Abstract
The ENCODE and genome-wide association projects have shown that much of the genome is transcribed into RNA and much less is translated into protein. These and other functional studies suggest that the druggable transcriptome is much larger than the druggable proteome. This review highlights approaches to define druggable RNA targets and structure–activity relationships across genomic RNA. Binding compounds can be identified and optimized into structure-specific ligands by using sequence-based design with various modes of action, for example, inhibiting translation or directing pre-mRNA splicing outcomes. In addition, strategies to direct protein activity against an RNA of interest via chemically induced proximity is a burgeoning area that has been validated both in cells and in preclinical animal models, and we describe that it may allow rapid access to new avenues to affect RNA biology. These approaches and the unique modes of action suggest that more RNAs are potentially amenable to targeting than proteins.
Keywords: RNA, Small molecules, Induced proximity, Cancer, Microsatellite disorders, Chemical biology, Transcriptome-wide design
Introduction
Although ~80% of the human genome is transcribed into RNA, only 1% is translated into protein [1], suggesting that there are more opportunities to affect biology at the transcriptional level (Fig. 1). Furthermore, RNA has diverse functions tied to its structure [2], including regulating pre-mRNA splicing outcomes [3], cellular localization [4], phase separation [5], and microRNA (miRNA) biogenesis [6,7]. Indeed, RNA-targeting oligonucleotide-based modalities have garnered Food and Drug Administration (FDA) approval, including those that target RNAs associated with central nervous system disorders such as spinal muscular atrophy (SMA) [8] and liver disease [9]. Although these medicines have transformed patients’ lives, their transcriptome-wide implementation has been limited to tissues readily amenable to oligonucleotide targeting, that is, direct delivery to the central nervous system, liver, or kidney.
Figure 1: Disparity in Drug Development Against Protein and RNA Targets.
The human genome, transcriptome, and proteome are shown to scale, with current clinical targets[1] highlighted as a small slice (bright red) of the druggable proteome. Current RNA therapeutics, including antisense oligonucleotides, currently account for ~1% FDA approved therapies, shown to scale as a purple pixel within all FDA-approved small molecules.
Highly structured regions in RNA are often functional [10], and the three-dimensional shapes they adopt could provide small-molecule binding pockets [11], occupied by compounds with complementarity of size, shape, stacking, and charge. Although there are challenges associated with small-molecule targeting of RNA, small molecules can be penetrant to a variety of tissues, and often, lead compounds can be medicinally optimized to enhance target engagement [12] and minimize suboptimal features [13]. Herein, we discuss lead drug and chemical probe discovery against RNA targets on a genome-wide scale to elicit effects on the proteome (Table 1): downregulation of ‘undruggable’ proteins by targeting RNA structures in untranslated regions, upregulation of proteins by targeting miRNAs that repress their expression, and altering the ratio of protein isoforms by influencing pre-mRNA splicing outcomes. Collectively, targeting RNA structures with small molecules can indeed be an effective way to study and control biology.
Table 1:
Compounds from Highlighted References
Compound Name {ref #} | Chemical Structure | RNA target {Disease} | Potency (EC50) or change {Cell line or Model} | Selectivity {off-target} | Structure | Development Stage |
---|---|---|---|---|---|---|
Risdiplam {8/83/85} |
![]() |
SMN2 Splicing {SMA} | 4nm (EC1.5X) {SMA patient derived Fibroblasts} |
16.75nm {F0XM1 Splicing} |
Yes | FDA Approved |
pyrido[2,3-d]pyrimidine {29} |
![]() |
r(CUG) {DM1} |
** Foci Reduction {DM1 patient cells} |
NR | MD Only | NA |
DB1246 {49} |
![]() |
r(G4C2)exp {c9ALS/FTD} | ** Foci Reduction {iPSC Cortical Neurons} | NR | No | NA |
bPGN {70} |
![]() |
Pre-miR-21 {Colorectal cancer} |
0.035 ± 0.003 μM (GI50) {HTC-116: Colorectal cancer cells } |
>10μM {CRL-1790: Normal Colon} |
No | NR |
Compound 5 {95} |
![]() |
MALAT triple helix {Metastasis and Proliferation} |
*** Branching Morphogenesis {MMTV-PyMT tumors} |
No significant differences {Neat 1} |
MD Only | NR |
TGP-21-C1–3 {97} |
![]() |
Pre-miR-21 {Triple Negative Breast Cancer} |
** Cell Invasion {MCF-10A + miR-21 transfection} |
No significant differences {miRNA Profiling} |
MD Only | NR |
NR = Not reported
NA = Not applicable
Drug discovery on a genome-wide scale
Defining small-molecule binding landscapes for RNA structures
Targeting RNA with small molecules is not without its challenges, particularly garnering sufficient selectivity and potency to elicit a biological outcome. Selectivity is a composite of (i) the selectivity of the small molecule for the desired RNA structure, (ii) the presence of that structure elsewhere in the transcriptome, (iii) the relative expression levels of all targets with said structure, and (iv) the structure’s functionality. We developed a method, two-dimensional combinatorial screening (2DCS), that studies the binding capacity and selectivity of small molecules for RNA structures, defining binding landscapes and structure–activity relationships on a transcriptome-wide scale. Coupling 2DCS with the accurate modeling of the RNA’s structure from sequence [14, 15, 16, 17] has enabled the development of small molecules against RNA targets on a genome-wide scale.
2DCS probes the binding potential of small molecules against a library of discrete RNA structures, wherein compounds from a chemical library are immobilized onto functionalized agarose microarrays. RNA structures that bind a small molecule are identified by high-throughput RNA sequencing (RNA-seq). The resulting RNA-seq data are analyzed to calculate the statistical confidence in the enrichment of RNA structures from a 2DCS selection vs. the starting RNA pool (library) [18]; that is, the analysis accounts for biases that may arise during in vitro transcription to generate the RNA library, reverse transcription, polymerase chain reaction amplification, and sequencing. Statistical confidence is a metric of binding fitness, including both affinity and selectivity. Logos [19] and DiffLogos [20] can then be used to visualize privileged sequences and relate structural similarities and differences between molecules, elucidating structure-activity relationships (SAR).
SAR on a transcriptome-wide scale
Fortuitously, RNA secondary structure can be readily and accurately modeled from sequence. RNA’s limited number of building blocks and the large contribution of base pairing energy to its stability enable rapid secondary structure prediction. This ability to model RNA structure facilitates mapping of potential small-molecule binding pockets across the transcriptome and insight into compound selectivity, a function of the number of structures that a compound binds, its relative affinity for each, and the presence of these structural elements across the transcriptome. Notably, the folding landscape for RNA can be dynamic, wherein interconversion between states is necessary for function in some cases [10]. Although labor-intensive, RNA conformational dynamics can be modeled and used to identify lead molecules, as demonstrated with a dynamic ensemble of HIV trans-activation response element (TAR) RNA [11]. Likewise, Hong et al [21, 22] and Dohno and Nakatani [21, 22] have exploited the base pairing propensity of unpaired and dynamic RNA structures by developing ligands to induce higher order interactions within target RNAs.
To integrate RNA structure prediction and small-molecule design on a transcriptome-wide scale, our laboratory has developed a pipeline named Inforna [23,24]. This pipeline starts with computational prediction of the target RNA’s secondary structure based on its genomic sequence alone, which can be further refined by analyzing sequence conservation [25] and by experiment [26]. The structures within an RNA target are then compared with the RNA structure–small molecule interactions identified by 2DCS or other methods. In the following section, we describe how a fundamental understanding of the molecular recognition of RNA structures by small molecules enables the development of compounds with various modes of action (MOAs) that influence the content of the proteome (Table 1).
Affecting RNA biology: binding small molecules
As coding and noncoding RNAs adopt 3D folds that influence their biological roles, there are likely many ways to modulate their function. This section describes how simple binding compounds can alter cellular protein content by various mechanisms (Fig. 2).
Figure 2: RNA Structure-Function Relationships.
Hubs of RNA structure are essential for many cellular processes[10] and provide targetable sites for small molecule binding. Shown are a subset of important RNA structural motifs and their associated biological processes, both in health and disease. RNA-binding molecules, shown in dark blue, have been developed to target a number of these structures and alter their function. Primary and precursor microRNA structures are shown on the bottom right, which are recognized by the microRNA biogenesis machinery by their specific structural motifs and the distances between them.
RNA repeat expansions
Microsatellite disorders are a class of more than 40 inherited diseases [27] caused by RNA repeat expansions, perhaps ideal targets for small molecules. In general, repeat length is correlated with disease onset and/or severity, and disease mechanism is dependent on the location of the repeat within the transcript. Once the repeat expansion reaches a pathogenic length, which is unique to each disease, the RNA folds into a periodic array of loops that form small-molecule binding pockets. These pockets are absent in transcripts lacking the repeat expansion, and the structural differences between pathogenic and short repeats afford the possibility of developing allele-specific small molecules. Furthermore, the repeating nature of these 3D structures enables the design of dimers that bind two adjacent loops simultaneously, which increases affinity, specificity, and potency [28,29]. Notably, repeat expansions, owing to the highly thermodynamically stable structures that they form, are not amenable to antisense oligonucleotide (ASO) targeting, which are not allele specific as they recognize the repeat sequence present in both mutant and wild-type alleles as well as in other transcripts.
The loops formed by RNA repeat expansions can be toxic via gain-of-function mechanisms, such as binding and sequestration of RNA-binding proteins. Such is the case in myotonic dystrophy 1 [DM1; r(CUG)exp] and myotonic dystrophy type 2 [r(CCUG)exp], the two most common forms of adult-onset muscular dystrophy. Indeed, many laboratories have developed ligands that bind r(CUG)exp, although some have mixed MOAs. In early work, the Miller lab used a dynamic combinatorial screening method to identify compounds that bind a model of r(CUG)exp [30], while Nakatani [31] and Zimmerman [32,33] laboratories synthesized dimeric compounds to bind r(CUG)exp and inhibit the sequestration of MBNL1. A peptide has also been developed to bind r(CUG)exp and alleviate defects in a Drosophila model of DM1 [34].
A dimer that binds two adjacent loops in r(CUG)exp developed by our laboratory is allele selective, occupying the target in DM1-affected cells while not affecting mRNAs that contained these shorter repeats [35]. This selectivity can be traced to the lack of structure adopted by short repeats in their corresponding transcripts [29]. The dimer was later appended with bleomycin [36,37], affording a chimeric molecule that cleaved the mutant allele selectively over short repeats found in the wild-type allele and other transcripts in patient-derived muscle cells and in a mouse model with no detectable off-targets [38].
This notion that expanded repeats can be selectively targeted by small molecules over short, nonpathogenic repeats has been explored for other repeat expansions [39], for example, r(CAG)exp, which causes Huntington disease [40, 41, 42, 43]; r(CGG)exp, associated with fragile X syndrome and fragile X-associated tremor/ataxia syndrome [44, 45*, 46, 47]; and r(G4C2)exp, which causes C9orf72 frontotemporal dementia and amyotrophic lateral sclerosis (c9ALS/FTD). The latter repeat has been shown to adopt various structures [48, 49, 50*] (an array of internal loops and G-quadruplexes) that have been targeted with small molecules for therapeutic benefit [45,50*, 51, 52*].
MicroRNAs
Small, but highly structured, miRNAs suppress translation of complementary mRNAs, acting as genetic ‘dimmer switches.’ Expression levels of a subset of proteins can therefore be tuned by modifying miRNA abundance, that is, miRNA biogenesis. Primary and precursor miRNAs form well-defined structures that are recognized by processing enzymes, affording the mature miRNA. Small molecules that bind structures located in processing sites and thus inhibit miRNA biogenesis have been discovered through various methods, including microarrays [53] and a click chemistry approach that assays for Dicer inhibition [54]. The Duca lab developed aminoglycoside conjugates and polyamines to show specific interaction with miR-372 [55,56], while many studies have focused on inhibiting the biogenesis of miR-21 [57*, 58, 59, 60, 61*, 62**, 63] as its upregulation has been linked to cardiovascular disease, fibrosis, and many cancers.
The structures embedded in miRNA processing sites are not always unique in the transcriptome. In the following paragraphs, we describe the lead optimization of a small molecule that binds a structural element found in the Dicer processing sites of two different miRNAs, miR-421 and miR-377, to create a miR-377–selective inhibitor (Fig. 3) [64]. The lead molecule for the two Dicer sites was generated from the RNA-binding preferences of AstraZeneca’s small-molecule library, as determined by 2DCS. As expected, the lead molecule occupies both miRNAs in cells similarly and hence inhibits their biogeneses to similar extents. Target occupancy was studied by Chemical Cross-Linking and Isolation by Pull-down (Chem-CLIP), in which the RNA-binding small molecule is functionalized with a cross-linking module. RNAs cross-linked to the Chem-CLIP probe are isolated by cells and analyzed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) or RNA-seq.
Figure 3: 2D Combinatorial Screening and Fragment Assembly of RNA-Binding Small Molecules.
RNA sequence can be combined with folding simulations to reveal important structural motifs across the transcriptome. To demonstrate that binding preferences can inform design of a selective small molecule inhibitor, a degenerate Dicer site in pre-miR-377 and pre-miR-421 was used as a test case. A structural feature unique to miR-377 was exploited, affording a dimer that binds the degenerate and unique site simultaneously, abrogating binding and activity against pre-miR-421.
Thus, the question at hand was if the lead molecule could be optimized in facile fashion to selectively inhibit miR-377. Careful inspection of pre-miR-377 and pre-miR-421 revealed a structural feature unique to pre-miR-377, which fortuitously was nearby the Dicer site and could be exploited to enhance selectivity. Inforna identified a compound that binds the unique site, and tethering the two binding fragments together (a heterodimer) afforded selective inhibition of miR-377 biogenesis, as determined by miRNome-wide profiling and proteomics studies. Importantly, competitive Chem-CLIP studies, in which cells are cotreated with the Chem-CLIP probe and parent compound, showed that the pre-miR-377 target, but not pre-miR-421, was occupied.
Collectively, these studies show that degeneracy in RNA structural motifs in the genome does not preclude the development of selective small molecules that induce a biological effect. The combination of RNA structure modeling and small-molecule binding preferences, in the form of binding landscapes, can inform lead molecules and identify degenerate sites in the transcriptome. Furthermore, this degeneracy can be alleviated by exploiting structural differences in the RNAs with the degenerate site. Although these chimeric molecules lie outside traditional oral bioavailability guidelines, such as the Lipinski Rule of Five [65], there is growing evidence that therapeutic small molecules can indeed deviate from these standards.
Targeting pre-mRNA splicing with RNA-binding compounds, molecular glues that stabilize RNA–protein complexes
Small molecules can also affect the protein isoform generated from an mRNA by affecting alternative pre-mRNA splicing, with important proof-of-concept studies demonstrated with oligonucleotides [66]. The most extensive efforts have centered on alleviation of SMA, culminating in therapies such as risdiplam [8]. In SMA, the survival motor neuron 1 (SMN1) gene is defective, resulting in loss of SMN1 protein. Fortuitously, humans have a second SMN gene, SMN2, that varies by a single nucleotide, or single nucleotide polymorphism (SNP). This SNP changes SMN2’s splicing pattern, causing exon exclusion and producing a protein isoform with a reduced half-life. Risdiplam directs the splicing of the SMN2 gene to include SMN2 exon 7, affording the stable protein isoform that can compensate for loss of SMN1 [67].
Small molecules that influence splicing have been most commonly discovered by phenotypic screens and generally act by stabilizing RNA-protein interfaces [68]. More recently, drug/probe discovery efforts have focused on RNA-specific screening, as was the case for the discovery of the ‘5’ splice site bulge repair’ compound that directs the inclusion of SMN2 exon 7 [69].
Small molecules can also influence the protein isoforms generated from a transcript by effecting exon exclusion, as demonstrated for microtubule-associated protein tau (MAPT) [70, 71, 72]. In frontotemporal dementia with Parkinsonism-17, a splice site mutation enhances the interaction of a regulator element in MAPT with U1 snRNA, causing inclusion of exon 11 and an aggregation-prone protein isoform [73]. Small molecules that stabilize the mutated structure facilitate exon exclusion, shifting splicing toward the protein isoform that does not aggregate [70].
Targeting internal ribosome entry sites and G-quadruplexes
Small molecules can also alter the proteome by binding and occluding important structures in the encoding transcript that regulate protein expression. In particular, targeting internal ribosome entry sites sallows for gene-specific regulation of translation, for example, the internal ribosome entry site in c-MYC [74]. Likewise, inhibition of translation can be achieved by stabilization of other structures in the 5’ untranslated region (UTR), preventing cap-dependent translation. Of these UTR targets, G-quadruplex (G4) structures are often targeted as they are incredibly stable and are found in oncogenes KRAS [75], metalloproteinase 10 [76], NRAS [77], telomeric repeat-containing RNA [78,79], and others.
Small molecules that target metastasis-associated lung adenocarcinoma transcript 1, a long noncoding RNA
Long noncoding (lnc) RNAs are an emerging class of targets with a diverse array of functions, including epigenetic regulation, polycomb formation, nuclear localization, and others. A notable lnc RNA target, metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), causes tumorigenesis by forming a highly stable triple helix structure that enables nuclear localization and results in constitutive function. Indeed, small molecules that bind the triple helix reduce oncogenic potential [80].
Genome-wide design: fully functionalized fragments
Fragment-based drug discovery has been a key technology in the protein-targeting field. Unlike traditional screening methods of large small-molecule libraries (~106+), fragment-based approaches use a much smaller number (~103) of low-molecular-weight compounds. Indeed, a recent study combined fragment-based ligand discovery with a cross-linking and mass spectrometry strategy that uses fully functionalized fragments (FFFs) equipped with an alkyne and a photoreactive diazirine [81]. The strategy enabled study of target specificity for each fragment in living cells and allowed for optimization of highly homologous proteins while reducing the resources required for fruitful screening efforts.
This FFF screening strategy was applied to RNA targets using pre-miR-21 as a test case. In brief, the library was screened in the presence of a previously developed Dicer site–binding molecule to identify FFFs that bind other sites on the target RNA [82]. One FFF bound cooperatively with the RNA-binding module, the tethering of which afforded a low-molecular-weight dimer with improved affinity and specificity over its individual counterparts, validated by experiments that measured RNA target occupancy in cells and RNA-seq. Collectively, these studies provide a facile route to lead molecule optimization without substantially increasing the molecule’s molecular weight and hence maintaining druglikeness.
Moving forward, the cellular screening of FFFs against RNA could enable rapid targeting of functional sites in the RNA’s structure, simplifying the pipeline for transcriptome-wide targeting of RNA; that is, functionalized fragment screening of RNA targets strengthens structure-based targeting platforms by directly mapping small-molecule binding.
Affecting RNA biology: induced proximity
In addition to inhibiting RNA function through structure-specific targeting of tandem binding pockets, fragment assembly has enormous potential to enable design of small molecules with MOAs outside simple binding; that is, a structure-specific RNA-binding fragment can be coupled to a fragment that recruits enzymes or proteins that act on RNA, referred to as induced proximity. Originally developed for protein-targeted ligands by recruiting ubiquitin transferase [83] to target proteins for degradation, ‘protein targeting chimeric molecules,’ or PROTACs [84], were adopted for targeted RNA-induced degradation and similarly named ‘ribonuclease targeting chimeras’ or RIBOTACs [62,85,86]. These chimeric molecules comprise an RNA-targeting molecule attached to a molecule that selectively recruits ribonuclease (RNase) L to the desired RNA target. A miR-21–targeting RIBOTAC had enhanced activity and selectivity as compared with the parent binding molecule and selectively altered the proteome, short-circuiting oncogenic molecular pathways in triple-negative breast cancer cells and a mouse model of triple-negative breast cancer. The observed enhancement in selectivity was likely the combined effect of the RNA binder and the inherent substrate preferences of the RNase. A recent study showed that selectivity could be engineered into a promiscuous protein-binding small molecule by its conversion into a PROTAC [87]. Overall, these proof-of-concept studies of induced proximity directed at RNA targets illustrate the ability to recruit a cellular factor with desired function to a specific RNA using a small molecule. Importantly, induced proximity eliminates the requirement of RNA-targeted small molecules to bind functional sites. Although not induced proximity, a recent study showed that a bifunctional small molecule comprising an RNA binder and an inhibitor of Dicer blocked miR-21 biogenesis [88], an interesting approach that could be applied to other miRNAs.
Induced proximity has immense potential as other cellular factors could be recruited to specific RNA structures with a chimeric small molecule, expanding possible compound MOAs (Fig. 4). For example, induced proximity could be used to introduce site-specific modifications by recruiting adenosine deaminases that act on RNA (ADARs), supported by studies in which a guide RNA, delivered by an adeno-associated virus (AAV) vector, specifically edited an RNA target [89]. RNA lifetime could be altered by recruiting the polyadenylation machinery, deadenylating enzymes, terminal U transferase (TUTase) which targets an RNA for degradation, decapping enzymes, and so on. Induced proximity could unveil exciting new functions for RNA and further our understanding of RNA biology for therapeutic benefit.
Figure 4: Chemical Induced Proximity Expands the Mode of Action of RNA-targeted Small Molecules.
Using computational prediction and a fully functionalized fragment (FFF) screening strategy, structure-specific dimeric molecules can be used to reveal RNA function and downstream networks (red binding sites), for example the inhibition of miR-377 biogenesis[64]. In addition, induced proximity can be used to alter RNA lifetime (degradation by RNase L or TUTase[62,85] or change its sequence using editing enzymes such as ADAR (green), which could change the sequence of the encoded protein or alter splice site selection. Collectively, induced proximity has the potential to impart new modes of action for RNA-targeting small molecules.
Summary and outlook
There is vast potential to alter the proteome by targeting RNA, not only for RNA-related diseases but also for ‘undruggable’ proteins. RNA’s limited number of building blocks and diverse folding landscapes come with unique and challenging problems. However, its hierarchical folding and the accurate modeling thereof can be exploited to design lead molecules. With additional methodology such as Chem-CLIP and Chem-CLIP Fragment Mapping (Chem-CLIP-Frag-Map), small-molecule specificity can be directly measured in cells, validating the target and informing off-target effects. These methodologies can be synergistically combined into a platform for drugging the RNA transcriptome by sequence alone.
As genome-wide association studies provide a stream of RNA targets, many questions remain, such as how relatively small changes in sequence or repeat length cause large changes in structure [90], the uniqueness of RNA structures throughout the transcriptome, if small molecules can lock dynamic structures in a cellular RNA into a single conformation, and so on. Induced proximity could be an invaluable tool for future RNA research. While initial work demonstrated that removing a specific RNA through the recruitment of an RNase is indeed feasbile, induced proximity could provide a more extensive understanding of RNA localization, modification, lifetime, and epigenetic effects. Indeed, recent studies on RNA methylation [91] and sequence modification [89] illustrate its biological diversity. As we anticipate the discovery of new and important RNA functions, we believe that RNA-targeted therapies will become a pillar of small-molecule development.
Acknowledgements
Funding was provided by the National Institutes of Health (R01 CA249180, P01 NS09914, R35 NS116846, and UG3 NS116921 to MDD), the Department of Defense (W81XWH-18-0718 and W81XWH-19-PRMRP-IIRA to MDD), and the Huntington’s Disease Society of America (JTB).
Footnotes
Declaration of competing interest
The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: MDD is a founder of Expansion Therapeutics.
REFERENCES
- 1.Santos R, Ursu O, Gaulton A, Bento AP, Donadi RS, Bologa CG, Karlsson A, Al-Lazikani B, Hersey A, Oprea TI, et al. : A comprehensive map of molecular drug targets. Nat Rev Drug Discov 2017, 16:19–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Esteller M: Non-coding RNAs in human disease. Nat Rev Genet 2011, 12:861–874. [DOI] [PubMed] [Google Scholar]
- 3.Taliaferro JM, Lambert NJ, Sudmant PH, Dominguez D, Merkin JJ, Alexis MS, Bazile C, Burge CB: RNA sequence context effects measured in vitro predict in vivo protein binding and regulation. Mol Cell 2016, 64:294–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Sun L, Fazal FM, Li P, Broughton JP, Lee B, Tang L, Huang W, Kool ET, Chang HY, Zhang QC: RNA structure maps across mammalian cellular compartments. Nat Struct Mol Biol 2019, 26:322–330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Langdon EM, Qiu Y, Ghanbari Niaki A, McLaughlin GA, Weidmann CA, Gerbich TM, Smith JA, Crutchley JM, Termini CM, Weeks KM, et al. : mRNA structure determines specificity of a polyQ-driven phase separation. Science 2018, 360:922–927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kwon SC, Baek SC, Choi YG, Yang J, Lee YS, Woo JS, Kim VN: Molecular basis for the single-nucleotide precision of primary microRNA processing. Mol Cell 2019, 73:505–518 e505. [DOI] [PubMed] [Google Scholar]
- 7.Nam Y, Chen C, Gregory RI, Chou JJ, Sliz P: Molecular basis for interaction of let-7 microRNAs with Lin28. Cell 2011, 147:1080–1091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Ratni H, Ebeling M, Baird J, Bendels S, Bylund J, Chen KS, Denk N, Feng Z, Green L, Guerard M, et al. : Discovery of risdiplam, a selective survival of motor neuron-2 ( SMN2) gene splicing modifier for the treatment of spinal muscular atrophy (SMA). J Med Chem 2018, 61:6501–6517. (**) The authors discovered risdiplam, a pharmacophology optimized compound, that directs SMN2 splicing to alleviate SMA. Through medicinal chemistry efforts, they achieved better potency, alleviated phototoxicity risk, lowered basicity, and prevented active metabolites. With these modifications, risdiplam successfully underwent clinical trials and garnered FDA-approval.
- 9.Stein CA, Castanotto D: FDA-approved oligonucleotide therapies in 2017. Mol Ther 2017, 25:1069–1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ganser LR, Kelly ML, Herschlag D, Al-Hashimi HM: The roles of structural dynamics in the cellular functions of RNAs. Nat Rev Mol Cell Biol 2019, 20:474–489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Ganser LR, Lee J, Rangadurai A, Merriman DK, Kelly ML, Kansal AD, Sathyamoorthy B, Al-Hashimi HM: High-performance virtual screening by targeting a high-resolution RNA dynamic ensemble. Nat Struct Mol Biol 2018, 25:425–434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Rizvi NF, Santa Maria JP Jr., Nahvi A, Klappenbach J, Klein DJ, Curran PJ, Richards MP, Chamberlin C, Saradjian P, Burchard J, et al. : Targeting RNA with small molecules: Identification of selective, RNA-binding small molecules occupying drug-like chemical space. SLAS Discov 2020, 25:384–396. (**) The authors use mass spectroscopy and an automated ligand system to screen a diverse chemical library for binding across 42 RNA targets across various classes/functions. These screening efforts were used to build an RNA-focused library containing ~3700 small molecules that occupy drug-like chemical space.
- 13.Jimenez-Moreno E, Montalvillo-Jimenez L, Santana AG, Gomez AM, Jimenez-Oses G, Corzana F, Bastida A, Jimenez-Barbero J, Canada FJ, Gomez-Pinto I, et al. : Finding the right candidate for the right position: A fast NMR-assisted combinatorial method for optimizing nucleic acids binders. J Am Chem Soc 2016, 138:6463–6474. [DOI] [PubMed] [Google Scholar]
- 14.Zuker M: Mfold web server for nucleic acid folding and hybridization prediction. Nucleic Acids Res 2003, 31:3406–3415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Andrews RJ, Roche J, Moss WN: ScanFold: An approach for genome-wide discovery of local RNA structural elements-applications to Zika virus and HIV. PeerJ 2018, 6:e6136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Mathews DH: Revolutions in RNA secondary structure prediction. J Mol Biol 2006, 359:526–532. [DOI] [PubMed] [Google Scholar]
- 17.Parisien M, Major F: The MC-Fold and MC-Sym pipeline infers RNA structure from sequence data. Nature 2008, 452:51–55. [DOI] [PubMed] [Google Scholar]
- 18.Velagapudi SP, Luo Y, Tran T, Haniff HS, Nakai Y, Fallahi M, Martinez GJ, Childs-Disney JL, Disney MD: Defining RNA-small molecule affinity landscapes enables design of a small molecule inhibitor of an oncogenic noncoding RNA. ACS Cent Sci 2017, 3:205–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Schneider TD, Stephens RM: Sequence logos: A new way to display consensus sequences. Nucleic Acids Res 1990, 18:6097–6100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Nettling M, Treutler H, Grau J, Keilwagen J, Posch S, Grosse I: Difflogo: A comparative visualization of sequence motifs. BMC Bioinformatics 2015, 16:387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hong C, Otabe T, Matsumoto S, Dohno C, Murata A, Hagihara M, Nakatani K: Formation of a ligand-assisted complex of two RNA hairpin loops. Chemistry 2014, 20:5282–5287. [DOI] [PubMed] [Google Scholar]
- 22.Dohno C, Nakatani K: Molecular glue for RNA: Regulating RNA structure and function through synthetic RNA binding molecules. Chembiochem 2019, 20:2903–2910. [DOI] [PubMed] [Google Scholar]
- 23.Velagapudi SP, Gallo SM, Disney MD: Sequence-based design of bioactive small molecules that target precursor microRNAs. Nat Chem Biol 2014, 10:291–297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Disney MD, Winkelsas AM, Velagapudi SP, Southern M, Fallahi M, Childs-Disney JL: Inforna 2.0: A platform for the sequence-based design of small molecules targeting structured RNAs. ACS Chem Biol 2016, 11:1720–1728. The authors improved a databse for RNA structures preferred to bind by a library of small molecules. In addition to data gathered from a library-vs library selection method, or 2DCS, this database includes all other reported information on RNA motif-small molecule binding parters. Together, these selected interactions form an optimized basis set to inform lead small molecules and lead RNA targets.
- 25.Su H, Peng Z, Yang J: Recognition of small molecule-RNA binding sites using RNA sequence and structure. Bioinformatics 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mitchell D 3rd, Assmann SM, Bevilacqua PC: Probing RNA structure in vivo. Curr Opin Struct Biol 2019, 59:151–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Paulson H: Repeat expansion diseases. Handb Clin Neurol 2018, 147:105–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Ondono R, Lirio A, Elvira C, Alvarez-Marimon E, Provenzano C, Cardinali B, Perez-Alonso M, Peralvarez-Marin A, Borrell JI, Falcone G, et al. : Design of novel small molecule base-pair recognizers of toxic CUG RNA transcripts characteristics of DM1. Comput Struct Biotechnol J 2021, 19:51–61. The authors used computational modeling to evaluate ligands that established a Janus-Wedge between the U•U bulge in r(CUG). They identified a pyrido[2,3-d]pyrimidine as as Janus-Wedge binder and optimized scaffolds to created a homodimeric r(CUG) binding molecule.
- 29.Angelbello AJ, Rzuczek SG, McKee KK, Chen JL, Olafson H, Cameron MD, Moss WN, Wang ET, Disney MD: Precise small-molecule cleavage of an r(CUG) repeat expansion in a myotonic dystrophy mouse model. Proc Natl Acad Sci U S A 2019, 116:7799–7804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Gareiss PC, Sobczak K, McNaughton BR, Palde PB, Thornton CA, Miller BL: Dynamic combinatorial selection of molecules capable of inhibiting the (CUG) repeat RNA-MBNL1 interaction in vitro: Discovery of lead compounds targeting myotonic dystrophy (DM1). J Am Chem Soc 2008, 130:16254–16261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Li J, Nakamori M, Matsumoto J, Murata A, Dohno C, Kiliszek A, Taylor K, Sobczak K, Nakatani K: A dimeric 2,9-diamino-1,10-phenanthroline derivative improves alternative splicing in myotonic dystrophy type 1 cell and mouse models. Chemistry 2018, 24:18115–18122. [DOI] [PubMed] [Google Scholar]
- 32.Lee J, Bai Y, Chembazhi UV, Peng S, Yum K, Luu LM, Hagler LD, Serrano JF, Chan HYE, Kalsotra A, et al. : Intrinsically cell-penetrating multivalent and multitargeting ligands for myotonic dystrophy type 1. Proc Natl Acad Sci U S A 2019, 116:8709–8714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hagler LD, Luu LM, Tonelli M, Lee J, Hayes SM, Bonson SE, Vergara JI, Butcher SE, Zimmerman SC: Expanded DNA and RNA trinucleotide repeats in myotonic dystrophy type 1 select their own multitarget, sequence-selective inhibitors. Biochemistry 2020, 59:3463–3472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Garcia-Lopez A, Llamusi B, Orzaez M, Perez-Paya E, Artero RD: In vivo discovery of a peptide that prevents CUG-RNA hairpin formation and reverses RNA toxicity in myotonic dystrophy models. Proc Natl Acad Sci U S A 2011, 108:11866–11871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Rzuczek SG, Colgan LA, Nakai Y, Cameron MD, Furling D, Yasuda R, Disney MD: Precise small-molecule recognition of a toxic CUG RNA repeat expansion. Nat Chem Biol 2017, 13:188–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Boger DL, Cai H: Bleomycin: Synthetic and mechanistic studies. Angew Chem Int Ed Engl 1999, 38:448–476. [DOI] [PubMed] [Google Scholar]
- 37.Carter BJ, de Vroom E, Long EC, van der Marel GA, van Boom JH, Hecht SM: Site-specific cleavage of RNA by FE(ii).Bleomycin. Proc Natl Acad Sci U S A 1990, 87:9373–9377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Angelbello AJ, DeFeo ME, Glinkerman CM, Boger DL, Disney MD: Precise targeted cleavage of a r(CUG) repeat expansion in cells by using a small-molecule-deglycobleomycin conjugate. ACS Chem Biol 2020, 15:849–855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Angelbello AJ, Chen JL, Disney MD: Small molecule targeting of RNA structures in neurological disorders. Ann N Y Acad Sci 2020, 1471:57–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Khan E, Tawani A, Mishra SK, Verma AK, Upadhyay A, Kumar M, Sandhir R, Mishra A, Kumar A: Myricetin reduces toxic level of CAG repeats RNA in Huntington’s Disease (HD) and spino cerebellar ataxia (SCAS). ACS Chem Biol 2018, 13:180–188. [DOI] [PubMed] [Google Scholar]
- 41.Khan E, Mishra SK, Mishra R, Mishra A, Kumar A: Discovery of a potent small molecule inhibiting Huntington’s Disease (HD) pathogenesis via targeting CAG repeats RNA and poly Q protein. Sci Rep 2019, 9:16872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Li J, Sakata A, He H, Bai LP, Murata A, Dohno C, Nakatani K: Naphthyridine-benzoazaquinolone: Evaluation of a tricyclic system for the binding to (CAG)n repeat DNA and RNA. Chem Asian J 2016, 11:1971–1981. [DOI] [PubMed] [Google Scholar]
- 43.Matthes F, Massari S, Bochicchio A, Schorpp K, Schilling J, Weber S, Offermann N, Desantis J, Wanker E, Carloni P, et al. : Reducing mutant huntingtin protein expression in living cells by a newly identified RNA CAG binder. ACS Chem Neurosci 2018, 9:1399–1408. [DOI] [PubMed] [Google Scholar]
- 44.Hagerman RJ, Leehey M, Heinrichs W, Tassone F, Wilson R, Hills J, Grigsby J, Gage B, Hagerman PJ: Intention tremor, parkinsonism, and generalized brain atrophy in male carriers of fragile X. Neurology 2001, 57:127–130. [DOI] [PubMed] [Google Scholar]
- 45. Green KM, Sheth UJ, Flores BN, Wright SE, Sutter AB, Kearse MG, Barmada SJ, Ivanova MI, Todd PK: High-throughput screening yields several small-molecule inhibitors of repeat-associated non-AUG translation. J Biol Chem 2019, 294:18624–18638. The authors discovered five small molecules that inhibition repeat-associated non-AUG (RAN) but not canonical translation of the RNA repeat expansion that causes FXTAS, r(CGG)exp. The activities of three of the molecules were traced to binding the RNA repeat expansion and interestingly also inhibit RNA translation from the structurally related repeat expansion that causes C9ALS/FTD.
- 46.Verma AK, Khan E, Mishra SK, Jain N, Kumar A: Piperine modulates protein mediated toxicity in fragile X-associated tremor/ataxia syndrome through interacting expanded CGG repeat r((CGG)exp) RNA. ACS Chem Neurosci 2019, 10:3778–3788. [DOI] [PubMed] [Google Scholar]
- 47.Verma AK, Khan E, Mishra SK, Mishra A, Charlet-Berguerand N, Kumar A: Curcumin regulates the r((CGG)exp) RNA hairpin structure and ameliorate defects in fragile X-associated tremor ataxia syndrome. Front Neurosci 2020, 14:295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Zhou B, Liu C, Geng Y, Zhu G: Topology of a G-quadruplex DNA formed by C9orf72 hexanucleotide repeats associated with ALS and FTD. Sci Rep 2015, 5:16673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Wang ZF, Ursu A, Childs-Disney JL, Guertler R, Yang WY, Bernat V, Rzuczek SG, Fuerst R, Zhang YJ, Gendron TF, et al. : The hairpin form of r((G4C2)exp) in C9ALS/FTD is repeat-associated non-ATG translated and a target for bioactive small molecules. Cell Chem Biol 2019, 26:179–190 e112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Simone R, Balendra R, Moens TG, Preza E, Wilson KM, Heslegrave A, Woodling NS, Niccoli T, Gilbert-Jaramillo J, Abdelkarim S, et al. : G-quadruplex-binding small molecules ameliorate C9orf72 FTD/ALS pathology in vitro and in vivo. EMBO Mol Med 2018, 10:22–31. (*) The authors identified an selective binder to RNA G-quadruplex formed by the RNA repeat expansion, r(G4C2)exp that caues C9ALS/FTD. Stabilization of the G-quadruplex structure by the small molecule was cofirmed by FRET and circular dichroism studies. These compounds were then shown to reduce RNA foci levels in c9ALS patient-derived iPSCs and a Drosophila model and levels of toxic dipeptide repeats in the former.
- 51.Wortman MJ, Dagdanova AV, Clark AM, Godfrey EW, Pascal SM, Johnson EM, Daniel DC: A synthetic Pur-based peptide binds and alters G-quadruplex secondary structure present in the expanded RNA repeat of C9orf72 ALS/FTD. Biochim Biophys Acta Mol Cell Res 2020, 1867:118674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Zamiri B, Reddy K, Macgregor RB Jr., Pearson CE: Tmpyp4 porphyrin distorts RNA G-quadruplex structures of the disease-associated r(GGGGCC)n repeat of the C9orf72 gene and blocks interaction of RNA-binding proteins. J Biol Chem 2014, 289:4653–4659. The authors show that the RNA repeat expansion that causes C9ALS/FTD folds into a G-quadruplex structure. The C9ALS/FTD quadruplex binds to the known G-quadruplex binding-ligand TmPyP4 and inhibits its interactions with various RNA-binding proteins. These studies were one of the first to show that the C9ALS/FTD repeat expansion is ligandable and hence might be a target for therapeutic intervention.
- 53.Connelly CM, Boer RE, Moon MH, Gareiss P, Schneekloth JS Jr.: Discovery of inhibitors of microRNA-21 processing using small molecule microarrays. ACS Chem Biol 2017, 12:435–443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Lorenz DA, Vander Roest S, Larsen MJ, Garner AL: Development and implementation of an HTS-compatible assay for the discovery of selective small-molecule ligands for pre-microRNAs. SLAS Discov 2018, 23:47–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Staedel C, Tran TPA, Giraud J, Darfeuille F, Di Giorgio A, Tourasse NJ, Salin F, Uriac P, Duca M: Modulation of oncogenic miRNA biogenesis using functionalized polyamines. Sci Rep 2018, 8:1667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Vo DD, Becquart C, Tran TPA, Di Giorgio A, Darfeuille F, Staedel C, Duca M: Building of neomycin-nucleobase-amino acid conjugates for the inhibition of oncogenic miRNAs biogenesis. Org Biomol Chem 2018, 16:6262–6274. [DOI] [PubMed] [Google Scholar]
- 57. Matarlo JS, Krumpe LRH, Heinz WF, Oh D, Shenoy SR, Thomas CL, Goncharova EI, Lockett SJ, O’Keefe BR: The natural product butylcycloheptyl prodiginine binds pre-miR-21, inhibits Dicer-mediated processing of pre-miR-21, and blocks cellular proliferation. Cell Chem Biol 2019, 26:1133–1142 e1134. The authors used high throughput screening to identify natural products that bind the precursor of miR-21. They describe butyl-cyclohepyl prodiginine, which inhibits Dicer processing by binding pre-miR-21 with a 0.41μM Kd. They show that this compound modulates miR-21 processing, causing downregulation of downstream miR-21 targets PTEN and Pdcd4.
- 58.Naro Y, Ankenbruck N, Thomas M, Tivon Y, Connelly CM, Gardner L, Deiters A: Small molecule inhibition of microRNA miR-21 rescues chemosensitivity of renal-cell carcinoma to topotecan. J Med Chem 2018, 61:5900–5909. [DOI] [PubMed] [Google Scholar]
- 59.Jiang CS, Wang XM, Zhang SQ, Meng LS, Zhu WH, Xu J, Lu SM: Discovery of 4-benzoylamino-N-(prop-2-yn-1-yl)benzamides as novel microRNA-21 inhibitors. Bioorg Med Chem 2015, 23:6510–6519. [DOI] [PubMed] [Google Scholar]
- 60.Naro Y, Thomas M, Stephens MD, Connelly CM, Deiters A: Aryl amide small-molecule inhibitors of microRNA miR-21 function. Bioorg Med Chem Lett 2015, 25:4793–4796. [DOI] [PubMed] [Google Scholar]
- 61. Shortridge MD, Walker MJ, Pavelitz T, Chen Y, Yang W, Varani G: A macrocyclic peptide ligand binds the oncogenic microRNA-21 precursor and suppresses Dicer processing. ACS Chem Biol 2017, 12:1611–1620. Here, the authors show that peptide-based ligands, in particular a cyclic β-hairpin peptidomimeitc, can bind to the Dicer site of oncogenic pre-miR-21 specifically and inhibit its processing by Dicer in vitro and in cells. MiRNA precursors are likely particularly well suited for this type of modality that bind RNA hairpins and can be customized/designed to afford selective binding. Indeed, these studies also report the NMR solution structure, which could inform optimization of the lead molecule.
- 62. Costales MG, Aikawa H, Li Y, Childs-Disney JL, Abegg D, Hoch DG, Pradeep Velagapudi S, Nakai Y, Khan T, Wang KW, et al. : Small-molecule targeted recruitment of a nuclease to cleave an oncogenic RNA in a mouse model of metastatic cancer. Proc Natl Acad Sci U S A 2020, 117:2406–2411. (**) The authors show that ribonuclease targeting chimeras (RIBOTACs) can selectively target and degrade an oncogenic miRNA in cells and in a mouse model of metastatic breast cancer by recruiting endogenous RNase L. Importantly, the chimeras comprises a small molecule that binds oncogenic pre-miR-21 and a heterocycle, rather than 2’−5’A4, to recruit RNase L.
- 63.Diaz JP, Chirayil R, Chirayil S, Tom M, Head KJ, Luebke KJ: Association of a peptoid ligand with the apical loop of pri-miR-21 inhibits cleavage by Drosha. RNA 2014, 20:528–539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Haniff HS, Knerr L, Liu X, Crynen G, Bostrom J, Abegg D, Adibekian A, Lekah E, Wang KW, Cameron MD, et al. : Design of a small molecule that stimulates vascular endothelial growth factor a enabled by screening RNA fold-small molecule interactions. Nat Chem 2020, 12:952–961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Lipinski CA, Lombardo F, Dominy BW, Feeney PJ: Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev 2001, 46:3–26. [DOI] [PubMed] [Google Scholar]
- 66.Dominski Z, Kole R: Restoration of correct splicing in thalassemic pre-mRNA by antisense oligonucleotides. Proc Natl Acad Sci U S A 1993, 90:8673–8677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Naryshkin NA, Weetall M, Dakka A, Narasimhan J, Zhao X, Feng Z, Ling KK, Karp GM, Qi H, Woll MG, et al. : Motor neuron disease. SMN2 splicing modifiers improve motor function and longevity in mice with spinal muscular atrophy. Science 2014, 345:688–693. [DOI] [PubMed] [Google Scholar]
- 68.Salani M, Urbina F, Brenner A, Morini E, Shetty R, Gallagher CS, Law EA, Sunshine S, Finneran DJ, Johnson G, et al. : Development of a screening platform to identify small molecules that modify ELP1 pre-mRNA splicing in familial dysautonomia. SLAS Discov 2019, 24:57–67. [DOI] [PubMed] [Google Scholar]
- 69. Campagne S, Boigner S, Rudisser S, Moursy A, Gillioz L, Knorlein A, Hall J, Ratni H, Clery A, Allain FH: Structural basis of a small molecule targeting RNA for a specific splicing correction. Nat Chem Biol 2019, 15:1191–1198. (*) The authors describe a 5’ splice site bulge repairing small molecule. Further, a solution structure of the target RNA in complex with the splicing modifier revealed how the drug selectively promotes 5’ splicing. This thorough investigation of RNA structure stabilization illustrates the potential for RNA targeted splicing modifiers, enabling specific inclusion of SMN2 exon 7.
- 70.Chen JL, Zhang P, Abe M, Aikawa H, Zhang L, Frank AJ, Zembryski T, Hubbs C, Park H, Withka J, et al. : Design, optimization, and study of small molecules that target tau pre-mRNA and affect splicing. J Am Chem Soc 2020, 142:8706–8727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Lopez-Senin P, Gomez-Pinto I, Grandas A, Marchan V: Identification of ligands for the tau exon 10 splicing regulatory element RNA by using dynamic combinatorial chemistry. Chemistry 2011, 17:1946–1953. [DOI] [PubMed] [Google Scholar]
- 72.Zheng S, Chen Y, Donahue CP, Wolfe MS, Varani G: Structural basis for stabilization of the tau pre-mRNA splicing regulatory element by novantrone (mitoxantrone). Chem Biol 2009, 16:557–566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Brunden KR, Trojanowski JQ, Lee VM: Advances in tau-focused drug discovery for alzheimer’s disease and related tauopathies. Nat Rev Drug Discov 2009, 8:783–793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Vaklavas C, Meng Z, Choi H, Grizzle WE, Zinn KR, Blume SW: Small molecule inhibitors of IRES-mediated translation. Cancer Biol Ther 2015, 16:1471–1485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Miglietta G, Cogoi S, Marinello J, Capranico G, Tikhomirov AS, Shchekotikhin A, Xodo LE: RNA G-quadruplexes in kirsten ras (KRAS) oncogene as targets for small molecules inhibiting translation. J Med Chem 2017, 60:9448–9461. [DOI] [PubMed] [Google Scholar]
- 76.Dai J, Liu ZQ, Wang XQ, Lin J, Yao PF, Huang SL, Ou TM, Tan JH, Li D, Gu LQ, et al. : Discovery of small molecules for up-regulating the translation of antiamyloidogenic secretase, a disintegrin and metalloproteinase 10 (ADAM10), by binding to the G-quadruplex-forming sequence in the 5’ untranslated region (UTR) of its mRNA. J Med Chem 2015, 58:3875–3891. [DOI] [PubMed] [Google Scholar]
- 77.Katsuda Y, Sato S, Asano L, Morimura Y, Furuta T, Sugiyama H, Hagihara M, Uesugi M: A small molecule that represses translation of G-quadruplex-containing mRNA. J Am Chem Soc 2016, 138:9037–9040. [DOI] [PubMed] [Google Scholar]
- 78.Collie GW, Sparapani S, Parkinson GN, Neidle S: Structural basis of telomeric RNA quadruplex--acridine ligand recognition. J Am Chem Soc 2011, 133:2721–2728. [DOI] [PubMed] [Google Scholar]
- 79.Garavis M, Lopez-Mendez B, Somoza A, Oyarzabal J, Dalvit C, Villasante A, Campos-Olivas R, Gonzalez C: Discovery of selective ligands for telomeric RNA G-quadruplexes (TERRA) through 19F-NMR based fragment screening. ACS Chem Biol 2014, 9:1559–1566. [DOI] [PubMed] [Google Scholar]
- 80. Abulwerdi FA, Xu W, Ageeli AA, Yonkunas MJ, Arun G, Nam H, Schneekloth JS Jr., Dayie TK, Spector D, Baird N, et al. : Selective small-molecule targeting of a triple helix encoded by the long noncoding RNA, MALAT1. ACS Chem Biol 2019, 14:223–235. The authors show that small molecules can indeed selectively recognize structures formed by long noncoding RNAs and affect function, in this case the 3′ terminal stability element for nuclear expression (ENE) found in oncogenic MALAT1. Indeed, two compounds that bind the triple helix reduced MALAT1 levels in a mammary tumor model.
- 81.Parker CG, Galmozzi A, Wang Y, Correia BE, Sasaki K, Joslyn CM, Kim AS, Cavallaro CL, Lawrence RM, Johnson SR, et al. : Ligand and target discovery by fragment-based screening in human cells. Cell 2017, 168:527–541 e529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Suresh BM, Li W, Zhang P, Wang KW, Yildirim I, Parker CG, Disney MD: A general fragment-based approach to identify and optimize bioactive ligands targeting RNA. Proc Natl Acad Sci U S A 2020, 117:33197–33203. (*) The authors reported the adoption of the FFF approach to RNA ligands. They showed that FFF could be used to determine cooperative binding among heterodimeric compounds. Assembly of dimers improved affinity and specificity, while still occluding the function site within miR-21 required for biogenesis. These studies were then validated in cells with RNA-seq experiemts, directly confirming target RNA engagement.
- 83.Gosink MM, Vierstra RD: Redirecting the specificity of ubiquitination by modifying ubiquitin-conjugating enzymes. Proc Natl Acad Sci U S A 1995, 92:9117–9121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Sakamoto KM, Kim KB, Kumagai A, Mercurio F, Crews CM, Deshaies RJ: Protacs: Chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradation. Proc Natl Acad Sci U S A 2001, 98:8554–8559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Costales MG, Matsumoto Y, Velagapudi SP, Disney MD: Small molecule targeted recruitment of a nuclease to RNA. J Am Chem Soc 2018, 140:6741–6744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Costales MG, Suresh B, Vishnu K, Disney MD: Targeted degradation of a hypoxia-associated non-coding RNA enhances the selectivity of a small molecule interacting with RNA. Cell Chem Biol 2019, 26:1180–1186.e1185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Bondeson DP, Smith BE, Burslem GM, Buhimschi AD, Hines J, Jaime-Figueroa S, Wang J, Hamman BD, Ishchenko A, Crews CM: Lessons in PROTAC design from selective degradation with a promiscuous warhead. Cell Chem Biol 2018, 25:78–87.e75. These pioneering studies showed that the selectivity of a promiscuous kinase inhibitor (affects >100 kinases) can be vastly improved by its conversion into a proteolysis targeting chimera, or Protac. Further, these studies showed that Protac activity is traced to stabilization of the ternary complex formed by the Protac, target protein, and recruited enzyme.
- 88.Yan H, Liang FS: miRNA inhibition by proximity-enabled Dicer inactivation. Methods 2019, 167:117–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Qu L, Yi Z, Zhu S, Wang C, Cao Z, Zhou Z, Yuan P, Yu Y, Tian F, Liu Z, et al. : Programmable RNA editing by recruiting endogenous ADAR using engineered RNAs. Nat Biotechnol 2019, 37:1059–1069. [DOI] [PubMed] [Google Scholar]
- 90.Wan Y, Qu K, Zhang QC, Flynn RA, Manor O, Ouyang Z, Zhang J, Spitale RC, Snyder MP, Segal E, et al. : Landscape and variation of RNA secondary structure across the human transcriptome. Nature 2014, 505:706–709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Liu J, Dou X, Chen C, Chen C, Liu C, Xu MM, Zhao S, Shen B, Gao Y, Han D, et al. : N (6)-methyladenosine of chromosome-associated regulatory RNA regulates chromatin state and transcription. Science 2020, 367:580–586. [DOI] [PMC free article] [PubMed] [Google Scholar]