Next Article in Journal
Comparative Analysis of Complete Chloroplast Genomes and Phylogenetic Relationships of 21 Sect. Camellia (Camellia L.) Plants
Next Article in Special Issue
Cell Cycle-Based Molecular Features via Synthetic Lethality and Non-Coding RNA Interactions in Cancer
Previous Article in Journal
Sickle Cell Disease in the Islands of Zanzibar: Patients’ Characteristics, Management, and Clinical Outcomes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Plethora of RNA–Protein Interactions Model a Basis for RNA Therapies

Department of Chemistry, The RNA Institute, University at Albany, SUNY, 1400 Washington Ave Extension, Albany, NY 12222, USA
*
Author to whom correspondence should be addressed.
Genes 2025, 16(1), 48; https://doi.org/10.3390/genes16010048
Submission received: 30 November 2024 / Revised: 25 December 2024 / Accepted: 27 December 2024 / Published: 2 January 2025
(This article belongs to the Special Issue Feature Papers: RNA)

Abstract

:
The notion of RNA-based therapeutics has gained wide attractions in both academic and commercial institutions. RNA is a polymer of nucleic acids that has been proven to be impressively versatile, dating to its hypothesized RNA World origins, evidenced by its enzymatic roles in facilitating DNA replication, mRNA decay, and protein synthesis. This is underscored through the activities of riboswitches, spliceosomes, ribosomes, and telomerases. Given its broad range of interactions within the cell, RNA can be targeted by a therapeutic or modified as a pharmacologic scaffold for diseases such as nucleotide repeat disorders, infectious diseases, and cancer. RNA therapeutic techniques that have been researched include, but are not limited to, CRISPR/Cas gene editing, anti-sense oligonucleotides (ASOs), siRNA, small molecule treatments, and RNA aptamers. The knowledge gleaned from studying RNA-centric mechanisms will inevitably improve the design of RNA-based therapeutics. Building on this understanding, we explore the physiological diversity of RNA functions, examine specific dysfunctions, such as splicing errors and viral interactions, and discuss their therapeutic implications.

1. Part I: RNA–Protein Evolution and Biology

Darwin’s Origin of Species postulated that all life descended from one primordial form. The earliest genetic material is believed to be RNA, which, according to the RNA World Theory, preceded the time of protein synthesis and function as enzymes [1]. Crick postulated that early ribosomes could entirely be made of RNA [2], and polypeptide synthesis without proteins has since been demonstrated as possible, albeit much less efficiently [2,3,4], casting the ribosome itself as a ribozyme. Proteins may facilitate this peptidyl transfer reaction by guiding the ribosome to a fold conducive for catalysis [5].
Still, the notion that RNA evolved independently of peptides remains ambivalent. The purported self-replication ability of ribozymes is yet to be shown experimentally [6], though Synak et al. (2020) reported the possibility of evolutionary selection for such a polymerase [7]. Moreover, Tagami et al. (2023) observed certain varieties of peptides that facilitate RNA synthesis through either their binding to RNA polymerase or formation of the ribozyme’s native core [6]. Such prebiotic peptides could have begun as noncoded amino acid chains from Miller’s primordial soup [8] that were amenable to polyribonucleotides in this prebiotic environment [9].
Amino acid-binding riboswitches may very well have laid the foundation for the deliberate formation of peptides [10], which later became intertwined with cellular signaling networks as the first RNA-binding proteins (RBPs) [11]. Thus, whether RNA and proteins evolved concurrently or sequentially, numerous RNA molecules and structures, including nucleotide-derived coenzymes, self-processing ribozymes, metabolite-binding riboswitches, and ribosomes, play vital roles in gene expression and cellular physiology [12].

1.1. Riboswitch Folding and Dynamics

As the name suggests, riboswitches are non-coding RNAs [13] that function as genetic switches [14]. Consisting of an aptamer and expression platform, the former operates as a sensor so as to cue its downstream component in response to the intracellular environment [15]. Such tuning of gene expression [16], per small metabolite, elemental ion, or other effector-types [17], harkens to the RNA World Theory [18]. To this point, riboswitches are capable of binding their target or otherwise adopting their prescribed regulatory structures without proteins, though, again, proteins may support these molecules’ structural versatility [19].
Although some level of conformational selection in the apo state has been observed [20], the dynamics among these regulatory folds are nonetheless crucial [21] for governing mRNA transcription, translation, and decay [22]. Indeed, distinct conformations were observed among the apo and bound states of the S-adenosylmethionine (SAM)-responsive riboswitches [23]. General conformational reorganizations involve loop–loop interactions and coaxial stacking while retaining intrinsically disordered regions for effector accessibility to the binding site [18].
Superseding the established two-state “on-off” system, Reining et al. (2013) investigated the adenine sensing riboswitch in Vibrio vulnificus using 1H-15N heteronuclear chemical exchange NMR spectroscopy, demonstrating a third conformation inherent to its mechanism [24]. Single-molecule FRET (smFRET) not only corroborated a kissing loop fold of this three-way junctional purine riboswitch aptamer, but further conveyed its persistence through docked and undocked conformations [25]. Thus, in-tandem, NMR and smFRET are powerful tools for comparing folded conformations and measuring their exchange dynamics [26].
Some riboswitches operate via the alternative pairing between nucleotides separated by some intramolecular distance [27], thereby bringing them together and facilitating the removal of an intron [28]. Such a regulation of alternative splicing by riboswitches is common across eukaryotes [29,30], and the knowledge of the binding site and splicing machinery can be exploited to engineer a riboswitch that upregulates a given protein [31].

1.2. Spliceosomes and Pre-mRNA Processing

The spliceosome is a ribonucleoprotein (RNP) complex that catalyzes pre-mRNA splicing to help pre-mRNA mature into mRNA by removing introns [32]. Exclusive to eukaryotic genes, the spliceosome’s structure is highly conserved among yeasts and metazoans [33]. Microorganisms, such as Escherichia coli or the unicellular eukaryotic Tetrahymena thermophila, have self-splicing capabilities [34].
Roughly 200 proteins and five snRNAs constitute the spliceosome, and the recognition of the splice site by multiple RNAs and proteins affords a high accuracy to the splicing process [33]. The two types of spliceosomes, major and minor, splice different exons, but can work together to influence mRNA processing where there are minor introns [35].
The major spliceosome splices 99.5% of introns with GU at the 5′ splice site and AG at the 3′ splice site [36]. The major spliceosome is composed of the snRNAs predominant in metazoans, plants, and fungi [36,37]. This spliceosome removes introns with GT-AT and occasionally AT-AC and GC-AG boundaries [36], as illustrated in Figure 1. The major spliceosome works by interactions between snRNAs and spliceosome proteins, in addition to their interactions with the pre-RNA substrate [36].
The minor spliceosome only splices 0.5% of introns and specializes in severing atypical U12-type introns [35]. Both the major and minor spliceosomes share an overall common secondary structure and span most eukaryotic lineages [35,36].

1.2.1. Small Nuclear (sn) RNAs and Pre-mRNA Form the Spliceosome

A number of factors interact with pre-mRNA to form the spliceosome and distribute the pre-mRNAs’ reactive groups for catalysis. Of these are various snRNPs and non-snRNPs alike, each of the former consisting of its own snRNAs [37]. The rearrangements among these snRNAs throughout the splicing process imparts structural fluidity among the many RNA–RNA and RNA–protein interactions that constitute a spliceosome [38]. The subsequent and spontaneous dissociation of these molecules from the spliceosome primes it for another round of splicing [39].

1.2.2. Polyadenylation in mRNA Stability and Translation

Adding a poly(A) tail to post-transcriptional mRNA can increase the stability in mRNA in cells [40]. Most poly(A) tails are located on the 3′ untranslated regions (3′ UTRs). The tail is recognized by nuclear export proteins, which help mature mRNA enter the cytoplasm from the nucleus [40]. The tail also binds to poly(A)-binding proteins (PABP), which are involved in translation efficiency, thereby linking the 5′ end to the 3′ end in mRNA and facilitating ribosomal attachment and recruitment [41].

1.3. RNA Stability and Decay

Gene expression is dependent on the stability of both coding and non-coding RNAs [42,43], as the former are receptive to environmental stimuli so as to modulate protein production accordingly [44]. Mechanistically, mRNA decay rates are dictated by bound proteins and non-coding RNAs [45] that facilitate mRNA degradation through various pathways [46,47].
A common approach among eukaryotes is the removal of the poly(A) tail in a process known as deadenylation and the subsequent exonucleolytic degradation in either the 5′ or 3′ direction [48]. Funakoshi et al. (2007) have demonstrated the eukaryotic elongation factor (eRF3) to work in concert with PABPC1 and the deadenylation complexes Pan2-Pan3 and Caf1-Ccr4 [49] in a sequence-specific manner [50]. These shortened poly(A) tails are then recognized by the Lsm1-7/Pat1 complex, which signal for the removal of the 5′ cap by the decapping enzyme complex Dcp1p/Dcp2p [51]. Consequently, mRNA degradation in the 5′-3′ direction ensues [52], though Audebert et al. (2024) recently demonstrated an independence between deadenylation and decapping [53].
The presence of AU-rich elements (AREs) in the 3′UTR of unstable mRNAs offers an alternate means of regulation over gene expression [54,55,56], and Park-Lee et al. (2003) demonstrated the efficacy of these regions in recruiting their binding partners of as little as 13 residues [57]. Moreover, two RNA-recognition motif (RRM) domains constituting HuD have been crystalized in complex with a short oligonucleotide fragment of class II AREs [58]. These Hu proteins belong to the ELAV family of RBPs, which comprises four members in humans: HuR occurs is all proliferating cell-types while Hel-N1, HuC, and HuD are confined to terminally differentiated neurons [59].
The overlap or close proximity among 3′ UTR regulatory sequences bound by RBPs and miRNAs suggests a concentration-dependent accessibility and, thus, competition among regulatory processes [60,61].

1.4. Telomerase RNA Component (TERC) and Cell Senescence

Telomeres are important for delaying the senescence of cells because every time cells split, the ends of DNA strands are excised [62]. Telomerase replicates telomeres and TERC is involved in synthesizing telomerase on chromosomal ends [63]. Non-coding RNAs are important in delaying senescence. Flores et al. (2001) demonstrated that TERC was restored in mice with short telomere length, increasing chromosomal stability; reviving telomere activity reversed some effects of aging in mice [64]. Moreover, Rudolph et al. (2024) showed that hepatic damage in the liver damages TERC length, resulting in senescence in liver cells [63]. The effects of senescence were shown to be reversible by TERC transfer, indicating the possible therapeutic use of TERC transfer, in the future, to combat liver cirrhosis [64,65]. Therapeutics in the pipeline targeting TERC, such as a possible gene therapy, has been postulated to prevent cancer [65]. For instance, Koscioleck et al. (2003) found that siRNAs targeting TERC or TERT (telomerase reverse transcriptase) in cancer cells attenuate telomerase activity [64,66].

1.5. Ribosomes in Translation and Regulation

Ribosomes present with structural dissimilarities, suggesting subspecialties based on their own binding specificities [67]. Such a heterogeneity is conducive to the rapid changes in protein expression, suggesting that ribosomal subpopulations govern the translation of their assigned mRNAs [68]. This has been evidenced in diploid studies using Saccharomyces cerevisiae, whereby the depletion of one copy of a ribosomal protein gene compromised the translation of localized ASH1 mRNA [69].
Ribosomal proteins (RPs) are instrumental in DNA repair, cellular development, and cellular differentiation [70]. Given the ribosome’s many enzymatic components, it has been referred to as the ribozyme, whereby ribosomal RNAs (rRNAs) have catalytic activity [3]. What separates eukaryotic rRNAs from their prokaryotic counterparts are their expansion segments and RP-specificity. This enhances their dual role during the translation of decoding genetic information and catalyzing peptide bond formation [71,72].
Small and large ribosomal subunits are synthesized in the nucleolus from the precursor 28S, 18S, and 5.8S rRNAs and their assembly with ribosomal proteins [73]. Following their export to the cytoplasm, these ribosomal subunits are further processed into mature 40S and 60S ribosomal subunits [74].
The activity of various ribosomal proteins has been shown to be dependent upon non-ribosomal proteins. Fuchs et al. (2011) investigated this dependence: By separately purifying actively translating and non-translating ribosomes in HeLa cells, a mass spectrometry analysis revealed glycogen synthase 1 (GYS1) to be specifically associated with polysomes [69]. As a member of the family of glycosyltransferases, this protein catalyzes the rate-limiting step during glycogen biosynthesis [75].

Ribosome Biosynthesis and Ribosomopathies

Aside from their role in viral replication, ribosomes are also believed to play a role in the cell’s stress response [76]. Ribosome biogenesis, facilitated by nutrient rich conditions and hindered under cellular stress, is a necessary component in regulating cell growth and proliferation [77]. The disruption of this process is believed to cause ribosomopathies associated with various cancers and metabolic disorders, thus initiating the p53 signaling pathway and, consequently, apoptosis [78].

2. Part II: RNA Dysfunction, Disease, and Drug Targets

RNA dysfunction leads to numerous diseases. Some dysfunctions include transcriptional errors where the mRNA formed is faulty due to mutations in the transcription factors, promotors, or enhancers [79]. An example of such a disease is the Fragile X Syndrome [79]. The failure to polyadenylate pre-mRNA could result in oculopharyngeal muscular dystrophy (OCMD), thalassemias, and neurodevelopmental disorders [79]. Alternative splicing dysregulation leads to diseases such as myotonic dystrophy. Viruses and bacteria can exploit RNA-based cellular machinery. However, suitable drug targets and therapeutics can be derived from studying malformed RNA, which will be further elaborated on in Part III of this review.

2.1. Dysregulation in Alternative Splicing and Trinucleotide-Repeat Disorders

As already described, the purpose of the spliceosome is to facilitate alternative splicing, which expands the coding repertoire of our genes. In turn, this allows for the environment-specific expression of the various possible isoforms. The ratio of isoforms becomes altered during diseases, including cancers, muscular dystrophies, and neurological diseases [80]. Calibrating the concentration of alternative splicing factors to given isoform ratios is a current area of investigation [80], as in the case of myotonic muscular dystrophy.

2.1.1. CUG-Repeats in DM1

Myotonic dystrophy type 1 (DM1) is the most prevalent form of adult-onset muscular dystrophy but lacks a disease-targeting treatment [81]. Its cause has been attributed to an extended CTG repeat tract in the 3′ UTR of the dystrophia myotonica protein kinase (DMPK) gene [82]. Consequently, a cytotoxic CUG-repeat RNA is transcribed and retained in the nucleus [83], disrupting the function of RBPs of the muscleblind-like (MBNL) and CUG-BP Elav-like (CELF) families [71,84].
The subsequent sequestration of MBNL proteins and upregulation of CELF proteins perturb alternative splicing, translation, polyadenylation, microRNA processing, and mRNA localization. Such a dysregulation of alternative splicing espouses variable disease symptoms, including the mis-splicing of muscle-specific chloride channel (CLCN1), cardiac troponin T (TNNT2) and insulin receptor (IR) mRNAs, resulting in myotonia, cardiac defects, and insulin resistance, respectively [71,72]. A schematic of this CTG repeat-mediated disruption of RNA–protein interactions is shown in Figure 2.
Berglund et al. (2016) linked levels of RNA-binding protein MBNL1 to its role in splicing events crucial for muscle, cardiovascular, and nervous system development [80]. Imbalances in MBNL1-RNA complexes, along with interactions with MBNL2 and MBNL3, can result in myotonic dystrophy. Specifically, MBNL proteins binding to the YGCY clusters of toxic RNA leads to both myotonic dystrophy types 1 and 2. Moreover, altering the YGCY clusters of the intron splicing silencer (ISS) immediately upstream allowed for the MBNL dose-dependent splicing activity to be analyzed [80]. By studying 44 DM1 patients and 11 healthy controls, it was discovered that the variability in splicing events between DM1 patients could be biomarkers for disease severity [80].
Some small molecule inhibitors of the MBNL–CUG interaction have been developed to modulate the production and stability of CUG repeats. Alternate drug targets that modulate these levels are kinases [71]. For example, the anti-sense oligonucleotide Balisforsen decreases levels of cytotoxic RNA repeats by targeting DMPK [85].
Exploiting the multistep pathogenic mechanism of DM1, Berglund et al. sought to combine the antibiotic erythromycin and the trypanicide furamidine as a multifaceted DM1 treatment [71]. Both small molecules have previously been shown to reverse mis-splicing in DM1 mouse models and are believed to work by displacing and thereby freeing MBNL from its complex with CUG RNAs. A combination treatment of each, in a transgenic mouse expressing the human skeletal actin (HSA) with 220 CUG repeats, demonstrated additive effects three times greater than either stand-alone treatment [71].

2.1.2. Microsatellite Aggregates Form Stable, Cytotoxic RNA Foci

The aforementioned short-tandem repeats are microsatellites and constitute 2–12 base pair stretches of DNA. Found in both coding and non-coding regions, these tracts are intrinsically unstable and display high variability in gene site, length, and function. The latter is not limited to regulatory roles, DNA repair, and non-canonical translation. However, when such repeats occur in the thousands, either loss- or gain-of-function mutations occur with ensuing pathology [86].
Most diseases of this etiology are inherited in a dominant fashion, and the accumulation of expanded-repeat RNAs within the cell forms complexes called RNA foci [87]. These foci gain their stability from both Watson–Crick and non-canonical base pairing, forming secondary structures such as imperfect stem-loops [88]. These include single A-form double helices for shorter repeats that accommodate U–U mismatches, while longer repeat RNAs comprise an ensemble of stem–loop structures [86,87,88].
RNA foci were first identified in DM1-diseased fibroblasts and myofibrils, which contain nuclear clumps of dystrophia myotonica protein kinase (DMPK) mRNA with the expanded CUG repeats [89]. Although RNA normally supports RNP phase separations for regulatory purposes, repeat expansions exaggerate this transition with cytotoxic consequences [34,86].

2.1.3. CCTG-Repeats and DM2

Myotonic dystrophy II (DM2) is a disorder caused by an RNA gain-of-function: An unstable CCTG repeat expansion on intron 1 of the CNBP gene in chromosome 3q21 results in the formation of hairpin structures [90]. The disorder is, in most cases, less debilitating than DM1 and lacks a clear common presentation at birth [91]. The CNBP gene codes for a zinc-finger RNA binding protein that mediates transcriptional repression [91,92].
DM2 presents possible RNA drug targets because the disorder involves the inability of MBNL1 to participate in alternative splicing [93]. Therefore, therapeutics involving small molecule treatment and anti-sense oligonucleotides are worth investigating because of their potential ability to disrupt the sequestration of MBNL1.

2.1.4. CGG-Repeats and FXTAS

Fragile X Syndrome (FXTAS) is another RNA gain-of-function disorder caused by CGG repeat expansions in the FMR1 gene on the X chromosome [94]. When the amount of CGG repeats exceed 200, the ensuing hypermethylation leads to the transcriptional silencing of the FMRP gene promoter and a consequent deficiency in this protein [95]. The inherited disorder is known to be the most prevalent cause of mild-to-severe intellectual disabilities and has high variance in presentation among such patients [94,95]. This hallmark of FXTAS manifests because the FMRP protein is normally tasked with removing interfering proteins around synapses. Thus, scant FMRP greatly diminishes synaptic plasticity [96].

2.1.5. CTG*CAG-Repeats and SCA8

SCA8 is a disease involving slowly intensifying ataxia that occurs when the two overlapping genes, ATXN8OS/ATXN8, include abnormal (CTG*CAG)n repeat expansions identified through a proband, a type of molecular genetic testing [97,98]. Like the previous disorders discussed in this section, SCA8 expansion occurs in the non-coding region, resulting in toxic RNA that can disrupt normal RNA functions by sequestering functional RNA [99]. One result of overexpression in the SCA8 mutation is the production of polyglutamine proteins and CUG/CAG transcripts [100]. This leads to the sequestration of the MBNL1 gene, thereby creating motor deficits [97,101].

2.2. Viruses

Viruses infect hosts by evading their immune response and hijacking replication machinery, namely, ribosomes and translation factors, in order to synthesize viral proteins [102].
An invading virus can have a genome comprising either DNA or RNA surrounded by a protein shell forming a capsid [103]. Normally, cellular mRNAs are transcribed in the nucleus, facilitated at their 5′ end by a 7-methyl-guanosine (m7G) cap binding to the cap-binding protein eIF4E (eukaryotic translation initiation factor 4E) [103]. Translation can occur cap-dependently or cap-independently, and the latter is often employed by viral mRNAs [76].

2.2.1. Cap-Independent Translation Targets Ribosome

Some viruses have their own RNA structural elements upstream of the open reading frame. Such internal ribosomal entry sites (IRESs) operate independent of the 5′ methyl cap and are, thus, referred to as cap-independent translation [104]. This direct means of targeting ribosomes and translation factors alters both their availability and associated posttranslational modifications [105]. The latter was made possible by the viral IRES’s affinity to certain ribosomal proteins such as RACK1 [105].
RACK1 is a 40S ribosomal subunit protein required for the replication of certain viruses [105]. Its function during IRES-mediated translation appears distinct from its eS25 counterpart, as illustrated by the inability of ribosomes lacking eS25 to bind to the IRES and the consequent translational inhibition [106]. However, a loss of RACK1 did not have the same effect, and the wide array of RACK1 functions may impart both proviral and antiviral outcomes [105]. The dependency of ribosomal recruitment to the IRES is illustrated in Figure 3.
Specifically, Cryo-EM places RPS25 on the 40S subunit near mRNA entry, and Fuchs et al. (2015) used fluorescent labeling to study the RPS25 recruitment to an HCV IRES [107]. Introduction of their RPS25-SNAP fusion protein within RPS25 KO cells restored translation. Moreover, dynamic FRET revealed flexibility in this protein and its interaction with the HCV IRES [107].

2.2.2. Viruses Target Post-Translational Modifications

Post-translational modifications encompass nature’s repertoire of 20 amino acids, primarily eliciting methylation, phosphorylation, and acetylation at the amino acid chain’s N- or C-termini. These modifications to host cell proteins and factors can be imparted by viral enzymes, thereby disrupting protein folding and cell signaling. For instance, the acetylation of eL24 at K27 was found to decrease the association of the modified eL24 with polysomes [105]. Moreover, a comparison of 31 ribosomal proteins by Yu et al. (2005) revealed inconsistent modifications between IRES-bound and native 40S ribosomal subunits [106,107].
Viruses exploit host post-translational modifications by various methods including ubiquitination and less common approaches, such as ISGylation, SUMOylation, and ADP ribosylation [108]. The occurrences of these processes attenuating and promoting viral infections have been reported. An example of the former is the ubiquitination of Dengue (DENV) NS3, which tags the virus for proteolytic degradation [109]. The latter involves ubiquitination, as seen with the Ebola virus protein VP35; the indiscriminate ubiquitination of the viral polymerase subunit inadvertently enhances its enzymatic activity [110].
The cap-independent method employs the virus’ IRES within the 5′ untranslated region of their genome. However, the mechanistic details of this IRES-mediated translation are unknown, something compounded by the evolution of IRES across different viruses. One commonality shared among viruses is their need for the ribosomal protein eS25 (RPS25) in order to initiate translation [106,111].

2.2.3. tRNA Modifications in Antibiotic Design

Understanding the mechanisms of translational modifications offers a means of antibiotic design [112,113]. One site of particular interest along the codon–anticodon complex is position-34, also known as the wobble position [114,115]. As the third base pair along the three-letter codon, the stability of the first two base pairs affords some flexibility [116], thereby tolerating non-canonical base pairs [117] essential for advancing the ribosome properly along the reading frame [118]. The absence of these modifications in humans can induce frameshift mutations [119], but disrupting bacterial translation machinery provides an antimicrobial approach. Bacterial tRNA-2-selenouridine synthase (SelU) is a drug target responsible for derivatizing 2-uridine of this anticodon position with either a geranyl group or selenium [118].

2.2.4. SelU-Mediated Geranylation and Selenation of tRNA Affects Translational Fidelity

These two modifications are centered around the role of selenium, namely, that of 2-selenouridine in tRNA anticodons [120]. Most seleno-tRNAs code for lysine, glutamate, and glutamine with their isoacceptors containing 5-methylaminomethyl 2-selenouridine in the wobble position of their anticodons [120]. This process begins as the original tRNA transcript’s uridine is modified with sulfur and yields 2-selenouridine upon the substitution of sulfur with selenium, as illustrated in Figure 4 [121].
Upon treating the RNA isolated from bacteria with nuclease P1, Dumelin et al. (2012) found eight species that contained the modified nucleobases mnm5s2U and cmnm5s2U [122]. Geranyl modifications, specifically at 2-thiouridine, were found in certain bacterial species but not in others, and eukaryotic RNA and the presence of the gene sufY was correlated with geranylation across species [121]. The addition of this isoprenoid also has been attributed to SelU and often occurs in tRNA containing a carboxymethylaminomethyl (cmnmn) at 5-uridine, suggesting a coordination of these 5- and 2-uridine modifications through multienzyme biosynthetic pathways [122]. Of particular interest is the rhodanese homology domain of SelU, given the utility, in such motifs, in sulfurtransferase catalysis [122,123]. These conserved residues may provide insights into the rational design of an antibiotic.
In addition, aminoacyl tRNA synthetases have been proposed as potential drug targets for malaria and tuberculosis [124]. These enzymes catalyze the aminoacylation of tRNAs with cognate amino acids [125]. Aspartyl-tRNA synthetase has been explored as a potential drug target because of its structural differences from that of humans [126].
Several of the 36 Plasmodium falciparum aminoacyl-tRNA synthetases have been investigated as drug targets, including prolyl-tRNA synthetase (PRS) [126]. Jain et al. (2014) obtained a crystal structure of this parasite’s prolyl-tRNA synthetase and found some contrasts with the human glutamyl-prolyl-tRNA synthetase [127]. The former’s structure in 3 Å resolution revealed an open state upon ATP-binding and an asymmetric dimer interface, owing to a flexible segment extending into these ATP-binding pockets [127]. Structural analyses have revealed key differences between parasite and human enzymes that can be exploited for drug design.

3. Part III: RNA as a Therapeutic

The biological versatility of RNA, including its interactions with nucleic acids and proteins, makes it suitable as a novel class of therapeutics. Several RNA-based treatments have been approved, including anti-sense oligonucleotides (ASOs), RNA interference (RNAi), and CRISPR/Cas gene therapies [128,129]. Other approaches under investigation are RNA aptamers and the rational design of small molecule mimetics [130,131].
RNA folding patterns impart further functionality enzymatically and during translation.
Single-stranded RNA has been folded into varying conformations, performing biological tasks [132]. RNA can form distinct secondary and tertiary structures through complex intramolecular base-pairing and complementary interactions between different strands [133].

3.1. ASOs and siRNAs Silence Genes

Antisense oligonucleotides are short synthetic strands of DNA oligonucleotides designed to bind RNA and induce gene silencing [134,135]. Both in vitro and in vivo studies have demonstrated the efficacy of this approach [136,137]. ASOs bind to RNA in an antisense fashion, forming an RNA/DNA heteroduplex. The resulting double-stranded region of RNA is now a substrate to RNase H–an enzyme that removes RNA strands [138]. ASOs are also effective at manipulating alternative splicing through either exon skipping or exon inclusions [139]. The former can be useful for correcting frameshift mutations [140], whereby an ASO binds its target pre-mRNA and corrects the mutation, producing a short functional protein [141]. Conversely, exon exclusion methods involve ASOs attaching to the pre-mRNA to prevent spliceosomes and splicing factors from binding [142].
Despite the promise of gene therapies targeting disease at their source rather than upstream pathways [136,137], greater headway needs to be made prior to ASOs becoming a mainstay treatment plan.
RNA interference (RNAi) is another approach to silence disease-related genes, employing siRNAs [143]. The process involves introducing double-stranded siRNAs into cells, where they are incorporated into the RNA-induced silencing protein complex (RISC) [144]. Next, the strands are separated, and the guide strand ushers the RISC to its mRNA target for enzymatic digestion [145]. This technique can be applied to genetic disorders such as hypercholesterolemia, in which the drug Inclisiran targets PCSK9 mRNA to lower LDL cholesterol levels through the recycling of LDL receptors [146,147].
Considerations in the delivery of such RNA therapeutics include maintaining its structural integrity while routing its intracellular target. The chemical modifications of ASOs, such as the thiolation of the phosphate backbone at non-binding sites, adding 2′O-methyl/methoxyethyl groups [148], and employing gapmers, which also imparts stability by using DNA as central segments, facilitate cellular uptake [149].
Two of the three FDA-approved siRNA therapeutics are N-Acetylgalactosamine (GalNac)-based, meaning amino-sugar derivatives are conjugated to these siRNAs, designating them for the liver [150]. There, Givosiran alleviates acute hepatic porphyria and Inclisiran reduces cholesterol levels [151]. The other FDA-approved therapeutic of this class, Patisiran, uses a lipid nanoparticle (LNP) delivery method to treat hereditary transthyretin amyloidosis [152].

3.2. CRISPR/Cas9 and Gene Editing

CRISPR/Cas is a popular genome editing tool that can alter RNA as well as DNA [153]. RNA is the preferrable target because it bypasses the ethical concerns of permanently altering the genetic code [154]. Zhang et al. (2019) developed the RNA base-pair editing systems, REPAIR, which enables A to I(G) replacement, and RESCUE, which replaces C with U [154,155].
Therapeutic attempts at attenuating toxic RNA in DM1 mouse models include the CRISPR/Cas9-mediated genome editing of the expanded CTG repeats [156]. Another approach inserts polyadenylation signals upstream of the CTG repeats to reduce the expression of CUG-repeat-containing transcripts [157].
CRISPR/Cas can also be useful in fighting viral infections. For example, PAC-MAN is a CRISPR/Cas13 system has shown promise in targeting and destroying RNA in influenza and coronaviruses [158]. Similarly, CasRx is also an artificial enzyme that can target RNA; however, it can make specific cuts to RNA instead of outrightly destroying RNA [159,160].
The modes of delivering this gene editing complex include viral vectors [161], exosomes [162], various nanoparticles, and lipid nanoparticles (LNPs) [163], in which the Cas mRNA and gRNA are encapsulated to facilitate delivery [164]. For example, Pfizer’s COVID-19 vaccine is an mRNA administered within an ionizable lipid known as ALC-0315 [165].

3.3. Small Molecules as Inhibitors

Small molecules are readily finetuned through derivatization with various functional groups, and their small size leads them to targeting biomolecules implicated in RNA-based diseases [166,167]. RNA itself has multiple levels of structural organization, thereby affording a high specificity to a properly tailored small molecule [168]. Consequently, small molecules can disrupt RNA–protein interactions to prevent metastasis [169] or inhibit bacterial translation to stop infection, as referenced earlier with regards to SelU. Examples of the former include targeting splicing factors, such as SF3B1, PRPF8, and U2AF1, which could potentially correct aberrant splicing in cancer cells, thereby preventing further oncological effects [170,171,172].
Currently, the drugs Risdiplam, Branaplam, and Indisulam, among others, are under consideration for their potential in cancer treatments [173,174]. Indisulam is especially promising due to its ability to kill cancer cells directly by degrading the splicing factor, RBM39 [174].
The convenience of the oral administration and shorter half-lives of small molecules, as well as their longer shelf-life, lower manufacturing costs, and conduciveness to traditional high-throughput screening, highlights their therapeutic potential [175].
Aiding in the delivery of small molecules into cells or specific tissue-types are polymeric nanoparticles, lipid-based systems [176], and GalNAc, which are tailored to the asialoglycoprotein receptors (ASGPR) of liver hepatocytes [177,178].

3.4. RNA Aptamers Offer Variability

While the majority of this review has, thus far, focused on RNA therapeutics as targeting genes or other nucleic acid targets in an antisense fashion, these are not the limits of their potential. Most notably, RNA aptamers are a promising therapeutic that can bind not only to nucleic acids, but also to organic compounds, proteins, and even whole cells [130,179]. They can target a variety of molecules with high affinity and specificity in a similar fashion to antibodies. Furthermore, by modifying the backbone or nucleic acids, one can manipulate an aptamer’s half-life, affinity, and even its function [180,181].
RNA aptamers work by intramolecularly hybridizing into a unique 3-dimensional structure, depending on the nucleic acid sequence [182]. Once formed, they can bind to their target and induce a variety of effects, such as inhibition or marking for destruction [183]. Due to their immense variability in structure, aptamers for a particular target must be selected for through the systematic evolution of ligands by exponential enrichment (SELEX) [184]. While there are many different types, the basic idea is that a large pool of RNA oligonucleotides are allowed to fold into a defined 3-dimensional structure, after which they are exposed to the target of interest [185,186]. The subset of oligonucleotides that bind is purified and amplified as potential aptamers [186]. This process is illustrated schematically in Figure 5. Because the 3-dimensional structures of aptamers vary with nucleic acid sequences, it is theoretically possible to design an aptamer against any molecular target [130,187,188].
Currently, there is only one FDA approved aptamer, under the name Pegnatib, to treat neovascularization, which is responsible for 90% of vision loss in age-related macular degeneration (AMD) [189,190]. Neovascularization is caused by the production of too much of the vascular endothelial growth factor (VEGF), which increases the proliferation of new blood vessels in the choroid, leading to vision loss [191]. Pregnatib acts as a potent inhibitor to VEGF165, the isoform primarily responsible for neovascularization, and has been shown to slow, and even prevent, further visual damage [192]. Other aptamers that act to inhibit alternate isoforms of VEGF responsible for more types of vision loss, such as that in diabetes, are being developed [193,194]. Many others could act as potential viral or cancer therapeutics. Pegnatib is the only RNA aptamer approved by the FDA because many of the same challenges of other RNA therapeutics, such as degradation by nucleases and cell permeability, apply to aptamers [195]. However, aptamers show incredible therapeutic potential [196].
The delivery is consistent with other oligonucleotide therapies, with the advantage of using SELEX for optimization of modifications, of which 2′-O methyl, 2′-fluoro, and phosphorothioate-linkage modifications proved efficacious [197,198]. Moreover, the protection from nucleases is afforded by enclosing the modified oligonucleotide within liposomes or LNPs [199]. Alternate methods of delivery include viral vectors [200], polymeric carriers [201], and metallic nanoparticles [202].

4. Conclusions

The evolution of RNA from its role as life’s original catalyst and genetic material into a behemoth of RNA-types and protein-coding processes has opened an avenue to its therapeutic development. This may entail targeting certain enzymes involved in splicing, RNA-folding, or post-translational modifications, or employing RNA itself as a therapeutic to modulate gene expression. RNA aptamers may even act as chemical antibodies. The flexibility of this molecule offers advantages over the traditional small molecule approach to pharmaceuticals, including their nucleotide-mediated selectivity and variety of applications by modification. This has implications in modulating gene expression and the treatment of a wide variety diseases.
Investigations into treating these disorders have illustrated the breadth of RNA-based therapeutic approaches at our disposal once the relevant biomarkers and mechanisms are understood. These include gene editing, the polyadenylation-mediated reduction in the expression of CUG-repeat-containing transcripts, the degradation of CUG-repeat RNA using antisense oligonucleotides or siRNAs, the targeting of small molecule inhibitors to the MBNL–CUG RNA interaction, and combinatorial antibody therapy.
The same repertoire of techniques can be applied to other pathologies, especially those pertaining to ribosome biosynthesis and its role in translation. The former is ramped up during cancer, rendering its mechanism of production an appealing means to deny cancerous cells a necessary commodity. Likewise, viruses commandeer the latter by imparting post-translational modifications on host-proteins, making the plethora of ribosomal and associated proteins plausible targets. Further, the pattern of RNA-folds and tRNA-modifications necessary to their functions can also provide insights into a therapeutic’s rational design.

Author Contributions

Conceptualization—S.J.D. and J.S.; writing—original draft preparation, S.J.D.; writing—review and editing, S.J.D., H.C., R.P.D. and J.S.; funding acquisition—J.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by National Institutes of Health, grant number 1R01GM143749-22A1.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We are grateful to NIH (GM143749) to J.S. for the financial support.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Alberts, B.; Johnson, A.; Lewis, J.; Raff, M.; Roberts, K.; Walter, P. The RNA world and the origins of life. In Molecular Biology of the Cell, 4th ed.; Garland Science: New York, NY, USA, 2002. [Google Scholar]
  2. Noller, H.F. Evolution of protein synthesis from an RNA world. Cold Spring Harb. Perspect. Biol. 2012, 4, a003681. [Google Scholar] [CrossRef] [PubMed]
  3. Cech, T.R. The ribosome is a ribozyme. Science 2000, 289, 878–879. [Google Scholar] [CrossRef] [PubMed]
  4. Lilley, D.M. The ribosome functions as a ribozyme. Chembiochem 2001, 2, 31–35. [Google Scholar] [CrossRef] [PubMed]
  5. Steitz, T.A.; Moore, P.B. RNA, the first macromolecular catalyst: The ribosome is a ribozyme. Trends Biochem. Sci. 2003, 28, 411–418. [Google Scholar] [CrossRef]
  6. Tagami, S.; Li, P. The origin of life: RNA and protein co-evolution on the ancient earth. Dev. Growth Differ. 2023, 65, 167–174. [Google Scholar] [CrossRef]
  7. Synak, J.; Rybarczyk, A.; Blazewicz, J. Multi-agent approach to sequence structure simulation in the RNA World hypothesis. PLoS ONE 2020, 15, e0238253. [Google Scholar] [CrossRef]
  8. Lazcano, A.; Bada, J.L. The 1953 Stanley L. Miller experiment: Fifty years of prebiotic organic chemistry. Orig. Life Evol. Biosph. 2003, 33, 235–242. [Google Scholar] [CrossRef]
  9. Fine, J.L.; Pearlman, R.E. On the origin of life: An RNA-focused synthesis and narrative. RNA 2023, 29, 1085–1098. [Google Scholar] [CrossRef]
  10. da Silva, J.A. From the RNA world to the RNA/protein world: Contribution of some riboswitch-binding species? J. Theor. Biol. 2015, 370, 197–201. [Google Scholar] [CrossRef]
  11. Spirov, A. Evolution of the RNA world: From signals to codes. Biosystems 2023, 234, 105043. [Google Scholar] [CrossRef]
  12. Nelson, J.W.; Breaker, R.R. The lost language of the RNA World. Sci. Signal. 2017, 10, eaam8812. [Google Scholar] [CrossRef] [PubMed]
  13. Kaikkonen, M.U.; Lam, M.T.; Glass, C.K. Non-coding RNAs as regulators of gene expression and epigenetics. Cardiovasc. Res. 2011, 90, 430–440. [Google Scholar] [CrossRef] [PubMed]
  14. Serganov, A.; Nudler, E. A decade of riboswitches. Cell 2013, 152, 17–24. [Google Scholar] [CrossRef] [PubMed]
  15. Tyrrell, J.; McGinnis, J.L.; Weeks, K.M.; Pielak, G.J. The cellular environment stabilizes adenine riboswitch RNA structure. Biochemistry 2013, 52, 8777–8785. [Google Scholar] [CrossRef]
  16. Beisel, C.L.; Smolke, C.D. Design principles for riboswitch function. PLoS Comput. Biol. 2009, 5, e1000363. [Google Scholar] [CrossRef]
  17. Breaker, R.R. The biochemical landscape of riboswitch ligands. Biochemistry 2022, 61, 137–149. [Google Scholar] [CrossRef]
  18. Garst, A.D.; Edwards, A.L.; Batey, R.T. Riboswitches: Structures and mechanisms. Cold Spring Harb. Perspect. Biol. 2011, 3, a003533. [Google Scholar] [CrossRef]
  19. Breaker, R.R. Riboswitches and the RNA world. Cold Spring Harb. Perspect. Biol. 2012, 4, a003566. [Google Scholar] [CrossRef]
  20. Liberman, J.A.; Wedekind, J.E. Riboswitch structure in the ligand-free state. Wiley Interdiscip. Rev. RNA 2012, 3, 369–384. [Google Scholar] [CrossRef]
  21. Savinov, A.; Perez, C.F.; Block, S.M. Single-molecule studies of riboswitch folding. Biochim. Biophys. Acta (BBA)-Gene Regul. Mech. 2014, 1839, 1030–1045. [Google Scholar] [CrossRef]
  22. Richards, J.; Belasco, J.G. Riboswitch control of bacterial RNA stability. Mol. Microbiol. 2021, 116, 361–365. [Google Scholar] [CrossRef] [PubMed]
  23. Wilson, R.C.; Smith, A.M.; Fuchs, R.T.; Kleckner, I.R.; Henkin, T.M.; Foster, M.P. Tuning riboswitch regulation through conformational selection. J. Mol. Biol. 2011, 405, 926–938. [Google Scholar] [CrossRef] [PubMed]
  24. Reining, A.; Nozinovic, S.; Schlepckow, K.; Buhr, F.; Fürtig, B.; Schwalbe, H. Three-state mechanism couples ligand and temperature sensing in riboswitches. Nature 2013, 499, 355–359. [Google Scholar] [CrossRef] [PubMed]
  25. Warhaut, S.; Mertinkus, K.R.; Höllthaler, P.; Fürtig, B.; Heilemann, M.; Hengesbach, M.; Schwalbe, H. Ligand-modulated folding of the full-length adenine riboswitch probed by NMR and single-molecule FRET spectroscopy. Nucleic Acids Res. 2017, 45, 5512–5522. [Google Scholar] [CrossRef]
  26. Bains, J.K.; Blechar, J.; de Jesus, V.; Meiser, N.; Zetzsche, H.; Fürtig, B.; Schwalbe, H.; Hengesbach, M. Combined smFRET and NMR analysis of riboswitch structural dynamics. Methods 2019, 153, 22–34. [Google Scholar] [CrossRef]
  27. Li, S.; Breaker, R.R. Eukaryotic TPP riboswitch regulation of alternative splicing involving long-distance base pairing. Nucleic Acids Res. 2013, 41, 3022–3031. [Google Scholar] [CrossRef]
  28. Cheah, M.T.; Wachter, A.; Sudarsan, N.; Breaker, R.R. Control of alternative RNA splicing and gene expression by eukaryotic riboswitches. Nature 2007, 447, 497–500. [Google Scholar] [CrossRef]
  29. Breaker, R.R. Riboswitches and translation control. Cold Spring Harb. Perspect. Biol. 2018, 10, a032797. [Google Scholar] [CrossRef]
  30. Wachter, A. Riboswitch-mediated control of gene expression in eukaryotes. RNA Biol. 2010, 7, 67–76. [Google Scholar] [CrossRef]
  31. Yamauchi, T.; Miyoshi, D.; Kubodera, T.; Ban, M.; Nishimura, A.; Sugimoto, N. Riboswitches for enhancing target gene expression in eukaryotes. ChemBioChem 2008, 9, 1040–1043. [Google Scholar] [CrossRef]
  32. Lamond, A.I. The spliceosome. Bioessays 1993, 15, 595–603. [Google Scholar] [CrossRef] [PubMed]
  33. Brenner, S.; Miller, J.H. Brenner’s Encyclopedia of Genetics; Elsevier Science: Amsterdam, The Netherlands, 2014. [Google Scholar]
  34. Zaug, A.J.; Cech, T.R. Self-splicing RNA and an RNA enzyme in Tetrahymena. J. Protozool. 1987, 34, 416–417. [Google Scholar] [CrossRef]
  35. Olthof, A.M.; White, A.K.; Mieruszynski, S.; Doggett, K.; Lee, M.F.; Chakroun, A.; Aleem, A.K.A.; Rousseau, J.; Magnani, C.; Roifman, C.M.; et al. Disruption of exon-bridging interactions between the minor and major spliceosomes results in alternative splicing around minor introns. Nucleic Acids Res. 2021, 49, 3524–3545. [Google Scholar] [CrossRef] [PubMed]
  36. Bajan, S.; Hutvagner, G. RNA-based therapeutics: From antisense oligonucleotides to miRNAs. Cells 2020, 9, 137. [Google Scholar] [CrossRef]
  37. Will, C.L.; Lührmann, R. Spliceosome structure and function. Cold Spring Harb. Perspect. Biol. 2011, 3, a003707. [Google Scholar] [CrossRef]
  38. Ares, M., Jr.; Weiser, B. Rearrangement of snRNA structure during assembly and function of the spliceosome. Prog. Nucleic Acid Res. Mol. Biol. 1995, 50, 131–159. [Google Scholar]
  39. Atkins, J.F.; Gesteland, R.F.; Cech, T. (Eds.) RNA Worlds: From Life’s Origins to Diversity in Gene Regulation; Cold Spring Harbor Laboratory Press: New York, NY, USA, 2011; pp. 483–503. [Google Scholar]
  40. Xu, S.-M.; Curry-Hyde, A.; Sytnyk, V.; Janitz, M. RNA polyadenylation patterns in the human transcriptome. Gene 2022, 816, 146133. [Google Scholar] [CrossRef]
  41. Antson, A.A. Single stranded RNA binding proteins. Curr. Opin. Struct. Biol. 2000, 10, 87–94. [Google Scholar] [CrossRef]
  42. Wu, Q.; Bazzini, A.A. Translation and mRNA stability control. Annu. Rev. Biochem. 2023, 92, 227–245. [Google Scholar] [CrossRef]
  43. Radhakrishnan, A.; Green, R. Connections underlying translation and mRNA stability. J. Mol. Biol. 2016, 428, 3558–3564. [Google Scholar] [CrossRef]
  44. Wu, X.; Brewer, G. The regulation of mRNA stability in mammalian cells: 2.0. Gene 2012, 500, 10–21. [Google Scholar] [CrossRef] [PubMed]
  45. Mitchell, P.; Tollervey, D. mRNA stability in eukaryotes. Curr. Opin. Genet. Dev. 2000, 10, 193–198. [Google Scholar] [CrossRef] [PubMed]
  46. Decker, C.J.; Parker, R. Mechanisms of mRNA degradation in eukaryotes. Trends Biochem. Sci. 1994, 19, 336–340. [Google Scholar] [CrossRef] [PubMed]
  47. Houseley, J.; Tollervey, D. The many pathways of RNA degradation. Cell 2009, 136, 763–776. [Google Scholar] [CrossRef]
  48. Goldstrohm, A.C.; Wickens, M. Multifunctional deadenylase complexes diversify mRNA control. Nat. Rev. Mol. Cell Biol. 2008, 9, 337–344. [Google Scholar] [CrossRef]
  49. Funakoshi, Y.; Doi, Y.; Hosoda, N.; Uchida, N.; Osawa, M.; Shimada, I.; Tsujimoto, M.; Suzuki, T.; Katada, T.; Hoshino, S.-I. Mechanism of mRNA deadenylation: Evidence for a molecular interplay between translation termination factor eRF3 and mRNA deadenylases. Genes Dev. 2007, 21, 3135–3148. [Google Scholar] [CrossRef]
  50. Paillard, L.; Omilli, F.; Legagneux, V.; Bassez, T.; Maniey, D.; Osborne, H. EDEN and EDEN-BP, a cis element and an associated factor that mediate sequence-specific mRNA deadenylation in Xenopus embryos. EMBO J. 1998, 17, 278–287. [Google Scholar] [CrossRef]
  51. Tharun, S.; Parker, R. Targeting an mRNA for decapping: Displacement of translation factors and association of the Lsm1p–7p complex on deadenylated yeast mRNAs. Mol. Cell 2001, 8, 1075–1083. [Google Scholar] [CrossRef]
  52. Bouveret, E.; Rigaut, G.; Shevchenko, A.; Wilm, M.; Séraphin, B. A Sm-like protein complex that participates in mRNA degradation. EMBO J. 2000, 19, 1661–1671. [Google Scholar] [CrossRef]
  53. Audebert, L.; Feuerbach, F.; Zedan, M.; Schürch, A.P.; Decourty, L.; Namane, A.; Permal, E.; Weis, K.; Badis, G.; Saveanu, C. RNA degradation triggered by decapping is largely independent of initial deadenylation. EMBO J. 2024, 43, 6496–6525. [Google Scholar] [CrossRef]
  54. Shaw, G.; Kamen, R. A conserved AU sequence from the 3′ untranslated region of GM-CSF mRNA mediates selective mRNA degradation. Cell 1986, 46, 659–667. [Google Scholar] [CrossRef] [PubMed]
  55. Zubiaga, A.M.; Belasco, J.G.; Greenberg, M.E. The nonamer UUAUUUAUU is the key AU-rich sequence motif that mediates mRNA degradation. Mol. Cell. Biol. 1995, 15, 2219–2230. [Google Scholar] [CrossRef] [PubMed]
  56. Chen, C.-Y.A.; Shyu, A.-B. AU-rich elements: Characterization and importance in mRNA degradation. Trends Biochem. Sci. 1995, 20, 465–470. [Google Scholar] [CrossRef] [PubMed]
  57. Park-Lee, S.; Kim, S.; Laird-Offringa, I.A. Characterization of the interaction between neuronal RNA-binding protein HuD and AU-rich RNA. J. Biol. Chem. 2003, 278, 39801–39808. [Google Scholar] [CrossRef]
  58. Wang, X.; Hall, T.M.T. Structural basis for recognition of AU-rich element RNA by the HuD protein. Nat. Struct. Biol. 2001, 8, 141–145. [Google Scholar] [CrossRef]
  59. Peng, S.S.Y.; Chen, C.Y.A.; Xu, N.; Shyu, A.B. RNA stabilization by the AU-rich element binding protein, HuR, an ELAV protein. EMBO J. 1998, 17, 3461–3470. [Google Scholar] [CrossRef]
  60. Plass, M.; Rasmussen, S.H.; Krogh, A. Highly accessible AU-rich regions in 3′ untranslated regions are hotspots for binding of regulatory factors. PLoS Comput. Biol. 2017, 13, e1005460. [Google Scholar] [CrossRef]
  61. Vakalopoulou, E.; Schaack, J.; Shenk, T. A 32-kilodalton protein binds to AU-rich domains in the 3′ untranslated regions of rapidly degraded mRNAs. Mol. Cell. Biol. 1991, 11, 3355–3364. [Google Scholar]
  62. Hong, J.; Yun, C.-O. Telomere gene therapy: Polarizing therapeutic goals for treatment of various diseases. Cells 2019, 8, 392. [Google Scholar] [CrossRef]
  63. Zhu, Y.; Liu, X.; Ding, X.; Wang, F.; Geng, X. Telomere and its role in the aging pathways: Telomere shortening, cell senescence and mitochondria dysfunction. Biogerontology 2019, 20, 1–16. [Google Scholar] [CrossRef]
  64. Samper, E.; Flores, J.M.; Blasco, M.A. Restoration of telomerase activity rescues chromosomal instability and premature aging in Terc−/− mice with short telomeres. EMBO Rep. 2001, 2, 800–807. [Google Scholar] [CrossRef] [PubMed]
  65. Rudolph, K.L.; Chang, S.; Millard, M.; Schreiber-Agus, N.; DePinho, R.A. Inhibition of experimental liver cirrhosis in mice by telomerase gene delivery. Science 2000, 287, 1253–1258. [Google Scholar] [CrossRef] [PubMed]
  66. Kosciolek, B.A.; Kalantidis, K.; Tabler, M.; Rowley, P.T. Inhibition of telomerase activity in human cancer cells by RNA interference. Mol. Cancer Ther. 2003, 2, 209–216. [Google Scholar]
  67. Schierholz, L.; Brown, C.R.; Helena-Bueno, K.; Uversky, V.N.; Hirt, R.P.; Barandun, J.; Melnikov, S.V. A conserved ribosomal protein has entirely dissimilar structures in different organisms. Mol. Biol. Evol. 2024, 41, msad254. [Google Scholar] [CrossRef]
  68. Shi, Z.; Fujii, K.; Kovary, K.M.; Genuth, N.R.; Röst, H.L.; Teruel, M.N.; Barna, M. Heterogeneous ribosomes preferentially translate distinct subpools of mRNAs genome-wide. Mol. Cell 2017, 67, 71–83. [Google Scholar] [CrossRef]
  69. Fuchs, G.; Diges, C.; Kohlstaedt, L.A.; Wehner, K.A.; Sarnow, P. Proteomic analysis of ribosomes: Translational control of mRNA populations by glycogen synthase GYS1. J. Mol. Biol. 2011, 410, 118–130. [Google Scholar] [CrossRef]
  70. Kang, J.; Brajanovski, N.; Chan, K.T.; Xuan, J.; Pearson, R.B.; Sanij, E. Ribosomal proteins and human diseases: Molecular mechanisms and targeted therapy. Signal Transduct. Target. Ther. 2021, 6, 323. [Google Scholar] [CrossRef]
  71. Jenquin, J.R.; Yang, H.; Huigens, R.W., III; Nakamori, M.; Berglund, J.A. Combination treatment of erythromycin and furamidine provides additive and synergistic rescue of mis-splicing in myotonic dystrophy type 1 models. ACS Pharmacol. Transl. Sci. 2019, 2, 247–263. [Google Scholar] [CrossRef]
  72. Wang, E.T.; Ward, A.J.; Cherone, J.M.; Giudice, J.; Wang, T.T.; Treacy, D.J.; Lambert, N.J.; Freese, P.; Saxena, T.; Cooper, T.A.; et al. Antagonistic regulation of mRNA expression and splicing by CELF and MBNL proteins. Genome Res. 2015, 25, 858–871. [Google Scholar] [CrossRef]
  73. Cooper, G.M.; Ganem, D. The cell: A molecular approach. Nat. Med. 1997, 3, 1042. [Google Scholar]
  74. Boulon, S.; Westman, B.J.; Hutten, S.; Boisvert, F.-M.; Lamond, A.I. The nucleolus under stress. Mol. Cell 2010, 40, 216–227. [Google Scholar] [CrossRef] [PubMed]
  75. Ullman, J.C.; Mellem, K.T.; Xi, Y.; Ramanan, V.; Merritt, H.; Choy, R.; Gujral, T.; Young, L.E.; Blake, K.; Tep, S.; et al. Small-molecule inhibition of glycogen synthase 1 for the treatment of Pompe disease and other glycogen storage disorders. Sci. Transl. Med. 2024, 16, eadf1691. [Google Scholar] [CrossRef] [PubMed]
  76. Vind, A.C.; Genzor, A.V.; Bekker-Jensen, S. Ribosomal stress-surveillance: Three pathways is a magic number. Nucleic Acids Res. 2020, 48, 10648–10661. [Google Scholar] [CrossRef] [PubMed]
  77. Jiao, L.; Liu, Y.; Yu, X.-Y.; Pan, X.; Zhang, Y.; Tu, J.; Song, Y.-H.; Li, Y. Ribosome biogenesis in disease: New players and therapeutic targets. Signal Transduct. Target. Ther. 2023, 8, 15. [Google Scholar] [CrossRef] [PubMed]
  78. Wang, W.; Nag, S.; Zhang, X.; Wang, M.; Wang, H.; Zhou, J.; Zhang, R. Ribosomal proteins and human diseases: Pathogenesis, molecular mechanisms, and therapeutic implications. Med. Res. Rev. 2015, 35, 225–285. [Google Scholar] [CrossRef]
  79. Cooper, T.A.; Wan, L.; Dreyfuss, G. RNA and disease. Cell 2009, 136, 777–793. [Google Scholar] [CrossRef]
  80. Wagner, S.D.; Struck, A.J.; Gupta, R.; Farnsworth, D.R.; Mahady, A.E.; Eichinger, K.; Thornton, C.A.; Wang, E.T.; Berglund, J.A. Dose-dependent regulation of alternative splicing by MBNL proteins reveals biomarkers for myotonic dystrophy. PLoS Genet. 2016, 12, e1006316. [Google Scholar] [CrossRef]
  81. Vydra, D.G.; Rayi, A. Myotonic dystrophy. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  82. Huichalaf, C.; Sakai, K.; Jin, B.; Jones, K.; Wang, G.; Schoser, B.; Schneider-Gold, C.; Sarkar, P.; Pereira-Smith, O.M.; Timchenko, N.; et al. Expansion of CUG RNA repeats causes stress and inhibition of translation in myotonic dystrophy 1 (DM1) cells. FASEB J. 2010, 24, 3706–3719. [Google Scholar] [CrossRef]
  83. Brook, J.D.; McCurrach, M.E.; Harley, H.G.; Buckler, A.J.; Church, D.; Aburatani, H.; Hunter, K.; Stanton, V.P.; Thirion, J.-P.; Hudson, T.; et al. Molecular basis of myotonic dystrophy: Expansion of a trinucleotide (CTG) repeat at the 3′ end of a transcript encoding a protein kinase family member. Cell 1992, 68, 799–808. [Google Scholar] [CrossRef]
  84. Lin, X.; Miller, J.W.; Mankodi, A.; Kanadia, R.N.; Yuan, Y.; Moxley, R.T.; Swanson, M.S.; Thornton, C.A. Failure of MBNL1-dependent post-natal splicing transitions in myotonic dystrophy. Hum. Mol. Genet. 2006, 15, 2087–2097. [Google Scholar] [CrossRef]
  85. Thornton, C.A.; Moxley, R.T.; Eichinger, K.; Heatwole, C.; Mignon, L.; Arnold, W.D.; Ashizawa, T.; Day, J.W.; Dent, G.; Tanner, M.K.; et al. Antisense oligonucleotide targeting DMPK in patients with myotonic dystrophy type 1: A multicentre, randomised, dose-escalation, placebo-controlled, phase 1/2a trial. Lancet Neurol. 2023, 22, 218–228. [Google Scholar] [CrossRef] [PubMed]
  86. Malik, I.; Kelley, C.P.; Wang, E.T.; Todd, P.K. Molecular mechanisms underlying nucleotide repeat expansion disorders. Nat. Rev. Mol. Cell Biol. 2021, 22, 589–607. [Google Scholar] [CrossRef] [PubMed]
  87. Mooers, B.H.M.; Logue, J.S.; Berglund, J.A. The structural basis of myotonic dystrophy from the crystal structure of CUG repeats. Proc. Natl. Acad. Sci. USA 2005, 102, 16626–16631. [Google Scholar] [CrossRef]
  88. Tian, B.; White, R.J.; Xia, T.; Welle, S.; Turner, D.H.; Mathews, M.B.; Thornton, C.A. Expanded CUG repeat RNAs form hairpins that activate the double-stranded RNA-dependent protein kinase PKR. RNA 2000, 6, 79–87. [Google Scholar] [CrossRef]
  89. Xia, G.; Ashizawa, T. Dynamic changes of nuclear RNA foci in proliferating DM1 cells. Histochem. Cell Biol. 2015, 143, 557–564. [Google Scholar] [CrossRef]
  90. Dere, R.; Napierala, M.; Ranum, L.P.; Wells, R.D. Hairpin structure-forming propensity of the (CCTG·CAGG) tetranucleotide repeats contributes to the genetic instability associated with myotonic dystrophy type 2. J. Biol. Chem. 2004, 279, 41715–41726. [Google Scholar] [CrossRef]
  91. Meola, G. Myotonic dystrophy type 2: The 2020 update. Acta Myol. 2020, 39, 222. [Google Scholar]
  92. Benhalevy, D.; Gupta, S.K.; Danan, C.H.; Ghosal, S.; Sun, H.-W.; Kazemier, H.G.; Paeschke, K.; Hafner, M.; Juranek, S.A. The human CCHC-type zinc finger nucleic acid-binding protein binds G-rich elements in target mRNA coding sequences and promotes translation. Cell Rep. 2017, 18, 2979–2990. [Google Scholar] [CrossRef]
  93. Radunović, D.; Prelević, V.; Tomović, F.; Domislović, M.; Jelaković, B. Glucagon-like peptide-1 (GLP-1) receptor agonist as cardio-and nephroprotection in a patient with diabetic kidney disease and proximal myotonic myopathy type 2 (PROMM MD2). Rad Hrvat. Akad. Znan. Umjetnosti. Med. Znan. 2024, 563, 112–115. [Google Scholar]
  94. Stone, W.L.; Basit, H.; Shah, M.; Los, E. Fragile X Syndrome. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar] [PubMed]
  95. Nobile, V.; Pucci, C.; Chiurazzi, P.; Neri, G.; Tabolacci, E. DNA methylation, mechanisms of FMR1 inactivation and therapeutic perspectives for fragile X syndrome. Biomolecules 2021, 11, 296. [Google Scholar] [CrossRef]
  96. Sidorov, M.S.; Auerbach, B.D.; Bear, M.F. Fragile X mental retardation protein and synaptic plasticity. Mol. Brain 2013, 6, 15. [Google Scholar] [CrossRef] [PubMed]
  97. Daughters, R.S.; Tuttle, D.L.; Gao, W.; Ikeda, Y.; Moseley, M.L.; Ebner, T.J.; Swanson, M.S.; Ranum, L.P. RNA gain-of-function in spinocerebellar ataxia type 8. PLoS Genet. 2009, 5, e1000600. [Google Scholar] [CrossRef] [PubMed]
  98. Cleary, J.D.; Subramony, S.; Ranum, L.P. Spinocerebellar ataxia type 8. In Literature Cited; University of Washington: Seattle, WA, USA, 1993. [Google Scholar] [PubMed]
  99. Merienne, K.; Trottier, Y. SCA8 CAG/CTG Expansions, a Tale of Two TOXIC ities: A Unique or Common Case? PLoS Genet. 2009, 5, e1000593. [Google Scholar] [CrossRef] [PubMed]
  100. Moseley, M.L.; Zu, T.; Ikeda, Y.; Gao, W.; Mosemiller, A.K.; Daughters, R.S.; Chen, G.; Weatherspoon, M.R.; Clark, H.B.; Ebner, T.J. Bidirectional expression of CUG and CAG expansion transcripts and intranuclear polyglutamine inclusions in spinocerebellar ataxia type 8. Nat. Genet. 2006, 38, 758–769. [Google Scholar] [CrossRef]
  101. Tan, H.; Xu, Z.; Jin, P. Role of noncoding RNAs in trinucleotide repeat neurodegenerative disorders. Exp. Neurol. 2012, 235, 469–475. [Google Scholar] [CrossRef]
  102. Gelderblom, H.R. Structure and classification of viruses. In Medical Microbiology, 4th ed.; Baron, S., Ed.; University of Texas Medical Branch at Galveston: Galveston, TX, USA, 1996. [Google Scholar]
  103. Montero, H.; García-Román, R.; Mora, S.I. eIF4E as a control target for viruses. Viruses 2015, 7, 739–750. [Google Scholar] [CrossRef]
  104. Ding, P.; Summers, M.F. Sequestering the 5′-cap for viral RNA packaging. Bioessays 2022, 44, 2200104. [Google Scholar] [CrossRef]
  105. Miller, C.M.; Selvam, S.; Fuchs, G. Fatal attraction: The roles of ribosomal proteins in the viral life cycle. Wiley Interdiscip. Rev. RNA 2021, 12, e1613. [Google Scholar] [CrossRef]
  106. Leary, J.A. Mass spectrometric analysis of the human 40S ribosomal subunit: Native and HCV IRES-bound complexes. Protein Sci. 2005, 14, 1438–1446. [Google Scholar]
  107. Fuchs, G.; Petrov, A.N.; Marceau, C.D.; Popov, L.M.; Chen, J.; O’Leary, S.E.; Wang, R.; Carette, J.E.; Sarnow, P.; Puglisi, J.D. Kinetic pathway of 40S ribosomal subunit recruitment to hepatitis C virus internal ribosome entry site. Proc. Natl. Acad. Sci. USA 2015, 112, 319–325. [Google Scholar] [CrossRef]
  108. Kumar, R.; Mehta, D.; Mishra, N.; Nayak, D.; Sunil, S. Role of host-mediated post-translational modifications (PTMs) in RNA virus pathogenesis. Int. J. Mol. Sci. 2020, 22, 323. [Google Scholar] [CrossRef] [PubMed]
  109. Byk, L.A.; Iglesias, N.G.; De Maio, F.A.; Gebhard, L.G.; Rossi, M.; Gamarnik, A.V. Dengue virus genome uncoating requires ubiquitination. mBio 2016, 7, e00804-16. [Google Scholar] [CrossRef] [PubMed]
  110. Leung, D.W.; Prins, K.C.; Basler, C.F.; Amarasinghe, G.K. Ebolavirus VP35 is a multifunctional virulence factor. Virulence 2010, 1, 526–531. [Google Scholar] [CrossRef] [PubMed]
  111. Valerdi, K.M.; Hage, A.; van Tol, S.; Rajsbaum, R.; Giraldo, M.I. The role of the host ubiquitin system in promoting replication of emergent viruses. Viruses 2021, 13, 369. [Google Scholar] [CrossRef]
  112. Muteeb, G.; Rehman, M.T.; Shahwan, M.; Aatif, M. Origin of antibiotics and antibiotic resistance, and their impacts on drug development: A narrative review. Pharmaceuticals 2023, 16, 1615. [Google Scholar] [CrossRef]
  113. Sheng, J. Sheng Lab, University at Albany Department of Chemistry: Home. 2022. Available online: https://www.albany.edu/rna/sheng-lab (accessed on 1 July 2024).
  114. Berg, M.D.; Brandl, C.J. Transfer RNAs: Diversity in form and function. RNA Biol. 2021, 18, 316–339. [Google Scholar] [CrossRef]
  115. Alberts, B.; Johnson, A.; Lewis, J.; Raff, M.; Roberts, K.; Walter, P. From RNA to protein. In Molecular Biology of the Cell, 4th ed.; Garland Science: New York, NY, USA, 2002. [Google Scholar]
  116. Agris, P.F.; Eruysal, E.R.; Narendran, A.; Väre, V.Y.; Vangaveti, S.; Ranganathan, S.V. Celebrating wobble decoding: Half a century and still much is new. RNA Biol. 2018, 15, 537–553. [Google Scholar] [CrossRef]
  117. Olspert, A.; Chung, B.Y.-W.; Atkins, J.F.; Carr, J.P.; Firth, A.E. Transcriptional slippage in the positive-sense RNA virus family Potyviridae. EMBO Rep. 2015, 16, 995–1004. [Google Scholar] [CrossRef]
  118. Zheng, Y.Y.; Wu, Y.; Begley, T.J.; Sheng, J. Sulfur modification in natural RNA and therapeutic oligonucleotides. RSC Chem. Biol. 2021, 2, 990–1003. [Google Scholar] [CrossRef]
  119. Jäger, G.; Chen, P.; Björk, G.R. Transfer RNA bound to MnmH protein is enriched with geranylated tRNA—A possible intermediate in its selenation? PLoS ONE 2016, 11, e0153488. [Google Scholar] [CrossRef]
  120. Kulik, K.; Sadowska, K.; Wielgus, E.; Pacholczyk-Sienicka, B.; Sochacka, E.; Nawrot, B. 2-Selenouridine, a modified nucleoside of bacterial tRNAs, its reactivity in the presence of oxidizing and reducing reagents. Int. J. Mol. Sci. 2022, 23, 7973. [Google Scholar] [CrossRef] [PubMed]
  121. Veres, Z.; Stadtman, T.C. A purified selenophosphate-dependent enzyme from Salmonella typhimurium catalyzes the replacement of sulfur in 2-thiouridine residues in tRNAs with selenium. Proc. Natl. Acad. Sci. USA 1994, 91, 8092–8096. [Google Scholar] [CrossRef] [PubMed]
  122. Dumelin, C.E.; Chen, Y.; Leconte, A.M.; Chen, Y.G.; Liu, D.R. Discovery and biological characterization of geranylated RNA in bacteria. Nat. Chem. Biol. 2012, 8, 913–919. [Google Scholar] [CrossRef] [PubMed]
  123. Čavužić, M.; Liu, Y. Biosynthesis of sulfur-containing tRNA modifications: A comparison of bacterial, archaeal, and eukaryotic pathways. Biomolecules 2017, 7, 27. [Google Scholar] [CrossRef] [PubMed]
  124. Chakraborti, S.; Chhibber-Goel, J.; Sharma, A. Drug targeting of aminoacyl-tRNA synthetases in Anopheles species and Aedes aegypti that cause malaria and dengue. Parasites Vectors 2021, 14, 605. [Google Scholar] [CrossRef]
  125. Gomez, M.A.R.; Ibba, M. Aminoacyl-tRNA synthetases. RNA 2020, 26, 910–936. [Google Scholar] [CrossRef]
  126. Rathnayake, U.M.; Hendrickson, T.L. Bacterial aspartyl-tRNA synthetase has glutamyl-tRNA synthetase activity. Genes 2019, 10, 262. [Google Scholar] [CrossRef]
  127. Jain, V.; Kikuchi, H.; Oshima, Y.; Sharma, A.; Yogavel, M. Structural and functional analysis of the anti-malarial drug target prolyl-tRNA synthetase. J. Struct. Funct. Genom. 2014, 15, 181–190. [Google Scholar] [CrossRef]
  128. Zhu, Y.; Zhu, L.; Wang, X.; Jin, H. RNA-based therapeutics: An overview and prospectus. Cell Death Dis. 2022, 13, 644. [Google Scholar] [CrossRef]
  129. Elbashir, S.M.; Harborth, J.; Lendeckel, W.; Yalcin, A.; Weber, K.; Tuschl, T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 2001, 411, 494–498. [Google Scholar] [CrossRef]
  130. Germer, K.; Leonard, M.; Zhang, X. RNA aptamers and their therapeutic and diagnostic applications. Int. J. Biochem. Mol. Biol. 2013, 4, 27. [Google Scholar]
  131. Juru, A.U.; Hargrove, A.E. Frameworks for targeting RNA with small molecules. J. Biol. Chem. 2021, 296, 100191. [Google Scholar]
  132. Suga, K.; Tanabe, T.; Tomita, H.; Shimanouchi, T.; Umakoshi, H. Conformational change of single-stranded RNAs induced by liposome binding. Nucleic Acids Res. 2011, 39, 8891–8900. [Google Scholar] [CrossRef] [PubMed]
  133. Quemener, A.M.; Bachelot, L.; Forestier, A.; Donnou-Fournet, E.; Gilot, D.; Galibert, M.D. The powerful world of antisense oligonucleotides: From bench to bedside. Wiley Interdiscip. Rev. RNA 2020, 11, e1594. [Google Scholar] [CrossRef]
  134. Kilanowska, A.; Studzińska, S. In vivo and in vitro studies of antisense oligonucleotides—A review. RSC Adv. 2020, 10, 34501–34516. [Google Scholar] [CrossRef]
  135. Aartsma-Rus, A.; Garanto, A.; van Roon-Mom, W.; McConnell, E.M.; Suslovitch, V.; Yan, W.X.; Watts, J.K.; Yu, T.W.; Collaborative, N. Consensus guidelines for the design and in vitro preclinical efficacy testing N-of-1 exon skipping antisense oligonucleotides. Nucleic Acid Ther. 2023, 33, 17–25. [Google Scholar] [CrossRef]
  136. Collotta, D.; Bertocchi, I.; Chiapello, E.; Collino, M. Antisense oligonucleotides: A novel Frontier in pharmacological strategy. Front. Pharmacol. 2023, 14, 1304342. [Google Scholar] [CrossRef]
  137. Roberts, T.C.; Langer, R.; Wood, M.J. Advances in oligonucleotide drug delivery. Nat. Rev. Drug Discov. 2020, 19, 673–694. [Google Scholar] [CrossRef]
  138. Kaburagi, H.; Nagata, T.; Enomoto, M.; Hirai, T.; Ohyagi, M.; Ihara, K.; Yoshida-Tanaka, K.; Ebihara, S.; Asada, K.; Yokoyama, H. Systemic DNA/RNA heteroduplex oligonucleotide administration for regulating the gene expression of dorsal root ganglion and sciatic nerve. Mol. Ther.-Nucleic Acids 2022, 28, 910–919. [Google Scholar] [CrossRef]
  139. Hua, Y.; Krainer, A.R. Antisense-Mediated Exon Inclusion. In Exon Skipping; Aartsma-Rus, A., Ed.; Methods in Molecular Biology; Humana Press: Totowa, NJ, USA, 2012; Volume 867. [Google Scholar] [CrossRef]
  140. Happi Mbakam, C.; Roustant, J.; Rousseau, J.; Yameogo, P.; Lu, Y.; Bigot, A.; Mamchaoui, K.; Mouly, V.; Lamothe, G.; Tremblay, J.P. Prime editing strategies to mediate exon skipping in DMD gene. Front. Med. 2023, 10, 1128557. [Google Scholar] [CrossRef]
  141. Levin, A.A. Treating disease at the RNA level with oligonucleotides. N. Engl. J. Med. 2019, 380, 57–70. [Google Scholar] [CrossRef] [PubMed]
  142. Liang, X.-H.; Nichols, J.G.; De Hoyos, C.L.; Crooke, S.T. Some ASOs that bind in the coding region of mRNAs and induce RNase H1 cleavage can cause increases in the pre-mRNAs that may blunt total activity. Nucleic Acids Res. 2020, 48, 9840–9858. [Google Scholar] [CrossRef] [PubMed]
  143. Agrawal, N.; Dasaradhi, P.; Mohmmed, A.; Malhotra, P.; Bhatnagar, R.K.; Mukherjee, S.K. RNA interference: Biology, mechanism, and applications. Microbiol. Mol. Biol. Rev. 2003, 67, 657–685. [Google Scholar] [CrossRef] [PubMed]
  144. Dana, H.; Chalbatani, G.M.; Mahmoodzadeh, H.; Karimloo, R.; Rezaiean, O.; Moradzadeh, A.; Mehmandoost, N.; Moazzen, F.; Mazraeh, A.; Marmari, V. Molecular mechanisms and biological functions of siRNA. Int. J. Biomed. Sci. IJBS 2017, 13, 48. [Google Scholar] [CrossRef]
  145. van den Berg, A.; Mols, J.; Han, J. RISC-target interaction: Cleavage and translational suppression. Biochim. Biophys. Acta (BBA)-Gene Regul. Mech. 2008, 1779, 668–677. [Google Scholar] [CrossRef]
  146. Wołowiec, Ł.; Osiak, J.; Wołowiec, A.; Wijata, A.; Grześk, E.; Kozakiewicz, M.; Banach, J.; Nowaczyk, A.; Nowaczyk, J.; Grześk, G. Inclisiran—Safety and Effectiveness of Small interfering RNA in inhibition of PCSK-9. Pharmaceutics 2023, 15, 323. [Google Scholar] [CrossRef]
  147. Albosta, M.S.; Grant, J.K.; Taub, P.; Blumenthal, R.S.; Martin, S.S.; Michos, E.D. Inclisiran: A new strategy for LDL-C lowering and prevention of atherosclerotic cardiovascular disease. Vasc. Health Risk Manag. 2023, 19, 421–431. [Google Scholar] [CrossRef]
  148. Crooke, S.T.; Vickers, T.A.; Liang, X.-H. Phosphorothioate modified oligonucleotide–protein interactions. Nucleic Acids Res. 2020, 48, 5235–5253. [Google Scholar] [CrossRef]
  149. Anderson, B.A.; Freestone, G.C.; Low, A.; De-Hoyos, C.L.; Drury, W.J., III; Østergaard, M.E.; Migawa, M.T.; Fazio, M.; Wan, W.B.; Berdeja, A.; et al. Towards next generation antisense oligonucleotides: Mesylphosphoramidate modification improves therapeutic index and duration of effect of gapmer antisense oligonucleotides. Nucleic Acids Res. 2021, 49, 9026–9041. [Google Scholar] [CrossRef]
  150. Zhang, L.; Liang, Y.; Liang, G.; Tian, Z.; Zhang, Y.; Liu, Z.; Ji, X. The therapeutic prospects of N-acetylgalactosamine-siRNA conjugates. Front. Pharmacol. 2022, 13, 1090237. [Google Scholar] [CrossRef]
  151. An, G. Pharmacokinetics and Pharmacodynamics of GalNAc-Conjugated siRNAs. J. Clin. Pharmacol. 2024, 64, 45–57. [Google Scholar] [CrossRef] [PubMed]
  152. Zhang, X.; Goel, V.; Robbie, G.J. Pharmacokinetics of Patisiran, the first approved RNA interference therapy in patients with hereditary transthyretin-mediated amyloidosis. J. Clin. Pharmacol. 2020, 60, 573–585. [Google Scholar] [CrossRef] [PubMed]
  153. Xu, Y.; Li, Z. Overview, innovations and applications in human disease research and gene therapy. Comput. Struct. Biotechnol. J. 2020, 18, 2401–2415. [Google Scholar] [CrossRef] [PubMed]
  154. Gunitseva, N.; Evteeva, M.; Borisova, A.; Patrushev, M.; Subach, F. RNA-dependent RNA targeting by CRISPR-cas systems: Characterizations and applications. Int. J. Mol. Sci. 2023, 24, 6894. [Google Scholar] [CrossRef]
  155. Cox, D.B.; Gootenberg, J.S.; Abudayyeh, O.O.; Franklin, B.; Kellner, M.J.; Joung, J.; Zhang, F. RNA editing with CRISPR-Cas13. Science 2017, 358, 1019–1027. [Google Scholar] [CrossRef]
  156. Scrudato, M.L.; Poulard, K.; Sourd, C.; Tomé, S.; Klein, A.F.; Corre, G.; Huguet, A.; Furling, D.; Gourdon, G.; Buj-Bello, A. Genome editing of expanded CTG repeats within the human DMPK gene reduces nuclear RNA foci in the muscle of DM1 mice. Mol. Ther. 2019, 27, 1372–1388. [Google Scholar] [CrossRef]
  157. Marsh, S.; Hanson, B.; Wood, M.J.; Varela, M.A.; Roberts, T.C. Application of CRISPR-Cas9-mediated genome editing for the treatment of myotonic dystrophy type 1. Mol. Ther. 2020, 28, 2527–2539. [Google Scholar] [CrossRef]
  158. Abbott, T.R.; Dhamdhere, G.; Liu, Y.; Lin, X.; Goudy, L.; Zeng, L.; Chemparathy, A.; Chmura, S.; Heaton, N.S.; Debs, R. Development of CRISPR as an antiviral strategy to combat SARS-CoV-2 and influenza. Cell 2020, 181, 865–876.e12. [Google Scholar] [CrossRef]
  159. Konermann, S.; Lotfy, P.; Brideau, N.J.; Oki, J.; Shokhirev, M.N.; Hsu, P.D. Transcriptome engineering with RNA-targeting type VI-D CRISPR effectors. Cell 2018, 173, 665–676.e14. [Google Scholar] [CrossRef]
  160. Guo, Y.; Han, L.; Han, S.; Tang, H.; Wang, S.; Cui, C.; Chen, B.; Li, H.; Shu, Y. Specific knockdown of Htra2 by CRISPR-CasRx prevents acquired sensorineural hearing loss in mice. Mol. Ther.-Nucleic Acids 2022, 28, 643–655. [Google Scholar] [CrossRef]
  161. Asmamaw Mengstie, M. Viral vectors for the in vivo delivery of CRISPR components: Advances and challenges. Front. Bioeng. Biotechnol. 2022, 10, 895713. [Google Scholar] [CrossRef] [PubMed]
  162. Lu, Y.; Godbout, K.; Lamothe, G.; Tremblay, J.P. CRISPR/Cas9 delivery strategies with engineered extracellular vesicles. Mol. Ther.-Nucleic Acids 2023, 34, 102040. [Google Scholar] [CrossRef] [PubMed]
  163. Kazemian, P.; Yu, S.-Y.; Thomson, S.B.; Birkenshaw, A.; Leavitt, B.R.; Ross, C.J. Lipid-nanoparticle-based delivery of CRISPR/Cas9 genome-editing components. Mol. Pharm. 2022, 19, 1669–1686. [Google Scholar] [CrossRef] [PubMed]
  164. Chen, F.; Alphonse, M.; Liu, Q. Strategies for nonviral nanoparticle-based delivery of CRISPR/Cas9 therapeutics. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnology 2020, 12, e1609. [Google Scholar] [CrossRef]
  165. Saadati, F.; Cammarone, S.; Ciufolini, M.A. A Route to Lipid ALC-0315: A Key Component of a COVID-19 mRNA Vaccine. Chem. A Eur. J. 2022, 28, e202200906. [Google Scholar] [CrossRef]
  166. Meyer, S.M.; Williams, C.C.; Akahori, Y.; Tanaka, T.; Aikawa, H.; Tong, Y.; Childs-Disney, J.L.; Disney, M.D. Small molecule recognition of disease-relevant RNA structures. Chem. Soc. Rev. 2020, 49, 7167–7199. [Google Scholar] [CrossRef]
  167. Esteller, M. Non-coding RNAs in human disease. Nat. Rev. Genet. 2011, 12, 861–874. [Google Scholar] [CrossRef]
  168. Baisden, J.T.; Childs-Disney, J.L.; Ryan, L.S.; Disney, M.D. Affecting RNA biology genome-wide by binding small molecules and chemically induced proximity. Curr. Opin. Chem. Biol. 2021, 62, 119–129. [Google Scholar] [CrossRef]
  169. Jaiswal, A.K.; Thaxton, M.L.; Scherer, G.M.; Sorrentino, J.P.; Garg, N.K.; Rao, D.S. Small molecule inhibition of RNA binding proteins in haematologic cancer. RNA Biol. 2024, 21, 276–289. [Google Scholar] [CrossRef]
  170. Webb, T.R.; Joyner, A.S.; Potter, P.M. The development and application of small molecule modulators of SF3b as therapeutic agents for cancer. Drug Discov. Today 2013, 18, 43–49. [Google Scholar] [CrossRef]
  171. López-Cánovas, J.L.; Hermán-Sánchez, N.; del Rio-Moreno, M.; Fuentes-Fayos, A.C.; Lara-López, A.; Sánchez-Frias, M.E.; Amado, V.; Ciria, R.; Briceño, J.; de la Mata, M. PRPF8 increases the aggressiveness of hepatocellular carcinoma by regulating FAK/AKT pathway via fibronectin 1 splicing. Exp. Mol. Med. 2023, 55, 132–142. [Google Scholar] [CrossRef] [PubMed]
  172. Nian, Q.; Li, Y.; Li, J.; Zhao, L.; Rodrigues Lima, F.; Zeng, J.; Liu, R.; Ye, Z. U2AF1 in various neoplastic diseases and relevant targeted therapies for malignant cancers with complex mutations. Oncol. Rep. 2023, 51, 5. [Google Scholar] [CrossRef]
  173. Ratni, H.; Scalco, R.S.; Stephan, A.H. Risdiplam, the first approved small molecule splicing modifier drug as a blueprint for future transformative medicines. ACS Med. Chem. Lett. 2021, 12, 874–877. [Google Scholar] [CrossRef] [PubMed]
  174. Nijhuis, A.; Sikka, A.; Yogev, O.; Herendi, L.; Balcells, C.; Ma, Y.; Poon, E.; Eckold, C.; Valbuena, G.N.; Xu, Y. Indisulam targets RNA splicing and metabolism to serve as a therapeutic strategy for high-risk neuroblastoma. Nat. Commun. 2022, 13, 1380. [Google Scholar] [CrossRef]
  175. Jenquin, J.R.; Coonrod, L.A.; Silverglate, Q.A.; Pellitier, N.A.; Hale, M.A.; Xia, G.; Nakamori, M.; Berglund, J.A. Furamidine rescues myotonic dystrophy type I associated mis-splicing through multiple mechanisms. ACS Chem. Biol. 2018, 13, 2708–2718. [Google Scholar] [CrossRef]
  176. Devulapally, R.; Paulmurugan, R. Polymer nanoparticles for drug and small silencing RNA delivery to treat cancers of different phenotypes. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2014, 6, 40–60. [Google Scholar] [CrossRef]
  177. Ramírez-Cortés, F.; Ménová, P. Hepatocyte targeting via the asialoglycoprotein receptor. RSC Med. Chem. 2025. ahead of print. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  178. Cui, H.; Zhu, X.; Li, S.; Wang, P.; Fang, J. Liver-targeted delivery of oligonucleotides with N-acetylgalactosamine conjugation. ACS Omega 2021, 6, 16259–16265. [Google Scholar] [CrossRef]
  179. Dua, P.; Kim, S.; Lee, D.-K. Nucleic acid aptamers targeting cell-surface proteins. Methods 2011, 54, 215–225. [Google Scholar] [CrossRef]
  180. Du, J.; Dartawan, R.; Rice, W.; Gao, F.; Zhou, J.H.; Sheng, J. Fluorescent platforms for RNA chemical biology research. Genes 2022, 13, 1348. [Google Scholar] [CrossRef]
  181. Zhang, Y.; Zhang, H.; Chan, D.W.H.; Ma, Y.; Lu, A.; Yu, S.; Zhang, B.; Zhang, G. Strategies for developing long-lasting therapeutic nucleic acid aptamer targeting circulating protein: The present and the future. Front. Cell Dev. Biol. 2022, 10, 1048148. [Google Scholar] [CrossRef] [PubMed]
  182. Panigaj, M.; Johnson, M.B.; Ke, W.; McMillan, J.; Goncharova, E.A.; Chandler, M.; Afonin, K.A. Aptamers as modular components of therapeutic nucleic acid nanotechnology. Ther. RNA Nanotechnol. 2021, 13, 825–882. [Google Scholar] [CrossRef] [PubMed]
  183. Catuogno, S.; Rienzo, A.; Di Vito, A.; Esposito, C.L.; de Franciscis, V. Selective delivery of therapeutic single strand antimiRs by aptamer-based conjugates. J. Control. Release 2015, 210, 147–159. [Google Scholar] [CrossRef] [PubMed]
  184. Ellington, A.D.; Szostak, J.W. In vitro selection of RNA molecules that bind specific ligands. Nature 1990, 346, 818–822. [Google Scholar] [CrossRef]
  185. Tuerk, C.; Gold, L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science 1990, 249, 505–510. [Google Scholar] [CrossRef]
  186. Ni, X.; Castanares, M.; Mukherjee, A.; Lupold, S.E. Nucleic acid aptamers: Clinical applications and promising new horizons. Curr. Med. Chem. 2011, 18, 4206–4214. [Google Scholar] [CrossRef]
  187. Buglak, A.A.; Samokhvalov, A.V.; Zherdev, A.V.; Dzantiev, B.B. Methods and applications of in silico aptamer design and modeling. Int. J. Mol. Sci. 2020, 21, 8420. [Google Scholar] [CrossRef]
  188. Mahmoudian, F.; Ahmari, A.; Shabani, S.; Sadeghi, B.; Fahimirad, S.; Fattahi, F. Aptamers as an approach to targeted cancer therapy. Cancer Cell Int. 2024, 24, 108. [Google Scholar] [CrossRef]
  189. Nagpal, M.; Nagpal, K.; Nagpal, P. A comparative debate on the various anti-vascular endothelial growth factor drugs: Pegaptanib sodium (Macugen), ranibizumab (Lucentis) and bevacizumab (Avastin). Indian J. Ophthalmol. 2007, 55, 437–439. [Google Scholar] [CrossRef]
  190. Ng, E.; Shima, D.; Calias, P.; Cunningham, E.T., Jr.; Guyer, D.R.; Adamis, A.P. Pegaptanib, a targeted anti-VEGF aptamer for ocular vascular disease. Nat. Rev. Drug Discov. 2006, 5, 123–132. [Google Scholar] [CrossRef]
  191. Penn, J.; Madan, A.; Caldwell, R.B.; Bartoli, M.; Caldwell, R.; Hartnett, M. Vascular endothelial growth factor in eye disease. Prog. Retin. Eye Res. 2008, 27, 331–371. [Google Scholar] [CrossRef] [PubMed]
  192. Vinores, S.A. Pegaptanib in the treatment of wet, age-related macular degeneration. Int. J. Nanomed. 2006, 1, 263–268. [Google Scholar]
  193. Gupta, N.; Mansoor, S.; Sharma, A.; Sapkal, A.; Sheth, J.; Falatoonzadeh, P.; Kuppermann, B.; Kenney, M. Diabetic retinopathy and VEGF. Open Ophthalmol. J. 2013, 7, 4. [Google Scholar] [CrossRef]
  194. Macugen Diabetic Retinopathy Study Group. A phase II randomized double-masked trial of pegaptanib, an Anti–Vascular endothelial growth factor aptamer, for diabetic macular edema. Ophthalmology 2005, 112, 1747–1757. [Google Scholar] [CrossRef]
  195. Lakhin, A.; Tarantul, V.Z.; Gening, L. Aptamers: Problems, solutions and prospects. Acta Nat. 2013, 5, 34–43. [Google Scholar] [CrossRef]
  196. Nimjee, S.M.; White, R.R.; Becker, R.C.; Sullenger, B.A. Aptamers as therapeutics. Annu. Rev. Pharmacol. Toxicol. 2017, 57, 61–79. [Google Scholar] [CrossRef]
  197. Maio, G.E.; Enweronye, O.; Zumrut, H.E.; Batool, S.; Van, N.A.; Mallikaratchy, P.R. Systematic optimization and modification of a DNA aptamer with 2′-O-methyl RNA analogues. ChemistrySelect 2017, 2, 2335–2340. [Google Scholar] [CrossRef]
  198. Volk, D.E.; Lokesh, G.L. Development of phosphorothioate DNA and DNA thioaptamers. Biomedicines 2017, 5, 41. [Google Scholar] [CrossRef]
  199. Wang, Y.; Miao, L.; Satterlee, A.; Huang, L. Delivery of oligonucleotides with lipid nanoparticles. Adv. Drug Deliv. Rev. 2015, 87, 68–80. [Google Scholar] [CrossRef]
  200. Puzzo, F.; Zhang, C.; Gray, B.P.; Zhang, F.; Sullenger, B.A.; Kay, M.A. Aptamer-programmable adeno-associated viral vectors as a novel platform for cell-specific gene transfer. Mol. Ther.-Nucleic Acids 2023, 31, 383–397. [Google Scholar] [CrossRef]
  201. Mirón-Barroso, S.; Correia, J.S.; Frampton, A.E.; Lythgoe, M.P.; Clark, J.; Tookman, L.; Ottaviani, S.; Castellano, L.; Porter, A.E.; Georgiou, T.K. Polymeric carriers for delivery of RNA cancer therapeutics. Non-Coding RNA 2022, 8, 58. [Google Scholar] [CrossRef] [PubMed]
  202. Jo, H.; Ban, C. Aptamer–nanoparticle complexes as powerful diagnostic and therapeutic tools. Exp. Mol. Med. 2016, 48, e230. [Google Scholar] [CrossRef]
Figure 1. A ribonucleoprotein complex constitutes the spliceosome. The spliceosome, a ribonucleoprotein complex consisting of snRNAs U1-6, is instrumental in converting pre-mRNA into mature-mRNA via the excision of non-coding intron sequences, typically bordered by GT and AT nucleotides, of the mRNA. This allows the expressed regions, or exons, to merge into a single, mature mRNA.
Figure 1. A ribonucleoprotein complex constitutes the spliceosome. The spliceosome, a ribonucleoprotein complex consisting of snRNAs U1-6, is instrumental in converting pre-mRNA into mature-mRNA via the excision of non-coding intron sequences, typically bordered by GT and AT nucleotides, of the mRNA. This allows the expressed regions, or exons, to merge into a single, mature mRNA.
Genes 16 00048 g001
Figure 2. Toxic CUG-repeats disrupt MBNL–RNA interactions. A buildup of CTG repeats in the 3′ UTR of the DMPK gene leads to increased CUG–RNA interactions following transcription. Subsequently, otherwise necessary, MBNL–RNA interactions are disrupted, perturbing alternative splicing. This manifests as myotonic muscular dystrophy.
Figure 2. Toxic CUG-repeats disrupt MBNL–RNA interactions. A buildup of CTG repeats in the 3′ UTR of the DMPK gene leads to increased CUG–RNA interactions following transcription. Subsequently, otherwise necessary, MBNL–RNA interactions are disrupted, perturbing alternative splicing. This manifests as myotonic muscular dystrophy.
Genes 16 00048 g002
Figure 3. Viral IRES attracts 40S subunit via lysine acetylation of RPS25. Lysine acetyltransferases neutralize the positive charge on lysines, thereby allowing RPS25, along with RACK1, to bind the 40S ribosomal subunit. This complex facilitates the so-called cap-independent translation of the viral genome by freeing it of eukaryotic transcription factors that target the 5′ cap. Instead, the small ribosomal complex interacts with the virus’ IRES and, consequently, translates its genome using host machinery.
Figure 3. Viral IRES attracts 40S subunit via lysine acetylation of RPS25. Lysine acetyltransferases neutralize the positive charge on lysines, thereby allowing RPS25, along with RACK1, to bind the 40S ribosomal subunit. This complex facilitates the so-called cap-independent translation of the viral genome by freeing it of eukaryotic transcription factors that target the 5′ cap. Instead, the small ribosomal complex interacts with the virus’ IRES and, consequently, translates its genome using host machinery.
Genes 16 00048 g003
Figure 4. SelU causes selenation at tRNA’s 5′ wobble position. Modifications of tRNA nucleosides, especially around the wobble and anticodon positions, can induce mutations. In particular, 2-selenouridine transferase catalyzes the conversion of sulfur to selenium, leading to a frameshift mutation and the associated pathology.
Figure 4. SelU causes selenation at tRNA’s 5′ wobble position. Modifications of tRNA nucleosides, especially around the wobble and anticodon positions, can induce mutations. In particular, 2-selenouridine transferase catalyzes the conversion of sulfur to selenium, leading to a frameshift mutation and the associated pathology.
Genes 16 00048 g004
Figure 5. Schematic of SELEX procedure.
Figure 5. Schematic of SELEX procedure.
Genes 16 00048 g005
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

Dansereau, S.J.; Cui, H.; Dartawan, R.P.; Sheng, J. The Plethora of RNA–Protein Interactions Model a Basis for RNA Therapies. Genes 2025, 16, 48. https://doi.org/10.3390/genes16010048

AMA Style

Dansereau SJ, Cui H, Dartawan RP, Sheng J. The Plethora of RNA–Protein Interactions Model a Basis for RNA Therapies. Genes. 2025; 16(1):48. https://doi.org/10.3390/genes16010048

Chicago/Turabian Style

Dansereau, Stephen J., Hua Cui, Ricky P. Dartawan, and Jia Sheng. 2025. "The Plethora of RNA–Protein Interactions Model a Basis for RNA Therapies" Genes 16, no. 1: 48. https://doi.org/10.3390/genes16010048

APA Style

Dansereau, S. J., Cui, H., Dartawan, R. P., & Sheng, J. (2025). The Plethora of RNA–Protein Interactions Model a Basis for RNA Therapies. Genes, 16(1), 48. https://doi.org/10.3390/genes16010048

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