1
|
Nematollahzadeh S, Athukorala A, Donnelly CM, Pavan S, Atelie-Djossou V, Di Iorio E, Nath B, Helbig KJ, McSharry BP, Forwood JK, Sarker S, Alvisi G. Mechanistic Insights Into an Ancient Adenovirus Precursor Protein VII Show Multiple Nuclear Import Receptor Pathways. Traffic 2024; 25:e12953. [PMID: 39301720 DOI: 10.1111/tra.12953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/10/2024] [Accepted: 07/30/2024] [Indexed: 09/22/2024]
Abstract
Adenoviral pVII proteins are multifunctional, highly basic, histone-like proteins that can bind to and transport the viral genome into the host cell nucleus. Despite the identification of several nuclear localization signals (NLSs) in the pVII protein of human adenovirus (HAdV)2, the mechanistic details of nuclear transport are largely unknown. Here we provide a full characterization of the nuclear import of precursor (Pre-) pVII protein from an ancient siadenovirus, frog siadenovirus 1 (FrAdV1), using a combination of structural, functional, and biochemical approaches. Two strong NLSs (termed NLSa and NLSd) interact with importin (IMP)β1 and IMPα, respectively, and are the main drivers of nuclear import. A weaker NLS (termed NLSb) also contributes, together with an additional signal (NLSc) which we found to be important for nucleolar targeting and intranuclear binding. Expression of wild-type and NLS defective derivatives Pre-pVII in the presence of selective inhibitors of different nuclear import pathways revealed that, unlike its human counterpart, FrAdV1 Pre-pVII nuclear import is dependent on IMPα/β1 and IMPβ1, but not on transportin-1 (IMPβ2). Clearly, AdVs evolved to maximize the nuclear import pathways for the pVII proteins, whose subcellular localization is the result of a complex process. Therefore, our results pave the way for an evolutionary comparison of the interaction of different AdVs with the host cell nuclear transport machinery.
Collapse
Affiliation(s)
| | - Ajani Athukorala
- Department of Microbiology, Anatomy, Physiology, and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria, Australia
| | - Camilla M Donnelly
- School of Dentistry and Medical Sciences Biomedical Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia
- Gulbali Institute, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - Silvia Pavan
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | - Enzo Di Iorio
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Babu Nath
- School of Dentistry and Medical Sciences Biomedical Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - Karla J Helbig
- Department of Microbiology, Anatomy, Physiology, and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria, Australia
| | - Brian P McSharry
- School of Dentistry and Medical Sciences Biomedical Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia
- Gulbali Institute, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - Jade K Forwood
- School of Dentistry and Medical Sciences Biomedical Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia
- Gulbali Institute, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - Subir Sarker
- Department of Microbiology, Anatomy, Physiology, and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria, Australia
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Queensland, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia
| | - Gualtiero Alvisi
- Department of Molecular Medicine, University of Padua, Padua, Italy
| |
Collapse
|
2
|
Cross-regulation of viral kinases with cyclin A secures shutoff of host DNA synthesis. Nat Commun 2020; 11:4845. [PMID: 32973148 PMCID: PMC7518283 DOI: 10.1038/s41467-020-18542-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 08/24/2020] [Indexed: 12/25/2022] Open
Abstract
Herpesviruses encode conserved protein kinases (CHPKs) to stimulate phosphorylation-sensitive processes during infection. How CHPKs bind to cellular factors and how this impacts their regulatory functions is poorly understood. Here, we use quantitative proteomics to determine cellular interaction partners of human herpesvirus (HHV) CHPKs. We find that CHPKs can target key regulators of transcription and replication. The interaction with Cyclin A and associated factors is identified as a signature of β-herpesvirus kinases. Cyclin A is recruited via RXL motifs that overlap with nuclear localization signals (NLS) in the non-catalytic N termini. This architecture is conserved in HHV6, HHV7 and rodent cytomegaloviruses. Cyclin A binding competes with NLS function, enabling dynamic changes in CHPK localization and substrate phosphorylation. The cytomegalovirus kinase M97 sequesters Cyclin A in the cytosol, which is essential for viral inhibition of cellular replication. Our data highlight a fine-tuned and physiologically important interplay between a cellular cyclin and viral kinases.
Collapse
|
3
|
Zhang S, Williamson NA, Duvick L, Lee A, Orr HT, Korlin-Downs A, Yang P, Mok YF, Jans DA, Bogoyevitch MA. The ataxin-1 interactome reveals direct connection with multiple disrupted nuclear transport pathways. Nat Commun 2020; 11:3343. [PMID: 32620905 PMCID: PMC7334205 DOI: 10.1038/s41467-020-17145-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 06/09/2020] [Indexed: 11/21/2022] Open
Abstract
The expanded polyglutamine (polyQ) tract form of ataxin-1 drives disease progression in spinocerebellar ataxia type 1 (SCA1). Although known to form distinctive intranuclear bodies, the cellular pathways and processes that polyQ-ataxin-1 influences remain poorly understood. Here we identify the direct and proximal partners constituting the interactome of ataxin-1[85Q] in Neuro-2a cells, pathways analyses indicating a significant enrichment of essential nuclear transporters, pointing to disruptions in nuclear transport processes in the presence of elevated levels of ataxin-1. Our direct assessments of nuclear transporters and their cargoes confirm these observations, revealing disrupted trafficking often with relocalisation of transporters and/or cargoes to ataxin-1[85Q] nuclear bodies. Analogous changes in importin-β1, nucleoporin 98 and nucleoporin 62 nuclear rim staining are observed in Purkinje cells of ATXN1[82Q] mice. The results highlight a disruption of multiple essential nuclear protein trafficking pathways by polyQ-ataxin-1, a key contribution to furthering understanding of pathogenic mechanisms initiated by polyQ tract proteins.
Collapse
Affiliation(s)
- Sunyuan Zhang
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Nicholas A Williamson
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Lisa Duvick
- Institute of Translational Neuroscience, and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Alexander Lee
- Nuclear Signalling Lab., Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Harry T Orr
- Institute of Translational Neuroscience, and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Austin Korlin-Downs
- Institute of Translational Neuroscience, and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Praseuth Yang
- Institute of Translational Neuroscience, and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Yee-Foong Mok
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - David A Jans
- Nuclear Signalling Lab., Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia.
| | - Marie A Bogoyevitch
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| |
Collapse
|
4
|
Tavolieri MV, Droppelmann CA, Campos-Melo D, Volkening K, Strong MJ. A novel overlapping NLS/NES region within the PH domain of Rho Guanine Nucleotide Exchange Factor (RGNEF) regulates its nuclear-cytoplasmic localization. Eur J Cell Biol 2019; 98:27-35. [PMID: 30482479 DOI: 10.1016/j.ejcb.2018.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 10/08/2018] [Accepted: 11/13/2018] [Indexed: 10/27/2022] Open
Abstract
Rho Guanine Nucleotide Exchange Factor (RGNEF) is a 190 kDa protein implicated in both amyotrophic lateral sclerosis (ALS) and cancer. Under normal physiological conditions, RGNEF is predominantly cytoplasmic with moderate levels of nuclear localization. We have identified a 23-amino acid region containing a bipartite nuclear localization signal (NLS) within the Pleckstrin Homology (PH) domain of RGNEF, which when deleted or mutated abolishes the nuclear localization of this protein. Fusion proteins containing only the PH domain demonstrated that this region by itself is able to translocate a 160 kDa protein to the nucleus. Interestingly, we also detected a nuclear export signal (NES) within the linker region of this bipartite NLS which is able to export from the nucleus a fusion protein containing two NLSs. Experiments using Leptomycin-B -an inhibitor of nuclear export- confirmed that this region promotes nuclear export in an exportin-1 dependent manner. This study is the first report demonstrating either of these signals embedded within a PH domain. Notably, this is also the first description of a functional overlapped NLS/NES signal.
Collapse
Affiliation(s)
- Michael V Tavolieri
- Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada.
| | - Cristian A Droppelmann
- Molecular Medicine Group, Robarts Research Institute, Western University, London, Ontario, Canada.
| | - Danae Campos-Melo
- Molecular Medicine Group, Robarts Research Institute, Western University, London, Ontario, Canada.
| | - Kathryn Volkening
- Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; Molecular Medicine Group, Robarts Research Institute, Western University, London, Ontario, Canada.
| | - Michael J Strong
- Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; Molecular Medicine Group, Robarts Research Institute, Western University, London, Ontario, Canada.
| |
Collapse
|
5
|
Pieters T, Goossens S, Haenebalcke L, Andries V, Stryjewska A, De Rycke R, Lemeire K, Hochepied T, Huylebroeck D, Berx G, Stemmler MP, Wirth D, Haigh JJ, van Hengel J, van Roy F. p120 Catenin-Mediated Stabilization of E-Cadherin Is Essential for Primitive Endoderm Specification. PLoS Genet 2016; 12:e1006243. [PMID: 27556156 PMCID: PMC4996431 DOI: 10.1371/journal.pgen.1006243] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/14/2016] [Indexed: 12/15/2022] Open
Abstract
E-cadherin-mediated cell-cell adhesion is critical for naive pluripotency of cultured mouse embryonic stem cells (mESCs). E-cadherin-depleted mESC fail to downregulate their pluripotency program and are unable to initiate lineage commitment. To further explore the roles of cell adhesion molecules during mESC differentiation, we focused on p120 catenin (p120ctn). Although one key function of p120ctn is to stabilize and regulate cadherin-mediated cell-cell adhesion, it has many additional functions, including regulation of transcription and Rho GTPase activity. Here, we investigated the role of mouse p120ctn in early embryogenesis, mESC pluripotency and early fate determination. In contrast to the E-cadherin-null phenotype, p120ctn-null mESCs remained pluripotent, but their in vitro differentiation was incomplete. In particular, they failed to form cystic embryoid bodies and showed defects in primitive endoderm formation. To pinpoint the underlying mechanism, we undertook a structure-function approach. Rescue of p120ctn-null mESCs with different p120ctn wild-type and mutant expression constructs revealed that the long N-terminal domain of p120ctn and its regulatory domain for RhoA were dispensable, whereas its armadillo domain and interaction with E-cadherin were crucial for primitive endoderm formation. We conclude that p120ctn is not only an adaptor and regulator of E-cadherin, but is also indispensable for proper lineage commitment.
Collapse
Affiliation(s)
- Tim Pieters
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Steven Goossens
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Lieven Haenebalcke
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
| | - Vanessa Andries
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
| | - Agata Stryjewska
- Department of Development and Regeneration, Laboratory of Molecular Biology (Celgen), University of Leuven, Leuven, Belgium
| | - Riet De Rycke
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
| | - Kelly Lemeire
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
| | - Tino Hochepied
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
| | - Danny Huylebroeck
- Department of Development and Regeneration, Laboratory of Molecular Biology (Celgen), University of Leuven, Leuven, Belgium
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Geert Berx
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Marc P. Stemmler
- Department of Molecular Embryology, Max-Planck Institute of Immunobiology, Freiburg, Germany
- Department of Experimental Medicine I, Nikolaus-Fiebiger Center for Molecular Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Dagmar Wirth
- Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Jody J. Haigh
- Mammalian Functional Genetics Laboratory, Division of Blood Cancers, Australian Centre for Blood Diseases, Department of Clinical Haematology, Monash University and Alfred Health Alfred Centre, Melbourne, Victoria, Australia
| | - Jolanda van Hengel
- Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- * E-mail: (JvH); (FvR)
| | - Frans van Roy
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- * E-mail: (JvH); (FvR)
| |
Collapse
|
6
|
Spencer NY, Yan Z, Cong L, Zhang Y, Engelhardt JF, Stanton RC. Definitive localization of intracellular proteins: Novel approach using CRISPR-Cas9 genome editing, with glucose 6-phosphate dehydrogenase as a model. Anal Biochem 2016; 494:55-67. [PMID: 26576833 PMCID: PMC4695245 DOI: 10.1016/j.ab.2015.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 10/27/2015] [Accepted: 11/02/2015] [Indexed: 01/23/2023]
Abstract
Studies to determine subcellular localization and translocation of proteins are important because subcellular localization of proteins affects every aspect of cellular function. Such studies frequently utilize mutagenesis to alter amino acid sequences hypothesized to constitute subcellular localization signals. These studies often utilize fluorescent protein tags to facilitate live cell imaging. These methods are excellent for studies of monomeric proteins, but for multimeric proteins, they are unable to rule out artifacts from native protein subunits already present in the cells. That is, native monomers might direct the localization of fluorescent proteins with their localization signals obliterated. We have developed a method for ruling out such artifacts, and we use glucose 6-phosphate dehydrogenase (G6PD) as a model to demonstrate the method's utility. Because G6PD is capable of homodimerization, we employed a novel approach to remove interference from native G6PD. We produced a G6PD knockout somatic (hepatic) cell line using CRISPR-Cas9 mediated genome engineering. Transfection of G6PD knockout cells with G6PD fluorescent mutant proteins demonstrated that the major subcellular localization sequences of G6PD are within the N-terminal portion of the protein. This approach sets a new gold standard for similar studies of subcellular localization signals in all homodimerization-capable proteins.
Collapse
Affiliation(s)
- Netanya Y Spencer
- Joslin Diabetes Center, 1 Joslin Place, Boston, MA, 02215, USA; Harvard Medical School, 25 Shattuck St., Boston, MA, 02115, USA.
| | - Ziying Yan
- Department of Anatomy and Cell Biology, The University of Iowa, 51 Newton Rd., Iowa City, IA, 52242, USA
| | - Le Cong
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA, 02142, USA
| | - Yulong Zhang
- Department of Anatomy and Cell Biology, The University of Iowa, 51 Newton Rd., Iowa City, IA, 52242, USA
| | - John F Engelhardt
- Department of Anatomy and Cell Biology, The University of Iowa, 51 Newton Rd., Iowa City, IA, 52242, USA
| | - Robert C Stanton
- Joslin Diabetes Center, 1 Joslin Place, Boston, MA, 02215, USA; Harvard Medical School, 25 Shattuck St., Boston, MA, 02115, USA; Beth Israel Deaconess Medical Center, 330 Brookline Ave., Boston, MA, 02215, USA
| |
Collapse
|
7
|
Hochrainer K, Pejanovic N, Olaseun VA, Zhang S, Iadecola C, Anrather J. The ubiquitin ligase HERC3 attenuates NF-κB-dependent transcription independently of its enzymatic activity by delivering the RelA subunit for degradation. Nucleic Acids Res 2015; 43:9889-904. [PMID: 26476452 PMCID: PMC4787756 DOI: 10.1093/nar/gkv1064] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/06/2015] [Indexed: 11/21/2022] Open
Abstract
Activation of NF-κB-dependent transcription represents an important hallmark of inflammation. While the acute inflammatory response is per se beneficial, it can become deleterious if its spatial and temporal profile is not tightly controlled. Classically, NF-κB activity is limited by cytoplasmic retention of the NF-κB dimer through binding to inhibitory IκB proteins. However, increasing evidence suggests that NF-κB activity can also be efficiently contained by direct ubiquitination of NF-κB subunits. Here, we identify the HECT-domain ubiquitin ligase HERC3 as novel negative regulator of NF-κB activity. We find that HERC3 restricts NF-κB nuclear import and DNA binding without affecting IκBα degradation. Instead HERC3 indirectly binds to the NF-κB RelA subunit after liberation from IκBα inhibitor leading to its ubiquitination and protein destabilization. Remarkably, the regulation of RelA activity by HERC3 is independent of its inherent ubiquitin ligase activity. Rather, we show that HERC3 and RelA are part of a multi-protein complex containing the proteasome as well as the ubiquitin-like protein ubiquilin-1 (UBQLN1). We present evidence that HERC3 and UBQLN1 provide a link between NF-κB RelA and the 26S proteasome, thereby facilitating RelA protein degradation. Our findings establish HERC3 as novel candidate regulating the inflammatory response initiated by NF-κB.
Collapse
Affiliation(s)
- Karin Hochrainer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY10065, USA
| | - Nadja Pejanovic
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY10065, USA Instituto Gulbenkian de Ciência, Apartado 14, Oeiras, Portugal
| | | | - Sheng Zhang
- Institute of Biotechnology and Life Sciences Biotechnologies, Cornell University, Ithaca, NY14853, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY10065, USA
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY10065, USA
| |
Collapse
|
8
|
Róna G, Borsos M, Ellis JJ, Mehdi AM, Christie M, Környei Z, Neubrandt M, Tóth J, Bozóky Z, Buday L, Madarász E, Bodén M, Kobe B, Vértessy BG. Dynamics of re-constitution of the human nuclear proteome after cell division is regulated by NLS-adjacent phosphorylation. Cell Cycle 2015; 13:3551-64. [PMID: 25483092 DOI: 10.4161/15384101.2014.960740] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Phosphorylation by the cyclin-dependent kinase 1 (Cdk1) adjacent to nuclear localization signals (NLSs) is an important mechanism of regulation of nucleocytoplasmic transport. However, no systematic survey has yet been performed in human cells to analyze this regulatory process, and the corresponding cell-cycle dynamics have not yet been investigated. Here, we focused on the human proteome and found that numerous proteins, previously not identified in this context, are associated with Cdk1-dependent phosphorylation sites adjacent to their NLSs. Interestingly, these proteins are involved in key regulatory events of DNA repair, epigenetics, or RNA editing and splicing. This finding indicates that cell-cycle dependent events of genome editing and gene expression profiling may be controlled by nucleocytoplasmic trafficking. For in-depth investigations, we selected a number of these proteins and analyzed how point mutations, expected to modify the phosphorylation ability of the NLS segments, perturb nucleocytoplasmic localization. In each case, we found that mutations mimicking hyper-phosphorylation abolish nuclear import processes. To understand the mechanism underlying these phenomena, we performed a video microscopy-based kinetic analysis to obtain information on cell-cycle dynamics on a model protein, dUTPase. We show that the NLS-adjacent phosphorylation by Cdk1 of human dUTPase, an enzyme essential for genomic integrity, results in dynamic cell cycle-dependent distribution of the protein. Non-phosphorylatable mutants have drastically altered protein re-import characteristics into the nucleus during the G1 phase. Our results suggest a dynamic Cdk1-driven mechanism of regulation of the nuclear proteome composition during the cell cycle.
Collapse
Key Words
- Cdc28, cyclin-dependent protein kinase (Cdk) encoded by CDC28
- Cdk1, cyclin-dependent kinase 1
- GO, gene ontology
- NES, nuclear export signal
- NLS, nuclear localization signal
- SNP, single nucleotide polymorphisms
- SV40, Simian virus 40
- UBA1, Ubiquitin-activating enzyme E1
- UNG2, Human Uracil-DNA glycosylase 2
- cNLS, classical nuclear localization signal
- cell cycle
- dNTP, deoxyribonucleotide triphosphate
- dTTP, deoxythymidine triphosphate
- dUMP, deoxyuridine monophosphate
- dUTP, deoxyuridine triphosphate
- dUTPase
- importin
- phosphorylation
- trafficking
Collapse
Affiliation(s)
- Gergely Róna
- a Institute of Enzymology; RCNS; Hungarian Academy of Sciences ; Budapest , Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Olmos Y, Hodgson L, Mantell J, Verkade P, Carlton JG. ESCRT-III controls nuclear envelope reformation. Nature 2015; 522:236-9. [PMID: 26040713 PMCID: PMC4471131 DOI: 10.1038/nature14503] [Citation(s) in RCA: 263] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 04/27/2015] [Indexed: 12/12/2022]
Abstract
During telophase, the nuclear envelope (NE) reforms around daughter nuclei to ensure proper segregation of nuclear and cytoplasmic contents. NE reformation requires the coating of chromatin by membrane derived from the endoplasmic reticulum, and a subsequent annular fusion step to ensure that the formed envelope is sealed. How annular fusion is accomplished is unknown, but it is thought to involve the p97 AAA-ATPase complex and bears a topological equivalence to the membrane fusion event that occurs during the abscission phase of cytokinesis. Here we show that the endosomal sorting complex required for transport-III (ESCRT-III) machinery localizes to sites of annular fusion in the forming NE in human cells, and is necessary for proper post-mitotic nucleo-cytoplasmic compartmentalization. The ESCRT-III component charged multivesicular body protein 2A (CHMP2A) is directed to the forming NE through binding to CHMP4B, and provides an activity essential for NE reformation. Localization also requires the p97 complex member ubiquitin fusion and degradation 1 (UFD1). Our results describe a novel role for the ESCRT machinery in cell division and demonstrate a conservation of the machineries involved in topologically equivalent mitotic membrane remodelling events.
Collapse
Affiliation(s)
- Yolanda Olmos
- Division of Cancer Studies, Section of Cell Biology and Imaging King’s College London London SE1 1UL
| | - Lorna Hodgson
- School of Biochemistry University of Bristol Medical Sciences Building University Walk Bristol BS8 1TD
| | - Judith Mantell
- School of Biochemistry University of Bristol Medical Sciences Building University Walk Bristol BS8 1TD
- Wolfson Bioimaging Facility University of Bristol Medical Sciences Building University Walk Bristol BS8 1TD
| | - Paul Verkade
- School of Biochemistry University of Bristol Medical Sciences Building University Walk Bristol BS8 1TD
- Wolfson Bioimaging Facility University of Bristol Medical Sciences Building University Walk Bristol BS8 1TD
- School of Physiology & Pharmacology University of Bristol Medical Sciences Building University Walk Bristol BS8 1TD
| | - Jeremy G Carlton
- Division of Cancer Studies, Section of Cell Biology and Imaging King’s College London London SE1 1UL
| |
Collapse
|
10
|
Salemi LM, Loureiro SO, Schild-Poulter C. Characterization of RanBPM molecular determinants that control its subcellular localization. PLoS One 2015; 10:e0117655. [PMID: 25659156 PMCID: PMC4319831 DOI: 10.1371/journal.pone.0117655] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 12/30/2014] [Indexed: 12/14/2022] Open
Abstract
RanBPM/RanBP9 is a ubiquitous, nucleocytoplasmic protein that is part of an evolutionary conserved E3 ubiquitin ligase complex whose function and targets in mammals are still unknown. RanBPM itself has been implicated in various cellular processes that involve both nuclear and cytoplasmic functions. However, to date, little is known about how RanBPM subcellular localization is regulated. We have conducted a systematic analysis of RanBPM regions that control its subcellular localization using RanBPM shRNA cells to examine ectopic RanBPM mutant subcellular localization without interference from the endogenously expressed protein. We show that several domains and motifs regulate RanBPM nuclear and cytoplasmic localization. In particular, RanBPM comprises two motifs that can confer nuclear localization, one proline/glutamine-rich motif in the extreme N-terminus which has a dominant effect on RanBPM localization, and a second motif in the C-terminus which minimally contributes to RanBPM nuclear targeting. We also identified a nuclear export signal (NES) which mutation prevented RanBPM accumulation in the cytoplasm. Likewise, deletion of the central RanBPM conserved domains (SPRY and LisH/CTLH) resulted in the relocalization of RanBPM to the nucleus, suggesting that RanBPM cytoplasmic localization is also conferred by protein-protein interactions that promote its cytoplasmic retention. Indeed we found that in the cytoplasm, RanBPM partially colocalizes with microtubules and associates with α-tubulin. Finally, in the nucleus, a significant fraction of RanBPM is associated with chromatin. Altogether, these analyses reveal that RanBPM subcellular localization results from the combined effects of several elements that either confer direct transport through the nucleocytoplasmic transport machinery or regulate it indirectly, likely through interactions with other proteins and by intramolecular folding.
Collapse
Affiliation(s)
- Louisa M. Salemi
- Robarts Research Institute and Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Sandra O. Loureiro
- Robarts Research Institute and Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Caroline Schild-Poulter
- Robarts Research Institute and Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
11
|
Thomas M, Zielke B, Reuter N, Stamminger T. Methods to study the nucleocytoplasmic transport of macromolecules with respect to their impact on the regulation of human cytomegalovirus gene expression. Methods Mol Biol 2014; 1119:197-216. [PMID: 24639225 DOI: 10.1007/978-1-62703-788-4_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
One defining feature of eukaryotic cells is their compartmentalization into nucleus and cytoplasm which provides sophisticated opportunities for the regulation of gene expression. Accurate subcellular localization is crucial for the effective function of most viral macromolecules, and nuclear translocation is central to the function of herpesviral proteins that are involved in processes such as transcription or DNA replication. Human cytomegalovirus (HCMV) encodes several transactivator proteins which stimulate viral gene expression either on the transcriptional or posttranscriptional level. In this chapter, we focus on nucleocytoplasmic transport mechanisms of either proteins or RNA that are utilized during HCMV infection. We describe commonly used assays to determine the subcellular localization of a protein, its nucleocytoplasmic shuttling activity, its capacity to export unspliced RNA from the nucleus, and its association with RNA in vivo.
Collapse
Affiliation(s)
- Marco Thomas
- Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | | | | |
Collapse
|
12
|
Held C, Webel R, Palmisano R, Hutterer C, Marschall M, Wittenberg T. Using multi-channel level sets to measure the cytoplasmic localization of HCMV pUL97 in GFP-B-gal fusion constructs. J Virol Methods 2014; 199:61-7. [PMID: 24445057 DOI: 10.1016/j.jviromet.2013.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 11/26/2013] [Accepted: 12/16/2013] [Indexed: 10/25/2022]
Abstract
Human cytomegalovirus UL97-encoded protein kinase (pUL97) phosphorylates cellular and viral proteins and is critical for viral replication. To quantify the efficiency of nuclear translocation and to elucidate the role of putative nuclear localization signal (NLS) elements, immunofluorescence analysis of different pUL97 expression constructs was performed. Since manual quantitation of respective expression levels lacks objectivity and reproducibility, and is time-consuming as well, a computer-based model is established. This model enables objective quantitation of the degree of cytoplasmic localization λ. To determine the degree of cytoplasmic localization of different pUL97-GFP-β-gal fusion proteins automatically, a multi-channel segmentation of the nucleus and cytoplasm of transfected HeLa cells is performed in DAPI and GFP micrographs. A watershed transform-based segmentation scheme is used for the segmentation of the cell nuclei. Subsequently, the cytoplasm is segmented using a fast marching level set method. Based on the segmentation of cell nuclei and cytoplasm, λ can be determined for each HeLa cell by quantitation of the ratio of average signal intensity outside and inside the nucleus. The degree of cytoplasmic localization of an individual construct is then determined by evaluating the average and standard deviation of λ for the corresponding HeLa cells. Evaluation demonstrates that nuclear transport of pUL97 is a multilayered mechanism resulting in different efficiencies of nuclear translocation between a small and a large isoform and objective quantitation of the cytoplasmic localization is possible with a high accuracy (96.7% and 94.3%).
Collapse
Affiliation(s)
- Christian Held
- Department of Image Processing and Biomedical Engineering, Fraunhofer Institute for Integrated Circuits IIS, Erlangen, Germany.
| | - Rike Webel
- Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg, Germany.
| | - Ralf Palmisano
- Optical Imaging Center Erlangen, OICE, Erlangen, Germany.
| | - Corina Hutterer
- Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg, Germany.
| | - Manfred Marschall
- Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg, Germany.
| | - Thomas Wittenberg
- Department of Image Processing and Biomedical Engineering, Fraunhofer Institute for Integrated Circuits IIS, Erlangen, Germany.
| |
Collapse
|
13
|
Davies RG, Wagstaff KM, McLaughlin EA, Loveland KL, Jans DA. The BRCA1-binding protein BRAP2 can act as a cytoplasmic retention factor for nuclear and nuclear envelope-localizing testicular proteins. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:3436-3444. [DOI: 10.1016/j.bbamcr.2013.05.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 05/10/2013] [Accepted: 05/13/2013] [Indexed: 01/07/2023]
|
14
|
Korb E, Wilkinson CL, Delgado RN, Lovero KL, Finkbeiner S. Arc in the nucleus regulates PML-dependent GluA1 transcription and homeostatic plasticity. Nat Neurosci 2013; 16:874-83. [PMID: 23749147 PMCID: PMC3703835 DOI: 10.1038/nn.3429] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 05/12/2013] [Indexed: 02/06/2023]
Abstract
The activity-regulated cytoskeletal protein Arc (also known as Arg3.1) is required for long-term memory formation and synaptic plasticity. Arc expression is robustly induced by activity, and Arc protein localizes to both active synapses and the nucleus. Whereas its synaptic function has been examined, it is not clear why or how Arc is localized to the nucleus. We found that murine Arc nuclear expression is regulated by synaptic activity in vivo and in vitro. We identified distinct regions of Arc that control its localization, including a nuclear localization signal, a nuclear retention domain and a nuclear export signal. Arc localization to the nucleus promotes an activity-induced increase in the expression of promyelocytic leukemia nuclear bodies, which decreases GluA1 (also called Gria1) transcription and synaptic strength. We further show that Arc nuclear localization regulates homeostatic plasticity. Thus, Arc mediates the homeostatic response to increased activity by translocating to the nucleus, increasing promyelocytic leukemia protein expression and decreasing GluA1 transcription, ultimately downscaling synaptic strength.
Collapse
Affiliation(s)
- Erica Korb
- Gladstone Institutes of Neurological Disease, San Francisco, California, USA
| | | | | | | | | |
Collapse
|
15
|
Schmeiser C, Borst E, Sticht H, Marschall M, Milbradt J. The cytomegalovirus egress proteins pUL50 and pUL53 are translocated to the nuclear envelope through two distinct modes of nuclear import. J Gen Virol 2013; 94:2056-2069. [PMID: 23740483 DOI: 10.1099/vir.0.052571-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The nucleocytoplasmic export of cytomegaloviral capsids is regulated by formation of a multi-component nuclear egress complex (NEC), essentially based on viral proteins pUL50 and pUL53. In this study, the generation of recombinant human cytomegaloviruses, expressing tagged versions of pUL50 and pUL53, enabled the investigation of NEC formation in infected primary fibroblasts. For these recombinant viruses, a wild-type-like mode of pUL50-pUL53 interaction and recruitment of both proteins to the nuclear envelope could be demonstrated. Importantly, pUL50 was translocated from an initial cytoplasmic distribution to the nuclear rim, whereas pUL53 accumulated in the nucleus before attaining overall rim colocalization with pUL50. Specified experimental settings illustrated that pUL50 and pUL53 were subject to different pathways of intracellular trafficking. Importantly, a novel nuclear localization signal (NLS) could be identified and functionally verified for pUL53 (amino acids 18-27), whereas no NLS was present in pUL50. Analysis of amino acid replacement mutants further illustrated the differential modes of nuclear import of the two essential viral egress proteins. Taken together, our findings suggest a combination of classical nuclear import (pUL53) and interaction-mediated recruitment (pUL50) as the driving forces for core NEC formation and viral nuclear egress.
Collapse
Affiliation(s)
- Cathrin Schmeiser
- Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Eva Borst
- Department of Virology, Hannover Medical School, Hannover, Germany
| | - Heinrich Sticht
- Division of Bioinformatics, Institute of Biochemistry, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Manfred Marschall
- Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Jens Milbradt
- Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
16
|
Wu J, Wang Y, Qiao X, Saiyin H, Zhao S, Qiao S, Wu Y. Cloning and characterization of a novel human BRMS1 transcript variant in hepatocellular carcinoma cells. Cancer Lett 2013; 337:266-75. [PMID: 23643861 DOI: 10.1016/j.canlet.2013.04.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 04/19/2013] [Accepted: 04/26/2013] [Indexed: 12/22/2022]
Abstract
Breast cancer metastasis suppressor 1 (BRMS1) is able to suppress tumor metastasis without affecting primary tumor growth in various cancers. Here, we report a novel transcript variant of human BRMS1, termed BRMS1.vh. BRMS1.vh is identical to the major BRMS1 variant (BRMS1.v1) except for missing base pairs 683-775, encoding a 215-amino acid protein lacking a functional nuclear localization sequence. Expression of BRMS1.vh in hepatocellular carcinoma (HCC) cells suppressed NF-κB signaling pathway, sensitized cells to apoptotic stimuli, leading to suppressed tumor growth. Taken together, our results suggest a potential role for BRMS1.vh in regulating cell apoptosis and tumor growth in HCC.
Collapse
Affiliation(s)
- Jun Wu
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200433, PR China
| | | | | | | | | | | | | |
Collapse
|
17
|
Gonzalo H, Brieva L, Tatzber F, Jové M, Cacabelos D, Cassanyé A, Lanau-Angulo L, Boada J, Serrano JCE, González C, Hernández L, Peralta S, Pamplona R, Portero-Otin M. Lipidome analysis in multiple sclerosis reveals protein lipoxidative damage as a potential pathogenic mechanism. J Neurochem 2012; 123:622-34. [PMID: 22924648 DOI: 10.1111/j.1471-4159.2012.07934.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 08/18/2012] [Accepted: 08/19/2012] [Indexed: 11/30/2022]
Abstract
Metabolomic and lipidomic analyses have been used for the profiling of neurodegenerative processes, both in targeted and untargeted approaches. In this work we have applied these techniques to the study of CSF samples of multiple sclerosis (MS) patients (n = 9), compared with samples of non-MS individuals (n = 9) using mass-spectrometry. We have used western-blot and analyzed cell culture to confirm pathogenic pathways suggested by mass-spectrometric measurements. The results of the untargeted approach of metabolomics and lipidomics suggest the existence of several metabolites and lipids discriminating both populations. Applying targeted lipidomic analyses focused to a pathogenic pathway in MS, oxidative stress, reveal that the lipid peroxidation marker 8-iso-prostaglandin F2α is increased in CSF from MS patients. Furthermore, as lipid peroxidation exerts its pathogenical effects through protein modification, we studied the incidence of protein lipoxidation, revealing specific increases in carboxymethylated, neuroketal and malondialdehyde-mediated protein modifications in proteins of CSF from MS patients, despite the absence of their precursors glyoxal and methylglyoxal. Finally, we report that the level of neuroketal-modified proteins correlated with a hitherto unknown increased amount of autoantibodies against lipid peroxidation-modified proteins in CSF, without compensation by signaling induced by lipid peroxidation via peroxisome proliferator-activated receptor γ (PPARγ). The results, despite the limitation of being obtained in a small population, strongly suggest that autoimmunity against in situ produced epitopes derived from lipid peroxidation can be a relevant pathogenic factor in MS.
Collapse
Affiliation(s)
- Hugo Gonzalo
- Department of Experimental Medicine, PCiTAL-Universitat de Lleida-IRBLLEIDA, Lleida, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Desmond CR, Atwal RS, Xia J, Truant R. Identification of a karyopherin β1/β2 proline-tyrosine nuclear localization signal in huntingtin protein. J Biol Chem 2012; 287:39626-33. [PMID: 23012356 DOI: 10.1074/jbc.m112.412379] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Among the known pathways of protein nuclear import, the karyopherin β2/transportin pathway is only the second to have a defined nuclear localization signal (NLS) consensus. Huntingtin, a 350-kDa protein, has defined roles in the nucleus, as well as a CRM1/exportin-dependent nuclear export signal; however, the NLS and exact pathway of import have remained elusive. Here, using a live cell assay and affinity chromatography, we show that huntingtin has a karyopherin β2-dependent proline-tyrosine (PY)-NLS in the amino terminus of the protein. This NLS comprises three consensus components: a basic charged sequence, a downstream conserved arginine, and a PY sequence. Unlike the classic PY-NLS, which has an unstructured intervening sequence between the consensus components, we show that a β sheet structured region separating the consensus elements is critical for huntingtin NLS function. The huntingtin PY-NLS is also capable of import through the importin/karyopherin β1 pathway but was not functional in all cell types tested. We propose that this huntingtin PY-NLS may comprise a new class of multiple import factor-dependent NLSs with an internal structural component that may regulate NLS activity.
Collapse
Affiliation(s)
- Carly R Desmond
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada L8N3Z5
| | | | | | | |
Collapse
|
19
|
Munsie LN, Desmond CR, Truant R. Cofilin nuclear-cytoplasmic shuttling affects cofilin-actin rod formation during stress. J Cell Sci 2012; 125:3977-88. [PMID: 22623727 DOI: 10.1242/jcs.097667] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cofilin protein is involved in regulating the actin cytoskeleton during typical steady state conditions, as well as during cell stress conditions where cofilin saturates F-actin, forming cofilin-actin rods. Cofilin can enter the nucleus through an active nuclear localization signal (NLS), accumulating in nuclear actin rods during stress. Here, we characterize the active nuclear export of cofilin through a leptomycin-B-sensitive, CRM1-dependent, nuclear export signal (NES). We also redefine the NLS of cofilin as a bipartite NLS, with an additional basic epitope required for nuclear localization. Using fluorescence lifetime imaging microscopy (FLIM) and Förster resonant energy transfer (FRET) between cofilin moieties and actin, as well as automated image analysis in live cells, we have defined subtle mutations in the cofilin NLS that allow cofilin to bind actin in vivo and affect cofilin dynamics during stress. We further define the requirement of cofilin-actin rod formation in a system of cell stress by temporal live-cell imaging. We propose that cofilin nuclear shuttling is critical for the cofilin-actin rod stress response with cofilin dynamically communicating between the nucleus and cytoplasm during cell stress.
Collapse
Affiliation(s)
- Lise Nicole Munsie
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1200 Main Street West, Hamilton, Ontario, Canada, L8N 3Z5
| | | | | |
Collapse
|
20
|
Webel R, Solbak SMØ, Held C, Milbradt J, Groß A, Eichler J, Wittenberg T, Jardin C, Sticht H, Fossen T, Marschall M. Nuclear import of isoforms of the cytomegalovirus kinase pUL97 is mediated by differential activity of NLS1 and NLS2 both acting through classical importin-α binding. J Gen Virol 2012; 93:1756-1768. [PMID: 22552943 DOI: 10.1099/vir.0.040592-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The multifunctional protein kinase pUL97 of human cytomegalovirus (HCMV) strongly determines the efficiency of virus replication. Previously, the existence of two pUL97 isoforms that arise from alternative translational initiation and show a predominant nuclear localization was described. Two bipartite nuclear localization sequences, NLS1 and NLS2, were identified in the N terminus of the large isoform, whilst the small isoform exclusively contained NLS2. The current study found the following: (i) pUL97 nuclear localization in HCMV-infected primary fibroblasts showed accumulations in virus replication centres and other nuclear sections; (ii) in a quantitative evaluation system for NLS activity, the large isoform showed higher efficiency of nuclear translocation than the small isoform; (iii) NLS1 was mapped to aa 6-35 and NLS2 to aa 190-213; (iv) using surface plasmon resonance spectroscopy, the binding of both NLS1 and NLS2 to human importin-α was demonstrated, stressing the importance of individual arginine residues in the bipartite consensus motifs; (v) nuclear magnetic resonance spectroscopy of pUL97 peptides confirmed an earlier statement about the functional requirement of NLS1 embedding into an intact α-helical structure; and (vi) a bioinformatics investigation of the solvent-accessible surface suggested a high accessibility of NLS1 and an isoform-specific, variable accessibility of NLS2 for interaction with importin-α. Thus, the nucleocytoplasmic transport mechanism of the isoforms appeared to be differentially regulated, and this may have consequences for isoform-dependent functions of pUL97 during virus replication.
Collapse
Affiliation(s)
- Rike Webel
- Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg, Schlossgarten 4, 91054 Erlangen, Germany
| | - Sara M Ø Solbak
- Centre of Pharmacy and Department of Chemistry, University of Bergen, Bergen, Norway
| | - Christian Held
- Department for Image Processing and Biomedical Engineering, Fraunhofer Institute for Integrated Circuits IIS, Erlangen, Germany
| | - Jens Milbradt
- Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg, Schlossgarten 4, 91054 Erlangen, Germany
| | - Andrea Groß
- Department Medicinal Chemistry, University of Erlangen-Nuremberg, Germany
| | - Jutta Eichler
- Department Medicinal Chemistry, University of Erlangen-Nuremberg, Germany
| | - Thomas Wittenberg
- Department for Image Processing and Biomedical Engineering, Fraunhofer Institute for Integrated Circuits IIS, Erlangen, Germany
| | - Christophe Jardin
- Division of Bioinformatics, Institute of Biochemistry, University of Erlangen-Nuremberg, Germany
| | - Heinrich Sticht
- Division of Bioinformatics, Institute of Biochemistry, University of Erlangen-Nuremberg, Germany
| | - Torgils Fossen
- Centre of Pharmacy and Department of Chemistry, University of Bergen, Bergen, Norway
| | - Manfred Marschall
- Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg, Schlossgarten 4, 91054 Erlangen, Germany
| |
Collapse
|
21
|
Roth DM, Moseley GW, Pouton CW, Jans DA. Mechanism of microtubule-facilitated "fast track" nuclear import. J Biol Chem 2011; 286:14335-51. [PMID: 21339293 DOI: 10.1074/jbc.m110.210302] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Although the microtubule (MT) cytoskeleton has been shown to facilitate nuclear import of specific cancer-regulatory proteins including p53, retinoblastoma protein, and parathyroid hormone-related protein (PTHrP), the MT association sequences (MTASs) responsible and the nature of the interplay between MT-dependent and conventional importin (IMP)-dependent nuclear translocation are unknown. Here we used site-directed mutagenesis, live cell imaging, and direct IMP and MT binding assays to map the MTAS of PTHrP for the first time, finding that it is within a short modular region (residues 82-108) that overlaps with the IMPβ1-recognized nuclear localization signal (residues 66-108) of PTHrP. Importantly, fluorescence recovery after photobleaching experiments indicated that disruption of the MT network or mutation of the MTAS of PTHrP decreases the rate of nuclear import by 2-fold. Moreover, MTAS functions depend on mutual exclusivity of binding of PTHrP to MTs and IMPβ1 such that, following MT-dependent trafficking toward the nucleus, perinuclear PTHrP can be displaced from MTs by IMPβ1 prior to import into the nucleus. This is the first molecular definition of an MTAS that facilitates protein nuclear import as well as the first delineation of the mechanism whereby cargo is transferred directly from the cytoskeleton to the cellular nuclear import apparatus. The results have broad significance with respect to fundamental processes regulating cell physiology/transformation.
Collapse
Affiliation(s)
- Daniela Martino Roth
- Nuclear Signalling Laboratory, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | | | | | | |
Collapse
|
22
|
Webel R, Milbradt J, Auerochs S, Schregel V, Held C, Nöbauer K, Razzazi-Fazeli E, Jardin C, Wittenberg T, Sticht H, Marschall M. Two isoforms of the protein kinase pUL97 of human cytomegalovirus are differentially regulated in their nuclear translocation. J Gen Virol 2010; 92:638-49. [PMID: 21084499 DOI: 10.1099/vir.0.026799-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The pUL97 protein kinase encoded by human cytomegalovirus is a multifunctional determinant of the efficiency of viral replication and phosphorylates viral as well as cellular substrate proteins. Here, we report that pUL97 is expressed in two isoforms with molecular masses of approximately 90 and 100 kDa. ORF UL97 comprises an unusual coding strategy in that five in-frame ATG start codons are contained within the N-terminal 157 aa. Site-directed mutagenesis, transient expression of point and deletion mutants and proteomic analyses accumulated evidence that the formation of the large and small isoforms result from alternative initiation of translation, with the start points being at amino acids 1 and 74, respectively. In vitro kinase assays demonstrated that catalytic activity, in terms of autophosphorylation and histone substrate phosphorylation, was indistinguishable for the two isoforms. An analysis of the intracellular distribution of pUL97 by confocal laser-scanning microscopy demonstrated that both isoforms have a pronounced nuclear localization. Surprisingly, mapping experiments performed to identify the nuclear localization signal (NLS) of pUL97 strongly suggest that the mechanism of nuclear transport is distinct for the two isoforms. While the extreme N terminus (large isoform) comprises a highly efficient, bipartite NLS (amino acids 6-35), a second sequence apparently conferring a less efficient mode of nuclear translocation was identified downstream of amino acid 74 (small and large isoforms). Taken together, the findings argue for a complex mechanism of nuclear translocation for pUL97 which might be linked with fine-regulatory differences between the two isoforms.
Collapse
Affiliation(s)
- Rike Webel
- Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Key motifs in EBV (Epstein-Barr virus)-encoded protein kinase for phosphorylation activity and nuclear localization. Biochem J 2010; 431:227-35. [PMID: 20704565 DOI: 10.1042/bj20100558] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A sole EBV (Epstein-Barr virus)-encoded protein kinase (EBV-PK) (the BGLF4 gene product) plays important roles in viral infection. Although a number of targets of this protein have been identified, the kinase itself remains largely unstudied with regard to its enzymology and structure. In the present study, site-directed mutagenesis has been employed to generate mutations targeting residues involved in nuclear localization of the EBV-PK, core residues in subdomain III of the protein kinase domain conserved in most protein kinases or residues in subdomain VIa conserved only within the HPK (herpesvirus-encoded protein kinase) group. Deletion of amino acids 389-391 resulted in exclusive cytoplasmic localization of the protein, indicating the involvement of this region in nuclear translocation of the EBV-PK. Mutations at the amino acids Glu113 (core component), Phe175, Leu178, Phe184, Leu185 and Asn186 (conserved in HPKs) resulted in loss of EBV-PK autophosphorylation, protein substrate [EBV EA-D (early antigen diffused)] phosphorylation, and ability to facilitate ganciclovir phosphorylation. These results reiterate the unique features of this group of kinases and present an opportunity for designing more specific antiviral compounds.
Collapse
|
24
|
Gal J, Zhang J, Kwinter DM, Zhai J, Jia H, Jia J, Zhu H. Nuclear localization sequence of FUS and induction of stress granules by ALS mutants. Neurobiol Aging 2010; 32:2323.e27-40. [PMID: 20674093 DOI: 10.1016/j.neurobiolaging.2010.06.010] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Accepted: 06/16/2010] [Indexed: 12/12/2022]
Abstract
Mutations in fused in sarcoma (FUS) have been reported to cause a subset of familial amyotrophic lateral sclerosis (ALS) cases. Wild-type FUS is mostly localized in the nuclei of neurons, but the ALS mutants are partly mislocalized in the cytoplasm and can form inclusions. We demonstrate that the C-terminal 32 amino acid residues of FUS constitute an effective nuclear localization sequence (NLS) as it targeted beta-galactosidase (LacZ, 116 kDa) to the nucleus. Deletion of or the ALS mutations within the NLS caused cytoplasmic mislocalization of FUS. Moreover, we identified the poly-A binding protein (PABP1), a stress granule marker, as an interacting partner of FUS. Large PABP1-positive cytoplasmic foci (i.e. stress granules) colocalized with the mutant FUS inclusions but were absent in wild-type FUS-expressing cells. Processing bodies, which are functionally related to stress granules, were adjacent to but not colocalized with the mutant FUS inclusions. Our results suggest that the ALS mutations in FUS NLS can impair FUS nuclear localization, induce cytoplasmic inclusions and stress granules, and potentially perturb RNA metabolism.
Collapse
Affiliation(s)
- Jozsef Gal
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Bański P, Mahboubi H, Kodiha M, Shrivastava S, Kanagaratham C, Stochaj U. Nucleolar targeting of the chaperone hsc70 is regulated by stress, cell signaling, and a composite targeting signal which is controlled by autoinhibition. J Biol Chem 2010; 285:21858-67. [PMID: 20457599 DOI: 10.1074/jbc.m110.117291] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hsc70s are constitutively synthesized members of the 70-kDa chaperone family; they are essential for viability and conserved among all organisms. When eukaryotic cells recover from stress, hsc70s accumulate in nucleoli by an unknown mechanism. Our studies were undertaken to characterize the signaling events and the targeting sequence required to concentrate hsc70 in the nucleoli of human cells. Here, we show that pharmacological inhibitors of phosphatidylinositol (PI) 3-kinase and MEK kinases as well as protein-tyrosine phosphatases abolished the stress-dependent nucleolar accumulation of hsc70. Furthermore, to identify the hsc70 nucleolar targeting sequence, green fluorescent protein-tagged fusion proteins with defined segments of hsc70 were generated and their subcellular distribution was analyzed in growing cells. These studies demonstrated that residues 225 to 297 serve as a heat-inducible nucleolar targeting signal. This segment directs green fluorescent protein to nucleoli in response to stress, but fails to do so under nonstress conditions. Fine mapping of the nucleolar targeting signal revealed that it has two separable functions. First, residues 225 to 262 direct reporter proteins constitutively to nucleoli, even without stress. Second, segment 263 to 287 functions as an autoinhibitory element that prevents hsc70 from concentrating in nucleoli when cells are not stressed. Taken together, PI 3-kinase and MEK kinase signaling as well as tyrosine dephosphorylation are essential for the accumulation of hsc70 in nucleoli of stressed cells. This process relies on a stress-dependent composite targeting signal that combines multiple functions.
Collapse
Affiliation(s)
- Piotr Bański
- Department of Physiology, McGill University, Montreal H3G 1Y6, Canada
| | | | | | | | | | | |
Collapse
|
26
|
Lahaye F, Lespinasse F, Staccini P, Palin L, Paquis-Flucklinger V, Santucci-Darmanin S. hMSH5 is a nucleocytoplasmic shuttling protein whose stability depends on its subcellular localization. Nucleic Acids Res 2010; 38:3655-71. [PMID: 20185565 PMCID: PMC2887964 DOI: 10.1093/nar/gkq098] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
MSH5 is a MutS-homologous protein required for meiotic DNA recombination. In addition, recent studies suggest that the human MSH5 protein (hMSH5) participates to mitotic recombination and to the cellular response to DNA damage and thus raise the possibility that a tight control of hMSH5 function(s) may be important for genomic stability. With the aim to characterize mechanisms potentially involved in the regulation of hMSH5 activity, we investigated its intracellular trafficking properties. We demonstrate that hMSH5 possesses a CRM1-dependent nuclear export signal (NES) and a nuclear localization signal that participates to its nuclear targeting. Localization analysis of various mutated forms of hMSH5 by confocal microscopy indicates that hMSH5 shuttles between the nucleus and the cytoplasm. We also provide evidence suggesting that hMSH5 stability depends on its subcellular compartmentalization, hMSH5 being much less stable in the nucleus than in the cytoplasm. Together, these data suggest that hMSH5 activity may be regulated by nucleocytoplasmic shuttling and nuclear proteasomal degradation, both of these mechanisms contributing to the control of nuclear hMSH5 content. Moreover, data herein also support that in tissues where both hMSH5 and hMSH4 proteins are expressed, hMSH5 might be retained in the nucleus through masking of its NES by binding of hMSH4.
Collapse
Affiliation(s)
- François Lahaye
- FRE 3086 Instabilité génétique: Maladies rares et cancers, Université de Nice Sophia-Antipolis, CNRS, Nice Cedex 2, France
| | | | | | | | | | | |
Collapse
|
27
|
Melzer N, Villmann C, Becker K, Harvey K, Harvey RJ, Vogel N, Kluck CJ, Kneussel M, Becker CM. Multifunctional basic motif in the glycine receptor intracellular domain induces subunit-specific sorting. J Biol Chem 2009; 285:3730-3739. [PMID: 19959465 DOI: 10.1074/jbc.m109.030460] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The strychnine-sensitive glycine receptor (GlyR) is a ligand-gated ion channel that mediates fast synaptic inhibition in the vertebrate central nervous system. As a member of the family of Cys-loop receptors, it assembles from five homologous subunits (GlyRalpha1-4 and -beta). Each subunit contains an extracellular ligand binding domain, four transmembrane domains (TM), and an intracellular domain, formed by the loop connecting TM3 and TM4 (TM3-4 loop). The TM3-4 loops of the subunits GlyRalpha1 and -alpha3 harbor a conserved basic motif, which is part of a potential nuclear localization signal. When tested for functionality by live cell imaging of green fluorescent protein and beta-galactosidase-tagged domain constructs, the TM3-4 loops of GlyRalpha1 and -alpha3, but not of GlyRalpha2 and -beta, exhibited nuclear sorting activity. Subunit specificity may be attributed to slight amino acid alterations in the basic motif. In yeast two-hybrid screening and GST pulldown assays, karyopherin alpha3 and alpha4 were found to interact with the TM3-4 loop, providing a molecular mechanism for the observed intracellular trafficking. These results indicate that the multifunctional basic motif of the TM3-4 loop is capable of mediating a karyopherin-dependent intracellular sorting of full-length GlyRs.
Collapse
Affiliation(s)
- Nima Melzer
- From the Institut für Biochemie (Emil-Fischer-Zentrum), Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Carmen Villmann
- From the Institut für Biochemie (Emil-Fischer-Zentrum), Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Kristina Becker
- From the Institut für Biochemie (Emil-Fischer-Zentrum), Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Kirsten Harvey
- the Department of Pharmacology, The School of Pharmacy, London WC1N 1AX, United Kingdom, and
| | - Robert J Harvey
- the Department of Pharmacology, The School of Pharmacy, London WC1N 1AX, United Kingdom, and
| | - Nico Vogel
- From the Institut für Biochemie (Emil-Fischer-Zentrum), Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Christoph J Kluck
- From the Institut für Biochemie (Emil-Fischer-Zentrum), Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Matthias Kneussel
- the Zentrum für Molekulare Neurobiologie Hamburg, ZMNH, Universität Hamburg, Hamburg 20251, Germany
| | - Cord-Michael Becker
- From the Institut für Biochemie (Emil-Fischer-Zentrum), Universität Erlangen-Nürnberg, Erlangen 91054, Germany.
| |
Collapse
|
28
|
Antony PMA, Mäntele S, Mollenkopf P, Boy J, Kehlenbach RH, Riess O, Schmidt T. Identification and functional dissection of localization signals within ataxin-3. Neurobiol Dis 2009; 36:280-92. [DOI: 10.1016/j.nbd.2009.07.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Revised: 07/22/2009] [Accepted: 07/27/2009] [Indexed: 10/20/2022] Open
|
29
|
Leong V, Lorenowicz J, Kozij N, Guarné A. Nuclear import of human MLH1, PMS2, and MutLalpha: redundancy is the key. Mol Carcinog 2009; 48:742-50. [PMID: 19148896 DOI: 10.1002/mc.20514] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
DNA mismatch repair maintains genomic stability by correcting errors that have escaped polymerase proofreading. Defects on mismatch repair genes lead to an increased mutation rate, microsatellite instability and predisposition to human non-polyposis colorectal cancer (HNPCC). Human MutLalpha is a heterodimer formed by the interaction of MLH1 and PMS2 that coordinates a series of key events in mismatch repair. It has been proposed that nuclear import of MutLalpha may be the first regulatory step on the activation of the mismatch repair pathway. Using confocal microscopy and mismatch repair deficient cells, we have identified the sequence determinants that drive nuclear import of human MLH1, PMS2, and MutLalpha. Transient transfection of the individual proteins reveals that MLH1 has a bipartite and PMS2 has a single monopartite nuclear localization signal. Although dimerization is not required for nuclear localization, the MutLalpha heterodimer is imported more efficiently than the MLH1 or PMS2 monomers. Interestingly, the bipartite localization signal of MLH1 can direct import of MutLalpha even when PMS2 encompasses a mutated localization signal. Hence we conclude that the presence of redundant nuclear localization signals guarantees nuclear transport of MutLalpha and, consequently, efficient mismatch repair.
Collapse
Affiliation(s)
- Vivian Leong
- Department of Biochemistry and Biomedical Sciences, McMaster University, Ontario, Canada
| | | | | | | |
Collapse
|
30
|
Rivera J, Megías D, Navas C, Bravo J. Identification of essential sequences for cellular localization in BRMS1 metastasis suppressor. PLoS One 2009; 4:e6433. [PMID: 19649328 PMCID: PMC2713406 DOI: 10.1371/journal.pone.0006433] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2009] [Accepted: 06/30/2009] [Indexed: 12/15/2022] Open
Abstract
Background Breast cancer metastasis suppressor 1 (BRMS1) reduces the number and the size of secondary tumours in a mouse model without affecting the growth of the primary foci upon its re-expression. Knockdown of BRMS1 expression associates with metastasis. The molecular details on BRMS1 mechanism of action include its ability to function as a transcriptional co-repressor and consistently BRMS1 has been described as a predominantly nuclear protein. Since cellular distribution could represent a potential mechanism of regulation, we wanted to characterize BRMS1 sequence motifs that might regulate its cellular distribution. According to its amino acids sequence, BRMS1 contain two putative nuclear localization signals, however none of them has been proved to work so far. Methodology/Principal Findings By using well known in vivo assays to detect both nuclear import and export signal, we have characterized, in the present study, one functional nuclear localisation signal as necessary and sufficient to promote nuclear transport. Additionally, the outcome of a directed yeast two-hybrid assay identify importin α6 as a specific partner of BRMS1 thus speculating that BRMS1 nuclear import could be specifically mediated by the reported nuclear transporter. Besides, the combination of a computational searching approach along the utilization of a nuclear export assay, identified a functional motif within the BRMS1 sequence responsible for its nuclear export, that resulted not affected by the highly specific CRM1 inhibitor Leptomycin-B. Interspecies heterokaryon assay demonstrate the capability of BRMS1 to shuttle between the nuclear and cytosolic compartments Conclusions/Significance Our results show for the first time that BRMS1 contains both nuclear import and export signals enabling its nucleo-cytoplasmic shuttling. These findings contributes new data for the understanding of the BRMS1 functions and allow us to speculate that this phenomenon could represent a novel mechanism for regulating the activity of BRMS1 or its associated cytosolic partners
Collapse
Affiliation(s)
- José Rivera
- Signal Transduction Group, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
- * E-mail: (JR); (JB)
| | - Diego Megías
- Confocal Microscopy Unit, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Carolina Navas
- Signal Transduction Group, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Jerónimo Bravo
- Signal Transduction Group, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
- * E-mail: (JR); (JB)
| |
Collapse
|
31
|
Analysis of influenza B Virus NS1 protein trafficking reveals a novel interaction with nuclear speckle domains. J Virol 2008; 83:701-11. [PMID: 18987144 DOI: 10.1128/jvi.01858-08] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Many proteins that function in the transcription, maturation, and export of metazoan mRNAs are concentrated in nuclear speckle domains, indicating that the compartment is important for gene expression. Here, we show that the NS1 protein of influenza B virus (B/NS1) accumulates in nuclear speckles and causes rounding and morphological changes of the domains, indicating a disturbance in their normal functions. This property was located within the N-terminal 90 amino acids of the B/NS1 protein and was shown to be independent of any other viral gene product. Within this protein domain, we identified a monopartite importin alpha binding nuclear localization signal. Reverse-genetic analysis of this motif indicated that nuclear import and speckle association of the B/NS1 protein are required for the full replication capacity of the virus. In the late phase of virus infection, the B/NS1 protein relocated to the cytoplasm, which occurred in a CRM1-independent manner. The interaction of the B/NS1 protein with nuclear speckles may reflect a recruitment function to promote viral-gene expression. To our knowledge, this is the first functional description of a speckle-associated protein that is encoded by a negative-strand RNA virus.
Collapse
|
32
|
Stoller JZ, Degenhardt KR, Huang L, Zhou DD, Lu MM, Epstein JA. Cre reporter mouse expressing a nuclear localized fusion of GFP and beta-galactosidase reveals new derivatives of Pax3-expressing precursors. Genesis 2008; 46:200-4. [PMID: 18395835 DOI: 10.1002/dvg.20384] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
A new Cre-reporter strain of mouse has been developed that expresses a fusion protein derived from the lacZ gene fused to GFP with a nuclear localization signal. This construct is expressed from the ROSA26 locus upon Cre-mediated recombination that removes a loxP-flanked PGK-neo cassette, thus allowing for detection of Cre activity in all tissues. This reporter strain, which is similar to prior R26R and R26EGFP strains, has certain advantages related to the nuclear expression and the combined expression of both beta-galactosidase and GFP activities. We show that the use of this newly developed reporter line allows for enhanced resolution, detection and co-localization. Thus, we report a previously unrecognized subset of venous endothelial cells derived from Pax3 expressing precursors.
Collapse
|
33
|
Bengtsson L, Otto H. LUMA interacts with emerin and influences its distribution at the inner nuclear membrane. J Cell Sci 2008; 121:536-48. [DOI: 10.1242/jcs.019281] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We present here a first characterization of LUMA, an unique integral inner nuclear membrane (INM) protein. LUMA is a highly conserved protein even in some bacteria and shares a PFAM domain of unknown function with orthologs from many species. Assessing LUMA topology by using protease protection of membrane-inserted LUMA and antibody epitope accessibility assays reveals that LUMA contains four transmembrane domains and a large hydrophilic domain located between membrane spans 1 and 2. The large hydrophilic domain is exposed to the perinuclear space whereas both LUMA termini reside cyto- or nucleoplasmically. Nuclear envelope targeting of LUMA mainly depends on the membrane spans. LUMA's transmembrane domains also promote homooligomerization. LUMA binds A- and B-type lamins and depends on A-type lamins for its INM localization. Furthermore, it interacts with emerin. Both downregulation of LUMA and overexpression of dominant-negative acting LUMA fragments causes redistribution of emerin. We propose that LUMA functions as a tetraspanin-like membrane organizer and has the potential to contribute to the pathomechanism of dystrophic diseases, such as Emery-Dreifuss muscular dystrophy.
Collapse
Affiliation(s)
- Luiza Bengtsson
- Freie Universität Berlin, Institut für Chemie und Biochemie, Thielallee 63, 14195 Berlin, Germany
| | - Henning Otto
- Freie Universität Berlin, Institut für Chemie und Biochemie, Thielallee 63, 14195 Berlin, Germany
| |
Collapse
|
34
|
Tkach JM, Glover JR. Nucleocytoplasmic trafficking of the molecular chaperone Hsp104 in unstressed and heat-shocked cells. Traffic 2007; 9:39-56. [PMID: 17973656 DOI: 10.1111/j.1600-0854.2007.00666.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Hsp104 is a molecular chaperone in yeast that restores solubility and activity to inactivated proteins after severe heat shock. We investigated the mechanisms that influence Hsp104 subcellular distribution in both unstressed and heat-shocked cells. In unstressed cells, Hsp104 and a green fluorescent protein-Hsp104 fusion protein were detected in both the nucleus and the cytoplasm. We demonstrate that a 17-amino-acid sequence of Hsp104 nuclear localization sequence 17 (NLS17) is sufficient to target a reporter molecule to the nucleus and is also necessary for normal Hsp104 subcellular distribution. The nuclear targeting function of NLS17 is genetically dependent on KAP95 and KAP121. In addition, wild-type Hsp104, but not an NLS17-mutated Hsp104 variant, accumulated in the nucleus of cells depleted for the general export factor Xpo1. Interestingly, severe, nonlethal heat shock enhances the nuclear levels of Hsp104 in an NLS17-independent manner. Under these conditions, we demonstrate that karyopherin-mediated nuclear transport is impaired, while the integrity of the nuclear-cytoplasmic barrier remains intact. Based on these observations, we propose that Hsp104 continues to access the nucleus during severe heat shock using a karyopherin-independent mechanism.
Collapse
Affiliation(s)
- Johnny M Tkach
- Department of Biochemistry, University of Toronto, Room 5302, Medical Sciences Building, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | | |
Collapse
|
35
|
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
|
36
|
Cattaruzzi G, Altamura S, Tessari MA, Rustighi A, Giancotti V, Pucillo C, Manfioletti G. The second AT-hook of the architectural transcription factor HMGA2 is determinant for nuclear localization and function. Nucleic Acids Res 2007; 35:1751-60. [PMID: 17324944 PMCID: PMC1874589 DOI: 10.1093/nar/gkl1106] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
High Mobility Group A (HMGA) is a family of architectural nuclear factors which play an important role in neoplastic transformation. HMGA proteins are multifunctional factors that associate both with DNA and nuclear proteins that have been involved in several nuclear processes including transcription. HMGA localization is exclusively nuclear but, to date, the mechanism of nuclear import for these proteins remains unknown. Here, we report the identification and characterization of a nuclear localization signal (NLS) for HMGA2, a member of the HMGA family. The NLS overlaps with the second of the three AT-hooks, the DNA-binding domains characteristic for this group of proteins. The functionality of this NLS was demonstrated by its ability to target a heterologous β-galactosidase/green fluorescent protein fusion protein to the nucleus. Mutations to alanine of basic residues within the second AT-hook resulted in inhibition of HMGA2 nuclear localization and impairment of its function in activating the cyclin A promoter. In addition, HMGA2 was shown to directly interact with the nuclear import receptor importin-α2 via the second AT-hook. HMGA proteins are overexpressed and rearranged in a variety of tumors; our findings can thus help elucidating their role in neoplastic transformation.
Collapse
Affiliation(s)
- Giacomo Cattaruzzi
- Dipartimento di Scienze e Tecnologie Biomediche, University of Udine, P.le Kolbe, 4 - 33100 Udine, Italy Dipartimento di Biochimica, Biofisica e Chimica delle Macromolecole, University of Trieste, via Giorgieri, 1 - 34127 Trieste, Italy and Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie, AREA Science Park, 34012 Trieste, Italy
| | - Sandro Altamura
- Dipartimento di Scienze e Tecnologie Biomediche, University of Udine, P.le Kolbe, 4 - 33100 Udine, Italy Dipartimento di Biochimica, Biofisica e Chimica delle Macromolecole, University of Trieste, via Giorgieri, 1 - 34127 Trieste, Italy and Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie, AREA Science Park, 34012 Trieste, Italy
| | - Michela A. Tessari
- Dipartimento di Scienze e Tecnologie Biomediche, University of Udine, P.le Kolbe, 4 - 33100 Udine, Italy Dipartimento di Biochimica, Biofisica e Chimica delle Macromolecole, University of Trieste, via Giorgieri, 1 - 34127 Trieste, Italy and Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie, AREA Science Park, 34012 Trieste, Italy
| | - Alessandra Rustighi
- Dipartimento di Scienze e Tecnologie Biomediche, University of Udine, P.le Kolbe, 4 - 33100 Udine, Italy Dipartimento di Biochimica, Biofisica e Chimica delle Macromolecole, University of Trieste, via Giorgieri, 1 - 34127 Trieste, Italy and Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie, AREA Science Park, 34012 Trieste, Italy
| | - Vincenzo Giancotti
- Dipartimento di Scienze e Tecnologie Biomediche, University of Udine, P.le Kolbe, 4 - 33100 Udine, Italy Dipartimento di Biochimica, Biofisica e Chimica delle Macromolecole, University of Trieste, via Giorgieri, 1 - 34127 Trieste, Italy and Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie, AREA Science Park, 34012 Trieste, Italy
| | - Carlo Pucillo
- Dipartimento di Scienze e Tecnologie Biomediche, University of Udine, P.le Kolbe, 4 - 33100 Udine, Italy Dipartimento di Biochimica, Biofisica e Chimica delle Macromolecole, University of Trieste, via Giorgieri, 1 - 34127 Trieste, Italy and Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie, AREA Science Park, 34012 Trieste, Italy
| | - Guidalberto Manfioletti
- Dipartimento di Scienze e Tecnologie Biomediche, University of Udine, P.le Kolbe, 4 - 33100 Udine, Italy Dipartimento di Biochimica, Biofisica e Chimica delle Macromolecole, University of Trieste, via Giorgieri, 1 - 34127 Trieste, Italy and Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie, AREA Science Park, 34012 Trieste, Italy
- *To whom correspondence should be addressed. +39 040 5583675+39 040 5583694
| |
Collapse
|
37
|
Stallings CL, Silverstein SJ. Posttranslational modification and cell type-specific degradation of varicella-zoster virus ORF29p. J Virol 2006; 80:10836-46. [PMID: 16956951 PMCID: PMC1641786 DOI: 10.1128/jvi.00966-06] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The ORF29 gene of varicella-zoster virus encodes a single-stranded DNA binding protein that is predominantly nuclear during lytic infection but appears to be restricted to the cytoplasm of latently infected neurons. Following reactivation, ORF29p accumulates in the nuclei of neurons, suggesting that its confinement to the cytosol may be critical for maintaining quiescence. When autonomously expressed, ORF29p accumulates in the nuclei of fibroblasts and the cytoplasm of cells (guinea pig enteric neurons) and cell lines (U373MG) of neuronal origin. Inhibition of the 26S proteasome redirects the accumulation of ORF29p to the nucleus in cells of neuronal origin. Here, we show that ORF29p is ubiquitinated and sumoylated in 293T cells and subsequently degraded from the N terminus. Ubiquitinated ORF29p accumulates in both the nuclei and the cytoplasm of fibroblasts, but degradation products are seen primarily in the cytoplasm. Modification and degradation of ORF29p occurs in 293T, U373MG, and MeWo cells. Therefore, these processes are ubiquitous; however, the robustness of the degradation process is cell type specific. The proteasome-mediated mechanism of nuclear exclusion in U373MG cells is an active process that is not specific for the endogenous ORF29p nuclear localization signal but can be saturated by protein stabilization or overexpression, which leads to nuclear accumulation of ORF29p. The evidence for ORF29p ubiquitination and previous data regarding the effect of proteasome inhibitors on the abundance and distribution of ORF29p implicate the 26S proteasome in influencing the protein's cell type-specific localization.
Collapse
|
38
|
Alvisi G, Ripalti A, Ngankeu A, Giannandrea M, Caraffi SG, Dias MM, Jans DA. Human cytomegalovirus DNA polymerase catalytic subunit pUL54 possesses independently acting nuclear localization and ppUL44 binding motifs. Traffic 2006; 7:1322-32. [PMID: 16911590 DOI: 10.1111/j.1600-0854.2006.00477.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The catalytic subunit of human cytomegalovirus (HCMV) DNA polymerase pUL54 is a 1242-amino-acid protein, whose function, stimulated by the processivity factor, phosphoprotein UL44 (ppUL44), is essential for viral replication. The C-terminal residues (amino acids 1220-1242) of pUL54 have been reported to be sufficient for ppUL44 binding in vitro. Although believed to be important for functioning in the nuclei of infected cells, no data are available on either the interaction of pUL54 with ppUL44 in living mammalian cells or the mechanism of pUL54 nuclear transport and its relationship with that of ppUL44. The present study examines for the first time the nuclear import pathway of pUL54 and its interaction with ppUL44 using dual color, quantitative confocal laser scanning microscopy on live transfected cells and quantitative gel mobility shift assays. We showed that of two nuclear localization signals (NLSs) located at amino acids 1153-1159 (NLSA) and 1222-1227 (NLSB), NLSA is sufficient to confer nuclear localization on green fluorescent protein (GFP) by mediating interaction with importin alpha/beta. We also showed that pUL54 residues 1213-1242 are sufficient to confer ppUL44 binding abilities on GFP and that pUL54 and ppUL44 can be transported to the nucleus as a complex. Our work thus identified distinct sites within the HCMV DNA polymerase, which represent potential therapeutic targets and establishes the molecular basis of UL54 nuclear import.
Collapse
Affiliation(s)
- Gualtiero Alvisi
- Dipartimento di Medicina Clinica Specialistica e Sperimentale, Sezione di Microbiologia, Università degli Studi di Bologna, via Massarenti 9, 40138 Bologna, Italy.
| | | | | | | | | | | | | |
Collapse
|
39
|
Alvisi G, Jans DA, Guo J, Pinna LA, Ripalti A. A protein kinase CK2 site flanking the nuclear targeting signal enhances nuclear transport of human cytomegalovirus ppUL44. Traffic 2006; 6:1002-13. [PMID: 16190981 DOI: 10.1111/j.1600-0854.2005.00331.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The processivity factor of the human cytomegalovirus (HCMV) DNA polymerase phosphoprotein ppUL44 plays an essential role in viral replication, showing nuclear localization in infected cells. The present study examines ppUL44's nuclear import pathway for the first time, ectopic expression of ppUL44 revealing a strong nuclear localization in transfected COS-7 and other cell types, implying that no other HCMV proteins are required for nuclear transportation and retention. We show that of the two potential nuclear localization signals (NLSs) located at amino acids 162-168 (NLS1) and 425-431 (NLS2), NLS2 is necessary and sufficient to confer nuclear localization. Moreover, using enzyme-linked immunosorbent assays and gel mobility shift assays, we show that NLS2 is recognized with high affinity by the importin (IMP) alpha/beta heterodimer. Using gel mobility shift and transient transfection assays, we find that flanking sequences containing a cluster of potential phosphorylation sites, including a consensus site for protein kinase CK2 (CK2) at Ser413 upstream of the NLS, increase NLS2-dependent IMP binding and nuclear localization, suggesting a role for these sites in enhancing UL44 nuclear transport. Results from site-directed mutagenic analysis and live-cell imaging of green fluorescent protein (GFP)-UL44 fusion protein-expressing cells treated with the CK2-specific inhibitor 4,5,6,7-tetrabromobenzotriazole are consistent with phosphorylation of Ser413 enhancing ppUL44 nuclear transport.
Collapse
Affiliation(s)
- Gualtiero Alvisi
- Dipartimento di Medicina Clinica Specialistica e Sperimentale, Sezione di Microbiologia, Università degli Studi di Bologna, Bologna, Italy
| | | | | | | | | |
Collapse
|
40
|
Kelly KF, Otchere AA, Graham M, Daniel JM. Nuclear import of the BTB/POZ transcriptional regulator Kaiso. J Cell Sci 2005; 117:6143-52. [PMID: 15564377 DOI: 10.1242/jcs.01541] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Kaiso is a BTB/POZ transcription factor that functions in vitro as a transcriptional repressor of the matrix metalloproteinase gene matrilysin and the non-canonical Wnt signaling gene Wnt-11, and as an activator of the acetylcholine-receptor-clustering gene rapsyn. Similar to other BTB/POZ proteins (e.g. Bcl-6, PLZF, HIC-1), endogenous Kaiso localizes predominantly to the nuclei of mammalian cells. To date, however, the mechanism of nuclear import for most POZ transcription factors, including Kaiso, remain unknown. Here, we report the identification and characterization of a highly basic nuclear localization signal (NLS) in Kaiso. The functionality of this NLS was verified by its ability to target a heterologous beta-galactosidase/green-fluorescent-protein fusion protein to nuclei. The mutation of one positively charged lysine to alanine in the NLS of full-length Kaiso significantly inhibited its nuclear localization in various cell types. In addition, wild-type Kaiso, but not NLS-defective Kaiso, interacted directly with the nuclear import receptor Importin-alpha2 both in vitro and in vivo. Finally, minimal promoter assays using a sequence-specific Kaiso-binding-site fusion with luciferase as reporter demonstrated that the identified NLS was crucial for Kaiso-mediated transcriptional repression. The identification of a Kaiso NLS thus clarifies the mechanism by which Kaiso translocates to the nucleus to regulate transcription of genes with diverse roles in cell growth and development.
Collapse
Affiliation(s)
- Kevin F Kelly
- Department of Biology, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | | | | | | |
Collapse
|
41
|
Zhu J, Qiu Z, Wiese C, Ishii Y, Friedrichsen J, Rajashekara G, Splitter GA. Nuclear and Mitochondrial Localization Signals Overlap within Bovine Herpesvirus 1 Tegument Protein VP22. J Biol Chem 2005; 280:16038-44. [PMID: 15705574 DOI: 10.1074/jbc.m500054200] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
VP22, a tegument protein of bovine herpesvirus 1, accumulates in the nucleus of infected and transiently transfected cells. Previous studies indicated a possible regulatory function of VP22 within nuclei, but how VP22 enters nuclei is unknown. Despite the abundance of basic residues within this protein, no classic nuclear localization signal (NLS) motif has been identified. To identify the signal directing nuclear accumulation, a series of truncations, internal deletions, and point mutations were constructed. Fluorescence microscopy of cells transfected with VP22 constructs indicated that a sequence of 103 residues is necessary and sufficient for nuclear localization. This NLS sequence is conformation-sensitive in contrast to a classical sequential NLS. Energy depletion assays and co-immunoprecipitation suggested that this NLS sequence also binds histone H4, resulting in nuclear retention of VP22. In addition, a mitochondrial targeting sequence was identified at the C-terminal 49 amino acids, which overlapped the sequence required for nuclear targeting. Our findings demonstrate the diversity of VP22 protein to localize within the cell and provide the opportunity for VP22 to direct cargo specifically to different subcellular compartments.
Collapse
Affiliation(s)
- Jun Zhu
- Department of Animal Health, University of Wisconsin, Madison, Wisconsin 53706, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Stoller JZ, Epstein JA. Identification of a novel nuclear localization signal in Tbx1 that is deleted in DiGeorge syndrome patients harboring the 1223delC mutation. Hum Mol Genet 2005; 14:885-92. [PMID: 15703190 DOI: 10.1093/hmg/ddi081] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
DiGeorge syndrome (DGS) is the most common human chromosomal deletion syndrome and is frequently associated with deletions on chromosome 22q11. Approximately 17% of patients with the phenotypic features of this syndrome have no detectable genomic deletion. Animal studies using mouse models have implicated Tbx1 as a critical gene within the commonly deleted region, and several mutations in TBX1 have been identified recently in non-deleted patients, including missense and frameshift mutations. The mechanisms by which these mutations cause disease have remained unclear. We have identified a previously unrecognized and novel nuclear localization signal (NLS) at the C-terminus of Tbx1 that is deleted by the 1223delC mutation, thus explaining the mechanism of disease in these patients. This NLS is conserved across species, among a subfamily of T-box proteins including Brachyury and Tbx10, and among additional nuclear proteins. By providing functional data to indicate loss-of-function produced by the 1223delC TBX1 mutation, our results provide strong support for the conclusion that TBX1 mutations can cause DGS in humans.
Collapse
Affiliation(s)
- Jason Z Stoller
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | |
Collapse
|
43
|
Kodiha M, Chu A, Matusiewicz N, Stochaj U. Multiple mechanisms promote the inhibition of classical nuclear import upon exposure to severe oxidative stress. Cell Death Differ 2005; 11:862-74. [PMID: 15088071 DOI: 10.1038/sj.cdd.4401432] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
In growing HeLa cells, severe stress elicited by the oxidant hydrogen peroxide inhibits classical nuclear import. Oxidant treatment collapses the nucleocytoplasmic Ran concentration gradient, thereby elevating cytoplasmic GTPase levels. The Ran gradient dissipates in response to a stress-induced depletion of RanGTP and a decreased efficiency of Ran nuclear import. In addition, oxidative stress induces a relocation of the nucleoporin Nup153 as well as the nuclear carrier importin-beta, and docking of the importin-alpha/beta/cargo complex at the nuclear envelope is reduced. Moreover, Ran, importin-beta and Nup153 undergo proteolysis upon oxidative stress. Caspases and the proteasome degrade Ran and importin-beta; however, ubiquitination of these transport factors is not observed. Inhibition of caspases in stressed cells alleviates the mislocalization of importin-beta, but does not restore the Ran concentration gradient or classical import. In summary, inhibition of classical nuclear import by hydrogen peroxide is caused by a combination of multiple mechanisms that target different components of the transport apparatus.
Collapse
Affiliation(s)
- M Kodiha
- Department of Physiology, McGill University, 3655 Promenade Sir William Osler, Montreal, PQ, H3G 1Y6, Canada
| | | | | | | |
Collapse
|
44
|
MacDonald JIS, Kubu CJ, Meakin SO. Nesca, a novel adapter, translocates to the nuclear envelope and regulates neurotrophin-induced neurite outgrowth. ACTA ACUST UNITED AC 2004; 164:851-62. [PMID: 15024033 PMCID: PMC2172297 DOI: 10.1083/jcb.200309081] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
We provide the first characterization of a novel signaling adapter, Nesca, in neurotrophic signal transduction. Nesca contains a RUN domain, a WW domain, a leucine zipper, a carboxyl-terminal SH3 domain, and several proline-rich regions. Nesca is highly expressed in the brain, is serine phosphorylated, and mobilizes from the cytoplasm to the nuclear membrane in response to neurotrophin, but not epidermal growth factor, stimulation in a MEK-dependent process. Overexpression studies in PC12 cells indicate that Nesca facilitates neurotrophin-dependent neurite outgrowth at nonsaturating doses of nerve growth factor (NGF). Similarly, short interfering RNA studies significantly reduce NGF-dependent neuritogenesis in PC12 cells. Mutational analyses demonstrate that the RUN domain is an important structural determinant for the nuclear translocation of Nesca and that the nuclear redistribution of Nesca is essential to its neurite outgrowth-promoting properties. Collectively, these works provide the first functional characterization of Nesca in the context of neurotrophin signaling and suggest that Nesca serves a novel, nuclear-dependent role in neurotrophin-dependent neurite outgrowth.
Collapse
Affiliation(s)
- James I S MacDonald
- Laboratory of Neural Signalling, Cell Biology Group, The Robarts Research Institute, 100 Perth Dr., London, Ontario, N6A 5K8 Canada
| | | | | |
Collapse
|
45
|
Kelly KF, Spring CM, Otchere AA, Daniel JM. NLS-dependent nuclear localization of p120ctnis necessary to relieve Kaiso-mediated transcriptional repression. J Cell Sci 2004; 117:2675-86. [PMID: 15138284 DOI: 10.1242/jcs.01101] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The Armadillo catenin p120ctn regulates cadherin adhesive strength at the plasma membrane and interacts with the novel BTB/POZ transcriptional repressor Kaiso in the nucleus. The dual localization of p120ctn at cell-cell junctions and in the nucleus suggests that its nucleocytoplasmic trafficking is tightly regulated. Here we report on the identification of a specific and highly basic nuclear localization signal (NLS) in p120ctn. The functionality of the NLS was validated by its ability to direct the nuclear localization of a heterologous β-galactosidase-GFP fusion protein. Mutating two key positively charged lysines to neutral alanines in the NLS of full-length p120ctn inhibited both p120ctn nuclear localization as well as the characteristic p120ctn-induced branching phenotype that correlates with increased cell migration. However, while these findings and others suggested that nuclear localization of p120ctn was crucial for the p120ctn-induced branching phenotype, we found that forced nuclear localization of both wild-type and NLS-mutated p120ctn did not induce branching. Recently, we also found that one role of p120ctn was to regulate Kaiso-mediated transcriptional repression. However, it remained unclear whether p120ctn sequestered Kaiso in the cytosol or directly inhibited Kaiso transcriptional activity in the nucleus. Using minimal promoter assays, we show here that the regulatory effect of p120ctn on Kaiso transcriptional activity requires the nuclear translocation of p120ctn. Therefore, an intact NLS in p120ctn is requisite for its first identified regulatory role of the transcriptional repressor Kaiso.
Collapse
Affiliation(s)
- Kevin F Kelly
- Department of Biology, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | | | | | | |
Collapse
|
46
|
Lemrow SM, Anderson KA, Joseph JD, Ribar TJ, Noeldner PK, Means AR. Catalytic activity is required for calcium/calmodulin-dependent protein kinase IV to enter the nucleus. J Biol Chem 2003; 279:11664-71. [PMID: 14701808 DOI: 10.1074/jbc.m312613200] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Calcium/calmodulin-dependent protein kinase IV (CaMKIV) is a nuclear protein kinase that responds to acute rises in intracellular calcium by phosphorylating and activating proteins involved in transcription. Consistent with these roles, CaMKIV is found predominantly in the nucleus of cells in which it is expressed. Here we evaluate nuclear entry of CaMKIV and demonstrate that the protein kinase homology domain is both necessary and sufficient for nuclear localization. Unexpectedly, although catalytic activity is required for nuclear translocation, it is not required for CaMKIV to interact with the nuclear adaptor protein, importin-alpha. Because the catalytically inactive molecules remain in the cytoplasm, these data suggest that this interaction is not sufficient for nuclear entry. We evaluated a role for other proteins known to interact with CaMKIV in regulation of its nuclear entry. Although our data do not support a role for calmodulin or protein phosphatase 2A, the catalytically inactive CaMKIV proteins interact more avidly with CaM-dependent protein kinase kinase (CaMKK), which is restricted to the cytoplasm. We find that the catalytically inactive proteins do not inhibit nuclear entry of wild-type CaMKIV but do inhibit the ability of the wild-type protein kinase to stimulate cyclic AMP response element-binding protein-mediated transcription. Because activation loop phosphorylation is required for the transcriptional roles of CaMKIV, these data suggest that CaMKK phosphorylation of CaMKIV may occur in the cytoplasm. We propose that sequestration of CaMKK may be the molecular mechanism by which catalytically inactive mutants of CaMKIV exert their "dominant-negative" functions within the cell.
Collapse
Affiliation(s)
- Shannon M Lemrow
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | |
Collapse
|
47
|
Nielsen J, Adolph SK, Rajpert-De Meyts E, Lykke-Andersen J, Koch G, Christiansen J, Nielsen FC. Nuclear transit of human zipcode-binding protein IMP1. Biochem J 2003; 376:383-91. [PMID: 12921532 PMCID: PMC1223771 DOI: 10.1042/bj20030943] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2003] [Revised: 08/04/2003] [Accepted: 08/18/2003] [Indexed: 11/17/2022]
Abstract
The human IMPs (insulin-like growth factor II mRNA-binding proteins) belong to a vertebrate zipcode-binding protein family consisting of two RNA recognition motifs and four K homology domains and have been implicated in cytoplasmic mRNA localization, turnover and translational control. In the present study, we show that IMP1 is capable of translocating into nuclei of NIH 3T3 fibroblasts and its immunoreactivity is present in the nuclei of human spermatogenic cells. IMP1 does not contain a simple import signal, but nuclear entry was facilitated by disruption of RNA binding and cytoplasmic granule formation. IMP1 contains two NESs (nuclear export signals) within the RNA-binding K homology domains 2 and 4. The former is a leucine-rich leptomycin B-sensitive NES, whereas the latter is a leptomycin B-insensitive NES. Taken together, these results indicate that IMP1 may attach to its target mRNAs in the nucleus and thereby define the cytoplasmic fate of the transcripts.
Collapse
Affiliation(s)
- Jacob Nielsen
- Institute of Molecular Biology, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | |
Collapse
|
48
|
Masson M, Hindelang C, Sibler AP, Schwalbach G, Travé G, Weiss E. Preferential nuclear localization of the human papillomavirus type 16 E6 oncoprotein in cervical carcinoma cells. J Gen Virol 2003; 84:2099-2104. [PMID: 12867640 DOI: 10.1099/vir.0.18961-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The E6 protein of the high-risk human papillomavirus type 16 (HPV-16) is involved in the tumorigenesis of human cervical cells by targeting numerous cellular proteins. We characterized new anti-E6 monoclonal antibodies and used them for precise localization of the E6 oncoprotein within carcinoma cells. Overexpressed E6 protein was predominantly detected in the nucleus of transiently transfected HaCaT cells. While mostly localized at the periphery of condensed chromatin, E6 was also associated with nuclear ribonucleoproteic ultrastructures and with some ribosomal areas in the cytoplasm of SiHa and CaSki cells. The chimeric beta-galactosidase-E6 protein expressed in transfected HeLa cells was essentially localized in the nuclear compartment. Together, these data indicate that the E6 sequence of HPV-16 may encode a nuclear localization signal. The preferential nuclear distribution of this viral oncoprotein in HPV-transformed cells correlates with its activities at the transcriptional level.
Collapse
Affiliation(s)
- Murielle Masson
- UMR 7100, Ecole Supérieure de Biotechnologie de Strasbourg, Boulevard Sébastien Brant, BP 10413, 67412 Illkirch cedex, France
| | - Colette Hindelang
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Service de Microscopie Electronique, 1 rue Laurent Fries, 67400 Illkirch cedex, France
| | - Annie-Paule Sibler
- UMR 7100, Ecole Supérieure de Biotechnologie de Strasbourg, Boulevard Sébastien Brant, BP 10413, 67412 Illkirch cedex, France
| | - Georges Schwalbach
- UMR 7100, Ecole Supérieure de Biotechnologie de Strasbourg, Boulevard Sébastien Brant, BP 10413, 67412 Illkirch cedex, France
| | - Gilles Travé
- UMR 7100, Ecole Supérieure de Biotechnologie de Strasbourg, Boulevard Sébastien Brant, BP 10413, 67412 Illkirch cedex, France
| | - Etienne Weiss
- UMR 7100, Ecole Supérieure de Biotechnologie de Strasbourg, Boulevard Sébastien Brant, BP 10413, 67412 Illkirch cedex, France
| |
Collapse
|
49
|
Sibler AP, Nordhammer A, Masson M, Martineau P, Travé G, Weiss E. Nucleocytoplasmic shuttling of antigen in mammalian cells conferred by a soluble versus insoluble single-chain antibody fragment equipped with import/export signals. Exp Cell Res 2003; 286:276-87. [PMID: 12749856 DOI: 10.1016/s0014-4827(03)00093-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The ectopic expression of antibody fragments within mammalian cells is a challenging approach for interfering with or even blocking the biological function of the intracellular target. For this purpose, single-chain Fv (scFv) fragments are generally preferred. Here, by transfecting several mammalian cell lines, we compared the intracellular behavior of two scFvs (13R4 and 1F4) that strongly differ in their requirement of disulphide bonding for the formation of active molecules in bacteria. The scFv 13R4, which is correctly folded in the bacterial cytoplasm, was solubly expressed in all cell lines tested and was distributed in their cytoplasm and nucleus, as well. In addition, by appending to the 13R4 molecules the SV40 T-antigen nuclear localisation signal (NLS) tag, cytoplasmic-coexpressed antigen was efficiently retargeted to the nucleus. Compared to the scFv 13R4, the scFv 1F4, which needs to be secreted in bacteria for activity, accumulated, even with the NLS tag, as insoluble aggregates within the cytoplasm of the transfected cells, thereby severely disturbing fundamental functions of cell physiology. Furthermore, by replacing the NLS tag with a leucine-rich nuclear export signal (NES), the scFv 13R4 was exclusively located in the cytoplasm, whereas the similarly modified scFv 1F4 still promoted cell death. Coexpression of NES-tagged 13R4 fragments with nuclear antigen promoted its efficient retargeting to the cytoplasm. This dominant effect of the NES tag was also observed after exchange of the nuclear signals between the scFv 13R4 and its antigen. Taken together, the results indicate that scFvs that are active in the cytoplasm of bacteria may behave similarly in mammalian cells and that the requirement of their conserved disulphide bridges for activity is a limiting factor for mediating the nuclear import/export of target in a mammalian cell context. The described shuttling effect of antigen conferred by a soluble scFv may represent the basis of a reliable in vivo assay of effective protein- protein interactions.
Collapse
Affiliation(s)
- Annie-Paule Sibler
- Biotechnologie des Interactions Macromoléculaires, UMR 7100, Ecole Supérieure de Biotechnologie de Strasbourg, boulevard Sébastien Brant, 67400 Illkirch, France
| | | | | | | | | | | |
Collapse
|
50
|
Lischka P, Sorg G, Kann M, Winkler M, Stamminger T. A nonconventional nuclear localization signal within the UL84 protein of human cytomegalovirus mediates nuclear import via the importin alpha/beta pathway. J Virol 2003; 77:3734-48. [PMID: 12610148 PMCID: PMC149505 DOI: 10.1128/jvi.77.6.3734-3748.2003] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The open reading frame UL84 of human cytomegalovirus encodes a multifunctional regulatory protein which is required for viral DNA replication and binds with high affinity to the immediate-early transactivator IE2-p86. Although the exact role of pUL84 in DNA replication is unknown, the nuclear localization of this protein is a prerequisite for this function. To investigate whether the activities of pUL84 are modulated by cellular proteins we used the Saccharomyces cerevisiae two-hybrid system to screen a cDNA-library for interacting proteins. Strong interactions were found between pUL84 and four members of the importin alpha protein family. These interactions could be confirmed in vitro by pull down experiments and in vivo by coimmunoprecipitation analysis from transfected cells. Using in vitro transport assays we showed that the pUL84 nuclear import required importin alpha, importin beta, and Ran, thus following the classical importin-mediated import pathway. Deletion mutagenesis of pUL84 revealed a domain of 282 amino acids which is required for binding to the importin alpha proteins. Its function as a nuclear localization signal (NLS) was confirmed by fusion to heterologous proteins. Although containing a cluster of basic amino acids similar to classical NLSs, this cluster did not contain the NLS activity. Thus, a complex structure appears to be essential for importin alpha binding and import activity.
Collapse
Affiliation(s)
- Peter Lischka
- Institut für Klinische und Molekulare Virologie der Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | | | | | | | | |
Collapse
|