bims-nucpor Biomed News
on Nuclear pore complex and nucleoporins in stress, aging and disease
Issue of 2022‒05‒15
fourteen papers selected by
Sara Mingu
Johannes Gutenberg University


  1. Cells. 2022 Apr 25. pii: 1456. [Epub ahead of print]11(9):
      Nuclear pore complexes (NPCs) are the only transport channels that cross the nuclear envelope. Constructed from ~500-1000 nucleoporin proteins each, they are among the largest macromolecular assemblies in eukaryotic cells. Thanks to advances in structural analysis approaches, the construction principles and architecture of the NPC have recently been revealed at submolecular resolution. Although the overall structure and inventory of nucleoporins are conserved, NPCs exhibit significant compositional and functional plasticity even within single cells and surprising variability in their assembly pathways. Once assembled, NPCs remain seemingly unexchangeable in post-mitotic cells. There are a number of as yet unresolved questions about how the versatility of NPC assembly and composition is established, how cells monitor the functional state of NPCs or how they could be renewed. Here, we review current progress in our understanding of the key aspects of NPC architecture and lifecycle.
    Keywords:  Brl1; FG repeats; NPC; Ran; ageing; aggregation; amphipathic helix; assembly factor; autophagy; lipids; membrane fusion; neurodegneration; nuclear pore complex; nuclear transport receptor; nucleoporin
    DOI:  https://doi.org/10.3390/cells11091456
  2. Cell Cycle. 2022 May 13. 1-10
      Nup50 is nuclear pore complex component localized to the nuclear side of the pore and in the nucleoplasm. It has been characterized as an auxiliary factor in nuclear transport reactions. Our recent work indicates that it interacts with and stimulates RCC1, the sole guanine nucleotide exchange factor for the GTPase Ran. Here, we discuss how this interaction might contribute to Nup50 function in nuclear transport but also its other functions like control of gene expression, cell cycle and DNA damage repair.
    Keywords:  DNA repair; Nup50; RCC1; cell cycle; nuclear pore assembly; nuclear trafficking; p27Kip1
    DOI:  https://doi.org/10.1080/15384101.2022.2074742
  3. FASEB J. 2022 May;36 Suppl 1
      Nuclear pore complexes (NPCs) mediate nucleocytoplasmic exchange controlling the flow of molecules into and out of the nucleus. The selective filter properties of NPCs enable translocation of specific molecules known as nuclear transport factors (NTRs) and their cargo. Central to this selectivity barrier is a group of largely intrinsically disordered nucleoporins (Nups) that contain multiple phenylalanine-glycine repeats, termed FG Nups. The interactions between FG Nups and NTRs enable NTRs to translocate rapidly yet selectively through the NPC in what is referred to as the "transport paradox". FG Nups in the NPC have a small folded anchor region tethering them to the NPC outer ring and otherwise are fully disordered, random coil polymers that remain predominantly disordered while engaged to NTRs. FG Nups interact with NTRs using mainly their FG motifs and minimally their intervening spacer residues. The overall enthalpy of the interaction increases as multivalency increases the frequency of individually weak FG-NTR contacts. Tight binding is limited by an entropy penalty that disfavors simultaneous engagements of FG motifs. Small angle neutron scattering (SANS) shows that the entropy loss is partly due to the local rigidity of an FG motif in the interacting state(Fig 1). All-atom molecular dynamics (MD) simulation indicates that spacers between the FG motifs behave as "entropic springs", disfavoring any static binding of the FG repeats. The dynamics of FG Nups enables the FG motifs to slide along the hydrophobic patches of NTRs enabling FG motifs to be easily displaced by other competing FG motifs (Fig. 2). This explanation provides a simple hypothesis for the rapid exchange of FG motif contacts during transport, focusing on the entropic exclusion of non-NTRs, and 'solubilization' of NTR complexes, in contrast to possible condensate formation which would provide a compartmentalization of components. These results reveal fundamental aspects of the functioning mechanisms underlying NPC transport at high structural resolution, something lacking in current models of nuclear transport. 1. Sparks, S., et al., Analysis of Multivalent IDP Interactions: Stoichiometry, Affinity, and Local Concentration Effect Measurements. Methods Mol Biol, 2020. 2141: p. 463-475. 2. Sparks, S., et al., Deciphering the "Fuzzy" Interaction of FG Nucleoporins and Transport Factors Using Small-Angle Neutron Scattering. Structure, 2018. 26(3): p. 477-484 e4. 3. Hayama, R., et al., Thermodynamic characterization of the multivalent interactions underlying rapid and selective translocation through the nuclear pore complex. J Biol Chem, 2018. 293(12): p. 4555-4563. 4. Hough, L.E., et al., The molecular mechanism of nuclear transport revealed by atomic-scale measurements. Elife, 2015. 4. 5. Raveh, B., et al., Slide-and-exchange mechanism for rapid and selective transport through the nuclear pore complex. Proc Natl Acad Sci U S A, 2016. 113(18): p. E2489-97.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.R4086
  4. FASEB J. 2022 May;36 Suppl 1
      The mechanisms by which intrinsically disordered proteins (IDPs) engage in rapid and highly selective binding is a subject of considerable interest and represents a central paradigm to nuclear pore complex (NPC) function. Nuclear transport receptors (NTRs) can move through the central channel of the NPC which is filled with hundreds of phenylalanine-glycine-rich nucleoporins (FG-Nups) reaching millimolar concentrations with elusive conformational plasticity. Since site-specific labeling of proteins with small but highly photostable fluorescent dyes inside cells remains the major bottleneck for directly studying protein dynamics in the cellular interior, we have now developed a semi-synthetic strategy based on novel artificial amino acids that are easily and site-specifically introduced into any protein by the natural machinery of the living cell via a newly developed thin-film synthetic organelle that equips the living cell with up to three genetic codes. This allowed us to develop an experimental approach combining site-specific fluorescent labeling of IDPs in non-fixed cells with fluorescent lifetime imaging microscopy (FLIM) to directly decipher the plasticity of FG-Nups via FRET. Our study enabled a conformational look on the condensated IDPs in the sub-resolution (roughly (50 nm)3 small cavity) cavity of the NPC. By measuring the end-to-end distances of different segments of the labeled FG-Nups using FLIM-FRET, we can extract the scaling exponent, which directly describes the conformations of FG-Nups at their functional status as well as the solvent quality in the cellular and even inner NPC environment. Reinkemeier CD, Lemke EA. Dual film-like organelles enable spatial separation of orthogonal eukaryotic translation. Cell. 2021 Sep 16;184(19):4886-4903.e21 Celetti G, Paci G, Caria J, VanDelinder V, Bachand G, Lemke EA. The liquid state of FG-nucleoporins mimics permeability barrier properties of nuclear pore complexes. J Cell Bio. (2020) Jan 6;219(1). Reinkemeier CD, Estrada Girona G, Lemke EA, 2019 Designer membraneless organelles enable codon reassignment of selected mRNAs in eukaryotes. Science, Mar 29;363(6434) Nikić I, Estrada Girona G, Kang JH, Paci G, rei S, Koehler C, Shymanska NV, Ventura Santos C, Spitz D, Lemke EA, Debugging Eukaryotic Genetic Code Expansion for Site-Specific Click-PAINT Super-Resolution Microscopy. Angew Chem Int Ed Engl. (2016) Dec 23;55(52):16172-16176.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.0I109
  5. FASEB J. 2022 May;36 Suppl 1
      DNA viruses that replicate in the nuclei of infected cells must transport to and deliver their viral genomes into the nucleus to cause infection. How polyomavirus (PyV), a tumor virus that causes debilitating disease in humans, accomplishes this essential step in infection is not well-understood. During entry, PyV sorts from the cell surface to the endoplasmic reticulum (ER) where it penetrates the ER membrane to reach the cytosol. From there, the virus is transported to the nucleus for entry through the narrow nuclear pore complex (NPC). The incoming viral genome is harbored within an icosahedral capsid that is approximately 45 nm in diameter and larger than the NPC cargo limit. Using the archetype PyV, simian virus 40 (SV40), which shares both structural and genetic organization with human PyVs as well as the same infectious life cycle, we recently reported that the virus is disassembled in the cytosol by the bicaudal D2 (BICD2) dynein cargo adaptor to generate a subviral particle competent for transport through the NPC. How this disassembled particle is subsequently targeted to the nuclear membrane as well as the exact mechanism of nuclear import is unclear. In mammalian cells, intracellular transport to the nucleus is facilitated largely by the cytoplasmic dynein motor, which moves cargo along microtubules towards the center of the cell. Interestingly, BICD2 was shown to mediate the recruitment of dynein to the nuclear membrane through its association with either the linker of nucleoskeleton and cytoskeleton (LINC) complex component Nesprin-2 or the NPC cytoplasmic filament RanBP2. Our preliminary experiments reveal that loss of Nesprin-2 (and not RanBP2) blocks nuclear entry, suggesting that SV40 is recruited to the nucleus by a dynein-BICD2-Nesprin-2 nexus. Moreover, SV40 associates with Nesprin-2 and this interaction appears to mediate a second, previously unreported, disassembly step at the nuclear membrane. We are currently investigating the role of BICD2/Nesprin-2 in nuclear arrival of SV40 as well as the functional relationship between Nesprin-2 and the NPC during nuclear import. Our studies will clarify the mechanism responsible for driving these final steps of viral entry, and more broadly, further our understanding of host cellular nucleocytoplasmic transport.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.R4656
  6. FASEB J. 2022 May;36 Suppl 1
      Nuclear pore complexes (NPCs) fuse the inner and outer nuclear membranes and mediate nucleocytoplasmic exchange. They are made of 30 different nucleoporins and form a cylindrical architecture around an aqueous central channel. This architecture is highly dynamic in space and time. Variations in NPC diameter were reported, but the physiological circumstances and the molecular details remained unknown. We combined cryo-electron tomography (cryo-ET) with integrative structural modeling to capture the respective large-scale conformational changes in cellulo. While NPCs of exponentially growing cells adopt a dilated conformation, they reversibly constrict upon cellular energy depletion or conditions of hypertonic osmotic stress. We propose a model in which nuclear envelope membrane tension regulates the conformation of the NPC.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.0I102
  7. FASEB J. 2022 May;36 Suppl 1
      Nuclear pore complexes (NPCs) are conduits between the cytoplasm and the nucleus. NPCs consist of roughly 35 distinct nucleoporins with filaments extending from both the nuclear and cytoplasmic faces of the pore. Nucleoporins regulate many processes, including nucleocytoplasmic trafficking, protein modification, chromatin remodeling and transcription, as well as mRNA processing and export. Our current research is focused on basket nucleoporin TPR, which works as a docking platform for other proteins, including transport receptors and subunits of the TRanscription and EXport 2 (TREX-2) complex. We found that Auxin Inducible Degron (AID)-mediated TPR elimination specifically disrupts TREX-2 complex recruitment to NPCs, and results in rapid and profound changes in transcriptomic profiles of TREX-2-dependent RNAs and retention of those RNAs within the nucleus. The molecular mechanism through which TPR controls TREX-2-dependent gene expression is not clear. We are currently working in two ways to understand TREX-2-dependent RNA export and how TPR facilitates this pathway. First, we are examining the components of TREX-2. GANP is a major scaffold subunit of the TREX-2 complex, and loss of GANP disrupts the association of TREX-2 complex subunits to the NPC. We have AID-tagged other TREX-2 complex members (PCID2, ENY2, CETN2) and are analyzing how their loss impacts TREX-2 targeting, transcriptomic profiles and RNA retention. Our findings suggest that GANP or PCID2 depletion rapidly leads to polyA RNA retention inside the nucleus, but this was not the case after depletion of ENY2 or CETN2. Further comparison of TREX-2 complex subunit profiles to each other should elucidate the sequence of their actions during mRNP maturation. Second, we are investigating the point(s) at which TPR controls TREX-2-dependent gene expression: TPR may help to remodel mRNP particles within the nucleus, to recruit ready-to-export mRNP particles to the nuclear envelope, or it may perform both of these functions. To distinguish between these possibilities, we have analyzed a TPR mutant that lacks an NPC recruitment domain, to see whether it can rescue any part of TPR's function in this pathway without recruiting TREX-2 to the NPC. We found that this TPR mutant resides inside the nucleus and can not restore normal export of polyA RNA. We speculate that TPR preferentially acts as a landing platform for TREX-2-bound mRNAs at the nuclear pore and may help with quality control of the transcripts. To explore if TPR-dependent transcripts have unique signatures, we are analyzing mRNA length, GC content and intron context in other AID-tagged nucleoporins and comparing transcriptomic profiles after their loss in comparison to TPR. Alltogether, these data will help us to unfold the role of TREX2 complex subunits in targeting GANP-TPR dependent transcripts to the pore and to gain a better understanding of how TPR controls gene expression.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.R1915
  8. FASEB J. 2022 May;36 Suppl 1
      Ran Binding Protein 2 (RanBP2) contributes to the efficiency of nuclear import and export from its position at the cytoplasmic filaments of the nuclear pore complex. Here it can recruit import receptors, mediate efficient loading of cargo proteins onto importins, and serve as a docking site to efficiently recycle import and export complexes. Loss of function mutations in the RanBP2 gene are observed in many familial cases of acute necrotizing encephalopathy (ANE), a rare, sometimes fatal brain disease often occurring in infancy or early childhood following a viral infection. The influence of the RanBP2 mutation on the pathogenesis of ANE is unknown, but research points to a connection to the associated cytokine storm resulting from abnormal cytokine production. Specifically, the hyperactivation of the IL-6/JAK/STAT3 cytokine signaling pathway has been observed in patients with ANE. We aimed to investigate a possible relationship between RanBP2 and STAT3 nuclear transport to determine if a loss of RanBP2 could influence the cellular localization of the transcription factor STAT3. Due to RanBP2's importance in maintaining the efficiency of nuclear transport, its absence could impact either the nuclear or cytoplasmic localization of STAT3. siRNA knockdown of RanBP2 caused a 27% increase of STAT3 in the nucleus, suggesting increased STAT3 induced cytokine gene expression may occur without RanBP2. Our ongoing studies involve testing RanBP2 knockdown in conjunction with IL-6 treatment to determine if there is an additional effect on the cellular localization of STAT3 with the activation of signaling. Accumulation of STAT3 in the nucleus alone could contribute to the onset of ANE, however STAT3 typically requires activation of signaling to increase gene expression. Our current results established increased nuclear STAT3 as a potential mechanism for mutation of RanBP2 to influence escalated cytokine production, perpetuating cytokine storm in ANE.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.R2980
  9. FASEB J. 2022 May;36 Suppl 1
      The GTPase, Ran, is a major regulator of nuclear transport and disruption of its established gradient disrupts nuclear export and leads to cell death through the accumulation of pro-apoptotic factors. The chemotherapy 5-Fluorouracil (5-FU) increases nuclear pore permeability causing Ran to leak into the cytoplasm, disrupting the gradient and in turn nuclear export, leading to cell death of HeLa cervical cancer cells. Our previous work demonstrated that combining 5-FU with topoisomerase inhibitors can have a greater total impact on disruption of the Ran gradient and accumulates proteins in the nucleus. Gemcitabine (GEM) is widely used when treating pancreatic cancer; yet a resistance is common. However, combination of GEM with nuclear export inhibitors can help overcome this problem. Therefore, we sought to determine if combination of 5-FU and GEM could impact nuclear export to help overcome GEM resistance. Our initial studies found that 5-FU+GEM further decreased nuclear Ran in HeLa cells and lead to increased cell death in combination over GEM alone. Therefore, we transitioned to exploring the effects of 5-FU+GEM on disrupting the Ran gradient in PANC1 pancreatic cells. Both 5-FU and GEM individually and in combination decreased the nuclear Ran levels by 15%, 17%, and 23% respectively. This disruption of nuclear export may help overcome GEM resistance in PANC1 cells. Specifically, our ongoing work is exploring the impact of 5FU+GEM on the nuclear retention of the tumor suppressors p27 and p21 as well as cell viability to identify a possible mechanism for the impact of the 5-FU+GEM combination on apoptosis. Our work helps support a possible mechanism for overcoming GEM resistance in Panc1 cells and identifies a potential combination treatment with 5-FU for use against pancreatic cancer.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.R2155
  10. Int J Mol Sci. 2022 Apr 21. pii: 4627. [Epub ahead of print]23(9):
      DYT1 dystonia is a debilitating neurological movement disorder that arises upon Torsin ATPase deficiency. Nuclear envelope (NE) blebs that contain FG-nucleoporins (FG-Nups) and K48-linked ubiquitin are the hallmark phenotype of Torsin manipulation across disease models of DYT1 dystonia. While the aberrant deposition of FG-Nups is caused by defective nuclear pore complex assembly, the source of K48-ubiquitylated proteins inside NE blebs is not known. Here, we demonstrate that the characteristic K48-ubiquitin accumulation inside blebs requires p97 activity. This activity is highly dependent on the p97 adaptor UBXD1. We show that p97 does not significantly depend on the Ufd1/Npl4 heterodimer to generate the K48-ubiquitylated proteins inside blebs, nor does inhibiting translation affect the ubiquitin sequestration in blebs. However, stimulating global ubiquitylation by heat shock greatly increases the amount of K48-ubiquitin sequestered inside blebs. These results suggest that blebs have an extraordinarily high capacity for sequestering ubiquitylated protein generated in a p97-dependent manner. The p97/UBXD1 axis is thus a major factor contributing to cellular DYT1 dystonia pathology and its modulation represents an unexplored potential for therapeutic development.
    Keywords:  DYT1; ERAD; TorsinA; UBXD1; Ufd1/Npl4; YOD1; dystonia; p97; ubiquitin
    DOI:  https://doi.org/10.3390/ijms23094627
  11. FASEB J. 2022 May;36 Suppl 1
      PCI-domain containing protein 2 (PCID2) plays a key role in facilitating the nuclear export of various proteins. PCID2 further functions by localizing to the centrosome, where it likely helps to regulate the process of centrosome duplication. Nucleophosmin (NPM) is a nucleolar protein that undergoes Crm1-dependent nuclear export and also associates with centrosomes to prevent premature cell division. Other proteins reliant on Crm1 for nuclear export, including BRCA1, further require the presence of PCID2 for efficient nuclear export and subsequent centrosomal localization. Here, we show that NPM is also reliant on PCID2 for its nuclear export through the use of siRNA knockdown on HeLa cells. Immunofluorescence revealed a 34% increase in nuclear fluorescence of NPM without PCID2, while centrosomal localization of NPM exhibited no direct impact due to removal of PCID2. These results identify a supporting role for PCID2 in the regulation of centrosome duplication through the facilitation of NPM nuclear export to the cytoplasm. Ongoing studies seek to identify NPM and PCID2 complexes via immunoprecipitation as well as exploring the possible effects of PCID2 knockdown on the structure or composition of the nucleolus and on NPM subnuclear localization. Our research has identified a role for PCID2 in the regulation of NPM nuclear export, providing potential insight into the development of tumorigenesis due to centrosome amplification and possible changes to nucleolar structure and composition.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.R2292
  12. FASEB J. 2022 May;36 Suppl 1
      PCI-domain containing protein 2 (PCID2) assists in the export of proteins from the nucleus to the cytoplasm. PCID2 also localizes to the centrosome, where it is present more frequently in cells exhibiting an abnormally high number of centrosomes, an aberration associated with the development of cancer. Several proteins that negatively regulate the centrosome cycle are typically found in the nucleus, and we have previously discovered that PCID2 is involved in the nuclear export and centrosomal localization of one key centrosome cycle regulator, BRCA1. Therefore, PCID2 may play a role in the regulation of the centrosome cycle by delivering key regulators of duplication. BRCA2 is a nuclear tumor suppressor that travels to the centrosome, where it helps to arrest the centrosome cycle and prevent overduplication. BRCA2 mutations are associated with the development of breast and ovarian cancers, both of which are also associated with centrosomal overduplication. We proposed that PCID2 aids in the transport of BRCA2 from the nucleus to the centrosome. Using siRNA knockdown of PCID2 and subsequent immunofluorescence studies of BRCA2 and γ-Tubulin, we found that the loss of PCID2 significantly decreased the amount of BRCA2 able to be exported from the nucleus to the cytoplasm, with Hs578T breast cancer cells depleted of PCID2 demonstrating a 37% increase in nuclear BRCA2 retention. However, the loss of PCID2 demonstrated no significant impact on the ability of BRCA2 to localize to centrosomes. Our results indicate that while PCID2 affects the cellular localization of BRCA2 by assisting its nuclear transport, it likely does not directly influence the recruitment of cytoplasmic BRCA2 to the centrosome. Ongoing studies are exploring the relationship of PCID2 to BRCA2 export and centrosomal localization in ovarian cancer cells and are attempting to confirm the presence of a PCID2-BRCA2 nuclear export complexes in breast and ovarian cancer cells. These findings will shed light on the process by which PCID2 role in nuclear transport and the centrosome cycle, leading to a better understanding of centrosomal amplification and tumorigenesis in a variety of cancer types.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.R3021
  13. FASEB J. 2022 May;36 Suppl 1
      Karyopherin-β2 (Kapβ2) is a nuclear import receptor (NIR) that localizes proteins bearing a proline-tyrosine nuclear localization signal (PY-NLS) to the nucleus, including the RNA-binding protein (RBP) FUS. Recent work has demonstrated that NIRs can chaperone RBPs in vitro and in cells, preventing and reversing their self-assembly and aggregation. However, when the PY-NLS is mutated, Kapβ2 no longer efficiently chaperones its cargo. Here, we focus on the case of FUSP525L , an RBP NLS mutant associated with a highly aggressive form of juvenile ALS. Using a structural approach, we have engineered Kapβ2 to recognize mutant FUS and demonstrate improved chaperone activity in vitro, in human cells, and in mice.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.R2282
  14. FASEB J. 2022 May;36 Suppl 1
      NUP98 fusion oncoproteins (FOs) are drivers in pediatric leukemias and many transform hematopoietic cells. Most NUP98 FOs harbor an intrinsically disordered region from NUP98 that is prone to liquid-liquid phase separation (LLPS) in vitro. A predominant class of NUP98 FOs, including NUP98-HOXA9 (NHA9), retains a DNA-binding homeodomain, whereas others harbor other types of DNA- or chromatin-binding domains. NUP98 FOs have long been known to form puncta, but long-standing questions are how nuclear puncta form, and how they drive leukemogenesis. We will discuss our studies of NHA9 condensates, showing that homotypic interactions and different types of heterotypic interactions are required to form nuclear puncta, which are associated with aberrant transcriptional activity and transformation of hematopoietic stem and progenitor cells. We also show that three other leukemia-associated NUP98 FOs form nuclear puncta and transform hematopoietic cells. To extend our findings, we tested ~150 additional fusion oncoproteins associate with a wide range of human cancers for puncta formation in cells. We will discuss our observation that >50% of these formed cellular condensates, with this behavior for some linked with aberrant gene expression and cell transformation. Our findings indicate that LLPS is critical for leukemogenesis by NUP98 FOs and likely contributes to oncogenesis driven by many other fusion oncoproteins.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.0I161