Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Res Sq ; 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38463957

ABSTRACT

DEV is an obligatory lytic Pseudomonas phage of the N4-like genus, recently reclassified as Schitoviridae. The DEV genome encodes 91 ORFs, including a 3,398 amino acid virion-associated RNA polymerase. Here, we describe the complete architecture of DEV, determined using a combination of cryo-electron microscopy localized reconstruction, biochemical methods, and genetic knockouts. We built de novo structures of all capsid factors and tail components involved in host attachment. We demonstrate that DEV long tail fibers are essential for infection of Pseudomonas aeruginosa and dispensable for infecting mutants with a truncated lipopolysaccharide devoid of the O-antigen. We identified DEV ejection proteins and, unexpectedly, found that the giant DEV RNA polymerase, the hallmark of the Schitoviridae family, is an ejection protein. We propose that DEV ejection proteins form a genome ejection motor across the host cell envelope and that these structural principles are conserved in all Schitoviridae.

2.
Sci Adv ; 10(2): eadi7606, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38198557

ABSTRACT

Nuclear import of the hepatitis B virus (HBV) nucleocapsid is essential for replication that occurs in the nucleus. The ~360-angstrom HBV capsid translocates to the nuclear pore complex (NPC) as an intact particle, hijacking human importins in a reaction stimulated by host kinases. This paper describes the mechanisms of HBV capsid recognition by importins. We found that importin α1 binds a nuclear localization signal (NLS) at the far end of the HBV coat protein Cp183 carboxyl-terminal domain (CTD). This NLS is exposed to the capsid surface through a pore at the icosahedral quasi-sixfold vertex. Phosphorylation at serine-155, serine-162, and serine-170 promotes CTD compaction but does not affect the affinity for importin α1. The binding of 30 importin α1/ß1 augments HBV capsid diameter to ~620 angstroms, close to the maximum size trafficable through the NPC. We propose that phosphorylation favors CTD externalization and prompts its compaction at the capsid surface, exposing the NLS to importins.


Subject(s)
Hepatitis B virus , Nucleocapsid , Humans , Active Transport, Cell Nucleus , Karyopherins , Capsid Proteins , Immunologic Factors , Serine
3.
J Mol Biol ; 435(24): 168365, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37952769

ABSTRACT

Bacteriophage P22 is a prototypical member of the Podoviridae superfamily. Since its discovery in 1952, P22 has become a paradigm for phage transduction and a model for icosahedral viral capsid assembly. Here, we describe the complete architecture of the P22 tail apparatus (gp1, gp4, gp10, gp9, and gp26) and the potential location and organization of P22 ejection proteins (gp7, gp20, and gp16), determined using cryo-EM localized reconstruction, genetic knockouts, and biochemical analysis. We found that the tail apparatus exists in two equivalent conformations, rotated by ∼6° relative to the capsid. Portal protomers make unique contacts with coat subunits in both conformations, explaining the 12:5 symmetry mismatch. The tail assembles around the hexameric tail hub (gp10), which folds into an interrupted ß-propeller characterized by an apical insertion domain. The tail hub connects proximally to the dodecameric portal protein and head-to-tail adapter (gp4), distally to the trimeric tail needle (gp26), and laterally to six trimeric tailspikes (gp9) that attach asymmetrically to gp10 insertion domain. Cryo-EM analysis of P22 mutants lacking the ejection proteins gp7 or gp20 and biochemical analysis of purified recombinant proteins suggest that gp7 and gp20 form a molecular complex associated with the tail apparatus via the portal protein barrel. We identified a putative signal transduction pathway from the tailspike to the tail needle, mediated by three flexible loops in the tail hub, that explains how lipopolysaccharide (LPS) is sufficient to trigger the ejection of the P22 DNA in vitro.


Subject(s)
Bacteriophage P22 , Salmonella typhimurium , Bacteriophage P22/genetics , Bacteriophage P22/chemistry , Bacteriophage P22/metabolism , Capsid Proteins/chemistry , Salmonella typhimurium/virology , Viral Tail Proteins/genetics
4.
Nat Commun ; 14(1): 4052, 2023 07 08.
Article in English | MEDLINE | ID: mdl-37422479

ABSTRACT

E217 is a Pseudomonas phage used in an experimental cocktail to eradicate cystic fibrosis-associated Pseudomonas aeruginosa. Here, we describe the structure of the whole E217 virion before and after DNA ejection at 3.1 Å and 4.5 Å resolution, respectively, determined using cryogenic electron microscopy (cryo-EM). We identify and build de novo structures for 19 unique E217 gene products, resolve the tail genome-ejection machine in both extended and contracted states, and decipher the complete architecture of the baseplate formed by 66 polypeptide chains. We also determine that E217 recognizes the host O-antigen as a receptor, and we resolve the N-terminal portion of the O-antigen-binding tail fiber. We propose that E217 design principles presented in this paper are conserved across PB1-like Myoviridae phages of the Pbunavirus genus that encode a ~1.4 MDa baseplate, dramatically smaller than the coliphage T4.


Subject(s)
Pseudomonas Phages , Pseudomonas Phages/genetics , Cryoelectron Microscopy , O Antigens , Microscopy, Electron , Myoviridae , Bacteriophage T4/chemistry
5.
Viruses ; 14(10)2022 10 08.
Article in English | MEDLINE | ID: mdl-36298770

ABSTRACT

The genome packaging motor of bacteriophages and herpesviruses is built by two terminase subunits, known as large (TerL) and small (TerS), both essential for viral genome packaging. TerL structure, composition, and assembly to an empty capsid, as well as the mechanisms of ATP-dependent DNA packaging, have been studied in depth, shedding light on the chemo-mechanical coupling between ATP hydrolysis and DNA translocation. Instead, significantly less is known about the small terminase subunit, TerS, which is dispensable or even inhibitory in vitro, but essential in vivo. By taking advantage of the recent revolution in cryo-electron microscopy (cryo-EM) and building upon a wealth of crystallographic structures of phage TerSs, in this review, we take an inventory of known TerSs studied to date. Our analysis suggests that TerS evolved and diversified into a flexible molecular framework that can conserve biological function with minimal sequence and quaternary structure conservation to fit different packaging strategies and environmental conditions.


Subject(s)
Bacteriophages , Virus Assembly , Virus Assembly/genetics , Cryoelectron Microscopy , Viral Proteins/genetics , DNA, Viral/chemistry , DNA Packaging , Endodeoxyribonucleases/genetics , Bacteriophages/genetics , Bacteriophages/chemistry , Adenosine Triphosphate
6.
J Mol Biol ; 434(20): 167799, 2022 10 30.
Article in English | MEDLINE | ID: mdl-36007626

ABSTRACT

Pseudomonas phages are increasingly important biomedicines for phage therapy, but little is known about how these viruses package DNA. This paper explores the terminase subunits from the Myoviridae E217, a Pseudomonas-phage used in an experimental cocktail to eradicate P. aeruginosa in vitro and in animal models. We identified the large (TerL) and small (TerS) terminase subunits in two genes ∼58 kbs away from each other in the E217 genome. TerL presents a classical two-domain architecture, consisting of an N-terminal ATPase and C-terminal nuclease domain arranged into a bean-shaped tertiary structure. A 2.05 Å crystal structure of the C-terminal domain revealed an RNase H-like fold with two magnesium ions in the nuclease active site. Mutations in TerL residues involved in magnesium coordination had a dominant-negative effect on phage growth. However, the two ions identified in the active site were too far from each other to promote two-metal-ion catalysis, suggesting a conformational change is required for nuclease activity. We also determined a 3.38 Å cryo-EM reconstruction of E217 TerS that revealed a ring-like decamer, departing from the most common nonameric quaternary structure observed thus far. E217 TerS contains both N-terminal helix-turn-helix motifs enriched in basic residues and a central channel lined with basic residues large enough to accommodate double-stranded DNA. Overexpression of TerS caused a more than a 4-fold reduction of E217 burst size, suggesting a catalytic amount of the protein is required for packaging. Together, these data expand the molecular repertoire of viral terminase subunits to Pseudomonas-phages used for phage therapy.


Subject(s)
Endodeoxyribonucleases , Myoviridae , Pseudomonas Phages , Pseudomonas aeruginosa , Viral Proteins , Adenosine Triphosphatases/metabolism , DNA, Viral/metabolism , Endodeoxyribonucleases/chemistry , Magnesium/chemistry , Myoviridae/enzymology , Pseudomonas Phages/enzymology , Pseudomonas aeruginosa/virology , Ribonuclease H/chemistry , Viral Proteins/chemistry
7.
Cell Rep ; 39(13): 111007, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35767952

ABSTRACT

Cytoplasmic mislocalization of the TAR-DNA binding protein of 43 kDa (TDP-43) leads to large, insoluble aggregates that are a hallmark of amyotrophic lateral sclerosis and frontotemporal dementia. Here, we study how importin α1/ß recognizes TDP-43 bipartite nuclear localization signal (NLS). We find that the NLS makes extensive contacts with importin α1, especially at the minor NLS-binding site. NLS binding results in steric clashes with the C terminus of importin α1 that disrupts the TDP-43 N-terminal domain (NTD) dimerization interface. A putative phosphorylation site in the proximity of TDP-43 R83 at the minor NLS site destabilizes binding to importins by reducing the NLS backbone dynamics. Based on these data, we explain the pathogenic role of several post-translational modifications and mutations in the proximity of TDP-43 minor NLS site that are linked to disease and shed light on the chaperone activity of importin α1/ß.


Subject(s)
Nuclear Localization Signals , beta Karyopherins , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Nuclear Localization Signals/metabolism , alpha Karyopherins/genetics , alpha Karyopherins/metabolism , beta Karyopherins/genetics , beta Karyopherins/metabolism
8.
J Mol Biol ; 434(9): 167537, 2022 05 15.
Article in English | MEDLINE | ID: mdl-35278476

ABSTRACT

Portal proteins are dodecameric assemblies that occupy a unique 5-fold vertex of the icosahedral capsid of tailed bacteriophages and herpesviruses. The portal vertex interrupts the icosahedral symmetry, and in vivo, its assembly and incorporation in procapsid are controlled by the scaffolding protein. Ectopically expressed portal oligomers are polymorphic in solution, and portal rings built by a different number of subunits have been documented in the literature. In this paper, we describe the cryo-EM structure of the portal protein from the Pseudomonas-phage PaP3, which we determined at 3.4 Å resolution. Structural analysis revealed a dodecamer with helical rather than rotational symmetry, which we hypothesize is kinetically trapped. The helical assembly was stabilized by local mispairing of portal subunits caused by the slippage of crown and barrel helices that move like a lever with respect to the portal body. Removing the C-terminal barrel promoted assembly of undecameric and dodecameric rings with quasi-rotational symmetry, suggesting that the barrel contributes to subunits mispairing. However, ΔC-portal rings were intrinsically asymmetric, with most particles having one open portal subunit interface. Together, these data expand the structural repertoire of viral portal proteins to Pseudomonas-phages and shed light on the unexpected plasticity of the portal protein quaternary structure.


Subject(s)
Capsid Proteins , Capsid , Pseudomonas Phages , Capsid/chemistry , Capsid Proteins/chemistry , Cryoelectron Microscopy , Protein Conformation , Pseudomonas Phages/chemistry , Virus Assembly
9.
Nat Commun ; 13(1): 1207, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35260573

ABSTRACT

Nuclear translocation of the p50/p65 heterodimer is essential for NF-κB signaling. In unstimulated cells, p50/p65 is retained by the inhibitor IκBα in the cytoplasm that masks the p65-nuclear localization sequence (NLS). Upon activation, p50/p65 is translocated into the nucleus by the adapter importin α3 and the receptor importin ß. Here, we describe a bipartite NLS in p50/p65, analogous to nucleoplasmin NLS but exposed in trans. Importin α3 accommodates the p50- and p65-NLSs at the major and minor NLS-binding pockets, respectively. The p50-NLS is the predominant binding determinant, while the p65-NLS induces a conformational change in the Armadillo 7 of importin α3 that stabilizes a helical conformation of the p65-NLS. Neither conformational change was observed for importin α1, which makes fewer bonds with the p50/p65 NLSs, explaining the preference for α3. We propose that importin α3 discriminates between the transcriptionally active p50/p65 heterodimer and p50/p50 and p65/65 homodimers, ensuring fidelity in NF-κB signaling.


Subject(s)
Karyopherins , NF-kappa B , Cell Nucleus/metabolism , Karyopherins/genetics , Karyopherins/metabolism , NF-kappa B/metabolism , Nuclear Localization Signals/metabolism , beta Karyopherins/metabolism
10.
STAR Protoc ; 2(4): 100960, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34825220

ABSTRACT

Bacteriophages of the Podoviridae family densely package their genomes into precursor capsids alongside internal virion proteins called ejection proteins. In phage T7 these proteins (gp14, gp15, and gp16) are ejected into the host envelope forming a DNA-ejectosome for genome delivery. Here, we describe the purification and characterization of recombinant gp14, gp15, and gp16. This protocol was used for high-resolution cryo-EM structure analysis of the T7 periplasmic tunnel and can be adapted to study ejection proteins from other phages. For complete details on the use and execution of this protocol, please refer to Swanson et al. (2021).


Subject(s)
Bacteriophage T7 , Cryoelectron Microscopy/methods , Recombinant Proteins , Viral Proteins , Bacteriophage T7/genetics , Bacteriophage T7/metabolism , Escherichia coli/genetics , Periplasm/chemistry , Periplasm/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/isolation & purification , Viral Proteins/metabolism
11.
Mol Cell ; 81(15): 3145-3159.e7, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34214465

ABSTRACT

Hershey and Chase used bacteriophage T2 genome delivery inside Escherichia coli to demonstrate that DNA, not protein, is the genetic material. Seventy years later, our understanding of viral genome delivery in prokaryotes remains limited, especially for short-tailed phages of the Podoviridae family. These viruses expel mysterious ejection proteins found inside the capsid to form a DNA-ejectosome for genome delivery into bacteria. Here, we reconstitute the phage T7 DNA-ejectosome components gp14, gp15, and gp16 and solve the periplasmic tunnel structure at 2.7 Å resolution. We find that gp14 forms an outer membrane pore, gp15 assembles into a 210 Å hexameric DNA tube spanning the host periplasm, and gp16 extends into the host cytoplasm forming a ∼4,200 residue hub. Gp16 promotes gp15 oligomerization, coordinating peptidoglycan hydrolysis, DNA binding, and lipid insertion. The reconstituted gp15:gp16 complex lacks channel-forming activity, suggesting that the pore for DNA passage forms only transiently during genome ejection.


Subject(s)
Bacteriophage T7/genetics , DNA, Viral/chemistry , Periplasm/chemistry , Viral Core Proteins/chemistry , Computational Biology , Cryoelectron Microscopy , Cytoplasm/chemistry , DNA, Viral/metabolism , Lipid Bilayers/metabolism , Periplasm/genetics , Periplasm/metabolism , Podoviridae/chemistry , Podoviridae/genetics , Viral Core Proteins/metabolism
12.
Nucleic Acids Res ; 48(20): 11721-11736, 2020 11 18.
Article in English | MEDLINE | ID: mdl-33125059

ABSTRACT

The genome packaging motor of tailed bacteriophages and herpesviruses is a powerful nanomachine built by several copies of a large (TerL) and a small (TerS) terminase subunit. The motor assembles transiently at the portal vertex of an empty precursor capsid (or procapsid) to power genome encapsidation. Terminase subunits have been studied in-depth, especially in classical bacteriophages that infect Escherichia coli or Salmonella, yet, less is known about the packaging motor of Pseudomonas-phages that have increasing biomedical relevance. Here, we investigated the small terminase subunit from three Podoviridae phages that infect Pseudomonas aeruginosa. We found TerS is polymorphic in solution but assembles into a nonamer in its high-affinity heparin-binding conformation. The atomic structure of Pseudomonas phage PaP3 TerS, the first complete structure for a TerS from a cos phage, reveals nine helix-turn-helix (HTH) motifs asymmetrically arranged around a ß-stranded channel, too narrow to accommodate DNA. PaP3 TerS binds DNA in a sequence-specific manner in vitro. X-ray scattering and molecular modeling suggest TerS adopts an open conformation in solution, characterized by dynamic HTHs that move around an oligomerization core, generating discrete binding crevices for DNA. We propose a model for sequence-specific recognition of packaging initiation sites by lateral interdigitation of DNA.


Subject(s)
DNA/metabolism , Endodeoxyribonucleases/chemistry , Pseudomonas Phages/enzymology , Viral Proteins/chemistry , Base Sequence , DNA/chemistry , Endodeoxyribonucleases/metabolism , Helix-Turn-Helix Motifs , Models, Molecular , Protein Binding , Pseudomonas aeruginosa/virology , Viral Proteins/metabolism
13.
Acta Crystallogr D Struct Biol ; 76(Pt 9): 876-888, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32876063

ABSTRACT

The genome-packaging motor of tailed bacteriophages and herpesviruses is a multisubunit protein complex formed by several copies of a large (TerL) and a small (TerS) terminase subunit. The motor assembles transiently at the portal protein vertex of an empty precursor capsid to power the energy-dependent packaging of viral DNA. Both the ATPase and nuclease activities associated with genome packaging reside in TerL. Structural studies of TerL from bacteriophage P22 have been hindered by the conformational flexibility of this enzyme and its susceptibility to proteolysis. Here, an unbiased, synthetic phage-display Fab library was screened and a panel of high-affinity Fabs against P22 TerL were identified. This led to the discovery of a recombinant antibody fragment, Fab4, that binds a 33-amino-acid α-helical hairpin at the N-terminus of TerL with an equilibrium dissociation constant Kd of 71.5 nM. A 1.51 Šresolution crystal structure of Fab4 bound to the TerL epitope (TLE) together with a 1.15 Šresolution crystal structure of the unliganded Fab4, which is the highest resolution ever achieved for a Fab, elucidate the principles governing the recognition of this novel helical epitope. TLE adopts two different conformations in the asymmetric unit and buries as much as 1250 Å2 of solvent-accessible surface in Fab4. TLE recognition is primarily mediated by conformational changes in the third complementarity-determining region of the Fab4 heavy chain (CDR H3) that take place upon epitope binding. It is demonstrated that TLE can be introduced genetically at the N-terminus of a target protein, where it retains high-affinity binding to Fab4.


Subject(s)
Bacteriophage P22/enzymology , Endodeoxyribonucleases , Immunoglobulin Fab Fragments , Viral Proteins , Endodeoxyribonucleases/chemistry , Helix-Turn-Helix Motifs , Immunoglobulin Fab Fragments/chemistry , Models, Molecular , Peptide Library , Protein Binding , Viral Proteins/chemistry
14.
Nat Commun ; 11(1): 2013, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32332720

ABSTRACT

The long external filament of bacterial flagella is composed of several thousand copies of a single protein, flagellin. Here, we explore the role played by lysine methylation of flagellin in Salmonella, which requires the methylase FliB. We show that both flagellins of Salmonella enterica serovar Typhimurium, FliC and FljB, are methylated at surface-exposed lysine residues by FliB. A Salmonella Typhimurium mutant deficient in flagellin methylation is outcompeted for gut colonization in a gastroenteritis mouse model, and methylation of flagellin promotes bacterial invasion of epithelial cells in vitro. Lysine methylation increases the surface hydrophobicity of flagellin, and enhances flagella-dependent adhesion of Salmonella to phosphatidylcholine vesicles and epithelial cells. Therefore, posttranslational methylation of flagellin facilitates adhesion of Salmonella Typhimurium to hydrophobic host cell surfaces, and contributes to efficient gut colonization and host infection.


Subject(s)
Bacterial Adhesion , Flagellin/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Salmonella Infections/pathology , Salmonella typhimurium/pathogenicity , Animals , Cell Line , Disease Models, Animal , Epithelial Cells , Flagella/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Methylation , Mice , NIH 3T3 Cells , Protein Processing, Post-Translational , Salmonella Infections/microbiology , Salmonella typhimurium/metabolism
15.
Nat Commun ; 10(1): 2747, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31227715

ABSTRACT

Many intracellular bacteria, including Chlamydia, establish a parasitic membrane-bound organelle inside the host cell that is essential for the bacteria's survival. Chlamydia trachomatis forms inclusions that are decorated with poorly characterized membrane proteins known as Incs. The prototypical Inc, called IncA, enhances Chlamydia pathogenicity by promoting the homotypic fusion of inclusions and shares structural and functional similarity to eukaryotic SNAREs. Here, we present the atomic structure of the cytoplasmic domain of IncA, which reveals a non-canonical four-helix bundle. Structure-based mutagenesis, molecular dynamics simulation, and functional cellular assays identify an intramolecular clamp that is essential for IncA-mediated homotypic membrane fusion during infection.


Subject(s)
Bacterial Proteins/ultrastructure , Chlamydia Infections/microbiology , Chlamydia trachomatis/pathogenicity , Inclusion Bodies/microbiology , Membrane Fusion , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Chlamydia trachomatis/genetics , Chlamydia trachomatis/metabolism , Crystallography, X-Ray , Gene Knockout Techniques , HeLa Cells , Humans , Molecular Dynamics Simulation , Mutagenesis , Protein Conformation, alpha-Helical , Protein Domains/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , SNARE Proteins/chemistry
16.
Biochemistry ; 58(6): 534-545, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30548067

ABSTRACT

Siw14 is a recently discovered inositol phosphatase implicated in suppressing prion propagation in Saccharomyces cerevisiae. In this paper, we used hybrid structural methods to decipher Siw14 molecular architecture. We found the protein exists in solution as an elongated monomer that is ∼140 Šin length, containing an acidic N-terminal domain and a basic C-terminal dual-specificity phosphatase (DSP) domain, structurally similar to the glycogen phosphatase laforin. The two domains are connected by a protease susceptible linker and do not interact in vitro. The crystal structure of Siw14-DSP reveals a highly basic phosphate-binding loop and an ∼10 Šdeep substrate-binding crevice that evolved to dephosphorylate pyro-phosphate moieties. A pseudoatomic model of the full-length phosphatase generated from solution, crystallographic, biochemical, and modeling data sheds light on the interesting zwitterionic nature of Siw14, which we hypothesized may play a role in discriminating negatively charged inositol phosphates.


Subject(s)
Protein Conformation , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Protein Folding , Saccharomyces cerevisiae/growth & development
17.
Biochemistry ; 57(50): 6878-6887, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30452242

ABSTRACT

Smk1 is a mitogen-activated protein kinase (MAPK) family member in the yeast Saccharomyces cerevisiae that controls the postmeiotic program of spore formation. Ssp2 is a meiosis-specific protein that activates Smk1 and triggers the autophosphorylation of its activation loop. A fragment of Ssp2 that is sufficient to activate Smk1 contains two segments that resemble RNA recognition motifs (RRMs). Mutations in either of these motifs eliminated Ssp2's ability to activate Smk1. In contrast, deletions and insertions within the segment linking the RRM-like motifs only partially reduced the activity of Ssp2. Moreover, when the two RRM-like motifs were expressed as separate proteins in bacteria, they activated Smk1. We also find that both motifs can be cross-linked to Smk1 and that at least one of the motifs binds near the ATP-binding pocket of the MAPK. These findings demonstrate that motifs related to RRMs can directly activate protein kinases.


Subject(s)
Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , RNA Recognition Motif/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Binding Sites/genetics , Enzyme Activation/genetics , Meiosis/genetics , Mitogen-Activated Protein Kinases/chemistry , Models, Molecular , Mutation , Protein Conformation , RNA, Fungal/genetics , RNA, Fungal/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Spores, Fungal/genetics , Spores, Fungal/metabolism
18.
Cell ; 174(5): 1200-1215.e20, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30100187

ABSTRACT

Nuclear pore complexes (NPCs) regulate nuclear-cytoplasmic transport, transcription, and genome integrity in eukaryotic cells. However, their functional roles in cancer remain poorly understood. We interrogated the evolutionary transcriptomic landscape of NPC components, nucleoporins (Nups), from primary to advanced metastatic human prostate cancer (PC). Focused loss-of-function genetic screen of top-upregulated Nups in aggressive PC models identified POM121 as a key contributor to PC aggressiveness. Mechanistically, POM121 promoted PC progression by enhancing importin-dependent nuclear transport of key oncogenic (E2F1, MYC) and PC-specific (AR-GATA2) transcription factors, uncovering a pharmacologically targetable axis that, when inhibited, decreased tumor growth, restored standard therapy efficacy, and improved survival in patient-derived pre-clinical models. Our studies molecularly establish a role of NPCs in PC progression and give a rationale for NPC-regulated nuclear import targeting as a therapeutic strategy for lethal PC. These findings may have implications for understanding how NPC deregulation contributes to the pathogenesis of other tumor types.


Subject(s)
E2F1 Transcription Factor/metabolism , Membrane Glycoproteins/metabolism , Nuclear Pore/physiology , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Carcinogenesis , Cell Nucleus/metabolism , Cell Proliferation , GATA2 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Humans , Male , Nuclear Envelope , Nuclear Pore Complex Proteins , Signal Transduction
19.
Nat Commun ; 8(1): 979, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29042532

ABSTRACT

Active nuclear import of Ran exchange factor RCC1 is mediated by importin α3. This pathway is essential to generate a gradient of RanGTP on chromatin that directs nucleocytoplasmic transport, mitotic spindle assembly and nuclear envelope formation. Here we identify the mechanisms of importin α3 selectivity for RCC1. We find this isoform binds RCC1 with one order of magnitude higher affinity than the generic importin α1, although the two isoforms share an identical NLS-binding groove. Importin α3 uses its greater conformational flexibility to wedge the RCC1 ß-propeller flanking the NLS against its lateral surface, preventing steric clashes with its Armadillo-core. Removing the ß-propeller, or inserting a linker between NLS and ß-propeller, disrupts specificity for importin α3, demonstrating the structural context rather than NLS sequence determines selectivity for isoform 3. We propose importin α3 evolved to recognize topologically complex NLSs that lie next to bulky domains or are masked by quaternary structures.Importin α3 facilitates the nuclear transport of the Ran guanine nucleotide exchange factor RCC1. Here the authors reveal the molecular basis for the selectivity of RCC1 for importin α3 vs the generic importin α1 and discuss the evolution of importin α isoforms.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/metabolism , Nuclear Localization Signals/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , alpha Karyopherins/metabolism , Cell Cycle Proteins/genetics , Cell Nucleus/chemistry , Cell Nucleus/genetics , Cell Nucleus/metabolism , Guanine Nucleotide Exchange Factors/genetics , Humans , Models, Molecular , Nuclear Localization Signals/chemistry , Nuclear Localization Signals/genetics , Nuclear Proteins/genetics , Protein Binding , Protein Conformation , Protein Transport , alpha Karyopherins/chemistry , alpha Karyopherins/genetics
20.
Sci Rep ; 7(1): 11381, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28900157

ABSTRACT

The influenza A virus nucleoprotein (NP) is an essential multifunctional protein that encapsidates the viral genome and functions as an adapter between the virus and the host cell machinery. NPs from all strains of influenza A viruses contain two nuclear localization signals (NLSs): a well-studied monopartite NLS1 and a less-characterized NLS2, thought to be bipartite. Through site-directed mutagenesis and functional analysis, we found that NLS2 is also monopartite and is indispensable for viral infection. Atomic structures of importin α bound to two variants of NLS2 revealed NLS2 primarily binds the major-NLS binding site of importin α, unlike NLS1 that associates with the minor NLS-pocket. Though peptides corresponding to NLS1 and NLS2 bind weakly to importin α, the two NLSs synergize in the context of the full length NP to confer high avidity for importin α7, explaining why the virus efficiently replicates in the respiratory tract that exhibits high levels of this isoform. This study, the first to functionally characterize NLS2, demonstrates NLS2 plays an important and unexpected role in influenza A virus infection. We propose NLS1 and NLS2 form a bipartite NLS in trans, which ensures high avidity for importin α7 while preventing non-specific binding to viral RNA.


Subject(s)
Influenza A virus/genetics , Influenza A virus/metabolism , Nuclear Localization Signals , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Viral Core Proteins/genetics , Viral Core Proteins/metabolism , alpha Karyopherins/genetics , alpha Karyopherins/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Cell Line , Humans , Influenza, Human/metabolism , Influenza, Human/virology , Models, Molecular , Molecular Conformation , Mutation , Nucleocapsid Proteins , Protein Binding , RNA-Binding Proteins/chemistry , Viral Core Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...