1
|
Serganov AA, Udi Y, Stein ME, Patel V, Fridy PC, Rice CM, Saeed M, Jacobs EY, Chait BT, Rout MP. Proteomic elucidation of the targets and primary functions of the picornavirus 2A protease. J Biol Chem 2022; 298:101882. [PMID: 35367208 PMCID: PMC9168619 DOI: 10.1016/j.jbc.2022.101882] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 03/04/2022] [Accepted: 03/06/2022] [Indexed: 11/19/2022] Open
Abstract
Picornaviruses are small RNA viruses that hijack host cell machinery to promote their replication. During infection, these viruses express two proteases, 2Apro and 3Cpro, which process viral proteins. They also subvert a number of host functions, including innate immune responses, host protein synthesis, and intracellular transport, by utilizing poorly understood mechanisms for rapidly and specifically targeting critical host proteins. Here, we used proteomic tools to characterize 2Apro interacting partners, functions, and targeting mechanisms. Our data indicate that, initially, 2Apro primarily targets just two cellular proteins: eukaryotic translation initiation factor eIF4G (a critical component of the protein synthesis machinery) and Nup98 (an essential component of the nuclear pore complex, responsible for nucleocytoplasmic transport). The protease appears to employ two different cleavage mechanisms; it likely interacts with eIF3L, utilizing the eIF3 complex to proteolytically access the eIF4G protein but also directly binds and degrades Nup98. This Nup98 cleavage results in only a marginal effect on nuclear import of proteins, while nuclear export of proteins and mRNAs were more strongly affected. Collectively, our data indicate that 2Apro selectively inhibits protein translation, key nuclear export pathways, and cellular mRNA localization early in infection to benefit viral replication at the expense of particular cell functions.
Collapse
Affiliation(s)
- Artem A Serganov
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA
| | - Yael Udi
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA.
| | - Milana E Stein
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA
| | - Valay Patel
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA
| | - Peter C Fridy
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, USA
| | - Mohsan Saeed
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, USA; Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA; National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston University, Massachusetts, USA.
| | - Erica Y Jacobs
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, New York, USA; Chemistry Department, St John's University, Queens, New York, USA.
| | - Brian T Chait
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, New York, USA.
| | - Michael P Rout
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA.
| |
Collapse
|
2
|
Wang Y, Liu R, Liao J, Jiang L, Jeong GH, Zhou L, Polite M, Duong D, Seyfried NT, Wang H, Kiyokawa H, Yin J. Orthogonal ubiquitin transfer reveals human papillomavirus E6 downregulates nuclear transport to disarm interferon-γ dependent apoptosis of cervical cancer cells. FASEB J 2021; 35:e21986. [PMID: 34662469 DOI: 10.1096/fj.202101232rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 02/05/2023]
Abstract
The E6 protein of the human papillomavirus (HPV) underpins important protein interaction networks between the virus and host to promote viral infection. Through its interaction with E6AP, a host E3 ubiquitin (UB) ligase, E6 stirs the protein ubiquitination pathways toward the oncogenic transformation of the infected cells. For a systematic measurement of E6 reprogramming of the substrate pool of E6AP, we performed a proteomic screen based on "orthogonal UB transfer (OUT)" that allowed us to identify the ubiquitination targets of E6AP dependent on the E6 protein of HPV-16, a high-risk viral subtype for the development of cervical cancer. The OUT screen identified more than 200 potential substrates of the E6-E6AP pair based on the transfer of UB from E6AP to the substrate proteins. Among them, we verified that E6 would induce E6AP-catalyzed ubiquitination of importin proteins KPNA1-3, protein phosphatase PGAM5, and arginine methyltransferases CARM1 to trigger their degradation by the proteasome. We further found that E6 could significantly reduce the cellular level of KPNA1 that resulted in the suppression of nuclear transport of phosphorylated STAT1 and the inhibition of interferon-γ-induced apoptosis in cervical cancer cells. Overall, our work demonstrates OUT as a powerful proteomic platform to probe the interaction of E6 and host cells through protein ubiquitination and reveals a new role of E6 in down-regulating nuclear transport proteins to attenuate tumor-suppressive signaling.
Collapse
Affiliation(s)
- Yiyang Wang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Ruochuan Liu
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Jia Liao
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Lucen Jiang
- Department of Pathology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Geon H Jeong
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Li Zhou
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Monica Polite
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Duc Duong
- Integrated Proteomics Core, Emory University, Atlanta, Georgia, USA
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Huadong Wang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, China
| | - Hiroaki Kiyokawa
- Department of Pharmacology, Northwestern University, Chicago, Illinois, USA
| | - Jun Yin
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| |
Collapse
|
3
|
Mehmood R, Jibiki K, Shibazaki N, Yasuhara N. Molecular profiling of nucleocytoplasmic transport factor genes in breast cancer. Heliyon 2021; 7:e06039. [PMID: 33553736 PMCID: PMC7851789 DOI: 10.1016/j.heliyon.2021.e06039] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 12/14/2020] [Accepted: 01/14/2021] [Indexed: 11/24/2022] Open
Abstract
Transport of functional molecules across the nuclear membrane of a eukaryotic cell is regulated by a dedicated set of transporter proteins that carry molecules into the nucleus or out of the nucleus to the cytoplasm for homeostasis of the cell. One of the categories of cargo molecules these transporters carry are the molecules for cell cycle regulation. Therefore, their role is critical in terms of cancer development. Any misregulation of the transport factors would means aberrant abundance of cell cycle regulators and might have consequences in cell cycle progression. While earlier studies have focussed on individual transport related molecules, a collective overview of how these molecules may be dysregulated in breast cancer is lacking. Using genomic and transcriptomic datasets from TCGA (The Cancer Genome Atlas) and microarray platforms, we carried out bioinformatic analysis and provide a genetic and molecular profile of all the molecules directly related to nucleocytoplasmic shuttling of proteins and RNAs. Interestingly, we identified that many of these molecules are either mutated or have dysregulated expression in breast cancer. Strikingly, some of the molecules, namely, KPNA2, KPNA3, KPNA5, IPO8, TNPO1, XPOT, XPO7 and CSE1L were correlated with poor patient survival. This study provides a comprehensive genetic and molecular landscape of nucleocytoplasmic factors in breast cancer and points to the important roles of various nucleocytoplasmic factors in cancer progression. This data might have implications in prognosis and therapeutic targeting in breast cancer.
Collapse
Affiliation(s)
- Rashid Mehmood
- Department of Life Sciences, College of Science and General Studies, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
| | - Kazuya Jibiki
- Graduate School of Integrated Basic Sciences, Nihon University, Setagaya-ku, Tokyo, Japan
| | - Noriko Shibazaki
- Graduate School of Integrated Basic Sciences, Nihon University, Setagaya-ku, Tokyo, Japan
| | - Noriko Yasuhara
- Graduate School of Integrated Basic Sciences, Nihon University, Setagaya-ku, Tokyo, Japan
| |
Collapse
|
4
|
Smith KM, Tsimbalyuk S, Edwards MR, Cross EM, Batra J, Soares da Costa TP, Aragão D, Basler CF, Forwood JK. Structural basis for importin alpha 3 specificity of W proteins in Hendra and Nipah viruses. Nat Commun 2018; 9:3703. [PMID: 30209309 PMCID: PMC6135763 DOI: 10.1038/s41467-018-05928-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 06/29/2018] [Indexed: 12/14/2022] Open
Abstract
Seven human isoforms of importin α mediate nuclear import of cargo in a tissue- and isoform-specific manner. How nuclear import adaptors differentially interact with cargo harbouring the same nuclear localisation signal (NLS) remains poorly understood, as the NLS recognition region is highly conserved. Here, we provide a structural basis for the nuclear import specificity of W proteins in Hendra and Nipah viruses. We determine the structural interfaces of these cargo bound to importin α1 and α3, identifying a 2.4-fold more extensive interface and > 50-fold higher binding affinity for importin α3. Through the design of importin α1 and α3 chimeric and mutant proteins, together with structures of cargo-free importin α1 and α3 isoforms, we establish that the molecular basis of specificity resides in the differential positioning of the armadillo repeats 7 and 8. Overall, our study provides mechanistic insights into a range of important nucleocytoplasmic transport processes reliant on isoform adaptor specificity.
Collapse
Affiliation(s)
- Kate M Smith
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia
| | - Sofiya Tsimbalyuk
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia
| | - Megan R Edwards
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Emily M Cross
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia
| | - Jyoti Batra
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Tatiana P Soares da Costa
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - David Aragão
- Australian Synchrotron, Australian Nuclear Science and Technology Organisation, 800 Blackburn Road, Clayton, VIC, 3168, Australia
| | - Christopher F Basler
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA.
| | - Jade K Forwood
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia.
| |
Collapse
|
5
|
Cabot B, Cabot RA. Chromatin remodeling in mammalian embryos. Reproduction 2018; 155:R147-R158. [PMID: 29339454 DOI: 10.1530/rep-17-0488] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 01/12/2018] [Indexed: 12/28/2022]
Abstract
The mammalian embryo undergoes a dramatic amount of epigenetic remodeling during the first week of development. In this review, we discuss several epigenetic changes that happen over the course of cleavage development, focusing on covalent marks (e.g., histone methylation and acetylation) and non-covalent remodeling (chromatin remodeling via remodeling complexes; e.g., SWI/SNF-mediated chromatin remodeling). Comparisons are also drawn between remodeling events that occur in embryos from a variety of mammalian species.
Collapse
Affiliation(s)
- Birgit Cabot
- Department of Animal SciencesPurdue University, West Lafayette, Indiana, USA
| | - Ryan A Cabot
- Department of Animal SciencesPurdue University, West Lafayette, Indiana, USA
| |
Collapse
|
6
|
Koyama M, Matsuura Y. Crystal structure of importin-α3 bound to the nuclear localization signal of Ran-binding protein 3. Biochem Biophys Res Commun 2017; 491:609-613. [DOI: 10.1016/j.bbrc.2017.07.155] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 07/27/2017] [Indexed: 12/12/2022]
|
7
|
Abstract
The human genome encodes seven isoforms of importin α which are grouped into three subfamilies known as α1, α2 and α3. All isoforms share a fundamentally conserved architecture that consists of an N-terminal, autoinhibitory, importin-β-binding (IBB) domain and a C-terminal Arm (Armadillo)-core that associates with nuclear localization signal (NLS) cargoes. Despite striking similarity in amino acid sequence and 3D structure, importin-α isoforms display remarkable substrate specificity in vivo. In the present review, we look at key differences among importin-α isoforms and provide a comprehensive inventory of known viral and cellular cargoes that have been shown to associate preferentially with specific isoforms. We illustrate how the diversification of the adaptor importin α into seven isoforms expands the dynamic range and regulatory control of nucleocytoplasmic transport, offering unexpected opportunities for pharmacological intervention. The emerging view of importin α is that of a key signalling molecule, with isoforms that confer preferential nuclear entry and spatiotemporal specificity on viral and cellular cargoes directly linked to human diseases.
Collapse
|
8
|
Disruption of the ran system by cysteine oxidation of the nucleotide exchange factor RCC1. Mol Cell Biol 2014; 35:566-81. [PMID: 25452301 DOI: 10.1128/mcb.01133-14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Transport regulation by the Ran GTPase requires its nuclear localization and GTP loading by the chromatin-associated exchange factor RCC1. These reactions generate Ran protein and Ran nucleotide gradients between the nucleus and the cytoplasm. Cellular stress disrupts the Ran gradients, but the specific mechanisms underlying this disruption have not been elucidated. We used biochemical approaches to determine how oxidative stress disrupts the Ran system. RCC1 exchange activity was reduced by diamide-induced oxidative stress and restored with dithiothreitol. Using mass spectrometry, we found that multiple solvent-exposed cysteines in RCC1 are oxidized in cells treated with diamide. The cysteines oxidized in RCC1 included Cys93, which is solvent exposed and unique because it becomes buried upon contact with Ran. A Cys93Ser substitution dramatically reduced exchange activity through an effect on RCC1 binding to RanGDP. Diamide treatment reduced the size of the mobile fraction of RCC1-green fluorescent protein in cells and inhibited nuclear import in digitonin-permeabilized cell assays. The Ran protein gradient was also disrupted by UV-induced stress but without affecting RCC1 exchange activity. Our data suggest that stress can disrupt the Ran gradients through RCC1-dependent and RCC1-independent mechanisms, possibly dependent on the particular stress condition.
Collapse
|
9
|
Kulshreshtha V, Ayalew LE, Islam A, Tikoo SK. Conserved arginines of bovine adenovirus-3 33K protein are important for transportin-3 mediated transport and virus replication. PLoS One 2014; 9:e101216. [PMID: 25019945 PMCID: PMC4096500 DOI: 10.1371/journal.pone.0101216] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 06/04/2014] [Indexed: 01/14/2023] Open
Abstract
The L6 region of bovine adenovirus (BAdV)-3 encodes a spliced protein designated 33K. The 33K specific sera detected five major proteins and three minor proteins in transfected or virus infected cells, which could arise by internal initiation of translation and alternative splicing. The 33K protein is predominantly localized to the nucleus of BAdV-3 infected cells. The 33K nuclear transport utilizes both classical importin-α/-β and importin-β dependent nuclear import pathways and preferentially binds to importin-α5 and transportin-3 receptors, respectively. Analysis of mutant 33K proteins demonstrated that amino acids 201–240 of the conserved C-terminus of 33K containing RS repeat are required for nuclear localization and, binding to both importin-α5 and transportin-3 receptors. Interestingly, the arginine residues of conserved RS repeat are required for binding to transportin-3 receptor but not to importin-α5 receptor. Moreover, mutation of arginines residues of RS repeat proved lethal for production of progeny virus. Our results suggest that arginines of RS repeat are required for efficient nuclear transport of 33K mediated by transportin-3, which appears to be essential for replication and production of infectious virion.
Collapse
Affiliation(s)
- Vikas Kulshreshtha
- VIDO-InterVac, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Veterinary Microbiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Lisanework E. Ayalew
- VIDO-InterVac, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Veterinary Microbiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Azharul Islam
- VIDO-InterVac, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Suresh K. Tikoo
- VIDO-InterVac, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Veterinary Microbiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Vaccinology & Immunotherapeutics Program, School of Public Health, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- * E-mail:
| |
Collapse
|
10
|
Ayalew LE, Gaba A, Kumar P, Tikoo SK. Conserved regions of bovine adenovirus-3 pVIII contain functional domains involved in nuclear localization and packaging in mature infectious virions. J Gen Virol 2014; 95:1743-1754. [PMID: 24854002 DOI: 10.1099/vir.0.065763-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Adenoviruses are non-enveloped DNA viruses that replicate in the nucleus of infected cells. One of the core proteins, named pVIII, is a minor capsid protein connecting the core with the inner surface of the capsid. Here, we report the characterization of minor capsid protein pVIII encoded by the L6 region of bovine adenovirus (BAdV)-3. Anti-pVIII serum detected a 24 kDa protein at 12-48 h post-infection and an additional 8 kDa protein at 24-48 h post-infection. While the 24 kDa protein was detected in empty capsids, only the C-terminal-cleaved 8 kDa protein was detected in the mature virion, suggesting that amino acids147-216 of the conserved C-terminus of BAdV-3 pVIII are incorporated in mature virions. Detection of hexon protein associated with both precursor (24 kDa) and cleaved (8 kDa) forms of pVIII suggest that the C-terminus of pVIII interacts with the hexon. The pVIII protein predominantly localizes to the nucleus of BAdV-3-infected cells utilizing the classical importin α/β dependent nuclear import pathway. Analysis of mutant pVIII demonstrated that amino acids 52-72 of the conserved N-terminus bind to importin α-3 with high affinity and are required for the nuclear localization.
Collapse
Affiliation(s)
- Lisanework E Ayalew
- Veterinary Microbiology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E3, Canada.,VIDO-InterVac, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E3, Canada
| | - Amit Gaba
- Veterinary Microbiology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E3, Canada.,VIDO-InterVac, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E3, Canada
| | - Pankaj Kumar
- VIDO-InterVac, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E3, Canada
| | - Suresh K Tikoo
- Vaccinology & Immunotherapeutics Program, School of Public Health, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E3, Canada.,Veterinary Microbiology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E3, Canada.,VIDO-InterVac, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E3, Canada
| |
Collapse
|
11
|
Thomas S, Rai J, John L, Schaefer S, Pützer BM, Herchenröder O. Chikungunya virus capsid protein contains nuclear import and export signals. Virol J 2013; 10:269. [PMID: 23984714 PMCID: PMC3765696 DOI: 10.1186/1743-422x-10-269] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 08/26/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chikungunya virus (CHIKV) is an alphavirus of the Togaviridae family. After autoproteolytic cleavage, the CHIKV capsid protein (CP) is involved in RNA binding and assembly of the viral particle. The monomeric CP is approximately 30 kDa in size and is small enough for passive transport through nuclear pores. Some alphaviruses are found to harbor nuclear localization signals (NLS) and transport of these proteins between cellular compartments was shown to be energy dependent. The active nuclear import of cytoplasmic proteins is mediated by karyopherins and their export by exportins. As nuclear and cytoplasmic trafficking may play a role in the life cycle of CHIKV, we have sought to identify nuclear localization and nuclear export signals in CHIKV CP in a virus-free system. METHODS EGFP-fusion proteins of CHIKV CP and mutants thereof were created and used to monitor their intracellular localization. Binding of cellular proteins was confirmed in pull-down assays with purified CP using co-immuoprecipitation. Nuclear localization was demonstrated in a virus-free system using fluorescence microscopy. RESULTS Here we show that CHIKV CP is a nuclear-cytoplasmic shuttling protein with an active NLS that binds to karyopherin α (Karα) for its nuclear translocation. We also found that the Karα4 C-terminal NLS binding site is sufficient for this interaction. We further demonstrate that CHIKV CP interacts directly with the export receptor CRM1 to transport this viral protein out of the nucleus via a nuclear export signal (NES). The CHIKV CP NES was mapped between amino acids 143 and 155 of CP. Deduced from in silico analyses we found that the NES has a mode of binding similar to the snurportin-1 CRM1 complex. CONCLUSIONS We were able to show that in a virus-free system that the CHIKV capsid protein contains both, a NLS and a NES, and that it is actively transported between the cytoplasma and the nucleus. We conclude that CHIKV CP has the ability to shuttle via interaction with karyopherins for its nuclear import and, vice versa, by CRM1-dependent nuclear export.
Collapse
Affiliation(s)
- Saijo Thomas
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, Schillingallee 69, 18057, Rostock, Germany.
| | | | | | | | | | | |
Collapse
|
12
|
Ye W, Lin W, Tartakoff AM, Ma Q, Tao T. Nuclear import of aristaless-related homeobox protein via its NLS1 regulates its transcriptional function. Mol Cell Biochem 2013; 381:221-31. [PMID: 23771350 DOI: 10.1007/s11010-013-1706-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Accepted: 05/24/2013] [Indexed: 10/26/2022]
Abstract
Nucleocytoplasmic transport of transcription factors is essential in eukaryotes. We previously reported the presence of two functional NLSs in the homeodomain protein, aristaless-related homeobox (Arx) protein, which is a key transcriptional repressor of LMO1, SHOX2, and PAX4 during development. NLS2, that overlaps the homeodomain, is recognized directly by multiple importin βs, but not by importin αs. In this study, we found that the N-terminal NLS1 of Arx is targeted by multiple importin α proteins, including importin α3 and α5. Both in vivo and in vitro assays demonstrated that nuclear import of Arx via NLS1 is mediated by the importin α/β pathway. Mutagenesis analysis indicated that two basic amino acids, (84)K and (87)R, are essential to the function of NLS1, and that their mutation prevents interactions of Arx with importin αs. Interestingly, inhibition of nuclear import of Arx via NLS1 clearly attenuates its ability of transcriptional repression, suggesting that nuclear import of Arx via NLS1 contributes to its transcriptional function.
Collapse
Affiliation(s)
- Wenduo Ye
- Xiamen University School of Life Sciences, Xiamen, Fujian, China
| | | | | | | | | |
Collapse
|
13
|
The polycomb group protein EED varies in its ability to access the nucleus in porcine oocytes and cleavage stage embryos. Anim Reprod Sci 2012; 133:198-204. [DOI: 10.1016/j.anireprosci.2012.07.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 06/08/2012] [Accepted: 07/18/2012] [Indexed: 11/17/2022]
|
14
|
Paterson CP, Ayalew LE, Tikoo SK. Mapping of nuclear import signal and importin α3 binding regions of 52K protein of bovine adenovirus-3. Virology 2012; 432:63-72. [PMID: 22739443 DOI: 10.1016/j.virol.2012.05.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 04/24/2012] [Accepted: 05/24/2012] [Indexed: 11/19/2022]
Abstract
The L1 region of bovine adenovirus (BAdV)-3 encodes a non-structural protein designated 52K. Anti-52K serum detected a protein of 40kDa, which localized to the nucleus but not to the nucleolus in BAdV-3-infected or transfected cells. Analysis of mutant 52K proteins suggested that three basic residues ((105)RKR(107)) of the identified domain (amino acids (102)GMPRKRVLT(110)) are essential for nuclear localization of 52K. The nuclear import of a GST-52K fusion protein utilizes the classical importin α/β-dependent nuclear transport pathway. The 52K protein is preferentially bound to the cellular nuclear import receptor importin α3. Although deletion of amino acid 102-110 is sufficient to abrogate the nuclear localization of 52K, amino acid 90-133 are required for interaction with importin-α3 and localizing a cytoplasmic protein to the nucleus. These results suggest that 52K contains a bipartite NLS, which preferentially utilize an importin α3 nuclear import receptor-mediated pathway to transport 52K to the nucleus.
Collapse
Affiliation(s)
- Carolyn P Paterson
- Vaccine & Infectious Disease Organization-International Vaccine Center, University of Saskatchewan, Saskatoon, SK S7N 5E3 Canada
| | | | | |
Collapse
|
15
|
Park KE, Inerowicz HD, Wang X, Li Y, Koser S, Cabot RA. Identification of karyopherin α1 and α7 interacting proteins in porcine tissue. PLoS One 2012; 7:e38990. [PMID: 22720010 PMCID: PMC3374799 DOI: 10.1371/journal.pone.0038990] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 05/17/2012] [Indexed: 11/19/2022] Open
Abstract
Specialized trafficking systems in eukaryotic cells serve a critical role in partitioning intracellular proteins between the nucleus and cytoplasm. Cytoplasmic proteins (including chromatin remodeling enzymes and transcription factors) must gain access to the nucleus to exert their functions to properly program fundamental cellular events ranging from cell cycle progression to gene transcription. Knowing that nuclear import mediated by members of the karyopherin α family of transport receptors plays a critical role in regulating development and differentiation, we wanted to determine the identity of proteins that are trafficked by this karyopherin α pathway. To this end, we performed a GST pull-down assay using porcine orthologs of karyopherin α1 (KPNA1) and karyopherin α7 (KPNA7) and prey protein derived from porcine fibroblast cells and used a liquid chromatography and tandem mass spectrometry (LC-MS/MS) approach to determine the identity of KPNA1 and KPNA7 interacting proteins. Our screen revealed that the proteins that interact with KPNA1 and KPNA7 are generally nuclear proteins that possess nuclear localization signals. We further validated two candidate proteins from this screen and showed that they are able to be imported into the nucleus in vivo and also interact with members of the karyopherin α family of proteins in vitro. Our results also reveal the utility of using a GST pull-down approach coupled with LC-MS/MS to screen for protein interaction partners in a non-traditional model system.
Collapse
Affiliation(s)
- Ki-Eun Park
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - H. Dorota Inerowicz
- Bindley Biosciences Center, Proteomics Core Facility, Purdue University, West Lafayette, Indiana, United States of America
| | - Xin Wang
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Yanfang Li
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Stephanie Koser
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Ryan A. Cabot
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, United States of America
| |
Collapse
|
16
|
Depping R, Schindler SG, Jacobi C, Kirschner KM, Scholz H. Nuclear transport of Wilms' tumour protein Wt1 involves importins α and β. Cell Physiol Biochem 2012; 29:223-32. [PMID: 22415091 DOI: 10.1159/000337603] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2011] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND/AIMS Wilms' tumour protein, Wt1, is a zinc finger molecule, which is required for normal embryonic development. Mutations of the WT1 gene can give rise to childhood cancer of the kidneys. Different Wt1 isoforms exist, which function either as transcription factors or have a presumed role in mRNA processing. Previous studies suggested that Wt1 undergoes nucleocytoplasmic shuttling, and cytoplasmic Wt1 was higher in malignant than in normal cells. The aim of this study was to analyse the molecular pathways along which Wt1 shuttles between the cytoplasm and nucleus. METHODS Interaction of Wt1 protein with various importin α subtypes and importin β was assessed in pull-down assays and co-immunoprecipitation experiments. Nuclear localisation signals (NLS) were identified by combining site-directed mutagenesis with subcellular immunodetection of the transfected Wt1 variants. RESULTS Wt1(+/-KTS) proteins were found to interact with importin α1 and importin β in vitro and in living cells in vivo. A NLS that was necessary and sufficient for nuclear import could be mapped to the third Wt1 zinc finger. Mutation of this NLS strongly weakened binding of Wt1 to importins. CONCLUSION Nuclear translocation of Wilms' tumour protein involves importins α and β, and a NLS in the third zinc finger.
Collapse
Affiliation(s)
- Reinhard Depping
- Institut für Physiologie, Zentrum für Medizinische Struktur- und Zellbiologie, Universität zu Lübeck, Lübeck, Germany
| | | | | | | | | |
Collapse
|
17
|
Wang X, Park KE, Koser S, Liu S, Magnani L, Cabot RA. KPNA7, an oocyte- and embryo-specific karyopherin?subtype, is required for porcine embryo development. Reprod Fertil Dev 2012; 24:382-91. [DOI: 10.1071/rd11119] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 06/15/2011] [Indexed: 01/26/2023] Open
Abstract
Coordinated partitioning of intracellular cargoes between nuclear and cytoplasmic compartments is critical for cell survival and differentiation. The karyopherin α/β heterodimer functions to import cytoplasmic proteins that possess classical nuclear localisation signals into the nucleus. Seven karyopherin α subtypes have been identified in mammals. The aim of this study was to determine the relative abundance of transcripts encoding seven karyopherin α subtypes in porcine oocytes and embryos at discrete stages of cleavage development, and to determine the developmental requirements of karypopherin α 7 (KPNA7), an oocyte and cleavage stage embryo-specific karyopherin α subtype. We hypothesised that knockdown of KPNA7 would negatively affect porcine cleavage development. To test this hypothesis, in vitro matured and fertilised porcine oocytes were injected with a double-stranded interfering RNA molecule that targeted KPNA7; nuclei were counted in all embryos 6 days after fertilisation. Embryos injected with KPNA7-interfering RNAs possessed significantly lower cell numbers than their respective control groups (P < 0.05). In vitro binding assays also suggest that KPNA7 may transport intracellular proteins that possess unique nuclear localisation signals. Our data suggest that embryos have differential requirements for individual karyopherin α subtypes and that these karyopherin α subtypes differentially transport intracellular cargo during cleavage development.
Collapse
|
18
|
Mehmood R, Yasuhara N, Fukumoto M, Oe S, Tachibana T, Yoneda Y. Cross-talk between distinct nuclear import pathways enables efficient nuclear import of E47 in conjunction with its partner transcription factors. Mol Biol Cell 2011; 22:3715-24. [PMID: 21832153 PMCID: PMC3183024 DOI: 10.1091/mbc.e10-10-0809] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Study of the nuclear import behavior of E47 in conjunction with its partner transcription factors shows that although the nuclear import of E47 is importin α dependent, it is capable of accumulating in the nucleus under importin α–blocked conditions by virtue of its interaction with its binding partners NeuroD1 and MyoD. Nuclear import of karyophilic proteins is carried out by a variety of mechanisms. We previously showed that two basic helix-loop-helix proteins, NeuroD1 and E47, synergistically affect each other's nuclear import. In this study, we dissected the molecular pathways underlying nuclear import of the NeuroD1/E47 heterodimer. In vitro nuclear import assays indicated that importin α family members are the major nuclear import receptors for E47. However, inhibition of importin α resulted in cytoplasmic retention of E47 that could be rescued by its binding partner, NeuroD1, through heterodimerization. In addition, nuclear import of NeuroD1 was importin α independent but importin β1 dependent. In primary neurons, localization of endogenous E47 was not affected by importin α inhibition, suggesting that neuronal E47 could be imported into the nucleus as a heterodimer with NeuroD1 by using importin β1 alone. We also found that E47 had similar nuclear import characteristics in C2C12 cells, where E47 heterodimerized with MyoD, another helix-loop-helix protein, suggesting functional conservation within the same family of transcription factors. Collectively, our data reveal that E47 is imported into the nucleus via multiple pathways, depending on the molecular binding mode, establishing a previously uncharacterized cross-talk between two distinct nuclear import pathways.
Collapse
Affiliation(s)
- Rashid Mehmood
- Department of Frontier Biosciences, Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan
| | | | | | | | | | | |
Collapse
|
19
|
Holt JE, Ly-Huynh JD, Efthymiadis A, Hime GR, Loveland KL, Jans DA. Regulation of Nuclear Import During Differentiation; The IMP alpha Gene Family and Spermatogenesis. Curr Genomics 2011; 8:323-34. [PMID: 19384428 PMCID: PMC2652405 DOI: 10.2174/138920207782446151] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2007] [Revised: 06/09/2007] [Accepted: 06/10/2007] [Indexed: 11/30/2022] Open
Abstract
Access to nuclear genes in eukaryotes is provided by members of the importin (IMP) superfamily of proteins, which are of α- or β-types, the best understood nuclear import pathway being mediated by a heterodimer of an IMP α and IMP β1. IMP α recognises specific targeting signals on cargo proteins, while IMP β1 mediates passage into, and release within, the nucleus by interacting with other components of the transport machinery, including the monomeric guanine nucleotide binding protein Ran. In this manner, hundreds of different proteins can be targeted specifically into the nucleus in a tightly regulated fashion. The IMP α gene family has expanded during evolution, with only a single IMP α (Srp1p) gene in budding yeast, and three (IMP α1, 2/pendulin and 3) and five (IMP α1, -2, -3, -4 and -6) IMP α genes in Drosophila melanogaster and mouse respectively, which fall into three phylogenetically distinct groups. The fact that IMP α3 and IMP α2 are only present in metazoans implies that they emerged during the evolution of multicellular animals to perform specialised roles in particular cells and tissues. This review describes what is known of the IMP α gene family in mouse and in D. melanogaster, including a comparitive examination of their mRNA expression profiles in a highly differentiated tissue, the testis. The clear implication of their highly regulated synthesis during the course of spermatogenesis is that the different IMP αs have distinct expression patterns during cellular differentiation, implying tissue/cell type-specific roles.
Collapse
Affiliation(s)
- J E Holt
- Monash University, Department of Biochemistry and Molecular Biology, Nuclear Signalling Laboratory, Clayton, Australia
| | | | | | | | | | | |
Collapse
|
20
|
Moriyama T, Nagai M, Oka M, Ikawa M, Okabe M, Yoneda Y. Targeted disruption of one of the importin α family members leads to female functional incompetence in delivery. FEBS J 2011; 278:1561-72. [PMID: 21371262 DOI: 10.1111/j.1742-4658.2011.08079.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Importin α mediates the nuclear import of proteins through nuclear pore complexes in eukaryotic cells, and is common to all eukaryotes. Previous reports identified at least six importin α family genes in mice. Although these isoforms show differential binding to various import cargoes in vitro, the in vivo physiological roles of these mammalian importin α isoforms remain unknown. Here, we generated and examined importin α5 knockout (impα5(-/-)) mice. These mice developed normally, and showed no gross histological abnormalities in most major organs. However, the ovary and uterus of impα5(-/-) female mice exhibited hypoplasia. Furthermore, we found that impα5(-/-) female mice had a 50% decrease in serum progesterone levels and a 57% decrease in progesterone receptor mRNA levels in the ovary. Additionally, impα5(-/-) uteruses that were treated with exogenous gonadotropins displayed hypertrophy, similarly to progesterone receptor-deficient mice. Although these mutant female mice could become pregnant, the total number of pups was significantly decreased, and some of the pups were dead at birth. These results suggest that importin α5 has essential roles in the mammalian female reproductive organs.
Collapse
Affiliation(s)
- Tetsuji Moriyama
- Department of Frontier Biosciences, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | | | | | | | | | | |
Collapse
|
21
|
Langer K, Dian C, Rybin V, Müller CW, Petosa C. Insights into the function of the CRM1 cofactor RanBP3 from the structure of its Ran-binding domain. PLoS One 2011; 6:e17011. [PMID: 21364925 PMCID: PMC3045386 DOI: 10.1371/journal.pone.0017011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Accepted: 01/18/2011] [Indexed: 02/03/2023] Open
Abstract
Proteins bearing a leucine-rich nuclear export signal (NES) are exported from the nucleus by the transport factor CRM1, which forms a cooperative ternary complex with the NES-bearing cargo and with the small GTPase Ran. CRM1-mediated export is regulated by RanBP3, a Ran-interacting nuclear protein. Unlike the related proteins RanBP1 and RanBP2, which promote disassembly of the export complex in the cytosol, RanBP3 acts as a CRM1 cofactor, enhancing NES export by stabilizing the export complex in the nucleus. RanBP3 also alters the cargo selectivity of CRM1, promoting recognition of the NES of HIV-1 Rev and of other cargos while deterring recognition of the import adaptor protein Snurportin1. Here we report the crystal structure of the Ran-binding domain (RBD) from RanBP3 and compare it to RBD structures from RanBP1 and RanBP2 in complex with Ran and CRM1. Differences among these structures suggest why RanBP3 binds Ran with unusually low affinity, how RanBP3 modulates the cargo selectivity of CRM1, and why RanBP3 promotes assembly rather than disassembly of the export complex. The comparison of RBD structures thus provides an insight into the functional diversity of Ran-binding proteins.
Collapse
Affiliation(s)
- Karla Langer
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Cyril Dian
- Institut de Biologie Structurale Jean-Pierre Ebel, Unité Mixte de Recherche 5075 (Commissariat à L'Energie Atomique et aux Energies Alternatives/Centre National de la Recherche Scientifique/Université Joseph Fourier), Grenoble, France
| | - Vladimir Rybin
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Christoph W. Müller
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Carlo Petosa
- Institut de Biologie Structurale Jean-Pierre Ebel, Unité Mixte de Recherche 5075 (Commissariat à L'Energie Atomique et aux Energies Alternatives/Centre National de la Recherche Scientifique/Université Joseph Fourier), Grenoble, France
| |
Collapse
|
22
|
Kelley JB, Talley AM, Spencer A, Gioeli D, Paschal BM. Karyopherin alpha7 (KPNA7), a divergent member of the importin alpha family of nuclear import receptors. BMC Cell Biol 2010; 11:63. [PMID: 20701745 PMCID: PMC2929220 DOI: 10.1186/1471-2121-11-63] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Accepted: 08/11/2010] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Classical nuclear localization signal (NLS) dependent nuclear import is carried out by a heterodimer of importin alpha and importin beta. NLS cargo is recognized by importin alpha, which is bound by importin beta. Importin beta mediates translocation of the complex through the central channel of the nuclear pore, and upon reaching the nucleus, RanGTP binding to importin beta triggers disassembly of the complex. To date, six importin alpha family members, encoded by separate genes, have been described in humans. RESULTS We sequenced and characterized a seventh member of the importin alpha family of transport factors, karyopherin alpha 7 (KPNA7), which is most closely related to KPNA2. The domain of KPNA7 that binds Importin beta (IBB) is divergent, and shows stronger binding to importin beta than the IBB domains from of other importin alpha family members. With regard to NLS recognition, KPNA7 binds to the retinoblastoma (RB) NLS to a similar degree as KPNA2, but it fails to bind the SV40-NLS and the human nucleoplasmin (NPM) NLS. KPNA7 shows a predominantly nuclear distribution under steady state conditions, which contrasts with KPNA2 which is primarily cytoplasmic. CONCLUSION KPNA7 is a novel importin alpha family member in humans that belongs to the importin alpha2 subfamily. KPNA7 shows different subcellular localization and NLS binding characteristics compared to other members of the importin alpha family. These properties suggest that KPNA7 could be specialized for interactions with select NLS-containing proteins, potentially impacting developmental regulation.
Collapse
Affiliation(s)
- Joshua B Kelley
- Center for Cell Signaling, University of Virginia, Charlottesville, VA, 22908, USA
| | - Ashley M Talley
- Center for Cell Signaling, University of Virginia, Charlottesville, VA, 22908, USA
| | - Adam Spencer
- Center for Cell Signaling, University of Virginia, Charlottesville, VA, 22908, USA
| | - Daniel Gioeli
- Department of Microbiology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Bryce M Paschal
- Center for Cell Signaling, University of Virginia, Charlottesville, VA, 22908, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22908, USA
| |
Collapse
|
23
|
Importin alpha3 interacts with HIV-1 integrase and contributes to HIV-1 nuclear import and replication. J Virol 2010; 84:8650-63. [PMID: 20554775 DOI: 10.1128/jvi.00508-10] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
HIV-1 employs the cellular nuclear import machinery to actively transport its preintegration complex (PIC) into the nucleus for integration of the viral DNA. Several viral karyophilic proteins and cellular import factors have been suggested to contribute to HIV-1 PIC nuclear import and replication. However, how HIV interacts with different cellular machineries to ensure efficient nuclear import of its preintegration complex in dividing and nondividing cells is still not fully understood. In this study, we have investigated different importin alpha (Impalpha) family members for their impacts on HIV-1 replication, and we demonstrate that short hairpin RNA (shRNA)-mediated Impalpha3 knockdown (KD) significantly impaired HIV infection in HeLa cells, CD4(+) C8166 T cells, and primary macrophages. Moreover, quantitative real-time PCR analysis revealed that Impalpha3-KD resulted in significantly reduced levels of viral 2-long-terminal repeat (2-LTR) circles but had no effect on HIV reverse transcription. All of these data indicate an important role for Impalpha3 in HIV nuclear import. In an attempt to understand how Impalpha3 participates in HIV nuclear import and replication, we first demonstrated that the HIV-1 karyophilic protein integrase (IN) was able to interact with Impalpha3 both in a 293T cell expression system and in HIV-infected CD4(+) C8166 T cells. Deletion analysis suggested that a region (amino acids [aa] 250 to 270) in the C-terminal domain of IN is involved in this viral-cellular protein interaction. Overall, this study demonstrates for the first time that Impalpha3 is an HIV integrase-interacting cofactor that is required for efficient HIV-1 nuclear import and replication in both dividing and nondividing cells.
Collapse
|
24
|
Merényi G, Kónya E, Vértessy BG. Drosophila proteins involved in metabolism of uracil-DNA possess different types of nuclear localization signals. FEBS J 2010; 277:2142-56. [DOI: 10.1111/j.1742-4658.2010.07630.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
25
|
Tejomurtula J, Lee KB, Tripurani SK, Smith GW, Yao J. Role of Importin Alpha8, a New Member of the Importin Alpha Family of Nuclear Transport Proteins, in Early Embryonic Development in Cattle1. Biol Reprod 2009; 81:333-42. [DOI: 10.1095/biolreprod.109.077396] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
26
|
Dai F, Lin X, Chang C, Feng XH. Nuclear export of Smad2 and Smad3 by RanBP3 facilitates termination of TGF-beta signaling. Dev Cell 2009; 16:345-57. [PMID: 19289081 PMCID: PMC2676691 DOI: 10.1016/j.devcel.2009.01.022] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Revised: 12/31/2008] [Accepted: 01/30/2009] [Indexed: 10/21/2022]
Abstract
Smad2 and Smad3 (Smad2/3) are key intracellular signal transducers for TGF-beta signaling, and their transcriptional activities are controlled through reversible phosphorylation and nucleocytoplasmic shuttling. However, the precise mechanism underlying nuclear export of Smad2/3 remains elusive. Here we report the essential function of RanBP3 in selective nuclear export of Smad2/3 in the TGF-beta pathway. RanBP3 directly recognizes dephosphorylated Smad2/3, which results from the activity of nuclear Smad phosphatases, and mediates nuclear export of Smad2/3 in a Ran-dependent manner. As a result, increased expression of RanBP3 inhibits TGF-beta signaling in mammalian cells and Xenopus embryos. Conversely, depletion of RanBP3 expression or dominant-negative inhibition of RanBP3 enhances TGFbeta-induced antiproliferative and transcriptional responses. In conclusion, our study supports a definitive role for RanBP3 in mediating Smad2/3 nuclear export and terminating TGF-beta signaling.
Collapse
Affiliation(s)
- Fangyan Dai
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX77030, USA
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX77030, USA
- The Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX77030, USA
| | - Xia Lin
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX77030, USA
- The Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX77030, USA
| | - Chenbei Chang
- Department of Cell Biology, University of Alabama, Birmingham, AL35294, USA
| | - Xin-Hua Feng
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX77030, USA
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX77030, USA
- The Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX77030, USA
| |
Collapse
|
27
|
Nuclear localization signal-dependent and -independent movements of Drosophila melanogaster dUTPase isoforms during nuclear cleavage. Biochem Biophys Res Commun 2009; 381:271-5. [PMID: 19232319 DOI: 10.1016/j.bbrc.2009.02.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Accepted: 02/09/2009] [Indexed: 11/20/2022]
Abstract
Two dUTPase isoforms (23 kDa and 21 kDa) are present in the fruitfly with the sole difference of an N-terminal extension. In Drosophila embryo, both isoforms are detected inside the nucleus. Here, we investigated the function of the N-terminal segment using eYFP-dUTPase constructs. In Schneider 2 cells, only the 23 kDa construct showed nuclear localization arguing that it may contain a nuclear localization signal (NLS). Sequence comparisons identified a lysine-rich nonapeptide with similarity to the human c-myc NLS. In Drosophila embryos during nuclear cleavages, the 23 kDa isoform showed the expected localization shifts. Contrariwise, although the 21 kDa isoform was excluded from the nuclei during interphase, it was shifted to the nucleus during prophase and forthcoming mitotic steps. The observed dynamic localization character showed strict timing to the nuclear cleavage phases and explained how both isoforms can be present within the nuclear microenvironment, although at different stages of cell cycle.
Collapse
|
28
|
Sorokin AV, Kim ER, Ovchinnikov LP. Nucleocytoplasmic transport of proteins. BIOCHEMISTRY (MOSCOW) 2008; 72:1439-57. [PMID: 18282135 DOI: 10.1134/s0006297907130032] [Citation(s) in RCA: 163] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In eukaryotic cells, the movement of macromolecules between the nucleus and cytoplasm occurs through the nuclear pore complex (NPC)--a large protein complex spanning the nuclear envelope. The nuclear transport of proteins is usually mediated by a family of transport receptors known as karyopherins. Karyopherins bind to their cargoes via recognition of nuclear localization signal (NLS) for nuclear import or nuclear export signal (NES) for export to form a transport complex. Its transport through NPC is facilitated by transient interactions between the karyopherins and NPC components. The interactions of karyopherins with their cargoes are regulated by GTPase Ran. In the current review, we describe the NPC structure, NLS, and NES, as well as the model of classic Ran-dependent transport, with special emphasis on existing alternative mechanisms; we also propose a classification of the basic mechanisms of protein transport regulation.
Collapse
Affiliation(s)
- A V Sorokin
- Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia
| | | | | |
Collapse
|
29
|
Abstract
In a recent issue of Molecular Cell, Yoon et al. provide evidence for the control of nucleocytoplasmic transport by protein kinase signaling pathways through phosphorylation of RanBP3, an accessory factor in the Ran GTPase system. This mechanism may coordinate nucleocytoplasmic transport with other mitogenic effects of these pathways.
Collapse
Affiliation(s)
- Paul R Clarke
- Biomedical Research Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, United Kingdom.
| |
Collapse
|
30
|
Yoon SO, Shin S, Liu Y, Ballif BA, Woo MS, Gygi SP, Blenis J. Ran-binding protein 3 phosphorylation links the Ras and PI3-kinase pathways to nucleocytoplasmic transport. Mol Cell 2008; 29:362-75. [PMID: 18280241 PMCID: PMC2266693 DOI: 10.1016/j.molcel.2007.12.024] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2007] [Revised: 08/17/2007] [Accepted: 12/15/2007] [Indexed: 10/22/2022]
Abstract
The major participants of the Ras/ERK and PI3-kinase (PI3K) pathways are well characterized. The cellular response to activation of these pathways, however, can vary dramatically. How differences in signal strength, timing, spatial location, and cellular context promote specific cell-fate decisions remains unclear. Nuclear transport processes can have a major impact on the determination of cell fate; however, little is known regarding how nuclear transport is regulated by or regulates these pathways. Here we show that RSK and Akt, which are activated downstream of Ras/ERK and PI3K, respectively, modulate the Ran gradient and nuclear transport by interacting with, phosphorylating, and regulating Ran-binding protein 3 (RanBP3) function. Our findings highlight an important link between two major cell-fate determinants: nuclear transport and the Ras/ERK/RSK and PI3K/Akt signaling pathways.
Collapse
Affiliation(s)
- Sang-Oh Yoon
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Sejeong Shin
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Yuzhen Liu
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Bryan A. Ballif
- Department of Biology, University of Vermont, 109 Carrigan Drive, Burlington, VT 05405, USA
| | | | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - John Blenis
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
31
|
Kelley JB, Paschal BM. Hyperosmotic stress signaling to the nucleus disrupts the Ran gradient and the production of RanGTP. Mol Biol Cell 2007; 18:4365-76. [PMID: 17761537 PMCID: PMC2043571 DOI: 10.1091/mbc.e07-01-0089] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The RanGTP gradient depends on nucleocytoplasmic shuttling of Ran and its nucleotide exchange in the nucleus. Here we show that hyperosmotic stress signaling induced by sorbitol disrupts the Ran protein gradient and reduces the production of RanGTP. Ran gradient disruption is rapid and is followed by early (10-20 min) and late (30-60 min) phases of recovery. Results from SB203580 and siRNA experiments suggest the stress kinase p38 is important for Ran gradient recovery. NTF2 and Mog1, which are transport factors that regulate the nuclear localization of Ran, showed kinetics of delocalization and recovery similar to Ran. Microinjection of a nuclear localization signal reporter protein revealed that sorbitol stress decreases the rate of nuclear import. Sorbitol stress also slowed RCC1 mobility in the nucleus, which is predicted to reduce RCC1 dissociation from chromatin and RanGTP production. This was tested using a FRET biosensor that registers nuclear RanGTP levels, which were reduced in response to sorbitol stress. Although sorbitol alters nucleotide levels, we show that inverting the GTP/GDP ratio in cells is not sufficient to disrupt the Ran gradient. Thus, the Ran system is a target of hyperosmotic stress signaling, and cells use protein localization-based mechanisms as part of a rapid stress response.
Collapse
Affiliation(s)
- Joshua B. Kelley
- Center for Cell Signaling, Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908
| | - Bryce M. Paschal
- Center for Cell Signaling, Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
32
|
Grundt K, Haga IV, Huitfeldt HS, Ostvold AC. Identification and characterization of two putative nuclear localization signals (NLS) in the DNA-binding protein NUCKS. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:1398-406. [PMID: 17604136 DOI: 10.1016/j.bbamcr.2007.05.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2006] [Revised: 05/21/2007] [Accepted: 05/24/2007] [Indexed: 10/23/2022]
Abstract
Immunofluorescence analyses show that the vertebrate specific and DNA-binding protein NUCKS is distributed throughout the cytoplasm in mitotic cells and targeted to the reforming nuclei in late telophase of the cell cycle. Computer analysis of the primary structure of NUCKS revealed the presence of two regions of highly charged, basic residues, which were identified as potential nuclear localization signals (NLSs). One of these signals (NLS1) is highly conserved between the species investigated, and fits to the description of being a classical bipartite NLS. The other amino acid motif (NLS2) is less conserved and does not constitute a classical bipartite NLS consensus sequence. We have shown that each of the two putative NLSs is capable of translocating green fluorescent protein (GFP) into the nucleus. The highly conserved NLS1 is monopartite, resembling the signals of c-Myc and RanBP3. Surprisingly, a natural occurring splice variant of NUCKS lacking 40 amino acids including NLS1, is not capable of translocating a corresponding NUCKS-GFP fusion protein into the nucleus, indicating that NLS1 is the main nuclear localization signal in NUCKS. This is also confirmed by site-directed mutagenesis of the full-length protein. By GFP-immunoprecipitation and GST-pull down experiments, we show that NUCKS binds to importin alpha3 and importin alpha5 in vitro, suggesting that the nuclear targeting of NUCKS follows a receptor-mediated and energy-dependent import mechanism.
Collapse
Affiliation(s)
- Kirsten Grundt
- Department of Biochemistry, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1112, Blindern, 0317 Oslo, Norway
| | | | | | | |
Collapse
|
33
|
Donaldson NS, Daniel Y, Kelly KF, Graham M, Daniel JM. Nuclear trafficking of the POZ-ZF protein Znf131. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:546-55. [PMID: 17306895 DOI: 10.1016/j.bbamcr.2006.12.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2006] [Revised: 12/06/2006] [Accepted: 12/08/2006] [Indexed: 01/21/2023]
Abstract
Znf131 is a member of the BTB/POZ family of transcription factors with roles in development and carcinogenesis. Like many members of this protein family, Znf131 displays robust nuclear localization in cultured cells, but the mechanism(s) of Znf131 nuclear trafficking is unknown. Here, we report the mechanism of Znf131 nuclear localization. Visual inspection of the Znf131 amino acid sequence revealed three basic regions (BR-1, -2 and -3) with the potential to serve as nuclear localization signals (NLS). Of the three basic regions, only BR-1 functioned independently to efficiently target heterologous beta-gal-GFP fusion proteins to HeLa cell nuclei. However, a Znf131 truncation mutant containing BR-2 and BR-3 efficiently targeted heterologous beta-gal-GFP fusion proteins to HeLa cell nuclei. Mutational analysis of full-length GFP-tagged Znf131 revealed that loss of any one BR alone did not prevent Znf131 nuclear localization. This apparent redundancy in NLS activity was due to the fact that intact BR-1 or BR-2 alone could target full-length Znf131 to nuclei. Consequently, simultaneous mutation of BR-1 and BR-2 abolished full-length Znf131 nuclear localization. Therefore, BR-1 and BR-2 are functional NLSs for Znf131 and as such are designated NLS-1 and NLS-2. Finally, wild type Znf131, and not a Znf131 NLS-defective mutant (NLS-1m/NLS-2m) interacted preferentially with the nuclear import receptor Importin-alpha3 in vitro.
Collapse
Affiliation(s)
- Nickett S Donaldson
- Department of Biology, LSB-331 McMaster University, 1280 Main Street West Hamilton, Canada ON L8S 4K1
| | | | | | | | | |
Collapse
|
34
|
Friedrich B, Quensel C, Sommer T, Hartmann E, Köhler M. Nuclear localization signal and protein context both mediate importin alpha specificity of nuclear import substrates. Mol Cell Biol 2006; 26:8697-709. [PMID: 17000757 PMCID: PMC1636818 DOI: 10.1128/mcb.00708-06] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The "classical" nuclear protein import pathway depends on importin alpha and importin beta. Importin alpha binds nuclear localization signal (NLS)-bearing proteins and functions as an adapter to access the importin beta-dependent import pathway. In humans, only one importin beta is known to interact with importin alpha, while six alpha importins have been described. Various experimental approaches provided evidence that several substrates are transported specifically by particular alpha importins. Whether the NLS is sufficient to mediate importin alpha specificity is unclear. To address this question, we exchanged the NLSs of two well-characterized import substrates, the seven-bladed propeller protein RCC1, preferentially transported into the nucleus by importin alpha3, and the less specifically imported substrate nucleoplasmin. In vitro binding studies and nuclear import assays revealed that both NLS and protein context contribute to the specificity of importin alpha binding and transport.
Collapse
Affiliation(s)
- Beate Friedrich
- The Max Delbrueck Center for Molecular Medicine, Robert Roessle Strasse 10, 13125 Berlin, Germany
| | | | | | | | | |
Collapse
|
35
|
Abstract
Accurate cellular localization is crucial for the effective function of most signalling molecules and nuclear translocation is central to the function of transcription factors. The passage of large molecules between the cytoplasm and nucleus is restricted, and this restriction affords a mechanism to regulate transcription by controlling the access of transcription factors to the nucleus. In this Review, we focus on the signal transducer and activator of transcription (STAT) family of transcription factors. The regulation of the nuclear trafficking of STAT-family members is diverse. Some STAT proteins constitutively shuttle between the nucleus and cytoplasm, whereas others require tyrosine phosphorylation for nuclear localization. In either case, the regulation of nuclear trafficking can provide a target for therapeutic intervention.
Collapse
Affiliation(s)
- Nancy C Reich
- Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11777, USA.
| | | |
Collapse
|
36
|
Nishijima H, Nakayama JI, Yoshioka T, Kusano A, Nishitani H, Shibahara KI, Nishimoto T. Nuclear RanGAP is required for the heterochromatin assembly and is reciprocally regulated by histone H3 and Clr4 histone methyltransferase in Schizosaccharomyces pombe. Mol Biol Cell 2006; 17:2524-36. [PMID: 16540522 PMCID: PMC1474784 DOI: 10.1091/mbc.e05-09-0893] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Although the Ran GTPase-activating protein RanGAP mainly functions in the cytoplasm, several lines of evidence indicate a nuclear function of RanGAP. We found that Schizosaccharomyces pombe RanGAP, SpRna1, bound the core of histone H3 (H3) and enhanced Clr4-mediated H3-lysine 9 (K9) methylation. This enhancement was not observed for methylation of the H3-tail containing K9 and was independent of SpRna1-RanGAP activity, suggesting that SpRna1 itself enhances Clr4-mediated H3-K9 methylation via H3. Although most SpRna1 is in the cytoplasm, some cofractionated with H3. Sprna1(ts) mutations caused decreases in Swi6 localization and H3-K9 methylation at all three heterochromatic regions of S. pombe. Thus, nuclear SpRna1 seems to be involved in heterochromatin assembly. All core histones bound SpRna1 and inhibited SpRna1-RanGAP activity. In contrast, Clr4 abolished the inhibitory effect of H3 on the RanGAP activity of SpRna1 but partially affected the other histones. SpRna1 formed a trimeric complex with H3 and Clr4, suggesting that nuclear SpRna1 is reciprocally regulated by histones, especially H3, and Clr4 on the chromatin to function for higher order chromatin assembly. We also found that SpRna1 formed a stable complex with Xpo1/Crm1 plus Ran-GTP, in the presence of H3.
Collapse
Affiliation(s)
- Hitoshi Nishijima
- *Department of Molecular Biology, Graduate School of Medical Science, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
- Department of Integrated Genetics, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan; and
| | - Jun-ichi Nakayama
- Laboratory for Chromatin Dynamics, Center for Developmental Biology, RIKEN, Kobe 650-0047, Japan
| | - Tomoko Yoshioka
- *Department of Molecular Biology, Graduate School of Medical Science, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ayumi Kusano
- *Department of Molecular Biology, Graduate School of Medical Science, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hideo Nishitani
- *Department of Molecular Biology, Graduate School of Medical Science, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kei-ichi Shibahara
- Department of Integrated Genetics, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan; and
| | - Takeharu Nishimoto
- *Department of Molecular Biology, Graduate School of Medical Science, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
- Address correspondence to: Takeharu Nishimoto (
)
| |
Collapse
|
37
|
Aratani S, Oishi T, Fujita H, Nakazawa M, Fujii R, Imamoto N, Yoneda Y, Fukamizu A, Nakajima T. The nuclear import of RNA helicase A is mediated by importin-α3. Biochem Biophys Res Commun 2006; 340:125-33. [PMID: 16375861 DOI: 10.1016/j.bbrc.2005.11.161] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Accepted: 11/29/2005] [Indexed: 10/25/2022]
Abstract
RNA helicase A (RHA), an ATPase/helicase, regulates the gene expression at various steps including transcriptional activation and RNA processing. RHA is known to shuttle between the nucleus and cytoplasm. We identified the nuclear localization signal (NLS) of RHA and analyzed the nuclear import mechanisms. The NLS of RHA (RHA-NLS) consisting of 19 amino acid residues is highly conserved through species and does not have the consensus classical NLS. In vitro nuclear import assays revealed that the nuclear import of RHA was Ran-dependent and mediated with the classical importin-alpha/beta-dependent pathway. The binding assay indicated that the basic residues in RHA-NLS were used for interaction with importin-alpha. Furthermore, the nuclear import of RHA-NLS was supported by importin-alpha1 and preferentially importin-alpha3. Our results indicate that the nuclear import of RHA is mediated by the importin-alpha3/importin-beta-dependent pathway and suggest that the specificity for importin may regulate the functions of cargo proteins.
Collapse
Affiliation(s)
- Satoko Aratani
- Department of Genome Science, Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Wang Y, Nakashima N, Sekiguchi T, Nishimoto T. Saccharomyces cerevisiae GTPase complex: Gtr1p-Gtr2p regulates cell-proliferation through Saccharomyces cerevisiae Ran-binding protein, Yrb2p. Biochem Biophys Res Commun 2005; 336:639-45. [PMID: 16143306 DOI: 10.1016/j.bbrc.2005.08.108] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2005] [Accepted: 08/16/2005] [Indexed: 11/25/2022]
Abstract
A Gtr1p GTPase, the GDP mutant of which suppresses both temperature-sensitive mutants of Saccharomyces cerevisiae RanGEF/Prp20p and RanGAP/Rna1p, was presently found to interact with Yrb2p, the S. cerevisiae homologue of mammalian Ran-binding protein 3. Gtr1p bound the Ran-binding domain of Yrb2p. In contrast, Gtr2p, a partner of Gtr1p, did not bind Yrb2p, although it bound Gtr1p. A triple mutant: yrb2delta gtr1delta gtr2delta was lethal, while a double mutant: gtr1delta gtr2delta survived well, indicating that Yrb2p protected cells from the killing effect of gtr1delta gtr2delta. Recombinant Gtr1p and Gtr2p were purified as a complex from Escherichia coli. The resulting Gtr1p-Gtr2p complex was comprised of an equal amount of Gtr1p and Gtr2p, which inhibited the Rna1p/Yrb2 dependent RanGAP activity. Thus, the Gtr1p-Gtr2p cycle was suggested to regulate the Ran cycle through Yrb2p.
Collapse
Affiliation(s)
- Yonggang Wang
- Department of Molecular Biology, Graduate School of Medical Science, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | | |
Collapse
|
39
|
Quensel C, Friedrich B, Sommer T, Hartmann E, Kohler M. In vivo analysis of importin alpha proteins reveals cellular proliferation inhibition and substrate specificity. Mol Cell Biol 2005; 24:10246-55. [PMID: 15542834 PMCID: PMC529023 DOI: 10.1128/mcb.24.23.10246-10255.2004] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The "classical" nuclear import pathway depends on importins alpha and beta. Humans have only one importin beta, while six alpha importins have been described. Whether or not distinct alpha importins are essential for specific import pathways in living human cells is unclear. We used RNA interference technology to specifically down-regulate the expression of ubiquitously expressed human alpha importins in HeLa cells. Down-regulation of importins alpha3, alpha5, alpha7, and beta strongly inhibited HeLa cell proliferation, while down-regulation of importins alpha1 and alpha4 had only a minor effect or no effect. Nucleoplasmin import was not prevented by down-regulation of any alpha importin, indicating that the importin alpha/beta pathway was generally not affected. In contrast, importin alpha3 or alpha5 down-regulation specifically inhibited the nuclear import of the Ran guanine nucleotide exchange factor, RCC1. Coinjection of recombinant alpha importins and RCC1 into down-regulated cells demonstrated that these transport defects were specifically caused by the limited availability of importin alpha3 in both cases. Thus, importin alpha3 is the only alpha importin responsible for the classical nuclear import of RCC1 in living cells.
Collapse
Affiliation(s)
- Christina Quensel
- Osteeklinik Damp GmbH, Sente-Deern-Ring 30, 24351 Osteebad Damp, Germany
| | | | | | | | | |
Collapse
|
40
|
Chen T, Brownawell AM, Macara IG. Nucleocytoplasmic shuttling of JAZ, a new cargo protein for exportin-5. Mol Cell Biol 2004; 24:6608-19. [PMID: 15254228 PMCID: PMC444848 DOI: 10.1128/mcb.24.15.6608-6619.2004] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2004] [Revised: 03/24/2004] [Accepted: 05/06/2004] [Indexed: 11/20/2022] Open
Abstract
Exportin-5 is a nuclear export receptor for certain classes of double-stranded RNA (dsRNA), including pre-micro-RNAs, viral hairpin RNAs, and some tRNAs. It can also export the RNA binding proteins ILF3 and elongation factor EF1A. However, the rules that determine which RNA binding proteins are exportin-5 cargoes remain unclear. JAZ possesses an unusual dsRNA binding domain consisting of multiple C2H2 zinc fingers. We found that JAZ binds to exportin-5 in a Ran-GTP- and dsRNA-dependent manner. Exportin-5 stimulates JAZ shuttling, and gene silencing of exportin-5 reduces shuttling. Recombinant exportin-5 also stimulates nuclear export of JAZ in permeabilized cells. JAZ also binds to ILF3, and surprisingly, this interaction is RNA independent, even though it requires the dsRNA binding domains of ILF3. Exportin-5, JAZ, and ILF3 can form a heteromeric complex with Ran-GTP and dsRNA, and JAZ increases ILF3 binding to exportin-5. JAZ does not contain a classical nuclear localization signal, and in digitonin-permeabilized cells, nuclear accumulation of JAZ does not require energy or cytosol. Nonetheless, low temperatures prevent JAZ import, suggesting that nuclear entry does not occur via simple diffusion. Together, these data suggest that JAZ is exported by exportin-5 but translocates back into nuclei by a facilitated diffusion mechanism.
Collapse
Affiliation(s)
- Ting Chen
- Center for Cell Signaling, Department of Microbiology, Health Sciences Center, University of Virginia School of Medicine, Charlottesville, VA 22908-0577, USA
| | | | | |
Collapse
|
41
|
Békési A, Zagyva I, Hunyadi-Gulyás E, Pongrácz V, Kovári J, Nagy AO, Erdei A, Medzihradszky KF, Vértessy BG. Developmental regulation of dUTPase in Drosophila melanogaster. J Biol Chem 2004; 279:22362-70. [PMID: 14996835 DOI: 10.1074/jbc.m313647200] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
dUTPase prevents uracil incorporation into DNA by strict regulation of the cellular dUTP:dTTP ratio. Lack of the enzyme initiates thymineless cell death, prompting studies on enzyme regulation. We investigated expression pattern and localization of Drosophila dUTPase. Similarly to human, two isoforms of the fly enzyme were identified at both mRNA and protein levels. During larval stages, a drastic decrease of dUTPase expression was demonstrated at the protein level. In contrast, dUTPase mRNAs display constitutive character throughout development. A putative nuclear localization signal was identified in one of the two isoforms. However, immunohistochemistry of ovaries and embryos did not show a clear correlation between the presence of this signal and subcellular localization of the protein, suggesting that the latter may be perturbed by additional factors. Results are in agreement with a multilevel regulation of dUTPase in the Drosophila proteome, possibly involving several interacting protein partners of the enzyme. Using independent approaches, the existence of such macromolecular partners was verified.
Collapse
Affiliation(s)
- Angéla Békési
- Institute of Enzymology, Biological Research Center, Hungarian Academy of Sciences, H-1518 Budapest, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Sakaguchi N, Miyamoto Y, Yoneda Y, Ogino K, Tachibana T. Generation of a Rat Monoclonal Antibody Specific for Importinα3/Qip1. ACTA ACUST UNITED AC 2003; 22:397-400. [PMID: 14683601 DOI: 10.1089/153685903771797110] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Importin alpha, which mediates the nuclear import of nuclear localization signal (NLS)-containing proteins, is a member of nuclear transport factors. Importin alpha binds directly NLS and functions as an adapter for accessing the importin beta-dependent import pathway. To date, several isoforms of importin alpha have been identified and classified into three subfamilies in higher eukaryotes. In this study, we report on the production of a rat monoclonal antibody (MAb) against importin alpha3/Qip1, a member of the importin alpha family, using a rat medial iliac lymph node method. The MAb 3D10 produced, reacted with both recombinant and endogenous importin alpha 3/Qip1. Immunoblotting analysis revealed that MAb 3D10 exclusively recognizes importin alpha3/Qip1 among members of the importin alpha family, in various mammalian cells.
Collapse
Affiliation(s)
- Naoko Sakaguchi
- Department of Applied & Bioapplied Chemistry, Graduate School of Engineering, Osaka City University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka 558-8585, Japan
| | | | | | | | | |
Collapse
|
43
|
Mason DA, Máthé E, Fleming RJ, Goldfarb DS. The Drosophila melanogaster importin α3 Locus Encodes an Essential Gene Required for the Development of Both Larval and Adult Tissues. Genetics 2003; 165:1943-58. [PMID: 14704178 PMCID: PMC1462915 DOI: 10.1093/genetics/165.4.1943] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Abstract
The nuclear transport of classical nuclear localization signal (cNLS)-containing proteins is mediated by the cNLS receptor importin α. The conventional importin α gene family in metazoan animals is composed of three clades that are conserved between flies and mammals and are referred to here as α1, α2, and α3. In contrast, plants and fungi contain only α1 genes. In this study we report that Drosophila importin α3 is required for the development of both larval and adult tissues. Importin α3 mutant flies die around the transition from first to second instar larvae, and homozygous importin α3 mutant eyes are defective. The transition to second instar larvae was rescued with importin α1, α2, or α3 transgenes, indicating that Importin α3 is normally required at this stage for an activity shared by all three importin α's. In contrast, an α3-specific biochemical activity(s) of Importin α3 is probably required for development to adults and photoreceptor cell development, since only an importin α3 transgene rescued these processes. These results are consistent with the view that the importin α's have both overlapping and distinct functions and that their role in animal development involves the spatial and temporal control of their expression.
Collapse
Affiliation(s)
- D Adam Mason
- Department of Biology, University of Rochester, Rochester, New York 14627, USA
| | | | | | | |
Collapse
|
44
|
Hakata Y, Yamada M, Shida H. A multifunctional domain in human CRM1 (exportin 1) mediates RanBP3 binding and multimerization of human T-cell leukemia virus type 1 Rex protein. Mol Cell Biol 2003; 23:8751-61. [PMID: 14612415 PMCID: PMC262658 DOI: 10.1128/mcb.23.23.8751-8761.2003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2003] [Revised: 05/19/2003] [Accepted: 08/29/2003] [Indexed: 11/20/2022] Open
Abstract
Human CRM1 (hCRM1) functions in the Rex-mediated mRNA export of human T-cell leukemia virus type 1 (HTLV-1) as an export receptor and as an inducing factor for Rex multimerization on its cognate RNA. Although there are only 24 amino acid differences between hCRM1 and rat CRM1 (rCRM1), rCRM1 can hardly support Rex activity, suggesting a role for rCRM1 as a determinant restricting the host range of HTLV-1. Here, we used a series of mutants, which were generated by interchanging residues of these CRM1s, to examine the relationship of hCRM1 functions. The functions for Rex multimerization and binding to nuclear export signals are mapped to different amino acid residues, and these are separable, suggesting that CRM1 not only functions as an export receptor but also participates in the formation of the RNA export complex through higher-ordered interaction with Rex. The region for the interaction with RanBP3, comprising four residues (amino acids [aa] 411, 414, 474, and 481), and the region for Rex multimerization, including two residues (aa 411 and 414), form an overlapped domain. Our results provide the molecular basis underlying the species-specific ability of HTLV-1 to propagate in human cells.
Collapse
Affiliation(s)
- Yoshiyuki Hakata
- Institute for Genetic Medicine, Hokkaido University, Kita-ku, Sapporo 060-0815, Japan
| | | | | |
Collapse
|
45
|
Andrade R, Alonso R, Peña R, Arlucea J, Aréchaga J. Localization of importin alpha (Rch1) at the plasma membrane and subcellular redistribution during lymphocyte activation. Chromosoma 2003; 112:87-95. [PMID: 12883947 DOI: 10.1007/s00412-003-0247-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2003] [Revised: 06/17/2003] [Accepted: 06/23/2003] [Indexed: 10/26/2022]
Abstract
Rch1 belongs to the importin alpha subfamily and works as an adapter between karyophilic proteins and the nuclear import machinery. Its level of expression varies among species and tissues, and depends on the state of cellular metabolism. In the present study we examined the level of expression of nuclear envelope and nuclear transport proteins (Rch1, importin beta, lamins A/C, lamin B, gp210, p62 and transportin) after human lymphocyte activation with phytohemagglutinin. We observed that the level of Rch1 increases dramatically, especially in larger lymphocytes, in response to activation. Moreover, using immunoelectron microscopy, this nuclear transport factor was found to be localized at the plasma membrane and also in tracks from the cytoplasm through the nuclear envelope into the nucleus. Similar localization was also observed in the human melanoma cell line A375. In addition, metabolic activation led to a redistribution of Rch1 from the cytoplasm to both the plasma membrane and the nuclear interior. These results suggest that, during lymphocyte activation, Rch1 may be involved in a signal transduction pathway that involves the shuttling of karyophilic proteins from the plasma membrane to the nucleus.
Collapse
Affiliation(s)
- Ricardo Andrade
- Department of Cell Biology and Histology, University of the Basque Country, Medical School, 48940 Leioa, Vizcaya, Spain
| | | | | | | | | |
Collapse
|
46
|
Cabot RA, Prather RS. Cleavage stage porcine embryos may have differing developmental requirements for karyopherins alpha2 and alpha3. Mol Reprod Dev 2003; 64:292-301. [PMID: 12548662 DOI: 10.1002/mrd.10238] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Numerous cellular proteins are able to localize to the nucleus due to the fact that they possess a nuclear localization signal (NLS) in their amino acid sequence. Nuclear localization sequences recognized by the importin alpha/beta heterodimer are found in cellular proteins capable of performing many diverse functions, ranging from chromatin remodeling to cell cycle regulation. Evidence has been presented that suggests individual importin alpha homologues are present at varying levels in different adult tissues. Other data have shown that specific subsets of NLSs found in different cellular proteins are recognized by individual importin alpha homologues with varying affinities. This evidence led us to hypothesize that due to the specific cargoes they carry, the mammalian embryo has different developmental requirements for individual importin alpha homologues. The results of the studies presented here indicate that importin alpha/beta-mediated import occurs throughout early cleavage in the porcine embryo, as determined by a reporter protein microinjection assay, and that multiple importin alpha homologues are present throughout early cleavage, as determined by immunocytochemical analysis. An RNA interference approach was used in an attempt to determine the developmental requirements for specific importin alpha homologues during early cleavage in the porcine embryo. Results from this study showed that fertilized porcine embryos injected with double stranded RNA (dsRNA) corresponding to the importin alpha homologue karyopherin alpha3 had significantly fewer nuclei following four days of culture than did embryos injected with dsRNA for another importin alpha homologue, karyopherin alpha2, or two control groups. This is the first report indicating that mammalian embryos may have differential developmental requirements for specific nuclear trafficking pathways.
Collapse
Affiliation(s)
- Ryan A Cabot
- Department of Animal Sciences, University of Missouri, 162 Animal Science Research Center, Columbia, Missouri 65211, USA
| | | |
Collapse
|
47
|
Nakashima K, Hagiwara T, Yamada M. Nuclear localization of peptidylarginine deiminase V and histone deimination in granulocytes. J Biol Chem 2002; 277:49562-8. [PMID: 12393868 DOI: 10.1074/jbc.m208795200] [Citation(s) in RCA: 273] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Peptidylarginine deiminase (PAD) deiminates arginine residues in proteins to citrulline residues Ca(2+) dependently. There are four types of PADs, I, II, III, and V, in humans. We studied the subcellular distribution of PAD V in HL-60 granulocytes and peripheral blood granulocytes. Expression of green fluorescent protein-tagged PADs in HeLa cells revealed that PAD V is localized in the nucleus, whereas PAD I, II, and III are localized in the cytoplasm. PAD V deletion mutants indicated that the sequence residues 45-74 have a nuclear localization signal (NLS). A sequence feature of this NLS is a three-lysine residue cluster preceded by a proline residue and is not found in the three other PADs. Substitution of the lysine cluster by an alanine cluster abrogated the nuclear import activity. These results suggested that the NLS is a classical monopartite NLS. HL-60 granulocytes, neutrophils, and eosinophils stained with antibody specific for PAD V exhibited distinct positive signals in the nucleus. Subcellular fractionation of HL-60 granulocytes also showed the nuclear localization of the enzyme. When neutrophils were stimulated with calcium ionophore, protein deimination occurred in the nucleus. The major deiminated proteins were identified as histones H2A, H3, and H4. The implication of PAD V in histone modifications is discussed.
Collapse
Affiliation(s)
- Katsuhiko Nakashima
- Graduate School of Integrated Science, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan
| | | | | |
Collapse
|
48
|
Steggerda SM, Paschal BM. Regulation of nuclear import and export by the GTPase Ran. INTERNATIONAL REVIEW OF CYTOLOGY 2002; 217:41-91. [PMID: 12019565 DOI: 10.1016/s0074-7696(02)17012-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This review focuses on the control of nuclear import and export pathways by the small GTPase Ran. Transport of signal-containing cargo substrates is mediated by receptors that bind to the cargo proteins and RNAs and deliver them to the appropriate cellular compartment. Ran is an evolutionarily conserved member of the Ras superfamily that regulates all receptor-mediated transport between the nucleus and the cytoplasm. We describe the identification and characterization of the RanGTPase and its binding partners: the guanine nucleotide exchange factor, RanGEF; the GTPase activating protein, RanGAP; the soluble import and export receptors; Ran-binding domain-(RBD) containing proteins; and NTF2 and related factors.
Collapse
Affiliation(s)
- Susanne M Steggerda
- Center for Cell Signaling and Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville 22908, USA
| | | |
Collapse
|
49
|
Lindsay ME, Plafker K, Smith AE, Clurman BE, Macara IG. Npap60/Nup50 is a tri-stable switch that stimulates importin-alpha:beta-mediated nuclear protein import. Cell 2002; 110:349-60. [PMID: 12176322 DOI: 10.1016/s0092-8674(02)00836-x] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Many nuclear-targeted proteins are transported through the nuclear pore complex (NPC) by the importin-alpha:beta receptor. We now show that Npap60 (also called Nup50), a protein previously believed to be a structural component of the NPC, is a Ran binding protein and a cofactor for importin-alpha:beta-mediated import. Npap60 is a tri-stable switch that alternates between binding modes. The C terminus binds importin-beta through RanGTP. The N terminus binds the C terminus of importin-alpha, while a central domain binds importin-beta. Npap60:importin-alpha:beta binds cargo and can stimulate nuclear import. Endogenous Npap60 can shuttle and is accessible from the cytoplasmic side of the nuclear envelope. These results identify Npap60 as a cofactor for importin-alpha:beta nuclear import and as a previously unidentified subunit of the importin complex.
Collapse
Affiliation(s)
- Mark E Lindsay
- Center for Cell Signaling, Department of Microbiology, University of Virginia School of Medicine, Charlottesville 22908, USA
| | | | | | | | | |
Collapse
|
50
|
Romanelli MG, Morandi C. Importin alpha binds to an unusual bipartite nuclear localization signal in the heterogeneous ribonucleoprotein type I. EUROPEAN JOURNAL OF BIOCHEMISTRY 2002; 269:2727-34. [PMID: 12047381 DOI: 10.1046/j.1432-1033.2002.02942.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The heterogeneous nuclear ribonucleoprotein (hnRNP) type I, a modulator of alternative splicing, localizes in the nucleoplasm of mammalian cells and in a discrete perinucleolar structure. HnRNP I contains a novel type of bipartite nuclear localization signal (NLS) at the N-terminus of the protein that we have previously named nuclear determinant localization type I (NLD-I). Recently, a neural counterpart of hnRNP I has been identified that contains a putative NLS with two strings of basic amino acids separated by a spacer of 30 residues. In the present study we show that the neural hnRNP I NLS is necessary and sufficient for nuclear localization and represents a variant of the novel bipartite NLS present in the NLD-I domain. Furthermore, we demonstrate that the NLD-I is transported into the nucleus by cytoplasmic factor(s) with active transport modality. Binding assays using recombinant importin alpha show an interaction with NLD-I similar to that of SV40 large T antigen NLS. Deletion analysis indicates that both stretches of basic residues are necessary for binding to importin alpha. The above experimental results lead to the conclusion that importin alpha acts as cytoplasmic receptor for proteins characterized by a bipartite NLS signal that extends up to 37 residues.
Collapse
Affiliation(s)
- Maria G Romanelli
- Department of Mother and Child, Biology and Genetics, University of Verona, Italy.
| | | |
Collapse
|