Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.009
Filter
1.
Nat Commun ; 15(1): 3992, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734767

ABSTRACT

Visual proteomics attempts to build atlases of the molecular content of cells but the automated annotation of cryo electron tomograms remains challenging. Template matching (TM) and methods based on machine learning detect structural signatures of macromolecules. However, their applicability remains limited in terms of both the abundance and size of the molecular targets. Here we show that the performance of TM is greatly improved by using template-specific search parameter optimization and by including higher-resolution information. We establish a TM pipeline with systematically tuned parameters for the automated, objective and comprehensive identification of structures with confidence 10 to 100-fold above the noise level. We demonstrate high-fidelity and high-confidence localizations of nuclear pore complexes, vaults, ribosomes, proteasomes, fatty acid synthases, lipid membranes and microtubules, and individual subunits inside crowded eukaryotic cells. We provide software tools for the generic implementation of our method that is broadly applicable towards realizing visual proteomics.


Subject(s)
Cryoelectron Microscopy , Electron Microscope Tomography , Proteasome Endopeptidase Complex , Proteomics , Ribosomes , Software , Electron Microscope Tomography/methods , Cryoelectron Microscopy/methods , Ribosomes/ultrastructure , Ribosomes/metabolism , Proteasome Endopeptidase Complex/ultrastructure , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/chemistry , Humans , Proteomics/methods , Nuclear Pore/ultrastructure , Nuclear Pore/metabolism , Microtubules/ultrastructure , Microtubules/metabolism , Fatty Acid Synthases/metabolism , Machine Learning , Imaging, Three-Dimensional/methods , Algorithms , Image Processing, Computer-Assisted/methods
2.
Nat Commun ; 15(1): 4358, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38778058

ABSTRACT

3C-based methods have significantly advanced our understanding of 3D genome organization. However, it remains a formidable task to precisely capture long-range chromosomal interactions between individual loci, such as those between promoters and distal enhancers. Here, we present Methyltransferase Targeting-based chromosome Architecture Capture (MTAC), a method that maps the contacts between a target site (viewpoint) and the rest of the genome in budding yeast with high resolution and sensitivity. MTAC detects hundreds of intra- and inter-chromosomal interactions within nucleosome-depleted regions (NDRs) that cannot be captured by 4C, Hi-C, or Micro-C. By applying MTAC to various viewpoints, we find that (1) most long-distance chromosomal interactions detected by MTAC reflect tethering by the nuclear pore complexes (NPCs), (2) genes co-regulated by methionine assemble into inter-chromosomal clusters near NPCs upon activation, (3) mediated by condensin, the mating locus forms a highly specific interaction with the recombination enhancer (RE) in a mating-type specific manner, and (4) correlation of MTAC signals among NDRs reveal spatial mixing and segregation of the genome. Overall, these results demonstrate MTAC as a powerful tool to resolve fine-scale long-distance chromosomal interactions and provide insights into the 3D genome organization.


Subject(s)
Chromosomes, Fungal , DNA Methylation , Nucleosomes , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Nucleosomes/metabolism , Nucleosomes/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Chromosomes, Fungal/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Chromosome Mapping/methods , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/genetics , Genome, Fungal , Promoter Regions, Genetic/genetics , Multiprotein Complexes/metabolism , Multiprotein Complexes/genetics , Nuclear Pore/metabolism , Nuclear Pore/genetics , Methyltransferases/metabolism , Methyltransferases/genetics
3.
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
4.
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
5.
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
6.
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
7.
Biophys J ; 123(9): 1085-1097, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38640928

ABSTRACT

As the main gatekeeper of the nucleocytoplasmic transport in eukaryotic cells, the nuclear pore complex (NPC) faces the daunting task of facilitating the bidirectional transport of a high volume of macromolecular cargoes while ensuring the selectivity, speed, and efficiency of this process. The competition between opposing nuclear import and export fluxes passing through the same channel is expected to pose a major challenge to transport efficiency. It has been suggested that phase separation-like radial segregation of import and export fluxes within the assembly of intrinsically disordered proteins that line the NPC pore could be a mechanism for ensuring efficient bidirectional transport. We examine the impact of radial segregation on the efficiency of bidirectional transport through the NPC using a coarse-grained computational model of the NPC. We find little evidence that radial segregation improves transport efficiency. By contrast, surprisingly, we find that NTR crowding may enhance rather than impair the efficiency of bidirectional transport although it decreases the available space in the pore. We identify mechanisms of this novel crowding-induced transport cooperativity through the self-regulation of cargo density and flux in the pore. These findings explain how the functional architecture of the NPC resolves the problem of efficient bidirectional transport, and provide inspiration for the alleviation of clogging in artificial selective nanopores.


Subject(s)
Nuclear Pore , Nuclear Pore/metabolism , Nuclear Pore/chemistry , Kinetics , Active Transport, Cell Nucleus , Models, Biological
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.
J Cell Biol ; 223(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38683248

ABSTRACT

Nucleocytoplasmic transport (NCT), the facilitated diffusion of cargo molecules between the nucleus and cytoplasm through nuclear pore complexes (NPCs), enables numerous fundamental eukaryotic cellular processes. Ran GTPase uses cellular energy in the direct form of GTP to create a gradient across the nuclear envelope (NE) that drives the majority of NCT. We report here that changes in GTP availability resulting from altered cellular physiology modulate the rate of NCT, as monitored using synthetic and natural cargo, and the dynamics of Ran itself. Cell migration, cell spreading, and/or modulation of the cytoskeleton or its connection to the nucleus alter GTP availability and thus rates of NCT, regulating RNA export and protein synthesis. These findings support a model in which changes in cellular physiology that alter GTP availability can regulate the rate of NCT, impacting fundamental cellular processes that extensively utilize NCT.


Subject(s)
Active Transport, Cell Nucleus , Guanosine Triphosphate , ran GTP-Binding Protein , Guanosine Triphosphate/metabolism , ran GTP-Binding Protein/metabolism , ran GTP-Binding Protein/genetics , Humans , Cell Nucleus/metabolism , Cell Movement , Nuclear Pore/metabolism , Nuclear Pore/genetics , Animals , Nuclear Envelope/metabolism , Cytoskeleton/metabolism , Protein Biosynthesis , Cytoplasm/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.
Front Immunol ; 15: 1330738, 2024.
Article in English | MEDLINE | ID: mdl-38449868

ABSTRACT

Nucleoporins (NUPs) are cellular effectors of human immunodeficiency virus-1 (HIV-1) replication that support nucleocytoplasmic trafficking of viral components. However, these also non-canonically function as positive effectors, promoting proviral DNA integration into the host genome and viral gene transcription, or as negative effectors by associating with HIV-1 restriction factors, such as MX2, inhibiting the replication of HIV-1. Here, we investigated the regulatory role of NUP98 on HIV-1 as we observed a lowering of its endogenous levels upon HIV-1 infection in CD4+ T cells. Using complementary experiments in NUP98 overexpression and knockdown backgrounds, we deciphered that NUP98 negatively affected HIV-1 long terminal repeat (LTR) promoter activity and lowered released virus levels. The negative effect on promoter activity was independent of HIV-1 Tat, suggesting that NUP98 prevents the basal viral gene expression. ChIP-qPCR showed NUP98 to be associated with HIV-1 LTR, with the negative regulatory element (NRE) of HIV-1 LTR playing a dominant role in NUP98-mediated lowering of viral gene transcription. Truncated mutants of NUP98 showed that the attenuation of HIV-1 LTR-driven transcription is primarily contributed by its N-terminal region. Interestingly, the virus generated from the producer cells transiently expressing NUP98 showed lower infectivity, while the virus generated from NUP98 knockdown CD4+ T cells showed higher infectivity as assayed in TZM-bl cells, corroborating the anti-HIV-1 properties of NUP98. Collectively, we show a new non-canonical function of a nucleoporin adding to the list of moonlighting host factors regulating viral infections. Downregulation of NUP98 in a host cell upon HIV-1 infection supports the concept of evolutionary conflicts between viruses and host antiviral factors.


Subject(s)
HIV-1 , Nuclear Pore Complex Proteins , Humans , Nuclear Pore Complex Proteins/genetics , Nuclear Pore/genetics , HIV Long Terminal Repeat/genetics , Gene Expression
12.
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
13.
J Cell Biol ; 223(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38456969

ABSTRACT

Coordination between nucleus and mitochondria is essential for cell survival, and thus numerous communication routes have been established between these two organelles over eukaryotic cell evolution. One route for organelle communication is via membrane contact sites, functional appositions formed by molecular tethers. We describe a novel nuclear-mitochondrial membrane contact site in the protozoan Toxoplasma gondii. We have identified specific contacts occurring at the nuclear pore and demonstrated an interaction between components of the nuclear pore and the mitochondrial protein translocon, highlighting them as molecular tethers. Genetic disruption of the nuclear pore or the TOM translocon components, TgNup503 or TgTom40, respectively, result in contact site reduction, supporting their potential involvement in this tether. TgNup503 depletion further leads to specific mitochondrial morphology and functional defects, supporting a role for nuclear-mitochondrial contacts in mediating their communication. The discovery of a contact formed through interaction between two ancient mitochondrial and nuclear complexes sets the ground for better understanding of mitochondrial-nuclear crosstalk in eukaryotes.


Subject(s)
Cell Nucleus , Mitochondria , Toxoplasma , Eukaryotic Cells , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria Associated Membranes , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Toxoplasma/cytology , Cell Nucleus/metabolism , Nuclear Envelope/metabolism , Nuclear Pore/metabolism , Protozoan 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.
Elife ; 122024 Feb 20.
Article in English | MEDLINE | ID: mdl-38376900

ABSTRACT

The nuclear pore complex (NPC) regulates the selective transport of large biomolecules through the nuclear envelope. As a model system for nuclear transport, we construct NPC mimics by functionalizing the pore walls of freestanding palladium zero-mode waveguides with the FG-nucleoporin Nsp1. This approach enables the measurement of single-molecule translocations through individual pores using optical detection. We probe the selectivity of Nsp1-coated pores by quantitatively comparing the translocation rates of the nuclear transport receptor Kap95 to the inert probe BSA over a wide range of pore sizes from 35 nm to 160 nm. Pores below 55 ± 5 nm show significant selectivity that gradually decreases for larger pores. This finding is corroborated by coarse-grained molecular dynamics simulations of the Nsp1 mesh within the pore, which suggest that leakage of BSA occurs by diffusion through transient openings within the dynamic mesh. Furthermore, we experimentally observe a modulation of the BSA permeation when varying the concentration of Kap95. The results demonstrate the potential of single-molecule fluorescence measurements on biomimetic NPCs to elucidate the principles of nuclear transport.


Subject(s)
Nanopores , Nuclear Pore , Humans , Nuclear Envelope , Biomimetics , Diffusion , Translocation, Genetic
17.
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
18.
Int J Mol Sci ; 25(2)2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38255786

ABSTRACT

Polymer-based membranes represent an irreplaceable group of materials that can be applied in a wide range of key industrial areas, from packaging to high-end technologies. Increased selectivity to transport properties or the possibility of controlling membrane permeability by external stimuli represents a key issue in current material research. In this work, we present an unconventional approach with the introduction of silver nanoparticles (AgNPs) into membrane pores, by immobilising them onto the surface of polyethyleneterephthalate (PET) foil with subsequent physical modification by means of laser and plasma radiation prior to membrane preparation. Our results showed that the surface characteristics of AgNP-decorated PET (surface morphology, AgNP content, and depth profile) affected the distribution and concentration of AgNPs in subsequent ion-track membranes. We believe that the presented approach affecting the redistribution of AgNPs in the polymer volume may open up new possibilities for the preparation of metal nanoparticle-filled polymeric membranes. The presence of AgNPs on the pore walls can facilitate the grafting of stimuli-responsive molecules onto these active sites and may contribute to the development of intelligent membranes with controllable transport properties.


Subject(s)
Metal Nanoparticles , Nuclear Pore , Silver , Polymers , Positron-Emission Tomography
19.
Curr Opin Genet Dev ; 84: 102150, 2024 02.
Article in English | MEDLINE | ID: mdl-38215626

ABSTRACT

Nuclear organization has emerged as a critical layer in the coordination of DNA repair activities. Distinct types of DNA lesions have notably been shown to relocate at the vicinity of nuclear pore complexes (NPCs), where specific repair pathways are favored, ultimately safeguarding genome integrity. Here, we review the most recent progress in this field, notably highlighting the increasingly diverse types of DNA structures undergoing repositioning, and the signaling pathways involved. We further discuss our growing knowledge of the molecular mechanisms underlying the choice of repair pathways at NPCs, and their conservation - or divergences. Intriguingly, a series of recent findings suggest that DNA metabolism may be coupled to NPC biogenesis and specialization, challenging our initial vision of these processes.


Subject(s)
DNA Repair , Nuclear Pore , Humans , Nuclear Pore/genetics , DNA Repair/genetics , DNA Damage/genetics , Genomic Instability/genetics , DNA/metabolism , Nuclear Envelope
20.
Nucleus ; 15(1): 2299632, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38238284

ABSTRACT

The nuclear envelope (NE) regulates nuclear functions, including transcription, nucleocytoplasmic transport, and protein quality control. While the outer membrane of the NE is directly continuous with the endoplasmic reticulum (ER), the NE has an overall distinct protein composition from the ER, which is crucial for its functions. During open mitosis in higher eukaryotes, the NE disassembles during mitotic entry and then reforms as a functional territory at the end of mitosis to reestablish nucleocytoplasmic compartmentalization. In this review, we examine the known mechanisms by which the functional NE reconstitutes from the mitotic ER in the continuous ER-NE endomembrane system during open mitosis. Furthermore, based on recent findings indicating that the NE possesses unique lipid metabolism and quality control mechanisms distinct from those of the ER, we explore the maintenance of NE identity and homeostasis during interphase. We also highlight the potential significance of membrane junctions between the ER and NE.


Subject(s)
Nuclear Envelope , Nuclear Pore , Nuclear Envelope/metabolism , Nuclear Pore/metabolism , Endoplasmic Reticulum/metabolism , Mitosis , Active Transport, Cell Nucleus
SELECTION OF CITATIONS
SEARCH DETAIL
...