Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.860
Filter
1.
Sci Adv ; 10(23): eadn8963, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38838144

ABSTRACT

Nucleoporins, the components of nuclear pore complexes (NPCs), can play cell type- and tissue-specific functions. Yet, the physiological roles and mechanisms of action for most NPC components have not yet been established. We report that Nup358, a nucleoporin linked to several myeloid disorders, is required for the developmental progression of early myeloid progenitors. We found that Nup358 ablation in mice results in the loss of myeloid-committed progenitors and mature myeloid cells and the accumulation of myeloid-primed multipotent progenitors (MPPs) in bone marrow. Accumulated MPPs in Nup358 knockout mice are greatly restricted to megakaryocyte/erythrocyte-biased MPP2, which fail to progress into committed myeloid progenitors. Mechanistically, we found that Nup358 is required for histone deacetylase 3 (HDAC3) nuclear import and function in MPP2 cells and established that this nucleoporin regulates HDAC3 nuclear translocation in a SUMOylation-independent manner. Our study identifies a critical function for Nup358 in myeloid-primed MPP2 differentiation and uncovers an unexpected role for NPCs in the early steps of myelopoiesis.


Subject(s)
Cell Differentiation , Histone Deacetylases , Mice, Knockout , Nuclear Pore Complex Proteins , Animals , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Nuclear Pore Complex Proteins/metabolism , Nuclear Pore Complex Proteins/genetics , Mice , Molecular Chaperones/metabolism , Molecular Chaperones/genetics , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/cytology , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Multipotent Stem Cells/metabolism , Multipotent Stem Cells/cytology , Myeloid Cells/metabolism , Myeloid Cells/cytology , Sumoylation , Myelopoiesis/genetics
2.
Methods Mol Biol ; 2807: 141-151, 2024.
Article in English | MEDLINE | ID: mdl-38743226

ABSTRACT

To integrate with host chromatin and establish a productive infection, HIV-1 must translocate the viral Ribonucleoprotein (RNP) complex through the nuclear pore complex (NPC). Current assay to measure HIV-1 nuclear import relies on a transient byproduct of HIV-1 integration failure called 2-LTR circles. However, 2-LTR circles require complete or near-complete reverse transcription and association with the non-homologous end joining (NHEJ) machinery in the nucleus, which can complicate interpretation of 2-LTR circle formation as a measure of nuclear import kinetics. Here, we describe an approach to measure nuclear import of infectious HIV-1 particles. This involves chemically induced dimerization of Nup62, a central FG containing nucleoporin. Using this technique, nuclear import of infectious particles can be monitored in both primary and cell culture models. In response to host factor depletion or restriction factors, changes in HIV-1 nuclear import can be effectively measured using the nuclear import kinetics (NIK) assay.


Subject(s)
Active Transport, Cell Nucleus , HIV-1 , Nuclear Pore Complex Proteins , Nuclear Pore , HIV-1/metabolism , HIV-1/physiology , Humans , Nuclear Pore/metabolism , Nuclear Pore Complex Proteins/metabolism , Kinetics , Cell Nucleus/metabolism , HIV Infections/virology , HIV Infections/metabolism , Virus Integration
3.
Nature ; 629(8014): 1126-1132, 2024 May.
Article in English | MEDLINE | ID: mdl-38750356

ABSTRACT

Plants exposed to incidences of excessive temperatures activate heat-stress responses to cope with the physiological challenge and stimulate long-term acclimation1,2. The mechanism that senses cellular temperature for inducing thermotolerance is still unclear3. Here we show that TWA1 is a temperature-sensing transcriptional co-regulator that is needed for basal and acquired thermotolerance in Arabidopsis thaliana. At elevated temperatures, TWA1 changes its conformation and allows physical interaction with JASMONATE-ASSOCIATED MYC-LIKE (JAM) transcription factors and TOPLESS (TPL) and TOPLESS-RELATED (TPR) proteins for repressor complex assembly. TWA1 is a predicted intrinsically disordered protein that has a key thermosensory role functioning through an amino-terminal highly variable region. At elevated temperatures, TWA1 accumulates in nuclear subdomains, and physical interactions with JAM2 and TPL appear to be restricted to these nuclear subdomains. The transcriptional upregulation of the heat shock transcription factor A2 (HSFA2) and heat shock proteins depended on TWA1, and TWA1 orthologues provided different temperature thresholds, consistent with the sensor function in early signalling of heat stress. The identification of the plant thermosensors offers a molecular tool for adjusting thermal acclimation responses of crops by breeding and biotechnology, and a sensitive temperature switch for thermogenetics.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Intrinsically Disordered Proteins , Temperature , Thermosensing , Thermotolerance , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis/physiology , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Nucleus/metabolism , Gene Expression Regulation, Plant , Heat Shock Transcription Factors/metabolism , Heat Shock Transcription Factors/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Nuclear Pore Complex Proteins/metabolism , Repressor Proteins/metabolism , Thermosensing/genetics , Thermosensing/physiology , Thermotolerance/genetics , Thermotolerance/physiology , Transcription Factors/metabolism , Signal Transduction
4.
J Chem Inf Model ; 64(10): 4149-4157, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38713459

ABSTRACT

A substantial portion of various organisms' proteomes comprises intrinsically disordered proteins (IDPs) that lack a defined three-dimensional structure. These IDPs exhibit a diverse array of conformations, displaying remarkable spatiotemporal heterogeneity and exceptional conformational flexibility. Characterizing the structure or structural ensemble of IDPs presents significant conceptual and methodological challenges owing to the absence of a well-defined native structure. While databases such as the Protein Ensemble Database (PED) provide IDP ensembles obtained through a combination of experimental data and molecular modeling, the absence of reaction coordinates poses challenges in comprehensively understanding pertinent aspects of the system. In this study, we leverage the energy landscape visualization method (JCTC, 6482, 2019) to scrutinize four IDP ensembles sourced from PED. ELViM, a methodology that circumvents the need for a priori reaction coordinates, aids in analyzing the ensembles. The specific IDP ensembles investigated are as follows: two fragments of nucleoporin (NUL: 884-993 and NUS: 1313-1390), yeast sic 1 N-terminal (1-90), and the N-terminal SH3 domain of Drk (1-59). Utilizing ELViM enables the comprehensive validation of ensembles, facilitating the detection of potential inconsistencies in the sampling process. Additionally, it allows for identifying and characterizing the most prevalent conformations within an ensemble. Moreover, ELViM facilitates the comparative analysis of ensembles obtained under diverse conditions, thereby providing a powerful tool for investigating the functional mechanisms of IDPs.


Subject(s)
Intrinsically Disordered Proteins , Protein Conformation , Intrinsically Disordered Proteins/chemistry , Thermodynamics , Models, Molecular , Databases, Protein , Saccharomyces cerevisiae Proteins/chemistry , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor Proteins
5.
Nat Commun ; 15(1): 3797, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714656

ABSTRACT

Nucleoporins rich in phenylalanine/glycine (FG) residues form the permeability barrier within the nuclear pore complex and are implicated in several pathological cellular processes, including oncogenic fusion condensates. The self-association of FG-repeat proteins and interactions between FG-repeats play a critical role in these activities by forming hydrogel-like structures. Here we show that mutation of specific FG repeats of Nup98 can strongly decrease the protein's self-association capabilities. We further present a cryo-electron microscopy structure of a Nup98 peptide fibril with higher stability per residue compared with previous Nup98 fibril structures. The high-resolution structure reveals zipper-like hydrophobic patches which contain a GLFG motif and are less compatible for binding to nuclear transport receptors. The identified distinct molecular properties of different regions of the nucleoporin may contribute to spatial variations in the self-association of FG-repeats, potentially influencing transport processes through the nuclear pore.


Subject(s)
Cryoelectron Microscopy , Nuclear Pore Complex Proteins , Nuclear Pore Complex Proteins/metabolism , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/ultrastructure , Humans , Mutation , Nuclear Pore/metabolism , Nuclear Pore/ultrastructure , Nuclear Pore/chemistry , Glycine/chemistry , Glycine/metabolism , Phenylalanine/chemistry , Phenylalanine/metabolism , Repetitive Sequences, Amino Acid , Protein Binding , Models, Molecular , Hydrophobic and Hydrophilic Interactions
6.
Cell Host Microbe ; 32(4): 441-442, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38604120

ABSTRACT

The size of the nuclear pore should, in principle, prevent HIV-1 entry. However, HIV-1 capsid is able to gain nuclear pore entry. In a recent issue of Nature, Fu et al. and Dickson et al. demonstrate that the HIV-1 capsid mimics the nuclear transport protein karyopherins to access host nuclei.


Subject(s)
HIV Infections , Nuclear Pore , Humans , Capsid/metabolism , Capsid Proteins/metabolism , HIV Infections/metabolism , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Nuclear Pore Complex Proteins/metabolism
7.
Nucleus ; 15(1): 2310452, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38605598

ABSTRACT

The nuclear envelope (NE) separates translation and transcription and is the location of multiple functions, including chromatin organization and nucleocytoplasmic transport. The molecular basis for many of these functions have diverged between eukaryotic lineages. Trypanosoma brucei, a member of the early branching eukaryotic lineage Discoba, highlights many of these, including a distinct lamina and kinetochore composition. Here, we describe a cohort of proteins interacting with both the lamina and NPC, which we term lamina-associated proteins (LAPs). LAPs represent a diverse group of proteins, including two candidate NPC-anchoring pore membrane proteins (POMs) with architecture conserved with S. cerevisiae and H. sapiens, and additional peripheral components of the NPC. While many of the LAPs are Kinetoplastid specific, we also identified broadly conserved proteins, indicating an amalgam of divergence and conservation within the trypanosome NE proteome, highlighting the diversity of nuclear biology across the eukaryotes, increasing our understanding of eukaryotic and NPC evolution.


Subject(s)
Nuclear Envelope , Trypanosoma , Humans , Nuclear Envelope/metabolism , Nuclear Pore/metabolism , Saccharomyces cerevisiae/metabolism , Nuclear Pore Complex Proteins/metabolism , Trypanosoma/metabolism
8.
J Cell Biol ; 223(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38652117

ABSTRACT

Assembly of macromolecular complexes at correct cellular sites is crucial for cell function. Nuclear pore complexes (NPCs) are large cylindrical assemblies with eightfold rotational symmetry, built through hierarchical binding of nucleoporins (Nups) forming distinct subcomplexes. Here, we uncover a role of ubiquitin-associated protein 2-like (UBAP2L) in the assembly and stability of properly organized and functional NPCs at the intact nuclear envelope (NE) in human cells. UBAP2L localizes to the nuclear pores and facilitates the formation of the Y-complex, an essential scaffold component of the NPC, and its localization to the NE. UBAP2L promotes the interaction of the Y-complex with POM121 and Nup153, the critical upstream factors in a well-defined sequential order of Nups assembly onto NE during interphase. Timely localization of the cytoplasmic Nup transport factor fragile X-related protein 1 (FXR1) to the NE and its interaction with the Y-complex are likewise dependent on UBAP2L. Thus, this NPC biogenesis mechanism integrates the cytoplasmic and the nuclear NPC assembly signals and ensures efficient nuclear transport, adaptation to nutrient stress, and cellular proliferative capacity, highlighting the importance of NPC homeostasis at the intact NE.


Subject(s)
Carrier Proteins , Nuclear Envelope , Nuclear Pore , Humans , Active Transport, Cell Nucleus , HeLa Cells , Homeostasis , Membrane Glycoproteins , Nuclear Envelope/metabolism , Nuclear Pore/metabolism , Nuclear Pore Complex Proteins/metabolism , Carrier Proteins/metabolism
9.
ACS Chem Neurosci ; 15(10): 1967-1989, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38657106

ABSTRACT

Disturbances in protein phase transitions promote protein aggregation─a neurodegeneration hallmark. The modular Ran-binding protein 2 (Ranbp2) is a cytosolic molecular hub for rate-limiting steps of phase transitions of Ran-GTP-bound protein ensembles exiting nuclear pores. Chaperones also regulate phase transitions and proteostasis by suppressing protein aggregation. Ranbp2 haploinsufficiency promotes the age-dependent neuroprotection of the chorioretina against phototoxicity by proteostatic regulations of neuroprotective substrates of Ranbp2 and by suppressing the buildup of polyubiquitylated substrates. Losses of peptidyl-prolyl cis-trans isomerase (PPIase) and chaperone activities of the cyclophilin domain (CY) of Ranbp2 recapitulate molecular effects of Ranbp2 haploinsufficiency. These CY impairments also stimulate deubiquitylation activities and phase transitions of 19S cap subunits of the 26S proteasome that associates with Ranbp2. However, links between CY moonlighting activity, substrate ubiquitylation, and proteostasis remain incomplete. Here, we reveal the Ranbp2 regulation of small heat shock chaperones─crystallins in the chorioretina by proteomics of mice with total or selective modular deficits of Ranbp2. Specifically, loss of CY PPIase of Ranbp2 upregulates αA-Crystallin, which is repressed in adult nonlenticular tissues. Conversely, impairment of CY's chaperone activity opposite to the PPIase pocket downregulates a subset of αA-Crystallin's substrates, γ-crystallins. These CY-dependent effects cause age-dependent and chorioretinal-selective declines of ubiquitylated substrates without affecting the chorioretinal morphology. A model emerges whereby inhibition of Ranbp2's CY PPIase remodels crystallins' expressions, subdues molecular aging, and preordains the chorioretina to neuroprotection by augmenting the chaperone capacity and the degradation of polyubiquitylated substrates against proteostatic impairments. Further, the druggable Ranbp2 CY holds pan-therapeutic potential against proteotoxicity and neurodegeneration.


Subject(s)
Cyclophilins , Molecular Chaperones , Nuclear Pore Complex Proteins , Peptidylprolyl Isomerase , Proteostasis , Animals , Molecular Chaperones/metabolism , Mice , Cyclophilins/metabolism , Proteostasis/physiology , Peptidylprolyl Isomerase/metabolism , Nuclear Pore Complex Proteins/metabolism , Crystallins/metabolism
10.
EMBO J ; 43(11): 2198-2232, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38649536

ABSTRACT

Nuclear pore complex (NPC) biogenesis is a still enigmatic example of protein self-assembly. We now introduce several cross-reacting anti-Nup nanobodies for imaging intact nuclear pore complexes from frog to human. We also report a simplified assay that directly tracks postmitotic NPC assembly with added fluorophore-labeled anti-Nup nanobodies. During interphase, NPCs are inserted into a pre-existing nuclear envelope. Monitoring this process is challenging because newly assembled NPCs are indistinguishable from pre-existing ones. We overcame this problem by inserting Xenopus-derived NPCs into human nuclear envelopes and using frog-specific anti-Nup nanobodies for detection. We further asked whether anti-Nup nanobodies could serve as NPC assembly inhibitors. Using a selection strategy against conserved epitopes, we obtained anti-Nup93, Nup98, and Nup155 nanobodies that block Nup-Nup interfaces and arrest NPC assembly. We solved structures of nanobody-target complexes and identified roles for the Nup93 α-solenoid domain in recruiting Nup358 and the Nup214·88·62 complex, as well as for Nup155 and the Nup98 autoproteolytic domain in NPC scaffold assembly. The latter suggests a checkpoint linking pore formation to the assembly of the Nup98-dominated permeability barrier.


Subject(s)
Nuclear Pore Complex Proteins , Nuclear Pore , Single-Domain Antibodies , Nuclear Pore Complex Proteins/metabolism , Nuclear Pore/metabolism , Humans , Single-Domain Antibodies/metabolism , Animals , Xenopus , Xenopus laevis , HeLa Cells
11.
EMBO Rep ; 25(5): 2391-2417, 2024 May.
Article in English | MEDLINE | ID: mdl-38605278

ABSTRACT

ELYS is a nucleoporin that localizes to the nuclear side of the nuclear pore complex (NPC) in interphase cells. In mitosis, it serves as an assembly platform that interacts with chromatin and then with nucleoporin subcomplexes to initiate post-mitotic NPC assembly. Here we identify ELYS as a major binding partner of the membrane protein VAPB during mitosis. In mitosis, ELYS becomes phosphorylated at many sites, including a predicted FFAT (two phenylalanines in an acidic tract) motif, which mediates interaction with the MSP (major sperm protein)-domain of VAPB. Binding assays using recombinant proteins or cell lysates and co-immunoprecipitation experiments show that VAPB binds the FFAT motif of ELYS in a phosphorylation-dependent manner. In anaphase, the two proteins co-localize to the non-core region of the newly forming nuclear envelope. Depletion of VAPB results in prolonged mitosis, slow progression from meta- to anaphase and in chromosome segregation defects. Together, our results suggest a role of VAPB in mitosis upon recruitment to or release from ELYS at the non-core region of the chromatin in a phosphorylation-dependent manner.


Subject(s)
Mitosis , Protein Binding , Humans , Phosphorylation , HeLa Cells , Chromatin/metabolism , Transcription Factors/metabolism , Chromosome Segregation , Nuclear Pore Complex Proteins/metabolism , Nuclear Envelope/metabolism , Membrane Proteins/metabolism , Anaphase
12.
PLoS Pathog ; 20(3): e1011830, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38512975

ABSTRACT

Human myxovirus resistance 2 (MX2/MXB) is an interferon-induced GTPase that inhibits human immunodeficiency virus-1 (HIV-1) infection by preventing nuclear import of the viral preintegration complex. The HIV-1 capsid (CA) is the major viral determinant for sensitivity to MX2, and complex interactions between MX2, CA, nucleoporins (Nups), cyclophilin A (CypA), and other cellular proteins influence the outcome of viral infection. To explore the interactions between MX2, the viral CA, and CypA, we utilized a CRISPR-Cas9/AAV approach to generate CypA knock-out cell lines as well as cells that express CypA from its endogenous locus, but with specific point mutations that would abrogate CA binding but should not affect enzymatic activity or cellular function. We found that infection of CypA knock-out and point mutant cell lines with wild-type HIV-1 and CA mutants recapitulated the phenotypes observed upon cyclosporine A (CsA) addition, indicating that effects of CsA treatment are the direct result of blocking CA-CypA interactions and are therefore independent from potential interactions between CypA and MX2 or other cellular proteins. Notably, abrogation of GTP hydrolysis by MX2 conferred enhanced antiviral activity when CA-CypA interactions were abolished, and this effect was not mediated by the CA-binding residues in the GTPase domain, or by phosphorylation of MX2 at position T151. We additionally found that elimination of GTPase activity also altered the Nup requirements for MX2 activity. Our data demonstrate that the antiviral activity of MX2 is affected by CypA-CA interactions in a virus-specific and GTPase activity-dependent manner. These findings further highlight the importance of the GTPase domain of MX2 in regulation of substrate specificity and interaction with nucleocytoplasmic trafficking pathways.


Subject(s)
Capsid , Nuclear Pore Complex Proteins , Humans , Capsid/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Cyclophilin A/genetics , Cyclophilin A/metabolism , GTP Phosphohydrolases/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Antiviral Agents/metabolism , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/metabolism
13.
Mol Biol Cell ; 35(5): ar62, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38507240

ABSTRACT

The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) accessory protein Orf6 works as an interferon antagonist, in part, by inhibiting the nuclear import activated p-STAT1, an activator of interferon-stimulated genes, and the export of the poly(A) RNA. Insight into the transport regulatory function of Orf6 has come from the observation that Orf6 binds to the nuclear pore complex (NPC) components: Rae1 and Nup98. To gain further insight into the mechanism of Orf6-mediated transport inhibition, we examined the role of Rae1 and Nup98. We show that Rae1 alone is not necessary to support p-STAT1 import or nuclear export of poly(A) RNA. Moreover, the loss of Rae1 suppresses the transport inhibitory activity of Orf6. We propose that the Rae1/Nup98 complex strategically positions Orf6 within the NPC where it alters FG-Nup interactions and their ability to support nuclear transport. In addition, we show that Rae1 is required for normal viral protein production during SARS-CoV-2 infection presumably through its role in supporting Orf6 function.


Subject(s)
Active Transport, Cell Nucleus , COVID-19 , Nuclear Pore , Nucleocytoplasmic Transport Proteins , SARS-CoV-2 , Humans , COVID-19/metabolism , Interferons/metabolism , Nuclear Pore/metabolism , Nuclear Pore Complex Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , RNA, Messenger/metabolism , SARS-CoV-2/metabolism , Viral Proteins/metabolism , Nuclear Matrix-Associated Proteins/metabolism
14.
Genetics ; 226(4)2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38302116

ABSTRACT

The nuclear pore complex (NPC) mediates the selective exchange of macromolecules between the nucleus and the cytoplasm. Neurodegenerative diseases such as amyotrophic lateral sclerosis are characterized by mislocalization of nucleoporins (Nups), transport receptors, and Ras-related nuclear proteins into nucleoplasmic or cytosolic aggregates, underscoring the importance of precise assembly of the NPC. The assembly state of large protein complexes is strictly monitored by the protein quality control system. The ubiquitin-proteasome system may eliminate aberrant, misfolded, and/or orphan components; however, the involvement of the ubiquitin-proteasome system in the degradation of nonnative Nups in the NPC remains unclear. Here, we show that in Saccharomyces cerevisiae, although Nup1 (the FG-Nup component of the central core of the NPC) was stable, C-terminally green fluorescent protein-tagged Nup1, which had been incorporated into the NPC, was degraded by the proteasome especially under heat stress conditions. The degradation was dependent on the San1 ubiquitin ligase and Cdc48/p97, as well as its cofactor Doa1. We also demonstrate that San1 weakly but certainly contributes to the degradation of nontagged endogenous Nup1 in cells defective in NPC biogenesis by the deletion of NUP120. In addition, the overexpression of SAN1 exacerbated the growth defect phenotype of nup120Δ cells, which may be caused by excess degradation of defective Nups due to the deletion of NUP120. These biochemical and genetic data suggest that San1 is involved in the degradation of nonnative Nups generated by genetic mutation or when NPC biogenesis is impaired.


Subject(s)
Proteasome Endopeptidase Complex , Saccharomyces cerevisiae Proteins , Nuclear Pore/genetics , Nuclear Pore/chemistry , Nuclear Pore/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin/analysis , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
15.
Nucleus ; 15(1): 2314297, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38383349

ABSTRACT

The separation of genetic material from bulk cytoplasm has enabled the evolution of increasingly complex organisms, allowing for the development of sophisticated forms of life. However, this complexity has created new categories of dysfunction, including those related to the movement of material between cellular compartments. In eukaryotic cells, nucleocytoplasmic trafficking is a fundamental biological process, and cumulative disruptions to nuclear integrity and nucleocytoplasmic transport are detrimental to cell survival. This is particularly true in post-mitotic neurons, where nuclear pore injury and errors to nucleocytoplasmic trafficking are strongly associated with neurodegenerative disease. In this review, we summarize the current understanding of nuclear pore biology in physiological and pathological contexts and discuss potential therapeutic approaches for addressing nuclear pore injury and dysfunctional nucleocytoplasmic transport.


Subject(s)
Neurodegenerative Diseases , Nuclear Pore , Humans , Nuclear Pore/metabolism , Cell Nucleus/metabolism , Neurodegenerative Diseases/metabolism , Active Transport, Cell Nucleus/physiology , Nuclear Pore Complex Proteins/metabolism
16.
Nature ; 626(8000): 843-851, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38267583

ABSTRACT

HIV-1 infection requires nuclear entry of the viral genome. Previous evidence suggests that this entry proceeds through nuclear pore complexes (NPCs), with the 120 × 60 nm capsid squeezing through an approximately 60-nm-wide central channel1 and crossing the permeability barrier of the NPC. This barrier can be described as an FG phase2 that is assembled from cohesively interacting phenylalanine-glycine (FG) repeats3 and is selectively permeable to cargo captured by nuclear transport receptors (NTRs). Here we show that HIV-1 capsid assemblies can target NPCs efficiently in an NTR-independent manner and bind directly to several types of FG repeats, including barrier-forming cohesive repeats. Like NTRs, the capsid readily partitions into an in vitro assembled cohesive FG phase that can serve as an NPC mimic and excludes much smaller inert probes such as mCherry. Indeed, entry of the capsid protein into such an FG phase is greatly enhanced by capsid assembly, which also allows the encapsulated clients to enter. Thus, our data indicate that the HIV-1 capsid behaves like an NTR, with its interior serving as a cargo container. Because capsid-coating with trans-acting NTRs would increase the diameter by 10 nm or more, we suggest that such a 'self-translocating' capsid undermines the size restrictions imposed by the NPC scaffold, thereby bypassing an otherwise effective barrier to viral infection.


Subject(s)
Capsid Proteins , Capsid , Glycine , HIV-1 , Nuclear Pore Complex Proteins , Nuclear Pore , Phenylalanine , Humans , Active Transport, Cell Nucleus , Capsid/chemistry , Capsid/metabolism , Glycine/metabolism , HIV-1/chemistry , HIV-1/genetics , HIV-1/metabolism , Nuclear Pore/chemistry , Nuclear Pore/metabolism , Nuclear Pore/virology , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/metabolism , Permeability , Phenylalanine/metabolism , Solubility , Virus Internalization , Capsid Proteins/chemistry , Capsid Proteins/metabolism
17.
Proc Natl Acad Sci U S A ; 121(4): e2313737121, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38241438

ABSTRACT

Nuclear import and uncoating of the viral capsid are critical steps in the HIV-1 life cycle that serve to transport and release genomic material into the nucleus. Viral core import involves translocating the HIV-1 capsid at the nuclear pore complex (NPC). Notably, the central channel of the NPC appears to often accommodate and allow passage of intact HIV-1 capsid, though mechanistic details of the process remain to be fully understood. Here, we investigate the molecular interactions that operate in concert between the HIV-1 capsid and the NPC that regulate capsid translocation through the central channel. To this end, we develop a "bottom-up" coarse-grained (CG) model of the human NPC from recently released cryo-electron tomography structure and then construct composite membrane-embedded CG NPC models. We find that successful translocation from the cytoplasmic side to the NPC central channel is contingent on the compatibility of the capsid morphology and channel dimension and the proper orientation of the capsid approach to the channel from the cytoplasmic side. The translocation dynamics is driven by maximizing the contacts between phenylalanine-glycine nucleoporins at the central channel and the capsid. For the docked intact capsids, structural analysis reveals correlated striated patterns of lattice disorder likely related to the intrinsic capsid elasticity. Uncondensed genomic material inside the docked capsid augments the overall lattice disorder of the capsid. Our results suggest that the intrinsic "elasticity" can also aid the capsid to adapt to the stress and remain structurally intact during translocation.


Subject(s)
Capsid , HIV-1 , Humans , Capsid/metabolism , HIV-1/genetics , Nuclear Pore/metabolism , Capsid Proteins/genetics , Active Transport, Cell Nucleus , Nuclear Pore Complex Proteins/metabolism , Translocation, Genetic , Elasticity
18.
Cell Death Dis ; 15(1): 7, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38177114

ABSTRACT

Manipulation of the subcellular localization of transcription factors by preventing their shuttling via the nuclear pore complex (NPC) emerges as a novel therapeutic strategy against cancer. One transmembrane component of the NPC is POM121, encoded by a tandem gene locus POM121A/C on chromosome 7. Overexpression of POM121 is associated with metabolic diseases (e.g., diabetes) and unfavorable clinical outcome in patients with colorectal cancer (CRC). Peroxisome proliferator-activated receptor-gamma (PPARγ) is a transcription factor with anti-diabetic and anti-tumoral efficacy. It is inhibited by export from the nucleus to the cytosol via the RAS-RAF-MEK1/2-ERK1/2 signaling pathway, a major oncogenic driver of CRC. We therefore hypothesized that POM121 participates in the transport of PPARγ across the NPC to regulate its transcriptional activity on genes involved in metabolic and tumor control. We found that POM121A/C mRNA was enriched and POM121 protein co-expressed with PPARγ in tissues from CRC patients conferring poor prognosis. Its interactome was predicted to include proteins responsible for tumor metabolism and immunity, and in-silico modeling provided insights into potential 3D structures of POM121. A peptide region downstream of the nuclear localization sequence (NLS) of POM121 was identified as a cytoplasmic interactor of PPARγ. POM121 positivity correlated with the cytoplasmic localization of PPARγ in patients with KRAS mutant CRC. In contrast, POM121A/C silencing by CRISPR/Cas9 sgRNA or siRNA enforced nuclear accumulation of PPARγ and activated PPARγ target genes promoting lipid metabolism and cell cycle arrest resulting in reduced proliferation of human CRC cells. Our data suggest the POM121-PPARγ axis as a potential drugable target in CRC.


Subject(s)
Neoplasms , Nuclear Pore , Humans , Nuclear Pore/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , RNA, Guide, CRISPR-Cas Systems , Nuclear Pore Complex Proteins/metabolism , Transcription Factors/metabolism , Neoplasms/metabolism , Membrane Glycoproteins/metabolism
19.
Dev Cell ; 59(4): 482-495.e6, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38272027

ABSTRACT

Mutations or dysregulation of nucleoporins (Nups) are strongly associated with neural developmental diseases, yet the underlying mechanisms remain poorly understood. Here, we show that depletion of Nup Seh1 in radial glial progenitors results in defective neural progenitor proliferation and differentiation that ultimately manifests in impaired neurogenesis and microcephaly. This loss of stem cell proliferation is not associated with defects in the nucleocytoplasmic transport. Rather, transcriptome analysis showed that ablation of Seh1 in neural stem cells derepresses the expression of p21, and knockdown of p21 partially restored self-renewal capacity. Mechanistically, Seh1 cooperates with the NuRD transcription repressor complex at the nuclear periphery to regulate p21 expression. Together, these findings identified that Nups regulate brain development by exerting a chromatin-associated role and affecting neural stem cell proliferation.


Subject(s)
Neocortex , Neural Stem Cells , Animals , Mice , Cell Differentiation , Gene Expression , Neocortex/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism
20.
Nature ; 626(8000): 836-842, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38267582

ABSTRACT

HIV can infect non-dividing cells because the viral capsid can overcome the selective barrier of the nuclear pore complex and deliver the genome directly into the nucleus1,2. Remarkably, the intact HIV capsid is more than 1,000 times larger than the size limit prescribed by the diffusion barrier of the nuclear pore3. This barrier in the central channel of the nuclear pore is composed of intrinsically disordered nucleoporin domains enriched in phenylalanine-glycine (FG) dipeptides. Through multivalent FG interactions, cellular karyopherins and their bound cargoes solubilize in this phase to drive nucleocytoplasmic transport4. By performing an in vitro dissection of the nuclear pore complex, we show that a pocket on the surface of the HIV capsid similarly interacts with FG motifs from multiple nucleoporins and that this interaction licences capsids to penetrate FG-nucleoporin condensates. This karyopherin mimicry model addresses a key conceptual challenge for the role of the HIV capsid in nuclear entry and offers an explanation as to how an exogenous entity much larger than any known cellular cargo may be able to non-destructively breach the nuclear envelope.


Subject(s)
Capsid Proteins , Glycine , HIV , Karyopherins , Molecular Mimicry , Nuclear Pore Complex Proteins , Nuclear Pore , Phenylalanine , Humans , Active Transport, Cell Nucleus , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Diffusion , Dipeptides/chemistry , Dipeptides/metabolism , Glycine/metabolism , HIV/chemistry , HIV/metabolism , In Vitro Techniques , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Karyopherins/metabolism , Nuclear Pore/chemistry , Nuclear Pore/metabolism , Nuclear Pore/virology , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/metabolism , Permeability , Phenylalanine/metabolism , Solubility , Virus Internalization , Capsid/chemistry , Capsid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...