1
|
Li J, Xu T, Zheng Y, Liu D, Zhang C, Li J, Wang ZA, Du Y. In Silico Study on a Binding Mechanism of ssDNA Aptamers Targeting Glycosidic Bond-Containing Small Molecules. Anal Chem 2024; 96:5056-5064. [PMID: 38497564 DOI: 10.1021/acs.analchem.4c00927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Aptamer-based detection targeting glycoconjugates has attracted significant attention for its remarkable potential in identifying structural changes in saccharides in different stages of various diseases. However, the challenges in screening aptamers for small carbohydrates or glycoconjugates, which contain highly flexible and diverse glycosidic bonds, have hindered their application and commercialization. In this study, we investigated the binding conformations between three glycosidic bond-containing small molecules (GlySMs; glucose, N-acetylneuraminic acid, and neomycin) and their corresponding aptamers in silico, and analyzed factors contributing to their binding affinities. Based on the findings, a novel binding mechanism was proposed, highlighting the central role of the stem structure of the aptamer in binding and recognizing GlySMs and the auxiliary role of the mismatched bases in the adjacent loop. Guided by this binding mechanism, an aptamer with a higher 6'-sialyllactose binding affinity was designed, achieving a KD value of 4.54 ± 0.64 μM in vitro through a single shear and one mutation. The binding mechanism offers crucial guidance for designing high-affinity aptamers, enhancing the virtual screening efficiency for GlySMs. This streamlined workflow filters out ineffective binding sites, accelerating aptamer development and providing novel insights into glycan-nucleic acid interactions.
Collapse
Affiliation(s)
- Jiaqing Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, 1 North second Street, Zhongguancun, Haidian District, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, 1 North second Street, Zhongguancun, Haidian District, Beijing 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, No.19A Yuquan Road, Shijingshan District, Beijing 100049, China
| | - Tong Xu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, 1 North second Street, Zhongguancun, Haidian District, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, 1 North second Street, Zhongguancun, Haidian District, Beijing 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, No.19A Yuquan Road, Shijingshan District, Beijing 100049, China
| | - Yalan Zheng
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Dongdong Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, 1 North second Street, Zhongguancun, Haidian District, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, 1 North second Street, Zhongguancun, Haidian District, Beijing 100190, China
| | - Chen Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, 1 North second Street, Zhongguancun, Haidian District, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, 1 North second Street, Zhongguancun, Haidian District, Beijing 100190, China
| | - Jianjun Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, 1 North second Street, Zhongguancun, Haidian District, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, 1 North second Street, Zhongguancun, Haidian District, Beijing 100190, China
| | - Zhuo A Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, 1 North second Street, Zhongguancun, Haidian District, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, 1 North second Street, Zhongguancun, Haidian District, Beijing 100190, China
| | - Yuguang Du
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, 1 North second Street, Zhongguancun, Haidian District, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, 1 North second Street, Zhongguancun, Haidian District, Beijing 100190, China
| |
Collapse
|
2
|
Wang K, Zhou R, Wu Y, Li M. RLBind: a deep learning method to predict RNA-ligand binding sites. Brief Bioinform 2023; 24:6832814. [PMID: 36398911 DOI: 10.1093/bib/bbac486] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/28/2022] [Accepted: 10/14/2022] [Indexed: 11/19/2022] Open
Abstract
Identification of RNA-small molecule binding sites plays an essential role in RNA-targeted drug discovery and development. These small molecules are expected to be leading compounds to guide the development of new types of RNA-targeted therapeutics compared with regular therapeutics targeting proteins. RNAs can provide many potential drug targets with diverse structures and functions. However, up to now, only a few methods have been proposed. Predicting RNA-small molecule binding sites still remains a big challenge. New computational model is required to better extract the features and predict RNA-small molecule binding sites more accurately. In this paper, a deep learning model, RLBind, was proposed to predict RNA-small molecule binding sites from sequence-dependent and structure-dependent properties by combining global RNA sequence channel and local neighbor nucleotides channel. To our best knowledge, this research was the first to develop a convolutional neural network for RNA-small molecule binding sites prediction. Furthermore, RLBind also can be used as a potential tool when the RNA experimental tertiary structure is not available. The experimental results show that RLBind outperforms other state-of-the-art methods in predicting binding sites. Therefore, our study demonstrates that the combination of global information for full-length sequences and local information for limited local neighbor nucleotides in RNAs can improve the model's predictive performance for binding sites prediction. All datasets and resource codes are available at https://github.com/KailiWang1/RLBind.
Collapse
Affiliation(s)
- Kaili Wang
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Renyi Zhou
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Yifan Wu
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Min Li
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| |
Collapse
|
3
|
G R, Mitra A, Pk V. Predicting functional riboSNitches in the context of alternative splicing. Gene X 2022; 837:146694. [PMID: 35738445 DOI: 10.1016/j.gene.2022.146694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 05/11/2022] [Accepted: 06/17/2022] [Indexed: 11/19/2022] Open
Abstract
RNAs are the major regulators of gene expression, and their secondary structures play crucial roles at different levels. RiboSNitches are disease-associated SNPs that cause changes in the pre-mRNA secondary structural ensemble. Several riboSNitches have been detected in the 5' and 3' untranslated regions and lncRNA. Although cases of secondary structural elements playing a regulatory role in alternative splicing are known, regions specific to splicing events, such as splice junctions have not received much attention. We tested splice-site mutations for their efficiency in disrupting the secondary structure and hypothesized that these could play a crucial role in alternative splicing. Multiple riboSNitch prediction methods were applied to obtain overlapping results that are potentially more reliable. Putative riboSNitches were identified from aberrant 5' and 3' splice site mutations, cancer-causing somatic mutations, and genes that harbor the regulatory RNA secondary structural elements. Our workflow for predicting riboSNitches associated with alternative splicing is novel and paves the way for subsequent experimental validation.
Collapse
Affiliation(s)
- Ramya G
- Center for Computational Natural Sciences and Bioinformatics, International Institute of Information Technology, Gachibowli, Hyderabad, Telangana 500032, India.
| | - Abhijit Mitra
- Center for Computational Natural Sciences and Bioinformatics, International Institute of Information Technology, Gachibowli, Hyderabad, Telangana 500032, India.
| | - Vinod Pk
- Center for Computational Natural Sciences and Bioinformatics, International Institute of Information Technology, Gachibowli, Hyderabad, Telangana 500032, India.
| |
Collapse
|
4
|
Przewodowska D, Marzec W, Madetko N. Novel Therapies for Parkinsonian Syndromes-Recent Progress and Future Perspectives. Front Mol Neurosci 2021; 14:720220. [PMID: 34512258 PMCID: PMC8427499 DOI: 10.3389/fnmol.2021.720220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 07/23/2021] [Indexed: 11/23/2022] Open
Abstract
Background: Atypical parkinsonian syndromes are rare, fatal neurodegenerative diseases associated with abnormal protein accumulation in the brain. Examples of these syndromes include progressive supranuclear palsy, multiple system atrophy, and corticobasal degeneration. A common clinical feature in parkinsonism is a limited improvement with levodopa. So far, there are no disease-modifying treatments to address these conditions, and therapy is only limited to the alleviation of symptoms. Diagnosis is devastating for patients, as prognosis is extremely poor, and the disease tends to progress rapidly. Currently, potential causes and neuropathological mechanisms involved in these diseases are being widely investigated. Objectives: The goal of this review is to summarize recent advances and gather emerging disease-modifying therapies that could slow the progression of atypical parkinsonian syndromes. Methods: PubMed and Google Scholar databases were searched regarding novel perspectives for atypical parkinsonism treatment. The following medical subject headings were used: "atypical parkinsonian syndromes-therapy," "treatment of atypical parkinsonian syndromes," "atypical parkinsonian syndromes-clinical trial," "therapy of tauopathy," "alpha-synucleinopathy treatment," "PSP therapy/treatment," "CBD therapy/treatment," "MSA therapy/treatment," and "atypical parkinsonian syndromes-disease modifying." All search results were manually reviewed prior to inclusion in this review. Results: Neuroinflammation, mitochondrial dysfunction, microglia activation, proteasomal impairment, and oxidative stress play a role in the neurodegenerative process. Ongoing studies and clinical trials target these components in order to suppress toxic protein accumulation. Various approaches such as stem cell therapy, anti-aggregation/anti-phosphorylation agent administration, or usage of active and passive immunization appear to have promising results. Conclusion: Presently, disease-modifying strategies for atypical parkinsonian syndromes are being actively explored, with encouraging preliminary results. This leads to an assumption that developing accurate, safe, and progression-halting treatment is not far off. Nevertheless, the further investigation remains necessary.
Collapse
Affiliation(s)
- Dominika Przewodowska
- Students' Scientific Association of the Department of Neurology, Medical University of Warsaw, Warsaw, Poland
| | - Weronika Marzec
- Students' Scientific Association of the Department of Neurology, Medical University of Warsaw, Warsaw, Poland
| | - Natalia Madetko
- Department of Neurology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
5
|
Angelbello AJ, Chen JL, Disney MD. Small molecule targeting of RNA structures in neurological disorders. Ann N Y Acad Sci 2020; 1471:57-71. [PMID: 30964958 PMCID: PMC6785366 DOI: 10.1111/nyas.14051] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 12/11/2022]
Abstract
Aberrant RNA structure and function operate in neurological disease progression and severity. As RNA contributes to disease pathology in a complex fashion, that is, via various mechanisms, it has become an attractive therapeutic target for small molecules and oligonucleotides. In this review, we discuss the identification of RNA structures that cause or contribute to neurological diseases as well as recent progress toward the development of small molecules that target them, including small molecule modulators of pre-mRNA splicing and RNA repeat expansions that cause microsatellite disorders such as Huntington's disease and amyotrophic lateral sclerosis. The use of oligonucleotide-based modalities is also discussed. There are key differences between small molecule and oligonucleotide targeting of RNA. The former targets RNA structure, while the latter prefers unstructured regions. Thus, some targets will be preferentially targeted by oligonucleotides and others by small molecules.
Collapse
Affiliation(s)
| | - Jonathan L Chen
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida
| | - Matthew D Disney
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida
| |
Collapse
|
6
|
Chen JL, Zhang P, Abe M, Aikawa H, Zhang L, Frank AJ, Zembryski T, Hubbs C, Park H, Withka J, Steppan C, Rogers L, Cabral S, Pettersson M, Wager TT, Fountain MA, Rumbaugh G, Childs-Disney JL, Disney MD. Design, Optimization, and Study of Small Molecules That Target Tau Pre-mRNA and Affect Splicing. J Am Chem Soc 2020; 142:8706-8727. [PMID: 32364710 PMCID: PMC7357857 DOI: 10.1021/jacs.0c00768] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Approximately 95% of human genes are alternatively spliced, and aberrant splicing events can cause disease. One pre-mRNA that is alternatively spliced and linked to neurodegenerative diseases is tau (microtubule-associated protein tau), which can cause frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) and can contribute to Alzheimer's disease. Here, we describe the design of structure-specific lead small molecules that directly target tau pre-mRNA from sequence. This was followed by hit expansion and analogue synthesis to further improve upon these initial lead molecules. The emergent compounds were assessed for functional activity in a battery of assays, including binding assays and an assay that mimics molecular recognition of tau pre-mRNA by a U1 small nuclear ribonucleoprotein (snRNP) splicing factor. Compounds that emerged from these studies had enhanced potency and selectivity for the target RNA relative to the initial hits, while also having significantly improved drug-like properties. The compounds are shown to directly target tau pre-mRNA in cells, via chemical cross-linking and isolation by pull-down target profiling, and to rescue disease-relevant splicing of tau pre-mRNA in a variety of cellular systems, including primary neurons. More broadly, this study shows that lead, structure-specific compounds can be designed from sequence and then further optimized for their physicochemical properties while at the same time enhancing their activity.
Collapse
Affiliation(s)
- Jonathan L. Chen
- Department of Chemistry and Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Peiyuan Zhang
- Department of Chemistry and Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Masahito Abe
- Department of Chemistry and Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Haruo Aikawa
- Department of Chemistry and Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Liying Zhang
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, United States
| | - Alexander J. Frank
- Department of Chemistry & Biochemistry, State University of New York at Fredonia, Fredonia, New York 14063, United States
| | - Timothy Zembryski
- Department of Chemistry & Biochemistry, State University of New York at Fredonia, Fredonia, New York 14063, United States
| | - Christopher Hubbs
- Department of Chemistry and Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - HaJeung Park
- Department of Chemistry and Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Jane Withka
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, United States
| | - Claire Steppan
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Lucy Rogers
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Shawn Cabral
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Martin Pettersson
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, United States
| | - Travis T. Wager
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, United States
| | - Matthew A. Fountain
- Department of Chemistry & Biochemistry, State University of New York at Fredonia, Fredonia, New York 14063, United States
| | - Gavin Rumbaugh
- Department of Chemistry and Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Jessica L. Childs-Disney
- Department of Chemistry and Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Matthew D. Disney
- Department of Chemistry and Neuroscience, The Scripps Research Institute, Jupiter
| |
Collapse
|
7
|
Charak S, Shandilya M, Mehrotra R. RNA targeting by an anthracycline drug: spectroscopic and in silico evaluation of epirubicin interaction with tRNA. J Biomol Struct Dyn 2019; 38:1761-1771. [PMID: 31084352 DOI: 10.1080/07391102.2019.1617786] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Anthracyclines are putative anticancer agents used to treat a wide range of cancers. Among these anthracyclines, epirubicin is derived from the doxorubicin by the subtle difference in the orientation of C4-hydroxyl group at sugar molecule. Epirubicin has great significance as it has propitious anticancer potential with lesser cardiotoxicity and faster elimination from the body. The present study is done to understand the molecular aspect of epirubicin binding to tRNA. We have used various spectroscopic techniques like Fourier transform infrared spectroscopy (FTIR), absorption spectroscopy and circular dichroism to illustrate the binding sites, the extent of binding and conformational changes associated with tRNA after interacting with epirubicin. From infrared studies, we infer that epirubicin interacts with guanine and uracil bases of tRNA. Results obtained from infrared and CD studies suggest that epirubicin complexation with tRNA does not result in any conformational change in tRNA structure. Binding constant (2.1 × 103 M-1) calculated from the absorbance data illustrates that epirubicin has a weak interaction with tRNA molecule. These spectroscopic results like the binding site of epirubicin and binding energy of epirubicin-tRNA complex were also verified by the molecular docking. Results of the present study provide information that aids in the development of efficient RNA targeted drugs from the existing drugs by certain chemical modification in their structure resulting in lesser side effects and better efficacy.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sonika Charak
- Physico Mechanical Metrology Division, CSIR-National Physical Laboratory, New Delhi, India
| | - Manish Shandilya
- Amity School of Applied Sciences, Amity University Haryana, Gurgaon, India
| | - Ranjana Mehrotra
- Physico Mechanical Metrology Division, CSIR-National Physical Laboratory, New Delhi, India
| |
Collapse
|
8
|
Tan J, Yang L, Ong AAL, Shi J, Zhong Z, Lye ML, Liu S, Lisowiec-Wachnicka J, Kierzek R, Roca X, Chen G. A Disease-Causing Intronic Point Mutation C19G Alters Tau Exon 10 Splicing via RNA Secondary Structure Rearrangement. Biochemistry 2019; 58:1565-1578. [DOI: 10.1021/acs.biochem.9b00001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Jiazi Tan
- Division of Chemistry & Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 637371 Singapore
| | - Lixia Yang
- Division of Chemistry & Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 637371 Singapore
| | - Alan Ann Lerk Ong
- Division of Chemistry & Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 637371 Singapore
| | - Jiahao Shi
- Division of Chemistry & Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 637371 Singapore
| | - Zhensheng Zhong
- Division of Chemistry & Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 637371 Singapore
| | - Mun Leng Lye
- Division of Chemistry & Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 637371 Singapore
| | - Shiyi Liu
- Division of Chemistry & Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 637371 Singapore
| | - Jolanta Lisowiec-Wachnicka
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Ryszard Kierzek
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Xavier Roca
- School of Biological Sciences, Nanyang Technological University, 637551 Singapore
| | - Gang Chen
- Division of Chemistry & Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 637371 Singapore
| |
Collapse
|
9
|
Wales DJ, Disney MD, Yildirim I. Computational Investigation of RNA A-Bulges Related to the Microtubule-Associated Protein Tau Causing Frontotemporal Dementia and Parkinsonism. J Phys Chem B 2019; 123:57-65. [PMID: 30517788 PMCID: PMC6465094 DOI: 10.1021/acs.jpcb.8b09139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mutations in the human tau gene result in alternative splicing of the tau protein, which causes frontotemporal dementia and Parkinsonism. One disease mechanism is linked to the stability of a hairpin within the microtubule-associated protein tau (MAPT) mRNA, which contains an A-bulge. Here we employ computational methods to investigate the structural and thermodynamic properties of several A-bulge RNAs with different closing base-pairs. We find that the current amber RNA force field has a preference to overstabilize base-triple over stacked states, even though some of the A-bulges are known to prefer stacked states according to NMR studies. We further determined that if the neighboring base-pairs of A-bulges are AU, this situation can lead to base slippage. However, when the 3'-side of the A-bulge has an UA base-pair, the stacked state is stabilized by an extra interaction that is not observed in the other sequences. We suggest that these A-bulge RNA systems could be used as benchmarks to improve the current RNA force fields.
Collapse
Affiliation(s)
- David J. Wales
- Department of Chemistry, University of Cambridge, Cambridge, Cambridgeshire CB2 1EW, U.K
| | - Matthew D. Disney
- Department of Chemistry, Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Ilyas Yildirim
- Department of Chemistry, Scripps Research Institute, Jupiter, Florida 33458, United States
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, Florida 33458, United States
| |
Collapse
|
10
|
Feng C, Chan D, Joseph J, Muuronen M, Coldren WH, Dai N, Corrêa IR, Furche F, Hadad CM, Spitale RC. Light-activated chemical probing of nucleobase solvent accessibility inside cells. Nat Chem Biol 2018; 14:276-283. [PMID: 29334380 PMCID: PMC6203945 DOI: 10.1038/nchembio.2548] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 11/15/2017] [Indexed: 12/30/2022]
Abstract
The discovery of functional RNAs that are critical for normal and disease physiology continues to expand at a breakneck pace. Many RNA functions are controlled by the formation of specific structures, and an understanding of each structural component is necessary to elucidate its function. Measuring solvent accessibility intracellularly with experimental ease is an unmet need in the field. Here, we present a novel method for probing nucleobase solvent accessibility, Light Activated Structural Examination of RNA (LASER). LASER depends on light activation of a small molecule, nicotinoyl azide (NAz), to measure solvent accessibility of purine nucleobases. In vitro, this technique accurately monitors solvent accessibility and identifies rapid structural changes resulting from ligand binding in a metabolite-responsive RNA. LASER probing can further identify cellular RNA-protein interactions and unique intracellular RNA structures. Our photoactivation technique provides an adaptable framework to structurally characterize solvent accessibility of RNA in many environments.
Collapse
Affiliation(s)
- Chao Feng
- Department of Pharmaceutical Sciences, University of California, Irvine. Irvine, California 92697
| | - Dalen Chan
- Department of Pharmaceutical Sciences, University of California, Irvine. Irvine, California 92697
| | - Jojo Joseph
- Department of Chemistry and Biochemistry, The Ohio State University, 100 West 18 Avenue, Columbus Ohio 43210
| | - Mikko Muuronen
- Department of Chemistry, University of California, Irvine. Irvine, California 92697
| | - William H. Coldren
- Department of Chemistry and Biochemistry, The Ohio State University, 100 West 18 Avenue, Columbus Ohio 43210
| | - Nan Dai
- New England Biolabs, 240 County Road, Ipswich, Massachusetts 01938, USA
| | - Ivan R. Corrêa
- New England Biolabs, 240 County Road, Ipswich, Massachusetts 01938, USA
| | - Filipp Furche
- Department of Chemistry, University of California, Irvine. Irvine, California 92697
| | - Christopher M. Hadad
- Department of Chemistry and Biochemistry, The Ohio State University, 100 West 18 Avenue, Columbus Ohio 43210
| | - Robert C. Spitale
- Department of Pharmaceutical Sciences, University of California, Irvine. Irvine, California 92697
- Department of Chemistry, University of California, Irvine. Irvine, California 92697
| |
Collapse
|
11
|
Kang HJ, Cui Y, Yin H, Scheid A, Hendricks WPD, Schmidt J, Sekulic A, Kong D, Trent JM, Gokhale V, Mao H, Hurley LH. A Pharmacological Chaperone Molecule Induces Cancer Cell Death by Restoring Tertiary DNA Structures in Mutant hTERT Promoters. J Am Chem Soc 2016; 138:13673-13692. [PMID: 27643954 DOI: 10.1021/jacs.6b07598] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Activation of human telomerase reverse transcriptase (hTERT) is necessary for limitless replication in tumorigenesis. Whereas hTERT is transcriptionally silenced in normal cells, most tumor cells reactivate hTERT expression by alleviating transcriptional repression through diverse genetic and epigenetic mechanisms. Transcription-activating hTERT promoter mutations have been found to occur at high frequencies in multiple cancer types. These mutations have been shown to form new transcription factor binding sites that drive hTERT expression, but this model cannot fully account for differences in wild-type (WT) and mutant promoter activation and has not yet enabled a selective therapeutic strategy. Here, we demonstrate a novel mechanism by which promoter mutations activate hTERT transcription, which also sheds light on a unique therapeutic opportunity. Promoter mutations occur in a core promoter region that forms tertiary structures consisting of a pair of G-quadruplexes involved in transcriptional silencing. We show that promoter mutations exert a detrimental effect on the folding of one of these G-quadruplexes, resulting in a nonfunctional silencer element that alleviates transcriptional repression. We have also identified a small drug-like pharmacological chaperone (pharmacoperone) molecule, GTC365, that acts at an early step in the G-quadruplex folding pathway to redirect mutant promoter G-quadruplex misfolding, partially reinstate the correct folding pathway, and reduce hTERT activity through transcriptional repression. This transcription-mediated repression produces cancer cell death through multiple routes including both induction of apoptosis through inhibition of hTERT's role in regulating apoptosis-related proteins and induction of senescence by decreasing telomerase activity and telomere length. We demonstrate the selective therapeutic potential of this strategy in melanoma cells that overexpress hTERT.
Collapse
Affiliation(s)
- Hyun-Jin Kang
- University of Arizona , College of Pharmacy, 1703 East Mabel Street, Tucson, Arizona 85721, United States
| | - Yunxi Cui
- Department of Chemistry and Biochemistry and School of Biomedical Sciences, Kent State University , Kent, Ohio 44242, United States
| | - Holly Yin
- Translational Genomics Research Institute , 445 North Fifth Street, Phoenix, Arizona 85004, United States
| | - Amy Scheid
- College of Science, University of Arizona , 1040 East Fourth Street, Tucson, Arizona 85721, United States
| | - William P D Hendricks
- Translational Genomics Research Institute , 445 North Fifth Street, Phoenix, Arizona 85004, United States
| | - Jessica Schmidt
- Department of Dermatology, Mayo Clinic , 13400 East Shea Boulevard, Scottsdale, Arizona 85259, United States
| | - Aleksandar Sekulic
- Department of Dermatology, Mayo Clinic , 13400 East Shea Boulevard, Scottsdale, Arizona 85259, United States
| | - Deming Kong
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University , Tianjin 300071, People's Republic of China
| | - Jeffrey M Trent
- Translational Genomics Research Institute , 445 North Fifth Street, Phoenix, Arizona 85004, United States
| | - Vijay Gokhale
- BIO5 Institute , 1657 East Helen Street, Tucson, Arizona 85721, United States
| | - Hanbin Mao
- Department of Chemistry and Biochemistry and School of Biomedical Sciences, Kent State University , Kent, Ohio 44242, United States
| | - Laurence H Hurley
- University of Arizona , College of Pharmacy, 1703 East Mabel Street, Tucson, Arizona 85721, United States.,BIO5 Institute , 1657 East Helen Street, Tucson, Arizona 85721, United States.,Arizona Cancer Center , 1515 North Campbell Avenue, Tucson, Arizona 85724, United States
| |
Collapse
|
12
|
Wu S, Mao G, Kirsebom LA. Inhibition of Bacterial RNase P RNA by Phenothiazine Derivatives. Biomolecules 2016; 6:biom6030038. [PMID: 27618117 PMCID: PMC5039424 DOI: 10.3390/biom6030038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/24/2016] [Accepted: 08/26/2016] [Indexed: 12/19/2022] Open
Abstract
There is a need to identify novel scaffolds and targets to develop new antibiotics. Methylene blue is a phenothiazine derivative, and it has been shown to possess anti-malarial and anti-trypanosomal activities. Here, we show that different phenothiazine derivatives and pyronine G inhibited the activities of three structurally different bacterial RNase P RNAs (RPRs), including that from Mycobacterium tuberculosis, with Ki values in the lower μM range. Interestingly, three antipsychotic phenothiazines (chlorpromazine, thioridazine, and trifluoperazine), which are known to have antibacterial activities, also inhibited the activity of bacterial RPRs, albeit with higher Ki values than methylene blue. Phenothiazines also affected lead(II)-induced cleavage of bacterial RPR and inhibited yeast tRNA(Phe), indicating binding of these drugs to functionally important regions. Collectively, our findings provide the first experimental data showing that long, noncoding RNAs could be targeted by different phenothiazine derivatives.
Collapse
Affiliation(s)
- Shiying Wu
- Department of Cell and Molecular Biology, Box 596, Biomedical Centre, Uppsala SE-751 24, Sweden.
| | - Guanzhong Mao
- Department of Cell and Molecular Biology, Box 596, Biomedical Centre, Uppsala SE-751 24, Sweden.
| | - Leif A Kirsebom
- Department of Cell and Molecular Biology, Box 596, Biomedical Centre, Uppsala SE-751 24, Sweden.
| |
Collapse
|
13
|
Lisowiec J, Magner D, Kierzek E, Lenartowicz E, Kierzek R. Structural determinants for alternative splicing regulation of the MAPT pre-mRNA. RNA Biol 2015; 12:330-42. [PMID: 25826665 DOI: 10.1080/15476286.2015.1017214] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Alternative splicing at the MAPT gene exon 10 yields similar levels of the 3R and 4R tau protein isoforms. (1) The presence of mutations, particularly in exon 10 and intron 10-11, changes the quantity of tau isoforms. Domination each of the isoform yields tau protein aggregation and frontotemporal dementia and Parkinsonism linked to chromosome 17 (FTDP-17). Here, we report for the first time the secondary structure of the 194/195 nucleotide region for the wild type (WT) and 10 mutants of the MAPT gene pre-mRNA determined using both chemical and microarray mapping. Thermodynamic analyses indicate that single nucleotide mutations in the splicing regulatory element (SRE) that form a hairpin affect its stability by up to 4 and 7 kcal/mol. Moreover, binding the regulatory hairpin of small molecule ligands (neomycin, kanamycin, tobramycin and mitoxantrone) enhance its stability depending on the nature of the ligands and the RNA mutations. Experiments using the cos-7 cell line indicate that the presence of ligands and modified antisense oligonucleotides affect the quantity of 3R and 4R isoforms. This finding correlates with the thermodynamic stability of the regulatory hairpin. An alternative splicing regulation mechanism for exon 10 is postulated based on our experimental data and on published data.
Collapse
Key Words
- AD, Alzheimer disease
- DMS, dimethyl sulfide
- ESE, exonic splicing enhancer
- ESS, exonic splicing silencer
- FTD, frontotemporal dementia
- FTDP-17, frontotemporal dementia and Parkinsonism linked to chromosome 17
- ISM, intronic splicing modulator
- ISS, intronic splicing silencer
- MAPT, microtubule-associated protein tau
- NMIA, N-methylisotoic anhydride
- NMR, nuclear magnetic resonance
- PPE, polypurine enhancer
- RNA structure
- RNA thermodynamics
- RT-PCR, reverse transcription polymerase chain reaction
- SHAPE, selective 2′-hydroxyl acylation analyzed by primer extension
- SMA, spinal muscular atrophy
- SRE, splicing regulatory element
- U1 snRNP, U1 small nuclear ribonucleoprotein
- WT, wild type
- alternative splicing regulation
- antisense oligonucleotides
- neurodegradation
- pre-mRNA, pre-messenger RNA
- small molecule binding
Collapse
Affiliation(s)
- Jolanta Lisowiec
- a Institute of Bioorganic Chemistry; Polish Academy of Sciences ; Noskowskiego, Poland
| | | | | | | | | |
Collapse
|
14
|
Nahar S, Ranjan N, Ray A, Arya DP, Maiti S. Potent inhibition of miR-27a by neomycin-bisbenzimidazole conjugates. Chem Sci 2015; 6:5837-5846. [PMID: 29861909 PMCID: PMC5947510 DOI: 10.1039/c5sc01969a] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 07/07/2015] [Indexed: 12/27/2022] Open
Abstract
miRNAs are important components of regulatory networks that control gene expression and have implications in various diseases including cancer. Targeting oncogenic miRNAs with small molecules is currently being explored to develop cancer therapeutics. Here, we report the development of dual binding neomycin-bisbenzimidazole conjugates that target oncogenic miR-27a with high affinity (Ka = 1.2 to 7.4 × 108 M-1). These conjugates bring significant reduction (∼65% at 5 μM) in mature miRNA levels and penetrate easily in the cells where they localise both in the cytoplasm and the nucleus. Cell cycle analysis showed significant increase in the G0/G1 phase (∼15%) and decrease in the S phase (∼7%) upon treatment with neomycin-bisbenzimidazole conjugates, suggesting inhibition of cell proliferation. Using the conjugation approach, we show that moderately binding ligands can be covalently combined into high affinity binders. This study also highlights the role of linker optimization in designing high affinity ligands for miR-27a targeting.
Collapse
Affiliation(s)
- Smita Nahar
- Academy of Scientific and Innovative Research (AcSIR) , Anusandhan Bhawan, 2 Rafi Marg , New Delhi-110001 , India
- CSIR-Institute of Genomics and Integrative Biology , Mathura Road , Delhi-110020 , India . ; ; Tel: +91-11-2766-6156
| | - Nihar Ranjan
- Department of Chemistry , Clemson University , Clemson , SC 29634 , USA
| | - Arjun Ray
- Academy of Scientific and Innovative Research (AcSIR) , Anusandhan Bhawan, 2 Rafi Marg , New Delhi-110001 , India
- CSIR-Institute of Genomics and Integrative Biology , Mathura Road , Delhi-110020 , India . ; ; Tel: +91-11-2766-6156
| | - Dev P Arya
- Department of Chemistry , Clemson University , Clemson , SC 29634 , USA
| | - Souvik Maiti
- Academy of Scientific and Innovative Research (AcSIR) , Anusandhan Bhawan, 2 Rafi Marg , New Delhi-110001 , India
- CSIR-Institute of Genomics and Integrative Biology , Mathura Road , Delhi-110020 , India . ; ; Tel: +91-11-2766-6156
- CSIR-National Chemical Laboratory , Dr. Homi Bhabha Road , Pune , 411008 , India . ;
| |
Collapse
|
15
|
McCarthy A, Lonergan R, Olszewska DA, O'Dowd S, Cummins G, Magennis B, Fallon EM, Pender N, Huey ED, Cosentino S, O'Rourke K, Kelly BD, O'Connell M, Delon I, Farrell M, Spillantini MG, Rowland LP, Fahn S, Craig P, Hutton M, Lynch T. Closing the tau loop: the missing tau mutation. Brain 2015; 138:3100-9. [PMID: 26297556 DOI: 10.1093/brain/awv234] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 06/27/2015] [Indexed: 12/30/2022] Open
Abstract
Frontotemporal lobar degeneration comprises a group of disorders characterized by behavioural, executive, language impairment and sometimes features of parkinsonism and motor neuron disease. In 1994 we described an Irish-American family with frontotemporal dementia linked to chromosome 17 associated with extensive tau pathology. We named this disinhibition-dementia-parkinsonism-amyotrophy complex. We subsequently identified mutations in the MAPT gene. Eleven MAPT gene splice site stem loop mutations were identified over time except for 5' splice site of exon 10. We recently identified another Irish family with autosomal dominant early amnesia and behavioural change or parkinsonism associated with the 'missing' +15 mutation at the intronic boundary of exon 10. We performed a clinical, neuropsychological and neuroimaging study on the proband and four siblings, including two affected siblings. We sequenced MAPT and performed segregation analysis. We looked for a biological effect of the tau variant by performing real-time polymerase chain reaction analysis of RNA extracted from human embryonic kidney cells transfected with exon trapping constructs. We found a c.915+15A>C exon 10/intron 10 stem loop mutation in all affected subjects but not in the unaffected. The c.915+15A>C variant caused a shift in tau splicing pattern to a predominantly exon 10+ pattern presumably resulting in predominant 4 repeat tau and little 3 repeat tau. This strongly suggests that the c.915+15A>C variant is a mutation and that it causes frontotemporal dementia linked to chromosome 17 in this pedigree by shifting tau transcription and translation to +4 repeat tau. Tau (MAPT) screening should be considered in families where amnesia or atypical parkinsonism coexists with behavioural disturbance early in the disease process. We describe the final missing stem loop tau mutation predicted 15 years ago. Mutations have now been identified at all predicted sites within the 'stem' when the stem-loop model was first proposed and no mutations have been found within the 'loop' region as expected. Therefore we 'close the tau loop' having 'opened the loop' 21 years ago.
Collapse
Affiliation(s)
- Allan McCarthy
- 1 The Dublin Neurological Institute at the Mater Misericordiae University Hospital, 57 Eccles Street, Dublin 7, Ireland
| | - Roisin Lonergan
- 1 The Dublin Neurological Institute at the Mater Misericordiae University Hospital, 57 Eccles Street, Dublin 7, Ireland
| | - Diana A Olszewska
- 1 The Dublin Neurological Institute at the Mater Misericordiae University Hospital, 57 Eccles Street, Dublin 7, Ireland
| | - Sean O'Dowd
- 1 The Dublin Neurological Institute at the Mater Misericordiae University Hospital, 57 Eccles Street, Dublin 7, Ireland
| | - Gemma Cummins
- 2 Department of Clinical Neuroscience, Centre for Brain Repair, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK
| | - Brian Magennis
- 1 The Dublin Neurological Institute at the Mater Misericordiae University Hospital, 57 Eccles Street, Dublin 7, Ireland
| | - Emer M Fallon
- 1 The Dublin Neurological Institute at the Mater Misericordiae University Hospital, 57 Eccles Street, Dublin 7, Ireland
| | - Niall Pender
- 3 Department of Psychology, Beaumont Hospital, Beaumont Rd, Dublin 9, Ireland, Department of Psychology, Royal College of Surgeons in Ireland
| | - Edward D Huey
- 4 Departments of Psychiatry and Neurology, College of Physicians and Surgeons, University Medical Centre, 630 West 168th Street, New York, NY 10032, USA
| | - Stephanie Cosentino
- 5 Cognitive Neuroscience Section, Department of Neurology, Columbia University Medical Centre, 630 West 168th Street, New York, NY 10032, USA
| | - Killian O'Rourke
- 1 The Dublin Neurological Institute at the Mater Misericordiae University Hospital, 57 Eccles Street, Dublin 7, Ireland
| | - Brendan D Kelly
- 6 Department of Psychiatry, Mater Misericordiae University Hospital, 63 Eccles Street, Dublin 7, Ireland
| | - Martin O'Connell
- 7 Department of Radiology, Mater Misericordiae University Hospital, North Circular Road, Dublin 7, Ireland
| | - Isabelle Delon
- 8 Medical Genetics Service, Cambridge University Hospital NHS Foundation Trust, Addenbrooke's Treatment Centre, Hills Road, Cambridge, CB2 0QQ, UK
| | - Michael Farrell
- 9 Department of Neuropathology, Beaumont Hospital, Beaumont Road, Dublin, Ireland
| | - Maria Grazia Spillantini
- 10 Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge, CB2 0AH, UK
| | - Lewis P Rowland
- 11 The Neurological Institute, Columbia University, 710 West 168th Street, New York, NY 10032-3784, USA
| | - Stanley Fahn
- 11 The Neurological Institute, Columbia University, 710 West 168th Street, New York, NY 10032-3784, USA
| | - Peter Craig
- 12 Eli Lilly, Erl Wood Manor, Windlesham, Surrey, GU20 6PH, UK
| | - Michael Hutton
- 12 Eli Lilly, Erl Wood Manor, Windlesham, Surrey, GU20 6PH, UK
| | - Tim Lynch
- 1 The Dublin Neurological Institute at the Mater Misericordiae University Hospital, 57 Eccles Street, Dublin 7, Ireland
| |
Collapse
|
16
|
Leeder WM, Reuss AJ, Brecht M, Kratz K, Wachtveitl J, Göringer HU. Charge reduction and thermodynamic stabilization of substrate RNAs inhibit RNA editing. PLoS One 2015; 10:e0118940. [PMID: 25742417 PMCID: PMC4350841 DOI: 10.1371/journal.pone.0118940] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 01/07/2015] [Indexed: 01/04/2023] Open
Abstract
African trypanosomes cause a parasitic disease known as sleeping sickness. Mitochondrial transcript maturation in these organisms requires a RNA editing reaction that is characterized by the insertion and deletion of U-nucleotides into otherwise non-functional mRNAs. Editing represents an ideal target for a parasite-specific therapeutic intervention since the reaction cycle is absent in the infected host. In addition, editing relies on a macromolecular protein complex, the editosome, that only exists in the parasite. Therefore, all attempts to search for editing interfering compounds have been focused on molecules that bind to proteins of the editing machinery. However, in analogy to other RNA-driven biochemical pathways it should be possible to stall the reaction by targeting its substrate RNAs. Here we demonstrate inhibition of editing by specific aminoglycosides. The molecules bind into the major groove of the gRNA/pre-mRNA editing substrates thereby causing a stabilization of the RNA molecules through charge compensation and an increase in stacking. The data shed light on mechanistic details of the editing process and identify critical parameters for the development of new trypanocidal compounds.
Collapse
Affiliation(s)
- W.-Matthias Leeder
- Molecular Genetics, Darmstadt University of Technology, Darmstadt, Germany
| | - Andreas J. Reuss
- Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Michael Brecht
- Molecular Genetics, Darmstadt University of Technology, Darmstadt, Germany
| | - Katja Kratz
- Molecular Genetics, Darmstadt University of Technology, Darmstadt, Germany
| | - Josef Wachtveitl
- Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - H. Ulrich Göringer
- Molecular Genetics, Darmstadt University of Technology, Darmstadt, Germany
- * E-mail:
| |
Collapse
|
17
|
Artigas G, López-Senín P, González C, Escaja N, Marchán V. Ametantrone-based compounds as potential regulators of Tau pre-mRNA alternative splicing. Org Biomol Chem 2014; 13:452-64. [PMID: 25372055 DOI: 10.1039/c4ob01925c] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Tau pre-mRNA contains a stem-loop structure involved in the regulation of the alternative splicing of tau protein. We describe here a new family of Tau RNA ligands selected by dynamic combinatorial chemistry based on the combination of ametantrone with small RNA-binding molecules. The most promising compound results from derivatization of one of the side chains of the anthraquinone ring with the small aminoglycoside neamine through a short spacer. This compound binds the RNA target with a high affinity in a preferred binding site, in which the heteroaromatic moiety intercalates in the bulged region of the stem-loop and its side chains and neamine interact with the major groove of the RNA. Importantly, binding of this compound to mutated RNA sequences involved in the onset of some tauopathies such as FTDP-17 restores their thermodynamic stability to a similar or even higher levels than that of the wild-type sequence, thereby revealing its potential as a modulator of Tau pre-mRNA splicing.
Collapse
Affiliation(s)
- Gerard Artigas
- Departament de Química Orgànica and IBUB, Universitat de Barcelona, Martí i Franquès 1-11, E-08028 Barcelona, Spain.
| | | | | | | | | |
Collapse
|
18
|
Luo Y, Disney MD. Bottom-up design of small molecules that stimulate exon 10 skipping in mutant MAPT pre-mRNA. Chembiochem 2014; 15:2041-4. [PMID: 25115866 DOI: 10.1002/cbic.201402069] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Indexed: 11/07/2022]
Abstract
One challenge in chemical biology is to develop small molecules that control cellular protein content. The amount and identity of proteins are influenced by the RNAs that encode them; thus, protein content in a cell could be affected by targeting mRNA. However, RNA has been traditionally difficult to target with small molecules. In this report, we describe controlling the protein products of the mutated microtubule-associated protein tau (MAPT) mature mRNA with a small molecule. MAPT mutations in exon 10 are associated with inherited frontotemporal dementia and Parkinsonism linked to chromosome 17 (FTDP-17), an incurable disease that is directly caused by increased inclusion of exon 10 in MAPT mRNA. Recent studies have shown that mutations within a hairpin at the MAPT exon 10-intron junction decrease the thermodynamic stability of the RNA, increasing binding to U1 snRNP and thus exon 10 inclusion. Therefore, we designed small molecules that bind and stabilize a mutant MAPT by using Inforna, a computational approach based on information about RNA-small-molecule interactions. The optimal compound selectively bound the mutant MAPT hairpin and thermodynamically stabilized its folding, facilitating exon 10 exclusion.
Collapse
Affiliation(s)
- Yiling Luo
- Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, 3A1, Jupiter, FL 33458 (USA)
| | | |
Collapse
|
19
|
Wolfe MS. Targeting mRNA for Alzheimer's and related dementias. SCIENTIFICA 2014; 2014:757549. [PMID: 24876993 PMCID: PMC4020195 DOI: 10.1155/2014/757549] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 03/20/2014] [Indexed: 06/03/2023]
Abstract
Brain deposition of the amyloid beta-protein (A β ) and tau are characteristic features in Alzheimer's disease (AD). Mutations in the A β precursor protein (APP) and a protease involved in A β production from APP strongly argue for a pathogenic role of A β in AD, while mutations in tau are associated with related disorders collectively called frontotemporal lobar degeneration (FTLD). Despite intense effort, therapeutic strategies that target A β or tau have not yet yielded medications, suggesting that alternative approaches should be pursued. In recent years, our laboratory has studied the role of mRNA in AD and FTLD, specifically those encoding tau and the A β -producing protease BACE1. As many FTLD-causing tau mutations destabilize a hairpin structure that regulates RNA splicing, we have targeted this structure with small molecules, antisense oligonucleotides, and small molecule-antisense conjugates. We have also discovered that microRNA interaction with the 3'-untranslated region of tau regulates tau expression. Regarding BACE1, we found that alternative splicing leads to inactive splice isoforms and antisense oligonucleotides shift splicing toward these inactive isoforms to decrease A β production. In addition, a G-quadruplex structure in the BACE1 mRNA plays a role in splice regulation. The prospects for targeting tau and BACE1 mRNAs as therapeutic strategies will be discussed.
Collapse
Affiliation(s)
- Michael S. Wolfe
- Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, H.I.M. 754, Boston, MA 02115, USA
| |
Collapse
|
20
|
Artigas G, Marchán V. Synthesis of Janus compounds for the recognition of G-U mismatched nucleobase pairs. J Org Chem 2013; 78:10666-77. [PMID: 24087986 DOI: 10.1021/jo401684j] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The design and synthesis of two Janus-type heterocycles with the capacity to simultaneously recognize guanine and uracyl in G-U mismatched pairs through complementary hydrogen bond pairing is described. Both compounds were conveniently functionalized with a carboxylic function and efficiently attached to a tripeptide sequence by using solid-phase methodologies. Ligands based on the derivatization of such Janus compounds with a small aminoglycoside, neamine, and its guanidinylated analogue have been synthesized, and their interaction with Tau RNA has been investigated by using several biophysical techniques, including UV-monitored melting curves, fluorescence titration experiments, and (1)H NMR. The overall results indicated that Janus-neamine/guanidinoneamine showed some preference for the +3 mutated RNA sequence associated with the development of some tauopathies, although preliminary NMR studies have not confirmed binding to G-U pairs. Moreover, a good correlation has been found between the RNA binding affinity of such Janus-containing ligands and their ability to stabilize this secondary structure upon complexation.
Collapse
Affiliation(s)
- Gerard Artigas
- Departament de Química Orgànica, Facultat de Química, Universitat de Barcelona Martí i Franquès 1-11, E-08028 Barcelona, Spain
| | | |
Collapse
|
21
|
Maeda Y, Iwata R, Wada T. Synthesis and properties of cationic oligopeptides with different side chain lengths that bind to RNA duplexes. Bioorg Med Chem 2013; 21:1717-23. [DOI: 10.1016/j.bmc.2013.01.053] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 01/22/2013] [Accepted: 01/23/2013] [Indexed: 10/27/2022]
|
22
|
Shen M, Bellaousov S, Hiller M, de La Grange P, Creamer TP, Malina O, Sperling R, Mathews DH, Stoilov P, Stamm S. Pyrvinium pamoate changes alternative splicing of the serotonin receptor 2C by influencing its RNA structure. Nucleic Acids Res 2013; 41:3819-32. [PMID: 23393189 PMCID: PMC3616728 DOI: 10.1093/nar/gkt063] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 01/08/2013] [Accepted: 01/15/2013] [Indexed: 01/17/2023] Open
Abstract
The serotonin receptor 2C plays a central role in mood and appetite control. It undergoes pre-mRNA editing as well as alternative splicing. The RNA editing suggests that the pre-mRNA forms a stable secondary structure in vivo. To identify substances that promote alternative exons inclusion, we set up a high-throughput screen and identified pyrvinium pamoate as a drug-promoting exon inclusion without editing. Circular dichroism spectroscopy indicates that pyrvinium pamoate binds directly to the pre-mRNA and changes its structure. SHAPE (selective 2'-hydroxyl acylation analysed by primer extension) assays show that part of the regulated 5'-splice site forms intramolecular base pairs that are removed by this structural change, which likely allows splice site recognition and exon inclusion. Genome-wide analyses show that pyrvinium pamoate regulates >300 alternative exons that form secondary structures enriched in A-U base pairs. Our data demonstrate that alternative splicing of structured pre-mRNAs can be regulated by small molecules that directly bind to the RNA, which is reminiscent to an RNA riboswitch.
Collapse
MESH Headings
- Alternative Splicing/drug effects
- Base Sequence
- Exons
- HEK293 Cells
- High-Throughput Screening Assays
- Humans
- Molecular Sequence Data
- Nucleic Acid Conformation
- Phylogeny
- Pyrvinium Compounds/metabolism
- Pyrvinium Compounds/pharmacology
- RNA Editing
- RNA Precursors/metabolism
- RNA, Double-Stranded/chemistry
- RNA, Double-Stranded/drug effects
- RNA, Messenger/chemistry
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Receptor, Serotonin, 5-HT2C/genetics
- Receptor, Serotonin, 5-HT2C/metabolism
- Ribonucleoprotein, U1 Small Nuclear/metabolism
- Spliceosomes/metabolism
Collapse
Affiliation(s)
- Manli Shen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, 741 South Limestone, Lexington, KY 40536, USA, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany and Max Planck Institute for the Physics of Complex Systems, 01187 Dresden, Germany GenoSplice technology, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, 75010 Paris, France, Department of Biochemistry and Biophysics,University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA, Department of Biochemistry, West Virginia University, Morgantown, P.O. Box 9142, WV 26506, USA, Department of Genetics, The Hebrew University of Jerusalem, Jerusalem 91904, Israel and Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Stanislav Bellaousov
- Department of Molecular and Cellular Biochemistry, University of Kentucky, 741 South Limestone, Lexington, KY 40536, USA, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany and Max Planck Institute for the Physics of Complex Systems, 01187 Dresden, Germany GenoSplice technology, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, 75010 Paris, France, Department of Biochemistry and Biophysics,University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA, Department of Biochemistry, West Virginia University, Morgantown, P.O. Box 9142, WV 26506, USA, Department of Genetics, The Hebrew University of Jerusalem, Jerusalem 91904, Israel and Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Michael Hiller
- Department of Molecular and Cellular Biochemistry, University of Kentucky, 741 South Limestone, Lexington, KY 40536, USA, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany and Max Planck Institute for the Physics of Complex Systems, 01187 Dresden, Germany GenoSplice technology, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, 75010 Paris, France, Department of Biochemistry and Biophysics,University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA, Department of Biochemistry, West Virginia University, Morgantown, P.O. Box 9142, WV 26506, USA, Department of Genetics, The Hebrew University of Jerusalem, Jerusalem 91904, Israel and Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Pierre de La Grange
- Department of Molecular and Cellular Biochemistry, University of Kentucky, 741 South Limestone, Lexington, KY 40536, USA, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany and Max Planck Institute for the Physics of Complex Systems, 01187 Dresden, Germany GenoSplice technology, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, 75010 Paris, France, Department of Biochemistry and Biophysics,University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA, Department of Biochemistry, West Virginia University, Morgantown, P.O. Box 9142, WV 26506, USA, Department of Genetics, The Hebrew University of Jerusalem, Jerusalem 91904, Israel and Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Trevor P. Creamer
- Department of Molecular and Cellular Biochemistry, University of Kentucky, 741 South Limestone, Lexington, KY 40536, USA, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany and Max Planck Institute for the Physics of Complex Systems, 01187 Dresden, Germany GenoSplice technology, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, 75010 Paris, France, Department of Biochemistry and Biophysics,University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA, Department of Biochemistry, West Virginia University, Morgantown, P.O. Box 9142, WV 26506, USA, Department of Genetics, The Hebrew University of Jerusalem, Jerusalem 91904, Israel and Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Orit Malina
- Department of Molecular and Cellular Biochemistry, University of Kentucky, 741 South Limestone, Lexington, KY 40536, USA, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany and Max Planck Institute for the Physics of Complex Systems, 01187 Dresden, Germany GenoSplice technology, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, 75010 Paris, France, Department of Biochemistry and Biophysics,University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA, Department of Biochemistry, West Virginia University, Morgantown, P.O. Box 9142, WV 26506, USA, Department of Genetics, The Hebrew University of Jerusalem, Jerusalem 91904, Israel and Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Ruth Sperling
- Department of Molecular and Cellular Biochemistry, University of Kentucky, 741 South Limestone, Lexington, KY 40536, USA, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany and Max Planck Institute for the Physics of Complex Systems, 01187 Dresden, Germany GenoSplice technology, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, 75010 Paris, France, Department of Biochemistry and Biophysics,University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA, Department of Biochemistry, West Virginia University, Morgantown, P.O. Box 9142, WV 26506, USA, Department of Genetics, The Hebrew University of Jerusalem, Jerusalem 91904, Israel and Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - David H. Mathews
- Department of Molecular and Cellular Biochemistry, University of Kentucky, 741 South Limestone, Lexington, KY 40536, USA, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany and Max Planck Institute for the Physics of Complex Systems, 01187 Dresden, Germany GenoSplice technology, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, 75010 Paris, France, Department of Biochemistry and Biophysics,University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA, Department of Biochemistry, West Virginia University, Morgantown, P.O. Box 9142, WV 26506, USA, Department of Genetics, The Hebrew University of Jerusalem, Jerusalem 91904, Israel and Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Peter Stoilov
- Department of Molecular and Cellular Biochemistry, University of Kentucky, 741 South Limestone, Lexington, KY 40536, USA, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany and Max Planck Institute for the Physics of Complex Systems, 01187 Dresden, Germany GenoSplice technology, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, 75010 Paris, France, Department of Biochemistry and Biophysics,University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA, Department of Biochemistry, West Virginia University, Morgantown, P.O. Box 9142, WV 26506, USA, Department of Genetics, The Hebrew University of Jerusalem, Jerusalem 91904, Israel and Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Stefan Stamm
- Department of Molecular and Cellular Biochemistry, University of Kentucky, 741 South Limestone, Lexington, KY 40536, USA, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany and Max Planck Institute for the Physics of Complex Systems, 01187 Dresden, Germany GenoSplice technology, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, 75010 Paris, France, Department of Biochemistry and Biophysics,University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA, Department of Biochemistry, West Virginia University, Morgantown, P.O. Box 9142, WV 26506, USA, Department of Genetics, The Hebrew University of Jerusalem, Jerusalem 91904, Israel and Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| |
Collapse
|
23
|
Wolfe MS. The role of tau in neurodegenerative diseases and its potential as a therapeutic target. SCIENTIFICA 2012; 2012:796024. [PMID: 24278740 PMCID: PMC3820460 DOI: 10.6064/2012/796024] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 11/05/2012] [Indexed: 06/01/2023]
Abstract
The abnormal deposition of proteins in and around neurons is a common pathological feature of many neurodegenerative diseases. Among these pathological proteins, the microtubule-associated protein tau forms intraneuronal filaments in a spectrum of neurological disorders. The discovery that dominant mutations in the MAPT gene encoding tau are associated with familial frontotemporal dementia strongly supports abnormal tau protein as directly involved in disease pathogenesis. This and other evidence suggest that tau is a worthwhile target for the prevention or treatment of tau-associated neurodegenerative diseases, collectively called tauopathies. However, it is critical to understand the normal biological roles of tau, the specific molecular events that induce tau to become neurotoxic, the biochemical nature of pathogenic tau, the means by which pathogenic tau exerts neurotoxicity, and how tau pathology propagates. Based on known differences between normal and abnormal tau, a number of approaches have been taken toward the discovery of potential therapeutics. Key questions still remain open, such as the nature of the connection between the amyloid- β protein of Alzheimer's disease and tau pathology. Answers to these questions should help better understand the nature of tauopathies and may also reveal new therapeutic targets and strategies.
Collapse
Affiliation(s)
- Michael S. Wolfe
- Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, H.I.M. 754, Boston, MA 02115, USA
| |
Collapse
|
24
|
Diez-García F, Chakrabartty A, González C, Laurents DV. An Arg-rich putative prebiotic protein is as stable as its Lys-rich variant. Arch Biochem Biophys 2012; 528:118-26. [DOI: 10.1016/j.abb.2012.09.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 09/14/2012] [Accepted: 09/15/2012] [Indexed: 10/27/2022]
|
25
|
López-Senín P, Artigas G, Marchán V. Exploring the effect of aminoglycoside guanidinylation on ligands for Tau exon 10 splicing regulatory element RNA. Org Biomol Chem 2012; 10:9243-54. [DOI: 10.1039/c2ob26623g] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
26
|
Ray P, Kar A, Fushimi K, Havlioglu N, Chen X, Wu JY. PSF suppresses tau exon 10 inclusion by interacting with a stem-loop structure downstream of exon 10. J Mol Neurosci 2011; 45:453-66. [PMID: 21881826 DOI: 10.1007/s12031-011-9634-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 08/17/2011] [Indexed: 01/24/2023]
Abstract
Microtubule binding protein Tau has been implicated in a wide range of neurodegenerative disorders collectively classified as tauopathies. Exon 10 of the human tau gene, which codes for a microtubule binding repeat region, is alternatively spliced to form Tau protein isoforms containing either four or three microtubule binding repeats, Tau4R and Tau3R, respectively. The levels of different Tau splicing isoforms are fine-tuned by alternative splicing with the ratio of Tau4R/Tau3R maintained approximately at one in adult neurons. Mutations that disrupt tau exon 10 splicing regulation cause an imbalance of different tau splicing isoforms and have been associated with tauopathy. To search for factors interacting with tau pre-messenger RNA (pre-mRNA) and regulating tau exon 10 alternative splicing, we performed a yeast RNA-protein interaction screen and identified polypyrimidine tract binding protein associated splicing factor (PSF) as a candidate tau exon 10 splicing regulator. UV crosslinking experiments show that PSF binds to the stem-loop structure at the 5' splice site downstream of tau exon 10. This PSF-interacting RNA element is distinct from known PSF binding sites previously identified in other genes. Overexpression of PSF promotes tau exon 10 exclusion, whereas down-regulation of the endogenous PSF facilitates exon 10 inclusion. Immunostaining shows that PSF is expressed in the human brain regions affected by tauopathy. Our data reveal a new player in tau exon 10 alternative splicing regulation and uncover a previously unknown mechanism of PSF in regulating tau pre-mRNA splicing.
Collapse
Affiliation(s)
- Payal Ray
- Department of Neurology, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | | | | | |
Collapse
|
27
|
RNA helicase p68 (DDX5) regulates tau exon 10 splicing by modulating a stem-loop structure at the 5' splice site. Mol Cell Biol 2011; 31:1812-21. [PMID: 21343338 DOI: 10.1128/mcb.01149-10] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Regulation of tau exon 10 splicing plays an important role in tauopathy. One of the cis elements regulating tau alternative splicing is a stem-loop structure at the 5' splice site of tau exon 10. The RNA helicase(s) modulating this stem-loop structure was unknown. We searched for splicing regulators interacting with this stem-loop region using an RNA affinity pulldown-coupled mass spectrometry approach and identified DDX5/RNA helicase p68 as an activator of tau exon 10 splicing. The activity of p68 in stimulating tau exon 10 inclusion is dependent on RBM4, an intronic splicing activator. RNase H cleavage and U1 protection assays suggest that p68 promotes conformational change of the stem-loop structure, thereby increasing the access of U1snRNP to the 5' splice site of tau exon 10. This study reports the first RNA helicase interacting with a stem-loop structure at the splice site and regulating alternative splicing in a helicase-dependent manner. Our work uncovers a previously unknown function of p68 in regulating tau exon 10 splicing. Furthermore, our experiments reveal functional interaction between two splicing activators for tau exon 10, p68 binding at the stem-loop region and RBM4 interacting with the intronic splicing enhancer region.
Collapse
|
28
|
López-Senín P, Gómez-Pinto I, Grandas A, Marchán V. Identification of ligands for the Tau exon 10 splicing regulatory element RNA by using dynamic combinatorial chemistry. Chemistry 2011; 17:1946-53. [PMID: 21274946 DOI: 10.1002/chem.201002065] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Indexed: 01/25/2023]
Abstract
We describe the use of dynamic combinatorial chemistry (DCC) to identify ligands for the stem-loop structure located at the exon 10-5'-intron junction of Tau pre-mRNA, which is involved in the onset of several tauopathies including frontotemporal dementia with Parkinsonism linked to chromosome 17 (FTDP-17). A series of ligands that combine the small aminoglycoside neamine and heteroaromatic moieties (azaquinolone and two acridines) have been identified by using DCC. These compounds effectively bind the stem-loop RNA target (the concentration required for 50% RNA response (EC(50)): 2-58 μM), as determined by fluorescence titration experiments. Importantly, most of them are able to stabilize both the wild-type and the +3 and +14 mutated sequences associated with the development of FTDP-17 without producing a significant change in the overall structure of the RNA (as analyzed by circular dichroism (CD) spectroscopy), which is a key factor for recognition by the splicing regulatory machinery. A good correlation has been found between the affinity of the ligands for the target and their ability to stabilize the RNA secondary structure.
Collapse
Affiliation(s)
- Paula López-Senín
- Departament de Química Orgànica and IBUB, Universitat de Barcelona, Martí i Franquès 1-11, 08028 Barcelona, Spain
| | | | | | | |
Collapse
|
29
|
López-Alonso JP, Pardo-Cea MA, Gómez-Pinto I, Fernández I, Chakrabartty A, Pedroso E, González C, Laurents DV. Putative one-pot prebiotic polypeptides with ribonucleolytic activity. Chemistry 2010; 16:5314-23. [PMID: 20232309 DOI: 10.1002/chem.200903207] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
KIA7, a peptide with a highly restricted set of amino acids (Lys, Ile, Ala, Gly and Tyr), adopts a specifically folded structure. Some amino acids, including Lys, Ile, Ala, Gly and His, form under the same putative prebiotic conditions, whereas different conditions are needed for producing Tyr, Phe and Trp. Herein, we report the 3D structure and conformational stability of the peptide KIA7H, which is composed of only Lys, Ile, Ala, Gly and His. When the imidazole group is neutral, this 20-mer peptide adopts a four-helix bundle with a specifically packed hydrophobic core. Therefore, one-pot prebiotic proteins with well-defined structures might have arisen early in chemical evolution. The Trp variant, KIA7W, was also studied. It adopts a 3D structure similar to that of KIA7H and its previously studied Tyr and Phe variants, but is remarkably more stable. When tested for ribonucleolytic activity, KIA7H, KIA7W and even short, unstructured peptides rich in His and Lys, in combination with Mg(++), Mn(++) or Ni(++) (but not Cu(++), Zn(++) or EDTA) specifically cleave the single-stranded region in an RNA stem-loop. This suggests that prebiotic peptide-divalent cation complexes with ribonucleolytic activity might have co-inhabited the RNA world.
Collapse
Affiliation(s)
- Jorge P López-Alonso
- Instituto de Química Física Rocasolano C.S.I.C. Serrano 119, 28006, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Stamelou M, de Silva R, Arias-Carrión O, Boura E, Höllerhage M, Oertel WH, Müller U, Höglinger GU. Rational therapeutic approaches to progressive supranuclear palsy. Brain 2010; 133:1578-90. [PMID: 20472654 DOI: 10.1093/brain/awq115] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Progressive supranuclear palsy is a sporadic and progressive neurodegenerative disease, most often presenting as a symmetric, akinetic-rigid syndrome with postural instability, vertical supranuclear gaze palsy and frontal lobe deficits. It belongs to the family of tauopathies and involves both cortical and subcortical structures. Although the exact pathophysiology is not yet fully understood, several lines of evidence point to a crucial contribution from both genetic predisposition and mitochondrial dysfunction. Recently gained insights into the pathophysiology of this disease have led to several hypothesis-driven therapeutic approaches aiming at disease-modification rather than mere symptomatic neurotransmitter-replacement therapy. Agents targeting mitochondrial dysfunction have already shown a positive effect in a phase II study and further studies to verify and expand these results are ongoing. Clinical studies with agents targeting tau dysfunction such as tau-kinase inhibitors, tau-aggregation inhibitors and microtubule stabilizers are in preparation or ongoing. This review presents the current pathophysiological concepts driving these exciting therapeutic developments.
Collapse
Affiliation(s)
- Maria Stamelou
- Department of Neurology, Philipps University, Rudolf-Bultmann Str. 8, D-35033 Marburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Kondo J, Westhof E. Base pairs and pseudo pairs observed in RNA-ligand complexes. J Mol Recognit 2010; 23:241-52. [PMID: 19701919 DOI: 10.1002/jmr.978] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Previously, a geometric nomenclature was proposed in which RNA base pairs were classified by their interaction edges (Watson-Crick, Hoogsteen or sugar-edge) and the glycosidic bond orientations relative to the hydrogen bonds formed (cis or trans). Here, base pairs and pseudo pairs observed in RNA-ligand complexes are classified in a similar manner. Twenty-one basic geometric families are geometrically possible (18 for base pairs formed between a nucleic acid base and a ligand containing heterocycle and 3 families for pseudo pairs). Of those, 16 of them have been observed in X-ray and/or NMR structures.
Collapse
Affiliation(s)
- Jiro Kondo
- Architecture et Réactivité de l'ARN, Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire, CNRS, 15 rue René Descartes, 67084 Strasbourg, France
| | | |
Collapse
|
32
|
Scheunemann AE, Graham WD, Vendeix FAP, Agris PF. Binding of aminoglycoside antibiotics to helix 69 of 23S rRNA. Nucleic Acids Res 2010; 38:3094-105. [PMID: 20110260 PMCID: PMC2875026 DOI: 10.1093/nar/gkp1253] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aminoglycosides antibiotics negate dissociation and recycling of the bacterial ribosome’s subunits by binding to Helix 69 (H69) of 23S rRNA. The differential binding of various aminoglycosides to the chemically synthesized terminal domains of the Escherichia coli and human H69 has been characterized using spectroscopy, calorimetry and NMR. The unmodified E. coli H69 hairpin exhibited a significantly higher affinity for neomycin B and tobramycin than for paromomycin (Kds = 0.3 ± 0.1, 0.2 ± 0.2 and 5.4 ± 1.1 µM, respectively). The binding of streptomycin was too weak to assess. In contrast to the E. coli H69, the human 28S rRNA H69 had a considerable decrease in affinity for the antibiotics, an important validation of the bacterial target. The three conserved pseudouridine modifications (Ψ1911, Ψ1915, Ψ1917) occurring in the loop of the E. coli H69 affected the dissociation constant, but not the stoichiometry for the binding of paromomycin (Kd = 2.6 ± 0.1 µM). G1906 and G1921, observed by NMR spectrometry, figured predominantly in the aminoglycoside binding to H69. The higher affinity of the E. coli H69 for neomycin B and tobramycin, as compared to paromomycin and streptomycin, indicates differences in the efficacy of the aminoglycosides.
Collapse
Affiliation(s)
- Ann E Scheunemann
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC 27695-7622, USA
| | | | | | | |
Collapse
|
33
|
Liu Y, Peacey E, Dickson J, Donahue CP, Zheng S, Varani G, Wolfe MS. Mitoxantrone analogues as ligands for a stem-loop structure of tau pre-mRNA. J Med Chem 2009; 52:6523-6. [PMID: 19839622 DOI: 10.1021/jm9013407] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A series of mitoxantrone (MTX) analogues have been designed, synthesized, and evaluated for binding to and stabilizing a stem-loop structure that serves as a splicing regulatory element in the pre-mRNA of tau, which is involved in Alzheimer's and other neurodegenerative diseases. Several compounds showed significantly improved binding activity relative to the original screening hit mitoxantrone. These findings establish essential structure-activity relationships to further optimize the activity of this promising class of compounds.
Collapse
Affiliation(s)
- Yang Liu
- Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School,H.I.M. 754, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Zheng S, Chen Y, Donahue CP, Wolfe MS, Varani G. Structural basis for stabilization of the tau pre-mRNA splicing regulatory element by novantrone (mitoxantrone). ACTA ACUST UNITED AC 2009; 16:557-66. [PMID: 19477420 DOI: 10.1016/j.chembiol.2009.03.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2008] [Revised: 02/20/2009] [Accepted: 03/11/2009] [Indexed: 10/20/2022]
Abstract
Some familial neurodegenerative diseases are associated with mutations that destabilize a putative stem-loop structure within an intronic region of the tau pre-messenger RNA (mRNA) and alter the production of tau protein isoforms by alternative splicing. Because stabilization of the stem loop reverses the splicing pattern associated with neurodegeneration, small molecules that stabilize this stem loop would provide new ways to dissect the mechanism of neurodegeneration and treat tauopathies. The anticancer drug mitoxantrone was recently identified in a high throughput screen to stabilize the tau pre-mRNA stem loop. Here we report the solution structure of the tau mRNA-mitoxantrone complex, validated by the structure-activity relationship of existing mitoxantrone analogs. The structure describes the molecular basis for their interaction with RNA and provides a rational basis to optimize the activity of this new class of RNA-binding molecules.
Collapse
Affiliation(s)
- Suxin Zheng
- Department of Chemistry, University of Washington, Seattle, 98195, USA
| | | | | | | | | |
Collapse
|
35
|
The secondary structure of the human immunodeficiency virus type 1 transcript modulates viral splicing and infectivity. J Virol 2008; 82:8038-50. [PMID: 18550660 DOI: 10.1128/jvi.00721-08] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Splicing of human immunodeficiency virus type 1 (HIV-1) exon 6D is regulated by the presence of a complex splicing regulatory element (SRE) sequence that interacts with the splicing factors hnRNP H and SC35. In this work, we show that, in the context of the wild-type viral sequence, hnRNP H acts as a repressor of exon 6D inclusion independent of its binding to the SRE. However, hnRNP H binding to the SRE acts as an enhancer of exon 6D inclusion in the presence of a critical T-to-C mutation. These seemingly contrasting functional properties of hnRNP H appear to be caused by a change in the RNA secondary structure induced by the T-to-C mutation that affects the spatial location of bound hnRNP H with respect to the exon 6D splicing determinants. We propose a new regulatory mechanism mediated by RNA folding that may also explain the dual properties of hnRNP H in splicing regulation.
Collapse
|
36
|
González-Díaz H, González-Díaz Y, Santana L, Ubeira FM, Uriarte E. Proteomics, networks and connectivity indices. Proteomics 2008; 8:750-78. [DOI: 10.1002/pmic.200700638] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
37
|
Donahue CP, Ni J, Rozners E, Glicksman MA, Wolfe MS. Identification of tau stem loop RNA stabilizers. ACTA ACUST UNITED AC 2007; 12:789-99. [PMID: 17525136 DOI: 10.1177/1087057107302676] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Alternative splicing of tau exon 10 produces tau isoforms with either 3 (3R) or 4 (4R) repeated microtubule-binding domains. Increased ratios of 4R to 3R tau expression, above the physiological 1:1, leads to neurofibrillary tangles and causes neurodegenerative disease. An RNA stem loop structure plays a significant role in determining the ratio, with decreasing stability correlating with an increase in 4R tau mRNA expression. Recent studies have shown that aminoglycosides are able to bind and stabilize the tau stem loop in vitro, suggesting that other druglike small molecules could be identified and that such molecules might lead to decreased exon 10 splicing in vivo. The authors have developed a fluorescent high-throughput fluorescent binding assay and screened a library of approximately 110,000 compounds to identify candidate drugs that will bind the tau stem loop in vitro. In addition, they have developed a fluorescent-based RNA probe to assay the stabilizing effects of candidate drugs on the tau stem loop RNA. These assays should be applicable to the general problem of identifying small molecules that interact with mRNA secondary structures.
Collapse
Affiliation(s)
- Christine P Donahue
- Center for Neurologic Diseases, Brigham & Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
38
|
Wu JY, Kar A, Kuo D, Yu B, Havlioglu N. SRp54 (SFRS11), a regulator for tau exon 10 alternative splicing identified by an expression cloning strategy. Mol Cell Biol 2006; 26:6739-47. [PMID: 16943417 PMCID: PMC1592875 DOI: 10.1128/mcb.00739-06] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The tau gene encodes a microtubule-associated protein that is critical for neuronal survival and function. Splicing defects in the human tau gene lead to frontotemporal dementia with Parkinsonism linked to chromosome 17 (FTDP-17), an autosomal dominant neurodegenerative disorder. Genetic mutations associated with FTDP-17 often affect tau exon 10 alternative splicing. To investigate mechanisms regulating tau exon 10 alternative splicing, we have developed a green fluorescent protein reporter for tau exon 10 skipping and an expression cloning strategy to identify splicing regulators. A role for SRp54 (also named SFRS11) as a tau exon 10 splicing repressor has been uncovered using this strategy. The overexpression of SRp54 suppresses tau exon 10 inclusion. RNA interference-mediated knock-down of SRp54 increases exon 10 inclusion. SRp54 interacts with a purine-rich element in exon 10 and antagonizes Tra2beta, an SR-domain-containing protein that enhances exon 10 inclusion. Deletion of this exonic element eliminates the activity of SRp54 in suppressing exon 10 inclusion. Our data support a role of SRp54 in regulating tau exon 10 splicing. These experiments also establish a generally useful approach for identifying trans-acting regulators of alternative splicing by expression cloning.
Collapse
Affiliation(s)
- Jane Y Wu
- Northwestern University Feinberg School of Medicine, Center for Genetic Medicine, 303 E. Superior St., Lurie 6-117, Chicago, IL 60611, USA.
| | | | | | | | | |
Collapse
|
39
|
Donahue CP, Muratore C, Wu JY, Kosik KS, Wolfe MS. Stabilization of the tau exon 10 stem loop alters pre-mRNA splicing. J Biol Chem 2006; 281:23302-6. [PMID: 16782711 PMCID: PMC2072867 DOI: 10.1074/jbc.c600143200] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neurofibrillary tangles containing filaments of the microtubule-associated protein tau are found in a variety of neurodegenerative diseases. Mutations in the tau gene itself cause frontotemporal dementia with parkinsonism, demonstrating the critical role of tau in pathogenesis. Many of these mutations in tau are silent, are found at the 5'-splice site of exon 10, and lead to increased inclusion of exon 10. These silent mutations are predicted to destabilize a stem loop structure at the exon 10 5'-splice site; however, the existence of this stem loop under physiological conditions and its role in splice regulation are controversial. Here we show that base changes that stabilize this stem loop in vitro substantially decrease exon 10 inclusion in a wild type tau minigene and rescue the increase in exon 10 splicing caused by a dementia-causing point mutation. Moreover, we probed the intracellular structure of the tau stem loop with antisense RNA and demonstrate that the stability of the stem loop dictates antisense effectiveness. Together these results validate the stem loop as a bona fide structure regulating tau exon 10 splicing.
Collapse
Affiliation(s)
- Christine P. Donahue
- Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Christina Muratore
- Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Jane Y. Wu
- Center for Genetic Medicine, Northwestern University Feinberg Medical School, Chicago, Illinois 60611
| | - Kenneth S. Kosik
- Neuroscience Research Institute, University of California, Santa Barbara, California 93106
| | - Michael S. Wolfe
- Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
40
|
Kar A, Havlioglu N, Tarn WY, Wu JY. RBM4 interacts with an intronic element and stimulates tau exon 10 inclusion. J Biol Chem 2006; 281:24479-88. [PMID: 16777844 PMCID: PMC2072872 DOI: 10.1074/jbc.m603971200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tau protein, which binds to and stabilizes microtubules, is critical for neuronal survival and function. In the human brain, tau pre-mRNA splicing is regulated to maintain a delicate balance of exon 10-containing and exon 10-skipping isoforms. Splicing mutations affecting tau exon 10 alternative splicing lead to tauopathies, a group of neurodegenerative disorders including dementia. Molecular mechanisms regulating tau alternative splicing remain to be elucidated. In this study, we have developed an expression cloning strategy to identify splicing factors that stimulate tau exon 10 inclusion. Using this expression cloning approach, we have identified a previously unknown tau exon 10 splicing regulator, RBM4 (RNA binding motif protein 4). In cells transfected with a tau minigene, RBM4 overexpression leads to an increased inclusion of exon 10, whereas RBM4 down-regulation decreases exon 10 inclusion. The activity of RBM4 in stimulating tau exon 10 inclusion is abolished by mutations in its RNA-binding domain. A putative intronic splicing enhancer located in intron 10 of the tau gene is required for the splicing stimulatory activity of RBM4. Immunohistological analyses reveal that RBM4 is expressed in the human brain regions affected in tauopathy, including the hippocampus and frontal cortex. Our study demonstrates that RBM4 is involved in tau exon 10 alternative splicing. Our work also suggests that down-regulating tau exon 10 splicing activators, such as RBM4, may be of therapeutic potential in tauopathies involving excessive tau exon 10 inclusion.
Collapse
Affiliation(s)
- Amar Kar
- Department of Neurology, Lurie Comprehensive Cancer Center, Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - Necat Havlioglu
- Department of Pathology, Saint Louis University, St. Louis, Missouri 63103
| | - Woan-Yuh Tarn
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Jane Y. Wu
- Department of Neurology, Lurie Comprehensive Cancer Center, Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
- To whom correspondence should be addressed: Northwestern University Feinberg School of Medicine, Center for Genetic Medicine, 303 E. Superior St., Lurie 6-117, Chicago, IL 60611. Tel.: 312-503-0684; Fax: 312-503-5603; E-mail:
| |
Collapse
|
41
|
Kuyumcu-Martinez NM, Cooper TA. Misregulation of alternative splicing causes pathogenesis in myotonic dystrophy. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2006; 44:133-59. [PMID: 17076268 PMCID: PMC4127983 DOI: 10.1007/978-3-540-34449-0_7] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Myotonic dystrophy (DM), the most common form of adult onset muscular dystrophy, affects skeletal muscle, heart, and the central nervous system (CNS). Mortality results primarily from muscle wasting and cardiac arrhythmias. There are two forms of the disease: DM1 and DM2. DM1, which constitutes 98% of cases, is caused by a CTG expansion in the 3' untranslated region (UTR) of the DMPK gene. DM2 is caused by a CCTG expansion in the first intron of the ZNF9 gene. RNA containing CUG- or CCUG-expanded repeats are transcribed but are retained in the nucleus in foci. Disease pathogenesis results primarily from a gain of function of the expanded RNAs, which alter developmentally regulated alternative splicing as well as pathways of muscle differentiation. The toxic RNA has been implicated in sequestration of splicing regulators and transcription factors thereby causing specific symptoms of the disease. Here we review the proposed mechanisms for the toxic effects of the expanded repeats and discuss the molecular mechanisms of splicing misregulation and disease pathogenesis.
Collapse
|
42
|
Abstract
A number of neurodegenerative diseases are characterized by the presence of abundant deposits containing Tau protein. Expression of the human tau gene is under complex regulation. Mutations in the tau gene have been identified in patients with frontotemporal lobe dementia. These mutations affect either biochemical/biophysical properties or the delicate balance of different splicing isoforms. In this review, we summarize recent advances in our understanding of genetics and molecular pathogenesis of tauopathies with the focus on frontotemporal lobe dementia. We review published studies on tau pre-mRNA splicing regulation. Understanding molecular mechanisms of tauopathies may help in developing effective therapies for neurodegenerative tauopathies and related disorders, including Alzheimer disease.
Collapse
Affiliation(s)
- Amar Kar
- Center for Molecular Neuroscience, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David Kuo
- Department of Pediatrics, John F. Kennedy Center for Research on Human Development, Department of Cell and Developmental Biology, Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Rongqiao He
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jiawei Zhou
- Institute of Biochemistry & Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Jane Y. Wu
- Center for Molecular Neuroscience, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Pediatrics, John F. Kennedy Center for Research on Human Development, Department of Cell and Developmental Biology, Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
43
|
Abstract
Tau is the major component of the intracellular filamentous deposits that define a number of neurodegenerative diseases, including the largely sporadic Alzheimer's disease, progressive supranuclear palsy, corticobasal degeneration, Pick's disease, and argyrophilic grain disease, as well as the inherited frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17). For a long time, it was unclear whether the dysfunction of tau protein follows disease or whether disease follows the dysfunction of tau protein. The identification of mutations in Tau as the cause of FTDP-17 has resolved this issue. About half of the known mutations have their primary effect at the protein level, and they reduce the ability of tau protein to interact with microtubules and increase its propensity to assemble into abnormal filaments. The other mutations have their primary effect at the RNA level, thus perturbing the normal ratio of three-repeat to four-repeat tau isoforms. Where studied, this resulted in the relative overproduction of tau protein with four microtubule-binding repeats in brain. Several Tau mutations give rise to diseases that resemble progressive supranuclear palsy, corticobasal degeneration, or Pick's disease. Moreover, the H1 haplotype of Tau has been identified as a significant risk factor for progressive supranuclear palsy and corticobasal degeneration.
Collapse
Affiliation(s)
- Michel Goedert
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK.
| |
Collapse
|
44
|
Goedert M, Jakes R. Mutations causing neurodegenerative tauopathies. Biochim Biophys Acta Mol Basis Dis 2005; 1739:240-50. [PMID: 15615642 DOI: 10.1016/j.bbadis.2004.08.007] [Citation(s) in RCA: 295] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2004] [Accepted: 08/17/2004] [Indexed: 12/25/2022]
Abstract
Tau is the major component of the intracellular filamentous deposits that define a number of neurodegenerative diseases. They include the largely sporadic Alzheimer's disease (AD), progressive supranuclear palsy, corticobasal degeneration, Pick's disease and argyrophilic grain disease, as well as the inherited frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17). For a long time, it was unclear whether the dysfunction of tau protein follows disease or whether disease follows tau dysfunction. This was resolved when mutations in Tau were found to cause FTDP-17. Currently, 32 different mutations have been identified in over 100 families. About half of the known mutations have their primary effect at the protein level. They reduce the ability of tau protein to interact with microtubules and increase its propensity to assemble into abnormal filaments. The other mutations have their primary effect at the RNA level and perturb the normal ratio of three-repeat to four-repeat tau isoforms. Where studied, this resulted in a relative overproduction of tau protein with four microtubule-binding domains in the brain. Individual Tau mutations give rise to diseases that resemble progressive supranuclear palsy, corticobasal degeneration or Pick's disease. Moreover, the H1 haplotype of Tau has been identified as a significant risk factor for progressive supranuclear palsy and corticobasal degeneration. At a practical level, the new work is leading to the production of experimental animal models that reproduce the essential molecular and cellular features of the human tauopathies, including the formation of abundant filaments made of hyperphosphorylated tau protein and nerve cell degeneration.
Collapse
Affiliation(s)
- Michel Goedert
- MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB2 2QH, UK.
| | | |
Collapse
|
45
|
Affiliation(s)
- Emanuele Buratti
- International Centre for Genetic Engineering and Biotechnology, Padriciano 99, 34012 Trieste, Italy
| | | |
Collapse
|
46
|
Kondo S, Yamamoto N, Murakami T, Okumura M, Mayeda A, Imaizumi K. Tra2 beta, SF2/ASF and SRp30c modulate the function of an exonic splicing enhancer in exon 10 of tau pre-mRNA. Genes Cells 2004; 9:121-30. [PMID: 15009090 DOI: 10.1111/j.1356-9597.2004.00709.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Some of mutations in the tau gene, which were found in frontotemporal dementia with Parkinsonism linked to chromosome 17 (FTDP-17), affect alternative splicing of its exon 10 which encodes one of four microtubule-binding motifs. To examine the molecular mechanisms responsible for aberrant splicing of the tau gene containing mutations linked to FTDP-17, we performed Exon trapping and binding assay using tau exon 10 pre-mRNA and nuclear extracts of neuroblastoma cell lines and in vitro splicing using dsx-substrate. We determined that 5' site of tau exon 10 (nucleotides 12-45) possesses exonic splicing enhancer (ESE) activities in vitro splicing and the FTDP-17-linked mutations affect the ESE activities and alter the splicing patterns of tau exon 10. Tra2 beta directly and ASF/SF2 indirectly associated with the ESE of wild tau exon 10. The binding amounts of these SR proteins to tau exon 10 bearing N279K mutation increased and they enhanced splicing the mutant tau exon 10. SRp30c also enhanced the splicing of tau exon 10. These results suggest that mutations in tau exon 10 that are linked to FTDP-17 affect the ESE activities by altering the binding of some SR proteins to its pre-mRNA.
Collapse
Affiliation(s)
- Shinichi Kondo
- Division of Structural Cellular Biology, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0101, Japan
| | | | | | | | | | | |
Collapse
|
47
|
Detering C, Varani G. Validation of automated docking programs for docking and database screening against RNA drug targets. J Med Chem 2004; 47:4188-201. [PMID: 15293991 DOI: 10.1021/jm030650o] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The increasing awareness of the essential role of RNA in controlling viral replication and in bacterial protein synthesis emphasizes the potential of ribonucleoproteins as targets for developing new antibacterial and antiviral drugs. RNA forms well defined three-dimensional structures with clefts and binding pockets reminiscent of the active sites of proteins. Furthermore, it precedes proteins in the translation pathway; inhibiting the function of a single RNA molecule would result in inhibition of multiple proteins. Thus, small molecules that bind RNA specifically would combine the advantages of antisense and RNAi strategies with the much more favorable medicinal chemistry of small-molecule therapeutics. The discovery of small-molecule inhibitors of RNA with attractive pharmacological potential would be facilitated if we had available effective computational tools of structure-based drug design. Here, we systematically test automated docking tools developed for proteins using existing three-dimensional structures of RNA-small molecule complexes. The results show that the native structures can generally be reproduced to within 2.5 angstroms more than 50-60% of the time. For more than half of the test complexes, the native ligand ranked among the top 10% compounds in a database-scoring test. Through this work, we provide parameters for the validated application of automated docking tools to the discovery of new inhibitors of RNA function.
Collapse
Affiliation(s)
- Carsten Detering
- Departments of Chemistry and Biochemistry, University of Washington, Seattle, Washington 98195-1700, USA
| | | |
Collapse
|
48
|
McPike MP, Goodisman J, Dabrowiak JC. Specificity of neomycin analogues bound to the packaging region of human immunodeficiency virus type 1 RNA. Bioorg Med Chem 2004; 12:1835-43. [PMID: 15051052 DOI: 10.1016/j.bmc.2004.01.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2003] [Revised: 01/28/2004] [Accepted: 01/28/2004] [Indexed: 11/24/2022]
Abstract
The packaging region of HIV-1 RNA contains a number of structural features which are important in the life cycle of the virus, making this segment of RNA a potential target for new types of AIDS-directed drugs. We studied the binding of three neomycin analogues (neo-guanidino, neo-acridine, and neo-neo) to a 171-mer RNA molecule from the packaging region of HIV-1 using quantitative footprinting and circular dichroism. Neo-guanidino produced footprinting patterns and effects on the CD similar to those observed for neomycin and paromomycin, indicating that all three compounds bind to the same regions of the 171-mer. Neo-guanidino binds to SL 1 where it joins the large internal loop, near a bulge in the stem of SL 1, and on SL 2. Neo-acridine, which has an acridine attached to neomycin, and neo-neo, which has two neomycins linked by a flexible tether, bind bivalently, and give very different footprinting and CD results from the other compounds. The neomycin portion of neo-acridine binds to the same sites as neomycin, while the attached acridine group appears to bind to a duplex region in the main stem of the folded 171-mer. Since the footprinting data for this analogue show few enhancements, bivalent binding of neo-acridine appears to stabilize the folded structure of RNA by effectively 'stapling' parts of the structure together. Neo-neo induces significant structural changes in RNA where neomycin binds. This may be related to the inability of both neomycins of neo-neo it find optimal binding sites adjacent to one another without changing RNA structure. The intensity of a strong negative CD band in the spectrum of psi-RNA at 208 nm is sensitive to drug-induced changes in RNA structure. Neo-guanidino and neo-neo (also neomycin and paromomycin), which change RNA structure, cause an increase in intensity while neo-acridine, which induces little distortion to RNA, causes a decrease in intensity. Molecular modeling analysis shows that C-5' of ribose of neo-acridine and neo-neo must be directed away from the binding pocket when these analogues are bivalently bound to RNA. This study showed how variations in the structure of aminoglycosides lead to different binding specificity to part of the packaging region of HIV-1. Such knowledge will be important in design of drugs to target this region.
Collapse
Affiliation(s)
- Mark P McPike
- Department of Chemistry, Center for Science and Technology, R 1-014, Syracuse University, Syracuse, New York 13244-4100, USA
| | | | | |
Collapse
|
49
|
Jiang Z, Tang H, Havlioglu N, Zhang X, Stamm S, Yan R, Wu JY. Mutations in tau gene exon 10 associated with FTDP-17 alter the activity of an exonic splicing enhancer to interact with Tra2 beta. J Biol Chem 2003; 278:18997-9007. [PMID: 12649279 PMCID: PMC2140226 DOI: 10.1074/jbc.m301800200] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Mutations in the human tau gene leading to aberrant splicing have been identified in FTDP-17, an autosomal dominant hereditary neurodegenerative disorder. Molecular mechanisms by which such mutations cause tau aberrant splicing were not understood. We characterized two mutations in exon 10 of the tau gene, N279K and Del280K. Our results revealed an exonic splicing enhancer element located in exon 10. The activity of this AG-rich splicing enhancer was altered by N279K and Del280K mutations. This exonic enhancer element interacts with human Tra2 beta protein. The interaction between Tra2 beta and the exonic splicing enhancer correlates with the activity of this enhancer element in stimulating splicing. Biochemical studies including in vitro splicing and RNA interference experiments in transfected cells support a role for Tra2 beta protein in regulating alternative splicing of human tau gene. Our results implicate the human tau gene as a target gene for the alternative splicing regulator Tra2 beta, suggesting that Tra2 beta may play a role in aberrant tau exon 10 alternative splicing and in the pathogenesis of tauopathies.
Collapse
Affiliation(s)
- Zhihong Jiang
- Departments of Pediatrics and of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Hao Tang
- Departments of Pediatrics and of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Necat Havlioglu
- Departments of Pediatrics and of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Xiaochun Zhang
- Departments of Pediatrics and of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Stefan Stamm
- Universitate Erlangen-Nurenberg, Institute of Biochemistry, Fahrstrasse 17, Erlangen 91054, Germany
| | - Riqiang Yan
- Department of Cell and Molecular Biology, Pharmacia Corp., Kalamazoo, Michigan 49007
| | - Jane Y. Wu
- Departments of Pediatrics and of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, Missouri 63110
- To whom correspondence should be addressed. Tel.: 314−286−2798; Fax: 314−286−2892; E-mail:
| |
Collapse
|
50
|
Affiliation(s)
- Nuno André Faustino
- Department of Pathology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | |
Collapse
|