Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters

Publication year range
1.
Cell ; 176(4): 805-815.e8, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30639102

ABSTRACT

Early embryogenesis is accompanied by reductive cell divisions requiring that subcellular structures adapt to a range of cell sizes. The interphase nucleus and mitotic spindle scale with cell size through both physical and biochemical mechanisms, but control systems that coordinately scale intracellular structures are unknown. We show that the nuclear transport receptor importin α is modified by palmitoylation, which targets it to the plasma membrane and modulates its binding to nuclear localization signal (NLS)-containing proteins that regulate nuclear and spindle size in Xenopus egg extracts. Reconstitution of importin α targeting to the outer boundary of extract droplets mimicking cell-like compartments recapitulated scaling relationships observed during embryogenesis, which were altered by inhibitors that shift levels of importin α palmitoylation. Modulation of importin α palmitoylation in human cells similarly affected nuclear and spindle size. These experiments identify importin α as a conserved surface area-to-volume sensor that scales intracellular structures to cell size.


Subject(s)
Cell Division/physiology , alpha Karyopherins/metabolism , alpha Karyopherins/physiology , Active Transport, Cell Nucleus , Animals , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cell Size , Cytoplasm/metabolism , Lipoylation , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Ovum/cytology , Spindle Apparatus/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/metabolism
2.
PLoS Pathog ; 14(1): e1006823, 2018 01.
Article in English | MEDLINE | ID: mdl-29304174

ABSTRACT

Herpesviruses are large DNA viruses which depend on many nuclear functions, and therefore on host transport factors to ensure specific nuclear import of viral and host components. While some import cargoes bind directly to certain transport factors, most recruit importin ß1 via importin α. We identified importin α1 in a small targeted siRNA screen to be important for herpes simplex virus (HSV-1) gene expression. Production of infectious virions was delayed in the absence of importin α1, but not in cells lacking importin α3 or importin α4. While nuclear targeting of the incoming capsids, of the HSV-1 transcription activator VP16, and of the viral genomes were not affected, the nuclear import of the HSV-1 proteins ICP4 and ICP0, required for efficient viral transcription, and of ICP8 and pUL42, necessary for DNA replication, were reduced. Furthermore, quantitative electron microscopy showed that fibroblasts lacking importin α1 contained overall fewer nuclear capsids, but an increased proportion of mature nuclear capsids indicating that capsid formation and capsid egress into the cytoplasm were impaired. In neurons, importin α1 was also not required for nuclear targeting of incoming capsids, but for nuclear import of ICP4 and for the formation of nuclear capsid assembly compartments. Our data suggest that importin α1 is specifically required for the nuclear localization of several important HSV1 proteins, capsid assembly, and capsid egress into the cytoplasm, and may become rate limiting in situ upon infection at low multiplicity or in terminally differentiated cells such as neurons.


Subject(s)
Capsid Proteins/metabolism , Cell Nucleus/metabolism , Fibroblasts/virology , Herpesvirus 1, Human/physiology , Neurons/virology , Virus Assembly/genetics , alpha Karyopherins/physiology , Active Transport, Cell Nucleus/genetics , Animals , Capsid/metabolism , Cell Line , Cell Nucleus/virology , Cricetinae , Fibroblasts/metabolism , HEK293 Cells , HeLa Cells , Herpesvirus 1, Human/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , alpha Karyopherins/genetics
3.
Acta Biochim Biophys Sin (Shanghai) ; 51(3): 285-292, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30883648

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most aggressive cancers worldwide. Identification of the molecular mechanisms underlying the development and progression of HCC is particularly important. Here, we demonstrated the expression pattern, clinical significance, and function of Karyopherin α2 (KPNA2) in HCC. The expression of KPNA2 was upregulated in tumor tissue and negatively associated with the survival time, and a significant correlation between KPNA2 expression and aggressive clinical characteristics was established. Both in vitro and in vivo experiments demonstrated that knockdown of KPNA2 reduced migration and proliferation capacities of HCC cells, while over-expression of KPNA2 increased these malignant characteristics. The analysis of the Cancer Genome Atlas cohorts also reveals that high-KPNA2 expression is associated with poor outcome in multiple cancer types. In addition, gene sets enrichment analysis exhibited cell cycle and DNA replication as the top altered pathways in the high-KPNA2 expression group in HCC and other two cancer types. Overall, this study identified KPNA2 as a potential diagnostic and prognostic biomarker in HCC and other neoplasms, probably by regulating cell cycle and DNA replication.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , alpha Karyopherins/physiology , Adult , Aged , Animals , Carcinoma, Hepatocellular/mortality , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Humans , Liver Neoplasms/mortality , Mice , Middle Aged , Prognosis , Up-Regulation , alpha Karyopherins/genetics
4.
Biochem Biophys Res Commun ; 493(4): 1555-1559, 2017 12 02.
Article in English | MEDLINE | ID: mdl-28988109

ABSTRACT

In the absence of approved therapeutics, Zika virus (ZIKV)'s recent prolific outbreaks in the Americas, together with impacts on unborn fetuses of infected mothers, make it a pressing human health concern worldwide. Although a key player in viral replication in the infected host cell cytoplasm, ZIKV non-structural protein 5 (NS5) appears to contribute integrally to pathogenesis by localising in the host cell nucleus, in similar fashion to NS5 from Dengue virus (DENV). We show here for the first time that ZIKV NS5 is recognized with high nanomolar affinity by the host cell importin α/ß1 heterodimer, and that this interaction can be blocked by the novel DENV NS5 targeting inhibitor N-(4-hydroxyphenyl) retinamide (4-HPR). Importantly, we show that 4-HPR has potent anti-ZIKV activity at low µM concentrations. With an established safety profile for human use, 4-HPR represents an exciting possibility as an anti-ZIKV agent.


Subject(s)
Antiviral Agents/pharmacology , Fenretinide/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Zika Virus/drug effects , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Amino Acid Sequence , Animals , Chlorocebus aethiops , Conserved Sequence , Humans , Vero Cells , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/physiology , Virus Replication/drug effects , Zika Virus/genetics , Zika Virus/physiology , Zika Virus Infection/drug therapy , Zika Virus Infection/prevention & control , Zika Virus Infection/virology , alpha Karyopherins/physiology , beta Karyopherins/physiology
5.
J Proteome Res ; 14(4): 1739-51, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-25728791

ABSTRACT

Karyopherin alpha 2 (KPNA2) is overexpressed in various human cancers and is associated with cancer invasiveness and poor prognosis. Herein, to understand the essential role of KPNA2 protein complexes in cancer progression, we applied stable isotope labeling with amino acids in cell culture (SILAC)-based quantitative proteomic strategy combined with immunoprecipitation (IP) to investigate the differential KPNA2 protein complexes in lung adenocarcinoma cell lines with different invasiveness potentials. We found that 64 KPNA2-interaction proteins displayed a 2-fold difference in abundance between CL1-5 (high invasiveness) and CL1-0 (low invasiveness) cells. Pathway map analysis revealed that the formation of complexes containing KPNA2 and cytoskeleton-remodeling-related proteins, including actin, beta tubulin, tubulin heterodimers, vimentin, keratin 8, keratin 18, and plectin, was associated with cancer invasiveness. IP demonstrated that the levels of KPNA2-vimentin-pErk complexes were significantly higher in CL1-5 cells than in CL1-0 cells. The KPNA2-vimentin-pErk complex was also up-regulated in the advanced stage compared with the early-stage lung adenocarcinoma tissues. Importantly, the levels of pErk as well as cell migration ability were significantly reduced in KPNA2-knockdown cells; however, migration was restored by treatment with pErk phosphatase inhibitors. Collectively, our results demonstrate the usefulness of a SILAC-based proteomic strategy for identifying invasiveness-associated KPNA2 protein complexes and provide new insight into the KPNA2-mediated modulation of cell migration.


Subject(s)
Cell Movement/physiology , Lung Neoplasms/metabolism , Multiprotein Complexes/metabolism , Neoplasm Invasiveness/physiopathology , Signal Transduction/physiology , alpha Karyopherins/metabolism , Cell Movement/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Phosphorylation , Proteomics/methods , Signal Transduction/genetics , Vimentin/metabolism , alpha Karyopherins/physiology
6.
J Biol Chem ; 289(15): 10270-10275, 2014 Apr 11.
Article in English | MEDLINE | ID: mdl-24596094

ABSTRACT

Mature microRNAs (miRNAs), ∼ 22-nucleotide noncoding RNAs regulating target gene expression at the post-transcriptional level, have been recently shown to be transported into the nucleus where they modulate the biogenesis of other miRNAs or their own expression. However, the mechanism that governs the transport of mature miRNAs from cytoplasm to nucleus remains unknown. Here, we report that importin 8 (IPO8), a member of the karyopherin ß (also named the protein import receptor importin ß) family, plays a critical role in mediating the cytoplasm-to-nucleus transport of mature miRNAs. Specifically knocking down IPO8 but not other karyopherin ß family proteins via siRNA significantly decreases the nuclear transport of various known nucleus-enriched miRNAs without affecting their total cellular levels. IPO8-mediated nuclear transport of mature miRNAs is also dependent on the association of IPO8 with the Argonaute 2 (Ago2) complex. Cross-immunoprecipitation and Western blot analysis show that IPO8 is physically associated with Ago2. Knocking down IPO8 via siRNA markedly decreases the nuclear transport of Ago2 but does not affect the total cellular Ago2 level. Furthermore, dissociating the binding of miRNAs with Ago2 by trypaflavine strongly reduces the IPO8-mediated nuclear transport of miRNAs.


Subject(s)
Argonaute Proteins/metabolism , Cell Nucleus/metabolism , MicroRNAs/metabolism , alpha Karyopherins/physiology , Active Transport, Cell Nucleus , Animals , Cell Line , Cytoskeleton/metabolism , Gene Regulatory Networks , Mice , Microscopy, Fluorescence , Protein Binding , RNA, Small Interfering/metabolism , alpha Karyopherins/chemistry
7.
FASEB J ; 28(8): 3480-93, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24790034

ABSTRACT

Importin α proteins function as adaptors to connect a cargo protein and importin ß1 in the classical nuclear import pathway. Here we measure for the first time the stoichiometry of importins α2, α3, α4, and ß1 in primary cells corresponding to 2 successive stages of rat spermatogenesis: meiotic spermatocytes and haploid round spermatids. Importin α2 levels were more than 2-fold higher in spermatocytes than in spermatids, while importins α4 and ß1 levels did not differ significantly. We performed a comprehensive proteomics analysis to identify binding proteins in spermatocytes and spermatids using recombinant importin α2 and α4 proteins. Among the 100 candidate partners, 42 contained a strong classical nuclear localization signal (cNLS; score of>6 by cNLS Mapper), while 8 nuclear proteins lacked any cNLS. In addition, we developed a new strategy to predict which cargoes bind to importin α through the conserved C-terminal acidic domain (ARM repeats 9-10), and provided functional validation of a predicted importin α C-terminal binding segment in Senataxin and Smarca4. Evaluation of this set of candidate binding partners from spermatogenic cells using several bioinformatics approaches provides new evidence that individual importin αs may serve unique and nonredundant roles in mediating cellular differentiation.


Subject(s)
Active Transport, Cell Nucleus/physiology , Spermatogenesis/physiology , alpha Karyopherins/physiology , Amino Acid Motifs , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Cell Compartmentation , DNA Helicases/chemistry , Male , Meiosis , Mice , Molecular Sequence Data , Nuclear Proteins/chemistry , Pachytene Stage , Peptide Fragments/metabolism , Protein Binding , Protein Isoforms/physiology , Protein Structure, Tertiary , Proteomics , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/metabolism , Spermatids/metabolism , Spermatids/ultrastructure , Transcription Factors/chemistry , alpha Karyopherins/analysis , beta Karyopherins/analysis , beta Karyopherins/physiology
8.
Exp Cell Res ; 322(1): 159-67, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24275456

ABSTRACT

Pancreatic cancer is an aggressive malignancy and one of the leading causes of cancer deaths. The high mortality rate is mostly due to the lack of appropriate tools for early detection of the disease and a shortage of effective therapies. We have previously shown that karyopherin alpha 7 (KPNA7), the newest member of the alpha karyopherin family of nuclear import receptors, is frequently amplified and overexpressed in pancreatic cancer. Here, we report that KPNA7 expression is absent in practically all normal human adult tissues but elevated in several pancreatic cancer cell lines. Inhibition of KPNA7 expression in AsPC-1 and Hs700T pancreatic cancer cells led to a reduction in cell growth and decreased anchorage independent growth, as well as increased autophagy. The cell growth effects were accompanied by an induction of the cell cycle regulator p21 and a G1 arrest of the cell cycle. Interestingly, the p21 induction was caused by increased mRNA synthesis and not defective nuclear transport. These data strongly demonstrate that KPNA7 silencing inhibits the malignant properties of pancreatic cancer cells in vitro and thereby provide the first evidence on the functional role for KPNA7 in human cancer.


Subject(s)
Cell Transformation, Neoplastic/genetics , Pancreatic Neoplasms/pathology , alpha Karyopherins/physiology , Active Transport, Cell Nucleus , Adult , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Pancreatic Neoplasms/genetics , RNA Interference/physiology , RNA, Small Interfering/pharmacology , Receptors, Cytoplasmic and Nuclear/physiology , Tumor Cells, Cultured , alpha Karyopherins/antagonists & inhibitors
9.
Biochim Biophys Acta ; 1833(10): 2348-56, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23773962

ABSTRACT

The importin (IMP) superfamily of nuclear transport proteins is essential to key developmental pathways, including in the murine testis where expression of the 6 distinct IMPα proteins is highly dynamic. Present predominantly from the spermatocyte stage onwards, IMPα4 is unique in showing a striking nuclear localization, a property we previously found to be linked to maintenance of pluripotency in embryonic stem cells and to the cellular stress response in cultured cells. Here we examine the role of IMPα4 in vivo for the first time using a novel transgenic mouse model in which we overexpress an IMPα4-EGFP fusion protein from the protamine 1 promoter to recapitulate endogenous testicular germ cell IMPα4 expression in spermatids. IMPα4 overexpression did not affect overall fertility, testis morphology/weight or spermatogenic progression under normal conditions, but conferred significantly (>30%) increased resistance to oxidative stress specifically in the spermatid subpopulation expressing the transgene. Consistent with a cell-specific role for IMPα4 in protecting against oxidative stress, haploid germ cells from IMPα4 null mice were significantly (c. 30%) less resistant to oxidative stress than wild type controls. These results from two unique and complementary mouse models demonstrate a novel protective role for IMPα4 in stress responses specifically within haploid male germline cells, with implications for male fertility and genetic integrity.


Subject(s)
Active Transport, Cell Nucleus/genetics , Cell Nucleus/genetics , Germ Cells/metabolism , Oxidative Stress , Spermatids/metabolism , Spermatogenesis , Testis/metabolism , alpha Karyopherins/physiology , Animals , Blotting, Southern , Blotting, Western , Cell Differentiation , Cell Nucleus/metabolism , Cell Proliferation , DNA/genetics , Fertility , Flow Cytometry , Germ Cells/cytology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoenzyme Techniques , Immunoprecipitation , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Promoter Regions, Genetic/genetics , Protamines/genetics , Real-Time Polymerase Chain Reaction , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spermatids/cytology , Testis/cytology
10.
Kidney Int ; 85(3): 624-35, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24284509

ABSTRACT

Glomerular endothelial cells (GEnCs) contribute to renal injuries in IgA nephropathy (IgAN). Here we profiled microRNAs (miRNAs) in GEnCs treated with conditioned medium from human mesangial cells in vitro. Levels of miR-223 in GEnCs decreased after incubation with the medium prepared with pIgA from patients with glomerular endothelial proliferation and were also decreased in the glomerular tissues of patients with glomerular endothelial proliferation. Mesangial-derived IL-6 caused miR-223 levels to decrease. The addition of exogenous miR-223 inhibited cell proliferation, ICAM-1 expression, and monocyte adhesion. The NF-κB and STAT3 signaling pathways collaborate during the activation process. MiR-223 mimics inhibited the nuclear localization and DNA binding of p65 and STAT3 but had no effect on the expression of upstream molecules. Instead, importin α4 and α5 (multipurpose nuclear transport receptors), validated as targets of miR-223, were responsible for the nuclear transport of p65 and STAT3. Importin α4 and α5 siRNA inhibited the nuclear localization of p65 and STAT3 and prevented cell proliferation and monocyte adhesion. The level of miR-223 in circulating endothelial cells was decreased and related to the clinical and pathological parameters. Thus, miR-223 downregulation promotes glomerular endothelial cell activation by upregulating importin α4 and α5 in IgAN. Monitoring the level of miR-223 in circulating endothelial cells may provide a noninvasive method for evaluating the severity of IgAN.


Subject(s)
Glomerulonephritis, IGA/pathology , Kidney Glomerulus/pathology , MicroRNAs/physiology , alpha Karyopherins/physiology , Adult , Cell Proliferation , Down-Regulation , Endothelial Cells/physiology , Female , Humans , Interleukin-6/pharmacology , Male , MicroRNAs/blood , NF-kappa B/physiology , STAT3 Transcription Factor/physiology , Up-Regulation
11.
J Biol Chem ; 286(17): 15126-31, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21454664

ABSTRACT

Snail, a zinc finger-containing transcriptional regulator, migrates into the nucleus where it controls gene expression. We demonstrated previously that importin ß1 directly recognizes the zinc finger domain of Snail and transports it into the nucleus. Here, using in vitro and in vivo assays, we show that importin α, an adaptor protein for importin ß1, negatively regulates the nuclear import of Snail mediated by importin ß1. In vitro binding assays indicated that importin α interacted with the zinc finger domain of Snail to compete with the binding of importin ß1 and that Snail did not form a ternary complex with importin α/importin ß1. Overexpression of importin α in A549 cells reduced the endogenous Snail protein level, which was restored by inhibitors of the proteasome and glycogen synthase kinase 3ß. Furthermore, knockdown of importin α by siRNA treatment increased the endogenous Snail protein level in several cancer cell lines. This study provides a novel regulatory mechanism of the nuclear protein import process by importin α and gives an implication to control Snail activity by inhibiting its nuclear localization.


Subject(s)
Cell Nucleus/metabolism , Transcription Factors/metabolism , alpha Karyopherins/physiology , Active Transport, Cell Nucleus , Binding Sites , Cell Line, Tumor , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinase 3 beta , Humans , Proteasome Endopeptidase Complex , Protein Binding , Snail Family Transcription Factors , Zinc Fingers , beta Karyopherins/metabolism
12.
Biochim Biophys Acta ; 1813(9): 1562-77, 2011 Sep.
Article in English | MEDLINE | ID: mdl-20977914

ABSTRACT

Although proteins are translated on cytoplasmic ribosomes, many of these proteins play essential roles in the nucleus, mediating key cellular processes including but not limited to DNA replication and repair as well as transcription and RNA processing. Thus, understanding how these critical nuclear proteins are accurately targeted to the nucleus is of paramount importance in biology. Interaction and structural studies in the recent years have jointly revealed some general rules on the specificity determinants of the recognition of nuclear targeting signals by their specific receptors, at least for two nuclear import pathways: (i) the classical pathway, which involves the classical nuclear localization sequences (cNLSs) and the receptors importin-α/karyopherin-α and importin-ß/karyopherin-ß1; and (ii) the karyopherin-ß2 pathway, which employs the proline-tyrosine (PY)-NLSs and the receptor transportin-1/karyopherin-ß2. The understanding of specificity rules allows the prediction of protein nuclear localization. We review the current understanding of the molecular determinants of the specificity of nuclear import, focusing on the importin-α•cargo recognition, as well as the currently available databases and predictive tools relevant to nuclear localization. This article is part of a Special Issue entitled: Regulation of Signaling and Cellular Fate through Modulation of Nuclear Protein Import.


Subject(s)
Active Transport, Cell Nucleus/physiology , Nuclear Localization Signals/physiology , Amino Acid Sequence , Animals , Binding Sites , Databases, Protein , Humans , Mice , Models, Biological , Models, Molecular , Molecular Sequence Data , Nuclear Localization Signals/chemistry , Nuclear Localization Signals/genetics , Parathyroid Hormone-Related Protein/chemistry , Parathyroid Hormone-Related Protein/physiology , Protein Interaction Domains and Motifs , Signal Transduction/physiology , Sterol Regulatory Element Binding Protein 2/chemistry , Sterol Regulatory Element Binding Protein 2/physiology , alpha Karyopherins/chemistry , alpha Karyopherins/physiology , beta Karyopherins/chemistry , beta Karyopherins/physiology
13.
Biol Reprod ; 86(3): 84, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22156475

ABSTRACT

This study arose from our finding that SubH2Bv, a histone H2B variant residing in the subacrosomal compartment of mammalian spermatozoa, contains a bipartite nuclear localization signal (bNLS) but in spite of this did not enter the spermatid nucleus. Instead, it associated with proacrosomic and acrosomic vesicles, which were targeted to the nuclear surface to form the acrosome. On this basis we proposed that SubH2Bv targets proacrosomic/acrosomic vesicles from the Golgi apparatus to the nuclear envelope by utilizing the classical bipartite/karyopherin alpha (KPNA) nuclear import pathway. To test the protein's nuclear targeting ability, SubH2Bv, with and without targeted mutations of the basic residues of bNLS, as well as bNLS alone, were transfected into mammalian cells as GFP-fusion proteins. Only the intact bNLS conferred nuclear entry. Subsequently, we showed that a KPNA, most likely KPNA6, occupies the same sperm head compartment and follows the same pattern of acrosomal association during spermiogenesis as SubH2Bv. Sperm head fractionation combined with Western blotting located this KPNA to the subacrosomal layer of the perinuclear theca, while immunocytochemistry of testicular sections showed that it associates with the surface of proacrosomic/acrosomic vesicles during acrosomal biogenesis. The identical sperm-localization and testicular-expression patterns between KPNA and SubH2Bv suggested a potential binding interaction between these proteins. This was supported by recombinant SubH2Bv affinity pull-down assays on germ cell extracts. The results of this study provide a compelling argument that these two nuclear homing proteins work in concert to direct the acrosomic vesicle to the nucleus. Their final residence in the subacrosomal layer of the perinuclear theca of spermatozoa indicates a role for SubH2Bv and KPNA in acrosomal-nuclear docking.


Subject(s)
Acrosome/physiology , Active Transport, Cell Nucleus/physiology , Signal Transduction/physiology , Spermatogenesis/physiology , alpha Karyopherins/physiology , Animals , Cattle , Green Fluorescent Proteins/genetics , Histones/genetics , Histones/physiology , Male , Mice , Mutation/genetics , Sperm Head/physiology , Transfection
14.
FASEB J ; 25(11): 3958-65, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21840941

ABSTRACT

The cellular repertoire of importin (IMP) proteins that mediates nuclear import of transcription factors and chromatin remodeling agents is critical to processes such as differentiation and transformation. This study identifies for the first time independent roles for specific IMPαs in murine embryonic stem cells (mESCs), showing that mESC differentiation is accompanied by dynamic changes in the levels of transcripts encoding the IMPs, IMPα3, IMPα4, IMPß1, and IPO5. Of these, only IMPα4 was maintained at higher levels in differentiating mESCs, correlating with the finding that IMPα4 overexpression induced a significant decrease in Oct3/4 protein levels compared to control transfections. In parallel, IMPα4 protein showed a unique and striking shift in subcellular localization from the nucleus to the cytoplasm during differentiation, which is consistent with activation of a role in nuclear import of differentiation factors. Overexpression of a dominant-negative IMPα2 isoform, when assessed against adjacent untransfected or IMPα2 transfected cells, led to both a significant reduction in endogenous Oct3/4 protein levels and inhibition of Oct3/4 nuclear localization, suggesting that IMPα2-mediated delivery of Oct3/4 to the nucleus contributes directly to maintenance of mESC pluripotency. These findings implicate IMPα2 and IMPα4 in specific but distinct roles in the fate choice between pluripotency and commitment to differentiation.


Subject(s)
Embryonic Stem Cells/metabolism , Karyopherins/physiology , Nuclear Proteins/physiology , Octamer Transcription Factor-3/metabolism , alpha Karyopherins/physiology , Active Transport, Cell Nucleus/genetics , Animals , Cell Differentiation/physiology , Cell Line , Mice , Nuclear Proteins/biosynthesis , Octamer Transcription Factor-3/biosynthesis , alpha Karyopherins/biosynthesis , beta Karyopherins/physiology
15.
Reprod Fertil Dev ; 24(2): 382-91, 2012.
Article in English | MEDLINE | ID: mdl-22281085

ABSTRACT

Coordinated partitioning of intracellular cargoes between nuclear and cytoplasmic compartments is critical for cell survival and differentiation. The karyopherin α/ß heterodimer functions to import cytoplasmic proteins that possess classical nuclear localisation signals into the nucleus. Seven karyopherinαsubtypes have been identified in mammals. The aim of this study was to determine the relative abundance of transcripts encoding seven karyopherinαsubtypes in porcine oocytes and embryos at discrete stages of cleavage development, and to determine the developmental requirements of karypopherinα7 (KPNA7), an oocyte and cleavage stage embryo-specific karyopherinαsubtype. We hypothesised that knockdown of KPNA7 would negatively affect porcine cleavage development. To test this hypothesis, in vitro matured and fertilised porcine oocytes were injected with a double-stranded interfering RNA molecule that targeted KPNA7; nuclei were counted in all embryos 6 days after fertilisation. Embryos injected with KPNA7-interfering RNAs possessed significantly lower cell numbers than their respective control groups (P<0.05). In vitro binding assays also suggest that KPNA7 may transport intracellular proteins that possess unique nuclear localisation signals. Our data suggest that embryos have differential requirements for individual karyopherinαsubtypes and that these karyopherinαsubtypes differentially transport intracellular cargo during cleavage development.


Subject(s)
Embryonic Development/genetics , Swine/embryology , Swine/genetics , alpha Karyopherins/genetics , alpha Karyopherins/physiology , Animals , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cells, Cultured , Embryo, Mammalian/metabolism , Embryonic Development/drug effects , Embryonic Development/physiology , Female , Gene Expression Regulation, Developmental/drug effects , Nuclear Localization Signals/metabolism , Oocytes/metabolism , Organ Specificity/drug effects , Organ Specificity/genetics , Pregnancy , Protein Transport/genetics , RNA, Small Interfering/pharmacology , Swine/physiology , alpha Karyopherins/antagonists & inhibitors , alpha Karyopherins/metabolism
16.
Neuron ; 56(5): 793-806, 2007 Dec 06.
Article in English | MEDLINE | ID: mdl-18054857

ABSTRACT

The organization of neuronal wiring into layers and columns is a common feature of both vertebrate and invertebrate brains. In the Drosophila visual system, each R7 photoreceptor axon projects within a single column to a specific layer of the optic lobe. We refer to the restriction of terminals to single columns as tiling. In a genetic screen based on an R7-dependent behavior, we identified the Activin receptor Baboon and the nuclear import adaptor Importin-alpha3 as being required to prevent R7 axon terminals from overlapping with the terminals of R7s in neighboring columns. This tiling function requires the Baboon ligand, dActivin, the transcription factor, dSmad2, and retrograde transport from the growth cone to the R7 nucleus. We propose that dActivin is an autocrine signal that restricts R7 growth cone motility, and we demonstrate that it acts in parallel with a paracrine signal that mediates repulsion between R7 terminals.


Subject(s)
Activins/physiology , Axons/physiology , Brain/physiology , Signal Transduction/physiology , Vision, Ocular/physiology , Animals , Cell Movement/physiology , Cells, Cultured , Drosophila , Growth Cones/physiology , Immunohistochemistry , In Situ Hybridization , Mutation/physiology , Paracrine Communication/physiology , Photoreceptor Cells, Invertebrate/physiology , Presynaptic Terminals/physiology , Smad2 Protein/genetics , Smad2 Protein/physiology , Ultraviolet Rays , alpha Karyopherins/genetics , alpha Karyopherins/physiology
17.
J Cell Biol ; 171(1): 19-25, 2005 Oct 10.
Article in English | MEDLINE | ID: mdl-16203861

ABSTRACT

Transport receptors of the importin beta family continuously shuttle between the nucleus and cytoplasm. We previously reported that the nuclear export of importin beta involves energy-requiring step(s) in living cells. Here, we show that the in vitro nuclear export of importin beta also requires energy input. Cytosol, depleted of ATP-binding proteins, did not support the sufficient nuclear export of importin beta. Further purification revealed that the active component in the absorbed fraction was a 70-kD heat shock cognate protein (hsc70). The addition of recombinant hsc70, but not an ATPase-deficient hsc70 mutant, to the depleted cytosol restored the export activity. In living cells, depletion of hsc70 caused the significant nuclear accumulation of importin beta. These effects of hsc70 were observed in the nuclear export of importin beta, but also for other import receptors, transportin and importin alpha. These results suggest that hsc70 broadly modulates nucleocytoplasmic transport systems by regulating the nuclear export of receptor proteins.


Subject(s)
Active Transport, Cell Nucleus/physiology , HSC70 Heat-Shock Proteins/metabolism , Nuclear Proteins/metabolism , Adenosine Triphosphatases/metabolism , Cell Line, Tumor , Cell Nucleus/chemistry , Cytosol/chemistry , Cytosol/enzymology , HeLa Cells , Humans , Karyopherins/physiology , Recombinant Proteins/analysis , Recombinant Proteins/genetics , alpha Karyopherins/physiology , beta Karyopherins/physiology
18.
Science ; 369(6505): 842-846, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32792398

ABSTRACT

How is neuropathic pain regulated in peripheral sensory neurons? Importins are key regulators of nucleocytoplasmic transport. In this study, we found that importin α3 (also known as karyopherin subunit alpha 4) can control pain responsiveness in peripheral sensory neurons in mice. Importin α3 knockout or sensory neuron-specific knockdown in mice reduced responsiveness to diverse noxious stimuli and increased tolerance to neuropathic pain. Importin α3-bound c-Fos and importin α3-deficient neurons were impaired in c-Fos nuclear import. Knockdown or dominant-negative inhibition of c-Fos or c-Jun in sensory neurons reduced neuropathic pain. In silico screens identified drugs that mimic importin α3 deficiency. These drugs attenuated neuropathic pain and reduced c-Fos nuclear localization. Thus, perturbing c-Fos nuclear import by importin α3 in peripheral neurons can promote analgesia.


Subject(s)
Chronic Pain/physiopathology , Neuralgia/physiopathology , Sensory Receptor Cells/physiology , alpha Karyopherins/physiology , Active Transport, Cell Nucleus , Animals , Benzophenones/pharmacology , Chronic Pain/genetics , Gene Expression Profiling , Gene Knockdown Techniques , Isoxazoles/pharmacology , Mice , Mice, Inbred C57BL , Neuralgia/genetics , Proto-Oncogene Proteins c-fos/antagonists & inhibitors , Proto-Oncogene Proteins c-fos/metabolism , Transcription Factor AP-1/metabolism , alpha Karyopherins/genetics
19.
Biochim Biophys Acta ; 1783(3): 394-404, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18187047

ABSTRACT

Hypoxia-inducible factors are the key elements in the essential process of oxygen homeostasis of vertebrate cells. Stabilisation and subsequent nuclear localisation of HIF-alpha subunits results in the activation of target genes such as vegf, epo and glut1. The passage of transcription factors e.g. HIF-1alpha into the nucleus through the nuclear pore complex is regulated by nuclear transport receptors. Therefore nucleocytoplasmic shuttling can regulate transcriptional activity by facilitating the cellular traffic of transcription factors between both compartments. Here, we report on the identification of specific interactions of hypoxia-inducible factors with nuclear transport receptors importin alpha/beta. HIF-1alpha, -1beta, and HIF-2alpha are binding to importin alpha1, alpha3, alpha5, and alpha7. The direct interaction of HIF-1alpha to alpha importins is dependent on a functional nuclear localisation signal within the C-terminal region of the protein. In contrast, the supposed N-terminal NLS is not effective. Our findings provide new insight into the mechanism of the regulation of nuclear transport of hypoxia-inducible factors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Nucleus/metabolism , Hypoxia-Inducible Factor 1/metabolism , alpha Karyopherins/physiology , beta Karyopherins/physiology , Active Transport, Cell Nucleus , Amino Acid Sequence , Binding Sites , Cells, Cultured , HeLa Cells , Humans , Nuclear Localization Signals/chemistry , Nuclear Localization Signals/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Isoforms/metabolism , Signal Transduction
20.
BMC Cell Biol ; 10: 74, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19835572

ABSTRACT

BACKGROUND: Tpr is a large protein with an extended coiled-coil domain that is localized within the nuclear basket of the nuclear pore complex. Previous studies 1 involving antibody microinjection into mammalian cells suggested a role for Tpr in nuclear export of proteins via the CRM1 export receptor. In addition, Tpr was found to co-immunoprecipitate with importins alpha and beta from Xenopus laevis egg extracts 2, although the function of this is unresolved. Yeast Mlp1p and Mlp2p, which are homologous to vertebrate Tpr, have been implicated in mRNA surveillance to retain unspliced mRNAs in the nucleus34. To augment an understanding of the role of Tpr in nucleocytoplasmic trafficking, we explored the interactions of recombinant Tpr with the karyopherins CRM1, importin beta and importin alpha by solid phase binding assays. We also investigated the conditions required for nuclear import of Tpr using an in vitro assay. RESULTS: We found that Tpr binds strongly and specifically to importin alpha, importin beta, and a CRM1 containing trimeric export complex, and that the binding sites for importins alpha and beta are distinct. We also determined that the nuclear import of Tpr is dependent on cytosolic factors and energy and is efficiently mediated by the importin alpha/beta import pathway. CONCLUSION: Based on the binding and nuclear import assays, we propose that Tpr is imported into the nucleus by the importin alpha/beta heterodimer. In addition, we suggest that Tpr can serve as a nucleoporin binding site for importin beta during import of importin beta cargo complexes and/or importin beta recycling. Our finding that Tpr bound preferentially to CRM1 in an export complex strengthens the notion that Tpr is involved in protein export.


Subject(s)
Nuclear Pore Complex Proteins/metabolism , alpha Karyopherins/metabolism , beta Karyopherins/metabolism , Binding Sites , Karyopherins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Proteins/metabolism , alpha Karyopherins/physiology , beta Karyopherins/physiology , Exportin 1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL