Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 178
Filter
Add more filters










Publication year range
1.
mBio ; 15(2): e0313423, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38171004

ABSTRACT

Vaccinia virus assembly in the cytoplasm of infected cells involves the formation of a biconcave viral core inside the maturing viral particle. The boundary of the core is defined by a pseudohexagonal palisade layer, composed of trimers projecting from an inner wall. To understand the assembly of this complex core architecture, we obtained a subnanometer structure of the palisade trimer by cryo-electron tomography and subtomogram averaging of purified intact virions. Using AlphaFold2 structure predictions, we determined that the palisade is formed from trimers of the proteolytically processed form of the viral protein A10. In addition, we found that each A10 protomer associates with an α-helix (residues 24-66) of A4. Cellular localization assays outside the context of infection demonstrate that the A4 N-terminus is necessary and sufficient to interact with A10. The interaction between A4 and A10 provides insights into how the palisade layer might become tightly associated with the viral membrane during virion maturation. Reconstruction of the palisade layer reveals that, despite local hexagonal ordering, the A10/A4 trimers are widely spaced, suggesting that additional components organize the lattice. This spacing would, however, allow the adoption of the characteristic biconcave shape of the viral core. Finally, we also found that the palisade incorporates multiple copies of a hexameric portal structure. We suggest that these portals are formed by E6, a viral protein that is essential for virion assembly and required to release viral mRNA from the core early in infection.IMPORTANCEPoxviruses such as variola virus (smallpox) and monkeypox cause diseases in humans. Other poxviruses, including vaccinia and modified vaccinia Ankara, are used as vaccine vectors. Given their importance, a greater structural understanding of poxvirus virions is needed. We now performed cryo-electron tomography of purified intact vaccinia virions to study the structure of the palisade, a protein lattice that defines the viral core boundary. We identified the main viral proteins that form the palisade and their interaction surfaces and provided new insights into the organization of the viral core.


Subject(s)
Benzeneacetamides , Piperidones , Vaccinia virus , Vaccinia , Humans , Vaccinia virus/chemistry , Virus Assembly , Virion/genetics , Viral Proteins/metabolism
2.
J Virol ; 97(12): e0134323, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-37975688

ABSTRACT

IMPORTANCE: Vaccinia virus infection requires virus-cell membrane fusion to complete entry during endocytosis; however, it contains a large viral fusion protein complex of 11 viral proteins that share no structure or sequence homology to all the known viral fusion proteins, including type I, II, and III fusion proteins. It is thus very challenging to investigate how the vaccinia fusion complex works to trigger membrane fusion with host cells. In this study, we crystallized the ectodomain of vaccinia H2 protein, one component of the viral fusion complex. Furthermore, we performed a series of mutational, biochemical, and molecular analyses and identified two surface loops containing 170LGYSG174 and 125RRGTGDAW132 as the A28-binding region. We also showed that residues in the N-terminal helical region (amino acids 51-90) are also important for H2 function.


Subject(s)
Membrane Fusion , Vaccinia virus , Viral Fusion Proteins , Virus Internalization , Vaccinia virus/chemistry , Vaccinia virus/genetics , Vaccinia virus/metabolism , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism
3.
mBio ; 14(5): e0113523, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37646531

ABSTRACT

IMPORTANCE: An outstanding problem in the understanding of poxvirus biology is the molecular structure of the mature virion. Via deep learning methods combined with chemical cross-linking mass spectrometry, we have addressed the structure and assembly pathway of P4a, a key poxvirus virion core component.


Subject(s)
Deep Learning , Poxviridae , Vaccinia , Humans , Vaccinia virus/chemistry , Virion/metabolism
4.
Sci Rep ; 12(1): 13007, 2022 07 29.
Article in English | MEDLINE | ID: mdl-35906465

ABSTRACT

Viruses of the giant virus family are characterized by a structurally conserved scaffold-capsid protein that shapes the icosahedral virion. The vaccinia virus (VACV) scaffold protein D13, however, transiently shapes the newly assembled viral membrane in to a sphere and is absent from the mature brick-shaped virion. In infected cells D13, a 62 kDa polypeptide, forms trimers that arrange in hexamers and a honey-comb like lattice. Membrane association of the D13-lattice may be mediated by A17, an abundant 21 kDa viral membrane protein. Whether membrane binding mediates the formation of the honey-comb lattice or if other factors are involved, remains elusive. Here we show that H7, a 17 kDa protein conserved among poxviruses, mediates proper formation of D13-hexamers, and hence the honey comb lattice and spherical immature virus. Without H7 synthesis D13 trimers assemble into a large 3D network rather than the typical well organized scaffold layer observed in wild-type infection, composed of short D13 tubes of discrete length that are tightly associated with the endoplasmic reticulum (ER). The data show an unexpected role for H7 in D13 organization and imply that formation of the honey-comb, hexagonal, lattice is essential for VACV membrane assembly and production of infectious progeny. The data are discussed with respect to scaffold proteins of other giant viruses.


Subject(s)
Vaccinia virus , Vaccinia , Humans , Vaccinia virus/chemistry , Viral Proteins/metabolism , Virion/metabolism , Virus Assembly
5.
Viruses ; 14(2)2022 01 20.
Article in English | MEDLINE | ID: mdl-35215792

ABSTRACT

Vaccinia virus (VACV) belongs to the genus Orthopoxvirus of the family Poxviridae. There are four different forms of infectious virus particles: intracellular mature virus (IMV), intracellular en-veloped virus (IEV), cell-associated enveloped virus (CEV) and extracellular enveloped virus (EEV). The F13 protein occupies the inner side of the CEV- and EEV-membranes and the outer side of the IEV-membranes. It plays an important role in wrapping progress and EEV production. We constructed a human single-chain fragment variable (scFv) library with a diversity of ≥4 × 108 independent colonies using peripheral blood from four vaccinated donors. One anti-F13 scFv was isolated and characterised after three rounds of panning. In Western blotting assays, the scFv 3E2 reacted with the recombinant F13VACV protein with a reduction of binding under denatured and reduced conditions. Two antigenic binding sites (139-GSIHTIKTLGVYSDY-153 and 169-AFNSAKNSWLNL-188) of scFv 3E2 were mapped using a cellulose membrane encompassing 372 15-mere peptides with 12 overlaps covering the whole F13 protein. No neutralisation capa-bilities were observed either in the presence or absence of complement. In conclusion, the con-struction of recombinant immunoglobulin libraries is a promising strategy to isolate specific scFvs to enable the study of the host-pathogen interaction.


Subject(s)
Antibodies, Viral/immunology , Single-Chain Antibodies/immunology , Vaccinia virus/immunology , Amino Acid Sequence , Antibodies, Viral/chemistry , Antibodies, Viral/genetics , Epitope Mapping , Gene Library , Humans , Neutralization Tests , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Vaccinia virus/chemistry , Vaccinia virus/genetics
6.
J Gen Virol ; 103(1)2022 01.
Article in English | MEDLINE | ID: mdl-35020582

ABSTRACT

The morphogenesis of vaccinia virus (VACV, family Poxviridae), the smallpox vaccine, is a complex process involving multiple distinct cellular membranes and resulting in multiple different forms of infectious virion. Efficient release of enveloped virions, which promote systemic spread of infection within hosts, requires the VACV protein E2 but the molecular basis of E2 function remains unclear and E2 lacks sequence homology to any well-characterised family of proteins. We solved the crystal structure of VACV E2 to 2.3 Å resolution, revealing that it comprises two domains with novel folds: an N-terminal annular (ring) domain and a C-terminal globular (head) domain. The C-terminal head domain displays weak structural homology with cellular (pseudo)kinases but lacks conserved surface residues or kinase features, suggesting that it is not enzymatically active, and possesses a large surface basic patch that might interact with phosphoinositide lipid headgroups. Recent deep learning methods have revolutionised our ability to predict the three-dimensional structures of proteins from primary sequence alone. VACV E2 is an exemplar 'difficult' viral protein target for structure prediction, being comprised of multiple novel domains and lacking sequence homologues outside Poxviridae. AlphaFold2 nonetheless succeeds in predicting the structures of the head and ring domains with high and moderate accuracy, respectively, allowing accurate inference of multiple structural properties. The advent of highly accurate virus structure prediction marks a step-change in structural virology and beckons a new era of structurally-informed molecular virology.


Subject(s)
Poxviridae/metabolism , Vaccinia virus/chemistry , Vaccinia virus/physiology , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Replication , Binding Sites , Crystallography, X-Ray , Protein Binding , Protein Conformation , Vaccinia virus/genetics , Viral Proteins/genetics
7.
Nat Struct Mol Biol ; 28(10): 779-788, 2021 10.
Article in English | MEDLINE | ID: mdl-34556871

ABSTRACT

Poxviruses express their genes in the cytoplasm of infected cells using a virus-encoded multi-subunit polymerase (vRNAP) and unique transcription factors. We present cryo-EM structures that uncover the complete transcription initiation phase of the poxvirus vaccinia. In the pre-initiation complex, the heterodimeric early transcription factor VETFs/l adopts an arc-like shape spanning the polymerase cleft and anchoring upstream and downstream promoter elements. VETFI emerges as a TBP-like protein that inserts asymmetrically into the DNA major groove, triggers DNA melting, ensures promoter recognition and enforces transcription directionality. The helicase VETFs fosters promoter melting and the phospho-peptide domain (PPD) of vRNAP subunit Rpo30 enables transcription initiation. An unprecedented upstream promoter scrunching mechanism assisted by the helicase NPH-I probably fosters promoter escape and transition into elongation. Our structures shed light on unique mechanisms of poxviral gene expression and aid the understanding of thus far unexplained universal principles in transcription.


Subject(s)
Transcription Initiation, Genetic , Vaccinia virus/chemistry , Vaccinia virus/genetics , Viral Proteins/chemistry , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Cryoelectron Microscopy , DNA Helicases/chemistry , DNA Helicases/genetics , DNA, Viral/chemistry , DNA, Viral/metabolism , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Gene Expression Regulation, Viral , HeLa Cells , Humans , Models, Molecular , Promoter Regions, Genetic , Protein Domains , Protein Subunits , Transcription Factors/chemistry , Transcription Factors/genetics , Viral Proteins/genetics
8.
mBio ; 12(1)2021 01 19.
Article in English | MEDLINE | ID: mdl-33468698

ABSTRACT

Immunotherapy for cervical cancer should target high-risk human papillomavirus types 16 and 18, which cause 50% and 20% of cervical cancers, respectively. Here, we describe the construction and characterization of the pBI-11 DNA vaccine via the addition of codon-optimized human papillomavirus 18 (HPV18) E7 and HPV16 and 18 E6 genes to the HPV16 E7-targeted DNA vaccine pNGVL4a-SigE7(detox)HSP70 (DNA vaccine pBI-1). Codon optimization of the HPV16/18 E6/E7 genes in pBI-11 improved fusion protein expression compared to that in DNA vaccine pBI-10.1 that utilized the native viral sequences fused 3' to a signal sequence and 5' to the HSP70 gene of Mycobacterium tuberculosis Intramuscular vaccination of mice with pBI-11 DNA better induced HPV antigen-specific CD8+ T cell immune responses than pBI-10.1 DNA. Furthermore, intramuscular vaccination with pBI-11 DNA generated stronger therapeutic responses for C57BL/6 mice bearing HPV16 E6/E7-expressing TC-1 tumors. The HPV16/18 antigen-specific T cell-mediated immune responses generated by pBI-11 DNA vaccination were further enhanced by boosting with tissue-antigen HPV vaccine (TA-HPV). Combination of the pBI-11 DNA and TA-HPV boost vaccination with PD-1 antibody blockade significantly improved the control of TC-1 tumors and extended the survival of the mice. Finally, repeat vaccination with clinical-grade pBI-11 with or without clinical-grade TA-HPV was well tolerated in vaccinated mice. These preclinical studies suggest that the pBI-11 DNA vaccine may be used with TA-HPV in a heterologous prime-boost strategy to enhance HPV 16/18 E6/E7-specific CD8+ T cell responses, either alone or in combination with immune checkpoint blockade, to control HPV16/18-associated tumors. Our data serve as an important foundation for future clinical translation.IMPORTANCE Persistent expression of high-risk human papillomavirus (HPV) E6 and E7 is an obligate driver for several human malignancies, including cervical cancer, wherein HPV16 and HPV18 are the most common types. PD-1 antibody immunotherapy helps a subset of cervical cancer patients, and its efficacy might be improved by combination with active vaccination against E6 and/or E7. For patients with HPV16+ cervical intraepithelial neoplasia grade 2/3 (CIN2/3), the precursor of cervical cancer, intramuscular vaccination with a DNA vaccine targeting HPV16 E7 and then a recombinant vaccinia virus expressing HPV16/18 E6-E7 fusion proteins (TA-HPV) was safe, and half of the patients cleared their lesions in a small study (NCT00788164). Here, we sought to improve upon this therapeutic approach by developing a new DNA vaccine that targets E6 and E7 of HPV16 and HPV18 for administration prior to a TA-HPV booster vaccination and for application against cervical cancer in combination with a PD-1-blocking antibody.


Subject(s)
Human papillomavirus 16/genetics , Human papillomavirus 18/genetics , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/genetics , Uterine Cervical Neoplasms/prevention & control , Vaccines, DNA/genetics , Animals , Antibodies, Monoclonal/administration & dosage , Bacterial Proteins/genetics , Bacterial Proteins/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Female , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/immunology , Human papillomavirus 16/drug effects , Human papillomavirus 16/immunology , Human papillomavirus 18/drug effects , Human papillomavirus 18/immunology , Humans , Immunization, Secondary/methods , Mice , Mice, Inbred C57BL , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/genetics , Papillomavirus Infections/immunology , Papillomavirus Infections/mortality , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Protein Engineering/methods , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Repressor Proteins/genetics , Repressor Proteins/immunology , Survival Analysis , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/mortality , Vaccination/methods , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Vaccinia virus/chemistry , Vaccinia virus/immunology
9.
J Virol ; 94(20)2020 09 29.
Article in English | MEDLINE | ID: mdl-32759313

ABSTRACT

Robust priming of CD8+ T cells by viruses is considered to require infection and de novo expression of viral antigens. A corollary of this is that inactivated viruses are thought of as being inevitably poor vaccines for eliciting these responses. In contrast to this dogma, we found that some antigens present in vaccinia virus (VACV) virions prime strong CD8+ T cell responses when the virus was rendered noninfectious by heat. More surprisingly, in some cases these responses were similar in magnitude to those primed by infectious virus administered at an equivalent dose. Next, we tested whether this was a special property of particular antigens and their epitopes and found that foreign epitopes tagged onto three different VACV virion proteins were able to elicit CD8+ T cell responses irrespective of whether the virus was viable or heat killed. Further, the polyfunctionality and cytotoxic ability of the CD8+ T cells primed by these VACVs was equivalent irrespective of whether they were administered to mice as inactivated or live viruses. Finally, we used these VACVs in prime-boost combinations of inactivated and live virus and found that priming with dead virus before a live booster was the most immunogenic regime. We conclude that VACV virions can be efficient vectors for targeting antigens to dendritic cells for effective priming of CD8+ T cells, even when rendered noninfectious and speculate that this might also be the case for other viruses.IMPORTANCE The design of viral vectored vaccines is often considered to require a trade-off between efficacy and safety. This is especially the case for vaccines that aim to induce killer (CD8+) T cells, where there is a well-established dogma that links infection in vaccinated individuals with effective induction of immunity. However, we found that some proteins of vaccinia virus generate strong CD8+ T cell responses even when the virus preparation was inactivated by heat prior to administration as a vaccine. We took advantage of this finding by engineering a new vaccine vector virus that could be used as an inactivated vaccine. These results suggest that vaccinia virus may be a more versatile vaccine vector than previously appreciated and that in some instances safety can be prioritized by the complete elimination of viral replication without a proportional loss of immunogenicity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Hot Temperature , Immunization, Secondary , Vaccinia virus , Virion , Virus Inactivation , Animals , Cell Line , Mice , Vaccinia virus/chemistry , Vaccinia virus/immunology , Viral Proteins/chemistry , Viral Proteins/immunology , Virion/chemistry , Virion/immunology
10.
J Biol Chem ; 294(16): 6416-6429, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30819806

ABSTRACT

BTB-Kelch proteins are substrate-specific adaptors for cullin-3 (Cul3) RING-box-based E3 ubiquitin ligases, mediating protein ubiquitylation for subsequent proteasomal degradation. Vaccinia virus encodes three BTB-Kelch proteins: A55, C2, and F3. Viruses lacking A55 or C2 have altered cytopathic effects in cultured cells and altered pathology in vivo Previous studies have shown that the ectromelia virus orthologue of A55 interacts with Cul3 in cells. We report that the N-terminal BTB-BACK (BB) domain of A55 binds directly to the Cul3 N-terminal domain (Cul3-NTD), forming a 2:2 complex in solution. We solved the structure of an A55BB/Cul3-NTD complex from anisotropic crystals diffracting to 2.3/3.7 Å resolution in the best/worst direction, revealing that the overall interaction and binding interface closely resemble the structures of cellular BTB/Cul3-NTD complexes, despite low sequence identity between A55 and cellular BTB domains. Surprisingly, despite this structural similarity, the affinity of Cul3-NTD for A55BB was stronger than for cellular BTB proteins. Glutamate substitution of the A55 residue Ile-48, adjacent to the canonical φX(D/E) Cul3-binding motif, reduced affinity of A55BB for Cul3-NTD by at least 2 orders of magnitude. Moreover, Ile-48 and the φX(D/E) motif are conserved in A55 orthologues from other poxviruses, but not in the vaccinia virus proteins C2 or F3. The high-affinity interaction between A55BB and Cul3-NTD suggests that, in addition to directing the Cul3-RING E3 ligase complex to degrade cellular/viral target proteins that are normally unaffected, A55 may also sequester Cul3 from cellular adaptor proteins, thereby protecting substrates of these cellular adaptors from ubiquitylation and degradation.


Subject(s)
Cullin Proteins/chemistry , Multiprotein Complexes/chemistry , Vaccinia virus/chemistry , Viral Proteins/chemistry , Amino Acid Substitution , Cullin Proteins/genetics , Cullin Proteins/metabolism , HEK293 Cells , Humans , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutation, Missense , Protein Domains , Protein Structure, Quaternary , Proteolysis , Ubiquitination/genetics , Vaccinia/genetics , Vaccinia/metabolism , Vaccinia virus/genetics , Vaccinia virus/metabolism , Viral Proteins/genetics
11.
J Proteome Res ; 18(3): 900-911, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30629447

ABSTRACT

Protective cellular and humoral immune responses require previous recognition of viral antigenic peptides complexed with human leukocyte antigen (HLA) class II molecules on the surface of the antigen presenting cells. The HLA class II-restricted immune response is important for the control and the clearance of poxvirus infection including vaccinia virus (VACV), the vaccine used in the worldwide eradication of smallpox. In this study, a mass spectrometry analysis was used to identify VACV ligands bound to HLA-DR and -DP class II molecules present on the surface of VACV-infected cells. Twenty-six naturally processed viral ligands among the tens of thousands of cell peptides bound to HLA class II proteins were identified. These viral ligands arose from 19 parental VACV proteins: A4, A5, A18, A35, A38, B5, B13, D1, D5, D7, D12, D13, E3, E8, H5, I2, I3, J2, and K2. The majority of these VACV proteins yielded one HLA ligand and were generated mainly, but not exclusively, by the classical HLA class II antigen processing pathway. Medium-sized and abundant proteins from the virion core and/or involved in the viral gene expression were the major source of VACV ligands bound to HLA-DR and -DP class II molecules. These findings will help to understand the effectiveness of current poxvirus-based vaccines and will be important in the design of new ones.


Subject(s)
Histocompatibility Antigens Class II/metabolism , Ligands , Proteomics/methods , Vaccinia virus/chemistry , Viral Structural Proteins , Virion/chemistry , Cells, Cultured , Gene Expression , Humans , Mass Spectrometry , Poxviridae/immunology , Vaccinia/immunology , Viral Proteins/immunology , Viral Structural Proteins/immunology , Viral Vaccines
12.
Sci Rep ; 8(1): 16807, 2018 11 14.
Article in English | MEDLINE | ID: mdl-30429486

ABSTRACT

The poxvirus F9 protein is a component of the vaccinia virus entry fusion complex (EFC) which consists of 11 proteins. The EFC forms a unique apparatus among viral fusion proteins and complexes. We solved the atomic structure of the F9 ectodomain at 2.10 Å. A structural comparison to the ectodomain of the EFC protein L1 indicated a similar fold and organization, in which a bundle of five α-helices is packed against two pairs of ß-strands. However, instead of the L1 myristoylation site and hydrophobic cavity, F9 possesses a protruding loop between α-helices α3 and α4 starting at Gly90. Gly90 is conserved in all poxviruses except Salmon gill poxvirus (SGPV) and Diachasmimorpha longicaudata entomopoxvirus. Phylogenetic sequence analysis of all Poxviridae F9 and L1 orthologs revealed the SGPV genome to contain the most distantly related F9 and L1 sequences compared to the vaccinia proteins studied here. The structural differences between F9 and L1 suggest functional adaptations during evolution from a common precursor that underlie the present requirement for each protein.


Subject(s)
Membrane Fusion , Poxviridae/chemistry , Viral Proteins/physiology , Virus Internalization , Amino Acid Sequence , Animals , Conserved Sequence , Evolution, Molecular , Phylogeny , Protein Conformation , Vaccinia virus/chemistry , Viral Proteins/analysis , Viral Proteins/chemistry
13.
Anal Chem ; 90(22): 13236-13242, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30352503

ABSTRACT

A laser-induced rf plasma (LIRFP) ion source was developed to ionize submicrometer-sized particles for the first time. The LIRFP ion source can increase the charge of those particles to several thousand charges via charge exchange reactions so that those particles can be trapped and analyzed with a charge detection quadrupole ion trap-mass spectrometer (CD QIT-MS). Different reagent gases for charge exchange reaction were investigated, viz. argon, nitrogen, oxygen, methane, helium, krypton, xenon, argon/methane (with ratios of 10:1 and 2:1), argon/nitrogen (with a ratio of 1:1), nitrogen/oxygen (10:1), krypton/methane (10:1), and air. The average charge of 0.75 µm polystyrene particles could reach 1631 using an argon/methane mixture with a ratio of ∼10:1. The average charges for freeze-dried Escherichia coli EC11303, Escherichia coli strain W, and Staphylococcus aureus were 842, 1112, and 971, respectively, with a mass-to-charge ratio ( m/ z) range from 107 to 108; and the average masses were 3.5 × 1010 Da, 6.0 × 1010 Da, and 5.6 × 1010 Da, respectively. The average mass and charge of the vaccinia virus were ∼9.1 × 109 Da and ∼708 with a m/ z of ∼107. This LIRFP CD QIT-MS method was rapid with only 20 min for each sample measurement.


Subject(s)
Gases/chemistry , Ions/chemistry , Escherichia coli/chemistry , Lasers , Mass Spectrometry/instrumentation , Mass Spectrometry/methods , Particle Size , Polystyrenes/chemistry , Radio Waves , Staphylococcus aureus/chemistry , Static Electricity , Vaccinia virus/chemistry
14.
J Biol Chem ; 293(41): 16057-16068, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30171073

ABSTRACT

IFNϵ and IFNκ are interferons that induce microbial immunity at mucosal surfaces and in the skin. They are members of the type-I interferon (IFN) family, which consists of 16 different IFNs, that all signal through the common IFNAR1/IFNAR2 receptor complex. Although IFNϵ and IFNκ have unique expression and functional properties, their biophysical properties have not been extensively studied. In this report, we describe the expression, purification, and characterization of recombinant human IFNϵ and IFNκ. In cellular assays, IFNϵ and IFNκ exhibit ∼1000-fold lower potency than IFNα2 and IFNω. The reduced potency of IFNϵ and IFNκ are consistent with their weak affinity for the IFNAR2 receptor chain. Despite reduced IFNAR2-binding affinities, IFNϵ and IFNκ exhibit affinities for the IFNAR1 chain that are similar to other IFN subtypes. As observed for cellular IFNAR2 receptor, the poxvirus antagonist, B18R, also exhibits reduced affinity for IFNϵ and IFNκ, relative to the other IFNs. Taken together, our data suggest IFNϵ and IFNκ are specialized IFNs that have evolved to weakly bind to the IFNAR2 chain, which allows innate protection of the mucosa and skin and limits neutralization of IFNϵ and IFNκ biological activities by viral IFN antagonists.


Subject(s)
Interferon Type I/metabolism , Interferons/metabolism , Receptor, Interferon alpha-beta/metabolism , Viral Proteins/metabolism , Cell Line , Gene Expression , Humans , Interferon Type I/genetics , Interferons/genetics , Models, Molecular , Mutation , Protein Binding , Vaccinia virus/chemistry
15.
Proc Natl Acad Sci U S A ; 115(27): 7028-7032, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29915071

ABSTRACT

Cellular membranes are maintained as closed compartments, broken up only transiently during membrane reorganization or lipid transportation. However, open-ended membranes, likely derived from scissions of the endoplasmic reticulum, persist in vaccinia virus-infected cells during the assembly of the viral envelope. A group of viral membrane assembly proteins (VMAPs) were identified as essential for this process. To understand the mechanism of VMAPs, we determined the 2.2-Å crystal structure of the largest member, named A6, which is a soluble protein with two distinct domains. The structure of A6 displays a novel protein fold composed mainly of alpha helices. The larger C-terminal domain forms a unique cage that encloses multiple glycerophospholipids with a lipid bilayer-like configuration. The smaller N-terminal domain does not bind lipid but negatively affects lipid binding by A6. Mutations of key hydrophobic residues lining the lipid-binding cage disrupt lipid binding and abolish viral replication. Our results reveal a protein modality for enclosing the lipid bilayer and provide molecular insight into a viral machinery involved in generating and/or stabilizing open-ended membranes.


Subject(s)
Lipid Bilayers/chemistry , Membrane Proteins/chemistry , Vaccinia virus/chemistry , Viral Proteins/chemistry , Crystallography, X-Ray , Membrane Proteins/genetics , Vaccinia virus/genetics , Viral Proteins/genetics
16.
Vaccine ; 36(9): 1174-1182, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29398273

ABSTRACT

Inactivated influenza vaccines are not approved for use in infants less than 6 months of age due to poor immunogenicity in that population. While the live attenuated influenza vaccine has the potential to be more immunogenic, it is not an option for infants and other vulnerable populations, including the elderly and immunocompromised individuals due to safety concerns. In an effort to improve the immunogenicity of the inactivated vaccine for use in vulnerable populations, we have used an approach of chemically crosslinking the Toll-like receptor (TLR) 7/8 agonist R848 directly to virus particles. We have reported previously that an R848-conjugated, inactivated vaccine is more effective at inducing adaptive immune responses and protecting against lung pathology in influenza challenged neonatal African green monkeys than is the unmodified counterpart. In the current study, we describe a second generation vaccine that utilizes an amide-sulfhydryl crosslinker with different spacer chemistry and length to couple R848 to virions. The new vaccine has significantly enhanced immunostimulatory activity for murine macrophages and importantly for monocyte derived human dendritic cells. Demonstration of the significant differences in stimulatory activity afforded by modest changes in linker impacts our fundamental view of the design of TLR agonist-antigen vaccines.


Subject(s)
Imidazoles/immunology , Influenza Vaccines/immunology , Vaccines, Conjugate/immunology , Vaccines, Inactivated/immunology , Adaptive Immunity/drug effects , Animals , Dendritic Cells/immunology , Dendritic Cells/virology , Humans , Imidazoles/chemistry , Influenza A virus , Influenza Vaccines/chemistry , Mice , RAW 264.7 Cells , Vaccines, Conjugate/chemistry , Vaccines, Inactivated/chemistry , Vaccinia virus/chemistry
17.
Int J Mol Sci ; 18(8)2017 Aug 16.
Article in English | MEDLINE | ID: mdl-28813018

ABSTRACT

Certain viruses have the ability to subvert the mammalian immune response, including interference in the chemokine system. Poxviruses produce the chemokine binding protein vCCI (viral CC chemokine inhibitor; also called 35K), which tightly binds to CC chemokines. To facilitate the study of vCCI, we first provide a protocol to produce folded vCCI from Escherichia coli (E. coli.) It is shown here that vCCI binds with unusually high affinity to viral Macrophage Inflammatory Protein-II (vMIP-II), a chemokine analog produced by the virus, human herpesvirus 8 (HHV-8). Fluorescence anisotropy was used to investigate the vCCI:vMIP-II complex and shows that vCCI binds to vMIP-II with a higher affinity than most other chemokines, having a Kd of 0.06 ± 0.006 nM. Nuclear magnetic resonance (NMR) chemical shift perturbation experiments indicate that key amino acids used for binding in the complex are similar to those found in previous work. Molecular dynamics were then used to compare the vCCI:vMIP-II complex with the known vCCI:Macrophage Inflammatory Protein-1ß/CC-Chemokine Ligand 4 (MIP-1ß/CCL4) complex. The simulations show key interactions, such as those between E143 and D75 in vCCI/35K and R18 in vMIP-II. Further, in a comparison of 1 µs molecular dynamics (MD) trajectories, vMIP-II shows more overall surface binding to vCCI than does the chemokine MIP-1ß. vMIP-II maintains unique contacts at its N-terminus to vCCI that are not made by MIP-1ß, and vMIP-II also makes more contacts with the vCCI flexible acidic loop (located between the second and third beta strands) than does MIP-1ß. These studies provide evidence for the basis of the tight vCCI:vMIP-II interaction while elucidating the vCCI:MIP-1ß interaction, and allow insight into the structure of proteins that are capable of broadly subverting the mammalian immune system.


Subject(s)
Chemokine CXCL2/chemistry , Fluorescence Polarization , Herpesvirus 8, Human/chemistry , Molecular Dynamics Simulation , Multiprotein Complexes/chemistry , Vaccinia virus/chemistry , Viral Proteins/chemistry , Chemokine CXCL2/genetics , Herpesvirus 8, Human/genetics , Multiprotein Complexes/genetics , Protein Structure, Quaternary , Vaccinia virus/genetics , Viral Proteins/genetics
18.
Biologicals ; 49: 39-45, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28754514

ABSTRACT

In the present investigation, the thermostability of a live attenuated buffalopox vaccine prepared with an indigenous baffalopox virus isolate (BPXV Vij/96) and freeze-dried under conventional lyophilizing conditions is described. Three different stabilizer combinations like LS (lactalbumin hydralysate + sucrose), LHT (lactalbumin hydralysate + Trehalose dihydrate) and TAA (Trehalose dihydrate + l- Alanine + l-Histidine) were used to prepare the vaccine. The study indicated that the LS stabilizer was found to be the stabilizer of choice followed by LHT and TAA for buffalopox vaccine at all temperatures studied. The presence of stabilizers has beneficial influence in preserving the keeping quality of the vaccine. Further, among the diluents used to reconstitute the freeze-dried buffalopox vaccine, double distilled water, 0.85% normal saline solution and phosphate buffer saline were the choice of diluents in that order. However, 1M MgSO4 did not perform well at higher temperatures. Investigation suggests for using LS as a stabilizer for freeze-drying and any of the three diluents except 1MgSO4 for reconstitution of buffalopox vaccine.


Subject(s)
Excipients/chemistry , Vaccinia virus/chemistry , Viral Vaccines/chemistry , Animals , Chlorocebus aethiops , Freeze Drying , Vero Cells
19.
J Immunol ; 198(10): 3835-3845, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28363906

ABSTRACT

CD8+ T cell immunosurveillance is based on recognizing oligopeptides presented by MHC class I molecules. Despite decades of study, the importance of protein ubiquitylation to peptide generation remains uncertain. In this study, we examined the ability of MLN7243, a recently described ubiquitin-activating enzyme E1 inhibitor, to block overall cytosolic peptide generation and generation of specific peptides from vaccinia- and influenza A virus-encoded proteins. We show that MLN7243 rapidly inhibits ubiquitylation in a variety of cell lines and can profoundly reduce the generation of cytosolic peptides. Kinetic analysis of specific peptide generation reveals that ubiquitylation of defective ribosomal products is rate limiting in generating class I peptide complexes. More generally, our findings demonstrate that the requirement for ubiquitylation in MHC class I-restricted Ag processing varies with class I allomorph, cell type, source protein, and peptide context. Thus, ubiquitin-dependent and -independent pathways robustly contribute to MHC class I-based immunosurveillance.


Subject(s)
Antigen Presentation , Histocompatibility Antigens Class I/immunology , Nucleosides/pharmacology , Peptides/immunology , Sulfonamides/pharmacology , T-Lymphocytes/immunology , Animals , Cell Line , Cytosol/chemistry , Cytosol/immunology , Enzyme Inhibitors/pharmacology , Histocompatibility Antigens Class I/metabolism , Humans , Influenza A virus/chemistry , Influenza A virus/immunology , Kinetics , Ligands , Mice , Monitoring, Immunologic , Peptides/metabolism , Pyrazoles , Pyrimidines , Sulfides , Ubiquitination , Vaccinia virus/chemistry , Vaccinia virus/immunology
20.
Subcell Biochem ; 83: 103-126, 2017.
Article in English | MEDLINE | ID: mdl-28271474

ABSTRACT

Vaccinia Virus (VACV) is an enveloped double stranded DNA virus and the active ingredient of the smallpox vaccine. The systematic administration of this vaccine led to the eradication of circulating smallpox (variola virus, VARV) from the human population. As a tribute to its success, global immunization was ended in the late 1970s. The efficacy of the vaccine is attributed to a robust production of protective antibodies against several envelope proteins of VACV, which cross-protect against infection with pathogenic VARV. Since global vaccination was ended, most children and young adults do not possess immunity against smallpox. This is a concern, since smallpox is considered a potential bioweapon. Although the smallpox vaccine is considered the gold standard of all vaccines and the targeted antigens have been widely studied, the epitopes that are targeted by the protective antibodies and their mechanism of binding had been, until recently, poorly characterized. Understanding the precise interaction between the antibodies and their epitopes will be helpful in the design of better vaccines against other diseases. In this review we will discuss the structural basis of recognition of the immunodominant VACV antigens A27, A33, D8, and L1 by protective antibodies and discuss potential implications regarding their protective capacity.


Subject(s)
Antibodies, Viral/immunology , Immunodominant Epitopes/immunology , Vaccinia virus/immunology , Viral Envelope Proteins/immunology , Humans , Immunodominant Epitopes/chemistry , Smallpox Vaccine/immunology , Vaccinia virus/chemistry , Viral Envelope Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...