55 results on '"Kylie M. Wagstaff"'
Search Results
2. Conservation of Importin α Function in Apicomplexans: Ivermectin and GW5074 Target
- Author
-
Sujata B, Walunj, Chunxiao, Wang, Kylie M, Wagstaff, Swati, Patankar, and David A, Jans
- Subjects
alpha Karyopherins ,Mammals ,Mice ,Ivermectin ,Plasmodium falciparum ,Nuclear Localization Signals ,Animals ,Parasites ,Protein Binding - Abstract
Signal-dependent transport into and out of the nucleus mediated by members of the importin (IMP) superfamily of nuclear transporters is critical to the eukaryotic function and a point of therapeutic intervention with the potential to limit disease progression and pathogenic outcomes. Although the apicomplexan parasites
- Published
- 2022
3. The nuclear transporter importin 13 is critical for cell survival during embryonic stem cell differentiation
- Author
-
Shadma Fatima, Kylie M. Wagstaff, Julia C. Young, Sue Mei Lim, David A. Jans, and Jose M. Polo
- Subjects
0301 basic medicine ,Brachyury ,Mesoderm ,Cell Survival ,Biophysics ,Apoptosis ,Ectoderm ,Importin ,Karyopherins ,Biology ,Biochemistry ,Gene Knockout Techniques ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Proliferation Marker ,Molecular Biology ,Embryoid Bodies ,Embryonic Stem Cells ,Cell Proliferation ,Cell Differentiation ,Cell Biology ,Embryonic stem cell ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,embryonic structures ,Nuclear transport ,Endoderm ,Octamer Transcription Factor-3 - Abstract
Nuclear transporter Importin (Imp, Ipo) 13 is known to transport various mammalian cargoes into/out of the nucleus, but its role in directing cell-fate is unclear. Here we examine the role of Imp13 in the maintenance of pluripotency and differentiation of embryonic stem cells (ESCs) for the first time, using an embryonic body (EB)-based model. When induced to differentiate, Ipo13-/- ESCs displayed slow proliferation, reduced EB size, and lower expression of the proliferation marker KI67, concomitant with an increase in the number of TUNEL+ nuclei compared to wildtype ESCs. At days 5 and 10 of differentiation, Ipo13-/- EBs also showed enhanced loss of the pluripotency transcript OCT3/4, and barely detectable clusters of OCT3/4 positive cells. Day 5 Ipo13-/- EBs further exhibited reduced levels of the mesodermal markers Brachyury and Mixl1, correlating with reduced numbers of haemoglobinised cells generated. Our findings suggest that Imp13 is critical to ESC survival as well as early post-gastrulation differentiation.
- Published
- 2021
- Full Text
- View/download PDF
4. The broad spectrum host-directed agent ivermectin as an antiviral for SARS-CoV-2 ?
- Author
-
Kylie M. Wagstaff and David A. Jans
- Subjects
alpha Karyopherins ,0301 basic medicine ,viruses ,Biophysics ,Pseudorabies ,Dengue virus ,medicine.disease_cause ,Antiviral Agents ,Biochemistry ,Article ,Virus ,Zika virus ,Dengue fever ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Chikungunya ,Antiviral ,Molecular Biology ,Ivermectin ,Antiparasitic Agents ,biology ,SARS-CoV-2 ,COVID-19 ,virus diseases ,Cell Biology ,biology.organism_classification ,medicine.disease ,Virology ,COVID-19 Drug Treatment ,Integrase ,030104 developmental biology ,030220 oncology & carcinogenesis ,Venezuelan equine encephalitis virus ,biology.protein - Abstract
FDA approved for parasitic indications, the small molecule ivermectin has been the focus of growing attention in the last 8 years due to its potential as an antiviral. We first identified ivermectin in a high throughput compound library screen as an agent potently able to inhibit recognition of the nuclear localizing Human Immunodeficiency Virus-1 (HIV-1) integrase protein by the host importin (IMP) α/β1 heterodimer, and recently demonstrated its ability to bind directly to IMPα to cause conformational changes that prevent its function in nuclear import of key viral as well as host proteins. Cell culture experiments have shown robust antiviral action towards a whole range of viruses, including HIV-1, dengue, Zika and West Nile Virus, Venezuelan equine encephalitis virus, Chikungunya, pseudorabies virus, adenovirus, and SARS-CoV-2 (COVID-19). Close to 70 clinical trials are currently in progress worldwide for SARS-CoV-2. Although few of these studies have been completed, the results that are available, as well as those from observational/retrospective studies, indicate clinical benefit. Here we discuss the case for ivermectin as a host-directed broad-spectrum antiviral agent, including for SARS-CoV-2.
- Published
- 2021
- Full Text
- View/download PDF
5. High-Throughput Screening to Identify Inhibitors of
- Author
-
Sujata B, Walunj, Manisha M, Dias, Chhaminder, Kaur, Kylie M, Wagstaff, Vishakha, Dey, Caroline, Hick, Swati, Patankar, and David A, Jans
- Subjects
alpha Karyopherins ,Nuclear Localization Signals ,Plasmodium falciparum ,High-Throughput Screening Assays ,Protein Binding - Abstract
The global burden of malaria and toxoplasmosis has been limited by the use of efficacious anti-parasitic agents, however, emerging resistance in
- Published
- 2022
6. Selective Targeting of Protein Kinase C (PKC)-θ Nuclear Translocation Reduces Mesenchymal Gene Signatures and Reinvigorates Dysfunctional CD8
- Author
-
Jenny, Dunn, Robert D, McCuaig, Abel H Y, Tan, Wen Juan, Tu, Fan, Wu, Kylie M, Wagstaff, Anjum, Zafar, Sayed, Ali, Himanshu, Diwakar, Jane E, Dahlstrom, Elaine G, Bean, Jade K, Forwood, Sofiya, Tsimbalyuk, Emily M, Cross, Kristine, Hardy, Amanda L, Bain, Elizabeth, Ahern, Riccardo, Dolcetti, Roberta, Mazzieri, Desmond, Yip, Melissa, Eastgate, Laeeq, Malik, Peter, Milburn, David A, Jans, and Sudha, Rao
- Abstract
Protein kinase C (PKC)-θ is a serine/threonine kinase with both cytoplasmic and nuclear functions. Nuclear chromatin-associated PKC-θ (nPKC-θ) is increasingly recognized to be pathogenic in cancer, whereas its cytoplasmic signaling is restricted to normal T-cell function. Here we show that nPKC-θ is enriched in circulating tumor cells (CTCs) in patients with triple-negative breast cancer (TNBC) brain metastases and immunotherapy-resistant metastatic melanoma and is associated with poor survival in immunotherapy-resistant disease. To target nPKC-θ, we designed a novel PKC-θ peptide inhibitor (nPKC-θi2) that selectively inhibits nPKC-θ nuclear translocation but not PKC-θ signaling in healthy T cells. Targeting nPKC-θ reduced mesenchymal cancer stem cell signatures in immunotherapy-resistant CTCs and TNBC xenografts. PKC-θ was also enriched in the nuclei of CD8
- Published
- 2022
7. Transcriptomic profile dataset of embryonic stem cells (Wild-type and IPO13-Knock Out) with and without oxidative stress
- Author
-
Katarzyna A. Gajewska, Mirana Ramialison, Kylie M. Wagstaff, and David A. Jans
- Subjects
Multidisciplinary - Abstract
The transcriptional response to cellular stress relies upon trafficking of regulators of transcription between the nuclear and cytoplasmic compartments, which occurs through action of members of the importin (IPO) superfamily. As a result of stresses such as oxidative or osmotic stress, one consequence is that importins become mislocalised, leading to inhibition of conventional nuclear transport. Here, we examine IPO13, which has a number of nonconventional characteristics, in the context of cell stress. We used Next Generation RNA Sequencing using the Illumina platform to compare the transcriptomes of Wild-type (WT) and IPO13-Knockout (KO) mouse embryonic stem cells in the absence and presence of oxidative stress. Differences in the mRNA expression profiles were observed between the cell lines in the absence and in the presence of stress. This data will be a key resource to enable characterization of the contribution of nuclear transporter IPO13 to cellular transcription in the absence and presence of oxidative stress, as well as more broadly, in the study of stem cell biology and effect of stress on embryonic stem cell transcription.
- Published
- 2022
8. Inhibitors of nuclear transport
- Author
-
David A. Jans, Kylie M. Wagstaff, and Alexander J. Martin
- Subjects
Active Transport, Cell Nucleus ,Receptors, Cytoplasmic and Nuclear ,Importin ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Exportin-1 ,medicine ,Animals ,Humans ,Receptor ,Nuclear export signal ,030304 developmental biology ,Cell Nucleus ,0303 health sciences ,Cancer ,Transporter ,Cell Biology ,medicine.disease ,Transport protein ,Protein Transport ,Virus Diseases ,Host-Pathogen Interactions ,Cancer research ,Nuclear transport ,030217 neurology & neurosurgery - Abstract
Central to eukaryotic cell function, transport into and out of the nucleus is largely mediated by members of the Importin (IMP) superfamily of transporters of α- and β-types. The first inhibitor of nuclear transport, leptomycin B (LMB), was shown to be a specific inhibitor of the IMPβ homologue Exportin 1 (EXP1) almost 20 years ago, but it has only been in the last five or so years that new inhibitors of nuclear export as well as import have been identified and characterised. Of utility in biological research, these inhibitors include those that target-specific EXPs/IMPs, with accompanying toxicity profiles, as well as agents that specifically target particular nuclear import cargoes. Both types of inhibitors have begun to be tested in preclinical/clinical studies, with particular focus on limiting various types of cancer or treating viral infection, and the most advanced agent targeting EXP1 (Selinexor) has progressed successfully through >40 clinical trials for a range of high-grade cancers and is approaching FDA approval for a number of indications. Selectively inhibiting the nucleocytoplasmic trafficking of specific proteins of interest remains a challenge, but progress in the area of the host-pathogen interface holds promise for the future.
- Published
- 2019
- Full Text
- View/download PDF
9. Internalized Functional DNA Aptamers as Alternative Cancer Therapies
- Author
-
Kylie M. Wagstaff and Morgan L Marshall
- Subjects
0301 basic medicine ,tumor-specific ,precision medicine ,Aptamer ,media_common.quotation_subject ,DNA-aptamer ,Review ,Computational biology ,Disease ,DNA Aptamers ,Biology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,nanoparticle-free ,Pharmacology (medical) ,Internalization ,media_common ,Pharmacology ,lcsh:RM1-950 ,Cancer ,Precision medicine ,medicine.disease ,Small molecule ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,030220 oncology & carcinogenesis ,intracellular targeting ,Intracellular - Abstract
Despite major advances, cancer remains one of the largest burdens of disease worldwide. One reason behind this is that killing tumor cells without affecting healthy surrounding tissue remains a largely elusive prospect, despite the widespread availability of cytotoxic chemotherapeutic agents. To meet these modern healthcare requirements, it is essential to develop precision therapeutics that minimise off-target side-effects for various cancer types. To this end, highly specific molecular targeting agents against cancer are of great interest. These agents may work by targeting intracellular signalling pathways following receptor binding, or via internalization and targeting to specific subcellular compartments. DNA aptamers represent a promising molecular tool in this arena that can be used for both specific cell surface targeting and subsequent internalization and can also elicit a functional effect upon internalization. This review examines various cancer targeting cell-internalizing aptamers, with a particular focus towards functional aptamers that do not require additional conjugation to nanoparticles or small molecules to elicit a biological response. With a deeper understanding and precise exploitation of cancer specific molecular pathways, functional intracellular DNA aptamers may be a powerful step towards more widespread development of precision therapeutics.
- Published
- 2020
10. Selective Targeting of Protein Kinase C (PKC)-θ Nuclear Translocation Reduces Mesenchymal Gene Signatures and Reinvigorates Dysfunctional CD8+ T Cells in Immunotherapy-Resistant and Metastatic Cancers
- Author
-
Jenny Dunn, Robert D. McCuaig, Abel H. Y. Tan, Wen Juan Tu, Fan Wu, Kylie M. Wagstaff, Anjum Zafar, Sayed Ali, Himanshu Diwakar, Jane E. Dahlstrom, Elaine G. Bean, Jade K. Forwood, Sofiya Tsimbalyuk, Emily M. Cross, Kristine Hardy, Amanda L. Bain, Elizabeth Ahern, Riccardo Dolcetti, Roberta Mazzieri, Desmond Yip, Melissa Eastgate, Laeeq Malik, Peter Milburn, David A. Jans, and Sudha Rao
- Subjects
Cancer Research ,Oncology ,breast cancer ,cancer stem cell ,epithelial-to-mesenchymal transition ,immunotherapy ,melanoma ,metastasis ,nuclear translocation ,protein kinase C (PKC)-θ ,T cell ,resistance - Abstract
Protein kinase C (PKC)-θ is a serine/threonine kinase with both cytoplasmic and nuclear functions. Nuclear chromatin-associated PKC-θ (nPKC-θ) is increasingly recognized to be pathogenic in cancer, whereas its cytoplasmic signaling is restricted to normal T-cell function. Here we show that nPKC-θ is enriched in circulating tumor cells (CTCs) in patients with triple-negative breast cancer (TNBC) brain metastases and immunotherapy-resistant metastatic melanoma and is associated with poor survival in immunotherapy-resistant disease. To target nPKC-θ, we designed a novel PKC-θ peptide inhibitor (nPKC-θi2) that selectively inhibits nPKC-θ nuclear translocation but not PKC-θ signaling in healthy T cells. Targeting nPKC-θ reduced mesenchymal cancer stem cell signatures in immunotherapy-resistant CTCs and TNBC xenografts. PKC-θ was also enriched in the nuclei of CD8+ T cells isolated from stage IV immunotherapy-resistant metastatic cancer patients. We show for the first time that nPKC-θ complexes with ZEB1, a key repressive transcription factor in epithelial-to-mesenchymal transition (EMT), in immunotherapy-resistant dysfunctional PD1+/CD8+ T cells. nPKC-θi2 inhibited the ZEB1/PKC-θ repressive complex to induce cytokine production in CD8+ T cells isolated from patients with immunotherapy-resistant disease. These data establish for the first time that nPKC-θ mediates immunotherapy resistance via its activity in CTCs and dysfunctional CD8+ T cells. Disrupting nPKC-θ but retaining its cytoplasmic function may offer a means to target metastases in combination with chemotherapy or immunotherapy.
- Published
- 2022
- Full Text
- View/download PDF
11. Nuclear localization and secretion competence are conserved among henipavirus matrix proteins
- Author
-
Kim G. Lieu, Lin-Fa Wang, Hans J Netter, Glenn A. Marsh, Gregory W. Moseley, Kylie M. Wagstaff, Alexander Lee, David A. Jans, and Elisabeth C McLinton
- Subjects
0301 basic medicine ,Virosomes ,Paramyxoviridae ,viruses ,Viral budding ,Nuclear Localization Signals ,030106 microbiology ,Active Transport, Cell Nucleus ,Viral Matrix Proteins ,03 medical and health sciences ,VP40 ,Virus-like particle ,Virology ,Animals ,Humans ,Hendra Virus ,Henipavirus ,Microscopy, Confocal ,Viral matrix protein ,biology ,biology.organism_classification ,Protein Transport ,030104 developmental biology ,Microscopy, Fluorescence ,Viral replication - Abstract
Viruses of the genus Henipavirus of the family Paramyxoviridae are zoonotic pathogens, which have emerged in Southeast Asia, Australia and Africa. Nipah virus (NiV) and Hendra virus are highly virulent pathogens transmitted from bats to animals and humans, while the henipavirus Cedar virus seems to be non-pathogenic in infection studies. The full replication cycle of the Paramyxoviridae occurs in the host cell's cytoplasm, where viral assembly is orchestrated by the matrix (M) protein. Unexpectedly, the NiV-M protein traffics through the nucleus as an essential step to engage the plasma membrane in preparation for viral budding/release. Comparative studies were performed to assess whether M protein nuclear localization is a common feature of the henipaviruses, including the recently sequenced (although not yet isolated) Ghanaian bat henipavirus (Kumasi virus, GH-M74a virus) and Mojiang virus. Live-cell confocal microscopy revealed that nuclear translocation of GFP-fused M protein is conserved between henipaviruses in both human- and bat-derived cell lines. However, the efficiency of M protein nuclear localization and virus-like particle budding competency varied. Additionally, Cedar virus-, Kumasi virus- and Mojiang virus-M proteins were mutated in a bipartite nuclear localization signal, indicating that a key lysine residue is essential for nuclear import, export and induction of budding events, as previously reported for NiV-M. The results of this study suggest that the M proteins of henipaviruses may utilize a similar nucleocytoplasmic trafficking pathway as an essential step during viral replication in both humans and bats.
- Published
- 2017
- Full Text
- View/download PDF
12. Interactome of the inhibitory isoform of the nuclear transporter Importin 13
- Author
-
Yasuka L. Yamaguchi, Rebecca G. Davies, David A. Jans, Patrick P.L. Tam, Shadma Fatima, Kate L Loveland, Kylie M. Wagstaff, Kim G. Lieu, and Satomi S. Tanaka
- Subjects
Male ,0301 basic medicine ,Gene isoform ,Immunoprecipitation ,Importin ,Karyopherins ,Biology ,Mice ,03 medical and health sciences ,Receptors, Glucocorticoid ,Eukaryotic translation ,Spermatocytes ,Two-Hybrid System Techniques ,Protein Interaction Mapping ,Testis ,Animals ,Humans ,Protein Isoforms ,Initiation factor ,Spermatogenesis ,Molecular Biology ,Gene Library ,Cell Nucleus ,Genetics ,030102 biochemistry & molecular biology ,cDNA library ,High Mobility Group Proteins ,Gene Expression Regulation, Developmental ,Cell Biology ,Spermatids ,Cell biology ,Protein Transport ,030104 developmental biology ,High-mobility group ,Nuclear transport ,Eukaryotic Initiation Factor-4G ,Protein Binding ,Signal Transduction - Abstract
Importin 13 (Imp13) is a bidirectional nuclear transporter of proteins involved in a range of important cellular processes, with an N-terminally truncated inhibitory isoform (tImp13) specifically expressed in testis. To gain insight into tImp13 function, we performed a yeast-2-hybrid screen from a human testis cDNA library, identifying for the first time a suite of interactors with roles in diverse cellular process. We validated the interaction of tImp13 with Eukaryotic translation initiation factor 4γ2 (EIF4G2) and High mobility group containing protein 20A (HMG20A), benchmarking that with glucocorticoid receptor (GR), a known Imp13 interactor expressed in testis. Coimmunoprecipitation assays indicated association of both tImp13 and Imp13 with EIF4G2, HMG20A and GR. Quantitative confocal microscopic analysis revealed the ability of tImp13 to inhibit the nuclear localisation of EIF4G2, HMG20A and GR, as well as that of Imp13 to act as a nuclear exporter for both EIF4G2 and HMG20A, and as a nuclear importer for GR. The physiological relevance of these results was highlighted by the cytoplasmic localisation of EIF4G2, HMG20A and GR in pachytene spermatocytes/round spermatids in the murine testis where tImp13 is present at high levels, in contrast to the nuclear localisation of HMG20A and GR in spermatogonia, where tImp13 is largely absent. Interestingly, Imp13, EIF4G2, HMG20A and GR were found together in the acrosome vesicle of murine epididymal spermatozoa. Collectively, our findings show, for the first time, that tImp13 may have a functional role in the mature spermatozoa, in addition to that in the meiotic germ cells of the testis.
- Published
- 2017
- Full Text
- View/download PDF
13. Adenovirus Terminal Protein Contains a Bipartite Nuclear Localisation Signal Essential for Its Import into the Nucleus
- Author
-
David R. Thomas, Hareth Al-Wassiti, Kylie M. Wagstaff, Colin W. Pouton, Angus P. R. Johnston, David A. Jans, and Stewart A. Fabb
- Subjects
0301 basic medicine ,Nuclear Localization Signals ,DNA binding proteins (DBP) ,terminal protein (TP) ,Chromosomal translocation ,medicine.disease_cause ,lcsh:Chemistry ,Cytosol ,nuclear localisation signal (NLS) ,preterminal protein (pTP) ,lcsh:QH301-705.5 ,Spectroscopy ,Infectivity ,Microscopy, Confocal ,Chemistry ,viral genome ,adenovirus ,General Medicine ,Transfection ,beta Karyopherins ,Computer Science Applications ,Cell biology ,medicine.anatomical_structure ,Protein Binding ,alpha Karyopherins ,Green Fluorescent Proteins ,Active Transport, Cell Nucleus ,Genome, Viral ,Importin ,Article ,Catalysis ,Adenoviridae ,Inorganic Chemistry ,Viral Proteins ,03 medical and health sciences ,Escherichia coli ,medicine ,Humans ,NLS ,Physical and Theoretical Chemistry ,Molecular Biology ,Cell Nucleus ,030102 biochemistry & molecular biology ,Organic Chemistry ,DNA ,HEK293 Cells ,030104 developmental biology ,lcsh:Biology (General) ,lcsh:QD1-999 ,Nuclear transport ,DNA viruses ,Nucleus ,HeLa Cells - Abstract
Adenoviruses contain dsDNA covalently linked to a terminal protein (TP) at the 5′end. TP plays a pivotal role in replication and long-lasting infectivity. TP has been reported to contain a nuclear localisation signal (NLS) that facilitates its import into the nucleus. We studied the potential NLS motifs within TP using molecular and cellular biology techniques to identify the motifs needed for optimum nuclear import. We used confocal imaging microscopy to monitor the localisation and nuclear association of GFP fusion proteins. We identified two nuclear localisation signals, PV(R)6VP and MRRRR, that are essential for fully efficient TP nuclear entry in transfected cells. To study TP–host interactions further, we expressed TP in Escherichia coli (E. coli). Nuclear uptake of purified protein was determined in digitonin-permeabilised cells. The data confirmed that nuclear uptake of TP requires active transport using energy and shuttling factors. This mechanism of nuclear transport was confirmed when expressed TP was microinjected into living cells. Finally, we uncovered the nature of TP binding to host nuclear shuttling proteins, revealing selective binding to Imp β, and a complex of Imp α/β but not Imp α alone. TP translocation to the nucleus could be inhibited using selective inhibitors of importins. Our results show that the bipartite NLS is required for fully efficient TP entry into the nucleus and suggest that this translocation can be carried out by binding to Imp β or Imp α/β. This work forms the biochemical foundation for future work determining the involvement of TP in nuclear delivery of adenovirus DNA.
- Published
- 2021
- Full Text
- View/download PDF
14. The FDA-approved drug ivermectin inhibits the replication of SARS-CoV-2 in vitro
- Author
-
Mike Catton, Julian Druce, Kylie M. Wagstaff, David A. Jans, and Leon Caly
- Subjects
0301 basic medicine ,viruses ,Pneumonia, Viral ,030106 microbiology ,Virus Replication ,medicine.disease_cause ,Antiviral Agents ,Approved drug ,Virus ,Betacoronavirus ,03 medical and health sciences ,Ivermectin ,Virology ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Drug Approval ,Pandemics ,Vero Cells ,Coronavirus ,Pharmacology ,SARS-CoV-2 ,business.industry ,Australia ,COVID-19 ,Outbreak ,In vitro ,Clinical trial ,030104 developmental biology ,Vero cell ,Coronavirus Infections ,business ,medicine.drug - Abstract
Although several clinical trials are now underway to test possible therapies, the worldwide response to the COVID-19 outbreak has been largely limited to monitoring/containment. We report here that Ivermectin, an FDA-approved anti-parasitic previously shown to have broad-spectrum anti-viral activity in vitro, is an inhibitor of the causative virus (SARS-CoV-2), with a single addition to Vero-hSLAM cells 2 h post infection with SARS-CoV-2 able to effect ~5000-fold reduction in viral RNA at 48 h. Ivermectin therefore warrants further investigation for possible benefits in humans.
- Published
- 2020
- Full Text
- View/download PDF
15. Author Correction: Novel inhibitors targeting Venezuelan equine encephalitis virus capsid protein identified using In Silico Structure-Based-Drug-Design
- Author
-
Aaron DeBono, David A. Jans, Lindsay Lundberg, Sharon Shechter, Kylie M. Wagstaff, David R. Thomas, Kylene Kehn-Hall, Shih-Chao Lin, and Chelsea Pinkham
- Subjects
alpha Karyopherins ,Drug ,Nucleocytoplasmic Transport Proteins ,In silico ,media_common.quotation_subject ,Active Transport, Cell Nucleus ,lcsh:Medicine ,Biology ,Virus Replication ,medicine.disease_cause ,Antiviral Agents ,Encephalitis Virus, Venezuelan Equine ,Capsid ,Chlorocebus aethiops ,Drug Discovery ,medicine ,Animals ,Humans ,Computer Simulation ,Author Correction ,lcsh:Science ,Vero Cells ,media_common ,Cell Nucleus ,Multidisciplinary ,lcsh:R ,beta Karyopherins ,Virology ,Drug Design ,Venezuelan equine encephalitis virus ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,Structure based ,Capsid Proteins ,lcsh:Q - Abstract
Therapeutics are currently unavailable for Venezuelan equine encephalitis virus (VEEV), which elicits flu-like symptoms and encephalitis in humans, with an estimated 14% of cases resulting in neurological disease. Here we identify anti-VEEV agents using in silico structure-based-drug-design (SBDD) for the first time, characterising inhibitors that block recognition of VEEV capsid protein (C) by the host importin (IMP) α/β1 nuclear transport proteins. From an initial screen of 1.5 million compounds, followed by in silico refinement and screening for biological activity in vitro, we identified 21 hit compounds which inhibited IMPα/β1:C binding with IC
- Published
- 2018
- Full Text
- View/download PDF
16. Nuclear transporter Importin-13 plays a key role in the oxidative stress transcriptional response
- Author
-
K. A. Gajewska, Kylie M. Wagstaff, David A. Jans, H. Lescesen, and M. Ramialison
- Subjects
Embryonic stem cells ,animal structures ,Science ,Active Transport, Cell Nucleus ,Kruppel-Like Transcription Factors ,General Physics and Astronomy ,Importin ,Biology ,Karyopherins ,environment and public health ,General Biochemistry, Genetics and Molecular Biology ,Article ,Transcriptome ,Kruppel-Like Factor 4 ,Mice ,Nuclear receptors ,Animals ,Humans ,Transport receptors ,Nuclear export signal ,Transcription factor ,Mice, Knockout ,Gene knockdown ,Multidisciplinary ,Protein transport ,RNA sequencing ,General Chemistry ,Hydrogen Peroxide ,Embryonic stem cell ,Cell biology ,Oxidative Stress ,Nuclear receptor ,KLF4 ,embryonic structures ,Protein Kinases ,HeLa Cells ,Transcription Factors - Abstract
The importin superfamily member Importin-13 is a bidirectional nuclear transporter. To delineate its functional roles, we performed transcriptomic analysis on wild-type and Importin-13-knockout mouse embryonic stem cells, revealing enrichment of differentially expressed genes involved in stress responses and apoptosis regulation. De novo promoter motif analysis on 277 Importin-13-dependent genes responsive to oxidative stress revealed an enrichment of motifs aligned to consensus sites for the transcription factors specificity protein 1, SP1, or Kruppel like factor 4, KLF4. Analysis of embryonic stem cells subjected to oxidative stress revealed that Importin-13-knockout cells were more resistant, with knockdown of SP1 or KLF4 helping protect wild-type embryonic stem cells against stress-induced death. Importin-13 was revealed to bind to SP1 and KLF4 in a cellular context, with a key role in oxidative stress-dependent nuclear export of both transcription factors. The results are integral to understanding stress biology, highlighting the importance of Importin-13 in the stress response., Importin superfamily member IPO13 mediates nuclear transport bidirectionally. Here the authors delineate the transcriptome of wild-type and IPO13-knockout mouse embryonic stem cells, revealing IPO13’s key role in the oxidative stress response through targeted transport of specific transcription factors.
- Published
- 2018
17. Novel RU486 (mifepristone) analogues with increased activity against Venezuelan Equine Encephalitis Virus but reduced progesterone receptor antagonistic activity
- Author
-
Aaron DeBono, Christine L. Clarke, Kylene Kehn-Hall, David A. Jans, Lindsay Lundberg, David R. Thomas, Ying Cao, Kylie M. Wagstaff, Sharon Shechter, Chelsea Pinkham, and J. Dinny Graham
- Subjects
0301 basic medicine ,Active Transport, Cell Nucleus ,lcsh:Medicine ,Alphavirus ,Pharmacology ,medicine.disease_cause ,Article ,Encephalitis Virus, Venezuelan Equine ,03 medical and health sciences ,Structure-Activity Relationship ,0302 clinical medicine ,Receptors, Glucocorticoid ,Progesterone receptor ,medicine ,Humans ,Receptor ,lcsh:Science ,Abortifacient ,Cell Nucleus ,Multidisciplinary ,biology ,business.industry ,lcsh:R ,Mifepristone ,medicine.disease ,biology.organism_classification ,3. Good health ,Molecular Docking Simulation ,030104 developmental biology ,Toxicity ,Venezuelan equine encephalitis virus ,lcsh:Q ,Capsid Proteins ,business ,Receptors, Progesterone ,030217 neurology & neurosurgery ,Encephalitis ,medicine.drug ,HeLa Cells ,Protein Binding - Abstract
There are currently no therapeutics to treat infection with the alphavirus Venezuelan equine encephalitis virus (VEEV), which causes flu-like symptoms leading to neurological symptoms in up to 14% of cases. Large outbreaks of VEEV can result in 10,000 s of human cases and mass equine death. We previously showed that mifepristone (RU486) has anti-VEEV activity (EC50 = 20 μM) and only limited cytotoxicity (CC50 > 100 μM), but a limitation in its use is its abortifacient activity resulting from its ability to antagonize the progesterone receptor (PR). Here we generate a suite of new mifepristone analogues with enhanced antiviral properties, succeeding in achieving >11-fold improvement in anti-VEEV activity with no detectable increase in toxicity. Importantly, we were able to derive a lead compound with an EC50 of 7.2 µM and no detectable PR antagonism activity. Finally, based on our SAR analysis we propose avenues for the further development of these analogues as safe and effective anti-VEEV agents.
- Published
- 2018
18. Novel Antivirals For Venezuelan Equine Encephalitis
- Author
-
Jonathan B. Baell, Lindsay Lundberg, and David A. Jans, Kylie M. Wagstaff, David R. Thomas, and Kylene Kehn-Hall
- Subjects
Biology ,Equine Encephalitis ,Virology - Published
- 2018
- Full Text
- View/download PDF
19. Tumor cell-specific photothermal killing by SELEX-derived DNA aptamer-targeted gold nanorods
- Author
-
Ramya Chandrasekaran, Lim Wei Yap, Wenlong Cheng, Alexander Lee, Kylie M. Wagstaff, and David A. Jans
- Subjects
Programmed cell death ,Aptamer ,Breast Neoplasms ,02 engineering and technology ,Biology ,010402 general chemistry ,01 natural sciences ,chemistry.chemical_compound ,Cell Line, Tumor ,Humans ,Cytotoxic T cell ,General Materials Science ,Nanotubes ,SELEX Aptamer Technique ,Hyperthermia, Induced ,Aptamers, Nucleotide ,Phototherapy ,Photothermal therapy ,021001 nanoscience & nanotechnology ,Molecular biology ,0104 chemical sciences ,Carcinoma, Ductal ,chemistry ,Cell culture ,Cancer research ,Female ,Gold ,0210 nano-technology ,DNA ,Systematic evolution of ligands by exponential enrichment - Abstract
Despite widespread availability of cytotoxic chemotherapeutic agents, the killing of tumour cells without affecting healthy surrounding tissue remains elusive, although recent developments in terms of plasmonic nanoparticles capable of photothermal killing have some promise. Here we describe novel DNA aptamer-tethered gold nanorods (GNRs) that act as efficient photothermal therapeutics against tumour cells, but not their isogenic normal cell counterparts. A modified Cell-SELEX process was developed to select a novel DNA aptamer (KW16-13) that specifically recognised and was internalised by cells of the MCF10CA1h human breast ductal carcinoma line but not by those of its isogenic normal counterpart (MCF10A). GNRs conjugated to KW16-13 were readily internalized by the MCF10CA1h tumour cells with minimal uptake by MCF10A normal cells. Upon near infrared (NIR) light irradiation, tumour cell death of96%, could be effected, compared to1% in the normal cells or cells incubated with GNRs alone, our KW16-13 aptamer-targeted GNRs thus showing71-fold tumor cell death than GNRs-targeted with a previously described aptamer. This demonstrates the significant potential for aptamer functionalised-GNRs to be used effective and above all selective anti-cancer photothermal therapeutics.
- Published
- 2016
- Full Text
- View/download PDF
20. PKA-site phosphorylation of importin13 regulates its subcellular localization and nuclear transport function
- Author
-
Xujie Liu, David A. Jans, Kylie M. Wagstaff, Rebecca G. Davies, Xiuyu Shi, Tao Tao, and Wenbo Lin
- Subjects
0301 basic medicine ,Cytoplasm ,PAX6 Transcription Factor ,Active Transport, Cell Nucleus ,Importin ,Karyopherins ,environment and public health ,Biochemistry ,03 medical and health sciences ,Humans ,Phosphorylation ,Nuclear export signal ,Protein kinase A ,Molecular Biology ,Fluorescence loss in photobleaching ,Cell Nucleus ,Chemistry ,Fluorescence recovery after photobleaching ,Cell Biology ,Cyclic AMP-Dependent Protein Kinases ,Cell biology ,030104 developmental biology ,Nucleocytoplasmic Transport ,Nuclear transport ,HeLa Cells - Abstract
Importin 13 (IPO13) is a key member of the importin β superfamily, which can transport cargoes both into and out of the nucleus to contribute to a variety of important cellular processes. IPO13 is known to undergo phosphorylation, but the impact of this on function has not been investigated. Here, we show for the first time that IPO13 is phosphorylated by cAMP-dependent protein kinase A specifically at serine 193. Results from fluorescence recovery after photobleaching and fluorescence loss in photobleaching approaches establish that negative charge at serine 193 through phosphorylation or point mutation both reduces IPO13 nuclear import and increases its nuclear export. Importantly, phosphorylation also appears to enhance cargo interaction on the part of IPO13, with significant impact on localization, as shown for the Pax6 homeobox-containing transcription partner. This is the first report that IPO13 can be phosphorylated at Ser193 and that this modification regulates IPO13 subcellular localization and nucleocytoplasmic transport function, with important implications for IPO13's role in development and other processes.
- Published
- 2018
21. Recognition by host nuclear transport proteins drives disorder-to-order transition in Hendra virus V
- Author
-
Gregory W. Moseley, Natalie A. Borg, Sarah C. Atkinson, David A. Jans, Michelle D. Audsley, Ashley M. Buckle, Jason J. Paxman, Glenn A. Marsh, Kylie M. Wagstaff, Steven M. Heaton, Kim G. Lieu, and David R. Thomas
- Subjects
Models, Molecular ,0301 basic medicine ,Nucleocytoplasmic Transport Proteins ,Immunoprecipitation ,viruses ,Molecular Conformation ,lcsh:Medicine ,Receptors, Cytoplasmic and Nuclear ,Importin ,Karyopherins ,Intrinsically disordered proteins ,Antiviral Agents ,Article ,Hendra Virus ,Structure-Activity Relationship ,Viral Proteins ,03 medical and health sciences ,Drug Discovery ,Humans ,Protein Interaction Domains and Motifs ,lcsh:Science ,Cell Nucleus ,Henipavirus Infections ,Multidisciplinary ,Chemistry ,lcsh:R ,virus diseases ,Transporter ,digestive system diseases ,3. Good health ,Cell biology ,Protein Transport ,030104 developmental biology ,Interaction with host ,Cytoplasm ,Gene Knockdown Techniques ,Host-Pathogen Interactions ,lcsh:Q ,Nuclear transport ,Protein Binding - Abstract
Hendra virus (HeV) is a paramyxovirus that causes lethal disease in humans, for which no vaccine or antiviral agent is available. HeV V protein is central to pathogenesis through its ability to interact with cytoplasmic host proteins, playing key antiviral roles. Here we use immunoprecipitation, siRNA knockdown and confocal laser scanning microscopy to show that HeV V shuttles to and from the nucleus through specific host nuclear transporters. Spectroscopic and small angle X-ray scattering studies reveal HeV V undergoes a disorder-to-order transition upon binding to either importin α/β1 or exportin-1/Ran-GTP, dependent on the V N-terminus. Importantly, we show that specific inhibitors of nuclear transport prevent interaction with host transporters, and reduce HeV infection. These findings emphasize the critical role of host-virus interactions in HeV infection, and potential use of compounds targeting nuclear transport, such as the FDA-approved agent ivermectin, as anti-HeV agents.
- Published
- 2018
- Full Text
- View/download PDF
22. Identification of novel antivirals inhibiting recognition of Venezuelan equine encephalitis virus capsid protein by the Importin α/β1 heterodimer through high-throughput screening
- Author
-
Chelsea Pinkham, Kylene Kehn-Hall, Caroline A. Hick, David R. Thomas, Jonathan B. Baell, Kylie M. Wagstaff, David A. Jans, Lindsay Lundberg, Sharon Shechter, and Aaron DeBono
- Subjects
0301 basic medicine ,Cell Survival ,High-throughput screening ,Active Transport, Cell Nucleus ,Importin ,Alphavirus ,Karyopherins ,medicine.disease_cause ,Virus Replication ,Antiviral Agents ,Encephalitis Virus, Venezuelan Equine ,Small Molecule Libraries ,03 medical and health sciences ,Inhibitory Concentration 50 ,Virology ,Chlorocebus aethiops ,medicine ,Animals ,Vero Cells ,Host cell nucleus ,Pharmacology ,030102 biochemistry & molecular biology ,biology ,Molecular Structure ,Encephalomyelitis, Venezuelan Equine ,biology.organism_classification ,High-Throughput Screening Assays ,030104 developmental biology ,Viral replication ,Capsid ,Venezuelan equine encephalitis virus ,Host-Pathogen Interactions ,Capsid Proteins ,Nuclear transport ,Protein Binding - Abstract
Although the alphavirus Venezuelan equine encephalitis virus (VEEV) has been the cause of multiple outbreaks resulting in extensive human and equine mortality and morbidity, there are currently no anti-VEEV therapeutics available. VEEV pathogenicity is largely dependent on targeting of the viral capsid protein (CP) to the host cell nucleus through the nuclear transporting importin (Imp) α/β1 heterodimer. Here we perform a high-throughput screen, combined with nested counterscreens to identify small molecules able to inhibit the Impα/β1:CP interaction for the first time. Several compounds were able to significantly reduce viral replication in infected cells. Compound G281-1564 in particular could inhibit VEEV replication at low μM concentration, while showing minimal toxicity, with steady state and dynamic quantitative microscopic measurements confirming its ability to inhibit CP nuclear import. This study establishes the principle that inhibitors of CP nucleocytoplasmic trafficking can have potent antiviral activity against VEEV, and represents a platform for future development of safe anti-VEEV compounds with high efficacy and specificity.
- Published
- 2017
23. Nucleocytoplasmic shuttling of the West Nile virus RNA-dependent RNA polymerase NS5 is critical to infection
- Author
-
Peter A. White, Adam J. Lopez-Denman, David A. Jans, Kylie M. Wagstaff, Jason M. Mackenzie, and Alice G. Russo
- Subjects
0301 basic medicine ,viruses ,030106 microbiology ,Immunology ,Nuclear Localization Signals ,RNA-dependent RNA polymerase ,Viral Plaque Assay ,Biology ,Viral Nonstructural Proteins ,Virus Replication ,Microbiology ,03 medical and health sciences ,chemistry.chemical_compound ,Kunjin virus ,Virology ,Chlorocebus aethiops ,Animals ,Enzyme Inhibitors ,Nuclear export signal ,Vero Cells ,virus diseases ,Leptomycin ,biology.organism_classification ,Flavivirus ,Protein Transport ,030104 developmental biology ,chemistry ,Viral replication ,Fatty Acids, Unsaturated ,Mutagenesis, Site-Directed ,Nuclear transport ,West Nile virus ,Nuclear localization sequence - Abstract
West Nile virus (WNV) is a single-stranded, positive sense RNA virus of the family Flaviviridae and is a significant pathogen of global medical importance. Flavivirus replication is known to be exclusively cytoplasmic, but we show here for the first time that access to the nucleus of the WNV strain Kunjin (WNVKUN ) RNA-dependent RNA polymerase (protein NS5) is central to WNVKUN virus production. We show that treatment of cells with the specific nuclear export inhibitor leptomycin B (LMB) results in increased NS5 nuclear accumulation in WNVKUN -infected cells and NS5-transfected cells, indicative of nucleocytoplasmic shuttling under normal conditions. We used site-directed mutagenesis to identify the nuclear localisation sequence (NLS) responsible for WNVKUN NS5 nuclear targeting, observing that mutation of this NLS resulted in exclusively cytoplasmic accumulation of NS5 even in the presence of leptomycin B. Introduction of NS5 NLS mutations into FLSDX, an infectious clone of WNVKUN , resulted in lethality, suggesting that the ability of NS5 to traffic into the nucleus in integral to WNVKUN replication. This study thus shows for the first time that NLS-dependent trafficking into the nucleus during infection of WNVKUN NS5 is critical for viral replication. Excitingly, specific inhibitors of NS5 nuclear import reduce WNVKUN virus production, proving the principle that inhibition of WNVKUN NS5 nuclear import is a viable therapeutic avenue for antiviral drug development in the future.
- Published
- 2017
24. Nucleocytoplasmic shuttling of the Duchenne muscular dystrophy gene product dystrophin Dp71d is dependent on the importin α/β and CRM1 nuclear transporters and microtubule motor dynein
- Author
-
Kim G. Lieu, A. Vásquez-Limeta, R. Suárez-Sánchez, Paulina Margarita Azuara-Medina, Kylie M. Wagstaff, O. Hernández-Hernández, David A. Jans, Pablo Gomez, A. Aguilar, Bulmaro Cisneros, and G. Velez
- Subjects
alpha Karyopherins ,Dystrophin Dp71 ,Dynein ,Emerin ,Receptors, Cytoplasmic and Nuclear ,Importin ,Karyopherins ,Biology ,Microtubules ,Cell Line ,Dystrophin ,Mice ,Exportin Crm1 ,Microtubule ,Importins ,Animals ,Nuclear protein ,ZZ domain ,Molecular Biology ,DNA Primers ,Cell Nucleus ,Base Sequence ,Dyneins ,Cell Biology ,beta Karyopherins ,Rats ,Cell biology ,Muscular Dystrophy, Duchenne ,Protein Transport ,Biochemistry ,Nucleoporin ,Nuclear transport ,Nuclear localization sequence - Abstract
Even though the Duchenne muscular dystrophy (DMD) gene product Dystrophin Dp71d is involved in various key cellular processes through its role as a scaffold for structural and signalling proteins at the plasma membrane as well as the nuclear envelope, its subcellular trafficking is poorly understood. Here we map the nuclear import and export signals of Dp71d by truncation and point mutant analysis, showing for the first time that Dp71d shuttles between the nucleus and cytoplasm mediated by the conventional nuclear transporters, importin (IMP) α/β and the exportin CRM1. Binding was confirmed in cells using pull-downs, while in vitro binding assays showed direct, high affinity (apparent dissociation coefficient of c. 0.25nM) binding of Dp71d to IMPα/β. Interestingly, treatment of cells with the microtubule depolymerizing reagent nocodazole or the dynein inhibitor EHNA both decreased Dp71d nuclear localization, implying that Dp71d nuclear import may be facilitated by microtubules and the motor protein dynein. The role of Dp71d in the nucleus appears to relate in part to interaction with the nuclear envelope protein emerin, and maintenance of the integrity of the nuclear architecture. The clear implication is that Dp71d's previously unrecognised nuclear transport properties likely contribute to various, important physiological roles.
- Published
- 2014
- Full Text
- View/download PDF
25. Novel Flavivirus Antiviral That Targets the Host Nuclear Transport Importin α/β1 Heterodimer
- Author
-
Belinda Maher, David A. Jans, Sarah C. Atkinson, Noelia Roman, Chunxiao Wang, Natalie A. Borg, Johanna E. Fraser, Sundy N.Y. Yang, Kylie M. Wagstaff, and Jade K. Forwood
- Subjects
Models, Molecular ,alpha Karyopherins ,0301 basic medicine ,Indoles ,importins ,viruses ,Population ,Protein domain ,Active Transport, Cell Nucleus ,nuclear transport inhibitors ,Importin ,Dengue virus ,medicine.disease_cause ,Antiviral Agents ,Article ,Cell Line ,Inhibitory Concentration 50 ,03 medical and health sciences ,Phenols ,Protein Domains ,flavivirus ,medicine ,Animals ,education ,lcsh:QH301-705.5 ,Cell Nucleus ,education.field_of_study ,dengue virus ,030102 biochemistry & molecular biology ,biology ,Protein Stability ,Chemistry ,virus diseases ,General Medicine ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,In vitro ,3. Good health ,Cell biology ,Flavivirus ,030104 developmental biology ,lcsh:Biology (General) ,Cell culture ,viral infection ,Protein Multimerization ,Nuclear transport - Abstract
Dengue virus (DENV) threatens almost 70% of the world&rsquo, s population, with effective vaccine or therapeutic currently available. A key contributor to infection is nuclear localisation in the infected cell of DENV nonstructural protein 5 (NS5) through the action of the host importin (IMP) &alpha, /&beta, 1 proteins. Here, we used a range of microscopic, virological and biochemical/biophysical approaches to show for the first time that the small molecule GW5074 has anti-DENV action through its novel ability to inhibit NS5&ndash, IMP&alpha, 1 interaction in vitro as well as NS5 nuclear localisation in infected cells. Strikingly, GW5074 not only inhibits IMP&alpha, binding to IMP&beta, 1, but can dissociate preformed IMP&alpha, 1 heterodimer, through targeting the IMP&alpha, armadillo (ARM) repeat domain to impact IMP&alpha, thermal stability and &alpha, helicity, as shown using analytical ultracentrifugation, thermostability analysis and circular dichroism measurements. Importantly, GW5074 has strong antiviral activity at low µ, M concentrations against not only DENV-2, but also zika virus and West Nile virus. This work highlights DENV NS5 nuclear targeting as a viable target for anti-flaviviral therapeutics.
- Published
- 2019
- Full Text
- View/download PDF
26. Nuclear trafficking of proteins from RNA viruses: Potential target for antivirals?
- Author
-
Leon Caly, Kylie M. Wagstaff, and David A. Jans
- Subjects
Pharmacology ,viruses ,Active Transport, Cell Nucleus ,Biology ,medicine.disease ,medicine.disease_cause ,Antiviral Agents ,Virology ,Virus ,Microbiology ,Dengue fever ,Viral Proteins ,RNA Virus Infections ,Veterinary virology ,Venezuelan equine encephalitis virus ,medicine ,Animals ,Humans ,RNA Viruses ,Antibody-dependent enhancement ,Nuclear protein ,Oncovirus ,Host cell nucleus - Abstract
A key aspect of the infectious cycle of many viruses is the transport of specific viral proteins into the host cell nucleus to perturb the antiviral response. Examples include a number of RNA viruses that are significant human pathogens, such as human immunodeficiency virus (HIV)-1, influenza A, dengue, respiratory syncytial virus and rabies, as well agents that predominantly infect livestock, such as Rift valley fever virus and Venezuelan equine encephalitis virus. Inhibiting the nuclear trafficking of viral proteins as a therapeutic strategy offers an attractive possibility, with important recent progress having been made with respect to HIV-1 and dengue. The results validate nuclear protein import as an antiviral target, and suggest the identification and development of nuclear transport inhibitors as a viable therapeutic approach for a range of human and zoonotic pathogenic viruses.
- Published
- 2012
- Full Text
- View/download PDF
27. Ivermectin is a specific inhibitor of importin α/β-mediated nuclear import able to inhibit replication of HIV-1 and dengue virus
- Author
-
David Harrich, Haran Sivakumaran, Steven M. Heaton, Kylie M. Wagstaff, and David A. Jans
- Subjects
NES, nuclear export signal ,DENV, dengue virus ,Dengue virus ,Imp, importin ,Virus Replication ,medicine.disease_cause ,Biochemistry ,PIC, pre-integration complex ,0302 clinical medicine ,VSV-G, vesicular stomatitis virus glycoprotein ,Polymerase ,GFP, green fluorescent protein ,0303 health sciences ,DMEM, Dulbecco's modified Eagle's medium ,SUMO, small ubiquitin-related modifier ,hCMV, human cytomegalovirus ,IN, integrase ,3. Good health ,Integrase ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,CLSM, confocal laser-scanning microscopy ,T-ag, large tumour antigen ,PTHrP, parathyroid hormone-related protein ,TRF1, telomeric repeat factor-binding protein 1 ,NS5, non-structural protein 5 ,Accelerated Publication ,Importin ,SRY, sex-determining region of the Y chromosome ,Karyopherins ,Biology ,ivermectin ,Tat, transactivator of transcription ,03 medical and health sciences ,FBS, fetal bovine serum ,importin α/β1 ,parasitic diseases ,medicine ,Humans ,SV40, simian virus 40 ,Molecular Biology ,030304 developmental biology ,dengue virus ,HIV ,Cell Biology ,nuclear import ,Virology ,sulfo-NHS-biotin, sulfo-N-hydroxysuccinimide biotin ,NLS, nuclear localization signal ,Cytoplasm ,HIV-1 ,biology.protein ,Nuclear transport ,Carcinogenesis ,Nucleus ,LMB, leptomycin B ,HeLa Cells - Abstract
The movement of proteins between the cytoplasm and nucleus mediated by the importin superfamily of proteins is essential to many cellular processes, including differentiation and development, and is critical to disease states such as viral disease and oncogenesis. We recently developed a high-throughput screen to identify specific and general inhibitors of protein nuclear import, from which ivermectin was identified as a potential inhibitor of importin α/β-mediated transport. In the present study, we characterized in detail the nuclear transport inhibitory properties of ivermectin, demonstrating that it is a broad-spectrum inhibitor of importin α/β nuclear import, with no effect on a range of other nuclear import pathways, including that mediated by importin β1 alone. Importantly, we establish for the first time that ivermectin has potent antiviral activity towards both HIV-1 and dengue virus, both of which are strongly reliant on importin α/β nuclear import, with respect to the HIV-1 integrase and NS5 (non-structural protein 5) polymerase proteins respectively. Ivermectin would appear to be an invaluable tool for the study of protein nuclear import, as well as the basis for future development of antiviral agents.
- Published
- 2012
- Full Text
- View/download PDF
28. An AlphaScreen®-Based Assay for High-Throughput Screening for Specific Inhibitors of Nuclear Import
- Author
-
Kylie M. Wagstaff, Anna C. Hearps, Stephen M. Rawlinson, and David A. Jans
- Subjects
alpha Karyopherins ,viruses ,High-throughput screening ,Active Transport, Cell Nucleus ,Human immunodeficiency virus (HIV) ,HIV Integrase ,Importin ,medicine.disease_cause ,Sensitivity and Specificity ,Biochemistry ,Analytical Chemistry ,Protein–protein interaction ,Hormone Antagonists ,Viral life cycle ,Drug Discovery ,medicine ,Humans ,Cell Nucleus ,Ivermectin ,Antiparasitic Agents ,biology ,Reproducibility of Results ,beta Karyopherins ,Virology ,High-Throughput Screening Assays ,Integrase ,Mifepristone ,biology.protein ,Molecular Medicine ,Nuclear transport ,HeLa Cells ,Protein Binding ,Biotechnology - Abstract
Specific viral proteins enter the nucleus of infected cells to perform essential functions, as part of the viral life cycle. The integrase (IN) molecule of human immunodeficiency virus (HIV)-1 is of particular interest in this context due to its integral role in integrating the HIV genome into that of the infected host cell. Most IN-based antiviral compounds target the IN/DNA interaction, but since IN must first enter the nucleus before it can perform these critical functions, nuclear transport of IN is also an attractive target for therapeutic intervention. Here the authors describe a novel high-throughput screening assay for identifying inhibitors of nuclear import, particularly IN, based on amplified luminescent proximity homogeneous assay (AlphaScreen(®)) technology, which is high throughput, requires low amounts of material, and is efficient and cost-effective. The authors use the assay to screen for specific inhibitors of the interaction between IN and its nuclear transport receptor importin α/β, successfully identifying several inhibitors of the IN/importin α/β interaction. Importantly, they demonstrate that one of the identified compounds, mifepristone, is effective in preventing active nuclear transport of IN in transfected cells and hence may represent a useful anti-HIV therapeutic. The screen also identified broad-spectrum importin α/β inhibitors such as ivermectin, which may represent useful tools for nuclear transport research in the future. The authors validate the activity and specificity of mifepristone and ivermectin in inhibiting nuclear protein import in living cells, underlining the utility of the screening approach.
- Published
- 2011
- Full Text
- View/download PDF
29. Molecular dissection of an inhibitor targeting the HIV integrase dependent preintegration complex nuclear import
- Author
-
David Tyssen, Anna C. Hearps, Gilda Tachedjian, Stephen J. Headey, David R. Thomas, Kylie M. Wagstaff, David A. Jans, and Sushama Telwatte
- Subjects
Virus Integration ,Immunology ,Active Transport, Cell Nucleus ,HIV Integrase ,Plasma protein binding ,Biology ,Virus Replication ,Microbiology ,Cell Line ,Core domain ,Structure-Activity Relationship ,03 medical and health sciences ,Virology ,medicine ,Animals ,Humans ,Structure–activity relationship ,HIV Integrase Inhibitors ,Budesonide ,Research Articles ,030304 developmental biology ,0303 health sciences ,030306 microbiology ,HIV ,Rats ,3. Good health ,Integrase ,Cell nucleus ,medicine.anatomical_structure ,Fluocinolone Acetonide ,Viral replication ,Cell culture ,biology.protein ,Nuclear transport ,Research Article ,Protein Binding - Abstract
Human immunodeficiency virus (HIV) continues to be a major contributor to morbidity and mortality worldwide, particularly in developing nations where high cost and logistical issues severely limit the use of current HIV therapeutics. This, combined HIV's high propensity to develop resistance, means that new antiviral agents against novel targets are still urgently required. We previously identified novel anti‐HIV agents directed against the nuclear import of the HIV integrase (IN) protein, which plays critical roles in the HIV lifecycle inside the cell nucleus, as well as in transporting the HIV preintegration complex (PIC) into the nucleus. Here we investigate the structure activity relationship of a series of these compounds for the first time, including a newly identified anti‐IN compound, budesonide, showing that the extent of binding to the IN core domain correlates directly with the ability of the compound to inhibit IN nuclear transport in a permeabilised cell system. Importantly, compounds that inhibited the nuclear transport of IN were found to significantly decrease HIV viral replication, even in a dividing cell system. Significantly, budesonide or its analogue flunisolide, were able to effect a significant reduction in the presence of specific nuclear forms of the HIV DNA (2‐LTR circles), suggesting that the inhibitors work though blocking IN, and potentially PIC, nuclear import. The work presented here represents a platform for further development of these specific inhibitors of HIV replication with therapeutic and prophylactic potential.
- Published
- 2018
- Full Text
- View/download PDF
30. Nuclear drug delivery to target tumour cells
- Author
-
Kylie M. Wagstaff and David A. Jans
- Subjects
Genetic enhancement ,medicine.medical_treatment ,Active Transport, Cell Nucleus ,Antineoplastic Agents ,Context (language use) ,Targeted therapy ,Drug Delivery Systems ,Neoplasms ,Bystander effect ,Animals ,Humans ,Medicine ,Cytotoxic T cell ,Cell Nucleus ,Pharmacology ,business.industry ,Bystander Effect ,Genetic Therapy ,Cell nucleus ,medicine.anatomical_structure ,Drug delivery ,Immunology ,Cancer research ,Nuclear transport ,business ,Signal Transduction - Abstract
Cancer remains one of the leading causes of death worldwide. Although enticing, the concept of a chemotherapeutic treatment directed towards a single target that kills tumour cells, without any harmful side effects or death of neighbouring cells is probably naïve, due to the fact that tumour cells arise from normal cells and share many common biological features with them. Various means to damage/destroy tumour cells preferentially have been developed, but as yet, none are truly selective. However, by combining numerous tumour-specific/-enhanced targeting signals into a single modular multifaceted approach, it may prove possible some time in the future to achieve the desired outcome, without any unwanted bystander effects, with the delivery of cytotoxic drugs/DNA directly to the nucleus specifically within tumour cells of great interest in this context. To achieve this, modules such as the tumour-specific nuclear targeting signal of the chicken anemia virus Apoptin protein represent exciting possibilities. The present review discusses the question of nuclear delivery of bioactive molecules and its associated problems, as well as recent progress towards the development of tumour-specific modular recombinant transporters as viable anti-cancer therapeutics. In particular we focus upon the incorporation of tumour cell-selective nuclear targeting into these systems as a means to deliver cytotoxic genes or drugs to the very heart of tumour cells.
- Published
- 2009
- Full Text
- View/download PDF
31. Importins and Beyond: Non-Conventional Nuclear Transport Mechanisms
- Author
-
Kylie M. Wagstaff and David A. Jans
- Subjects
Cytoplasm ,Nuclear Localization Signals ,Active Transport, Cell Nucleus ,Importin ,Karyopherins ,Biology ,Biochemistry ,Structural Biology ,Genetics ,medicine ,Animals ,Humans ,Nuclear pore ,Nuclear export signal ,Molecular Biology ,Cell Nucleus ,Cell Biology ,biochemical phenomena, metabolism, and nutrition ,Cell biology ,Nuclear Pore Complex Proteins ,enzymes and coenzymes (carbohydrates) ,Cell nucleus ,medicine.anatomical_structure ,bacteria ,Nucleoporin ,Nuclear transport ,Nuclear localization sequence - Abstract
The movement of proteins between the cytoplasm and the nucleus conventionally involves the recognition of nuclear targeting signals by members of the importin (Imp) superfamily of nuclear transporters, followed by translocation through the nuclear envelope-embedded nuclear pore complexes (NPCs). It is becoming increasingly apparent, however, that distinct alternative pathways for nuclear transport exist and are relatively abundant. This review examines several of these novel pathways, including facilitation of Imp-dependent transport by microtubule motors, and Imp-independent pathways involving either other transport molecules such as the calcium-binding protein calmodulin or through direct binding to the components of the NPC. The existence of these pathways and the fact that many proteins appear to possess separate Imp-dependent and -independent nuclear import mechanisms ensure that the cell can function under conditions in which Imp-dependent transport is inhibited and/or modulate the efficiency of Imp-dependent transport itself, according to the need.
- Published
- 2009
- Full Text
- View/download PDF
32. The C-terminus of apoptin represents a unique tumor cell-enhanced nuclear targeting module
- Author
-
Gualtiero Alvisi, David A. Jans, Henna Veera Kuusisto, and Kylie M. Wagstaff
- Subjects
Cancer Research ,Viral protein ,viruses ,Molecular Sequence Data ,Nuclear Localization Signals ,Context (language use) ,nuclear transport ,Biology ,medicine.disease_cause ,Drug Delivery Systems ,medicine ,Humans ,NLS ,antitumor activity ,Amino Acid Sequence ,Phosphorylation ,Nuclear export signal ,Osteosarcoma ,Microscopy, Confocal ,medicine.disease ,Virology ,Cell biology ,tumor specific phosphorylation ,Cell nucleus ,medicine.anatomical_structure ,Oncology ,Capsid Proteins ,Nuclear transport - Abstract
Chicken anemia virus viral protein 3 (VP3 or apoptin) localizes more efficiently in the nucleus of transformed than nontransformed cells. Although previous studies implicate the C-terminus of apoptin as being responsible, the molecular basis is controversial, and the extent to which altered nuclear transport efficiency in tumor cells may influence VP3 differential targeting unclear. Here we establish that the C-terminus of VP3 (residues 74-121), out of the context of the full-length protein, is indeed sufficient for tumor cell-enhanced nuclear targeting through phosphoinhibition of VP3 (74-121)-mediated nuclear export occurring exclusively in tumor cells. Importantly, we show that VP3 (74-121) is unique in showing tumor cell-enhanced nuclear targeting in that other NLS-containing proteins fail to show differential localization in human osteosarcoma cells compared to their normal isogenic counterparts. Thus, the C-terminus of VP3 represents a unique tumor cell-enhanced nuclear targeting module with potential application in tumor cell-specific drug delivery.
- Published
- 2008
- Full Text
- View/download PDF
33. The N-Terminal Basic Domain of the HIV-1 Matrix Protein Does Not Contain a Conventional Nuclear Localization Sequence But Is Required for DNA Binding and Protein Self-Association
- Author
-
Kylie M. Wagstaff, Anna C. Hearps, David A. Jans, and Sabine C. Piller
- Subjects
Viral matrix protein ,HIV Antigens ,Nuclear Localization Signals ,RNA ,DNA ,Importin ,Biology ,gag Gene Products, Human Immunodeficiency Virus ,Biochemistry ,Molecular biology ,Fusion protein ,Cell Line ,Rats ,Cell biology ,chemistry.chemical_compound ,chemistry ,Complementary DNA ,COS Cells ,Chlorocebus aethiops ,Animals ,Humans ,Nuclear transport ,Nuclear localization sequence ,Protein Binding - Abstract
The HIV p17 or matrix (MA) protein has long been implicated in the process of nuclear import of the HIV genome and thus the ability of the virus to infect nondividing cells such as macrophages. While it has been demonstrated that MA is not absolutely required for this process, debate continues to surround the subcellular targeting properties of MA and its potential contribution to nuclear import of the HIV cDNA. Through the use of in vitro techniques we have determined that, despite the ability of MA to interact with importins, the full-length protein fails to enter the nucleus of cells. While MA does contain a region of basic amino acids within its N-terminus which can confer nuclear accumulation of a fusion protein, we show that this is due to nuclear retention mediated by DNA binding and does not represent facilitated import. Importantly, we show that the 26 KK residues of MA, previously thought to be part of a nuclear localization sequence, are absolutely required for a number of MA's functions including its ability to bind DNA and RNA and its propensity to form high-order multimers/protein aggregates. The results presented here indicate that the N-terminal basic domain of MA does not appear likely to play a role in HIV cDNA nuclear import; rather this region appears to be a crucial structural and functional motif whose integrity is required for a number of other roles performed by MA during viral infection.
- Published
- 2008
- Full Text
- View/download PDF
34. Nucleocytoplasmic transport of DNA: enhancing non-viral gene transfer
- Author
-
David A. Jans and Kylie M. Wagstaff
- Subjects
Genetics ,Genes, Viral ,Genetic enhancement ,Cell Membrane ,Active Transport, Cell Nucleus ,Gene Transfer Techniques ,DNA ,Cell Biology ,Oncogenicity ,Biology ,Gene delivery ,Biochemistry ,Cell biology ,chemistry.chemical_compound ,chemistry ,Nucleocytoplasmic Transport ,Animals ,Humans ,Vector (molecular biology) ,Nuclear transport ,Molecular Biology ,Gene - Abstract
Gene therapy, the correction of dysfunctional or deleted genes by supplying the lacking component, has long been awaited as a means to permanently treat or reverse many genetic disorders. To achieve this, therapeutic DNA must be delivered to the nucleus of cells using a safe and efficient delivery vector. Although viral-based vectors have been utilized extensively due to their innate ability to deliver DNA to intact cells, safety considerations, such as pathogenicity, oncogenicity and the stimulation of an immunological response in the host, remain problematical. There has, however, been much progress in the development of safe non-viral gene-delivery vectors, although they remain less efficient than the viral counterparts. The major limitations of non-viral gene transfer reside in the fact that it must be tailored to overcome the intracellular barriers to DNA delivery that viruses already master, including the cellular and nuclear membranes. In particular, nuclear transport of the therapeutic DNA is known to be the rate-limiting step in the gene-delivery process. Despite this, much progress had been made in recent years in developing novel means to overcome these barriers and efficiently deliver DNA to the nuclei of intact cells. This review focuses on the nucleocytoplasmic delivery of DNA and mechanisms to enhance to non-viral-mediated gene transfer.
- Published
- 2007
- Full Text
- View/download PDF
35. Quantitative analysis of localization and nuclear aggregate formation induced by GFP-lamin A mutant proteins in living HeLa cells
- Author
-
David A. Jans, Stefan Hubner, J.E. Eam, and Kylie M. Wagstaff
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,animal structures ,Recombinant Fusion Proteins ,Green Fluorescent Proteins ,Mutant ,Active Transport, Cell Nucleus ,Mutation, Missense ,Laminopathy ,Biology ,Biochemistry ,Histones ,Progeria ,medicine ,Humans ,Intermediate filament ,Molecular Biology ,Nuclear Lamina ,integumentary system ,Wild type ,Cell Biology ,Lamin Type A ,medicine.disease ,Progerin ,Molecular biology ,Fusion protein ,Cell biology ,Microscopy, Fluorescence ,Mutagenesis ,embryonic structures ,Lamin ,HeLa Cells - Abstract
Although A-type lamins are ubiquitously expressed, their role in the tissue-specificity of human laminopathies remains enigmatic. In this study, we generate a series of transfection constructs encoding missense lamin A mutant proteins fused to green fluorescent protein and investigate their subnuclear localization using quantitative live cell imaging. The mutant constructs used included the laminopathy-inducing lamin A rod domain mutants N195K, E358K, M371K, R386K, the tail domain mutants G465D, R482L, and R527P, and the Hutchinson-Gilford progeria syndrome-causing deletion mutant, progerin (LaA delta50). All mutant derivatives induced nuclear aggregates, except for progerin, which caused a more lobulated phenotype of the nucleus. Quantitative analysis revealed that the frequency of nuclear aggregate formation was significantly higher (two to four times) for the mutants compared to the wild type, although the level of lamin fusion proteins within nuclear aggregates was not. The distribution of endogenous A-type lamins was altered by overexpression of the lamin A mutants, coexpression experiments revealing that aberrant localization of the N195K and R386K mutants had no effect on the subnuclear distribution of histones H2A or H2B, or on nuclear accumulation of H2A overexpressed as a DsRed2 fusion protein. The GFP-lamin fusion protein-expressing constructs will have important applications in the future, enabling live cell imaging of nuclear processes involving lamins and how this may relate to the pathogenesis of laminopathies.
- Published
- 2006
- Full Text
- View/download PDF
36. Fatty Acid-binding Proteins 1 and 2 Differentially Modulate the Activation of Peroxisome Proliferator-activated Receptor α in a Ligand-selective Manner
- Author
-
Maria Lourdes Regina Hughes, David A. Jans, Martin J. Scanlon, Tony Velkov, Kylie M. Wagstaff, Rahul Chandrakant Patil, Christopher J.H. Porter, Nigel W. Bunnett, Bonan Liu, and Michelle L. Halls
- Subjects
Peroxisome proliferator-activated receptor ,Importin ,Plasma protein binding ,Calorimetry ,Karyopherins ,Fatty Acid-Binding Proteins ,Ligands ,Biochemistry ,Fatty acid-binding protein ,Drug Delivery Systems ,Chlorocebus aethiops ,Protein Interaction Mapping ,Fluorescence Resonance Energy Transfer ,Animals ,Humans ,PPAR alpha ,Receptor ,Molecular Biology ,chemistry.chemical_classification ,Cell Nucleus ,biology ,Biological Transport ,Molecular Bases of Disease ,Cell Biology ,Lipids ,Cell biology ,Nuclear receptor ,chemistry ,Gene Expression Regulation ,Chaperone (protein) ,Drug Design ,COS Cells ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Nuclear localization sequence ,Plasmids ,Protein Binding - Abstract
Nuclear hormone receptors (NHRs) regulate the expression of proteins that control aspects of reproduction, development and metabolism, and are major therapeutic targets. However, NHRs are ubiquitous and participate in multiple physiological processes. Drugs that act at NHRs are therefore commonly restricted by toxicity, often at nontarget organs. For endogenous NHR ligands, intracellular lipid-binding proteins, including the fatty acid-binding proteins (FABPs), can chaperone ligands to the nucleus and promote NHR activation. Drugs also bind FABPs, raising the possibility that FABPs similarly regulate drug activity at the NHRs. Here, we investigate the ability of FABP1 and FABP2 (intracellular lipid-binding proteins that are highly expressed in tissues involved in lipid metabolism, including the liver and intestine) to influence drug-mediated activation of the lipid regulator peroxisome proliferator-activated receptor (PPAR) α. We show by quantitative fluorescence imaging and gene reporter assays that drug binding to FABP1 and FABP2 promotes nuclear localization and PPARα activation in a drug- and FABP-dependent manner. We further show that nuclear accumulation of FABP1 and FABP2 is dependent on the presence of PPARα. Nuclear accumulation of FABP on drug binding is driven largely by reduced nuclear egress rather than an increased rate of nuclear entry. Importin binding assays indicate that nuclear access occurs via an importin-independent mechanism. Together, the data suggest that specific drug-FABP complexes can interact with PPARα to effect nuclear accumulation of FABP and NHR activation. Because FABPs are expressed in a regionally selective manner, this may provide a means to tailor the patterns of NHR drug activation in a tissue-specific manner. Background: Fatty acid-binding proteins (FABPs) chaperone intracellular transport of lipophilic ligands. Results: FABP1 and FABP2 differentially promote drug activation of peroxisome proliferator-activated receptor α (PPARα) via ligand-dependent protein-protein interactions. Conclusion: Drug activation of PPARα is regulated by the presence of different FABPs. Significance: FABPs may act in a tissue-specific manner to enhance the selectivity of PPARα agonists.
- Published
- 2014
37. Nuclear localization of the dystrophin-associated protein α-dystrobrevin through importin α2/β1 is critical for interaction with the nuclear lamina/maintenance of nuclear integrity
- Author
-
David A. Jans, Areli Aguilar, Rocío Suárez-Sánchez, Kylie M. Wagstaff, Bulmaro Cisneros, and Samuel Zinker
- Subjects
alpha Karyopherins ,Blotting, Western ,Molecular Sequence Data ,Nuclear Localization Signals ,Active Transport, Cell Nucleus ,Fluorescent Antibody Technique ,Importin ,Real-Time Polymerase Chain Reaction ,Biochemistry ,Dystrophin ,Immunoenzyme Techniques ,Mice ,Genetics ,medicine ,Animals ,Humans ,Immunoprecipitation ,Amino Acid Sequence ,RNA, Messenger ,Nuclear protein ,Molecular Biology ,Cells, Cultured ,Cell Proliferation ,Cell Nucleus ,Microscopy, Confocal ,Nuclear Lamina ,Sequence Homology, Amino Acid ,Chemistry ,Reverse Transcriptase Polymerase Chain Reaction ,Cell Cycle ,Nuclear Proteins ,Flow Cytometry ,beta Karyopherins ,Dystrophin-associated protein ,Cell biology ,Cell nucleus ,medicine.anatomical_structure ,Dystrophin-Associated Proteins ,Mutation ,Mutagenesis, Site-Directed ,Nuclear lamina ,Nuclear transport ,Nuclear localization sequence ,Lamin ,Biotechnology ,Protein Binding - Abstract
Although α-dystrobrevin (DB) is assembled into the dystrophin-associated protein complex, which is central to cytoskeletal organization, it has also been found in the nucleus. Here we delineate the nuclear import pathway responsible for nuclear targeting of α-DB for the first time, together with the importance of nuclear α-DB in determining nuclear morphology. We map key residues of the nuclear localization signal of α-DB within the zinc finger domain (ZZ) using various truncated versions of the protein, and site-directed mutagenesis. Pulldown, immunoprecipitation, and AlphaScreen assays showed that the importin (IMP) α2/β1 heterodimer interacts with high affinity with the ZZ domain of α-DB. In vitro nuclear import assays using antibodies to specific importins, as well as in vivo studies using siRNA or a dominant negative importin construct, confirmed the key role of IMPα2/β1 in α-DB nuclear translocation. Knockdown of α-DB expression perturbed cell cycle progression in C2C12 myoblasts, with decreased accumulation of cells in S phase and, significantly, altered localization of lamins A/C, B1, and B2 with accompanying gross nuclear morphology defects. Because α-DB interacts specifically with lamin B1 in vivo and in vitro, nuclear α-DB would appear to play a key role in nuclear shape maintenance through association with the nuclear lamina.
- Published
- 2014
38. Nuclear Protein Import: Methods
- Author
-
Kylie M. Wagstaff and David A. Jans
- Subjects
Cytoplasm ,Live cell imaging ,Fluorescence recovery after photobleaching ,Nuclear protein ,Nuclear transport ,Biology ,Transcription factor ,Nuclear localization sequence ,Transport protein ,Cell biology - Abstract
Protein synthesis takes place predominantly in the cytoplasm, meaning that proteins that are needed in the nuclear compartment, such as those that control gene transcription, have to be transported from the cytoplasm to the nucleus. Analysis of the regulation of nuclear import, which is central to cell responses to signalling pathways, and stress responses such as viral infection, requires dynamic experimental systems able to provide quantitative kinetic information. This article describes an in vitro reconstituted system as well as quantitative live cell imaging approaches, including the technique of fluorescence recovery after photobleaching, that enable the rate and extent of nuclear import to be quantitatively determined, and assist mechanistic studies with respect to the nuclear transporters and targeting signals involved. This is critical to a full understanding of the importance of nuclear trafficking in biological systems. Key Concepts: The regulation of nuclear transport is critical to transcriptional control in response to cellular and developmental signals, viral infection, etc. Using quantitative confocal laser scanning microscopy (CLSM) can enable small changes in nuclear localisation to be identified. Systems to reconstitute nuclear transport in vitro can enable the delineation of nuclear transport mechanisms. The technique of fluorescence recovery after photobleaching can be used to determine nuclear transport kinetics in the living cell. Quantitative techniques to examine nuclear transport allow for sequences and mechanisms underpinning regulated nucleocytoplasmic protein transport to be dissected. Keywords: nuclear transport; nuclear envelope; transcription factors; in vitro reconstituted system; fluorescence recovery after photobleaching
- Published
- 2014
- Full Text
- View/download PDF
39. A nuclear transport inhibitor that modulates the unfolded protein response and provides in vivo protection against lethal dengue virus infection
- Author
-
Caroline A. Hick, David A. Jans, Kylie M. Wagstaff, Adam J. Lopez-Denman, Patrick M. Sexton, Satoru Watanabe, Subhash G. Vasudevan, Belinda Maher, Jason M. Mackenzie, Chunxiao Wang, Johanna E. Fraser, and Wing K K Chan
- Subjects
Fenretinide ,Active Transport, Cell Nucleus ,Tretinoin ,Dengue virus ,Viral Nonstructural Proteins ,medicine.disease_cause ,Virus Replication ,Antiviral Agents ,Cell Line ,Dengue ,Mice ,eIF-2 Kinase ,In vivo ,medicine ,Immunology and Allergy ,Animals ,Humans ,Protein kinase A ,EIF-2 kinase ,biology ,Endoplasmic reticulum ,Dengue Virus ,Virology ,Transport protein ,Disease Models, Animal ,Protein Transport ,Infectious Diseases ,biology.protein ,Unfolded protein response ,Unfolded Protein Response ,Carrier Proteins ,Ex vivo ,Protein Binding ,Signal Transduction - Abstract
Background Dengue virus (DENV) is estimated to cause 390 million infections each year, but there is no licensed vaccine or therapeutic currently available. Methods We describe a novel, high-throughput screen to identify compounds inhibiting the interaction between DENV nonstructural protein 5 and host nuclear transport proteins. We document the antiviral properties of a lead compound against all 4 serotypes of DENV, antibody-dependent enhanced (ADE) infection, and ex vivo and in vivo DENV infections. In addition, we use quantitative reverse-transcription polymerase chain reaction to examine cellular effects upon compound addition. Results We identify N-(4-hydroxyphenyl) retinamide (4-HPR) as effective in protecting against DENV-1-4 and DENV-1 ADE infections, with 50% effective concentrations in the low micromolar range. 4-HPR but not the closely related N-(4-methoxyphenyl) retinamide (4-MPR) could reduce viral RNA levels and titers when applied to an established infection. 4-HPR but not 4-MPR was found to specifically upregulate the protein kinase R-like endoplasmic reticulum kinase arm of the unfolded protein response. Strikingly, 4-HPR but not 4-MPR restricted infection in peripheral blood mononuclear cells and in a lethal ADE-infection mouse model. Conclusions 4-HPR is a novel antiviral that modulates the unfolded protein response, effective against DENV1-4 at concentrations achievable in the plasma in a clinical setting, and provides protection in a lethal mouse model.
- Published
- 2014
40. Overlapping binding sites for importin β1 and suppressor of fused (SuFu) on glioma-associated oncogene homologue 1 (Gli1) regulate its nuclear localization
- Author
-
Rebecca G. Davies, Gurpreet Kaur, Katarzyna Gajewska, Anette Szczepny, Manisha M Dias, Kate L Loveland, Chunxiao Wang, Kylie M. Wagstaff, Jennifer D. Ly-Huynh, and David A. Jans
- Subjects
Recombinant Fusion Proteins ,Importin ,Adenocarcinoma ,Biochemistry ,Binding, Competitive ,Zinc Finger Protein GLI1 ,GLI1 ,RNA interference ,Chlorocebus aethiops ,Animals ,Humans ,Protein Interaction Domains and Motifs ,Binding site ,RNA, Small Interfering ,Molecular Biology ,Cell Nucleus ,Binding Sites ,integumentary system ,biology ,Cell Biology ,beta Karyopherins ,Fusion protein ,Peptide Fragments ,Transport protein ,Neoplasm Proteins ,Repressor Proteins ,Luminescent Proteins ,Protein Transport ,COS Cells ,biology.protein ,RNA Interference ,Nuclear transport ,Nuclear localization sequence ,HeLa Cells ,Transcription Factors - Abstract
A key factor in oncogenesis is the transport into the nucleus of oncogenic signalling molecules, such as Gli1 (glioma-associated oncogene homologue 1), the central transcriptional activator in the Hedgehog signalling pathway. Little is known, however, how factors such as Gli are transported into the nucleus and how this may be regulated by interaction with other cellular factors, such as the negative regulator suppressor of fused (SuFu). In the present study we show for the first time that nuclear entry of Gli1 is regulated by a unique mechanism through mutually exclusive binding by its nuclear import factor Impβ1 (importin β1) and SuFu. Using quantitative live mammalian cell imaging, we show that nuclear accumulation of GFP–Gli1 fusion proteins, but not of a control protein, is specifically inhibited by co-expression of SuFu. Using a direct binding assay, we show that Impβ1 exhibits a high nanomolar affinity to Gli1, with specific knockdown of Impβ1 expression being able to inhibit Gli1 nuclear accumulation, thus implicating Impβ1 as the nuclear transporter for Gli1 for the first time. SuFu also binds to Gli1 with a high nanomolar affinity, intriguingly being able to compete with Impβ1 for binding to Gli1, through the fact that the sites for SuFu and Impβ1 binding overlap at the Gli1 N-terminus. The results indicate for the first time that the relative intracellular concentrations of SuFu and Impβ1 are likely to determine the localization of Gli1, with implications for its action in cancer, as well as in developmental systems.
- Published
- 2014
41. Investigating dengue virus nonstructural protein 5 (NS5) nuclear import
- Author
-
Johanna E, Fraser, Stephen M, Rawlinson, Chunxiao, Wang, David A, Jans, and Kylie M, Wagstaff
- Subjects
Cell Nucleus ,Ivermectin ,Reverse Transcriptase Polymerase Chain Reaction ,Active Transport, Cell Nucleus ,Fluorescent Antibody Technique ,Viral Plaque Assay ,Dengue Virus ,Viral Load ,Viral Nonstructural Proteins ,Transfection ,Aedes ,COS Cells ,Chlorocebus aethiops ,Image Processing, Computer-Assisted ,Animals ,Biotinylation ,Vero Cells ,Protein Binding - Abstract
Dengue virus (DENV) nonstructural protein 5 (NS5) plays a central role in viral replication in the cytoplasm of infected cells. Despite this, NS5 is predominantly located in the nucleus of infected cells where it is thought to play a role in suppression of the host antiviral response. We have investigated the nuclear localization of NS5 using immunofluorescent staining for NS5 in infected cells, showing that NS5 nuclear localization is significantly inhibited by Ivermectin, a general inhibitor of nuclear transport mediated by the cellular nuclear transport proteins importin α/β (IMPα/β). Experiments in living mammalian cells transfected to express green fluorescent protein (GFP)-tagged NS5 protein confirm that NS5 is predominantly nuclear and that this localization is inhibited by Ivermectin, demonstrating that NS5 contains an Ivermectin-sensitive IMPα/β-recognized nuclear localization signal [Pryor et al. Traffic 8:795-807, 2007]. Consistent with this observation, mutation of critical residues within the nuclear localization signal (the A2 mutant; [Pryor et al. Traffic 8:795-807, 2007]) results in an 80 % reduction in nuclear localization of NS5. Finally we demonstrate direct, high-affinity binding of NS5 to IMPα/β using an AlphaScreen protein-protein binding assay.
- Published
- 2014
42. Investigating Dengue Virus Nonstructural Protein 5 (NS5) Nuclear Import
- Author
-
Chunxiao Wang, Johanna E. Fraser, Stephen M. Rawlinson, Kylie M. Wagstaff, and David A. Jans
- Subjects
COS cells ,Chemistry ,viruses ,virus diseases ,Importin ,Dengue virus ,medicine.disease_cause ,Virology ,Cell biology ,Green fluorescent protein ,Viral replication ,Cytoplasm ,medicine ,Nuclear transport ,Nuclear localization sequence - Abstract
Dengue virus (DENV) nonstructural protein 5 (NS5) plays a central role in viral replication in the cytoplasm of infected cells. Despite this, NS5 is predominantly located in the nucleus of infected cells where it is thought to play a role in suppression of the host antiviral response. We have investigated the nuclear localization of NS5 using immunofluorescent staining for NS5 in infected cells, showing that NS5 nuclear localization is significantly inhibited by Ivermectin, a general inhibitor of nuclear transport mediated by the cellular nuclear transport proteins importin α/β (IMPα/β). Experiments in living mammalian cells transfected to express green fluorescent protein (GFP)-tagged NS5 protein confirm that NS5 is predominantly nuclear and that this localization is inhibited by Ivermectin, demonstrating that NS5 contains an Ivermectin-sensitive IMPα/β-recognized nuclear localization signal [Pryor et al. Traffic 8:795-807, 2007]. Consistent with this observation, mutation of critical residues within the nuclear localization signal (the A2 mutant; [Pryor et al. Traffic 8:795-807, 2007]) results in an 80 % reduction in nuclear localization of NS5. Finally we demonstrate direct, high-affinity binding of NS5 to IMPα/β using an AlphaScreen protein-protein binding assay.
- Published
- 2014
- Full Text
- View/download PDF
43. Nuclear import and export inhibitors alter capsid protein distribution in mammalian cells and reduce Venezuelan Equine Encephalitis Virus replication
- Author
-
Kylie M. Wagstaff, Chelsea Pinkham, Aarthi Narayanan, David A. Jans, Lindsay Lundberg, Alan Baer, Kylene Kehn-Hall, and Moushimi Amaya
- Subjects
alpha Karyopherins ,Virus Cultivation ,Viral protein ,Macromolecular Substances ,viruses ,Active Transport, Cell Nucleus ,Receptors, Cytoplasmic and Nuclear ,Importin ,Biology ,Astrocytoma ,Karyopherins ,medicine.disease_cause ,Virus Replication ,environment and public health ,Encephalitis Virus, Venezuelan Equine ,Capsid ,Cytopathogenic Effect, Viral ,Transcription (biology) ,Virology ,Cell Line, Tumor ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,RNA, Small Interfering ,Nuclear export signal ,Vero Cells ,Pharmacology ,Cell Nucleus ,Gene knockdown ,Ivermectin ,beta Karyopherins ,Cell Compartmentation ,Mifepristone ,Venezuelan equine encephalitis virus ,Fatty Acids, Unsaturated ,RNA Interference ,Nuclear transport - Abstract
Targeting host responses to invading viruses has been the focus of recent antiviral research. Venezuelan Equine Encephalitis Virus (VEEV) is able to modulate host transcription and block nuclear trafficking at least partially due to its capsid protein forming a complex with the host proteins importin α/β1 and CRM1. We hypothesized that disrupting the interaction of capsid with importin α/β1 or the interaction of capsid with CRM1 would alter capsid localization, thereby lowering viral titers in vitro. siRNA mediated knockdown of importin α, importin β1, and CRM1 altered capsid localization, confirming their role in modulating capsid trafficking. Mifepristone and ivermectin, inhibitors of importin α/β-mediated import, were able to reduce nuclear-associated capsid, while leptomycin B, a potent CRM1 inhibitor, confined capsid to the nucleus. In addition to altering the level and distribution of capsid, the three inhibitors were able to reduce viral titers in a relevant mammalian cell line with varying degrees of efficacy. The inhibitors were also able to reduce the cytopathic effects associated with VEEV infection, hinting that nuclear import inhibitors may be protecting cells from apoptosis in addition to disrupting the function of an essential viral protein. Our results confirm that VEEV uses host importins and exportins during part of its life cycle. Further, it suggests that temporarily targeting host proteins that are hijacked for use by viruses is a viable antiviral therapy.
- Published
- 2013
44. The BRCA1-binding protein BRAP2 can act as a cytoplasmic retention factor for nuclear and nuclear envelope-localizing testicular proteins
- Author
-
David A. Jans, Kylie M. Wagstaff, Kate L Loveland, Rebecca G. Davies, and Eileen A. McLaughlin
- Subjects
Male ,NuMA1 ,Nuclear Envelope ,Ubiquitin-Protein Ligases ,Cell Cycle Proteins ,Nerve Tissue Proteins ,Biology ,03 medical and health sciences ,Mice ,Nuclear Matrix-Associated Proteins ,Chlorocebus aethiops ,Testis ,Animals ,Humans ,Nuclear protein ,BRAP2 ,Molecular Biology ,Nuclear receptor co-repressor 1 ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,0303 health sciences ,BRCA1 Protein ,SYNE2 ,Binding protein ,030302 biochemistry & molecular biology ,Microfilament Proteins ,High Mobility Group Proteins ,Nuclear Proteins ,Antigens, Nuclear ,Cell Biology ,Subcellular localization ,Cell biology ,Chromatin ,Cytoplasmic retention factor ,Protein Transport ,Nuclear transport ,COS Cells ,HMG20A ,Nuclear lamina ,Nuclear localization sequence ,Protein Binding - Abstract
Regulation of nuclear protein import is central to many cellular processes such as development, with a key mechanism being factors that retain cargoes in the cytoplasm that normally localize in the nucleus. The breast cancer antigen BRCA1-binding protein BRAP2 has been reported as a novel negative regulator of nuclear import of various nuclear localization signal (NLS)-containing viral and cellular proteins, but although implicated in differentiation pathways and highly expressed in tissues including testis, the gamut of targets for BRAP2 action in a developmental context is unknown. As a first step towards defining the BRAP2 interactome, we performed a yeast-2-hybrid screen to identify binding partners of BRAP2 in human testis. Here we report characterization for the first time of three of these: the high mobility group (HMG)-box-domain-containing chromatin component HMG20A, nuclear mitotic apparatus protein NuMA1 and synaptic nuclear envelope protein SYNE2. Co-immunoprecipitation experiments indicate association of BRAP2 with HMG20A, NuMA1, and SYNE2 in testis, underlining the physiological relevance of the interactions, with immunohistochemistry showing that where BRAP2 is co-expressed with HMG20A and NuMA1, both are present in the cytoplasm, in contrast to their nuclear localization in other testicular cell types. Importantly, quantitative confocal microscopic analysis of cultured cells indicates that ectopic expression of BRAP2 inhibits nuclear localization of HMG20A and NuMA1, and prevents nuclear envelope accumulation of SYNE2, the first report of BRAP2 altering localization of a non-nuclear protein. These results imply for the first time that BRAP2 may have an important role in modulating subcellular localization during testicular development.
- Published
- 2013
45. Protein Transduction: Cell Penetrating Peptides and Their Therapeutic Applications
- Author
-
Kylie M. Wagstaff and David A. Jans
- Published
- 2012
- Full Text
- View/download PDF
46. Histone-mediated transduction as an efficient means for gene delivery
- Author
-
Kylie M. Wagstaff, David J. Tremethick, Dominic J. Glover, and David A. Jans
- Subjects
Recombinant Fusion Proteins ,Molecular Sequence Data ,Nuclear Localization Signals ,DNA, Recombinant ,Gene delivery ,Karyopherins ,Protein Engineering ,Cell Line ,Histones ,Histone H1 ,Transduction, Genetic ,Histone H2A ,Drug Discovery ,Chlorocebus aethiops ,medicine ,Histone H2B ,Genetics ,Animals ,Humans ,Amino Acid Sequence ,Molecular Biology ,Pharmacology ,Cell Nucleus ,biology ,Genetic Therapy ,Molecular biology ,Cell biology ,Rats ,Cell nucleus ,medicine.anatomical_structure ,Histone ,COS Cells ,biology.protein ,Molecular Medicine ,Nuclear transport ,Dimerization ,Nuclear localization sequence ,HeLa Cells ,Protein Binding - Abstract
Gene delivery into the nucleus of eukaryotic cells is inefficient, largely because of the significant barriers within the target cell of the plasma membrane and nuclear envelope. Recently, a group of basic proteins, including the HIV-1 Tat protein and the four core histones, have been shown to enter cells through a novel energy- and receptor-independent manner. Here, we show that engineered histone H2B proteins are able to mediate the efficient delivery of either green fluorescent protein or DNA into HeLa cells through the process of "Histone-Mediated Transduction" (HMT), with further enhancement achieved by utilizing a dimer of histones H2B and H2A. Subsequent nuclear delivery was accelerated approximately two-fold by the addition of an optimized nuclear localization signal to histone H2B, thereby increasing the affinity of interaction with components of the cellular nuclear import machinery, resulting in increased expression of a reporter gene. Further, we demonstrate that the domains responsible for this histone transduction are located in the N-terminal tail and globular regions of histone H2B. HMT represents a new, efficient, and technically non-demanding means to deliver DNA to the nucleus of intact cells, including embryonic stem cells, which has important applications in gene therapy and cancer therapeutics.
- Published
- 2007
47. Protein transduction: cell penetrating peptides and their therapeutic applications
- Author
-
Kylie M. Wagstaff and David A. Jans
- Subjects
Cell Membrane Permeability ,Biology ,Biochemistry ,DNA vaccination ,Cell Physiological Phenomena ,Transduction (genetics) ,Drug Delivery Systems ,Drug Discovery ,Vaccines, DNA ,Animals ,Humans ,Pharmacology ,Oligonucleotide ,Organic Chemistry ,Genetic transfer ,Cell Membrane ,Proteins ,Membrane transport ,Cell biology ,Protein Transport ,Drug delivery ,Cell-penetrating peptide ,Molecular Medicine ,Signal transduction ,Peptides - Abstract
Cell penetrating proteins or peptides (CPPs) have the ability to cross the plasma membranes of mammalian cells in an apparently energy- and receptor-independent fashion. Although there is much debate over the mechanism by which this "protein transduction" occurs, the ability of CPPs to translocate rapidly into cells is being exploited to deliver a broad range of therapeutics including proteins, DNA, antibodies, oligonucleotides, imaging agents and liposomes in a variety of situations and biological systems. The current review looks at the delivery of many such molecules by various CPPs, and their potential therapeutic application in a wide range of areas. CPP ability to deliver different cargoes in a relatively efficient and non-invasive manner has implications as far reaching as drug delivery, gene transfer, DNA vaccination and beyond. Although many questions remain to be answered and limitations on the use of CPPs exist, it is clear that this emerging technology has much to offer in a clinical setting.
- Published
- 2006
48. Nuclear import of the respiratory syncytial virus matrix protein is mediated by importin beta1 independent of importin alpha
- Author
-
Reena Ghildyal, Patricia Jans, Manisha M Dias, Kylie M. Wagstaff, Phillip G. Bardin, David A. Jans, Adeline Ho, and Chenoa Barton
- Subjects
alpha Karyopherins ,Molecular Sequence Data ,Active Transport, Cell Nucleus ,Importin ,Biology ,Biochemistry ,Viral Matrix Proteins ,Cell Line, Tumor ,medicine ,Importin-alpha ,Animals ,Amino Acid Sequence ,Nuclear protein ,Edetic Acid ,Chelating Agents ,Host cell membrane ,Cell Nucleus ,Viral matrix protein ,Nuclear Proteins ,beta Karyopherins ,Molecular biology ,Rats ,Respiratory Syncytial Viruses ,Cell nucleus ,medicine.anatomical_structure ,ran GTP-Binding Protein ,Ran ,Nuclear transport - Abstract
The matrix (M) protein of respiratory syncytial virus (RSV) plays an important role in virus assembly through specific interactions with RSV nucleocapsids and envelope glycoproteins in the cytoplasm as well as with the host cell membrane. We have previously shown that M localizes to the nucleus of infected cells at an early stage in the RSV infection cycle, where it may be instrumental in inhibiting host cell processes. The present study uses transient expression of M as well as a truncated green fluorescent protein (GFP) fusion derivative to show for the first time that M is able to localize in the nucleus in the absence of other RSV gene products, through the action of amino acids 110-183, encompassing the nucleic acid binding regions of the protein, that are sufficient to target GFP to the nucleus. Using native PAGE, ELISA-based binding assays, a novel Alphascreen assay, and an in vitro nuclear transport assay, we show that M is recognized directly by the importin beta1 nuclear import receptor, which mediates its nuclear import in concert with the guanine nucleotide-binding protein Ran. Retention of M in the nucleus through binding to nuclear components, probably mediated by the putative zinc finger domain of M, also contributes to M nuclear accumulation. This is the first report of the importin binding and nuclear import properties of a gene product from a negative sense RNA virus, with implications for the function of RSV M and possibly other viral M proteins in the nucleus of infected cells.
- Published
- 2005
49. Intramolecular masking of nuclear localization signals: analysis of importin binding using a novel AlphaScreen-based method
- Author
-
David A. Jans and Kylie M. Wagstaff
- Subjects
Viral protein ,viruses ,Recombinant Fusion Proteins ,Green Fluorescent Proteins ,Nuclear Localization Signals ,Biophysics ,Importin ,Biology ,Karyopherins ,medicine.disease_cause ,Biochemistry ,Sensitivity and Specificity ,Protein–protein interaction ,Green fluorescent protein ,medicine ,NLS ,Protein Isoforms ,Molecular Biology ,Cell Biology ,Molecular biology ,Fusion protein ,Cell biology ,Luminescent Measurements ,Capsid Proteins ,Nuclear transport ,Nuclear localization sequence ,Protein Binding - Abstract
Active nuclear import of proteins requires the recognition of a nuclear localization sequence (NLS) by members of the importin (IMP) family of proteins. We have developed a modified AlphaScreen-based assay able to estimate the solution binding affinities of such interactions using biotinylated IMPs and His6-tagged NLS-containing proteins. We describe this assay in detail as well as its application in documenting the phenomenon of intramolecular masking of NLSs using recombinant green fluorescent protein (GFP) fusion proteins containing sequences from the SV40 large tumor T antigen (T-ag). We also use it to examine, for the first time, IMP binding to the cancer cell-specific proapoptotic factor viral protein 3 (VP3) from the chicken anemia virus (CAV). High-affinity binding of the IMPalpha/beta heterodimer to the T-ag NLS was observed when the GFP tag was fused to its N terminus but not to its C terminus. Effects of flanking residues were also observed in GFP-T-ag fusion derivatives containing the Thr128 NLS-inactivating mutation, whereby the absence of flanking sequences N terminal to the T-ag NLS appeared to decrease the specificity of the mutation in terms of oblating IMPalpha/beta binding. IMPbeta, but not IMPalpha or the IMPalpha/beta heterodimer, was found to bind to CAV VP3 with high affinity. Interestingly, GFP-VP3(74-121) bound to IMPbeta with threefold higher affinity than the full-length protein, GFP-VP3(1-121), implying that the NLS is masked to a significant extent in the context of full-length protein. This may represent a regulatory mechanism to control nuclear import in a tumor cell-specific fashion.
- Published
- 2005
50. Nuclear Import of β-Dystroglycan Is Facilitated by Ezrin-Mediated Cytoskeleton Reorganization
- Author
-
Arturo Ortega, Dorothy H. Crouch, Bulmaro Cisneros, Kylie M. Wagstaff, A. Vásquez-Limeta, and David A. Jans
- Subjects
rho GTP-Binding Proteins ,lcsh:Medicine ,Actin Filaments ,Biochemistry ,environment and public health ,Muscular Dystrophies ,Mice ,0302 clinical medicine ,Ezrin ,Molecular Cell Biology ,Nuclear protein ,lcsh:Science ,Dystroglycans ,Cytoskeleton ,0303 health sciences ,Multidisciplinary ,beta Karyopherins ,Cellular Structures ,Cell biology ,Cell Motility ,Actin Cytoskeleton ,Neurology ,030220 oncology & carcinogenesis ,Medicine ,Cellular Types ,Research Article ,Signal Transduction ,Myoblasts, Skeletal ,Biophysics ,Active Transport, Cell Nucleus ,macromolecular substances ,Importin ,Biology ,Cell Line ,03 medical and health sciences ,Animals ,Biotinylation ,030304 developmental biology ,Cell Nucleus ,Muscle Cells ,lcsh:R ,Proteins ,Actin cytoskeleton ,Transmembrane Proteins ,Cytoskeletal Proteins ,lcsh:Q ,Nuclear transport ,Lamin ,Nuclear localization sequence - Abstract
The β-dystroglycan (β-DG) protein has the ability to target to multiple sites in eukaryotic cells, being a member of diverse protein assemblies including the transmembranal dystrophin-associated complex, and a nuclear envelope-localised complex that contains emerin and lamins A/C and B1. We noted that the importin α2/β1-recognised nuclear localization signal (NLS) of β-DG is also a binding site for the cytoskeletal-interacting protein ezrin, and set out to determine whether ezrin binding might modulate β-DG nuclear translocation for the first time. Unexpectedly, we found that ezrin enhances rather than inhibits β-DG nuclear translocation in C2C12 myoblasts. Both overexpression of a phosphomimetic activated ezrin variant (Ez-T567D) and activation of endogenous ezrin through stimulation of the Rho pathway resulted in both formation of actin-rich surface protrusions and significantly increased nuclear translocation of β-DG as shown by quantitative microscopy and subcellular fractionation/Western analysis. In contrast, overexpression of a nonphosphorylatable inactive ezrin variant (Ez-T567A) or inhibition of Rho signaling, decreased nuclear translocation of β-DG concomitant with a lack of cell surface protrusions. Further, a role for the actin cytoskeleton in ezrin enhancement of β-DG nuclear translocation was implicated by the observation that an ezrin variant lacking its actin-binding domain failed to enhance nuclear translocation of β-DG, while disruption of the actin cytoskeleton led to a reduction in β-DG nuclear localization. Finally, we show that ezrin-mediated cytoskeletal reorganization enhances nuclear translocation of the cytoplasmic but not the transmembranal fraction of β-DG. This is the first study showing that cytoskeleton reorganization can modulate nuclear translocation of β-DG, with the implication that β-DG can respond to cytoskeleton-driven changes in cell morphology by translocating from the cytoplasm to the nucleus to orchestrate nuclear processes in response to the functional requirements of the cell.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.