Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89.334
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 38: 673-703, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32340576

ABSTRACT

Development of improved approaches for HIV-1 prevention will likely be required for a durable end to the global AIDS pandemic. Recent advances in preclinical studies and early phase clinical trials offer renewed promise for immunologic strategies for blocking acquisition of HIV-1 infection. Clinical trials are currently underway to evaluate the efficacy of two vaccine candidates and a broadly neutralizing antibody (bNAb) to prevent HIV-1 infection in humans. However, the vast diversity of HIV-1 is a major challenge for both active and passive immunization. Here we review current immunologic strategies for HIV-1 prevention, with a focus on current and next-generation vaccines and bNAbs.


Subject(s)
AIDS Vaccines/immunology , Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/immunology , Host-Pathogen Interactions/immunology , AIDS Vaccines/administration & dosage , Animals , Clinical Trials as Topic , Disease Management , Genetic Variation , HIV Infections/virology , HIV-1/genetics , Humans , Immunization, Passive , RNA, Viral , Structure-Activity Relationship , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics
2.
Annu Rev Immunol ; 36: 667-694, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29677479

ABSTRACT

Pattern recognition receptors (PRRs) survey intra- and extracellular spaces for pathogen-associated molecular patterns (PAMPs) within microbial products of infection. Recognition and binding to cognate PAMP ligand by specific PRRs initiates signaling cascades that culminate in a coordinated intracellular innate immune response designed to control infection. In particular, our immune system has evolved specialized PRRs to discriminate viral nucleic acid from host. These are critical sensors of viral RNA to trigger innate immunity in the vertebrate host. Different families of PRRs of virus infection have been defined and reveal a diversity of PAMP specificity for wide viral pathogen coverage to recognize and extinguish virus infection. In this review, we discuss recent insights in pathogen recognition by the RIG-I-like receptors, related RNA helicases, Toll-like receptors, and other RNA sensor PRRs, to present emerging themes in innate immune signaling during virus infection.


Subject(s)
DEAD Box Protein 58/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Virus Diseases/etiology , Virus Diseases/metabolism , Viruses/immunology , Animals , DEAD-box RNA Helicases/metabolism , Humans , Protein Processing, Post-Translational , RNA Helicases/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Receptors, Immunologic , Signal Transduction , Toll-Like Receptors/metabolism
3.
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
4.
Cell ; 187(11): 2735-2745.e12, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38723628

ABSTRACT

Hepatitis B virus (HBV) is a small double-stranded DNA virus that chronically infects 296 million people. Over half of its compact genome encodes proteins in two overlapping reading frames, and during evolution, multiple selective pressures can act on shared nucleotides. This study combines an RNA-based HBV cell culture system with deep mutational scanning (DMS) to uncouple cis- and trans-acting sequence requirements in the HBV genome. The results support a leaky ribosome scanning model for polymerase translation, provide a fitness map of the HBV polymerase at single-nucleotide resolution, and identify conserved prolines adjacent to the HBV polymerase termination codon that stall ribosomes. Further experiments indicated that stalled ribosomes tether the nascent polymerase to its template RNA, ensuring cis-preferential RNA packaging and reverse transcription of the HBV genome.


Subject(s)
Hepatitis B virus , Reverse Transcription , Humans , Genome, Viral/genetics , Hepatitis B virus/genetics , Mutation , Ribosomes/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Cell Line
5.
Cell ; 187(9): 2236-2249.e17, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38614100

ABSTRACT

Unlike those of double-stranded DNA (dsDNA), single-stranded DNA (ssDNA), and ssRNA viruses, the mechanism of genome packaging of dsRNA viruses is poorly understood. Here, we combined the techniques of high-resolution cryoelectron microscopy (cryo-EM), cellular cryoelectron tomography (cryo-ET), and structure-guided mutagenesis to investigate genome packaging and capsid assembly of bluetongue virus (BTV), a member of the Reoviridae family of dsRNA viruses. A total of eleven assembly states of BTV capsid were captured, with resolutions up to 2.8 Å, with most visualized in the host cytoplasm. ATPase VP6 was found underneath the vertices of capsid shell protein VP3 as an RNA-harboring pentamer, facilitating RNA packaging. RNA packaging expands the VP3 shell, which then engages middle- and outer-layer proteins to generate infectious virions. These revealed "duality" characteristics of the BTV assembly mechanism reconcile previous contradictory co-assembly and core-filling models and provide insights into the mysterious RNA packaging and capsid assembly of Reoviridae members and beyond.


Subject(s)
Bluetongue virus , Capsid Proteins , Capsid , Cryoelectron Microscopy , RNA, Viral , Viral Genome Packaging , Bluetongue virus/genetics , Bluetongue virus/physiology , Bluetongue virus/metabolism , Capsid/metabolism , Capsid/ultrastructure , Capsid Proteins/metabolism , Capsid Proteins/genetics , Capsid Proteins/chemistry , Animals , RNA, Viral/metabolism , RNA, Viral/genetics , Genome, Viral/genetics , Virus Assembly , Electron Microscope Tomography , Virion/metabolism , Virion/genetics , Virion/ultrastructure , Models, Molecular , Cell Line , Cricetinae
6.
Cell ; 186(3): 646-661.e4, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36696902

ABSTRACT

Viroids and viroid-like covalently closed circular (ccc) RNAs are minimal replicators that typically encode no proteins and hijack cellular enzymes for replication. The extent and diversity of viroid-like agents are poorly understood. We developed a computational pipeline to identify viroid-like cccRNAs and applied it to 5,131 metatranscriptomes and 1,344 plant transcriptomes. The search yielded 11,378 viroid-like cccRNAs spanning 4,409 species-level clusters, a 5-fold increase compared to the previously identified viroid-like elements. Within this diverse collection, we discovered numerous putative viroids, satellite RNAs, retrozymes, and ribozy-like viruses. Diverse ribozyme combinations and unusual ribozymes within the cccRNAs were identified. Self-cleaving ribozymes were identified in ambiviruses, some mito-like viruses and capsid-encoding satellite virus-like cccRNAs. The broad presence of viroid-like cccRNAs in diverse transcriptomes and ecosystems implies that their host range is far broader than currently known, and matches to CRISPR spacers suggest that some cccRNAs replicate in prokaryotes.


Subject(s)
RNA, Catalytic , Viroids , RNA, Circular/metabolism , Viroids/genetics , Viroids/metabolism , RNA, Catalytic/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , Ecosystem , Plant Diseases
7.
Cell ; 186(15): 3291-3306.e21, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37413987

ABSTRACT

The number of sequenced viral genomes has surged recently, presenting an opportunity to understand viral diversity and uncover unknown regulatory mechanisms. Here, we conducted a screening of 30,367 viral segments from 143 species representing 96 genera and 37 families. Using a library of viral segments in 3' UTR, we identified hundreds of elements impacting RNA abundance, translation, and nucleocytoplasmic distribution. To illustrate the power of this approach, we investigated K5, an element conserved in kobuviruses, and found its potent ability to enhance mRNA stability and translation in various contexts, including adeno-associated viral vectors and synthetic mRNAs. Moreover, we identified a previously uncharacterized protein, ZCCHC2, as a critical host factor for K5. ZCCHC2 recruits the terminal nucleotidyl transferase TENT4 to elongate poly(A) tails with mixed sequences, delaying deadenylation. This study provides a unique resource for virus and RNA research and highlights the potential of the virosphere for biological discoveries.


Subject(s)
RNA , Regulatory Sequences, Nucleic Acid , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Base Sequence , Proteins/genetics , DNA-Directed DNA Polymerase/metabolism , RNA Stability , RNA, Viral/genetics , RNA, Viral/metabolism
8.
Cell ; 186(22): 4834-4850.e23, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37794589

ABSTRACT

Regulation of viral RNA biogenesis is fundamental to productive SARS-CoV-2 infection. To characterize host RNA-binding proteins (RBPs) involved in this process, we biochemically identified proteins bound to genomic and subgenomic SARS-CoV-2 RNAs. We find that the host protein SND1 binds the 5' end of negative-sense viral RNA and is required for SARS-CoV-2 RNA synthesis. SND1-depleted cells form smaller replication organelles and display diminished virus growth kinetics. We discover that NSP9, a viral RBP and direct SND1 interaction partner, is covalently linked to the 5' ends of positive- and negative-sense RNAs produced during infection. These linkages occur at replication-transcription initiation sites, consistent with NSP9 priming viral RNA synthesis. Mechanistically, SND1 remodels NSP9 occupancy and alters the covalent linkage of NSP9 to initiating nucleotides in viral RNA. Our findings implicate NSP9 in the initiation of SARS-CoV-2 RNA synthesis and unravel an unsuspected role of a cellular protein in orchestrating viral RNA production.


Subject(s)
COVID-19 , RNA, Viral , Humans , COVID-19/metabolism , Endonucleases/metabolism , RNA, Viral/metabolism , SARS-CoV-2/genetics , Virus Replication
9.
Annu Rev Biochem ; 91: 381-401, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35729072

ABSTRACT

The persistence of the coronavirus disease 2019 (COVID-19) pandemic has resulted in increasingly disruptive impacts, and it has become the most devastating challenge to global health in a century. The rapid emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants challenges the currently available therapeutics for clinical application. Nonstructural proteins (also known as replicase proteins) with versatile biological functions play central roles in viral replication and transcription inside the host cells, and they are the most conserved target proteins among the SARS-CoV-2 variants. Specifically, they constitute the replication-transcription complexes (RTCs) dominating the synthesis of viral RNA. Knowledge of themolecular mechanisms of nonstructural proteins and their assembly into RTCs will benefit the development of antivirals targeting them against existing or potentially emerging variants. In this review, we summarize current knowledge of the structures and functions of coronavirus nonstructural proteins as well as the assembly and functions of RTCs in the life cycle of the virus.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Humans , RNA, Viral/genetics , Virus Replication
10.
Cell ; 185(23): 4347-4360.e17, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36335936

ABSTRACT

Decoration of cap on viral RNA plays essential roles in SARS-CoV-2 proliferation. Here, we report a mechanism for SARS-CoV-2 RNA capping and document structural details at atomic resolution. The NiRAN domain in polymerase catalyzes the covalent link of RNA 5' end to the first residue of nsp9 (termed as RNAylation), thus being an intermediate to form cap core (GpppA) with GTP catalyzed again by NiRAN. We also reveal that triphosphorylated nucleotide analog inhibitors can be bonded to nsp9 and fit into a previously unknown "Nuc-pocket" in NiRAN, thus inhibiting nsp9 RNAylation and formation of GpppA. S-loop (residues 50-KTN-52) in NiRAN presents a remarkable conformational shift observed in RTC bound with sofosbuvir monophosphate, reasoning an "induce-and-lock" mechanism to design inhibitors. These findings not only improve the understanding of SARS-CoV-2 RNA capping and the mode of action of NAIs but also provide a strategy to design antiviral drugs.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase , Antiviral Agents/chemistry , Nucleotides/chemistry , Viral Nonstructural Proteins/metabolism
11.
Cell ; 185(2): 266-282.e15, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35026153

ABSTRACT

HIV-1-infected cells that persist despite antiretroviral therapy (ART) are frequently considered "transcriptionally silent," but active viral gene expression may occur in some cells, challenging the concept of viral latency. Applying an assay for profiling the transcriptional activity and the chromosomal locations of individual proviruses, we describe a global genomic and epigenetic map of transcriptionally active and silent proviral species and evaluate their longitudinal evolution in persons receiving suppressive ART. Using genome-wide epigenetic reference data, we show that proviral transcriptional activity is associated with activating epigenetic chromatin features in linear proximity of integration sites and in their inter- and intrachromosomal contact regions. Transcriptionally active proviruses were actively selected against during prolonged ART; however, this pattern was violated by large clones of virally infected cells that may outcompete negative selection forces through elevated intrinsic proliferative activity. Our results suggest that transcriptionally active proviruses are dynamically evolving under selection pressure by host factors.


Subject(s)
HIV-1/genetics , Proviruses/genetics , Transcription, Genetic , Aged , Base Sequence , Biological Evolution , Chromatin/metabolism , Clone Cells , DNA, Viral/genetics , Epigenesis, Genetic/drug effects , Female , Humans , Ionomycin/pharmacology , Male , Middle Aged , Phylogeny , Proviruses/drug effects , RNA, Viral/genetics , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic/drug effects , Virus Integration/genetics , Virus Latency/drug effects , Virus Latency/genetics
12.
Cell ; 184(7): 1895-1913.e19, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33657410

ABSTRACT

A dysfunctional immune response in coronavirus disease 2019 (COVID-19) patients is a recurrent theme impacting symptoms and mortality, yet a detailed understanding of pertinent immune cells is not complete. We applied single-cell RNA sequencing to 284 samples from 196 COVID-19 patients and controls and created a comprehensive immune landscape with 1.46 million cells. The large dataset enabled us to identify that different peripheral immune subtype changes are associated with distinct clinical features, including age, sex, severity, and disease stages of COVID-19. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA was found in diverse epithelial and immune cell types, accompanied by dramatic transcriptomic changes within virus-positive cells. Systemic upregulation of S100A8/A9, mainly by megakaryocytes and monocytes in the peripheral blood, may contribute to the cytokine storms frequently observed in severe patients. Our data provide a rich resource for understanding the pathogenesis of and developing effective therapeutic strategies for COVID-19.


Subject(s)
COVID-19/immunology , Megakaryocytes/immunology , Monocytes/immunology , RNA, Viral , SARS-CoV-2/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Child , China , Cohort Studies , Cytokines/metabolism , Female , Humans , Male , Middle Aged , RNA, Viral/blood , RNA, Viral/isolation & purification , Single-Cell Analysis , Transcriptome/immunology , Young Adult
13.
Cell ; 184(7): 1865-1883.e20, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33636127

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of the ongoing coronavirus disease 2019 (COVID-19) pandemic. Understanding of the RNA virus and its interactions with host proteins could improve therapeutic interventions for COVID-19. By using icSHAPE, we determined the structural landscape of SARS-CoV-2 RNA in infected human cells and from refolded RNAs, as well as the regulatory untranslated regions of SARS-CoV-2 and six other coronaviruses. We validated several structural elements predicted in silico and discovered structural features that affect the translation and abundance of subgenomic viral RNAs in cells. The structural data informed a deep-learning tool to predict 42 host proteins that bind to SARS-CoV-2 RNA. Strikingly, antisense oligonucleotides targeting the structural elements and FDA-approved drugs inhibiting the SARS-CoV-2 RNA binding proteins dramatically reduced SARS-CoV-2 infection in cells derived from human liver and lung tumors. Our findings thus shed light on coronavirus and reveal multiple candidate therapeutics for COVID-19 treatment.


Subject(s)
COVID-19 Drug Treatment , Drug Repositioning , RNA, Viral , RNA-Binding Proteins/antagonists & inhibitors , SARS-CoV-2 , Animals , Cell Line , Chlorocebus aethiops , Deep Learning , Humans , Nucleic Acid Conformation , RNA, Viral/chemistry , RNA-Binding Proteins/metabolism , SARS-CoV-2/drug effects , SARS-CoV-2/genetics
14.
Cell ; 184(9): 2276-2278, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33930293

ABSTRACT

Infection with SARS-CoV-2 sets off a molecular arms race between virus replication and host cell defense. In this issue of Cell, Flynn, Belk, et al. integrate an advanced large-scale RNA-centered approach with custom CRISPR screens to functionally characterize the interactome of the SARS-CoV-2 RNA genome during infection.


Subject(s)
COVID-19 , SARS-CoV-2 , Friends , Host-Pathogen Interactions/genetics , Humans , RNA, Viral/genetics
15.
Cell ; 184(18): 4713-4733.e22, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34352228

ABSTRACT

SARS-CoV-2 infection can cause severe respiratory COVID-19. However, many individuals present with isolated upper respiratory symptoms, suggesting potential to constrain viral pathology to the nasopharynx. Which cells SARS-CoV-2 primarily targets and how infection influences the respiratory epithelium remains incompletely understood. We performed scRNA-seq on nasopharyngeal swabs from 58 healthy and COVID-19 participants. During COVID-19, we observe expansion of secretory, loss of ciliated, and epithelial cell repopulation via deuterosomal cell expansion. In mild and moderate COVID-19, epithelial cells express anti-viral/interferon-responsive genes, while cells in severe COVID-19 have muted anti-viral responses despite equivalent viral loads. SARS-CoV-2 RNA+ host-target cells are highly heterogenous, including developing ciliated, interferon-responsive ciliated, AZGP1high goblet, and KRT13+ "hillock"-like cells, and we identify genes associated with susceptibility, resistance, or infection response. Our study defines protective and detrimental responses to SARS-CoV-2, the direct viral targets of infection, and suggests that failed nasal epithelial anti-viral immunity may underlie and precede severe COVID-19.


Subject(s)
COVID-19/immunology , COVID-19/virology , Immunity , SARS-CoV-2/physiology , Severity of Illness Index , Adult , Aged , Bystander Effect , COVID-19/genetics , Cohort Studies , Female , Humans , Male , Middle Aged , Nasopharynx/pathology , Nasopharynx/virology , RNA, Viral/analysis , RNA, Viral/genetics , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Transcription, Genetic , Viral Load
16.
Cell ; 184(2): 323-333.e9, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33306959

ABSTRACT

The December 2019 outbreak of a novel respiratory virus, SARS-CoV-2, has become an ongoing global pandemic due in part to the challenge of identifying symptomatic, asymptomatic, and pre-symptomatic carriers of the virus. CRISPR diagnostics can augment gold-standard PCR-based testing if they can be made rapid, portable, and accurate. Here, we report the development of an amplification-free CRISPR-Cas13a assay for direct detection of SARS-CoV-2 from nasal swab RNA that can be read with a mobile phone microscope. The assay achieved ∼100 copies/µL sensitivity in under 30 min of measurement time and accurately detected pre-extracted RNA from a set of positive clinical samples in under 5 min. We combined crRNAs targeting SARS-CoV-2 RNA to improve sensitivity and specificity and directly quantified viral load using enzyme kinetics. Integrated with a reader device based on a mobile phone, this assay has the potential to enable rapid, low-cost, point-of-care screening for SARS-CoV-2.


Subject(s)
COVID-19 Nucleic Acid Testing/methods , Cell Phone/instrumentation , Optical Imaging/methods , RNA, Viral/analysis , Viral Load/methods , Animals , COVID-19 Nucleic Acid Testing/economics , COVID-19 Nucleic Acid Testing/instrumentation , CRISPR-Cas Systems , Cell Line , Coronavirus Nucleocapsid Proteins/genetics , Humans , Nasopharynx/virology , Optical Imaging/instrumentation , Phosphoproteins/genetics , Point-of-Care Testing , RNA Interference , RNA, Viral/genetics , Sensitivity and Specificity , Viral Load/economics , Viral Load/instrumentation
17.
Cell ; 184(13): 3474-3485.e11, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34143953

ABSTRACT

The capping of mRNA and the proofreading play essential roles in SARS-CoV-2 replication and transcription. Here, we present the cryo-EM structure of the SARS-CoV-2 replication-transcription complex (RTC) in a form identified as Cap(0)-RTC, which couples a co-transcriptional capping complex (CCC) composed of nsp12 NiRAN, nsp9, the bifunctional nsp14 possessing an N-terminal exoribonuclease (ExoN) and a C-terminal N7-methyltransferase (N7-MTase), and nsp10 as a cofactor of nsp14. Nsp9 and nsp12 NiRAN recruit nsp10/nsp14 into the Cap(0)-RTC, forming the N7-CCC to yield cap(0) (7MeGpppA) at 5' end of pre-mRNA. A dimeric form of Cap(0)-RTC observed by cryo-EM suggests an in trans backtracking mechanism for nsp14 ExoN to facilitate proofreading of the RNA in concert with polymerase nsp12. These results not only provide a structural basis for understanding co-transcriptional modification of SARS-CoV-2 mRNA but also shed light on how replication fidelity in SARS-CoV-2 is maintained.


Subject(s)
Coronavirus RNA-Dependent RNA Polymerase/genetics , Exoribonucleases/genetics , Methyltransferases/genetics , SARS-CoV-2/genetics , Amino Acid Sequence , COVID-19/virology , Humans , RNA, Messenger/genetics , RNA, Viral/genetics , Sequence Alignment , Transcription, Genetic/genetics , Virus Replication/genetics
18.
Cell ; 184(13): 3410-3425.e17, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34062120

ABSTRACT

To control viral infection, vertebrates rely on both inducible interferon responses and less well-characterized cell-intrinsic responses composed of "at the ready" antiviral effector proteins. Here, we show that E3 ubiquitin ligase TRIM7 is a cell-intrinsic antiviral effector that restricts multiple human enteroviruses by targeting viral 2BC, a membrane remodeling protein, for ubiquitination and proteasome-dependent degradation. Selective pressure exerted by TRIM7 results in emergence of a TRIM7-resistant coxsackievirus with a single point mutation in the viral 2C ATPase/helicase. In cultured cells, the mutation helps the virus evade TRIM7 but impairs optimal viral replication, and this correlates with a hyperactive and structurally plastic 2C ATPase. Unexpectedly, the TRIM7-resistant virus has a replication advantage in mice and causes lethal pancreatitis. These findings reveal a unique mechanism for targeting enterovirus replication and provide molecular insight into the benefits and trade-offs of viral evolution imposed by a host restriction factor.


Subject(s)
Enterovirus/physiology , Enterovirus/pathogenicity , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Virus Replication/physiology , Adenosine Triphosphatases/metabolism , Animals , Cell Line , Female , Humans , Inflammation/pathology , Mice, Inbred C57BL , Mutation/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , RNA, Viral/metabolism , Ubiquitin/metabolism , Viral Proteins/genetics
19.
Cell ; 184(17): 4430-4446.e22, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34416147

ABSTRACT

Alphaviruses cause severe arthritogenic or encephalitic disease. The E1 structural glycoprotein is highly conserved in these viruses and mediates viral fusion with host cells. However, the role of antibody responses to the E1 protein in immunity is poorly understood. We isolated E1-specific human monoclonal antibodies (mAbs) with diverse patterns of recognition for alphaviruses (ranging from Eastern equine encephalitis virus [EEEV]-specific to alphavirus cross-reactive) from survivors of natural EEEV infection. Antibody binding patterns and epitope mapping experiments identified differences in E1 reactivity based on exposure of epitopes on the glycoprotein through pH-dependent mechanisms or presentation on the cell surface prior to virus egress. Therapeutic efficacy in vivo of these mAbs corresponded with potency of virus egress inhibition in vitro and did not require Fc-mediated effector functions for treatment against subcutaneous EEEV challenge. These studies reveal the molecular basis for broad and protective antibody responses to alphavirus E1 proteins.


Subject(s)
Alphavirus/immunology , Antibodies, Viral/immunology , Cross Reactions/immunology , Viral Proteins/immunology , Virus Release/physiology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/immunology , Antigens, Viral/immunology , Cell Line , Chikungunya virus/immunology , Encephalitis Virus, Eastern Equine/immunology , Encephalomyelitis, Equine/immunology , Encephalomyelitis, Equine/virology , Epitope Mapping , Female , Horses , Humans , Hydrogen-Ion Concentration , Joints/pathology , Male , Mice, Inbred C57BL , Models, Biological , Protein Binding , RNA, Viral/metabolism , Receptors, Fc/metabolism , Temperature , Virion/metabolism , Virus Internalization
20.
Cell ; 184(8): 2229-2238.e13, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33691138

ABSTRACT

The biosafety level 3 (BSL-3) requirement to culture severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a bottleneck for research. Here, we report a trans-complementation system that produces single-round infectious SARS-CoV-2 that recapitulates authentic viral replication. We demonstrate that the single-round infectious SARS-CoV-2 can be used at BSL-2 laboratories for high-throughput neutralization and antiviral testing. The trans-complementation system consists of two components: a genomic viral RNA containing ORF3 and envelope gene deletions, as well as mutated transcriptional regulator sequences, and a producer cell line expressing the two deleted genes. Trans-complementation of the two components generates virions that can infect naive cells for only one round but does not produce wild-type SARS-CoV-2. Hamsters and K18-hACE2 transgenic mice inoculated with the complementation-derived virions exhibited no detectable disease, even after intracranial inoculation with the highest possible dose. Thus, the trans-complementation platform can be safely used at BSL-2 laboratories for research and countermeasure development.


Subject(s)
COVID-19/virology , Containment of Biohazards/methods , SARS-CoV-2 , A549 Cells , Animals , Chlorocebus aethiops , Cricetinae , Genetic Complementation Test/methods , Genome, Viral , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic , RNA, Viral , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , SARS-CoV-2/physiology , Vero Cells , Virulence , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL