1
|
A Degradation Motif in STAU1 Defines a Novel Family of Proteins Involved in Inflammation. Int J Mol Sci 2022; 23:ijms231911588. [PMID: 36232890 PMCID: PMC9569955 DOI: 10.3390/ijms231911588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 11/21/2022] Open
Abstract
Cancer development is regulated by inflammation. Staufen1 (STAU1) is an RNA-binding protein whose expression level is critical in cancer cells as it is related to cell proliferation or cell death. STAU1 protein levels are downregulated during mitosis due to its degradation by the E3 ubiquitin ligase anaphase-promoting complex/cyclosome (APC/C). In this paper, we map the molecular determinant involved in STAU1 degradation to amino acids 38-50, and by alanine scanning, we shorten the motif to F39PxPxxLxxxxL50 (FPL-motif). Mutation of the FPL-motif prevents STAU1 degradation by APC/C. Interestingly, a search in databases reveals that the FPL-motif is shared by 15 additional proteins, most of them being involved in inflammation. We show that one of these proteins, MAP4K1, is indeed degraded via the FPL-motif; however, it is not a target of APC/C. Using proximity labeling with STAU1, we identify TRIM25, an E3 ubiquitin ligase involved in the innate immune response and interferon production, as responsible for STAU1 and MAP4K1 degradation, dependent on the FPL-motif. These results are consistent with previous studies that linked STAU1 to cancer-induced inflammation and identified a novel degradation motif that likely coordinates a novel family of proteins involved in inflammation. Data are available via ProteomeXchange with the identifier PXD036675.
Collapse
|
2
|
Gonzalez Quesada Y, Bonnet-Magnaval F, DesGroseillers L. Phosphomimicry on STAU1 Serine 20 Impairs STAU1 Posttranscriptional Functions and Induces Apoptosis in Human Transformed Cells. Int J Mol Sci 2022; 23:ijms23137344. [PMID: 35806349 PMCID: PMC9266326 DOI: 10.3390/ijms23137344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 12/22/2022] Open
Abstract
Staufen 1 (STAU1) is an RNA-binding protein that is essential in untransformed cells. In cancer cells, it is rather STAU1 overexpression that impairs cell proliferation. In this paper, we show that a modest increase in STAU1 expression in cancer cells triggers apoptosis as early as 12 h post-transfection and impairs proliferation in non-apoptotic cells for several days. Interestingly, a mutation that mimics the phosphorylation of STAU1 serine 20 is sufficient to cause these phenotypes, indicating that serine 20 is at the heart of the molecular mechanism leading to apoptosis. Mechanistically, phosphomimicry on serine 20 alters the ability of STAU1 to regulate translation and the decay of STAU1-bound mRNAs, indicating that the posttranscriptional regulation of mRNAs by STAU1 controls the balance between proliferation and apoptosis. Unexpectedly, the expression of RBD2S20D, the N-terminal 88 amino acids with no RNA-binding activity, is sufficient to induce apoptosis via alteration, in trans, of the posttranscriptional functions of endogenous STAU1. These results suggest that STAU1 is a sensor that controls the balance between cell proliferation and apoptosis, and, therefore, may be considered as a novel therapeutic target against cancer.
Collapse
|
3
|
Ramos H, Monette A, Niu M, Barrera A, López-Ulloa B, Fuentes Y, Guizar P, Pino K, DesGroseillers L, Mouland A, López-Lastra M. The double-stranded RNA-binding protein, Staufen1, is an IRES-transacting factor regulating HIV-1 cap-independent translation initiation. Nucleic Acids Res 2022; 50:411-429. [PMID: 34893869 PMCID: PMC8754648 DOI: 10.1093/nar/gkab1188] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/06/2021] [Accepted: 11/16/2021] [Indexed: 02/05/2023] Open
Abstract
Translation initiation of the viral genomic mRNA (vRNA) of human immunodeficiency virus-type 1 (HIV-1) can be mediated by a cap- or an internal ribosome entry site (IRES)-dependent mechanism. A previous report shows that Staufen1, a cellular double-stranded (ds) RNA-binding protein (RBP), binds to the 5'untranslated region (5'UTR) of the HIV-1 vRNA and promotes its cap-dependent translation. In this study, we now evaluate the role of Staufen1 as an HIV-1 IRES-transacting factor (ITAF). We first confirm that Staufen1 associates with both the HIV-1 vRNA and the Gag protein during HIV-1 replication. We found that in HIV-1-expressing cells, siRNA-mediated depletion of Staufen1 reduces HIV-1 vRNA translation. Using dual-luciferase bicistronic mRNAs, we show that the siRNA-mediated depletion and cDNA-mediated overexpression of Staufen1 acutely regulates HIV-1 IRES activity. Furthermore, we show that Staufen1-vRNA interaction is required for the enhancement of HIV-1 IRES activity. Interestingly, we find that only Staufen1 harboring an intact dsRNA-binding domain 3 (dsRBD3) rescues HIV-1 IRES activity in Staufen1 CRISPR-Cas9 gene edited cells. Finally, we show that the expression of Staufen1-dsRBD3 alone enhances HIV-1 IRES activity. This study provides evidence of a novel role for Staufen1 as an ITAF promoting HIV-1 vRNA IRES activity.
Collapse
Affiliation(s)
- Hade Ramos
- Laboratorio de Virología Molecular, Instituto Milenio de Inmunología e Inmunoterapia, Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, Escuela de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| | - Anne Monette
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, Québec H3T 1E2, Canada
| | - Meijuan Niu
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, Québec H3T 1E2, Canada
| | - Aldo Barrera
- Laboratorio de Virología Molecular, Instituto Milenio de Inmunología e Inmunoterapia, Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, Escuela de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| | - Brenda López-Ulloa
- Laboratorio de Virología Molecular, Instituto Milenio de Inmunología e Inmunoterapia, Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, Escuela de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| | - Yazmín Fuentes
- Laboratorio de Virología Molecular, Instituto Milenio de Inmunología e Inmunoterapia, Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, Escuela de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| | - Paola Guizar
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, Québec H3T 1E2, Canada
- Department of Medicine, McGill University, Montréal, Québec H4A 3J1, Canada
| | - Karla Pino
- Laboratorio de Virología Molecular, Instituto Milenio de Inmunología e Inmunoterapia, Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, Escuela de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| | - Luc DesGroseillers
- Department of Biochemistry and Molecular Medicine, University of Montreal, P.O. Box 6128, Station Centre Ville, Montreal, Québec H3C 3J7, Canada
| | - Andrew J Mouland
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, Québec H3T 1E2, Canada
- Department of Medicine, McGill University, Montréal, Québec H4A 3J1, Canada
| | - Marcelo López-Lastra
- Laboratorio de Virología Molecular, Instituto Milenio de Inmunología e Inmunoterapia, Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, Escuela de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| |
Collapse
|
4
|
Liang X, Jiao Y, Gong X, Gu H, Nuermaimaiti N, Meng X, Liu D, Guan Y. Staufen1 unwinds the secondary structure and facilitates the translation of fatty acid binding protein 4 mRNA during adipogenesis. Adipocyte 2021; 10:350-360. [PMID: 34224297 PMCID: PMC8259723 DOI: 10.1080/21623945.2021.1948165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Adipogenesis is regulated by genetic interactions, in which post-transcriptional regulation plays an important role. Staufen double-stranded RNA binding protein 1 (Staufen1 or STAU1) plays diverse roles in RNA processing and adipogenesis. Previously, we found that the downregulation of STAU1 affects the expression of fatty acid-binding protein 4 (FABP4) at the protein level but not at the mRNA level. This study aimed to determine the mechanism underlying the regulation of FABP4 expression by STAU1, explaining the inconsistency between FABP4 mRNA and protein levels. We used RNA interference, photoactivatable ribonucleoside enhanced cross-linking and immunoprecipitation, and an adeno-associated virus to examine the functions of STAU1 in adipogenesis. Our results indicate that STAU1 binds to the coding sequences of FABP4, thereby regulating the translation of FABP4 mRNA by unwinding the double-stranded structure. Furthermore, STAU1 mediates adipogenesis by regulating the secretion of free fatty acids. However, STAU1 knockdown decreases the fat weight/body weight ratio but does not affect the plasma triglyceride levels. These findings describe the mechanisms involved in STAU1-mediated regulation of FABP4 expression at the translational level during adipogenesis.
Collapse
Affiliation(s)
- Xiaodi Liang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, Preclinical Medicine College, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yi Jiao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, Preclinical Medicine College, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xueli Gong
- Department of Pathophysiology, Preclinical Medicine College, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Hao Gu
- Department of Laparoscopic Surgery, First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Nuerbiye Nuermaimaiti
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, Preclinical Medicine College, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xuanyu Meng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, Preclinical Medicine College, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Dihui Liu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, Preclinical Medicine College, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yaqun Guan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Biochemistry and Molecular Biology, Preclinical Medicine College, Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
5
|
Almasi S, Jasmin BJ. The multifunctional RNA-binding protein Staufen1: an emerging regulator of oncogenesis through its various roles in key cellular events. Cell Mol Life Sci 2021; 78:7145-7160. [PMID: 34633481 PMCID: PMC8629789 DOI: 10.1007/s00018-021-03965-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/19/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022]
Abstract
The double-stranded multifunctional RNA-binding protein (dsRBP) Staufen was initially discovered in insects as a regulator of mRNA localization. Later, its mammalian orthologs have been described in different organisms, including humans. Two human orthologues of Staufen, named Staufen1 (STAU1) and Staufen2 (STAU2), share some structural and functional similarities. However, given their different spatio-temporal expression patterns, each of these orthologues plays distinct roles in cells. In the current review, we focus on the role of STAU1 in cell functions and cancer development. Since its discovery, STAU1 has mostly been studied for its involvement in various aspects of RNA metabolism. Given the pivotal role of RNA metabolism within cells, recent studies have explored the mechanistic impact of STAU1 in a wide variety of cell functions ranging from cell growth to cell death, as well as in various disease states. In particular, there has been increasing attention on the role of STAU1 in neuromuscular disorders, neurodegeneration, and cancer. Here, we provide an overview of the current knowledge on the role of STAU1 in RNA metabolism and cell functions. We also highlight the link between STAU1-mediated control of cellular functions and cancer development, progression, and treatment. Hence, our review emphasizes the potential of STAU1 as a novel biomarker and therapeutic target for cancer diagnosis and treatment, respectively.
Collapse
Affiliation(s)
- Shekoufeh Almasi
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
- The Eric J. Poulin Centre for Neuromuscular Diseases, Faculty of Medicine, University of Ottawa, Ottawa, K1H 8M5, Canada.
| |
Collapse
|
6
|
Park SW, Yu KL, Bae JH, Kim GN, Kim HI, You JC. Investigation of the effect of Staufen1 overexpression on the HIV-1 virus production. BMB Rep 2021. [PMID: 34353428 PMCID: PMC8633522 DOI: 10.5483/bmbrep.2021.54.11.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this study, we investigated how Staufen1 influences the HIV-1 production. The overexpression of Staufen1 increased virus production without any negative affect on the viral infectivity. This increase was not caused by transcriptional activation; but by influencing post-transcriptional steps. Using multiple Gag protein derivatives, we confirmed that the zinc-finger domains of the HIV-1 nucleocapsid (NC) are important for its interaction with Staufen1. We also found that Staufen1 colocalized in stress granules with the mature form of the HIV-1 NC protein.
Collapse
Affiliation(s)
- Seong-won Park
- National Research Laboratory of Molecular Virology, Department of Pathology, School of Medicine, The Catholic University of Korea, Seoul 63071, Korea
| | - Kyung-Lee Yu
- National Research Laboratory of Molecular Virology, Department of Pathology, School of Medicine, The Catholic University of Korea, Seoul 63071, Korea
| | - Jun-Hyun Bae
- National Research Laboratory of Molecular Virology, Department of Pathology, School of Medicine, The Catholic University of Korea, Seoul 63071, Korea
| | - Ga-Na Kim
- National Research Laboratory of Molecular Virology, Department of Pathology, School of Medicine, The Catholic University of Korea, Seoul 63071, Korea
| | - Hae-In Kim
- National Research Laboratory of Molecular Virology, Department of Pathology, School of Medicine, The Catholic University of Korea, Seoul 63071, Korea
| | - Ji Chang You
- National Research Laboratory of Molecular Virology, Department of Pathology, School of Medicine, The Catholic University of Korea, Seoul 63071, Korea
| |
Collapse
|
7
|
Bonnet-Magnaval F, DesGroseillers L. The Staufen1-dependent cell cycle regulon or how a misregulated RNA-binding protein leads to cancer. Biol Rev Camb Philos Soc 2021; 96:2192-2208. [PMID: 34018319 DOI: 10.1111/brv.12749] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022]
Abstract
In recent years, an increasing number of reports have linked the RNA-binding protein Staufen1 (STAU1) to the control of cell decision making. In non-transformed cells, STAU1 balances the expression of messenger RNA (mRNA) regulons that regulate differentiation and well-ordered cell division. Misregulation of STAU1 expression and/or functions changes the fragile balance in the expression of pro- and anti-proliferative and apoptotic genes and favours a novel equilibrium that supports cell proliferation and cancer development. The misregulation of STAU1 functions causes multiple coordinated modest effects in the post-transcriptional regulation of many RNA targets that code for cell cycle regulators, leading to dramatic consequences at the cellular level. The new tumorigenic equilibrium in STAU1-mediated gene regulation observed in cancer cells can be further altered by a slight increase in STAU1 expression that favours expression of pro-apoptotic genes and cell death. The STAU1-dependent cell cycle regulon is a good model to study how abnormal expression of an RNA-binding protein promotes cell growth and provides an advantageous selection of malignant cells in the first step of cancer development.
Collapse
Affiliation(s)
- Florence Bonnet-Magnaval
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Édouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Luc DesGroseillers
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Édouard Montpetit, Montréal, QC, H3T 1J4, Canada
| |
Collapse
|
8
|
Zhang X, Li P, Zhong H, Yang F, Liu F, Yedid G, Zeng Y. Extending the L1 region in canonical double-stranded RNA-binding domains impairs their functions. Acta Biochim Biophys Sin (Shanghai) 2021; 53:463-471. [PMID: 33751023 DOI: 10.1093/abbs/gmab014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Indexed: 12/24/2022] Open
Abstract
A large number of proteins involved in RNA metabolism possess a double-stranded RNA-binding domain (dsRBD), whose sequence variations and functional versatilities are still being recognized. All dsRBDs have a similar structural fold: α1-L1-β1-L2-β2-L3-β3-L4-α2 (α represents an α-helix, β a β-sheet, and L a loop conformation between the well-defined secondary structures). Our recent work revealed that the dsRBD in Drosha, which is involved in animal microRNA (miRNA) biogenesis, differs from other dsRBDs by containing a short insertion in its L1 region and that this insertion is important for Drosha function. We asked why the same insertion is excluded in all other dsRBDs and proposed that a longer L1 may be detrimental to their functions. In this study, to test this hypothesis, we inserted the Drosha sequence into several well-known dsRBDs from various organisms. Gel mobility shift assay demonstrated that L1 extension invariably reduced RNA binding by these dsRBDs. In addition, such a mutation in Dicer, another protein involved in miRNA biogenesis, impaired Dicer's ability to process miRNAs, which led to de-repression of reporter expression, in human cells. Taken together, our results add to the growing appreciation of the diversity in dsRBDs and suggest that dsRBDs have intricate structures and functions that are sensitive to perturbations in the L1 region.
Collapse
Affiliation(s)
- Xiaoxiao Zhang
- Department of Zoology, College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Peng Li
- Department of Zoology, College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Huanhuan Zhong
- Department of Zoology, College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Fanming Yang
- Department of Zoology, College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Fanzhou Liu
- Department of Zoology, College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Gabriel Yedid
- Department of Zoology, College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Yan Zeng
- Department of Zoology, College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
9
|
Zheng D, Cho H, Wang W, Rambout X, Tian B, Maquat LE. 3'READS + RIP defines differential Staufen1 binding to alternative 3'UTR isoforms and reveals structures and sequence motifs influencing binding and polysome association. RNA (NEW YORK, N.Y.) 2020; 26:1621-1636. [PMID: 32796083 PMCID: PMC7566578 DOI: 10.1261/rna.076133.120] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/15/2020] [Indexed: 06/11/2023]
Abstract
Staufen1 (STAU1) is an RNA-binding protein (RBP) that interacts with double-stranded RNA structures and has been implicated in regulating different aspects of mRNA metabolism. Previous studies have indicated that STAU1 interacts extensively with RNA structures in coding regions (CDSs) and 3'-untranslated regions (3'UTRs). In particular, duplex structures formed within 3'UTRs by inverted-repeat Alu elements (IRAlus) interact with STAU1 through its double-stranded RNA-binding domains (dsRBDs). Using 3' region extraction and deep sequencing coupled to ribonucleoprotein immunoprecipitation (3'READS + RIP), together with reanalyzing previous STAU1 binding and RNA structure data, we delineate STAU1 interactions transcriptome-wide, including binding differences between alternative polyadenylation (APA) isoforms. Consistent with previous reports, RNA structures are dominant features for STAU1 binding to CDSs and 3'UTRs. Overall, relative to short 3'UTR counterparts, longer 3'UTR isoforms of genes have stronger STAU1 binding, most likely due to a higher frequency of RNA structures, including specific IRAlus sequences. Nevertheless, a sizable fraction of genes express transcripts showing the opposite trend, attributable to AU-rich sequences in their alternative 3'UTRs that may recruit antagonistic RBPs and/or destabilize RNA structures. Using STAU1-knockout cells, we show that strong STAU1 binding to mRNA 3'UTRs generally enhances polysome association. However, IRAlus generally have little impact on STAU1-mediated polysome association despite having strong interactions with the protein. Taken together, our work reveals complex interactions of STAU1 with its cognate RNA substrates. Our data also shed light on distinct post-transcriptional fates for the widespread APA isoforms in mammalian cells.
Collapse
Affiliation(s)
- Dinghai Zheng
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey 07103, USA
| | - Hana Cho
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, USA
- Center for RNA Biology, University of Rochester, Rochester, New York 14642, USA
| | - Wei Wang
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey 07103, USA
| | - Xavier Rambout
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, USA
- Center for RNA Biology, University of Rochester, Rochester, New York 14642, USA
| | - Bin Tian
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey 07103, USA
- Program in Gene Expression and Regulation, and Center for Systems and Computational Biology, Wistar Institute, Philadelphia, Pennsylvania 19104, USA
| | - Lynne E Maquat
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, USA
- Center for RNA Biology, University of Rochester, Rochester, New York 14642, USA
| |
Collapse
|
10
|
Hassine S, Bonnet-Magnaval F, Benoit Bouvrette LP, Doran B, Ghram M, Bouthillette M, Lecuyer E, DesGroseillers L. Staufen1 localizes to the mitotic spindle and controls the localization of RNA populations to the spindle. J Cell Sci 2020; 133:jcs247155. [PMID: 32576666 DOI: 10.1242/jcs.247155] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/07/2020] [Indexed: 12/20/2022] Open
Abstract
Staufen1 (STAU1) is an RNA-binding protein involved in the post-transcriptional regulation of mRNAs. We report that a large fraction of STAU1 localizes to the mitotic spindle in colorectal cancer HCT116 cells and in non-transformed hTERT-RPE1 cells. Spindle-associated STAU1 partly co-localizes with ribosomes and active sites of translation. We mapped the molecular determinant required for STAU1-spindle association within the first 88 N-terminal amino acids, a domain that is not required for RNA binding. Interestingly, transcriptomic analysis of purified mitotic spindles revealed that 1054 mRNAs and the precursor ribosomal RNA (pre-rRNA), as well as the long non-coding RNAs and small nucleolar RNAs involved in ribonucleoprotein assembly and processing, are enriched on spindles compared with cell extracts. STAU1 knockout causes displacement of the pre-rRNA and of 154 mRNAs coding for proteins involved in actin cytoskeleton organization and cell growth, highlighting a role for STAU1 in mRNA trafficking to spindle. These data demonstrate that STAU1 controls the localization of subpopulations of RNAs during mitosis and suggests a novel role of STAU1 in pre-rRNA maintenance during mitosis, ribogenesis and/or nucleoli reassembly.
Collapse
Affiliation(s)
- Sami Hassine
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Édouard Montpetit, Montréal, QC H3T 1J4, Canada
| | - Florence Bonnet-Magnaval
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Édouard Montpetit, Montréal, QC H3T 1J4, Canada
| | - Louis Philip Benoit Bouvrette
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Édouard Montpetit, Montréal, QC H3T 1J4, Canada
- Institut de Recherches Cliniques de Montréal, 110 Avenue des Pins Ouest, Montréal, QC H2W 1R7, Canada
| | - Bellastrid Doran
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Édouard Montpetit, Montréal, QC H3T 1J4, Canada
| | - Mehdi Ghram
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Édouard Montpetit, Montréal, QC H3T 1J4, Canada
| | - Mathieu Bouthillette
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Édouard Montpetit, Montréal, QC H3T 1J4, Canada
| | - Eric Lecuyer
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Édouard Montpetit, Montréal, QC H3T 1J4, Canada
- Institut de Recherches Cliniques de Montréal, 110 Avenue des Pins Ouest, Montréal, QC H2W 1R7, Canada
| | - Luc DesGroseillers
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Édouard Montpetit, Montréal, QC H3T 1J4, Canada
| |
Collapse
|
11
|
Ghram M, Bonnet-Magnaval F, Hotea DI, Doran B, Ly S, DesGroseillers L. Staufen1 is Essential for Cell-Cycle Transitions and Cell Proliferation Via the Control of E2F1 Expression. J Mol Biol 2020; 432:3881-3897. [DOI: 10.1016/j.jmb.2020.04.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/14/2020] [Accepted: 04/20/2020] [Indexed: 12/16/2022]
|
12
|
Yadav DK, Zigáčková D, Zlobina M, Klumpler T, Beaumont C, Kubíčková M, Vaňáčová Š, Lukavsky PJ. Staufen1 reads out structure and sequence features in ARF1 dsRNA for target recognition. Nucleic Acids Res 2020; 48:2091-2106. [PMID: 31875226 PMCID: PMC7038937 DOI: 10.1093/nar/gkz1163] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 11/22/2019] [Accepted: 12/05/2019] [Indexed: 12/13/2022] Open
Abstract
Staufen1 (STAU1) is a dsRNA binding protein mediating mRNA transport and localization, translational control and STAU1-mediated mRNA decay (SMD). The STAU1 binding site (SBS) within human ADP-ribosylation factor1 (ARF1) 3′UTR binds STAU1 and this downregulates ARF1 cytoplasmic mRNA levels by SMD. However, how STAU1 recognizes specific mRNA targets is still under debate. Our structure of the ARF1 SBS–STAU1 complex uncovers target recognition by STAU1. STAU1 dsRNA binding domain (dsRBD) 4 interacts with two pyrimidines and one purine from the minor groove side via helix α1, the β1–β2 loop anchors the dsRBD at the end of the dsRNA and lysines in helix α2 bind to the phosphodiester backbone from the major groove side. STAU1 dsRBD3 displays the same binding mode with specific recognition of one guanine base. Mutants disrupting minor groove recognition of ARF1 SBS affect in vitro binding and reduce SMD in vivo. Our data thus reveal how STAU1 recognizes minor groove features in dsRNA relevant for target selection.
Collapse
Affiliation(s)
- Deepak Kumar Yadav
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 62500, Brno, Czech Republic
| | - Dagmar Zigáčková
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 62500, Brno, Czech Republic
| | - Maria Zlobina
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 62500, Brno, Czech Republic
| | - Tomáš Klumpler
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 62500, Brno, Czech Republic
| | - Christelle Beaumont
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 62500, Brno, Czech Republic
| | - Monika Kubíčková
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 62500, Brno, Czech Republic
| | - Štěpánka Vaňáčová
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 62500, Brno, Czech Republic
| | - Peter J Lukavsky
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 62500, Brno, Czech Republic
| |
Collapse
|
13
|
Visentin S, Cannone G, Doutch J, Harris G, Gleghorn ML, Clifton L, Smith BO, Spagnolo L. A multipronged approach to understanding the form and function of hStaufen protein. RNA (NEW YORK, N.Y.) 2020; 26:265-277. [PMID: 31852734 PMCID: PMC7025507 DOI: 10.1261/rna.072595.119] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 12/09/2019] [Indexed: 05/09/2023]
Abstract
Staufen is a dsRNA-binding protein involved in many aspects of RNA regulation, such as mRNA transport, Staufen-mediated mRNA decay and the regulation of mRNA translation. It is a modular protein characterized by the presence of conserved consensus amino acid sequences that fold into double-stranded RNA binding domains (RBDs) as well as degenerated RBDs that are instead involved in protein-protein interactions. The variety of biological processes in which Staufen participates in the cell suggests that this protein associates with many diverse RNA targets, some of which have been identified experimentally. Staufen binding mediates the recruitment of effectors via protein-protein and protein-RNA interactions. The structural determinants of a number of these interactions, as well as the structure of full-length Staufen, remain unknown. Here, we present the first solution structure models for full-length hStaufen155, showing that its domains are arranged as beads-on-a-string connected by flexible linkers. In analogy with other nucleic acid-binding proteins, this could underpin Stau1 functional plasticity.
Collapse
Affiliation(s)
- Silvia Visentin
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow G12 8QQ, United Kingdom
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JQ, United Kingdom
- ISIS Pulsed Neutron and Muon Source, Science and Technology Facilities Council (STFC), Rutherford Appleton Laboratory, Didcot OX11 OQX, United Kingdom
| | - Giuseppe Cannone
- Institute of Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JQ, United Kingdom
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - James Doutch
- ISIS Pulsed Neutron and Muon Source, Science and Technology Facilities Council (STFC), Rutherford Appleton Laboratory, Didcot OX11 OQX, United Kingdom
| | - Gemma Harris
- Research Complex at Harwell, Rutherford Appleton Laboratory, Research Complex at Harwell, Didcot OX11 0FA, United Kingdom
| | - Michael L Gleghorn
- School of Chemistry and Materials Science, College of Science, Rochester Institute of Technology, Rochester, New York 14623, USA
| | - Luke Clifton
- ISIS Pulsed Neutron and Muon Source, Science and Technology Facilities Council (STFC), Rutherford Appleton Laboratory, Didcot OX11 OQX, United Kingdom
| | - Brian O Smith
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Laura Spagnolo
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| |
Collapse
|
14
|
Insights into the assembly and architecture of a Staufen-mediated mRNA decay (SMD)-competent mRNP. Nat Commun 2019; 10:5054. [PMID: 31699982 PMCID: PMC6838198 DOI: 10.1038/s41467-019-13080-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 10/18/2019] [Indexed: 02/07/2023] Open
Abstract
The mammalian Staufen proteins (Stau1 and Stau2) mediate degradation of mRNA containing complex secondary structures in their 3’-untranslated region (UTR) through a pathway known as Staufen-mediated mRNA decay (SMD). This pathway also involves the RNA helicase UPF1, which is best known for its role in the nonsense-mediated mRNA decay (NMD) pathway. Here we present a biochemical reconstitution of the recruitment and activation of UPF1 in context of the SMD pathway. We demonstrate the involvement of UPF2, a core NMD factor and a known activator of UPF1, in SMD. UPF2 acts as an adaptor between Stau1 and UPF1, stimulates the catalytic activity of UPF1 and plays a central role in the formation of an SMD-competent mRNP. Our study elucidates the molecular mechanisms of SMD and points towards extensive cross-talk between UPF1-mediated mRNA decay pathways in cells. The Staufen proteins recognize secondary structures in 3’-untranslated regions in mRNA transcripts and induce degradation of these mRNAs with the help of the RNA helicase UPF1. Here the authors report that the nonsense-mediated mRNA decay factor UPF2 mediates the interaction between Stau1 and UPF1 in Staufen-mediated mRNA decay.
Collapse
|
15
|
Oh D, Houston DW. RNA Localization in the Vertebrate Oocyte: Establishment of Oocyte Polarity and Localized mRNA Assemblages. Results Probl Cell Differ 2019; 63:189-208. [PMID: 28779319 PMCID: PMC6538070 DOI: 10.1007/978-3-319-60855-6_9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
RNA localization is a fundamental mechanism for controlling cell structure and function. Early development in fish and amphibians requires the localization of specific mRNAs to establish the initial differences in cell fates prior to the onset of zygotic genome activation. RNA localization in these oocytes (e.g., Xenopus and zebrafish) requires that animal-vegetal polarity be established early in oogenesis, mediated by formation of the Balbiani body/mitochondrial cloud. This structure serves as a platform for assembly and transport of germline determinants to the future vegetal pole and also sets up the machinery for the localization of non-germline transcripts later in oogenesis. Understanding these polarization and localization mechanisms is critical for understanding the basis for early embryonic development in these organisms and also for understanding the role of RNA compartmentalization in animal gametogenesis. Here we outline recent advances in elucidating the molecular basis for the establishment of oocyte polarity at the level of Balbiani body assembly as well as the formation of RNP assemblies for early and late pathway mRNA localization in the oocyte.
Collapse
Affiliation(s)
- Denise Oh
- Department of Biology, The University of Iowa, 257 BB, Iowa City, IA, 52242, USA
| | - Douglas W Houston
- Department of Biology, The University of Iowa, 257 BB, Iowa City, IA, 52242, USA.
| |
Collapse
|
16
|
Lazzaretti D, Bandholz-Cajamarca L, Emmerich C, Schaaf K, Basquin C, Irion U, Bono F. The crystal structure of Staufen1 in complex with a physiological RNA sheds light on substrate selectivity. Life Sci Alliance 2018; 1:e201800187. [PMID: 30456389 PMCID: PMC6238398 DOI: 10.26508/lsa.201800187] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/03/2018] [Accepted: 10/05/2018] [Indexed: 01/29/2023] Open
Abstract
Combination of in vitro and in vivo data show that RNA sequence influences Staufen target recognition and that protein–RNA base contacts are required for Staufen function in Drosophila. During mRNA localization, RNA-binding proteins interact with specific structured mRNA localization motifs. Although several such motifs have been identified, we have limited structural information on how these interact with RNA-binding proteins. Staufen proteins bind structured mRNA motifs through dsRNA-binding domains (dsRBD) and are involved in mRNA localization in Drosophila and mammals. We solved the structure of two dsRBDs of human Staufen1 in complex with a physiological dsRNA sequence. We identified interactions between the dsRBDs and the RNA sugar–phosphate backbone and direct contacts of conserved Staufen residues to RNA bases. Mutating residues mediating nonspecific backbone interactions only affected Staufen function in Drosophila when in vitro binding was severely reduced. Conversely, residues involved in base-directed interactions were required in vivo even when they minimally affected in vitro binding. Our work revealed that Staufen can read sequence features in the minor groove of dsRNA and suggests that these influence target selection in vivo.
Collapse
Affiliation(s)
| | | | | | - Kristina Schaaf
- Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Claire Basquin
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Uwe Irion
- Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Fulvia Bono
- Max Planck Institute for Developmental Biology, Tübingen, Germany.,Living Systems Institute, University of Exeter, Exeter, UK
| |
Collapse
|
17
|
Beaujois R, Ottoni E, Zhang X, Gagnon C, Hassine S, Mollet S, Viranaicken W, DesGroseillers L. The M-phase specific hyperphosphorylation of Staufen2 involved the cyclin-dependent kinase CDK1. BMC Cell Biol 2017; 18:25. [PMID: 28705199 PMCID: PMC5513041 DOI: 10.1186/s12860-017-0142-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/10/2017] [Indexed: 01/21/2023] Open
Abstract
Background Staufen2 (STAU2) is an RNA-binding protein involved in the post-transcriptional regulation of gene expression. This protein was shown to be required for organ formation and cell differentiation. Although STAU2 functions have been reported in neuronal cells, its role in dividing cells remains deeply uncharacterized. Especially, its regulation during the cell cycle is completely unknown. Results In this study, we showed that STAU2 isoforms display a mitosis-specific slow migration pattern on SDS-gels in all tested transformed and untransformed cell lines. Deeper analyses in hTert-RPE1 and HeLa cells further indicated that the slow migration pattern of STAU2 isoforms is due to phosphorylation. Time course studies showed that STAU2 phosphorylation occurs before prometaphase and terminates as cells exit mitosis. Interestingly, STAU2 isoforms were phosphorylated on several amino acid residues in the C-terminal half via the cyclin-dependent kinase 1 (Cdk1), an enzyme known to play crucial roles during mitosis. Introduction of phospho-mimetic or phospho-null mutations in STAU2 did not impair its RNA-binding capacity, its stability, its interaction with protein co-factors or its sub-cellular localization, suggesting that STAU2 phosphorylation in mitosis does not regulate these functions. Similarly, STAU2 phosphorylation is not likely to be crucial for cell cycle progression since expression of phosphorylation mutants in hTert-RPE1 cells did not impair cell proliferation. Conclusions Altogether, these results indicate that STAU2 isoforms are phosphorylated during mitosis and that the phosphorylation process involves Cdk1. The meaning of this post-translational modification is still elusive. Electronic supplementary material The online version of this article (doi:10.1186/s12860-017-0142-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rémy Beaujois
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Elizabeth Ottoni
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Xin Zhang
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Christina Gagnon
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Sami Hassine
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Stéphanie Mollet
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Wildriss Viranaicken
- Present address: UMR PIMIT, Processus Infectieux en Milieu Insulaire Tropical, Université de la Réunion, 97490 Sainte Clotilde, La Réunion, France
| | - Luc DesGroseillers
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada.
| |
Collapse
|
18
|
Hanke K, Hohn O, Bannert N. HERV-K(HML-2), a seemingly silent subtenant - but still waters run deep. APMIS 2016; 124:67-87. [PMID: 26818263 DOI: 10.1111/apm.12475] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/12/2015] [Indexed: 01/26/2023]
Abstract
A large proportion of the human genome consists of endogenous retroviruses, some of which are well preserved, showing transcriptional activity, and expressing retroviral proteins. The HERV-K(HML-2) family represents the most intact members of these elements, with some having open and intact reading frames for viral proteins and the ability to form virus-like particles. Although generally suppressed in most healthy tissues by a variety of epigenetic processes and antiviral mechanisms, there is evidence that some members of this family are (at least partly) still active - particularly in certain stem cells and various tumors. This raises the possibility of their involvement in tumor induction or in developmental processes. In recent years, many new insights into this fascinating field have been attained, and this review focuses on new discoveries about coevolutionary events and intracellular defense mechanisms against HERV-K(HML-2) activity. We also describe what might occur when these mechanisms fail or become modulated by viral proteins or other viruses and discuss the new vistas opened up by the reconstitution of ancestral viral proteins and even complete HML-2 viruses.
Collapse
Affiliation(s)
- Kirsten Hanke
- Department HIV and Other Retroviruses, Robert Koch Institute, Berlin, Germany
| | - Oliver Hohn
- Department HIV and Other Retroviruses, Robert Koch Institute, Berlin, Germany
| | - Norbert Bannert
- Department HIV and Other Retroviruses, Robert Koch Institute, Berlin, Germany
| |
Collapse
|
19
|
Zhang X, Trépanier V, Beaujois R, Viranaicken W, Drobetsky E, DesGroseillers L. The downregulation of the RNA-binding protein Staufen2 in response to DNA damage promotes apoptosis. Nucleic Acids Res 2016; 44:3695-712. [PMID: 26843428 PMCID: PMC4856980 DOI: 10.1093/nar/gkw057] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 01/21/2016] [Indexed: 12/14/2022] Open
Abstract
Staufen2 (Stau2) is an RNA-binding protein involved in cell fate decision by controlling several facets of mRNA processing including localization, splicing, translation and stability. Herein we report that exposure to DNA-damaging agents that generate replicative stress such as camptothecin (CPT), 5-fluoro-uracil (5FU) and ultraviolet radiation (UVC) causes downregulation of Stau2 in HCT116 colorectal cancer cells. In contrast, other agents such as doxorubicin and ionizing radiation had no effect on Stau2 expression. Consistently, Stau2 expression is regulated by the ataxia telangiectasia and Rad3-related (ATR) signaling pathway but not by the DNA-PK or ataxia telangiectasia mutated/checkpoint kinase 2 pathways. Stau2 downregulation is initiated at the level of transcription, independently of apoptosis induction. Promoter analysis identified a short 198 bp region which is necessary and sufficient for both basal and CPT-regulated Stau2 expression. The E2F1 transcription factor regulates Stau2 in untreated cells, an effect that is abolished by CPT treatment due to E2F1 displacement from the promoter. Strikingly, Stau2 downregulation enhances levels of DNA damage and promotes apoptosis in CPT-treated cells. Taken together our results suggest that Stau2 is an anti-apoptotic protein that could be involved in DNA replication and/or maintenance of genome integrity and that its expression is regulated by E2F1 via the ATR signaling pathway.
Collapse
Affiliation(s)
- Xin Zhang
- Département de Biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit Montréal, QC H3T 1J4, Canada
| | - Véronique Trépanier
- Département de Biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit Montréal, QC H3T 1J4, Canada
| | - Remy Beaujois
- Département de Biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit Montréal, QC H3T 1J4, Canada
| | - Wildriss Viranaicken
- Département de Biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit Montréal, QC H3T 1J4, Canada
| | - Elliot Drobetsky
- Département de Médecine, Université de Montréal and Centre de Recherche, Hôpital Maisonneuve Rosemont, Montréal, Québec, H1T 2M4, Canada
| | - Luc DesGroseillers
- Département de Biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit Montréal, QC H3T 1J4, Canada
| |
Collapse
|
20
|
Bondy-Chorney E, Crawford Parks TE, Ravel-Chapuis A, Klinck R, Rocheleau L, Pelchat M, Chabot B, Jasmin BJ, Côté J. Staufen1 Regulates Multiple Alternative Splicing Events either Positively or Negatively in DM1 Indicating Its Role as a Disease Modifier. PLoS Genet 2016; 12:e1005827. [PMID: 26824521 PMCID: PMC4733145 DOI: 10.1371/journal.pgen.1005827] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 01/05/2016] [Indexed: 01/10/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a neuromuscular disorder caused by an expansion of CUG repeats in the 3' UTR of the DMPK gene. The CUG repeats form aggregates of mutant mRNA, which cause misregulation and/or sequestration of RNA-binding proteins, causing aberrant alternative splicing in cells. Previously, we showed that the multi-functional RNA-binding protein Staufen1 (Stau1) was increased in skeletal muscle of DM1 mouse models and patients. We also showed that Stau1 rescues the alternative splicing profile of pre-mRNAs, e.g. the INSR and CLC1, known to be aberrantly spliced in DM1. In order to explore further the potential of Stau1 as a therapeutic target for DM1, we first investigated the mechanism by which Stau1 regulates pre-mRNA alternative splicing. We report here that Stau1 regulates the alternative splicing of exon 11 of the human INSR via binding to Alu elements located in intron 10. Additionally, using a high-throughput RT-PCR screen, we have identified numerous Stau1-regulated alternative splicing events in both WT and DM1 myoblasts. A number of these aberrant ASEs in DM1, including INSR exon 11, are rescued by overexpression of Stau1. However, we find other ASEs in DM1 cells, where overexpression of Stau1 shifts the splicing patterns away from WT conditions. Moreover, we uncovered that Stau1-regulated ASEs harbour Alu elements in intronic regions flanking the alternative exon more than non-Stau1 targets. Taken together, these data highlight the broad impact of Stau1 as a splicing regulator and suggest that Stau1 may act as a disease modifier in DM1.
Collapse
Affiliation(s)
- Emma Bondy-Chorney
- Department of Cellular and Molecular Medicine, University of Ottawa; Centre for Neuromuscular Disease, Ottawa, Ontario, Canada
| | - Tara E. Crawford Parks
- Department of Cellular and Molecular Medicine, University of Ottawa; Centre for Neuromuscular Disease, Ottawa, Ontario, Canada
| | - Aymeric Ravel-Chapuis
- Department of Cellular and Molecular Medicine, University of Ottawa; Centre for Neuromuscular Disease, Ottawa, Ontario, Canada
| | - Roscoe Klinck
- Département de microbiologie et d'infectiologie, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Lynda Rocheleau
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Martin Pelchat
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Benoit Chabot
- Département de microbiologie et d'infectiologie, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Bernard J. Jasmin
- Department of Cellular and Molecular Medicine, University of Ottawa; Centre for Neuromuscular Disease, Ottawa, Ontario, Canada
| | - Jocelyn Côté
- Department of Cellular and Molecular Medicine, University of Ottawa; Centre for Neuromuscular Disease, Ottawa, Ontario, Canada
- * E-mail:
| |
Collapse
|
21
|
Abstract
Messenger ribonucleoprotein (mRNP) granules are dynamic, self-assembling structures that harbor non-translating mRNAs bound by various proteins that regulate mRNA translation, localization, and turnover. Their importance in gene expression regulation is far reaching, ranging from precise spatial-temporal control of mRNAs that drive developmental programs in oocytes and embryos, to similarly exquisite control of mRNAs in neurons that underpin synaptic plasticity, and thus, memory formation. Analysis of mRNP granules in their various contexts has revealed common themes of assembly, disassembly, and modes of mRNA regulation, yet new studies continue to reveal unexpected and important findings, such as links between aberrant mRNP granule assembly and neurodegenerative disease. Continued study of these enigmatic structures thus promises fascinating new insights into cellular function, and may also suggest novel therapeutic strategies in various disease states.
Collapse
Affiliation(s)
- J Ross Buchan
- a Department of Molecular and Cellular Biology ; University of Arizona ; Tucson , AZ USA
| |
Collapse
|
22
|
Giner XC, Cotnoir-White D, Mader S, Lévesque D. Selective ligand activity at Nur/retinoid X receptor complexes revealed by dimer-specific bioluminescence resonance energy transfer-based sensors. FASEB J 2015; 29:4256-67. [PMID: 26148973 DOI: 10.1096/fj.14-259804] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 06/22/2015] [Indexed: 12/16/2022]
Abstract
Retinoid X receptors (RXRs) play a role as master regulators because of their capacity to form heterodimers with other nuclear receptors (NRs). Accordingly, retinoid signaling is involved in multiple biologic processes, including development, cell differentiation, metabolism, and cell death. However, the role and function of RXRs in different heterodimer complexes remain unidentified, mainly because most RXR drugs (called rexinoids) are not selective of specific heterodimer complexes. The lack of selectivity strongly limits the use of rexinoids for specific therapeutic approaches. To better characterize rexinoids at specific NR complexes, we have developed and optimized luciferase (Luc) protein complementation(PCA)-based bioluminescence resonance energy transfer (BRET) assays that can directly measure recruitment of a coactivator (CoA) motif fused to yellow fluorescent protein (YFP) by specific NR dimers. To validate the assays, we compared rexinoid modulation of CoA recruitment by the RXR homodimer and by the heterodimers Nur77/RXR and Nurr1/RXR. Results revealed that some rexinoids display selective CoA recruitment activities with homo- or heterodimer complexes. In particular, SR11237 (BMS649) has stronger potency for recruitment of CoA motif and transcriptional activity with the heterodimer Nur77/RXR than other complexes. This technology should be useful in identifying new compounds with specificity for individual dimeric species formed by NRs.
Collapse
Affiliation(s)
- Xavier C Giner
- *Faculté de Pharmacie and Groupe de Recherche Universitaire sur le Médicament, and Institut de Recherche en Immunologie et Cancérologie, Université de Montréal, Montréal, Québec, Canada
| | - David Cotnoir-White
- *Faculté de Pharmacie and Groupe de Recherche Universitaire sur le Médicament, and Institut de Recherche en Immunologie et Cancérologie, Université de Montréal, Montréal, Québec, Canada
| | - Sylvie Mader
- *Faculté de Pharmacie and Groupe de Recherche Universitaire sur le Médicament, and Institut de Recherche en Immunologie et Cancérologie, Université de Montréal, Montréal, Québec, Canada
| | - Daniel Lévesque
- *Faculté de Pharmacie and Groupe de Recherche Universitaire sur le Médicament, and Institut de Recherche en Immunologie et Cancérologie, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
23
|
Armando S, Quoyer J, Lukashova V, Maiga A, Percherancier Y, Heveker N, Pin JP, Prézeau L, Bouvier M. The chemokine CXC4 and CC2 receptors form homo- and heterooligomers that can engage their signaling G-protein effectors and βarrestin. FASEB J 2014; 28:4509-23. [PMID: 25053617 DOI: 10.1096/fj.13-242446] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
G-protein-coupled receptors have been shown to assemble at least as dimers early in the biosynthetic path, but some evidence suggests that they can also form larger oligomeric complexes. Using the human chemokine receptors CXCR4 and CCR2 as models, we directly probed the existence of higher order homo- and heterooligomers in human embryonic kidney cells. Combining bimolecular fluorescence and luminescence complementation (BiFC, BiLC) with bioluminescence resonance energy transfer (BRET) assays, we show that CXCR4 and CCR2 can assemble as homo- and heterooligomers, forming at least tetramers. Selective activation of CCR2 with the human monocyte chemotactic protein 1 (MCP-1) resulted in trans-conformational rearrangement of the CXCR4 dimer with an EC50 of 19.9 nM, compatible with a CCR2 action. Moreover, MCP-1 promoted the engagement of Gαi1, Gα13, Gαz, and βarrestin2 to the heterooligomer, resulting in calcium signaling that was synergistically potentiated on coactivation of CCR2 and CXCR4, demonstrating that complexes larger than dimers reach the cell surface as functional units. A mutation of CXCR4 (N119K), which prevents Gi activation, also affects the CCR2-promoted engagement of Gαi1 and βarrestin2 by the heterooligomer, supporting the occurrence of transprotomer regulation. Together, the results demonstrate that homo- and heteromultimeric CXCR4 and CCR2 can form functional signaling complexes that have unique properties.
Collapse
Affiliation(s)
- Sylvain Armando
- Department of Biochemistry, Institute for Research in Immunology and Cancer, and Institut de Génomique Fonctionnelle, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 5203, Institut National de la Santé et de la Recherche Médicale (INSERM) U661, University of Montpellier 1 and 2, Montpellier, France
| | - Julie Quoyer
- Department of Biochemistry, Institute for Research in Immunology and Cancer, and
| | - Viktorya Lukashova
- Department of Biochemistry, Institute for Research in Immunology and Cancer, and
| | - Arhamatoulaye Maiga
- Department of Biochemistry, Institute for Research in Immunology and Cancer, and
| | - Yann Percherancier
- Department of Biochemistry, Institute for Research in Immunology and Cancer, and
| | - Nikolaus Heveker
- Department of Biochemistry, Research Centre/Hôpital Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada; and
| | - Jean-Philippe Pin
- Institut de Génomique Fonctionnelle, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 5203, Institut National de la Santé et de la Recherche Médicale (INSERM) U661, University of Montpellier 1 and 2, Montpellier, France
| | - Laurent Prézeau
- Institut de Génomique Fonctionnelle, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 5203, Institut National de la Santé et de la Recherche Médicale (INSERM) U661, University of Montpellier 1 and 2, Montpellier, France
| | - Michel Bouvier
- Department of Biochemistry, Institute for Research in Immunology and Cancer, and
| |
Collapse
|
24
|
Gleghorn ML, Maquat LE. 'Black sheep' that don't leave the double-stranded RNA-binding domain fold. Trends Biochem Sci 2014; 39:328-40. [PMID: 24954387 DOI: 10.1016/j.tibs.2014.05.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 05/19/2014] [Accepted: 05/19/2014] [Indexed: 12/28/2022]
Abstract
The canonical double-stranded RNA (dsRNA)-binding domain (dsRBD) is composed of an α1-β1-β2-β3-α2 secondary structure that folds in three dimensions to recognize dsRNA. Recently, structural and functional studies of divergent dsRBDs revealed adaptations that include intra- and/or intermolecular protein interactions, sometimes in the absence of detectable dsRNA-binding ability. We describe here how discrete dsRBD components can accommodate pronounced amino-acid sequence changes while maintaining the core fold. We exemplify the growing importance of divergent dsRBDs in mRNA decay by discussing Dicer, Staufen (STAU)1 and 2, trans-activation responsive RNA-binding protein (TARBP)2, protein activator of protein kinase RNA-activated (PKR) (PACT), DiGeorge syndrome critical region (DGCR)8, DEAH box helicase proteins (DHX) 9 and 30, and dsRBD-like fold-containing proteins that have ribosome-related functions. We also elaborate on the computational limitations to discovering yet-to-be-identified divergent dsRBDs.
Collapse
Affiliation(s)
- Michael L Gleghorn
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA; Center for RNA Biology, University of Rochester, Rochester, NY 14642, USA
| | - Lynne E Maquat
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA; Center for RNA Biology, University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
25
|
Boulay K, Ghram M, Viranaicken W, Trépanier V, Mollet S, Fréchina C, DesGroseillers L. Cell cycle-dependent regulation of the RNA-binding protein Staufen1. Nucleic Acids Res 2014; 42:7867-83. [PMID: 24906885 PMCID: PMC4081104 DOI: 10.1093/nar/gku506] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Staufen1 (Stau1) is a ribonucleic acid (RNA)-binding protein involved in the post-transcriptional regulation of gene expression. Recent studies indicate that Stau1-bound messenger RNAs (mRNAs) mainly code for proteins involved in transcription and cell cycle control. Consistently, we report here that Stau1 abundance fluctuates through the cell cycle in HCT116 and U2OS cells: it is high from the S phase to the onset of mitosis and rapidly decreases as cells transit through mitosis. Stau1 down-regulation is mediated by the ubiquitin-proteasome system and the E3 ubiquitin ligase anaphase promoting complex/cyclosome (APC/C). Stau1 interacts with the APC/C co-activators Cdh1 and Cdc20 via its first 88 N-terminal amino acids. The importance of controlling Stau155 levels is underscored by the observation that its overexpression affects mitosis entry and impairs proliferation of transformed cells. Microarray analyses identified 275 Stau155-bound mRNAs in prometaphase cells, an early mitotic step that just precedes Stau1 degradation. Interestingly, several of these mRNAs are more abundant in Stau155-containing complexes in cells arrested in prometaphase than in asynchronous cells. Our results point out for the first time to the possibility that Stau1 participates in a mechanism of post-transcriptional regulation of gene expression that is linked to cell cycle progression in cancer cells.
Collapse
Affiliation(s)
- Karine Boulay
- Département de Biochimie, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Mehdi Ghram
- Département de Biochimie, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Wildriss Viranaicken
- Département de Biochimie, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Véronique Trépanier
- Département de Biochimie, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Stéphanie Mollet
- Département de Biochimie, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Céline Fréchina
- Département de Biochimie, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Luc DesGroseillers
- Département de Biochimie, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| |
Collapse
|
26
|
Thomas MG, Pascual ML, Maschi D, Luchelli L, Boccaccio GL. Synaptic control of local translation: the plot thickens with new characters. Cell Mol Life Sci 2014; 71:2219-39. [PMID: 24212248 PMCID: PMC11113725 DOI: 10.1007/s00018-013-1506-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Revised: 10/11/2013] [Accepted: 10/21/2013] [Indexed: 12/18/2022]
Abstract
The production of proteins from mRNAs localized at the synapse ultimately controls the strength of synaptic transmission, thereby affecting behavior and cognitive functions. The regulated transcription, processing, and transport of mRNAs provide dynamic control of the dendritic transcriptome, which includes thousands of messengers encoding multiple cellular functions. Translation is locally modulated by synaptic activity through a complex network of RNA-binding proteins (RBPs) and various types of non-coding RNAs (ncRNAs) including BC-RNAs, microRNAs, piwi-interacting RNAs, and small interference RNAs. The RBPs FMRP and CPEB play a well-established role in synaptic translation, and additional regulatory factors are emerging. The mRNA repressors Smaug, Nanos, and Pumilio define a novel pathway for local translational control that affects dendritic branching and spines in both flies and mammals. Recent findings support a role for processing bodies and related synaptic mRNA-silencing foci (SyAS-foci) in the modulation of synaptic plasticity and memory formation. The SyAS-foci respond to different stimuli with changes in their integrity thus enabling regulated mRNA release followed by translation. CPEB, Pumilio, TDP-43, and FUS/TLS form multimers through low-complexity regions related to prion domains or polyQ expansions. The oligomerization of these repressor RBPs is mechanistically linked to the aggregation of abnormal proteins commonly associated with neurodegeneration. Here, we summarize the current knowledge on how specificity in mRNA translation is achieved through the concerted action of multiple pathways that involve regulatory ncRNAs and RBPs, the modification of translation factors, and mRNA-silencing foci dynamics.
Collapse
Affiliation(s)
- María Gabriela Thomas
- Instituto Leloir, Av. Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- IIBBA-CONICET, C1405BWE Buenos Aires, Argentina
| | - Malena Lucía Pascual
- Instituto Leloir, Av. Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- IIBBA-CONICET, C1405BWE Buenos Aires, Argentina
- Facultad de Ciencias Exactas y Naturales, University of Buenos Aires, Buenos Aires, Argentina
| | - Darío Maschi
- Instituto Leloir, Av. Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- Present Address: Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO USA
| | - Luciana Luchelli
- Instituto Leloir, Av. Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- IIBBA-CONICET, C1405BWE Buenos Aires, Argentina
| | - Graciela Lidia Boccaccio
- Instituto Leloir, Av. Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
- IIBBA-CONICET, C1405BWE Buenos Aires, Argentina
- Facultad de Ciencias Exactas y Naturales, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
27
|
|
28
|
Staufen1 senses overall transcript secondary structure to regulate translation. Nat Struct Mol Biol 2013; 21:26-35. [PMID: 24336223 PMCID: PMC4605437 DOI: 10.1038/nsmb.2739] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 11/19/2013] [Indexed: 12/31/2022]
Abstract
Human Staufen1 (Stau1) is a double-stranded RNA (dsRNA)-binding protein implicated in multiple post-transcriptional gene-regulatory processes. Here we combined RNA immunoprecipitation in tandem (RIPiT) with RNase footprinting, formaldehyde cross-linking, sonication-mediated RNA fragmentation and deep sequencing to map Staufen1-binding sites transcriptome wide. We find that Stau1 binds complex secondary structures containing multiple short helices, many of which are formed by inverted Alu elements in annotated 3' untranslated regions (UTRs) or in 'strongly distal' 3' UTRs. Stau1 also interacts with actively translating ribosomes and with mRNA coding sequences (CDSs) and 3' UTRs in proportion to their GC content and propensity to form internal secondary structure. On mRNAs with high CDS GC content, higher Stau1 levels lead to greater ribosome densities, thus suggesting a general role for Stau1 in modulating translation elongation through structured CDS regions. Our results also indicate that Stau1 regulates translation of transcription-regulatory proteins.
Collapse
|
29
|
mRNA-mRNA duplexes that autoelicit Staufen1-mediated mRNA decay. Nat Struct Mol Biol 2013; 20:1214-20. [PMID: 24056942 PMCID: PMC3947523 DOI: 10.1038/nsmb.2664] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 08/08/2013] [Indexed: 12/28/2022]
Abstract
We report a new mechanism by which human mRNAs crosstalk: an Alu element in the 3'-untranslated region (3' UTR) of one mRNA can base-pair with a partially complementary Alu element in the 3' UTR of a different mRNA thereby creating a Staufen1 (STAU1)-binding site (SBS). STAU1 binding to a 3' UTR SBS was previously shown to trigger STAU1-mediated mRNA decay (SMD) by directly recruiting the ATP-dependent RNA helicase UPF1, which is also a key factor in the mechanistically related nonsense-mediated mRNA decay (NMD) pathway. In the case of a 3' UTR SBS created via mRNA–mRNA base-pairing, we show that SMD targets both mRNAs in the duplex provided that both mRNAs are translated. If only one mRNA is translated, then it alone is targeted for SMD. We demonstrate the importance of mRNA–mRNA-triggered SMD to the processes of cell migration and invasion.
Collapse
|
30
|
Elbarbary RA, Li W, Tian B, Maquat LE. STAU1 binding 3' UTR IRAlus complements nuclear retention to protect cells from PKR-mediated translational shutdown. Genes Dev 2013; 27:1495-510. [PMID: 23824540 PMCID: PMC3713430 DOI: 10.1101/gad.220962.113] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 05/29/2013] [Indexed: 11/24/2022]
Abstract
For a number of human genes that encode transcripts containing inverted repeat Alu elements (IRAlus) within their 3' untranslated region (UTR), product mRNA is efficiently exported to the cytoplasm when the IRAlus, which mediate nuclear retention, are removed by alternative polyadenylation. Here we report a new mechanism that promotes gene expression by targeting mRNAs that maintain their 3' UTR IRAlus: Binding of the dsRNA-binding protein Staufen1 (STAU1) to 3' UTR IRAlus inhibits nuclear retention so as to augment the nuclear export of 3' UTR IRAlus-containing mRNAs (IRAlus mRNAs). Moreover, we found that 3' UTR IRAlus-bound STAU1 enhances 3' UTR IRAlus mRNA translation by precluding protein kinase R (PKR) binding, which obviates PKR activation, eukaryotic translation initiation factor 2α (eIF2α) phosphorylation, and repression of global cell translation. Thus, STAU1 binding to 3' UTR IRAlus functions along with 3' UTR IRAlus-mediated nuclear retention to suppress the shutdown of cellular translation triggered by PKR binding to endogenous cytoplasmic dsRNAs. We also show that a changing STAU1/PKR ratio contributes to myogenesis via effects on the 3' UTR IRAlus of mRNA encoding the microRNA-binding protein LIN28.
Collapse
Affiliation(s)
- Reyad A. Elbarbary
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry
- Center for RNA Biology, University of Rochester, Rochester, New York 14642, USA
| | - Wencheng Li
- Department of Biochemistry and Molecular Biology, University of Medicine and Dentistry of New Jersey–New Jersey Medical School, Newark, New Jersey 07101, USA
| | - Bin Tian
- Department of Biochemistry and Molecular Biology, University of Medicine and Dentistry of New Jersey–New Jersey Medical School, Newark, New Jersey 07101, USA
| | - Lynne E. Maquat
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry
- Center for RNA Biology, University of Rochester, Rochester, New York 14642, USA
| |
Collapse
|
31
|
Park E, Maquat LE. Staufen-mediated mRNA decay. WILEY INTERDISCIPLINARY REVIEWS-RNA 2013; 4:423-35. [PMID: 23681777 DOI: 10.1002/wrna.1168] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 03/28/2013] [Accepted: 03/28/2013] [Indexed: 12/26/2022]
Abstract
Staufen1 (STAU1)-mediated mRNA decay (SMD) is an mRNA degradation process in mammalian cells that is mediated by the binding of STAU1 to a STAU1-binding site (SBS) within the 3'-untranslated region (3'-UTR) of target mRNAs. During SMD, STAU1, a double-stranded (ds) RNA-binding protein, recognizes dsRNA structures formed either by intramolecular base pairing of 3'-UTR sequences or by intermolecular base pairing of 3'-UTR sequences with a long-noncoding RNA (lncRNA) via partially complementary Alu elements. Recently, STAU2, a paralog of STAU1, has also been reported to mediate SMD. Both STAU1 and STAU2 interact directly with the ATP-dependent RNA helicase UPF1, a key SMD factor, enhancing its helicase activity to promote effective SMD. Moreover, STAU1 and STAU2 form homodimeric and heterodimeric interactions via domain-swapping. Because both SMD and the mechanistically related nonsense-mediated mRNA decay (NMD) employ UPF1; SMD and NMD are competitive pathways. Competition contributes to cellular differentiation processes, such as myogenesis and adipogenesis, placing SMD at the heart of various physiologically important mechanisms.
Collapse
Affiliation(s)
- Eonyoung Park
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, Center for RNA Biology, University of Rochester, Rochester, NY, USA
| | | |
Collapse
|
32
|
Abstract
Staufen1-mediated mRNA decay (SMD) degrades mRNAs that harbor a Staufen1-binding site (SBS) in their 3' untranslated regions (UTRs). Human SBSs can form by intermolecular base-pairing between a 3' UTR Alu element and an Alu element within a long noncoding RNA (lncRNA) called a ½-sbsRNA. Since Alu elements are confined to primates, it was unclear how SMD occurs in rodents. Here we identify mouse mRNA 3' UTRs and lncRNAs that contain a B1, B2, B4, or identifier (ID) element. We show that SMD occurs in mouse cells via mRNA-lncRNA base-pairing of partially complementary elements and that mouse ½-sbsRNA (m½-sbsRNA)-triggered SMD regulates C2C12 cell myogenesis. Our findings define new roles for lncRNAs as well as B and ID short interspersed elements (SINEs) in mice that undoubtedly influence many developmental and homeostatic pathways.
Collapse
Affiliation(s)
- Jiashi Wang
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, USA
| | | | | |
Collapse
|
33
|
Staufen1 dimerizes through a conserved motif and a degenerate dsRNA-binding domain to promote mRNA decay. Nat Struct Mol Biol 2013; 20:515-24. [PMID: 23524536 PMCID: PMC4096160 DOI: 10.1038/nsmb.2528] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 02/05/2013] [Indexed: 11/23/2022]
Abstract
Staufen (STAU)1-mediated mRNA decay (SMD) degrades mammalian-cell mRNAs that bind the double-stranded (ds)RNA-binding protein STAU1 in their 3′-untranslated region. We report a new motif, which typifies STAU homologs from all vertebrate classes, that is responsible for human (h)STAU1 homodimerization. Our crystal structure and mutagenesis analyses reveal that this motif, now named the Staufen-swapping motif (SSM), and dsRNA-binding domain 5 (‘RBD’5) mediate protein dimerization: the two SSM α-helices of one molecule interact primarily through a hydrophobic patch with the two ‘RBD’5 α-helices of a second molecule. ‘RBD’5 adopts the canonical α-β-β-β-α fold of a functional RBD, but it lacks residues and features needed to bind duplex RNA. In cells, SSM-mediated hSTAU1 dimerization increases the efficiency of SMD by augmenting hSTAU1 binding to the ATP-dependent RNA helicase hUPF1. Dimerization regulates keratinocyte-mediated wound-healing and, undoubtedly, many other cellular processes.
Collapse
|
34
|
Dassi E, Zuccotti P, Leo S, Provenzani A, Assfalg M, D’Onofrio M, Riva P, Quattrone A. Hyper conserved elements in vertebrate mRNA 3'-UTRs reveal a translational network of RNA-binding proteins controlled by HuR. Nucleic Acids Res 2013; 41:3201-16. [PMID: 23376935 PMCID: PMC3597683 DOI: 10.1093/nar/gkt017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 12/20/2012] [Accepted: 12/26/2012] [Indexed: 02/06/2023] Open
Abstract
Little is known regarding the post-transcriptional networks that control gene expression in eukaryotes. Additionally, we still need to understand how these networks evolve, and the relative role played in them by their sequence-dependent regulatory factors, non-coding RNAs (ncRNAs) and RNA-binding proteins (RBPs). Here, we used an approach that relied on both phylogenetic sequence sharing and conservation in the whole mapped 3'-untranslated regions (3'-UTRs) of vertebrate species to gain knowledge on core post-transcriptional networks. The identified human hyper conserved elements (HCEs) were predicted to be preferred binding sites for RBPs and not for ncRNAs, namely microRNAs and long ncRNAs. We found that the HCE map identified a well-known network that post-transcriptionally regulates histone mRNAs. We were then able to discover and experimentally confirm a translational network composed of RNA Recognition Motif (RRM)-type RBP mRNAs that are positively controlled by HuR, another RRM-type RBP. HuR shows a preference for these RBP mRNAs bound in stem-loop motifs, confirming its role as a 'regulator of regulators'. Analysis of the transcriptome-wide HCE distribution revealed a profile of prevalently small clusters separated by unconserved intercluster RNA stretches, which predicts the formation of discrete small ribonucleoprotein complexes in the 3'-UTRs.
Collapse
Affiliation(s)
- Erik Dassi
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, via delle Regole, 101 38123 Mattarello (TN) Italy, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, via Viotti, 3/5 20133 Milano, Italy, Laboratory of Genomic Screening, Centre for Integrative Biology, University of Trento, Trento, via delle Regole, 101 38123 Mattarello (TN) Italy and Department of Biotechnology, University of Verona, Province of Verona, Ca' Vignal 1, Strada Le Grazie 15 37134 Verona, Italy
| | - Paola Zuccotti
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, via delle Regole, 101 38123 Mattarello (TN) Italy, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, via Viotti, 3/5 20133 Milano, Italy, Laboratory of Genomic Screening, Centre for Integrative Biology, University of Trento, Trento, via delle Regole, 101 38123 Mattarello (TN) Italy and Department of Biotechnology, University of Verona, Province of Verona, Ca' Vignal 1, Strada Le Grazie 15 37134 Verona, Italy
| | - Sara Leo
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, via delle Regole, 101 38123 Mattarello (TN) Italy, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, via Viotti, 3/5 20133 Milano, Italy, Laboratory of Genomic Screening, Centre for Integrative Biology, University of Trento, Trento, via delle Regole, 101 38123 Mattarello (TN) Italy and Department of Biotechnology, University of Verona, Province of Verona, Ca' Vignal 1, Strada Le Grazie 15 37134 Verona, Italy
| | - Alessandro Provenzani
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, via delle Regole, 101 38123 Mattarello (TN) Italy, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, via Viotti, 3/5 20133 Milano, Italy, Laboratory of Genomic Screening, Centre for Integrative Biology, University of Trento, Trento, via delle Regole, 101 38123 Mattarello (TN) Italy and Department of Biotechnology, University of Verona, Province of Verona, Ca' Vignal 1, Strada Le Grazie 15 37134 Verona, Italy
| | - Michael Assfalg
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, via delle Regole, 101 38123 Mattarello (TN) Italy, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, via Viotti, 3/5 20133 Milano, Italy, Laboratory of Genomic Screening, Centre for Integrative Biology, University of Trento, Trento, via delle Regole, 101 38123 Mattarello (TN) Italy and Department of Biotechnology, University of Verona, Province of Verona, Ca' Vignal 1, Strada Le Grazie 15 37134 Verona, Italy
| | - Mariapina D’Onofrio
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, via delle Regole, 101 38123 Mattarello (TN) Italy, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, via Viotti, 3/5 20133 Milano, Italy, Laboratory of Genomic Screening, Centre for Integrative Biology, University of Trento, Trento, via delle Regole, 101 38123 Mattarello (TN) Italy and Department of Biotechnology, University of Verona, Province of Verona, Ca' Vignal 1, Strada Le Grazie 15 37134 Verona, Italy
| | - Paola Riva
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, via delle Regole, 101 38123 Mattarello (TN) Italy, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, via Viotti, 3/5 20133 Milano, Italy, Laboratory of Genomic Screening, Centre for Integrative Biology, University of Trento, Trento, via delle Regole, 101 38123 Mattarello (TN) Italy and Department of Biotechnology, University of Verona, Province of Verona, Ca' Vignal 1, Strada Le Grazie 15 37134 Verona, Italy
| | - Alessandro Quattrone
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, via delle Regole, 101 38123 Mattarello (TN) Italy, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, via Viotti, 3/5 20133 Milano, Italy, Laboratory of Genomic Screening, Centre for Integrative Biology, University of Trento, Trento, via delle Regole, 101 38123 Mattarello (TN) Italy and Department of Biotechnology, University of Verona, Province of Verona, Ca' Vignal 1, Strada Le Grazie 15 37134 Verona, Italy
| |
Collapse
|
35
|
Staufen2 functions in Staufen1-mediated mRNA decay by binding to itself and its paralog and promoting UPF1 helicase but not ATPase activity. Proc Natl Acad Sci U S A 2012; 110:405-12. [PMID: 23263869 DOI: 10.1073/pnas.1213508110] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Staufen (STAU)1-mediated mRNA decay (SMD) is a posttranscriptional regulatory mechanism in mammals that degrades mRNAs harboring a STAU1-binding site (SBS) in their 3'-untranslated regions (3' UTRs). We show that SMD involves not only STAU1 but also its paralog STAU2. STAU2, like STAU1, is a double-stranded RNA-binding protein that interacts directly with the ATP-dependent RNA helicase up-frameshift 1 (UPF1) to reduce the half-life of SMD targets that form an SBS by either intramolecular or intermolecular base-pairing. Compared with STAU1, STAU2 binds ~10-fold more UPF1 and ~two- to fivefold more of those SBS-containing mRNAs that were tested, and it comparably promotes UPF1 helicase activity, which is critical for SMD. STAU1- or STAU2-mediated augmentation of UPF1 helicase activity is not accompanied by enhanced ATP hydrolysis but does depend on ATP binding and a basal level of UPF1 ATPase activity. Studies of STAU2 demonstrate it changes the conformation of RNA-bound UPF1. These findings, and evidence for STAU1-STAU1, STAU2-STAU2, and STAU1-STAU2 formation in vitro and in cells, are consistent with results from tethering assays: the decrease in mRNA abundance brought about by tethering siRNA-resistant STAU2 or STAU1 to an mRNA 3' UTR is inhibited by downregulating the abundance of cellular STAU2, STAU1, or UPF1. It follows that the efficiency of SMD in different cell types reflects the cumulative abundance of STAU1 and STAU2. We propose that STAU paralogs contribute to SMD by "greasing the wheels" of RNA-bound UPF1 so as to enhance its unwinding capacity per molecule of ATP hydrolyzed.
Collapse
|
36
|
Patel S, Blose JM, Sokoloski JE, Pollack L, Bevilacqua PC. Specificity of the double-stranded RNA-binding domain from the RNA-activated protein kinase PKR for double-stranded RNA: insights from thermodynamics and small-angle X-ray scattering. Biochemistry 2012; 51:9312-22. [PMID: 23140277 DOI: 10.1021/bi300935p] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The interferon-inducible, double-stranded (ds) RNA-activated protein kinase (PKR) contains a dsRNA-binding domain (dsRBD) and plays key roles in viral pathogenesis and innate immunity. Activation of PKR is typically mediated by long dsRNA, and regulation of PKR is disfavored by most RNA imperfections, including bulges and internal loops. Herein, we combine isothermal titration calorimetry (ITC), electrophoretic mobility shift assays, and small-angle X-ray scattering (SAXS) to dissect the thermodynamic basis for the specificity of the dsRBD termed "p20" for various RNAs and to detect any RNA conformational changes induced upon protein binding. We monitor binding of p20 to chimeric duplexes containing terminal RNA-DNA hybrid segments and a central dsRNA segment, which was either unbulged ("perfect") or bulged. The ITC data reveal strong binding of p20 to the perfect duplex (K(d) ~ 30 nM) and weaker binding to the bulged duplex (K(d) ~ 2-5 μM). SAXS reconstructions and p(r) distance distribution functions further uncover that p20 induces no significant conformational change in perfect dsRNA but largely straightens bulged dsRNA. Together, these observations support the dsRBD's ability to tightly bind to only A-form RNA and suggest that in a noninfected cell, PKR may be buffered via weak interactions with various bulged and looped RNAs, which it may straighten. This work suggests that PKR-regulating RNAs with complex secondary and tertiary structures likely mimic dsRNA and/or engage portions of PKR outside of the dsRBD.
Collapse
Affiliation(s)
- Sunita Patel
- Department of Chemistry and Center for RNA Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | | | | | | | | |
Collapse
|
37
|
Milev MP, Ravichandran M, Khan MF, Schriemer DC, Mouland AJ. Characterization of staufen1 ribonucleoproteins by mass spectrometry and biochemical analyses reveal the presence of diverse host proteins associated with human immunodeficiency virus type 1. Front Microbiol 2012; 3:367. [PMID: 23125841 PMCID: PMC3486646 DOI: 10.3389/fmicb.2012.00367] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 09/27/2012] [Indexed: 12/02/2022] Open
Abstract
The human immunodeficiency virus type 1 (HIV-1) unspliced, 9 kb genomic RNA (vRNA) is exported from the nucleus for the synthesis of viral structural proteins and enzymes (Gag and Gag/Pol) and is then transported to sites of virus assembly where it is packaged into progeny virions. vRNA co-exists in the cytoplasm in the context of the HIV-1 ribonucleoprotein (RNP) that is currently defined by the presence of Gag and several host proteins including the double-stranded RNA-binding protein, Staufen1. In this study we isolated Staufen1 RNP complexes derived from HIV-1-expressing cells using tandem affinity purification and have identified multiple host protein components by mass spectrometry. Four viral proteins, including Gag, Gag/Pol, Env and Nef as well as >200 host proteins were identified in these RNPs. Moreover, HIV-1 induces both qualitative and quantitative differences in host protein content in these RNPs. 22% of Staufen1-associated factors are virion-associated suggesting that the RNP could be a vehicle to achieve this. In addition, we provide evidence on how HIV-1 modulates the composition of cytoplasmic Staufen1 RNPs. Biochemical fractionation by density gradient analyses revealed new facets on the assembly of Staufen1 RNPs. The assembly of dense Staufen1 RNPs that contain Gag and several host proteins were found to be entirely RNA-dependent but their assembly appeared to be independent of Gag expression. Gag-containing complexes fractionated into a lighter and another, more dense pool. Lastly, Staufen1 depletion studies demonstrated that the previously characterized Staufen1 HIV-1-dependent RNPs are most likely aggregates of smaller RNPs that accumulate at juxtanuclear domains. The molecular characterization of Staufen1 HIV-1 RNPs will offer important information on virus-host cell interactions and on the elucidation of the function of these RNPs for the transport of Gag and the fate of the unspliced vRNA in HIV-1-producing cells.
Collapse
Affiliation(s)
- Miroslav P Milev
- HIV-1 Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital Montréal, QC, Canada ; Division of Experimental Medicine, Department of Medicine, McGill University Montreal, QC, Canada
| | | | | | | | | |
Collapse
|
38
|
Ravel-Chapuis A, Bélanger G, Yadava RS, Mahadevan MS, DesGroseillers L, Côté J, Jasmin BJ. The RNA-binding protein Staufen1 is increased in DM1 skeletal muscle and promotes alternative pre-mRNA splicing. ACTA ACUST UNITED AC 2012; 196:699-712. [PMID: 22431750 PMCID: PMC3308689 DOI: 10.1083/jcb.201108113] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Staufen1 interacts with mRNAs with expanded CUG repeats and promotes their nuclear export and translation, while also promoting alternative splicing of other mRNAs. In myotonic dystrophy type 1 (DM1), dystrophia myotonica protein kinase messenger ribonucleic acids (RNAs; mRNAs) with expanded CUG repeats (CUGexp) aggregate in the nucleus and become toxic to cells by sequestering and/or misregulating RNA-binding proteins, resulting in aberrant alternative splicing. In this paper, we find that the RNA-binding protein Staufen1 is markedly and specifically increased in skeletal muscle from DM1 mouse models and patients. We show that Staufen1 interacts with mutant CUGexp mRNAs and promotes their nuclear export and translation. This effect is critically dependent on the third double-stranded RNA–binding domain of Staufen1 and shuttling of Staufen1 into the nucleus via its nuclear localization signal. Moreover, we uncover a new role of Staufen1 in splicing regulation. Overexpression of Staufen1 rescues alternative splicing of two key pre-mRNAs known to be aberrantly spliced in DM1, suggesting its increased expression represents an adaptive response to the pathology. Altogether, our results unravel a novel function for Staufen1 in splicing regulation and indicate that it may positively modulate the complex DM1 phenotype, thereby revealing its potential as a therapeutic target.
Collapse
Affiliation(s)
- Aymeric Ravel-Chapuis
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada
| | | | | | | | | | | | | |
Collapse
|
39
|
Lin CL, Huang YT, Richter JD. Transient CPEB dimerization and translational control. RNA (NEW YORK, N.Y.) 2012; 18:1050-1061. [PMID: 22456264 PMCID: PMC3334692 DOI: 10.1261/rna.031682.111] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Accepted: 02/16/2012] [Indexed: 05/29/2023]
Abstract
During oocyte development, the cytoplasmic polyadenylation element-binding protein (CPEB) nucleates a set of factors on mRNA that controls cytoplasmic polyadenylation and translation. The regulation of polyadenylation is mediated in part through serial phosphorylations of CPEB, which control both the dynamic integrity of the cytoplasmic polyadenylation apparatus and CPEB stability, events necessary for meiotic progression. Because the precise stoichiometry between CPEB and CPE-containing RNA is responsible for the temporal order of mRNA polyadenylation during meiosis, we hypothesized that, if CPEB production exceeded the amount required to bind mRNA, the excess would be sequestered in an inactive form. One attractive possibility for the sequestration is protein dimerization. We demonstrate that not only does CPEB form a dimer, but dimerization requires its RNA-binding domains. Dimer formation prevents CPEB from being UV cross-linked to RNA, which establishes a second pool of CPEB that is inert for polyadenylation and translational control. During oocyte maturation, the dimers are degraded much more rapidly than the CPEB monomers, due to their greater affinity for polo-like kinase 1 (plx1) and the ubiquitin E3 ligase β-TrCP. Because dimeric CPEB also binds cytoplasmic polyadenylation factors with greater affinity than monomeric CPEB, it may act as a hub or reservoir for the polyadenylation machinery. We propose that the balance between CPEB and its target mRNAs is maintained by CPEB dimerization, which inactivates spare proteins and prevents them from inducing polyadenylation of RNAs with low affinity binding sites. In addition, the dimers might serve as molecular hubs that release polyadenylation factors for translational activation upon CPEB dimer destruction.
Collapse
Affiliation(s)
- Chien-Ling Lin
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Yen-Tsung Huang
- Department of Epidemiology and
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | - Joel D. Richter
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| |
Collapse
|
40
|
Breton B, Sauvageau É, Zhou J, Bonin H, Le Gouill C, Bouvier M. Multiplexing of multicolor bioluminescence resonance energy transfer. Biophys J 2011; 99:4037-46. [PMID: 21156147 DOI: 10.1016/j.bpj.2010.10.025] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 09/30/2010] [Accepted: 10/15/2010] [Indexed: 10/18/2022] Open
Abstract
Bioluminescence resonance energy transfer (BRET) is increasingly being used to monitor protein-protein interactions and cellular events in cells. However, the ability to monitor multiple events simultaneously is limited by the spectral properties of the existing BRET partners. Taking advantage of newly developed Renilla luciferases and blue-shifted fluorescent proteins (FPs), we explored the possibility of creating novel BRET configurations using a single luciferase substrate and distinct FPs. Three new (to our knowledge) BRET assays leading to distinct color bioluminescence emission were generated and validated. The spectral properties of two of the FPs used (enhanced blue (EB) FP2 and mAmetrine) and the selection of appropriate detection filters permitted the concomitant detection of two independent BRET signals, without cross-interference, in the same cells after addition of a unique substrate for Renilla luciferase-II, coelentrazine-400a. Using individual BRET-based biosensors to monitor the interaction between G-protein-coupled receptors and G-protein subunits or activation of different G-proteins along with the production of a second messenger, we established the proof of principle that two new BRET configurations can be multiplexed to simultaneously monitor two dependent or independent cellular events. The development of this new multiplexed BRET configuration opens the way for concomitant monitoring of various independent biological processes in living cells.
Collapse
Affiliation(s)
- Billy Breton
- Department of Biochemistry, Institute for Research in Immunology and Cancer, and Groupe de Recherche Universitaire sur le Médicament, Université de Montréal, Montréal, Canada
| | | | | | | | | | | |
Collapse
|
41
|
Chung S, Takizawa PA. Multiple Myo4 motors enhance ASH1 mRNA transport in Saccharomyces cerevisiae. ACTA ACUST UNITED AC 2010; 189:755-67. [PMID: 20457760 PMCID: PMC2872910 DOI: 10.1083/jcb.200912011] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In Saccharomyces cerevisiae, ASH1 mRNA is transported to the bud tip by the class V myosin Myo4. In vivo, Myo4 moves RNA in a rapid and continuous fashion, but in vitro Myo4 is a nonprocessive, monomeric motor that forms a complex with She3. To understand how nonprocessive motors generate continuous transport, we used a novel purification method to show that Myo4, She3, and the RNA-binding protein She2 are the sole major components of an active ribonucleoprotein transport unit. We demonstrate that a single localization element contains multiple copies of Myo4 and a tetramer of She2, which suggests that She2 may recruit multiple motors to an RNA. Furthermore, we show that increasing the number of Myo4-She3 molecules bound to ASH1 RNA in the absence of She2 increases the efficiency of RNA transport to the bud. Our data suggest that multiple, nonprocessive Myo4 motors can generate continuous transport of mRNA to the bud tip.
Collapse
Affiliation(s)
- Sunglan Chung
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | |
Collapse
|