Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 356
Filtrar
1.
Nature ; 627(8005): 873-879, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38418882

RESUMEN

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.


Asunto(s)
Proteínas Nucleares , Nucleosomas , Nucleotidiltransferasas , Proteolisis , Ubiquitina-Proteína Ligasas , Animales , Humanos , Ratones , Núcleo Celular/metabolismo , Microscopía por Crioelectrón , Degrones , Infecciones por Virus ADN/inmunología , Virus ADN/inmunología , Virus ADN/metabolismo , ADN Viral/inmunología , ADN Viral/metabolismo , Inmunidad Innata , Reconocimiento de Inmunidad Innata , Interferón Tipo I/inmunología , Proteínas Nucleares/metabolismo , Nucleosomas/química , Nucleosomas/metabolismo , Nucleosomas/ultraestructura , Nucleotidiltransferasas/química , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/ultraestructura , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Especificidad por Sustrato , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/ultraestructura , Ubiquitinación
2.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34969857

RESUMEN

Type I interferons (IFNs) are the first frontline of the host innate immune response against invading pathogens. Herein, we characterized an unknown protein encoded by phospholipase A2 inhibitor and LY6/PLAUR domain-containing (PINLYP) gene that interacted with TBK1 and induced type I IFN in a TBK1- and IRF3-dependent manner. Loss of PINLYP impaired the activation of IRF3 and production of IFN-ß induced by DNA virus, RNA virus, and various Toll-like receptor ligands in multiple cell types. Because PINLYP deficiency in mice engendered an early embryonic lethality in mice, we generated a conditional mouse in which PINLYP was depleted in dendritic cells. Mice lacking PINLYP in dendritic cells were defective in type I IFN induction and more susceptible to lethal virus infection. Thus, PINLYP is a positive regulator of type I IFN innate immunity and important for effective host defense against viral infection.


Asunto(s)
Células Dendríticas/inmunología , Inhibidores Enzimáticos/inmunología , Inmunidad Innata , Interferón beta/inmunología , Animales , Línea Celular , Infecciones por Virus ADN/genética , Infecciones por Virus ADN/inmunología , Virus ADN/genética , Virus ADN/inmunología , Humanos , Interferón beta/genética , Ratones , Ratones Noqueados , Infecciones por Virus ARN/genética , Infecciones por Virus ARN/inmunología , Virus ARN/genética , Virus ARN/inmunología
3.
J Immunol ; 207(11): 2699-2709, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34750204

RESUMEN

IFN-γ-inducible protein 16 (IFI16) recognizes viral DNAs from both nucleus-replicating viruses and cytoplasm-replicating viruses. Isoform 2 of IFI16 (IFI16-iso2) with nuclear localization sequence (NLS) has been studied extensively as a well-known DNA sensor. However, the characteristics and functions of other IFI16 isoforms are almost unknown. Here, we find that IFI16-iso1, with exactly the same length as IFI16-iso2, lacks the NLS and locates in the cytoplasm. To distinguish the functions of IFI16-iso1 and IFI16-iso2, we have developed novel nuclear viral DNA mimics that can be recognized by the nuclear DNA sensors, including IFI16-iso2 and hnRNPA2B1. The hexanucleotide motif 5'-AGTGTT-3' DNA form of the nuclear localization sequence (DNLS) effectively drives cytoplasmic viral DNA nuclear translocation. These nuclear viral DNA mimics potently induce IFN-ß and antiviral IFN-stimulated genes in human A549 cells, HEK293T cells, and mouse macrophages. The subcellular location difference of IFI16 isoforms determines their differential functions in recognizing viral DNA and activating type I IFN-dependent antiviral immunity. IFI16-iso1 preferentially colocalizes with cytoplasmic HSV60mer and cytoplasm-replicating vaccinia virus (VACV), whereas IFI16-iso2 mainly colocalizes with nuclear HSV60-DNLS and nucleus-replicating HSV-1. Compared with IFI16-iso2, IFI16-iso1 induces more transcription of IFN-ß and IFN-stimulated genes, as well as stronger antiviral immunity upon HSV60mer transfection or VACV infection. IFI16-iso2, with the ability of nuclear-cytoplasmic shuttling, clears both invaded HSV type 1 and VACV significantly. However, IFI16-iso2 induces more type I IFN-dependent antiviral immunity than IFI16-iso1 upon HSV60-DNLS transfection or HSV type 1 infection. Our study has developed potent agonists for nuclear DNA sensors and also has demonstrated that IFI16 isoforms with cytoplasmic and nuclear locations play differential roles in innate immunity against DNA viruses.


Asunto(s)
Núcleo Celular/inmunología , Virus ADN/inmunología , Proteínas Nucleares/inmunología , Fosfoproteínas/inmunología , Células Cultivadas , Humanos , Isoformas de Proteínas/inmunología
4.
Exp Mol Med ; 53(11): 1647-1668, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34782737

RESUMEN

A harmonized balance between positive and negative regulation of pattern recognition receptor (PRR)-initiated immune responses is required to achieve the most favorable outcome for the host. This balance is crucial because it must not only ensure activation of the first line of defense against viral infection but also prevent inappropriate immune activation, which results in autoimmune diseases. Recent studies have shown how signal transduction pathways initiated by PRRs are positively and negatively regulated by diverse modulators to maintain host immune homeostasis. However, viruses have developed strategies to subvert the host antiviral response and establish infection. Viruses have evolved numerous genes encoding immunomodulatory proteins that antagonize the host immune system. This review focuses on the current state of knowledge regarding key host factors that regulate innate immune signaling molecules upon viral infection and discusses evidence showing how specific viral proteins counteract antiviral responses via immunomodulatory strategies.


Asunto(s)
Genoma Viral , Interacciones Huésped-Patógeno/inmunología , Evasión Inmune , Inmunidad Innata , Transducción de Señal , Virosis/etiología , Virosis/metabolismo , Animales , Biomarcadores , Virus ADN/genética , Virus ADN/inmunología , Resistencia a la Enfermedad , Susceptibilidad a Enfermedades/inmunología , Genoma Viral/inmunología , Humanos , Quinasa I-kappa B/metabolismo , Sistema Inmunológico , Factor 3 Regulador del Interferón/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas , Virus ARN/genética , Virus ARN/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Factor 3 Asociado a Receptor de TNF/metabolismo , Proteínas Virales/metabolismo
5.
Viruses ; 13(11)2021 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-34835121

RESUMEN

Abalone amyotrophia is a viral disease that causes mass mortality of juvenile Haliotis discus and H. madaka. Although the cause of this disease has yet to be identified, we had previously postulated a novel virus with partial genome sequence similarity to that of African swine fever virus is the causative agent and proposed abalone asfa-like virus (AbALV) as a provisional name. In this study, three species of juvenile abalone (H. gigantea, H. discus discus, and H. diversicolor) and four species of adult abalone (the above three species plus H. discus hannai) were experimentally infected, and their susceptibility to AbALV was investigated by recording mortality, quantitatively determining viral load by PCR, and conducting immunohistological studies. In the infection test using 7-month-old animals, H. gigantea, which was previously reported to be insusceptible to the disease, showed multiplication of the virus to the same extent as in H. discus discus, resulting in mass mortality. H. discus discus at 7 months old showed abnormal cell masses, notches in the edge of the shell and brown pigmentation inside of the shell, which are histopathological and external features of this disease, while H. gigantea did not show any of these characteristics despite suffering high mortality. Adult abalones had low mortality and viral replication in all species; however, all three species, except H. diversicolor, became carriers of the virus. In immunohistological observations, cells positive for viral antigens were detected predominantly in the gills of juvenile H. discus discus and H. gigantea, and mass mortality was observed in these species. In H. diversicolor, neither juvenile nor adult mortality from infection occurred, and the AbALV genome was not increased by experimental infection through cohabitation or injection. Our results suggest that H. gigantea, H. discus discus and H. discus hannai are susceptible to AbALV, while H. diversicolor is not. These results confirmed that AbALV is the etiological agent of abalone amyotrophia.


Asunto(s)
Enfermedades de los Animales , Virus ADN/inmunología , Moluscos , Virosis , Virus no Clasificados/inmunología , Enfermedades de los Animales/inmunología , Enfermedades de los Animales/virología , Animales , Moluscos/inmunología , Moluscos/virología , Virosis/inmunología , Virosis/virología
6.
Viruses ; 13(8)2021 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-34452478

RESUMEN

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


Asunto(s)
Desaminasas APOBEC-1/genética , Virus ADN/inmunología , Inmunidad Innata , Desaminasas APOBEC-1/clasificación , Desaminasas APOBEC-1/metabolismo , Animales , Virus ADN/metabolismo , Humanos , Ratones , Edición de ARN
7.
Viruses ; 13(7)2021 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-34199077

RESUMEN

Many viruses, especially RNA viruses, utilize programmed ribosomal frameshifting and/or stop codon readthrough in their expression, and in the decoding of a few a UGA is dynamically redefined to specify selenocysteine. This recoding can effectively increase viral coding capacity and generate a set ratio of products with the same N-terminal domain(s) but different C-terminal domains. Recoding can also be regulatory or generate a product with the non-universal 21st directly encoded amino acid. Selection for translation speed in the expression of many viruses at the expense of fidelity creates host immune defensive opportunities. In contrast to host opportunism, certain viruses, including some persistent viruses, utilize recoding or adventitious frameshifting as part of their strategy to evade an immune response or specific drugs. Several instances of recoding in small intensively studied viruses escaped detection for many years and their identification resolved dilemmas. The fundamental importance of ribosome ratcheting is consistent with the initial strong view of invariant triplet decoding which however did not foresee the possibility of transitory anticodon:codon dissociation. Deep level dynamics and structural understanding of recoding is underway, and a high level structure relevant to the frameshifting required for expression of the SARS CoV-2 genome has just been determined.


Asunto(s)
Virus ADN/genética , Virus ADN/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Evasión Inmune , Virus ARN/genética , Antivirales/farmacología , Codón de Terminación , Virus ADN/efectos de los fármacos , Sistema de Lectura Ribosómico , Antígenos de Histocompatibilidad Clase I/genética , Conformación de Ácido Nucleico , Péptidos/inmunología , Biosíntesis de Proteínas , Virus ARN/efectos de los fármacos , Virus ARN/inmunología
9.
Front Immunol ; 12: 689783, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34168656

RESUMEN

Interferon (IFN) system is considered as the first defense line against viral infection, and it has been extensively studied in vertebrates from fish to mammals. In invertebrates, Vagos from arthropod and IFN-like protein (CgIFNLP) from Crassostrea gigas appeared to function as IFN-like antiviral cytokines. In the present study, the CgIFNLP protein in hemocytes was observed to increase after Poly (I:C) stimulation. After CgIFNLP was knocked down by RNAi, the mRNA expression of IFN-stimulated genes (CgISGs) was significantly inhibited. Both cyclic GMP-AMP synthase (CgcGAS) and stimulator of interferon gene (CgSTING) identified from oyster were able to recognize the double-stranded nucleic acid [Poly (I:C) and dsDNA] and expressed at high level after Poly (I:C) stimulation. The expression of CgIFNLP and interferon regulatory factors (CgIRF1/8) and the nuclear translocation of CgIRF8 were all suppressed in CgcGAS-RNAi or CgSTING-RNAi oysters after Poly (I:C) stimulation. The expression level of CgSTING and TANK binding kinase1 (CgTBK1) did not decrease in CgcGAS-RNAi oysters. After CgSTING was knocked down, the high expression of CgTBK1 induced by Poly (I:C) was prevented significantly. These results indicated that there was a primitive IFN-like antiviral mechanism dependent on the cGAS/STING-TBK1-IRFs regulatory axis in mollusks, which was different from the classic cGAS-STING-TBK1 signal pathway in mammals.


Asunto(s)
Crassostrea/enzimología , Inmunidad , Factores Reguladores del Interferón/metabolismo , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Crassostrea/efectos de los fármacos , Crassostrea/inmunología , Crassostrea/virología , Virus ADN/inmunología , Interacciones Huésped-Patógeno , Inmunidad/efectos de los fármacos , Factores Reguladores del Interferón/genética , Proteínas de la Membrana/genética , Nucleotidiltransferasas/genética , Poli I-C/farmacología , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal
10.
J Immunol ; 206(11): 2668-2681, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34011520

RESUMEN

The antiviral innate immune responses are crucial steps during host defense and must be strictly regulated, but the molecular mechanisms of control remain unclear. In this study, we report increased expression of human ATPase Na+/K+ transporting subunit ß 1(ATP1B1) after DNA and RNA virus infections. We found that the expression of ATP1B1 can inhibit viral replication and increase the levels of IFNs, IFN-stimulated genes, and inflammatory cytokines. Knockdown of ATP1B1 by specific short hairpin RNA had the opposite effects. Upon viral infection, ATP1B1 was induced, interacted with TRAF3 and TRAF6, and potentiated the ubiquitination of these proteins, leading to increased phosphorylation of downstream molecules, including TGF-ß-activated kinase 1 (TAK1) and TANK-binding kinase 1 (TBK1). These results reveal a previously unrecognized role of ATP1B1 in antiviral innate immunity and suggest a novel mechanism for the induction of IFNs and proinflammatory cytokines during viral infection.


Asunto(s)
Inmunidad Innata/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , ATPasa Intercambiadora de Sodio-Potasio/inmunología , Factor 3 Asociado a Receptor de TNF/inmunología , Regulación hacia Arriba/inmunología , Animales , Células Cultivadas , Chlorocebus aethiops , Infecciones por Virus ADN/inmunología , Virus ADN/inmunología , Humanos , Infecciones por Virus ARN/inmunología , Virus ARN/inmunología , ATPasa Intercambiadora de Sodio-Potasio/genética , Ubiquitinación/inmunología , Replicación Viral
11.
Viruses ; 13(3)2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33801276

RESUMEN

The SAM and HD domain-containing protein 1 (SAMHD1) is a dNTP triphosphohydrolase that plays a crucial role for a variety of different cellular functions. Besides balancing intracellular dNTP concentrations, facilitating DNA damage repair, and dampening excessive immune responses, SAMHD1 has been shown to act as a major restriction factor against various virus species. In addition to its well-described activity against retroviruses such as HIV-1, SAMHD1 has been identified to reduce the infectivity of different DNA viruses such as the herpesviruses CMV and EBV, the poxvirus VACV, or the hepadnavirus HBV. While some viruses are efficiently restricted by SAMHD1, others have developed evasion mechanisms that antagonize the antiviral activity of SAMHD1. Within this review, we summarize the different cellular functions of SAMHD1 and highlight the countermeasures viruses have evolved to neutralize the restriction factor SAMHD1.


Asunto(s)
Infecciones por Virus ADN/inmunología , Interacciones Huésped-Patógeno/inmunología , Infecciones por Retroviridae/inmunología , Proteína 1 que Contiene Dominios SAM y HD/inmunología , Virus ADN/inmunología , Humanos , Retroviridae/inmunología , Interferencia Viral
12.
J Clin Immunol ; 41(5): 1031-1047, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33656624

RESUMEN

PURPOSE: The human antibody repertoire forms in response to infections, the microbiome, vaccinations, and environmental exposures. The specificity of such antibody responses was compared among a cohort of toddlers to identify differences between seropositive versus seronegative responses. METHODS: An assessment of the serum IgM and IgG antibody reactivities in 197 toddlers of 1- and 2-years of age was performed with a microfluidic array containing 110 distinct antigens. Longitudinal profiling was done from years 1 to 2. Seropositivity to RNA and DNA viruses; bacteria; live attenuated, inactive, and subunit vaccines; and autoantigens was compared. A stratification was developed based on quantitative variations in the IgG responses. Clinical presentations and previously known genetic risk alleles for various immune system conditions were investigated in relation to IgG responses. RESULTS: IgG reactivities stratified toddlers into low, moderate, and high responder groups. The high group (17%) had elevated IgG responses to multiple RNA and DNA viruses (e.g., respiratory syncytial virus, Epstein-Barr virus, adenovirus, Coxsackievirus) and this correlated with increased responses to live attenuated viral vaccines and certain autoantigens. This high group was more likely to be associated with gestational diabetes and an older age. Genetic analyses identified polymorphisms in the IL2RB, TNFSF4, and INS genes in two high responder individuals that were associated with their elevated cytokine levels and clinical history of eczema and asthma. CONCLUSION: Serum IgG profiling of toddlers reveals correlations between the magnitude of the antibody responses towards viruses, live attenuated vaccines, and certain autoantigens. A low responder group had much weaker responses overall, including against vaccines. The serum antibody screen also identifies individuals with IgG responses to less common infections (West Nile virus, parvovirus, tuberculosis). The characterization of the antibody responses in combination with the identification of genetic risk alleles provides an opportunity to identify children with increased risk of clinical disease.


Asunto(s)
Anticuerpos Antivirales/sangre , Autoantígenos/inmunología , Bacterias/inmunología , Virus ADN/inmunología , Inmunoglobulina G/sangre , Virus ARN/inmunología , Vacunas/inmunología , Preescolar , Citocinas/sangre , Femenino , Genotipo , Humanos , Inmunoglobulina M/sangre , Lactante , Masculino , Técnicas Analíticas Microfluídicas
13.
Front Immunol ; 11: 613039, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33329609

RESUMEN

DNA viruses are a source of great morbidity and mortality throughout the world by causing many diseases; thus, we need substantial knowledge regarding viral pathogenesis and the host's antiviral immune responses to devise better preventive and therapeutic strategies. The innate immune system utilizes numerous germ-line encoded receptors called pattern-recognition receptors (PRRs) to detect various pathogen-associated molecular patterns (PAMPs) such as viral nucleic acids, ultimately resulting in antiviral immune responses in the form of proinflammatory cytokines and type I interferons. The immune-stimulatory role of DNA is known for a long time; however, DNA sensing ability of the innate immune system was unraveled only recently. At present, multiple DNA sensors have been proposed, and most of them use STING as a key adaptor protein to exert antiviral immune responses. In this review, we aim to provide molecular and structural underpinnings on endosomal DNA sensor Toll-like receptor 9 (TLR9) and multiple cytosolic DNA sensors including cyclic GMP-AMP synthase (cGAS), interferon-gamma inducible 16 (IFI16), absent in melanoma 2 (AIM2), and DNA-dependent activator of IRFs (DAI) to provide new insights on their signaling mechanisms and physiological relevance. We have also addressed less well-understood DNA sensors such as DEAD-box helicase DDX41, RNA polymerase III (RNA pol III), DNA-dependent protein kinase (DNA-PK), and meiotic recombination 11 homolog A (MRE11). By comprehensive understanding of molecular and structural aspects of DNA-sensing antiviral innate immune signaling pathways, potential new targets for viral and autoimmune diseases can be identified.


Asunto(s)
Antivirales/inmunología , ADN Viral/inmunología , Inmunidad Innata/inmunología , Animales , Virus ADN/inmunología , Humanos , Transducción de Señal/inmunología , Receptor Toll-Like 9/inmunología
14.
Proc Natl Acad Sci U S A ; 117(35): 21568-21575, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32817552

RESUMEN

The DNA sensor cGMP-AMP synthase (cGAS) senses cytosolic microbial or self DNA to initiate a MITA/STING-dependent innate immune response. cGAS is regulated by various posttranslational modifications at its C-terminal catalytic domain. Whether and how its N-terminal unstructured domain is regulated by posttranslational modifications remain unknown. We identified the acetyltransferase KAT5 as a positive regulator of cGAS-mediated innate immune signaling. Overexpression of KAT5 potentiated viral-DNA-triggered transcription of downstream antiviral genes, whereas a KAT5 deficiency had the opposite effects. Mice with inactivated Kat5 exhibited lower levels of serum cytokines in response to DNA virus infection, higher viral titers in the brains, and more susceptibility to DNA-virus-induced death. Mechanistically, KAT5 catalyzed acetylation of cGAS at multiple lysine residues in its N-terminal domain, which promoted its DNA-binding ability. Our findings suggest that KAT5-mediated cGAS acetylation at its N terminus is important for efficient innate immune response to DNA virus.


Asunto(s)
Infecciones por Virus ADN/inmunología , Virus ADN/inmunología , Lisina Acetiltransferasa 5/inmunología , Nucleotidiltransferasas/inmunología , Acetilación , Animales , GMP Cíclico/metabolismo , Infecciones por Virus ADN/genética , Infecciones por Virus ADN/metabolismo , Virus ADN/genética , Femenino , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Interferón beta/inmunología , Lisina Acetiltransferasa 5/genética , Lisina Acetiltransferasa 5/metabolismo , Masculino , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Procesamiento Proteico-Postraduccional , Transducción de Señal , Proteínas Virales/metabolismo
15.
Mol Immunol ; 126: 153-164, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32853878

RESUMEN

Oyster production is an economic activity of great interest worldwide. Recently, oysters have been suffering significant mortalities from OsHV-1infection, which has resulted in substantial economic loses in several countries around the world. Understanding viral pathogenicity mechanisms is of central importance for the establishment of disease control measures. Thus, the present work aimed to identify and characterize miRNAs from OsHV-1 as well as to predict their target transcripts in the virus and the host. OsHV-1 genome was used for the in silico discovery of pre-miRNAs. Subsequently, viral and host target transcripts of the OsHV-1 miRNAs were predicted according to the base pairing interaction between mature miRNAs and mRNA 3' untranslated regions (UTRs). Six unique pre-miRNAs were found in different regions of the viral genome, ranging in length from 85 to 172 nucleotides. A complex network of self-regulation of viral gene expression mediated by the miRNAs was identified. These sequences also seem to have a broad ability to regulate the expression of host immune-related genes, especially those associated with pathogen recognition. Our results suggest that OsHV-1 encodes miRNAs with important functions in the infection process, inducing self-regulation of viral transcripts, as well as affecting the regulation of Pacific oyster transcripts related to immunity. Understanding the molecular basis of host-pathogen interactions can help mitigate the recurrent events of oyster mass mortalities by OsHV-1 observed worldwide.


Asunto(s)
Crassostrea/virología , Virus ADN/patogenicidad , Interacciones Huésped-Patógeno/genética , MicroARNs/metabolismo , ARN Viral/metabolismo , Animales , Acuicultura , Biología Computacional , Crassostrea/genética , Crassostrea/inmunología , Virus ADN/genética , Virus ADN/inmunología , Regulación de la Expresión Génica/inmunología , Redes Reguladoras de Genes/genética , Redes Reguladoras de Genes/inmunología , Interacciones Huésped-Patógeno/inmunología , MicroARNs/genética , MicroARNs/aislamiento & purificación , ARN Viral/genética , ARN Viral/aislamiento & purificación
16.
PLoS Pathog ; 16(3): e1008429, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32208449

RESUMEN

Chromatin dynamics regulated by epigenetic modification is crucial in genome stability and gene expression. Various epigenetic mechanisms have been identified in the pathogenesis of human diseases. Here, we examined the effects of ten epigenetic agents on pseudorabies virus (PRV) infection by using GFP-reporter assays. Inhibitors of bromodomain protein 4 (BRD4), which receives much more attention in cancer than viral infection, was found to exhibit substantial anti-viral activity against PRV as well as a range of DNA and RNA viruses. We further demonstrated that BRD4 inhibition boosted a robust innate immune response. BRD4 inhibition also de-compacted chromatin structure and induced the DNA damage response, thereby triggering the activation of cGAS-mediated innate immunity and increasing host resistance to viral infection both in vitro and in vivo. Mechanistically, the inhibitory effect of BRD4 inhibition on viral infection was mainly attributed to the attenuation of viral attachment. Our findings reveal a unique mechanism through which BRD4 inhibition restrains viral infection and points to its potent therapeutic value for viral infectious diseases.


Asunto(s)
Proteínas de Ciclo Celular/inmunología , Daño del ADN/inmunología , Virus ADN/inmunología , Inmunidad Innata , Proteínas Nucleares/inmunología , Virus ARN/inmunología , Factores de Transcripción/inmunología , Células A549 , Animales , Chlorocebus aethiops , Infecciones por Virus ADN/inmunología , Perros , Femenino , Células HEK293 , Células HeLa , Humanos , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Células 3T3 NIH , Células RAW 264.7 , Infecciones por Virus ARN/inmunología , Porcinos , Células Vero
17.
Biochem Biophys Res Commun ; 526(1): 199-205, 2020 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-32201077

RESUMEN

Upon detection of viral DNA, the cytoplasmic DNA sensor cyclic GMP-AMP (cGAMP) synthase (cGAS) utilizes GTP and ATP as substrates to synthesize the second messenger molecule 2'3'cyclic GMP-AMP (cGAMP), which binds to the ER-associated adaptor protein MITA/STING to signal innate antiviral response to DNA virus. How the cGAS-MITA pathways are post-translationally regulated is not fully understood. In this study, we identified the tyrosine kinase CSK as a positive regulator of cGAS-MITA mediated innate antiviral response. CSK-deficiency inhibits DNA virus-triggered induction of downstream antiviral effector genes. Following DNA virus infection, CSK phosphorylates MITA at Y240 and Y245, which is important for its activation. These results suggest that CSK plays a role in modulating innate immune response to DNA virus.


Asunto(s)
Proteína Tirosina Quinasa CSK/metabolismo , Virus ADN/inmunología , Inmunidad Innata , Proteínas de la Membrana/metabolismo , Animales , Proteína Tirosina Quinasa CSK/deficiencia , Línea Celular , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación
18.
Front Immunol ; 11: 26, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32117225

RESUMEN

In recent years, viruses similar to those that cause serious disease in humans and other mammals have been detected in apparently healthy bats. These include filoviruses, paramyxoviruses, and coronaviruses that cause severe diseases such as Ebola virus disease, Marburg haemorrhagic fever and severe acute respiratory syndrome (SARS) in humans. The evolution of flight in bats seem to have selected for a unique set of antiviral immune responses that control virus propagation, while limiting self-damaging inflammatory responses. Here, we summarize our current understanding of antiviral immune responses in bats and discuss their ability to co-exist with emerging viruses that cause serious disease in other mammals. We highlight how this knowledge may help us to predict viral spillovers into new hosts and discuss future directions for the field.


Asunto(s)
Quirópteros/inmunología , Quirópteros/virología , Virus ADN/inmunología , Adaptación al Huésped/inmunología , Sistema Inmunológico/virología , Virus ARN/inmunología , Inmunidad Adaptativa , Animales , Reservorios de Enfermedades/virología , Evolución Molecular , Inmunidad Innata , Interferones/metabolismo , Zoonosis Virales/inmunología , Zoonosis Virales/transmisión
19.
mBio ; 11(2)2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32156821

RESUMEN

Over the last decade, innate immune priming has been evidenced in many invertebrate phyla. If mechanistic models have been proposed, molecular studies aiming to substantiate these models have remained scarce. We reveal here the transcriptional signature associated with immune priming in the oyster Crassostrea gigas Oysters were fully protected against Ostreid herpesvirus 1 (OsHV-1), a major oyster pathogen, after priming with poly(I·C), which mimics viral double-stranded RNA. Global analysis through RNA sequencing of oyster and viral genes after immune priming and viral infection revealed that poly(I·C) induces a strong antiviral response that impairs OsHV-1 replication. Protection is based on a sustained upregulation of immune genes, notably genes involved in the interferon pathway and apoptosis, which control subsequent viral infection. This persistent antiviral alert state remains active over 4 months and supports antiviral protection in the long term. This acquired resistance mechanism reinforces the molecular foundations of the sustained response model of immune priming. It further opens the way to applications (pseudovaccination) to cope with a recurrent disease that causes dramatic economic losses in the shellfish farming industry worldwide.IMPORTANCE In the last decade, important discoveries have shown that resistance to reinfection can be achieved without a functional adaptive immune system, introducing the concept of innate immune memory in invertebrates. However, this field has been constrained by the limited number of molecular mechanisms evidenced to support these phenomena. Taking advantage of an invertebrate species, the Pacific oyster (Crassostrea gigas), in which we evidenced one of the longest and most effective periods of protection against viral infection observed in an invertebrate, we provide the first comprehensive transcriptomic analysis of antiviral innate immune priming. We show that priming with poly(I·C) induced a massive upregulation of immune-related genes, which control subsequent viral infection, and it was maintained for over 4 months after priming. This acquired resistant mechanism reinforces the molecular foundations of the sustained response model of immune priming. It opens the way to pseudovaccination to prevent the recurrent diseases that currently afflict economically or ecologically important invertebrates.


Asunto(s)
Crassostrea/genética , Crassostrea/inmunología , Infecciones por Virus ADN/inmunología , Virus ADN/inmunología , Inmunidad Innata , Animales , Infecciones por Virus ADN/genética , Virus ADN/patogenicidad , Perfilación de la Expresión Génica , Poli I-C/inmunología , Regulación hacia Arriba
20.
Fish Shellfish Immunol ; 100: 317-323, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32173450

RESUMEN

Largemouth bass ulcerative syndrome virus (LBUSV) is an important virus induce the mortality of largemouth bass (Micropterus Salmoides). In this study, we reported a single-walled carbon nanotubes (SWCNTs) containing LBUSV major capsid protein (MCP) subunit vaccine (SWCNTs-MCP) which was evaluated for its protective effect on largemouth bass by immersion immunization. We found an elevation in serum antibody levels, enzyme activities, complement C3 content and immune-related genes (IgM, TGF-ß, IL-1ß, IL-8, TNF-α and CD4) expression, in the SWCNTs-MCP immunized groups compared with the pure MCP group. The survival rates for control group, pure MCP protein groups (40 mg L-1) and four SWCNTs-MCP groups (5 mg L-1, 10 mg L-1, 20 mg L-1 and 40 mg L-1) were 0%, 27.78%, 30.56%, 50.00%, 66.67% and 80.56%, respectively. The results suggests that with the assistance of SWCNTs, the immune protection of the SWCNTs-MCP group (40 mg L-1) increased up 52.78%-80.1% compared with pure MCP group (40 mg L-1). Our results demonstrate that the single-walled carbon nanotube subunit vaccine can be used as a new immunization method against LBUSV showing protection following challenge with LBUSV. Taken together, our results demonstrate that the single-walled carbon nanotube subunit vaccine can be used as a new method against LBUSV and have a high immune protection during the largemouth bass farm.


Asunto(s)
Lubina/inmunología , Infecciones por Virus ADN/veterinaria , Virus ADN/inmunología , Enfermedades de los Peces/prevención & control , Inmunización/veterinaria , Nanotubos de Carbono/química , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Lubina/virología , Infecciones por Virus ADN/prevención & control , Enfermedades de los Peces/virología , Inmersión , Inmunización/métodos , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Vacunas Virales/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...