Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Proc Natl Acad Sci U S A ; 120(29): e2301199120, 2023 Jul 18.
Article En | MEDLINE | ID: mdl-37450495

Previously, we showed that the nuclear import receptor Importin-9 wraps around the H2A-H2B core to chaperone and transport it from the cytoplasm to the nucleus. However, unlike most nuclear import systems where RanGTP dissociates cargoes from their importins, RanGTP binds stably to the Importin-9•H2A-H2B complex, and formation of the ternary RanGTP•Importin-9•H2A-H2B complex facilitates H2A-H2B release to the assembling nucleosome. It was unclear how RanGTP and the cargo H2A-H2B can bind simultaneously to an importin, and how interactions of the three components position H2A-H2B for release. Here, we show cryo-EM structures of Importin-9•RanGTP and of its yeast homolog Kap114, including Kap114•RanGTP, Kap114•H2A-H2B, and RanGTP•Kap114•H2A-H2B, to explain how the conserved Kap114 binds H2A-H2B and RanGTP simultaneously and how the GTPase primes histone transfer to the nucleosome. In the ternary complex, RanGTP binds to the N-terminal repeats of Kap114 in the same manner as in the Kap114/Importin-9•RanGTP complex, and H2A-H2B binds via its acidic patch to the Kap114 C-terminal repeats much like in the Kap114/Importin-9•H2A-H2B complex. Ran binds to a different conformation of Kap114 in the ternary RanGTP•Kap114•H2A-H2B complex. Here, Kap114 no longer contacts the H2A-H2B surface proximal to the H2A docking domain that drives nucleosome assembly, positioning it for transfer to the assembling nucleosome or to dedicated H2A-H2B chaperones in the nucleus.


Nucleosomes , Saccharomyces cerevisiae Proteins , Nucleosomes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Protein Binding , Karyopherins/metabolism , Saccharomyces cerevisiae/metabolism , Molecular Chaperones/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism
2.
Structure ; 31(8): 924-934.e4, 2023 08 03.
Article En | MEDLINE | ID: mdl-37279758

The HNRNPH2 proline-tyrosine nuclear localization signal (PY-NLS) is mutated in HNRNPH2-related X-linked neurodevelopmental disorder, causing the normally nuclear HNRNPH2 to accumulate in the cytoplasm. We solved the cryoelectron microscopy (cryo-EM) structure of Karyopherin-ß2/Transportin-1 bound to the HNRNPH2 PY-NLS to understand importin-NLS recognition and disruption in disease. HNRNPH2 206RPGPY210 is a typical R-X2-4-P-Y motif comprising PY-NLS epitopes 2 and 3, followed by an additional Karyopherin-ß2-binding epitope, we term epitope 4, at residues 211DRP213; no density is present for PY-NLS epitope 1. Disease variant mutations at epitopes 2-4 impair Karyopherin-ß2 binding and cause aberrant cytoplasmic accumulation in cells, emphasizing the role of nuclear import defect in disease. Sequence/structure analysis suggests that strong PY-NLS epitopes 4 are rare and thus far limited to close paralogs of HNRNPH2, HNRNPH1, and HNRNPF. Epitope 4-binidng hotspot Karyopherin-ß2 W373 corresponds to close paralog Karyopherin-ß2b/Transportin-2 W370, a pathological variant site in neurodevelopmental abnormalities, suggesting that Karyopherin-ß2b/Transportin-2-HNRNPH2/H1/F interactions may be compromised in the abnormalities.


Karyopherins , Nuclear Localization Signals , Karyopherins/metabolism , Nuclear Localization Signals/metabolism , Epitopes/metabolism , Tyrosine/metabolism , Proline , Cryoelectron Microscopy , Active Transport, Cell Nucleus , beta Karyopherins/genetics , beta Karyopherins/chemistry , beta Karyopherins/metabolism , Cell Nucleus/metabolism
3.
Structure ; 31(8): 903-911.e3, 2023 08 03.
Article En | MEDLINE | ID: mdl-37379840

Imp9 is the primary importin for shuttling H2A-H2B from the cytoplasm to the nucleus. It employs an unusual mechanism where the binding of RanGTP is insufficient to release H2A-H2B. The resulting stable RanGTP·Imp9·H2A-H2B complex gains nucleosome assembly activity with H2A-H2B able to be deposited into an assembling nucleosome in vitro. Using hydrogen-deuterium exchange coupled with mass spectrometry (HDX), we show that Imp9 stabilizes H2A-H2B beyond the direct-binding site, like other histone chaperones. HDX also shows that binding of RanGTP releases H2A-H2B contacts at Imp9 HEAT repeats 4-5, but not 18-19. DNA- and histone-binding surfaces of H2A-H2B are exposed in the ternary complex, facilitating nucleosome assembly. We also reveal that RanGTP has a weaker affinity for Imp9 when H2A-H2B is bound. Imp9 thus provides a connection between the nuclear import of H2A-H2B and its deposition into chromatin.


Histones , Nucleosomes , Histones/metabolism , Nucleosome Assembly Protein 1/genetics , Chromatin , Karyopherins/metabolism
4.
bioRxiv ; 2023 May 10.
Article En | MEDLINE | ID: mdl-37214964

Histones are synthesized and processed in the cytoplasm and then transported into the nucleus for assembly into nucleosomes. H2A-H2B is imported into the S. cerevisiae nucleus by the importin Kap114, which also imports the most prominent H2A-H2B chaperone, Nap1. We understand how Kap114 recognizes H2A-H2B for nuclear import, but little is known about how it recognizes Nap1. Furthermore, the ternary complex of Nap1, H2A-H2B and Kap114 was previously detected in both the cytosol and the nucleus, but its role in nuclear import is unclear. Here, we present biophysical analysis of interactions between Nap1, H2A-H2B, Kap114 and RanGTP, and cryo-electron microscopy structures of ternary Kap114, Nap1 and H2A-H2B complexes. Kap114 binds Nap1 very weakly, but H2A-H2B enhances Kap114-Nap1 interaction to form a ternary Kap114/Nap1/H2A-H2B complex that is stable in the absence and presence of RanGTP. Cryogenic electron microscopy structures reveal two distinct ternary Kap114/Nap1/H2A-H2B complexes: a 3.2 Šresolution structure of Nap1 bound to H2A-H2B-bound Kap114 where Nap1 does not contact H2A-H2B, and a 3.5 Šresolution structure of H2A-H2B sandwiched between Nap1 and Kap114. Collectively, these results lead to a mechanistic model of how Nap1•H2A-H2B encounters Kap114 in the cytoplasm and how both H2A-H2B and Nap1 are chaperoned and co-imported by Kap114 into the nucleus. The model also suggests how RanGTP-binding stabilizes a quaternary RanGTP/Kap114/Nap1/H2A-H2B complex that facilitates hand-off of H2A-H2B from Kap114 to Nap1, the assembling nucleosome or other nuclear chaperone. Significance Statement: Free core histones are highly toxic and must be sequestered by other macromolecules in the cell. The mechanism of H3-H4 import by karyopherin Importin-4 in the presence of its chaperone ASF1 is understood, but the mechanism of how histone chaperone Nap1 influences H2A-H2B import is not resolved. We present biophysical interaction analysis and cryo-EM structures that reveal how Kap114, Nap1 and H2A-H2B assemble into an import complex. These results lead us to a structural mechanism of how Nap1 delivers H2A-H2B to Kap114 in the cytosol, how Nap1 and H2A-H2B are co-imported into the nucleus, and how RanGTP may influence Kap114/Nap1/H2A-H2B interactions to assemble nucleosomes in the nucleus.

5.
bioRxiv ; 2023 Jan 28.
Article En | MEDLINE | ID: mdl-36747879

Padavannil et al. 2019 show that Importin-9 (Imp9) transports Histones H2A-H2B from the cytoplasm to the nucleus using a non-canonical mechanism whereby binding of a GTP-bound Ran GTPase (RanGTP) fails to evict the H2A-H2B cargo. Instead, a stable complex forms, comprised of equimolar RanGTP, Imp9, and H2A-H2B. Unlike the binary Imp9•H2A-H2B complex, this RanGTP•Imp9•H2A-H2B ternary complex can release H2A-H2B to an assembling nucleosome. Here, we define the molecular basis for this RanGTP-activated nucleosome assembly by Imp9. We use hydrogen-deuterium exchange coupled with mass spectrometry and compare the dynamics and interfaces of the RanGTP•Imp9•H2A-H2B ternary complex to those in the Imp9•H2A-H2B or Imp9•RanGTP binary complexes. Our data are consistent with the Imp9•H2A-H2B structure by Padavannil et al. 2019 showing that Imp9 HEAT repeats 4-5 and 18-19 contact H2A-H2B, as well as many homologous importin•RanGTP structures showing that importin HEAT repeats 1 and 3, and the h8 loop, contact RanGTP. We show that Imp9 stabilizes H2A-H2B beyond the direct binding site, similar to other histone chaperones. Importantly, we reveal that binding of RanGTP releases H2A-H2B interaction at Imp9 HEAT repeats 4-5, but not 18-19. This exposes DNA- and histone-binding surfaces of H2A-H2B, thereby facilitating nucleosome assembly. We also reveal that RanGTP has a weaker affinity for Imp9 when H2A-H2B is bound. This may ensure that H2A-H2B is only released in high RanGTP concentrations near chromatin. We delineate the molecular link between the nuclear import of H2A-H2B and its deposition into chromatin by Imp9. Significance: Imp9 is the primary importin for shuttling H2A-H2B from the cytoplasm to the nucleus. It employs an unusual mechanism where the binding of RanGTP alone is insufficient to release H2A-H2B. The resulting stable RanGTP•Imp9•H2A-H2B complex gains nucleosome assembly activity as H2A-H2B can be deposited onto an assembling nucleosome. We show that H2A-H2B is allosterically stabilized via interactions with both N- and C-terminal portions of Imp9, reinforcing its chaperone-like behavior. RanGTP binding causes H2A-H2B release from the N-terminal portion of Imp9 only. The newly-exposed H2A-H2B surfaces can interact with DNA or H3-H4 in nucleosome assembly. Imp9 thus plays a multi-faceted role in histone import, storage, and deposition regulated by RanGTP, controlling histone supply in the nucleus and to chromatin.

6.
Proc Natl Acad Sci U S A ; 119(38): e2207177119, 2022 09 20.
Article En | MEDLINE | ID: mdl-36103578

IMPORTIN-4, the primary nuclear import receptor of core histones H3 and H4, binds the H3-H4 dimer and histone chaperone ASF1 prior to nuclear import. However, how H3-H3-ASF1 is recognized for transport cannot be explained by available crystal structures of IMPORTIN-4-histone tail peptide complexes. Our 3.5-Å IMPORTIN-4-H3-H4-ASF1 cryoelectron microscopy structure reveals the full nuclear import complex and shows a binding mode different from suggested by previous structures. The N-terminal half of IMPORTIN-4 clamps the globular H3-H4 domain and H3 αN helix, while its C-terminal half binds the H3 N-terminal tail weakly; tail contribution to binding energy is negligible. ASF1 binds H3-H4 without contacting IMPORTIN-4. Together, ASF1 and IMPORTIN-4 shield nucleosomal H3-H4 surfaces to chaperone and import it into the nucleus where RanGTP binds IMPORTIN-4, causing large conformational changes to release H3-H4-ASF1. This work explains how full-length H3-H4 binds IMPORTIN-4 in the cytoplasm and how it is released in the nucleus.


Histone Chaperones , Histones , Karyopherins , Membrane Transport Proteins , Molecular Chaperones , Saccharomyces cerevisiae Proteins , Cell Nucleus/metabolism , Cryoelectron Microscopy , Cytoplasm/metabolism , Histone Chaperones/chemistry , Histones/chemistry , Humans , Karyopherins/chemistry , Membrane Transport Proteins/chemistry , Molecular Chaperones/chemistry , Protein Conformation , Protein Multimerization , Saccharomyces cerevisiae Proteins/chemistry
7.
Methods Mol Biol ; 2502: 245-256, 2022.
Article En | MEDLINE | ID: mdl-35412243

CRM1 recognizes hundreds to thousands of protein cargoes by binding to the eight to fifteen residue-long nuclear export signals (NESs) within their polypeptide chains. Various assays to measure the binding affinity of NESs for CRM1 have been developed. CRM1 binds to NESs with a wide range of binding affinities, with dissociation constants that span from low nanomolar to tens of micromolar. An optimized binding affinity assay with improved throughput was recently developed to measure binding affinities of NES peptides for CRM1 in the presence of excess RanGTP. The assay can measure affinities, with multiple replicates, for up to seven different NES peptides per screening plate. Here, we present a protocol for the purification of the necessary proteins and for measuring CRM1-NES binding affinities.


Karyopherins , Nuclear Export Signals , Receptors, Cytoplasmic and Nuclear , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Karyopherins/chemistry , Karyopherins/metabolism , Peptides/metabolism , Protein Binding , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Exportin 1 Protein
8.
Methods Mol Biol ; 2502: 285-297, 2022.
Article En | MEDLINE | ID: mdl-35412246

The Karyopherin protein CRM1 or XPO1 is the major nuclear export receptor that regulates nuclear exit of thousands of macromolecules in the cell. CRM1 recognizes protein cargoes by binding to their 8-15 residue-long nuclear export signals (NESs). A ternary CRM1-Ran-RanBP1 complex engineered to be suitable for crystallization has enabled structure determination by X-ray crystallography of CRM1 bound to many NES peptides and small-molecule inhibitors. Here, we present a protocol for the purification of the individual proteins, formation of the ternary CRM1-Ran-RanBP1 complex and crystallization of this complex for X-ray crystallography.


Crystallography, X-Ray , Karyopherins , Nuclear Export Signals , Receptors, Cytoplasmic and Nuclear , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Crystallization/methods , Crystallography, X-Ray/methods , Karyopherins/chemistry , Karyopherins/metabolism , Protein Binding , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Exportin 1 Protein
9.
Nat Rev Mol Cell Biol ; 23(5): 307-328, 2022 05.
Article En | MEDLINE | ID: mdl-35058649

Efficient and regulated nucleocytoplasmic trafficking of macromolecules to the correct subcellular compartment is critical for proper functions of the eukaryotic cell. The majority of the macromolecular traffic across the nuclear pores is mediated by the Karyopherin-ß (or Kap) family of nuclear transport receptors. Work over more than two decades has shed considerable light on how the different Kap family members bring their respective cargoes into the nucleus or the cytoplasm in efficient and highly regulated manners. In this Review, we overview the main features and established functions of Kap family members, describe how Kaps recognize their cargoes and discuss the different ways in which these Kap-cargo interactions can be regulated, highlighting new findings and open questions. We also describe current knowledge of the import and export of the components of three large gene expression machines - the core replisome, RNA polymerase II and the ribosome - pointing out the questions that persist about how such large macromolecular complexes are trafficked to serve their function in a designated subcellular location.


Karyopherins , beta Karyopherins , Active Transport, Cell Nucleus/physiology , Cell Nucleus/metabolism , Karyopherins/metabolism , Nuclear Pore/metabolism , Receptors, Cytoplasmic and Nuclear , beta Karyopherins/metabolism
11.
Structure ; 28(3): 378-384.e4, 2020 03 03.
Article En | MEDLINE | ID: mdl-31995742

Tau is a neuronal microtubule (MT)-associated protein of significant interest due to its association with several neurodegenerative disorders. Tau's intrinsic disorder and the dynamic nature of its interactions with tubulin and MTs make its structural characterization challenging. Here, we use an environmentally sensitive fluorophore as a site-specific probe of tau bound to soluble tubulin. Comparison of our results with a recently published tau:MT cryoelectron microscopy model reveals structural similarities between tubulin- and MT-bound tau. Analysis of residues across the repeat regions reveals a hierarchy in tubulin occupancy, which may be relevant to tau's ability to differentiate between tubulin and MTs. As binding to soluble tubulin is a critical first step in MT polymerization, our characterization of the structural features of tau in dynamic, fuzzy tau:tubulin assemblies advances our understanding of how tau functions in the cell and how function may be disrupted in disease.


Tubulin/metabolism , tau Proteins/chemistry , tau Proteins/metabolism , Cryoelectron Microscopy , Humans , Microtubules/metabolism , Models, Molecular , Polymerization , Protein Binding , Protein Conformation
12.
Mol Biol Cell ; 29(17): 2037-2044, 2018 08 15.
Article En | MEDLINE | ID: mdl-29927350

CRM1 (Exportin1/XPO1) exports hundreds of broadly functioning protein cargoes out of the cell nucleus by binding to their classical nuclear export signals (NESs). The 8- to 15-amino-acid-long NESs contain four to five hydrophobic residues and are highly diverse in both sequence and CRM1-bound structure. Here we examine the relationship between nuclear export activities of 24 different NES peptides in cells and their CRM1-NES affinities. We found that binding affinity and nuclear export activity are linearly correlated for NESs with dissociation constants ( Kds) between tens of nanomolar to tens of micromolar. NESs with Kds outside this range have significantly reduced nuclear export activities. These include two unusually tight-binding peptides, one from the nonstructural protein 2 of murine minute virus (MVM NS2) and the other a mutant of the protein kinase A inhibitor (PKI) NES. The crystal structure of CRM1-bound MVM NS2NES suggests that extraordinarily tight CRM1 binding arises from intramolecular contacts within the NES that likely stabilizes the CRM1-bound conformation in free peptides. This mechanistic understanding led to the design of two novel peptide inhibitors that bind CRM1 with picomolar affinity.


Cell Nucleus/metabolism , Karyopherins/chemistry , Karyopherins/metabolism , Nuclear Export Signals , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Active Transport, Cell Nucleus/drug effects , Amino Acid Sequence , Cell Nucleus/drug effects , Cell Survival/drug effects , Crystallography, X-Ray , HeLa Cells , Humans , Peptides/chemistry , Peptides/pharmacology , Protein Binding/drug effects , Structure-Activity Relationship , Viral Proteins/chemistry , Exportin 1 Protein
13.
Sci Rep ; 8(1): 7083, 2018 05 04.
Article En | MEDLINE | ID: mdl-29728608

ALS (Amyotrophic Lateral Sclerosis) is a neurodegenerative disease characterized by the redistribution of the RNA binding protein TDP-43 in affected neurons: from predominantly nuclear to aggregated in the cytosol. However, the determinants of TDP-43 localization and the cellular insults that promote redistribution are incompletely understood. Here, we show that the putative Nuclear Export Signal (NES) is not required for nuclear egress of TDP-43. Moreover, when the TDP-43 domain which contains the putative NES is fused to a reporter protein, YFP, the presence of the NES is not sufficient to mediate nuclear exclusion of the fusion protein. We find that the previously studied "∆NES" mutant, in which conserved hydrophobic residues are mutated to alanines, disrupts both solubility and splicing function. We further show that nuclear export of TDP-43 is independent of the exportin XPO1. Finally, we provide evidence that nuclear egress of TDP-43 is size dependent; nuclear export of dTomato TDP-43 is significantly impaired compared to Flag TDP-43. Together, these results suggest nuclear export of TDP-43 is predominantly driven by passive diffusion.


Active Transport, Cell Nucleus , DNA-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line , DNA-Binding Proteins/chemistry , Genes, Reporter , Humans , Karyopherins/chemistry , Karyopherins/metabolism , Mice , Models, Molecular , Nuclear Export Signals , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Transport , Pyramidal Cells/metabolism , Rats , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Ribonucleoside Diphosphate Reductase/chemistry , Ribonucleoside Diphosphate Reductase/metabolism , Exportin 1 Protein
14.
Cell ; 173(3): 693-705.e22, 2018 04 19.
Article En | MEDLINE | ID: mdl-29677513

Liquid-liquid phase separation (LLPS) is believed to underlie formation of biomolecular condensates, cellular compartments that concentrate macromolecules without surrounding membranes. Physical mechanisms that control condensate formation/dissolution are poorly understood. The RNA-binding protein fused in sarcoma (FUS) undergoes LLPS in vitro and associates with condensates in cells. We show that the importin karyopherin-ß2/transportin-1 inhibits LLPS of FUS. This activity depends on tight binding of karyopherin-ß2 to the C-terminal proline-tyrosine nuclear localization signal (PY-NLS) of FUS. Nuclear magnetic resonance (NMR) analyses reveal weak interactions of karyopherin-ß2 with sequence elements and structural domains distributed throughout the entirety of FUS. Biochemical analyses demonstrate that most of these same regions also contribute to LLPS of FUS. The data lead to a model where high-affinity binding of karyopherin-ß2 to the FUS PY-NLS tethers the proteins together, allowing multiple, distributed weak intermolecular contacts to disrupt FUS self-association, blocking LLPS. Karyopherin-ß2 may act analogously to control condensates in diverse cellular contexts.


Active Transport, Cell Nucleus , Nuclear Localization Signals , RNA-Binding Protein FUS/chemistry , beta Karyopherins/chemistry , Binding Sites , Frontotemporal Lobar Degeneration/metabolism , Humans , Karyopherins/metabolism , Light , Liquid-Liquid Extraction , Macromolecular Substances , Magnetic Resonance Spectroscopy , Mutation , Nephelometry and Turbidimetry , Protein Binding , Protein Domains , RNA/chemistry , Scattering, Radiation , Temperature
15.
Curr Opin Struct Biol ; 49: 36-43, 2018 04.
Article En | MEDLINE | ID: mdl-29306779

Intrinsically disordered proteins (IDPs) have critical roles in a diverse array of cellular functions. Of relevance here is that they are components of macromolecular complexes, where their conformational flexibility helps mediate interactions with binding partners. IDPs often interact with their binding partners through short sequence motifs, commonly repeated within the disordered regions. As such, multivalent interactions are common for IDPs and their binding partners within macromolecular complexes. Here we discuss the importance of IDP multivalency in three very different macromolecular assemblies: biomolecular condensates, the nuclear pore, and the cytoskeleton.


Intrinsically Disordered Proteins/metabolism , Animals , Binding Sites , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Humans , Intrinsically Disordered Proteins/chemistry , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Nuclear Pore/chemistry , Nuclear Pore/metabolism , Protein Binding , Protein Conformation
16.
Elife ; 62017 03 10.
Article En | MEDLINE | ID: mdl-28282025

Nuclear export receptor CRM1 binds highly variable nuclear export signals (NESs) in hundreds of different cargoes. Previously we have shown that CRM1 binds NESs in both polypeptide orientations (Fung et al., 2015). Here, we show crystal structures of CRM1 bound to eight additional NESs which reveal diverse conformations that range from loop-like to all-helix, which occupy different extents of the invariant NES-binding groove. Analysis of all NES structures show 5-6 distinct backbone conformations where the only conserved secondary structural element is one turn of helix that binds the central portion of the CRM1 groove. All NESs also participate in main chain hydrogen bonding with human CRM1 Lys568 side chain, which acts as a specificity filter that prevents binding of non-NES peptides. The large conformational range of NES backbones explains the lack of a fixed pattern for its 3-5 hydrophobic anchor residues, which in turn explains the large array of peptide sequences that can function as NESs.


Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Karyopherins/chemistry , Nuclear Export Signals , Receptors, Cytoplasmic and Nuclear/chemistry , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Gene Expression , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Karyopherins/genetics , Karyopherins/metabolism , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Thermodynamics , Exportin 1 Protein
17.
Elife ; 42015 Sep 08.
Article En | MEDLINE | ID: mdl-26349033

The Chromosome Region of Maintenance 1 (CRM1) protein mediates nuclear export of hundreds of proteins through recognition of their nuclear export signals (NESs), which are highly variable in sequence and structure. The plasticity of the CRM1-NES interaction is not well understood, as there are many NES sequences that seem incompatible with structures of the NES-bound CRM1 groove. Crystal structures of CRM1 bound to two different NESs with unusual sequences showed the NES peptides binding the CRM1 groove in the opposite orientation (minus) to that of previously studied NESs (plus). Comparison of minus and plus NESs identified structural and sequence determinants for NES orientation. The binding of NESs to CRM1 in both orientations results in a large expansion in NES consensus patterns and therefore a corresponding expansion of potential NESs in the proteome.


Karyopherins/chemistry , Karyopherins/metabolism , Nuclear Export Signals , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Crystallography, X-Ray , Humans , Models, Molecular , Protein Binding , Protein Conformation , Exportin 1 Protein
18.
Nat Neurosci ; 18(4): 511-20, 2015 Apr.
Article En | MEDLINE | ID: mdl-25706475

Axonal damage has been associated with aberrant protein trafficking. We examined a newly characterized class of compounds that target nucleo-cytoplasmic shuttling by binding to the catalytic groove of the nuclear export protein XPO1 (also known as CRM1, chromosome region maintenance protein 1). Oral administration of reversible CRM1 inhibitors in preclinical murine models of demyelination significantly attenuated disease progression, even when started after the onset of paralysis. Clinical efficacy was associated with decreased proliferation of immune cells, characterized by nuclear accumulation of cell cycle inhibitors, and preservation of cytoskeletal integrity even in demyelinated axons. Neuroprotection was not limited to models of demyelination, but was also observed in another mouse model of axonal damage (that is, kainic acid injection) and detected in cultured neurons after knockdown of Xpo1, the gene encoding CRM1. A proteomic screen for target molecules revealed that CRM1 inhibitors in neurons prevented nuclear export of molecules associated with axonal damage while retaining transcription factors modulating neuroprotection.


Axons , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Karyopherins/metabolism , Neuroprotective Agents/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Acrylamides/administration & dosage , Acrylamides/pharmacokinetics , Acrylamides/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Axons/drug effects , Axons/metabolism , Axons/pathology , Cell Nucleus/metabolism , Cells, Cultured , Disease Models, Animal , Disease Progression , Drug Evaluation, Preclinical , Female , Karyopherins/antagonists & inhibitors , Karyopherins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacokinetics , Proteomics , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Thiazoles/administration & dosage , Thiazoles/pharmacokinetics , Thiazoles/pharmacology , Treatment Outcome , Exportin 1 Protein
19.
Semin Cancer Biol ; 27: 52-61, 2014 Aug.
Article En | MEDLINE | ID: mdl-24631835

CRM1 or XPO1 is the major nuclear export receptor in the cell, which controls the nuclear-cytoplasmic localization of many proteins and RNAs. CRM1 is also a promising cancer drug target as the transport receptor is overexpressed in many cancers where some of its cargos are misregulated and mislocalized to the cytoplasm. Atomic level understanding of CRM1 function has greatly facilitated recent drug discovery and development of CRM1 inhibitors to target a variety of malignancies. Numerous atomic resolution CRM1 structures are now available, explaining how the exporter recognizes nuclear export signals in its cargos, how RanGTP and cargo bind with positive cooperativity, how RanBP1 causes release of export cargos in the cytoplasm and how diverse inhibitors such as Leptomycin B and the new KPT-SINE compounds block nuclear export. This review summarizes structure-function studies that explain CRM1-cargo recognition, release and inhibition.


Carrier Proteins/chemistry , Carrier Proteins/metabolism , Karyopherins/chemistry , Karyopherins/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Humans , Karyopherins/antagonists & inhibitors , Protein Binding , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Structure-Activity Relationship , ran GTP-Binding Protein/metabolism , Exportin 1 Protein
20.
J Lipid Res ; 55(3): 540-8, 2014 Mar.
Article En | MEDLINE | ID: mdl-24474819

Previous studies demonstrated that L-Fabp KO mice are more susceptible to lithogenic diet (LD)-induced gallstones because of altered hepatic cholesterol metabolism and increased canalicular cholesterol secretion. Other studies demonstrated that liver-specific deletion of microsomal triglyceride transfer protein (Mttp-LKO) reduced LD-induced gallstone formation by increasing biliary phospholipid secretion. Here we show that mice with combined deletion (i.e., DKO mice) are protected from LD-induced gallstone formation. Following 2 weeks of LD feeding, 73% of WT and 100% of L-Fabp KO mice developed gallstones versus 18% of Mttp-LKO and 23% of DKO mice. This phenotype was recapitulated in both WT and L-Fabp KO mice treated with an Mttp antisense oligonucleotide (M-ASO). Biliary cholesterol secretion was increased in LD-fed L-Fabp KO mice and decreased in DKO mice. However, phospholipid secretion was unchanged in LD-fed Mttp-LKO and DKO mice as well as in M-ASO-treated mice. Expression of the canalicular export pump ABCG5/G8 was reduced in LD-fed DKO mice and in M-ASO-treated L-Fabp KO mice. We conclude that liver-specific Mttp deletion not only eliminates apical lipoprotein secretion from hepatocytes but also attenuates canalicular cholesterol secretion, which in turn decreases LD-induced gallstone susceptibility.


Carrier Proteins/metabolism , Fatty Acid-Binding Proteins/deficiency , Gallstones/metabolism , Liver/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Bile/drug effects , Bile/metabolism , Carrier Proteins/genetics , Cholesterol/blood , Cholesterol/metabolism , Diet/adverse effects , Fatty Acid-Binding Proteins/genetics , Gallstones/etiology , Gallstones/genetics , Gene Expression/drug effects , Genetic Predisposition to Disease/etiology , Genetic Predisposition to Disease/genetics , Immunoblotting , Lipids/analysis , Lipids/blood , Lipoproteins/genetics , Lipoproteins/metabolism , Liver/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/blood , Triglycerides/metabolism
...