[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (2,634)

Search Parameters:
Keywords = long non-coding RNAs

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
29 pages, 1061 KiB  
Review
Viroids and Retrozymes: Plant Circular RNAs Capable of Autonomous Replication
by Alexander A. Lezzhov, Anastasia K. Atabekova, Denis A. Chergintsev, Ekaterina A. Lazareva, Andrey G. Solovyev and Sergey Y. Morozov
Plants 2025, 14(1), 61; https://doi.org/10.3390/plants14010061 (registering DOI) - 27 Dec 2024
Abstract
Among the long non-coding RNAs that are currently recognized as important regulatory molecules influencing a plethora of processes in eukaryotic cells, circular RNAs (circRNAs) represent a distinct class of RNAs that are predominantly produced by back-splicing of pre-mRNA. The most studied regulatory mechanisms [...] Read more.
Among the long non-coding RNAs that are currently recognized as important regulatory molecules influencing a plethora of processes in eukaryotic cells, circular RNAs (circRNAs) represent a distinct class of RNAs that are predominantly produced by back-splicing of pre-mRNA. The most studied regulatory mechanisms involving circRNAs are acting as miRNA sponges, forming R-loops with genomic DNA, and encoding functional proteins. In addition to circRNAs generated by back-splicing, two types of circRNAs capable of autonomous RNA-RNA replication and systemic transport have been described in plants: viroids, which are infectious RNAs that cause a number of plant diseases, and retrozymes, which are transcripts of retrotransposon genomic loci that are capable of circularization due to ribozymes. Based on a number of common features, viroids and retrozymes are considered to be evolutionarily related. Here, we provide an overview of the biogenesis mechanisms and regulatory functions of non-replicating circRNAs produced by back-splicing and further discuss in detail the currently available data on viroids and retrozymes, focusing on their structural features, replication mechanisms, interaction with cellular components, and transport in plants. In addition, biotechnological approaches involving replication-capable plant circRNAs are discussed, as well as their potential applications in research and agriculture. Full article
(This article belongs to the Section Plant Genetics, Genomics and Biotechnology)
Show Figures

Figure 1

Figure 1
<p>Structural features of viroids and retrozymes. Schematic representation of the rod-like secondary structure of the <span class="html-italic">Potato spindle tuber viroid</span> (PSTVd, the family <span class="html-italic">Pospiviroidae</span>) genomic RNA shows location of the major PSTVd structural elements. The five structural domains indicated above the structure are as follows: the TL (terminal left), P (pathogenic), C (central), V (variable), and TR (terminal right) domains. A portion of the PSTVd structure shown in red indicates CCR (central conserved region); green and blue lines indicate HPI (Hairpin I) and HPII (Hairpin II), respectively. E indicates E-loop. The multibranched secondary structures of <span class="html-italic">Peach latent mosaic viroid</span> (PLMVd, the family <span class="html-italic">Avsunviroidae</span>) and <span class="html-italic">N. benthamiana</span> retrozyme 1 (NbRZ1) are shown with the positions of regions involved in the formation of hammerhead ribozyme structures in positive and negative polarity strands indicated by red and blue boxes, respectively. The sites of ribozyme self-cleavage are indicated by arrowheads. The NbRZ1 structure shown is a prediction; the structures of PSTVd and PLMVd are supported by experimental evidence.</p>
Full article ">Figure 2
<p>Proposed model for the retrozyme life cycle in plants. In the nucleus, activation of the retrozyme promoter located in the retrozyme (RZ) LTR and transcription of the RZ genomic locus results in the retrozyme transcript. The RZ transcript is cleaved by ribozymes located in the LTRs; the resulting RNA has a 5′-hydroxyl and a 2′,3′-cyclic phosphate end, which are ligated to give the retrozyme circular form. Since the site of retrozyme RNA replication is unknown, it has been proposed to occur either in the nucleus or, in analogy to Avsunviroids, in the chloroplast where the retrozyme RNA may be transported after exit from the nucleus to the cytoplasm, as indicated. In the nucleus, the retrozyme RNA can be replicated by Pol II as known for Pospiviroids, whereas in the chloroplast replication can depend on the RNA polymerase NEP, as known for Avsunviroids. In both cases, the replication products must then be transported to the cytoplasm for subsequent trafficking through plasmodesmata (PD) to neighboring cells and systemic leaves. It is likely that at this stage, the structured retrozyme RNA can be recognized by DCL proteins, which process it into small RNAs that can regulate the level of accumulation of the targeted mRNAs. Alternatively, the retrozyme circRNA may act as a template for protein expression or bind cellular miRNAs to act as a miRNA sponge.</p>
Full article ">
48 pages, 13373 KiB  
Review
Non-Coding RNAs in Breast Cancer: Diagnostic and Therapeutic Implications
by Roman Beňačka, Daniela Szabóová, Zuzana Guľašová and Zdenka Hertelyová
Int. J. Mol. Sci. 2025, 26(1), 127; https://doi.org/10.3390/ijms26010127 - 26 Dec 2024
Viewed by 148
Abstract
Breast cancer (BC) is one of the most prevalent forms of cancer globally, and has recently become the leading cause of cancer-related mortality in women. BC is a heterogeneous disease comprising various histopathological and molecular subtypes with differing levels of malignancy, and each [...] Read more.
Breast cancer (BC) is one of the most prevalent forms of cancer globally, and has recently become the leading cause of cancer-related mortality in women. BC is a heterogeneous disease comprising various histopathological and molecular subtypes with differing levels of malignancy, and each patient has an individual prognosis. Etiology and pathogenesis are complex and involve a considerable number of genetic alterations and dozens of alterations in non-coding RNA expression. Non-coding RNAs are part of an abundant family of single-stranded RNA molecules acting as key regulators in DNA replication, mRNA processing and translation, cell differentiation, growth, and overall genomic stability. In the context of breast cancer, non-coding RNAs are involved in cell cycle control and tumor cell migration and invasion, as well as treatment resistance. Alterations in non-coding RNA expression may contribute to the development and progression of breast cancer, making them promising biomarkers and targets for novel therapeutic approaches. Currently, the use of non-coding RNAs has not yet been applied to routine practice; however, their potential has been very well studied. The present review is a literature overview of current knowledge and its objective is to delineate the function of diverse classes of non-coding RNAs in breast cancer, with a particular emphasis on their potential utility as diagnostic and prognostic markers or as therapeutic targets and tools. Full article
(This article belongs to the Special Issue The Role of RNAs in Cancers: Recent Advances)
Show Figures

Figure 1

Figure 1
<p>Main histological forms of breast cancer and their molecular classification. On <span class="html-italic">the left</span> is a schematic representation of various cell masses in the breast, including benign tumor and malignant tumor subtypes (red). Molecular subtypes of breast cancer with essential markers, histological grade, therapeutic outline, and prognosis are shown on <span class="html-italic">the right</span>.</p>
Full article ">Figure 2
<p>Biogenesis of long non-coding RNAs (LncRNAs) and their role in breast cancer. (<b>A</b>) (1) Intergenic RNAs (LincRNAs) are transcripts of dsDNA between two protein-coding genes. (2) Antisense LncRNAs (asLncRNAs) are transcribed from complementary strands, either within the intronic or exonic region of protein-coding genes. (3) Intronic LncRNAs are transcripts of dsDNA from the intronic region of a protein-coding gene. Enhancer LncRNAs (eLnc RNAs) are transcribed from the dsDNA of enhancer regions of genes. (4) Bidirectional LncRNAs (biLncRNAs) originate from the bidirectional transcription of protein-coding genes. LncRNAs mediate the positioning of transcription factors in the promoters of protein-coding genes. (<b>B</b>) LncRNAs associated with different cellular processes that are important in cancerogenesis. (<b>C</b>) LncRNAs associated with each molecular subtype of breast cancer. Data are based on several sources [<a href="#B47-ijms-26-00127" class="html-bibr">47</a>,<a href="#B50-ijms-26-00127" class="html-bibr">50</a>,<a href="#B51-ijms-26-00127" class="html-bibr">51</a>,<a href="#B52-ijms-26-00127" class="html-bibr">52</a>,<a href="#B61-ijms-26-00127" class="html-bibr">61</a>,<a href="#B62-ijms-26-00127" class="html-bibr">62</a>].</p>
Full article ">Figure 3
<p>Biosynthesis and use of microRNA in breast cancer. (<b>A</b>) The primary transcript of microRNA (pri-miRNA) is processed in the nucleus into pre-miRNA by the RNAse III and self-assembles into double-stranded RNA with a small loop (hairpin shape). miRNA is exported from the nucleus into the cytosol (exportin 5), where a hairpin loop is cut off by Dicer endoribonuclease (RNA helicase), while the rest of the miRNA assembles into a complex called RNA-induced silencing complex (RISC) together with the Argonaute family of nucleoproteins (Ago). RISC binds to complementary motifs in the mRNA to cause post-transcriptional mRNA silencing by blocking the translation of the mRNA or degrading the mRNA into fragments. (<b>B</b>) Examples of upregulated (red) or downregulated (blue) miRNAs in ER+ and ER- breast cancer. (<b>C</b>) Regulatory role of miRNA in gene expression can be achieved by i) binding to mRNA and preventing translation or ii) binding to sponges made by lncRNA and/or circRNA. The synthesis of miRNAs can be epigenetically regulated by hyper- or hypomethylation. (<b>D</b>) Upregulated (red) and downregulated (blue) miRNAs in breast cancer, further subdivided according to their function in different cellular processes. Schematic visualization of acquired data on different miRNAs is based on the sources mentioned in the text and <a href="#ijms-26-00127-t001" class="html-table">Table 1</a>.</p>
Full article ">Figure 4
<p>Biogenesis and dysregulation of circRNAs in breast cancer. (<b>A</b>) CircRNAs can originate from intronic (i-circRNA), exonic (e-circRNA), or both intronic and exonic (ei-circRNA) transcripts of protein-coding genes. These transcripts can undergo either direct back-splitting (e.g., in e-circRNA formation) or the debranching of resistant intron variants (e.g., i-circRNA formation), intron-pairing-driven circularization, or exon skipping (e.g., ei-circRNA formation). CircRNA can act as an miRNA sponge by binding and suspending RNA-BPs (RNA binding proteins), or by interfering with mRNA (messenger RNA) translation. (<b>B</b>) CircRNAs associated with pro-oncogenic activity (<span class="html-italic">red</span>) and tumor suppressor activity (<span class="html-italic">blue</span>) and their role in breast cancer pathogenesis. The same circRNA can fall into several categories. (<b>C</b>) Distribution of circRNAs dysregulated in different BC subtypes (luminal A/BHR (+), hormone-positive, HER2+, and TNBC types). Image of breast tumor adapted from Servier Medical Art under license CC-BY-3.0.</p>
Full article ">Figure 5
<p>Biosynthesis of piRNAs and their alterations in breast cancer. (<b>A</b>) Formation of piRNA (PIWI-interacting RNA) occurs within and outside of the nucleus. Following transcription from genomic loci that contain transposon fragments, cluster transcripts are spliced into piRNA precursors (pre-piRNAs). The DNA loci responsible for producing piRNA precursors yield either single- or double-stranded molecules (sense and antisense transcripts). In the subsequent phase of piRNA biosynthesis (only the antisense precursor is illustrated in the figure), pre-piRNAs are transported to the perinuclear space (nuage) in close proximity to the mitochondria, where they are processed by the RNA helicase Armitage (Armi). Following despiralisation, the 5′ end of the precursor molecule is cleaved by the endonuclease Zucchini (Zuc), with the resulting 5′ fragment incorporated into PIWI proteins. The 3′ to 5′ exonuclease Nibbler (Nbr) then trims the piRNA to its final length. Concurrently, the small RNA 2′-O-methyltransferase Hen1 methylates the 2′-hydroxy group at the 3′ end. This process represents the primary biogenesis of piRNA (in the figure, this process is shown in red). The secondary biogenesis of piRNAs is called the ping-pong cycle and allows for amplification (arrows with dashed line; the DNA sequence with highlighted yellow background). The protein Aubergine (Aub) binds to antisense piRNAs and the complex cleaves sense piRNA precursors to give rise to sense piRNAs, which then form a complex with Ago3 (Argonaute3). The Ago3/piRNA complexes, in turn, cleave antisense piRNA precursors into pieces that form a complex with Aub. This cycle produces a large number of piRNAs in a short period of time. The piRNA-PIWI complexes return to the nucleus. The piRNA-PIWI complexes carry out their transposon-active activity with the help of DMTs (DNA methyl transferases) and HDACs (histone deacetylases). (<b>B</b>) Upregulated (<span class="html-italic">red</span>) and downregulated (<span class="html-italic">blue</span>) piRNAs in BC. Breast cancer image was adapted from Servier Medical Art under CC-BY-3.0 license. Data adapted from [<a href="#B222-ijms-26-00127" class="html-bibr">222</a>,<a href="#B228-ijms-26-00127" class="html-bibr">228</a>,<a href="#B230-ijms-26-00127" class="html-bibr">230</a>].</p>
Full article ">Figure 6
<p>Synthesis and use of siRNAs in breast cancer. (<b>A</b>) The precursors of siRNAs are long pieces of ds RNA or hairpin ss-RNA called pri-siRNAs. In cytosol, the molecule is cleaved by the Dicer enzyme into double-stranded siRNA (ds-siRNA) and, later, single-stranded siRNA (ss-siRNA), which form a RISC complex (RNA-induced silencing complex) together with RNPs (ribonucleoproteins) and Ago protein (Argonaute family). Inside the complex, siRNA binds to complementary motifs of the target mRNA to process the fragmentation. (<b>B</b>) Nanoparticles (NPs) (types in blue box) protect siRNAs from degradation, improve targeting and increasing the accumulation of s-siRNA in the tumor cells. Receptor-mediated endocytosis is initiated by the targeting of ligands or cationic components of NP. Following an endosomal escape, s-siRNA binds to the RISC, which enables the identification and degradation of complementary mRNA targets. (<b>C</b>) Experimental use of s-siRNA to target and inhibit mRNAs of the selected genes. For an explanation, see the text. Data adapted from Ngamcherdtrakul and Yantasee [<a href="#B247-ijms-26-00127" class="html-bibr">247</a>] and other resources [<a href="#B246-ijms-26-00127" class="html-bibr">246</a>,<a href="#B247-ijms-26-00127" class="html-bibr">247</a>,<a href="#B248-ijms-26-00127" class="html-bibr">248</a>,<a href="#B249-ijms-26-00127" class="html-bibr">249</a>,<a href="#B250-ijms-26-00127" class="html-bibr">250</a>,<a href="#B251-ijms-26-00127" class="html-bibr">251</a>,<a href="#B252-ijms-26-00127" class="html-bibr">252</a>,<a href="#B253-ijms-26-00127" class="html-bibr">253</a>,<a href="#B254-ijms-26-00127" class="html-bibr">254</a>,<a href="#B255-ijms-26-00127" class="html-bibr">255</a>,<a href="#B256-ijms-26-00127" class="html-bibr">256</a>,<a href="#B257-ijms-26-00127" class="html-bibr">257</a>,<a href="#B258-ijms-26-00127" class="html-bibr">258</a>,<a href="#B259-ijms-26-00127" class="html-bibr">259</a>,<a href="#B260-ijms-26-00127" class="html-bibr">260</a>,<a href="#B261-ijms-26-00127" class="html-bibr">261</a>,<a href="#B262-ijms-26-00127" class="html-bibr">262</a>]. <span class="html-italic">Abb.</span> CCR2, C Motif Chemokine Receptor 2; CXCR4, CXC Chemokine Receptor 4; DOPC, 1,2-dioleoyl-sn-glycero-3-phosphocholine; DANCR, Differentiation Antagonizing Non-Protein Coding RNA; DODAP-1,2-dioleoyl-3-dimethylammonium-propane; infMo, inflammatory monocytes; Lcn-2, Lipocalin-2; MIF, macrophage migration inhibitory factor; MSNP, mesoporous silica nanoparticle; mRNA, messenger RN; MTDH, metadherin; NP, nanoparticle; PAGA, Polyaminolated glycidyl methacrylate; PDPA, poly(2-(diisopropyl amino) ethyl methacrylate; PEG, Polyethyleneglycol; PEI, polyethyleneimine; Pgp, P-glycoprotein; PIGF, placental growth factor; PLK, Polo-like kinase; PLGA, poly (lactic-co-glycolic acid); PTPN, protein tyrosine phosphatase non-receptor; TAM, tumor-associated macrophage; T-Ly, T-cell; TME, tumor microenvironment; VEGF, vascular endothelial growth factor. Picture of breast cancer used from Servier Medical Art under CC-BY-3.0 license.</p>
Full article ">Figure 7
<p>Small nuclear (snRNA) and small nucleolar (sncRNA) molecules in breast cancer (BC). snRNA is a class of small nuclear RNA molecules which complex with small nuclear ribonucleoproteins (snRNP) to form various types of spliceosomes. These are involved in the post-transcriptional slicing of introns from the pre-messenger RNA (<span class="html-italic">pre-mRNA</span>) to form a mature mRNA (<span class="html-italic">left panel</span>). Certain components of snRNP and snRNA, e.g., U1, U2, U4, U5, and U6 (illustrated in color), were found to be overexpressed (<span class="html-italic">red</span>) in BC. The <span class="html-italic">right panel</span> shows sncRNAs that are upregulated (<span class="html-italic">red</span>) or downregulated (<span class="html-italic">blue</span>) in different types of breast cancer. <span class="html-italic">Abb. PRPF4</span>, core component of U4/U6 snRNP; <span class="html-italic">PRPF8</span>, core component of U4/U6-U5 tri-snRNP spliceosome complex; <span class="html-italic">SNRPC</span>, small nuclear ribonucleoprotein polypeptide C; <span class="html-italic">SNRNP200</span>, U5 small nuclear ribonucleoprotein. See the text for further explanation.</p>
Full article ">
36 pages, 2247 KiB  
Review
RNA Structure: Past, Future, and Gene Therapy Applications
by William A. Haseltine, Kim Hazel and Roberto Patarca
Int. J. Mol. Sci. 2025, 26(1), 110; https://doi.org/10.3390/ijms26010110 - 26 Dec 2024
Viewed by 234
Abstract
First believed to be a simple intermediary between the information encoded in deoxyribonucleic acid and that functionally displayed in proteins, ribonucleic acid (RNA) is now known to have many functions through its abundance and intricate, ubiquitous, diverse, and dynamic structure. About 70–90% of [...] Read more.
First believed to be a simple intermediary between the information encoded in deoxyribonucleic acid and that functionally displayed in proteins, ribonucleic acid (RNA) is now known to have many functions through its abundance and intricate, ubiquitous, diverse, and dynamic structure. About 70–90% of the human genome is transcribed into protein-coding and noncoding RNAs as main determinants along with regulatory sequences of cellular to populational biological diversity. From the nucleotide sequence or primary structure, through Watson–Crick pairing self-folding or secondary structure, to compaction via longer distance Watson–Crick and non-Watson–Crick interactions or tertiary structure, and interactions with RNA or other biopolymers or quaternary structure, or with metabolites and biomolecules or quinary structure, RNA structure plays a critical role in RNA’s lifecycle from transcription to decay and many cellular processes. In contrast to the success of 3-dimensional protein structure prediction using AlphaFold, RNA tertiary and beyond structures prediction remains challenging. However, approaches involving machine learning and artificial intelligence, sequencing of RNA and its modifications, and structural analyses at the single-cell and intact tissue levels, among others, provide an optimistic outlook for the continued development and refinement of RNA-based applications. Here, we highlight those in gene therapy. Full article
(This article belongs to the Special Issue Targeting RNA Molecules)
Show Figures

Figure 1

Figure 1
<p>Chemical structure of nitrogenous bases, which together with the sugar and phosphate backbone, form the helical structures of nucleic acids.</p>
Full article ">Figure 2
<p>Primary, secondary, and tertiary structural levels of the <span class="html-italic">Escherichia coli</span> phenylalanine tRNA. The messenger RNA and amino acid binding regions are highlighted in green and red. Tertiary structure image from PDB ID:6Y3G (<a href="https://www.wwpdb.org/pdb?id=pdb_00006y3g" target="_blank">https://www.wwpdb.org/pdb?id=pdb_00006y3g</a>; <a href="https://www.rcsb.org/sequence/6Y3G" target="_blank">https://www.rcsb.org/sequence/6Y3G</a>; both URLs accessed on 18 November 2024).</p>
Full article ">Figure 3
<p>(<b>A</b>). H-type pseudoknot. Complementarity regions are shown as overlapping boxes. (<b>B</b>). Pseudoknot-containing hammerhead ribozyme with synthetic construct bound to it at the top (5′ to 3′ in black. Nucleotide positions of the ribozyme are highlighted in red, as are its 5′ and 3′ termini. (<b>C</b>). RNA G-quadruplex. Guanine residues forming stacked tetrads are in red. Arrows follow the primary sequence in panels (<b>B</b>,<b>C</b>).</p>
Full article ">Figure 4
<p>Cyclodipeptide synthase RNA-binding protein from <span class="html-italic">Candidatus Glomeribacter gigasporarum</span> (red) bound to the <span class="html-italic">E. coli</span> Phe-tRNA<sup>Phe</sup> (green). Image from PDB ID:6Y4B (<a href="https://www.wwpdb.org/pdb?id=pdb_00006y4b" target="_blank">https://www.wwpdb.org/pdb?id=pdb_00006y4b</a>; accessed on 18 November 2024).</p>
Full article ">Figure 5
<p>RNA in gene therapy. RNA types used, challenges, and advantages.</p>
Full article ">
17 pages, 2740 KiB  
Article
Whole Genome Identification and Integrated Analysis of Long Non-Coding RNAs Responding ABA-Mediated Drought Stress in Panax ginseng C.A. Meyer
by Peng Chen, Cheng Chang and Lingyao Kong
Curr. Issues Mol. Biol. 2025, 47(1), 5; https://doi.org/10.3390/cimb47010005 - 25 Dec 2024
Viewed by 46
Abstract
Panax ginseng C.A. Meyer is a perennial herb that is used worldwide for a number of medical purposes. Long non-coding RNAs (lncRNAs) play a crucial role in diverse biological processes but still remain poorly understood in ginseng, which has limited the application of [...] Read more.
Panax ginseng C.A. Meyer is a perennial herb that is used worldwide for a number of medical purposes. Long non-coding RNAs (lncRNAs) play a crucial role in diverse biological processes but still remain poorly understood in ginseng, which has limited the application of molecular breeding in this plant. In this study, we identified 17,478 lncRNAs and 3106 novel mRNAs from ginseng by high-throughput illumine sequencing. 50 and 257 differentially expressed genes (DEGs) and DE lncRNAs (DELs) were detected under drought + ABA vs. drought conditions, respectively. The DEGs and DELs target genes main enrichment is focused on the “biosynthesis of secondary metabolites”, “starch and sucrose metabolism”, and “carbon metabolism” pathways under drought + ABA vs. drought conditions according to KEGG pathway enrichment analysis, suggesting that these secondary metabolites biosynthesis pathways might be crucial for ABA-mediated drought stress response in ginseng. Together, we identified drought stress response lncRNAs in ginseng for the first time and found that the target genes of these lncRNAs mainly regulate the biosynthesis of secondary metabolites pathway to response to drought stress. These findings also open up a new visual for molecular breeding in ginseng. Full article
Show Figures

Figure 1

Figure 1
<p>The RNA sequencing and bioinformatic analysis workflow in this study. The orange box shows the process of sequencing and bioinformatic analysis. The green box illustrates the data source and criteria quality control. LncRNAs and mRNAs identification were showed in the yellow box. Differential expression analysis and function enrichment analysis are displayed in the purple box.</p>
Full article ">Figure 2
<p>A comprehensive analysis of lncRNAs and mRNAs in ginseng roots. (<b>A</b>) Pie diagram shows the counts of different kind of lncRNAs. (<b>B</b>–<b>D</b>) Comparison of exon number, transcript length and ORF length between lncRNAs and mRNAs.</p>
Full article ">Figure 3
<p>Differentially expressed genes (DEGs) at the ginseng root tissue under different treatment conditions. D + A, represented drought + ABA treatment. (<b>A</b>) Volcano plots of DEGs in drought vs. CK. (<b>B</b>) Volcano plots of DEGs in D + A vs. CK. (<b>C</b>) Volcano plots of DEGs in D + A vs. drought. Upregulated, down-regulated, and non-differentially expressed genes are represented by red, green, and blue dots, respectively. (<b>D</b>) Heatmap and cluster analysis of the expression level of DEGs. CK1, CK2, CK3, D1, D2, D3, D + A1, D + A2, and D + A3 represent three repetitions of control, drought, and drought +ABA treatment, respectively.</p>
Full article ">Figure 4
<p>Comparison of Gene Ontology (GO) classification and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of differentially expressed genes (DEGs). (<b>A</b>–<b>C</b>) GO analysis of DEGs in drought vs. control, drought + ABA vs. control and drought + ABA vs. drought, respectively. (<b>D</b>) KEGG pathway enrichment analysis of DEGs in drought + ABA vs. drought. The size of the dot indicates the number of DEGs in the corresponding pathway. BP, CC, and MF represent biological processes (BP), cellular components (CC), and molecular functions (MF), respectively.</p>
Full article ">Figure 5
<p>Differentially expressed lncRNAs (DELs) at the ginseng root tissue under different treatment conditions. D + A, represented drought + ABA treatment. (<b>A</b>) Volcano plots of DELs in drought vs. CK. (<b>B</b>) Volcano plots of DELs in D + A vs. CK. (<b>C</b>) Volcano plots of DELs in D + A vs. drought. Up-regulated, down-regulated, and non-differentially expressed lncRNAs are represented by red, green, and blue dots, respectively. (<b>D</b>) Heatmap and cluster analysis of the expression level of DELs. CK1, CK2, CK3, D1, D2, D3, D + A1, D + A2, and D + A3 represent three repetitions of control, drought, and drought +ABA treatment, respectively.</p>
Full article ">Figure 6
<p>Comparison of Gene Ontology (GO) classification and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of the target genes of differentially expressed lnCRNAs (DELs). (<b>A</b>–<b>C</b>) GO analysis of these target genes in drought vs. control, drought + ABA vs. control and drought + ABA vs. drought, respectively. (<b>D</b>) KEGG pathway enrichment analysis of target genes of lncRNAs in drought + ABA vs. drought treatment. The size of the dot indicates the number of target genes in the corresponding pathway. BP, CC, and MF represent biological processes (BP), cellular components (CC), and molecular functions (MF), respectively.</p>
Full article ">
36 pages, 11803 KiB  
Article
Interplay of Transcriptomic Regulation, Microbiota, and Signaling Pathways in Lung and Gut Inflammation-Induced Tumorigenesis
by Beatriz Andrea Otálora-Otálora, César Payán-Gómez, Juan Javier López-Rivera, Natalia Belén Pedroza-Aconcha, Sally Lorena Arboleda-Mojica, Claudia Aristizábal-Guzmán, Mario Arturo Isaza-Ruget and Carlos Arturo Álvarez-Moreno
Cells 2025, 14(1), 1; https://doi.org/10.3390/cells14010001 - 24 Dec 2024
Viewed by 153
Abstract
Inflammation can positively and negatively affect tumorigenesis based on the duration, scope, and sequence of related events through the regulation of signaling pathways. A transcriptomic analysis of five pulmonary arterial hypertension, twelve Crohn’s disease, and twelve ulcerative colitis high throughput sequencing datasets using [...] Read more.
Inflammation can positively and negatively affect tumorigenesis based on the duration, scope, and sequence of related events through the regulation of signaling pathways. A transcriptomic analysis of five pulmonary arterial hypertension, twelve Crohn’s disease, and twelve ulcerative colitis high throughput sequencing datasets using R language specialized libraries and gene enrichment analyses identified a regulatory network in each inflammatory disease. IRF9 and LINC01089 in pulmonary arterial hypertension are related to the regulation of signaling pathways like MAPK, NOTCH, human papillomavirus, and hepatitis c infection. ZNF91 and TP53TG1 in Crohn’s disease are related to the regulation of PPAR, MAPK, and metabolic signaling pathways. ZNF91, VDR, DLEU1, SATB2-AS1, and TP53TG1 in ulcerative colitis are related to the regulation of PPAR, AMPK, and metabolic signaling pathways. The activation of the transcriptomic network and signaling pathways might be related to the interaction of the characteristic microbiota of the inflammatory disease, with the lung and gut cell receptors present in membrane rafts and complexes. The transcriptomic analysis highlights the impact of several coding and non-coding RNAs, suggesting their relationship with the unlocking of cell phenotypic plasticity for the acquisition of the hallmarks of cancer during lung and gut cell adaptation to inflammatory phenotypes. Full article
(This article belongs to the Topic Inflammatory Tumor Immune Microenvironment)
Show Figures

Figure 1

Figure 1
<p>Venn diagram with the transcriptomic metafirm in common and unique to each type of inflammatory disease. Created with BioRender.com.</p>
Full article ">Figure 2
<p>Coding (lilium and grey) and non-coding (purple) transcriptional regulatory network (cncTRN) of key upregulated transcription factors (TFs) and lncRNA in pulmonary arterial hypertension (PAH). Created with Cytoscape.</p>
Full article ">Figure 3
<p>Coding (lilium and grey) and non-coding (purple) transcriptional regulatory network (cncTRN) of key upregulated transcription factors in CD. Created with Cytoscape.</p>
Full article ">Figure 4
<p>Coding (lilium and grey) and non-coding (purple) transcriptional regulatory network (cncTRN) of key upregulated transcription factors in ulcerative colitis. Created with Cytoscape.</p>
Full article ">Figure 5
<p>Microbiome interaction with membrane receptor of PAH-related cells activating signaling pathways involved in transcriptional regulation during lung inflammation. In red are the upregulated genes and TFs; in black are the key upregulated TFs. Created with BioRender.com.</p>
Full article ">Figure 6
<p>Microbiome interaction with membrane receptor of CD-related cells, activating signaling pathways involved in transcriptional regulation during gut inflammation. In red are the upregulated genes and TFs; in black are the key upregulated TFs. Created with BioRender.com.</p>
Full article ">Figure 7
<p>Microbiome interaction with membrane receptor of UC-related cells, activating signaling pathways involved in transcriptional regulation during gut inflammation. In red are the upregulated genes and TFs; in black are the key upregulated TFs. Created with BioRender.com.</p>
Full article ">
35 pages, 12488 KiB  
Article
LncSL: A Novel Stacked Ensemble Computing Tool for Subcellular Localization of lncRNA by Amino Acid-Enhanced Features and Two-Stage Automated Selection Strategy
by Lun Zhu, Hong Chen and Sen Yang
Int. J. Mol. Sci. 2024, 25(24), 13734; https://doi.org/10.3390/ijms252413734 - 23 Dec 2024
Viewed by 156
Abstract
Long non-coding RNA (lncRNA) is a non-coding RNA longer than 200 nucleotides, crucial for functions like cell cycle regulation and gene transcription. Accurate localization prediction from sequence information is vital for understanding lncRNA’s biological roles. Computational methods offer an effective alternative to traditional [...] Read more.
Long non-coding RNA (lncRNA) is a non-coding RNA longer than 200 nucleotides, crucial for functions like cell cycle regulation and gene transcription. Accurate localization prediction from sequence information is vital for understanding lncRNA’s biological roles. Computational methods offer an effective alternative to traditional experimental methods for annotating lncRNA subcellular positions. Existing machine learning-based methods are limited and often overlook regions with coding potential that affect the function of lncRNA. Therefore, we propose a new model called LncSL. For feature encoding, both lncRNA sequences and amino acid sequences from open reading frames (ORFs) are employed. And we selected the most suitable features by CatBoost and integrated them into a new feature set. Additionally, a voting process with seven feature selection algorithms identified the higher contributive features for training our final stacked model. Additionally, an automatic model selection strategy is constructed to find a better performance meta-model for assembling LncSL. This study specifically focuses on predicting the subcellular localization of lncRNA in the nucleus and cytoplasm. On two benchmark datasets called S1 and S2 datasets, LncSL outperformed existing methods by 6.3% to 12.3% in the Matthew’s correlation coefficient on a balanced test dataset. On an unbalanced independent test dataset sourced from S1, LncSL improved by 4.7% to 18.6% in the Matthew’s correlation coefficient, which further demonstrates that LncSL is superior to other compared methods. In all, this study presents an effective method for predicting lncRNA subcellular localization through enhancing sequence information, which is always overlooked by traditional methods, and addressing contributive meta-model selection problems, which can offer new insights for other bioinformatics problems. Full article
Show Figures

Figure 1

Figure 1
<p>LncRNA subcellular location prediction technique.</p>
Full article ">Figure 2
<p>LncSL framework. Firstly, the dataset obtained is encoded by traditional nucleotide features and ORF-translated amino acid enhancement features; secondly, we conduct in-depth feature selection for the obtained high-dimensional feature matrix, select the features identified by more than four methods as our final feature space, train our feature space in the model pool, select the features through the output matrix composed of each output, and return the selected output to the model pool to find the four models that we finally use for stacking; thirdly, the four models selected by the model are used as the first basic classifier of the stack model, and the second meta classifier uses logical regression to form our LncSL model; Finally, LncSL is compared with the most advanced methods on several indicators.</p>
Full article ">Figure 3
<p>ROC curve and histogram of different nucleotide feature codes. (<b>A</b>,<b>C</b>) show performance bars for the different nucleotide feature encodings, and (<b>B</b>,<b>D</b>) are their ROC curves.</p>
Full article ">Figure 4
<p>ROC curve and histogram of different fusion amino acid feature codes. (<b>A</b>,<b>C</b>) show the performance bars of different amino acid fusion feature codes, and (<b>B</b>,<b>D</b>) are their ROC curves.</p>
Full article ">Figure 5
<p>Comparison of importance and correlation heatmaps before and after feature selection. (<b>A</b>,<b>B</b>) show the feature importance heat maps after feature selection of different datasets, and (<b>C</b>,<b>D</b>) are their feature correlation heat maps. For the feature importance graph, the horizontal coordinate represents the feature importance, where the redder the color, the higher the importance, and the grayer the color, the lower the importance, and the vertical coordinate represents each feature. For the feature correlation matrix, the horizontal and vertical coordinates represent the number of ten features, and the darker the color, the higher the correlation.</p>
Full article ">Figure 6
<p>Histogram and ROC curve of different single models and LncSL. (<b>A</b>,<b>B</b>) show the performance comparison between different single models and LncSL, and (<b>C</b>,<b>D</b>) are their ROC curves.</p>
Full article ">Figure 7
<p>Heat map of prediction results of different models on the S1 dataset. (<b>A</b>–<b>F</b>) show the predicted heat maps of different models on the S1 dataset.</p>
Full article ">Figure 8
<p>Heat map of prediction results of different models on the S2 dataset. (<b>A</b>–<b>F</b>) show the predicted heat maps of different models on the S2 dataset.</p>
Full article ">Figure 9
<p>Bar charts, ROC curves, and radar plots of different models. (<b>A</b>,<b>B</b>) show a bar chart comparing the predictions of different models. (<b>C</b>,<b>D</b>) are their ROC curves. (<b>E</b>,<b>F</b>) are their radar maps.</p>
Full article ">Figure 10
<p>ROC curves of different predictors on independent test dataset.</p>
Full article ">Figure 11
<p>Automatic model selection stacked integrated learning LncSL. Initially, seven types of lncRNA sequences were isolated by utilizing conventional nucleotide features and ORF-translated amino acid-improved features. Subsequently, deep feature selection was performed on the obtained high-dimensional feature space. Furthermore, the feature space underwent sequential training using a pool of 11 models, resulting in the formation of an output matrix composed of these 11 output vectors. Feature selection was performed on the output matrix, and the resulting matrix was then returned to the model pool to identify the four most appropriate models for the task. Next, the input for the stack model was the final feature space, and the first layer of the stack model utilized five-fold cross-validation with the four models chosen by the automatic model. Ultimately, the outcomes obtained from the initial layer were forwarded to the meta-learner, logistic regression, to obtain the probability of prediction for the outcome.</p>
Full article ">
17 pages, 3293 KiB  
Article
Comprehensive Transcriptome-Wide Profiling of 5-Methylcytosine Modifications in Long Non-Coding RNAs in a Rat Model of Traumatic Brain Injury
by Zhijun Xiang, Yixing Luo, Jiangtao Yu, Haoli Ma and Yan Zhao
Curr. Issues Mol. Biol. 2024, 46(12), 14497-14513; https://doi.org/10.3390/cimb46120871 - 23 Dec 2024
Viewed by 171
Abstract
Traumatic brain injury (TBI) poses a major global health challenge, leading to serious repercussions for those affected and imposing considerable financial strains on families and healthcare systems. RNA methylation, especially 5-methylcytosine (m5C), plays a crucial role as an epigenetic modification in [...] Read more.
Traumatic brain injury (TBI) poses a major global health challenge, leading to serious repercussions for those affected and imposing considerable financial strains on families and healthcare systems. RNA methylation, especially 5-methylcytosine (m5C), plays a crucial role as an epigenetic modification in regulating RNA at the level of post-transcriptional regulation. However, the impact of TBI on the m5C methylation profile of long non-coding RNAs (lncRNAs) remains unexplored. In the present study, we conducted a thorough transcriptome-wide examination of m5C methylation in lncRNAs in a rat TBI model utilizing MeRIP-Seq. Our results revealed significant differences in the amount and distribution of m5C methylation in lncRNAs between TBI and control groups, indicating profound changes in m5C methylation following TBI. Bioinformatic analyses linked these specifically methylated transcripts to pathways involved in immune response, neural repair, and lipid metabolism, providing insight into possible mechanisms underlying TBI pathology. These findings offer novel perspectives on the post-transcriptional modifications in lncRNA m5C methylation following TBI, which may contribute to understanding the disease mechanisms and developing targeted therapeutic strategies. Full article
(This article belongs to the Special Issue Chemical Biology of Nucleic Acid Modifications)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p><b>Experimental procedure:</b> (<b>A</b>). The general process of the experiment; (<b>B</b>). HE staining of coronal brain slices from the rat TBI model; and (<b>C</b>). HE staining of coronal sections of brain tissue from the sham group.</p>
Full article ">Figure 2
<p><b>Overview of lncRNA m<sup>5</sup>C methylation in both sham and TBI groups:</b> (<b>A</b>). Visualization of m<sup>5</sup>C peaks at the chromosomal level in TBI and sham groups. (<b>B</b>). Venn diagram showing the number of m<sup>5</sup>C methylation peaks detected in lncRNAs in TBI and sham groups. (<b>C</b>). Venn diagram showing the number of lncRNAs with m<sup>5</sup>C peaks in TBI and sham groups. (<b>D</b>). Pie charts depicting the distribution of methylated lncRNA sources in TBI and sham groups.</p>
Full article ">Figure 3
<p><b>Characteristics of post-TBI lncRNA m<sup>5</sup>C methylation and GO analysis:</b> (<b>A</b>). Number of significantly up-regulated and down-regulated m<sup>5</sup>C peaks in TBI rats (<span class="html-italic">p</span> &lt; 0.05, FC &gt; 2). (<b>B</b>). Number of m<sup>5</sup>C peaks on each lncRNA in TBI and sham rats (<span class="html-italic">p</span> &lt; 0.05, fold change &gt; 2). (<b>C</b>). Significantly enriched GO categories for hyper-methylated lncRNAs. <span class="html-italic">Incomplete GO term displayed: BP: Protein kinase C-activating G protein-coupled receptor signaling pathway; Positive regulation of vascular endothelial growth factor receptor signaling pathway.</span> (<b>D</b>). String diagrams showing the connections between different GO categories of hyper-methylated lncRNAs. (<b>E</b>). Significantly enriched GO categories for hypo-methylated lncRNAs. <span class="html-italic">Incomplete GO term displayed: MF: Beta-N-acetylglucosaminylglycopeptide beta-1,4-galactosyltransferase activity; Lysine N-acetyltransferase activity, acting on acetyl phosphate as donor. BP: positive regulation of cytokine-mediated signaling pathway</span>; positive regulation of phosphatidylinositol 3-kinase signaling pathway. (<b>F</b>). String diagrams showing the connections between different GO categories of hypo-methylated lncRNAs.</p>
Full article ">Figure 4
<p><b>Changes in lncRNA expression and GO analysis after TBI:</b> (<b>A</b>). Volcano plot displaying the lncRNAs that were significantly up-regulated and down-regulated after TBI (fold change &gt; 2, <span class="html-italic">p</span>-value &lt; 0.05). (<b>B</b>). Number of up-regulated and down-regulated lncRNAs (fold change &gt; 2, <span class="html-italic">p</span>-value &lt; 0.05). (<b>C</b>). Cluster analysis of differentially expressed lncRNAs. (<b>D</b>). Significantly enriched GO categories for up-regulated lncRNAs. <span class="html-italic">Incomplete GO term displayed: BP: positive regulation of cytokine-mediated signaling pathway.</span> (<b>E</b>). String diagrams showing the connections between different GO categories of the up-regulated lncRNAs. (<b>F</b>). Significantly enriched GO categories for down-regulated lncRNAs. <span class="html-italic">Incomplete GO terms displayed: BP: antigen processing and presentation of peptide antigen via MHC class I; antigen processing and presentation of endogenous peptide antigen via MHC class Ib; positive regulation of high voltage-gated calcium channel activity.</span> (<b>G</b>). String diagrams showing the connections between different GO categories of the down-regulated lncRNAs.</p>
Full article ">Figure 5
<p><b>Combined analysis of m<sup>5</sup>C methylation and lncRNA expression after TBI:</b> (<b>A</b>). The nine-quadrant plot illustrates the correlation between lncRNA m<sup>5</sup>C methylation and the corresponding lncRNA expression. Top-right quadrant: both methylation and gene expression are up-regulated (m<sup>5</sup>C-fold change &gt; 2 and Exp-fold change &gt; 2). Top-left quadrant: methylation is down-regulated while gene expression is up-regulated (m<sup>5</sup>C-fold change &lt; −2 and Exp-fold change &gt; 2). Bottom-left quadrant: both methylation and gene expression are down-regulated (m<sup>5</sup>C-fold change &lt; −2 and Exp-fold change &lt; −2). Bottom-right quadrant: methylation is down-regulated while gene expression is up-regulated (m<sup>5</sup>C-fold change &gt; 2 and Exp-fold change &lt; −2). (<b>B</b>). Venn diagrams showing the overlap between the number of differentially methylated genes and the number of differentially expressed lncRNAs.</p>
Full article ">
16 pages, 946 KiB  
Review
Host Long Noncoding RNAs as Key Players in Mycobacteria–Host Interactions
by Stephen K. Kotey, Xuejuan Tan, Audrey L. Kinser, Lin Liu and Yong Cheng
Microorganisms 2024, 12(12), 2656; https://doi.org/10.3390/microorganisms12122656 - 21 Dec 2024
Viewed by 425
Abstract
Mycobacterial infections, caused by various species within the Mycobacterium genus, remain one of the main challenges to global health across the world. Understanding the complex interplay between the host and mycobacterial pathogens is essential for developing effective diagnostic and therapeutic strategies. Host long [...] Read more.
Mycobacterial infections, caused by various species within the Mycobacterium genus, remain one of the main challenges to global health across the world. Understanding the complex interplay between the host and mycobacterial pathogens is essential for developing effective diagnostic and therapeutic strategies. Host long noncoding RNAs (lncRNAs) have emerged as key regulators in cellular response to bacterial infections within host cells. This review provides an overview of the intricate relationship between mycobacterial infections and host lncRNAs in the context of Mycobacterium tuberculosis and non-tuberculous mycobacterium (NTM) infections. Accumulation of evidence indicates that host lncRNAs play a critical role in regulating cellular response to mycobacterial infection within host cells, such as macrophages, the primary host cells for mycobacterial intracellular survival. The expression of specific host lncRNAs has been implicated in the pathogenesis of mycobacterial infections, providing potential targets for the development of novel host-directed therapies and biomarkers for TB diagnosis. In summary, this review aims to highlight the current state of knowledge regarding the involvement of host lncRNAs in mycobacterial infections. It also emphasizes their potential application as novel diagnostic biomarkers and therapeutic targets. Full article
(This article belongs to the Special Issue Editorial Board Members’ Collection Series: Bacterial Infection)
Show Figures

Figure 1

Figure 1
<p>Host lncRNAs in mycobacterial infections. PRC2: Polycomb repressive complex 2; DUSP4: Dual-specificity protein phosphatase 4; SATB1: Special AT-rich sequence-binding protein-1; NF-κβ: Nuclear factor kappa B; STAT3: Signal transducer and activator of transcription 3; LC3: Microtubule-associated protein 1A/1B-light chain 3 (MAP1LC3B); RHEB: Ras homolog enriched in brain; A20: TNF alpha induced protein 3 (TNFAIP3); hnRNPA2/B1: Heterogeneous nuclear ribonucleoproteins A2/B1; FUBP3: Far upstream element-binding protein 3; ULK1: Unc-51-like autophagy-activating kinases 1; mTOR: Mammalian target of rapamycin; TGF-β: Transforming growth factor beta; TRAF6: TNF receptor-associated factor 6.</p>
Full article ">
21 pages, 766 KiB  
Review
Epigenetic Biomarkers in Thrombophilia-Related Pregnancy Complications: Mechanisms, Diagnostic Potential, and Therapeutic Implications: A Narrative Review
by Claudia Ramona Bardan, Ioana Ioniță, Maria Iordache, Despina Călămar-Popovici, Violeta Todorescu, Roxana Popescu, Brenda Cristiana Bernad, Răzvan Bardan and Elena Silvia Bernad
Int. J. Mol. Sci. 2024, 25(24), 13634; https://doi.org/10.3390/ijms252413634 - 20 Dec 2024
Viewed by 293
Abstract
Pregnancy complications associated with thrombophilia represent significant risks for maternal and fetal health, leading to adverse outcomes such as pre-eclampsia, recurrent pregnancy loss, and intra-uterine growth restriction (IUGR). They are caused by disruptions in key physiological processes, including the coagulation cascade, trophoblast invasion, [...] Read more.
Pregnancy complications associated with thrombophilia represent significant risks for maternal and fetal health, leading to adverse outcomes such as pre-eclampsia, recurrent pregnancy loss, and intra-uterine growth restriction (IUGR). They are caused by disruptions in key physiological processes, including the coagulation cascade, trophoblast invasion, angiogenesis, and immune control. Recent advancements in epigenetics have revealed that non-coding RNAs, especially microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and extracellular vesicles (EVs) carrying these RNAs, play crucial roles in the regulation of these biological processes. This review aims to identify the epigenetic biomarkers that are the best candidates for evaluating thrombophilia-related pregnancy complications and for assessing the efficacy of anticoagulant and antiaggregant therapies. We emphasize their potential integration into personalized treatment plans, aiming to improve the risk assessment and therapy strategies for thrombophilic pregnancies. Future research should focus on validating these epigenetic biomarkers and establishing standardized protocols to enable their integration into clinical practice, paving the way for a precision medicine approach in obstetric care. Full article
Show Figures

Figure 1

Figure 1
<p>Key pathophysiological mechanisms of antiphospholipid syndrome during pregnancy (↑ = increase, PI3K = phopsphatidylinositol-3-kinase, AKT = protein kinase B).</p>
Full article ">Figure 2
<p>The most common modifications of the coagulation cascade induced by thrombophilias, including the key epigenetic biomarkers (modifications induced by thrombophilia are marked with red, while the epigenetic biomarkers are in yellow oval shapes; ↑ = elevation, ↓ = reduction).</p>
Full article ">
33 pages, 1875 KiB  
Review
New Relevant Evidence in Cholangiocarcinoma Biology and Characterization
by Nunzia Porro, Elena Spínola-Lasso, Mirella Pastore, Alessandra Caligiuri, Luca di Tommaso, Fabio Marra and Alessandra Gentilini
Cancers 2024, 16(24), 4239; https://doi.org/10.3390/cancers16244239 - 19 Dec 2024
Viewed by 298
Abstract
Among solid tumors, cholangiocarcinoma (CCA) emerges as one of the most difficult to eradicate. The silent and asymptomatic nature of this tumor, particularly in its early stages, as well as the high heterogeneity at genomic, epigenetic, and molecular levels delay the diagnosis, significantly [...] Read more.
Among solid tumors, cholangiocarcinoma (CCA) emerges as one of the most difficult to eradicate. The silent and asymptomatic nature of this tumor, particularly in its early stages, as well as the high heterogeneity at genomic, epigenetic, and molecular levels delay the diagnosis, significantly compromising the efficacy of current therapeutic options and thus contributing to a dismal prognosis. Extensive research has been conducted on the molecular pathobiology of CCA, and recent advances have been made in the classification and characterization of new molecular targets. Both targeted therapy and immunotherapy have emerged as effective and safe strategies for various types of cancers, demonstrating potential benefits in advanced CCA. Furthermore, the deeper comprehension of the cellular and molecular components in the tumor microenvironment (TME) has opened up possibilities for new innovative treatment methods. This review discusses recent evidence in the characterization and molecular biology of CCA, highlighting novel possible druggable targets. Full article
(This article belongs to the Special Issue Advanced Research in Oncology in 2024)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Representative histological images of CCA subtypes according to extent of the affected bile ducts. (<b>A</b>) A case of iCCA, small duct variant. The lesion is characterized by several homogeneous small glandular structures lacking overt cytological atypia and mucous production; the tumor border has an expansive pattern (H&amp;E, 20×); (<b>B</b>) a case of iCCA, large duct variant. The lesion is characterized by irregularly shaped and distributed mucin-secreting glands, intermingled with fibrous tissue; the lesion has a moderate degree of cytological atypia and shows infiltrative margins (H&amp;E, 20×).</p>
Full article ">Figure 2
<p>The tumor microenvironment in CCA. The CCA desmoplastic stroma is composed of extracellular components and different cell types that interact together as well as with components of the ECM and soluble factors within the TME to modulate CCA onset and progression. The immunosuppressive environment is mainly composed of TAMs, Tregs, and CAFs, which are mostly associated with worse outcomes. On the other side, cytotoxic CD8<sup>+</sup> T cells (FOXP3<sup>+</sup> and CTLA4<sup>+</sup>), GzmB-secreting B cells, and NK cells have antitumor effects.</p>
Full article ">Figure 3
<p>Dual role of cellular senescence in CCA. Under certain stimuli (i.e., telomere shortening, oncogenic activation, irradiation, or genotoxic drugs), cells can enter a state of sustained proliferation arrest, termed cellular senescence. In the context of CCA, cellular senescence constitutes a mechanism of tumor suppression by limiting the proliferation of pre-malignant lesions and tumors. Conversely, accumulation of senescent cells can contribute to tumor progression, metastasis, and increased resistance to therapy. SASP: senescent-associated secretory phenotype.</p>
Full article ">
17 pages, 1687 KiB  
Review
Current Insights into the Roles of LncRNAs and CircRNAs in Pulpitis: A Narrative Review
by Dulce Martha Fuchen-Ramos, Ana Gabriela Leija-Montoya, Javier González-Ramírez, Mario Isiordia-Espinoza, Fernando García-Arévalo, Viviana Pitones-Rubio, Carlos Olvera-Sandoval, Isis Mateos-Corral and Nicolás Serafín-Higuera
Int. J. Mol. Sci. 2024, 25(24), 13603; https://doi.org/10.3390/ijms252413603 - 19 Dec 2024
Viewed by 233
Abstract
Pulpitis, an inflammation of the dental pulp, is generated by bacterial invasion through different ways as caries. In the establishment and development of this disease, different biological processes are involved. Long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) are transcripts with regulatory capacity [...] Read more.
Pulpitis, an inflammation of the dental pulp, is generated by bacterial invasion through different ways as caries. In the establishment and development of this disease, different biological processes are involved. Long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) are transcripts with regulatory capacity participating in different biological functions and have been implicated in different diseases. The aim of this narrative review is to critically analyze available evidence on the biological role of lncRNAs and circRNAs in pulpitis and discuss possible new research prospects. LncRNAs and circRNAs involved in pulpitis were explored, addressing their expression, molecular mechanisms, targets and biological effects studied in animal and in vitro models, as well as in studies in human patients. LncRNAs and circRNAs are emerging as key regulators of diverse biological functions in pulpitis including apoptosis, proliferation, differentiation, oxidative stress, autophagy, ferroptosis, inflammation and immune response. The molecular mechanisms performed by these non-coding RNAs (ncRNAs) involved interactions with miRNAs and the formation of regulatory networks in the context of pulpitis. Further studies more deeply analyzing the participation of lncRNAs and circRNAs in pulpitis will reveal the potential applications of these ncRNAs as biomarkers or their use in therapeutic strategies in pulp inflammation. Full article
(This article belongs to the Special Issue The Role of Non‐coding RNAs in Human Health and Diseases)
Show Figures

Figure 1

Figure 1
<p>LncRNAs can modulate different biological processes in the context of pulpitis. LncRNAs expressed differentially and validated by qRT-PCR in pulpitis, including analyses in vitro and animal models, as well as studies in patients, are indicated. More lncRNAs have been analyzed in pulpitis; however, expression was not validated, and they were not included in this figure. Blue arrows indicate increased expression. Red arrows indicate decreased expression. The biological processes possibly regulated by validated lncRNAs in human pulpitis, as well as in models resembling some characteristics of this disease, are shown. Created with BioRender.com.</p>
Full article ">Figure 2
<p>CircRNAs can modulate diverse biological processes in the context of pulpitis. CircRNAs expressed differentially and validated by qRT-PCR in human pulpitis and in vitro models, resembling some characteristics of this disease, are indicated. Blue arrows indicate increased expression. Red arrow indicates decreased expression. Additionally, these circRNAs could be involved in regulation of different cellular processes. Created with BioRender.com.</p>
Full article ">
17 pages, 7699 KiB  
Systematic Review
Long Non-Coding RNAs as Diagnostic Biomarkers for Ischemic Stroke: A Systematic Review and Meta-Analysis
by Jianwei Pan, Weijian Fan, Chenjie Gu, Yongmei Xi, Yu Wang and Peter Wang
Genes 2024, 15(12), 1620; https://doi.org/10.3390/genes15121620 - 18 Dec 2024
Viewed by 333
Abstract
Ischemic stroke is a serious cerebrovascular disease, highlighting the urgent need for reliable biomarkers for early diagnosis. Recent reports suggest that long non-coding RNAs (lncRNAs) can be potential biomarkers for ischemic stroke. Therefore, our study seeks to investigate the potential diagnostic value of [...] Read more.
Ischemic stroke is a serious cerebrovascular disease, highlighting the urgent need for reliable biomarkers for early diagnosis. Recent reports suggest that long non-coding RNAs (lncRNAs) can be potential biomarkers for ischemic stroke. Therefore, our study seeks to investigate the potential diagnostic value of lncRNAs for ischemic stroke by analyzing existing research. A comprehensive literature search was conducted across the PubMed, ScienceDirect, Wiley Online Library, and Web of Science databases for articles published up to July 10, 2024. Statistical analyses were performed using Stata 17.0 software to calculate pooled sensitivity, specificity, positive likelihood ratio (PLR), diagnostic odds ratio (DOR), negative likelihood ratio (NLR), and area under the curve (AUC). Heterogeneity was explored with the Cochran-Q test and the I2 statistical test, and publication bias was assessed with Deeks’ funnel plot. A total of 44 articles were included, involving 4302 ischemic stroke patients and 3725 healthy controls. Results demonstrated that lncRNAs H19, GAS5, PVT1, TUG1, and MALAT1 exhibited consistent trends across multiple studies. The pooled sensitivity of lncRNAs in the diagnosis of ischemic stroke was 79% (95% CI: 73–84%), specificity was 88% (95% CI: 77–94%), PLR was 6.63 (95% CI: 3.11–14.15), NLR was 0.23 (95% CI: 0.16–0.33), DOR was 28.5 (95% CI: 9.88–82.21), and AUC was 0.88 (95% CI: 0.85–0.90). Furthermore, the results of subgroup analysis indicated that lncRNA H19 had superior diagnostic performance. LncRNAs demonstrated strong diagnostic accuracy in distinguishing ischemic stroke patients from healthy controls, underscoring their potential as reliable biomarkers. Because most of the articles included in this study originate from China, large-scale, high-quality, multi-country prospective studies are required to further validate the reliability of lncRNAs as biomarkers for ischemic stroke. Full article
(This article belongs to the Special Issue The Epigenetic Roles of lncRNAs)
Show Figures

Figure 1

Figure 1
<p>Flow diagram of study search and selection.</p>
Full article ">Figure 2
<p>Risk of bias assessment of eligible studies using QUADAS-2. (<b>A</b>) Summary of bias risk items in the QUADAS-2 quality assessment. (<b>B</b>) Percentile of risk of bias in the QUADAS-2 quality assessment [<a href="#B18-genes-15-01620" class="html-bibr">18</a>,<a href="#B19-genes-15-01620" class="html-bibr">19</a>,<a href="#B21-genes-15-01620" class="html-bibr">21</a>,<a href="#B25-genes-15-01620" class="html-bibr">25</a>,<a href="#B28-genes-15-01620" class="html-bibr">28</a>,<a href="#B30-genes-15-01620" class="html-bibr">30</a>,<a href="#B32-genes-15-01620" class="html-bibr">32</a>,<a href="#B38-genes-15-01620" class="html-bibr">38</a>,<a href="#B41-genes-15-01620" class="html-bibr">41</a>,<a href="#B43-genes-15-01620" class="html-bibr">43</a>,<a href="#B51-genes-15-01620" class="html-bibr">51</a>,<a href="#B52-genes-15-01620" class="html-bibr">52</a>,<a href="#B53-genes-15-01620" class="html-bibr">53</a>,<a href="#B56-genes-15-01620" class="html-bibr">56</a>,<a href="#B57-genes-15-01620" class="html-bibr">57</a>,<a href="#B58-genes-15-01620" class="html-bibr">58</a>,<a href="#B59-genes-15-01620" class="html-bibr">59</a>].</p>
Full article ">Figure 3
<p>(<b>A</b>) Forest plot showing the pooled sensitivity and specificity of lncRNAs in diagnosing ischemic stroke. Squares represent individual studies, while line segments indicate the 95% confidence interval (CI) for each study. The center of the diamond and the red dashed line represent the pooled effect size, and the width of the diamond corresponds to the 95% CI of the pooled results. (<b>B</b>) Summary receiver operating characteristic (SROC) curve with the 95% confidence and prediction contours. The <span class="html-italic">Y</span>-axis represents sensitivity, and the <span class="html-italic">X</span>-axis represents specificity. Numbers represent individual studies, and the curves depict combined diagnostic performance.</p>
Full article ">Figure 4
<p>Fagan nomogram (<b>A</b>) and likelihood ratio scattergram (<b>B</b>) are illustrated. (<b>A</b>) If two values are known, the nomogram can be used to calculate a third value. (<b>B</b>) The ordinate represents the positive likelihood ratio, indicating the likelihood of a positive result in a patient compared to a non-patient. The abscissa represents the negative likelihood ratio, indicating the likelihood of a negative result in a patient compared to a non-patient.</p>
Full article ">Figure 5
<p>Deeks’ funnel plot for publication bias analysis. A <span class="html-italic">p</span>-value &gt; 0.05 indicates no significant publication bias.</p>
Full article ">Figure 6
<p>LncRNAs as diagnostic markers for ischemic stroke. Created using <a href="https://BioRender.com" target="_blank">https://BioRender.com</a> (accessed on 3 December 2024).</p>
Full article ">
19 pages, 17567 KiB  
Article
Whole-Genome Sequencing and Genome Annotation of Pathogenic Elsinoë batatas Causing Stem and Foliage Scab Disease in Sweet Potato
by Yuan Xu, Yuqing Liu, Yihan Wang, Yi Liu and Guopeng Zhu
J. Fungi 2024, 10(12), 882; https://doi.org/10.3390/jof10120882 - 18 Dec 2024
Viewed by 433
Abstract
A pathogen strain responsible for sweet potato stem and foliage scab disease was isolated from sweet potato stems. Through a phylogenetic analysis based on the rDNA internal transcribed spacer (ITS) region, combined with morphological methods, the isolated strain was identified as Elsinoë batatas. [...] Read more.
A pathogen strain responsible for sweet potato stem and foliage scab disease was isolated from sweet potato stems. Through a phylogenetic analysis based on the rDNA internal transcribed spacer (ITS) region, combined with morphological methods, the isolated strain was identified as Elsinoë batatas. To comprehensively analyze the pathogenicity of the isolated strain from a genetic perspective, the whole-genome sequencing of E. batatas HD-1 was performed using both the PacBio and Illumina platforms. The genome of E. batatas HD-1 is about 26.31 Mb long in 167 scaffolds, with a GC content of 50.81%, and 7898 protein-coding genes, 131 non-coding RNAs, and 1954 interspersed repetitive sequences were predicted. Functional annotation revealed that 408 genes encode virulence factors involved in plant disease (DFVF—Plant). Notably, twenty-eight of these virulence genes encode secretory carbohydrate-active enzymes (CAZymes), including two endo-1,4-β-xylanase genes and seven cutinase genes, which suggested that endo-1,4-β-xylanase and cutinase play a vital role in the pathogenicity of E. batatas HD-1 within sweet potato. In total, twelve effectors were identified, including five LysM effectors and two CDIP effectors, suggesting that LysM and CDIP effectors play significant roles in the interaction between E. batatas HD-1 and sweet potato. Additionally, our analysis of biosynthetic gene clusters (BGCs) showed that two gene clusters are involved in melanin and choline metabolism. This study enriches the genomic resources of E. batatas and provides a theoretical foundation for future investigations into the pathogenic mechanisms of its infection in sweet potatoes, as well as potential targets for disease control. Full article
Show Figures

Figure 1

Figure 1
<p>“Fucaishu 1830-11” typical diseased plant.</p>
Full article ">Figure 2
<p>Morphology of filamentous fungus: (<b>a</b>) the front view of the colony morphological characteristics of strain HD-1 cultured for 30d; (<b>b</b>) the back view of the colony morphological characteristics of strain HD-1 cultured for 30d; and (<b>c</b>) conidia image (bar = 10 µm).</p>
Full article ">Figure 3
<p>The neighbor-joining (NJ) phylogenetic tree was constructed based on ITS sequences.</p>
Full article ">Figure 4
<p>Symptoms of infection in sweet potato “Fucaishu1830-11” plants after 10 d of infection.</p>
Full article ">Figure 5
<p>The circular graphical map of the whole <span class="html-italic">E. batatas</span> HD-1 genome.</p>
Full article ">Figure 6
<p>The GC_depth distribution diagram of <span class="html-italic">E. batatas</span> HD-1.</p>
Full article ">Figure 7
<p>The COG function classification of HD-1 genes.</p>
Full article ">Figure 8
<p>The GO function classification of HD-1 genes.</p>
Full article ">Figure 9
<p>The KEGG function classification of HD-1 genes.</p>
Full article ">Figure 10
<p>Analysis of genes encoding DFVF—Plant in the HD-1 genome: (<b>a</b>) genes predicted to encode DFVF—Plant. (<b>b</b>) GO functional annotation of genes encoding DFVF—Plant.</p>
Full article ">Figure 11
<p>Analysis of genes encoding secretory CAZymes associated with plant diseases in the HD-1 genome: (<b>a</b>) Venn diagram of genes encoding DFVF—Plant, CAZymes, and secretome; (<b>b</b>) numbers of genes for different types of CAZymes involved in plant diseases; and (<b>c</b>) GO functional annotation of genes encoding secretory CAZymes associated with plant diseases.</p>
Full article ">Figure 12
<p>PHI functional classification of proteins encoded in the HD-1 genome.</p>
Full article ">Figure 13
<p>Analysis of secondary metabolite biosynthetic gene clusters.</p>
Full article ">Figure 14
<p>The synteny analyses between the <span class="html-italic">E. batatas</span> CRI-CJ2 and HD-1 genome. The colored area between two black dividing lines represents the collinear region aligned. Each connecting line represents a comparison record, where if it aligns positively, the outer ring colors of the comparison area are the same; if it aligns in reverse, the outer ring colors are different.</p>
Full article ">
20 pages, 12418 KiB  
Article
LncRNA-MSTRG.19083.1 Targets NTRK2 as a miR-429-y Sponge to Regulate Circadian Rhythm via the cAMP Pathway in Yak Testis and Cryptorchidism
by Tianan Li, Qiu Yan, Jinghong Nan, Xue Huang, Ruiqing Wang, Yong Zhang, Xingxu Zhao and Qi Wang
Int. J. Mol. Sci. 2024, 25(24), 13553; https://doi.org/10.3390/ijms252413553 - 18 Dec 2024
Viewed by 237
Abstract
Long noncoding RNAs (LncRNAs) play essential roles in numerous biological processes in mammals, such as reproductive physiology and endocrinology. Cryptorchidism is a common male reproductive disease. Circadian rhythms are actively expressed in the reproductive system. In this study, a total of 191 LncRNAs [...] Read more.
Long noncoding RNAs (LncRNAs) play essential roles in numerous biological processes in mammals, such as reproductive physiology and endocrinology. Cryptorchidism is a common male reproductive disease. Circadian rhythms are actively expressed in the reproductive system. In this study, a total of 191 LncRNAs were obtained from yak testes and cryptorchids. Then, we identified NTRK2’s relationship to circadian rhythm and behavioral processes. Meanwhile, the ceRNA (LncRNA-MSTRG.19083.1/miR-429-y/NTRK2) network was constructed, and its influence on circadian rhythm was revealed. The results showed that NTRK2 and LncRNA-MSTRG.19083.1 were significantly upregulated, and miR-429-y was obviously decreased in cryptorchid tissue; NTRK2 protein was mainly distributed in the Leydig cells of the testis. In addition, the upregulation of the expression level of miR-429-y resulted in the significant downregulation of LncRNA and NTRK2 levels, while the mRNA and protein levels of CREB, CLOCK, and BMAL1 were significantly upregulated; the knockdown of miR-429-y resulted in the opposite changes. Our findings suggested that LncRNA-MSTRG.19083.1 competitively binds to miR-429-y to target NTRK2 to regulate circadian rhythm through the cAMP pathway. Taken together, the results of our study provide a comprehensive understanding of how the LncRNA-miRNA-mRNA networks operate when yak cryptorchidism occurs. Knowledge of circadian-rhythm-associated mRNAs and LncRNAs could be useful for better understanding the relationship between circadian rhythm and reproduction. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

Figure 1
<p>Analysis of differentially expressed LncRNA: (<b>A</b>) LncRNA distribution in the testis group and the cryptorchidism group; (T, testis; C, cryptorchidism); (<b>B</b>) number of LncRNA types; (<b>C</b>) total number of LncRNAs that were differentially expressed in the testis and the cryptorchidism group; the red color represents the number of LncRNAs upregulated in cryptorchidism compared to normal testes, the green color represents the number of LncRNAs decreased. (<b>D</b>) cluster heat map analysis of testis and cryptorchidism; (<b>E</b>) LncRNA differentially expressed volcano map; (<b>F</b>) GO analysis of 191 differentially expressed LncRNAs; (<b>G</b>) KEGG enrichment analysis.</p>
Full article ">Figure 2
<p>GO and KEGG analysis of differentially expressed genes (circadian rhythms): (<b>A</b>) genes involved in circadian activity were analyzed in testis and cryptorchids; (<b>B</b>) analysis of differentially expressed genes involved in circadian rhythms in testicular and cryptorchidism; (<b>C</b>) biological process analysis of circadian rhythm-related differential genes; (<b>D</b>) screening for circadian rhythm-related differential genes using Veen map; (<b>E</b>) interaction gene analysis of NTRK2; (<b>F</b>) functional analysis of NTRK2 involved in the GO enrichment process; (<b>G</b>) NTRK2 is involved in the enrichment signaling pathway.</p>
Full article ">Figure 3
<p>Verification of differentially expressed LncRNAs in the testis and cryptorchids: (<b>A</b>) differentially expressed LncRNAs were detected with qRT-PCR, <span class="html-italic">n</span> = 3, mean ± SD, <span class="html-italic">** p</span> &lt; 0.01; (<b>B</b>) LncRNA sequencing analysis; (<b>C</b>) differentially expressed miRNAs were detected with qRT-PCR, <span class="html-italic">n</span> = 3, mean ± SD, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>Differential expression of genes was verified in the testis and cryptorchids of the yaks: (<b>A</b>) differentially expressed mRNAs were detected with qRT-PCR, <span class="html-italic">n</span> = 3, mean ± SD, <span class="html-italic">** p</span> &lt; 0.01; (<b>B</b>) mRNA sequencing analysis.</p>
Full article ">Figure 5
<p>NTRK2 expression pattern analysis in testis and cryptorchids: (<b>A</b>–<b>C</b>) mRNA and protein expression levels of NTRK2 were analyzed using qPCR and Western blot, <span class="html-italic">n</span> = 3, mean ± SD, <span class="html-italic">** p</span> &lt; 0.01; (<b>D</b>) H&amp;E staining was used to analyze the morphology and structure of testis and cryptorchids; (<b>E</b>,<b>F</b>) protein distribution of NTRK2 was stained by immunohistochemistry and immunofluorescence in testis and cryptorchids. LC: Leydig cells, SC: Sertoli cells, SP: spermatogonium, PS: primary spermatocyte, ST: seminiferous tubule, PMC: peritubular myoid cells.</p>
Full article ">Figure 6
<p>Targeting relationship between <span class="html-italic">NTRK2</span> and LncRNAs/miRNAs: (<b>A</b>) the scatter plot revealed the expression level of LncRNAs; (<b>B</b>,<b>C</b>) the network map and mulberry map reveal the targeting relationship between NTRK2 and LncRNAs and miRNAs. –– means Gene unknown.</p>
Full article ">Figure 7
<p>Between target gene NTRK2 and LncRNA and miR-429-y, (<b>A</b>) IF staining identified isolated yak LCs using antibodies against HSD3β (green) and β-tubulin (red), with magnification, 20×; (<b>B</b>,<b>C</b>) binding site of LncRNA-MSTRG.19083.1, NTRK2, and miRNA-429-y; (<b>D</b>) Luciferase activity in 293T cells after co-transfection with mimics of miRNA-429-y (100 nM) or mimic NC (100 nM) and pmirGLO-LncRNA-MSTRG.19083.1/NTRK2 3′-UTR-WT (400 ng) or pmirGLO-LncRNA-MSTRG.19 083.1/NTRK2 3′-UTR-MUT (400 ng). Values represent mean ± SD; <span class="html-italic">n</span> = 3, <span class="html-italic">** p</span> &lt; 0.01.</p>
Full article ">Figure 8
<p>Validation of the targeting relationship between miR-429-y and LncRNA-MSTRG.19083.1 and NTRK2. (<b>A</b>,<b>B</b>) Localization of NTRK2 protein after transfection with mimic-miR-429-y/inhibitor-miR-429-y in LCs was analyzed by immunofluorescence staining. NTRK2 was colored green, HSD3β is shown in red, and nuclei were counterstained with DAPI (blue); magnification, 20×. (<b>C</b>–<b>F</b>) mRNA expression of miR-429-y after transfection of 100 nM mimic/inhibitor into LCs for 48 h. Values represent mean ± SD; <span class="html-italic">n</span> = 3. ** <span class="html-italic">p</span> &lt; 0.01. (<b>D</b>,<b>E</b>,<b>G</b>,<b>H</b>) the mRNA expression of LncRNA-MSTRG.19083.1 and NTRK2 after transfection of 100 nM mimic/inhibitor into LCs for 48 h. ** <span class="html-italic">p</span> &lt; 0.01. (<b>I</b>–<b>K</b>) Protein expression of NTRK2 was assessed by Western blotting after transfection of 100 nM mimic/inhibitor into LCs for 48 h (<span class="html-italic">n</span> = 3). ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 9
<p>LncRNA-MSTRG.19083.1/miR-429-y targets NTRK2 to mediate the cAMP signaling pathway to regulate circadian rhythm: (<b>A</b>–<b>E</b>) mRNA and protein expression of CREB, CLOCK, and BAML1 after transfection of 100 nM mimic into LCs for 48 h, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01; (<b>F</b>–<b>J</b>) mRNA and protein expression of CREB, CLOCK, and BAML1 after transfection of 100 nM inhibitor into LCs for 48 h, <span class="html-italic">** p</span> &lt; 0.01; (<b>K</b>) process model diagram of regulatory mechanism.</p>
Full article ">
13 pages, 526 KiB  
Review
Potential Regulation of the Long Non-Coding RNA Metastasis-Associated Lung Adenocarcinoma Transcript1 by Estrogen in Parkinson’s Disease
by Eman Adel and Maya Nicolas
Life 2024, 14(12), 1662; https://doi.org/10.3390/life14121662 - 16 Dec 2024
Viewed by 481
Abstract
Parkinson’s disease (PD) is the second-leading cause of death among neurodegenerative disease after Alzheimer’s disease (AD), affecting around 2% of the population. It is expected that the incidence of PD will exceed 12 million by 2040. Meanwhile, there is a recognized difference in [...] Read more.
Parkinson’s disease (PD) is the second-leading cause of death among neurodegenerative disease after Alzheimer’s disease (AD), affecting around 2% of the population. It is expected that the incidence of PD will exceed 12 million by 2040. Meanwhile, there is a recognized difference in the phenotypical expression of the disease and response to treatment between men and women. Men have twice the incidence of PD compared to women, who have a late onset and worse prognosis that is usually associated with menopause. In addition, the incidence of PD in women is associated with the cumulative estrogen levels in their bodies. These differences are suggested to be due to the protective effect of estrogen on the brain, which cannot be given in clinical practice to improve the symptoms of the disease because of its peripheral side effects, causing cancer in both males and females in addition to the feminizing effect it has on males. As PD pathophysiology involves alteration in the expression levels of multiple LncRNAs, including metastatic-associated lung adenocarcinoma transcript 1 (MALAT1), and as estrogen has been illustrated to control the expression of MALAT1 in multiple conditions, it is worth investigating the estrogen–MALAT1 interaction in Parkinson’s disease to mimic its protective effect on the brain while avoiding its peripheral side effects. The following literature review suggests the potential regulation of MALAT1 by estrogen in PD, which would enhance our understanding of the pathophysiology of the disease, improving the development of more tailored and effective treatments. Full article
(This article belongs to the Special Issue Revolutionizing Neuroregeneration)
Show Figures

Figure 1

Figure 1
<p><b>An illustration of the potential relationship between estrogen and MALAT1 in PD</b>. (<b>a</b>) The inhibitory effect of high-dose estrogen on the expression of MALAT1. (<b>b</b>) The activated expression of estrogen target genes in the absence of MALAT1 through favoring open chromatin remodeling. (<b>c</b>) The stimulatory effect of low-dose or no estrogen on the expression of MALAT1. (<b>d</b>) The inhibited expression of estrogen target genes in the presence of MALAT1 through favoring a closed chromatin remodeling. ER, estrogen receptor; ERE, estrogen response element; Me3 H3K27me3, trimethylation of lysine 27 on the histone H3 protein.</p>
Full article ">
Back to TopTop