Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
Microorganisms ; 10(3)2022 Feb 24.
Article in English | MEDLINE | ID: mdl-35336080

ABSTRACT

Bacterial viruses (or bacteriophages) have developed formidable ways to deliver their genetic information inside bacteria, overcoming the complexity of the bacterial-cell envelope. In short-tailed phages of the Podoviridae superfamily, genome ejection is mediated by a set of mysterious internal virion proteins, also called ejection or pilot proteins, which are required for infectivity. The ejection proteins are challenging to study due to their plastic structures and transient assembly and have remained less characterized than classical components such as the phage coat protein or terminase subunit. However, a spate of recent cryo-EM structures has elucidated key features underscoring these proteins' assembly and conformational gymnastics that accompany their expulsion from the virion head through the portal protein channel into the host. In this review, we will use a phage-T7-centric approach to critically review the status of the literature on ejection proteins, decipher the conformational changes of T7 ejection proteins in the pre- and post-ejection conformation, and predict the conservation of these proteins in other Podoviridae. The challenge is to relate the structure of the ejection proteins to the mechanisms of genome ejection, which are exceedingly complex and use the host's machinery.

4.
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
5.
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
6.
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
7.
Structure ; 26(12): 1565-1567, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30517883

ABSTRACT

In this issue of Structure, Dunne et al. (2018) unveil the architecture of Salmonella phage S16 adhesin. The structure unravels a beads-on-a-string topology consisting of three domains of which the C-terminal glycine-rich PGII domain, located at the distal tip of the long tail fiber, mediates cell surface attachment and host recognition.


Subject(s)
Bacteriophages , Salmonella Phages , Peptides , Virion
SELECTION OF CITATIONS
SEARCH DETAIL
...