Open access peer-reviewed chapter - ONLINE FIRST

Non-Coding RNAs Function as Diagnostic Biomarkers and Therapeutic Targets in Pulmonary Arterial Hypertension

Written By

Yingqi Wang, Daling Zhu and Hang Yu

Submitted: 25 February 2024 Reviewed: 03 March 2024 Published: 29 April 2024

DOI: 10.5772/intechopen.1005186

Unravelling Molecular Docking - From Theory to Practice IntechOpen
Unravelling Molecular Docking - From Theory to Practice Edited by Črtomir Podlipnik

From the Edited Volume

Unravelling Molecular Docking - From Theory to Practice [Working Title]

Dr. Črtomir Podlipnik

Chapter metrics overview

22 Chapter Downloads

View Full Metrics

Abstract

Pulmonary arterial hypertension is a disease characterized by complex pathogenesis and high mortality rates following diagnosis. Non-coding RNA plays a pivotal role in the development of pulmonary arterial hypertension, offering promising prospects as a diagnostic and therapeutic target for this condition. The utilization of nucleic acid drugs in disease treatment suggests the feasibility of packaging non-coding RNA into carrier systems and employing them in human pulmonary arterial hypertension (PAH) treatment through appropriate delivery routes. However, currently, no nucleic acid drugs are available for the clinical treatment of PAH. Identifying active regions within non-coding RNA through molecular docking analysis and developing suitable nucleic acid drugs hold great potential for advancing the field of PAH therapeutics.

Keywords

  • non-coding RNA
  • delivery
  • molecular docking
  • pulmonary arterial hypertension
  • clinical application

1. Introduction

1.1 Non-coding RNAs and pulmonary arterial hypertension

Pulmonary arterial hypertension (PAH) is a grave ailment characterized by the elevation in pulmonary artery pressure (mean pulmonary artery pressure ≥ 25 mmHg at rest) [1, 2], which results from the changes in the structure and function of the vessel wall due to aberrant proliferation, apoptosis, and migration of cells [3]. The causes of PAH can be broadly classified into primary and secondary causes. Currently, the pathogenesis of PAH is known to involve ion channels, vasoactive substances, immune factors, and genetic factors [4, 5, 6].

Recently, a growing body of evidence has highlighted the crucial regulatory roles played by non-coding RNAs (ncRNAs) in PAH development. It is worth noting that the majority of human genes (>95%) produce ncRNAs. MicroRNAs (miRNAs), which consist of 21–22 nucleotides, exert post-transcriptional control over gene expression [7]. Long non-coding RNAs (lncRNAs) are transcribed from intergenic or intragenic regions and typically exceed 200 nucleotides in length (Figure 1). They possess the ability to interact with proteins, RNA molecules, or DNA sequences to fulfill their regulatory functions [9]. Circular RNAs (circRNAs) represent a novel class of ncRNAs characterized by their closed-loop structure, which confers exceptional stability and enables interactions with both proteins and RNA molecules [10].

Figure 1.

RNA classification [8]. According to the size of non-coding RNAs (ncRNAs), they can be divided into >200 nucleotides (nt) long non-coding RNAs (lncRNAs) and small non-coding RNAs (sncRNAs) which are 18–200 nt. lncRNAs mainly include linear RNA and circular RNA. sncRNAs include small interfering RNA (siRNA), microRNA (miRNA), tRNA-derived small RNA (tsRNA), small nuclear RNA (snRNA), small nucleolar RNA (snoRNA), PIWI-interacting RNA (piRNA) and so on.

NcRNAs have been identified as regulators of multiple stages in gene expression. However, due to their vast quantity and diverse mechanisms, comprehensively understanding the roles of ncRNAs is challenging. NcRNAs often exhibit cell specificity in expression level and function, which are used for various disease therapy including PAH. A successful ncRNA-based strategy is based on a comprehensive understanding of the regulatory relationships involving ncRNAs [11].

1.2 Patential applications of non-coding RNAs

1.2.1 As therapeutic targets

With ever-increasing advancements in RNA structure determination methods and the ever-expanding discoveries of novel RNA structures and cellular functions, RNA-based therapeutics are emerging as promising approaches for treating human diseases. In general, RNA-based therapeutics are categorized into two types: (1) therapeutic RNAs: RNAs target RNA, DNA, or protein to modulate their function, such as RNA aptamers, antisense oligonucleotides (ASO), small interfering RNAs (siRNA), and guide RNAs (gRNA). Therapeutic RNAs are currently undergoing active development in the field of gene therapy [12, 13, 14, 15]. (2) Targeted RNAs: RNAs serve as a target for binding with drug-like small molecules [15, 16], which bears resemblance to drug discovery targeting proteins.

However, only approximately 1.5% of the human genome is responsible for protein coding [17, 18, 19, 20, 21]. Among these protein-encoding genes, only a small fraction of about 10–15% are associated with diseases [18, 19, 20, 22, 23, 24]. Therefore, RNAs exhibit a much broader druggability than proteins. NcRNAs play crucial roles in various human diseases. Targeting ncRNAs will provide huge opportunities for drug discovery.

Gene therapy is a complex approach to treating diseases, which was first proposed by Theodore Friedmann and Richard Roblin in 1972 in “Science”. RNAs, as a new class of treatments, may be an alternative to DNA therapy in preventing diseases. RNAs function in the cellular cytosol, avoiding the potential risks of insertion into patient genomes. RNAs have demonstrated therapeutic potential primarily through siRNA and microRNA (miRNA) applications, as evidenced by previous research efforts [25, 26]. In recent developments, mRNA vaccines have emerged as a powerful tool in combating infectious diseases such as COVID-19 [27, 28, 29].

NcRNAs have the potential to be PAH therapeutic targets. MiR-140-5p was found to be downregulated in PAH model rats; miR-140-5p mimics prevented the progression of established PAH in rats and suppressed the proliferation and migration of pulmonary artery smooth muscle cells (PASMCs) [30]. The results are promising. However, the clinical application of ncRNA therapy is currently constrained by several factors, including the diverse biological functions of ncRNAs and challenges in formulation and delivery approaches. As a therapeutic agent, ncRNAs may elicit adverse effects that could pose life-threatening risks or result in inadequate deposition within the deep lung tissues. Furthermore, large-scale, comprehensive, and well-designed clinical studies are indispensable for shedding more light on the potential applications of ncRNAs.

1.2.2 As biomarkers

NcRNAs have emerged as potential non-invasive and more sensitive diagnostic biomarkers for PAH. In the early diagnosis of PAH, hsa-miR-21-3p and hsa-miR-143-3p in peripheral plasma can be considered as potential biomarkers [31].

1.3 Status of pulmonary non-coding RNAs delivery

RNA therapeutics have been hailed as the medicine of the future, and this vision has now become our reality [32]. Currently, there are two mRNA vaccines available in the market, along with four siRNA-based therapeutics and five ASO-based drugs [33]. This signifies a true revolution in the field of RNA therapeutics; however, one major obstacle remains delivery [34]. Nanocarriers that are optimized for protecting and delivering payloads of RNA tend to accumulate primarily in the liver when administered intravenously [35]. To overcome this limitation and explore alternative administration routes beyond liver targeting, numerous clinical trials have investigated different approaches for RNA delivery [36]. Intravitreal injection (fomivirsen) and intrathecal injection (nusinersen) have demonstrated success, prompting further exploration of local administration routes such as intranasal or pulmonary delivery through ongoing clinical trials. However, no approved products utilizing these routes have emerged thus far [37]. The lung presents an array of currently untargetable sites that hold potential for treatment using ncRNA therapeutics.

In this chapter, we conducted a comprehensive literature review to identify functional ncRNAs, particularly miRNAs, circRNAs, and lncRNAs associated with PAH. Additionally, we identified potential untapped biomarkers and therapeutic targets for ncRNAs in PAH.

Advertisement

2. MicroRNAs

2.1 Introduction

MicroRNAs (miRNAs) are a class of ncRNAs, approximately 20–24 nucleotides in length, that possess regulatory functions. MiRNA genes are transcribed by RNA polymerase II or III into primary miRNA transcripts (pri-miRNA). These pri-miRNAs are subsequently cleaved by the Drosha-DGCR8 (DiGeorge syndrome critical region 8) complex [38], an RNase III enzyme. The resulting precursor miRNAs (pre-miRNAs) are then transported to the cytoplasm and processed into double-stranded mature miRNAs by the RNase Dicer [39]. This double-stranded form is rapidly loaded onto the RNA-induced silencing complex (RISC), where one of the single strands undergoes degradation while the other mature single-stranded miRNA molecule either degrades or translates its target gene through complementary pairing with the 3′UTR region of said target gene [40].

The stability of miRNAs is remarkably high in both DNA and RNA lysates, making them resistant to degradation. MiRNAs exhibit conserved sequences and functions across diverse cells, tissues, organs, and genera. The distinct expression profiles of miRNAs in various tissues and cells render them valuable molecular markers for specific cellular populations. Importantly, the presence of a particular miRNA at a specific stage within a given cell determines its differentiation direction and phase. Consequently, miRNAs play crucial roles as pivotal regulators governing cell timing and directional differentiation.

Numerous studies have demonstrated that miRNAs exert their biological function by participating in the regulation of downstream gene translation processes, with target mRNAs located within its 3′UTR [41]. In animals, a single miRNA can recognize multiple mRNA targets, and conversely, one mRNA target can be recognized by multiple miRNAs. It is estimated that approximately two-thirds of protein-coding genes in the human genome are regulated by miRNAs.

2.2 MiRNAs and PAH

It is widely acknowledged that miRNA dysfunction contributes to the enhanced proliferation of endothelial cells, smooth muscle cells, and outer epidermal cells in PAH. Increasing evidence suggests that various miRNAs and their expression changes (either upregulation or downregulation) are associated with PAH pathogenesis. MiRNAs regulate the proliferation and apoptosis of pulmonary artery endothelial cells (PAECs), PASMCs, and extrapulmonary fibroblasts through their target genes. They play roles in all aspects of pulmonary vasoconstriction and reconstruction. However, due to a large number of publications based on diverse experimental methods, it remains challenging to identify the most crucial miRNAs as potential therapeutic targets for PAH [42].

The level of miR-130/301 has been shown to be elevated in both pulmonary vessels and plasma in mammalian models as well as human patients with PAH. MiR-130/301 promotes PAH-related phenotypes through the apelin-miR-424/503-FGF2 signaling pathway in endothelial cells, while it utilizes the STAT3/miR-204 signaling pathway in smooth muscle cells. Administration of miR-130a oligonucleotide mimics enhances pathogenic effects related to PAH, whereas inhibition of miR-130/301 prevents the onset of PAH in mice treated with SU5416 under hypoxic conditions [43].

The miRNA-17/92 cluster exerts an impact on the proliferation of PAECs and PASMCs. The combination of miR-17/18a/19a/92a demonstrates the most potent induction effect on PCNA and α-SMA, while maintaining calponin and SM22α levels. Consequently, in SM-17 ~ 92−/− mice, miR-17 to 92 was reconstituted by administering a mixture of miR-17/18A/19a/92a mimics via intravenous injection through the tail vein. MiR-17 to 92 reconstitution restored elevated right ventricular systolic blood pressure (RVSP), RV/(LV + S) ratios, and pulmonary artery remodeling in hypoxia-exposed SM-17 ~ 92−/− mice without altering basal levels of RVSP, RV/(LV + S), or pulmonary artery wall thickness. These findings suggest that miR-17 ~ 92 plays a crucial role in the pathogenesis of PAH. By investigating PASMCs and lung tissue from PAH patients, it has been demonstrated that miRNA-17/92 promotes PASMC proliferation through the PDLIM5/TGF-beta/Smad pathway, offering novel insights for PAH treatment [44].

The expression of miR-495 is significantly upregulated in the mouse model of PAH induced by chronic hypoxia and hypoxia-Su5416 (HySu). Inhibition of miR-495 via adeno-associated virus type 9 (AAV9) effectively mitigates hemodynamic abnormalities and structural changes in the pulmonary vasculature in the mouse model of PAH, thereby improving vascular remodeling and angiogenesis. This represents a novel potential therapeutic approach for human pulmonary hypertension [45].

The serum level of miR-30a was found to be elevated in patients with pulmonary hypertension compared to the healthy control group. MiR-30a exhibits predominant expression in the pulmonary arteriole (PA). In animal models of HySu-induced and MCT-induced PAH, genetic knockout of the miR-30a gene effectively reduces right ventricular systolic blood pressure (RVSP), as well as attenuates remodeling of the pulmonary arteriole and hypertrophy of the right ventricle. Furthermore, pharmacological inhibition of miR-30a through intratracheal fluid perfusion (IT-L) administration strategy demonstrates high efficacy [46].

Our previous studies have demonstrated that circ-calm4 promotes pyroptosis in hypoxic PASMCs through the miR-124-3p/PDCD6 pathway [47]. Additionally, this circular RNA also enhances PASMC proliferation via the miR-337-3p/Myo10 pathway [48]. These findings suggest that circ-calm4 may represent a promising therapeutic target for treating pulmonary hypertension.

2.3 MiRNAs in clinical research

In terms of clinical diagnosis, accessing certain tissues such as the cardiac tissue can be challenging. However, miRNAs have emerged as potential biomarkers for disease states and prognostic markers in various diseases. An analysis conducted on plasma samples from 64 untreated patients with pulmonary arterial hypertension, along with 43 patients and healthy controls, demonstrated that miR-636 and miR-187-5p exhibit high diagnostic accuracy in predicting pulmonary hypertension [49]. The findings of these studies indicate that miRNAs have the potential to serve as biomarkers for the diagnosis of PAH.

A significant number of clinical trials involving ncRNAs primarily focus on miRNAs, with over 800 trials conducted, a substantial proportion of which are specifically targeted toward cancer research [50]. Growing evidence suggests that miRNAs play crucial roles in clinical diagnosis and therapy (Figure 2). The first miRNA to transition from basic research to clinical application is miR-122, which has been utilized for the treatment of hepatitis C virus (HCV) [52]. MRX34 represents the pioneering miRNA-based approach for cancer treatment, where it is encapsulated within lipid nanoparticles and administered via intravenous injection into the human body [53]. Furthermore, a nanoparticle formulation named TTX-MC138 has been developed to target anti-miR-10b for breast cancer treatment [54], while an ointment incorporating ionic liquid technology and a mimic of ds-miR-634 has been formulated for skin squamous cell carcinoma (cSCC) therapy [55].

Figure 2.

Several miRNA-related cancer therapy resistance [51]. There are four categories along with some regulators presented in the text, where the rectangle in green or in red shows whether the function of miRNAs in the mentioned cancer type is positive or negative for the corresponding therapy. ERα36: estrogen receptor alpha 36; CYP: cytochrome P450; ABCG2: ATP-binding cassette sub-family G member 2; MDR1: multidrug resistance 1; STAT3: signal transducer and transcriptional activating factor 3; MYC: myelocytomatosis oncogene; HnRNPA1: heterogeneous nuclear ribonucleoprotein A1; PKM2: the pyruvate kinase muscle 2; mTOR: mammalian target of rapamycin; RPA: replication protein A; PTEN: phosphatase and tensin homologous protein; ERCC1: excision repair cross-complementing 1.

The dysregulation of miRNA expression in exosomes has been demonstrated to be a potential therapeutic target for PAH, with the upregulation of miR-21 observed compared to the matched control group [56]. However, there is currently a lack of clinical studies on the therapeutic application of miRNAs in pulmonary hypertension. The future holds immense potential for nucleic acid therapy of pulmonary hypertension, as the combination of meticulous selection of miRNAs from fundamental research and efficient delivery routes will pave the way for groundbreaking advancements in this field.

Advertisement

3. Circular RNAs

3.1 Introduction

Circular RNAs (circRNAs) are a newly discovered class of ncRNAs that lack a 5′ terminal cap and a 3′ terminal poly(A) tail, instead forming a circular structure through covalent bonds [57, 58]. This unique circular conformation distinguishes circRNAs from conventional linear RNAs.

The categorization of circRNAs is based on their origin, which can be classified into three categories: exonic circRNAs derived from exons, circular intronic RNAs derived from introns, and retained-intron circRNAs composed of both exons and introns [59].

CircRNAs possess diverse biological capabilities, including the regulation of target gene expression and modulation of target gene activity through competitive binding with corresponding miRNAs, thereby influencing disease occurrence and progression. These circRNAs are commonly referred to as competitive endogenous RNAs (ceRNAs), acting as molecular “sponges” [60]. Furthermore, circRNAs interact with RNA-binding proteins [61], regulate mRNA stability [62], and serve as protein molecule sponges, with some circRNAs containing binding sites for one or more RNA-binding proteins [63]. Despite being classified as ncRNAs, a subset of circRNAs can encode peptides that exert regulatory functions through peptide encoding [64]. The findings of circRNAs pave the way for future investigations into circRNA biology, offering a novel perspective on our comprehension of gene expression regulation and disease pathogenesis.

3.2 CircRNAs and PAH

CircRNAs exhibit aberrant expression patterns in various diseases, including cancer, neurological disorders, and immune diseases [65, 66, 67, 68]. Additionally, they play a pivotal role in the pathogenesis of pulmonary hypertension.

CircRNAs exhibit aberrant expression in patients with PAH compared to healthy controls, and are involved in the processes of cell proliferation, angiogenesis, inflammation, and fibrosis in PAH patients. This suggests their potential role in disease progression [69, 70, 71]. The circHIPK3/miR-328-3p/STAT3 axis contributes to the pathogenesis of PAH by promoting proliferation, migration, and angiogenesis of human PAECs. The accelerated involvement of circHIPK3 indicates its potential value as a therapeutic target for treating PAH [72]. Hsa_circNFXL1_009 directly acts as a sponge for hsa-miR-29b-2-5p (miR-29b), positively regulating the expression of voltage-gated potassium (K+) channel subfamily B member 1 (KCNB1) at the mRNA level. Delivery of exogenous hsa_circNFXL1_009 attenuates hypoxia-induced proliferation, resistance to apoptosis, and migration of human PASMCs, suggesting its potential use as a therapeutic target for treating PAH [73].

3.3 CircRNAs in clinical research

CircRNAs have emerged as significant markers and targets for the diagnosis and treatment of numerous diseases owing to their distinctive structure, which effectively shields them from exonuclease degradation, rendering them relatively stable in terms of drug efficacy and biological activity (Figure 3).

Figure 3.

Clinical applications of circRNAs and exosomal circRNAs in tumors, including circRNA extraction, detection, as clinical markers and therapeutic strategies [74, 75].

The abundant presence and remarkable stability of circRNAs in bodily fluids render them an optimal non-invasive biomarker. The identification of circRNAs in serum or tissues holds the potential for early screening, disease monitoring, and prognosis evaluation.

It has been suggested that serum exosomal circCCDC66 secreted and released by pituitary adenoma cells can serve as a potential prognostic biomarker for monitoring tumor dynamics during treatment [76], which highlights the utility of circRNAs as biomarkers for early disease screening, disease monitoring and prognosis evaluation. Researchers have investigated the distinctive expression pattern of hsa_circ_0003416 in the plasma of pediatric patients with PAH resulting from congenital heart disease (CHD). It was observed that hsa_circ_0003416 exhibited a significant downregulation in children affected by PAH-CHD and demonstrated its potential as a biomarker for diagnosing PAH-CHD [77]. This finding implies that circRNAs could serve as valuable diagnostic markers for PAH in clinical settings.

CircRNAs have been extensively investigated in various diseases. For instance, researchers have observed that Steatohepatitis-associated circRNA ATP5B Regulator (SCAR), localized in mitochondria, exerts inhibitory effects on mitochondrial ROS (mROS) production and fibroblast activation. To explore the therapeutic potential of circRNA SCAR in vivo, they intravenously administered mito-NP containing circRNA SCAR, resulting in a significant increase of its hepatic levels in mice and attenuation of insulin resistance induced by high-fat diet-induced cirrhosis [78]. Additionally, it has been demonstrated that circNR3C2 negatively regulates triple-negative breast cancer metastasis through the circNR3C2/miR-513a-3p/HRD1/Vimentin axis, and this finding was validated using mouse models. Notably, circNR3C2 exhibits inhibitory effects on TNBC metastasis [79]. Collectively, these studies highlight the promising therapeutic potential of targeting circRNAs for clinical applications.

Our previous study discovered that the circ-calm4/miR-337-3p/Myo10 signaling pathway regulates the proliferation of PASMCs at the molecular level and validated this finding in mice. Administration of serotype 5 adenovirus-associated virus (AAV5) through nasal feeding revealed that silencing circ-calm4 alleviated symptoms associated with pulmonary hypertension [48]. Lihuang Su et al. reported that circ-Ntrk2 acts as a sponge for miR-296-5p and miR-296-5p, which bind to the 3′-untranslated region of transforming growth factor beta 1 (TGF β1) mRNA, thereby attenuating TGF β1 translation. Additionally, knocking down circ-Ntrk2 in mice resulted in improved symptoms of PAH [80]. These studies suggest that circRNAs hold potential as therapeutic targets for treating pulmonary hypertension.

Despite the potential of circRNAs in clinical treatment, there are still several challenges that need to be addressed. Antisense technology can selectively inhibit or degrade oncogenic circRNAs, but most loss-of-function studies have only been conducted on preclinical animal models using RNAi. Currently, no circRNA-targeted treatments have reached clinical trials yet [81]. However, delivery of circRNA expression cassettes can be achieved through encapsulation within lipid and/or polymer nanoparticles that can even target specific organelles like mitochondria [82]. Synthetic approaches have also been developed for producing and delivering preformed circRNAs to target cells using non-viral nanoparticles [83]. Additionally, researchers have synthesized artificial circRNA sponges called “circmiRs” in vitro to therapeutically target miRNAs [84, 85, 86, 87], while other studies show promising results in using circRNAs as vaccines for COVID-19 [88] and melanoma [89]. These findings provide valuable insights into the future application of circRNAs in treating PAH.

In summary, the current research on circRNA clinical application is still in its early stages. Its future prospects for development in scientific research and clinical investigations are extensive. Studies are required to validate its efficacy and reliability in clinical diagnosis. There is an urgent necessity to develop and enhance advanced methodologies for the design, synthesis, purification, delivery, and therapeutic applications of in vitro artificial circRNA vaccines or drugs. In the future, researchers need to strengthen collaboration, data acquisition, and resource utilization while promoting the translation of circRNA research findings into clinical practice to achieve improved treatment outcomes and enhanced quality of life for patients.

Advertisement

4. Long non-coding RNAs

4.1 Introduction

Long non-coding RNAs (lncRNAs) are characterized by their extensive nucleotide sequences, exceeding 200 nucleotides in length, making them a diverse and intriguing group of RNA molecules. LncRNAs play crucial roles in regulating various cellular processes [90].

The biogenesis of lncRNAs follows mechanisms similar to mRNA, involving transcription by RNA polymerase II, 5′ capping, splicing, and 3′ polyadenylation [91]. However, unlike mRNAs, lncRNAs rarely encode proteins and instead exert their functions through a myriad of intricate molecular mechanisms, including acting as molecular scaffolds [74, 75], guiding the assembly of protein complexes [92], serving as decoys by sequestering regulatory proteins [93], and functioning as enhancer-associated transcripts to modulate gene expression over long genomic distances [94].

One notable characteristic of lncRNAs is their specific expression patterns in different tissues and developmental stages [95]. These dynamic expression profiles reflect their involvement in essential biological processes, including embryonic development, cellular differentiation, and tissue homeostasis maintenance. The dysregulation of lncRNAs has increasingly been associated with various human diseases, highlighting their potential as diagnostic markers and therapeutic targets.

4.2 LncRNAs and PAH

LncRNAs play significant roles in cardiovascular disorders by influencing vascular function, cardiac development, and fibrosis [96, 97, 98, 99, 100]. An increasing body of evidence suggests that lncRNAs serve as crucial epigenetic modifiers in pulmonary hypertension. The dysregulation of lncRNAs may impact processes such as vascular remodeling [101], inflammation [102], and endothelial dysfunction [103], thereby contributing to the narrowing of pulmonary vessels observed in PAH [104].

A research from 11 control individuals and 12 pulmonary arterial hypertension patients suggests that JPX modulates RABEP1 (Rab GTPase-binding effector protein expression through miRNAs (miR-145, miR-146 ac, miR-216a, miR-216b, miR-24, miR-33ab, miR-129-5p), thereby impacting CD8+ T cells, NK cells, eosinophils, and other immune cells, ultimately influencing the immune processes associated with PAH. The finding reveals that lncRNA-mRNA pair (JPX-RABEP1) can serve as a potential biomarker in PAH [105]. The rs619586A > G polymorphism in MALAT1 plays a protective role in susceptibility to PAH within the Chinese Han population. This polymorphism has the potential to serve as a novel biomarker for PAH susceptibility and implicates lncRNAs in the etiology of PAH [106]. A study from 41 blood samples, SNHG12 is identified as a competing endogenous RNA (ceRNA) of CCR7 by sequestering hsa-let-7e-5p, which suggests the potential of CCR7, hsa-let-7e-5p, and SNHG12 as candidate biomarkers for PAH.

Although lncRNAs have been reported as potential biomarkers in exosomes or body fluids in diseases such as cancer, they are still in the developmental stage in the field of pulmonary arterial hypertension, but these studies may shed light on the discovery of biomarkers in pulmonary hypertension. Identification of lncRNAs as potential biomarkers holds promise for disease diagnosis, prognosis, and monitoring. The development of non-invasive diagnostic tools based on lncRNA expression profiles could significantly impact early disease detection and patient management.

A study from our team proves that m6A-induced degradation of FENDRR promotes pyroptosis in human pulmonary artery endothelial cells (HPAECs) by regulating the methylation of the DRP1 promoter, providing a potential novel target for hypoxia-induced pulmonary hypertension (HPAH) therapy. The overexpression of the conserved fragment TFO2 of FENDRR prevents HPAH in vivo [107]. The lncRNA PAXIP1-AS1 facilitated the proliferation and migration of human PASMCs through the ETS1/WIPF1/RhoA pathway. Inhibition of PAXIP1-AS1 via tail vein injection of lentiviral plasmid expressing stable short hairpin RNAs (sh-RNAs) specifically targeting PAXIP1-AS1 significantly attenuated the elevated right ventricular systolic pressure (RVSP) in MCT-induced pulmonary hypertension mice. Therefore, targeting lncRNA PAXIP1-AS1 may hold promise as a potential therapeutic strategy for PAH treatment [108]. The expression of lncRNA H19 is significantly elevated in rodent lungs induced by MCT and is upregulated by PDGF-BB. The H19/let-7b/AT-1R axis contributes to the pathogenesis of PAH by promoting the proliferation of pulmonary artery smooth muscle cells. Knockout of H19 plays a protective role in the development of PAH, suggesting that targeting H19 may hold potential for PAH treatment [102].

4.3 LncRNAs in clinical research

RNAs have demonstrated therapeutic potential primarily through small interfering RNA (siRNA) and microRNA (miRNA) applications [25, 26]. LncRNAs have gained significant attention in clinical research due to their diverse roles in various physiological and pathological processes. Advances in understanding the roles of lncRNAs may pave the way for developing novel therapeutic strategies. Targeting disease-associated lncRNAs could offer new avenues for precision medicine, especially in the treatment of pulmonary hypertension.

Given that lncRNA falls in size between siRNA and mRNA, its delivery vehicle may not differ significantly from those already employed in clinical settings for siRNA and mRNA delivery [109]. Therefore, leveraging existing delivery platforms could facilitate the translation of lncRNA-based therapeutics into clinical practice. However, the intricate secondary structure of lncRNAs poses challenges for their exogenous delivery, hindering the development of lncRNA-based drugs. As a result, instead of directly transferring lncRNAs, many lncRNA-based drugs opt to target lncRNAs to modulate their expression. The next is an exploration of diverse delivery systems utilized for RNA therapeutics, with some having undergone testing for lncRNA and nucleic acid delivery targeting lncRNAs.

Numerous studies have explored the potential of lncRNAs as diagnostic and prognostic biomarkers in various cancers, such as LINC01535 in breast cancer [110], MLETA1 in lung cancer [111], and HCG18 in cholangiocarcinoma [112]. LncRNAs ANRIL and HOTAIR can be used as biomarkers for cardiovascular disease risk prediction [113]. LncRNA PCA3 is highly expressed in prostate cancer and is considered useful for prostate cancer diagnosis [112]. However, lncRNAs as PAH biomarkers are still being researched.

Advertisement

5. PIWI-interacting RNA

5.1 Introduction

PiRNAs (PIWI-interacting RNA) are a class of small non-coding RNA molecules, typically ranging from 24 to 32 nucleotides in length [114]. PiRNAs exhibit highly conserved and diverse sequences. Unlike other types of small RNAs such as miRNAs and siRNAs, piRNAs are primarily generated through the interaction between transposon factors and the genome [115]. PiRNAs are mainly expressed in the reproductive system. They form complexes with PIWI proteins to form piRNA-induced silencing complexes (piRISCs) that play crucial roles in protecting genome stability during transposition, suppressing transposon activity, and regulating gene expression and epigenetic modifications [116]. The initial belief that piRNAs are exclusively involved in germ cell development has been challenged by recent research, which has demonstrated their expression in various tissues and cell types. Furthermore, these findings highlight the involvement of piRNAs in maintaining immune homeostasis and underscore their significance in diseases such as cancer [117].

5.2 PiRNAs and PAH

The involvement of piRNAs in crucial processes such as cardiovascular remodeling, inflammatory response, and cell proliferation is closely associated with the pathophysiological mechanisms underlying pulmonary arterial hypertension. PiRNA-63076 is upregulated in hypoxic PASMCs and the lung tissues of hypoxia-induced mice, which promotes the proliferation of PASMCs by regulating the interaction of acyl-CoA dehydrogenase (Acadm) with Piwi protein. PiRNA-63076 partially inhibits the expression of Acadm by mediating DNA methylation in the promoter region of Acadm, providing a new perspective on understanding the pathophysiological process of pulmonary arterial hypertension [118]. Some piRNAs from the extracellular vesicles of PAH patients exhibited different expression patterns correlated with the severity of PAH, indicating a potential role for piRNAs in the development and progression of PAH [119].

5.3 PiRNAs in clinical research

The expression of piRNAs demonstrates disease-specificity, rendering them potential diagnostic biomarkers. Researchers have already employed piRNAs for early tumor detection and prediction of patient prognosis, showcasing promising clinical applicability [120]. Assessing piRNAs levels in blood, urine, or other bodily fluids may be feasible to aid in the early diagnosis, classification, and prognostic evaluation of diseases.

PiRNAs also play a role in the treatment of diseases. PiRNA-54265 is upregulated in colorectal cancer. AntagopiR54265, the inhibitor of piRNA-54265, significantly suppressed the growth and metastasis of implanted tumors [121]. By targeting piRNAs as therapeutic targets, the regulation of relevant gene expression can be achieved through interference with specific interactions between piRNAs and Piwi proteins. Through the design and synthesis of specific piRNA ligands, it is possible to precisely modulate gene expression, signal pathways, and cellular processes, thereby intervening in disease development.

The research on PiRNAs is still in its nascent stage. The clinical application of piRNAs is still in the research stages, requiring further experiments and clinical studies to validate its accuracy, feasibility, and safety. PiRNAs hold promise as diagnostic biomarkers for various diseases, a powerful tool for targeted therapy, and a strategic approach toward cell therapy and regenerative medicine [120].

Advertisement

6. Small nucleolar RNA

6.1 Introduction

SnoRNAs (small nucleolar RNA) are a class of small non-coding RNA molecules that mainly exist in the nucleolus. They play regulatory roles in the nucleolus and are involved in the modification and maturation processes of ribosomal RNAs (rRNAs) [122]. SnoRNAs are typically composed of 60–300 nucleotides and are divided into two main categories: C/D box snoRNAs and H/ACA box snoRNAs. C/D box snoRNAs mainly form nucleotide methyltransferase complexes with proteins to carry out 2′-O-methylation modifications at specific positions in rRNAs. H/ACA box snoRNAs form complexes with proteins to maintain RNA stability and localization and guide post-transcriptional modifications in rRNAs [123].

SnoRNAs are involved in various cellular processes, such as proliferation, transcription, and RNA splicing, which are associated with tumor development and metastasis [124]. The abnormal expression or dysfunction of snoRNAs may play significant roles in the development of diseases, including cardiovascular diseases, congenital disorders, and genetic diseases [125].

6.2 SnoRNAs and PAH

A study from different types of pulmonary arterial hypertension patients (including idiopathic pulmonary arterial hypertension (IPAH), connective tissue disease-associated pulmonary arterial hypertension (CTD-PAH), and congenital heart disease-associated pulmonary arterial hypertension (CHD-PAH)) shows that there are 30 differentially expressed snoRNAs in lung tissues [126]. SNORD20, SCARNA4, and SNORD94 exhibit consistent differential expression in all three PAH, suggesting the three snoRNAs play important roles in the pathophysiological processes of PAH [126].

The relationship between snoRNAs and diseases still encompasses numerous unexplored areas. Further investigations are imperative to accumulate additional evidence elucidating the association between snoRNAs and diseases, thereby offering novel insights and approaches for disease prevention and therapy.

Advertisement

7. tRNA-derived small RNA

7.1 Introduction

TsRNA (tRNA-derived small RNA) refers to a class of small RNAs generated from tRNAs. TRNAs are a common type of RNA that primarily transports amino acids to the ribosome during protein synthesis. However, recent research has discovered that tRNAs participate in other cellular processes through various mechanisms and give rise to various tsRNAs [127]. The generation of tsRNAs mainly involves two mechanisms: tRNAs cleavage and modification and the generation of cleavage products through non-canonical functions of tRNAs. TsRNAs have diverse biological functions, including regulation of gene expression, translational control, and impact on cellular processes such as growth, division, and apoptosis [128]. Abnormal expression of tsRNAs may be associated with the occurrence and development of tumors. For example, tsRNAs are significantly increased or decreased in cancer cells, which may affect the expression of tumor suppressor genes and promote the growth and spread of tumor cells [129].

7.2 tsRNAs and PAH

An increasing number of studies are investigating the association of tsRNAs with cardiovascular diseases, such as pulmonary hypertension [32, 97, 99]. Altered expression of tsRNAs in the blood and lung tissue of PAH patients has been found. A study conducted a small RNA microarray analysis of tsRNAs in the plasma of pulmonary arterial hypertension patients and a healthy control group identified a total of 816 tsRNAs significantly regulated in PAH, with 243 upregulated and 573 downregulated [130]. Gene Ontology analysis showed that these tsRNAs might be involved in cellular metabolic conditions and other processes, suggesting that tsRNAs could potentially serve as novel biomarkers and therapeutic targets in idiopathic PAH [130].

Advertisement

8. Delivery tools for RNAs

Gene therapy is extensively utilized for the treatment of incurable disorders and has become a routine procedure in clinical practice. The main challenge in the development and application of RNA drugs lies in their delivery technology, as opposed to small molecule and protein drugs. Unlike these counterparts, RNA molecules are negatively charged, sensitive to ubiquitous RNases, and primarily act intracellularly. Therefore, the advancement of delivery systems is crucial for addressing existing issues in vivo RNA delivery and enhancing the efficacy of RNA drugs (Figure 4).

Figure 4.

Schematic overview of administration routes and features of miRNA therapeutics [131]. (A) miRNA therapeutics can be applied by systemic administration (e.g., by venous injection) or by cell−/tissue-directed administration, extracorporeal miRNA approaches, adoptive cell transfer, or implantation of 3D matrices releasing miRNA therapeutics. (B) Nucleic acid-based miRNA therapeutics show a low cell membrane permeability. Efficient intracellular delivery can be achieved by nano-sized carrier vehicles, viral transduction, introduction of chemical modifications, or conjugation to biomolecules that further a receptor-mediated uptake. Abbreviations: GalNAc, N-acetylgalactosamine; miRNA, microRNA.

8.1 Viral vector for RNA delivery

Viral vectors have emerged as indispensable components of gene therapy due to their inherent ability to infect mammalian cells and effectively deliver genetic material [132]. Adeno-associated virus (AAV), adenovirus (AdVs), and lentiviral vectors (LVV) are increasingly being recognized as fundamental therapeutic delivery systems owing to their usability and safety profile [133].

However, the utilization of viral vectors can give rise to two types of adverse effects: a response triggered by the virus’s entry or a reaction induced by the delivered genetic material. Any viral-based gene therapy approach necessitates thoroughly evaluating potential side effects and safety concerns.

AAV is a small, non-enveloped virus requiring helper viruses such as AdV, herpesvirus, and some others for replication. AAV is a natural human symbiont, and a weak immune response makes it a suitable vector for gene therapy [134, 135, 136].

AdV, a non-enveloped virus, contains protein-coding genes belonging to Adenoviridae [137, 138]. The wide applications of AdV are due to their high levels of gene expression, transduction efficiency, and ability to deliver transgenes of up to 8 kbp in size [139, 140].

Lentivirus is an enveloped virus belonging to the family Retroviridae. The advantages of LVVs are the packaging capacity of up to 10 kb [141], low cytotoxicity, and lack of viral replication after transduction.

8.2 Non-viral vectors for RNA delivery

The limitations of viral vector delivery systems such as immunogenicity and mutagenicity restrict their clinical application and prompt the development of non-viral gene carriers using different materials to achieve successful in vivo delivery of RNA (Figure 5).

Figure 5.

A schematic representation of some nanocarriers used for miRNA delivery [142].

8.2.1 Nanoparticles

8.2.1.1 Lipid nanoparticles (LNP)

LNPs stand as the sole FDA-approved carrier for macromolecular nucleic acid drugs, facilitating enhancements in clinical trials [143, 144]. Three LNP-based RNA drugs have been approved on the market, including Alnylam’s Patisiran (ONPATTRO™), Pfizer’s BNT162b2, and Moderna’s mRNA-1273. Despite LNPs exhibiting lower transfection efficiency compared to lentiviral vectors, their advanced, safe, relatively convenient, and cost-effective construction process renders them practical alternatives (Figure 6) [145].

Figure 6.

Illustration of targeted therapy using sophisticated RNA-LNPs designed to target specific tissues and organs [97, 98, 99]. (1) LNPs expressing (a) Tat and T7 peptides, (b) NT-lipidoids, and (c) aptamer for CCR5 can be preferentially delivered into the brain. (2) spray of aerosol RNA-LNPs can be preferentially delivered into the lungs. (3) RNA-LNPs expressing GALA peptide can be preferentially delivered into the lungs. (4) RNA-LNPs expressing MAdCAM-1 D1 and D2 peptides can be preferentially delivered into the gut. (5) RNA-LNPs expressing keratinocyte targeting peptide can be extensively delivered into the skin. (6) RNA-LNPs expressing retina targeting peptide can be extensively delivered into the neural retina (photoreceptor cells) in eyes. (7) RNA-LNPs composed of 50% DOTAP (positively charged lipid) can be preferentially delivered into lung. (8) RNA-LNPs containing C12-200 can be specifically delivered into bone. (9) RNA-LNPs containing 18PA (negatively charged lipid) can be specifically delivered into the spleen. (10) Most RNA-LNPs without specific LNP design features may be preferentially delivered into the liver.

8.2.1.2 GalNAc

GalNAc is a high-affinity targeting ligand of the Asialoglycoprotein receptor (ASGPR) [146]. When GalNAc binds to ASGPR, it can enter the endosome through clathrin-mediated endocytosis. Then, ASGPR dissociates from GalNAc-siRNA conjugate and circulates back to the surface of cells. The delivery strategy based on GalNAc offers more advantages compared to LNP due to its subcutaneous administration capability and ease of scaling up. Currently, a significant number of RNA drugs in clinical trials are utilizing GalNAc technology.

8.2.1.3 Lipid-based nanoparticles (LBNPs)

Lipid-based nanoparticles (LBNPs) encompass liposomes, solid lipid nanoparticles (SLN), and nanostructured lipid carriers (NLC). These nanoparticles possess the ability to transport both hydrophobic and hydrophilic molecules, exhibit minimal or no toxicity, and prolong drug action duration [147]. The advantages of LBNPs include excellent temporal and thermal stability, high loading capacity, ease of preparation, low production costs, and suitability for large-scale industrial production due to their natural source origin. LBNPs have been extensively evaluated in vitro for cancer therapy as well as in vivo, with promising outcomes observed in certain clinical trials [148]. The incorporation of chemotherapeutic agents into lipid nanoparticles reduces the required therapeutic dosage and associated toxicity while also diminishing drug resistance. Furthermore, it enhances drug levels within tumor tissue by minimizing accumulation in healthy tissue [148].

8.2.1.4 Polymeric nanoparticles

Polymeric nanoparticles (NPs) are particles ranging in size from 1 to 1000 nm and can be loaded with active compounds entrapped within or surface-adsorbed onto the polymeric core. Polymeric NPs have garnered significant interest due to their unique properties resulting from their small size [149, 150, 151]. As drug carriers, polymeric NPs can control release, protect drugs and other biologically active molecules against environmental factors, as well as improve bioavailability and therapeutic index [151, 152].

8.2.1.5 Aptamer

Aptamer is a single-strand oligonucleotide that can bind specifically to the target molecule, which forms a specific three-dimensional structure after adaptive folding through various interaction forces such as nucleotide base complementary pairing. This binding is comparable to that of monoclonal antibodies. Compared with antibodies, aptamers have many advantages, such as a wider range of target molecules, better thermal stability, smaller molecular weight, chemical synthesis, small batch differences, easy modification, and so on. Aptamers allowed targeted delivery of RNA therapeutics through cell-specific surface markers, and once inside the cell, the nanoparticles induced lysosomal leakage that released the RNA oligonucleotides into the cytosol to achieve gene silencing [153].

8.2.1.6 Biomimetic nanomaterials

Biomimetic nanomaterials are an intersection between traditional synthetic nanomedicine and biotechnologies, offering biocompatibility, safety, and scalability of formulations with complex behaviors. The composition of biomimetic nanomaterials involves the integration of synthetic nanomaterials with simplified designs and essential biological components, which are crucial for achieving the desired effect. This approach enables enhanced biocompatibility and scalability in production [154]. Among the various biomimetic approaches investigated for enhancing the pharmacokinetics of nanoparticles (NPs), cell membranes and their constituents have emerged as a captivating opportunity [155].

8.2.2 Exosomes

Exosomes are small vesicles released by cells, typically with diameters ranging from 40 to 160 nm, which elicit minimal immune responses and possess an ability to traverse biological barriers such as cellular membranes and the blood-brain barrier [156, 157]. Exosomes contain a diverse array of biomolecules, including proteins, lipids, nucleic acids, and cell surface receptors [158, 159] and protect these molecules from degradation and facilitate intercellular communication across different tissues within the organism [82]. The challenges of utilization of exosomes as carriers for RNA delivery are how to enable large-scale production and ensure consistent quality of exosomes [160, 161]

8.2.3 Hydrogels

Macroscopic hydrogels are soft and water-swollen, which possess remarkable features such as biodegradability, tunable physiochemical properties, and injectability (Figure 7). Hydrogels have attracted enormous attention for use in RNA therapy recently. RNA can be loaded into hydrogels either by direct inclusion of the naked RNA or by encapsulating RNA nanocarriers. Hydrogels could improve RNA stability, reduce unnecessary loss of therapeutics during systemic delivery, mitigate undesirable off-target toxicities, and avoid the necessity of multiple doses [162].

Figure 7.

Advantages of hydrogels as a platform for RNA delivery [162]. Hydrogels provide a unique strategy for local administration of RNA, overcoming some of the difficulties associated with systemic RNA delivery. They enable a localized, controlled, and sustained delivery of high levels of payloads, while maintaining RNA biological activity. Off-target effects and the need for multiple payload administrations in systemic delivery may thus be avoided.

8.2.4 Protein/peptide

A protein derived from retrovirus is found to transport RNAs outside the cell in virus-like particles (VLPs). This selective endogenous encapsidation for cellular delivery enables the delivery of exogenous mRNA cargos, such as Cre and Cas9, into cells in vitro without the use of non-human components [163]. As a fully human system, it may be a safer alternative to current methods, but it is still in its infancy.

8.2.5 Virus-like particles (VLPs)

VLPs possess protein coats resembling viruses, which are capable of adapting to fluctuations in PAH and temperature. Their surface can be customized with various peptides to target distinct cell types (Figure 8) [165].

Figure 8.

Delivery of mRNA cargo by virus-like particles derived from endogenous retroelements [164]. Schematic of the “selective endogenous encapsidation for cellular delivery” (SEND) system. PEG10, cargo RNA, and fusogen vectors are transfected into cells. Inside cells, the PEG10 proteins pack the cargo mRNA and assemble into virus-like particles (VLPs) that are secreted to the growth medium in extracellular vesicles. The medium is then collected, and the VLPs are isolated using ultracentrifugation. Finally, the target cells are transfected with the VLPs. Portions of this figure were created with BioRender.com.

Advertisement

9. Molecular docking in RNA-based therapeutics

Molecular docking is a key tool in structural molecular biology and computer-assisted drug design. The successful docking methods effectively explore high-dimensional spaces and employ a scoring function that accurately ranks candidate dockings. Docking can be utilized for virtual screening of extensive compound libraries, ranking the outcomes, and proposing structural hypotheses on how RNAs inhibit targets, which is invaluable in lead optimization [166].

The rapidly expanding therapeutic interest in RNA-targeted drug discovery has led to an increasing demand for computational tools that can predict interactions between RNA and ligands. Virtual screening remains a crucial initial step in the design of novel drugs, particularly when only information about the targeted RNA is available. Artificial intelligence (AI) is often portrayed as a new Industrial Revolution [167]. AI finds applications in various domains, including molecular simulation for drug discovery [17]. Various methods for docking (sampling) and scoring have been developed to expedite this process, thereby enhancing our understanding of the mechanisms underlying RNA-ligand binding. Physics-based and knowledge-based approaches have demonstrated promising success in predicting both ligand-binding poses and affinities [168]. The process of molecular docking verification, see Figure 9.

Figure 9.

The process of molecular docking verification.

It has been demonstrated that neglecting the flexibility of the protein (receptor) during protein-ligand docking can lead to inaccurate predictions of binding modes [169]. This issue becomes even more critical when dealing with ligand binding to RNA, which exhibits greater structural flexibility compared to proteins. RNA-Ligand DOCKing (RLDOCK) is a recently developed docking model for flexible ligands using a multiconformer approach [170, 171].

Successful computational drug discovery requires the integration of three essential components: (1) a methodology for identifying druggable RNA targets, (2) a computationally efficient sampling algorithm for exploring RNA conformations, ligand conformers, and ligand binding poses, and (3) accurate scoring functions to evaluate the structures of RNA-ligand complexes and assess their binding affinity [168].

Due to the limitations of the delivery system, it is currently restricted to small RNA delivery. However, emerging evidence highlights the crucial role of lncRNAs and circRNAs in disease pathogenesis. Given their extended length, current delivery systems are inadequate for introducing these RNAs into the body. Molecular docking analysis between RNA and target molecules enables the identification of active domains and nucleotide sites that exert local key functions. Consequently, short functional RNA sequences can be designed for these regions and introduced into the body using an appropriate delivery system. Upon demonstrating efficacy at cellular and animal levels, this approach holds promise for developing vaccines or nucleic acid therapeutics suitable for clinical applications.

Advertisement

10. Prospects

In order to improve the targeting and efficacy of RNA therapeutics, improving RNA stability, reducing RNA-induced immunological reactions, and improving the efficiency of RNA delivery systems are the main challenges facing the development of RNA drugs in the future. The integration of ncRNA identification against conserved regions within key genomic sites associated with PAH, coupled with the development of safe nanomedicines and the production of dry powders for inhalation to enhance storage stability, positions us favorably in our pursuit of RNA therapeutics.

Using computer-aided molecular docking technology to predict the interaction and affinity of ncRNAs with their targets through 3D structure, it is helpful to construct ncRNA drugs based on the finding of effective domains of ncRNAs. Then, the constructed RNA will play therapeutic roles through suitable in vivo delivery systems. With the development of various computational tools developed for RNA-targeted drug discovery, a CASP- and D3R-like [172, 173] community-wide events with blind tests and well-curated benchmark datasets, similar to those the benchmarks widely used in the protein-ligand modeling community, would be much needed [67174, 175, 176, 177, 178].

In addition, the discovery of specific markers for various cells and specific lipid components that can target the delivery of RNA-LNPs is crucial to improving the targeting effect of RNA delivery systems. The conjugation and expression of specific peptides or aptamers on RNA-LNPs are likely to be the most reliable strategy to recognize and deliver RNA-LNPs into specific organs and/or cells. In the future, advances in basic research will make it possible to achieve the precision and efficiency of ncRNA therapies.

Moreover, it is imperative to establish a correlation between the outcomes of our in vivo studies and computational as well as experimental parameters. Computational approaches can be employed to accurately predict efficient ncRNA nanocarriers. Looking ahead, we are actively developing a machine-learning algorithm that will facilitate expedited clinical trials with RNA therapeutics by effectively predicting the most suitable ncRNA nanocarriers.

In conclusion, the roles of ncRNAs in PAH have been continuously clarified. Treating PAH patients with ncRNAs through appropriate delivery systems will be the trend of PAH therapy in the future. The application of molecular docking technology will help ncRNAs play a targeted therapeutic role in clinical medicine.

References

  1. 1. Galiè N, Humbert M, Vachiery JL, et al. 2015 ESC/ERS guidelines for the diagnosis and treatment of pulmonary hypertension. Kardiologia Polska. 2015;73(12):1127-1206
  2. 2. Weber L, Rickli H, Haager PK, et al. Haemodynamic mechanisms and long-term prognostic impact of pulmonary hypertension in patients with severe aortic stenosis undergoing valve replacement. European Journal of Heart Failure. 2019;21(2):172-181
  3. 3. Bourgeois A, Lambert C, Habbout K, et al. FOXM1 promotes pulmonary artery smooth muscle cell expansion in pulmonary arterial hypertension. Journal of Molecular Medicine (Berlin, Germany). 2018;96(2):223-235
  4. 4. Bourgeois A, Omura J, Habbout K, Bonnet S, Boucherat O. Pulmonary arterial hypertension: New pathophysiological insights and emerging therapeutic targets. The International Journal of Biochemistry & Cell Biology. 2018;104:9-13
  5. 5. Chelladurai P, Seeger W, Pullamsetti SS. Epigenetic mechanisms in pulmonary arterial hypertension: The need for global perspectives. European Respiratory Review. 2016;25(140):135-140
  6. 6. Veith C, Schermuly RT, Brandes RP, Weissmann N. Molecular mechanisms of hypoxia-inducible factor-induced pulmonary arterial smooth muscle cell alterations in pulmonary hypertension. The Journal of Physiology. 2016;594(5):1167-1177
  7. 7. Wakiyama M, Yokoyama S. MicroRNA-mediated deadenylation in a mammalian cell-free system. Methods in Molecular Biology. 2014;1125:341-351
  8. 8. Xiao L, Wang J, Ju S, Cui M, Jing R. Disorders and roles of tsRNA, snoRNA, snRNA and piRNA in cancer. Journal of Medical Genetics. 2022;59(7):623-631
  9. 9. Botti G, Marra L, Malzone MG, et al. LncRNA HOTAIR as prognostic circulating marker and potential therapeutic target in patients with tumor diseases. Current Drug Targets. 2017;18(1):27-34
  10. 10. Di X, Jin X, Li R, Zhao M, Wang K. CircRNAs and lung cancer: Biomarkers and master regulators. Life Sciences. 2019;220:177-185
  11. 11. Correia de Sousa M, Gjorgjieva M, Dolicka D, Sobolewski C, Foti M. Deciphering miRNAs' action through miRNA editing. International Journal of Molecular Sciences. 2019;20(24):6249
  12. 12. Crooke ST, Witztum JL, Bennett CF, Baker BF. RNA-targeted therapeutics. Cell Metabolism. 2018;27(4):714-739
  13. 13. Yin W, Rogge M. Targeting RNA: A transformative therapeutic strategy. Clinical and Translational Science. 2019;12(2):98-112
  14. 14. Yu AM, Jian C, Yu AH, Tu MJ. RNA therapy: Are we using the right molecules? Pharmacology & Therapeutics. 2019;196:91-104
  15. 15. Yu AM, Choi YH, Tu MJ. RNA drugs and RNA targets for small molecules: Principles, progress, and challenges. Pharmacological Reviews. 2020;72(4):862-898
  16. 16. Connelly C, Moon M, Schneekloth J. The emerging role of RNA as a therapeutic target for small molecules. Cell Chemical Biology. 2016;23(9):1077-1090
  17. 17. Somody JC, MacKinnon SS, Windemuth A. Structural coverage of the proteome for pharmaceutical applications. Drug Discovery Today. 2017;22(12):1792-1799
  18. 18. Warner KD, Hajdin CE, Weeks KM. Principles for targeting RNA with drug-like small molecules. Nature Reviews Drug Discovery. 2018;17(8):547-558
  19. 19. Tessaro F, Scapozza L. How ‘protein-docking’ translates into the new emerging field of docking small molecules to nucleic acids? Molecules. 2020;25(12):2749-2764
  20. 20. Costales MG, Childs-Disney JL, Haniff HS, Disney MD. How we think about targeting RNA with small molecules. Journal of Medicinal Chemistry. 2020;63(17):8880-8900
  21. 21. Clamp M, Fry B, Kamal M, Xie X, Cuff J, Lin MF, et al. Distinguishing protein-coding and noncoding genes in the human genome. Proceedings of the National Academy of Sciences. 2007;104(49):19428-19433
  22. 22. Hopkins AL, Groom CR. The druggable genome. Nature Reviews Drug Discovery. 2002;1(9):727-730
  23. 23. Dixon SJ, Stockwell BR. Identifying druggable disease-modifying gene products. Current Opinion in Chemical Biology. 2009;13(5):549-555
  24. 24. Overington JP, Al-Lazikani B, Hopkins AL. How many drug targets are there? Nature Reviews Drug Discovery. 2006;5(12):993-996
  25. 25. Jordan PC, Stevens SK, Deval J. Nucleosides for the treatment of respiratory RNA virus infections. Antiviral Chemistry & Chemotherapy. 2018;26:2040206618764483
  26. 26. Sullenger BA, Nair S. From the RNA world to the clinic. Science. 2016;352(6292):1417-1420
  27. 27. Dorsett Y, Tuschl T. siRNAs: Applications in functional genomics and potential as therapeutics. Nature Reviews. Drug Discovery. 2004;3(4):318-329
  28. 28. Kifle ZD, Ayele AG, Enyew EF. Drug repurposing approach, potential drugs, and novel drug targets for COVID-19 treatment. Journal of Environmental and Public Health. 2021;2021:6631721
  29. 29. Liu C, Zhou Q , Li Y, et al. Research and Development on therapeutic agents and vaccines for COVID-19 and related human coronavirus diseases. ACS Central Science. 2020;6(3):315-331
  30. 30. Rothman AM, Arnold ND, Pickworth JA, et al. MicroRNA-140-5p and SMURF1 regulate pulmonary arterial hypertension. The Journal of Clinical Investigation. 2016;126(7):2495-2508
  31. 31. Düzgün Z, Kayıkçıoğlu LM, Aktan Ç, et al. Decreased circulating microRNA-21 and microRNA-143 are associated to pulmonary hypertension. Turkish Journal of Medical Sciences. 2023;53(1):130-141
  32. 32. Wang F, Zuroske T, Watts JK. RNA therapeutics on the rise. Nature Reviews. Drug Discovery. 2020;19(7):441-442
  33. 33. Roberts TC, Langer R, Wood M. Advances in oligonucleotide drug delivery. Nature Reviews. Drug Discovery. 2020;19(10):673-694
  34. 34. Kulkarni JA, Witzigmann D, Thomson SB, et al. The current landscape of nucleic acid therapeutics. Nature Nanotechnology. 2021;16(6):630-643
  35. 35. Merkel OM, Librizzi D, Pfestroff A, et al. Stability of siRNA polyplexes from poly(ethylenimine) and poly(ethylenimine)-g-poly(ethylene glycol) under in vivo conditions: Effects on pharmacokinetics and biodistribution measured by Fluorescence Fluctuation Spectroscopy and Single Photon Emission Computed Tomography (SPECT) imaging. Journal of Controlled Release. 2009;138(2):148-159
  36. 36. Titze-de-Almeida R, David C, Titze-de-Almeida SS. The race of 10 synthetic RNAi-based drugs to the pharmaceutical market. Pharmaceutical Research. 2017;34(7):1339-1363
  37. 37. Mehta A, Michler T, Merkel OM. siRNA therapeutics against respiratory viral infections-what have we learned for potential COVID-19 therapies. Advanced Healthcare Materials. 2021;10(7):e2001650
  38. 38. Winter J, Jung S, Keller S, Gregory RI, Diederichs S. Many roads to maturity: microRNA biogenesis pathways and their regulation. Nature Cell Biology. 2009;11(3):228-234
  39. 39. Ling H, Fabbri M, Calin GA. MicroRNAs and other non-coding RNAs as targets for anticancer drug development. Nature Reviews. Drug Discovery. 2013;12(11):847-865
  40. 40. Pritchard CC, Cheng HH, Tewari M. MicroRNA profiling: Approaches and considerations. Nature Reviews. Genetics. 2012;13(5):358-369
  41. 41. Gu S, Jin L, Zhang F, Sarnow P, Kay MA. Biological basis for restriction of microRNA targets to the 3′ untranslated region in mammalian mRNAs. Nature Structural & Molecular Biology. 2009;16(2):144-150
  42. 42. Thenappan T, Ormiston ML, Ryan JJ, Archer SL. Pulmonary arterial hypertension: Pathogenesis and clinical management. BMJ. 2018;360:j5492
  43. 43. Bertero T, Lu Y, Annis S, et al. Systems-level regulation of microRNA networks by miR-130/301 promotes pulmonary hypertension. The Journal of Clinical Investigation. 2014;124(8):3514-3528
  44. 44. Chen T, Zhou G, Zhou Q , et al. Loss of microRNA-17∼92 in smooth muscle cells attenuates experimental pulmonary hypertension via induction of PDZ and LIM domain 5. American Journal of Respiratory and Critical Care Medicine. 2015;191(6):678-692
  45. 45. Fu J, Bai P, Chen Y, Yu T, Li F. Inhibition of miR-495 improves both vascular Remodeling and angiogenesis in pulmonary hypertension. Journal of Vascular Research. 2019;56(2):97-106
  46. 46. Ma W, Qiu Z, Bai Z, et al. Inhibition of microRNA-30a alleviates vascular remodeling in pulmonary arterial hypertension. Molecular Therapy - Nucleic Acids. 2021;26:678-693
  47. 47. Jiang Y, Liu H, Yu H, et al. Circular RNA Calm4 regulates hypoxia-induced pulmonary arterial smooth muscle cells Pyroptosis via the Circ-Calm4/miR-124-3p/PDCD6 Axis. Arteriosclerosis, Thrombosis, and Vascular Biology. 2021;41(5):1675-1693
  48. 48. Zhang J, Li Y, Qi J, et al. Circ-calm4 serves as an miR-337-3p sponge to regulate Myo10 (myosin 10) and promote pulmonary artery smooth muscle proliferation. Hypertension. 2020;75(3):668-679
  49. 49. Errington N, Iremonger J, Pickworth JA, et al. A diagnostic miRNA signature for pulmonary arterial hypertension using a consensus machine learning approach. eBioMedicine. 2021;69:103444
  50. 50. Slack FJ, Chinnaiyan AM. The role of non-coding RNAs in oncology. Cell. 2019;179(5):1033-1055
  51. 51. Prakash S, Dhanushkodi NR, Zayou L, et al. Cross-protection induced by highly conserved human B, CD4+, and CD8+ T cell epitopes-based coronavirus vaccine against severe infection, disease, and death caused by multiple SARS-CoV-2 variants of concern. bioRxiv. 24 May 2023. 541850 [Preprint]
  52. 52. Hu J, Xu Y, Hao J, Wang S, Li C, Meng S. MiR-122 in hepatic function and liver diseases. Protein & Cell. 2012;3(5):364-371
  53. 53. Zhang L, Liao Y, Tang L. MicroRNA-34 family: A potential tumor suppressor and therapeutic candidate in cancer. Journal of Experimental & Clinical Cancer Research. 2019;38(1):53
  54. 54. Parrella P, Barbano R, Pasculli B, et al. Evaluation of microRNA-10b prognostic significance in a prospective cohort of breast cancer patients. Molecular Cancer. 2014;13:142
  55. 55. Inoue J, Inazawa J. Cancer-associated miRNAs and their therapeutic potential. Journal of Human Genetics. 2021;66(9):937-945
  56. 56. Chang WT, Lee WC, Lin YW, et al. Transpulmonary expression of exosomal microRNAs in idiopathic and congenital heart disease-related pulmonary arterial hypertension. Journal of the American Heart Association. 2023;12(23):e031435
  57. 57. Ali MK, Schimmel K, Zhao L, et al. The role of circular RNAs in pulmonary hypertension. The European Respiratory Journal. 2022;60(6):2200012
  58. 58. Yu CY, Kuo HC. The emerging roles and functions of circular RNAs and their generation. Journal of Biomedical Science. 2019;26(1):29
  59. 59. Li J, Xu Q , Huang ZJ, et al. CircRNAs: A new target for the diagnosis and treatment of digestive system neoplasms. Cell Death & Disease. 2021;12(2):205
  60. 60. Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi PP. A ceRNA hypothesis: The Rosetta stone of a hidden RNA language. Cell. 2011;146(3):353-358
  61. 61. Du WW, Yang W, Liu E, Yang Z, Dhaliwal P, Yang BB. Foxo3 circular RNA retards cell cycle progression via forming ternary complexes with p21 and CDK2. Nucleic Acids Research. 2016;44(6):2846-2858
  62. 62. Legnini I, Di Timoteo G, Rossi F, et al. Circ-ZNF609 is a circular RNA that can be translated and functions in myogenesis. Molecular Cell. 2017;66(1):22-37.e9
  63. 63. Conn SJ, Pillman KA, Toubia J, et al. The RNA binding protein quaking regulates formation of circRNAs. Cell. 2015;160(6):1125-1134
  64. 64. Pan Z, Cai J, Lin J, et al. A novel protein encoded by circFNDC3B inhibits tumor progression and EMT through regulating snail in colon cancer. Molecular Cancer. 2020;19(1):71
  65. 65. Hanan M, Soreq H, Kadener S. CircRNAs in the brain. RNA Biology. 2017;14(8):1028-1034
  66. 66. Kristensen LS, Hansen TB, Venø MT, Kjems J. Circular RNAs in cancer: Opportunities and challenges in the field. Oncogene. 2018;37(5):555-565
  67. 67. Li H, Li K, Lai W, et al. Comprehensive circular RNA profiles in plasma reveals that circular RNAs can be used as novel biomarkers for systemic lupus erythematosus. Clinica Chimica Acta. 2018;480:17-25
  68. 68. Vo JN, Cieslik M, Zhang Y, et al. The landscape of circular RNA in cancer. Cell. 2019;176(4):869-881.e13
  69. 69. Chouvarine P, Photiadis J, Cesnjevar R, et al. RNA expression profiles and regulatory networks in human right ventricular hypertrophy due to high pressure load. iScience. 2021;24(3):102232
  70. 70. Guo HM, Liu ZP. Up-regulation of circRNA_0068481 promotes right ventricular hypertrophy in PAH patients via regulating miR-646/miR-570/miR-885. Journal of Cellular and Molecular Medicine. 2021;25(8):3735-3743
  71. 71. Miao R, Wang Y, Wan J, et al. Microarray expression profile of circular RNAs in chronic thromboembolic pulmonary hypertension. Medicine (Baltimore). 2017;96(27):e7354
  72. 72. Hong L, Ma X, Liu J, et al. Circular RNA-HIPK3 regulates human pulmonary artery endothelial cells function and vessel growth by regulating microRNA-328-3p/STAT3 axis. Pulmonary Circulation. 2021;11(2):20458940211000234
  73. 73. Jin X, Xu Y, Guo M, et al. hsa_circNFXL1_009 modulates apoptosis, proliferation, migration, and potassium channel activation in pulmonary hypertension. Molecular Therapy - Nucleic Acids. 2021;23:1007-1019
  74. 74. Zhang F, Jiang J, Qian H, Yan Y, Xu W. Exosomal circRNA: Emerging insights into cancer progression and clinical application potential. Journal of Hematology & Oncology. 2023;16(1):67
  75. 75. Zhang W, Zhao J, Deng L, et al. INKILN is a novel long noncoding RNA promoting vascular smooth muscle inflammation via scaffolding MKL1 and USP10. Circulation. 2023;148(1):47-67
  76. 76. Yue X, Lan F, Liu W. Serum exosomal circCCDC66 as a potential diagnostic and prognostic biomarker for pituitary adenomas. Frontiers in Oncology. 2023;13:1268778
  77. 77. Huang Y, Su D, Ye B, et al. Expression and clinical significance of circular RNA hsa_circ_0003416 in pediatric pulmonary arterial hypertension associated with congenital heart disease. Journal of Clinical Laboratory Analysis. 2022;36(4):e24273
  78. 78. Zhao L, Gu C, Gan Y, Shao L, Chen H, Zhu H. Exosome-mediated siRNA delivery to suppress postoperative breast cancer metastasis. Journal of Controlled Release. 2020;318:1-15
  79. 79. Fan Y, Wang J, Jin W, et al. CircNR3C2 promotes HRD1-mediated tumor-suppressive effect via sponging miR-513a-3p in triple-negative breast cancer. Molecular Cancer. 2021;20(1):25
  80. 80. Su L, Li X, Mao X, et al. Circ-Ntrk2 acts as a miR-296-5p sponge to activate the TGF-β1/p38 MAPK pathway and promote pulmonary hypertension and vascular remodelling. Respiratory Research. 2023;24(1):78
  81. 81. Kristensen LS, Jakobsen T, Hager H, Kjems J. The emerging roles of circRNAs in cancer and oncology. Nature Reviews. Clinical Oncology. 2022;19(3):188-206
  82. 82. Zhao Q , Liu J, Deng H, et al. Targeting mitochondria-located circRNA SCAR alleviates NASH via reducing mROS output. Cell. 2020;183(1):76-93.e22
  83. 83. Wesselhoeft RA, Kowalski PS, Parker-Hale FC, Huang Y, Bisaria N, Anderson DG. RNA circularization diminishes immunogenicity and can extend translation duration In vivo. Molecular Cell. 2019;74(3):508-520.e4
  84. 84. Lavenniah A, Luu T, Li YP, et al. Engineered circular RNA sponges act as miRNA inhibitors to attenuate pressure overload-induced cardiac hypertrophy. Molecular Therapy. 2020;28(6):1506-1517
  85. 85. Liu X, Abraham JM, Cheng Y, et al. Synthetic circular RNA functions as a miR-21 sponge to suppress gastric carcinoma cell proliferation. Molecular Therapy - Nucleic Acids. 2018;13:312-321
  86. 86. Schreiner S, Didio A, Hung LH, Bindereif A. Design and application of circular RNAs with protein-sponge function. Nucleic Acids Research. 2020;48(21):12326-12335
  87. 87. Wang Z, Ma K, Cheng Y, et al. Synthetic circular multi-miR sponge simultaneously inhibits miR-21 and miR-93 in esophageal carcinoma. Laboratory Investigation. 2019;99(10):1442-1453
  88. 88. Qu L, Yi Z, Shen Y, et al. Circular RNA vaccines against SARS-CoV-2 and emerging variants. Cell. 2022;185(10):1728-1744.e16
  89. 89. Li H, Peng K, Yang K, et al. Circular RNA cancer vaccines drive immunity in hard-to-treat malignancies. Theranostics. 2022;12(14):6422-6436
  90. 90. Ponting CP, Oliver PL, Reik W. Evolution and functions of long noncoding RNAs. Cell. 2009;136(4):629-641
  91. 91. Wierzbicki AT, Haag JR, Pikaard CS. Noncoding transcription by RNA polymerase pol IVb/Pol V mediates transcriptional silencing of overlapping and adjacent genes. Cell. 2008;135(4):635-648
  92. 92. Malakar P, Shilo A, Mogilevsky A, et al. Long noncoding RNA MALAT1 promotes hepatocellular carcinoma development by SRSF1 upregulation and mTOR activation. Cancer Research. 2017;77(5):1155-1167
  93. 93. Rinn JL, Chang HY. Genome regulation by long noncoding RNAs. Annual Review of Biochemistry. 2012;81:145-166
  94. 94. Ørom UA, Shiekhattar R. Long noncoding RNAs usher in a new era in the biology of enhancers. Cell. 2013;154(6):1190-1193
  95. 95. Eptaminitaki GC, Stellas D, Bonavida B, Baritaki S. Long non-coding RNAs (lncRNAs) signaling in cancer chemoresistance: From prediction to druggability. Drug Resistance Updates. 2022;65:100866
  96. 96. Omura J, Habbout K, Shimauchi T, et al. Identification of long noncoding RNA H19 as a new biomarker and therapeutic target in right ventricular failure in pulmonary arterial hypertension. Circulation. 2020;142(15):1464-1484
  97. 97. Wang S, Luo Z, Yuan L, et al. tRNA-derived small RNAs: Novel insights into the pathogenesis and treatment of cardiovascular diseases. Journal of Cardiovascular Translational Research. 2023;16(2):300-309
  98. 98. Wang T, Sung TC, Yu T, et al. Next-generation materials for RNA-lipid nanoparticles: Lyophilization and targeted transfection. Journal of Materials Chemistry B. 2023;11(23):5083-5093
  99. 99. Wang Y, Chen D, Xie H, et al. LncRNA GAS5 suppresses TGF-β1-induced transformation of pulmonary pericytes into myofibroblasts by recruiting KDM5B and promoting H3K4me2/3 demethylation of the PDGFRα/β promoter. Molecular Medicine. 2023;29(1):32
  100. 100. Yu J, Wang W, Yang J, et al. LncRNA PSR regulates vascular remodeling through encoding a novel protein Arteridin. Circulation Research. 2022;131(9):768-787
  101. 101. Yang L, Liang H, Shen L, Guan Z, Meng X. LncRNA Tug1 involves in the pulmonary vascular remodeling in mice with hypoxic pulmonary hypertension via the microRNA-374c-mediated Foxc1. Life Sciences. 2019;237:116769
  102. 102. Su H, Xu X, Yan C, et al. LncRNA H19 promotes the proliferation of pulmonary artery smooth muscle cells through AT1R via sponging let-7b in monocrotaline-induced pulmonary arterial hypertension. Respiratory Research. 2018;19(1):254
  103. 103. Su H, Liu B, Chen H, et al. LncRNA ANRIL mediates endothelial dysfunction through BDNF downregulation in chronic kidney disease. Cell Death & Disease. 2022;13(7):661
  104. 104. Zahid KR, Raza U, Chen J, Raj UJ, Gou D. Pathobiology of pulmonary artery hypertension: Role of long non-coding RNAs. Cardiovascular Research. 2020;116(12):1937-1947
  105. 105. Gong Q , Hu Z, Jin Q , et al. Identification of JPX-RABEP1 pair as an immune-related biomarker and therapeutic target in pulmonary arterial hypertension by bioinformatics and experimental analyses. International Journal of Molecular Sciences. 2022;23(24)
  106. 106. Zhuo Y, Zeng Q , Zhang P, Li G, Xie Q , Cheng Y. Functional polymorphism of lncRNA MALAT1 contributes to pulmonary arterial hypertension susceptibility in Chinese people. Clinical Chemistry and Laboratory Medicine. 2017;55(1):38-46
  107. 107. Wang X, Li Q , He S, et al. LncRNA FENDRR with m6A RNA methylation regulates hypoxia-induced pulmonary artery endothelial cell pyroptosis by mediating DRP1 DNA methylation. Molecular Medicine. 2022;28(1):126
  108. 108. Song R, Lei S, Yang S, Wu SJ. LncRNA PAXIP1-AS1 fosters the pathogenesis of pulmonary arterial hypertension via ETS1/WIPF1/RhoA axis. Journal of Cellular and Molecular Medicine. 2021;25(15):7321-7334
  109. 109. Han S, Chen X, Huang L. The tumor therapeutic potential of long non-coding RNA delivery and targeting. Acta Pharmaceutica Sinica B. 2023;13(4):1371-1382
  110. 110. Peng Y, Huang X, Wang H. Serum lncRNA LINC01535 as biomarker of diagnosis, prognosis, and disease progression in breast cancer. Clinical Breast Cancer. 2023;23(6):620-627
  111. 111. Hsu XR, Wu JE, Wu YY, et al. Exosomal long noncoding RNA MLETA1 promotes tumor progression and metastasis by regulating the miR-186-5p/EGFR and miR-497-5p/IGF1R axes in non-small cell lung cancer. Journal of Experimental & Clinical Cancer Research. 2023;42(1):283
  112. 112. Bourdoumis A, Papatsoris AG, Chrisofos M, Efstathiou E, Skolarikos A, Deliveliotis C. The novel prostate cancer antigen 3 (PCA3) biomarker. International Brazilian Journal of Urology. 2010;36(6):665-668; discussion 669
  113. 113. Greco S, Zaccagnini G, Perfetti A, et al. Long noncoding RNA dysregulation in ischemic heart failure. Journal of Translational Medicine. 2016;14(1):183
  114. 114. Girard A, Sachidanandam R, Hannon GJ, Carmell MA. A germline-specific class of small RNAs binds mammalian Piwi proteins. Nature. 2006;442(7099):199-202
  115. 115. Aravin A, Gaidatzis D, Pfeffer S, et al. A novel class of small RNAs bind to MILI protein in mouse testes. Nature. 2006;442(7099):203-207
  116. 116. Halbach R, Miesen P, Joosten J, et al. A satellite repeat-derived piRNA controls embryonic development of Aedes. Nature. 2020;580(7802):274-277
  117. 117. Guo Z, Li Y, Ding SW. Small RNA-based antimicrobial immunity. Nature Reviews. Immunology. 2019;19(1):31-44
  118. 118. Ma C, Zhang L, Wang X, et al. piRNA-63076 contributes to pulmonary arterial smooth muscle cell proliferation through acyl-CoA dehydrogenase. Journal of Cellular and Molecular Medicine. 2020;24(9):5260-5273
  119. 119. Lipps C, Northe P, Figueiredo R, et al. Non-invasive approach for evaluation of pulmonary hypertension using extracellular vesicle-associated small non-coding RNA. Biomolecules. 2019;9(11):666
  120. 120. Liu Y, Dou M, Song X, et al. The emerging role of the piRNA/piwi complex in cancer. Molecular Cancer. 2019;18(1):123
  121. 121. Mai D, Ding P, Tan L, et al. PIWI-interacting RNA-54265 is oncogenic and a potential therapeutic target in colorectal adenocarcinoma. Theranostics. 2018;8(19):5213-5230
  122. 122. Esteller M. Non-coding RNAs in human disease. Nature Reviews. Genetics. 2011;12(12):861-874
  123. 123. Maxwell ES, Fournier MJ. The small nucleolar RNAs. Annual Review of Biochemistry. 1995;64:897-934
  124. 124. Mallardo M, Poltronieri P, D'Urso OF. Non-protein coding RNA biomarkers and differential expression in cancers: A review. Journal of Experimental & Clinical Cancer Research. 2008;27(1):19
  125. 125. Jaé N, Dimmeler S. Noncoding RNAs in vascular diseases. Circulation Research. 2020;126(9):1127-1145
  126. 126. Al Tabosh T, Liu H, Koça D, et al. Impact of heterozygous ALK1 mutations on the transcriptomic response to BMP9 and BMP10 in endothelial cells from hereditary hemorrhagic telangiectasia and pulmonary arterial hypertension donors. Angiogenesis. 2024;27(2):211-227
  127. 127. Chen Q , Yan M, Cao Z, et al. Sperm tsRNAs contribute to intergenerational inheritance of an acquired metabolic disorder. Science. 2016;351(6271):397-400
  128. 128. Waldron D. Non-coding RNA: Inheritance of diet-induced metabolic changes via tsRNAs. Nature Reviews. Genetics. 2016;17(3):128
  129. 129. Park J, Ahn SH, Shin MG, Kim HK, Chang S. tRNA-derived small RNAs: Novel epigenetic regulators. Cancers (Basel). 2020;12(10):2773
  130. 130. Chen Y, Tang Y, Hou S, et al. Differential expression spectrum and targeted gene prediction of tRNA-derived small RNAs in idiopathic pulmonary arterial hypertension. Frontiers in Molecular Biosciences. 2023;10:1204740
  131. 131. Diener C, Keller A, Meese E. Emerging concepts of miRNA therapeutics: From cells to clinic. Trends in Genetics. 2022;38(6):613-626
  132. 132. Finer M, Glorioso J. A brief account of viral vectors and their promise for gene therapy. Gene Therapy. 2017;24(1):1-2
  133. 133. Savenkova DA, Makarova AA, Shalik IK, Yudkin DV. miRNA pathway alteration in response to non-coding RNA delivery in viral vector-based gene therapy. International Journal of Molecular Sciences. 2022;23(23):14954
  134. 134. Samulski RJ, Muzyczka N. AAV-mediated gene therapy for research and therapeutic purposes. Annual Review of Virology. 2014;1(1):427-451
  135. 135. Saraiva J, Nobre RJ, Pereira de Almeida L. Gene therapy for the CNS using AAVs: The impact of systemic delivery by AAV9. Journal of Controlled Release. 2016;241:94-109
  136. 136. Verdera HC, Kuranda K, Mingozzi F. AAV vector immunogenicity in humans: A long journey to successful gene transfer. Molecular Therapy. 2020;28(3):723-746
  137. 137. Bulcha JT, Wang Y, Ma H, Tai P, Gao G. Viral vector platforms within the gene therapy landscape. Signal Transduction and Targeted Therapy. 2021;6(1):53
  138. 138. Ghebremedhin B. Human adenovirus: Viral pathogen with increasing importance. European Journal of Microbiology and Immunology. 2014;4(1):26-33
  139. 139. Allen RJ, Byrnes AP. Interaction of adenovirus with antibodies, complement, and coagulation factors. FEBS Letters. 2019;593(24):3449-3460
  140. 140. Lee CS, Bishop ES, Zhang R, et al. Adenovirus-mediated gene delivery: Potential applications for gene and cell-based therapies in the new era of personalized medicine. Genes & Diseases. 2017;4(2):43-63
  141. 141. Sweeney NP, Vink CA. The impact of lentiviral vector genome size and producer cell genomic to gag-pol mRNA ratios on packaging efficiency and titre. Molecular Therapy - Methods & Clinical Development. 2021;21:574-584
  142. 142. Moraes FC, Pichon C, Letourneur D, Chaubet F. miRNA delivery by nanosystems: State of the art and perspectives. Pharmaceutics. 2021;13(11):1901
  143. 143. Alameh MG, Tombácz I, Bettini E, et al. Lipid nanoparticles enhance the efficacy of mRNA and protein subunit vaccines by inducing robust T follicular helper cell and humoral responses. Immunity. 2021;54(12):2877-2892.e7
  144. 144. Chen Y, Li Z, Chen X, Zhang S. Long non-coding RNAs: From disease code to drug role. Acta Pharmaceutica Sinica B. 2021;11(2):340-354
  145. 145. Witzigmann D, Kulkarni JA, Leung J, Chen S, Cullis PR, van der Meel R. Lipid nanoparticle technology for therapeutic gene regulation in the liver. Advanced Drug Delivery Reviews. 2020;159:344-363
  146. 146. Springer AD, Dowdy SF. GalNAc-siRNA conjugates: Leading the way for delivery of RNAi therapeutics. Nucleic Acid Therapeutics. 2018;28(3):109-118
  147. 147. Ozpolat B, Sood AK, Lopez-Berestein G. Liposomal siRNA nanocarriers for cancer therapy. Advanced Drug Delivery Reviews. 2014;66:110-116
  148. 148. García-Pinel B, Porras-Alcalá C, Ortega-Rodríguez A, et al. Lipid-based nanoparticles: Application and recent advances in cancer treatment. Nanomaterials (Basel). 2019;9(4):638
  149. 149. Cano A, Ettcheto M, Chang JH, et al. Dual-drug loaded nanoparticles of Epigallocatechin-3-gallate (EGCG)/ascorbic acid enhance therapeutic efficacy of EGCG in a APPswe/PS1dE9 Alzheimer's disease mice model. Journal of Controlled Release. 2019;301:62-75
  150. 150. Cano A, Sánchez-López E, Ettcheto M, et al. Current advances in the development of novel polymeric nanoparticles for the treatment of neurodegenerative diseases. Nanomedicine (London, England). 2020;15(12):1239-1261
  151. 151. Soppimath KS, Aminabhavi TM, Kulkarni AR, Rudzinski WE. Biodegradable polymeric nanoparticles as drug delivery devices. Journal of Controlled Release. 2001;70(1-2):1-20
  152. 152. Owens DE 3rd, Peppas NA. Opsonization, biodistribution, and pharmacokinetics of polymeric nanoparticles. International Journal of Pharmaceutics. 2006;307(1):93-102
  153. 153. Tanno T, Zhang P, Bailey C, et al. A novel aptamer-based small RNA delivery platform and its application to cancer therapy. Genes & Diseases. 2023;10(3):1075-1089
  154. 154. Rampado R, Caliceti P, Agostini M. Latest advances in biomimetic cell membrane-coated and membrane-derived Nanovectors for biomedical applications. Nanomaterials (Basel). 2022;12(9):1543
  155. 155. Fang RH, Kroll AV, Gao W, Zhang L. Cell membrane coating nanotechnology. Advanced Materials. 2018;30(23):e1706759
  156. 156. Cui J, Wang X, Li J, et al. Immune exosomes loading self-assembled nanomicelles traverse the blood-brain barrier for chemo-immunotherapy against glioblastoma. ACS Nano. 2023 [Online ahead of print]
  157. 157. Myo AC, Kobayashi Y, Niki Y, Kamimoto H, Moriyama K. Exosome-mediated small interfering RNA delivery inhibits aberrant osteoblast differentiation in apert syndrome model mice. Archives of Oral Biology. 2023;153:105753
  158. 158. Arya SB, Collie SP, Parent CA. The ins-and-outs of exosome biogenesis, secretion, and internalization. Trends in Cell Biology. 2024;34(2):90-108
  159. 159. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367(6478):eaau6977
  160. 160. Darband SG, Mirza-Aghazadeh-Attari M, Kaviani M, et al. Exosomes: Natural nanoparticles as bio shuttles for RNAi delivery. Journal of Controlled Release. 2018;289:158-170
  161. 161. Li X, Yu Q , Zhao R, et al. Designer exosomes for targeted delivery of a novel therapeutic cargo to enhance sorafenib-mediated ferroptosis in hepatocellular carcinoma. Frontiers in Oncology. 2022;12:898156
  162. 162. Zhong R, Talebian S, Mendes BB, et al. Hydrogels for RNA delivery. Nature Materials. 2023;22(7):818-831
  163. 163. Segel M, Lash B, Song J, et al. Mammalian retrovirus-like protein PEG10 packages its own mRNA and can be pseudotyped for mRNA delivery. Science. 2021;373(6557):882-889
  164. 164. Gutkin A, Rosenblum D, Peer D. RNA delivery with a human virus-like particle. Nature Biotechnology. 2021;39(12):1514-1515
  165. 165. Mohsen MO, Gomes AC, Vogel M, Bachmann MF. Interaction of viral capsid-derived virus-like particles (VLPs) with the innate immune system. Vaccines (Basel). 2018;6(3):37
  166. 166. Morris GM, Lim-Wilby M. Molecular docking. Methods in Molecular Biology. 2008;443:365-382
  167. 167. Ezkurdia I, Juan D, Rodriguez JM, et al. Multiple evidence strands suggest that there may be as few as 19, 000 human protein-coding genes. Human Molecular Genetics. 2014;23(22):5866-5878
  168. 168. Zhou Y, Jiang Y, Chen SJ. RNA-ligand molecular docking: Advances and challenges. Wiley Interdisciplinary Reviews: Computational Molecular Science. 2022;12(3):e1571
  169. 169. Totrov M, Abagyan R. Flexible ligand docking to multiple receptor conformations: A practical alternative. Current Opinion in Structural Biology. 2008;18(2):178-184
  170. 170. Jiang Y, Chen SJ. RLDOCK method for predicting RNA-small molecule binding modes. Methods. 2022;197:97-105
  171. 171. Sun LZ, Jiang Y, Zhou Y, Chen SJ. RLDOCK: A new method for predicting RNA-ligand interactions. Journal of Chemical Theory and Computation. 2020;16(11):7173-7183
  172. 172. Gathiaka S, Liu S, Chiu M, et al. D3R grand challenge 2015: Evaluation of protein-ligand pose and affinity predictions. Journal of Computer-Aided Molecular Design. 2016;30(9):651-668
  173. 173. Moult J, Pedersen JT, Judson R, Fidelis K. A large-scale experiment to assess protein structure prediction methods. Proteins. 1995;23(3):ii-v
  174. 174. Huang N, Shoichet BK, Irwin JJ. Benchmarking sets for molecular docking. Journal of Medicinal Chemistry. 2006;49(23):6789-6801
  175. 175. Li Y, Han L, Liu Z, Wang R. Comparative assessment of scoring functions on an updated benchmark: 2. Evaluation methods and general results. Journal of Chemical Information and Modeling. 2014;54(6):1717-1736
  176. 176. Li Y, Su M, Liu Z, et al. Assessing protein-ligand interaction scoring functions with the CASF-2013 benchmark. Nature Protocols. 2018;13(4):666-680
  177. 177. Mysinger MM, Carchia M, Irwin JJ, Shoichet BK. Directory of useful decoys, enhanced (DUD-E): Better ligands and decoys for better benchmarking. Journal of Medicinal Chemistry. 2012;55(14):6582-6594
  178. 178. Su M, Yang Q , Du Y, et al. Comparative assessment of scoring functions: The CASF-2016 update. Journal of Chemical Information and Modeling. 2019;59(2):895-913

Written By

Yingqi Wang, Daling Zhu and Hang Yu

Submitted: 25 February 2024 Reviewed: 03 March 2024 Published: 29 April 2024