Importin-β1 and Nucleus Transporter Chromosome Region Maintenance 1 (CRM1) Constitute the Nucleocytoplasmic Transport System of STAT3 in Blood-Derived CLL Cells.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1059-1059
Author(s):  
Inbal Hazan-Halevy ◽  
David Harris ◽  
Zhiming Liu ◽  
Alessandra Ferrajoli ◽  
Michael J Keating ◽  
...  

Abstract B-cell chronic lymphocytic leukemia (CLL) is the most common adult leukemia in the Western hemisphere. Although several chromosomal and molecular abnormalities have been identified in CLL cells in recent years, the pathogenesis of CLL is still poorly understood. Signal transducer and activator of transcription 3 (STAT3) plays a major role in cellular physiology. Upon exposure to cytokines or growth factors, STAT3 is tyrosine-phosphorylated, migrates to the nucleus, and binds to DNA. Constitutive phosphorylation of STAT3 on tyrosine 705 residues has been found in several solid tumors and hematologic malignancies. Remarkably, CLL is the only disease in which STAT3 is constitutively phosphorylated (p) on serine rather than tyrosine residues (Frank et al. JCI100:3149, 1997). We have recently discovered that serine pSTAT3 translocates to the nucleus, binds to DNA, activates transcription, and plays a major role in the pathogenensis of CLL. Little is known about the transport mechanisms utilized by STAT molecules in fresh leukemia cells, and no data are available on the transport mechanism of serine pSTAT3. Therefore, we sought to identify the nucleocytoplasmic transport system of serine pSTAT3 in CLL cells. In other cellular systems, importin-α3 or -α6 binds to the nuclear localization signal in STAT3, the N terminus of importin-α is directly recognized by importin-β1, and the complex consisting of STAT3, importin-α3 and improtin-β1 transits through the nuclear pore complexes (NPC). To identify which nucleocytoplasmic transport mechanism of serine pSTAT3 is operative in CLL cells, we performed a series of immunoprecipitation experiments with antibodies to STAT3 and importin-β1. We found that STAT3 co-immunoprecipitated with importin-β1 in whole cell, cytoplasmic, and nuclear extracts. We could not determine which member of the importin-α family binds serine pSTAT3 to form a complex with importin-β1 because none of the investigated α-importins (importin-α1, -α3, -α5, -α6, and -α7) co-immunoprecipitated with STAT3. Similar results were obtained when importin-β1 was immunoprecipitated. Unlike the studied α-importins, serine pSTAT3 and STAT3 co-immunoprecipitated with importin-β1. Thus, either an α-importin binds serine pSTAT3 but failed to co-immunoprecipitate, or an unidentified transporter binds serine pSTAT3. After establishing that importin-β1 translocates serine pSTAT3 to the nucleus, we sought to identify the nuclear export mechanism. The established nuclear export mechanism of STAT3 consists of CRM1 that binds to the nuclear export signal on STAT3 and exports STAT3 through the NPC. Using an identical experimental design, we immunoprecipitated whole cell, cytoplasmic, and nuclear extracts with anti-CRM1 antibodies and found that STAT3 and serine pSTAT3 co-immunoprecipitated with CRM1. Then, we immunoprecipitated the cell extracts with anti-STAT3 antibodies. In these experiments, CRM1 co-immunoprecipitated with STAT3. To further elucidate the role of CRM1 in the STAT3 nuclear export system, we incubated CLL cells with increasing concentrations of the CRM1 inhibitor leptomycin B and assessed STAT3 protein levels in nuclear extracts by Western immunoblotting. We found that leptomycin B increased the accumulation of STAT3 in the nucleus in a dose dependent manner, further confirming that CRM1 exports STAT3 from the nucleus to the cytoplasm. Taken together, our data demonstrate for the first time that in CLL cells STAT3 and serine pSTAT3 are transported into the nucleus by importin-β1 and exported by CRM1. Targeting this nuclear trafficking system might provide a new therapeutic strategy for the treatment of CLL.

1999 ◽  
Vol 10 (3) ◽  
pp. 649-664 ◽  
Author(s):  
Katharine S. Ullman ◽  
Sundeep Shah ◽  
Maureen A. Powers ◽  
Douglass J. Forbes

The fundamental process of nucleocytoplasmic transport takes place through the nuclear pore. Peripheral pore structures are presumably poised to interact with transport receptors and their cargo as these receptor complexes first encounter the pore. One such peripheral structure likely to play an important role in nuclear export is the basket structure located on the nuclear side of the pore. At present, Nup153 is the only nucleoporin known to localize to the surface of this basket, suggesting that Nup153 is potentially one of the first pore components an RNA or protein encounters during export. In this study, anti-Nup153 antibodies were used to probe the role of Nup153 in nuclear export in Xenopus oocytes. We found that Nup153 antibodies block three major classes of RNA export, that of snRNA, mRNA, and 5S rRNA. Nup153 antibodies also block the NES protein export pathway, specifically the export of the HIV Rev protein, as well as Rev-dependent RNA export. Not all export was blocked; Nup153 antibodies did not impede the export of tRNA or the recycling of importin β to the cytoplasm. The specific antibodies used here also did not affect nuclear import, whether mediated by importin α/β or by transportin. Overall, the results indicate that Nup153 is crucial to multiple classes of RNA and protein export, being involved at a vital juncture point in their export pathways. This juncture point appears to be one that is bypassed by tRNA during its export. We asked whether a physical interaction between RNA and Nup153 could be observed, using homoribopolymers as sequence-independent probes for interaction. Nup153, unlike four other nucleoporins including Nup98, associated strongly with poly(G) and significantly with poly(U). Thus, Nup153 is unique among the nucleoporins tested in its ability to interact with RNA and must do so either directly or indirectly through an adaptor protein. These results suggest a unique mechanistic role for Nup153 in the export of multiple cargos.


1999 ◽  
Vol 19 (2) ◽  
pp. 1025-1037 ◽  
Author(s):  
Joanne G. A. Savory ◽  
Brian Hsu ◽  
Ian R. Laquian ◽  
Ward Giffin ◽  
Terry Reich ◽  
...  

ABSTRACT Glucocorticoid receptor (GR) cycles between a free liganded form that is localized to the nucleus and a heat shock protein (hsp)-immunophilin-complexed, unliganded form that is usually localized to the cytoplasm but that can also be nuclear. In addition, rapid nucleocytoplasmic exchange or shuttling of the receptor underlies its localization. Nuclear import of liganded GR is mediated through a well-characterized sequence, NL1, adjacent to the receptor DNA binding domain and a second, uncharacterized motif, NL2, that overlaps with the ligand binding domain. In this study we report that rapid nuclear import (half-life [t 1/2] of 4 to 6 min) of agonist- and antagonist-treated GR and the localization of unliganded, hsp-associated GRs to the nucleus in G0 are mediated through NL1 and correlate with the binding of GR to pendulin/importin α. By contrast, NL2-mediated nuclear transfer of GR occurred more slowly (t 1/2 = 45 min to 1 h), was agonist specific, and appeared to be independent of binding to importin α. Together, these results suggest that NL2 mediates the nuclear import of GR through an alternative nuclear import pathway. Nuclear export of GR was inhibited by leptomycin B, suggesting that the transfer of GR to the cytoplasm is mediated through the CRM1-dependent pathway. Inhibition of GR nuclear export by leptomycin B enhanced the nuclear localization of both unliganded, wild-type GR and hormone-treated NL1− GR. These results highlight that the subcellular localization of both liganded and unliganded GRs is determined, at least in part, by a flexible equilibrium between the rates of nuclear import and export.


2008 ◽  
Vol 19 (4) ◽  
pp. 1614-1626 ◽  
Author(s):  
Julia Dorfman ◽  
Ian G. Macara

LKB1, a serine/threonine kinase, regulates cell polarity, metabolism, and cell growth. The activity and cellular distribution of LKB1 are determined by cofactors, STRADα and MO25. STRADα induces relocalization of LKB1 from the nucleus to the cytoplasm and stimulates its catalytic activity. MO25 stabilizes the STRADα/LKB1 interaction. We investigated the mechanism of nucleocytoplasmic transport of LKB1 in response to its cofactors. Although LKB1 is imported into the nucleus by importin-α/β, STRADα and MO25 passively diffuse between the nucleus and the cytoplasm. STRADα induces nucleocytoplasmic shuttling of LKB1. STRADα facilitates nuclear export of LKB1 by serving as an adaptor between LKB1 and exportins CRM1 and exportin7. STRADα inhibits import of LKB1 by competing with importin-α for binding to LKB1. MO25 stabilizes the LKB1–STRADα complex but it does not facilitate its nucleocytoplasmic shuttling. Strikingly, the STRADβ, isoform which differs from STRADα in the N- and C-terminal domains that are responsible for interaction with export receptors, does not efficiently relocalize LKB1 from the nucleus to the cytoplasm. These results identify a multifactored mechanism to control LKB1 localization, and they suggest that the STRADβ-LKB1 complex might possess unique functions in the nucleus.


1998 ◽  
Vol 140 (3) ◽  
pp. 499-509 ◽  
Author(s):  
Michael J. Matunis ◽  
Jian Wu ◽  
Günter Blobel

RanGAP1 is the GTPase-activating protein for Ran, a small ras-like GTPase involved in regulating nucleocytoplasmic transport. In vertebrates, RanGAP1 is present in two forms: one that is cytoplasmic, and another that is concentrated at the cytoplasmic fibers of nuclear pore complexes (NPCs). The NPC-associated form of RanGAP1 is covalently modified by the small ubiquitin-like protein, SUMO-1, and we have recently proposed that SUMO-1 modification functions to target RanGAP1 to the NPC. Here, we identify the domain of RanGAP1 that specifies SUMO-1 modification and demonstrate that mutations in this domain that inhibit modification also inhibit targeting to the NPC. Targeting of a heterologous protein to the NPC depended on determinants specifying SUMO-1 modification and also on additional determinants in the COOH-terminal domain of RanGAP1. SUMO-1 modification and these additional determinants were found to specify interaction between the COOH-terminal domain of RanGAP1 and a region of the nucleoporin, Nup358, between Ran-binding domains three and four. Together, these findings indicate that SUMO-1 modification targets RanGAP1 to the NPC by exposing, or creating, a Nup358 binding site in the COOH-terminal domain of RanGAP1. Surprisingly, the COOH-terminal domain of RanGAP1 was also found to harbor a nuclear localization signal. This nuclear localization signal, and the presence of nine leucine-rich nuclear export signal motifs, suggests that RanGAP1 may shuttle between the nucleus and the cytoplasm.


2008 ◽  
Vol 284 (9) ◽  
pp. 5753-5762 ◽  
Author(s):  
Takashi Miki ◽  
Katsuya Okawa ◽  
Toshihiro Sekimoto ◽  
Yoshihiro Yoneda ◽  
Sadanori Watanabe ◽  
...  

2005 ◽  
Vol 169 (3) ◽  
pp. 415-424 ◽  
Author(s):  
Christian Faul ◽  
Stefan Hüttelmaier ◽  
Jun Oh ◽  
Virginie Hachet ◽  
Robert H. Singer ◽  
...  

14-3-3 proteins are phosphoserine/threonine-binding proteins that play important roles in many regulatory processes, including intracellular protein targeting. 14-3-3 proteins can anchor target proteins in the cytoplasm and in the nucleus or can mediate their nuclear export. So far, no role for 14-3-3 in mediating nuclear import has been described. There is also mounting evidence that nuclear import is regulated by the phosphorylation of cargo proteins, but the underlying mechanism remains elusive. Myopodin is a dual-compartment, actin-bundling protein that functions as a tumor suppressor in human bladder cancer. In muscle cells, myopodin redistributes between the nucleus and the cytoplasm in a differentiation-dependent and stress-induced fashion. We show that importin α binding and the subsequent nuclear import of myopodin are regulated by the serine/threonine phosphorylation-dependent binding of myopodin to 14-3-3. These results establish a novel paradigm for the promotion of nuclear import by 14-3-3 binding. They provide a molecular explanation for the phosphorylation-dependent nuclear import of nuclear localization signal-containing cargo proteins.


2011 ◽  
Vol 441 (1) ◽  
pp. 209-217 ◽  
Author(s):  
Iraia García-Santisteban ◽  
Sonia Bañuelos ◽  
Jose A. Rodríguez

The mechanisms that regulate the nucleocytoplasmic localization of human deubiquitinases remain largely unknown. The nuclear export receptor CRM1 binds to specific amino acid motifs termed NESs (nuclear export sequences). By using in silico prediction and experimental validation of candidate sequences, we identified 32 active NESs and 78 inactive NES-like motifs in human deubiquitinases. These results allowed us to evaluate the performance of three programs widely used for NES prediction, and to add novel information to the recently redefined NES consensus. The novel NESs identified in the present study reveal a subset of 22 deubiquitinases bearing motifs that might mediate their binding to CRM1. We tested the effect of the CRM1 inhibitor LMB (leptomycin B) on the localization of YFP (yellow fluorescent protein)- or GFP (green fluorescent protein)-tagged versions of six NES-bearing deubiquitinases [USP (ubiquitin-specific peptidase) 1, USP3, USP7, USP21, CYLD (cylindromatosis) and OTUD7B (OTU-domain-containing 7B)]. YFP–USP21 and, to a lesser extent, GFP–OTUD7B relocated from the cytoplasm to the nucleus in the presence of LMB, revealing their nucleocytoplasmic shuttling capability. Two sequence motifs in USP21 had been identified during our survey as active NESs in the export assay. Using site-directed mutagenesis, we show that one of these motifs mediates USP21 nuclear export, whereas the second motif is not functional in the context of full-length USP21.


Oncogene ◽  
1999 ◽  
Vol 18 (51) ◽  
pp. 7378-7386 ◽  
Author(s):  
Philip Smart ◽  
E Birgitte Lane ◽  
David P Lane ◽  
Carol Midgley ◽  
Borek Vojtesek ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document