Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 391
Filtrar
1.
Nature ; 627(8005): 873-879, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38418882

RESUMO

Cyclic GMP-AMP synthase (cGAS) senses aberrant DNA during infection, cancer and inflammatory disease, and initiates potent innate immune responses through the synthesis of 2'3'-cyclic GMP-AMP (cGAMP)1-7. The indiscriminate activity of cGAS towards DNA demands tight regulatory mechanisms that are necessary to maintain cell and tissue homeostasis under normal conditions. Inside the cell nucleus, anchoring to nucleosomes and competition with chromatin architectural proteins jointly prohibit cGAS activation by genomic DNA8-15. However, the fate of nuclear cGAS and its role in cell physiology remains unclear. Here we show that the ubiquitin proteasomal system (UPS) degrades nuclear cGAS in cycling cells. We identify SPSB3 as the cGAS-targeting substrate receptor that associates with the cullin-RING ubiquitin ligase 5 (CRL5) complex to ligate ubiquitin onto nuclear cGAS. A cryo-electron microscopy structure of nucleosome-bound cGAS in a complex with SPSB3 reveals a highly conserved Asn-Asn (NN) minimal degron motif at the C terminus of cGAS that directs SPSB3 recruitment, ubiquitylation and cGAS protein stability. Interference with SPSB3-regulated nuclear cGAS degradation primes cells for type I interferon signalling, conferring heightened protection against infection by DNA viruses. Our research defines protein degradation as a determinant of cGAS regulation in the nucleus and provides structural insights into an element of cGAS that is amenable to therapeutic exploitation.


Assuntos
Proteínas Nucleares , Nucleossomos , Nucleotidiltransferases , Proteólise , Ubiquitina-Proteína Ligases , Animais , Humanos , Camundongos , Núcleo Celular/metabolismo , Microscopia Crioeletrônica , 60652 , Infecções por Vírus de DNA/imunologia , Vírus de DNA/imunologia , Vírus de DNA/metabolismo , DNA Viral/imunologia , DNA Viral/metabolismo , Imunidade Inata , Reconhecimento da Imunidade Inata , Interferon Tipo I/imunologia , Proteínas Nucleares/metabolismo , Nucleossomos/química , Nucleossomos/metabolismo , Nucleossomos/ultraestrutura , Nucleotidiltransferases/química , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/ultraestrutura , Complexo de Endopeptidases do Proteassoma/metabolismo , Estabilidade Proteica , Especificidade por Substrato , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/ultraestrutura , Ubiquitinação
2.
ACS Chem Biol ; 18(8): 1808-1820, 2023 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-37498174

RESUMO

The constant and the sudden emergence of zoonotic human and animal viruses is a significant threat to human health, the world economy, and the world food supply. This has necessitated the development of broad-spectrum therapeutic strategies to combat these emerging pathogens. Mechanisms that are essential for viral replication and propagation have been successfully targeted in the past to develop broad-spectrum therapeutics that can be readily repurposed to combat new zoonotic pathogens. Because of the importance of viral RNA capping enzymes to viral replication and pathogenesis, as well as their presence in both DNA and RNA viruses, these viral proteins have been a long-standing therapeutic target. Here, we use genome sequencing information and yeast-based platforms (YeRC0M) to identify, characterize, and target viral genome-encoded essential RNA capping enzymes from emerging strains of DNA viruses, i.e., Monkeypox virus and African Swine Fever Virus, which are a significant threat to human and domestic animal health. We first identified and biochemically characterized these viral RNA capping enzymes and their necessary protein domains. We observed significant differences in functional protein domains and organization for RNA capping enzymes from emerging DNA viruses in comparison to emerging RNA viruses. We also observed several differences in the biochemical properties of these viral RNA capping enzymes using our phenotypic yeast-based approaches (YeRC0M) as compared to the previous in vitro studies. Further, using directed evolution, we were able to identify inactivation and attenuation mutations in these essential viral RNA capping enzymes; these data could have implications on virus biocontainment as well as live attenuated vaccine development. We also developed methods that would facilitate high-throughput phenotypic screening to identify broad-spectrum inhibitors that selectively target viral RNA capping enzymes over host RNA capping enzymes. As demonstrated here, our approaches to identify, characterize, and target viral genome-encoded essential RNA capping enzymes are highly modular and can be readily adapted for targeting emerging viral pathogens as well as their variants that emerge in the future.


Assuntos
Vírus da Febre Suína Africana , Vírus , Animais , Humanos , Suínos , Saccharomyces cerevisiae/metabolismo , Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/metabolismo , Vírus/genética , RNA Viral/genética , RNA Viral/metabolismo , Replicação Viral , Vírus de DNA/genética , Vírus de DNA/metabolismo
3.
Nature ; 617(7960): 409-416, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37138077

RESUMO

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


Assuntos
Vírus de DNA , Intestinos , Proteínas Virais , Vírion , Humanos , Capsídeo/química , Capsídeo/metabolismo , Capsídeo/ultraestrutura , Microscopia Crioeletrônica , Vírus de DNA/química , Vírus de DNA/classificação , Vírus de DNA/isolamento & purificação , Vírus de DNA/metabolismo , Vírus de DNA/ultraestrutura , Vírion/química , Vírion/metabolismo , Vírion/ultraestrutura , Montagem de Vírus , Intestinos/microbiologia , Intestinos/virologia , Proteínas Virais/química , Proteínas Virais/metabolismo , Proteínas Virais/ultraestrutura , Desdobramento de Proteína , Dobramento de Proteína
4.
Microbiol Mol Biol Rev ; 87(2): e0003522, 2023 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-37052496

RESUMO

Immune recognition of viral genome-derived double-stranded RNA (dsRNA) molecules and their subsequent processing into small interfering RNAs (siRNAs) in plants, invertebrates, and mammals trigger specific antiviral immunity known as antiviral RNA interference (RNAi). Immune sensing of viral dsRNA is sequence-independent, and most regions of viral RNAs are targeted by virus-derived siRNAs which extensively overlap in sequence. Thus, the high mutation rates of viruses do not drive immune escape from antiviral RNAi, in contrast to other mechanisms involving specific virus recognition by host immune proteins such as antibodies and resistance (R) proteins in mammals and plants, respectively. Instead, viruses actively suppress antiviral RNAi at various key steps with a group of proteins known as viral suppressors of RNAi (VSRs). Some VSRs are so effective in virus counter-defense that potent inhibition of virus infection by antiviral RNAi is undetectable unless the cognate VSR is rendered nonexpressing or nonfunctional. Since viral proteins are often multifunctional, resistance phenotypes of antiviral RNAi are accurately defined by those infection defects of VSR-deletion mutant viruses that are efficiently rescued by host deficiency in antiviral RNAi. Here, we review and discuss in vivo infection defects of VSR-deficient RNA and DNA viruses resulting from the actions of host antiviral RNAi in model systems.


Assuntos
Antivirais , Vírus de RNA , Animais , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Vírus de DNA/genética , Vírus de DNA/metabolismo , Vírus de RNA/genética , Mamíferos/genética , Mamíferos/metabolismo
5.
Viruses ; 15(3)2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36992355

RESUMO

The innate immune system is the first line of host defense sensing viral infection. Manganese (Mn) has recently been found to be involved in the activation of the innate immune DNA-sensing cGAS-STING pathway and subsequent anti-DNA virus function. However, it is still unclear whether Mn2+ mediates host defense against RNA viruses. In this study, we demonstrate that Mn2+ exhibited antiviral effects against various animal and human viruses, including RNA viruses such as PRRSVs and VSV, as well as DNA viruses such as HSV1, in a dose-dependent manner. Moreover, cGAS and STING were both investigated in the Mn2+ mediated antiviral roles using the knockout cells made by the CRISPR-Cas9 approach. Unexpectedly, the results revealed that neither cGAS knockout nor STING knockout had any effect on Mn2+-mediated antiviral functions. Nevertheless, we verified that Mn2+ promoted the activation of the cGAS-STING signaling pathway. These findings suggest that Mn2+ has broad-spectrum antiviral activities in a cGAS-STING pathway independent manner. This study also provides significant insights into redundant mechanisms participating in the Mn2+ antiviral functions, and also indicates a new target for Mn2+ antiviral therapeutics.


Assuntos
Antivirais , Imunidade Inata , Manganês , Animais , Humanos , Antivirais/farmacologia , Vírus de DNA/metabolismo , Manganês/farmacologia , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Transdução de Sinais , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo
6.
J Med Virol ; 95(1): e28310, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36377393

RESUMO

Cellular infections by DNA viruses trigger innate immune responses mediated by DNA sensors. The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon gene (STING) signaling pathway has been identified as a DNA-sensing pathway that activates interferons in response to viral infection and, thus, mediates host defense against viruses. Previous studies have identified oncogenes E7 and E1A of the DNA tumor viruses, human papillomavirus 18 (HPV18) and adenovirus, respectively, as inhibitors of the cGAS-STING pathway. However, the function of STING in infected cells and the mechanism by which HPV18 E7 antagonizes STING-induced Interferon beta production remain unknown. We report that HPV18 E7 selectively antagonizes STING-triggered nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation but not IRF3 activation. HPV18 E7 binds to STING in a region critical for NF-κB activation and blocks the nuclear accumulation of p65. Moreover, E7 inhibition of STING-triggered NF-κB activation is related to HPV pathogenicity but not E7-Rb binding. HPV18 E7, severe acute respiratory syndrome coronavirus-2 open reading frame 3a, human immunodeficiency virus-2 viral protein X, and Kaposi's sarcoma-associated herpesvirus KSHV viral interferon regulatory factor 1 selectively inhibited STING-triggered NF-κB or IRF3 activation, suggesting a convergent evolution among these viruses toward antagonizing host innate immunity. Collectively, selective suppression of the cGAS-STING pathway by viral proteins is likely to be a key pathogenic determinant, making it a promising target for treating oncogenic virus-induced tumor diseases.


Assuntos
COVID-19 , NF-kappa B , Humanos , NF-kappa B/metabolismo , Interferon beta/genética , Papillomavirus Humano 18/genética , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Imunidade Inata , DNA , Vírus de DNA/genética , Vírus de DNA/metabolismo , Proteínas Oncogênicas
7.
J Gen Virol ; 103(5)2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35510990

RESUMO

The antiviral role of innate immune responses mediated by the NF-κB family of transcription factors is well established in vertebrates but was for a long time less clear in insects. Insects encode two canonical NF-κB pathways, the Toll and Imd ('immunodeficiency') pathways, which are best characterised for their role in antibacterial and antifungal defence. An increasing body of evidence has also implicated NF-κB-mediated innate immunity in antiviral responses against some, but not all, viruses. Specific pattern recognition receptors (PRRs) and molecular events leading to NF-κB activation by viral pathogen-associated molecular patterns (PAMPs) have been elucidated for a number of viruses and insect species. Particularly interesting are recent findings indicating that the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway detects viral RNA to activate NF-κB-regulated gene expression. We summarise the literature on virus-NF-κB pathway interactions across the class Insecta, with a focus on the dipterans Drosophila melanogaster and Aedes aegypti. We discuss potential reasons for differences observed between different virus-host combinations, and highlight similarities and differences between cGAS-STING signalling in insects versus vertebrates. Finally, we summarise the increasing number of known molecular mechanisms by which viruses antagonise NF-κB responses, which suggest that NF-κB-mediated immunity exerts strong evolutionary pressures on viruses. These developments in our understanding of insect antiviral immunity have relevance to the large number of insect species that impact on humans through their transmission of human, livestock and plant diseases, exploitation as biotechnology platforms, and role as parasites, pollinators, livestock and pests.


Assuntos
Drosophila melanogaster , NF-kappa B , Animais , Antivirais , Vírus de DNA/metabolismo , Drosophila melanogaster/metabolismo , Imunidade Inata , Insetos , NF-kappa B/genética , NF-kappa B/metabolismo , Nucleotidiltransferases
8.
Commun Biol ; 5(1): 433, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35538147

RESUMO

The DNA sensor cGAS detects cytosolic DNA and instigates type I interferon (IFN) expression. Recent studies find that cGAS also localizes in the nucleus and binds the chromatin. Despite the mechanism controlling nuclear cGAS activation is well elucidated, whether nuclear cGAS participates in DNA sensing is unclear. Here, we report that herpes simplex virus 1 (HSV-1) infection caused the release of cGAS from the chromatin into the nuclear soluble fraction. Like its cytosolic counterpart, the leaked nuclear soluble cGAS also sensed viral DNA, produced cGAMP, and induced mRNA expression of type I IFN and interferon-stimulated genes. Consistently, the nuclear soluble cGAS limited HSV-1 infection. Furthermore, enzyme-deficient mutation (D307A) or cGAS inhibitor RU.251 abolished nuclear cGAS-mediated innate immune responses, suggesting that enzymatic activity is also required for nuclear soluble cGAS. Taken all together, our study demonstrates that nuclear soluble cGAS acts as a nuclear DNA sensor detecting nuclear-replicating DNA viruses.


Assuntos
Infecções por Vírus de DNA , Vírus de DNA , Nucleotidiltransferases , Cromatina , DNA/genética , DNA/metabolismo , Infecções por Vírus de DNA/genética , Infecções por Vírus de DNA/metabolismo , Infecções por Vírus de DNA/virologia , Vírus de DNA/genética , Vírus de DNA/metabolismo , Herpes Simples/genética , Humanos , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo
9.
Curr Opin Insect Sci ; 49: 85-92, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34974161

RESUMO

RNA interference pathways mediated by different types of small non-coding RNAs (siRNAs, miRNAs and piRNAs) are conserved biological responses to exotic stresses, including viral infection. Aside from the well-established siRNA pathway, the miRNA pathway and the piRNA pathway process viral sequences, exogenously or endogenously, into miRNAs and piRNAs, respectively. During the host-virus interaction, viral sequences, including both coding and non-coding sequences, can be integrated as endogenous viral elements (EVEs) and thereby become present within the germline of a non-viral organism. In recent years, significant progress has been made in characterizing the biogenesis and function of viruses and EVEs associated with snRNAs. Overall, the siRNA pathway acts as the primarily antiviral defense against a wide range of exogenous viruses; the miRNA pathways associated with viruses or EVEs function in antiviral response and host gene regulation; EVE derived piRNAs with a ping-pong signature have the potential to limit cognate viral infection.


Assuntos
Vírus de Insetos , MicroRNAs , Vírus , Animais , Antivirais , Vírus de DNA/genética , Vírus de DNA/metabolismo , Vírus de Insetos/genética , Insetos/genética , Insetos/metabolismo , MicroRNAs/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Vírus/genética
10.
Nat Commun ; 12(1): 4748, 2021 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-34362927

RESUMO

Encapsulins are a class of microbial protein compartments defined by the viral HK97-fold of their capsid protein, self-assembly into icosahedral shells, and dedicated cargo loading mechanism for sequestering specific enzymes. Encapsulins are often misannotated and traditional sequence-based searches yield many false positive hits in the form of phage capsids. Here, we develop an integrated search strategy to carry out a large-scale computational analysis of prokaryotic genomes with the goal of discovering an exhaustive and curated set of all HK97-fold encapsulin-like systems. We find over 6,000 encapsulin-like systems in 31 bacterial and four archaeal phyla, including two novel encapsulin families. We formulate hypotheses about their potential biological functions and biomedical relevance, which range from natural product biosynthesis and stress resistance to carbon metabolism and anaerobic hydrogen production. An evolutionary analysis of encapsulins and related HK97-type virus families shows that they share a common ancestor, and we conclude that encapsulins likely evolved from HK97-type bacteriophages.


Assuntos
Bacteriófagos/metabolismo , Capsídeo/metabolismo , Células Procarióticas/metabolismo , Células Procarióticas/virologia , Archaea/metabolismo , Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bacteriófagos/genética , Evolução Biológica , Vias Biossintéticas/genética , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Vírus de DNA/metabolismo , Filogenia , Virulência
11.
Viruses ; 13(8)2021 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-34452478

RESUMO

The apolipoprotein B mRNA editing enzyme, catalytic polypeptide (APOBEC) enzyme family in humans has 11 members with diverse functions in metabolism and immunity [...].


Assuntos
Desaminase APOBEC-1/genética , Vírus de DNA/imunologia , Imunidade Inata , Desaminase APOBEC-1/classificação , Desaminase APOBEC-1/metabolismo , Animais , Vírus de DNA/metabolismo , Humanos , Camundongos , Edição de RNA
12.
J Virol ; 95(21): e0081721, 2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-34406857

RESUMO

Redondoviridae is a newly established family of circular Rep-encoding single-stranded (CRESS) DNA viruses found in the human ororespiratory tract. Redondoviruses were previously found in ∼15% of respiratory specimens from U.S. urban subjects; levels were elevated in individuals with periodontitis or critical illness. Here, we report higher redondovirus prevalence in saliva samples: four rural African populations showed 61 to 82% prevalence, and an urban U.S. population showed 32% prevalence. Longitudinal, limiting-dilution single-genome sequencing revealed diverse strains of both redondovirus species (Brisavirus and Vientovirus) in single individuals, persistence over time, and evidence of intergenomic recombination. Computational analysis of viral genomes identified a recombination hot spot associated with a conserved potential DNA stem-loop structure. To assess the possible role of this site in recombination, we carried out in vitro studies which showed that this potential stem-loop was cleaved by the virus-encoded Rep protein. In addition, in reconstructed reactions, a Rep-DNA covalent intermediate was shown to mediate DNA strand transfer at this site. Thus, redondoviruses are highly prevalent in humans, found in individuals on multiple continents, heterogeneous even within individuals and encode a Rep protein implicated in facilitating recombination. IMPORTANCERedondoviridae is a recently established family of DNA viruses predominantly found in the human respiratory tract and associated with multiple clinical conditions. In this study, we found high redondovirus prevalence in saliva from urban North American individuals and nonindustrialized African populations in Botswana, Cameroon, Ethiopia, and Tanzania. Individuals on both continents harbored both known redondovirus species. Global prevalence of both species suggests that redondoviruses have long been associated with humans but have remained undetected until recently due to their divergent genomes. By sequencing single redondovirus genomes in longitudinally sampled humans, we found that redondoviruses persisted over time within subjects and likely evolve by recombination. The Rep protein encoded by redondoviruses catalyzes multiple reactions in vitro, consistent with a role in mediating DNA replication and recombination. In summary, we identify high redondovirus prevalence in humans across multiple continents, longitudinal heterogeneity and persistence, and potential mechanisms of redondovirus evolution by recombination.


Assuntos
Infecções por Vírus de DNA/virologia , Vírus de DNA/classificação , Vírus de DNA/genética , Vírus de DNA/metabolismo , Boca/virologia , Sistema Respiratório/virologia , Saliva/virologia , África/epidemiologia , Biodiversidade , Estado Terminal , Infecções por Vírus de DNA/epidemiologia , Proteínas de Ligação a DNA/metabolismo , Evolução Molecular , Genoma Viral , Humanos , Metagenômica , Periodontite/virologia , Filogenia , Prevalência , População Rural , Estados Unidos/epidemiologia , Proteínas Virais/metabolismo
13.
Cell ; 184(16): 4237-4250.e19, 2021 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-34297924

RESUMO

The organization of genomic DNA into defined nucleosomes has long been viewed as a hallmark of eukaryotes. This paradigm has been challenged by the identification of "minimalist" histones in archaea and more recently by the discovery of genes that encode fused remote homologs of the four eukaryotic histones in Marseilleviridae, a subfamily of giant viruses that infect amoebae. We demonstrate that viral doublet histones are essential for viral infectivity, localize to cytoplasmic viral factories after virus infection, and ultimately are found in the mature virions. Cryogenic electron microscopy (cryo-EM) structures of viral nucleosome-like particles show strong similarities to eukaryotic nucleosomes despite the limited sequence identify. The unique connectors that link the histone chains contribute to the observed instability of viral nucleosomes, and some histone tails assume structural roles. Our results further expand the range of "organisms" that require nucleosomes and suggest a specialized function of histones in the biology of these unusual viruses.


Assuntos
Vírus de DNA/metabolismo , Histonas/metabolismo , Nucleossomos/metabolismo , Amoeba/virologia , Corantes Fluorescentes/metabolismo , Histonas/química , Modelos Moleculares , Proteômica , Vírion/metabolismo
14.
Viruses ; 13(6)2021 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-34064125

RESUMO

To initiate infection, a virus enters a host cell typically via receptor-dependent endocytosis. It then penetrates a subcellular membrane, reaching a destination that supports transcription, translation, and replication of the viral genome. These steps lead to assembly and morphogenesis of the new viral progeny. The mature virus finally exits the host cell to begin the next infection cycle. Strikingly, viruses hijack host molecular chaperones to accomplish these distinct entry steps. Here we highlight how DNA viruses, including polyomavirus and the human papillomavirus, exploit soluble and membrane-associated chaperones to enter a cell, penetrating and escaping an intracellular membrane en route for infection. We also describe the mechanism by which RNA viruses-including flavivirus and coronavirus-co-opt cytosolic and organelle-selective chaperones to promote viral endocytosis, protein biosynthesis, replication, and assembly. These examples underscore the importance of host chaperones during virus infection, potentially revealing novel antiviral strategies to combat virus-induced diseases.


Assuntos
Vírus de DNA/fisiologia , Chaperonas Moleculares/metabolismo , Vírus de RNA/fisiologia , Citosol/metabolismo , Vírus de DNA/metabolismo , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/virologia , Endossomos/metabolismo , Endossomos/virologia , Interações Hospedeiro-Patógeno , Membranas Intracelulares/metabolismo , Vírus de RNA/metabolismo , Internalização do Vírus , Replicação Viral
15.
Nat Struct Mol Biol ; 28(5): 413-417, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33927388

RESUMO

Certain large DNA viruses, including those in the Marseilleviridae family, encode histones. Here we show that fused histone pairs Hß-Hα and Hδ-Hγ from Marseillevirus are structurally analogous to the eukaryotic histone pairs H2B-H2A and H4-H3. These viral histones form 'forced' heterodimers, and a heterotetramer of four such heterodimers assembles DNA to form structures virtually identical to canonical eukaryotic nucleosomes.


Assuntos
Vírus de DNA , DNA , Nucleossomos/metabolismo , DNA/química , DNA/metabolismo , Vírus de DNA/genética , Vírus de DNA/metabolismo , Histonas/química , Histonas/metabolismo , Ligação Proteica , Elementos Estruturais de Proteínas , Estrutura Terciária de Proteína
16.
Microbes Environ ; 36(1)2021.
Artigo em Inglês | MEDLINE | ID: mdl-33612562

RESUMO

Marseilleviridae is a family of large double-stranded DNA viruses that is currently divided into five subgroups, lineages A-E. Hokutovirus and kashiwazakivirus, both of which belong to lineage B, have been reported to induce host acanthamoeba cells to form aggregations called "bunches". This putatively results in increased opportunities to infect acanthamoeba cells, in contrast to lineage A, which has been reported to not form "bunches". In the present study, we isolated 14 virus strains of the family Marseilleviridae from several Japanese water samples, 11 of which were identified as lineage B viruses. All 11 lineage B strains caused infected amoeba cells to form bunches. We then investigated the involvement of monosaccharides in bunch formation by amoeba cells infected with hokutovirus. Galactose inhibited bunch formation, thereby allowing amoeba cells to delay the process, whereas mannose and glucose did not. A kinetic image analysis of hokutovirus-infected amoeba cells confirmed the inhibition of bunch formation by galactose. The number of hokutovirus-infected amoeba cells increased more rapidly than that of tokyovirus-infected cells, which belongs to lineage A. This result suggests that bunch formation by infected amoeba cells is advantageous for lineage B viruses.


Assuntos
Vírus de DNA/classificação , Galactose/metabolismo , Acanthamoeba/virologia , Vírus de DNA/genética , Vírus de DNA/isolamento & purificação , Vírus de DNA/metabolismo , Água Doce/virologia , Japão , Filogenia
17.
Viral Immunol ; 34(1): 41-48, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33074779

RESUMO

Nucleocytoplasmic large DNA viruses (NCLDVs) are a group of large viruses that infect a wide range of hosts, from animals to protists. These viruses are grouped together in NCLDV based on genomic sequence analyses. They share a set of essential genes for virion morphogenesis and replication. Most NCLDVs generally have large physical sizes while their morphologies vary in different families, such as icosahedral, brick, or oval shape, raising the question of the possible regulatory factor on their morphogenesis. The capsids of icosahedral NCLDVs are assembled from small building blocks, named capsomers, which are the trimeric form of the major capsid proteins. Note that the capsids of immature poxvirus are spherical even though they are assembled from capsomers that share high structural conservation with those icosahedral NCLDVs. The recently published high resolution structure of NCLDVs, Paramecium bursaria Chlorella virus 1 and African swine fever virus, described the intensive network of minor capsid proteins that are located underneath the capsomers. Among these minor proteins is the elongated tape measure protein (TmP) that spans from one icosahedral fivefold vertex to another. In this study, we focused on the critical roles that TmP plays in the assembly of icosahedral NCLDV capsids, answering a question raised in a previously proposed spiral mechanism. Interestingly, basic local alignment search on the TmPs showed no significant hits in poxviruses, which might be the factor that differentiates poxviruses and icosahedral NCLDVs in their morphogenesis.


Assuntos
Proteínas do Capsídeo/metabolismo , Capsídeo/química , Capsídeo/metabolismo , Vírus de DNA/química , Vírus de DNA/metabolismo , Montagem de Vírus , Vírus da Febre Suína Africana/química , Vírus da Febre Suína Africana/metabolismo , Animais , Chlorella/virologia , Suínos
18.
mSphere ; 5(6)2020 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-33177215

RESUMO

Double-stranded RNA (dsRNA) is the hallmark of many viral infections. dsRNA is produced either by RNA viruses during replication or by DNA viruses upon convergent transcription. Synthetic dsRNA is also able to mimic viral-induced activation of innate immune response and cell death. In this study, we employed a genome-wide CRISPR-Cas9 loss-of-function screen based on cell survival in order to identify genes implicated in the host response to dsRNA. By challenging HCT116 human cells with either synthetic dsRNA or Sindbis virus (SINV), we identified the heparan sulfate (HS) pathway as a crucial factor for dsRNA entry, and we validated SINV dependency on HS. Interestingly, we uncovered a novel role for COG4, a component of the conserved oligomeric Golgi (COG) complex, as a factor involved in cell survival to both dsRNA and SINV in human cells. We showed that COG4 knockout led to a decrease of extracellular HS that specifically affected dsRNA transfection efficiency and reduced viral production, which explains the increased cell survival of these mutants.IMPORTANCE When facing a viral infection, the organism has to put in place a number of defense mechanisms in order to clear the pathogen from the cell. At the early phase of this preparation for fighting against the invader, the innate immune response is triggered by the sensing of danger signals. Among those molecular cues, double-stranded RNA (dsRNA) is a very potent inducer of different reactions at the cellular level that can ultimately lead to cell death. Using a genome-wide screening approach, we set to identify genes involved in dsRNA entry, sensing, and apoptosis induction in human cells. This allowed us to determine that the heparan sulfate pathway and the conserved oligomeric Golgi complex are key determinants allowing entry of both dsRNA and viral nucleic acid leading to cell death.


Assuntos
Vírus de DNA/metabolismo , Vírus de RNA/metabolismo , RNA de Cadeia Dupla/metabolismo , Viroses/metabolismo , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Complexo de Golgi/metabolismo , Células HCT116 , Heparitina Sulfato/metabolismo , Humanos , Imunidade Inata , Viroses/patologia
19.
Nat Rev Microbiol ; 18(10): 559-570, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32533130

RESUMO

Eukaryotic gene expression is regulated not only by genomic enhancers and promoters, but also by covalent modifications added to both chromatin and RNAs. Whereas cellular gene expression may be either enhanced or inhibited by specific epigenetic modifications deposited on histones (in particular, histone H3), these epigenetic modifications can also repress viral gene expression, potentially functioning as a potent antiviral innate immune response in DNA virus-infected cells. However, viruses have evolved countermeasures that prevent the epigenetic silencing of their genes during lytic replication, and they can also take advantage of epigenetic silencing to establish latent infections. By contrast, the various covalent modifications added to RNAs, termed epitranscriptomic modifications, can positively regulate mRNA translation and/or stability, and both DNA and RNA viruses have evolved to utilize epitranscriptomic modifications as a means to maximize viral gene expression. As a consequence, both chromatin and RNA modifications could serve as novel targets for the development of antivirals. In this Review, we discuss how host epigenetic and epitranscriptomic processes regulate viral gene expression at the levels of chromatin and RNA function, respectively, and explore how viruses modify, avoid or utilize these processes in order to regulate viral gene expression.


Assuntos
Vírus de DNA/genética , Epigênese Genética , Regulação Viral da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Processamento Pós-Transcricional do RNA , Vírus de RNA/genética , Animais , Antivirais/farmacologia , Cromatina/química , Cromatina/metabolismo , Cromatina/virologia , Vírus de DNA/efeitos dos fármacos , Vírus de DNA/metabolismo , Células Eucarióticas/efeitos dos fármacos , Células Eucarióticas/metabolismo , Células Eucarióticas/virologia , Histonas/genética , Histonas/metabolismo , Humanos , Regiões Promotoras Genéticas , Biossíntese de Proteínas , Vírus de RNA/efeitos dos fármacos , Vírus de RNA/metabolismo , Transcriptoma , Latência Viral , Replicação Viral
20.
Curr Opin Struct Biol ; 65: 17-25, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32570107

RESUMO

Bacterial CRISPR-Cas systems provide adaptive immunity against viruses and other mobile genome elements. During the adaptation step cells become immunized by insertion of short fragments of foreign DNA, termed spacers, into the genomic region called a CRISPR array. Selection, processing and insertion of new spacers is an elaborate and precisely orchestrated reaction, which relies on the Cas1-Cas2 integrase complex and accessory proteins that vary among different types of CRISPR-Cas systems. This review focuses on CRISPR adaptation from the structural perspective, with the spotlight on adaptation proteins employed by type I and type II CRISPR-Cas systems.


Assuntos
Proteínas de Bactérias , Proteínas Associadas a CRISPR/metabolismo , Sistemas CRISPR-Cas , Vírus de DNA/metabolismo , Escherichia coli/genética , Vírus , Proteínas de Bactérias/metabolismo , Escherichia coli/virologia , Interações entre Hospedeiro e Microrganismos , Ligação Proteica , Vírus/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...