1
|
Kim H, Park H, Schulz ET, Azuma Y, Azuma M. EWSR1 prevents the induction of aneuploidy through direct regulation of Aurora B. Front Cell Dev Biol 2023; 11:987153. [PMID: 36875767 PMCID: PMC9975954 DOI: 10.3389/fcell.2023.987153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 01/09/2023] [Indexed: 02/17/2023] Open
Abstract
EWSR1 (Ewing sarcoma breakpoint region 1) was originally identified as a part of an aberrant EWSR1/FLI1 fusion gene in Ewing sarcoma, the second most common pediatric bone cancer. Due to formation of the EWSR1/FLI1 fusion gene in the tumor genome, the cell loses one wild type EWSR1 allele. Our previous study demonstrated that the loss of ewsr1a (homologue of human EWSR1) in zebrafish leads to the high incidence of mitotic dysfunction, of aneuploidy, and of tumorigenesis in the tp53 mutant background. To dissect the molecular function of EWSR1, we successfully established a stable DLD-1 cell line that enables a conditional knockdown of EWSR1 using an Auxin Inducible Degron (AID) system. When both EWSR1 genes of DLD-1 cell were tagged with mini-AID at its 5'-end using a CRISPR/Cas9 system, treatment of the (AID-EWSR1/AID-EWSR1) DLD-1 cells with a plant-based Auxin (AUX) led to the significant levels of degradation of AID-EWSR1 proteins. During anaphase, the EWSR1 knockdown (AUX+) cells displayed higher incidence of lagging chromosomes compared to the control (AUX-) cells. This defect was proceeded by a lower incidence of the localization of Aurora B at inner centromeres, and by a higher incidence of the protein at Kinetochore proximal centromere compared to the control cells during pro/metaphase. Despite these defects, the EWSR1 knockdown cells did not undergo mitotic arrest, suggesting that the cell lacks the error correction mechanism. Significantly, the EWSR1 knockdown (AUX+) cells induced higher incidence of aneuploidy compared to the control (AUX-) cells. Since our previous study demonstrated that EWSR1 interacts with the key mitotic kinase, Aurora B, we generated replacement lines of EWSR1-mCherry and EWSR1:R565A-mCherry (a mutant that has low affinity for Aurora B) in the (AID-EWSR1/AID-EWSR1) DLD-1 cells. The EWSR1-mCherry rescued the high incidence of aneuploidy of EWSR1 knockdown cells, whereas EWSR1-mCherry:R565A failed to rescue the phenotype. Together, we demonstrate that EWSR1 prevents the induction of lagging chromosomes, and of aneuploidy through the interaction with Aurora B.
Collapse
Affiliation(s)
| | | | | | | | - Mizuki Azuma
- Molecular Biosciences, University of Kansas, Lawrence, KS, United States
| |
Collapse
|
2
|
Kitagawa T, Kobayashi D, Baron B, Okita H, Miyamoto T, Takai R, Paudel D, Ohta T, Asaoka Y, Tokunaga M, Nakagawa K, Furutani-Seiki M, Araki N, Kuramitsu Y, Kobayashi M. AT-hook DNA-binding motif-containing protein one knockdown downregulates EWS-FLI1 transcriptional activity in Ewing's sarcoma cells. PLoS One 2022; 17:e0269077. [PMID: 36194562 PMCID: PMC9531837 DOI: 10.1371/journal.pone.0269077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 09/06/2022] [Indexed: 11/18/2022] Open
Abstract
Ewing's sarcoma is the second most common bone malignancy in children or young adults and is caused by an oncogenic transcription factor by a chromosomal translocation between the EWSR1 gene and the ETS transcription factor family. However, the transcriptional mechanism of EWS-ETS fusion proteins is still unclear. To identify the transcriptional complexes of EWS-ETS fusion transcription factors, we applied a proximal labeling system called BioID in Ewing's sarcoma cells. We identified AHDC1 as a proximal protein of EWS-ETS fusion proteins. AHDC1 knockdown showed a reduced cell growth and transcriptional activity of EWS-FLI1. AHDC1 knockdown also reduced BRD4 and BRG1 protein levels, both known as interacting proteins of EWS-FLI1. Our results suggest that AHDC1 supports cell growth through EWS-FLI1.
Collapse
Affiliation(s)
- Takao Kitagawa
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Hokkaido, Japan
- * E-mail:
| | - Daiki Kobayashi
- Department of Omics and Systems Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Department of Tumor Genetics and Biology, Faculty of Life Sciences, Kumamoto University, Kumamoto-Shi, Kumamoto, Japan
| | - Byron Baron
- Center for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Hajime Okita
- Division of Diagnostic Pathology, Keio University School of Medicine, Shinano, Shinjuku-ku, Tokyo, Japan
| | - Tatsuo Miyamoto
- Department of Molecular and Cellular Physiology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Rie Takai
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Hokkaido, Japan
| | - Durga Paudel
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Hokkaido, Japan
| | - Tohru Ohta
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Hokkaido, Japan
| | - Yoichi Asaoka
- Department of Systems Biochemistry in Pathology and Regeneration, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Masayuki Tokunaga
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Koji Nakagawa
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Hokkaido, Japan
| | - Makoto Furutani-Seiki
- Department of Systems Biochemistry in Pathology and Regeneration, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Norie Araki
- Department of Tumor Genetics and Biology, Faculty of Life Sciences, Kumamoto University, Kumamoto-Shi, Kumamoto, Japan
| | - Yasuhiro Kuramitsu
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Hokkaido, Japan
| | - Masanobu Kobayashi
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Hokkaido, Japan
| |
Collapse
|
3
|
Carron J, Torricelli C, Silva JK, Coser LDO, Lima CSP, Lourenço GJ. Intronic variants of MITF (rs7623610) and CREB1 (rs10932201) genes may enhance splicing efficiency in human melanoma cell line. Mutat Res 2021; 823:111763. [PMID: 34710701 DOI: 10.1016/j.mrfmmm.2021.111763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/24/2021] [Accepted: 10/17/2021] [Indexed: 11/20/2022]
Abstract
We previously reported that intronic single nucleotide variations (SNVs) in MITF (c.938-325G>A, rs7623610) and CREB1 (c.303+373G>A, rs10932201) genes were associated with risk, aggressiveness, and prognosis of cutaneous melanoma (CM). In this study, we investigated the influence of the above SNVs in splicing patterns and efficiency. We constructed minigenes with wild type and variant alleles from MITF and CREB1 to assess the effect of the SNVs on splicing. The minigenes were transfected in the human melanoma cell line (SK-MEL-28). RT-PCR and DNA sequencing investigated the constructs' splicing patterns. Minigenes constructs' splicing efficiency and HNRNPA1 and SF1 splicing genes' expression were investigated by qPCR. We found that MITF and CREB1 SNVs did not alter the splicing pattern, but they influenced the splicing efficiency. MITF-A (p= 0.03) and CREB1-A (p= 0.005) variant minigenes yielded an increase of mRNA generated from the constructions. Additionally, lower mRNA levels of HNRNPA1 and SF1 were seen in the variant minigenes MITF-A (p= 0.04) and CREB1-A (p= 0.005). We described for the first time the potential importance of MITF rs7623610 and CREB1 rs10932201 SNVs in splicing efficiency and its relationship with CM.
Collapse
Affiliation(s)
- Juliana Carron
- Laboratory of Cancer Genetics, School of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Caroline Torricelli
- Laboratory of Cancer Genetics, School of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Janet Keller Silva
- Laboratory of Cancer Genetics, School of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Lilian de Oliveira Coser
- Laboratory of Nerve Regeneration, Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| | - Carmen Silvia Passos Lima
- Laboratory of Cancer Genetics, School of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil; Department of Radiology, School of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Gustavo Jacob Lourenço
- Laboratory of Cancer Genetics, School of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil.
| |
Collapse
|
4
|
The HIV-1 Tat Protein Enhances Splicing at the Major Splice Donor Site. J Virol 2018; 92:JVI.01855-17. [PMID: 29743356 DOI: 10.1128/jvi.01855-17] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 04/25/2018] [Indexed: 12/23/2022] Open
Abstract
Transcription of the HIV-1 proviral DNA and subsequent processing of the primary transcript results in the production of a large set of unspliced and differentially spliced viral RNAs. The major splice donor site (5'ss) that is located in the untranslated leader of the HIV-1 transcript is used for the production of all spliced RNAs, and splicing at this site has to be tightly regulated to allow the balanced production of all viral RNAs and proteins. We demonstrate that the viral Tat protein, which is known to activate viral transcription, also stimulates splicing at the major 5'ss. As for the transcription effect, Tat requires the viral long terminal repeat promoter and the trans-acting responsive RNA hairpin for splicing regulation. These results indicate that HIV-1 transcription and splicing are tightly coupled processes through the coordinated action of the essential Tat protein.IMPORTANCE The HIV-1 proviral DNA encodes a single RNA transcript that is used as RNA genome and packaged into newly assembled virus particles. This full-length RNA is also used as mRNA for the production of structural and enzymatic proteins. Production of other essential viral proteins depends on alternative splicing of the primary transcript, which yields a large set of differentially spliced mRNAs. Optimal virus replication requires a balanced production of all viral RNAs, which means that the splicing process has to be strictly regulated. We show that the HIV-1 Tat protein, a factor that is well known for its transcription activating function, also stimulates splicing. Thus, Tat controls not only the level of the viral RNA but also the balance between spliced and unspliced RNAs.
Collapse
|
5
|
Nanan KK, Ocheltree C, Sturgill D, Mandler MD, Prigge M, Varma G, Oberdoerffer S. Independence between pre-mRNA splicing and DNA methylation in an isogenic minigene resource. Nucleic Acids Res 2017; 45:12780-12797. [PMID: 29244186 PMCID: PMC5727405 DOI: 10.1093/nar/gkx900] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 09/13/2017] [Accepted: 09/25/2017] [Indexed: 12/27/2022] Open
Abstract
Actively transcribed genes adopt a unique chromatin environment with characteristic patterns of enrichment. Within gene bodies, H3K36me3 and cytosine DNA methylation are elevated at exons of spliced genes and have been implicated in the regulation of pre-mRNA splicing. H3K36me3 is further responsive to splicing, wherein splicing inhibition led to a redistribution and general reduction over gene bodies. In contrast, little is known of the mechanisms supporting elevated DNA methylation at actively spliced genic locations. Recent evidence associating the de novo DNA methyltransferase Dnmt3b with H3K36me3-rich chromatin raises the possibility that genic DNA methylation is influenced by splicing-associated H3K36me3. Here, we report the generation of an isogenic resource to test the direct impact of splicing on chromatin. A panel of minigenes of varying splicing potential were integrated into a single FRT site for inducible expression. Profiling of H3K36me3 confirmed the established relationship to splicing, wherein levels were directly correlated with splicing efficiency. In contrast, DNA methylation was equivalently detected across the minigene panel, irrespective of splicing and H3K36me3 status. In addition to revealing a degree of independence between genic H3K36me3 and DNA methylation, these findings highlight the generated minigene panel as a flexible platform for the query of splicing-dependent chromatin modifications.
Collapse
Affiliation(s)
- Kyster K. Nanan
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cody Ocheltree
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David Sturgill
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mariana D. Mandler
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Maria Prigge
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Garima Varma
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shalini Oberdoerffer
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
6
|
Chatrikhi R, Wang W, Gupta A, Loerch S, Maucuer A, Kielkopf CL. SF1 Phosphorylation Enhances Specific Binding to U2AF 65 and Reduces Binding to 3'-Splice-Site RNA. Biophys J 2017; 111:2570-2586. [PMID: 28002734 DOI: 10.1016/j.bpj.2016.11.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/02/2016] [Accepted: 11/08/2016] [Indexed: 12/25/2022] Open
Abstract
Splicing factor 1 (SF1) recognizes 3' splice sites of the major class of introns as a ternary complex with U2AF65 and U2AF35 splicing factors. A conserved SPSP motif in a coiled-coil domain of SF1 is highly phosphorylated in proliferating human cells and is required for cell proliferation. The UHM kinase 1 (UHMK1), also called KIS, double-phosphorylates both serines of this SF1 motif. Here, we use isothermal titration calorimetry to demonstrate that UHMK1 phosphorylation of the SF1 SPSP motif slightly enhances specific binding of phospho-SF1 to its cognate U2AF65 protein partner. Conversely, quantitative fluorescence anisotropy RNA binding assays and isothermal titration calorimetry experiments establish that double-SPSP phosphorylation reduces phospho-SF1 and phospho-SF1-U2AF65 binding affinities for either optimal or suboptimal splice-site RNAs. Domain-substitution and mutagenesis experiments further demonstrate that arginines surrounding the phosphorylated SF1 loop are required for cooperative 3' splice site recognition by the SF1-U2AF65 complex (where cooperativity is defined as a nonadditive increase in RNA binding by the protein complex relative to the individual proteins). In the context of local, intracellular concentrations, the subtle effects of SF1 phosphorylation on its associations with U2AF65 and splice-site RNAs are likely to influence pre-mRNA splicing. However, considering roles for SF1 in pre-mRNA retention and transcriptional repression, as well as in splicing, future comprehensive investigations are needed to fully explain the requirement for SF1 SPSP phosphorylation in proliferating human cells.
Collapse
Affiliation(s)
- Rakesh Chatrikhi
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine, Rochester, New York
| | - Wenhua Wang
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine, Rochester, New York
| | - Ankit Gupta
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine, Rochester, New York
| | - Sarah Loerch
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine, Rochester, New York
| | | | - Clara L Kielkopf
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine, Rochester, New York.
| |
Collapse
|
7
|
Cai W, Xiong Chen Z, Rane G, Satendra Singh S, Choo Z, Wang C, Yuan Y, Zea Tan T, Arfuso F, Yap CT, Pongor LS, Yang H, Lee MB, Cher Goh B, Sethi G, Benoukraf T, Tergaonkar V, Prem Kumar A. Wanted DEAD/H or Alive: Helicases Winding Up in Cancers. J Natl Cancer Inst 2017; 109:2957323. [PMID: 28122908 DOI: 10.1093/jnci/djw278] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/08/2016] [Accepted: 10/20/2016] [Indexed: 12/23/2022] Open
Abstract
Cancer is one of the most studied areas of human biology over the past century. Despite having attracted much attention, hype, and investments, the search to find a cure for cancer remains an uphill battle. Recent discoveries that challenged the central dogma of molecular biology not only further increase the complexity but also demonstrate how various types of noncoding RNAs such as microRNA and long noncoding RNA, as well as their related processes such as RNA editing, are important in regulating gene expression. Parallel to this aspect, an increasing number of reports have focused on a family of proteins known as DEAD/H-box helicases involved in RNA metabolism, regulation of long and short noncoding RNAs, and novel roles as "editing helicases" and their association with cancers. This review summarizes recent findings on the roles of RNA helicases in various cancers, which are broadly classified into adult solid tumors, childhood solid tumors, leukemia, and cancer stem cells. The potential small molecule inhibitors of helicases and their therapeutic value are also discussed. In addition, analyzing next-generation sequencing data obtained from public portals and reviewing existing literature, we provide new insights on the potential of DEAD/H-box helicases to act as pharmacological drug targets in cancers.
Collapse
Affiliation(s)
- Wanpei Cai
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Zhi Xiong Chen
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Grishma Rane
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Shikha Satendra Singh
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Zhang'e Choo
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Chao Wang
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Yi Yuan
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Tuan Zea Tan
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Frank Arfuso
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Celestial T Yap
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Lorinc S Pongor
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Henry Yang
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Martin B Lee
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Boon Cher Goh
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Gautam Sethi
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Touati Benoukraf
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Vinay Tergaonkar
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| | - Alan Prem Kumar
- Affiliations of authors: Cancer Science Institute of Singapore, National University of Singapore, Singapore (WC, GR, SSS, CW, YY, TZT, HY, BCG, TB, APK); Departments of Pharmacology (WC, GR, SSS, CW, BCG, GS, APK), Physiology (ZXC, ZC, CTY), and Biochemistry (VT), Yong Loo Lin School of Medicine, National University of Singapore, Singapore; KK Women's and Children's Hospital, Singapore (ZXC); Stem Cell and Cancer Biology Laboratory (FA), School of Biomedical Sciences (GS, APK), Curtin Health Innovation Research Institute, Curtin Medical School (APK), Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore (CTY, BCG, APK); 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary (LSP); MTA TTK Lendület Cancer Biomarker Research Group, Research Centre for Natural Sciences, Budapest, Hungary (LSP); Department of Renal Medicine (MBL) and Department of Haematology-Oncology (BCG), National University Health System, Singapore; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore (VT); Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia (VT); Department of Biological Sciences, University of North Texas, Denton, TX (APK)
| |
Collapse
|
8
|
Heintz C, Doktor TK, Lanjuin A, Escoubas C, Zhang Y, Weir HJ, Dutta S, Silva-García CG, Bruun GH, Morantte I, Hoxhaj G, Manning BD, Andresen BS, Mair WB. Splicing factor 1 modulates dietary restriction and TORC1 pathway longevity in C. elegans. Nature 2017; 541:102-106. [PMID: 27919065 PMCID: PMC5361225 DOI: 10.1038/nature20789] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 11/15/2016] [Indexed: 12/31/2022]
Abstract
Ageing is driven by a loss of transcriptional and protein homeostasis and is the key risk factor for multiple chronic diseases. Interventions that attenuate or reverse systemic dysfunction associated with age therefore have the potential to reduce overall disease risk in the elderly. Precursor mRNA (pre-mRNA) splicing is a fundamental link between gene expression and the proteome, and deregulation of the splicing machinery is linked to several age-related chronic illnesses. However, the role of splicing homeostasis in healthy ageing remains unclear. Here we demonstrate that pre-mRNA splicing homeostasis is a biomarker and predictor of life expectancy in Caenorhabditis elegans. Using transcriptomics and in-depth splicing analysis in young and old animals fed ad libitum or subjected to dietary restriction, we find defects in global pre-mRNA splicing with age that are reduced by dietary restriction via splicing factor 1 (SFA-1; the C. elegans homologue of SF1, also known as branchpoint binding protein, BBP). We show that SFA-1 is specifically required for lifespan extension by dietary restriction and by modulation of the TORC1 pathway components AMPK, RAGA-1 and RSKS-1/S6 kinase. We also demonstrate that overexpression of SFA-1 is sufficient to extend lifespan. Together, these data demonstrate a role for RNA splicing homeostasis in dietary restriction longevity and suggest that modulation of specific spliceosome components may prolong healthy ageing.
Collapse
Affiliation(s)
- Caroline Heintz
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Thomas Koed Doktor
- Department of Biochemistry and Molecular Biology, and Villum Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | - Anne Lanjuin
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Caroline Escoubas
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
- Institute for Research on Cancer and Aging, Nice (IRCAN), CNRS, UMR7284, INSERM U1081, University of Nice Sophia Antipolis, Faculty of Medicine, Nice, France
| | - Yue Zhang
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Heather J Weir
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Sneha Dutta
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | | | - Gitte Hoffmann Bruun
- Department of Biochemistry and Molecular Biology, and Villum Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | - Ianessa Morantte
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Gerta Hoxhaj
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Brendan D Manning
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Brage S Andresen
- Department of Biochemistry and Molecular Biology, and Villum Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | - William B Mair
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
9
|
Abstract
Neurodegenerative disorders such as Alzheimer disease (AD), frontotemporal dementia (FTD), amyotrophic lateral sclerosis (ALS), Parkinson disease (PD), Huntington's disease (HD), and multiple sclerosis (MS) affect different neuronal cells, and have a variable age of onset, clinical symptoms, and pathological features. Despite the great progress in understanding the etiology of these disorders, the underlying mechanisms remain largely unclear. Among the processes affected in neurodegenerative diseases, alteration in RNA metabolism is emerging as a crucial player. RNA-binding proteins (RBPs) are involved at all stages of RNA metabolism and display a broad range of functions, including modulation of mRNA transcription, splicing, editing, export, stability, translation and localization and miRNA biogenesis, thus enormously impacting regulation of gene expression. On the other hand, aberrant regulation of RBP expression or activity can contribute to disease onset and progression. Recent reports identified mutations causative of neurological disorders in the genes encoding a family of RBPs named FET (FUS/TLS, EWS and TAF15). This review summarizes recent works documenting the involvement of FET proteins in the pathology of ALS, FTLD, essential tremor (ET) and other neurodegenerative diseases. Moreover, clinical implications of recent advances in FET research are critically discussed.
Collapse
Affiliation(s)
- Francesca Svetoni
- a University of Rome "Foro Italico," , Rome , Italy.,b Laboratory of Cellular and Molecular Neurobiology, Fondazione Santa Lucia , Rome , Italy
| | - Paola Frisone
- b Laboratory of Cellular and Molecular Neurobiology, Fondazione Santa Lucia , Rome , Italy
| | - Maria Paola Paronetto
- a University of Rome "Foro Italico," , Rome , Italy.,b Laboratory of Cellular and Molecular Neurobiology, Fondazione Santa Lucia , Rome , Italy
| |
Collapse
|
10
|
Regulation of Stem Cell Self-Renewal and Oncogenesis by RNA-Binding Proteins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 907:153-88. [PMID: 27256386 DOI: 10.1007/978-3-319-29073-7_7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Throughout their life span, multicellular organisms rely on stem cell systems. During development pluripotent embryonic stem cells give rise to all cell types that make up the organism. After birth, tissue stem cells maintain properly functioning tissues and organs under homeostasis as well as promote regeneration after tissue damage or injury. Stem cells are capable of self-renewal, which is the ability to divide indefinitely while retaining the potential of differentiation into multiple cell types. The ability to self-renew, however, is a double-edged sword; the molecular mechanisms of self-renewal can be a target of malignant transformation driving tumor development and progression. Growing lines of evidence have shown that RNA-binding proteins (RBPs) play pivotal roles in the regulation of self-renewal by modulating metabolism of coding and non-coding RNAs both in normal tissues and in cancers. In this review, we discuss our current understanding of tissue stem cell systems and how RBPs regulate stem cell fates as well as how the regulatory functions of RBPs contribute to oncogenesis.
Collapse
|
11
|
Shkreta L, Chabot B. The RNA Splicing Response to DNA Damage. Biomolecules 2015; 5:2935-77. [PMID: 26529031 PMCID: PMC4693264 DOI: 10.3390/biom5042935] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 09/20/2015] [Accepted: 10/16/2015] [Indexed: 12/29/2022] Open
Abstract
The number of factors known to participate in the DNA damage response (DDR) has expanded considerably in recent years to include splicing and alternative splicing factors. While the binding of splicing proteins and ribonucleoprotein complexes to nascent transcripts prevents genomic instability by deterring the formation of RNA/DNA duplexes, splicing factors are also recruited to, or removed from, sites of DNA damage. The first steps of the DDR promote the post-translational modification of splicing factors to affect their localization and activity, while more downstream DDR events alter their expression. Although descriptions of molecular mechanisms remain limited, an emerging trend is that DNA damage disrupts the coupling of constitutive and alternative splicing with the transcription of genes involved in DNA repair, cell-cycle control and apoptosis. A better understanding of how changes in splice site selection are integrated into the DDR may provide new avenues to combat cancer and delay aging.
Collapse
Affiliation(s)
- Lulzim Shkreta
- Microbiologie et d'Infectiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada.
| | - Benoit Chabot
- Microbiologie et d'Infectiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada.
| |
Collapse
|
12
|
Crisci A, Raleff F, Bagdiul I, Raabe M, Urlaub H, Rain JC, Krämer A. Mammalian splicing factor SF1 interacts with SURP domains of U2 snRNP-associated proteins. Nucleic Acids Res 2015; 43:10456-73. [PMID: 26420826 PMCID: PMC4666396 DOI: 10.1093/nar/gkv952] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 09/10/2015] [Indexed: 02/03/2023] Open
Abstract
Splicing factor 1 (SF1) recognizes the branch point sequence (BPS) at the 3′ splice site during the formation of early complex E, thereby pre-bulging the BPS adenosine, thought to facilitate subsequent base-pairing of the U2 snRNA with the BPS. The 65-kDa subunit of U2 snRNP auxiliary factor (U2AF65) interacts with SF1 and was shown to recruit the U2 snRNP to the spliceosome. Co-immunoprecipitation experiments of SF1-interacting proteins from HeLa cell extracts shown here are consistent with the presence of SF1 in early splicing complexes. Surprisingly almost all U2 snRNP proteins were found associated with SF1. Yeast two-hybrid screens identified two SURP domain-containing U2 snRNP proteins as partners of SF1. A short, evolutionarily conserved region of SF1 interacts with the SURP domains, stressing their role in protein–protein interactions. A reduction of A complex formation in SF1-depleted extracts could be rescued with recombinant SF1 containing the SURP-interaction domain, but only partial rescue was observed with SF1 lacking this sequence. Thus, SF1 can initially recruit the U2 snRNP to the spliceosome during E complex formation, whereas U2AF65 may stabilize the association of the U2 snRNP with the spliceosome at later times. In addition, these findings may have implications for alternative splicing decisions.
Collapse
Affiliation(s)
- Angela Crisci
- Department of Cell Biology, Faculty of Sciences, University of Geneva, CH-1211 Geneva 4, Switzerland
| | - Flore Raleff
- Department of Cell Biology, Faculty of Sciences, University of Geneva, CH-1211 Geneva 4, Switzerland
| | - Ivona Bagdiul
- Department of Cell Biology, Faculty of Sciences, University of Geneva, CH-1211 Geneva 4, Switzerland
| | - Monika Raabe
- Bioanalytical Mass Spectrometry, Max Planck Institute for Biophysical Chemistry, D-37077 Göttingen, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry, Max Planck Institute for Biophysical Chemistry, D-37077 Göttingen, Germany Bioanalytics, Institute for Clinical Chemistry, University Medical Center Göttingen, D-37075 Göttingen, Germany
| | | | - Angela Krämer
- Department of Cell Biology, Faculty of Sciences, University of Geneva, CH-1211 Geneva 4, Switzerland
| |
Collapse
|
13
|
Structure-function based molecular relationships in Ewing's sarcoma. BIOMED RESEARCH INTERNATIONAL 2015; 2015:798426. [PMID: 25688366 PMCID: PMC4320925 DOI: 10.1155/2015/798426] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 08/30/2014] [Accepted: 10/14/2014] [Indexed: 02/07/2023]
Abstract
Ewing's Sarcoma Oncogene (ews) on chromosome 22q12 is encoding a ubiquitously expressed RNA-binding protein (EWS) with unknown function that is target of tumor-specific chromosomal translocations in Ewing's sarcoma family of tumors. A model of transcription complex was proposed in which the heterodimer Rpb4/7 binds to EAD, connecting it to Core RNA Pol II. The DNA-binding domain, provided by EFP, is bound to the promoter. Rpb4/7 binds RNA, stabilizing the transcription complex. The complex Rpb4/7 can stabilize the preinitiation complexes by converting the conformation of RNA Pol II. EWS may change its conformation, so that NTD becomes accessible. Two different mechanisms of interaction between EWS and RNA Pol II are proposed: (I) an intermolecular EWS-EWS interaction between two molecules, pushing conformation from “closed” to “open” state, or (II) an intramolecular interaction inside the molecule of EWS, pushing conformation of the molecule from “closed” to “open” state. The modified forms of EWS may interact with Pol II subunits hsRpb5 and hsRpb7. The EWS and EFPs binding partners are described schematically in a model, an attempt to link the transcription with the splicing. The proposed model helps to understand the functional molecular interactions in cancer, to find new partners and ways to treat cancer.
Collapse
|
14
|
Yang HC, Chuang JY, Jeng WY, Liu CI, Wang AHJ, Lu PJ, Chang WC, Hung JJ. Pin1-mediated Sp1 phosphorylation by CDK1 increases Sp1 stability and decreases its DNA-binding activity during mitosis. Nucleic Acids Res 2014; 42:13573-87. [PMID: 25398907 PMCID: PMC4267622 DOI: 10.1093/nar/gku1145] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 10/27/2014] [Accepted: 10/27/2014] [Indexed: 11/15/2022] Open
Abstract
We have shown that Sp1 phosphorylation at Thr739 decreases its DNA-binding activity. In this study, we found that phosphorylation of Sp1 at Thr739 alone is necessary, but not sufficient for the inhibition of its DNA-binding activity during mitosis. We demonstrated that Pin1 could be recruited to the Thr739(p)-Pro motif of Sp1 to modulate the interaction between phospho-Sp1 and CDK1, thereby facilitating CDK1-mediated phosphorylation of Sp1 at Ser720, Thr723 and Thr737 during mitosis. Loss of the C-terminal end of Sp1 (amino acids 741-785) significantly increased Sp1 phosphorylation, implying that the C-terminus inhibits CDK1-mediated Sp1 phosphorylation. Binding analysis of Sp1 peptides to Pin1 by isothermal titration calorimetry indicated that Pin1 interacts with Thr739(p)-Sp1 peptide but not with Thr739-Sp1 peptide. X-ray crystallography data showed that the Thr739(p)-Sp1 peptide occupies the active site of Pin1. Increased Sp1 phosphorylation by CDK1 during mitosis not only stabilized Sp1 levels by decreasing interaction with ubiquitin E3-ligase RNF4 but also caused Sp1 to move out of the chromosomes completely by decreasing its DNA-binding activity, thereby facilitating cell cycle progression. Thus, Pin1-mediated conformational changes in the C-terminal region of Sp1 are critical for increased CDK1-mediated Sp1 phosphorylation to facilitate cell cycle progression during mitosis.
Collapse
Affiliation(s)
- Hang-Che Yang
- Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan
| | - Jian-Ying Chuang
- The PhD Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Wen-Yih Jeng
- Center for Bioscience and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan Core Facilities for Protein Structural Analysis, Academia Sinica, Taipei 115, Taiwan
| | - Chia-I Liu
- Core Facilities for Protein Structural Analysis, Academia Sinica, Taipei 115, Taiwan School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei 110, Taiwan
| | - Andrew H-J Wang
- Core Facilities for Protein Structural Analysis, Academia Sinica, Taipei 115, Taiwan Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Pei-Jung Lu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 70403, Taiwan
| | - Wen-Chang Chang
- Graduate Institute of Medical Sciences, College of Medicine, and Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 110, Taiwan
| | - Jan-Jong Hung
- Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei 110, Taiwan Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan Center for Infectious Disease and Signal Transduction Research, National Cheng Kung University, Tainan 701, Taiwan
| |
Collapse
|
15
|
Abstract
Members of the FET protein family, consisting of FUS, EWSR1, and TAF15, bind to RNA and contribute to the control of transcription, RNA processing, and the cytoplasmic fates of messenger RNAs in metazoa. FET proteins can also bind DNA, which may be important in transcription and DNA damage responses. FET proteins are of medical interest because chromosomal rearrangements of their genes promote various sarcomas and because point mutations in FUS or TAF15 can cause neurodegenerative diseases such as amyotrophic lateral sclerosis and frontotemporal lobar dementia. Recent results suggest that both the normal and pathological effects of FET proteins are modulated by low-complexity or prion-like domains, which can form higher-order assemblies with novel interaction properties. Herein, we review FET proteins with an emphasis on how the biochemical properties of FET proteins may relate to their biological functions and to pathogenesis.
Collapse
Affiliation(s)
- Jacob C Schwartz
- Howard Hughes Medical Institute, Department of Chemistry and Biochemistry, and BioFrontiers Institute, University of Colorado, Boulder, Colorado 80309; , ,
| | | | | |
Collapse
|
16
|
Campos-Melo D, Droppelmann CA, Volkening K, Strong MJ. RNA-binding proteins as molecular links between cancer and neurodegeneration. Biogerontology 2014; 15:587-610. [PMID: 25231915 DOI: 10.1007/s10522-014-9531-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 09/11/2014] [Indexed: 12/12/2022]
Abstract
For many years, epidemiological studies have suggested an association between cancer and neurodegenerative disorders-two disease processes that seemingly have little in common. Although these two disease processes share disruptions in a wide range of cellular pathways, including cell survival, cell death and the cell cycle, the end result is very divergent: uncontrolled cell survival and proliferation in cancer and progressive neuronal cell death in neurodegeneration. Despite the clinical data connecting these two disease processes, little is known about the molecular links between them. Among the mechanisms affected in cancer and neurodegenerative diseases, alterations in RNA metabolism are obtaining significant attention given the critical role for RNA transcription, maturation, transport, stability, degradation and translation in normal cellular function. RNA-binding proteins (RBPs) are integral to each stage of RNA metabolism through their participation in the formation of ribonucleoprotein complexes (RNPs). RBPs have a broad range of functions including posttranscriptional regulation of mRNA stability, splicing, editing and translation, mRNA export and localization, mRNA polyadenylation and miRNA biogenesis, ultimately impacting the expression of every single gene in the cell. In this review, we examine the evidence for RBPs as being key a molecular linkages between cancer and neurodegeneration.
Collapse
Affiliation(s)
- Danae Campos-Melo
- Molecular Medicine Group, Robarts Research Institute, Western University, London, ON, Canada
| | | | | | | |
Collapse
|
17
|
Paronetto M, Bernardis I, Volpe E, Bechara E, Sebestyén E, Eyras E, Valcárcel J. Regulation of FAS Exon Definition and Apoptosis by the Ewing Sarcoma Protein. Cell Rep 2014; 7:1211-26. [DOI: 10.1016/j.celrep.2014.03.077] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 02/16/2014] [Accepted: 03/31/2014] [Indexed: 12/18/2022] Open
|
18
|
Arieti F, Gabus C, Tambalo M, Huet T, Round A, Thore S. The crystal structure of the Split End protein SHARP adds a new layer of complexity to proteins containing RNA recognition motifs. Nucleic Acids Res 2014; 42:6742-52. [PMID: 24748666 PMCID: PMC4041450 DOI: 10.1093/nar/gku277] [Citation(s) in RCA: 32] [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
The Split Ends (SPEN) protein was originally discovered in Drosophila in the late 1990s. Since then, homologous proteins have been identified in eukaryotic species ranging from plants to humans. Every family member contains three predicted RNA recognition motifs (RRMs) in the N-terminal region of the protein. We have determined the crystal structure of the region of the human SPEN homolog that contains these RRMs—the SMRT/HDAC1 Associated Repressor Protein (SHARP), at 2.0 Å resolution. SHARP is a co-regulator of the nuclear receptors. We demonstrate that two of the three RRMs, namely RRM3 and RRM4, interact via a highly conserved interface. Furthermore, we show that the RRM3–RRM4 block is the main platform mediating the stable association with the H12–H13 substructure found in the steroid receptor RNA activator (SRA), a long, non-coding RNA previously shown to play a crucial role in nuclear receptor transcriptional regulation. We determine that SHARP association with SRA relies on both single- and double-stranded RNA sequences. The crystal structure of the SHARP–RRM fragment, together with the associated RNA-binding studies, extend the repertoire of nucleic acid binding properties of RRM domains suggesting a new hypothesis for a better understanding of SPEN protein functions.
Collapse
Affiliation(s)
- Fabiana Arieti
- Department of Molecular Biology, University of Geneva, Geneva 1211, Switzerland
| | - Caroline Gabus
- Department of Molecular Biology, University of Geneva, Geneva 1211, Switzerland
| | - Margherita Tambalo
- Department of Molecular Biology, University of Geneva, Geneva 1211, Switzerland
| | - Tiphaine Huet
- Department of Molecular Biology, University of Geneva, Geneva 1211, Switzerland
| | - Adam Round
- European Molecular Biology Laboratory, Grenoble Outstation and Unit for Virus Host-Cell Interactions, University Grenoble Alpes-EMBL-CNRS, Grenoble 38042, France
| | - Stéphane Thore
- Department of Molecular Biology, University of Geneva, Geneva 1211, Switzerland
| |
Collapse
|
19
|
Todorova R. Disordered binding regions of Ewing’s sarcoma fusion proteins. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2014. [DOI: 10.1134/s1068162014010117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
20
|
Kishore S, Piscuoglio S, Kovac MB, Gylling A, Wenzel F, Trapani F, Altermatt HJ, Mele V, Marra G, Peltomäki P, Terracciano L, Zavolan M, Heinimann K. 3'-UTR poly(T/U) tract deletions and altered expression of EWSR1 are a hallmark of mismatch repair-deficient cancers. Cancer Res 2013; 74:224-34. [PMID: 24158095 DOI: 10.1158/0008-5472.can-13-2100] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The genome-wide accumulation of DNA replication errors known as microsatellite instability (MSI) is the hallmark lesion of DNA mismatch repair (MMR)-deficient cancers. Although testing for MSI is widely used to guide clinical management, the contribution of MSI at distinct genic loci to the phenotype remains largely unexplored. Here, we report that a mononucleotide (T/U)16 tract located in the 3' untranslated region (3'-UTR) of the Ewing sarcoma breakpoint region 1 (EWSR1) gene is a novel MSI target locus that shows perfect sensitivity and specificity in detecting mismatch repair-deficient cancers in two independent populations. We further found a striking relocalization of the EWSR1 protein from nucleus to cytoplasm in MMR-deficient cancers and that the nonprotein-coding MSI target locus itself has a modulatory effect on EWSR1 gene expression through alternative 3' end processing of the EWSR1 gene. Our results point to a MSI target gene-specific effect in MMR-deficient cancers.
Collapse
Affiliation(s)
- Shivendra Kishore
- Authors' Affiliations: Biozentrum, University of Basel; Research Group Human Genetics, Dept of Biomedicine, University of Basel, and Division of Medical Genetics, University Hospital Basel; Institute for Surgical Research and Hospital Management and Department of Biomedicine, University of Basel; Institute of Pathology, University Hospital of Basel, Basel; Pathologie Laenggasse Bern, Bern; Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland; Department of Medical Genetics, Haartman Institute, University of Helsinki, Helsinki, Finland; and Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Ewing sarcoma protein: a key player in human cancer. Int J Cell Biol 2013; 2013:642853. [PMID: 24082883 PMCID: PMC3776376 DOI: 10.1155/2013/642853] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 07/26/2013] [Indexed: 01/04/2023] Open
Abstract
The Ewing sarcoma protein (EWS) is a well-known player in cancer biology for the specific translocations occurring in sarcomas. The EWS-FLI1 gene fusion is the prototypical translocation that encodes the aberrant, chimeric transcription factor, which is a landmark of Ewing tumors. In all described Ewing sarcoma oncogenes, the EWS RNA binding domains are completely missing; thus RNA binding properties are not retained in the hybrid proteins. However, it is currently unknown whether the absence of EWS function in RNA metabolism plays a role in oncogenic transformation or if EWS plays a role by itself in cancer development besides its contribution to the translocation. In this regard, recent reports have highlighted an essential role for EWS in the regulation of DNA damage response (DDR), a process that counteracts genome stability and is often deregulated in cancer cells. The first part of this review will describe the structural features of EWS and its multiple roles in the regulation of gene expression, which are exerted by coordinating different steps in the synthesis and processing of pre-mRNAs. The second part will examine the role of EWS in the regulation of DDR- and cancer-related genes, with potential implications in cancer therapies. Finally, recent advances on the involvement of EWS in neuromuscular disorders will be discussed. Collectively, the information reviewed herein highlights the broad role of EWS in bridging different cellular processes and underlines the contribution of EWS to genome stability and proper cell-cycle progression in higher eukaryotic cells.
Collapse
|
22
|
Dormann D, Haass C. Fused in sarcoma (FUS): an oncogene goes awry in neurodegeneration. Mol Cell Neurosci 2013; 56:475-86. [PMID: 23557964 DOI: 10.1016/j.mcn.2013.03.006] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 03/21/2013] [Accepted: 03/22/2013] [Indexed: 12/13/2022] Open
Abstract
Fused in sarcoma (FUS) is a nuclear DNA/RNA binding protein that regulates different steps of gene expression, including transcription, splicing and mRNA transport. FUS has been implicated in neurodegeneration, since mutations in FUS cause familial amyotrophic lateral sclerosis (ALS-FUS) and lead to the cytosolic deposition of FUS in the brain and spinal cord of ALS-FUS patients. Moreover, FUS and two related proteins of the same protein family (FET family) are co-deposited in cytoplasmic inclusions in a subset of patients with frontotemporal lobar degeneration (FTLD-FUS). Cytosolic deposition of these otherwise nuclear proteins most likely causes the loss of a yet unknown essential nuclear function and/or the gain of a toxic function in the cytosol. Here we summarize what is known about the physiological functions of the FET proteins in the nucleus and cytoplasm and review the distinctive pathomechanisms that lead to the deposition of only FUS in ALS-FUS, but all three FET proteins in FTLD-FUS. We suggest that ALS-FUS is caused by a selective dysfunction of FUS, while FTLD-FUS may be caused by a dysfunction of the entire FET family. This article is part of a Special Issue entitled 'RNA and splicing regulation in neurodegeneration'.
Collapse
Affiliation(s)
- Dorothee Dormann
- Adolf-Butenandt-Institute, Biochemistry, Ludwig-Maximilians-University, Schillerstr. 44, Munich 80336, Germany.
| | | |
Collapse
|
23
|
Abstract
The type I interferon system genes IKBKE and IFIH1 are associated with the risk of systemic lupus erythematosus (SLE). To identify the sequence variants that are able to account for the disease association, we resequenced the genes IKBKE and IFIH1. Eighty-six single-nucleotide variants (SNVs) with potentially functional effect or differences in allele frequencies between patients and controls determined by sequencing were further genotyped in 1140 SLE patients and 2060 controls. In addition, 108 imputed sequence variants in IKBKE and IFIH1 were included in the association analysis. Ten IKBKE SNVs and three IFIH1 SNVs were associated with SLE. The SNVs rs1539241 and rs12142086 tagged two independent association signals in IKBKE, and the haplotype carrying their risk alleles showed an odds ratio of 1.68 (P-value=1.0 × 10(-5)). The risk allele of rs12142086 affects the binding of splicing factor 1 in vitro and could thus influence its transcriptional regulatory function. Two independent association signals were also detected in IFIH1, which were tagged by a low-frequency SNV rs78456138 and a missense SNV rs3747517. Their joint effect is protective against SLE (odds ratio=0.56; P-value=6.6 × 10(-3)). In conclusion, we have identified new SLE-associated sequence variants in IKBKE and IFIH1, and proposed functional hypotheses for the association signals.
Collapse
|
24
|
Cattaruzza M, Nogoy N, Wojtowicz A, Hecker M. Zinc finger motif‐1 antagonizes PDGF‐BB‐induced growth and dediffer‐entiation of vascular smooth muscle cells. FASEB J 2012; 26:4864-75. [DOI: 10.1096/fj.12-210302] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Marco Cattaruzza
- Institute of Physiology and PathophysiologyDivision of Cardiovascular PhysiologyUniversity of HeidelbergGermany
| | - Nicole Nogoy
- Institute of Physiology and PathophysiologyDivision of Cardiovascular PhysiologyUniversity of HeidelbergGermany
| | - Agnieszka Wojtowicz
- Institute of Physiology and PathophysiologyDivision of Cardiovascular PhysiologyUniversity of HeidelbergGermany
| | - Markus Hecker
- Institute of Physiology and PathophysiologyDivision of Cardiovascular PhysiologyUniversity of HeidelbergGermany
| |
Collapse
|
25
|
Lim B, Jun HJ, Kim AY, Kim S, Choi J, Kim J. The TFG-TEC fusion gene created by the t(3;9) translocation in human extraskeletal myxoid chondrosarcomas encodes a more potent transcriptional activator than TEC. Carcinogenesis 2012; 33:1450-8. [PMID: 22581839 DOI: 10.1093/carcin/bgs164] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The t(3;9)(q11-q12;q22) translocation associated with human extraskeletal myxoid chondrosarcomas results in a chimeric molecule in which the N-terminal domain (NTD) of the TFG (TRK-fused gene) is fused to the TEC (Translocated in Extraskeletal Chondrosarcoma) gene. Little is known about the biological function of TFG-TEC. Because the NTDs of TFG-TEC and TEC are structurally different, and the TFG itself is a cytoplasmic protein, the functional consequences of this fusion in extraskeletal myxoid chondrosarcomas were examined. The results showed that the chimeric gene encoded a nuclear protein that bound DNA with the same sequence specificity as the parental TEC protein. Comparison of the transactivation properties of TFG-TEC and TEC indicated that the former has higher transactivation activity for a known target reporter containing TEC-binding sites. Additional reporter assays for TFG (NTD) showed that the TGF (NTD) of TFG-TEC induced a 12-fold increase in the activation of luciferase from a reporter plasmid containing GAL4 binding sites when fused to the DNA-binding domain of GAL4, indicating that the TFG (NTD) of the TFG-TEC protein has intrinsic transcriptional activation properties. Finally, deletion analysis of the functional domains of TFG (NTD) indicated that the PB1 (Phox and Bem1p) and SPYGQ-rich region of TFG (NTD) were capable of activating transcription and that full integrity of TFG (NTD) was necessary for full transactivation. These results suggest that the oncogenic effect of the t(3;9) translocation may be due to the TFG-TEC chimeric protein and that fusion of the TFG (NTD) to the TEC protein produces a gain-of-function chimeric product.
Collapse
Affiliation(s)
- Bobae Lim
- Laboratory of Molecular and Cellular Biology, Department of Life Science, Sogang University Seoul 121-742, Korea
| | | | | | | | | | | |
Collapse
|
26
|
Huang G, Zhou Z, Wang H, Kleinerman ES. CAPER-α alternative splicing regulates the expression of vascular endothelial growth factor₁₆₅ in Ewing sarcoma cells. Cancer 2011; 118:2106-16. [PMID: 22009261 DOI: 10.1002/cncr.26488] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 06/29/2011] [Accepted: 07/14/2011] [Indexed: 01/13/2023]
Abstract
BACKGROUND TC-71 Ewing sarcoma cells overexpress vascular endothelial growth factor (VEGF) with a shift from the 189 to the 165 isoform. METHODS The effect of CAPER-α on the expression of the VEGF isoforms, tumor growth, and vessel density was analyzed after transfection of TC-71 cells with CAPER-α cDNA or siRNA. RESULTS CAPER-α correlated inversely with the VEGF(165) /VEGF(189) mRNA ratio. Up-regulation of CAPER-α resulted in decreased tumor growth, tumor vessel density, and chemotactic activity of the cell's supernatant. CAPER-α expression was regulated by EWS/FLI-1 through a protein-protein interaction. CONCLUSIONS Increased VEGF(165) expression is secondary to the down-regulation of CAPER-α by EWS/FLI-1. CAPER-α mediates alternative splicing and controls the shift from VEGF(189) to VEGF(165) .
Collapse
Affiliation(s)
- Gangxiong Huang
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | |
Collapse
|
27
|
Paronetto M, Miñana B, Valcárcel J. The Ewing Sarcoma Protein Regulates DNA Damage-Induced Alternative Splicing. Mol Cell 2011; 43:353-68. [DOI: 10.1016/j.molcel.2011.05.035] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Revised: 04/14/2011] [Accepted: 05/25/2011] [Indexed: 01/20/2023]
|
28
|
Gupta A, Kielkopf CL. Purification, crystallization and preliminary X-ray crystallographic analysis of a central domain of human splicing factor 1. Acta Crystallogr Sect F Struct Biol Cryst Commun 2011; 67:486-90. [PMID: 21505248 PMCID: PMC3080157 DOI: 10.1107/s1744309111004623] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Accepted: 02/07/2011] [Indexed: 11/11/2022]
Abstract
Pre-mRNA splicing is an essential source of genetic diversity in eukaryotic organisms. In the early stages of splicing, splicing factor 1 (SF1) recognizes the pre-mRNA splice site as a complex with its partner, U2 auxiliary factor 65 kDa subunit (U2AF(65)). A central `mystery' domain of SF1 (SF1md) lacks detectable homology with known structures, yet is the region of highest phylogenetic sequence conservation among SF1 homologues. Here, steps towards determining the SF1md structure are described. Firstly, SF1md was expressed and purified. The presence of regular secondary structure was verified using circular dichroism spectroscopy and the SF1md protein was then crystallized. A native data set was collected and processed to 2.5 Å resolution. The SF1md crystals belonged to space group C2 and have most probable solvent contents of 64, 52 or 39% with three, four or five molecules per asymmetric unit, respectively. Mutually perpendicular peaks on the κ = 180° section of the self-rotation function support the presence of four molecules in the asymmetric unit.
Collapse
Affiliation(s)
- Ankit Gupta
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Clara L. Kielkopf
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| |
Collapse
|
29
|
Dr. Jekyll and Mr. Hyde: The Two Faces of the FUS/EWS/TAF15 Protein Family. Sarcoma 2010; 2011:837474. [PMID: 21197473 PMCID: PMC3005952 DOI: 10.1155/2011/837474] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 10/20/2010] [Accepted: 11/01/2010] [Indexed: 12/13/2022] Open
Abstract
FUS, EWS, and TAF15 form the FET family of RNA-binding proteins whose genes are found rearranged with various transcription factor genes predominantly in sarcomas and in rare hematopoietic and epithelial cancers. The resulting fusion gene products have attracted considerable interest as diagnostic and promising therapeutic targets. So far, oncogenic FET fusion proteins have been regarded as strong transcription factors that aberrantly activate or repress target genes of their DNA-binding fusion partners. However, the role of the transactivating domain in the context of the normal FET proteins is poorly defined, and, therefore, our knowledge on how FET aberrations impact on tumor biology is incomplete. Since we believe that a full understanding of aberrant FET protein function can only arise from looking at both sides of the coin, the good and the evil, this paper summarizes evidence for the central function of FET proteins in bridging RNA transcription, processing, transport, and DNA repair.
Collapse
|
30
|
Pastuszak AW, Joachimiak MP, Blanchette M, Rio DC, Brenner SE, Frankel AD. An SF1 affinity model to identify branch point sequences in human introns. Nucleic Acids Res 2010; 39:2344-56. [PMID: 21071404 PMCID: PMC3064769 DOI: 10.1093/nar/gkq1046] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Splicing factor 1 (SF1) binds to the branch point sequence (BPS) of mammalian introns and is believed to be important for the splicing of some, but not all, introns. To help identify BPSs, particularly those that depend on SF1, we generated a BPS profile model in which SF1 binding affinity data, validated by branch point mapping, were iteratively incorporated into computational models. We searched a data set of 117 499 human introns for best matches to the SF1 Affinity Model above a threshold, and counted the number of matches at each intronic position. After subtracting a background value, we found that 87.9% of remaining high-scoring matches identified were located in a region upstream of 3′-splice sites where BPSs are typically found. Since U2AF65 recognizes the polypyrimidine tract (PPT) and forms a cooperative RNA complex with SF1, we combined the SF1 model with a PPT model computed from high affinity binding sequences for U2AF65. The combined model, together with binding site location constraints, accurately identified introns bound by SF1 that are candidates for SF1-dependent splicing.
Collapse
Affiliation(s)
- Alexander W Pastuszak
- Department of Biochemistry and Biophysics, University of California, San Francisco, USA
| | | | | | | | | | | |
Collapse
|
31
|
Corioni M, Antih N, Tanackovic G, Zavolan M, Krämer A. Analysis of in situ pre-mRNA targets of human splicing factor SF1 reveals a function in alternative splicing. Nucleic Acids Res 2010; 39:1868-79. [PMID: 21062807 PMCID: PMC3061054 DOI: 10.1093/nar/gkq1042] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The conserved pre-mRNA splicing factor SF1 is implicated in 3' splice site recognition by binding directly to the intron branch site. However, because SF1 is not essential for constitutive splicing, its role in pre-mRNA processing has remained mysterious. Here, we used crosslinking and immunoprecipitation (CLIP) to analyze short RNAs directly bound by human SF1 in vivo. SF1 bound mainly pre-mRNAs, with 77% of target sites in introns. Binding to target RNAs in vitro was dependent on the newly defined SF1 binding motif ACUNAC, strongly resembling human branch sites. Surprisingly, the majority of SF1 binding sites did not map to the expected position near 3' splice sites. Instead, target sites were distributed throughout introns, and a smaller but significant fraction occurred in exons within coding and untranslated regions. These data suggest a more complex role for SF1 in splicing regulation. Indeed, SF1 silencing affected alternative splicing of endogenous transcripts, establishing a previously unexpected role for SF1 and branch site-like sequences in splice site selection.
Collapse
Affiliation(s)
- Margherita Corioni
- Department of Cell Biology, Faculty of Sciences, University of Geneva, 30 quai Ernest-Ansermet, CH-1211 Geneva
| | | | | | | | | |
Collapse
|
32
|
Romeo S, Dei Tos AP. Soft tissue tumors associated with EWSR1 translocation. Virchows Arch 2010; 456:219-34. [PMID: 19936782 DOI: 10.1007/s00428-009-0854-3] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Revised: 10/07/2009] [Accepted: 10/17/2009] [Indexed: 01/26/2023]
Abstract
The Ewing sarcoma breakpoint region 1 (EWSR1; also known as EWS) represents one of the most commonly involved genes in sarcoma translocations. In fact, it is involved in a broad variety of mesenchymal lesions which includes Ewing's sarcoma/peripheral neuroectodermal tumor, desmoplastic small round cell tumor,clear cell sarcoma, angiomatoid fibrous histiocytoma, extraskeletal myxoid chondrosarcoma, and a subset of myxoid liposarcoma. The fusion products between EWSR1 and partners usually results in fusion of the N-terminal transcription-activating domain of EWSR1 and the C-terminal DNA-binding domain of the fusion partner, eventually generating novel transcription factors. EWSR1 rearrangement can be visualized by the means of fluorescence in situ hybridization (FISH). As soft tissue sarcomas represent a diagnostically challenging group, FISH analysis is an extremely useful confirmatory diagnostic tool. However, as in most instances a split-apart approach is used, the results of molecular genetics must be evaluated in context with morphology.
Collapse
Affiliation(s)
- Salvatore Romeo
- Department of Pathology, General Hospital of Treviso, Piazza Ospedale 1, Treviso, Italy
| | | |
Collapse
|
33
|
Pahlich S, Quero L, Roschitzki B, Leemann-Zakaryan RP, Gehring H. Analysis of Ewing sarcoma (EWS)-binding proteins: interaction with hnRNP M, U, and RNA-helicases p68/72 within protein-RNA complexes. J Proteome Res 2010; 8:4455-65. [PMID: 19673543 DOI: 10.1021/pr900235t] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The human Ewing Sarcoma (EWS) protein belongs to the TET family of RNA-binding proteins and consists of an N-terminal transcriptional activation domain (EAD) and a C-terminal RNA-binding domain (RBD), which is extensively methylated at arginine residues. This multifunctional protein acts in transcriptional co-activation, DNA-recombination, -pairing and -repair, in splicing, and mRNA transport. The role of arginine methylation in these processes as well as the time and place of methylation within cells is still unclear. In this study, we show that methylation of recombinant EWS protein in HEK cells occurs immediately after or even during translation. Pull-down experiments with recombinant EWS protein as bait, followed by mass spectrometric analysis identified more than 30 interacting proteins independent of whether the EWS protein was methylated or not. The EWS protein interacts via its RBD with RNase-sensitive protein complexes consisting of mainly heterogeneous nuclear ribonucleoproteins (hnRNPs) and RNA helicases. HnRNP M and U, the RNA-helicases p68 and p72, but also actin and tubulin were found to interact directly with the EWS protein. Co-precipitation experiments with recombinant proteins confirmed the interaction of the EWS protein with p68 via its RBD. Colocalization of the EWS protein and the RNA-helicases in the nucleus of HEK cells was visualized by expressing labeled EWS protein and p68 or p72. When co-expressed, the labeled proteins relocated from the nucleoplasm to nucleolar capping structures. As arginine methylation within the RBD of the EWS protein are neither needed for its subcellular localization nor for its protein-protein interaction, a role of EWS protein methylation in RNA-binding and affecting the activation/repression activity or even in the stabilization of the EWS protein seems very likely.
Collapse
Affiliation(s)
- Steffen Pahlich
- Department of Biochemistry, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | | | | | | | | |
Collapse
|
34
|
Rabin SJ, Kim JMH, Baughn M, Libby RT, Kim YJ, Fan Y, Libby RT, La Spada A, Stone B, Ravits J. Sporadic ALS has compartment-specific aberrant exon splicing and altered cell-matrix adhesion biology. Hum Mol Genet 2009; 19:313-28. [PMID: 19864493 PMCID: PMC2796893 DOI: 10.1093/hmg/ddp498] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by progressive weakness from loss of motor neurons. The fundamental pathogenic mechanisms are unknown and recent evidence is implicating a significant role for abnormal exon splicing and RNA processing. Using new comprehensive genomic technologies, we studied exon splicing directly in 12 sporadic ALS and 10 control lumbar spinal cords acquired by a rapid autopsy system that processed nervous systems specifically for genomic studies. ALS patients had rostral onset and caudally advancing disease and abundant residual motor neurons in this region. We created two RNA pools, one from motor neurons collected by laser capture microdissection and one from the surrounding anterior horns. From each, we isolated RNA, amplified mRNA, profiled whole-genome exon splicing, and applied advanced bioinformatics. We employed rigorous quality control measures at all steps and validated findings by qPCR. In the motor neuron enriched mRNA pool, we found two distinct cohorts of mRNA signals, most of which were up-regulated: 148 differentially expressed genes (P ≤ 10−3) and 411 aberrantly spliced genes (P ≤ 10−5). The aberrantly spliced genes were highly enriched in cell adhesion (P ≤ 10−57), especially cell–matrix as opposed to cell–cell adhesion. Most of the enriching genes encode transmembrane or secreted as opposed to nuclear or cytoplasmic proteins. The differentially expressed genes were not biologically enriched. In the anterior horn enriched mRNA pool, we could not clearly identify mRNA signals or biological enrichment. These findings, perturbed and up-regulated cell–matrix adhesion, suggest possible mechanisms for the contiguously progressive nature of motor neuron degeneration. Data deposition: GeneChip raw data (CEL-files) have been deposited for public access in the Gene Expression Omnibus (GEO), www.ncbi.nlm.nih.gov/geo, accession number GSE18920.
Collapse
Affiliation(s)
- Stuart J Rabin
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Tan AY, Manley JL. The TET family of proteins: functions and roles in disease. J Mol Cell Biol 2009; 1:82-92. [PMID: 19783543 DOI: 10.1093/jmcb/mjp025] [Citation(s) in RCA: 204] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Translocated in liposarcoma, Ewing's sarcoma and TATA-binding protein-associated factor 15 constitute an interesting and important family of proteins known as the TET proteins. The proteins function in several aspects of cell growth control, including multiple different steps in gene expression, and they are also found mutated in a number of specific diseases. For example, all contain domains for binding nucleic acids and have been shown to function in both RNA polymerase II-mediated transcription and pre-mRNA splicing, possibly connecting these two processes. Chromosomal translocations in human sarcomas result in a fusion of the amino terminus of these proteins, which contains a transcription activation domain, to the DNA-binding domain of a transcription factor. Although the fusion proteins have been characterized in a clinical environment, the function of the cognate full-length protein in normal cells is a more recent topic of study. The first part of this review will describe the TET proteins, followed by detailed descriptions of their multiple roles in cells. The final sections will examine changes that occur in gene regulation in cells expressing the fusion proteins. The clinical implications and treatment of sarcomas will not be addressed but have recently been reviewed.
Collapse
Affiliation(s)
- Adelene Y Tan
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | | |
Collapse
|
36
|
Gene activation by dissociation of an inhibitor from a transcriptional activation domain. Mol Cell Biol 2009; 29:5604-10. [PMID: 19651897 DOI: 10.1128/mcb.00632-09] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Gal4 is a prototypical eukaryotic transcriptional activator whose recruitment function is inhibited in the absence of galactose by the Gal80 protein through masking of its transcriptional activation domain (AD). A long-standing nondissociation model posits that galactose-activated Gal3 interacts with Gal4-bound Gal80 at the promoter, yielding a tripartite Gal3-Gal80-Gal4 complex with altered Gal80-Gal4 conformation to enable Gal4 AD activity. Some recent data challenge this model, whereas other recent data support the model. To address this controversy, we imaged fluorescent-protein-tagged Gal80, Gal4, and Gal3 in live cells containing a novel GAL gene array. We find that Gal80 rapidly dissociates from Gal4 in response to galactose. Importantly, this dissociation is Gal3 dependent and concurrent with Gal4-activated GAL gene expression. When galactose-triggered dissociation is followed by galactose depletion, preexisting Gal80 reassociates with Gal4, indicating that sequestration of Gal80 by Gal3 contributes to the observed Gal80-Gal4 dissociation. Moreover, the ratio of nuclear Gal80 to cytoplasmic Gal80 decreases in response to Gal80-Gal3 interaction. Taken together, these and other results provide strong support for a GAL gene switch model wherein Gal80 rapidly dissociates from Gal4 through a mechanism that involves sequestration of Gal80 by galactose-activated Gal3.
Collapse
|
37
|
Makino H, Toyoda M, Matsumoto K, Saito H, Nishino K, Fukawatase Y, Machida M, Akutsu H, Uyama T, Miyagawa Y, Okita H, Kiyokawa N, Fujino T, Ishikawa Y, Nakamura T, Umezawa A. Mesenchymal to embryonic incomplete transition of human cells by chimeric OCT4/3 (POU5F1) with physiological co-activator EWS. Exp Cell Res 2009; 315:2727-40. [PMID: 19559696 DOI: 10.1016/j.yexcr.2009.06.016] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Revised: 06/15/2009] [Accepted: 06/16/2009] [Indexed: 11/30/2022]
Abstract
POU5F1 (more commonly known as OCT4/3) is one of the stem cell markers, and affects direction of differentiation in embryonic stem cells. To investigate whether cells of mesenchymal origin acquire embryonic phenotypes, we generated human cells of mesodermal origin with overexpression of the chimeric OCT4/3 gene with physiological co-activator EWS (product of the EWSR1 gene), which is driven by the potent EWS promoter by translocation. The cells expressed embryonic stem cell genes such as NANOG, lost mesenchymal phenotypes, and exhibited embryonal stem cell-like alveolar structures when implanted into the subcutaneous tissue of immunodeficient mice. Hierarchical analysis by microchip analysis and cell surface analysis revealed that the cells are subcategorized into the group of human embryonic stem cells and embryonal carcinoma cells. These results imply that cells of mesenchymal origin can be traced back to cells of embryonic phenotype by the OCT4/3 gene in collaboration with the potent cis-regulatory element and the fused co-activator. The cells generated in this study with overexpression of chimeric OCT4/3 provide us with insight into cell plasticity involving OCT4/3 that is essential for embryonic cell maintenance, and the complexity required for changing cellular identity.
Collapse
Affiliation(s)
- Hatsune Makino
- Department of Reproductive Biology, National Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Embree LJ, Azuma M, Hickstein DD. Ewing sarcoma fusion protein EWSR1/FLI1 interacts with EWSR1 leading to mitotic defects in zebrafish embryos and human cell lines. Cancer Res 2009; 69:4363-71. [PMID: 19417137 DOI: 10.1158/0008-5472.can-08-3229] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The mechanism whereby the fusion of EWSR1 with the ETS transcription factor FLI1 contributes to malignant transformation in Ewing sarcoma remains unclear. We show that injection of human or zebrafish EWSR1/FLI1 mRNA into developing zebrafish embryos leads to mitotic defects with multipolar and disorganized mitotic spindles. Expression of human EWSR1/FLI1 in HeLa cells also results in mitotic defects, along with mislocalization of Aurora kinase B, a key regulator of mitotic progression. Because these mitotic abnormalities mimic those observed with the knockdown of EWSR1 in zebrafish embryos and HeLa cells, we investigated whether EWSR1/FLI1 interacts with EWSR1 and interferes with its function. EWSR1 coimmunoprecipitates with EWSR1/FLI1, and overexpression of EWSR1 rescues the mitotic defects in EWSR1/FLI1-transfected HeLa cells. This interaction between EWSR1/FLI1 and EWSR1 in Ewing sarcoma may induce mitotic defects leading to genomic instability and subsequent malignant transformation.
Collapse
Affiliation(s)
- Lisa J Embree
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
39
|
Phosphorylation of Ewing's sarcoma protein (EWS) and EWS-Fli1 in response to DNA damage. Biochem J 2009; 418:625-34. [PMID: 19076070 DOI: 10.1042/bj20082097] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In Ewing's sarcomas, chromosomal translocations cause the N-terminal domain of the EWS (Ewing's sarcoma protein) to fuse with the DNA-binding domains of the Ets (E26 transformation-specific) family of transcription factors. Here we show that EWS and EWS-Fli1 (Friend leukaemia virus integration 1), the fusion most frequently found in Ewing's sarcomas, become phosphorylated at Thr(79) in response to either mitogens or DNA-damaging agents. The much weaker mitogen-induced phosphorylation of EWS is catalysed by the MAPKs (mitogen-activated protein kinases) ERK1 (extracellular signal-regulated kinase 1) and ERK2, whereas the much stronger phosphorylation of EWS induced by the DNA alkylating agent MMS (methyl methanesulphonate) can be catalysed by JNK (c-Jun N-terminal kinase) and at least one other protein kinase distinct from ERK1/ERK2. In contrast, the phosphorylation of EWS-Fli1 induced by MMS was largely mediated by p38alpha/p38beta MAPKs. MMS induced a much stronger phosphorylation of EWS-Fli1 than EWS in heterodimers comprising both proteins.
Collapse
|
40
|
TFIP11, CCNL1 and EWSR1 Protein-protein Interactions, and Their Nuclear Localization. Int J Mol Sci 2008; 9:1504-1514. [PMID: 19122807 PMCID: PMC2605624 DOI: 10.3390/ijms9081504] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Revised: 08/14/2008] [Accepted: 08/15/2008] [Indexed: 02/06/2023] Open
Abstract
Previous studies using the yeast two-hybrid assay (Y2H) have identified cyclin L1 (CCNL1) and Ewing sarcoma breakpoint region 1 protein (EWSR1) as being interacting partners of tuftelin-interacting protein 11 (TFIP11). All three proteins are functionally related to the spliceosome and involved in pre-mRNA splicing activities. The spliceosome is a dynamic ribonucleoprotein complex responsible for pre-mRNA splicing of intronic regions, and is composed of five small nuclear RNAs (snRNAs) and μ140 proteins. TFIP11 appears to play a role in spliceosome disassembly allowing for the release of the bound lariat-intron. The roles of CCNL1 and EWSR1 in the spliceosome are poorly understood. Using fluorescently-tagged proteins and confocal microscopy we show that TFIP11, CCNL1 and EWSR1 frequently co-localize to speckled nuclear domains. These data would suggest that all three proteins participate in a common cellular activity related to RNA splicing events.
Collapse
|
41
|
Cironi L, Riggi N, Provero P, Wolf N, Suvà ML, Suvà D, Kindler V, Stamenkovic I. IGF1 is a common target gene of Ewing's sarcoma fusion proteins in mesenchymal progenitor cells. PLoS One 2008; 3:e2634. [PMID: 18648544 PMCID: PMC2481291 DOI: 10.1371/journal.pone.0002634] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2008] [Accepted: 06/04/2008] [Indexed: 11/18/2022] Open
Abstract
Background The EWS-FLI-1 fusion protein is associated with 85–90% of Ewing's sarcoma family tumors (ESFT), the remaining 10–15% of cases expressing chimeric genes encoding EWS or FUS fused to one of several ets transcription factor family members, including ERG-1, FEV, ETV1 and ETV6. ESFT are dependent on insulin-like growth factor-1 (IGF-1) for growth and survival and recent evidence suggests that mesenchymal progenitor/stem cells constitute a candidate ESFT origin. Methodology/Principal Findings To address the functional relatedness between ESFT-associated fusion proteins, we compared mouse progenitor cell (MPC) permissiveness for EWS-FLI-1, EWS-ERG and FUS-ERG expression and assessed the corresponding expression profile changes. Whereas all MPC isolates tested could stably express EWS-FLI-1, only some sustained stable EWS-ERG expression and none could express FUS-ERG for more than 3–5 days. Only 14% and 4% of the total number of genes that were respectively induced and repressed in MPCs by the three fusion proteins were shared. However, all three fusion proteins, but neither FLI-1 nor ERG-1 alone, activated the IGF1 promoter and induced IGF1 expression. Conclusion/Significance Whereas expression of different ESFT-associated fusion proteins may require distinct cellular microenvironments and induce transcriptome changes of limited similarity, IGF1 induction may provide one common mechanism for their implication in ESFT pathogenesis.
Collapse
Affiliation(s)
- Luisa Cironi
- Division of Experimental Pathology, Institute of Pathology CHUV, University of Lausanne, Lausanne, Switzerland
| | - Nicolò Riggi
- Division of Experimental Pathology, Institute of Pathology CHUV, University of Lausanne, Lausanne, Switzerland
| | - Paolo Provero
- Department of Biology Genetics and Biochemistry, University of Turin, Turin, Italy
| | - Natalie Wolf
- Division of Experimental Pathology, Institute of Pathology CHUV, University of Lausanne, Lausanne, Switzerland
| | - Mario-Luca Suvà
- Division of Experimental Pathology, Institute of Pathology CHUV, University of Lausanne, Lausanne, Switzerland
| | - Domizio Suvà
- Department of Orthopedics, University Hospital, University of Geneva, Geneva, Switzerland
| | - Vincent Kindler
- Department of Orthopedics, University Hospital, University of Geneva, Geneva, Switzerland
| | - Ivan Stamenkovic
- Division of Experimental Pathology, Institute of Pathology CHUV, University of Lausanne, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
42
|
Hume DA, Sasmono T, Himes SR, Sharma SM, Bronisz A, Constantin M, Ostrowski MC, Ross IL. The Ewing Sarcoma Protein (EWS) Binds Directly to the Proximal Elements of the Macrophage-Specific Promoter of the CSF-1 Receptor (csf1r) Gene. THE JOURNAL OF IMMUNOLOGY 2008; 180:6733-42. [DOI: 10.4049/jimmunol.180.10.6733] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
43
|
Search for cellular partners of human papillomavirus type 16 E2 protein. Arch Virol 2008; 153:983-90. [PMID: 18305892 DOI: 10.1007/s00705-008-0061-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2007] [Accepted: 01/25/2008] [Indexed: 12/14/2022]
Abstract
Human papillomaviruses (HPVs) are small, double-stranded DNA viruses that infect cutaneous and mucosal epithelia. Type 16 (HPV16) displays tropism to genital epithelia, giving rise to genital warts and cervical intraepithelial neoplasia (CIN), which is a precursor lesion to invasive carcinoma of the cervix. The great majority of human cervical cancers contain integrated HPV DNA where the E2 gene is usually disrupted, suggesting that the loss of the E2 protein is an important step in HPV-induced carcinogenesis. The HPV16 E2 protein is a regulatory protein that seems to be essential for creating favourable conditions for establishment of infection and proper completion of the viral life cycle. Recently, diverse activities of the E2 proteins have been described, but the molecular basis of these processes has not beenfully elucidated. Using a yeast two-hybrid system, we have identified epithelial cellular proteins that bind to the E2 protein of HPV16.
Collapse
|
44
|
Riggi N, Cironi L, Suvà ML, Stamenkovic I. Sarcomas: genetics, signalling, and cellular origins. Part 1: The fellowship of TET. J Pathol 2007; 213:4-20. [PMID: 17691072 DOI: 10.1002/path.2209] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Sarcomas comprise some of the most aggressive solid tumours that, for the most part, respond poorly to chemo- and radiation therapy and are associated with a sombre prognosis when surgical removal cannot be performed or is incomplete. Partly because of their lower frequency, sarcomas have not been studied as intensively as carcinomas and haematopoietic malignancies, and the molecular mechanisms that underlie their pathogenesis are only beginning to be understood. Even more enigmatic is the identity of the primary cells from which these tumours originate. Over the past 25 years, however, several non-random chromosomal translocations have been found to be associated with defined sarcomas. Each of these translocations generates a fusion gene believed to be directly related to the pathogenesis of the sarcoma in which it is expressed. The corresponding fusion proteins provide a unique tool not only to study the process of sarcoma development, but also to identify cells that are permissive for their putative oncogenic properties. This is the first of two reviews that cover the mechanisms whereby specific fusion/mutant gene products participate in sarcoma development and the cellular context that may provide the necessary permissiveness for their expression and oncogenicity. Part 1 of the review focuses on sarcomas that express fusion genes containing TET gene family products, including EWSR1, TLS/FUS, and TAFII68. Part 2 (J Pathol 2007; DOI: 10.1002/path.2008) summarizes our current understanding of the genetic and cellular origins of sarcomas expressing fusion genes exclusive of TET family members; it also covers soft tissue malignancies harbouring specific mutations in RTK-encoding genes, the prototype of which are gastrointestinal stromal tumours (GIST).
Collapse
Affiliation(s)
- N Riggi
- Division of Experimental Pathology, Institute of Pathology, University of Lausanne, Lausanne, Switzerland
| | | | | | | |
Collapse
|
45
|
Li H, Watford W, Li C, Parmelee A, Bryant MA, Deng C, O’Shea J, Lee SB. Ewing sarcoma gene EWS is essential for meiosis and B lymphocyte development. J Clin Invest 2007; 117:1314-23. [PMID: 17415412 PMCID: PMC1838927 DOI: 10.1172/jci31222] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2006] [Accepted: 02/06/2007] [Indexed: 01/08/2023] Open
Abstract
Ewing sarcoma gene EWS encodes a putative RNA-binding protein with proposed roles in transcription and splicing, but its physiological role in vivo remains undefined. Here, we have generated Ews-deficient mice and demonstrated that EWS is required for the completion of B cell development and meiosis. Analysis of Ews(-/-) lymphocytes revealed a cell-autonomous defect in precursor B lymphocyte (pre-B lymphocyte) development. During meiosis, Ews-null spermatocytes were deficient in XY bivalent formation and showed reduced meiotic recombination, resulting in massive apoptosis and complete arrest in gamete maturation. Inactivation of Ews in mouse embryonic fibroblasts resulted in premature cellular senescence, and the mutant animals showed hypersensitivity to ionizing radiation. Finally, we showed that EWS interacts with lamin A/C and that loss of EWS results in a reduced lamin A/C expression. Our findings reveal essential functions for EWS in pre-B cell development and meiosis, with proposed roles in DNA pairing and recombination/repair mechanisms. Furthermore, we demonstrate a novel role of EWS in cellular senescence, possibly through its interaction and modulation of lamin A/C.
Collapse
Affiliation(s)
- Hongjie Li
- Genetics of Development and Disease Branch, NIDDK,
Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, and
Pathology Section, Diagnostic and Research Services Branch, Division of Veterinary Resources, and Office of Research Services, NIH, Bethesda, Maryland, USA
| | - Wendy Watford
- Genetics of Development and Disease Branch, NIDDK,
Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, and
Pathology Section, Diagnostic and Research Services Branch, Division of Veterinary Resources, and Office of Research Services, NIH, Bethesda, Maryland, USA
| | - Cuiling Li
- Genetics of Development and Disease Branch, NIDDK,
Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, and
Pathology Section, Diagnostic and Research Services Branch, Division of Veterinary Resources, and Office of Research Services, NIH, Bethesda, Maryland, USA
| | - Alissa Parmelee
- Genetics of Development and Disease Branch, NIDDK,
Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, and
Pathology Section, Diagnostic and Research Services Branch, Division of Veterinary Resources, and Office of Research Services, NIH, Bethesda, Maryland, USA
| | - Mark A. Bryant
- Genetics of Development and Disease Branch, NIDDK,
Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, and
Pathology Section, Diagnostic and Research Services Branch, Division of Veterinary Resources, and Office of Research Services, NIH, Bethesda, Maryland, USA
| | - Chuxia Deng
- Genetics of Development and Disease Branch, NIDDK,
Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, and
Pathology Section, Diagnostic and Research Services Branch, Division of Veterinary Resources, and Office of Research Services, NIH, Bethesda, Maryland, USA
| | - John O’Shea
- Genetics of Development and Disease Branch, NIDDK,
Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, and
Pathology Section, Diagnostic and Research Services Branch, Division of Veterinary Resources, and Office of Research Services, NIH, Bethesda, Maryland, USA
| | - Sean Bong Lee
- Genetics of Development and Disease Branch, NIDDK,
Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, and
Pathology Section, Diagnostic and Research Services Branch, Division of Veterinary Resources, and Office of Research Services, NIH, Bethesda, Maryland, USA
| |
Collapse
|
46
|
Neduva V, Russell RB. Proline-rich regions in transcriptional complexes: heading in many directions. ACTA ACUST UNITED AC 2007; 2007:pe1. [PMID: 17228057 DOI: 10.1126/stke.3692007pe1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Although many protein complexes are now known, the precise details of how each component operates are rarely understood. Through a combination of bioinformatics and analysis of gene-trapped mouse clones, Enkhmandakh et al. were able to deduce the modular function for a part of Ssdp1, a crucial component of the Ldb1 transcriptional complex, which plays a central role in mammalian head development. A proline-rich module from Ssdp1 is likely responsible for transactivation, and this region is curiously mobile, occurring in different proteins in different species. The results underscore the importance of protein modularity in complex organization, as well as the role of irregular or disordered parts of proteins in crucial biological processes.
Collapse
Affiliation(s)
- Victor Neduva
- European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | | |
Collapse
|
47
|
Martinez-Contreras R, Cloutier P, Shkreta L, Fisette JF, Revil T, Chabot B. hnRNP proteins and splicing control. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 623:123-47. [PMID: 18380344 DOI: 10.1007/978-0-387-77374-2_8] [Citation(s) in RCA: 282] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Proteins of the heterogeneous nuclear ribonucleoparticles (hnRNP) family form a structurally diverse group of RNA binding proteins implicated in various functions in metazoans. Here we discuss recent advances supporting a role for these proteins in precursor-messenger RNA (pre-mRNA) splicing. Heterogeneous nuclear RNP proteins can repress splicing by directly antagonizing the recognition of splice sites, or can interfere with the binding of proteins bound to enhancers. Recently, hnRNP proteins have been shown to hinder communication between factors bound to different splice sites. Conversely, several reports have described a positive role for some hnRNP proteins in pre-mRNA splicing. Moreover, cooperative interactions between bound hnRNP proteins may encourage splicing between specific pairs of splice sites while simultaneously hampering other combinations. Thus, hnRNP proteins utilize a variety of strategies to control splice site selection in a manner that is important for both alternative and constitutive pre-mRNA splicing.
Collapse
|
48
|
Zakaryan RP, Gehring H. Identification and characterization of the nuclear localization/retention signal in the EWS proto-oncoprotein. J Mol Biol 2006; 363:27-38. [PMID: 16965792 DOI: 10.1016/j.jmb.2006.08.018] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2006] [Revised: 08/03/2006] [Accepted: 08/03/2006] [Indexed: 10/24/2022]
Abstract
Ewing sarcoma (EWS) protein, a member of a large family of RNA-binding proteins, contains an N-terminal transcriptional activation domain (EAD) and a C-terminal RNA-binding domain (RBD). Due to its multifunctional properties EWS protein is involved in processes such as gene expression, RNA processing and transport, and cell signaling. Chimeric EWS proteins generated by chromosomal translocations cause malignant tumors. EWS protein is located predominantly in the nucleus, but was found also in the cytosol and associated with the cell membrane. The determinants responsible for the nuclear localization of the protein were as yet unknown. We identified the nuclear localization signal of EWS protein at its C terminus (C-NLS), which is required for the nuclear import and retention of the protein. The C-NLS sequence is conserved in related proto-oncoproteins suggesting an NLS function also in these proteins. Two arginine residues, due to their positive charge, a proline residue and a tyrosine residue are essential for C-NLS function. The nuclear localization of EWS protein is independent of the regions in RBD containing numerous arginine methylation sites, RNA-recognition and zinc finger motifs. Regions in EAD guide the subnuclear partition of EWS protein and contain another but different NLS that allows nucleocytoplasmic shuttling of the N-terminal domain.
Collapse
Affiliation(s)
- Rouzanna P Zakaryan
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | | |
Collapse
|
49
|
Manceau V, Swenson M, Le Caer JP, Sobel A, Kielkopf CL, Maucuer A. Major phosphorylation of SF1 on adjacent Ser-Pro motifs enhances interaction with U2AF65. FEBS J 2006; 273:577-87. [PMID: 16420481 PMCID: PMC1949809 DOI: 10.1111/j.1742-4658.2005.05091.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Protein phosphorylation ensures the accurate and controlled expression of the genome, for instance by regulating the activities of pre-mRNA splicing factors. Here we report that splicing factor 1 (SF1), which is involved in an early step of intronic sequence recognition, is highly phosphorylated in mammalian cells on two serines within an SPSP motif at the junction between its U2AF65 and RNA binding domains. We show that SF1 interacts in vitro with the protein kinase KIS, which possesses a 'U2AF homology motif' (UHM) domain. The UHM domain of KIS is required for KIS and SF1 to interact, and for KIS to efficiently phosphorylate SF1 on the SPSP motif. Importantly, SPSP phosphorylation by KIS increases binding of SF1 to U2AF65, and enhances formation of the ternary SF1-U2AF65-RNA complex. These results further suggest that this phosphorylation event has an important role for the function of SF1, and possibly for the structural rearrangements associated with spliceosome assembly and function.
Collapse
Affiliation(s)
- Valérie Manceau
- INSERM U706, Institut du Fer à Moulin, 17, rue du Fer à Moulin, F-75005 Paris, France; UPMC, F-75005 Paris, France
| | - Matthew Swenson
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21205,USA
| | - Jean-Pierre Le Caer
- Ecole Polytechnique, Laboratoire de Chimie des Mécanismes Réactionnels, Route de Saclay, F-91128 Palaiseau, France
| | - André Sobel
- INSERM U706, Institut du Fer à Moulin, 17, rue du Fer à Moulin, F-75005 Paris, France; UPMC, F-75005 Paris, France
| | - Clara L. Kielkopf
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21205,USA
| | - Alexandre Maucuer
- INSERM U706, Institut du Fer à Moulin, 17, rue du Fer à Moulin, F-75005 Paris, France; UPMC, F-75005 Paris, France
| |
Collapse
|
50
|
Abstract
Ewing tumors, which comprise Ewing's sarcoma and peripheral primitive neuroectodermal tumors, are highly aggressive and mostly affect children and adolescents. Their molecular signature is a chromosomal translocation leading to the generation of EWS-ETS (or very rarely FUS-ETS) fusion proteins that are capable of transforming cells. These oncoproteins act as aberrant transcription factors due to the fusion of an ETS DNA binding domain to a highly potent EWS (or FUS) transactivation domain. Accordingly, many EWS-ETS target genes have been identified whose dysregulation could contribute to the development of tumor formation. Furthermore, EWS-ETS oncoproteins may impact on RNA splicing or affect other proteins through disturbing their ability to form functional complexes. The molecular knowledge gained so far from studying EWS-ETS oncoproteins has not only broadened our understanding of Ewing tumors but also improved the diagnosis of these highly undifferentiated tumors. In addition, several potential prognostic markers have been uncovered and novel therapies are suggested that may improve the still dismal survival rate of Ewing tumor patients.
Collapse
Affiliation(s)
- Ralf Janknecht
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA.
| |
Collapse
|