downregulated lncRNA subgroup.
The third discovered motif in the downregulated lncRNA subgroup.
Human karyotype in which the percentage of the occurrence of motif 1 in each chromosome is presented.
Human karyotype in which the percentage of the occurrence of motif 2 in each chromosome is presented.
Human karyotype in which the percentage of the occurrence of motif 3 in each chromosome is presented.
Human karyotype in which the percentage of the occurrence of motif 4 in each chromosome is presented.
Human karyotype in which the percentage of the occurrence of motif 5 in each chromosome is presented.
Human karyotype in which the percentage of the occurrence of motif 6 in each chromosome is presented.
Chromosome plot for the 1,000 higher-scored loci of motif 1 in the human genome which are presented as red dots next to the corresponding chromosomal region where they are located.
Chromosome plot for the 1,000 higher-scored loci of motif 2 in the human genome which are presented as red dots next to the corresponding chromosomal region where they are located.
Chromosome plot for the 1,000 higher-scored loci of motif 3 in the human genome which are presented as red dots next to the corresponding chromosomal region where they are located.
Chromosome plot for the 1,000 higher-scored loci of motif 4 in the human genome which are presented as red dots next to the corresponding chromosomal region where they are located.
Chromosome plot for the 1,000 higher-scored loci of motif 5 in the human genome which are presented as red dots next to the corresponding chromosomal region where they are located.
Chromosome plot for the 1,000 higher-scored loci of motif 6 in the human genome which are presented as red dots next to the corresponding chromosomal region where they are located.
Multiple alignment of the sequences of motif 1 with AAVS1 site using Clustal Omega (51) visualized with JalView (52).
Multiple alignment of the sequences of motif 2 with AAVS1 site using Clustal Omega (51) visualized with JalView (52).
Multiple alignment of the sequences of motif 3 with AAVS1 site using Clustal Omega (51) visualized with JalView (52).
Multiple alignment of the sequences of motif 4 with AAVS1 site using Clustal Omega (51) visualized with JalView (52).
Multiple alignment of the sequences of motif 5 with AAVS1 site using Clustal Omega (51) visualized with JalView (52).
Multiple alignment of the sequences of motif 6 with AAVS1 site using Clustal Omega (51) visualized with JalView (52).
The biological pathways in which the lncRNAs that contain the motif 1 participate and have a regulatory function, according to GeneCards (https://www.genecards.org/) and NCBI Gene (https://www.ncbi.nlm.nih.gov/gene/) databases.
The biological pathways in which the lncRNAs that contain the motif 2 participate and have a regulatory function, according to GeneCards (https://www.genecards.org/) and NCBI Gene (https://www.ncbi.nlm.nih.gov/gene/) databases.
The biological pathways in which the lncRNAs that contain the motif 3 participate and have a regulatory function, according to GeneCards (https://www.genecards.org/) and NCBI Gene (https://www.ncbi.nlm.nih.gov/gene/) databases.
The biological pathways in which the lncRNAs that contain the motif 4 participate and have a regulatory function, according to GeneCards (https://www.genecards.org/) and NCBI Gene (https://www.ncbi.nlm.nih.gov/gene/) databases.
The biological pathways in which the lncRNAs that contain the motif 5 participate and have a regulatory function, according to GeneCards (https://www.genecards.org/) and NCBI Gene (https://www.ncbi.nlm.nih.gov/gene/) databases.
The biological pathways in which the lncRNAs that contain the motif 6 participate and have a regulatory function, according to GeneCards (https://www.genecards.org/) and NCBI Gene (https://www.ncbi.nlm.nih.gov/gene/) databases.
Data S1
Data S2
Data S1
The dataset of the lncRNAs that are either upregulated or downregulated following the activation of an NR.
List of 487 transcripts which are targets of hsa-miR-1908-3p according to the TargetScanS (Release 8.0) database.

Acknowledgements

Not applicable.

Funding

Funding: The authors would like to acknowledge funding from the following organizations: i) AdjustEBOVGP-Dx (RIA2018EF-2081): Biochemical Adjustments of native EBOV Glycoprotein in Patient Sample to Unmask target Epitopes for Rapid Diagnostic Testing. A European and Developing Countries Clinical Trials Partnership (EDCTP2) under the Horizon 2020 ‘Research and Innovation Actions’ DESCA; and ii) ‘MilkSafe: A novel pipeline to enrich formula milk using omics technologies’, a research co-financed by the European Regional Development Fund of the European Union and Greek national funds through the Operational Program Competitiveness, Entrepreneurship and Innovation, under the call RESEARCH-CREATE-INNOVATE (project code: T2EDK-02222).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

DV conceived the study. DV, GPC, EE and KP contributed to the overall study design. KP and LP developed the in-house algorithms in MATLAB. KP, EE and DV performed and evaluated the analysis using bioinformatics tools, as well as the statistical analysis. KP, LP, GPC, EE and DV wrote, drafted and critically revised the manuscript. DV, LP and KP confirm the authenticity of all the raw data. KP, LP, GPC, EE and DV wrote, drafted, revised, edited and reviewed the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

GPC is the Editor in Chief of the journal, and DV and EE are Editors of the journal. However, they had no personal involvement in the reviewing process, or any influence in terms of adjudicating on the final decision, for this article. The other authors declare that they have no competing interests.

References

1 

Carninci P, Kasukawa T, Katayama S, Gough J, Frith MC, Maeda N, Oyama R, Ravasi T, Lenhard B, Wells C, et al: The transcriptional landscape of the mammalian genome. Science. 309:1559–1563. 2005.PubMed/NCBI View Article : Google Scholar

2 

Mattick JS: RNA regulation: A new genetics? Nat Rev Genet. 5:316–323. 2004.PubMed/NCBI View Article : Google Scholar

3 

Frith MC, Pheasant M and Mattick JS: The amazing complexity of the human transcriptome. Eur J Hum Genet. 13:894–897. 2005.PubMed/NCBI View Article : Google Scholar

4 

Kaikkonen MU, Lam MTY and Glass CK: Non-coding RNAs as regulators of gene expression and epigenetics. Cardiovasc Res. 90:430–440. 2011.PubMed/NCBI View Article : Google Scholar

5 

Harrow J, Frankish A, Gonzalez JM, Tapanari E, Diekhans M, Kokocinski F, Aken BL, Barrell D, Zadissa A, Searle S, et al: GENCODE: The reference human genome annotation for The ENCODE project. Genome Res. 22:1760–1774. 2012.PubMed/NCBI View Article : Google Scholar

6 

Salviano-Silva A, Lobo-Alves SC, Almeida RC, Malheiros D and Petzl-Erler ML: Besides pathology: Long non-coding RNA in cell and tissue homeostasis. Noncoding RNA. 4(3)2018.PubMed/NCBI View Article : Google Scholar

7 

Ma L, Bajic VB and Zhang Z: On the classification of long non-coding RNAs. RNA Biol. 10:925–933. 2013.PubMed/NCBI View Article : Google Scholar

8 

Wang KC and Chang HY: Molecular mechanisms of long noncoding RNAs. Mol Cell. 43:904–914. 2011.PubMed/NCBI View Article : Google Scholar

9 

Hombach S and Kretz M: Non-coding RNAs: Classification, biology and functioning. Adv Exp Med Biol. 937:3–17. 2016.PubMed/NCBI View Article : Google Scholar

10 

Rinn JL and Chang HY: Genome regulation by long noncoding RNAs. Annu Rev Biochem. 81:145–166. 2012.PubMed/NCBI View Article : Google Scholar

11 

Francis GA, Fayard E, Picard F and Auwerx J: Nuclear receptors and the control of metabolism. Annu Rev Physiol. 65:261–311. 2003.PubMed/NCBI View Article : Google Scholar

12 

Olivares AM, Moreno-Ramos OA and Haider NB: Role of nuclear receptors in central nervous system development and associated diseases. J Exp Neurosci. 9 (Suppl 2):S93–S121. 2016.PubMed/NCBI View Article : Google Scholar

13 

Foulds CE, Panigrahi AK, Coarfa C, Lanz RB and O'Malley BW: Long noncoding RNAs as targets and regulators of nuclear receptors. Curr Top Microbiol Immunol. 394:143–176. 2016.PubMed/NCBI View Article : Google Scholar

14 

Volders PJ, Helsens K, Wang X, Menten B, Martens L, Gevaert K, Vandesompele J and Mestdagh P: LNCipedia: A database for annotated human lncRNA transcript sequences and structures. Nucleic Acids Res. 41 (Database Issue):D246–D251. 2013.PubMed/NCBI View Article : Google Scholar

15 

Bailey TL, Williams N, Misleh C and Li WW: MEME: Discovering and analyzing DNA and protein sequence motifs. Nucleic Acids Res. 34 (Web Server Issue):W369–W373. 2006.PubMed/NCBI View Article : Google Scholar

16 

Liu W and Wang X: Prediction of functional microRNA targets by integrative modeling of microRNA binding and target expression data. Genome Biol. 20(18)2019.PubMed/NCBI View Article : Google Scholar

17 

Agarwal V, Bell GW, Nam JW and Bartel DP: Predicting effective microRNA target sites in mammalian mRNAs. Elife. 4(e05005)2015.PubMed/NCBI View Article : Google Scholar

18 

Garcia DM, Baek D, Shin C, Bell GW, Grimson A and Bartel DP: Weak seed-pairing stability and high target-site abundance decrease the proficiency of lsy-6 and other microRNAs. Nat Struct Mol Biol. 18:1139–1146. 2011.PubMed/NCBI View Article : Google Scholar

19 

Grimson A, Farh KK, Johnston WK, Garrett-Engele P, Lim LP and Bartel DP: MicroRNA targeting specificity in mammals: Determinants beyond seed pairing. Mol Cell. 27:91–105. 2007.PubMed/NCBI View Article : Google Scholar

20 

Goecks J, Nekrutenko A and Taylor J: Galaxy Team. Galaxy: A comprehensive approach for supporting accessible, reproducible, and transparent computational research in the life sciences. Genome Biol. 11(R86)2010.PubMed/NCBI View Article : Google Scholar

21 

Afgan E, Baker D, van den Beek M, Blankenberg D, Bouvier D, Čech M, Chilton J, Clements D, Coraor N, Eberhard C, et al: The Galaxy platform for accessible, reproducible and collaborative biomedical analyses: 2016 update. Nucleic Acids Res. 44 (W1):W3–W10. 2016.PubMed/NCBI View Article : Google Scholar

22 

Durbin R, Eddy SR, Krogh A and Mitchison G: Biological sequence analysis: Probabilistic Models of Proteins and Nucleic Acids. Cambridge University Press, 1998.

23 

Ha M and Kim VN: Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol. 15:509–524. 2014.PubMed/NCBI View Article : Google Scholar

24 

Broughton JP, Lovci MT, Huang JL, Yeo GW and Pasquinelli AE: Pairing beyond the seed supports microRNA targeting specificity. Mol Cell. 64:320–333. 2016.PubMed/NCBI View Article : Google Scholar

25 

John B, Enright AJ, Aravin A, Tuschl T, Sander C and Marks DS: Human MicroRNA targets. PLoS Biol. 2(e363)2004.PubMed/NCBI View Article : Google Scholar

26 

Betel D, Koppal A, Agius P, Sander C and Leslie C: Comprehensive modeling of microRNA targets predicts functional non-conserved and non-canonical sites. Genome Biol. 11(R90)2010.PubMed/NCBI View Article : Google Scholar

27 

Krek A, Grün D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, MacMenamin P, da Piedade I, Gunsalus KC, Stoffel M and Rajewsky N: Combinatorial microRNA target predictions. Nat Genet. 37:495–500. 2005.PubMed/NCBI View Article : Google Scholar

28 

Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP and Burge CB: Prediction of mammalian microRNA targets. Cell. 115:787–798. 2003.PubMed/NCBI View Article : Google Scholar

29 

Lewis BP, Burge CB and Bartel DP: Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 120:15–20. 2005.PubMed/NCBI View Article : Google Scholar

30 

Miranda KC, Huynh T, Tay Y, Ang YS, Tam WL, Thomson AM, Lim B and Rigoutsos I: A pattern-based method for the identification of MicroRNA binding sites and their corresponding heteroduplexes. Cell. 126:1203–1217. 2006.PubMed/NCBI View Article : Google Scholar

31 

Kertesz M, Iovino N, Unnerstall U, Gaul U and Segal E: The role of site accessibility in microRNA target recognition. Nat Genet. 39:1278–1284. 2007.PubMed/NCBI View Article : Google Scholar

32 

Rehmsmeier M, Steffen P, Hochsmann M and Giegerich R: Fast and effective prediction of microRNA/target duplexes. RNA. 10:1507–1517. 2004.PubMed/NCBI View Article : Google Scholar

33 

Maragkakis M, Alexiou P, Papadopoulos GL, Reczko M, Dalamagas T, Giannopoulos G, Goumas G, Koukis E, Kourtis K, Simossis VA, et al: Accurate microRNA target prediction correlates with protein repression levels. BMC Bioinformatics. 10(295)2009.PubMed/NCBI View Article : Google Scholar

34 

Karagkouni D, Paraskevopoulou MD, Chatzopoulos S, Vlachos IS, Tastsoglou S, Kanellos I, Papadimitriou D, Kavakiotis I, Maniou S, Skoufos G, et al: DIANA-TarBase v8: A decade-long collection of experimentally supported miRNA-gene interactions. Nucleic Acids Res. 46 (D1):D239–D245. 2018.PubMed/NCBI View Article : Google Scholar

35 

McGeary SE, Lin KS, Shi CY, Pham TM, Bisaria N, Kelley GM and Bartel DP: The biochemical basis of microRNA targeting efficacy. Science. 366(eaav1741)2019.PubMed/NCBI View Article : Google Scholar

36 

Yang L, Shi CM, Chen L, Pang LX, Xu GF, Gu N, Zhu LJ, Guo XR, Ni YH and Ji CB: The biological effects of hsa-miR-1908 in human adipocytes. Mol Biol Rep. 42:927–935. 2015.PubMed/NCBI View Article : Google Scholar

37 

Kuang Q, Li J, You L, Shi C, Ji C, Guo X, Xu M and Ni Y: Identification and characterization of NF-kappaB binding sites in human miR-1908 promoter. Biomed Pharmacother. 74:158–163. 2015.PubMed/NCBI View Article : Google Scholar

38 

Bar M, Wyman SK, Fritz BR, Qi J, Garg KS, Parkin RK, Kroh EM, Bendoraite A, Mitchell PS, Nelson AM, et al: MicroRNA discovery and profiling in human embryonic stem cells by deep sequencing of small RNA libraries. Stem Cells. 26:2496–2505. 2008.PubMed/NCBI View Article : Google Scholar

39 

Pencheva N, Tran H, Buss C, Huh D, Drobnjak M, Busam K and Tavazoie SF: Convergent multi-miRNA targeting of ApoE drives LRP1/LRP8-dependent melanoma metastasis and angiogenesis. Cell. 151:1068–1082. 2012.PubMed/NCBI View Article : Google Scholar

40 

Xia X, Li Y, Wang W, Tang F, Tan J, Sun L, Li Q, Sun L, Tang B and He S: MicroRNA-1908 functions as a glioblastoma oncogene by suppressing PTEN tumor suppressor pathway. Mol Cancer. 14(154)2015.PubMed/NCBI View Article : Google Scholar

41 

Chai Z, Fan H, Li Y, Song L, Jin X, Yu J, Li Y, Ma C and Zhou R: miR-1908 as a novel prognosis marker of glioma via promoting malignant phenotype and modulating SPRY4/RAF1 axis. Oncol Rep. 38:2717–2726. 2017.PubMed/NCBI View Article : Google Scholar

42 

Jiang X, Yang L, Pang L, Chen L, Guo X, Ji C, Shi C and Ni Y: Expression of obesity-related miR-1908 in human adipocytes is regulated by adipokines, free fatty acids and hormones. Mol Med Rep. 10:1164–1169. 2014.PubMed/NCBI View Article : Google Scholar

43 

Grimwood J, Gordon LA, Olsen A, Terry A, Schmutz J, Lamerdin J, Hellsten U, Goodstein D, Couronne O, Tran-Gyamfi M, et al: The DNA sequence and biology of human chromosome 19. Nature. 428:529–535. 2004.PubMed/NCBI View Article : Google Scholar

44 

Kumar S, Stecher G, Suleski M and Hedges SB: TimeTree: A resource for timelines, timetrees, and divergence times. Mol Biol Evol. 34:1812–1819. 2017.PubMed/NCBI View Article : Google Scholar

45 

Kim SH, Elango N, Warden C, Vigoda E and Yi SV: Heterogeneous genomic molecular clocks in primates. PLoS Genet. 2(e163)2006.PubMed/NCBI View Article : Google Scholar

46 

Kotin RM, Menninger JC, Ward DC and Berns KI: Mapping and direct visualization of a region-specific viral DNA integration site on chromosome 19q13-qter. Genomics. 10:831–834. 1991.PubMed/NCBI View Article : Google Scholar

47 

Samulski RJ, Zhu X, Xiao X, Brook JD, Housman DE, Epstein N and Hunter LA: Targeted integration of adeno-associated virus (AAV) into human chromosome 19. EMBO J. 10:3941–3950. 1991.PubMed/NCBI View Article : Google Scholar

48 

Kotin RM, Linden RM and Berns KI: Characterization of a preferred site on human chromosome 19q for integration of adeno-associated virus DNA by non-homologous recombination. EMBO J. 11:5071–5078. 1992.PubMed/NCBI View Article : Google Scholar

49 

Aslanidis C, Jansen G, Amemiya C, Shutler G, Mahadevan M, Tsilfidis C, Chen C, Alleman J, Wormskamp NG, Vooijs M, et al: Cloning of the essential myotonic dystrophy region and mapping of the putative defect. Nature. 355:548–551. 1992.PubMed/NCBI View Article : Google Scholar

50 

Feichtinger W and Schmid M: Increased frequencies of sister chromatid exchanges at common fragile sites (1)(q42) and (19)(q13). Hum Genet. 83:145–147. 1989.PubMed/NCBI View Article : Google Scholar

51 

Sievers F and Higgins DG: Clustal Omega for making accurate alignments of many protein sequences. Protein Sci. 27:135–145. 2018.PubMed/NCBI View Article : Google Scholar

52 

Waterhouse AM, Procter JB, Martin DMA, Clamp M and Barton GJ: Jalview Version 2-a multiple sequence alignment editor and analysis workbench. Bioinformatics. 25:1189–1191. 2009.PubMed/NCBI View Article : Google Scholar

53 

Deaton AM and Bird A: CpG islands and the regulation of transcription. Genes Dev. 25:1010–1022. 2011.PubMed/NCBI View Article : Google Scholar

54 

Larsen F, Gundersen G, Lopez R and Prydz H: CpG islands as gene markers in the human genome. Genomics. 13:1095–1107. 1992.PubMed/NCBI View Article : Google Scholar

55 

Yamashita R, Suzuki Y, Sugano S and Nakai K: Genome-wide analysis reveals strong correlation between CpG islands with nearby transcription start sites of genes and their tissue specificity. Gene. 350:129–136. 2005.PubMed/NCBI View Article : Google Scholar

56 

Lamartina S, Sporeno E, Fattori E and Toniatti C: Characteristics of the adeno-associated virus preintegration site in human chromosome 19: Open chromatin conformation and transcription-competent environment. J Virol. 74:7671–7677. 2000.PubMed/NCBI View Article : Google Scholar

57 

Bhagwan JR, Collins E, Mosqueira D, Bakar M, Johnson BB, Thompson A, Smith JGW and Denning C: Variable expression and silencing of CRISPR-Cas9 targeted transgenes identifies the AAVS1 locus as not an entirely safe harbour. F1000Res. 8(1911)2019.PubMed/NCBI View Article : Google Scholar

58 

Ogata T, Kozuka T and Kanda T: Identification of an insulator in AAVS1, a preferred region for integration of adeno-associated virus DNA. J Virol. 77:9000–9007. 2003.PubMed/NCBI View Article : Google Scholar

59 

Gaspar HB, Cooray S, Gilmour KC, Parsley KL, Zhang F, Adams S, Bjorkegren E, Bayford J, Brown L, Davies EG, et al: Hematopoietic stem cell gene therapy for adenosine deaminase-deficient severe combined immunodeficiency leads to long-term immunological recovery and metabolic correction. Sci Transl Med. 3(97ra80)2011.PubMed/NCBI View Article : Google Scholar

60 

Cartier N, Hacein-Bey-Abina S, Bartholomae CC, Veres G, Schmidt M, Kutschera I, Vidaud M, Abel U, Dal-Cortivo L, Caccavelli L, et al: Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy. Science. 326:818–823. 2009.PubMed/NCBI View Article : Google Scholar

61 

Hacein-Bey-Abina S, Pai SY, Gaspar HB, Armant M, Berry CC, Blanche S, Bleesing J, Blondeau J, de Boer H, Buckland KF, et al: A modified γ-retrovirus vector for X-linked severe combined immunodeficiency. N Engl J Med. 371:1407–1417. 2014.PubMed/NCBI View Article : Google Scholar

62 

Martin DIK and Whitelaw E: The vagaries of variegating transgenes. Bioessays. 18:919–923. 1996.PubMed/NCBI View Article : Google Scholar

63 

Bestor TH: Gene silencing as a threat to the success of gene therapy. J Clin Invest. 105:409–411. 2000.PubMed/NCBI View Article : Google Scholar

64 

Papapetrou EP and Schambach A: Gene insertion into genomic safe harbors for human gene therapy. Mol Ther. 24:678–684. 2016.PubMed/NCBI View Article : Google Scholar

65 

Wu C and Dunbar CE: Stem cell gene therapy: The risks of insertional mutagenesis and approaches to minimize genotoxicity. Front Med. 5:356–371. 2011.PubMed/NCBI View Article : Google Scholar

66 

Lombardo A, Cesana D, Genovese P, Di Stefano B, Provasi E, Colombo DF, Neri M, Magnani Z, Cantore A, Lo Riso P, et al: Site-specific integration and tailoring of cassette design for sustainable gene transfer. Nat Methods. 8:861–869. 2011.PubMed/NCBI View Article : Google Scholar

67 

Klatt D, Cheng E, Hoffmann D, Santilli G, Thrasher AJ, Brendel C and Schambach A: Differential transgene silencing of myeloid-specific promoters in the AAVS1 safe harbor locus of induced pluripotent stem cell-derived myeloid cells. Hum Gene Ther. 31:199–210. 2019.PubMed/NCBI View Article : Google Scholar

68 

Papapetrou EP, Lee G, Malani N, Setty M, Riviere I, Tirunagari LM, Kadota K, Roth SL, Giardina P, Viale A, et al: Genomic safe harbors permit high β-globin transgene expression in thalassemia induced pluripotent stem cells. Nat Biotechnol. 29:73–78. 2011.PubMed/NCBI View Article : Google Scholar

69 

Sadelain M, Papapetrou EP and Bushman FD: Safe harbours for the integration of new DNA in the human genome. Nat Rev Cancer. 12:51–58. 2011.PubMed/NCBI View Article : Google Scholar

70 

Smith SD, Morgan R, Gemmell R, Amylon MD, Link MP, Linker C, Hecht BK, Warnke R, Glader BE and Hecht F: Clinical and biologic characterization of T-cell neoplasias with rearrangements of chromosome 7 band q34. Blood. 71:395–402. 1988.PubMed/NCBI

71 

Maes OC, Chertkow HM, Wang E and Schipper HM: MicroRNA: Implications for alzheimer disease and other human CNS disorders. Curr Genomics. 10:154–168. 2009.PubMed/NCBI View Article : Google Scholar

72 

Garofalo M, Condorelli G and Croce CM: MicroRNAs in diseases and drug response. Curr Opin Pharmacol. 8:661–667. 2008.PubMed/NCBI View Article : Google Scholar

73 

Juźwik CA, S Drake S, Zhang Y, Paradis-Isler N, Sylvester A, Amar-Zifkin A, Douglas C, Morquette B, Moore CS and Fournier AE: microRNA dysregulation in neurodegenerative diseases: A systematic review. Prog Neurobiol. 182(101664)2019.PubMed/NCBI View Article : Google Scholar

74 

Venneri M and Passantino A: MiRNA: What clinicians need to know. Eur J Intern Med. 113:6–9. 2023.PubMed/NCBI View Article : Google Scholar

75 

Zhang B, Pan X, Cobb GP and Anderson TA: microRNAs as oncogenes and tumor suppressors. Dev Biol. 302:1–12. 2007.PubMed/NCBI View Article : Google Scholar

76 

Small EM and Olson EN: Pervasive roles of microRNAs in cardiovascular biology. Nature. 469:336–342. 2011.PubMed/NCBI View Article : Google Scholar

77 

Romaine SPR, Tomaszewski M, Condorelli G and Samani NJ: MicroRNAs in cardiovascular disease: An introduction for clinicians. Heart. 101:921–928. 2015.PubMed/NCBI View Article : Google Scholar

78 

Bailey TL and Elkan C: Fitting a mixture model by expectation maximization to discover motifs in biopolymers. Proc Int Conf Intell Syst Mol Biol. 2:28–36. 1994.PubMed/NCBI

79 

Tirosh I and Barkai N: Two strategies for gene regulation by promoter nucleosomes. Genome Res. 18:1084–1091. 2008.PubMed/NCBI View Article : Google Scholar

80 

Bird A: DNA methylation patterns and epigenetic memory. Genes Dev. 16:6–21. 2002.PubMed/NCBI View Article : Google Scholar

81 

Li E, Bestor TH and Jaenisch R: Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell. 69:915–926. 1992.PubMed/NCBI View Article : Google Scholar

82 

Ulitsky I, Shkumatava A, Jan CH, Sive H and Bartel DP: Conserved function of lincRNAs in vertebrate embryonic development despite rapid sequence evolution. Cell. 147:1537–1550. 2011.PubMed/NCBI View Article : Google Scholar

83 

Liu L, Wang Z, Jia J, Shi Y, Lian T and Han X: Linc01230, transcriptionally regulated by PPARγ, is identified as a novel modifier in endothelial function. Biochem Biophys Res Commun. 507:369–376. 2018.PubMed/NCBI View Article : Google Scholar

84 

Cunningham TJ, Kumar S, Yamaguchi TP and Duester G: Wnt8a and Wnt3a cooperate in the axial stem cell niche to promote mammalian body axis extension. Dev Dyn. 244:797–807. 2015.PubMed/NCBI View Article : Google Scholar

85 

Okubo Y, Sugawara T, Abe-Koduka N, Kanno J, Kimura A and Saga Y: Lfng regulates the synchronized oscillation of the mouse segmentation clock via trans-repression of Notch signalling. Nat Commun. 3(1141)2012.PubMed/NCBI View Article : Google Scholar

86 

Pai SG, Carneiro BA, Mota JM, Costa R, Leite CA, Barroso-Sousa R, Kaplan JB, Chae YK and Giles FJ: Wnt/beta-catenin pathway: Modulating anticancer immune response. J Hematol Oncol. 10(101)2017.PubMed/NCBI View Article : Google Scholar

87 

Amsellem V, Dryden NH, Martinelli R, Gavins F, Almagro LO, Birdsey GM, Haskard DO, Mason JC, Turowski P and Randi AM: ICAM-2 regulates vascular permeability and N-cadherin localization through ezrin-radixin-moesin (ERM) proteins and Rac-1 signalling. Cell Commun Signal. 12(12)2014.PubMed/NCBI View Article : Google Scholar

88 

Overton HA, Fyfe MCT and Reynet C: GPR119, a novel G protein-coupled receptor target for the treatment of type 2 diabetes and obesity. Br J Pharmacol. 153 (Suppl 1):S76–S81. 2008.PubMed/NCBI View Article : Google Scholar

89 

Shang R, Wang M, Dai B, Du J, Wang J, Liu Z, Qu S, Yang X, Liu J, Xia C, et al: Long noncoding RNA SLC2A1-AS1 regulates aerobic glycolysis and progression in hepatocellular carcinoma via inhibiting the STAT3/FOXM1/GLUT1 pathway. Mol Oncol. 14:1381–1396. 2020.PubMed/NCBI View Article : Google Scholar

90 

Kim JJ, Lee SB, Jang J, Yi SY, Kim SH, Han SA, Lee JM, Tong SY, Vincelette ND, Gao B, et al: WSB1 promotes tumor metastasis by inducing pVHL degradation. Genes Dev. 29:2244–2257. 2015.PubMed/NCBI View Article : Google Scholar

91 

Shao X, Zhao T, Xi L, Zhang Y, He J, Zeng J and Deng L: LINC00565 promotes the progression of colorectal cancer by upregulating EZH2. Oncol Lett. 21(53)2021.PubMed/NCBI View Article : Google Scholar

92 

Chen C, Feng Y, Wang J, Liang Y and Zou W: Long non-coding RNA SNHG15 in various cancers: A meta and bioinformatic analysis. BMC Cancer. 20(1156)2020.PubMed/NCBI View Article : Google Scholar

93 

Qian C, Li H, Chang D, Wei B and Wang Y: Identification of functional lncRNAs in atrial fibrillation by integrative analysis of the lncRNA-mRNA network based on competing endogenous RNAs hypothesis. J Cell Physiol. 234:11620–11630. 2019.PubMed/NCBI View Article : Google Scholar

94 

Liu J, Xu R, Mai SJ, Ma YS, Zhang MY, Cao PS, Weng NQ, Wang RQ, Cao D, Wei W, et al: LncRNA CSMD1-1 promotes the progression of hepatocellular carcinoma by activating MYC signaling. Theranostics. 10:7527–7544. 2020.PubMed/NCBI View Article : Google Scholar

95 

Talwar D and Hammer MF: SCN8A epilepsy, developmental encephalopathy, and related disorders. Pediatr Neurol. 122:76–83. 2021.PubMed/NCBI View Article : Google Scholar

96 

Chen XW, Feng YQ, Hao CJ, Guo XL, He X, Zhou ZY, Guo N, Huang HP, Xiong W, Zheng H, et al: DTNBP1, a schizophrenia susceptibility gene, affects kinetics of transmitter release. J Cell Biol. 181:791–801. 2008.PubMed/NCBI View Article : Google Scholar

97 

Chen CL, Ke Q, Luo M, Gao ZY, Li ZJ, Luo ZG and Liu DB: Loss of LINC01939 expression predicts progression and poor survival in gastric cancer. Pathol Res Pract. 214:1539–1543. 2018.PubMed/NCBI View Article : Google Scholar

98 

Dosil M and Bustelo XR: Functional characterization of Pwp2, a WD family protein essential for the assembly of the 90 S pre-ribosomal particle. J Biol Chem. 279:37385–37397. 2004.PubMed/NCBI View Article : Google Scholar

99 

Li G, Ruan X, Auerbach RK, Sandhu KS, Zheng M, Wang P, Poh HM, Goh Y, Lim J, Zhang J, et al: Extensive promoter-centered chromatin interactions provide a topological basis for transcription regulation. Cell. 148:84–98. 2012.PubMed/NCBI View Article : Google Scholar

100 

Li Z, Li X, Jian W, Xue Q and Liu Z: Roles of long non-coding RNAs in the development of chronic pain. Front Mol Neurosci. 14(760964)2021.PubMed/NCBI View Article : Google Scholar

101 

Luo L, Martin SC, Parkington J, Cadena SM, Zhu J, Ibebunjo C, Summermatter S, Londraville N, Patora-Komisarska K, Widler L, et al: HDAC4 controls muscle homeostasis through deacetylation of myosin heavy chain, PGC-1α, and Hsc70. Cell Rep. 29:749–763.e12. 2019.PubMed/NCBI View Article : Google Scholar

102 

Mortison JD, Schenone M, Myers JA, Zhang Z, Chen L, Ciarlo C, Comer E, Natchiar SK, Carr SA, Klaholz BP and Myers AG: Tetracyclines modify translation by targeting key human rRNA substructures. Cell Chem Biol. 25:1506–1518.e13. 2018.PubMed/NCBI View Article : Google Scholar

103 

Nelson JK, Koenis DS, Scheij S, Cook EC, Moeton M, Santos A, Lobaccaro JA, Baron S and Zelcer N: EEPD1 is a novel LXR target gene in macrophages which regulates ABCA1 abundance and cholesterol efflux. Arterioscler Thromb Vasc Biol. 37:423–432. 2017.PubMed/NCBI View Article : Google Scholar

104 

Zhang T, Xia W, Song X, Mao Q, Huang X, Chen B, Liang Y, Wang H, Chen Y, Yu X, et al: Super-enhancer hijacking LINC01977 promotes malignancy of early-stage lung adenocarcinoma addicted to the canonical TGF-β/SMAD3 pathway. J Hematol Oncol. 15(114)2022.PubMed/NCBI View Article : Google Scholar

105 

Zeng J, Sun W, Chang J, Yi D, Zhu L, Zhang Y, Pan X, Zhou Y, Lai M, Bian G, et al: HOXC4 up-regulates NF-κB signaling and promotes the cell proliferation to drive development of human hematopoiesis, especially CD43+ cells. Blood Sci. 2:117–128. 2020.PubMed/NCBI View Article : Google Scholar

106 

Gao Y, Wang F, Zhang L, Kang M, Zhu L, Xu L, Liang W and Zhang W: LINC00311 promotes cancer stem-like properties by targeting miR-330-5p/TLR4 pathway in human papillary thyroid cancer. Cancer Med. 9:1515–1528. 2020.PubMed/NCBI View Article : Google Scholar

107 

Flosbach M, Oberle SG, Scherer S, Zecha J, von Hoesslin M, Wiede F, Chennupati V, Cullen JG, List M, Pauling JK, et al: PTPN2 deficiency enhances programmed T cell expansion and survival capacity of activated T cells. Cell Rep. 32(107957)2020.PubMed/NCBI View Article : Google Scholar

108 

Li Z, Chao TC, Chang KY, Lin N, Patil VS, Shimizu C, Head SR, Burns JC and Rana TM: The long noncoding RNA THRIL regulates TNFα expression through its interaction with hnRNPL. Proc Natl Acad Sci USA. 111:1002–1007. 2014.PubMed/NCBI View Article : Google Scholar

109 

Xu LB, Bo BX, Xiong J, Ren YJ, Han D, Wei SH and Ren XP: Long non-coding RNA LINC00887 promotes progression of lung carcinoma by targeting the microRNA-206/NRP1 axis. Oncol Lett. 21(87)2021.PubMed/NCBI View Article : Google Scholar

110 

Byun S, Affolter KE, Snow AK, Curtin K, Cannon AR, Cannon-Albright LA, Thota R and Neklason DW: Differential methylation of G-protein coupled receptor signaling genes in gastrointestinal neuroendocrine tumors. Sci Rep. 11(12303)2021.PubMed/NCBI View Article : Google Scholar

111 

Fang Z, Zhong M, Zhou L, Le Y, Wang H and Fang Z: Low-density lipoprotein receptor-related protein 8 facilitates the proliferation and invasion of non-small cell lung cancer cells by regulating the Wnt/β-catenin signaling pathway. Bioengineered. 13:6807–6818. 2022.PubMed/NCBI View Article : Google Scholar

112 

Huang R, Liu J, Chen X, Zhi Y, Ding S, Ming J, Li Y, Wang Y and Na J: A long non-coding RNA LncSync regulates mouse cardiomyocyte homeostasis and cardiac hypertrophy through coordination of miRNA actions. Protein Cell. 14:153–157. 2023.PubMed/NCBI View Article : Google Scholar

113 

Li N and Zhan X and Zhan X: The lncRNA SNHG3 regulates energy metabolism of ovarian cancer by an analysis of mitochondrial proteomes. Gynecol Oncol. 150:343–354. 2018.PubMed/NCBI View Article : Google Scholar

114 

Zhen H, Du P, Yi Q, Tang X and Wang T: LINC00958 promotes bladder cancer carcinogenesis by targeting miR-490-3p and AURKA. BMC Cancer. 21(1145)2021.PubMed/NCBI View Article : Google Scholar

115 

Majumdar R, Bandyopadhyay A, Deng H and Maitra U: Phosphorylation of mammalian translation initiation factor 5 (eIF5) in vitro and in vivo. Nucleic Acids Res. 30:1154–1162. 2002.PubMed/NCBI View Article : Google Scholar

116 

Xu L, Wu Q, Yan H, Shu C, Fan W, Tong X and Li Q: Long noncoding RNA KB-1460A1.5 inhibits glioma tumorigenesis via miR-130a-3p/TSC1/mTOR/YY1 feedback loop. Cancer Lett. 525:33–45. 2022.PubMed/NCBI View Article : Google Scholar

117 

Wang F, Peters R, Jia J, Mudd M, Salemi M, Allers L, Javed R, Duque TLA, Paddar MA, Trosdal ES, et al: ATG5 provides host protection acting as a switch in the atg8ylation cascade between autophagy and secretion. Dev Cell. 58:866–884.e8. 2023.PubMed/NCBI View Article : Google Scholar

118 

Cruickshank BM, Wasson MD, Brown JM, Fernando W, Venkatesh J, Walker OL, Morales-Quintanilla F, Dahn ML, Vidovic D, Dean CA, et al: LncRNA PART1 promotes proliferation and migration, is associated with cancer stem cells, and alters the miRNA landscape in triple-negative breast cancer. Cancers (Basel). 13(2644)2021.PubMed/NCBI View Article : Google Scholar

119 

Vogler M: BCL2A1: The underdog in the BCL2 family. Cell Death Differ. 19:67–74. 2012.PubMed/NCBI View Article : Google Scholar

120 

Barriocanal M, Carnero E, Segura V and Fortes P: Long non-coding RNA BST2/BISPR is induced by IFN and regulates the expression of the antiviral factor tetherin. Front Immunol. 5(655)2015.PubMed/NCBI View Article : Google Scholar

121 

Mohamed Haroon M, Lakshmanan V, Sarkar SR, Lei K, Vemula PK and Palakodeti D: Mitochondrial state determines functionally divergent stem cell population in planaria. Stem Cell Reports. 16:1302–1316. 2021.PubMed/NCBI View Article : Google Scholar

122 

Zhu W, Zhou BL, Rong LJ, Ye L, Xu HJ, Zhou Y, Yan XJ, Liu WD, Zhu B, Wang L, et al: Roles of PTBP1 in alternative splicing, glycolysis, and oncogensis. J Zhejiang Univ Sci B. 21:122–136. 2020.PubMed/NCBI View Article : Google Scholar

123 

Lee MY, Sumpter R Jr, Zou Z, Sirasanagandla S, Wei Y, Mishra P, Rosewich H, Crane DI and Levine B: Peroxisomal protein PEX13 functions in selective autophagy. EMBO Rep. 18:48–60. 2017.PubMed/NCBI View Article : Google Scholar

124 

L'Abbate A, Tolomeo D, De Astis F, Lonoce A, Lo Cunsolo C, Mühlematter D, Schoumans J, Vandenberghe P, Van Hoof A, Palumbo O, et al: t(15;21) translocations leading to the concurrent downregulation of RUNX1 and its transcription factor partner genes SIN3A and TCF12 in myeloid disorders. Mol Cancer. 14(211)2015.PubMed/NCBI View Article : Google Scholar

125 

Stelzer G, Rosen N, Plaschkes I, Zimmerman S, Twik M, Fishilevich S, Stein TI, Nudel R, Lieder I, Mazor Y, et al: The GeneCards suite: From gene data mining to disease genome sequence analyses. Curr Protoc Bioinformatics. 54:1.30.1–1.30.33. 2016.PubMed/NCBI View Article : Google Scholar

126 

Laudet V: Evolution of the nuclear receptor superfamily: Early diversification from an ancestral orphan receptor. J Mol Endocrinol. 19:207–226. 1997.PubMed/NCBI View Article : Google Scholar

127 

Letunic I and Bork P: Interactive tree of life (iTOL) v4: Recent updates and new developments. Nucleic Acids Res. 47 (W1):W256–W259. 2019.PubMed/NCBI View Article : Google Scholar

128 

Mouse Genome Sequencing Consortium. Waterston RH, Lindblad-Toh K, Birney E, Rogers J, Abril JF, Agarwal P, Agarwala R, Ainscough R, Alexandersson M, et al: Initial sequencing and comparative analysis of the mouse genome. Nature. 420:520–562. 2002.PubMed/NCBI View Article : Google Scholar

129 

Castoe TA, de Koning APJ, Hall KT, Card DC, Schield DR, Fujita MK, Ruggiero RP, Degner JF, Daza JM, Gu W, et al: The Burmese python genome reveals the molecular basis for extreme adaptation in snakes. Proc Natl Acad Sci USA. 110:20645–20650. 2013.PubMed/NCBI View Article : Google Scholar

130 

Howe K, Clark MD, Torroja CF, Torrance J, Berthelot C, Muffato M, Collins JE, Humphray S, McLaren K, Matthews L, et al: The zebrafish reference genome sequence and its relationship to the human genome. Nature. 496:498–503. 2013.PubMed/NCBI View Article : Google Scholar

131 

Adams MD, Celniker SE, Holt RA, Evans CA, Gocayne JD, Amanatides PG, Scherer SE, Li PW, Hoskins RA, Galle RF, et al: The genome sequence of Drosophila melanogaster. Science. 287:2185–2195. 2000.PubMed/NCBI View Article : Google Scholar

132 

Hodgkin J, Plasterk RH and Waterston RH: The nematode Caenorhabditis elegans and its genome. Science. 270:410–414. 1995.PubMed/NCBI View Article : Google Scholar

133 

Blattner FR, Plunkett G III, Bloch CA, Perna NT, Burland V, Riley M, Collado-Vides J, Glasner JD, Rode CK, Mayhew GF, et al: The complete genome sequence of Escherichia coli K-12. Science. 277:1453–1462. 1997.PubMed/NCBI View Article : Google Scholar

134 

Weng S, Dong Q, Balakrishnan R, Christie K, Costanzo M, Dolinski K, Dwight SS, Engel S, Fisk DG, Hong E, et al: Saccharomyces genome database (SGD) provides biochemical and structural information for budding yeast proteins. Nucleic Acids Res. 31:216–218. 2003.PubMed/NCBI View Article : Google Scholar

135 

Petersohn A, Brigulla M, Haas S, Hoheisel JD, Völker U and Hecker M: Global analysis of the general stress response of Bacillus subtilis. J Bacteriol. 183:5617–5631. 2001.PubMed/NCBI View Article : Google Scholar

136 

Ebenezer TE, Carrington M, Lebert M, Kelly S and Field MC: Euglena gracilis genome and transcriptome: Organelles, nuclear genome assembly strategies and initial features. Adv Exp Med Biol. 979:125–140. 2017.PubMed/NCBI View Article : Google Scholar

137 

Owen GI and Zelent A: Origins and evolutionary diversification of the nuclear receptor superfamily. Cell Mol Life Sci. 57:809–827. 2000.PubMed/NCBI View Article : Google Scholar

138 

Holzer G, Markov GV and Laudet V: Evolution of nuclear receptors and ligand signaling: Toward a soft key-lock model? Curr Top Dev Biol. 125:1–38. 2017.PubMed/NCBI View Article : Google Scholar

139 

Penvose A, Keenan JL, Bray D, Ramlall V and Siggers T: Comprehensive study of nuclear receptor DNA binding provides a revised framework for understanding receptor specificity. Nat Commun. 10(2514)2019.PubMed/NCBI View Article : Google Scholar

140 

Cotnoir-White D, Laperrière D and Mader S: Evolution of the repertoire of nuclear receptor binding sites in genomes. Mol Cell Endocrinol. 334:76–82. 2011.PubMed/NCBI View Article : Google Scholar

141 

Hanly D, Esteller M and Berdasco M: Altered long non-coding RNA expression in cancer: Potential biomarkers and therapeutic targets? In: Chemical Epigenetics. Mai A (ed). Topics in Medicinal Chemistry. Vol. 33. Springer, Cham, pp401-428, 2019.

142 

Fu D, Shi Y, Liu JB, Wu TM, Jia CY, Yang HQ, Zhang DD, Yang XL, Wang HM and Ma YS: Targeting long non-coding RNA to therapeutically regulate gene expression in cancer. Mol Ther Nucleic Acids. 21:712–724. 2020.PubMed/NCBI View Article : Google Scholar

143 

Zhang L, Peng D, Sood AK, Dang CV and Zhong X: Shedding light on the dark cancer genomes: Long noncoding RNAs as novel biomarkers and potential therapeutic targets for cancer. Mol Cancer Ther. 17:1816–1823. 2018.PubMed/NCBI View Article : Google Scholar

144 

Fatima R, Akhade VS, Pal D and Rao SM: Long noncoding RNAs in development and cancer: Potential biomarkers and therapeutic targets. Mol Cell Ther. 3(5)2015.PubMed/NCBI View Article : Google Scholar

Related Articles

  • Abstract
  • View
  • Download
Copy and paste a formatted citation
Spandidos Publications style
Pierouli K, Papageorgiou L, Chrousos GP, Eliopoulos E and Vlachakis D: Role of lncRNAs related to NRs in the regulation of gene expression. Int J Epigen 5: 3, 2025.
APA
Pierouli, K., Papageorgiou, L., Chrousos, G.P., Eliopoulos, E., & Vlachakis, D. (2025). Role of lncRNAs related to NRs in the regulation of gene expression. International Journal of Epigenetics, 5, 3. https://doi.org/10.3892/ije.2025.26
MLA
Pierouli, K., Papageorgiou, L., Chrousos, G. P., Eliopoulos, E., Vlachakis, D."Role of lncRNAs related to NRs in the regulation of gene expression". International Journal of Epigenetics 5.1 (2025): 3.
Chicago
Pierouli, K., Papageorgiou, L., Chrousos, G. P., Eliopoulos, E., Vlachakis, D."Role of lncRNAs related to NRs in the regulation of gene expression". International Journal of Epigenetics 5, no. 1 (2025): 3. https://doi.org/10.3892/ije.2025.26
Copy and paste a formatted citation
x
Spandidos Publications style
Pierouli K, Papageorgiou L, Chrousos GP, Eliopoulos E and Vlachakis D: Role of lncRNAs related to NRs in the regulation of gene expression. Int J Epigen 5: 3, 2025.
APA
Pierouli, K., Papageorgiou, L., Chrousos, G.P., Eliopoulos, E., & Vlachakis, D. (2025). Role of lncRNAs related to NRs in the regulation of gene expression. International Journal of Epigenetics, 5, 3. https://doi.org/10.3892/ije.2025.26
MLA
Pierouli, K., Papageorgiou, L., Chrousos, G. P., Eliopoulos, E., Vlachakis, D."Role of lncRNAs related to NRs in the regulation of gene expression". International Journal of Epigenetics 5.1 (2025): 3.
Chicago
Pierouli, K., Papageorgiou, L., Chrousos, G. P., Eliopoulos, E., Vlachakis, D."Role of lncRNAs related to NRs in the regulation of gene expression". International Journal of Epigenetics 5, no. 1 (2025): 3. https://doi.org/10.3892/ije.2025.26
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team