Next Article in Journal
Clinical Features of Families with a Novel Pathogenic Mutation in Sepiapterin Reductase
Next Article in Special Issue
Long Non-Coding TP73-AS1: A Potential Biomarker and Therapeutic Target in Cancer
Previous Article in Journal
Control of Conformational Transitions by the Conserved GX9P Motif in the Fifth Transmembrane Domain of Neurotransmitter Sodium Symporters
Previous Article in Special Issue
Single-Cell RNA Sequencing Reveals Extensive Heterogeneity and Unique Gene Trajectories in Non-Transformed and Transformed Human Lung Epithelial Cells: Insights into the Role of LncRNAs in Tumor Heterogeneity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Application of Non-Coding RNAs as Biomarkers, Therapies, and Novel Vaccines in Diseases

1
Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
2
Institute of Scientific Research, Guangxi University, Nanning 530004, China
3
College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(7), 3055; https://doi.org/10.3390/ijms26073055
Submission received: 31 January 2025 / Revised: 22 March 2025 / Accepted: 24 March 2025 / Published: 26 March 2025
(This article belongs to the Special Issue Roles and Mechanisms of Non-Coding RNAs in Human Health and Disease)

Abstract

Non-coding RNAs (ncRNAs) are a class of RNAs that largely lack the capacity to encode proteins. They have garnered significant attention due to their central regulatory functions across numerous cellular and physiological processes at transcriptional, post-transcriptional, and translational levels. Over the past decade, ncRNA-based therapies have gained considerable attention in the diagnosis, treatment, and prevention of diseases, and many studies have revealed a significant relationship between ncRNAs and diseases. At the same time, due to their tissue specificity, an increasing number of projects have focused on the application of ncRNAs as biomarkers in diseases, as well as the design and development of novel ncRNA-based vaccines and therapies for clinical use. These ncRNAs may also drive research into the potential molecular mechanisms and complex pathogenesis of related diseases. However, new biomarkers need to be validated for their clinical effectiveness. Additionally, to produce safe and stable RNA products, factors such as purity, precise dosage, and effective delivery methods must be ensured to achieve optimal bioactivity. These challenges remain key issues in the clinical application of ncRNAs. This review summarizes the prospects of ncRNAs as potential biomarkers, as well as the current research status and clinical applications of ncRNAs in therapies and vaccines, and discusses the challenges and expectations of ncRNAs in disease diagnosis and drug therapy.

1. Introduction

In the past few years, with the advancement and progress of deep RNA sequencing technology, research on RNAs has gone beyond just mRNA, which encodes proteins. Meanwhile, non-coding RNAs (ncRNAs) have received increased attention and investigation due to their diverse roles in multiple biological activities, such as cell survival [1], immune response [2], disease development, and so on [3]. Typically, ncRNAs are classified into two major categories based on their functions: “Housekeeping ncRNAs”, which are widely expressed within cells and primarily regulate basic cellular functions that are essential for cell survival. These ncRNAs maintain relatively stable levels and encompass transfer RNAs (tRNAs), ribosomal RNAs (rRNAs), small nuclear RNAs (snRNAs), small nucleolar RNAs (snoRNAs), and telomerase RNAs (TERCs). The other class of ncRNAs possesses regulatory functions, capable of modulating gene expression at both the transcriptional and post-transcriptional levels. This category includes microRNAs (miRNAs), circular RNAs (circRNAs), long non-coding RNAs (lncRNAs), small interfering RNAs (siRNAs), and PIWI-interacting RNAs (piRNAs). These ncRNAs have different structures and functions, which serve as the basis for their classification (Table 1).
There is an inseparable relationship between ncRNAs and diseases. In addition to influencing the occurrence and development of diseases, many of these regulatory ncRNAs have also been described as biomarkers that can be used to predict and diagnose various diseases, including cancers [4], cardiovascular diseases [3], and neurological disorders [3,5]. Their emergence provides more non-invasive and specific options for disease diagnosis. In therapeutics, an increasing number of valuable studies have shown that ncRNAs can regulate gene expression networks, providing new directions for targeted therapy. Some ncRNA-based therapies have been applied to cancer, cardiovascular diseases, genetic disorders, and metabolic diseases [6]. Additionally, in recent years, circRNA vaccines have shown great potential in infectious disease prevention and immunotherapy. They are more stable than mRNA vaccines and can induce longer-lasting immune responses [7]. Although ncRNAs have been increasingly applied in diseases, their clinical translation still faces many challenges. For example, as a biomarker, the threshold of expression levels needs to be considered; as a drug, safety and efficacy must be evaluated; and as a vaccine, synthesis and delivery require careful consideration. Based on recent studies, this review primarily discusses the research on ncRNAs as potential biomarkers in diseases and their breakthroughs and applications in novel vaccines and therapies, as well as the challenges they face and future prospects, which aims to provide new insights into the research related to the diagnosis, treatment, and prevention of diseases using ncRNAs.
Table 1. Common types of ncRNAs and their descriptions.
Table 1. Common types of ncRNAs and their descriptions.
Types of RNAFeatures and IntroductionReferences
tRNAsThese possess a cloverleaf structure, deliver the correct amino acids to the ribosome, and facilitate the formation of polypeptide chains[8,9]
rRNAsThe structural components of the ribosome[10]
snRNAsA class of small RNA molecules in the nucleus of eukaryotic cells that are capable of processing pre-mRNAs[11]
snoRNAsRegulatory factors promoting rRNA maturation[12]
TERCsThe RNA component of telomerase, which provides a structural scaffold for the assembly of the telomerase complex[13,14]
miRNAsWith a length of approximately 19~24 nucleotides (nts), these serve as post-transcriptional regulatory factors of gene expression[15]
circRNAsThese lack free ends and have covalently closed loop structures, and can function as miRNA sponges, RNA-binding protein sponges, and translation regulatory factors[16,17]
lncRNAsWith a length exceeding 200 nts, these mediate the post-transcriptional control of signal transduction pathways, translation processes, and gene expression[18]
siRNAsThese are primarily involved in RNA interference to regulate gene expression[19]
piRNAsThese have diverse functions including gene regulation, transposon suppression, epigenetic programming, and antiviral defense, among others[20,21]

2. NcRNAs as Diagnostic Biomarkers

NcRNAs serve as important potential biomarkers in the diagnosis and prognosis of diseases. They exhibit stability and universality, being expressed in various tissues, blood, urine, and saliva [22,23,24], and also possess certain disease specificity [25,26], which makes them highly suitable as biomarkers for disease diagnosis, prognosis, and treatment. In current research on ncRNAs, studies on miRNAs are the most extensive, while research on circRNAs and lncRNAs is also progressing further. Therefore, this section discusses the applications of miRNAs, circRNAs, and lncRNAs in diseases.

2.1. MiRNAs as Biomarkers

MiRNAs are the most abundant small RNAs [27], playing crucial roles in cell differentiation [28,29], apoptosis [30,31], organ development [32], metabolism [33,34], and others. They regulate target gene expression by binding to the 3′ untranslated region (3′UTR) of mRNA, leading to translation inhibition or degradation [35]. Tissue-specific miRNAs are often associated with diseases related to specific tissues, and changes in their expression patterns have been confirmed in various human diseases, including cancer, cardiovascular diseases, neurological disorders, genetic diseases, metabolic disorders, and viral pathogenesis. Moreover, the dysregulation of miRNAs has been proven to be a pathogenic factor in disease progression [36].
The differential expression of certain miRNAs in various stages of different diseases has led to their continued investigation as potential biomarkers. For example, miR-21 and miR-22 are two highly frequent miRNAs in cancer and are abnormally expressed in various types of cancer [37,38]. In a study, by comparing the expression levels of miR-21 and miR-22 in the serum of 80 epithelial ovarian cancer (EOC) patients and 80 healthy volunteers, researchers found that these two miRNAs exhibited different numerical changes in different stages of epithelial EOC. In the late stage of EOC, the fold change in miR-21 expression was higher than in the early stage, while miR-22 exhibited the opposite trend [39]. This opposite trend allows these two miRNAs to serve as diagnostic biomarkers in the early stages of EOC and enables the prediction of ovarian cancer progression over time by monitoring their fold change values. Moreover, in recent research, Duan et al. [40] discovered through sequencing that the expression levels of miR-92a-3p, miR-425-5p, and miR-185-5p were significantly elevated in the urine of patients with IgA nephropathy (IgAN). Gene Ontology (GO), the Kyoto Encyclopedia of Genes and Genomes (KEGG), and subsequent dual-luciferase reporter assays confirmed that in renal tubular epithelial cells, the target gene of miR-185-5p is tight junction protein 1 (TJP1). In vitro experiments also indicated that miR-185-5p mimics can reduce the expression of TJP1 in human kidney 2 (HK-2) cells and promote the transition of renal tubular epithelial cells to a fibrotic phenotype. During the progression of IgAN, tubulointerstitial fibrosis is an important histological marker [41]. This study provides strong evidence that the expression levels of miR-92a-3p, miR-425-5p, and miR-185-5p in urine can serve as biomarkers for IgAN. This non-invasive detection method is undoubtedly simpler and faster, aiding in early diagnosis. Additionally, miRNAs can also serve as biomarkers for liver injury and rejection after liver transplantation [42]. Plasma miRNAs, including miR-181a-5p, miR-155-5p, and miR-122-5p, are significantly upregulated in patients with acute T-cell-mediated rejection (TCMAR) and subclinical rejection (SCR) after liver transplantation. Before transplantation, the expression levels of miR-155-5p and miR-181a-5p exhibit different trends between the two groups: miR-155-5p is significantly higher in TCMAR patients than in SCR patients, whereas miR-181a-5p is significantly higher in SCR patients than in TCMAR patients. This difference provides valuable insights for diagnosis and prognosis before and after liver transplantation, making these miRNAs potential candidates for a biomarker panel in liver injury following transplantation [43].

2.2. CircRNAs as Biomarkers

CircRNAs are covalently closed loops formed by single-stranded RNA without free ends. The majority of circRNAs originate from exons and are highly expressed in organisms. Their circular and distinct structure renders circRNAs more stable and less prone to degradation by RNA exonucleases when compared to linear RNAs [44]. This high stability provides favorable conditions for their use as biomarkers. CircRNAs serve multiple functions; they can act as competitive endogenous RNAs or miRNA sponges [45], and they can also interact with RNA-binding proteins [46]. Furthermore, some circRNAs have the capability to undergo translation to produce proteins [47] and regulate gene transcription [48]. Similarly, circRNAs exhibit differential expression in various diseases, making them potential biomarkers or therapeutic targets for these conditions [25].
The oral glucose tolerance test (OGTT) and overnight fasting plasma glucose (FPG) have long been the primary methods for detecting diabetes [49]. However, these methods involve many uncertainties, are time-consuming, and are complex. Using specific circRNAs in the blood as biomarkers allows for lower-cost, more convenient sampling and detection, enabling a faster determination of disease progression. For example, Zhao et al. [50] extracted whole blood from three groups: healthy individuals, individuals with prediabetes, and those with type 2 diabetes (T2DM). RNA was extracted and the relative expression levels of circRNAs were measured using q-PCR. The results showed that the expression levels of hsa_circ_0054633 and hsa_circ_0068087 were significantly different among the three groups and increased sequentially in the healthy control group, prediabetes group, and T2DM group. They then used a microarray analysis comparing the expression of circRNAs in the peripheral blood of six healthy individuals and six T2DM patients. They found that hsa_circ_0054633 had the largest area under the curve (AUC) and a lower p-value, indicating its potential diagnostic capability for prediabetes and T2DM. Additionally, pancreatic ductal adenocarcinoma (PDAC) is a cancer with a high mortality rate [51]. Patients often do not exhibit prominent symptoms clinically, and its diagnosis is usually made incidentally during routine imaging. This often leads to many patients being diagnosed in an advanced stage or after distant metastasis has occurred [52]. Recently, Xu et al. [53] developed a circular circRNA biomarker panel for the identification of PDAC. They identified five circRNAs in plasma for the active detection of PDAC and combined the circRNA-based panel with the serum tumor marker, cancer antigen 19-9 (CA19-9) glycoprotein, which enhances the diagnostic performance for early-stage PDAC. Additionally, circRNAs are widely involved in the regulation of bone metabolism [54]. Hsa_circ_0006859 is one of the most significantly upregulated circRNAs in serum exosomes of postmenopausal osteoporosis patients. It can promote the expression of ROCK1 through the miR-431-5p/ROCK1 axis, thereby inhibiting osteoblast differentiation and promoting adipogenesis. These results suggest that Hsa_circ_0006859 is a potential biomarker for postmenopausal osteoporosis and provide a direction for the targeted therapy of related diseases [55].

2.3. LncRNAs as Biomarkers

LncRNAs are defined by their length, which exceeds 200 nucleotides. Based on their relative position to coding genes on the chromosome, lncRNAs can be broadly categorized into five classes: antisense, sense, intronic, intergenic, and bidirectional. They are primarily transcribed by RNA polymerase II and lack apparent open reading frames [56]. LncRNAs have various functions based on their subcellular localization, including gene imprinting [57], histone modification [58], chromatin remodeling [58], transcriptional activation [59], transcriptional interference [60], and cell cycle regulation [61].
LncRNAs can also serve as potential biomarkers. For example, PCA3 is a well-known prostate cancer-specific lncRNA [62]. It is overexpressed in prostate cancer (PCa) but not associated with other prostate conditions [63,64]. This makes PCA3 a suitable potential biomarker for PCa. Previously, serum prostate-specific antigen (PSA) was an important indicator for prostate cancer diagnosis, while the histopathological evaluation of prostate biopsy was the decisive factor for confir mation [65]. PCA3 can be collected from urine and plasma, which undoubtedly reduces the difficulty of sample collection and shortens the testing time. Additionally, inflammatory bowel disease (IBD) is a chronic, relapsing condition that includes Crohn’s disease (CD) and ulcerative colitis (UC) [66]. Heydari et al. [67] compared five previously reported lncRNAs related to IBD (including H19 [68], CDKN2B-AS1 [69], TUG1 [70], GAS5 [71], and CRNDE [72]). By analyzing the expression of each lncRNA in patient tissue samples and plasma extracellular vesicles (EVs), they found that the expression level of H19 was significantly higher in IBD patients than in the healthy control group. Additionally, compared to rheumatoid arthritis (RA) and irritable bowel syndrome (IBS) patients, H19 expression in plasma EVs was significantly elevated, confirming that H19 can serve as a potential biomarker for IBD with strong discriminatory ability. In addition, lnc-GAS5 is involved in the development of atherosclerosis and coronary heart disease (CHD), with miR-21 as its target [73]. Jiang et al. [74] detected the expression levels of lnc-GAS5 and miR-21 in CHD patients and the control group using RT-qPCR. They found that the expression level of lnc-GAS5 was significantly increased in CHD patients, while miR-21 showed an opposite trend, with a negative correlation between the two in CHD patients. Additionally, lnc-GAS5 was positively correlated with the Gensini score, which reflects the severity of coronary artery stenosis. These results suggest that lnc-GAS5 and its target, miR-21, could serve as potential biomarkers for CHD.
Some examples of ncRNAs with the potential to be used as disease biomarkers are summarized in Table 2.

3. NcRNAs in Target Therapy

The ability of ncRNAs to regulate the expression of target genes represents the great potential for target therapies. Currently, various RNA-based therapies have been developed, including antisense oligonucleotides (ASOs), short hairpin RNAs (shRNAs), small interfering RNAs (siRNAs), anti-microRNAs (anti-miRNAs), aptamers, and others [6]. In 1998, Fomivirsen, the world’s first antisense oligonucleotide-based medicine was approved by the United States Food and Drug Administration (FDA) and the European Medicines Agency (EMA) to treat cytomegalovirus (CMV) retinitis in patients with acquired immunodeficiency syndrome (AIDS) [104]. This drug consists of 21 thio-oligonucleotides with the nucleotide sequence 5′-GCGTTTGCTCTTCTTCTTGCG-3′, which can complement the mRNA of the major immediate-early region proteins of human cytomegalovirus, and it has an approximate half-life of about 55 h in the human body, making it an effective antiviral medication targeting CMV retinitis [105]. Since then, an increasing number of drugs based on ncRNAs have been approved and utilized. In 2018, Patisiran became the first siRNA drug to be approved by the FDA. It reduces mutant and wild-type transthyretin protein production by targeting the 3′ untranslated region of transthyretin mRNA for the treatment of hereditary transthyretin-mediated amyloidosis [106].
In addition, there are other ncRNA therapies under exploration, and different ncRNAs have shown varying effects in clinical trials. miRNA-based therapies offer several advantages. On one hand, miRNAs naturally exist in the human body, and they have established mechanisms and can selectively target downstream targets [107]. On the other hand, they can affect multiple genes within the same pathway, leading to broader yet specific responses [108]. Previously, the Kasinski team published a study where they developed a novel cancer therapy using a modified form of miR-34a to halt cancer cell division. In preclinical trials with mice, treatment using fully modified miR-34a targeting tumors resulted in the cessation of tumor growth within a 21-day assessment period, with some animals achieving complete remission. By contrast, untreated tumors doubled in size during the same period. These findings have the potential to rejuvenate miR-34a as an anticancer agent and provide robust support for clinical trials [109].
In another study, Liu et al. [110] identified a lncRNA enriched in cardiomyocytes, named LncHrt. Knocking out LncHrt impairs cardiac homeostasis, whereas overexpressing it using an AAV9 vector protects the heart from myocardial infarction and rescues the transcriptome in infarcted hearts. This indicates the therapeutic potential of LncHrt in myocardial infarction, providing a theoretical foundation for LncHrt as a novel treatment for ischemic heart disease.
Table 3 lists current RNA therapies currently approved by the FDA and/or EMA.

4. NcRNAs in Novel Vaccines

The RNA vaccines have the advantages of high efficacy, strong adaptability, and good tolerability [128]. For example, mRNA vaccines can directly encode antigens after entering the cytoplasm, characterized by their non-integrating, non-infectious nature and good tolerability [129,130]. However, mRNA vaccines are highly unstable in terms of storage and intracellular delivery, prone to degradation by nucleases, which significantly increase the risks and production costs associated with mRNA vaccines. Therefore, seeking stable alternative types of RNA vaccines to replace them is a new research direction.
Currently, breakthrough progress has been made in vaccine research based on ncRNAs, with the focus primarily on circRNAs. This is because circRNA-based vaccines are mainly derived from linear RNA precursors (pre-circRNAs) that are cyclized in vitro and then rely on the host organism’s translation system to generate antigens, thereby stimulating immune responses [131]. Compared to linear mRNAs, circRNAs have a covalently closed loop structure, which makes them highly stable and resistant to degradation by exonuclease-mediated cleavage [7]. Additionally, circRNAs exhibit lower immunogenicity and cytotoxicity, thereby reducing side effects [132]. Furthermore, circRNAs can elicit more durable immune responses [132,133]. These advantages demonstrate the immense potential of circRNAs in vaccine research.
The synthesis of linear RNA precursors is typically carried out using the in vitro transcription (IVT) method. In this process, plasmid DNA is used as the template, and bacteriophage RNA polymerase is employed to generate large numbers of RNA molecules. Subsequently, chemical, enzymatic, or ribozyme-mediated methods are used to circularize the linear RNA precursors [134,135,136]. The chemical strategy most commonly employs condensation reagents such as hydrogen bromide (BrCN) or 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC) to ligate two RNA strands with terminal 5′-phosphate and 3′-hydroxyl groups [137,138]. The enzymatic strategy typically uses T4 DNA ligase (T4 Dnl), T4 RNA ligase 1 (T4 Rnl1), or T4 RNA ligase 2 (T4 Rnl2) to catalyze the ligation of RNA ends with 5′-phosphate and 3′-hydroxyl groups [136]. Ribozyme-mediated ligation is the most common approach, with Group I and Group II introns exhibiting ribozyme RNase activity to connect linear RNA precursors into circRNA through self-catalyzed intron splicing reactions [131,139,140]. The methods for in vitro synthesis of circRNA are shown in Figure 1.
The successful delivery of circRNAs is crucial in vaccination. CircRNAs are typically large molecules, which makes it difficult for them to diffuse in and out of cells. In the delivery of mRNA vaccines, electroporation was one of the earliest methods [141], and techniques such as gene gun [142] and microinjection [143] can also deliver mRNA antigens. Similarly, these methods can also be applied to the delivery of circRNA vaccines. With the advancement of delivery strategies, lipid nanoparticles (LNPs), including solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs), have become the most widely used. They exhibit more complex structures, enhanced physical stability, easier synthesis, and greater loading capacity [140,144,145]. Previous delivery methods and the structure of LNP are shown in Figure 2.
Research on circRNA vaccines is gradually innovating and advancing. Previous studies have shown that tumor-specific antigens (TSAs) primarily originate from non-coding regions, and that the non-coding genome may encode peptides that bind to human leukocyte antigens (HLAs) as cryptic antigens to stimulate adaptive immunity [146,147]. Building on this basis, Huang et al. [148] compared the mass spectrometry analysis of the HLA class I (HLA-I) peptidome in human breast cancer samples with ribosome sequencing. They confirmed that circFAM53B can undergo non-canonical translation as a tumor-specific circRNA to produce cryptic antigenic peptides that bind to HLA-I. These cryptic peptides can initiate naive CD4 and CD8 T cells in an antigen-specific manner, thereby inducing an anti-tumor immune response. This suggests that using tumor-specific circRNA as a vaccine against malignant tumors holds considerable therapeutic potential.
In addition, synthetic circRNA vaccines have found more applications. For instance, during the global coronavirus disease 2019 (COVID-19) pandemic, Qu et al. [149] utilized an IVT strategy to generate SARS-CoV-2 circRNARBD, which can express the trimeric receptor-binding domain (RBD) of the spike protein. They validated its ability to induce sustained immune responses and neutralize antibodies in mice and rhesus monkeys.
Li et al. [150] developed a lipid nanoparticle (LNP) system for both the in vitro and in vivo delivery of circRNAs to study the functionality of circRNA vaccines. They prepared circRNAOVA-luc-LNP (OVA [257-264]-luciferase-coding circRNA) vaccines for subsequent research. The results showed that circRNA-LNPs elicited effective innate immune responses and significant antigen-specific responses, demonstrating good anti-tumor effects in various mouse tumor models.
Research on circRNA vaccines is still developing, and their use in humans still requires a lot of clinical practice. Table 4 summarizes some circRNA vaccines that have been developed or are in development.

5. Discussions: Challenges and Opportunities

Although an increasing number of studies have demonstrated the great potential of ncRNAs in biomarkers, therapeutic targets, and novel vaccines, their transition from clinical trials to widespread application still faces significant challenges.
Discovering a new ncRNA biomarker can undoubtedly bring great convenience to disease diagnosis and prognosis, but identifying a new biomarker is not an easy task. Firstly, the threshold value of ncRNAs as biomarkers is a crucial indicator. Establishing a clear threshold for disease diagnosis requires a large number of experimental samples. At the same time, factors such as age differences, gender differences, and regional variations also need to be considered. Secondly, there are currently very few single biomarkers that can truly be used for disease diagnosis, and some ncRNAs exhibit similar expression patterns across different diseases [159], making them less suitable as specific disease markers. By contrast, combinational biomarkers can provide higher sensitivity and specificity. For example, in previous reports, although PCA3 has been approved as a PCa diagnostic marker, it has certain limitations, such as low sensitivity in patients with low-grade prostate cancer. Studies have shown that the elevated expression of the ERG oncogene in prostate cancer patients is due to its fusion with transmembrane protease serine 2 (TMPRSS2) [160]. Combining PCA3 with the TMPRSS2-ERG gene fusion for joint analysis can further improve diagnostic accuracy [161,162].
Additionally, the development and application of ncRNA therapies face numerous challenges. In the same disease, different types of ncRNAs may exhibit abnormal expression. Investigating disease pathogenesis and identifying specific ncRNAs require extensive time for exploration. Meanwhile, ensuring that the therapy targets only the intended ncRNAs without affecting other ncRNAs or gene expression remains a significant challenge. Furthermore, factors such as maintaining an effective drug concentration in the body, preventing drug accumulation in non-target tissues, and assessing safety and toxicity must be carefully considered, necessitating extensive clinical trials. Finally, therapies involving gene regulation also require ethical evaluations. These challenges and issues call for further experimentation, refinement, and optimization.
In recent years, circRNA vaccines have gained increasing attention. It is undeniable that while the circular structure of circRNA vaccines enhances stability, the processes of circularization and purification remain complex, and residual non-target sequences may introduce unnecessary immunogenicity. This has also driven more researchers to explore simpler synthesis methods. Furthermore, the efficient delivery system for circRNA vaccines still requires optimization to ensure precise delivery to target cells while avoiding degradation. Similarly to ncRNA therapies, circRNA vaccines also require extensive clinical trials to establish dosage standards, assess safety and side effects, and ensure their efficacy and reliability.
Moreover, to promote the application of ncRNAs as biomarkers, therapeutics, and novel vaccines, obtaining regulatory approval is the first step. Additionally, reducing production costs, simplifying drug synthesis methods, and addressing potential adverse reactions must also be considered.
In conclusion, ncRNAs present a promising strategy and significant opportunities for disease treatment but also face inevitable challenges. With more in-depth and valuable research, these issues will eventually be overcome.

6. Conclusions

NcRNAs play a crucial role in the regulation of gene expression and have increasingly been studied in recent years as disease biomarkers, innovative therapeutic approaches, and novel vaccines. The exploration of ncRNAs has opened new horizons for understanding disease mechanisms, preventing diseases, and developing treatments. As biomarkers, ncRNAs have demonstrated significant diagnostic and prognostic value in various diseases. With further research, some dysregulated ncRNAs in diseases may mediate changes in target gene expression levels, providing greater insights into disease pathogenesis and aiding in the development of targeted therapies. Concurrently, RNA therapies that regulate gene expression by synthesizing or inhibiting ncRNA molecules offer new possibilities for precise treatments. Furthermore, ncRNAs have shown great potential in eliciting specific immune responses as emerging vaccines.
In spite of the remarkable insights and promises ncRNAs bring, challenges remain. Firstly, accurately measuring and quantifying the expression levels of ncRNAs remains a complex issue, necessitating the development of more sensitive and specific detection methods. Secondly, we need a deeper understanding of the mechanisms of action of different types of ncRNAs in various diseases to ensure their reliability and accuracy. Additionally, ensuring the stability, delivery efficiency, and safety of ncRNA therapeutics is also a critical concern. Despite these challenges, an increasing number of studies and clinical trials indicate that ncRNAs offers more solutions in disease diagnosis, prevention, and treatment, providing new opportunities for the future development of precision medicine.
In conclusion, ncRNAs have revealed promising strategies and immense potential in disease diagnosis and treatment. The future of ncRNAs in medicine is expected to be transformative, heralding advanced, personalized, and effective strategies for managing and eradicating diseases in the coming years.

Author Contributions

L.-X.Y. and H.L. conceptualized the study and authored most part of the manuscript. Z.-H.C. and H.-Y.S. added content to the manuscript. J.-P.H., Z.-P.L. and X.-X.L. organized materials and revised the manuscript. Z.-G.H. and J.W. contributed to the work design and the manuscript revision. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Guangxi Natural Science Foundation (Grant Nos. 2024GXNSFBA010422, 2025GXNSFBA069202, and 2024GXNSFFA010018), the National Natural Science Foundation of China (Grant Nos. 32472863 and 32102514), China Agriculture Research System of MOF and MARA (CARS-42-2) and the National Key Research and Development Program of China (2023YFD1300303).

Conflicts of Interest

All authors declare that there are no conflicts of interest.

References

  1. Wang, W.; Chen, R.; Droll, S.; Barber, E.; Saleh, L.; Corrigan-Cummins, M.; Trick, M.; Anastas, V.; Hawk, N.V.; Zhao, Z.; et al. MiR-181c regulates MCL1 and cell survival in GATA2 deficient cells. J. Leukoc. Biol. 2022, 111, 805–816. [Google Scholar] [CrossRef] [PubMed]
  2. Xie, N.; Liu, G. NcRNA-regulated immune response and its role in inflammatory lung diseases. Am. J. Physiol. Lung Cell. Mol. Physiol. 2015, 309, L1076–L1087. [Google Scholar] [CrossRef]
  3. Esteller, M. Non-coding RNAs in human disease. Nat. Rev. Genet. 2011, 12, 861–874. [Google Scholar] [CrossRef] [PubMed]
  4. Yan, H.; Bu, P. Non-coding RNA in cancer. Essays Biochem. 2021, 65, 625–639. [Google Scholar] [CrossRef] [PubMed]
  5. Das, T.; Das, T.K.; Khodarkovskaya, A.; Dash, S. Non-coding RNAs and their bioengineering applications for neurological diseases. Bioengineered 2021, 12, 11675–11698. [Google Scholar] [CrossRef]
  6. Winkle, M.; El-Daly, S.M.; Fabbri, M.; Calin, G.A. Noncoding RNA therapeutics—Challenges and potential solutions. Nat. Rev. Drug Discov. 2021, 20, 629–651. [Google Scholar] [CrossRef]
  7. Kristensen, L.S.; Andersen, M.S.; Stagsted, L.V.W.; Ebbesen, K.K.; Hansen, T.B.; Kjems, J. The biogenesis, biology and characterization of circular RNAs. Nat. Rev. Genet. 2019, 20, 675–691. [Google Scholar] [CrossRef]
  8. Biela, A.; Hammermeister, A.; Kaczmarczyk, I.; Walczak, M.; Koziej, L.; Lin, T.Y.; Glatt, S. The diverse structural modes of tRNA binding and recognition. J. Biol. Chem. 2023, 299, 104966. [Google Scholar] [CrossRef]
  9. Mathew, B.A.; Katta, M.; Ludhiadch, A.; Singh, P.; Munshi, A. Role of tRNA-Derived Fragments in Neurological Disorders: A Review. Mol. Neurobiol. 2023, 60, 655–671. [Google Scholar] [CrossRef]
  10. Denisenko, O. Epigenetics of Ribosomal RNA Genes. Biochemistry 2022, 87, S103–S131. [Google Scholar] [CrossRef]
  11. Matera, A.G.; Wang, Z. A day in the life of the spliceosome. Nat. Rev. Mol. Cell Biol. 2014, 15, 108–121. [Google Scholar] [CrossRef] [PubMed]
  12. van der Werf, J.; Chin, C.V.; Fleming, N.I. SnoRNA in Cancer Progression, Metastasis and Immunotherapy Response. Biology 2021, 10, 809. [Google Scholar] [CrossRef]
  13. Rubtsova, M.; Dontsova, O. Human Telomerase RNA: Telomerase Component or More? Biomolecules 2020, 10, 873. [Google Scholar] [CrossRef]
  14. Nguyen, T.H.D.; Tam, J.; Wu, R.A.; Greber, B.J.; Toso, D.; Nogales, E.; Collins, K. Cryo-EM structure of substrate-bound human telomerase holoenzyme. Nature 2018, 557, 190–195. [Google Scholar] [CrossRef] [PubMed]
  15. Bhaskaran, M.; Mohan, M. MicroRNAs: History, biogenesis, and their evolving role in animal development and disease. Vet. Pathol. 2014, 51, 759–774. [Google Scholar] [CrossRef]
  16. Zhang, Z.; Yang, T.; Xiao, J. Circular RNAs: Promising Biomarkers for Human Diseases. EBioMedicine 2018, 34, 267–274. [Google Scholar] [CrossRef] [PubMed]
  17. Patop, I.L.; Wüst, S.; Kadener, S. Past, present, and future of circRNAs. EMBO J. 2019, 38, e100836. [Google Scholar] [CrossRef]
  18. Bridges, M.C.; Daulagala, A.C.; Kourtidis, A. LNCcation: lncRNA localization and function. J. Cell Biol. 2021, 220, e202009045. [Google Scholar] [CrossRef]
  19. Alshaer, W.; Zureigat, H.; Al Karaki, A.; Al-Kadash, A.; Gharaibeh, L.; Hatmal, M.M.; Aljabali, A.A.A.; Awidi, A. SiRNA: Mechanism of action, challenges, and therapeutic approaches. Eur. J. Pharmacol. 2021, 905, 174178. [Google Scholar] [CrossRef]
  20. Mani, S.R.; Juliano, C.E. Untangling the web: The diverse functions of the PIWI/piRNA pathway. Mol. Reprod. Dev. 2013, 80, 632–664. [Google Scholar] [CrossRef]
  21. Ku, H.Y.; Lin, H. PIWI proteins and their interactors in piRNA biogenesis, germline development and gene expression. Natl. Sci. Rev. 2014, 1, 205–218. [Google Scholar] [CrossRef] [PubMed]
  22. Mitchell, P.S.; Parkin, R.K.; Kroh, E.M.; Fritz, B.R.; Wyman, S.K.; Pogosova-Agadjanyan, E.L.; Peterson, A.; Noteboom, J.; O’Briant, K.C.; Allen, A.; et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc. Natl. Acad. Sci. USA 2008, 105, 10513–10518. [Google Scholar] [CrossRef] [PubMed]
  23. Li, Y.; Zhao, J.; Yu, S.; Wang, Z.; He, X.; Su, Y.; Guo, T.; Sheng, H.; Chen, J.; Zheng, Q.; et al. Extracellular Vesicles Long RNA Sequencing Reveals Abundant mRNA, circRNA, and lncRNA in Human Blood as Potential Biomarkers for Cancer Diagnosis. Clin. Chem. 2019, 65, 798–808. [Google Scholar] [CrossRef] [PubMed]
  24. Arantes, L.; De Carvalho, A.C.; Melendez, M.E.; Lopes Carvalho, A. Serum, plasma and saliva biomarkers for head and neck cancer. Expert Rev. Mol. Diagn. 2018, 18, 85–112. [Google Scholar] [CrossRef] [PubMed]
  25. Lei, B.; Tian, Z.; Fan, W.; Ni, B. Circular RNA: A novel biomarker and therapeutic target for human cancers. Int. J. Med. Sci. 2019, 16, 292–301. [Google Scholar] [CrossRef]
  26. Jin, Z.Q. MicroRNA targets and biomarker validation for diabetes-associated cardiac fibrosis. Pharmacol. Res. 2021, 174, 105941. [Google Scholar] [CrossRef]
  27. Felekkis, K.; Touvana, E.; Stefanou, C.; Deltas, C. MicroRNAs: A newly described class of encoded molecules that play a role in health and disease. Hippokratia 2010, 14, 236–240. [Google Scholar] [PubMed]
  28. Martin, E.C.; Qureshi, A.T.; Dasa, V.; Freitas, M.A.; Gimble, J.M.; Davis, T.A. MicroRNA regulation of stem cell differentiation and diseases of the bone and adipose tissue: Perspectives on miRNA biogenesis and cellular transcriptome. Biochimie 2016, 124, 98–111. [Google Scholar] [CrossRef]
  29. Ding, S.; Liu, G.; Jiang, H.; Fang, J. MicroRNA Determines the Fate of Intestinal Epithelial Cell Differentiation and Regulates Intestinal Diseases. Curr. Protein Pept. Sci. 2019, 20, 666–673. [Google Scholar] [CrossRef]
  30. He, J.F.; Luo, Y.M.; Wan, X.H.; Jiang, D. Biogenesis of MiRNA-195 and its role in biogenesis, the cell cycle, and apoptosis. J. Biochem. Mol. Toxicol. 2011, 25, 404–408. [Google Scholar] [CrossRef]
  31. Jing, R.; Zhong, Q.Q.; Long, T.Y.; Pan, W.; Qian, Z.X. Downregulated miRNA-26a-5p induces the apoptosis of endothelial cells in coronary heart disease by inhibiting PI3K/AKT pathway. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 4940–4947. [Google Scholar] [CrossRef] [PubMed]
  32. Ha, M.; Kim, V.N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol. 2014, 15, 509–524. [Google Scholar] [CrossRef] [PubMed]
  33. Agbu, P.; Carthew, R.W. MicroRNA-mediated regulation of glucose and lipid metabolism. Nat. Rev. Mol. Cell Biol. 2021, 22, 425–438. [Google Scholar] [CrossRef]
  34. Vienberg, S.; Geiger, J.; Madsen, S.; Dalgaard, L.T. MicroRNAs in metabolism. Acta Physiol. 2017, 219, 346–361. [Google Scholar] [CrossRef]
  35. Qadir, M.I.; Bukhat, S.; Rasul, S.; Manzoor, H.; Manzoor, M. RNA therapeutics: Identification of novel targets leading to drug discovery. J. Cell. Biochem. 2020, 121, 898–929. [Google Scholar] [CrossRef] [PubMed]
  36. Saliminejad, K.; Khorram Khorshid, H.R.; Soleymani Fard, S.; Ghaffari, S.H. An overview of microRNAs: Biology, functions, therapeutics, and analysis methods. J. Cell. Physiol. 2019, 234, 5451–5465. [Google Scholar] [CrossRef] [PubMed]
  37. Bautista-Sánchez, D.; Arriaga-Canon, C.; Pedroza-Torres, A.; De La Rosa-Velázquez, I.A.; González-Barrios, R.; Contreras-Espinosa, L.; Montiel-Manríquez, R.; Castro-Hernández, C.; Fragoso-Ontiveros, V.; Álvarez-Gómez, R.M.; et al. The Promising Role of miR-21 as a Cancer Biomarker and Its Importance in RNA-Based Therapeutics. Mol. Ther. Nucleic Acids 2020, 20, 409–420. [Google Scholar] [CrossRef]
  38. Wang, J.; Li, Y.; Ding, M.; Zhang, H.; Xu, X.; Tang, J. Molecular mechanisms and clinical applications of miR-22 in regulating malignant progression in human cancer (Review). Int. J. Oncol. 2017, 50, 345–355. [Google Scholar] [CrossRef]
  39. Paliwal, N.; Vashist, M.; Chauhan, M. Evaluation of miR-22 and miR-21 as diagnostic biomarkers in patients with epithelial ovarian cancer. 3 Biotech 2020, 10, 142. [Google Scholar] [CrossRef]
  40. Duan, Z.Y.; Bu, R.; Liang, S.; Chen, X.Z.; Zhang, C.; Zhang, Q.Y.; Li, J.J.; Chen, X.M.; Cai, G.Y. Urinary miR-185-5p is a biomarker of renal tubulointerstitial fibrosis in IgA nephropathy. Front. Immunol. 2024, 15, 1326026. [Google Scholar] [CrossRef]
  41. Roberts, I.S. Pathology of IgA nephropathy. Nat. Rev. Nephrol. 2014, 10, 445–454. [Google Scholar] [CrossRef]
  42. Koch, P.F.; Ludwig, K.; Krenzien, F.; Hillebrandt, K.H.; Schöning, W.; Pratschke, J.; Raschzok, N.; Sauer, I.M.; Moosburner, S. miRNA as potential biomarkers after liver transplantation: A systematic review. Transplant. Rev. 2024, 38, 100831. [Google Scholar] [CrossRef]
  43. Millán, O.; Ruiz, P.; Orts, L.; Ferré, P.; Crespo, G.; Santana, M.; Fortuna, V.; Quintairos, L.; Navasa, M.; Brunet, M. Monitoring of miR-181a-5p and miR-155-5p Plasmatic Expression as Prognostic Biomarkers for Acute and Subclinical Rejection in De Novo Adult Liver Transplant Recipients. Front. Immunol. 2019, 10, 873. [Google Scholar] [CrossRef]
  44. Jeck, W.R.; Sorrentino, J.A.; Wang, K.; Slevin, M.K.; Burd, C.E.; Liu, J.; Marzluff, W.F.; Sharpless, N.E. Circular RNAs are abundant, conserved, and associated with ALU repeats. RNA 2013, 19, 141–157. [Google Scholar] [CrossRef]
  45. Panda, A.C. Circular RNAs Act as miRNA Sponges. Adv. Exp. Med. Biol. 2018, 1087, 67–79. [Google Scholar] [CrossRef] [PubMed]
  46. Zang, J.; Lu, D.; Xu, A. The interaction of circRNAs and RNA binding proteins: An important part of circRNA maintenance and function. J. Neurosci. Res. 2020, 98, 87–97. [Google Scholar] [CrossRef] [PubMed]
  47. Zhou, W.Y.; Cai, Z.R.; Liu, J.; Wang, D.S.; Ju, H.Q.; Xu, R.H. Circular RNA: Metabolism, functions and interactions with proteins. Mol. Cancer 2020, 19, 172. [Google Scholar] [CrossRef]
  48. Li, Z.; Huang, C.; Bao, C.; Chen, L.; Lin, M.; Wang, X.; Zhong, G.; Yu, B.; Hu, W.; Dai, L.; et al. Exon-intron circular RNAs regulate transcription in the nucleus. Nat. Struct. Mol. Biol. 2015, 22, 256–264. [Google Scholar] [CrossRef]
  49. Bartoli, E.; Fra, G.P.; Carnevale Schianca, G.P. The oral glucose tolerance test (OGTT) revisited. Eur. J. Intern. Med. 2011, 22, 8–12. [Google Scholar] [CrossRef]
  50. Zhao, Z.; Li, X.; Jian, D.; Hao, P.; Rao, L.; Li, M. Hsa_circ_0054633 in peripheral blood can be used as a diagnostic biomarker of pre-diabetes and type 2 diabetes mellitus. Acta Diabetol. 2017, 54, 237–245. [Google Scholar] [CrossRef]
  51. Kolbeinsson, H.M.; Chandana, S.; Wright, G.P.; Chung, M. Pancreatic Cancer: A Review of Current Treatment and Novel Therapies. J. Investig. Surg. 2023, 36, 2129884. [Google Scholar] [CrossRef]
  52. Singhi, A.D.; Koay, E.J.; Chari, S.T.; Maitra, A. Early Detection of Pancreatic Cancer: Opportunities and Challenges. Gastroenterology 2019, 156, 2024–2040. [Google Scholar] [CrossRef] [PubMed]
  53. Xu, C.; Jun, E.; Okugawa, Y.; Toiyama, Y.; Borazanci, E.; Bolton, J.; Taketomi, A.; Kim, S.C.; Shang, D.; Von Hoff, D.; et al. A Circulating Panel of circRNA Biomarkers for the Noninvasive and Early Detection of Pancreatic Ductal Adenocarcinoma. Gastroenterology 2024, 166, 178–190.e116. [Google Scholar] [CrossRef] [PubMed]
  54. Chen, W.; Zhang, B.; Chang, X. Emerging roles of circular RNAs in osteoporosis. J. Cell. Mol. Med. 2021, 25, 9089–9101. [Google Scholar] [CrossRef]
  55. Zhi, F.; Ding, Y.; Wang, R.; Yang, Y.; Luo, K.; Hua, F. Exosomal hsa_circ_0006859 is a potential biomarker for postmenopausal osteoporosis and enhances adipogenic versus osteogenic differentiation in human bone marrow mesenchymal stem cells by sponging miR-431-5p. Stem Cell Res. Ther. 2021, 12, 157. [Google Scholar] [CrossRef] [PubMed]
  56. Zhang, H.; Zhu, J.K. Emerging roles of RNA processing factors in regulating long non-coding RNAs. Rna Biol. 2014, 11, 793–797. [Google Scholar] [CrossRef]
  57. Kanduri, C. Long noncoding RNAs: Lessons from genomic imprinting. Biochim. Biophys. Acta-Gene Regul. Mech. 2016, 1859, 102–111. [Google Scholar] [CrossRef]
  58. Nainar, S.; Feng, C.; Spitale, R.C. Chemical Tools for Dissecting the Role of lncRNAs in Epigenetic Regulation. ACS Chem. Biol. 2016, 11, 2091–2100. [Google Scholar] [CrossRef]
  59. González-Moro, I.; Garcia-Etxebarria, K.; Mendoza, L.M.; Fernández-Jiménez, N.; Mentxaka, J.; Olazagoitia-Garmendia, A.; Arroyo, M.N.; Sawatani, T.; Moreno-Castro, C.; Vinci, C.; et al. LncRNA ARGI Contributes to Virus-Induced Pancreatic β Cell Inflammation Through Transcriptional Activation of IFN-Stimulated Genes. Adv. Sci. 2023, 10, e2300063. [Google Scholar] [CrossRef]
  60. Fang, Y.; Fullwood, M.J. Roles, Functions, and Mechanisms of Long Non-coding RNAs in Cancer. Genom. Proteom. Bioinform. 2016, 14, 42–54. [Google Scholar] [CrossRef]
  61. Su, M.; Wang, H.; Wang, W.; Wang, Y.; Ouyang, L.; Pan, C.; Xia, L.; Cao, D.; Liao, Q. LncRNAs in DNA damage response and repair in cancer cells. Acta Biochim. Biophys. Sin. 2018, 50, 433–439. [Google Scholar] [CrossRef] [PubMed]
  62. Ghafouri-Fard, S.; Khoshbakht, T.; Hussen, B.M.; Baniahmad, A.; Taheri, M.; Rashnoo, F. A review on the role of PCA3 lncRNA in carcinogenesis with an especial focus on prostate cancer. Pathol. Res. Pract. 2022, 231, 153800. [Google Scholar] [CrossRef] [PubMed]
  63. Bourdoumis, A.; Papatsoris, A.G.; Chrisofos, M.; Efstathiou, E.; Skolarikos, A.; Deliveliotis, C. The novel prostate cancer antigen 3 (PCA3) biomarker. Int. Braz. J. Urol. 2010, 36, 665–668, discussion 669. [Google Scholar] [CrossRef] [PubMed]
  64. Hessels, D.; Schalken, J.A. The use of PCA3 in the diagnosis of prostate cancer. Nat. Rev. Urol. 2009, 6, 255–261. [Google Scholar] [CrossRef]
  65. Venderbos, L.D.; Luiting, H.; Hogenhout, R.; Roobol, M.J. Interaction of MRI and active surveillance in prostate cancer: Time to re-evaluate the active surveillance inclusion criteria. Urol. Oncol. Semin. Orig. Investig. 2023, 41, 82–87. [Google Scholar] [CrossRef]
  66. Zhang, Y.Z.; Li, Y.Y. Inflammatory bowel disease: Pathogenesis. World J. Gastroenterol. 2014, 20, 91–99. [Google Scholar] [CrossRef]
  67. Heydari, R.; Fayazzadeh, S.; Shahrokh, S.; Shekari, F.; Farsad, F.; Meyfour, A. Plasma Extracellular Vesicle LncRNA H19 as a Potential Diagnostic Biomarker for Inflammatory Bowel Diseases. Inflamm. Bowel Dis. 2024, 30, 795–807. [Google Scholar] [CrossRef]
  68. Chen, S.W.; Wang, P.Y.; Liu, Y.C.; Sun, L.; Zhu, J.; Zuo, S.; Ma, J.; Li, T.Y.; Zhang, J.L.; Chen, G.W.; et al. Effect of Long Noncoding RNA H19 Overexpression on Intestinal Barrier Function and Its Potential Role in the Pathogenesis of Ulcerative Colitis. Inflamm. Bowel Dis. 2016, 22, 2582–2592. [Google Scholar] [CrossRef]
  69. Benshoshan, M.; Sosnovski, K.E.; Braun, T.; Amir, A.; Anikster, Y.; Haberman Ziv, Y. P041 CDKN2B-AS1 (ANRIL) expression is decreased in Inflammatory Bowel Disease epithelia and in Celiac, and its reduction is linked with induced cells proliferation. J. Crohn’s Colitis 2021, 15, S152. [Google Scholar] [CrossRef]
  70. Han, J.; Li, Y.; Zhang, B.; Liu, H.; Wu, M.; Zhang, X. lncRNA TUG1 regulates ulcerative colitis through miR-142-5p/SOCS1 axis. Microb. Pathog. 2020, 143, 104139. [Google Scholar] [CrossRef]
  71. Lucafò, M.; Pugnetti, L.; Bramuzzo, M.; Curci, D.; Di Silvestre, A.; Marcuzzi, A.; Bergamo, A.; Martelossi, S.; Villanacci, V.; Bozzola, A.; et al. Long Non-Coding RNA GAS5 and Intestinal MMP2 and MMP9 Expression: A Translational Study in Pediatric Patients with IBD. Int. J. Mol. Sci. 2019, 20, 5280. [Google Scholar] [CrossRef] [PubMed]
  72. Yang, F.; Li, X.F.; Cheng, L.N.; Li, X.L. Long non-coding RNA CRNDE promotes cell apoptosis by suppressing miR-495 in inflammatory bowel disease. Exp. Cell Res. 2019, 382, 111484. [Google Scholar] [CrossRef] [PubMed]
  73. Song, J.; Ahn, C.; Chun, C.H.; Jin, E.J. A long non-coding RNA, GAS5, plays a critical role in the regulation of miR-21 during osteoarthritis. J. Orthop. Res. 2014, 32, 1628–1635. [Google Scholar] [CrossRef]
  74. Jiang, Y.; Du, T. Relation of circulating lncRNA GAS5 and miR-21 with biochemical indexes, stenosis severity, and inflammatory cytokines in coronary heart disease patients. J. Clin. Lab. Anal. 2022, 36, e24202. [Google Scholar] [CrossRef] [PubMed]
  75. Samsonov, R.; Burdakov, V.; Shtam, T.; Radzhabova, Z.; Vasilyev, D.; Tsyrlina, E.; Titov, S.; Ivanov, M.; Berstein, L.; Filatov, M.; et al. Plasma exosomal miR-21 and miR-181a differentiates follicular from papillary thyroid cancer. Tumor Biol. 2016, 37, 12011–12021. [Google Scholar] [CrossRef]
  76. Fang, T.; Lv, H.; Lv, G.; Li, T.; Wang, C.; Han, Q.; Yu, L.; Su, B.; Guo, L.; Huang, S.; et al. Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nat. Commun. 2018, 9, 191. [Google Scholar] [CrossRef]
  77. Luo, T.; Liu, Q.; Tan, A.; Duan, L.; Jia, Y.; Nong, L.; Tang, J.; Zhou, W.; Xie, W.; Lu, Y.; et al. Mesenchymal Stem Cell-Secreted Exosome Promotes Chemoresistance in Breast Cancer via Enhancing miR-21-5p-Mediated S100A6 Expression. Mol. Ther.-Oncolyt. 2020, 19, 283–293. [Google Scholar] [CrossRef]
  78. Gao, M.; Yu, T.; Liu, D.; Shi, Y.; Yang, P.; Zhang, J.; Wang, J.; Liu, Y.; Zhang, X. Sepsis plasma-derived exosomal miR-1-3p induces endothelial cell dysfunction by targeting SERP1. Clin. Sci. 2021, 135, 347–365. [Google Scholar] [CrossRef]
  79. Ibrahim, A.G.; Cheng, K.; Marbán, E. Exosomes as critical agents of cardiac regeneration triggered by cell therapy. Stem Cell Rep. 2014, 2, 606–619. [Google Scholar] [CrossRef]
  80. Liao, Z.; Chen, Y.; Duan, C.; Zhu, K.; Huang, R.; Zhao, H.; Hintze, M.; Pu, Q.; Yuan, Z.; Lv, L.; et al. Cardiac telocytes inhibit cardiac microvascular endothelial cell apoptosis through exosomal miRNA-21-5p-targeted cdip1 silencing to improve angiogenesis following myocardial infarction. Theranostics 2021, 11, 268–291. [Google Scholar] [CrossRef]
  81. Gámez-Valero, A.; Campdelacreu, J.; Vilas, D.; Ispierto, L.; Reñé, R.; Álvarez, R.; Armengol, M.P.; Borràs, F.E.; Beyer, K. Exploratory study on microRNA profiles from plasma-derived extracellular vesicles in Alzheimer’s disease and dementia with Lewy bodies. Transl. Neurodegener. 2019, 8, 31. [Google Scholar] [CrossRef] [PubMed]
  82. Li, C.; Wang, F.; Miao, P.; Yan, L.; Liu, S.; Wang, X.; Jin, Z.; Gu, Z. miR-138 Increases Depressive-Like Behaviors by Targeting SIRT1 in Hippocampus. Neuropsychiatr. Dis. Treat. 2020, 16, 949–957. [Google Scholar] [CrossRef] [PubMed]
  83. Gartz, M.; Beatka, M.; Prom, M.J.; Strande, J.L.; Lawlor, M.W. Cardiomyocyte-produced miR-339-5p mediates pathology in Duchenne muscular dystrophy cardiomyopathy. Hum. Mol. Genet. 2021, 30, 2347–2361. [Google Scholar] [CrossRef] [PubMed]
  84. Shan, L.; Yang, D.; Feng, F.; Zhu, D.; Li, X. miR-3146 induces neutrophil extracellular traps to aggravate gout flare. J. Clin. Lab. Anal. 2021, 35, e24032. [Google Scholar] [CrossRef]
  85. Lu, J.; Wang, Y.H.; Yoon, C.; Huang, X.Y.; Xu, Y.; Xie, J.W.; Wang, J.B.; Lin, J.X.; Chen, Q.Y.; Cao, L.L.; et al. Circular RNA circ-RanGAP1 regulates VEGFA expression by targeting miR-877-3p to facilitate gastric cancer invasion and metastasis. Cancer Lett. 2020, 471, 38–48. [Google Scholar] [CrossRef] [PubMed]
  86. Huang, X.Y.; Huang, Z.L.; Huang, J.; Xu, B.; Huang, X.Y.; Xu, Y.H.; Zhou, J.; Tang, Z.Y. Exosomal circRNA-100338 promotes hepatocellular carcinoma metastasis via enhancing invasiveness and angiogenesis. J. Exp. Clin. Cancer Res. 2020, 39, 20. [Google Scholar] [CrossRef]
  87. Pan, Z.; Zhao, R.; Li, B.; Qi, Y.; Qiu, W.; Guo, Q.; Zhang, S.; Zhao, S.; Xu, H.; Li, M.; et al. EWSR1-induced circNEIL3 promotes glioma progression and exosome-mediated macrophage immunosuppressive polarization via stabilizing IGF2BP3. Mol. Cancer 2022, 21, 16. [Google Scholar] [CrossRef]
  88. Sun, R.; Liu, W.; Zhao, Y.; Chen, H.; Wang, Z.; Zhang, Y.; Sun, X.; Cui, X. Exosomal circRNA as a novel potential therapeutic target for multiple myeloma-related myocardial damage. Cancer Cell Int. 2021, 21, 311. [Google Scholar] [CrossRef]
  89. Pan, W.; Hu, Y.; Wang, L.; Li, J. Circ_0003611 acts as a miR-885-5p sponge to aggravate the amyloid-β-induced neuronal injury in Alzheimer’s disease. Metab. Brain Dis. 2022, 37, 961–971. [Google Scholar] [CrossRef]
  90. Xiong, L.; Chen, L.; Wu, L.; He, W.; Chen, D.; Peng, Z.; Li, J.; Zhu, X.; Su, L.; Li, Y.; et al. Lipotoxicity-induced circGlis3 impairs beta cell function and is transmitted by exosomes to promote islet endothelial cell dysfunction. Diabetologia 2022, 65, 188–205. [Google Scholar] [CrossRef]
  91. Yang, J.; Cheng, M.; Gu, B.; Wang, J.; Yan, S.; Xu, D. CircRNA_09505 aggravates inflammation and joint damage in collagen-induced arthritis mice via miR-6089/AKT1/NF-κB axis. Cell Death Dis. 2020, 11, 833. [Google Scholar] [CrossRef] [PubMed]
  92. Guo, G.; Wang, H.; Ye, L.; Shi, X.; Yan, K.; Lin, K.; Huang, Q.; Li, B.; Lin, Q.; Zhu, L.; et al. Hsa_circ_0000479 as a Novel Diagnostic Biomarker of Systemic Lupus Erythematosus. Front. Immunol. 2019, 10, 2281. [Google Scholar] [CrossRef]
  93. Jiang, Z.; Huang, L.; Chen, L.; Zhou, J.; Liang, B.; Bai, X.; Wu, L.; Huang, H. Circular RNA profile in Graves’ disease and potential function of hsa_circ_0090364. Endocr. Connect. 2022, 11, e220030. [Google Scholar] [CrossRef] [PubMed]
  94. Jiang, Y.; Zhao, H.; Chen, Y.; Li, K.; Li, T.; Chen, J.; Zhang, B.; Guo, C.; Qing, L.; Shen, J.; et al. Exosomal long noncoding RNA HOXD-AS1 promotes prostate cancer metastasis via miR-361-5p/FOXM1 axis. Cell Death Dis. 2021, 12, 1129. [Google Scholar] [CrossRef]
  95. Ni, C.; Fang, Q.Q.; Chen, W.Z.; Jiang, J.X.; Jiang, Z.; Ye, J.; Zhang, T.; Yang, L.; Meng, F.B.; Xia, W.J.; et al. Breast cancer-derived exosomes transmit lncRNA SNHG16 to induce CD73+γδ1 Treg cells. Signal Transduct. Target. Ther. 2020, 5, 41. [Google Scholar] [CrossRef]
  96. Zheng, H.; Chen, C.; Luo, Y.; Yu, M.; He, W.; An, M.; Gao, B.; Kong, Y.; Ya, Y.; Lin, Y.; et al. Tumor-derived exosomal BCYRN1 activates WNT5A/VEGF-C/VEGFR3 feedforward loop to drive lymphatic metastasis of bladder cancer. Clin. Transl. Med. 2021, 11, e497. [Google Scholar] [CrossRef]
  97. Sun, L.; He, X.; Zhang, T.; Tao, G.; Wang, X. Knockdown of lnc-KCNC3-3:1 Alleviates the Development of Atherosclerosis via Downregulation of JAK1/STAT3 Signaling Pathway. Front. Cardiovasc. Med. 2021, 8, 701058. [Google Scholar] [CrossRef]
  98. Sato, M.; Kadomatsu, T.; Miyata, K.; Warren, J.S.; Tian, Z.; Zhu, S.; Horiguchi, H.; Makaju, A.; Bakhtina, A.; Morinaga, J.; et al. The lncRNA Caren antagonizes heart failure by inactivating DNA damage response and activating mitochondrial biogenesis. Nat. Commun. 2021, 12, 2529. [Google Scholar] [CrossRef]
  99. Wang, Q.; Han, C.L.; Wang, K.L.; Sui, Y.P.; Li, Z.B.; Chen, N.; Fan, S.Y.; Shimabukuro, M.; Wang, F.; Meng, F.G. Integrated analysis of exosomal lncRNA and mRNA expression profiles reveals the involvement of lnc-MKRN2-42:1 in the pathogenesis of Parkinson’s disease. CNS Neurosci. Ther. 2020, 26, 527–537. [Google Scholar] [CrossRef]
  100. Wang, D.; Wang, P.; Bian, X.; Xu, S.; Zhou, Q.; Zhang, Y.; Ding, M.; Han, M.; Huang, L.; Bi, J.; et al. Elevated plasma levels of exosomal BACE1-AS combined with the volume and thickness of the right entorhinal cortex may serve as a biomarker for the detection of Alzheimer’s disease. Mol. Med. Rep. 2020, 22, 227–238. [Google Scholar] [CrossRef]
  101. Tian, F.; Wang, J.; Zhang, Z.; Yang, J. LncRNA SNHG7/miR-34a-5p/SYVN1 axis plays a vital role in proliferation, apoptosis and autophagy in osteoarthritis. Biol. Res. 2020, 53, 9. [Google Scholar] [CrossRef] [PubMed]
  102. Kuai, X.Y.; Yu, S.Y.; Cui, X.F.; Zhao, X.J.; Mao, X.Q.; Yu, Y.; Hu, T.; Zhang, H.J.; Zhou, C.L. LncRNA LUCAT1 as a Plasma Biomarker for Assessing Disease Activity in Adult Patients with Crohn’s Disease. Gastroenterol. Res. Pract. 2021, 2021, 5557357. [Google Scholar] [CrossRef]
  103. Ji, X.; Sun, J.; Wang, Z. High level of lncRNA NR2F1-AS1 predict the onset and progression of diabetic retinopathy in type 2 diabetes. Exp. Eye Res. 2022, 219, 109069. [Google Scholar] [CrossRef]
  104. Group, V.S. A randomized controlled clinical trial of intravitreous fomivirsen for treatment of newly diagnosed peripheral cytomegalovirus retinitis in patients with AIDS. Am. J. Ophthalmol. 2002, 133, 467–474. [Google Scholar] [CrossRef]
  105. Geary, R.S.; Henry, S.P.; Grillone, L.R. Fomivirsen: Clinical pharmacology and potential drug interactions. Clin. Pharmacokinet. 2002, 41, 255–260. [Google Scholar] [CrossRef] [PubMed]
  106. Adams, D.; Gonzalez-Duarte, A.; O’Riordan, W.D.; Yang, C.C.; Ueda, M.; Kristen, A.V.; Tournev, I.; Schmidt, H.H.; Coelho, T.; Berk, J.L.; et al. Patisiran, an RNAi Therapeutic, for Hereditary Transthyretin Amyloidosis. N. Engl. J. Med. 2018, 379, 11–21. [Google Scholar] [CrossRef]
  107. Shah, M.Y.; Ferrajoli, A.; Sood, A.K.; Lopez-Berestein, G.; Calin, G.A. microRNA Therapeutics in Cancer—An Emerging Concept. EBioMedicine 2016, 12, 34–42. [Google Scholar] [CrossRef]
  108. Ling, H.; Fabbri, M.; Calin, G.A. MicroRNAs and other non-coding RNAs as targets for anticancer drug development. Nat. Rev. Drug Discov. 2013, 12, 847–865. [Google Scholar] [CrossRef]
  109. Abdelaal, A.M.; Sohal, I.S.; Iyer, S.; Sudarshan, K.; Kothandaraman, H.; Lanman, N.A.; Low, P.S.; Kasinski, A.L. A first-in-class fully modified version of miR-34a with outstanding stability, activity, and anti-tumor efficacy. Oncogene 2023, 42, 2985–2999. [Google Scholar] [CrossRef]
  110. Liu, N.; Kataoka, M.; Wang, Y.; Pu, L.; Dong, X.; Fu, X.; Zhang, F.; Gao, F.; Liang, T.; Pei, J.; et al. LncRNA LncHrt preserves cardiac metabolic homeostasis and heart function by modulating the LKB1-AMPK signaling pathway. Basic Res. Cardiol. 2021, 116, 48. [Google Scholar] [CrossRef]
  111. Gragoudas, E.S.; Adamis, A.P.; Cunningham, E.T., Jr.; Feinsod, M.; Guyer, D.R.; VEGF Inhibition Study in Ocular Neovascularization Clinical Trial Group. Pegaptanib for neovascular age-related macular degeneration. N. Engl. J. Med. 2004, 351, 2805–2816. [Google Scholar] [CrossRef] [PubMed]
  112. Santos, R.D.; Raal, F.J.; Donovan, J.M.; Cromwell, W.C. Mipomersen preferentially reduces small low-density lipoprotein particle number in patients with hypercholesterolemia. J. Clin. Lipidol. 2015, 9, 201–209. [Google Scholar] [CrossRef]
  113. Mendell, J.R.; Rodino-Klapac, L.R.; Sahenk, Z.; Roush, K.; Bird, L.; Lowes, L.P.; Alfano, L.; Gomez, A.M.; Lewis, S.; Kota, J.; et al. Eteplirsen for the treatment of Duchenne muscular dystrophy. Ann. Neurol. 2013, 74, 637–647. [Google Scholar] [CrossRef]
  114. Mercuri, E.; Darras, B.T.; Chiriboga, C.A.; Day, J.W.; Campbell, C.; Connolly, A.M.; Iannaccone, S.T.; Kirschner, J.; Kuntz, N.L.; Saito, K.; et al. Nusinersen versus Sham Control in Later-Onset Spinal Muscular Atrophy. N. Engl. J. Med. 2018, 378, 625–635. [Google Scholar] [CrossRef]
  115. Benson, M.D.; Waddington-Cruz, M.; Berk, J.L.; Polydefkis, M.; Dyck, P.J.; Wang, A.K.; Planté-Bordeneuve, V.; Barroso, F.A.; Merlini, G.; Obici, L.; et al. Inotersen Treatment for Patients with Hereditary Transthyretin Amyloidosis. N. Engl. J. Med. 2018, 379, 22–31. [Google Scholar] [CrossRef] [PubMed]
  116. Balwani, M.; Sardh, E.; Ventura, P.; Peiró, P.A.; Rees, D.C.; Stölzel, U.; Bissell, D.M.; Bonkovsky, H.L.; Windyga, J.; Anderson, K.E.; et al. Phase 3 Trial of RNAi Therapeutic Givosiran for Acute Intermittent Porphyria. N. Engl. J. Med. 2020, 382, 2289–2301. [Google Scholar] [CrossRef]
  117. Scaglioni, D.; Catapano, F.; Ellis, M.; Torelli, S.; Chambers, D.; Feng, L.; Beck, M.; Sewry, C.; Monforte, M.; Harriman, S.; et al. The administration of antisense oligonucleotide golodirsen reduces pathological regeneration in patients with Duchenne muscular dystrophy. Acta Neuropathol. Commun. 2021, 9, 7. [Google Scholar] [CrossRef]
  118. Kolovou, G.; Kolovou, V.; Katsiki, N. Volanesorsen: A New Era in the Treatment of Severe Hypertriglyceridemia. J. Clin. Med. 2022, 11, 982. [Google Scholar] [CrossRef] [PubMed]
  119. Roshmi, R.R.; Yokota, T. Pharmacological Profile of Viltolarsen for the Treatment of Duchenne Muscular Dystrophy: A Japanese Experience. Clin. Pharmacol. 2021, 13, 235–242. [Google Scholar] [CrossRef]
  120. Garrelfs, S.F.; Frishberg, Y.; Hulton, S.A.; Koren, M.J.; O’Riordan, W.D.; Cochat, P.; Deschênes, G.; Shasha-Lavsky, H.; Saland, J.M.; Van’t Hoff, W.G.; et al. Lumasiran, an RNAi Therapeutic for Primary Hyperoxaluria Type 1. N. Engl. J. Med. 2021, 384, 1216–1226. [Google Scholar] [CrossRef]
  121. Zakeri, S.E.; Pradeep, S.P.; Kasina, V.; Laddha, A.P.; Manautou, J.E.; Bahal, R. Casimersen for the treatment of Duchenne muscular dystrophy. Trends Pharmacol. Sci. 2022, 43, 607–608. [Google Scholar] [CrossRef]
  122. Samuel, E.; Watford, M.; Egolum, U.O.; Ombengi, D.N.; Ling, H.; Cates, D.W. Inclisiran: A First-in-Class siRNA Therapy for Lowering Low-Density Lipoprotein Cholesterol. Ann. Pharmacother. 2023, 57, 317–324. [Google Scholar] [CrossRef]
  123. Nie, T.; Heo, Y.A.; Shirley, M. Vutrisiran: A Review in Polyneuropathy of Hereditary Transthyretin-Mediated Amyloidosis. Drugs 2023, 83, 1425–1432. [Google Scholar] [CrossRef] [PubMed]
  124. Baum, M.A.; Langman, C.; Cochat, P.; Lieske, J.C.; Moochhala, S.H.; Hamamoto, S.; Satoh, H.; Mourani, C.; Ariceta, G.; Torres, A.; et al. PHYOX2: A pivotal randomized study of nedosiran in primary hyperoxaluria type 1 or 2. Kidney Int. 2023, 103, 207–217. [Google Scholar] [CrossRef] [PubMed]
  125. Coelho, T.; Marques, W., Jr.; Dasgupta, N.R.; Chao, C.C.; Parman, Y.; França, M.C., Jr.; Guo, Y.C.; Wixner, J.; Ro, L.S.; Calandra, C.R.; et al. Eplontersen for Hereditary Transthyretin Amyloidosis With Polyneuropathy. JAMA 2023, 330, 1448–1458. [Google Scholar] [CrossRef] [PubMed]
  126. Miller, T.M.; Cudkowicz, M.E.; Genge, A.; Shaw, P.J.; Sobue, G.; Bucelli, R.C.; Chiò, A.; Van Damme, P.; Ludolph, A.C.; Glass, J.D.; et al. Trial of Antisense Oligonucleotide Tofersen for SOD1 ALS. N. Engl. J. Med. 2022, 387, 1099–1110. [Google Scholar] [CrossRef]
  127. Bergmark, B.A.; Marston, N.A.; Prohaska, T.A.; Alexander, V.J.; Zimerman, A.; Moura, F.A.; Murphy, S.A.; Goodrich, E.L.; Zhang, S.; Gaudet, D.; et al. Olezarsen for Hypertriglyceridemia in Patients at High Cardiovascular Risk. N. Engl. J. Med. 2024, 390, 1770–1780. [Google Scholar] [CrossRef]
  128. Zhou, W.; Jiang, L.; Liao, S.; Wu, F.; Yang, G.; Hou, L.; Liu, L.; Pan, X.; Jia, W.; Zhang, Y. Vaccines’ New Era-RNA Vaccine. Viruses 2023, 15, 1760. [Google Scholar] [CrossRef]
  129. He, Q.; Gao, H.; Tan, D.; Zhang, H.; Wang, J.Z. mRNA cancer vaccines: Advances, trends and challenges. Acta Pharm. Sin. B 2022, 12, 2969–2989. [Google Scholar] [CrossRef]
  130. Vishweshwaraiah, Y.L.; Dokholyan, N.V. mRNA vaccines for cancer immunotherapy. Front. Immunol. 2022, 13, 1029069. [Google Scholar] [CrossRef]
  131. Niu, D.; Wu, Y.; Lian, J. Circular RNA vaccine in disease prevention and treatment. Signal Transduct. Target. Ther. 2023, 8, 341. [Google Scholar] [CrossRef] [PubMed]
  132. Wesselhoeft, R.A.; Kowalski, P.S.; Parker-Hale, F.C.; Huang, Y.; Bisaria, N.; Anderson, D.G. RNA Circularization Diminishes Immunogenicity and Can Extend Translation Duration In Vivo. Mol. Cell 2019, 74, 508–520.e504. [Google Scholar] [CrossRef]
  133. Kameda, S.; Ohno, H.; Saito, H. Synthetic circular RNA switches and circuits that control protein expression in mammalian cells. Nucleic Acids Res. 2023, 51, e24. [Google Scholar] [CrossRef]
  134. Usman, N.; Cedergren, R. Exploiting the chemical synthesis of RNA. Trends Biochem. Sci. 1992, 17, 334–339. [Google Scholar] [CrossRef] [PubMed]
  135. Rong, M.; He, B.; McAllister, W.T.; Durbin, R.K. Promoter specificity determinants of T7 RNA polymerase. Proc. Natl. Acad. Sci. USA 1998, 95, 515–519. [Google Scholar] [CrossRef] [PubMed]
  136. Müller, S.; Appel, B. In vitro circularization of RNA. Rna Biol. 2017, 14, 1018–1027. [Google Scholar] [CrossRef] [PubMed]
  137. Sokolova, N.I.; Ashirbekova, D.T.; Dolinnaya, N.G.; Shabarova, Z.A. Chemical reactions within DNA duplexes cyanogen bromide as an effective oligodeoxyribonucleotide coupling agent. FEBS Lett. 1988, 232, 153–155. [Google Scholar] [CrossRef]
  138. Petkovic, S.; Müller, S. RNA circularization strategies in vivo and in vitro. Nucleic Acids Res. 2015, 43, 2454–2465. [Google Scholar] [CrossRef]
  139. Bai, Y.; Liu, D.; He, Q.; Liu, J.; Mao, Q.; Liang, Z. Research progress on circular RNA vaccines. Front. Immunol. 2022, 13, 1091797. [Google Scholar] [CrossRef]
  140. Xie, J.; Ye, F.; Deng, X.; Tang, Y.; Liang, J.Y.; Huang, X.; Sun, Y.; Tang, H.; Lei, J.; Zheng, S.; et al. Circular RNA: A promising new star of vaccine. J. Transl. Intern. Med. 2023, 11, 372–381. [Google Scholar] [CrossRef]
  141. Neumann, E.; Schaefer-Ridder, M.; Wang, Y.; Hofschneider, P.H. Gene transfer into mouse lyoma cells by electroporation in high electric fields. EMBO J. 1982, 1, 841–845. [Google Scholar] [CrossRef] [PubMed]
  142. Klein, R.M.; Wolf, E.D.; Wu, R.; Sanford, J.C. High-velocity microprojectiles for delivering nucleic acids into living cells. 1987. Biotechnology 1992, 24, 384–386. [Google Scholar] [PubMed]
  143. Sun, X.; Zeng, L.; Huang, Y. Transcutaneous delivery of DNA/mRNA for cancer therapeutic vaccination. J. Gene Med. 2019, 21, e3089. [Google Scholar] [CrossRef] [PubMed]
  144. Tenchov, R.; Bird, R.; Curtze, A.E.; Zhou, Q. Lipid Nanoparticles─From Liposomes to mRNA Vaccine Delivery, a Landscape of Research Diversity and Advancement. ACS Nano 2021, 15, 16982–17015. [Google Scholar] [CrossRef]
  145. Cullis, P.R.; Felgner, P.L. The 60-year evolution of lipid nanoparticles for nucleic acid delivery. Nat. Rev. Drug Discov. 2024, 23, 709–722. [Google Scholar] [CrossRef]
  146. Laumont, C.M.; Vincent, K.; Hesnard, L.; Audemard, É.; Bonneil, É.; Laverdure, J.P.; Gendron, P.; Courcelles, M.; Hardy, M.P.; Côté, C.; et al. Noncoding regions are the main source of targetable tumor-specific antigens. Sci. Transl. Med. 2018, 10, eaau5516. [Google Scholar] [CrossRef]
  147. Chong, C.; Coukos, G.; Bassani-Sternberg, M. Identification of tumor antigens with immunopeptidomics. Nat. Biotechnol. 2022, 40, 175–188. [Google Scholar] [CrossRef]
  148. Huang, D.; Zhu, X.; Ye, S.; Zhang, J.; Liao, J.; Zhang, N.; Zeng, X.; Wang, J.; Yang, B.; Zhang, Y.; et al. Tumour circular RNAs elicit anti-tumour immunity by encoding cryptic peptides. Nature 2024, 625, 593–602. [Google Scholar] [CrossRef]
  149. Qu, L.; Yi, Z.; Shen, Y.; Lin, L.; Chen, F.; Xu, Y.; Wu, Z.; Tang, H.; Zhang, X.; Tian, F.; et al. Circular RNA vaccines against SARS-CoV-2 and emerging variants. Cell 2022, 185, 1728–1744.e1716. [Google Scholar] [CrossRef]
  150. Li, H.; Peng, K.; Yang, K.; Ma, W.; Qi, S.; Yu, X.; He, J.; Lin, X.; Yu, G. Circular RNA cancer vaccines drive immunity in hard-to-treat malignancies. Theranostics 2022, 12, 6422–6436. [Google Scholar] [CrossRef]
  151. Seephetdee, C.; Bhukhai, K.; Buasri, N.; Leelukkanaveera, P.; Lerdwattanasombat, P.; Manopwisedjaroen, S.; Phueakphud, N.; Kuhaudomlarp, S.; Olmedillas, E.; Saphire, E.O.; et al. A circular mRNA vaccine prototype producing VFLIP-X spike confers a broad neutralization of SARS-CoV-2 variants by mouse sera. Antivir. Res. 2022, 204, 105370. [Google Scholar] [CrossRef] [PubMed]
  152. Yang, J.; Zhu, J.; Sun, J.; Chen, Y.; Du, Y.; Tan, Y.; Wu, L.; Zhai, M.; Wei, L.; Li, N.; et al. Intratumoral delivered novel circular mRNA encoding cytokines for immune modulation and cancer therapy. Mol. Ther. Nucleic Acids 2022, 30, 184–197. [Google Scholar] [CrossRef]
  153. Zhou, J.; Ye, T.; Yang, Y.; Li, E.; Zhang, K.; Wang, Y.; Chen, S.; Hu, J.; Zhang, K.; Liu, F.; et al. Circular RNA vaccines against monkeypox virus provide potent protection against vaccinia virus infection in mice. Mol. Ther. 2024, 32, 1779–1789. [Google Scholar] [CrossRef]
  154. Wang, F.; Cai, G.; Wang, Y.; Zhuang, Q.; Cai, Z.; Li, Y.; Gao, S.; Li, F.; Zhang, C.; Zhao, B.; et al. Circular RNA-based neoantigen vaccine for hepatocellular carcinoma immunotherapy. MedComm 2024, 5, e667. [Google Scholar] [CrossRef]
  155. Wan, J.; Wang, Z.; Wang, L.; Wu, L.; Zhang, C.; Zhou, M.; Fu, Z.F.; Zhao, L. Circular RNA vaccines with long-term lymph node-targeting delivery stability after lyophilization induce potent and persistent immune responses. MBio 2024, 15, e0177523. [Google Scholar] [CrossRef]
  156. Xu, S.; Xu, Y.; Solek, N.C.; Chen, J.; Gong, F.; Varley, A.J.; Golubovic, A.; Pan, A.; Dong, S.; Zheng, G.; et al. Tumor-Tailored Ionizable Lipid Nanoparticles Facilitate IL-12 Circular RNA Delivery for Enhanced Lung Cancer Immunotherapy. Adv. Mater. 2024, 36, e2400307. [Google Scholar] [CrossRef]
  157. Yue, X.; Zhong, C.; Cao, R.; Liu, S.; Qin, Z.; Liu, L.; Zhai, Y.; Luo, W.; Lian, Y.; Zhang, M.; et al. CircRNA based multivalent neuraminidase vaccine induces broad protection against influenza viruses in mice. Npj Vaccines 2024, 9, 170. [Google Scholar] [CrossRef] [PubMed]
  158. Liu, X.; Li, Z.; Li, X.; Wu, W.; Jiang, H.; Zheng, Y.; Zhou, J.; Ye, X.; Lu, J.; Wang, W.; et al. A single-dose circular RNA vaccine prevents Zika virus infection without enhancing dengue severity in mice. Nat. Commun. 2024, 15, 8932. [Google Scholar] [CrossRef]
  159. Jenike, A.E.; Halushka, M.K. miR-21: A non-specific biomarker of all maladies. Biomark. Res. 2021, 9, 18. [Google Scholar] [CrossRef]
  160. Tomlins, S.A.; Bjartell, A.; Chinnaiyan, A.M.; Jenster, G.; Nam, R.K.; Rubin, M.A.; Schalken, J.A. ETS gene fusions in prostate cancer: From discovery to daily clinical practice. Eur. Urol. 2009, 56, 275–286. [Google Scholar] [CrossRef]
  161. Yang, Z.; Yu, L.; Wang, Z. PCA3 and TMPRSS2-ERG gene fusions as diagnostic biomarkers for prostate cancer. Chin. J. Cancer Res. 2016, 28, 65–71. [Google Scholar] [CrossRef] [PubMed]
  162. Warli, S.M.; Warli, M.H.; Prapiska, F.F. PCA3 and TMPRSS2: ERG Urine Level as Diagnostic Biomarker of Prostate Cancer. Res. Rep. Urol. 2023, 15, 149–155. [Google Scholar] [CrossRef] [PubMed]
Figure 1. In vitro synthesis methods of circRNAs. (A) The chemical strategy. (B) The enzymatic strategy. (C) Group I intron-based permuted intron–exon (PIE) system. The sequence to be circularized is inserted into the exon regions (E1 and E2). In the presence of guanosine triphosphate (GTP), self-splicing is initiated, resulting in circularization, and two half-intron fragments are released. The circularized circRNA contains E1 and E2 (referred to as the scar sequence), which may affect the target sequence. (D) Group II intron-based PIE system. This system has no redundant sequences. After the lariat intron is removed, the ends of the exon are joined to form a circle.
Figure 1. In vitro synthesis methods of circRNAs. (A) The chemical strategy. (B) The enzymatic strategy. (C) Group I intron-based permuted intron–exon (PIE) system. The sequence to be circularized is inserted into the exon regions (E1 and E2). In the presence of guanosine triphosphate (GTP), self-splicing is initiated, resulting in circularization, and two half-intron fragments are released. The circularized circRNA contains E1 and E2 (referred to as the scar sequence), which may affect the target sequence. (D) Group II intron-based PIE system. This system has no redundant sequences. After the lariat intron is removed, the ends of the exon are joined to form a circle.
Ijms 26 03055 g001
Figure 2. Delivery methods of circRNA vaccines. (A) Early experimental delivery methods include electroporation, gene gun, and microinjection. (B) The structure of using LNP to deliver circRNA vaccines.
Figure 2. Delivery methods of circRNA vaccines. (A) Early experimental delivery methods include electroporation, gene gun, and microinjection. (B) The structure of using LNP to deliver circRNA vaccines.
Ijms 26 03055 g002
Table 2. Summary of ncRNAs as potential disease biomarkers.
Table 2. Summary of ncRNAs as potential disease biomarkers.
Related DiseasesBiofluidPotential BiomarkersTarget Genes/Pathways/Mechanistic ApproachesReferences
Thyroid CancerPlasma miR-21 and miR-181a-5pN/A[75]
Liver CancerCCM and serummiR-1247-3pB4GALT3[76]
Breast CancerMSC CCMmiR-21-5pS100A6[77]
SepsisPlasmamiR-1-3pSERP1[78]
Heart FailurePlasmamiR-146a IRAK-1, TRAF6, NOX-4
SMAD4, and TGF-β
[79]
Myocardial InfarctionCardiac telocyte CCMmiR-21-5pCDIP1[80]
Alzheimer’s DiseasePlasmamiR-451a and miR-21-5pN/A[81]
Depressive DisorderCortical neuron CCMmiR-138SIRT1[82]
Duchenne Muscular DystrophyDMD cardiomyocytes CCMmiR-339-5p MDM2, GSK3A and MAP2K3[83]
GoutPlasmamiR-3146Mediates NETs formation[84]
Gastric CancerPlasmacirc-RanGAP1miR-877-3p/VEGFA axis[85]
Hepatocellular CarcinomaHCC CCMcircRNA-100338N/A[86]
GliomaGBM CCMcircNEIL3Stabilizing IGF2BP3[87]
Myeloma-Related Myocardial DamageSerumcirc-G042080miR-4268/TLR4 axis[88]
Alzheimer’s DiseaseSerumcirc_0003611miR-885-5p/KREMEN1 axis[89]
Type 2 DiabetesSerumcircGlis3Regulates GMEB1 degradation and HSP27 phosphorylation[90]
Rheumatoid ArthritisPlasmacircRNA_09505miR-6089/AKT1/NF-κB axis[91]
Systemic Lupus ErythematosusPlasmahsa_circ_0000479Metabolic and the Wnt signaling pathway[92]
Graves’ DiseasePlasmahsa_circ_0090364hsa-miR-378a-3p/IL-6ST/IL21R axis[93]
Prostate CancerSerumHOXD-AS1miR-361-5p/FOXM1 axis[94]
Breast CancerSerumSNHG16miR-16–5p/SMAD5 axis[95]
Bladder CancerUrinelncBCYRN1Activates WNT5A/VEGF-C/VEGFR3 feedforward loop[96]
AtherosclerosisHUVEC CCMlnc-KCNC3-3:1JAK1/STAT3 signaling pathway[97]
Heart FailurePlasmalncRNA-NRFN/A[98]
Parkinson’s DiseasePlasmalnc-MKRN2-42:1N/A[99]
Alzheimer’s DiseasePlasmaBACE1-ASN/A[100]
OsteoarthritisOA CCMSNHG7miR-34a-5p/SYVN1 axis[101]
Crohn’s DiseasePlasmaLUCAT1NA[102]
Diabetic Retinopathy in Type 2 DiabetesPlasmaNR1F1-AS2Moderates EndMT[103]
CCM: cell culture media; B4GALT3: beta-1,4-galactosyltransferase 3; MSC: mesenchymal stem cell; S100A6: S100 calcium binding protein A6; SERP1: stress-associated endoplasmic reticulum protein 1; HCC: hepatocellular carcinoma; IRAK-1: interleukin 1 receptor-associated kinase 1; TRAF6: TNF receptor-associated factor 6; NOX-4: NADPH oxidase 4; SMAD4: SMAD family member 4; TGF-β: transforming growth factor beta; CDIP1: cell death-inducing P53 target 1; SIRT1: sirtuin 1; DMD: Duchenne muscular dystrophy; MDM2: MDM2 proto-oncogene; GSK3A: glycogen synthase kinase 3 alpha; MAP2K3: mitogen-activated protein kinase kinase 3; NETs: neutrophil extracellular traps; VEGFA: vascular endothelial growth factor A; GBM: glioblastoma multiforme; IGF2BP3: insulin-like growth factor 2 MRNA binding protein 3; TLR4: toll-like receptor 4; KREMEN1: kringle-containing transmembrane protein 1; GMEB1: glucocorticoid modulatory element binding protein 1; HSP27: heat shock protein 27; AKT1: AKT serine/threonine kinase 1; NF-κB: nuclear factor kappa B; IL-6ST: interleukin 6 cytokine family signal transducer; IL21R: interleukin 21 receptor; FOXM1: forkhead box M1; SMAD5: SMAD family member 5; WNT5A: wnt family member 5A; VEGF-C: vascular endothelial growth factor C; VEGFR3: vascular endothelial growth factor receptor 3; JAK1: Janus kinase 1; STAT3: signal transducer and activator of transcription 3; SYVN1: synoviolin 1; EndMT: endothelial-to-mesenchymal transition.
Table 3. RNA therapeutics approved by the FDA and/or EMA.
Table 3. RNA therapeutics approved by the FDA and/or EMA.
DrugsTypesRoute of AdministrationTarget OrganMechanism of ActionRelated DiseasesFDA and/or EMA Approval YearReferences
FomivirsenASOIVTEyeTargeting and silencing the mRNA of CMV IE2 proteinCMV retinitis in immunocompromised patientsFDA (1998)
EMA (1999)
[104]
PegaptanibPhosphate oligonucleotide aptamerIVTEyeInhibiting VEGF-165nAMDFDA (2004)[111]
MipomersenASOSCLiverTargeting and silencing the mRNA of apolipoprotein B to reduce LDL levelsHoFHFDA (2013)
EMA (2012)
[112]
EteplirsenASOIVMuscleTargeting and splicing the pre-mRNA of defective DMD dystrophin proteinDMDFDA (2016)[113]
NusinersenASOITHCentral nervous systemTargeting and splicing the pre-mRNA of defective SMN proteinSMAFDA (2016)
EMA (2017)
[114]
PatisiransiRNAIVLiverTargeting and silencing the mRNA of TTR protein to prevent the production of TTR proteinATTR amyloidosisFDA (2018)[106]
InotersenASOSCLiverTargeting and silencing the mRNA of TTR protein to prevent the production of TTR proteinATTR amyloidosisFDA (2018)
EMA (2018)
[115]
GivosiransiRNASCLiverTargeting and silencing the mRNA of ALAS1 to reduce ALAS1 levelsAHPFDA (2019)
EMA (2020)
[116]
GolodirsenASOIVMuscleTargeting the splicing of DMD pre-mRNA (exon 53 skipping)DMDFDA (2019)[117]
VolanesorsenASOSCLiverTargeting and silencing the mRNA of APOC3 to reduce triglyceride productionFCSEMA (2019)[118]
ViltolarsenASOIVMuscleTargeting the splicing of DMD pre-mRNA (exon 53 skipping)DMDFDA (2020)[119]
LumasiransiRNASCLiverTargeting and silencing the mRNA of HAO1 to reduce GO levelsPH1FDA (2020)
EMA (2020)
[120]
CasimersenASOIVMuscleTargeting the splicing of DMD pre-mRNA (exon 45 skipping)DMDFDA (2021)[121]
InclisiransiRNASCLiverTargeting PCSK9 to inhibit its synthesis and reduce LDL-C levelsASCVDFDA (2021)
EMA (2020)
[122]
VutrisiransiRNASCLiverTargeting and silencing the mRNA of TTR protein to prevent the production of TTR proteinATTR amyloidosisFDA (2022)
EMA (2022)
[123]
NedosiransiRNASCLiverTargeting liver LDH mRNA to reduce the expression of LDHPH1FDA (2023)[124]
EplontersenASOSCLiverTargeting and silencing the mRNA of TTR protein to prevent the production of TTR proteinATTR amyloidosisFDA (2023)[125]
TofersenASOITHMuscleTargeting SOD1 mRNA to reduce the synthesis of SOD1 proteinSOD1-ALSFDA (2023)
EMA (2023)
[126]
OlezarsenASOSCLiverReducing hepatic synthesis of apolipoprotein C-III to lower plasma triglyceride levelsFCSFDA (2024)[127]
ASO: antisense oligonucleotide; siRNA: small interfering RNA; IVT: intravitreal; SC: subcutaneous; IV: intravenous; ITH: intrathecal; CMV: cytomegalovirus; IE2: immediate Early 2; VEGF-165: vascular endothelial growth factor 165; LDL: low-density lipoprotein; DMD: Duchenne muscular dystrophy; SMN: survival motor neuron; TTR: transthyretin; ALAS1: 5′-aminolevulinate synthase 1; APOC3: apolipoprotein CIII; HAO1: hydroxyacid oxidase 1; GO: glycolate oxidase; PCSK9: proprotein convertase subtilisin/kexin Type 9; LDL-C: low-density lipoprotein cholesterol; nAMD: neovascular age-related macular degeneration; HoFH: homozygous familial hypercholesterolemia; SMA: spinal muscular atrophy; ATTR: transthyretin amyloidosis; AHP: acute hepatic porphyria; ASCVD: atherosclerotic cardiovascular disease; PH1: primary hyperoxaluria type 1; SOD1-ALS: amyotrophic lateral sclerosis (ALS) associated with a mutation in the superoxide dismutase 1 (SOD1) gene; FCS: familial chylomicronemia syndrome.
Table 4. Summary of circRNA vaccines.
Table 4. Summary of circRNA vaccines.
VaccinesCyclization StrategyDeliveryAntigenReferences
circRNARBDRibozymatic autocatalysisLNPSARS-CoV-2 RBD antigen[149]
VFLIP-XT4 RNA ligaseLNPSARS-CoV-2 spiking protein[151]
circRNAOVA-luc-LNPRibozymatic autocatalysisLNPOVA [257-264]-luciferase[150]
CircRNA encoding cytokinesRibozymatic autocatalysisLNPActive IL-15\IL-12\GM-CSF\IFN-α 2b[152]
cirA29L, cirA35R, cirB6R, and cirM1RRibozymatic autocatalysisLNPMPXV proteins A29L, A35R, B6R, and M1R[153]
circRNA3×PTPN2 Ribozymatic autocatalysisLNPPTPN2[154]
circRNA-GRibozymatic autocatalysisLNPGlycoproteins of the RABV vaccine strain SAD-L16[155]
IL12-circRNARibozymatic autocatalysisLNPIL-12[156]
circRNA-NARibozymatic autocatalysisLNPNA[157]
EDIII-Fc circRNARibozymatic autocatalysisLNPDimeric EDIII-Fc fusion[158]
RBD: receptor-binding domain; SARS: severe acute respiratory syndrome; CoV: coronavirus; OVA: ovalbumin; IL: interleukin; GM-CSF: granulocyte-macrophage colony-stimulating factor; IFN: interferon; MPXV: monkeypox virus; PTPN2: protein Tyrosine Phosphatase Non-Receptor Type 2; RABV: rabies virus; NA: multi-subtype neuraminidases; EDIII: domain III of Zika virus envelope protein.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yang, L.-X.; Li, H.; Cheng, Z.-H.; Sun, H.-Y.; Huang, J.-P.; Li, Z.-P.; Li, X.-X.; Hu, Z.-G.; Wang, J. The Application of Non-Coding RNAs as Biomarkers, Therapies, and Novel Vaccines in Diseases. Int. J. Mol. Sci. 2025, 26, 3055. https://doi.org/10.3390/ijms26073055

AMA Style

Yang L-X, Li H, Cheng Z-H, Sun H-Y, Huang J-P, Li Z-P, Li X-X, Hu Z-G, Wang J. The Application of Non-Coding RNAs as Biomarkers, Therapies, and Novel Vaccines in Diseases. International Journal of Molecular Sciences. 2025; 26(7):3055. https://doi.org/10.3390/ijms26073055

Chicago/Turabian Style

Yang, Lu-Xuan, Hui Li, Zhi-Hui Cheng, He-Yue Sun, Jie-Ping Huang, Zhi-Peng Li, Xin-Xin Li, Zhi-Gang Hu, and Jian Wang. 2025. "The Application of Non-Coding RNAs as Biomarkers, Therapies, and Novel Vaccines in Diseases" International Journal of Molecular Sciences 26, no. 7: 3055. https://doi.org/10.3390/ijms26073055

APA Style

Yang, L.-X., Li, H., Cheng, Z.-H., Sun, H.-Y., Huang, J.-P., Li, Z.-P., Li, X.-X., Hu, Z.-G., & Wang, J. (2025). The Application of Non-Coding RNAs as Biomarkers, Therapies, and Novel Vaccines in Diseases. International Journal of Molecular Sciences, 26(7), 3055. https://doi.org/10.3390/ijms26073055

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop