Affecting RNA biology genome-wide by binding small molecules and chemically induced proximity

https://doi.org/10.1016/j.cbpa.2021.03.006Get rights and content

Abstract

The ENCODE and genome-wide association projects have shown that much of the genome is transcribed into RNA and much less is translated into protein. These and other functional studies suggest that the druggable transcriptome is much larger than the druggable proteome. This review highlights approaches to define druggable RNA targets and structure–activity relationships across genomic RNA. Binding compounds can be identified and optimized into structure-specific ligands by using sequence-based design with various modes of action, for example, inhibiting translation or directing pre-mRNA splicing outcomes. In addition, strategies to direct protein activity against an RNA of interest via chemically induced proximity is a burgeoning area that has been validated both in cells and in preclinical animal models, and we describe that it may allow rapid access to new avenues to affect RNA biology. These approaches and the unique modes of action suggest that more RNAs are potentially amenable to targeting than proteins.

Introduction

Although ∼80% of the human genome is transcribed into RNA, only 1% is translated into protein [1], suggesting that there are more opportunities to affect biology at the transcriptional level (Fig. 1). Furthermore, RNA has diverse functions tied to its structure [2], including regulating pre-mRNA splicing outcomes [3], cellular localization [4], phase separation [5], and microRNA (miRNA) biogenesis [6,7]. Indeed, RNA-targeting oligonucleotide-based modalities have garnered Food and Drug Administration (FDA) approval, including those that target RNAs associated with central nervous system disorders such as spinal muscular atrophy (SMA) [8] and liver disease [9]. Although these medicines have transformed patients' lives, their transcriptome-wide implementation has been limited to tissues readily amenable to oligonucleotide targeting, that is, direct delivery to the central nervous system, liver, or kidney.

Highly structured regions in RNA are often functional [10], and the three-dimensional shapes they adopt could provide small-molecule binding pockets [11], occupied by compounds with complementarity of size, shape, stacking, and charge. Although there are challenges associated with small-molecule targeting of RNA, small molecules can be penetrant to a variety of tissues, and often, lead compounds can be medicinally optimized to enhance target engagement [12] and minimize suboptimal features [13]. Herein, we discuss lead drug and chemical probe discovery against RNA targets on a genome-wide scale to elicit effects on the proteome (Table 1): downregulation of ‘undruggable’ proteins by targeting RNA structures in untranslated regions, upregulation of proteins by targeting miRNAs that repress their expression, and altering the ratio of protein isoforms by influencing pre-mRNA splicing outcomes. Collectively, targeting RNA structures with small molecules can indeed be an effective way to study and control biology.

Section snippets

Defining small-molecule binding landscapes for RNA structures

Targeting RNA with small molecules is not without its challenges, particularly garnering sufficient selectivity and potency to elicit a biological outcome. Selectivity is a composite of (i) the selectivity of the small molecule for the desired RNA structure, (ii) the presence of that structure elsewhere in the transcriptome, (iii) the relative expression levels of all targets with said structure, and (iv) the structure's functionality. We developed a method, two-dimensional combinatorial

Affecting RNA biology: binding small molecules

As coding and noncoding RNAs adopt 3D folds that influence their biological roles, there are likely many ways to modulate their function. This section describes how simple binding compounds can alter cellular protein content by various mechanisms (Fig. 2).

Genome-wide design: fully functionalized fragments

Fragment-based drug discovery has been a key technology in the protein-targeting field. Unlike traditional screening methods of large small-molecule libraries (∼106+), fragment-based approaches use a much smaller number (∼103) of low-molecular-weight compounds. Indeed, a recent study combined fragment-based ligand discovery with a cross-linking and mass spectrometry strategy that uses fully functionalized fragments (FFFs) equipped with an alkyne and a photoreactive diazirine [81]. The strategy

Affecting RNA biology: induced proximity

In addition to inhibiting RNA function through structure-specific targeting of tandem binding pockets, fragment assembly has enormous potential to enable design of small molecules with MOAs outside simple binding; that is, a structure-specific RNA-binding fragment can be coupled to a fragment that recruits enzymes or proteins that act on RNA, referred to as induced proximity. Originally developed for protein-targeted ligands by recruiting ubiquitin transferase [83] to target proteins for

Summary and outlook

There is vast potential to alter the proteome by targeting RNA, not only for RNA-related diseases but also for ‘undruggable’ proteins. RNA's limited number of building blocks and diverse folding landscapes come with unique and challenging problems. However, its hierarchical folding and the accurate modeling thereof can be exploited to design lead molecules. With additional methodology such as Chem-CLIP and Chem-CLIP Fragment Mapping (Chem-CLIP-Frag-Map), small-molecule specificity can be

Declaration of competing interest

The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: MDD is a founder of Expansion Therapeutics.

Acknowledgements

Funding was provided by the National Institutes of Health (R01 CA249180, P01 NS09914, R35 NS116846, and UG3 NS116921 to MDD), the Department of Defense (W81XWH-18-0718 and W81XWH-19-PRMRP-IIRA to MDD), and the Huntington's Disease Society of America (JTB).

References (91)

  • Y. Naro et al.

    Small molecule inhibition of microRNA miR-21 rescues chemosensitivity of renal-cell carcinoma to topotecan

    J Med Chem

    (2018)
  • M.D. Shortridge et al.

    A macrocyclic peptide ligand binds the oncogenic microRNA-21 precursor and suppresses Dicer processing

    ACS Chem Biol

    (2017)
  • J.P. Diaz et al.

    Association of a peptoid ligand with the apical loop of pri-miR-21 inhibits cleavage by Drosha

    RNA

    (2014)
  • N.A. Naryshkin et al.

    Motor neuron disease. SMN2 splicing modifiers improve motor function and longevity in mice with spinal muscular atrophy

    Science

    (2014)
  • M. Salani et al.

    Development of a screening platform to identify small molecules that modify ELP1 pre-mRNA splicing in familial dysautonomia

    SLAS Discov

    (2019)
  • G. Miglietta et al.

    RNA G-quadruplexes in kirsten ras (KRAS) oncogene as targets for small molecules inhibiting translation

    J Med Chem

    (2017)
  • G.W. Collie et al.

    Structural basis of telomeric RNA quadruplex--acridine ligand recognition

    J Am Chem Soc

    (2011)
  • M.M. Gosink et al.

    Redirecting the specificity of ubiquitination by modifying ubiquitin-conjugating enzymes

    Proc Natl Acad Sci U S A

    (1995)
  • R. Santos et al.

    A comprehensive map of molecular drug targets

    Nat Rev Drug Discov

    (2017)
  • M. Esteller

    Non-coding RNAs in human disease

    Nat Rev Genet

    (2011)
  • L. Sun et al.

    RNA structure maps across mammalian cellular compartments

    Nat Struct Mol Biol

    (2019)
  • E.M. Langdon et al.

    mRNA structure determines specificity of a polyQ-driven phase separation

    Science

    (2018)
  • S.C. Kwon et al.

    Molecular basis for the single-nucleotide precision of primary microRNA processing

    Mol Cell

    (2019)
  • H. Ratni et al.

    Discovery of risdiplam, a selective survival of motor neuron-2 ( SMN2) gene splicing modifier for the treatment of spinal muscular atrophy (SMA)

    J Med Chem

    (2018)
  • C.A. Stein et al.

    FDA-approved oligonucleotide therapies in 2017

    Mol Ther

    (2017)
  • L.R. Ganser et al.

    High-performance virtual screening by targeting a high-resolution RNA dynamic ensemble

    Nat Struct Mol Biol

    (2018)
  • N.F. Rizvi et al.

    Targeting RNA with small molecules: identification of selective, RNA-binding small molecules occupying drug-like chemical space

    SLAS Discov

    (2020)
  • M. Zuker

    Mfold web server for nucleic acid folding and hybridization prediction

    Nucleic Acids Res

    (2003)
  • R.J. Andrews et al.

    ScanFold: an approach for genome-wide discovery of local RNA structural elements-applications to Zika virus and HIV

    PeerJ

    (2018)
  • D.H. Mathews

    Revolutions in RNA secondary structure prediction

    J Mol Biol

    (2006)
  • M. Parisien et al.

    The MC-Fold and MC-Sym pipeline infers RNA structure from sequence data

    Nature

    (2008)
  • T.D. Schneider et al.

    Sequence logos: a new way to display consensus sequences

    Nucleic Acids Res

    (1990)
  • M. Nettling et al.

    Difflogo: a comparative visualization of sequence motifs

    BMC Bioinf

    (2015)
  • C. Hong et al.

    Formation of a ligand-assisted complex of two RNA hairpin loops

    Chemistry

    (2014)
  • C. Dohno et al.

    Molecular glue for RNA: regulating RNA structure and function through synthetic RNA binding molecules

    Chembiochem

    (2019)
  • S.P. Velagapudi et al.

    Sequence-based design of bioactive small molecules that target precursor microRNAs

    Nat Chem Biol

    (2014)
  • M.D. Disney et al.

    Inforna 2.0: a platform for the sequence-based design of small molecules targeting structured RNAs

    ACS Chem Biol

    (2016)
  • H. Su et al.

    Recognition of small molecule-RNA binding sites using RNA sequence and structure

    Bioinformatics

    (2021)
  • D. Mitchell et al.

    Probing RNA structure in vivo

    Curr Opin Struct Biol

    (2019)
  • H. Paulson

    Repeat expansion diseases

    Handb Clin Neurol

    (2018)
  • R. Ondono et al.

    Design of novel small molecule base-pair recognizers of toxic CUG RNA transcripts characteristics of DM1

    Comput Struct Biotechnol J

    (2021)
  • J. Lee et al.

    Intrinsically cell-penetrating multivalent and multitargeting ligands for myotonic dystrophy type 1

    Proc Natl Acad Sci U S A

    (2019)
  • L.D. Hagler et al.

    Expanded DNA and RNA trinucleotide repeats in myotonic dystrophy type 1 select their own multitarget, sequence-selective inhibitors

    Biochemistry

    (2020)
  • A. Garcia-Lopez et al.

    In vivo discovery of a peptide that prevents CUG-RNA hairpin formation and reverses RNA toxicity in myotonic dystrophy models

    Proc Natl Acad Sci U S A

    (2011)
  • S.G. Rzuczek et al.

    Precise small-molecule recognition of a toxic CUG RNA repeat expansion

    Nat Chem Biol

    (2017)
  • View full text