Xueyi Yang’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 (8)


Figure 2. TAMRA labeling assay to determine cross-linking events. ( A ) Scheme of the cross-linking and TAMRA labeling assay. ( B -D ) Dose responses of the reactions of probes 2a and 4a (chlorambucil-based) ( B ) 2b and 4b (chlorambucil-based) ( C ); and 3 and 5 (diazirine-based) ( D ) with Aptamer 21 (1 μM),
Figure 3. Identifying cross-linked (binding) sites using capillary electrophoresis. ( A ) Scheme of Chem-CLIP and RT stop analysis using capillary electrophoresis. ( B ) R epresentativ e fragment analyzer trace to identify cross-linking sites of 2a (chlorambucil-based Chem-CLIP probe) in Aptamer 21 by RT stops (indicated with arrows; RT stops in DMSO and 2a-treated samples, angled arrows; full-length RNA, horizontal arrows), with or without affinity enrichment. Only cross-linked RNAs remain after the enrichment step. ( C ) R epresentativ e fragment analyzer traces identified unique, UV-dependent RT stop sites for 3 (indicated with arrows; RT stops in 3-treated samples, angled arrows; full-length RNA, horizontal arrows), thus identifying HAP binding sites. ( D ) Secondary str uct ure of Aptamer 21 with cross-linked nucleotides indicated with arrows (2a, hollow arrows; 3, solid arrows).
Figure 4. Studying the read-through of various reverse transcriptases to enable binding site mapping by mutational profiling. ( A ) Schematic str uct ures of different modifications of Aptamer 21 after reaction with various Chem-CLIP probes. 2a-Ak: HAP-binding module with an alkyne handle; 2a-S-CAM (HAP-S-carbamidometh yl): HAP alkyne click ed with disulfide biotin azide, clea v ed b y TCEP and capped with iodoacet amide; 2a-Biotin, HAP alk yne clicked with disulfide biotin azide without clea v age. ( B ) Identification of RT enzymes capable of reading through a compound adduct using capillary electrophoresis. "+B" indicates that Betaine was added to the RT reaction. ( C ) Representative fragment analyzer traces and quantification thereof to assess RT enzyme processivity through adducts of different sizes. "+B" indicates that betaine was added to the RT reaction. * P < .05; ** P < .01; *** P < .001; and **** P < .0001, as determined by a one-way Analysis of Variance (ANOVA) with multiple comparisons.
Figure 5. Mutational mapping f ollo w ed b y nanopore sequencing to identify cross-link ed sites within an RNA target. ( A ) Scheme of Chem-CLIP -MaP -Seq w orkflo w. ( B ) R epresentativ e mutational profile of Aptamer 21 cross-linked to 2a obtained from nanopore sequencing. R elativ e mutation rate of 2a-treated samples at each nucleotide position was calculated by subtracting the mutational rate observed in 4a-treated samples. ( C ) Representative mutational profile of Aptamer 21 cross-linked to diazirine probe 3 obtained from nanopore sequencing. R elativ e mutation rate of 3-treated samples at each nucleotide position was calculated by subtracting the mutational rate observed in 5-treated samples. ( D ) Secondary str uct ure of Aptamer 21 o v erlaid with cross-linked sites for 2a (hollow arrows) and 3 (solid arrows) from mutational profiling studies. Note that affinity enrichment is k e y f or probes in which the extent of cross-linking is < 5%.
Mapping small molecule-RNA binding sites via Chem-CLIP synergized with capillary electrophoresis and nanopore sequencing
  • Article
  • Full-text available

March 2025

·

30 Reads

Nucleic Acids Research

Xueyi Yang

·

Jielei Wang

·

Noah A Springer

·

[...]

·

Target validation and identification of binding sites are keys to the development of bioactive small molecules that target RNA. Herein, we describe optimized protocols to profile small molecule–RNA interactions and to define binding sites of the small molecules in RNAs using covalent chemistry. Various reactive modules appended to an RNA-binding small molecule were studied for cross-linking to the RNA target. Electrophilic modules, whether N-chloroethyl aniline or diazirine, have reactive profiles consistent with induced proximity; however, probes with N-chloroethyl aniline were more reactive and more specific than those with a diazirine cross-linking moiety. Depending upon the identity of the cross-linking module, covalent adducts with different nucleotides that are proximal to a small molecule’s binding site were formed. The nucleotides where cross-linking occurred were elucidated by using two different platforms: (i) automated capillary electrophoresis that identified a binding site by impeding reverse transcriptase, or “RT stops”; and (ii) nanopore sequencing where the cross-link produces mutations in the corresponding complementary DNA formed by reverse transcriptase-polymerase chain reaction amplification of the cross-linked RNA. These approaches are broadly applicable to aid in the advancement of chemical probes targeting RNA, including identifying binding sites and using covalent chemistry to screen for RNA-binding molecules in a high throughput format.

Download

Figure 3. Schematic of the Chem-CLIP workflow for transcriptome-wide mapping of small molecule-RNA binding sites in cells
Figure 4. Example of RNA length distribution before and after random fragmentation to identify small molecule binding sites transcriptome-wide Top, expected length distribution prior to fragmentation, as determined by Bioanalyzer analysis. Bottom, expected length distribution after fragmentation ($150-200 nt), as measured by Bioanalyzer (steps 42-47). Please refer to troubleshooting problem 7 if the length distribution is shorter or longer than expected.
Figure 5. The example skyline plot of the enriched target and the mapped binding site
Protocol for transcriptome-wide mapping of small-molecule RNA-binding sites in live cells

August 2024

·

63 Reads

·

4 Citations

STAR Protocols

Small molecules targeting RNA can be valuable chemical probes and potential therapeutics. The interactions between small molecules, particularly fragments, and RNA, however, can be difficult to detect due to their modest affinities and short residence times. Here, we present a protocol for mapping the molecular fingerprints of small molecules in vitro and throughout the human transcriptome in live cells. We describe steps for compound treatment, cross-linking, RNA extraction, fragmentation, and pull-down. We then detail procedures for RNA sequencing and data analysis. For complete details on the use and execution of this protocol, please refer to Tong et al.¹


Transcriptome-wide mapping of small-molecule RNA-binding sites in live cells

May 2024

·

27 Reads

Small molecules targeting RNA can be valuable chemical probes and potential therapeutics. The interactions between small molecules, particularly fragments, and RNA, however, can be difficult to detect due to their modest affinities and short residence times. Here, we describe the procedures for mapping the molecular fingerprints of small molecules in vitro and throughout the human transcriptome in live cells, identifying both the targets bound by the small molecule and the sites of binding therein. For complete details on the use and execution of this protocol, please refer to 1.


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

January 2024

·

122 Reads

·

21 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.


DNA‐Encoded Libraries and Their Application to RNA

July 2023

·

52 Reads

·

6 Citations

Israel Journal of Chemistry

The functional roles of structured RNAs in the regulation of biological processes, and hence RNA's potential as an effective therapeutic target, have only recently been appreciated. Robust and high‐throughput methods that identify potent RNA ligands are critical to the development of chemical probes and therapeutics. DNA‐encoded libraries (DEL) technology has emerged as a powerful tool for protein ligand discovery, and its ability to generate large, custom‐tailored, and novel chemical space offers unprecedented opportunities to discover the rules of RNA ligand design. In this review, we discuss the basic principles of DEL selection, current progress on the application of DEL to RNA targets, and the outlook of targeting RNA by DEL.


DNA-Encoded Library Screening To Inform Design of a Ribonuclease Targeting Chimera (RiboTAC)

November 2022

·

73 Reads

·

47 Citations

Journal of the American Chemical Society

Ribonuclease targeting chimeras (RiboTACs) induce degradation of an RNA target by facilitating an interaction between an RNA and a ribonuclease (RNase). We describe the screening of a DNA-encoded library (DEL) to identify binders of monomeric RNase L to provide a compound that induced dimerization of RNase L, activating its ribonuclease activity. This compound was incorporated into the design of a next-generation RiboTAC that targeted the microRNA-21 (miR-21) precursor and alleviated a miR-21-associated cellular phenotype in triple-negative breast cancer cells. The RNA-binding module in the RiboTAC is Dovitinib, a known receptor tyrosine kinase (RTK) inhibitor, which was previously identified to bind miR-21 as an off-target. Conversion of Dovitinib into this RiboTAC reprograms the known drug to selectively affect the RNA target. This work demonstrates that DEL can be used to identify compounds that bind and recruit proteins with effector functions in heterobifunctional compounds.


Targeting RNA structures with small molecules

August 2022

·

163 Reads

·

382 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 (4)


... 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

... Fragment-based drug design enables the rapid development of tight and specific small-molecule binders for a target from weak but highly specific low-molecular-weight ligands 32,33 . In recent years, fragment-based technologies have been broadly applied to RNA targeting 34 , including NMR 35,36 , equilibrium dialysis 37 , selective 2′-hydroxyl acylation analysed by primer extension and mutational profiling (SHAPE-MaP) 38 , surface plasmon resonance (SPR) 39 , MS 40 and DNA encoded library (DEL)-based screening 41 . We performed a computational ligandability analysis of the Mango II TO-binding pocket to identify hydrophobic regions, pocket volumes and areas that were well-suited to serve as binding sites for small-molecule fragments. ...

DNA-Encoded Library Screening To Inform Design of a Ribonuclease Targeting Chimera (RiboTAC)
  • Citing Article
  • November 2022

Journal of the American Chemical Society

... Developing scoring methods that generalize beyond the available training data is especially important for the next generation of drugs. Recent advances in drug discovery have expanded interest beyond the traditional druggable proteome, targeting intrinsically disordered proteins [38], protein-protein interactions [33], and RNA tertiary structures [12] as promising avenues for nextgeneration therapeutics. The ability to efficiently identify small-molecule binders for these novel targets could greatly accelerate the development of new treatments. ...

Targeting RNA structures with small molecules
  • Citing Article
  • August 2022

Nature Reviews Drug Discovery

... Since RNA targets are smaller than protein targets, RNA therapeutics have an advantage over protein therapies. Small compounds can change the function of mRNA in solution and in cultured human cells by binding to specific locations in the IRE RNA (Yang et al., 2023;Childs-Disney et al., 2022). These findings demonstrated that the small RNA binding molecules had the same selectivity in binding to folded target RNA structures in solution as they do when they enter living cells. ...

A meditation on accelerating the development of small molecule medicines targeting RNA
  • Citing Article
  • June 2022