Abstract
Ribonucleases (RNases) cleave and process RNAs, thereby regulating the biogenesis, metabolism, and degradation of coding and noncoding RNAs. Thus, small molecules targeting RNases have the potential to perturb RNA biology, and RNases have been studied as therapeutic targets of antibiotics, antivirals, and agents for autoimmune diseases and cancers. Additionally, the recent advances in chemically induced proximity approaches have led to the discovery of bifunctional molecules that target RNases to achieve RNA degradation or inhibit RNA processing. Here, we summarize the efforts that have been made to discover small-molecule inhibitors and activators targeting bacterial, viral, and human RNases. We also highlight the emerging examples of RNase-targeting bifunctional molecules and discuss the trends in developing such molecules for both biological and therapeutic applications.
Introduction
Ribonucleases (RNases) are RNA-cleaving proteins that regulate the metabolism of RNAs. The two main classes of RNases are endoribonucleases and exoribonucleases. In humans, RNases belong to a superfamily constituted of at least 122 proteins that not only act as simple RNA degraders but are also involved in essential nondigestive cellular functions involved in immune response, RNA maturation, and angiogenesis.1−3 Distinction of these processes relies on the differences in RNA-substrate sequence of RNases and specificity for single- or double-stranded RNAs. RNases either function in a processive manner or cleave their substrate only once after binding.4 The nucleases are master regulators of RNA-dependent pathways and play indispensable roles in RNA biogenesis.5,6 Therefore, small molecules modulating RNase activities are useful tools for studying the regulatory mechanisms involving both human and pathogenic RNases and are potential candidates for the development of therapeutics. Representative examples of such small molecules include inhibitors of viral RNases influenza polymerase acidic (PA) endonuclease and human immunodeficiency virus (HIV) RNase H, and several human RNases. The potential of RNases as therapeutic targets was discussed in a previous opinion article;1 however, a review summarizing the scattered examples of these RNase-targeting small molecules is currently missing. To note, in addition to small molecules, RNases have been targeted by the emerging class of proximity-inducing bifunctional molecules for various chemical biology applications. Therefore, in this Review, we highlight two aspects of the current landscape in RNase targeting: small molecules for the development of therapeutics and bifunctional molecules that modulate RNA biogenesis and metabolism (Figure 1). The former aspect is divided into three target families, bacterial RNases, viral RNases, and human RNases. We also discuss trends and future directions to explore RNases as drug targets for small and bifunctional molecules.
Figure 1.
Current RNase-targeting strategies using small molecules and bifunctional molecules. (A) Small-molecule modulators include inhibitors that bind to the RNase active site, inhibitors and activators that bind to allosteric sites, or binders to adjacent domains. (B) Described bifunctional modulators enable targeted RNA degradation by the recruitment of RNase L or inhibition of Dicer-mediated RNA cleavage by coupling a weak Dicer inhibitor to an RNA binder.
SMALL-MOLECULE INHIBITORS OF BACTERIAL RNASES
RNase P
Bacterial ribonuclease P (RNase P) is a unique RNase since it is a ribonucleoprotein functioning as a ribozyme. RNase P catalyzes the maturation of tRNA (tRNA) by the cleavage of the 5′ end of precursor tRNA. Inhibitors of RNase P enzyme function, which is crucial for bacterial survival, could potentially serve as antibiotics. The only FDA-approved inhibitors of RNase P to date are antibiotic aminoglycosides such as neomycin. To note, RNase P inhibition is, however, not the sole mechanism of action for aminoglycoside antibiotics since their interaction with rRNA is known to cause errors in translation.7 The development of fluorescence real-time tRNA cleavage assays enabled increased throughput screening for inhibitors.8,9 A fluorescence polarization assay using a fluorophore at the 5′-end of the precursor tRNA confirmed neomycin B as an RNase P inhibitor (Figure 2). A screening of 2880 compounds discovered iriginol hexaacetate that inhibited RNase P with an IC50 of 0.8 μM (Figure 2).8 A Förster resonance energy transfer (FRET) assay-based screening retrieved purpurin as an RNase P inhibitor (Figure 2). Purpurin bound to RNase P with a KD of 13 μM and its binding to the protein component of the enzyme was confirmed by crystallization of a purpurin-RNase P complex.9 Purpurin carries a three-oxygen pharmacophore that was often observed to coordinate the divalent metal ions of RNases.9,10 However, data on the antibacterial effect of purpurin was not reported.9 Additionally, RNPA2000 was reported to show weak RNase P inhibitory activity (IC50: 125 μM, Figure 2).11 A concern for the identified RNase P inhibitors, including RNPA2000, iriginol hexaacetate, and purpurin, was that they acted as aggregators and are likely unspecific RNase P inhibitors.12,13 In addition to small molecules, a FRET assay was employed to evaluate rationally designed and modified oligonucleotides as inhibitors of RNase P. Antisense oligonucleotides targeting the RNA component of RNase P were coupled to cell-penetrating peptides to yield conjugates that inhibited bacterial growth. The best-performed conjugates showed IC50 values of ∼100 nM, being the most potent RNase P inhibitors reported to date.14
Figure 2.
Small-molecule inhibitors of bacterial RNase P (the red color indicates the metal-chelating pharmacophore of purpurin).
RNase E
RNase E is another bacterial RNase that functions as an endonuclease involved in ribosomal maturation and RNA turnover. RNase E is a central component in the formation of the degradosome complex in E. coli, and small-molecule inhibitors were identified with the aim of studying the cellular functions of RNase E. A virtual screening predicted small-molecule binders of RNase E, which were evaluated by biochemical assays and SPR, albeit with weak millimolar activities.15 In summary, although inhibitors against the bacterial RNases such as RNase P and E are potential antibiotics, no potent and selective small-molecule inhibitors have been described so far. Even for the reported inhibitors listed here, their effects in inhibiting bacterial growth mainly were not reported or investigated.
SMALL-MOLECULE INHIBITORS TARGETING VIRAL RNASES
Small molecules have been reported for viral ribonucleases as antiviral therapeutics, such as those targeting HIV and HBV RNase H, influenza virus PA endonuclease, and the RNases nsp-14-ExoN and nsp15 of SARS-CoV-2.16−18
RNase H
RNase H is part of viral reverse transcriptase that removes RNA template strands to allow the synthesis of double-stranded DNA from viral genomic RNA, leading to the incorporation of the double-stranded DNA into the host genome by integrases.19,20 To date, HIV is treated by a combination of drugs in highly active antiretroviral therapy (HAART) and all HIV enzymatic activities except RNase H can be targeted effectively.21,22 Despite numerous studies aiming to identify RNase H inhibitors, current FDA-approved HIV-1 reverse transcriptase inhibitors all target the polymerase activity, but not the RNase H activity of the enzyme.23,20 RNase H has been extensively studied as a drug target already since the 1990s, as reviewed recently by Tramontano et al.16,24−26 Screenings for RNase H inhibitors were historically performed by denaturing gel-based assays with isotope-labeled RNA substrates in limited throughput.24,25,27 More recent studies used fluorescent assays to identify active compounds, e.g., a FRET assay using an RNA-DNA hybrid in which the DNA was labeled at both ends with a fluorophore quencher pair. Hydrolysis of the RNA led to DNA-hairpin formation and induced fluorescence quenching.28 Another assay used a fluorophore-labeled DNA annealed with a quencher-labeled RNA, leading to a fluorescent signal upon RNA cleavage.29
In general, three classes of RNase H inhibitors were described, metal-chelating active-site binders, allosteric inhibitors, and dual inhibitors against both reverse transcription and RNase activities.16 Active-site binding molecules mostly carry a three-oxygen pharmacophore that allows chelation of the magnesium ions of the DEDD (Asp-Glu-Asp-Asp) family endonuclease RNase H. Small molecules with diverse scaffolds have been identified as active-site inhibitors and evaluated in structure–activity relationship studies to optimize affinities (Figure 3). Most molecules were reported to show low micromolar IC50 values in biochemical assays, with some showing nanomolar potency.16 Several molecules, such as the pyrimidinol carboxylic acid 11 that showed an IC50 of 0.18 μM against HIV RNase H domain did not inhibit viral replication in HIV infectivity assays.30 Complex structures of HIV RNase H domain and small molecules have been solved and clearly showed the binding mode involving the three-oxygen pharmacophore interacting with the metal ions.16,31−33 The RNase H domain active site is a shallow pocket difficult to be drugged without metal chelation.16 Active-site analysis revealed homology to the catalytic center of HIV integrase that contains two divalent metal ions and is responsible for inserting viral DNA into host genomic DNA.34 Modification of integrase inhibitors led to the identification of dual inhibitors of RNase H and HIV integrase, such as one of the first described RNase H inhibitors, 4-[5-(benzoylamino)thien-2-yl]-2,4-dioxobutanoic acid (BTDBA).35,36 Dual inhibitors against two enzymatic functions of the same virus boosted the activity of the molecules against HIV replication and thus were further investigated.36 One of the most potent HIV RNase H domain inhibitors reported to date is the N-hydroxypyrimidinedione 45 with an IC50 of 25 nM against RNase H and an IC50 of 21 nM against HIV integrase. Compound 45 showed an EC50 of 15 nM in an antiviral replication assay.37 Another promising RNase H inhibitor is the N-hydroxypyrimidinedione 13j with an IC50 of 5 nM against RNase H, ∼1000-fold less activity against HIV integrase (IC50 4 μM), and an EC50 of 7.7 μM in the antiviral replication assay. The large difference in biochemical and cellular activities could be explained by high substrate abundance and the fact that small molecules could only bind to the RNase H domain while substrate binding is highly dependent on its interaction with the polymerase domain. Thus, it is a challenge for small molecules to outcompete the substrates to achieve potent inhibition.38,39 Active-site inhibitors of HIV RNase H were tested for their inhibitory activity against HBV RNase H, given that the HIV and HBV RNase H share 23% amino acid sequence homology. Among compounds that showed moderate activity, the N-hydroxypyridinediones were investigated in a further SAR study.28,40 Other reported HBV RNase H inhibitors either showed weak inhibitory potency or suffered from cytotoxicity.40−42
Figure 3.
Structures of inhibitors of HIV RNase (the red color indicates metal-chelating pharmacophore).
While active-site RNase H inhibitors all carry a chelating-triad pharmacophore, inhibitors without this motif were characterized by crystallography and NMR as allosteric inhibitors. Among them was the acylhydrazone BHMP03 (IC50 0.4 μM) binding to the substrate handle region of RNase H.16 Vinylogous ureas such as NSC727447 were identified as allosteric RNase H inhibitors by screening 230,000 natural compounds and led to the development of 3′,4′-dihydroxyphenyl-containing thienopyrimidinones (compound 9 and analogues) with submicromolar activity in biochemical assays and low micromolar activities in antiviral replication assays.16,43 Further SAR studies led to the identification of benzothienooxazinone compound 22 as a dual inhibitor of RNase H and reverse transcriptase activities of the HIV enzyme with IC50 values of 0.53 and 2.90 μM, respectively.44 Other dual inhibitors were based on the structure of the dihydroxy benzoyl naphthyl hydrazone DHBNH but did not show improved activity in comparison with that of compound 22.45,46
Nsp14 and Nsp15
Recently, the first inhibitors targeting SARS-CoV-2 ribonucleases nsp14 and nsp15 were reported. Nsp15 is an endoribonuclease cleaving RNA 3′ of uridines to prevent host recognition of the virus. Virtual screening of approved drugs and drugs under investigation for approval identified dutasteride and tasosartan as inhibitors of nsp15, but biochemical assays revealed only 40% inhibition at 600 μM concentrations.17 Further screenings led to the identification of kinase inhibitor NSC95397 as an inhibitor of ribonuclease nsp15 with micromolar activity but without effect in antiviral replication assays (Figure 4A).47−49 Uracil derivative tipiracil is a low-micromolar (IC50 7.5 μM) nsp15 inhibitor with antiviral activity.50
Figure 4.
(A) Structure of small-molecule inhibitors of nsp14 and nsp15 of SARS-CoV-2 and PA endonuclease. The red-colored structure indicates the metal-chelating pharmacophore. (B) Crystal structure of baloxavir acid bound to the PA endonuclease of influenza A virus showing the interaction of the compound with the metal ions (top) and a surface representation (bottom) (PDB 6FS6).
Nsp14 is a proofreading exonuclease enhancing replication fidelity, rendering nucleoside analogues ineffective as drugs against SARS-CoV-2. Nsp14 inhibition was proposed as a strategy to enable nucleoside analogues to be active.18 First reports of nsp14 drug discovery efforts identified micromolar inhibitors from a FRET assay using fluorophore-labeled annealed RNA strands as a probe. Nsp14 was identified to belong to the DEDD superfamily of ribonucleases harboring divalent metal ions.51 Thus, it is unsurprising that one of the identified inhibitors, compound 79, harbored the chelating triad pharmacophore as described for RNase H inhibitors (4).52 A SAMDI assay screening of 10,240 small molecules detecting RNA-cleavage by mass spectrometry led to the identification of an nsp14 inhibitor with an IC50 of 5.7 μM.53 Fragments with potential allosteric binding mode were discovered by fragment screening and could serve as a starting point for future nsp14 inhibitors.54
PA Endonuclease
Another well-studied viral nuclease target is the influenza virus PA protein possessing endonuclease activity that was reported to be active against RNA and DNA substrates. It cleaves host-cell mRNA caps that are then used as primers for the transcription of viral mRNA.55,56 The enzymatic function is crucial for viral replication, which makes the influenza PA endonuclease a prime target for antiviral therapeutics. Like RNase H, PA endonuclease has two crucial divalent metal ions (Mn2+ or Mg2+) in its active site.56 Therefore, structures of reported inhibitors of influenza PA endonuclease are similar to inhibitors of viral RNase H. Nearly all inhibitors harbor a three-oxygen metal-chelating pharmacophore combined with hydrophobic moieties (Figure 4A).57 The metal-chelating pharmacophore for inhibition of the cap-snatching endonuclease was identified via screening already in the 1990s. One identified inhibitor was L-735,882 which inhibited influenza endonuclease with an IC50 of 1.1 μM.58 SAR exploration improved the activities (IC50 < 1 μM).57 Recently, a fragment-based approach starting with metal-chelating compound 1 led to the discovery of compound 23 which displayed an IC50 of 47 pm. However, the measured EC50 of compound 23 in the antiviral replication assay was in the low micromolar range.59 The most successful PA endonuclease inhibitor is baloxavir marboxil, which the US FDA approved in 2018 for the treatment of the influenza virus. Baloxavir was developed based on a rational design from the approved HIV integrase inhibitor dolutegravir (Figure 4), as the HIV integrase active site shares structural similarity with influenza PA.60,61 Baloxavir marboxil is a prodrug derived from the potent inhibitor baloxavir acid by shielding the hydroxyl group to increase bioavailability.60,62,63 Baloxavir acid had single-digit nanomolar or subnanomolar potency against 22 different influenza A and B strains and showed an improved effect in clinical trials compared with the results induced by the treatment of the neuraminidase inhibitor oseltamivir.64
Taken together, viral nucleases have been studied as antiviral drug targets for more than three decades. The influenza endonuclease inhibitor baloxavir is a representative example that has been approved for antiviral clinical usage. Besides influenza endonuclease, small molecules have been reported for the HIV RNase H, but still with limited potency in inhibiting virus replication, so RNase H remains the only HIV enzymatic function that has not been successfully addressed clinically. The Covid-19 pandemic promoted the study of RNases of SARS-CoV-2, and emerging inhibitors targeting nsp 14 and nsp15 are being reported.
SMALL MOLECULES TARGETING HUMAN RIBONUCLEASES
Human ribonucleases are essential effectors in various cellular pathways responsible for immune responses, apoptosis, and inflammation. Thus, the modulation of human RNases with small molecules has been studied as a promising strategy for developing therapeutics and various biological applications. For example, RNase A family ribonucleases RNase 1 and Angiogenin are secretory enzymes involved in host defense, tightly regulated by RNase inhibitor protein.65,66 First reported RNase A family inhibitors were derived from nucleotide structures and showed nanomolar activities, followed by a few small-molecule inhibitors with moderate activities.67−69
Dicer
The RNases Drosha and Dicer are central regulators of the maturation of noncoding RNAs. The enzymes specifically cleave primary and precursor miRNA transcripts to generate mature miRNAs. Both Drosha and Dicer are conserved processors of canonical miRNAs. No small-molecule inhibitor of Drosha has been reported to date. Small-molecule modulators of Dicer activity allow studying Dicer biology or serve as starting points for the development of anticancer agents. To identify Dicer inhibitors, a fluorescence quenching assay using double-stranded fluorophore- and quencher-labeled RNA substrate was developed to monitor Dicer activity, which identified the aminoglycoside kanamycin that inhibited Dicer processing by 40% at a concentration of 100 μM.70
Caf1 and PARN
Ribonucleases involved in mRNA post-transcriptional regulation and turnover are mRNA deadenylases such as Caf1/CNOT7, which is part of the deadenylating Ccr4-Not complex, and Poly(A)-specific ribonuclease (PARN). Both enzymes have two divalent metal ions in the catalytic site coordinated by a DEDD motif. Caf1 inhibitors were discovered to develop probes to assist the biological understanding of the RNase. A FRET assay employing a fluorophore-labeled RNA probe was applied. The RNA probe hybridized with a fluorescently labeled DNA when it was not cleaved by Caf1.71 The most potent Caf1 inhibitors with low micromolar to submicromolar IC50 values were inspired by HIV RNase H inhibitors and harbor the metal chelating three-oxygen pharmacophore, such as compound 8j that also showed activity against PARN (Figure 5).72 Other published PARN inhibitors are mainly nucleoside analogues and aminoglycosides.73−76
Figure 5.
Structures of inhibitors of human RNases Caf1 and RNase H2 (the red color indicates the metal-chelating pharmacophore).
RNase H2
The RNase H2 is a DEDD superfamily metalloenzyme that cleaves the RNA strand of DNA–RNA duplexes.77 RNase H dysregulation in humans is associated with the genetic autoimmune disease Aicardi-Goutières syndrome (AGS) that causes neurological dysfunction early during infancy.78 A screening of 47,520 compounds employing a fluorescent assay using fluorophore- and quencher-labeled RNA-DNA duplex substrates led to the identification of isothiazolidinone R11/ebsulfur with an IC50 of 0.02 μM against RNase H2 (Figure 5).79
RNase L
The latent ribonuclease (RNase L) is another human RNase implicated in AGS. RNase L is endogenously expressed in human cells and is central to innate immune and antiviral responses.3 The oligoadenylate synthase recognizes and binds double-stranded RNA upon viral infections and produces 2′-5′-linked oligoadenylates (2′-5′A).80 2′-5′A bind to RNase L to induce dimerization to form the catalytically active, dimeric form of RNase L.81 Activated RNase L cleaves host and viral single-stranded RNA leading to a global translational arrest.82,83 RNase L consists of an N-terminal ankyrin repeat domain which is mainly responsible for 2′-5′A binding, a catalytically inactive kinase domain that binds ATP, and a C-terminal ribonuclease domain that cleaves the substrate RNA upon dimerization.81 Both small-molecule inhibitors and activators of RNase L have been reported (Figure 6). Inhibitors could serve as candidates for the treatment of AGS, while activators were developed as antiviral compounds.84,85 The kinase domain of RNase L shares high sequence homology with that of dsRNA-dependent protein kinase R (PKR) and inositol-requiring enzyme 1α (IRE1), and therefore, reported RNase L inhibitors share structural similarity of small-molecule kinase inhibitors that target the kinase domain, such as the FDA-approved receptor tyrosine kinase inhibitor sunitinib.86 Sunitinib was tested with varied IC50 values against RNase L ranging from 1.4 to 33 μM and showed an enhanced effect of oncolytic viruses against tumors in mouse models.87−89 The complex structure of sunitinib and RNase L confirmed the binding at the kinase domain and suggested dimer destabilization as the inhibition mechanism. Exchange of the fluorine substituent to chlorine improved the inhibitory activity by ∼4 fold.89 Screenings of 500 kinase inhibitors and 840 fragments using a FRET-based assay employing a single-stranded RNA probe labeled with a fluorophore quencher pair resulted in the identification of ellagic acid (IC50 73.5 nM) and hyperoside (IC50 1.63 μM) as RNase L inhibitors.84,90 The discovery of RNase L activators was based on similar FRET assays used to evaluate inhibitors, albeit without the addition of the natural activator 2′-5′A.91−94 First, thiophenone C1 and thienopyrimidinone C2 were retrieved as RNase L activators out of a library of 30,000 small molecules with EC50 of 26 and 22 μM, respectively, with a proposed activating mechanism by binding to the 2′-5′A-binding site.91 A more recent study optimized the thiophenone scaffold of C1 and yielded compound C1–3 that showed 48% activation at 130 μM compared with 2′-5′A at 100 nM.92 Combination of the 2-aminothiopheonone scaffold of C1–3 with that of the pyrrole scaffold of sunitinib led to RNase L inhibitors with a hybridized scaffold.95 An extensive SAR study focusing on the aminothiophene core scaffold and a screening of 240,000 small molecules resulted in small-molecule binders that stabilized RNase L but without significant improvement on the RNase L activating potency.93,94 Very recently, compound 2 was identified by DNA-encoded library-based screening as an RNase L activator that induced RNase L dimerization in micromolar potency.96
Figure 6.
Small molecules targeting human RNases. RNase L inhibitors (gray background), RNase L activators (green background), and IRE1 modulators (blue background). The full-length structure of RNase L is shown (PDB 4O1P) together with the kinase and RNase domains of IRE1 (PDB 6W3C).
IRE1
The serine/threonine-protein kinase/endoribonuclease IRE1 regulates the unfolded protein response located on the membrane of the endoplasmic reticulum (ER).84 IRE1 consists of an N-terminal ER-lumenal domain involved in the unfolded-protein detection, a transmembrane region, a kinase domain, and an RNase domain.97 Kinase and RNase domains are structurally related to those of RNase L, and the RNase domain fold is distinct from other proteins (Figure 6).98 Unfolded proteins induce dimerization of IRE1 and downstream signaling, including autophosphorylation and RNase activation.97,99 Activated RNase cleaves ER-bound RNA for its decay and specifically cuts X-box binding protein-1 (XBP1) mRNA in a nonconventional splicing mechanism, leading to the expression of a potent transcription factor.86,100 One reported downstream target of spliced XBP1 is the oncogene MYC. Thus, IRE1 activity is associated with cancer progression, aggressiveness, and poor prognosis.101−104 In addition to being studied as an anticancer target, IRE1 has also been implicated in diabetes and angiogenesis regulation.105,106 IRE1-targeting modulators were developed against the kinase and RNase domains (Figure 6).86 Similar to RNase L, reported kinase inhibitors were repurposed for IRE1 inhibition.97,107 Interestingly, some kinase inhibitors did not inhibit RNase function but activated IRE1 RNase activity instead.106,108,109 In general, type I kinase inhibitors stabilized the active kinase conformation and activated IRE1 RNase activity promoting oligomerization, while type II kinase inhibitors stabilized the inactive kinase conformation and inhibited RNase enzymatic activities.107,110,111 Assays for the identification of IRE1 modulators comprise splicing assays, fluorescence-based cleavage assays using fluorophore- and quencher-labeled XBP1 RNA substrate, and phosphorylation assays that focused on kinase activity instead of RNase activity.107,109,112,113 IRE1 inhibitor imidazo[1,5-α]pyrazine-8-amine compound 3 was identified based on screening of known type II kinase inhibitors, and further modification led to the development of compounds KIRA6 and KIRA7.107,109,114 Both KIRA6 and KIRA7 were potent inhibitors in biochemical and cellular evaluations, but a photoaffinity labeling approach revealed low selectivity of the imidazo[1,5-α]pyrazine-8-amine scaffold toward IRE1.109,114,115 The sulfonamide KIRA8 was an optimized hit with an IC50 of 0.014 μM in biochemical assays, but it did not affect cancer cell viability when screened against more than 300 different cancer cell lines.105,116 GSK2850163 is a screening-identified selective IRE1 inhibitor that bound to the kinase domain and displaced the kinase activation loop, thereby inhibiting RNase activity with an IC50 of 200 nM.117 Compound 31 was further identified as an IRE1-selective inhibitor (IC50 80 nM) that stabilized the inactive kinase conformation allosterically.118 Unfolded protein response inhibitor UPRM8 is a covalent inhibitor targeting the IRE1 kinase domain of IRE1 by reacting to a conserved cysteine residue in the active site.119 In contrast to the type II kinase inhibitors, type I kinase inhibitors activated the RNase domain of IRE1. Examples of such activators with poor selectivity include the receptor tyrosine kinase inhibitor sunitinib and the promiscuous kinase inhibitor APY29.97 In contrast, the pyrazolopyridine compound G-1749 activated unphosphorylated IRE1 via the modulation of the activation loop with an EC50 below 0.1 μM and showed a favorable selectivity profile.108 The RNase domain of IRE1 was directly targeted by covalent modifiers of lysine 907, such as hydroxy-aryl-aldehydes represented by compounds MKC9989.120 The lysine residue is not present in the same position in RNase L, making such Lysine-based covalent-targeting approach selective for IRE1. Furthermore, a docking study addressing the dimer interface of the RNase domain identified neomycin (IC50 0.33 μM) as an IRE1 inhibitor.112
BIFUNCTIONAL MOLECULES TARGETING RNASES
Bifunctional Dicer Inhibitors
An emerging approach to target ribonucleases is the design of bifunctional molecules to activate, bind, or inhibit an RNase in a specific context (Figure 7). The metal-dependent RNase Dicer is involved in the maturation of noncoding miRNAs. Bifunctional Dicer inhibitors allow specific inhibition of processing of a selected target RNA, avoiding affecting all canonically generated miRNAs by Dicer inhibition. The bifunctional inhibitors consist of an RNA-binding molecule and a weak Dicer inhibitor.121−124 The reported Dicer inhibitors were derived from pharmacophores of other endoribonuclease III inhibitors initially developed against influenza endonuclease and harbor the typical metal-chelating three oxygen pharmacophore.125 Aminoglycosides neomycin and kanamycin were employed as the RNA-binding molecules to target the oncogenic miRNA miR-21.122,123 The specificity of such bifunctional Dicer inhibitors was increased using antisense oligonucleotides, which alone did not inhibit the processing of pre-miR-21 by Dicer.121 A following study showed the possibility of incorporating a light-cleavable linker in the bifunctional molecule to deactivate the inhibitory activity by light.124
Figure 7.
Bifunctional molecules targeting RNases. General structures and representative examples of Dicer-targeting bifunctional molecules and RNA-targeting RIBOTACs, instead of all reported examples with full structures, were shown for clarity. ASO, Antisense oligonucleotide.
PROTAC
In general, the primary type of proximity-inducing bifunctional molecules is proteolysis targeting chimeras (PROTACs) that have gained substantial attention in the past decade to achieve degradation of protein targets of interest via ubiquitination and proteasome-mediate degradation pathway.126 Until now, no PROTAC that degrades an RNase has been developed, while a screening hit APL-16-5 against influenza PA endonuclease was confirmed to have a PROTAC-like mechanism. APL-16-5 is a microbial metabolite of Aspergillus sp. CPCC 400735 with an EC50 of 0.28 μM in antiviral assays that was shown to prevent the lethality of influenza infections in mice. Evaluation of its mode of action revealed the gluing mechanism of binding to the PA endonuclease and the E3-ligase TRIM25 to induce ubiquitination and thus endonuclease degradation.127
RIBOTACs
In comparison to the protein-degrading PROTACs, proximity-inducing bifunctional molecules have been reported by the Disney lab to induce targeted degradation of RNAs via the recruitment of RNase L, i.e., by ribonuclease targeting chimeras (RIBOTACs). So far, RNAs with different structured elements have been successfully degraded via RIBOTACs that recruit RNase L.92,96,128−134 The pioneering type of RIBOTACs used 2′-5′-linked oligoadenylates, the natural RNase L activator, as the RNase L recruiting component to be coupled to a dimeric miR-96 binder. The bifunctional molecule reduced amounts of mature miR-96 in cells.128 Small-molecule-based RIBOTACs using the aminothiophenone compound C1-3 as the RNase L recruiter were then reported to induce miR-21 degradation and led to reduced metastasis in mice breast cancer models.92 The same RNase L recruiter was used for the design of RIBOTACs degrading oncogenic miR-17, miR-18a, and miR-20a cluster, degrading SARS-CoV-2 attenuator hairpin involved in frameshifting of the ribosome (C5-RIBOTAC), for isoform-specific targeting of the mRNA of quiescin sulfhydryl oxidase 1 isoform a, and degrading an expanded G4C2 RNA repeat which is associated with amyotrophic lateral sclerosis and frontotemporal dementia.129−131,133 Small-molecule RNA binders that do not convey biologically active interactions were conjugated to the same aminothiophenone RNase L recruiter to form RIBOTAC degraders targeting oncogenic pre-miR-155, JUN mRNA and MYC mRNA.134 Recently, a biphenyl RNase L activator identified via screening of a DNA-encoded library was used in building the dovitinib-RIBOTAC 7 that degraded miR-21 and deactivated a miR-21-mediated cancer circuit in MDA-MB-231 cells.96 Apart from RNase L recruiters, bifunctional molecules with a chemical degrader of RNA including imidazole or a bleomycin derivative were developed.135−137
Both small-molecule RNase L recruiters and the Dicer inhibitor that were used as the building blocks for the bifunctional molecules are weak activators or inhibitors of their target RNases. The bifunctional molecule approach allows the local increase of RNase concentration or a local modulation of the RNase, affecting only the target RNA of interest.
SUMMARY AND PERSPECTIVES
RNases are ubiquitous RNA-cleaving and modifying proteins that regulate RNA biology and metabolism. RNases play essential roles in various cellular functions, including immune responses, antiviral pathways, apoptosis, and inflammation reactions. Therefore, RNases have been the targets for the development of small-molecule modulators for both biological and therapeutic applications. To date, small-molecule inhibitors of bacterial, viral, and human RNases have been reported, together with limited examples of small-molecule activators targeting a few selected human RNases. Successful examples, such as the approved agent baloxavir marboxil that inhibits the influenza PA endonuclease indicate the potential to develop RNase-targeting small molecules for therapeutic purposes. Most addressed ribonucleases are metalloenzymes, and thus, the three-oxygen pharmacophore discussed in the above-mentioned metal chelating inhibitors is highly abundant. The prodrug approach of baloxavir marboxil allows for higher cellular availability and thus provides a feasible approach to yield molecules with improved effectivity. The prodrug design to shield the three-oxygen pharmacophore of many RNase-targeting small molecules can potentially improve the limited cellular activity of many reported inhibitors and will be useful for discovering further optimized molecules. It is noteworthy that many reported RNase-targeting small molecules feature structures that may interfere with assay readouts,13 the same applies for small-molecule inhibitors of RNA-binding proteins in general.138 Therefore, careful validations via orthogonal assays and evaluations on the targeting profile and mechanisms of inhibition/activation are needed to identify robust molecules to be studied as either drug candidates or useful probes. In addition to synthetic small molecules, aminoglycosides such as neomycin were repeatedly described as inhibitors against a broad range of different RNases with generally low activity and selectivity, and aminoglycosides are also known to bind to RNAs. RNase L is one of the few RNase targets for which both inhibitors and activators have been reported by targeting different structural domains. Covalent inhibitors binding to the ribonuclease or adjacent domains were reported for IRE1. Reported covalent compounds showed improved efficiency in targeting shallow binding sites and thus hold great potential for developing potent RNase-targeting small molecules.120,139−141 Of note, cytotoxic RNases such as Ranpirnase and Barnase have also been studied for their direct use as anticancer agents in gene therapy.142,143 A new perspective in the field is the emerging class of bifunctional molecules that target RNases Dicer and RNase L for specific inhibition or activation of the RNases, achieving either inhibition of RNA biogenesis or induced RNA degradation. Concerning the biological relevance of RNases and their close associations with pathological states, it is reasonable to expect that more RNases will be subject both as the protein targets of interest for bifunctional molecules and as the effector protein components to be utilized by bifunctional molecules. These RNase-targeting molecules will contribute to probe an improved understanding of RNase biology at both the transcriptional and translational levels and provide new perspectives in developing small-molecule-based therapeutics.
Acknowledgments
We thank the financial support from the AstraZeneca, Merck KGaA, Pfizer Inc., and the Max Planck Society. P.W. thanks for funding from the Volkswagen Foundation on ribonuclease-targeting research (99535). The research in the Wu group is further supported by Boehringer Ingelheim Foundation (Exploration Grant to P.W.) and the Federal Ministry of Education and Research, Germany (BMBF). The authors thank Prof. Herbert Waldmann for his support and Yang Liu for proofreading the manuscript. L.B. acknowledges the International Max Planck Research School for Living Matter.
Glossary
ABBREVIATIONS
- AGS
Aicardi-Goutières syndrome
- BTDBA
4-[5-(benzoylamino)thien-2-yl]-2,4-dioxobutanoic acid
- CC50
Half maximal cytotoxic concentration
- Covid
Coronavirus disease
- EC50
Half maximal effective concentration
- ENCODE
Encyclopedia of DNA Elements
- FDA
United States food and drug administration
- FRET
Förster resonance energy transfer
- HAART
Highly active antiretroviral therapy
- HBV
Hepatitis B virus
- HIV
Human immunodeficiency virus
- IC50
Half maximal inhibitory concentration
- IRE1
Serine/threonine-protein kinase/endoribonuclease IRE1
- PA
Polymerase acidic
- PAGE
Polyacrylamide gel electrophoresis
- PAINS
Pan-assay interference compounds
- PARN
Poly(A)-specific ribonuclease
- PCR
Polymerase chain reaction
- PKR
Interferon-induced, double-stranded RNA-activated protein kinase
- RIBOTAC
Ribonuclease targeting chimera
- RT-PCR
Real-time polymerase chain reaction
- SAMDI
self-assembled monolayers for matrix-assisted laser desorption ionization
- SAR
Structure–activity relationship
- SARS-CoV-2
Severe acute respiratory syndrome coronavirus 2
- SPR
Surface plasmon resonance
- tRNA
tRNA
- XBP1
X-box binding protein-1
KEYWORDS
- Bifunctional molecule
a molecule with two functional entities. Synthetic heterobifunctional molecules often incorporate a linker structure between the two functional entities (e.g., two small molecules) interacting with two different biomacromolecules or domains
- DEDD motif
a common motif in nucleases consists of the four acidic amino acids Asp, Glu, Asp, and Asp, coordinating two divalent metal ions that coordinate the substrate phosphate in the active site of the nuclease
- noncoding RNAs
RNA molecules that are not translated into proteins but usually play important regulatory roles in various biological processes and diverse cellular activities
- PROTAC
proteolysis targeting chimeras are bifunctional molecules coupling a protein-binding moiety to an E3-ligase binder targeting a protein of interest for degradation
- Proximity-inducing molecule
a molecule that induces post-translational modifications or temporal control of biological processes by bringing two biomarcomolecules (that would not normally interact) in close proximity
- Ribonuclease
an enzyme that cleaves the phosphodiester bonds of ribonucleic acids
- Ribonuclease L (RNase L)
an antiviral interferon-induced ribonuclease that is activated depending on 2′-5′ oligoadenylates binding, resulting in cleavage and degradation of cellular RNAs
- RIBOTAC
ribonuclease targeting chimeras are bifunctional molecules consisting of a functional unit that binds RNase L linked to an RNA-binding moiety. The RNase is recruited to the RNA of interest, inducing specific degradation of the RNA
Supporting Information Available
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acschembio.3c00191.
Summary of molecular weights of depicted small molecules and bifunctional molecules (PDF)
Open access funded by Max Planck Society.
The authors declare no competing financial interest.
Special Issue
Published as part of ACS Chemical Biologyvirtual special issue “Nucleic Acid Regulation”.
Supplementary Material
References
- Canestrari E.; Paroo Z. Ribonucleases as Drug Targets. Trends Pharmacol. Sci. 2018, 39, 855–866. 10.1016/j.tips.2018.07.005. [DOI] [PubMed] [Google Scholar]
- Sorrentino S.; Libonati M. Structure-function relationships in human ribonucleases: Main distinctive features of the major RNase types. FEBS Lett. 1997, 404, 1–5. 10.1016/S0014-5793(97)00086-0. [DOI] [PubMed] [Google Scholar]
- Silverman R. H. A scientific journey through the 2–5A/RNase L system. Cytokine Growth Factor Rev. 2007, 18, 381–388. 10.1016/j.cytogfr.2007.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Worrall J. A.; Luisi B. F. Information available at cut rates: structure and mechanism of ribonucleases. Curr. Opin. Struct. Biol. 2007, 17, 128–137. 10.1016/j.sbi.2006.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gusho E.; Baskar D.; Banerjee S. New advances in our understanding of the “unique” RNase L in host pathogen interaction and immune signaling. Cytokine 2020, 133, 153847. 10.1016/j.cyto.2016.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MacRae I. J.; Doudna J. A. Ribonuclease revisited: structural insights into ribonuclease III family enzymes. Curr. Opin. Struct. Biol. 2007, 17, 138–145. 10.1016/j.sbi.2006.12.002. [DOI] [PubMed] [Google Scholar]
- Mikkelsen N. E.; Brännvall M.; Virtanen A.; Kirsebom L. A. Inhibition of RNase P RNA cleavage by aminoglycosides. Proc. Natl. Acad. Sci. U. S. A. 1999, 96, 6155–6160. 10.1073/pnas.96.11.6155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu X.; Chen Y.; Fierke C. A. A real-time fluorescence polarization activity assay to screen for inhibitors of bacterial ribonuclease P. Nucleic Acids Res. 2014, 42, e159 10.1093/nar/gku850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Madrigal-Carrillo E. A.; Díaz-Tufinio C. A.; Santamaría-Suárez H. A.; Arciniega M.; Torres-Larios A. A screening platform to monitor RNA processing and protein-RNA interactions in ribonuclease P uncovers a small molecule inhibitor. Nucleic Acids Res. 2019, 47, 6425–6438. 10.1093/nar/gkz285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kazantsev A. V.; Pace N. R. Bacterial RNase P: A new view of an ancient enzyme. Nat. Rev. Microbiol. 2006, 4, 729–740. 10.1038/nrmicro1491. [DOI] [PubMed] [Google Scholar]
- Eidem T. M.; et al. Small-molecule inhibitors of Staphylococcus aureus RnpA-mediated RNA turnover and tRNA processing. Antimicrob. Agents Chemother. 2015, 59, 2016–2028. 10.1128/AAC.04352-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schencking I.; Schafer E. M.; Scanlan J. H. W.; Wenzel B. M.; Emmerich R. E.; Steinmetzer T.; Diederich W. E.; Schlitzer M.; Hartmann R. K. RNase P Inhibitors Identified as Aggregators. Antimicrob. Agents Chemother. 2021, 65, e00300-21 10.1128/AAC.00300-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baell J. B. Feeling Nature’s PAINS: Natural Products, Natural Product Drugs, and Pan Assay Interference Compounds (PAINS). J. Nat. Prod. 2016, 79, 616–628. 10.1021/acs.jnatprod.5b00947. [DOI] [PubMed] [Google Scholar]
- Novopashina D.; Vorobyeva M.; Nazarov A.; Davydova A.; Danilin N.; Koroleva L.; Matveev A.; Bardasheva A.; Tikunova N.; Kupryushkin M.; Pyshnyi D.; Altman S.; Venyaminova A. Novel peptide conjugates of modified oligonucleotides for inhibition of bacterial RNase P. Front. Pharmacol. 2019, 10, 1–8. 10.3389/fphar.2019.00813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kime L.; Vincent H. A.; Gendoo D. M. A.; Jourdan S. S.; Fishwick C. W. G.; Callaghan A. J.; McDowall K. J. The first small-molecule inhibitors of members of the ribonuclease E family. Sci. Rep. 2015, 5, 9781. 10.1038/srep08028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tramontano E.; Corona A.; Menéndez-Arias L. Ribonuclease H, an unexploited target for antiviral intervention against HIV and hepatitis B virus. Antiviral Res. 2019, 171, 104613. 10.1016/j.antiviral.2019.104613. [DOI] [PubMed] [Google Scholar]
- Saramago M.; et al. The nsp15 Nuclease as a Good Target to Combat SARS-CoV-2: Mechanism of Action and Its Inactivation with FDA-Approved Drugs. Microorganisms 2022, 10, 342. 10.3390/microorganisms10020342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tahir M. Coronavirus genomic nsp14-ExoN, structure, role, mechanism, and potential application as a drug target. J. Med. Virol. 2021, 93, 4258–4264. 10.1002/jmv.27009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woehrl B. M.; Moelling K. Interaction of HIV-1 ribonuclease H with polypurine tract containing RNA-DNA hybrids. Biochemistry 1990, 29, 10141–10147. 10.1021/bi00496a001. [DOI] [PubMed] [Google Scholar]
- Moelling K.; Broecker F.; Kerrigan J. E.. RNase H: Specificity, Mechanisms of Action, and Antiviral Target. In Human Retroviruses. Methods in Molecular Biology; Vicenzi E., Poli G., Eds; Springer, 2014; pp 71–84. 10.1007/978-1-62703-670-2_7. [DOI] [PubMed] [Google Scholar]
- Himmel D. M.; et al. Structure of a dihydroxycoumarin active-site inhibitor in complex with the RNase H domain of HIV-1 reverse transcriptase and structure-activity analysis of inhibitor analogs. J. Mol. Biol. 2014, 426, 2617–2631. 10.1016/j.jmb.2014.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corona A.; et al. Targeting HIV-1 RNase H: N’-(2-Hydroxy-benzylidene)-3,4,5-Trihydroxybenzoylhydrazone as Selective Inhibitor Active against NNRTIs-Resistant Variants. Viruses 2020, 12, 729. 10.3390/v12070729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li G.; Wang Y.; De Clercq E. Approved HIV reverse transcriptase inhibitors in the past decade. Acta Pharm. Sin. B 2022, 12, 1567–1590. 10.1016/j.apsb.2021.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Loya S.; Tal R.; Kashman Y.; Hizi A. Illimaquinone, a selective inhibitor of the RNase H activity of human immunodeficiency virus type 1 reserve transcriptase. Antimicrob. Agents Chemother. 1990, 34, 2009–2012. 10.1128/AAC.34.10.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hafkemeyer P.; et al. HP 0.35, a cephalosporin degradation product is a specific inhibitor of lentiviral RNAses H. Nucleic Acids Res. 1991, 19, 4059–4065. 10.1093/nar/19.15.4059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Andréola M. L.; Tharaud D.; Litvak S.; Tarrago-Litvak L. The ribonuclease H activity of HIV-1 reverse transcriptase: Further biochemical characterization and search of inhibitors. Biochimie 1993, 75, 127–134. 10.1016/0300-9084(93)90034-P. [DOI] [PubMed] [Google Scholar]
- Tavis J. E.; et al. The Hepatitis B Virus Ribonuclease H Is Sensitive to Inhibitors of the Human Immunodeficiency Virus Ribonuclease H and Integrase Enzymes. PLoS Pathog. 2013, 9, e1003125 10.1371/journal.ppat.1003125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Edwards T. C.; et al. Inhibition of hepatitis B virus replication by N-hydroxyisoquinolinediones and related polyoxygenated heterocycles. Antiviral Res. 2017, 143, 205–217. 10.1016/j.antiviral.2017.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corona A.; et al. Inhibition of foamy virus reverse transcriptase by human immunodeficiency virus type 1 RNase H inhibitors. Antimicrob. Agents Chemother. 2014, 58, 4086–4093. 10.1128/AAC.00056-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kirschberg T. A.; et al. RNase H active site inhibitors of human immunodeficiency virus type 1 reverse transcriptase: Design, biochemical activity, and structural information. J. Med. Chem. 2009, 52, 5781–5784. 10.1021/jm900597q. [DOI] [PubMed] [Google Scholar]
- Tajwar R.; Bradley D. P.; Ponzar N. L.; Tavis J. E. Predicted structure of the hepatitis B virus polymerase reveals an ancient conserved protein fold. Protein Sci. 2022, 31, 1–18. 10.1002/pro.4421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu H.; et al. Computational and Crystallographic Analysis of Binding Structures of Inhibitory Compounds for HIV-1 RNase H Activity. J. Chem. Inf. Model. 2022, 62, 6762–6774. 10.1021/acs.jcim.2c00537. [DOI] [PubMed] [Google Scholar]
- Kankanala J.; et al. Design, Synthesis, and Biological Evaluations of Hydroxypyridonecarboxylic Acids as Inhibitors of HIV Reverse Transcriptase Associated RNase H. J. Med. Chem. 2016, 59, 5051–5062. 10.1021/acs.jmedchem.6b00465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dyda F.; et al. Crystal structure of the catalytic domain of HIV-1 integrase: Similarity to other polynucleotidyl transferases. Science (80-.). 1994, 266, 1981–1986. 10.1126/science.7801124. [DOI] [PubMed] [Google Scholar]
- Shaw-Reid C. A.; et al. Inhibition of HIV-1 ribonuclease H by a novel diketo acid, 4-[5-(benzoylamino)thien-2-yl]-2,4-dioxobutanoic acid. J. Biol. Chem. 2003, 278, 2777–2780. 10.1074/jbc.C200621200. [DOI] [PubMed] [Google Scholar]
- Cuzzucoli Crucitti G.; et al. Structure-activity relationship of pyrrolyl diketo acid derivatives as dual inhibitors of HIV-1 integrase and reverse transcriptase ribonuclease H domain. J. Med. Chem. 2015, 58, 1915–1928. 10.1021/jm501799k. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu B.; et al. 3-Hydroxypyrimidine-2,4-dione-5-N-benzylcarboxamides Potently Inhibit HIV-1 Integrase and RNase H. J. Med. Chem. 2016, 59, 6136–6148. 10.1021/acs.jmedchem.6b00040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang L.; Sarafianos S. G.; Wang Z. Cutting into the Substrate Dominance: Pharmacophore and Structure-Based Approaches toward Inhibiting Human Immunodeficiency Virus Reverse Transcriptase-Associated Ribonuclease H. Acc. Chem. Res. 2020, 53, 218–230. 10.1021/acs.accounts.9b00450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang L.; et al. 6-Arylthio-3-hydroxypyrimidine-2,4-diones potently inhibited HIV reverse transcriptase-associated RNase H with antiviral activity. Eur. J. Med. Chem. 2018, 156, 652–665. 10.1016/j.ejmech.2018.07.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Edwards T. C.; et al. Inhibition of HBV replication by N-hydroxyisoquinolinedione and N-hydroxypyridinedione ribonuclease H inhibitors. Antiviral Res. 2019, 164, 70–80. 10.1016/j.antiviral.2019.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu G.; et al. Hydroxylated tropolones inhibit hepatitis B virus replication by blocking viral ribonuclease H activity. Antimicrob. Agents Chemother. 2015, 59, 1070–1079. 10.1128/AAC.04617-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lomonosova E.; et al. Efficacy and cytotoxicity in cell culture of novel α-hydroxytropolone inhibitors of hepatitis B virus ribonuclease H. Antiviral Res. 2017, 144, 164–172. 10.1016/j.antiviral.2017.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Masaoka T.; et al. Exploiting drug-resistant enzymes as tools to identify thienopyrimidinone inhibitors of human immunodeficiency virus reverse transcriptase-associated ribonuclease H. J. Med. Chem. 2013, 56, 5436–5445. 10.1021/jm400405z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Massari S.; et al. From cycloheptathiophene-3-carboxamide to oxazinone-based derivatives as allosteric HIV-1 ribonuclease H inhibitors. J. Enzyme Inhib. Med. Chem. 2019, 34, 55–74. 10.1080/14756366.2018.1523901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Distinto S.; et al. Identification of HIV-1 reverse transcriptase dual inhibitors by a combined shape-, 2D-fingerprint- and pharmacophore-based virtual screening approach. Eur. J. Med. Chem. 2012, 50, 216–229. 10.1016/j.ejmech.2012.01.056. [DOI] [PubMed] [Google Scholar]
- Meleddu R.; et al. Isatin thiazoline hybrids as dual inhibitors of HIV-1 reverse transcriptase. J. Enzyme Inhib. Med. Chem. 2017, 32, 130–136. 10.1080/14756366.2016.1238366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vijayan R.; Gourinath S. Structure-based inhibitor screening of natural products against NSP15 of SARS-CoV-2 revealed thymopentin and oleuropein as potent inhibitors. J. Proteins Proteomics 2021, 12, 71–80. 10.1007/s42485-021-00059-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu L.-C.; Ding C.-H.; Li H.-Y.; Li Z.-Z.; Chen Y.; Li L.-P.; Li W.-Z.; Liu W.-S. Identification of potential target endoribonuclease NSP15 inhibitors of SARS-COV-2 from natural products through high-throughput virtual screening and molecular dynamics simulation. J. Food Biochem. 2022, 46, 1–14. 10.1111/jfbc.14085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Canal B.; et al. Identifying SARS-CoV-2 antiviral compounds by screening for small molecule inhibitors of nsp15 endoribonuclease. Biochem. J. 2021, 478, 2465–2479. 10.1042/BCJ20210199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim Y.; Wower J.; Maltseva N.; Chang C.; Jedrzejczak R.; Wilamowski M.; Kang S.; Nicolaescu V.; Randall G.; Michalska K.; Joachimiak A. Tipiracil binds to uridine site and inhibits Nsp15 endoribonuclease NendoU from SARS-CoV-2. Commun. Biol. 2021, 4, 1–11. 10.1038/s42003-021-01735-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saramago M.; et al. New targets for drug design: importance of nsp14/nsp10 complex formation for the 3′-5′ exoribonucleolytic activity on SARS-CoV-2. FEBS J. 2021, 288, 5130–5147. 10.1111/febs.15815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rona G.; et al. The NSP14/NSP10 RNA repair complex as a Pan-coronavirus therapeutic target. Cell Death Differ. 2022, 29, 285–292. 10.1038/s41418-021-00900-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scholle M. D.; Liu C.; Deval J.; Gurard-Levin Z. A. Label-Free Screening of SARS-CoV-2 NSP14 Exonuclease Activity Using SAMDI Mass Spectrometry. SLAS Discovery 2021, 26, 766–774. 10.1177/24725552211008854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Imprachim N.; Yosaatmadja Y.; Newman J. A. Crystal structures and fragment screening of SARS-CoV-2 NSP14 reveal details of exoribonuclease activation and mRNA capping and provide starting points for antiviral drug development. Nucleic Acids Res. 2023, 51, 475–487. 10.1093/nar/gkac1207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clohisey S.; Parkinson N.; Wang B.; Bertin N.; Wise H.; Tomoiu A.; Summers K. M.; Hendry R. W.; Carninci P.; Forrest A. R. R.; Hayashizaki Y.; Digard P.; Hume D. A.; Baillie J. K. Comprehensive Characterization of Transcriptional Activity during Influenza A Virus Infection Reveals Biases in Cap-Snatching of Host RNA Sequences. J. Virol. 2020, 94, e01720-19 10.1128/JVI.01720-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dias A.; et al. The cap-snatching endonuclease of influenza virus polymerase resides in the PA subunit. Nature 2009, 458, 914–918. 10.1038/nature07745. [DOI] [PubMed] [Google Scholar]
- Ju H.; et al. Inhibitors of Influenza Virus Polymerase Acidic (PA) Endonuclease: Contemporary Developments and Perspectives. J. Med. Chem. 2017, 60, 3533–3551. 10.1021/acs.jmedchem.6b01227. [DOI] [PubMed] [Google Scholar]
- Tomassini J.; et al. Inhibition of cap (m7GpppXm)-dependent endonuclease of influenza virus by 4-substituted 2,4-dioxobutanoic acid compounds. Antimicrob. Agents Chemother. 1994, 38, 2827–2837. 10.1128/AAC.38.12.2827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Credille C. V.; et al. SAR Exploration of Tight-Binding Inhibitors of Influenza Virus PA Endonuclease. J. Med. Chem. 2019, 62, 9438–9449. 10.1021/acs.jmedchem.9b00747. [DOI] [PubMed] [Google Scholar]
- Noshi T.; et al. In vitro characterization of baloxavir acid, a first-in-class cap-dependent endonuclease inhibitor of the influenza virus polymerase PA subunit. Antiviral Res. 2018, 160, 109–117. 10.1016/j.antiviral.2018.10.008. [DOI] [PubMed] [Google Scholar]
- Omoto S.; Speranzini V.; Hashimoto T.; Noshi T.; Yamaguchi H.; Kawai M.; Kawaguchi K.; Uehara T.; Shishido T.; Naito A.; Cusack S. Characterization of influenza virus variants induced by treatment with the endonuclease inhibitor baloxavir marboxil. Sci. Rep. 2018, 8, 9633. 10.1038/s41598-018-27890-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heo Y. A. Baloxavir: First Global Approval. Drugs 2018, 78, 693–697. 10.1007/s40265-018-0899-1. [DOI] [PubMed] [Google Scholar]
- Taoda Y.; et al. Dihydrodibenzothiepine: Promising hydrophobic pharmacophore in the influenza cap-dependent endonuclease inhibitor. Bioorg. Med. Chem. Lett. 2020, 30, 127547. 10.1016/j.bmcl.2020.127547. [DOI] [PubMed] [Google Scholar]
- Ren Y.; Chen Y.; Cao S. Targeted inhibition of the endonuclease activity of influenza polymerase acidic proteins. Future Med. Chem. 2022, 14, 571–586. 10.4155/fmc-2021-0264. [DOI] [PubMed] [Google Scholar]
- Rosenberg H. F. RNase A ribonucleases and host defense: an evolving story. J. Leukoc. Biol. 2008, 83, 1079–1087. 10.1189/jlb.1107725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun D.; Han C.; Sheng J. The role of human ribonuclease A family in health and diseases: A systematic review. iScience 2022, 25, 105284. 10.1016/j.isci.2022.105284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Russo A.; Acharya K. R.; Shapiro R. Small Molecule Inhibitors of RNase A and Related Enzymes. Methods Enzymol. 2001, 341, 629–648. 10.1016/S0076-6879(01)41181-5. [DOI] [PubMed] [Google Scholar]
- Jenkins J. L.; Shapiro R. Identification of small-molecule inhibitors of human angiogenin and characterization of their binding interactions guided by computational docking. Biochemistry 2003, 42, 6674–6687. 10.1021/bi034164e. [DOI] [PubMed] [Google Scholar]
- Kao R. Y. T.; et al. A small-molecule inhibitor of the ribonucleolytic activity of human angiogenin that possesses antitumor activity. Proc. Natl. Acad. Sci. U. S. A. 2002, 99, 10066–10071. 10.1073/pnas.152342999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davies B. P.; Arenz C. A homogenous assay for micro RNA maturation. Angew. Chemie - Int. Ed. 2006, 45, 5550–5552. 10.1002/anie.200601332. [DOI] [PubMed] [Google Scholar]
- Maryati M.; Kaur I.; Jadhav G. P.; Olotu-Umoren L.; Oveh B.; Hashmi L.; Fischer P. M.; Winkler G. S. A fluorescence-based assay suitable for quantitative analysis of deadenylase enzyme activity. Nucleic Acids Res. 2014, 42, e30 10.1093/nar/gkt972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jadhav G. P.; et al. Discovery, synthesis and biochemical profiling of purine-2,6-dione derivatives as inhibitors of the human poly(A)-selective ribonuclease Caf1. Bioorg. Med. Chem. Lett. 2015, 25, 4219–4224. 10.1016/j.bmcl.2015.07.095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ren Y. G.; Martínez J.; Kirsebom L. A.; Virtanen A. Inhibition of Klenow DNA polymerase and poly(A)-specific ribonuclease by aminoglycosides. Rna 2002, 8, 1393–1400. 10.1017/S1355838202021015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Balatsos N. A. A.; et al. Competitive inhibition of human poly(A)-specific ribonuclease (PARN) by synthetic fluoro-pyranosyl nucleosides. Biochemistry 2009, 48, 6044–6051. 10.1021/bi900236k. [DOI] [PubMed] [Google Scholar]
- Balatsos N. A. A.; Anastasakis D.; Stathopoulos C. Inhibition of human poly(A)-specific ribonuclease (PARN) by purine nucleotides: Kinetic analysis. J. Enzyme Inhib. Med. Chem. 2009, 24, 516–523. 10.1080/14756360802218763. [DOI] [PubMed] [Google Scholar]
- Vlachakis D.; et al. An Integrated In Silico Approach to Design Specific Inhibitors Targeting Human Poly(A)-Specific Ribonuclease. PLoS One 2012, 7, e51113 10.1371/journal.pone.0051113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nowotny M.; Gaidamakov S. A.; Crouch R. J.; Yang W. Crystal structures of RNase H bound to an RNA/DNA hybrid: Substrate specificity and metal-dependent catalysis. Cell 2005, 121, 1005–1016. 10.1016/j.cell.2005.04.024. [DOI] [PubMed] [Google Scholar]
- Crow Y. J.; et al. Mutations in genes encoding ribonuclease H2 subunits cause Aicardi-Goutières syndrome and mimic congenital viral brain infection. Nat. Genet. 2006, 38, 910–916. 10.1038/ng1842. [DOI] [PubMed] [Google Scholar]
- White R.; Saxty B.; Large J.; Kettleborough C. A.; Jackson A. P. Identification of small-molecule inhibitors of the ribonuclease H2 enzyme. J. Biomol. Screen. 2013, 18, 610–620. 10.1177/1087057113476550. [DOI] [PubMed] [Google Scholar]
- Donovan J.; Dufner M.; Korennykh A. Structural basis for cytosolic double-stranded RNA surveillance by human oligoadenylate synthetase 1. Proc. Natl. Acad. Sci. U. S. A. 2013, 110, 1652–1657. 10.1073/pnas.1218528110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang H.; et al. Dimeric Structure of Pseudokinase RNase L Bound to 2–5A Reveals a Basis for Interferon-Induced Antiviral Activity. Mol. Cell 2014, 53, 221–234. 10.1016/j.molcel.2013.12.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rath S.; et al. Concerted 2–5A-Mediated mRNA Decay and Transcription Reprogram Protein Synthesis in the dsRNA Response. Mol. Cell 2019, 75, 1218–1228. 10.1016/j.molcel.2019.07.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Donovan J.; Rath S.; Kolet-Mandrikov D.; Korennykh A. Rapid RNase L-driven arrest of protein synthesis in the dsRNA response without degradation of translation machinery. RNA 2017, 23, 1660–1671. 10.1261/rna.062000.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Daou S.; et al. A phenolic small molecule inhibitor of RNase L prevents cell death from ADAR1 deficiency. Proc. Natl. Acad. Sci. U. S. A. 2020, 117, 24802–24812. 10.1073/pnas.2006883117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crow Y. J.; Manel N. Aicardi-Goutières syndrome and the type I interferonopathies. Nat. Rev. Immunol. 2015, 15, 429–440. 10.1038/nri3850. [DOI] [PubMed] [Google Scholar]
- Langlais T.; et al. Structural and molecular bases to IRE1 activity modulation. Biochem. J. 2021, 478, 2953–2975. 10.1042/BCJ20200919. [DOI] [PubMed] [Google Scholar]
- Jha B. K.; et al. Inhibition of RNase L and RNA-dependent protein kinase (PKR) by sunitinib impairs antiviral innate immunity. J. Biol. Chem. 2011, 286, 26319–26326. 10.1074/jbc.M111.253443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jha B. K.; Dong B.; Nguyen C. T.; Polyakova I.; Silverman R. H. Suppression of antiviral innate immunity by sunitinib enhances oncolytic virotherapy. Mol. Ther. 2013, 21, 1749–1757. 10.1038/mt.2013.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang J.; et al. Sunitinib inhibits RNase L by destabilizing its active dimer conformation. Biochem. J. 2020, 477, 3387–3399. 10.1042/BCJ20200260. [DOI] [PubMed] [Google Scholar]
- Tang J.; et al. Identification of Small Molecule Inhibitors of RNase L by Fragment-Based Drug Discovery. J. Med. Chem. 2022, 65, 1445–1457. 10.1021/acs.jmedchem.1c01156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thakur C. S.; et al. Small-molecule activators of RNase L with broad-spectrum antiviral activity. Proc. Natl. Acad. Sci. U. S. A. 2007, 104, 9585–9590. 10.1073/pnas.0700590104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Costales M. G.; 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. 10.1073/pnas.1914286117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hwang J.; et al. Synthesis and evaluation of RNase L-binding 2-aminothiophenes as anticancer agents. Bioorg. Med. Chem. 2022, 58, 116653. 10.1016/j.bmc.2022.116653. [DOI] [PubMed] [Google Scholar]
- Borgelt L.; et al. Small-molecule screening of ribonuclease L binders for RNA degradation. Biomed. Pharmacother. 2022, 154, 113589. 10.1016/j.biopha.2022.113589. [DOI] [PubMed] [Google Scholar]
- Hwang J.; et al. Rational design and evaluation of 2-((pyrrol-2-yl)methylene)thiophen-4-ones as RNase L inhibitors. Eur. J. Med. Chem. 2023, 256, 115439. 10.1016/j.ejmech.2023.115439. [DOI] [PubMed] [Google Scholar]
- Meyer S. M.; et al. DNA-Encoded Library Screening To Inform Design of a Ribonuclease Targeting Chimera (RiboTAC). J. Am. Chem. Soc. 2022, 144, 21096–21102. 10.1021/jacs.2c07217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Korennykh A. V.; et al. The unfolded protein response signals through high-order assembly of Ire1. Nature 2009, 457, 687–693. 10.1038/nature07661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee K. P. K.; et al. Structure of the Dual Enzyme Ire1 Reveals the Basis for Catalysis and Regulation in Nonconventional RNA Splicing. Cell 2008, 132, 89–100. 10.1016/j.cell.2007.10.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shamu C. E.; Walter P. Oligomerization phosphorylation of the Ire1p kinase during intracellular signaling from the endoplasmic reticulum to the nucleus. EMBO J. 1996, 15, 3028–3039. 10.1002/j.1460-2075.1996.tb00666.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raymundo D. P.; et al. Pharmacological Targeting of IRE1 in Cancer. Trends in Cancer 2020, 6, 1018–1030. 10.1016/j.trecan.2020.07.006. [DOI] [PubMed] [Google Scholar]
- Madden E.; Logue S. E.; Healy S. J.; Manie S.; Samali A. The role of the unfolded protein response in cancer progression: From oncogenesis to chemoresistance. Biol. Cell 2019, 111, 1–17. 10.1111/boc.201800050. [DOI] [PubMed] [Google Scholar]
- Logue S. E.; McGrath E. P.; Cleary P.; Greene S.; Mnich K.; Almanza A.; Chevet E.; Dwyer R. M.; Oommen A.; Legembre P.; et al. Inhibition of IRE1 RNase activity modulates the tumor cell secretome and enhances response to chemotherapy. Nat. Commun. 2018, 9, 3267. 10.1038/s41467-018-05763-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao N.; et al. Pharmacological targeting of MYC-regulated IRE1/XBP1 pathway suppresses MYC-driven breast cancer Find the latest version : Pharmacological targeting of MYC-regulated IRE1/XBP1 pathway suppresses MYC-driven breast cancer. J. Clin. Invest. 2018, 128, 1283–1299. 10.1172/JCI95873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sheng X.; Nenseth H. Z.; Qu S.; Kuzu O. F.; Frahnow T.; Simon L.; Greene S.; Zeng Q.; Fazli L.; Rennie P. S.; et al. IRE1α-XBP1s pathway promotes prostate cancer by activating c-MYC signaling. Nat. Commun. 2019, 10, 1–12. 10.1038/s41467-018-08152-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morita S.; et al. Targeting ABL-IRE1α Signaling Spares ER-Stressed Pancreatic β Cells to Reverse Autoimmune Diabetes. Cell Metab. 2017, 25, 883–897. 10.1016/j.cmet.2017.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fan F.; et al. Salvianolic acid B, a new type I IRE1 kinase inhibitor, abrogates AngII-induced angiogenesis by interacting with IRE1 in its active conformation. Clin. Exp. Pharmacol. Physiol. 2023, 50, 82–95. 10.1111/1440-1681.13726. [DOI] [PubMed] [Google Scholar]
- Wang L.; et al. Divergent allosteric control of the IRE1α endoribonuclease using kinase inhibitors. Nat. Chem. Biol. 2012, 8, 982–989. 10.1038/nchembio.1094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferri E.; Le Thomas A.; Wallweber H. A.; Day E. S.; Walters B. T.; Kaufman S. E.; Braun M.-G.; Clark K. R.; Beresini M. H.; Mortara K.; et al. Activation of the IRE1 RNase through remodeling of the kinase front pocket by ATP-competitive ligands. Nat. Commun. 2020, 11, 1–15. 10.1038/s41467-020-19974-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ghosh R.; et al. Allosteric inhibition of the IRE1α RNase preserves cell viability and function during endoplasmic reticulum stress. Cell 2014, 158, 534–548. 10.1016/j.cell.2014.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feldman H. C.; et al. Structural and Functional Analysis of the Allosteric Inhibition of IRE1α with ATP-Competitive Ligands. ACS Chem. Biol. 2016, 11, 2195–2205. 10.1021/acschembio.5b00940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goebel G. L.; Qiu X.; Wu P. Kinase-targeting small-molecule inhibitors and emerging bifunctional molecules. Trends Pharmacol. Sci. 2022, 43, 866–881. 10.1016/j.tips.2022.04.006. [DOI] [PubMed] [Google Scholar]
- Amarasinghe K. N.; et al. Sensor dimer disruption as a new mode of action to block the IRE1-mediated unfolded protein response. Comput. Struct. Biotechnol. J. 2022, 20, 1584–1592. 10.1016/j.csbj.2022.03.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cross B. C. S.; Bond P. J.; Sadowski P. G.; Jha B. K.; Zak J.; Goodman J. M.; Silverman R. H.; Neubert T. A.; Baxendale I. R.; Ron D.; Harding H. P. The molecular basis for selective inhibition of unconventional mRNA splicing by an IRE1-binding small molecule. Proc. Natl. Acad. Sci. U. S. A. 2012, 109, 869–878. 10.1073/pnas.1115623109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thamsen M.; Ghosh R.; Auyeung V. C.; Brumwell A.; Chapman H. A.; Backes B. J.; Perara G.; Maly D. J.; Sheppard D.; Papa F. R. Small molecule inhibition of IRE1α kinase/ RNase has anti-fibrotic effects in the lung. PLoS One 2019, 14, e0209824. 10.1371/journal.pone.0209824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Korovesis D.; Rufo N.; Derua R.; Agostinis P.; Verhelst S. H. L. Kinase Photoaffinity Labeling Reveals Low Selectivity Profile of the IRE1 Targeting Imidazopyrazine-Based KIRA6 Inhibitor. ACS Chem. Biol. 2020, 15, 3106–3111. 10.1021/acschembio.0c00802. [DOI] [PubMed] [Google Scholar]
- Harrington P. E.; et al. Unfolded protein response in cancer: IRE1α inhibition by selective kinase ligands does not impair tumor cell viability. ACS Med. Chem. Lett. 2015, 6, 68–72. 10.1021/ml500315b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Concha N. O.; et al. Long-range inhibitor-induced conformational regulation of human IRE1α endoribonuclease activity. Mol. Pharmacol. 2015, 88, 1011–1023. 10.1124/mol.115.100917. [DOI] [PubMed] [Google Scholar]
- Colombano G.; et al. Binding to an Unusual Inactive Kinase Conformation by Highly Selective Inhibitors of Inositol-Requiring Enzyme 1α Kinase-Endoribonuclease. J. Med. Chem. 2019, 62, 2447–2465. 10.1021/acs.jmedchem.8b01721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Waller D. D.; et al. A Covalent Cysteine-Targeting Kinase Inhibitor of Ire1 Permits Allosteric Control of Endoribonuclease Activity. ChemBioChem. 2016, 17, 843–851. 10.1002/cbic.201500485. [DOI] [PubMed] [Google Scholar]
- Sanches M.; et al. Structure and mechanism of action of the hydroxy-aryl-aldehyde class of IRE1 endoribonuclease inhibitors. Nat. Commun. 2014, 5, 4202. 10.1038/ncomms5202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bhattarai U.; et al. Bifunctional small molecule-oligonucleotide hybrid as microRNA inhibitor. Bioorg. Med. Chem. 2020, 28, 115394. 10.1016/j.bmc.2020.115394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yan H.; Bhattarai U.; Guo Z.-F.; Liang F.-S. Regulating miRNA-21 Biogenesis by Bifunctional Small Molecules. J. Am. Chem. Soc. 2017, 139, 4987–4990. 10.1021/jacs.7b00610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yan H.; Liang F.-S. miRNA inhibition by proximity-enabled Dicer inactivation. Methods 2019, 167, 117–123. 10.1016/j.ymeth.2019.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yan H.; Bhattarai U.; Song Y.; Liang F.-S. Design, synthesis and activity of light deactivatable microRNA inhibitor. Bioorg. Chem. 2018, 80, 492–497. 10.1016/j.bioorg.2018.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parkes K. E. B.; et al. Use of a pharmacophore model to discover a new class of influenza endonuclease inhibitors. J. Med. Chem. 2003, 46, 1153–1164. 10.1021/jm020334u. [DOI] [PubMed] [Google Scholar]
- Békés M.; Langley D. R.; Crews C. M. PROTAC targeted protein degraders: the past is prologue. Nat. Rev. Drug Discovery 2022, 21, 181–200. 10.1038/s41573-021-00371-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao J.; Wang J.; Pang X.; Liu Z.; Li Q.; Yi D.; Zhang Y.; Fang X.; Zhang T.; Zhou R.; et al. An anti-influenza A virus microbial metabolite acts by degrading viral endonuclease PA. Nat. Commun. 2022, 13, 2079. 10.1038/s41467-022-29690-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Costales M. G.; Matsumoto Y.; Velagapudi S. P.; Disney M. D. Small Molecule Targeted Recruitment of a Nuclease to RNA. J. Am. Chem. Soc. 2018, 140, 6741–6744. 10.1021/jacs.8b01233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu X.; et al. Targeted Degradation of the Oncogenic MicroRNA 17–92 Cluster by Structure-Targeting Ligands. J. Am. Chem. Soc. 2020, 142, 6970–6982. 10.1021/jacs.9b13159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haniff H. S.; et al. Targeting the SARS-CoV-2 RNA Genome with Small Molecule Binders and Ribonuclease Targeting Chimera (RIBOTAC) Degraders. ACS Cent. Sci. 2020, 6, 1713–1721. 10.1021/acscentsci.0c00984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tong Y.; et al. Transcriptome-Wide Mapping of Small-Molecule RNA-Binding Sites in Cells Informs an Isoform-Specific Degrader of QSOX1 mRNA. J. Am. Chem. Soc. 2022, 144, 11620–11625. 10.1021/jacs.2c01929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang P.; et al. Reprogramming of Protein-Targeted Small-Molecule Medicines to RNA by Ribonuclease Recruitment. J. Am. Chem. Soc. 2021, 143, 13044–13055. 10.1021/jacs.1c02248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bush J. A.; Aikawa H.; Fuerst R.; Li Y.; Ursu A.; Meyer S. M.; Benhamou R. I.; Chen J. L.; Khan T.; Wagner-Griffin S.; et al. Ribonuclease recruitment using a small molecule reduced c9ALS/FTD r(G 4 C 2) repeat expansion in vitro and in vivo ALS models. Sci. Transl. Med. 2021, 13, 1–14. 10.1126/scitranslmed.abd5991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tong Y.; et al. Programming inactive RNA-binding small molecules into bioactive degraders. Nature 2023, 618, 169–179. 10.1038/s41586-023-06091-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martin C.; Bonnet M.; Patino N.; Azoulay S.; Di Giorgio A.; Duca M. Design, Synthesis, and Evaluation of Neomycin-Imidazole Conjugates for RNA Cleavage. ChemPlusChem. 2022, 87, e202200250 10.1002/cplu.202200250. [DOI] [PubMed] [Google Scholar]
- Mikutis S.; et al. Proximity-Induced Nucleic Acid Degrader (PINAD) Approach to Targeted RNA Degradation Using Small Molecules. ACS Cent. Sci. 2023, 9, 892. 10.1021/acscentsci.3c00015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benhamou R. I.; et al. Structure-Specific Cleavage of an RNA Repeat Expansion with a Dimeric Small Molecule Is Advantageous over Sequence-Specific Recognition by an Oligonucleotide. ACS Chem. Biol. 2020, 15, 485–493. 10.1021/acschembio.9b00958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu P. Inhibition of RNA-binding proteins with small molecules. Nat. Rev. Chem. 2020, 4, 441–458. 10.1038/s41570-020-0201-4. [DOI] [PubMed] [Google Scholar]
- Sutanto F.; Konstantinidou M.; Dömling A. Covalent inhibitors: A rational approach to drug discovery. RSC Med. Chem. 2020, 11, 876–884. 10.1039/D0MD00154F. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Billamboz M.; et al. Magnesium chelating 2-hydroxyisoquinoline-1,3(2H, 4H)-diones, as inhibitors of HIV-1 integrase and/or the HIV-1 reverse transcriptase ribonuclease H domain: Discovery of a novel selective inhibitor of the ribonuclease H function. J. Med. Chem. 2011, 54, 1812–1824. 10.1021/jm1014692. [DOI] [PubMed] [Google Scholar]
- Koslover D. J.; et al. The Crystal Structure of the Escherichia coli RNase E Apoprotein and a Mechanism for RNA Degradation. Structure 2008, 16, 1238–1244. 10.1016/j.str.2008.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee J. E.; Raines R. T. Ribonucleases as novel chemotherapeutics: The ranpirnase example. BioDrugs 2008, 22, 53–58. 10.2165/00063030-200822010-00006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shilova O.; Kotelnikova P.; Proshkina G.; Shramova E.; Deyev S. Barnase-barstar pair: Contemporary application in cancer research and nanotechnology. Molecules 2021, 26, 6785. 10.3390/molecules26226785. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.