Quentin M. R. Gibaut’s research while affiliated with The Scripps Research Institute and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (11)


The Far Upstream Element Binding Protein 3 enhances HIV-1 transcriptional activity and regulates immune response pathways in T cells
  • Article

March 2025

·

18 Reads

Molecular Therapy — Nucleic Acids

Quentin M.R. Gibaut

·

·

Anqi Cheng

·

[...]

·

Susana T. Valente

Far-upstream element-binding protein 3 (FUBP3) was identified at actively transcribing HIV promoters through chromatin affinity purification and mass spectrometry. Known for regulating cellular processes such as transcription and translation by binding to DNAs and RNAs, FUBP3’s role in HIV transcriptional regulation was previously unrecognized. This study reveals that FUBP3 enhances HIV-1 transcriptional activation by interacting with Tat and trans-activation response (TAR)-RNA, critical for boosting viral transcription through recruitment of activating factors that promote RNA polymerase II (RNAPII) elongation. Transcriptomic analysis, chromatin immunoprecipitation, and biochemical assays demonstrated that FUBP3 associates with and stabilizes TAR-RNA, in a Tat-dependent manner, and enhances Tat steady-state levels via interaction with Tat’s basic domain. Suppressing FUBP3 decreased HIV-1 transcription and altered expression of host genes linked to T cell activation and inflammation, underscoring its broad regulatory impact. Additionally, FUBP3 was enriched at active promoters, confirming its role in transcriptional regulation at specific genomic locations. These findings highlight FUBP3’s critical role in the HIV-1 life cycle and suggest its potential as a therapeutic target in HIV-1 infection. Additionally, this study expands our understanding of FUBP3’s functions in oncogenic and inflammatory pathways.


Fig. 2. Synucleozid-2.0 inhibits translation of SNCA mRNA and is cytoprotective in cells by selectively binding to the SNCA IRE region. (A) Effect of Synucleozid-2.0 (48 h) on translation of a luciferase reporter gene in HeLa cells with or without the SNCA 5′ UTR that harbors the IRE (n = 3 biological replicates for all conditions except vehicle-treated Control Luciferase reporter where n = 4). (B) Effect of Synucleozid-2.0 (48 h) on SNCA mRNA levels in native SH-SY5Y cells (n = 4 biological replicates for vehicle (0 µM) and n = 3 for Synucleozid-2.0 (2 µM)). (C) Cytoprotective effect after 48 h of Synucleozid-2.0 treatment of SH-SY5Y cells against 50 ng/µL human preformed fibrils (PFFs), a model of α-synuclein toxicity, measured by LDH assay (n = 4 biological replicates). PFFs act as seeds and recruit endogenous α-synuclein to aggregate. (D) Representative Western blot analysis for α-synuclein and other proteins that have IREs in the UTRs of their respective encoding mRNAs, including ferritin, TfR, and APP after treatment with Synucleozid-2.0 for 48 h. (E) Quantification of Western blot data, including those shown in D (n = 2 biological replicates). *P < 0.05; **P < 0.01; ***P < 0.001, as determined by two-tailed Student t test. Error bars indicate SD.
Fig. 5. Synucleozid-2.0 and Syn-RiboTAC are selective on the proteome and reduce α-synuclein protein abundance in PD patient-derived dopaminergic neurons. (A) Global proteomics for SH-SY5Y cells treated with Synucleozid-1.0 (1.5 µM), Synucleozid-2.0 (2 µM), and Syn-RIBOTAC (2 µM) for 48 h (n = 3 biological replicates). Dotted lines indicate a false discovery rate (FDR) of 1%. Among 2,813 proteins detected, Synucleozid-2.0 affected 150 (0.53%) proteins, with 55 proteins down-regulated and 95 proteins up-regulated. Syn-RiboTAC showed similar selectivity by affecting 194 (0.56%) proteins among 3,436 total detected, with 114 proteins downregulated and 80 up-regulated. (B) Protein expression levels detectable by proteomics (shown on Y axis) of seven genes enriched by Syn-ChemCLIP after a 16-h treatment period (shown on X axis) were not affected by Synucleozid-2.0 (2 µM) treatment (n = 3 biological replicates). (C) Protein expression levels detectable by proteomics of seven genes enriched by Syn-ChemCLIP are not affected by Syn-RiboTAC (2 µM) treatment (n = 3 biological replicates). (D) Synucleozid-2.0 and Syn-RiboTAC decreased α-synuclein protein levels in human iPSC-derived dopaminergic neurons as determined by ELISA (48 h treatment; n = 3 biological replicates). (E) Syn-RiboTAC decreased SNCA mRNA levels while Synucleozid-2.0 had no effect in iPSCderived dopaminergic neurons (48 h treatment; n = 3 biological replicates except for vehicle-treated control dopaminergic neurons where n = 4 biological replicates). (F) Syn-RiboTAC normalized expression level of about half of the genes in dopaminergic neurons differentiated from PD patient-derived iPSC cells by converging them toward the levels observed in neurons from a control individual after a 48-h treatment period (n = 3 biological replicates). *P < 0.05; **P < 0.01; ***P < 0.001, as determined by two-tailed Student t test. Error bars indicate SD.
Decreasing the intrinsically disordered protein α-synuclein levels by targeting its structured mRNA with a ribonuclease-targeting chimera
  • Article
  • Full-text available

January 2024

·

122 Reads

·

19 Citations

Proceedings of the National Academy of Sciences

α-Synuclein is an important drug target for the treatment of Parkinson’s disease (PD), but it is an intrinsically disordered protein lacking typical small-molecule binding pockets. In contrast, the encoding SNCA mRNA has regions of ordered structure in its 5′ untranslated region (UTR). Here, we present an integrated approach to identify small molecules that bind this structured region and inhibit α-synuclein translation. A drug-like, RNA-focused compound collection was studied for binding to the 5′ UTR of SNCA mRNA, affording Synucleozid-2.0, a drug-like small molecule that decreases α-synuclein levels by inhibiting ribosomes from assembling onto SNCA mRNA. This RNA-binding small molecule was converted into a ribonuclease-targeting chimera (RiboTAC) to degrade cellular SNCA mRNA. RNA-seq and proteomics studies demonstrated that the RiboTAC (Syn-RiboTAC) selectively degraded SNCA mRNA to reduce its protein levels, affording a fivefold enhancement of cytoprotective effects as compared to Synucleozid-2.0. As observed in many diseases, transcriptome-wide changes in RNA expression are observed in PD. Syn-RiboTAC also rescued the expression of ~50% of genes that were abnormally expressed in dopaminergic neurons differentiated from PD patient–derived iPSCs. These studies demonstrate that the druggability of the proteome can be expanded greatly by targeting the encoding mRNAs with both small molecule binders and RiboTAC degraders.

Download

HIV-1 transcriptional modulation: novel host factors and prospective therapeutic strategies

July 2023

·

40 Reads

·

7 Citations

Current Opinion in HIV and AIDS

Purpose of review: This review highlights advances in HIV transcription and epigenetic latency mechanisms and outlines current therapeutic approaches to eliminate or block the HIV-1 latent reservoir. Recent findings: Novel host factors have been reported to modulate HIV-1 transcription and latency. Chromatin affinity purification strategies followed by mass spectrometry (ChAP-MS) identified the chaperone protein p32 to play an important role in HIV-1 transcriptional regulation via interactions with the viral transcriptional activator Tat. Similarly, an shRNA screen identified the methyltransferase SMYD5 contributing to HIV-1 transcriptional activation also by modulating Tat activity. These new factors, among others, represent potential druggable targets that could be explored in the 'block-and-lock' or 'shock-and-kill' approaches. Summary: The HIV-1 latent reservoir is established early after infection, persists during antiretroviral therapy, and is the source of viral rebound after treatment interruption. An HIV cure requires either eliminating this reservoir or blocking latent proviral reactivation in the absence of antiretroviral therapy (ART). Understanding the mechanisms and key-players modulating HIV transcriptional and reactivation may facilitate therapeutic advancements. Here we summarize, the latest findings on host factors' roles in HIV transcriptional regulation.


Transcriptome-Wide Studies of RNA-Targeted Small Molecules Provide a Simple and Selective r(CUG) exp Degrader in Myotonic Dystrophy

June 2023

·

44 Reads

·

12 Citations

ACS Central Science

Myotonic dystrophy type 1 (DM1) is caused by a highly structured RNA repeat expansion, r(CUG)exp, harbored in the 3' untranslated region (3' UTR) of dystrophia myotonica protein kinase (DMPK) mRNA and drives disease through a gain-of-function mechanism. A panel of low-molecular-weight fragments capable of reacting with RNA upon UV irradiation was studied for cross-linking to r(CUG)expin vitro, affording perimidin-2-amine diazirine (1) that bound to r(CUG)exp. The interactions between the small molecule and RNA were further studied by nuclear magnetic resonance (NMR) spectroscopy and molecular modeling. Binding of 1 in DM1 myotubes was profiled transcriptome-wide, identifying 12 transcripts including DMPK that were bound by 1. Augmenting the functionality of 1 with cleaving capability created a chimeric degrader that specifically targets r(CUG)exp for elimination. The degrader broadly improved DM1-associated defects as assessed by RNA-seq, while having limited effects on healthy myotubes. This study (i) provides a platform to investigate molecular recognition of ligands directly in disease-affected cells; (ii) illustrates that RNA degraders can be more specific than the binders from which they are derived; and (iii) suggests that repeating transcripts can be selectively degraded due to the presence of multiple ligand binding sites.


Library-versus-library screening defines new RNA-binding small molecules and druggable targets
a, 2DCS analysis of more than 61 million theoretical interactions, identifying new interactions between small molecules and RNA motifs. b, The newly identified small-molecule RNA binders (n = 344) included 156 different scaffolds that fall into 79 major classes based on scaffold similarities. Among the top 10 most abundant classes (collectively covering 59.6% of all hits), six are new classes (green). c, Motif distribution from a 3 × 3 randomized RNA library used for 2DCS screening. As expected, 3 × 3 and 2 × 2 internal loops comprise the majority (85.4% total) of the library. Motifs that bound to C1–C6 showed a significant enrichment for 3 × 3 internal loops (P < 0.001) and one-nucleotide bulges (P < 0.001). A total of 1,044 motifs bound to C1–C20 with Zobs > 8. Preference for 3 × 3 internal loops and one-nucleotide bulges was collectively observed for these compounds. Of these 1,044 motifs, only 23 (2.2%) are present in highly expressed human transcripts (n = 2,712 total motifs), and 375 are new motifs with no previously known small-molecule binder. Inforna contains over 100,000 RNA–small molecule interactions and 6,453 unique RNA motifs of various types. d, Although around 6% of all miRNAs can be bound by C1–C6, only about 30% of targetable sites within them are functional (Drosha or Dicer processing site) and are therefore predicted to induce a biological effect. The other approximately 70% are unproductive interactions that are predicted to be biologically silent. We identified that 48% of miRNAs that have ligandable non-functional sites are potential substrates for RNase L, which could be targeted by RIBOTACs. Thus, biologically inert binders can be converted into bioactive RIBOTACs that provoke targeted degradation. Statistical significance referred to in c was calculated using two-tailed Student’s t-tests.
Pre-miR-155-RIBOTAC selectively degrades pre-miR-155 in an RNase-L-dependent manner in breast cancer cells
a, Schematic of converting an inert binder engaging pre-miR-155 into an active RIBOTAC degrader. b, Structures of the compounds used to target pre-miR-155. c, The effects of pre-miR-155-RIBOTAC on mature (mat) (n = 4 biological replicates), pre- (n = 3 biological replicates) and pri- (n = 3 biological replicates) miR-155 levels, competed by increasing concentrations of pre-miR-155-binder in MDA-MB-231 cells. d, The effect of siRNA knockdown of RNase L on pre-miR-155-RIBOTAC-mediated cleavage of pre-miR-155 in MDA-MB-231 cells, as determined using RT–qPCR. n = 3 biological replicates. e, Immunoprecipitation of pre-miR-155 using an anti-RNase L antibody in the presence of pre-miR-155-RIBOTAC in MDA-MB-231 cells (n = 3 biological replicates). f, The effect of pre-miR-155-amide-binder (left; 100 nM; n = 4 biological replicates) and pre-miR-155-RIBOTAC (right; 100 nM; n = 3 biological replicates) on the levels of the 373 miRNAs expressed in MDA-MB-231 cells. FC, fold change. g, Western blot analysis of SOCS1, a direct target of miR-155, after treatment of MDA-MB-231 cells with pre-miR-155-RIBOTAC (n = 3 biological replicates). h, The effect of pre-miR-155-RIBOTAC on the activity of a SOCS1 3′ UTR-luciferase reporter transfected into HEK293T cells, establishing both dose (left) and time dependence (right; n = 4 biological replicates). Data are mean ± s.d. (c–e, g and h). Statistical significance was determined using two-tailed Student’s t-tests (c–e, g and h) or a Wald’s test (f).
Source data
Pre-miR-155-RIBOTAC selectively degrades pre-miR-155 and reduces lung colonization in vivo
a, Left, proteome-wide changes in MDA-MB-231 cells treated with pre-miR-155-RIBOTAC (100 nM) versus vehicle (n = 3 biological replicates). Right, pre-miR-155-RIBOTAC significantly upregulated miR-155 related proteins (n = 98 proteins), as indicated by a Kolmogorov–Smirnov analysis (right) of their levels versus all proteins (n = 3 biological replicates). b, The effect of pre-miR-155-RIBOTAC on MDA-MB-231 cell migration (n = 3 biological replicates); 2 fields of view were quantified per replicate. Scale bars, 0.5 mm. c, pre-miR-155-RIBOTAC suppresses lung colonization in vivo, as determined by counting lung nodules (n = 5 mice) and by haematoxylin and eosin (H&E) staining (n = 5 mice; 2 fields of view were quantified per replicate). Scale bars, 1 mm (left) and 0.2 mm (right). d, The effect of pre-miR-155-amide-binder and pre-miR-155-RIBOTAC on pre-miR-155 levels in vivo, as determined by RT–qPCR using primers selective for human pre-miR-155 (n = 3 mice). Data are mean ± s.d. (b and d). Statistical significance was determined using a Wald’s test (a) or two-tailed Student’s t-tests (b–d).
Source data
JUN-RIBOTAC impairs pancreatic tumour cell proliferation and migration by selectively degrading JUN mRNA
a, Schematic of JUN degradation by targeting the JUN IRES. b, The structures of compounds used to target JUN mRNA. c, The effect of JUN-RIBOTAC and JUN-binder on JUN mRNA levels in MIA PaCa-2 cells after treatment for 72 h, as determined using RT–qPCR (n = 6 biological replicates). d, The effect of JUN-RIBOTAC on JUN protein levels in MIA PaCa-2 cells (n = 4 biological replicates). e, The effect of JUN-RIBOTAC on JUN mRNA levels in MIA PaCa-2 cells in which RNase L was knocked down by CRISPR (n = 3 biological replicates) and in the corresponding MIA PaCa-2 control cell line in which CRISPR editing was performed using a scrambled guide RNA (n = 4 biological replicates), as determined using RT–qPCR. f, The effect of JUN-RIBOTAC on the proliferation of MIA PaCa-2 cells (n = 6 biological replicates). g, The effect of JUN-RIBOTAC on the invasiveness of MIA PaCa-2 cells, as determined using a Boyden chamber assay (n = 2 biological replicates; 2 fields of view were quantified per replicate). Data are mean ± s.d. (c–f). Statistical significance was determined using two-tailed Student’s t-tests (d–f) and one-way analysis of variance (ANOVA) adjusted for multiple comparisons (c).
Source data
MYC-RIBOTAC selectively targets MYC in an RNase-L-dependent manner
a, Schematic of the targeted degradation of the MYC IRES. b, Compound structures. c, The effect of MYC-binder and MYC-RIBOTAC on MYC mRNA levels in HeLa cells, as determined using RT–qPCR. n = 3 biological replicates. d, The effect of MYC-RIBOTAC on MYC protein levels in HeLa cells (n = 3 biological replicates). e, The effect of MYC-RIBOTAC on the proliferation (left) and apoptosis (right) of HeLa cells (n = 3 biological replicates). f, The effect of MYC-RIBOTAC on MYC IRES luciferase reporter in HEK293T cells (left) or on a control reporter lacking the IRES (right)(n = 3 biological replicates). g, Transcriptome-wide changes in HeLa cells treated with MYC-RIBOTAC (10 μM) after treatment for 48 h (n = 3 biological replicates). EGR1 is a well-known downstream target of MYC⁵⁰. h, Cumulative distribution analysis of the effect of MYC-RIBOTAC and a MYC-selective siRNA on 87 well-validated downstream targets of MYC, or on the downstream targets of HIF-1α, as indicated by a Kolmogorov–Smirnov analysis of their levels relative to all proteins (n = 3 biological replicates). i, The effect of MYC-Ctr and MYC-RIBOTAC on MYC mRNA levels in Namalwa Burkitt lymphoma cells (n = 3 biological replicates) compared with the vehicle (n = 6 biological replicates). j, The effect of MYC-RIBOTAC on MYC protein levels in Namalwa cells (n = 2 biological replicates). k, The effect of MYC-RIBOTAC on the cell cycle of Namalwa cells. n = 2 biological replicates. l, The ability of MYC-RIBOTAC or MYC-Ctr to induce apoptosis of Namalwa cells (n = 2 biological replicates). m, The effect of MYC-RIBOTAC on colony formation of Namalwa cells (n = 2 biological replicates). Data are mean ± s.d. (c–f and i). Statistical significance was determined using a one-way ANOVA adjusted for multiple comparisons (c), two-tailed Student’s t-tests (d–i), Wald’s test (g), or Kolmogorov–Smirnov test (h).
Source data
Programming inactive RNA-binding small molecules into bioactive degraders

May 2023

·

621 Reads

·

88 Citations

Nature

Target occupancy is often insufficient to elicit biological activity, particularly for RNA, compounded by the longstanding challenges surrounding the molecular recognition of RNA structures by small molecules. Here we studied molecular recognition patterns between a natural-product-inspired small-molecule collection and three-dimensionally folded RNA structures. Mapping these interaction landscapes across the human transcriptome defined structure–activity relationships. Although RNA-binding compounds that bind to functional sites were expected to elicit a biological response, most identified interactions were predicted to be biologically inert as they bind elsewhere. We reasoned that, for such cases, an alternative strategy to modulate RNA biology is to cleave the target through a ribonuclease-targeting chimera, where an RNA-binding molecule is appended to a heterocycle that binds to and locally activates RNase L¹. Overlay of the substrate specificity for RNase L with the binding landscape of small molecules revealed many favourable candidate binders that might be bioactive when converted into degraders. We provide a proof of concept, designing selective degraders for the precursor to the disease-associated microRNA-155 (pre-miR-155), JUN mRNA and MYC mRNA. Thus, small-molecule RNA-targeted degradation can be leveraged to convert strong, yet inactive, binding interactions into potent and specific modulators of RNA function.



Fig. 2. Compound 1 selectively alleviates disease-associated defects in patient-derived cells. (A) Effect of 1 on poly(GP) abundance in protein lysate extracted from c9ALS patient-derived LCLs (n = 3 C9orf72 LCLs, three replicates per concentration in each line). (B) Effect of 1 on poly(GP) abundance in protein lysate extracted from c9ALS patient-derived iPSCs (n = 4 C9orf72 iPSC lines, three replicates per concentration in each line). (C) Effect of 1 on poly(GP) abundance in protein lysate extracted from patient-derived iPSNs (n = 3 C9orf72 iPSN lines, three replicates per concentration in each line). (D) Effect of 1 on C9orf72 intron 1 abundance, which harbors r(G 4 C 2 ) exp , in c9ALS patient-derived LCLs, as determined by qRT-PCR with intron 1-specific primers (n = 3 C9orf72 LCLs, three replicates per concentration in each line). (E) Effect of 1 on C9orf72 intron 1 abundance in c9ALS patient-derived iPSCs, as determined by qRT-PCR with intron 1-specific primers (n = 4 C9orf72 iPSC lines, three replicates per concentration in each line). (F) Effect of 1 on C9orf72 intron 1 abundance in c9ALS patient-derived iPSNs, as determined by qRT-PCR with intron 1-specific primers primers (n = 3 C9orf72 iPSN lines, three replicates per concentration in each line). (G) Effect of 1 on C9orf72 intron 1 3′ splice in a c9ALS patient-derived LCL, as determined by qRT-PCR with primers spanning the intron 1-exon 2 junction (n = 1 C9orf72 LCL, three replicates per concentration). (H) Effect of 1 on C9orf72 intron 1 3′ splice in a c9ALS patient-derived iPSC line, as determined by qRT-PCR with primers spanning the intron 1-exon 2 junction (n = 1 C9orf72 iPSC line, three replicates per concentration). (I) Effect of 1 on C9orf72 intron 1 3′ splice in a c9ALS patient-derived iPSN, as determined by RT-qPCR with primers spanning the intron 1-exon 2 junction (n = 1 C9orf72 iPSN line, three replicates per concentration). RNA quantification was measured relative to GAPDH. Vehicle indicates 0.1% (v/v) dimethyl sulfoxide (DMSO). *P < 0.05, **P < 0.01, ***P < 0.001 , ****P < 0.0001, as determined by a One Way ANOVA with multiple comparisons. Error bars are reported as SD.
Fig. 3. Compound 1 selectively degrades C9orf72 intron 1 via the nuclear exosome and RNA carrier proteins. (A) Effect of co-treating a c9ALS patient-derived iPSC line with 1 and an siRNA targeting hnRNP H, on the abundance of C9orf72 intron 1, as determined by qRT-PCR using intron 1-specific primers (n = 1 c9ALS iPSC line, five replicates per concentration). (B) Left: Representative images of r(G 4 C 2 ) exp foci in a c9ALS patient-derived LCL imaged by confocal microscopy treated with vehicle or 1. Right: Quantification of relative number of r(G 4 C 2 ) exp foci per cell (n = 1 c9ALS LCL, three replicates; 200 nuclei counted per biological sample). (C) Effect of co-treating c9ALS patient-derived iPSCs with 1 and an siRNA targeting CWC22, on the abundance of C9orf72 intron 1, as determined by qRT-PCR using intron 1-specific primers (n = 1 c9ALS iPSC line, five replicates per condition). (D) Effect of co-treating c9ALS patient-derived iPSCs with 1, and siRNAs targeting either XRN1 or XRN2 on the abundance of C9orf72 intron 1, as determined by qRT-PCR using intron 1-specific primers (n = 1 c9ALS iPSC line, five replicates per condition). (E) Effect of treating c9ALS patient-derived iPSCs with 1, siRNAs targeting NEXTPAXT, or both, on the abundance of C9orf72 intron 1, as determined by qRT-PCR using intron 1-specific primers (n = 1 c9ALS iPSC line, five replicates per condition). (F) Effect of co-treating c9ALS patient-derived iPSCs with 1 and siRNAs targeting various components of the exosome (hRRP6, hRRP40, or hRRP44), on the abundance of C9orf72 intron 1, as determined by qRT-PCR using intron 1-specific primers (n = 1 c9ALS iPSC line, five replicates per condition). (G) Schematic representation of the RNA decay mechanism of action upon treatment with 1. RNA abundance was measured and quantified relative to GAPDH. Vehicle indicates 0.1% (v/v) DMSO. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001, as determined by an Unpaired t test with Welch's correction. Error bars are repoted as SD.
A blood–brain penetrant RNA-targeted small molecule triggers elimination of r(G 4 C 2 ) exp in c9ALS/FTD via the nuclear RNA exosome

November 2022

·

159 Reads

·

18 Citations

Proceedings of the National Academy of Sciences

A hexanucleotide repeat expansion in intron 1 of the C9orf72 gene is the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia, or c9ALS/FTD. The RNA transcribed from the expansion, r(G 4 C 2 ) exp , causes various pathologies, including intron retention, aberrant translation that produces toxic dipeptide repeat proteins (DPRs), and sequestration of RNA-binding proteins (RBPs) in RNA foci. Here, we describe a small molecule that potently and selectively interacts with r(G 4 C 2 ) exp and mitigates disease pathologies in spinal neurons differentiated from c9ALS patient-derived induced pluripotent stem cells (iPSCs) and in two c9ALS/FTD mouse models. These studies reveal a mode of action whereby a small molecule diminishes intron retention caused by the r(G 4 C 2 ) exp and allows the liberated intron to be eliminated by the nuclear RNA exosome, a multi-subunit degradation complex. Our findings highlight the complexity of mechanisms available to RNA-binding small molecules to alleviate disease pathologies and establishes a pipeline for the design of brain penetrant small molecules targeting RNA with novel modes of action in vivo.


Study of an RNA-Focused DNA-Encoded Library Informs Design of a Degrader of a r(CUG) Repeat Expansion

November 2022

·

52 Reads

·

23 Citations

Journal of the American Chemical Society

A solid-phase DNA-encoded library (DEL) was studied for binding the RNA repeat expansion r(CUG)exp, the causative agent of the most common form of adult-onset muscular dystrophy, myotonic dystrophy type 1 (DM1). A variety of uncharged and novel RNA binders were identified to selectively bind r(CUG)exp by using a two-color flow cytometry screen. The cellular activity of one binder was augmented by attaching it with a module that directly cleaves r(CUG)exp. In DM1 patient-derived muscle cells, the compound specifically bound r(CUG)exp and allele-specifically eliminated r(CUG)exp, improving disease-associated defects. The approaches herein can be used to identify and optimize ligands and bind RNA that can be further augmented for functionality including degradation.


Low-Molecular Weight Small Molecules Can Potently Bind RNA and Affect Oncogenic Pathways in Cells

November 2022

·

59 Reads

·

17 Citations

Journal of the American Chemical Society

RNA is challenging to target with bioactive small molecules, particularly those of low molecular weight that bind with sufficient affinity and specificity. In this report, we developed a platform to address this challenge, affording a novel bioactive interaction. An RNA-focused small-molecule fragment collection (n = 2500) was constructed by analyzing features in all publicly reported compounds that bind RNA, the largest collection of RNA-focused fragments to date. The RNA-binding landscape for each fragment was studied by using a library-versus-library selection with an RNA library displaying a discrete structural element, probing over 12.8 million interactions, the greatest number of interactions between fragments and biomolecules probed experimentally. Mining of this dataset across the human transcriptome defined a drug-like fragment that potently and specifically targeted the microRNA-372 hairpin precursor, inhibiting its processing into the mature, functional microRNA and alleviating invasive and proliferative oncogenic phenotypes in gastric cancer cells. Importantly, this fragment has favorable properties, including an affinity for the RNA target of 300 ± 130 nM, a molecular weight of 273 Da, and quantitative estimate of drug-likeness (QED) score of 0.8. (For comparison, the mean QED of oral medicines is 0.6 ± 0.2). Thus, these studies demonstrate that a low-molecular weight, fragment-like compound can specifically and potently modulate RNA targets.


Targeting RNA structures with small molecules

August 2022

·

161 Reads

·

368 Citations

Nature Reviews Drug Discovery

RNA adopts 3D structures that confer varied functional roles in human biology and dysfunction in disease. Approaches to therapeutically target RNA structures with small molecules are being actively pursued, aided by key advances in the field including the development of computational tools that predict evolutionarily conserved RNA structures, as well as strategies that expand mode of action and facilitate interactions with cellular machinery. Existing RNA-targeted small molecules use a range of mechanisms including directing splicing — by acting as molecular glues with cellular proteins (such as branaplam and the FDA-approved risdiplam), inhibition of translation of undruggable proteins and deactivation of functional structures in noncoding RNAs. Here, we describe strategies to identify, validate and optimize small molecules that target the functional transcriptome, laying out a roadmap to advance these agents into the next decade. The potential of therapeutically targeting RNA structures with small molecules is being increasingly recognized. Here, Disney and colleagues review strategies to identify, validate and optimize small-molecule RNA binders. Examples of existing RNA-targeted small molecules, as well as challenges and future directions in the field, are discussed.


Citations (9)


... Administration of iron chelators reduce the mRNA expression of α-syn in HEK293 cells (Febbraro et al. 2012). Research by Tong et al. identified Synucleozid-2.0, a small molecule that targets the IRE region of α-syn mRNA, promoting its degradation and reducing α-syn levels (Tong and Zhang 2024). These findings suggest that iron overload facilitates IRP dissociation from α-syn mRNA, enhancing α-syn translation and promoting its aggregation, which worsens PD pathology. ...

Reference:

The interplay of iron, oxidative stress, and α-synuclein in Parkinson’s disease progression
Decreasing the intrinsically disordered protein α-synuclein levels by targeting its structured mRNA with a ribonuclease-targeting chimera

Proceedings of the National Academy of Sciences

... Around 76% of PLWH receive antiretroviral therapy (ART), which has had remarkable success at reducing mortality and morbidity. However, despite its effectiveness, ART is not a cure, and PLWH must maintain lifelong treatment [2][3][4][5]. This necessity arises because HIV persists in a latent state within CD4 + T cells, even in individuals who have been on ART for extended periods. ...

HIV-1 transcriptional modulation: novel host factors and prospective therapeutic strategies
  • Citing Article
  • July 2023

Current Opinion in HIV and AIDS

... [84] (b) Overview of the binders linked to a bleomycin unit utilized to target different RNAs and their K d , highlighted in red. References for Bl-2, Bl-3, Bl-4, Bl-5 and Bl-6 are, [93,46,46,94,95] and, [96] respectively. ...

Transcriptome-Wide Studies of RNA-Targeted Small Molecules Provide a Simple and Selective r(CUG) exp Degrader in Myotonic Dystrophy
  • Citing Article
  • June 2023

ACS Central Science

... Thus, positional reactivity could be a general strategy for improving the selectivity of binding small molecules, as observed for targeted degradation approaches, either directly 100 or via enzymatic recruitment. 101 was not certified by peer review) is the author/funder. All rights reserved. ...

Programming inactive RNA-binding small molecules into bioactive degraders

Nature

... r(GGGGCC) n , in particular, has previously been shown to arrange into hairpin and G-quadruplex (G4) structures, both of which have exhibited potential involvement in disease progression 19 . Indeed, the potential of these structures as therapeutic targets is demonstrated by using small molecule probes that bind to and stabilise them, which leads to amelioration in disease models 11,[19][20][21][22][23] . In particular, the use of ligands to bind G4s has been shown to ameliorate ALS phenotypes in neuronal cells 10,19,20,22 and targeting of the hairpin with a small molecule inhibited repeat-associated non-ATG (RAN) translation and subsequent generation of toxic dipeptide repeats from the C9orf72 gene mutation 11 . ...

A blood–brain penetrant RNA-targeted small molecule triggers elimination of r(G 4 C 2 ) exp in c9ALS/FTD via the nuclear RNA exosome

Proceedings of the National Academy of Sciences

... All chemical materials were purchased from commercial sources and did not need further purification. The ligands HL 1 and HL 2 were synthesized according to the reported procedure [36]. The Fourier-transform infrared (FT-IR) spectrum was measured on Bruker Equinox 55 in 4000-400-cm −1 region with KBr pellets. ...

Study of an RNA-Focused DNA-Encoded Library Informs Design of a Degrader of a r(CUG) Repeat Expansion
  • Citing Article
  • November 2022

Journal of the American Chemical Society

... Notably, Hanabusa, [9][10][11][12] Feringa, [13][14][15][16][17][18] and Tomioka [19][20][21] and other research groups [22][23][24][25][26][27][28][29][30][31][32][33] successfully constructed gelators for organic liquids. However, a more accurate and deeper understanding of these interactions is necessary to design more efficient gelators and apply these materials. ...

Low-Molecular Weight Small Molecules Can Potently Bind RNA and Affect Oncogenic Pathways in Cells
  • Citing Article
  • November 2022

Journal of the American Chemical Society

... The formation of a covalent bond between an RNA and a small molecule binder enables direct target engagement and identification of the binding site, as often employed in the protein-targeting field [ 22 , 23 ]. Dubbed Chemical Cross-Linking and Isolation by Pull-down (Chem-CLIP), an RNA binder is appended with a cross-linking module, such as chlorambucil [ 16 , 20 , 21 ] or photoreactive diazirine [24][25][26], and a purification tag. The latter enables the pull-down of probe-modified transcripts with streptavidin beads or by direct copper-catalyzed azide-alkyne cycloaddition (CuAAC) [ 27 , 28 ] to azide-functionalized beads. ...

Transcriptome-Wide Mapping of Small-Molecule RNA-Binding Sites in Cells Informs an Isoform-Specific Degrader of QSOX1 mRNA
  • Citing Article
  • June 2022

Journal of the American Chemical Society