
Noncoding RNAs in periodontitis: Progress and perspectives (Review)
- Authors:
- Published online on: August 11, 2025 https://doi.org/10.3892/ijmm.2025.5607
- Article Number: 166
-
Copyright: © Feng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Periodontitis is an epidemic disease whose global prevalence increased by 99.0% between 1990 and 2019, reaching 1 billion in the latter year (1,2). Periodontitis reduces patients' quality of life and places an economic burden on society, making exploration of its pathogenesis and achieving a cure urgent (3). As an inflammatory disease, periodontitis is affected by a number of factors such as genomics, epigenetic modification and environmental conditions. Due to the complexity of the disease's etiology and mechanism, conventional treatment for periodontitis cannot completely help regenerate lost periodontal tissue. In recent years, due to the rapid development of sequencing technology, researchers have gained broader and deeper understanding of the transcriptome in periodontitis, which lays a foundation for exploring the disease's pathogenesis and potential targeted therapies (4).
In the past, the linear model of deoxyribonucleic acid (DNA) to ribonucleic acid (RNA) to protein was often regarded as the central dogma of molecular biology, with the greatest attention on coding RNAs (5). However, as sequencing technology matured, researchers found that the number of noncoding RNAs (ncRNAs), previously considered 'dark transcriptomes' or 'genomic dark matter', far exceeds that of coding RNAs (6). Moreover, ncRNAs play important regulatory roles in the occurrence and development of a number of inflammatory diseases (7). At present, there is no unified system of classification for ncRNAs. Commonly, they are divided into those shorter than and those longer than 200 nucleotides (nt) and respectively called short noncoding RNAs (sncRNAs) and long noncoding RNAs (lncRNAs). The former group includes microRNAs (miRNAs), small nucleolar RNAs (snoRNAs), small nuclear RNAs (snRNAs), piwi-interaction RNAs (piRNAs), small interfering RNAs (siRNAs), transfer RNA (tRNAs), tRNA-derived fragments (tRFs) and Y RNA fragments (YRFs). The last can be divided into long intergenic RNAs (lincRNAs), antisense RNAs (asRNAs), intronic noncoding RNAs (iRNAs), enhancer RNAs (eRNAs) and pseudogenes (8). In addition, circular RNAs (circRNAs) have emerged onto researchers' radar and are considered a new class of ncRNAs with regulatory potential (9).
The continuous advancement of high-throughput sequencing technology and computational technology has made significant contributions to the research on ncRNAs (10). In particular, long-read sequencing has enhanced our understanding of ncRNAs (11). Sequencing analysis involves multiple key steps, starting from sample preparation to high-throughput sequencing. Initially, samples are processed based on the specific type of target ncRNAs (such as small RNA, lncRNA, or circRNA) and the species being studied. Magnetic columns are then used to enrich or remove interfering RNAs. Subsequently, libraries are constructed, which involves connector connection, reverse transcription and PCR amplification. Finally, sequencing is performed using a high-throughput platform, followed by bioinformatics analysis tools (12). These computational tools are essential for revealing the relationship between RNA structure and function (13).
In inflammatory diseases, ncRNAs are abnormally expressed and regulate the occurrence and development of disease (14). The pathogenesis of periodontitis, as a nonspecific inflammatory disease, involves different types of ncRNAs (15). For example, the function of ncRNAs depends on their sequence, secondary and tertiary structures, subcellular localization and interactions with other molecules. Commonly, ncRNAs bind to microRNAs or RNA-binding sites, thereby affecting gene expression. The present review briefly summarized the reports of ncRNAs in periodontal immune inflammation, cell proliferation and bone homeostasis, subsequently moving on to descriptions of new ncRNA research areas. Finally, it addressed the potential of ncRNAs in the diagnosis and treatment of periodontitis, the limitations of this research field at present and its future prospects (Fig. 1). Micro (mi)RNA is a common ncRNA and previous comprehensive articles have summarized the mechanism and function of miRNAs in periodontitis (16). However, there are a number of research reports on lncRNA and circRNA in periodontitis and no systematic review summary has been seen yet. Therefore, this review instead mainly discusses the relevance of newly emerging lncRNAs and circRNAs to the field of periodontitis.
LncRNAs in periodontitis
LncRNAs regulate immune inflammation in periodontitis
Periodontitis is a chronic inflammatory disease. Most tissue damage during the progression of periodontitis has been proven to be caused mainly by the host's immune response to infection, rather than from the direct effects of microorganisms. Activation of the inflammatory process is a double-edged sword. On the one hand, a moderate inflammatory response can maintain the health of the body; on the other, activation and continuous stimulation of relevant inflammatory pathways harm the host, resulting in the occurrence or aggravation of rheumatoid arthritis, atherosclerosis (AS), kidney injury, periodontitis and other diseases (17-19). In immunoinflammatory studies, related genes mainly pass through the Janus-associated kinase/signal transducer and activator of transcription/interleukin-6 (IL-6), nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB), Toll-like receptor (TLR), B-cell receptor and T-cell receptor signaling pathways to regulate disease progression (20,21). The same is true of lncRNAs involved in periodontitis.
TLR4 is mainly expressed in cells involved in the host defense function. It can conduct signal recognition and transmission by recognizing specific antigenic components of pathogen-related molecular patterns, a capability considered to be closely related to immune inflammation (22). One study on periodontitis demonstrated that the lncRNA papillary thyroid carcinoma susceptibility candidate 3 (PTCSC3) can downregulate TLR4 (23), while another found that the lncRNA MFG-AS1 can upregulate TLR4 (24). The two studies indicated that lncRNAs could affect the progression of periodontitis by regulating TLR4. Conversely, TLR4 could also regulate lncRNAs and thereby affect periodontitis. For example, in macrophages of periodontitic tissues, the response of the lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) to TLR4 activation is time dependent and contributes to macrophage polarization (25).
LncRNAs also play an important role in the regulation of periodontitis by acting on interleukin, a common inflammatory factor. Knockdown of the lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) can downregulate IL-6, IL-1β and tumor necrosis factor-α (TNF-α); reduce the ratio of B-cell lymphoma 2 (Bcl-2)-like protein 4 (BAX) to Bcl-2 through the microRNA (miR)-200C-3p/TNF receptor-associated factor 6 (TRAF6) axis; and reduce inflammation and apoptosis in periodontal-ligament cells (PDLCs), thereby helping alleviate the progression of chronic periodontitis (26). Lysyl oxidase homolog 1 (LOXL1)-AS1 inhibits IL-1β expression in periodontal ligament stem cells (PDLSCs) to reduce the severity of periodontitis (27). The lncRNA zinc finger Y-chromosomal protein (ZFY)-AS1 inhibits the release of inflammatory factors in periodontitis, such as IL-6 and IL-1B, by regulating the miRNA-129-5P/DEAD-box helicase 3 X-linked (DDX3X) axis (28). The lncRNA small nucleolar RNA host gene 5 (SNHG5) mediates periodontal inflammation by influencing the expression of inflammatory cytokines through the NF-κB signaling pathway (29). In lipopolysaccharide (LPS)-induced PDLSCs, Linc00616 promotes ferroptosis through the miRNA-370/transferrin receptor (TfR) axis, aggravating the progression of periodontitis (30). Linc01126 suppressed proliferation while promoting apoptosis and the secretion of inflammatory cytokines IL-1β, IL-6, IL-8 and TNF-α in human PDLCs (hPDLCs) under hypoxia by sponging miR-518a-5p (31). A recent study found that Linc01126 acts a sponge for miR-655-3p, inhibiting its binding to IL-6 mRNA and thereby promoting inflammation progression and JAK2/STAT3 pathway activation in periodontitis (32) (Fig. 2).
LncRNAs regulate bone homeostasis in periodontitis
In healthy periodontal tissue, the rates of bone matrix degradation and bone resorption are balanced by those of matrix mineralization and new-bone deposition, a state known as periodontal-bone homeostasis. When periodontal tissue is infected with pathogens, this balance is disturbed and the rates of matrix degradation and bone resorption exceed those of matrix mineralization and new-bone deposition. This results in periodontal-tissue inflammation, bone absorption and even tooth loosening and loss (33).
Existing studies have shown that lncRNAs play important roles in bone metabolism, regulating the occurrence and development of various bone diseases (34,35). Therefore, studying the mechanisms of ncRNAs in periodontal homeostasis can provide new ideas for exploring the pathogenesis and clinical treatment of periodontitis. Studies on the role of lncRNAs in periodontal homeostasis are mainly limited to their effects on osteogenic differentiation; in terms of mechanism of action (36,37), the present review mainly focused on the competitive binding of lncRNAs to miRNAs, which affects the expression of target genes and promotes osteogenesis. The bone morphogenetic protein/Smad, Wingless/Integrated (Wnt)/β-catenin, Notch, Hedgehog, mitogen-activated protein kinase (MAPK) and fibroblast growth factor (FGF) signaling pathways are the most critical ones in the process of osteogenic differentiation. Our research group previously constructed the competing endogenous RNA (ceRNA) regulatory network of PDLSCs osteogenic differentiation, preliminarily verified that Loc1001302640/miR-1469 and miR-1469/Smad6 may play important roles in the process of osteogenic differentiation, predicted their possible binding sites and screened out that the MAPK and transforming growth factor-β (TGF-β) pathways play important roles in the process of osteogenic differentiation (38). In addition, studies have shown that the lncRNAs maternally expressed gene 3 (MEG3) (39), AS ncRNA in the Inhibitor of cyclin-dependent kinase 4 (CDK4; INK4) locus (ANRIL) (40), JmjC domain-containing histone demethylation protein 1D (JHDM1D)-AS1 (41), Linc01126 (31), dendrocyte-expressed seven-transmembrane protein (DCSTAMP) domain-containing protein 1 (DCST1)-AS1 (42), MALAT1 (43), Linc00707 (44), LncRNA00638 (45), distal-less homeobox 2 (DLX2)-AS1 (46) and Opa interacting protein 5 (OIP5)-AS1 (47) and Linc01133 (36) regulate the osteogenic differentiation of PDLSCs by adsorbing miRNAs as ceRNAs. In addition to the key pathways, MEG3 was found to regulate different miRNAs to affect osteogenic differentiation of PDLSCs through the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) or Akt/inhibitor of NF-κB kinase (IKK) pathways (37,39).
Some researchers have focused on another aspect of lncRNA regulation of periodontal homeostasis: Bone resorption. Activation of osteoprotegerin (OPG)/receptor activator of nuclear factor κ-β ligand (RANKL)/TNF receptor superfamily member 11a (TNFRSF11A, or RANK), a key bone resorption pathway, promotes osteoclastic differentiation and disrupts the balance between bone degradation and bone formation, leading to fracture and related functional loss (48). NEAT1 in myeloid-derived suppressor cells (MDSCs) activates the classic nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain containing 3 (NLRP3) inflammasome activation pathway, leading to the exacerbation of bone destruction in periodontitis (49). In a rat orthodontic tooth movement (OTM) model, the lncRNA differentiation antagonizing non-protein coding RNA (DANCR) could disturb the balance of periodontal-bone metabolism by upregulating Jagged1 to increase stress-induced osteoclast formation and root absorption (50). SNHG5 affects the secretion of RANKL and inhibits differentiation of osteoclasts during OTM by combining with CCAAT-enhancer-binding protein-β (C/EBP-β), a regulator of osteoclasts (51). The lncRNA noncoding repressor of nuclear factor of activated T cells (NRON) can effectively promote nuclear transport of NF-κB inhibitor, inhibit osteoclastic- and alveolar-bone resorption and correct the imbalance of bone metabolism in periodontitis (52) (Fig. 3). Much study remains to be done on lncRNAs in bone metabolism in periodontitis compared with other diseases, especially in osteoclasts.
LncRNAs regulate the proliferation of PDLSCs/PDLCs. Excessive proliferation of PDLSCs is an abnormal phenotype in periodontitis that is considered to be related to the disease's pathogenesis. It is generally considered that abnormal proliferation of PDLSCs in periodontitis can disrupt the balance between proliferation and apoptosis, leading to aggravation of periodontal inflammation. Various lncRNAs participate in the regulation of PDLSC proliferation and affect disease progression by acting on different targets. The lncRNA erythrocyte membrane protein band 4.1-like 4A (EPB41L4A)-AS1 directly targets the expression of miR-214-3p, resulting in changes in Yamaguchi sarcoma virus oncogene (Yes1)-associated transcriptional regulatory factor (YAP1) levels and ultimately enhancing the proliferation and osteogenic differentiation of hPDLCs (53). MALAT1 promotes proliferation of PDLSCs in periodontitis as well as disease progression by upregulating FGF2 (54). The lncRNA mortal obligate RNA transcript (MORT) inhibits PDLSC proliferation and reduces recurrence of periodontitis (55). LncRNA activator of myogenesis (linc-RAM) was found to be the downstream regulatory factor of FGF2; FGF2 can promote proliferation of PDLSCs by inhibiting expression of linc-RAM in periodontitic pathogenesis (56). The lncRNA DCST1-AS1 upregulates periodontal-ligament-associated protein-1 (PLAP-1) by combining with miR-21 precursor to reduce expression of CDK4, CDK6 and cyclin D1 (CCND-1) and inhibit PDLC proliferation, thereby alleviating the inflammatory response in periodontal tissues (40). Similarly, LOXL1-AS1 might inhibit the proliferation of periodontitic PDLSCs and downregulate IL-1β to improve periodontitis (27). The lncRNA taurine upregulated 1 (TUG1) mediates LPS-induced proliferative inhibition of hPDLCs by sponging miR-132 (57) (Fig. 4). However, few studies have been conducted on the exact mechanism of lncRNAs in regulating cell proliferation and apoptosis in periodontitis and further exploration is still needed.
CircRNAs as emerging players in periodontitis
CircRNAs regulate immune inflammation in periodontitis
In periodontitis, research into circRNAs in immune inflammation is mainly focused on regulation of downstream target genes by sponging miRNA. Researchers collected gingival tissues from healthy and periodontitic patients for high-throughput sequencing and constructed a circRNA-miRNA-mRNA regulatory network through bioinformatics (58). Similarly, another researcher collected inflamed gingiva and sequenced them to construct a circRNA-miRNA interaction network (59). The key circRNAs in the pathological process of periodontitis were screened out of the interaction network, including Circ_0095812, Circ_0120299, Circ_0125699, Circ_0062491 and Circ_0043115. Circ_0062491 sponges miR-584 in THP-1 cells treated with P. gingivalis, which enhances the anti-inflammatory effect of lactoferrin receptor. This suggests that circRNAs play a regulatory role in the pathological process of periodontitis. A recent study found that in LPS-induced human PDLCs, Circ_0062491 targeted miR-498/SOCS6 to alleviate periodontitis (60). These two studies showed that the same circRNA can sponge different miRNAs to function as a ceRNA in different periodontal cells. CircRNA plasmacytoma variant translocation 1 (circPVT1) promotes periodontitic progression by modulating the nuclear factor (erythroid-derived 2)-like 2 (Nrf-2)/heme oxygenase-1 (HO-1) pathway via the miR-24-3p/hypoxia inducible factor 1 subunit α inhibitor (HIF1AN) axis (61). However, some studies have also reported on the protective effect of circRNA in periodontitis. Silencing Circ_0099630 alleviates LPS-induced PDLC injury by targeting miR-940/TRAF6/NF-κB in periodontitis (62). In another RNA-seq study of gingiva from patients with chronic periodontitis, Circ_0003948 was markedly downregulated; this downregulation conferred protection through the miR-144-3p/ nuclear receptor subfamily 2 group F member 2 (NR2F2)/phosphatase and tensin homolog (PTEN) axis (63). However, in human gingival fibroblasts, Circ_0138959 was confirmed to inhibit cell viability and promote pyroptosis through the miR-527/cysteine-aspartic acid protease/proteinase 5 (Caspase-5) axis, aggravating periodontitis (64) (Fig. 2).
CircRNAs regulate bone homeostasis in periodontitis
At present, studies of the mechanism by which circRNAs regulate bone homeostasis in periodontitis are still relatively limited. PDLSCs, as important seed cells for periodontal regeneration, are the main objects of research into periodontal-bone homeostasis. RNA-seq was performed on osteogenesis-induced PDLSCs to construct a circRNA-ceRNA network. It was predicted that the circRNAs B-cell translocation gene 3 (BTG3)-associated nuclear protein (BANP) and itchy E3 ubiquitin protein ligase (ITCH) can bind to miR-34a and miR-146a, respectively and both eventually participate in the MAPK pathway (65). Circ_0003072 can promote osteogenic differentiation of PDLSCs through the Circ_0003072/miR-488-3p/chordin-like 1 (CHRDL1) pathway; Circ_0003072 acted as a sponge for miR-488-3p, thereby upregulating CHRDL1 levels (66). Furthermore, researchers found that the circRNA spermatogenesis-associated 13 (circ-SPATA13) functions as a molecular sponge for miR-485-5p_R+1, in turn targeting BMP7 to promote osteogenic differentiation of PDLSCs (67). In another study, the circRNA FAT1 was involved in regulating PDLSC osteogenic differentiation through the ceRNA network of miR-4781-3p/Smad5 (68). Similarly, Circ_0062491 has been found to promote PDLSC osteogenic differentiation through the miR-142-5p/insulin-like growth factor 1 (IGF1) axis (69). Hsa-miR-6783-3p could act as a sponge of the circRNA protein kinase D3 (circPRKD3) to indirectly regulate osteogenic differentiation of mechanically stimulated PDLSCs (70). Moreover, it has been found that the circRNA low-density lipoprotein receptor-related protein 6 (Lrp6) promotes cementoblastic differentiation through the miR-145A-5p/zinc finger E-box-binding homeobox 2 (Zeb2) axis and plays an important role in the regeneration of periodontal tissues (71). However, it has also been reported that hsa_Circ_0003489 inhibits osteogenic differentiation of PDLSCs by triggering activation of autophagy in the hypoxic periodontitic microenvironment (72). In addition to its role in the regulation of alveolar-bone remodeling through the ceRNA network, circRNA can directly regulate bone homeostasis by influencing signaling pathways. Exosomal Circ_0000722 derived from PDLSCs undergoing osteogenic differentiation might promote osteoclastogenesis by upregulating TRAF6 expression and activating the downstream NF-κB and Akt signaling pathways (73) (Fig. 3).
CircRNAs regulate the proliferation of periodontal-ligament cells and stem cells. The functions of various circRNAs in regulating cell proliferation are inconsistent. For example, a study found that Circ_0099630 curbed PDLC proliferation and osteogenic differentiation through elevating sprouty rhoketin signaling antagonist 1 (SPRY1) expression via sponging miR-212-5p in periodontitis (74). The circRNA MAP3K11 (Circ_002284) enhances the proliferation and migration of human PDLSCs (hPDLSCs) and reduces their apoptosis through miR-511/TLR4 (75). Although Circ_0003764 regulates osteogenic differentiation of PDLSCs, it can also inhibit the proliferation of PDLSCs (76). Moreover, the circRNA sponge for miR-7 (CiRS-7, also known as CDR1-AS) binds to miR-7 to activate the extracellular signal-regulated kinase signaling pathway and mediates the inhibitory effect of LPS on PDLSC proliferation (77). In addition to its regulation of bone homeostasis and PDLSC proliferation (77,78), the binding of CDR1-AS to miR-7 in PDLSCs has been confirmed to inhibit Krüppel-like factor 4 (KLF4) expression to maintain expression of stemness-related genes (79). As previous studies reported, CDR1-AS/miR-7 affects different target genes to regulate the biological functions of PDLSCs via a ceRNA mechanism (62,78,79). Furthermore, CDR1-AS can also combine with miR-7 to act on different targets and participate in the regulation of such diseases as pulmonary hypertension (80), abdominal aortic aneurysm (81), breast cancer (82) and hepatocellular carcinoma (83). Therefore, the interaction of CDR1-AS with miR-7 could be an interesting research focus; more studies are needed (Fig. 4).
In general, recent studies have demonstrated that circRNAs play important roles in the regulation of periodontitis. Research on circRNAs in periodontitic pathogenesis has important clinical significance. Most studies have revealed that ceRNA mechanisms underlie the regulatory role of circRNAs; no other regulatory mechanisms of circRNAs have been found in periodontitis. Furthermore, previous studies on the ceRNA mechanism of circRNAs often did not assess cellular localization of circRNAs; circRNA in cytoplasm can often bind to miRNAs and then regulate target genes. The multiple mechanisms of circRNAs in periodontitis deserve further exploration.
Other ncRNAs in periodontitis
In addition to lncRNAs and circRNAs, other ncRNAs are also involved in the progression of periodontitis. Through ncRNA analysis of exosomes in blood samples from healthy individuals and patients with periodontitis, both before and after treatment, it was found that small nucleolar RNAs (SNORD57 and SNODB177) exhibited significant differences (84). This indicates that the expression level of small non-coding RNAs (sRNAs) may serve as a diagnostic indicator for periodontitis in the future. In 2015, Duran-Pinedo et al (85) conducted a comprehensive analysis of bacterial sRNAs in the intergenic regions of the oral microbiome in healthy individuals and periodontitis patients. This sequencing analysis found that sRNAs were an important factor in regulating the transformation of the oral microbiome from a symbiotic to a dysbiotic one. These bacterial sRNAs affect bacterial toxicity by regulating changes in the oral microbiome, while at the same time they exert the host's immune response. Therefore, bacterial sRNAs may serve as key regulatory elements in the pathogenesis of periodontitis.
New areas of ncRNA research
Secondary structures of ncRNAs
NcRNAs can directly physically bind to other nucleic acid targets or sequence-specific recognition of RNA-binding proteins (RBPs) through base complementary pairing. A ncRNA can also bind with cellular partners through its secondary or tertiary structure (6), which is composed of local stem loops and helices.
In periodontitis, it is common for ncRNAs to directly bind to proteins or genes through their structural features, thereby affecting protein function or the expression of downstream genes. For example, apoptotic vesicles containing miR-143-3p derived from macrophages disrupt periodontal bone homeostasis by binding to insulin-like growth factor-binding protein 5 (IGFBP5) in bone marrow mesenchymal stem cells (BMSCs) (86). Similarly, circLRP6 acts as a binding target of miR-145a-5p, while Zeb2 is a downstream target of the same miRNA during the cementoblastic differentiation process (71). Therefore, circLRP6 regulates the differentiation of cemontoblasts in periodontitis by antagonizing the function of miR-145a-5p. The structure of ncRNA determines the ncRNA's mode of interaction with cellular partners; by changing the ncRNA's interactions with proteins or other RNAs, it can perform regulatory functions (87). H19, as one of the earliest-discovered lncRNAs, has been found to have secondary structural changes that might damage lncRNA function, leading to complex diseases such as heart failure (88). The development of sequencing technologies and in silico and experimental methods has allowed further exploration of the effects of variations in ncRNAs. For instance, single-nucleotide polymorphisms and indels in lncRNA genes and regulatory regions alter RNA secondary structures, expression levels and target recognition and then confer a predisposition toward cancer on carriers (89). Currently, techniques for RNA structure detection can be divided into two categories, one based on small-molecule modification and the other based on crosslinking and proximity linking. Sequencing is an important detection method used to interpret RNA structure (90): it provides crucial technical support for detecting the secondary structure of ncRNAs, reveals their mysterious spatial structure and proves the close relationship between RNA structure and function. Researchers have even developed RNA structural analyses that enable structural profiling at the transcriptomic scale in living cells (91).
NcRNA-protein interaction
Post-transcriptional regulation (PTR) mediated by ncRNAs is closely related to RBP expression. RBPs, which are critical for maintaining the transcriptome, can also regulate RNA processing and transportation through PTR, including regulation of RNA splicing, polyadenylation, mRNA stability, mRNA localization and translation (92). Recently, accumulating evidence has demonstrated that ncRNA-RBP interactions modulate the pathogenesis of periodontitis. The lncRNA solute carrier family 30, member A4 (SLC30A4)-AS1 binds serine and arginine rich splicing factor 3 (SRSF3) protein to affect the senescence of PDLSCs and the osteogenic-differentiation ability of PDLSCs (93). LincRNA-EPS could inhibit the expression of Caspase-11/NLRP3 inflammasome components in periodontal inflammation by compromising the activation of the NF-κB signaling pathway via interacting with transformation-associated recombination (TAR) DNA-binding protein 43 (TDP43) (94). The lncRNA X-inactive specific transcript (Xist) recruited the enrichment of upstream transcription factor 2, C-Fos interacting (USF2) to the WD repeat-containing protein 72 (WDR72) promoter region to promote its transcription and facilitate osteogenic differentiation of PDLSCs (95).
To uncover the interaction between RNA and RBPs, researchers have developed various experimental methods and predictive tools. In the process of experimental research, a larger range can be narrowed down to screen out candidate RBPs with greater potential. For example, the binding of the lncRNA gastric-cancer-associated transcript 2 (GACAT2) to pyruvate kinase muscle isozyme M1/M2 (PKM1/2) proteins was tested using chromatin isolation by RNA purification (ChIRP) sequencing (96). In a recent study on periodontitis, an RNA pull-down assay identified the RBP to the lncRNA NR_045147 as integrin subunit β3 binding protein (ITGB3BP). Co-immunoprecipitation (CoIP) and ubiquitination-IP assays found that NR_045147 maintains protein stability by competing with ubiquitination (97). Lu et al (98) used the catRAPID algorithm to predict the potential protein partners of lncRNA Lockd and RNA pull-down to confirm Lockd's interaction with SUZ12 polycomb repressive complex 2 subunit (SUZ12), thereby affecting the binding of SUZ12 to the key regulatory regions of osteogenic genes in the inflammatory microenvironment. This experimental approach might very well reflect the interactions of lncRNAs and proteins in vivo. The aforementioned methods have important reference value in exploring regulation of interactions between ncRNA and RBP in periodontitis.
NcRNA-encoded peptides or proteins
The traditional view is that ncRNA does not encode proteins. In fact, however, small open reading frames (sORFs) can encode small bioactive peptides (99,100). NcRNAs can have one or more sORFs that can be translated into peptides of <100 amino acids. However, by default, the traditional gene annotation process filters out proteins with <100 amino acids and treats them as noise or false positives, leading some ncRNAs with coding functions to be ignored. With the advancement of ribosome sequencing and protein translation techniques, as well as improvements in precision and accuracy, a number of ncRNAs have been found to translate into bioactive peptides or proteins (101). In the last decade, an increasing number of ncRNAs have been identified as encoding and functioning through peptides; these ncRNAs play important roles in a number of diseases (102,103). For example, human leukocyte antigen complex P5 (HCP5)-132aa, a microprotein encoded by HCP5 harboring this ORF, is highly expressed in gastric-cancer (GC) cells and tissues and can promote proliferation of GC cells by inhibiting ferroptosis (104). The lncRNA 181-Rik is translated into the conserved micropeptide short transmembrane mitochondrial protein 1 (Stmp1), located in mitochondria, which regulates retinal ischemia/reperfusion-induced retinal ganglion cell (RGC) death by modulating microglial inflammatory response (105). Cell division cycle 42 (CDC42)-165aa protein, encoded by circCDC42, promotes excessive activation of the pyrin inflammasome and aggravates lung injury caused by pyroptosis of alveolar macrophages (106).
At present, multiple bioassay software applications have been developed to predict the coding capacity of ncRNAs. Liang et al (107) developed the MicroProteinDB database, which offers and visualizes extensive knowledge to aid retrieval and analysis of computationally predicted and experimentally validated microproteins originating from various ncRNA types. Although these applications can predict coding capacity using different algorithms, experimental verification is the most convincing method. Researchers can construct an ncRNA-ORF mutant fusion protein vector with green fluorescent protein (GFP) to detect whether GFP fusion protein can be expressed and verify the protein coding ability of an ncRNA (108). Antibodies (Abs) that bind specifically to ncRNA-translated peptides can also be designed and prepared to verify whether ncRNA-translated peptides can be detected (109). For example, anti-Linc00263-P Ab was customized to explore the function of the new microprotein encoded by Linc00263 in promoting osteolytic bone metastasis in breast cancer (110). Although there is currently no relevant report in the field of periodontitis, the regulation of periodontitis by ncRNAs can be further explored based on the mechanisms of ncRNA-encoded proteins.
NcRNAs and single-cell sequencing
Unlike traditional sequencing technology, single-cell sequencing technology can sequence the genome or transcriptome of a single cell type, more accurately detect cell clustering and identify cell heterogeneity, which is of great significance for disease prevention, diagnosis and individualized treatment (111,112). The use of single-cell sequencing to detect ncRNA in periodontal tissues could offer new perspectives on the epigenetic regulation of periodontitic pathogenesis. Single-cell RNA sequencing of periodontitic gingival tissue revealed that in lncR APDC knockout mice, the proportions and functions of immune cells changed, including T and B cells, macrophages and neutrophils (113). However, there are relatively few reports on using single-cell sequencing to detect ncRNAs in periodontal tissues. Exploring the expression profile of ncRNAs at the single-cell level could reveal unique cell clusters in periodontitis and provide insights into the heterogeneity of the disease. In summary, as a new technology, single-cell sequencing can more accurately analyze the transcriptome profile of a single cell and enable researchers to further study the molecular characteristics of the heterogeneity of periodontitis.
Diagnostic potential of ncRNAs in periodontitis
NcRNA expression level is closely related to disease progression and has tissue specificity. NcRNAs in tissues or body fluids are emerging as potential new biomarkers because the levels of free ncRNAs in body fluids are stable and renewable. Numerous studies have indicated that ncRNAs have outstanding advantages as diagnostic markers, while lncRNAs and circRNAs in particular show high diagnostic value (114,115).
In recent years, the receiver operating characteristic (ROC) curve has been widely used to find prognostic disease markers and evaluate the reliability of diagnostic markers. In studies of ncRNAs as diagnostic markers, the research flowchart mainly includes screening differentially expressed genes by sequencing, predicting target genes using relevant databases, performing functional clustering and analysis via Gene Ontology/Kyoto Encyclopedia of Genes and Genomes and evaluating diagnostic markers using an ROC curve. Finally, verification methods such as quantitative reverse-transcription polymerase chain reaction (RT-qPCR) and western blotting are used to evaluate the accuracy of these markers (Fig. 5).
Serum and urine are becoming the main sample sources due to the noninvasiveness of their collection and their renewability. Changes in blood components are closely related to systemic diseases. Several studies have proven the potential of lncRNAs in serum as diagnostic tools in multiple systemic diseases (116). The lncRNA cholesterol cytochrome P450 family 1 subfamily B member 1 (CYP1B1)-AS1 promotes inflammation and apoptosis by targeting miR-18a-5p; CYP1B1-AS1 in serum serves as a biomarker for sepsis diagnosis and poor prognosis (117). Expression of circulating Linc01094 as detected by RT-qPCR can differentiate osteoporotic patients from healthy ones and those with osteoporotic fractures from those without (118). LncRNAs in urine are also extremely important in the diagnosis of systemic diseases. The exosomal lncRNA MALAT1 in urine is regarded as a diagnostic marker for Wilms tumor and high expression of SNHG16 can potentially serve as a diagnostic biological marker for bladder cancer (119,120). Expression of circRNAs is abundant, stable and conservative and specific to tissue and developmental stage, making them ideal biomarkers (121). CircRNAs in bodily fluids show strong potential as diagnostic markers in systemic diseases, including autoimmune diseases, cardiovascular diseases, cancers and ischemic stroke (122,123). Therefore, related studies mainly have focused on systemic diseases or those that pose a great threat to life. In summary, exploring the diagnostic ability of ncRNAs can aid early prevention, guide treatment and assess prognosis. Furthermore, it can improve survival rates and reduce social burdens.
At present, the diagnosis of periodontitis is mainly based on clinical measurements, including bleeding assessment, periodontal-pocket depth and X-ray. However, these indicators have certain deficiencies in the early diagnosis, staging, progression prediction and prognosis evaluation of the disease. In addition to serum, diagnostic markers for periodontitis can be found in saliva and gingival crevicular fluid (GCF). These samples are simple to obtain, can be taken noninvasively way to prevent further tissue damage during clinical diagnosis and are plentiful (124).
Current studies on ncRNAs as diagnostic markers for periodontitis are mainly limited to miRNAs, whose ability to serve as such has been proven (15,125). Due to their stable properties, the use of circRNAs as diagnostic markers for periodontitis warrants further exploration (Fig. 6).
Therapeutic potential of ncRNAs in periodontitis
NcRNAs play important regulatory roles in systemic diseases, but discovering the mechanisms underlying these roles should not be the endpoint of relevant research. The ultimate goal is to transform ncRNAs into clinical treatment, with the expectation of achieving safe, accurate gene therapy. There are mainly two strategies in ncRNA treatment: Targeting ncRNAs to inhibit their expression and function; and restoring the expression or function of the target ncRNA. Techniques may involve antisense oligonucleotides (ASOs), ASO therapeutic circRNAs, anti-microRNAs (anti-miRs), siRNAs, miRNA sponges, miRNA mimics, short hairpin RNAs and clustered regularly interspaced short palindromic repeats-Caspase-9-based gene editing (126).
Clinical application of ncRNAs must be accurate and safe. At present, the limited bioavailability of ncRNAs in vivo presents a major challenge to said clinical application (127). Therefore, selecting the appropriate ncRNA vector and system is vitally important. Nanoparticles (NPs), ncRNA modification and the oncolytic-adenovirus strategy have become research hotspots. A number of studies have focused on gene therapy using NPs to deliver relevant ncRNAs, including diseases of the digestive, respiratory and circulatory systems (128-130). Currently, siRNA or ASO drugs are the focus of research into nucleic acid drugs; no miRNA, lncRNA, or circRNA has been clinically employed. At present, gene intervention studies using biological vectors (adenovirus, adeno-associated virus and lentivirus) are limited to laboratory studies due to safety issues such as immune origin and gene recombination; a number of problems remain to be solved before human experiments can be conducted.
To date, synthetic carriers mainly include lipid, polymer and inorganic NPs, which have been explored for delivery of siRNA to the target site. Lipid NPs are excellent nucleic acid delivery systems, with strong tissue penetration and low cytotoxicity and immunogenicity (131). Tong et al (132) have optimized a lipid NP (LNP) formulation (C2) and modified the LNPs with the Angiopep-2 peptide. This work demonstrates the efficacy of a brain-targeted siRNA delivery system in glioblastoma multiforme treatment.
Polymer NPs can effectively wrap negatively charged nucleic acids mainly through their positive charge; their ability to alter charge, degradability and molecular weight makes them excellent delivery systems. Construction of both engineered exosomes and polymers as nucleic acid carriers should have the advantages of i) biocompatibility, ii) effective delivery of nucleic acids and avoidance of nucleic acid degradation, iii) delivery to target organs, iv) biodegradability, v) stable properties and vi) commercial production. Inorganic nanocarrier delivery systems mainly include gold, magnetic, mesoporous silica and calcium phosphate NPs.
As aforementioned, lncRNAs and circRNAs as nucleic acid drugs remain in the animal research stage. Administration of Lnc-RAINY encapsulated in Food and Drug Administration (FDA)-approved NPs into a lung cancer patient-derived xenograft model markedly reduces tumor progression, demonstrating therapeutic potential (133). An intranasal LNP delivery system carrying the circRNA Scm polycomb group protein homolog 1 (circSCMH1) promoted and enhanced sensorimotor and cognitive-function recovery in post-stroke mice (134). In recent years, nanomedicine has used biomaterials and biomedical-engineering innovations to overcome biological barriers and patient heterogeneity to achieve precise treatment and improve patient-specific therapeutic responses. At the same time, research on NP delivery systems and in particular nucleic acid therapy have entered a period of rapid development (135). In particular, the construction of the ASP8-PU lncRNA NRON system targeting osteoclasts is highly valuable (52). In this system, NRON mainly serves to inhibit osteoclast formation and reduce bone resorption. PU is a nanoscale polymer that can encapsulate nucleic acids via electrostatic interactions, offering excellent biocompatibility and effective transport capacity. The loading of asp-8 enables nanodrugs to directly target the surface of the absorbed bone, achieving periodontitis treatment in mice (136). Nevertheless, nucleic acid therapy is still not widely used in clinical settings, as it has failed to achieve safe and efficient treatment in a number of diseases and research into using it for periodontitis has also not progressed. Although the regulatory mechanism of new ncRNAs in periodontitis has not been fully explored, the mainstream view is that local administration is the most suitable method of treatment. Consequently, the study of an appropriate NP delivery system to achieve targeted treatment of periodontitis has obvious clinical-application prospects (Fig. 7).
Perspectives
Due to continuous innovations in sequencing technology, research on the periodontitis transcriptome has been greatly expanded. A number of ncRNA regulatory mechanisms have been revealed, unknown ncRNAs have been discovered and identified, a ceRNA network under different disease states has been constructed and a more comprehensive profile of ncRNA has been drawn. Research on ncRNAs in periodontitis is gradually increasing. However, the current dataset on the role of ncRNA in periodontitis has various limitations, such as incomplete clinical information, differences in disease progression and incomplete data on different types of periodontitis. No single database contains all ncRNA datasets relevant to this disease. A relatively complete and large database, The Cancer Genome Atlas database (TCGA), has been constructed in the field of tumor research. TCGA contains the genome maps of almost all human cancers. Systematic analysis of this database allows key cancer genes to be found, which is of great significance for understanding the mechanisms governing the occurrence and development of cancer cells and realizing early diagnosis and targeted therapy. It is necessary to build a similar gene information system for periodontitis. As more studies are conducted, additional key gene information in periodontitis is expected to be revealed and exploration of same will help researchers gain a deeper understanding of the disease's pathogenesis and further guide clinical diagnosis and treatment.
LncRNAs and circRNAs, newly discovered types of ncRNAs, are known to play key roles in systemic diseases through a variety of molecules and pathways. However, studies on their mechanisms in the field of periodontitis are limited to those ncRNAs that play regulatory roles as ceRNAs; additional mechanisms of action remain to be explored. Summaries of the ncRNAs relevant to the pathogenesis of the periodontitis are provided in Tables I and II. As ncRNAs regulate a variety of complex biological behaviors in systemic diseases, research on these RNAs in the field of periodontitis must be expanded beyond immune inflammation, autophagy, bone homeostasis and cell proliferation. It is worth affirming that the regulatory mechanisms of ncRNA in periodontitis are being explored more widely and deeply. Greater future recognition of ncRNAs' clinical applications as diagnostic markers and therapeutic targets in periodontitis is needed.
As the role of ncRNAs in periodontitis becomes more detailed, researchers have begun to pay attention to their potential as biomarkers and nucleic acid drugs in clinical applications. At present, the diagnosis of periodontitis is mainly based on clinical symptoms, which is relatively subjective and thus deficient for early screening and diagnosis. As biomarkers, ncRNAs can overcome the limitations of these clinical indicators and provide more objective, accurate and timely results. Using ncRNA in saliva or GCF as a diagnostic marker not only is noninvasive but also facilitates objective evaluation of patient progress. As aforementioned, ncRNAs are considered to have good application prospects as targets for the early diagnosis and treatment of periodontitis.
Currently, nucleic acid drugs approved by the FDA are mainly siRNAs or ASOs; no miRNA, lncRNA, or circRNA has been applied in clinical treatment. As nucleic acid drugs, ncRNAs have great potential and improved advantages in the treatment of various diseases. As naturally occurring molecules in human cells, they have all the required mechanisms for processing and downstream target selection. In addition, ncRNAs work by targeting multiple genes in a single pathway, leading to a broader and more specific response. At present, the clinical application of ncRNA drugs is still limited by the problems of off-target effects, delivery failure and tolerance. Successful application of ncRNA-based therapies requires a multidisciplinary effort, including technological advances in molecular biology, immunology, pharmacology, chemistry and nanotechnology. Despite the challenges, ncRNA-based drug therapies will eventually become available as technology and research advance in the treatment of periodontitis.
Availability of data and materials
Not applicable.
Authors' contributions
YF reviewed the literature and wrote the manuscript. XG collected and analyzed the existing data from the previously published research articles. YY and WQ wrote and revised the manuscript and constructed figures. ZC and FF conceived the study and revised the manuscript. Data authentication is not applicable. All authors read and approved the final manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Acknowledgments
Not applicable.
Funding
The present review was supported by the National Natural Science Foundation of China (grant nos. 82470975, 82270982), the Natural Science Foundation of Guangdong Province (grant no. 2024A1515010840) and Science and Technology Projects in Guangzhou (grant no. 2024B03J0667).
References
GBD 2021 Oral Disorders Collaborators: Trends in the global, regional, and national burden of oral conditions from 1990 to 2021: A systematic analysis for the global burden of disease study 2021. Lancet. 405:897–910. 2025. View Article : Google Scholar : PubMed/NCBI | |
Wu L, Zhang SQ, Zhao L, Ren ZH and Hu CY: Global, regional, and national burden of periodontitis from 1990 to 2019: Results from the global burden of disease study 2019. J Periodontol. 93:1445–1454. 2022. View Article : Google Scholar : PubMed/NCBI | |
Cui Y, Tian G, Li R, Shi Y, Zhou T and Yan Y: Epidemiological and sociodemographic transitions of severe periodontitis incidence, prevalence, and disability-adjusted life years for 21 world regions and globally from 1990 to 2019: An age-period-cohort analysis. J Periodontol. 94:193–203. 2023. View Article : Google Scholar | |
Valverde A, George A, Nares S and Naqvi AR: Emerging therapeutic strategies targeting bone signaling pathways in periodontitis. J Periodontal Res. 60:101–120. 2025. View Article : Google Scholar : | |
Crick F: Central dogma of molecular biology. Nature. 227:561–563. 1970. View Article : Google Scholar : PubMed/NCBI | |
Fabbri M, Girnita L, Varani G and Calin GA: Decrypting noncoding RNA interactions, structures, and functional networks. Genome Res. 29:1377–1388. 2019. View Article : Google Scholar : PubMed/NCBI | |
Alghazali T, Ahmed AT, Hussein UAR, Sanghvi G, Uthirapathy S, Edan RT, Lal M, Shit D, Naidu KS and Al-Hamairy AK: Noncoding RNA (ncRNA)-mediated regulation of TLRs: Critical regulator of inflammation in tumor microenvironment. Med Oncol. 42:1442025. View Article : Google Scholar : PubMed/NCBI | |
Fang F, Zhang K, Chen Z and Wu B: Noncoding RNAs: New insights into the odontogenic differentiation of dental tissue-derived mesenchymal stem cells. Stem Cell Res Ther. 10:2972019. View Article : Google Scholar : PubMed/NCBI | |
Kristensen L, Sandersen MS, Stagsted LVW, Ebbesen KK, Hansen TB and Kjems J: The biogenesis, biology and characterization of circular RNAs. Nat Rev Genet. 20:675–691. 2019. View Article : Google Scholar : PubMed/NCBI | |
Chen Q, Meng X, Liao Q and Chen M: Versatile interactions and bioinformatics analysis of noncoding RNAs. Brief Bioinform. 20:1781–1794. 2019. View Article : Google Scholar | |
Micheel J, Safrastyan A and Wollny D: Advances in non-coding RNA sequencing. Noncoding RNA. 7:702021.PubMed/NCBI | |
Wolfien M, Brauer DL, Bagnacani A and Wolkenhauer O: Workflow development for the functional characterization of ncRNAs. Methods Mol Biol. 1912:111–132. 2019. View Article : Google Scholar : PubMed/NCBI | |
Rinaldi S, Moroni E, Rozza R and Magistrato A: Frontiers and challenges of computing ncRNAs biogenesis, function and modulation. J Chem Theory Comput. 20:993–1018. 2024. View Article : Google Scholar : PubMed/NCBI | |
Pekáčová A, Baloun J, Švec X and Šenolt L: Non-coding RNAs in diseases with a focus on osteoarthritis. Wiley Interdiscip Rev RNA. 14:e17562023. View Article : Google Scholar | |
Adamouli D, Marasli C and Bobetsis YA: The expression patterns of non-coding RNAs in periodontal disease. Dent J (Basel). 12:1592024. View Article : Google Scholar : PubMed/NCBI | |
Taheri M, Khoshbakht T, Hussen BM, Abdullah ST, Ghafouri-Fard S and Sayad A: Emerging role of miRNAs in the pathogenesis of periodontitis. Curr Stem Cell Res Ther. 19:427–448. 2024. View Article : Google Scholar | |
Ni H, Ge Y, Zhuge Y, Liu X, Chen H, Liu J, Li W, Wang X, Shen G, Wang Q, et al: LncRNA MIR181A1HG deficiency attenuates vascular inflammation and atherosclerosis. Circ Res. 136:862–883. 2025. View Article : Google Scholar : PubMed/NCBI | |
Shi W, Song J, Weiner JM III, Chopra A, Dommisch H, Beule D and Schaefer AS: lncRNA CDKN2B-AS1 regulates collagen expression. Hum Genet. 143:907–919. 2024. View Article : Google Scholar : PubMed/NCBI | |
Huang W, Li X, Huang C, Tang Y, Zhou Q and Chen W: LncRNAs and rheumatoid arthritis: From identifying mechanisms to clinical investigation. Front Immunol. 12:8077382022. View Article : Google Scholar : PubMed/NCBI | |
Carpenter S: Long noncoding RNA: Novel links between gene expression and innate immunity. Virus Res. 212:137–145. 2016. View Article : Google Scholar | |
Amroodi MN, Maghsoudloo M, Amiri S, Mokhtari K, Mohseni P, Pourmarjani A, Jamali B, Khosroshahi EM, Asadi S, Tabrizian P, et al: Unraveling the molecular and immunological landscape: Exploring signaling pathways in osteoporosis. Biomed Pharmacother. 177:1169542024. View Article : Google Scholar : PubMed/NCBI | |
Luo R, Yao Y, Chen Z and Sun X: An examination of the LPS-TLR4 immune response through the analysis of molecular structures and protein-protein interactions. Cell Commun Signal. 23:1422025. View Article : Google Scholar : PubMed/NCBI | |
Liu W, Zheng Y, Chen B, Ke T and Shi Z: LncRNA papillary thyroid carcinoma susceptibility candidate 3 (PTCSC3) regulates the proliferation of human periodontal ligament stem cells and toll-like receptor 4 (TLR4) expression to improve periodontitis. BMC Oral Health. 19:1082019. View Article : Google Scholar : PubMed/NCBI | |
Wangzhou K, Gong L, Liu C, Tan Y, Chen J, Li C, Lai Z and Hao C: LncRNA MAFG-AS1 regulates human periodontal ligament stem cell proliferation and Toll-like receptor 4 expression. Oral Dis. 26:1302–1307. 2020. View Article : Google Scholar : PubMed/NCBI | |
Ahmad I, Naqvi RA, Valverde A and Naqvi AR: LncRNA MALAT1/microRNA-30b axis regulates macrophage polarization and function. Front Immunol. 14:12148102023. View Article : Google Scholar : PubMed/NCBI | |
Zhang L, Lv H, Cui Y and Shi R: The role of long non-coding RNA (lncRNA) nuclear paraspeckle assembly transcript 1 (NEAT1) in chronic periodontitis progression. Bioengineered. 13:2336–2345. 2022. View Article : Google Scholar : PubMed/NCBI | |
Ruan D, Wu C and Zhang Y and Zhang Y: LncRNA LOXL1-AS1 inhibits proliferation of PDLSCs and downregulates IL-1β in periodontitis patients. J Periodontal Res. 57:324–331. 2022. View Article : Google Scholar | |
Cheng L, Fan Y, Cheng J, Wang J, Liu Q and Feng Z: Long non-coding RNA ZFY-AS1 represses periodontitis tissue inflammation and oxidative damage via modulating microRNA-129-5p/DEAD-Box helicase 3 X-linked axis. Bioengineered. 13:12691–12705. 2022. View Article : Google Scholar : PubMed/NCBI | |
Han Y, Huang Y, Yang Q, Jia L, Zheng Y and Li W: Long non-coding RNA SNHG5 mediates periodontal inflammation through the NF-κB signalling pathway. J Clin Periodontol. 49:1038–1051. 2022. View Article : Google Scholar : PubMed/NCBI | |
Wang H, Qiao X, Zhang C, Hou J and Qi S: Long non-coding RNA LINC00616 promotes ferroptosis of periodontal ligament stem cells via the microRNA-370/transferrin receptor axis. Bioengineered. 13:13070–13081. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zhou M, Hu H, Han Y, Li J, Zhang Y, Tang S, Yuan Y and Zhang X: Long non-coding RNA 01126 promotes periodontitis pathogenesis of human periodontal ligament cells via miR-518a-5p/HIF-1α/MAPK pathway. Cell Prolif. 54:e129572021. View Article : Google Scholar | |
Tang S, Zhong Y, Li J, Ji P and Zhang X: Long intergenic non-coding RNA 01126 activates IL-6/JAK2/STAT3 pathway to promote periodontitis pathogenesis. Oral Dis. 31:193–205. 2025. View Article : Google Scholar | |
Li L, Qin W, Ye T, Wang C, Qin Z, Ma Y, Mu Z, Jiao K, Tay FR, Niu W and Niu L: Bioactive Zn-V-Si-Ca glass nanoparticle hydrogel microneedles with antimicrobial and antioxidant properties for bone regeneration in diabetic periodontitis. ACS Nano. 19:7981–7995. 2025. View Article : Google Scholar : PubMed/NCBI | |
Sun R, Duan D and Li R: Transcriptome sequencing identifies abnormal lncRNAs and mRNAs and reveals potentially hub immune-related mRNA in osteoporosis with vertebral fracture. Clin Interv Aging. 19:203–217. 2024. View Article : Google Scholar : PubMed/NCBI | |
Li H, Zheng F, Tao A, Wu T, Zhan X, Tang H, Cui X, Ma Z, Li C, Jiang J and Wang Y: LncRNA H19 promotes osteoclast differentiation by sponging miR-29c-3p to increase expression of cathepsin K. Bone. 192:1173402025. View Article : Google Scholar | |
Li Q, Zhou H, Wang C and Zhu Z: Long non-coding RNA Linc01133 promotes osteogenic differentiation of human periodontal ligament stem cells via microRNA-30c/bone gamma-carboxyglutamate protein axis. Bioengineered. 13:9602–9612. 2022.PubMed/NCBI | |
Dong Y, Feng S and Dong F: Maternally-expressed gene 3 (MEG3)/miR-143-3p regulates injury to periodontal ligament cells by mediating the AKT/inhibitory κB kinase (IKK) pathway. Med Sci Monit. 26:e9224862020. View Article : Google Scholar | |
Lai L, Wang Z, Ge Y, Qiu W, Wu B, Fang F, Xu H and Chen Z: Comprehensive analysis of the long noncoding RNA-associated competitive endogenous RNA network in the osteogenic differentiation of periodontal ligament stem cells. BMC Genomics. 23:12022. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Liu C, Zhang A, Yin S, Wang T, Wang Y, Wang M, Liu Y, Ying Q, Sun J, et al: Down-regulation of long non-coding RNA MEG3 suppresses osteogenic differentiation of periodontal ligament stem cells (PDLSCs) through miR-27a-3p/IGF1 axis in periodontitis. Aging (Albany NY). 11:5334–5350. 2019. View Article : Google Scholar : PubMed/NCBI | |
Bian M, Yu Y, Li Y, Zhou Z, Wu X, Ye X and Yu J: Upregulating the expression of LncRNA ANRIL promotes osteogenesis via the miR-7-5p/IGF-1R axis in the inflamed periodontal ligament stem cells. Front Cell Dev Biol. 9:6044002021. View Article : Google Scholar : PubMed/NCBI | |
Zhong X and Wang H: LncRNA JHDM1D-AS1 promotes osteogenic differentiation of periodontal ligament cells by targeting miR-532-5p to activate IGF1R signaling. J Periodontal Res. 59:220–230. 2024. View Article : Google Scholar | |
Wang X and Wang Y: LncRNA DCST1-AS1 inhibits PDLCs' proliferation in periodontitis and may bind with miR-21 precursor to upregulate PLAP-1. J Periodontal Res. 56:256–264. 2021. View Article : Google Scholar : PubMed/NCBI | |
Gu Y and Bai Y: LncRNA MALAT1 promotes osteogenic differentiation through the miR-93-5p/SMAD5 axis. Oral Dis. 30:2398–2409. 2024. View Article : Google Scholar | |
Guo J and Zheng M: The regulation mechanism of LINC00707 on the osteogenic differentiation of human periodontal ligament stem cells. J Mol Histol. 53:13–26. 2022. View Article : Google Scholar | |
Zhang X, Yan Q, Liu X, Gao J, Xu Y, Jin Z and Qin W: LncRNA00638 promotes the osteogenic differentiation of periodontal mesenchymal stem cells from periodontitis patients under static mechanical strain. Stem Cell Res Ther. 14:1772023. View Article : Google Scholar : PubMed/NCBI | |
Wang Z, Wang D, Guo S, Zhuo Q, Jiang D and Yu Z: Long noncoding RNA distal-less homeobox 2 antisense 1 restrains inflammatory response and apoptosis of periodontal ligament cells by binding with microRNA-330-3p to regulate Ro60, Y RNA binding protein. Arch Oral Biol. 133:1052982022. View Article : Google Scholar | |
Wang S and Duan Y: LncRNA OIP5-AS1 inhibits the lipopolysaccharide-induced inflammatory response and promotes osteogenic differentiation of human periodontal ligament cells by sponging miR-92a-3p. Bioengineered. 13:12055–12066. 2022. View Article : Google Scholar : PubMed/NCBI | |
Yao S, Lu H, Zhou T, Jiang Q, Jiang C, Hu W, Li M, Tan CP, Feng Y, Du Q, et al: Sciadonic acid attenuates high-fat diet-induced bone metabolism disorders in mice. Food Funct. 15:4490–4502. 2024. View Article : Google Scholar : PubMed/NCBI | |
Zhou Z, Zhan C, Li W, Luo W, Liu Y, He F, Tian Y, Lin Z and Song Z: Monocytic myeloid-derived suppressor cells contribute to the exacerbation of bone destruction in periodontitis. J Transl Med. 23:2172025. View Article : Google Scholar : PubMed/NCBI | |
Zhang X, Zhao Y, Zhao Z, Han X and Chen Y: Knockdown of DANCR reduces osteoclastogenesis and root resorption induced by compression force via Jagged1. Cell Cycle. 18:1759–1769. 2019. View Article : Google Scholar : PubMed/NCBI | |
Feng J, Tan A, Li W and Zheng Y: Small nucleolar RNA host gene 5 plays a role in orthodontic tooth movement by inhibiting osteoclast differentiation. Orthod Craniofac Res. 27:775–784. 2024. View Article : Google Scholar : PubMed/NCBI | |
Jin F, Li J, Zhang YB, Liu X, Cai M, Liu M, Li M, Ma C, Yue R, Zhu Y, et al: A functional motif of long noncoding RNA Nron against osteoporosis. Nat Commun. 12:33192021. View Article : Google Scholar : PubMed/NCBI | |
Li R, Huang Z and Chen M: Long non-coding RNA EPB41L4A-AS1 serves as a diagnostic marker for chronic periodontitis and regulates periodontal ligament injury and osteogenic differentiation by targeting miR-214-3p/YAP1. J Inflamm Res. 18:2483–2497. 2025. View Article : Google Scholar : PubMed/NCBI | |
Chen P, Huang Y, Wang Y, Li S, Chu H and Rong M: MALAT1 overexpression promotes the proliferation of human periodontal ligament stem cells by upregulating fibroblast growth factor 2. Exp Ther Med. 18:1627–1632. 2019.PubMed/NCBI | |
Wang Y, Sun Y, Zheng P, Cai C, Jiang Y, Zhang H, Li Z and Cai Q: Long non-coding RNAs mortal obligate RNA transcript regulates the proliferation of human periodontal ligament stem cells and affects the recurrence of periodontitis. Arch Oral Biol. 105:1–4. 2019. View Article : Google Scholar : PubMed/NCBI | |
Wu X, Cao Z, Chen H, Ou Q, Huang X and Wang Y: Downregulation of Linc-RNA activator of myogenesis lncRNA participates in FGF2-mediated proliferation of human periodontal ligament stem cells. J Periodontol. 91:422–427. 2020. View Article : Google Scholar | |
Han Y, Wang F, Shao L, Huang P and Xu Y: LncRNA TUG1 mediates lipopolysaccharide-induced proliferative inhibition and apoptosis of human periodontal ligament cells by sponging miR-132. Acta Biochim Biophys Sin (Shanghai). 51:1208–1215. 2019. View Article : Google Scholar : PubMed/NCBI | |
Yu W, Gu Q, Wu D, Zhang W, Li G, Lin L, Lowe JM, Hu S, Li TW, Zhou Z, et al: Identification of potentially functional circRNAs and prediction of circRNA-miRNA-mRNA regulatory network in periodontitis: Bridging the gap between bioinformatics and clinical needs. J Periodontal Res. 57:594–614. 2022. View Article : Google Scholar : PubMed/NCBI | |
Li J and Xie R: Circular RNA expression profile in gingival tissues identifies circ_0062491 and circ_0095812 as potential treatment targets. J Cell Biochem. 120:14867–14874. 2019. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Li Y, Hong F and Ning H: Circ_0062491 alleviates LPS-induced apoptosis and inflammation in periodontitis by regulating miR-498/SOCS6 axis. Innate Immun. 28:174–184. 2022. View Article : Google Scholar : PubMed/NCBI | |
Bian Y, Yu J, Liu Y, Shi Y, Hou Y and Liu X: CircPVT1 promotes periodontitis progression by regulating miR-24-3p/HIF1AN pathway. J Stomatol Oral Maxillofac Surg. 126:1021982025. View Article : Google Scholar : PubMed/NCBI | |
Zhao XQ, Ao CB and Yan YT: The circular RNA circ_0099630/miR-940/receptor-associated factor 6 regulation cascade modulates the pathogenesis of periodontitis. J Dent Sci. 17:1566–1576. 2022. View Article : Google Scholar : PubMed/NCBI | |
Li W, Zhang Z, Li Y and Wang Z: Abnormal hsa_circ_0003948 expression affects chronic periodontitis development by regulating miR-144-3p/NR2F2/PTEN signaling. J Periodontal Res. 57:316–323. 2022. View Article : Google Scholar | |
Pan J, Zhao L, Liu J and Wang G: Inhibition of circular RNA circ_0138959 alleviates pyroptosis of human gingival fibroblasts via the microRNA-527/caspase-5 axis. Bioengineered. 13:1908–1920. 2022. View Article : Google Scholar : PubMed/NCBI | |
Gu X, Li M, Jin Y, Liu D and Wei F: Identification and integrated analysis of differentially expressed lncRNAs and circRNAs reveal the potential ceRNA networks during PDLSC osteogenic differentiation. BMC Genet. 18:1002017. View Article : Google Scholar : PubMed/NCBI | |
Qi Y, Lin C, Zhao C and Wu Y: Circ_0003072 mediates the pro-osteogenic differentiation effect of betulinic acid on human periodontal ligament stem cells. Int Dent J. 75:1390–1399. 2025. View Article : Google Scholar : | |
Xiao T, Shi Y, Ye Y, Wang J, Wang W, Yu H, Yan M and Yu J: Circ-SPATA13 regulates the osteogenic differentiation of human periodontal ligament stem cells through the miR-485-5p_R + 1/BMP7 axis. Cell Signal. 127:1115612025. View Article : Google Scholar | |
Ye Y, Ke Y, Liu L, Xiao T and Yu J: CircRNA FAT1 regulates osteoblastic differentiation of periodontal ligament stem cells via miR-4781-3p/SMAD5 pathway. Stem Cells Int. 2021:51774882021. View Article : Google Scholar | |
Wang C, Gong J, Li D and Xing X: circ_0062491 alleviates periodontitis via the miR-142-5p/IGF1 axis. Open Med (Wars). 17:638–647. 2022. View Article : Google Scholar : PubMed/NCBI | |
Liu J, Liu R, Wang H, Zhang Z, Wang J and Wei F: CircPRKD3/miR-6783-3p responds to mechanical force to facilitate the osteogenesis of stretched periodontal ligament stem cells. J Orthop Surg Res. 19:2572024. View Article : Google Scholar : PubMed/NCBI | |
Li M, Du M, Wang Y, Zhu J, Pan J, Cao Z and He H: CircRNA Lrp6 promotes cementoblast differentiation via miR-145a-5p/Zeb2 axis. J Periodontal Res. 56:1200–1212. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zheng J, Zhu X, He Y, Hou S, Liu T, Zhi K, Hou T and Gao L: CircCDK8 regulates osteogenic differentiation and apoptosis of PDLSCs by inducing ER stress/autophagy during hypoxia. Ann N Y Acad Sci. 1485:56–70. 2021. View Article : Google Scholar | |
Xie L, Ren X, Yang Z, Zhou T, Zhang M, An W and Guan Z: Exosomal circ_0000722 derived from periodontal ligament stem cells undergoing osteogenic differentiation promotes osteoclastogenesis. Int Immunopharmacol. 128:1115202024. View Article : Google Scholar : PubMed/NCBI | |
Wang J, Wang Z, Huang M, Zhang Y and Xu L: Circ_0099630 participates in SPRY1-mediated repression in periodontitis. Int Dent J. 73:136–143. 2023. View Article : Google Scholar : | |
Yu B, Hu J, Li Q and Wang F: CircMAP3K11 contributes to proliferation, apoptosis and migration of human periodontal ligament stem cells in inflammatory microenvironment by regulating TLR4 via miR-511 sponging. Front Pharmacol. 12:6333532021. View Article : Google Scholar : PubMed/NCBI | |
Wang H, Gao S and Dissanayaka WL: Circ_0003764 regulates the osteogenic differentiation of periodontal ligament stem cells. Int Dent J. 74:1110–1119. 2024. View Article : Google Scholar : PubMed/NCBI | |
Wang F, Chen X, Han Y, Xi S and Wu G: circRNA CDR1as regulated the proliferation of human periodontal ligament stem cells under a lipopolysaccharide-induced inflammatory condition. Mediators Inflamm. 2019:16253812019. View Article : Google Scholar : PubMed/NCBI | |
Li X, Zheng Y, Zheng Y, Huang Y, Zhang Y, Jia L and Li W: Circular RNA CDR1as regulates osteoblastic differentiation of periodontal ligament stem cells via the miR-7/GDF5/SMAD and p38 MAPK signaling pathway. Stem Cell Res Ther. 9:2322018. View Article : Google Scholar : PubMed/NCBI | |
Gu X, Li X, Jin Y, Zhang Z, Li M, Liu D and Wei F: CDR1as regulated by hnRNPM maintains stemness of periodontal ligament stem cells via miR-7/KLF4. J Cell Mol Med. 25:4501–4515. 2021. View Article : Google Scholar : PubMed/NCBI | |
Ma C, Gu R, Wang X, He S, Bai J, Zhang L, Zhang J, Li Q, Qu L, Xin W, et al: circRNA CDR1as promotes pulmonary artery smooth muscle cell calcification by upregulating CAMK2D and CNN3 via sponging miR-7-5p. Mol Ther Nucleic Acids. 22:530–541. 2020. View Article : Google Scholar : PubMed/NCBI | |
Zhao F, Chen T and Jiang N: CDR1as/miR-7/CKAP4 axis contributes to the pathogenesis of abdominal aortic aneurysm by regulating the proliferation and apoptosis of primary vascular smooth muscle cells. Exp Ther Med. 19:3760–3766. 2020.PubMed/NCBI | |
Yang W, Yang X, Wang X, Gu J, Zhou D, Wang Y, Yin B, Guo J and Zhou M: Silencing CDR1as enhances the sensitivity of breast cancer cells to drug resistance by acting as a miR-7 sponge to down-regulate REGγ. J Cell Mol Med. 23:4921–4932. 2019. View Article : Google Scholar : PubMed/NCBI | |
Yu L, Gong X, Sun L, Zhou Q, Lu B and Zhu L: The circular RNA Cdr1as Act as an oncogene in hepatocellular carcinoma through targeting miR-7 expression. PLoS One. 11:e01583472016. View Article : Google Scholar : PubMed/NCBI | |
Kwon EJ, Kim HJ, Woo BH, Joo JY, Kim YH and Park HR: Profiling of plasma-derived exosomal RNA expression in patients with periodontitis: A pilot study. Oral Dis. 29:1726–1737. 2023. View Article : Google Scholar | |
Duran-Pinedo AE, Yost S and Frias-Lopez J: Small RNA transcriptome of the oral microbiome during periodontitis progression. Appl Environ Microbiol. 81:6688–6699. 2015. View Article : Google Scholar : PubMed/NCBI | |
Xiao J, Deng Y, Xie J, Liu H, Yang Q, Zhang Y, Huang X and Cao Z: Apoptotic vesicles from macrophages exacerbate periodontal bone resorption in periodontitis via delivering miR-143-3p targeting Igfbp5. J Nanobiotechnology. 22:6582024. View Article : Google Scholar : PubMed/NCBI | |
Wang XW, Liu CX, Chen LL and Zhang QC: RNA structure probing uncovers RNA structure-dependent biological functions. Nat Chem Biol. 17:755–766. 2021. View Article : Google Scholar : PubMed/NCBI | |
Martens L, Rühle F, Witten A, Meder B, Katus HA, Arbustini E, Hasenfuß G, Sinner MF, Kääb S, Pankuweit S, et al: A genetic variant alters the secondary structure of the lncRNA H19 and is associated with dilated cardiomyopathy. RNA Biol. 18(Suppl 1): S409–S415. 2021. View Article : Google Scholar | |
Zhou H, Hao X, Zhang P and He S: Noncoding RNA mutations in cancer. Wiley Interdiscip Rev RNA. 14:e18122023. View Article : Google Scholar : PubMed/NCBI | |
Bose E, Xiong S and Jones AN: Probing RNA structure and dynamics using nanopore and next generation sequencing. J Biol Chem. 300:1073172024. View Article : Google Scholar : PubMed/NCBI | |
Takizawa N: RNA structure determination by high-throughput structural analysis. Methods Mol Biol. 2586:217–231. 2023. View Article : Google Scholar : PubMed/NCBI | |
Wang S, Sun Z, Lei Z and Zhang HT: RNA-binding proteins and cancer metastasis. Semin Cancer Biol. 86:748–768. 2022. View Article : Google Scholar : PubMed/NCBI | |
Xu M, Gan D, Zhang XY, He XT, Wu RX, Yin Y, Jin R, Li L, Tan YJ, Chen FM, et al: SLC30A4-AS1 mediates the senescence of periodontal ligament stem cells in inflammatory environments via the alternative splicing of TP53BP1. Cell Prolif. 58:e137782025. View Article : Google Scholar | |
Hu A, Xiao F, Wu W, Xu H and Su J: LincRNA-EPS inhibits caspase-11 and NLRP3 inflammasomes in gingival fibroblasts to alleviate periodontal inflammation. Cell Prolif. 57:e135392024. View Article : Google Scholar | |
Xu K, Li YD, Ren LY, Song HL, Yang QY and Xu DL: Long non-coding RNA X-inactive specific transcript (XIST) interacting with USF2 promotes osteogenic differentiation of periodontal ligament stem cells through regulation of WDR72 transcription. J Periodontal Res. 58:1235–1247. 2023. View Article : Google Scholar : PubMed/NCBI | |
Li X, Tian BM, Deng DK, Liu F, Zhou H, Kong DQ, Qu HL, Sun LJ, He XT and Chen FM: LncRNA GACAT2 binds with protein PKM1/2 to regulate cell mitochondrial function and cementogenesis in an inflammatory environment. Bone Res. 10:292022. View Article : Google Scholar : PubMed/NCBI | |
Long L, Zhang C, He Z, Liu O, Yang H and Fan Z: LncRNA NR_045147 modulates osteogenic differentiation and migration in PDLSCs via ITGB3BP degradation and mitochondrial dysfunction. Stem Cells Transl Med. 14:szae0882025. View Article : Google Scholar : | |
Lu Y, Ruan X, Xiao G, Dai Y, Li G, Cai G, Zheng L, Guan Z, Sun W and Wang H: Lockd enhances mandibular mesenchymal stem cell proliferation while inhibiting osteogenic capability via binding with SUZ12 in the inflammatory microenvironment. J Clin Periodontol. 52:171–185. 2025. View Article : Google Scholar | |
Yan H and Bu P: Non-coding RNA in cancer. Essays Biochem. 65:625–639. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wang J, Zhu S, Meng N, He Y, Lu R and Yan GR: ncRNA-encoded peptides or proteins and cancer. Mol Ther. 27:1718–1725. 2019. View Article : Google Scholar : PubMed/NCBI | |
Tan S, Yang W, Ren Z, Peng Q, Xu X, Jiang X, Wu Z, Oyang L, Luo X, Lin J, et al: Noncoding RNA-encoded peptides in cancer: biological functions, posttranslational modifications and therapeutic potential. J Hematol Oncol. 18:202025. View Article : Google Scholar : PubMed/NCBI | |
Lin Y, Wang Y, Li L and Zhang K: Coding circular RNA in human cancer. Genes Dis. 12:1013472024. View Article : Google Scholar | |
Zhang Y: LncRNA-encoded peptides in cancer. J Hematol Oncol. 17:662024. View Article : Google Scholar : PubMed/NCBI | |
Li Q, Guo G, Chen Y, Lu L, Li H, Zhou Z, Guo J, Gan X, Hu Y, Li Q, et al: HCP5 derived novel microprotein triggers progression of gastric cancer through regulating ferroptosis. Adv Sci (Weinh). 11:e24070122024. View Article : Google Scholar : PubMed/NCBI | |
Zheng X, Wang M, Liu S, Chen H, Li Y, Yuan F, Yang L, Qiu S, Wang H, Xie Z and Xiang M: A lncRNA-encoded mitochondrial micropeptide exacerbates microglia-mediated neuroinflammation in retinal ischemia/reperfusion injury. Cell Death Dis. 14:1262023. View Article : Google Scholar : PubMed/NCBI | |
Xu N, Jiang J, Jiang F, Dong G, Meng L, Wang M, Chen J, Li C, Shi Y, He S and Li R: CircCDC42-encoded CDC42-165aa regulates macrophage pyroptosis in Klebsiella pneumoniae infection through Pyrin inflammasome activation. Nat Commun. 15:57302024. View Article : Google Scholar : PubMed/NCBI | |
Liang Y, Lv D, Liu K, Yang L, Shu H, Wen L, Lv C, Sun Q, Yin J, Liu H, et al: MicroProteinDB: A database to provide knowledge on sequences, structures and function of ncRNA-derived microproteins. Comput Biol Med. 177:1086602024. View Article : Google Scholar : PubMed/NCBI | |
He S, Yang S, Zhang Y, Li X, Gao D, Zhong Y, Cao L, Ma H, Liu Y, Li G, et al: LncRNA ODIR1 inhibits osteogenic differentiation of hUC-MSCs through the FBXO25/H2BK120ub/H3K4me3/OSX axis. Cell Death Dis. 10:9472019. View Article : Google Scholar : PubMed/NCBI | |
Pang Y, Liu Z, Han H, Wang B, Li W, Mao C and Liu S: Peptide SMIM30 promotes HCC development by inducing SRC/YES1 membrane anchoring and MAPK pathway activation. J Hepatol. 73:1155–1169. 2020. View Article : Google Scholar : PubMed/NCBI | |
Chen S, Tang M, Yu X, Qian W, Xu Y, Li J, Wu G and Zhang S: A microprotein encoded by LINC00263 promotes breast cancer osteolytic bone metastasis by inducing osteoclastogenesis and inhibiting osteoclast ferroptosis. Oncogene. 44:2201–2216. 2025. View Article : Google Scholar : PubMed/NCBI | |
Gonzalez-Fernandez J, Zaragozano S, Monteagudo-Sánchez A, Simon C and Vilella F: Single-cell technology: The key to an improved understanding of the human endometrium in health and disease. Am J Obstet Gynecol. 232(Suppl 4): S43–S53. 2025. View Article : Google Scholar : PubMed/NCBI | |
Gulati GS, D'Silva JP, Liu Y, Wang L and Newman AM: Profiling cell identity and tissue architecture with single-cell and spatial transcriptomics. Nat Rev Mol Cell Biol. 26:11–31. 2025. View Article : Google Scholar | |
Zhu ZX, Liu Y, Wang J, Xie Y, Li RY, Ma Q, Tu Q, Melhem NA, Couldwell S, El-Araby RE, et al: A novel lncRNA-mediated epigenetic regulatory mechanism in periodontitis. Int J Biol Sci. 19:5187–5203. 2023. View Article : Google Scholar : PubMed/NCBI | |
Yang Q, Wang M, Xu J, Yu D, Li Y, Chen Y and Zhang X, Zhang J, Gu J and Zhang X: LINC02159 promotes non-small cell lung cancer progression via ALYREF/YAP1 signaling. Mol Cancer. 22:1222023. View Article : Google Scholar : PubMed/NCBI | |
Sur S, Pal JK, Shekhar S, Bafna P and Bhattacharyya R: Emerging role and clinical applications of circular RNAs in human diseases. Funct Integr Genomics. 25:772025. View Article : Google Scholar : PubMed/NCBI | |
Jiang X, Liu Z, Wang H and Wang L: Discovery of lncRNA-based ProsRISK score in serum as potential biomarkers for improved accuracy of prostate cancer detection. J Cell Mol Med. 29:e705552025. View Article : Google Scholar : PubMed/NCBI | |
Xu L, Li J, Li L, Zhang Q, Feng Q and Bai L: LncRNA CYP1B1-AS1 as a clinical biomarker exacerbates sepsis inflammatory response via targeting miR-18a-5p. BMC Immunol. 26:322025. View Article : Google Scholar | |
Xu J, Tian Z, Huang L and Yu Y: LINC01094 as a diagnostic marker of osteoporotic fractures is involved in fracture healing. J Endocrinol. 265:e2500082025. View Article : Google Scholar : PubMed/NCBI | |
Liu C, Xu P, Shao S, Wang F, Zheng Z, Li S, Liu W and Li G: The value of urinary exosomal lncRNA SNHG16 as a diagnostic biomarker for bladder cancer. Mol Biol Rep. 50:8297–8304. 2023. View Article : Google Scholar : PubMed/NCBI | |
Sharma D, Singh A, Wilson C, Swaroop P, Kumar S, Yadav DK, Jain V, Agarwala S, Husain M and Sharawat SK: Exosomal long non-coding RNA MALAT1: A candidate of liquid biopsy in monitoring of Wilms' tumor. Pediatr Surg Int. 40:572024. View Article : Google Scholar : PubMed/NCBI | |
Tang C, He X, Jia L and Zhang X: Circular RNAs in glioma: Molecular functions and pathological implications. Noncoding RNA Res. 9:105–115. 2023. View Article : Google Scholar : PubMed/NCBI | |
Zhao RJ, Zhang WY and Fan XX: Circular RNAs: Potential biomarkers and therapeutic targets for autoimmune diseases. Heliyon. 10:e236942023. View Article : Google Scholar | |
Zhang X, Wan M, Min X, Chu G, Luo Y, Han Z, Li W, Xu R, Luo J, Li W, et al: Circular RNA as biomarkers for acute ischemic stroke: A systematic review and meta-analysis. CNS Neurosci Ther. 29:2086–2100. 2023. View Article : Google Scholar : PubMed/NCBI | |
Yılmaz B and Emingil G: Validating proteomic biomarkers in saliva: Distinguishing between health and periodontal diseases. Expert Rev Proteomics. 21:417–429. 2024. View Article : Google Scholar | |
Schmalz G, Li S, Burkhardt R, Rinke S, Krause F, Haak R and Ziebolz D: MicroRNAs as salivary markers for periodontal diseases: A new diagnostic approach? Biomed Res Int. 2016:10275252016. View Article : Google Scholar : PubMed/NCBI | |
Nappi F: Non-coding RNA-targeted therapy: A state-of-the-art review. Int J Mol Sci. 25:36302024. View Article : Google Scholar : PubMed/NCBI | |
Nemeth K, Bayraktar R, Ferracin M and Calin GA: Non-coding RNAs in disease: From mechanisms to therapeutics. Nat Rev Genet. 25:211–232. 2024. View Article : Google Scholar | |
Kim M, Jeong M, Hur S, Cho Y, Park J, Jung H, Seo Y, Woo HA, Nam KT, Lee K and Lee H: Engineered ionizable lipid nanoparticles for targeted delivery of RNA therapeutics into different types of cells in the liver. Sci Adv. 7:eabf43982021. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Yang X, Li J, Luo H, Huang Q, Yang W, Lei T, Lui S, Gong Q, Li H, et al: A nasally administrated reactive oxygen species-responsive carrier-free gene delivery nanosystem for Alzheimer's disease combination therapy. J Control Release. 381:1136042025. View Article : Google Scholar : PubMed/NCBI | |
Jiang Y, Jiang R, Xia Z, Guo M, Fu Y, Wang X and Xie J: Engineered neutrophil membrane-camouflaged nanocomplexes for targeted siRNA delivery against myocardial ischemia reperfusion injury. J Nanobiotechnology. 23:1342025. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Ma Z, Jiang L, Bojan N, Sha Y, Huang B, Ming L, Shen J and Pang W: Specific muscle targeted delivery of miR-130a loaded lipid nanoparticles: A novel approach to inhibit lipid accumulation in skeletal muscle and obesity. J Nanobiotechnology. 23:1592025. View Article : Google Scholar : PubMed/NCBI | |
Tong H, Ma Z, Yu J, Li D, Zhu Q, Shi H, Wu Y, Yang H, Zheng Y, Sun D, et al: Optimizing peptide-conjugated lipid nanoparticles for efficient siRNA delivery across the blood-brain barrier and treatment of glioblastoma multiforme. ACS Chem Biol. 20:942–952. 2025. View Article : Google Scholar : PubMed/NCBI | |
Westemeier-Rice ES, Winters MT, Rawson TW, Patel KJ, McHugh O, Ward S, McLaughlin S, Stewart A, Misra B, Dziadowicz S, et al: Lnc-RAINY regulates genes involved in radiation susceptibility through DNA:DNA:RNA triplex-forming interactions and has tumor therapeutic potential in lung cancers. Noncoding RNA Res. 12:152–166. 2024. View Article : Google Scholar | |
Jia Y, Xu L, Leng S, Sun Y, Huang X, Wang Y, Ren H, Li G, Bai Y, Zhang Z, et al: Nose-to-brain delivery of circular RNA SCMH1-loaded lipid nanoparticles for ischemic stroke therapy. Adv Mater. 37:e25005982025. View Article : Google Scholar : PubMed/NCBI | |
Kulkarni JA, Witzigmann D, Thomson SB, Chen S, Leavitt BR, Cullis PR and van der Meel R: The current landscape of nucleic acid therapeutics. Nat Nanotechnol. 16:630–643. 2021. View Article : Google Scholar : PubMed/NCBI | |
Li J, Jin F, Cai M, Lin T, Wang X and Sun Y: LncRNA nron inhibits bone resorption in periodontitis. J Dent Res. 101:187–195. 2022. View Article : Google Scholar |