1
|
Harris SE, Hu Y, Bridges K, Cavazos FF, Martyr JG, Guzmán BB, Murn J, Aleman MM, Dominguez D. Dissecting RNA selectivity mediated by tandem RNA-binding domains. J Biol Chem 2025; 301:108435. [PMID: 40120682 DOI: 10.1016/j.jbc.2025.108435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
RNA-protein interactions are pivotal to proper gene regulation. Many RNA-binding proteins possess multiple RNA-binding domains; however, how these domains interplay to select and regulate RNA targets remains poorly understood. Here, we investigate three multidomain proteins, Musashi-1, Musashi-2, and unkempt, which share a high degree of RNA specificity, a common feature across RNA-binding proteins. We used massively parallel in vitro assays with unprecedented depth with random or naturally derived RNA sequences and find that individual domains within a protein can have differing affinities, specificities, and motif spacing preferences. We conducted large scale competition assays between these proteins and determined how individual protein specificities and affinities influence competitive binding. Integration of binding and regulation in cells with in vitro specificities showed that target selection involves a combination of the protein intrinsic specificities described here, but cellular context is critical to drive these proteins to motifs in specific transcript regions. Finally, evolutionarily conserved RNA regions displayed evidence of binding multiple RBPs in cultured cells, and these RNA regions represent the highest affinity targets. This work emphasizes the importance of in vitro and in cultured cells studies to fully profile RNA-binding proteins and highlights the complex modes of RNA-protein interactions and the contributing factors in target selection.
Collapse
Affiliation(s)
- Sarah E Harris
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina, USA; Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Yue Hu
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Kaitlin Bridges
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Francisco F Cavazos
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Justin G Martyr
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Bryan B Guzmán
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jernej Murn
- Department of Biochemistry, University of California, Riverside, California, USA; Division of Biomedical Sciences, Center for RNA Biology and Medicine, Riverside, California, USA
| | - Maria M Aleman
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA; RNA Discovery Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Daniel Dominguez
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina, USA; Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA; RNA Discovery Center, University of North Carolina, Chapel Hill, North Carolina, USA.
| |
Collapse
|
2
|
Goel K, Saraogi I. Harnessing RNA-Protein Interactions for Therapeutic Interventions. Chem Asian J 2025; 20:e202401117. [PMID: 39714962 DOI: 10.1002/asia.202401117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
Interactions between RNAs and proteins play a crucial role in various diseases, including viral infections and cancer. Hence, understanding and inhibiting these interactions are important for the development of novel therapeutics. However, the identification of drugs targeting RNA-protein interactions with high specificity and affinity is challenged by our limited molecular understanding of these interactions. Recent focus on structural and biochemical characterization, coupled with high-throughput screening technologies and computational modeling, have accelerated the identification of new RBPs and optimization of potential inhibitors. This review discusses key examples of inhibitors developed over the past decade that effectively disrupt pathogenic RNA-protein interactions. We focus on small molecule and peptide-based inhibitors that have shown promise in disrupting crucial RNA-protein interactions in eukaryotes, prokaryotes, and viruses. We also present the challenges and future directions in this field, emphasizing the need to achieve improved specificity and reduce the off-target effects of the inhibitors. This review aims to contribute to ongoing efforts towards the development of novel therapeutic agents targeting RNA-protein interactions by providing an in-depth analysis of significant developments and emerging trends in this rapidly growing field.
Collapse
Affiliation(s)
- Khushboo Goel
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal Bhauri, Bhopal Bypass Road, Bhopal, Madhya Pradesh, 462066, India
| | - Ishu Saraogi
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal Bhauri, Bhopal Bypass Road, Bhopal, Madhya Pradesh, 462066, India
| |
Collapse
|
3
|
Walters K, Sajek MP, Murphy E, Issaian A, Baldwin A, Harrison E, Daniels M, Reisz JA, Hansen K, D'Alessandro A, Mukherjee N. Small-molecule Ro-08-2750 interacts with many RNA-binding proteins and elicits MUSASHI2-independent phenotypes. RNA (NEW YORK, N.Y.) 2023; 29:1458-1470. [PMID: 37369529 PMCID: PMC10578479 DOI: 10.1261/rna.079605.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023]
Abstract
RNA-binding proteins (RBPs) are key regulators of gene expression. Small molecules targeting these RBP-RNA interactions are a rapidly emerging class of therapeutics for treating a variety of diseases. Ro-08-2750 (Ro) is a small molecule identified as a competitive inhibitor of Musashi (MSI)-RNA interactions. Here, we show that multiple Ro-dependent cellular phenotypes, specifically adrenocortical steroid production and cell viability, are Musashi-2 (MSI2)-independent. Using an unbiased proteome-wide approach, we discovered Ro broadly interacts with RBPs, many containing RRM domains. To confirm this finding, we leveraged the large-scale ENCODE data to identify a subset of RBPs whose depletion phenocopies Ro inhibition, indicating Ro is a promiscuous inhibitor of multiple RBPs. Consistent with broad disruption of ribonucleoprotein complexes, Ro treatment leads to stress granule formation. This strategy represents a generalizable framework for validating the specificity and identifying targets of RBP inhibitors in a cellular context.
Collapse
Affiliation(s)
- Kathryn Walters
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Marcin Piotr Sajek
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland
| | - Elisabeth Murphy
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Aaron Issaian
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Amber Baldwin
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Evan Harrison
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Miles Daniels
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
- Howard University Karsh STEM Scholars Program, Washington DC 20059, USA
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Kirk Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Neelanjan Mukherjee
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| |
Collapse
|
4
|
Sicking M, Falke I, Löblein MT, Eich HT, Götte M, Greve B, Troschel FM. The Musashi RNA-binding proteins in female cancers: insights on molecular mechanisms and therapeutic relevance. Biomark Res 2023; 11:76. [PMID: 37620963 PMCID: PMC10463710 DOI: 10.1186/s40364-023-00516-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023] Open
Abstract
RNA-binding proteins have increasingly been identified as important regulators of gene expression given their ability to bind distinct RNA sequences and regulate their fate. Mounting evidence suggests that RNA-binding proteins are involved in the onset and progression of multiple malignancies, prompting increasing interest in their potential for therapeutic intervention.The Musashi RNA binding proteins Musashi-1 and Musashi-2 were initially identified as developmental factors of the nervous system but have more recently been found to be ubiquitously expressed in physiological tissues and may be involved in pathological cell behavior. Both proteins are increasingly investigated in cancers given dysregulation in multiple tumor entities, including in female malignancies. Recent data suggest that the Musashi proteins serve as cancer stem cell markers as they contribute to cancer cell proliferation and therapy resistance, prompting efforts to identify mechanisms to target them. However, as the picture remains incomplete, continuous efforts to elucidate their role in different signaling pathways remain ongoing.In this review, we focus on the roles of Musashi proteins in tumors of the female - breast, endometrial, ovarian and cervical cancer - as we aim to summarize current knowledge and discuss future perspectives.
Collapse
Affiliation(s)
- Mark Sicking
- Department of Radiation Oncology, University Hospital Münster, Albert Schweitzer-Campus 1, 48149, Münster, Germany
| | - Isabel Falke
- Department of Radiation Oncology, University Hospital Münster, Albert Schweitzer-Campus 1, 48149, Münster, Germany
| | - Maria T Löblein
- Department of Radiation Oncology, University Hospital Münster, Albert Schweitzer-Campus 1, 48149, Münster, Germany
| | - Hans Th Eich
- Department of Radiation Oncology, University Hospital Münster, Albert Schweitzer-Campus 1, 48149, Münster, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Burkhard Greve
- Department of Radiation Oncology, University Hospital Münster, Albert Schweitzer-Campus 1, 48149, Münster, Germany
| | - Fabian M Troschel
- Department of Radiation Oncology, University Hospital Münster, Albert Schweitzer-Campus 1, 48149, Münster, Germany.
| |
Collapse
|
5
|
Matalkah F, Jeong B, Sheridan M, Horstick E, Ramamurthy V, Stoilov P. The Musashi proteins direct post-transcriptional control of protein expression and alternate exon splicing in vertebrate photoreceptors. Commun Biol 2022; 5:1011. [PMID: 36153373 PMCID: PMC9509328 DOI: 10.1038/s42003-022-03990-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 09/13/2022] [Indexed: 11/23/2022] Open
Abstract
The Musashi proteins, MSI1 and MSI2, are conserved RNA binding proteins with a role in the maintenance and renewal of stem cells. Contrasting with this role, terminally differentiated photoreceptor cells express high levels of MSI1 and MSI2, pointing to a role for the two proteins in vision. Combined knockout of Msi1 and Msi2 in mature photoreceptor cells abrogated the retinal response to light and caused photoreceptor cell death. In photoreceptor cells the Musashi proteins perform distinct nuclear and cytoplasmic functions. In the nucleus, the Musashi proteins promote splicing of photoreceptor-specific alternative exons. Surprisingly, conserved photoreceptor-specific alternative exons in genes critical for vision proved to be dispensable, raising questions about the selective pressures that lead to their conservation. In the cytoplasm MSI1 and MSI2 activate protein expression. Loss of Msi1 and Msi2 lead to reduction in the levels of multiple proteins including proteins required for vision and photoreceptor survival. The requirement for MSI1 and MSI2 in terminally differentiated photoreceptors alongside their role in stem cells shows that, depending on cellular context, these two proteins can control processes ranging from cell proliferation to sensory perception.
Collapse
Affiliation(s)
- Fatimah Matalkah
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, USA
| | - Bohye Jeong
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, USA
| | - Macie Sheridan
- Undergraduate Program in Biochemistry, West Virginia University, Morgantown, WV, USA
| | - Eric Horstick
- Department of Biology, West Virginia University, Morgantown, WV, USA
- Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | - Visvanathan Ramamurthy
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, USA
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV, USA
| | - Peter Stoilov
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
6
|
Darai N, Mahalapbutr P, Wolschann P, Lee VS, Wolfinger MT, Rungrotmongkol T. Theoretical studies on RNA recognition by Musashi 1 RNA-binding protein. Sci Rep 2022; 12:12137. [PMID: 35840700 PMCID: PMC9287312 DOI: 10.1038/s41598-022-16252-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 07/07/2022] [Indexed: 01/12/2023] Open
Abstract
The Musashi (MSI) family of RNA-binding proteins, comprising the two homologs Musashi-1 (MSI1) and Musashi-2 (MSI2), typically regulates translation and is involved in cell proliferation and tumorigenesis. MSI proteins contain two ribonucleoprotein-like RNA-binding domains, RBD1 and RBD2, that bind single-stranded RNA motifs with a central UAG trinucleotide with high affinity and specificity. The finding that MSI also promotes the replication of Zika virus, a neurotropic Flavivirus, has triggered further investigations of the biochemical principles behind MSI–RNA interactions. However, a detailed molecular understanding of the specificity of MSI RBD1/2 interaction with RNA is still missing. Here, we performed computational studies of MSI1–RNA association complexes, investigating different RNA pentamer motifs using molecular dynamics simulations with binding free energy calculations based on the solvated interaction energy method. Simulations with Alphafold2 suggest that predicted MSI protein structures are highly similar to experimentally determined structures. The binding free energies show that two out of four RNA pentamers exhibit a considerably higher binding affinity to MSI1 RBD1 and RBD2, respectively. The obtained structural information on MSI1 RBD1 and RBD2 will be useful for a detailed functional and mechanistic understanding of this type of RNA–protein interactions.
Collapse
Affiliation(s)
- Nitchakan Darai
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Panupong Mahalapbutr
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Peter Wolschann
- Department of Theoretical Chemistry, University of Vienna, Währinger Strasse 17, 1090, Vienna, Austria
| | - Vannajan Sanghiran Lee
- Department of Chemistry, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Michael T Wolfinger
- Department of Theoretical Chemistry, University of Vienna, Währinger Strasse 17, 1090, Vienna, Austria. .,Research Group Bioinformatics and Computational Biology, Faculty of Computer Science, University of Vienna, Währinger Strasse 29, 1090, Vienna, Austria.
| | - Thanyada Rungrotmongkol
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok, 10330, Thailand. .,Center of Excellence in Biocatalyst and Sustainable Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
7
|
Wang J, Lan L, Wu X, Xu L, Miao Y. Mechanism of RNA recognition by a Musashi RNA-binding protein. Curr Res Struct Biol 2021; 4:10-20. [PMID: 34988468 PMCID: PMC8695263 DOI: 10.1016/j.crstbi.2021.12.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 10/31/2021] [Accepted: 12/07/2021] [Indexed: 12/21/2022] Open
Abstract
The Musashi RNA-binding proteins (RBPs) regulate translation of target mRNAs and maintenance of cell stemness and tumorigenesis. Musashi-1 (MSI1), long considered as an intestinal and neural stem cell marker, has been more recently found to be over expressed in many cancers. It has served as an important drug target for treating acute myeloid leukemia and solid tumors such as ovarian, colorectal and bladder cancer. One of the reported binding targets of MSI1 is Numb, a negative regulator of the Notch signaling. However, the dynamic mechanism of Numb RNA binding to MSI1 remains unknown, largely hindering effective drug design targeting this critical interaction. Here, we have performed extensive all-atom microsecond-timescale simulations using a robust Gaussian accelerated molecular dynamics (GaMD) method, which successfully captured multiple times of spontaneous and highly accurate binding of the Numb RNA from bulk solvent to the MSI1 protein target site. GaMD simulations revealed that Numb RNA binding to MSI1 involved largely induced fit in both the RNA and protein. The simulations also identified important low-energy intermediate conformational states during RNA binding, in which Numb interacted mainly with the β2-β3 loop and C terminus of MSI1. The mechanistic understanding of RNA binding obtained from our GaMD simulations is expected to facilitate rational structure-based drug design targeting MSI1 and other RBPs.
Collapse
Affiliation(s)
- Jinan Wang
- Center for Computational Biology, University of Kansas, Lawrence, KS, 66047, USA
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66047, USA
| | - Lan Lan
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66047, USA
| | - Xiaoqing Wu
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66047, USA
| | - Liang Xu
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66047, USA
- Department of Radiation Oncology, The University of Kansas Cancer Center, Kansas City, KS, 66160, USA
| | - Yinglong Miao
- Center for Computational Biology, University of Kansas, Lawrence, KS, 66047, USA
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66047, USA
| |
Collapse
|
8
|
Heyes E, Schmidt L, Manhart G, Eder T, Proietti L, Grebien F. Identification of gene targets of mutant C/EBPα reveals a critical role for MSI2 in CEBPA-mutated AML. Leukemia 2021; 35:2526-2538. [PMID: 33623142 PMCID: PMC7611617 DOI: 10.1038/s41375-021-01169-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 01/12/2021] [Accepted: 01/28/2021] [Indexed: 01/31/2023]
Abstract
Mutations in the gene encoding the transcription factor CCAAT/enhancer-binding protein alpha (C/EBPα) occur in 10-15% of acute myeloid leukemia (AML). Frameshifts in the CEBPA N-terminus resulting in exclusive expression of a truncated p30 isoform represent the most prevalent type of CEBPA mutations in AML. C/EBPα p30 interacts with the epigenetic machinery, but it is incompletely understood how p30-induced changes cause leukemogenesis. We hypothesized that critical effector genes in CEBPA-mutated AML are dependent on p30-mediated dysregulation of the epigenome. We mapped p30-associated regulatory elements (REs) by ATAC-seq and ChIP-seq in a myeloid progenitor cell model for p30-driven AML that enables inducible RNAi-mediated knockdown of p30. Concomitant p30-dependent changes in gene expression were measured by RNA-seq. Integrative analysis identified 117 p30-dependent REs associated with 33 strongly down-regulated genes upon p30-knockdown. CRISPR/Cas9-mediated mutational disruption of these genes revealed the RNA-binding protein MSI2 as a critical p30-target. MSI2 knockout in p30-driven murine AML cells and in the CEBPA-mutated human AML cell line KO-52 caused proliferation arrest and terminal myeloid differentiation, and delayed leukemia onset in vivo. In summary, this work presents a comprehensive dataset of p30-dependent effects on epigenetic regulation and gene expression and identifies MSI2 as an effector of the C/EBPα p30 oncoprotein.
Collapse
Affiliation(s)
- Elizabeth Heyes
- University of Veterinary Medicine, Institute of Medical Biochemistry, Vienna, Austria
| | - Luisa Schmidt
- University of Veterinary Medicine, Institute of Medical Biochemistry, Vienna, Austria
| | - Gabriele Manhart
- University of Veterinary Medicine, Institute of Medical Biochemistry, Vienna, Austria
| | - Thomas Eder
- University of Veterinary Medicine, Institute of Medical Biochemistry, Vienna, Austria
| | - Ludovica Proietti
- University of Veterinary Medicine, Institute of Medical Biochemistry, Vienna, Austria
| | - Florian Grebien
- University of Veterinary Medicine, Institute of Medical Biochemistry, Vienna, Austria.
| |
Collapse
|
9
|
RNA-Targeted Therapies and High-Throughput Screening Methods. Int J Mol Sci 2020; 21:ijms21082996. [PMID: 32340368 PMCID: PMC7216119 DOI: 10.3390/ijms21082996] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 02/07/2023] Open
Abstract
RNA-binding proteins (RBPs) are involved in regulating all aspects of RNA metabolism, including processing, transport, translation, and degradation. Dysregulation of RNA metabolism is linked to a plethora of diseases, such as cancer, neurodegenerative diseases, and neuromuscular disorders. Recent years have seen a dramatic shift in the knowledge base, with RNA increasingly being recognised as an attractive target for precision medicine therapies. In this article, we are going to review current RNA-targeted therapies. Furthermore, we will scrutinise a range of drug discoveries targeting protein-RNA interactions. In particular, we will focus on the interplay between Lin28 and let-7, splicing regulatory proteins and survival motor neuron (SMN) pre-mRNA, as well as HuR, Musashi, proteins and their RNA targets. We will highlight the mechanisms RBPs utilise to modulate RNA metabolism and discuss current high-throughput screening strategies. This review provides evidence that we are entering a new era of RNA-targeted medicine.
Collapse
|
10
|
Lan L, Xing M, Kashipathy M, Douglas J, Gao P, Battaile K, Hanzlik R, Lovell S, Xu L. Crystal and solution structures of human oncoprotein Musashi-2 N-terminal RNA recognition motif 1. Proteins 2019; 88:573-583. [PMID: 31603583 PMCID: PMC7079100 DOI: 10.1002/prot.25836] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/16/2019] [Accepted: 09/28/2019] [Indexed: 01/03/2023]
Abstract
Musashi‐2 (MSI2) belongs to Musashi family of RNA binding proteins (RBP). Like Musashi‐1 (MSI1), it is overexpressed in a variety of cancers and is a promising therapeutic target. Both MSI proteins contain two N‐terminal RNA recognition motifs and play roles in posttranscriptional regulation of target mRNAs. Previously, we have identified several inhibitors of MSI1, all of which bind to MSI2 as well. In order to design MSI2‐specific inhibitors and compare the differences of binding mode of the inhibitors, we set out to solve the structure of MSI2‐RRM1, the key motif that is responsible for the binding. Here, we report the crystal structure and the first NMR solution structure of MSI2‐RRM1, and compare these to the structures of MSI1‐RBD1 and other RBPs. A high degree of structural similarity was observed between the crystal and solution NMR structures. MSI2‐RRM1 shows a highly similar overall folding topology to MSI1‐RBD1 and other RBPs. The structural information of MSI2‐RRM1 will be helpful for understanding MSI2‐RNA interaction and for guiding rational drug design of MSI2‐specific inhibitors.
Collapse
Affiliation(s)
- Lan Lan
- Department of Molecular Biosciences, The University of Kansas, Lawrence, Kansas
| | - Minli Xing
- Bio-NMR Core Facility, NIH COBRE in Protein Structure and Function, The University of Kansas, Lawrence, Kansas
| | - Maithri Kashipathy
- Protein Structure Laboratory, NIH COBRE in Protein Structure and Function, The University of Kansas, Lawrence, Kansas
| | - Justin Douglas
- Bio-NMR Core Facility, NIH COBRE in Protein Structure and Function, The University of Kansas, Lawrence, Kansas
| | - Philip Gao
- Protein Production Group, NIH COBRE in Protein Structure and Function, The University of Kansas, Lawrence, Kansas
| | - Kevin Battaile
- IMCA-CAT, Hauptman Woodward Medical Research Institute, Argonne, Illinois
| | - Robert Hanzlik
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas
| | - Scott Lovell
- Protein Structure Laboratory, NIH COBRE in Protein Structure and Function, The University of Kansas, Lawrence, Kansas
| | - Liang Xu
- Department of Molecular Biosciences, The University of Kansas, Lawrence, Kansas.,Department of Radiation Oncology, The University of Kansas Cancer Center, Kansas City, Kansas
| |
Collapse
|