Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 142
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 , Degrons , 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.
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
3.
Viruses ; 13(7)2021 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-34199077

RESUMO

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.


Assuntos
Vírus de DNA/genética , Vírus de DNA/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Evasão da Resposta Imune , Vírus de RNA/genética , Antivirais/farmacologia , Códon de Terminação , Vírus de DNA/efeitos dos fármacos , Mudança da Fase de Leitura do Gene Ribossômico , Antígenos de Histocompatibilidade Classe I/genética , Conformação de Ácido Nucleico , Peptídeos/imunologia , Biossíntese de Proteínas , Vírus de RNA/efeitos dos fármacos , Vírus de RNA/imunologia
4.
J Immunol ; 206(11): 2668-2681, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34011520

RESUMO

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.


Assuntos
Imunidade Inata/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , ATPase Trocadora de Sódio-Potássio/imunologia , Fator 3 Associado a Receptor de TNF/imunologia , Regulação para Cima/imunologia , Animais , Células Cultivadas , Chlorocebus aethiops , Infecções por Vírus de DNA/imunologia , Vírus de DNA/imunologia , Humanos , Infecções por Vírus de RNA/imunologia , Vírus de RNA/imunologia , ATPase Trocadora de Sódio-Potássio/genética , Ubiquitinação/imunologia , Replicação Viral
5.
Viruses ; 13(3)2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33801276

RESUMO

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.


Assuntos
Infecções por Vírus de DNA/imunologia , Interações Hospedeiro-Patógeno/imunologia , Infecções por Retroviridae/imunologia , Proteína 1 com Domínio SAM e Domínio HD/imunologia , Vírus de DNA/imunologia , Humanos , Retroviridae/imunologia , Interferência Viral
6.
J Clin Immunol ; 41(5): 1031-1047, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33656624

RESUMO

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.


Assuntos
Anticorpos Antivirais/sangue , Autoantígenos/imunologia , Bactérias/imunologia , Vírus de DNA/imunologia , Imunoglobulina G/sangue , Vírus de RNA/imunologia , Vacinas/imunologia , Pré-Escolar , Citocinas/sangue , Feminino , Genótipo , Humanos , Imunoglobulina M/sangue , Lactente , Masculino , Técnicas Analíticas Microfluídicas
7.
Front Immunol ; 11: 613039, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33329609

RESUMO

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.


Assuntos
Antivirais/imunologia , DNA Viral/imunologia , Imunidade Inata/imunologia , Animais , Vírus de DNA/imunologia , Humanos , Transdução de Sinais/imunologia , Receptor Toll-Like 9/imunologia
8.
PLoS Pathog ; 16(3): e1008429, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32208449

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/imunologia , Dano ao DNA/imunologia , Vírus de DNA/imunologia , Imunidade Inata , Proteínas Nucleares/imunologia , Vírus de RNA/imunologia , Fatores de Transcrição/imunologia , Células A549 , Animais , Chlorocebus aethiops , Infecções por Vírus de DNA/imunologia , Cães , Feminino , Células HEK293 , Células HeLa , Humanos , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Células NIH 3T3 , Células RAW 264.7 , Infecções por Vírus de RNA/imunologia , Suínos , Células Vero
9.
PLoS Pathog ; 16(1): e1008178, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31968013

RESUMO

Mediator of IRF3 activation (MITA, also known as stimulator of interferon genes, STING) senses the second messenger cyclic GMP-AMP (cGAMP) which is synthesized upon DNA virus infection and activates innate antiviral immune response. It has been demonstrated that the activity of MITA is delicately regulated by various post-translational modifications including polyubiquitination. In this study, we identified the deubiquitinating enzyme USP44 as a positive regulator of MITA. USP44 is recruited to MITA following DNA virus infection and removes K48-linked polyubiquitin moieties from MITA at K236, therefore prevents MITA from proteasome mediated degradation. USP44-deficiency results in acceleration of HSV-1-induced degradation of MITA and reduced induction of type I interferons (IFNs) and proinflammatory cytokines. Consistently, Usp44-/- mice are more susceptible to HSV-1 infection as indicated by higher tissue viral titers, greater tissue damage and lower survival rate. These findings suggest that USP44 plays a specific and critical role in the regulation of innate immune response against DNA viruses.


Assuntos
Vírus de DNA/imunologia , Imunidade Inata , Proteínas de Membrana/metabolismo , Ubiquitina Tiolesterase/metabolismo , Animais , Enzima Desubiquitinante CYLD/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Estabilidade Proteica , Transdução de Sinais , Ubiquitinação
10.
Front Immunol ; 11: 624556, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33505405

RESUMO

IFI16, hnRNPA2B1, and nuclear cGAS are nuclear-located DNA sensors that play important roles in initiating host antiviral immunity and modulating tumorigenesis. IFI16 triggers innate antiviral immunity, inflammasome, and suppresses tumorigenesis by recognizing double-stranded DNA (dsDNA), single-stranded DNA (ssDNA), damaged nuclear DNA, or cooperatively interacting with multiple tumor suppressors such as p53 and BRCA1. hnRNPA2B1 initiates interferon (IFN)-α/ß production and enhances STING-dependent cytosolic antiviral signaling by directly binding viral dsDNA from invaded viruses and facilitating N6 -methyladenosine (m6A) modification of cGAS, IFI16, and STING mRNAs. Nuclear cGAS is recruited to double-stranded breaks (DSBs), suppresses DNA repair, and promotes tumorigenesis. This review briefly describes the nuclear functions of IFI16, hnRNPA2B1, and cGAS, and summarizes the transcriptional, post-transcriptional, and post-translational regulation of these nuclear DNA sensors.


Assuntos
Transformação Celular Viral/imunologia , Infecções por Vírus de DNA/imunologia , Vírus de DNA/imunologia , DNA Viral/imunologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/imunologia , Proteínas Nucleares/imunologia , Nucleotidiltransferases/imunologia , Fosfoproteínas/imunologia , Infecções por Vírus de DNA/genética , Infecções por Vírus de DNA/patologia , Vírus de DNA/genética , DNA Viral/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/genética , Humanos , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon beta/genética , Interferon beta/imunologia , Proteínas Nucleares/genética , Nucleotidiltransferases/genética , Fosfoproteínas/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/imunologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/imunologia
11.
Viruses ; 11(9)2019 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-31450758

RESUMO

Autophagy is a catabolic biological process in the body. By targeting exogenous microorganisms and aged intracellular proteins and organelles and sending them to the lysosome for phagocytosis and degradation, autophagy contributes to energy recycling. When cells are stimulated by exogenous pathogenic microorganisms such as viruses, activation or inhibition of autophagy is often triggered. As autophagy has antiviral effects, many viruses may escape and resist the process by encoding viral proteins. At the same time, viruses can also use autophagy to enhance their replication or increase the persistence of latent infections. Here, we give a brief overview of autophagy and DNA viruses and comprehensively review the known interactions between human and animal DNA viruses and autophagy and the role and mechanisms of autophagy in viral DNA replication and DNA virus-induced innate and acquired immunity.


Assuntos
Autofagia/fisiologia , Vírus de DNA , Imunidade Adaptativa , Adenoviridae/crescimento & desenvolvimento , Adenoviridae/imunologia , Adenoviridae/metabolismo , Animais , Autofagossomos/metabolismo , Vírus de DNA/crescimento & desenvolvimento , Vírus de DNA/imunologia , Vírus de DNA/metabolismo , Herpesviridae/crescimento & desenvolvimento , Herpesviridae/imunologia , Herpesviridae/metabolismo , Interações entre Hospedeiro e Microrganismos , Humanos , Evasão da Resposta Imune , Imunidade Inata , Lisossomos/metabolismo , Papillomaviridae/crescimento & desenvolvimento , Papillomaviridae/imunologia , Papillomaviridae/metabolismo , Fagocitose/fisiologia , Transdução de Sinais , Proteínas Virais/imunologia , Proteínas Virais/metabolismo , Replicação Viral/imunologia
12.
Metallomics ; 11(4): 822-832, 2019 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-30843573

RESUMO

The mass mortality of molluscs caused by OsHV-1 infection has frequently occurred worldwide in recent years. Meanwhile the interaction between OsHV-1 and its host is largely unknown. Innate immunity mainly makes up the mollusc defense system, due to the lack of adaptive immunity in invertebrates. The iron limitation strategy is an indispensable facet of innate immunity across vertebrate and invertebrate species. In this study, an iron limitation strategy was interestingly found to contribute to mollusc innate immune responses against OsHV-1 infection. Firstly, ark clams, Scapharca broughtonii, were experimentally infected with OsHV-1, and serious hyperaemia in hepatopancreases and the erosion of gills were observed post OsHV-1 infection according to a histology assay. Meanwhile, based on quantification and Prussian blue staining, the process of iron efflux from ark clams was described post OsHV-1 infection. Secondly, ferritin, as an important iron storage protein, was characterized in ark clams and showed significant iron binding activity. According to the results of an immunohistochemistry assay, ferritin was supposed to be responsible for the iron translocation in ark clams post OsHV-1 infection. Its expression level was significantly fluctuant in response to OsHV-1 infection. Finally, oxidative stress was assessed by the analyses of H2O2 content, total antioxidant capacity and MDA level post OsHV-1 infection. Supplementary iron was found to promote ROS generation and death of hemocytes in vivo. These results highlighted that microenvironment changes in the essential nutrient iron should be an important aspect of the pathogenesis of OsHV-1 disease.


Assuntos
Infecções por Vírus de DNA/veterinária , Vírus de DNA/imunologia , Ferro/imunologia , Scapharca/imunologia , Scapharca/virologia , Animais , Infecções por Vírus de DNA/imunologia , Infecções por Vírus de DNA/virologia , Vírus de DNA/fisiologia , Interações Hospedeiro-Patógeno , Imunidade Inata
13.
Arch Immunol Ther Exp (Warsz) ; 67(1): 41-48, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30196473

RESUMO

Nuclear factor (NF)-κB is a major regulator of antiviral response. Viral pathogens exploit NF-κB activation pathways to avoid cellular mechanisms that eliminate the infection. Canonical (classical) NF-κB signaling, which regulates innate immune response, cell survival and inflammation, is often manipulated by viral pathogens that can counteract antiviral response. Oncogenic viruses can modulate not only canonical, but also non-canonical (alternative) NF-κB activation pathways. The non-canonical NF-κB signaling is responsible for adaptive immunity and plays a role in lymphoid organogenesis, B cell development, as well as bone metabolism. Thus, non-canonical NF-κB activation has been linked to lymphoid malignancies. However, some data strongly suggest that the non-canonical NF-κB activation pathway may also function in innate immunity and is modulated by certain non-oncogenic viruses. Collectively, these findings show the importance of studying the impact of different groups of viral pathogens on alternative NF-κB activation. This mini-review focuses on the influence of non-oncogenic viruses on the components of non-canonical NF-κB signaling.


Assuntos
Vírus de DNA/patogenicidade , NF-kappa B/metabolismo , Vírus de RNA/patogenicidade , Viroses/virologia , Imunidade Adaptativa , Animais , Vírus de DNA/imunologia , Vírus de DNA/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , NF-kappa B/imunologia , Vírus de RNA/imunologia , Vírus de RNA/metabolismo , Transdução de Sinais , Viroses/imunologia , Viroses/metabolismo
14.
Nat Commun ; 8(1): 945, 2017 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-29038422

RESUMO

Many double-stranded DNA viruses, such as Epstein-Barr virus, can establish persistent infection, but the underlying virus-host interactions remain poorly understood. Here we report that in human airway epithelial cells Epstein-Barr virus induces TRIM29, a member of the TRIM family of proteins, to inhibit innate immune activation. Knockdown of TRIM29 in airway epithelial cells enhances type I interferon production, and in human nasopharyngeal carcinoma cells results in almost complete Epstein-Barr virus clearance. TRIM29 is also highly induced by cytosolic double-stranded DNA in myeloid dendritic cells. TRIM29 -/- mice have lower adenovirus titers in the lung, and are resistant to lethal herpes simplex virus-1 infection due to enhanced production of type I interferon. Mechanistically, TRIM29 induces K48-linked ubiquitination of Stimulator of interferon genes, a key adaptor in double-stranded DNA-sensing pathway, followed by its rapid degradation. These data demonstrate that Epstein-Barr virus and possible other double-stranded DNA viruses use TRIM29 to suppress local innate immunity, leading to the persistence of DNA virus infections.Proteins of the TRIM family have regulatory functions in immune signaling, often via ubiquitination of target proteins. Here, the authors show that TRIM29 is induced upon infection with DNA viruses, resulting in degradation of STING, decreased interferon signaling and increased pathogenicity in mice.


Assuntos
Proteínas de Ligação a DNA/imunologia , Infecções por Vírus Epstein-Barr/imunologia , Herpesvirus Humano 4/imunologia , Imunidade Inata/imunologia , Fatores de Transcrição/imunologia , Animais , Linhagem Celular , Infecções por Vírus de DNA/imunologia , Infecções por Vírus de DNA/metabolismo , Infecções por Vírus de DNA/virologia , Vírus de DNA/imunologia , Vírus de DNA/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Infecções por Vírus Epstein-Barr/virologia , Inativação Gênica , Herpesvirus Humano 4/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata/genética , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Camundongos Knockout , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/virologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
15.
J Clin Oncol ; 35(31): 3547-3557, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28783452

RESUMO

Purpose Improvement of cure rates for patients treated with allogeneic hematopoietic stem-cell transplantation (HSCT) will require efforts to decrease treatment-related mortality from severe viral infections. Adoptively transferred virus-specific T cells (VSTs) generated from eligible, third-party donors could provide broad antiviral protection to recipients of HSCT as an immediately available off-the-shelf product. Patient and Methods We generated a bank of VSTs that recognized five common viral pathogens: Epstein-Barr virus (EBV), adenovirus (AdV), cytomegalovirus (CMV), BK virus (BKV), and human herpesvirus 6 (HHV-6). The VSTs were administered to 38 patients with 45 infections in a phase II clinical trial. Results A single infusion produced a cumulative complete or partial response rate of 92% (95% CI, 78.1% to 98.3%) overall and the following rates by virus: 100% for BKV (n = 16), 94% for CMV (n = 17), 71% for AdV (n = 7), 100% for EBV (n = 2), and 67% for HHV-6 (n = 3). Clinical benefit was achieved in 31 patients treated for one infection and in seven patients treated for multiple coincident infections. Thirteen of 14 patients treated for BKV-associated hemorrhagic cystitis experienced complete resolution of gross hematuria by week 6. Infusions were safe, and only two occurrences of de novo graft-versus host disease (grade 1) were observed. VST tracking by epitope profiling revealed persistence of functional VSTs of third-party origin for up to 12 weeks. Conclusion The use of banked VSTs is a feasible, safe, and effective approach to treat severe and drug-refractory infections after HSCT, including infections from two viruses (BKV and HHV-6) that had never been targeted previously with an off-the-shelf product. Furthermore, the multispecificity of the VSTs ensures extensive antiviral coverage, which facilitates the treatment of patients with multiple infections.


Assuntos
Infecções por Vírus de DNA/terapia , Vírus de DNA/imunologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/métodos , Imunoterapia Adotiva/métodos , Linfócitos T/imunologia , Linfócitos T/transplante , Adenovírus Humanos/imunologia , Adulto , Vírus BK/imunologia , Infecções por Vírus de DNA/etiologia , Infecções por Vírus de DNA/virologia , Feminino , Herpesvirus Humano 4/imunologia , Herpesvirus Humano 6/imunologia , Humanos , Masculino , Transplante Homólogo , Resultado do Tratamento
16.
Dev Comp Immunol ; 76: 334-342, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28684273

RESUMO

The recognition of pathogenic DNA is important to the initiation of antiviral responses. Here, we report the identification of the first avian DEAD (Asp-Glu-Ala-Asp) box polypeptide 41 (DDX41), an important DNA sensor, in chicken cells. In our study, we confirmed that chDDX41 is not an interferon-inducible gene. Knockdown of chDDX41 expression by shRNA blocked the ability of DF-1 cells to mount an IFN-ß response to DNA and associated viruses. ChDDX41 mRNAs could be upregulated by double-stranded DNA (dsDNA) analogue poly(dA:dT), but not by double-stranded RNA (dsRNA) analogue poly(I:C). In poly(dA:dT) stimulation assays, the immune molecules involved in the DDX41-mediated IFN-ß pathway in human cells were universally upregulated in chicken cells. Via coimmunoprecipitation (Co-IP) experiments, we found that chDDX41 could strongly interact with chicken stimulator of IFN genes (chSTING). Therefore, our results suggest that chDDX41 is involved in the dsDNA- and dsDNA virus-mediated chDDX41-chSTING-IFN-ß signaling pathway in chicken cells.


Assuntos
Proteínas Aviárias/metabolismo , RNA Helicases DEAD-box/metabolismo , Vírus de DNA/imunologia , Fibroblastos/metabolismo , Viroses/imunologia , Animais , Proteínas Aviárias/genética , Linhagem Celular , Galinhas , Clonagem Molecular , RNA Helicases DEAD-box/genética , Humanos , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Interferon gama/metabolismo , RNA Interferente Pequeno/genética , Transdução de Sinais
17.
Eur J Immunol ; 47(5): 780-796, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28383780

RESUMO

Pathogens have developed a plethora of strategies to undermine host immune defenses in order to guarantee their survival. For large DNA viruses, these immune evasion mechanisms frequently rely on the expression of genes acquired from host genomes. Horizontally transferred genes include members of the immunoglobulin superfamily, whose products constitute the most diverse group of proteins of vertebrate genomes. Their promiscuous immunoglobulin domains, which comprise the building blocks of these molecules, are involved in a large variety of functions mediated by ligand-binding interactions. The flexible structural nature of the immunoglobulin domains makes them appealing targets for viral capture due to their capacity to generate high functional diversity. Here, we present an up-to-date review of immunoglobulin superfamily gene homologs encoded by herpesviruses, poxviruses, and adenoviruses, that include CD200, CD47, Fc receptors, interleukin-1 receptor 2, interleukin-18 binding protein, CD80, carcinoembryonic antigen-related cell adhesion molecules, and signaling lymphocyte activation molecules. We discuss their distinct structural attributes, binding properties, and functions, shaped by evolutionary pressures to disarm specific immune pathways. We include several novel genes identified from extensive genome database surveys. An understanding of the properties and modes of action of these viral proteins may guide the development of novel immune-modulatory therapeutic tools.


Assuntos
Vírus de DNA/genética , Vírus de DNA/patogenicidade , Evasão da Resposta Imune , Imunoglobulinas/imunologia , Proteínas Virais/imunologia , Adenovírus Humanos/genética , Adenovírus Humanos/imunologia , Adenovírus Humanos/patogenicidade , Animais , Antígenos CD/imunologia , Vírus de DNA/imunologia , Evolução Molecular , Transferência Genética Horizontal , Genes de Imunoglobulinas , Herpesviridae/genética , Herpesviridae/imunologia , Herpesviridae/patogenicidade , Humanos , Imunoglobulinas/genética , Imunoglobulinas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Proteínas Virais/genética
18.
Viruses ; 8(10)2016 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-27782081

RESUMO

In recent years, it has been suggested that host cells exert intrinsic mechanisms to control nuclear replicating DNA viruses. This cellular response involves nuclear antiviral factors targeting incoming viral genomes. Herpes simplex virus-1 (HSV-1) is the best-studied model in this context, and it was shown that upon nuclear entry HSV-1 genomes are immediately targeted by components of promyelocytic leukemia nuclear bodies (PML-NBs) and the nuclear DNA sensor IFI16 (interferon gamma inducible protein 16). Based on HSV-1 studies, together with limited examples in other viral systems, these phenomena are widely believed to be a common cellular response to incoming viral genomes, although formal evidence for each virus is lacking. Indeed, recent studies suggest that the case may be different for adenovirus infection. Here we summarize the existing experimental evidence for the roles of nuclear antiviral factors against incoming viral genomes to better understand cellular responses on a virus-by-virus basis. We emphasize that cells seem to respond differently to different incoming viral genomes and discuss possible arguments for and against a unifying cellular mechanism targeting the incoming genomes of different virus families.


Assuntos
Núcleo Celular/imunologia , Núcleo Celular/virologia , Vírus de DNA/imunologia , Vírus de DNA/fisiologia , Interações Hospedeiro-Patógeno , Imunidade Inata , Animais , Vírus de DNA/genética , Humanos
19.
Immunity ; 45(3): 555-569, 2016 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-27637147

RESUMO

During viral infection, sensing of cytosolic DNA by the cyclic GMP-AMP synthase (cGAS) activates the adaptor protein STING and triggers an antiviral response. Little is known about the mechanisms that determine the kinetics of activation and deactivation of the cGAS-STING pathway, ensuring effective but controlled innate antiviral responses. Here we found that the ubiquitin ligase Trim38 targets cGas for sumoylation in uninfected cells and during the early phase of viral infection. Sumoylation of cGas prevented its polyubiquitination and degradation. Trim38 also sumoylated Sting during the early phase of viral infection, promoting both Sting activation and protein stability. In the late phase of infection, cGas and Sting were desumoylated by Senp2 and subsequently degraded via proteasomal and chaperone-mediated autophagy pathways, respectively. Our findings reveal an essential role for Trim38 in the innate immune response to DNA virus and provide insight into the mechanisms that ensure optimal activation and deactivation of the cGAS-STING pathway.


Assuntos
Vírus de DNA/imunologia , DNA/metabolismo , Nucleotídeos Cíclicos/metabolismo , Nucleotidiltransferases/metabolismo , Sumoilação/fisiologia , Viroses/metabolismo , Animais , Proteínas de Transporte/metabolismo , Cisteína Endopeptidases/metabolismo , Imunidade Inata/imunologia , Cinética , Proteínas de Membrana/metabolismo , Camundongos , Complexo de Endopeptidases do Proteassoma/metabolismo , Transdução de Sinais/imunologia , Transdução de Sinais/fisiologia , Sumoilação/imunologia , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases , Ubiquitinação/imunologia , Ubiquitinação/fisiologia
20.
Curr Opin Microbiol ; 32: 113-119, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27288760

RESUMO

Upon infection, both DNA and RNA viruses can be sensed by pattern recognition receptors (PRRs) in the cytoplasm or the nucleus to activate antiviral innate immunity. Sensing of viral products leads to the activation of a signaling cascade that ultimately results in transcriptional activation of type I and III interferons, as well as other antiviral genes that together mediate viral clearance and inhibit viral spread. Therefore, in order for viruses to replicate and spread efficiently, they must inhibit the host signaling pathways that induce the innate antiviral immune response. In this review, we will highlight recent advances in the understanding of the mechanisms by which viruses evade PRR detection, intermediate signaling molecule activation, transcription factor activation, and the actions of antiviral proteins.


Assuntos
Vírus de DNA/imunologia , Evasão da Resposta Imune/fisiologia , Imunidade Inata/imunologia , Interferon Tipo I/imunologia , Vírus de RNA/imunologia , Receptores de Reconhecimento de Padrão/imunologia , Proteína DEAD-box 58/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Receptores Imunológicos , Transdução de Sinais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA