Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Cell ; 171(7): 1692-1706.e18, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29153837

ABSTRACT

Methods for the targeted disruption of protein function have revolutionized science and greatly expedited the systematic characterization of genes. Two main approaches are currently used to disrupt protein function: DNA knockout and RNA interference, which act at the genome and mRNA level, respectively. A method that directly alters endogenous protein levels is currently not available. Here, we present Trim-Away, a technique to degrade endogenous proteins acutely in mammalian cells without prior modification of the genome or mRNA. Trim-Away harnesses the cellular protein degradation machinery to remove unmodified native proteins within minutes of application. This rapidity minimizes the risk that phenotypes are compensated and that secondary, non-specific defects accumulate over time. Because Trim-Away utilizes antibodies, it can be applied to a wide range of target proteins using off-the-shelf reagents. Trim-Away allows the study of protein function in diverse cell types, including non-dividing primary cells where genome- and RNA-targeting methods are limited.


Subject(s)
Antibodies/chemistry , Biochemistry/methods , Protein Transport , Proteolysis , Animals
2.
Nature ; 603(7902): 706-714, 2022 03.
Article in English | MEDLINE | ID: mdl-35104837

ABSTRACT

The SARS-CoV-2 Omicron BA.1 variant emerged in 20211 and has multiple mutations in its spike protein2. Here we show that the spike protein of Omicron has a higher affinity for ACE2 compared with Delta, and a marked change in its antigenicity increases Omicron's evasion of therapeutic monoclonal and vaccine-elicited polyclonal neutralizing antibodies after two doses. mRNA vaccination as a third vaccine dose rescues and broadens neutralization. Importantly, the antiviral drugs remdesivir and molnupiravir retain efficacy against Omicron BA.1. Replication was similar for Omicron and Delta virus isolates in human nasal epithelial cultures. However, in lung cells and gut cells, Omicron demonstrated lower replication. Omicron spike protein was less efficiently cleaved compared with Delta. The differences in replication were mapped to the entry efficiency of the virus on the basis of spike-pseudotyped virus assays. The defect in entry of Omicron pseudotyped virus to specific cell types effectively correlated with higher cellular RNA expression of TMPRSS2, and deletion of TMPRSS2 affected Delta entry to a greater extent than Omicron. Furthermore, drug inhibitors targeting specific entry pathways3 demonstrated that the Omicron spike inefficiently uses the cellular protease TMPRSS2, which promotes cell entry through plasma membrane fusion, with greater dependency on cell entry through the endocytic pathway. Consistent with suboptimal S1/S2 cleavage and inability to use TMPRSS2, syncytium formation by the Omicron spike was substantially impaired compared with the Delta spike. The less efficient spike cleavage of Omicron at S1/S2 is associated with a shift in cellular tropism away from TMPRSS2-expressing cells, with implications for altered pathogenesis.


Subject(s)
COVID-19/pathology , COVID-19/virology , Membrane Fusion , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Serine Endopeptidases/metabolism , Virus Internalization , Adult , Aged , Aged, 80 and over , Angiotensin-Converting Enzyme 2/metabolism , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19 Vaccines/immunology , Cell Line , Cell Membrane/metabolism , Cell Membrane/virology , Chlorocebus aethiops , Convalescence , Female , Humans , Immune Sera/immunology , Intestines/pathology , Intestines/virology , Lung/pathology , Lung/virology , Male , Middle Aged , Mutation , Nasal Mucosa/pathology , Nasal Mucosa/virology , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Tissue Culture Techniques , Virulence , Virus Replication
3.
Nature ; 599(7883): 114-119, 2021 11.
Article in English | MEDLINE | ID: mdl-34488225

ABSTRACT

The B.1.617.2 (Delta) variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was first identified in the state of Maharashtra in late 2020 and spread throughout India, outcompeting pre-existing lineages including B.1.617.1 (Kappa) and B.1.1.7 (Alpha)1. In vitro, B.1.617.2 is sixfold less sensitive to serum neutralizing antibodies from recovered individuals, and eightfold less sensitive to vaccine-elicited antibodies, compared with wild-type Wuhan-1 bearing D614G. Serum neutralizing titres against B.1.617.2 were lower in ChAdOx1 vaccinees than in BNT162b2 vaccinees. B.1.617.2 spike pseudotyped viruses exhibited compromised sensitivity to monoclonal antibodies to the receptor-binding domain and the amino-terminal domain. B.1.617.2 demonstrated higher replication efficiency than B.1.1.7 in both airway organoid and human airway epithelial systems, associated with B.1.617.2 spike being in a predominantly cleaved state compared with B.1.1.7 spike. The B.1.617.2 spike protein was able to mediate highly efficient syncytium formation that was less sensitive to inhibition by neutralizing antibody, compared with that of wild-type spike. We also observed that B.1.617.2 had higher replication and spike-mediated entry than B.1.617.1, potentially explaining the B.1.617.2 dominance. In an analysis of more than 130 SARS-CoV-2-infected health care workers across three centres in India during a period of mixed lineage circulation, we observed reduced ChAdOx1 vaccine effectiveness against B.1.617.2 relative to non-B.1.617.2, with the caveat of possible residual confounding. Compromised vaccine efficacy against the highly fit and immune-evasive B.1.617.2 Delta variant warrants continued infection control measures in the post-vaccination era.


Subject(s)
Immune Evasion , SARS-CoV-2/growth & development , SARS-CoV-2/immunology , Virus Replication/immunology , Antibodies, Neutralizing/immunology , COVID-19 Vaccines/immunology , Cell Fusion , Cell Line , Female , Health Personnel , Humans , India , Kinetics , Male , Spike Glycoprotein, Coronavirus/metabolism , Vaccination
4.
Proc Natl Acad Sci U S A ; 120(16): e2220557120, 2023 04 18.
Article in English | MEDLINE | ID: mdl-37040417

ABSTRACT

The mature HIV-1 capsid protects the viral genome and interacts with host proteins to travel from the cell periphery into the nucleus. To achieve this, the capsid protein, CA, constructs conical capsids from a lattice of hexamers and pentamers, and engages in and then relinquishes multiple interactions with cellular proteins in an orchestrated fashion. Cellular host factors including Nup153, CPSF6, and Sec24C engage the same pocket within CA hexamers. How CA assembles pentamers and hexamers of different curvatures, how CA oligomerization states or curvature might modulate host-protein interactions, and how binding of multiple cofactors to a single site is coordinated, all remain to be elucidated. Here, using single-particle cryoEM, we have determined the structure of the mature HIV-1 CA pentamer and hexamer from conical CA-IP6 polyhedra to ~3 Å resolution. We also determined structures of hexamers in the context of multiple lattice curvatures and number of pentamer contacts. Comparison of these structures, bound or not to host protein peptides, revealed two structural switches within HIV-1 CA that modulate peptide binding according to CA lattice curvature and whether CA is hexameric or pentameric. These observations suggest that the conical HIV-1 capsid has different host-protein binding properties at different positions on its surface, which may facilitate cell entry and represent an evolutionary advantage of conical morphology.


Subject(s)
Capsid , HIV-1 , Capsid/metabolism , Capsid Proteins/chemistry , HIV-1/genetics , Protein Binding , Cytoplasm/metabolism
5.
EMBO J ; 40(17): e108588, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34323299

ABSTRACT

The humoral immune response to SARS-CoV-2 results in antibodies against spike (S) and nucleoprotein (N). However, whilst there are widely available neutralization assays for S antibodies, there is no assay for N-antibody activity. Here, we present a simple in vitro method called EDNA (electroporated-antibody-dependent neutralization assay) that provides a quantitative measure of N-antibody activity in unpurified serum from SARS-CoV-2 convalescents. We show that N antibodies neutralize SARS-CoV-2 intracellularly and cell-autonomously but require the cytosolic Fc receptor TRIM21. Using EDNA, we show that low N-antibody titres can be neutralizing, whilst some convalescents possess serum with high titres but weak activity. N-antibody and N-specific T-cell activity correlates within individuals, suggesting N antibodies may protect against SARS-CoV-2 by promoting antigen presentation. This work highlights the potential benefits of N-based vaccines and provides an in vitro assay to allow the antibodies they induce to be tested.


Subject(s)
Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/isolation & purification , COVID-19/blood , SARS-CoV-2/isolation & purification , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19/virology , Humans , Nucleoproteins/blood , Nucleoproteins/immunology , SARS-CoV-2/pathogenicity
6.
EMBO J ; 40(5): e106228, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33258165

ABSTRACT

Nucleoprotein (N) is an immunodominant antigen in many enveloped virus infections. While the diagnostic value of anti-N antibodies is clear, their role in immunity is not. This is because while they are non-neutralising, they somehow clear infection by coronavirus, influenza and LCMV in vivo. Here, we show that anti-N immune protection is mediated by the cytosolic Fc receptor and E3 ubiquitin ligase TRIM21. Exploiting LCMV as a model system, we demonstrate that TRIM21 uses anti-N antibodies to target N for cytosolic degradation and generate cytotoxic T cells (CTLs) against N peptide. These CTLs rapidly eliminate N-peptide-displaying cells and drive efficient viral clearance. These results reveal a new mechanism of immune synergy between antibodies and T cells and highlights N as an important vaccine target.


Subject(s)
Antibodies, Viral/immunology , Immunity, Cellular , Lymphocytic choriomeningitis virus/immunology , Nucleocapsid Proteins/immunology , Ribonucleoproteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/genetics , Mice , Mice, Knockout , Nucleocapsid Proteins/genetics , Ribonucleoproteins/genetics , Viral Vaccines/genetics , Viral Vaccines/immunology
7.
Nat Immunol ; 14(4): 327-36, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23455675

ABSTRACT

During pathogen infection, antibodies can be carried into the infected cell, where they are detected by the ubiquitously expressed cytosolic antibody receptor TRIM21. Here we found that recognition of intracellular antibodies by TRIM21 activated immune signaling. TRIM21 catalyzed the formation of Lys63 (K63)-linked ubiquitin chains and stimulated the transcription factor pathways of NF-κB, AP-1, IRF3, IRF5 and IRF7. Activation resulted in the production of proinflammatory cytokines, modulation of natural killer stress ligands and induction of an antiviral state. Intracellular antibody signaling was abrogated by genetic deletion of TRIM21 and was restored by ectopic expression of TRIM21. The sensing of antibodies by TRIM21 was stimulated after infection by DNA or RNA nonenveloped viruses or intracellular bacteria. Thus, the antibody-TRIM21 detection system provides potent, comprehensive activation of the innate immune system independently of known pattern-recognition receptors.


Subject(s)
Antibodies, Viral/immunology , Intracellular Space/immunology , Intracellular Space/metabolism , Receptors, Fc/metabolism , Ribonucleoproteins/immunology , Signal Transduction , Viruses/immunology , Adenoviridae/immunology , Animals , Antibodies, Viral/chemistry , Antibodies, Viral/metabolism , Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/metabolism , Bacteria/immunology , Cell Line , Cross Reactions , Cytokines/biosynthesis , Humans , Inflammation Mediators/metabolism , Interferon Regulatory Factors/metabolism , Mice , Molecular Docking Simulation , NF-kappa B/metabolism , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Receptors, Pattern Recognition/metabolism , Ribonucleoproteins/chemistry , Ribonucleoproteins/metabolism , Transcription Factor AP-1/metabolism
8.
EMBO Rep ; 24(5): e56275, 2023 05 04.
Article in English | MEDLINE | ID: mdl-36970882

ABSTRACT

HIV-1 uses inositol hexakisphosphate (IP6) to build a metastable capsid capable of delivering its genome into the host nucleus. Here, we show that viruses that are unable to package IP6 lack capsid protection and are detected by innate immunity, resulting in the activation of an antiviral state that inhibits infection. Disrupting IP6 enrichment results in defective capsids that trigger cytokine and chemokine responses during infection of both primary macrophages and T-cell lines. Restoring IP6 enrichment with a single mutation rescues the ability of HIV-1 to infect cells without being detected. Using a combination of capsid mutants and CRISPR-derived knockout cell lines for RNA and DNA sensors, we show that immune sensing is dependent upon the cGAS-STING axis and independent of capsid detection. Sensing requires the synthesis of viral DNA and is prevented by reverse transcriptase inhibitors or reverse transcriptase active-site mutation. These results demonstrate that IP6 is required to build capsids that can successfully transit the cell and avoid host innate immune sensing.


Subject(s)
Capsid , HIV Infections , Humans , Capsid/metabolism , Host-Pathogen Interactions , Immunity, Innate , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism
9.
Cell ; 141(7): 1220-9, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20603002

ABSTRACT

The AP2 adaptor complex (alpha, beta2, sigma2, and mu2 subunits) crosslinks the endocytic clathrin scaffold to PtdIns4,5P(2)-containing membranes and transmembrane protein cargo. In the "locked" cytosolic form, AP2's binding sites for the two endocytic motifs, YxxPhi on the C-terminal domain of mu2 (C-mu2) and [ED]xxxL[LI] on sigma2, are blocked by parts of beta2. Using protein crystallography, we show that AP2 undergoes a large conformational change in which C-mu2 relocates to an orthogonal face of the complex, simultaneously unblocking both cargo-binding sites; the previously unstructured mu2 linker becomes helical and binds back onto the complex. This structural rearrangement results in AP2's four PtdIns4,5P(2)- and two endocytic motif-binding sites becoming coplanar, facilitating their simultaneous interaction with PtdIns4,5P(2)/cargo-containing membranes. Using a range of biophysical techniques, we show that the endocytic cargo binding of AP2 is driven by its interaction with PtdIns4,5P(2)-containing membranes.


Subject(s)
Adaptor Protein Complex 2/chemistry , Binding Sites , Cell Membrane/chemistry , Ligands , Models, Molecular , Phosphatidylinositols/chemistry , Protein Conformation
10.
Semin Cell Dev Biol ; 126: 99-107, 2022 06.
Article in English | MEDLINE | ID: mdl-34823983

ABSTRACT

The cytosolic antibody receptor and RING E3 ligase TRIM21 targets intracellular, antibody-coated immune complexes for degradation and activates the immune system. Here we review how TRIM21 degrades diverse targets and how this activity can be exploited in molecular biology and for the development of new therapeutics. In addition, we compare what is known about TRIM21's mechanism to other TRIM proteins and RING E3 ligases.


Subject(s)
Ribonucleoproteins , Ubiquitin-Protein Ligases , Antibodies , Cytosol/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
11.
Alzheimers Dement ; 20(2): 1013-1025, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37849026

ABSTRACT

INTRODUCTION: Signatures of a type-I interferon (IFN-I) response are observed in the post mortem brain in Alzheimer's disease (AD) and other tauopathies. However, the effect of the IFN-I response on pathological tau accumulation remains unclear. METHODS: We examined the effects of IFN-I signaling in primary neural culture models of seeded tau aggregation and P301S-tau transgenic mouse models in the context of genetic deletion of the IFN-I receptor (IFNAR). RESULTS: Polyinosinic:polycytidylic acid (PolyI:C), a synthetic analog of viral nucleic acids, evoked a potent cytokine response that enhanced seeded aggregation of tau in an IFN-I-dependent manner. IFN-I-induced vulnerability could be pharmacologically prevented and was intrinsic to neurons. Aged P301S-tau mice lacking Ifnar1 had significantly reduced tau pathology compared to mice with intact IFN signaling. DISCUSSION: We identify a critical role for IFN-I in potentiating tau aggregation. IFN-I is therefore identified as a potential therapeutic target in AD and other tauopathies. HIGHLIGHTS: Type-I IFN (IFN-I) promotes seeded tau aggregation in neural cultures. IFNAR inhibition prevents IFN-I driven sensitivity to tau aggregation. IFN-I driven vulnerability is intrinsic to neurons. Tau pathology is significantly reduced in aged P301S-tau mice lacking IFNAR.


Subject(s)
Alzheimer Disease , Interferon Type I , Tauopathies , Mice , Animals , tau Proteins/genetics , Interferon Type I/therapeutic use , Tauopathies/pathology , Mice, Transgenic , Alzheimer Disease/pathology , Disease Models, Animal
12.
EMBO J ; 38(21): e101365, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31468569

ABSTRACT

Inflammasomes are potent innate immune signalling complexes that couple cytokine release with pro-inflammatory cell death. However, pathogens have evolved strategies to evade this cell autonomous system. Here, we show how antibodies combine with innate sensors in primary human macrophages to detect viral infection and activate the inflammasome. Our data demonstrate that antibody opsonisation of virions can activate macrophages in multiple ways. In the first, antibody binding of adenovirus causes lysosomal damage, activating NLRP3 to drive inflammasome formation and IL-1ß release. Importantly, this mechanism enhances virion capture but not infection and is accompanied by cell death, denying the opportunity for viral replication. Unexpectedly, we also find that antibody-coated viruses, which successfully escape into the cytosol, trigger a second system of inflammasome activation. These viruses are intercepted by the cytosolic antibody receptor TRIM21 and the DNA sensor cGAS. Together, these sensors stimulate both NLRP3 inflammasome formation and NFκB activation, driving dose-dependent IL-1ß and TNF secretion, without inducing cell death. Our data highlight the importance of cooperativity between multiple sensing networks to expose viruses to the inflammasome pathway, which is particularly important for how our innate immune system responds to infection in the presence of pre-existing immunity.


Subject(s)
Adenoviridae Infections/immunology , Antibodies, Viral/immunology , Inflammasomes/immunology , Macrophages/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nucleotidyltransferases/metabolism , Ribonucleoproteins/metabolism , Virus Replication/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Adenoviridae Infections/metabolism , Adenoviridae Infections/virology , Animals , Cells, Cultured , Humans , Inflammasomes/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Macrophages/metabolism , Macrophages/virology , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Nucleotidyltransferases/genetics , Ribonucleoproteins/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
13.
PLoS Pathog ; 17(2): e1009164, 2021 02.
Article in English | MEDLINE | ID: mdl-33524070

ABSTRACT

The HIV capsid self-assembles a protective conical shell that simultaneously prevents host sensing whilst permitting the import of nucleotides to drive DNA synthesis. This is accomplished through the construction of dynamic, highly charged pores at the centre of each capsid multimer. The clustering of charges required for dNTP import is strongly destabilising and it is proposed that HIV uses the metabolite IP6 to coordinate the pore during assembly. Here we have investigated the role of inositol phosphates in coordinating a ring of positively charged lysine residues (K25) that forms at the base of the capsid pore. We show that whilst IP5, which can functionally replace IP6, engages an arginine ring (R18) at the top of the pore, the lysine ring simultaneously binds a second IP5 molecule. Dose dependent removal of K25 from the pore severely inhibits HIV infection and concomitantly prevents DNA synthesis. Cryo-tomography reveals that K25A virions have a severe assembly defect that inhibits the formation of mature capsid cones. Monitoring both the kinetics and morphology of capsids assembled in vitro reveals that while mutation K25A can still form tubes, the ability of IP6 to drive assembly of capsid cones has been lost. Finally, in single molecule TIRF microscopy experiments, capsid lattices in permeabilised K25 mutant virions are rapidly lost and cannot be stabilised by IP6. These results suggest that the coordination of IP6 by a second charged ring in mature hexamers drives the assembly of conical capsids capable of reverse transcription and infection.


Subject(s)
Capsid/metabolism , HIV-1/physiology , Lysine/metabolism , Phytic Acid/metabolism , Virus Assembly/physiology , Cell Line , DNA, Viral/biosynthesis , HIV-1/genetics , HIV-1/metabolism , Humans , Microscopy, Fluorescence , Nucleotides/metabolism
14.
PLoS Pathog ; 17(1): e1009246, 2021 01.
Article in English | MEDLINE | ID: mdl-33493182

ABSTRACT

Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) infects cells by binding to the host cell receptor ACE2 and undergoing virus-host membrane fusion. Fusion is triggered by the protease TMPRSS2, which processes the viral Spike (S) protein to reveal the fusion peptide. SARS-CoV-2 has evolved a multibasic site at the S1-S2 boundary, which is thought to be cleaved by furin in order to prime S protein for TMPRSS2 processing. Here we show that CRISPR-Cas9 knockout of furin reduces, but does not prevent, the production of infectious SARS-CoV-2 virus. Comparing S processing in furin knockout cells to multibasic site mutants reveals that while loss of furin substantially reduces S1-S2 cleavage it does not prevent it. SARS-CoV-2 S protein also mediates cell-cell fusion, potentially allowing virus to spread virion-independently. We show that loss of furin in either donor or acceptor cells reduces, but does not prevent, TMPRSS2-dependent cell-cell fusion, unlike mutation of the multibasic site that completely prevents syncytia formation. Our results show that while furin promotes both SARS-CoV-2 infectivity and cell-cell spread it is not essential, suggesting furin inhibitors may reduce but not abolish viral spread.


Subject(s)
Cell Fusion , Furin/genetics , Spike Glycoprotein, Coronavirus/chemistry , Virus Internalization , Animals , COVID-19 , CRISPR-Cas Systems , Chlorocebus aethiops , Gene Knockout Techniques , HEK293 Cells , Humans , Protein Structure, Tertiary , SARS-CoV-2 , Serine Endopeptidases , Vero Cells
15.
J Virol ; 95(15): e0020321, 2021 07 12.
Article in English | MEDLINE | ID: mdl-33963055

ABSTRACT

The majority of SARS-CoV-2 vaccines in use or advanced development are based on the viral spike protein (S) as their immunogen. S is present on virions as prefusion trimers in which the receptor binding domain (RBD) is stochastically open or closed. Neutralizing antibodies have been described against both open and closed conformations. The long-term success of vaccination strategies depends upon inducing antibodies that provide long-lasting broad immunity against evolving SARS-CoV-2 strains. Here, we have assessed the results of immunization in a mouse model using an S protein trimer stabilized in the closed state to prevent full exposure of the receptor binding site and therefore interaction with the receptor. We compared this with other modified S protein constructs, including representatives used in current vaccines. We found that all trimeric S proteins induced a T cell response and long-lived, strongly neutralizing antibody responses against 2019 SARS-CoV-2 and variants of concern P.1 and B.1.351. Notably, the protein binding properties of sera induced by the closed spike differed from those induced by standard S protein constructs. Closed S proteins induced more potent neutralizing responses than expected based on the degree to which they inhibit interactions between the RBD and ACE2. These observations suggest that closed spikes recruit different, but equally potent, immune responses than open spikes and that this is likely to include neutralizing antibodies against conformational epitopes present in the closed conformation. We suggest that closed spikes, together with their improved stability and storage properties, may be a valuable component of refined, next-generation vaccines. IMPORTANCE Vaccines in use against SARS-CoV-2 induce immune responses against the spike protein. There is intense interest in whether the antibody response induced by vaccines will be robust against new variants, as well as in next-generation vaccines for use in previously infected or immunized individuals. We assessed the use as an immunogen of a spike protein engineered to be conformationally stabilized in the closed state where the receptor binding site is occluded. Despite occlusion of the receptor binding site, the spike induces potently neutralizing sera against multiple SARS-CoV-2 variants. Antibodies are raised against a different pattern of epitopes to those induced by other spike constructs, preferring conformational epitopes present in the closed conformation. Closed spikes, or mRNA vaccines based on their sequence, can be a valuable component of next-generation vaccines.


Subject(s)
Angiotensin-Converting Enzyme 2 , Antibodies, Neutralizing , Antibodies, Viral , Epitopes , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/immunology , Animals , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , COVID-19 Vaccines/chemistry , COVID-19 Vaccines/immunology , Epitopes/chemistry , Epitopes/immunology , HEK293 Cells , Humans , Mice , Protein Stability , SARS-CoV-2/chemistry , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/immunology
16.
PLoS Pathog ; 16(8): e1008732, 2020 08.
Article in English | MEDLINE | ID: mdl-32750093

ABSTRACT

Rotavirus is a major cause of gastroenteritis in children, with infection typically inducing high levels of protective antibodies. Antibodies targeting the middle capsid protein VP6 are particularly abundant, and as VP6 is only exposed inside cells, neutralisation must be post-entry. However, while a system of poly immune globulin receptor (pIgR) transcytosis has been proposed for anti-VP6 IgAs, the mechanism by which VP6-specific IgG mediates protection remains less clear. We have developed an intracellular neutralisation assay to examine how antibodies neutralise rotavirus inside cells, enabling comparison between IgG and IgA isotypes. Unexpectedly we found that neutralisation by VP6-specific IgG was much more efficient than by VP6-specific IgA. This observation was highly dependent on the activity of the cytosolic antibody receptor TRIM21 and was confirmed using an in vivo model of murine rotavirus infection. Furthermore, mice deficient in only IgG and not other antibody isotypes had a serious deficit in intracellular antibody-mediated protection. The finding that VP6-specific IgG protect mice against rotavirus infection has important implications for rotavirus vaccination. Current assays determine protection in humans predominantly by measuring rotavirus-specific IgA titres. Measurements of VP6-specific IgG may add to existing mechanistic correlates of protection.


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/immunology , Capsid Proteins/immunology , Immunoglobulin G/immunology , Rotavirus Infections/immunology , Rotavirus/physiology , Animals , Antigens, Viral/genetics , Capsid Proteins/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rotavirus/genetics , Rotavirus Infections/virology , Species Specificity
17.
Nature ; 536(7616): 349-53, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27509857

ABSTRACT

During the early stages of infection, the HIV-1 capsid protects viral components from cytosolic sensors and nucleases such as cGAS and TREX, respectively, while allowing access to nucleotides for efficient reverse transcription. Here we show that each capsid hexamer has a size-selective pore bound by a ring of six arginine residues and a 'molecular iris' formed by the amino-terminal ß-hairpin. The arginine ring creates a strongly positively charged channel that recruits the four nucleotides with on-rates that approach diffusion limits. Progressive removal of pore arginines results in a dose-dependent and concomitant decrease in nucleotide affinity, reverse transcription and infectivity. This positively charged channel is universally conserved in lentiviral capsids despite the fact that it is strongly destabilizing without nucleotides to counteract charge repulsion. We also describe a channel inhibitor, hexacarboxybenzene, which competes for nucleotide binding and efficiently blocks encapsidated reverse transcription, demonstrating the tractability of the pore as a novel drug target.


Subject(s)
Capsid/metabolism , DNA Replication , DNA, Viral/biosynthesis , HIV-1/metabolism , Nucleotides/metabolism , Arginine/metabolism , Benzoates/pharmacology , Binding, Competitive/drug effects , Biological Transport, Active/drug effects , Capsid/chemistry , Capsid/drug effects , DNA Replication/drug effects , Diffusion , HEK293 Cells , HIV-1/drug effects , HIV-1/genetics , HIV-1/growth & development , HeLa Cells , Humans , Kinetics , Models, Molecular , Porosity/drug effects , Reverse Transcription/drug effects
18.
Proc Natl Acad Sci U S A ; 115(41): 10440-10445, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30209217

ABSTRACT

Adenovirus has enormous potential as a gene-therapy vector, but preexisting immunity limits its widespread application. What is responsible for this immune block is unclear because antibodies potently inhibit transgene expression without impeding gene transfer into target cells. Here we show that antibody prevention of adenoviral gene delivery in vivo is mediated by the cytosolic antibody receptor TRIM21. Genetic KO of TRIM21 or a single-antibody point mutation is sufficient to restore transgene expression to near-naïve immune levels. TRIM21 is also responsible for blocking cytotoxic T cell induction by vaccine vectors, preventing a protective response against subsequent influenza infection and an engrafted tumor. Furthermore, adenoviral preexisting immunity can lead to an augmented immune response upon i.v. administration of the vector. Transcriptomic analysis of vector-transduced tissue reveals that TRIM21 is responsible for the specific up-regulation of hundreds of immune genes, the majority of which are components of the intrinsic or innate response. Together, these data define a major mechanism underlying the preimmune block to adenovirus gene therapy and demonstrate that TRIM21 efficiently blocks gene delivery in vivo while simultaneously inducing a rapid program of immune transcription.


Subject(s)
Adenoviridae Infections/therapy , Adenoviridae/immunology , Antibodies/immunology , Fibrosarcoma/therapy , Genetic Therapy , Ribonucleoproteins/physiology , Vaccination , Adenoviridae Infections/genetics , Adenoviridae Infections/immunology , Animals , Fibrosarcoma/genetics , Fibrosarcoma/immunology , Gene Transfer Techniques , Genetic Vectors , Mice , Mice, Inbred C57BL , Mice, Knockout , Transgenes , Tumor Cells, Cultured
19.
Proc Natl Acad Sci U S A ; 114(3): 574-579, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28049840

ABSTRACT

Alzheimer's disease (AD) and other neurodegenerative disorders are associated with the cytoplasmic aggregation of microtubule-associated protein tau. Recent evidence supports transcellular transfer of tau misfolding (seeding) as the mechanism of spread within an affected brain, a process reminiscent of viral infection. However, whereas microbial pathogens can be recognized as nonself by immune receptors, misfolded protein assemblies evade detection, as they are host-derived. Here, we show that when misfolded tau assemblies enter the cell, they can be detected and neutralized via a danger response mediated by tau-associated antibodies and the cytosolic Fc receptor tripartite motif protein 21 (TRIM21). We developed fluorescent, morphology-based seeding assays that allow the formation of pathological tau aggregates to be measured in situ within 24 h in the presence of picomolar concentrations of tau seeds. We found that anti-tau antibodies accompany tau seeds into the cell, where they recruit TRIM21 shortly after entry. After binding, TRIM21 neutralizes tau seeds through the activity of the proteasome and the AAA ATPase p97/VCP in a similar manner to infectious viruses. These results establish that intracellular antiviral immunity can be redirected against host-origin endopathogens involved in neurodegeneration.


Subject(s)
Receptors, Fc/metabolism , Ribonucleoproteins/metabolism , tau Proteins/metabolism , Animals , Antibodies, Neutralizing/metabolism , Cells, Cultured , Cytosol/metabolism , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/immunology , Nerve Degeneration/metabolism , Nerve Degeneration/prevention & control , Neurons/immunology , Neurons/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Aggregates , Protein Aggregation, Pathological/immunology , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/prevention & control , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/prevention & control , Receptors, Fc/deficiency , Receptors, Fc/genetics , Ribonucleoproteins/deficiency , Ribonucleoproteins/genetics , tau Proteins/chemistry , tau Proteins/immunology
20.
PLoS Pathog ; 13(12): e1006758, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29261807

ABSTRACT

RNA-dependent RNA polymerases (RdRps) play a key role in the life cycle of RNA viruses and impact their immunobiology. The arenavirus lymphocytic choriomeningitis virus (LCMV) strain Clone 13 provides a benchmark model for studying chronic infection. A major genetic determinant for its ability to persist maps to a single amino acid exchange in the viral L protein, which exhibits RdRp activity, yet its functional consequences remain elusive. To unravel the L protein interactions with the host proteome, we engineered infectious L protein-tagged LCMV virions by reverse genetics. A subsequent mass-spectrometric analysis of L protein pulldowns from infected human cells revealed a comprehensive network of interacting host proteins. The obtained LCMV L protein interactome was bioinformatically integrated with known host protein interactors of RdRps from other RNA viruses, emphasizing interconnected modules of human proteins. Functional characterization of selected interactors highlighted proviral (DDX3X) as well as antiviral (NKRF, TRIM21) host factors. To corroborate these findings, we infected Trim21-/- mice with LCMV and found impaired virus control in chronic infection. These results provide insights into the complex interactions of the arenavirus LCMV and other viral RdRps with the host proteome and contribute to a better molecular understanding of how chronic viruses interact with their host.


Subject(s)
DEAD-box RNA Helicases/metabolism , Lymphocytic choriomeningitis virus/enzymology , Models, Molecular , RNA-Dependent RNA Polymerase/metabolism , Repressor Proteins/metabolism , Ribonucleoproteins/metabolism , Viral Proteins/metabolism , Animals , CRISPR-Cas Systems , Computational Biology , Crosses, Genetic , DEAD-box RNA Helicases/chemistry , Female , HEK293 Cells , Humans , Immunoprecipitation , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/veterinary , Male , Mice, Inbred C57BL , Mice, Knockout , Protein Interaction Domains and Motifs , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Repressor Proteins/chemistry , Ribonucleoproteins/chemistry , Ribonucleoproteins/genetics , Specific Pathogen-Free Organisms , Viral Proteins/chemistry , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL