Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 395
Filter
1.
Biochem Biophys Res Commun ; 728: 150334, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-38968773

ABSTRACT

Capsid-like poxvirus scaffold proteins self-assemble into semi-regular lattice that govern the formation of spherical immature virus particles. The scaffolding is a critical step in virus morphogenesis as exemplified by the drug rifampicin that impairs the recruitment of scaffold onto the viral membrane in vaccinia virus (VACV). Here we report cryo-electron microscopy structure of scaffolding protein Orfv075 of orf virus (ORFV) that causes smallpox-like diseases in sheep, goats and occasionally humans via zoonotic infection. We demonstrate that the regions that are involved in intertrimeric interactions for scaffold assembly are largely conserved in comparison to its VACV orthologue protein D13 whose intermediate assembly structures have been previously characterized. By contrast, less conserved regions are located away from these interfaces, indicating both viruses share similar assembly mechanisms. We also show that the phenylalanine-rich binding site of rifampicin in D13 is conserved in Orfv075, and molecular docking simulation confirms similar binding modes. Our study provides structural basis of scaffolding protein as a target for anti-poxvirus treatment across wide range of poxvirus genera.


Subject(s)
Cryoelectron Microscopy , Orf virus , Orf virus/chemistry , Orf virus/ultrastructure , Molecular Docking Simulation , Animals , Binding Sites , Protein Conformation , Models, Molecular , Amino Acid Sequence , Viral Proteins/chemistry , Viral Proteins/ultrastructure , Viral Proteins/metabolism , Viral Structural Proteins/chemistry , Viral Structural Proteins/ultrastructure , Viral Structural Proteins/metabolism , Rifampin/chemistry , Rifampin/pharmacology
2.
Nature ; 631(8021): 670-677, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38987591

ABSTRACT

In all organisms, regulation of gene expression must be adjusted to meet cellular requirements and frequently involves helix-turn-helix (HTH) domain proteins1. For instance, in the arms race between bacteria and bacteriophages, rapid expression of phage anti-CRISPR (acr) genes upon infection enables evasion from CRISPR-Cas defence; transcription is then repressed by an HTH-domain-containing anti-CRISPR-associated (Aca) protein, probably to reduce fitness costs from excessive expression2-5. However, how a single HTH regulator adjusts anti-CRISPR production to cope with increasing phage genome copies and accumulating acr mRNA is unknown. Here we show that the HTH domain of the regulator Aca2, in addition to repressing Acr synthesis transcriptionally through DNA binding, inhibits translation of mRNAs by binding conserved RNA stem-loops and blocking ribosome access. The cryo-electron microscopy structure of the approximately 40 kDa Aca2-RNA complex demonstrates how the versatile HTH domain specifically discriminates RNA from DNA binding sites. These combined regulatory modes are widespread in the Aca2 family and facilitate CRISPR-Cas inhibition in the face of rapid phage DNA replication without toxic acr overexpression. Given the ubiquity of HTH-domain-containing proteins, it is anticipated that many more of them elicit regulatory control by dual DNA and RNA binding.


Subject(s)
Bacteriophages , CRISPR-Cas Systems , DNA-Binding Proteins , Gene Expression Regulation, Viral , Helix-Turn-Helix Motifs , RNA-Binding Proteins , Viral Proteins , Bacteriophages/chemistry , Bacteriophages/genetics , Bacteriophages/metabolism , Bacteriophages/ultrastructure , Binding Sites , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , CRISPR-Associated Proteins/metabolism , Cryoelectron Microscopy , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/ultrastructure , Genes, Viral , Models, Molecular , Nucleic Acid Conformation , Pectobacterium carotovorum/virology , Protein Biosynthesis/genetics , Protein Domains , Ribosomes/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/ultrastructure , RNA, Viral/chemistry , RNA, Viral/genetics , RNA, Viral/metabolism , RNA, Viral/ultrastructure , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/ultrastructure , Substrate Specificity , Transcription, Genetic , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Proteins/ultrastructure
3.
Nat Commun ; 15(1): 4189, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760379

ABSTRACT

The viral polymerase complex, comprising the large protein (L) and phosphoprotein (P), is crucial for both genome replication and transcription in non-segmented negative-strand RNA viruses (nsNSVs), while structures corresponding to these activities remain obscure. Here, we resolved two L-P complex conformations from the mumps virus (MuV), a typical member of nsNSVs, via cryogenic-electron microscopy. One conformation presents all five domains of L forming a continuous RNA tunnel to the methyltransferase domain (MTase), preferably as a transcription state. The other conformation has the appendage averaged out, which is inaccessible to MTase. In both conformations, parallel P tetramers are revealed around MuV L, which, together with structures of other nsNSVs, demonstrates the diverse origins of the L-binding X domain of P. Our study links varying structures of nsNSV polymerase complexes with genome replication and transcription and points to a sliding model for polymerase complexes to advance along the RNA templates.


Subject(s)
Cryoelectron Microscopy , Mumps virus , Viral Proteins , Mumps virus/genetics , Mumps virus/ultrastructure , Mumps virus/metabolism , Viral Proteins/metabolism , Viral Proteins/ultrastructure , Viral Proteins/chemistry , Viral Proteins/genetics , Models, Molecular , RNA, Viral/metabolism , RNA, Viral/ultrastructure , RNA, Viral/genetics , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/ultrastructure , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/genetics , Protein Domains , Phosphoproteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/ultrastructure , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/ultrastructure , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Virus Replication , Transcription, Genetic , Protein Conformation
4.
Nature ; 631(8019): 224-231, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38811740

ABSTRACT

The prime editor system composed of Streptococcus pyogenes Cas9 nickase (nSpCas9) and engineered Moloney murine leukaemia virus reverse transcriptase (M-MLV RT) collaborates with a prime editing guide RNA (pegRNA) to facilitate a wide variety of precise genome edits in living cells1. However, owing to a lack of structural information, the molecular mechanism of pegRNA-guided reverse transcription by the prime editor remains poorly understood. Here we present cryo-electron microscopy structures of the SpCas9-M-MLV RTΔRNaseH-pegRNA-target DNA complex in multiple states. The termination structure, along with our functional analysis, reveals that M-MLV RT extends reverse transcription beyond the expected site, resulting in scaffold-derived incorporations that cause undesired edits at the target loci. Furthermore, structural comparisons among the pre-initiation, initiation and elongation states show that M-MLV RT remains in a consistent position relative to SpCas9 during reverse transcription, whereas the pegRNA-synthesized DNA heteroduplex builds up along the surface of SpCas9. On the basis of our structural insights, we rationally engineered pegRNA variants and prime-editor variants in which M-MLV RT is fused within SpCas9. Collectively, our findings provide structural insights into the stepwise mechanism of prime editing, and will pave the way for the development of a versatile prime editing toolbox.


Subject(s)
CRISPR-Associated Protein 9 , Gene Editing , Moloney murine leukemia virus , RNA, Guide, CRISPR-Cas Systems , RNA-Directed DNA Polymerase , Reverse Transcription , Streptococcus pyogenes , Humans , CRISPR-Associated Protein 9/chemistry , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/ultrastructure , Cryoelectron Microscopy , DNA/chemistry , DNA/metabolism , DNA/genetics , DNA/ultrastructure , Models, Molecular , Moloney murine leukemia virus/enzymology , Moloney murine leukemia virus/genetics , Ribonuclease H/deficiency , Ribonuclease H/genetics , RNA, Guide, CRISPR-Cas Systems/chemistry , RNA, Guide, CRISPR-Cas Systems/genetics , RNA, Guide, CRISPR-Cas Systems/metabolism , RNA, Guide, CRISPR-Cas Systems/ultrastructure , RNA-Directed DNA Polymerase/chemistry , RNA-Directed DNA Polymerase/metabolism , RNA-Directed DNA Polymerase/ultrastructure , Streptococcus pyogenes/enzymology , Streptococcus pyogenes/genetics , Viral Proteins/chemistry , Viral Proteins/metabolism , Viral Proteins/ultrastructure , Viral Proteins/genetics , HEK293 Cells
5.
Nucleic Acids Res ; 52(12): 7292-7304, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38806233

ABSTRACT

Herpes simplex virus 1 (HSV-1), a double-stranded DNA virus, replicates using seven essential proteins encoded by its genome. Among these, the UL30 DNA polymerase, complexed with the UL42 processivity factor, orchestrates leading and lagging strand replication of the 152 kb viral genome. UL30 polymerase is a prime target for antiviral therapy, and resistance to current drugs can arise in immunocompromised individuals. Using electron cryo-microscopy (cryo-EM), we unveil the dynamic changes of the UL30/UL42 complex with DNA in three distinct states. First, a pre-translocation state with an open fingers domain ready for nucleotide incorporation. Second, a halted elongation state where the fingers close, trapping dATP in the dNTP pocket. Third, a DNA-editing state involving significant conformational changes to allow DNA realignment for exonuclease activity. Additionally, the flexible UL30 C-terminal domain interacts with UL42, forming an extended positively charged surface binding to DNA, thereby enhancing processive synthesis. These findings highlight substantial structural shifts in the polymerase and its DNA interactions during replication, offering insights for future antiviral drug development.


Subject(s)
Cryoelectron Microscopy , DNA, Viral , DNA-Directed DNA Polymerase , Herpesvirus 1, Human , Viral Proteins , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/genetics , Viral Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/ultrastructure , Herpesvirus 1, Human/enzymology , Herpesvirus 1, Human/genetics , DNA, Viral/metabolism , DNA, Viral/biosynthesis , DNA Replication , Holoenzymes/chemistry , Holoenzymes/metabolism , Models, Molecular , Virus Replication , Protein Binding , Exodeoxyribonucleases
6.
Nucleic Acids Res ; 52(10): 6049-6065, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38709882

ABSTRACT

Severe fever with thrombocytopenia syndrome virus (SFTSV) is a human pathogen that is now endemic to several East Asian countries. The viral large (L) protein catalyzes viral transcription by stealing host mRNA caps via a process known as cap-snatching. Here, we establish an in vitro cap-snatching assay and present three high-quality electron cryo-microscopy (cryo-EM) structures of the SFTSV L protein in biologically relevant, transcription-specific states. In a priming-state structure, we show capped RNA bound to the L protein cap-binding domain (CBD). The L protein conformation in this priming structure is significantly different from published replication-state structures, in particular the N- and C-terminal domains. The capped-RNA is positioned in a way that it can feed directly into the RNA-dependent RNA polymerase (RdRp) ready for elongation. We also captured the L protein in an early-elongation state following primer-incorporation demonstrating that this priming conformation is retained at least in the very early stages of primer extension. This structural data is complemented by in vitro biochemical and cell-based assays. Together, these insights further our mechanistic understanding of how SFTSV and other bunyaviruses incorporate stolen host mRNA fragments into their viral transcripts thereby allowing the virus to hijack host cell translation machinery.


Subject(s)
Host Microbial Interactions , Models, Molecular , Phlebovirus , RNA Caps , Transcription, Genetic , Humans , Cryoelectron Microscopy , Phlebovirus/chemistry , Phlebovirus/genetics , Phlebovirus/ultrastructure , Protein Conformation , RNA Caps/chemistry , RNA Caps/metabolism , RNA Caps/ultrastructure , RNA, Viral/chemistry , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Viral Proteins/ultrastructure , Virus Replication/physiology , Host Microbial Interactions/physiology
7.
Nat Commun ; 15(1): 4620, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816392

ABSTRACT

Influenza viruses and thogotoviruses account for most recognized orthomyxoviruses. Thogotoviruses, exemplified by Thogoto virus (THOV), are capable of infecting humans using ticks as vectors. THOV transcribes mRNA without the extraneous 5' end sequences derived from cap-snatching in influenza virus mRNA. Here, we report cryo-EM structures to characterize THOV polymerase RNA synthesis initiation and elongation. The structures demonstrate that THOV RNA transcription and replication are able to start with short dinucleotide primers and that the polymerase cap-snatching machinery is likely non-functional. Triggered by RNA synthesis, asymmetric THOV polymerase dimers can form without the involvement of host factors. We confirm that, distinctive from influenza viruses, THOV-polymerase RNA synthesis is weakly dependent of the host factors ANP32A/B/E in human cells. This study demonstrates varied mechanisms in RNA synthesis and host factor utilization among orthomyxoviruses, providing insights into the mechanisms behind thogotoviruses' broad-infectivity range.


Subject(s)
Cryoelectron Microscopy , RNA, Viral , Thogotovirus , Transcription, Genetic , Virus Replication , Humans , Thogotovirus/genetics , Thogotovirus/metabolism , Thogotovirus/ultrastructure , RNA, Viral/metabolism , RNA, Viral/genetics , Virus Replication/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/chemistry , Viral Proteins/ultrastructure
8.
Annu Rev Biochem ; 93(1): 163-187, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38594919

ABSTRACT

Positive-strand RNA viruses encompass a variety of established and emerging eukaryotic pathogens. Their genome replication is confined to specialized cytoplasmic membrane compartments known as replication organelles (ROs). These ROs derive from host membranes, transformed into distinct structures such as invaginated spherules or intricate membrane networks including single- and/or double-membrane vesicles. ROs play a vital role in orchestrating viral RNA synthesis and evading detection by innate immune sensors of the host. In recent years, groundbreaking cryo-electron microscopy studies conducted with several prototypic viruses have significantly advanced our understanding of RO structure and function. Notably, these studies unveiled the presence of crown-shaped multimeric viral protein complexes that seem to actively participate in viral RNA synthesis and regulate the release of newly synthesized RNA into the cytosol for translation and packaging. These findings have shed light on novel viral functions and fascinating macromolecular complexes that delineate promising new avenues for future research.


Subject(s)
Cryoelectron Microscopy , RNA, Viral , Virus Replication , Cryoelectron Microscopy/methods , RNA, Viral/metabolism , RNA, Viral/genetics , RNA, Viral/chemistry , Humans , Positive-Strand RNA Viruses/metabolism , Positive-Strand RNA Viruses/genetics , Positive-Strand RNA Viruses/chemistry , Positive-Strand RNA Viruses/ultrastructure , Organelles/ultrastructure , Organelles/virology , Organelles/metabolism , Viral Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/ultrastructure , Animals , Viral Replication Compartments/metabolism , Viral Replication Compartments/ultrastructure
9.
J Virol ; 98(2): e0190023, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38289107

ABSTRACT

The endosomal sorting complex required for transport (ESCRT) is a conserved protein machine mediating membrane remodeling and scission. In the context of viral infection, different components of the ESCRT-III complex, which serve as the core machinery to catalyze membrane fission, are involved in diverse viruses' entry, replication, and/or budding. However, the interplay between ESCRT-III and viral factors in the virus life cycle, especially for that of large enveloped DNA viruses, is largely unknown. Recently, the ESCRT-III components Vps2B, Vps20, Vps24, Snf7, Vps46, and Vps60 were determined for entry and/or egress of the baculovirus Autographa californica multiple nucleopolyhedrovirus (AcMNPV). Here, we identified the final three ESCRT-III components Chm7, Ist1, and Vps2A of Spodoptera frugiperda. Overexpression of the dominant-negative forms of these proteins or RNAi downregulation of their transcripts significantly reduced infectious budded viruses (BVs) production of AcMNPV. Quantitative PCR together with confocal and transmission electron microscopy analysis revealed that these proteins were required for internalization and trafficking of BV during entry and egress of nucleocapsids. In infected Sf9 cells, nine ESCRT-III components were distributed on the nuclear envelope and plasma membrane, and except for Chm7, the other components were also localized to the intranuclear ring zone. Y2H and BiFC analysis revealed that 42 out of 64 BV-related proteins including 35 BV structural proteins and 7 non-BV structural proteins interacted with single or multiple ESCRT-III components. By further mapping the interactome of 64 BV-related proteins, we established the interaction networks of ESCRT-III and the viral protein complexes involved in BV entry and egress.IMPORTANCEFrom archaea to eukaryotes, the endosomal sorting complex required for transport (ESCRT)-III complex is hijacked by many enveloped and nonenveloped DNA or RNA viruses for efficient replication. However, the mechanism of ESCRT-III recruitment, especially for that of large enveloped DNA viruses, remains elusive. Recently, we found the ESCRT-III components Vps2B, Vps20, Vps24, Snf7, Vps46, and Vps60 are necessary for the entry and/or egress of budded viruses (BVs) of Autographa californica multiple nucleopolyhedrovirus. Here, we demonstrated that the other three ESCRT-III components Chm7, Ist1, and Vps2A play similar roles in BV infection. By determining the subcellular localization of ESCRT-III components in infected cells and mapping the interaction of nine ESCRT-III components and 64 BV-related proteins, we built the interaction networks of ESCRT-III and the viral protein complexes involved in BV entry and egress. These studies provide a fundamental basis for understanding the mechanism of the ESCRT-mediated membrane remodeling for replication of baculoviruses.


Subject(s)
Endosomal Sorting Complexes Required for Transport , Host Microbial Interactions , Nucleopolyhedroviruses , Spodoptera , Viral Proteins , Virus Internalization , Virus Release , Animals , Endosomal Sorting Complexes Required for Transport/chemistry , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomal Sorting Complexes Required for Transport/ultrastructure , Nucleopolyhedroviruses/metabolism , Nucleopolyhedroviruses/physiology , Nucleopolyhedroviruses/ultrastructure , Spodoptera/cytology , Spodoptera/metabolism , Spodoptera/ultrastructure , Spodoptera/virology , Viral Proteins/chemistry , Viral Proteins/metabolism , Viral Proteins/ultrastructure , Virus Replication , Biological Transport , Sf9 Cells
10.
Nature ; 617(7960): 409-416, 2023 05.
Article in English | MEDLINE | ID: mdl-37138077

ABSTRACT

CrAssphage and related viruses of the order Crassvirales (hereafter referred to as crassviruses) were originally discovered by cross-assembly of metagenomic sequences. They are the most abundant viruses in the human gut, are found in the majority of individual gut viromes, and account for up to 95% of the viral sequences in some individuals1-4. Crassviruses are likely to have major roles in shaping the composition and functionality of the human microbiome, but the structures and roles of most of the virally encoded proteins are unknown, with only generic predictions resulting from bioinformatic analyses4,5. Here we present a cryo-electron microscopy reconstruction of Bacteroides intestinalis virus ΦcrAss0016, providing the structural basis for the functional assignment of most of its virion proteins. The muzzle protein forms an assembly about 1 MDa in size at the end of the tail and exhibits a previously unknown fold that we designate the 'crass fold', that is likely to serve as a gatekeeper that controls the ejection of cargos. In addition to packing the approximately 103 kb of virus DNA, the ΦcrAss001 virion has extensive storage space for virally encoded cargo proteins in the capsid and, unusually, within the tail. One of the cargo proteins is present in both the capsid and the tail, suggesting a general mechanism for protein ejection, which involves partial unfolding of proteins during their extrusion through the tail. These findings provide a structural basis for understanding the mechanisms of assembly and infection of these highly abundant crassviruses.


Subject(s)
DNA Viruses , Intestines , Viral Proteins , Virion , Humans , Capsid/chemistry , Capsid/metabolism , Capsid/ultrastructure , Cryoelectron Microscopy , DNA Viruses/chemistry , DNA Viruses/classification , DNA Viruses/isolation & purification , DNA Viruses/metabolism , DNA Viruses/ultrastructure , Virion/chemistry , Virion/metabolism , Virion/ultrastructure , Virus Assembly , Intestines/microbiology , Intestines/virology , Viral Proteins/chemistry , Viral Proteins/metabolism , Viral Proteins/ultrastructure , Protein Unfolding , Protein Folding
11.
J Virol ; 97(3): e0158422, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36779755

ABSTRACT

Bacteriophages, viruses infecting bacteria, recognize their host with high specificity, binding to either saccharide motifs or proteins of the cell wall of their host. In the majority of bacteriophages, this host recognition is performed by receptor binding proteins (RBPs) located at the extremity of a tail. Interaction between the RBPs and the host is the trigger for bacteriophage infection, but the molecular details of the mechanisms are unknown for most bacteriophages. Here, we present the electron cryomicroscopy (cryo-EM) structure of bacteriophage T5 RBPpb5 in complex with its Escherichia coli receptor, the iron ferrichrome transporter FhuA. Monomeric RBPpb5 is located at the extremity of T5's long flexible tail, and its irreversible binding to FhuA commits T5 to infection. Analysis of the structure of RBPpb5 within the complex, comparison with its AlphaFold2-predicted structure, and its fit into a previously determined map of the T5 tail tip in complex with FhuA allow us to propose a mechanism of transmission of the RBPpb5 receptor binding to the straight fiber, initiating the cascade of events that commits T5 to DNA ejection. IMPORTANCE Tailed bacteriophages specifically recognize their bacterial host by interaction of their receptor binding protein(s) (RBPs) with saccharides and/or proteins located at the surface of their prey. This crucial interaction commits the virus to infection, but the molecular details of this mechanism are unknown for the majority of bacteriophages. We determined the structure of bacteriophage T5 RBPpb5 in complex with its E. coli receptor, FhuA, by cryo-EM. This first structure of an RBP bound to its protein receptor allowed us to propose a mechanism of transmission of host recognition to the rest of the phage, ultimately opening the capsid and perforating the cell wall and, thus, allowing safe channeling of the DNA into the host cytoplasm.


Subject(s)
Bacteriophages , Escherichia coli Proteins , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/ultrastructure , Bacteriophages/chemistry , Bacteriophages/metabolism , Escherichia coli/virology , Escherichia coli Proteins/chemistry , Protein Binding , Cryoelectron Microscopy , Viral Proteins/chemistry , Viral Proteins/metabolism , Viral Proteins/ultrastructure
13.
Nature ; 608(7922): 429-435, 2022 08.
Article in English | MEDLINE | ID: mdl-35922510

ABSTRACT

Bacteria encode myriad defences that target the genomes of infecting bacteriophage, including restriction-modification and CRISPR-Cas systems1. In response, one family of large bacteriophages uses a nucleus-like compartment to protect its replicating genomes by excluding host defence factors2-4. However, the principal composition and structure of this compartment remain unknown. Here we find that the bacteriophage nuclear shell assembles primarily from one protein, which we name chimallin (ChmA). Combining cryo-electron tomography of nuclear shells in bacteriophage-infected cells and cryo-electron microscopy of a minimal chimallin compartment in vitro, we show that chimallin self-assembles as a flexible sheet into closed micrometre-scale compartments. The architecture and assembly dynamics of the chimallin shell suggest mechanisms for its nucleation and growth, and its role as a scaffold for phage-encoded factors mediating macromolecular transport, cytoskeletal interactions, and viral maturation.


Subject(s)
Bacteria , Bacteriophages , Cell Compartmentation , Viral Proteins , Virus Assembly , Bacteria/cytology , Bacteria/immunology , Bacteria/metabolism , Bacteria/virology , Bacteriophages/chemistry , Bacteriophages/immunology , Bacteriophages/physiology , Bacteriophages/ultrastructure , Cryoelectron Microscopy , Viral Proteins/chemistry , Viral Proteins/metabolism , Viral Proteins/ultrastructure
14.
Sci Rep ; 12(1): 4631, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35301412

ABSTRACT

Using next generation sequencing technology, we identified a novel SARS-CoV-2 variant with a truncated ORF8 protein mutation near the end of the viral genome from nucleotides 27,878 to 27,958. This point mutation from C to T at nucleotide 27,956 changed the amino acid codon CAA (glutamine) to a stop codon, TAA, created a novel stop codon in ORF8 gene, resulting in a much smaller ORF8 protein (26 aa) than the wild type ORF8 protein (121 aa). This variant belongs to Pango lineage B.1.1291, which also contains the D614G mutation in the Spike (S) gene. The B.1.1291 lineage is predominantly circulated in the United States of America (97.18%), although it was also found in other counties (Russia, Canada, Latvia, Chile, India, Japan, Colombia, Germany, Greece, Mexico, and UK). A total of 340 closely related variants to this novel variant were identified in GISAID database with collection dates ranged from 3/6/2020 to 10/21/2020. In addition, a search within NCBI Genbank database found that 108,405 of 873,230 (12.4%) SAR-CoV-2 complete genomes contain this truncated ORF8 protein mutation, indicating this mutation may arise spontaneously in other lineages as well. The wide distribution of this mutation indicates that this truncated ORF8 protein mutation may provide the virus a growth advantage and adaptive evolution.


Subject(s)
COVID-19 , Chiroptera , SARS-CoV-2 , Viral Proteins , Animals , COVID-19/epidemiology , COVID-19/genetics , High-Throughput Nucleotide Sequencing , Humans , SARS-CoV-2/genetics , Viral Proteins/genetics , Viral Proteins/ultrastructure
16.
Structure ; 30(1): 55-68.e2, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34739839

ABSTRACT

Structural biologists provide direct insights into the molecular bases of human health and disease. The open-access Protein Data Bank (PDB) stores and delivers three-dimensional (3D) biostructure data that facilitate discovery and development of therapeutic agents and diagnostic tools. We are in the midst of a revolution in vaccinology. Non-infectious mRNA vaccines have been proven during the coronavirus disease 2019 (COVID-19) pandemic. This new technology underpins nimble discovery and clinical development platforms that use knowledge of 3D viral protein structures for societal benefit. The RCSB PDB supports vaccine designers through expert biocuration and rigorous validation of 3D structures; open-access dissemination of structure information; and search, visualization, and analysis tools for structure-guided design efforts. This resource article examines the structural biology underpinning the success of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) mRNA vaccines and enumerates some of the many protein structures in the PDB archive that could guide design of new countermeasures against existing and emerging viral pathogens.


Subject(s)
2019-nCoV Vaccine mRNA-1273/immunology , COVID-19/immunology , Computational Biology/methods , Databases, Protein , Protein Conformation , SARS-CoV-2/immunology , 2019-nCoV Vaccine mRNA-1273/administration & dosage , COVID-19/epidemiology , COVID-19/virology , Cryoelectron Microscopy , Crystallography, X-Ray , Drug Design , Humans , Internet , Models, Molecular , Pandemics/prevention & control , SARS-CoV-2/metabolism , SARS-CoV-2/physiology , Vaccination/methods , Vaccine Development/methods , Viral Proteins/chemistry , Viral Proteins/immunology , Viral Proteins/ultrastructure
17.
Front Immunol ; 12: 786617, 2021.
Article in English | MEDLINE | ID: mdl-34868073

ABSTRACT

Neuraminidase of influenza A and B viruses plays a critical role in the virus life cycle and is an important target of the host immune system. Here, we highlight the current understanding of influenza neuraminidase structure, function, antigenicity, immunogenicity, and immune protective potential. Neuraminidase inhibiting antibodies have been recognized as correlates of protection against disease caused by natural or experimental influenza A virus infection in humans. In the past years, we have witnessed an increasing interest in the use of influenza neuraminidase to improve the protective potential of currently used influenza vaccines. A number of well-characterized influenza neuraminidase-specific monoclonal antibodies have been described recently, most of which can protect in experimental challenge models by inhibiting the neuraminidase activity or by Fc receptor-dependent mechanisms. The relative instability of the neuraminidase poses a challenge for protein-based antigen design. We critically review the different solutions that have been proposed to solve this problem, ranging from the inclusion of stabilizing heterologous tetramerizing zippers to the introduction of inter-protomer stabilizing mutations. Computationally engineered neuraminidase antigens have been generated that offer broad, within subtype protection in animal challenge models. We also provide an overview of modern vaccine technology platforms that are compatible with the induction of robust neuraminidase-specific immune responses. In the near future, we will likely see the implementation of influenza vaccines that confront the influenza virus with a double punch: targeting both the hemagglutinin and the neuraminidase.


Subject(s)
Influenza Vaccines/immunology , Influenza, Human/prevention & control , Neuraminidase/immunology , Viral Proteins/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antigenic Drift and Shift , Antigens, Viral/immunology , Antigens, Viral/ultrastructure , Catalytic Domain/genetics , Catalytic Domain/immunology , Cross Protection , Evolution, Molecular , Humans , Immunogenicity, Vaccine , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza, Human/immunology , Influenza, Human/virology , Alphainfluenzavirus/enzymology , Alphainfluenzavirus/genetics , Alphainfluenzavirus/immunology , Betainfluenzavirus/enzymology , Betainfluenzavirus/genetics , Betainfluenzavirus/immunology , Mutation , Nanoparticles , Neuraminidase/administration & dosage , Neuraminidase/genetics , Neuraminidase/ultrastructure , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/ultrastructure , Viral Proteins/administration & dosage , Viral Proteins/genetics , Viral Proteins/ultrastructure
18.
Cells ; 10(11)2021 11 11.
Article in English | MEDLINE | ID: mdl-34831342

ABSTRACT

The regulation of the nucleocytoplasmic release of herpesviral capsids is defined by the process of nuclear egress. Due to their large size, nuclear capsids are unable to traverse via nuclear pores, so that herpesviruses evolved to develop a vesicular transport pathway mediating their transition through both leaflets of the nuclear membrane. This process involves regulatory proteins, which support the local distortion of the nuclear envelope. For human cytomegalovirus (HCMV), the nuclear egress complex (NEC) is determined by the pUL50-pUL53 core that initiates multicomponent assembly with NEC-associated proteins and capsids. Hereby, pUL50 serves as a multi-interacting determinant that recruits several viral and cellular factors by direct and indirect contacts. Recently, we generated an ORF-UL50-deleted recombinant HCMV in pUL50-complementing cells and obtained first indications of putative additional functions of pUL50. In this study, we produced purified ΔUL50 particles under both complementing (ΔUL50C) and non-complementing (ΔUL50N) conditions and performed a phenotypical characterization. Findings were as follows: (i) ΔUL50N particle preparations exhibited a clear replicative defect in qPCR-based infection kinetics compared to ΔUL50C particles; (ii) immuno-EM analysis of ΔUL50C did not reveal major changes in nuclear distribution of pUL53 and lamin A/C; (iii) mass spectrometry-based quantitative proteomics showed a large concordance of protein contents in the NIEP fractions of ΔUL50C and ΔUL50N particles, but virion fraction was close to the detection limit for ΔUL50N; (iv) confocal imaging of viral marker proteins of immediate early (IE) and later phases of ΔUL50N infection indicated a very low number of cells showing an onset of viral lytic protein expression; and, finally (v) quantitative measurements of encapsidated genomes provided evidence for a substantial reduction in the DNA contents in ΔUL50N compared to ΔUL50C particles. In summary, the results point to a complex and important regulatory role of the HCMV nuclear egress protein pUL50 in the maturation of infectious virus.


Subject(s)
Cell Nucleus/metabolism , Cytomegalovirus/pathogenicity , Viral Proteins/metabolism , Capsid/metabolism , Capsid/ultrastructure , Cell Line , Cytomegalovirus/genetics , Cytomegalovirus/ultrastructure , DNA Packaging/genetics , Fibroblasts/metabolism , Fibroblasts/virology , Gene Expression Regulation, Viral , Genes, Immediate-Early , Genome, Viral , Humans , Kinetics , Nuclear Envelope/metabolism , Proteomics , Viral Proteins/ultrastructure , Virion/metabolism , Virion/ultrastructure , Virus Replication/physiology
19.
Biomolecules ; 11(10)2021 09 29.
Article in English | MEDLINE | ID: mdl-34680060

ABSTRACT

Many biological and biotechnological processes are controlled by protein-protein and protein-solvent interactions. In order to understand, predict, and optimize such processes, it is important to understand how solvents affect protein structure during protein-solvent interactions. In this study, all-atom molecular dynamics are used to investigate the structural dynamics and energetic properties of a C-terminal domain of the Rift Valley Fever Virus L protein solvated in glycerol and aqueous glycerol solutions in different concentrations by molecular weight. The Generalized Amber Force Field is modified by including restrained electrostatic potential atomic charges for the glycerol molecules. The peptide is considered in detail by monitoring properties like the root-mean-squared deviation, root-mean-squared fluctuation, radius of gyration, hydrodynamic radius, end-to-end distance, solvent-accessible surface area, intra-potential energy, and solvent-peptide interaction energies for hundreds of nanoseconds. Secondary structure analysis is also performed to examine the extent of conformational drift for the individual helices and sheets. We predict that the peptide helices and sheets are maintained only when the modeling strategy considers the solvent with lower glycerol concentration. We also find that the solvent-peptide becomes more cohesive with decreasing glycerol concentrations. The density and radial distribution function of glycerol solvent calculated when modeled with the modified atomic charges show a very good agreement with experimental results and other simulations at 298.15K.


Subject(s)
Glycerol/chemistry , Rift Valley fever virus/ultrastructure , Viral Proteins/ultrastructure , Water/chemistry , Humans , Hydrogen Bonding , Molecular Dynamics Simulation , Peptides/chemistry , Protein Domains/genetics , Protein Structure, Secondary , Rift Valley fever virus/chemistry , Rift Valley fever virus/genetics , Solvents/chemistry , Viral Proteins/chemistry , Viral Proteins/genetics
20.
Biomolecules ; 11(9)2021 09 07.
Article in English | MEDLINE | ID: mdl-34572537

ABSTRACT

Henipaviruses are BSL-4 zoonotic pathogens responsible in humans for severe encephalitis. Their V protein is a key player in the evasion of the host innate immune response. We previously showed that the Henipavirus V proteins consist of a long intrinsically disordered N-terminal domain (NTD) and a ß-enriched C-terminal domain (CTD). These terminals are critical for V binding to DDB1, which is a cellular protein that is a component of the ubiquitin ligase E3 complex, as well as binding to MDA5 and LGP2, which are two host sensors of viral RNA. Here, we serendipitously discovered that the Hendra virus V protein undergoes a liquid-to-hydrogel phase transition and identified the V region responsible for this phenomenon. This region, referred to as PNT3 and encompassing residues 200-310, was further investigated using a combination of biophysical and structural approaches. Congo red binding assays, together with negative-staining transmisison electron microscopy (TEM) studies, show that PNT3 forms amyloid-like fibrils. Fibrillation abilities are dramatically reduced in a rationally designed PNT3 variant in which a stretch of three contiguous tyrosines, falling within an amyloidogenic motif, were replaced by three alanines. Worthy to note, Congo red staining experiments provided hints that these amyloid-like fibrils form not only in vitro but also in cellula after transfection or infection. The present results set the stage for further investigations aimed at assessing the functional role of phase separation and fibrillation by the Henipavirus V proteins.


Subject(s)
Amyloid/metabolism , Hendra Virus/metabolism , Phase Transition , Viral Proteins/chemistry , Viral Proteins/metabolism , Amino Acid Sequence , Congo Red/metabolism , HEK293 Cells , HSP70 Heat-Shock Proteins/metabolism , Humans , Hydrogels/chemistry , Magnetic Resonance Spectroscopy , Protein Domains , Scattering, Small Angle , Viral Proteins/ultrastructure , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL