Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Sci Immunol ; 9(97): eadp1139, 2024 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-39058762

RESUMO

Type I and III interferons (IFNs) are robustly induced during infections and protect cells against viral infection. Both type I and III IFNs are also produced at low levels in the thymus at steady state; however, their role in T cell development and immune tolerance is unclear. Here, we found that both type I and III IFNs were constitutively produced by a very small number of AIRE+ murine thymic epithelial cells, independent of microbial stimulation. Antigen-presenting cells were highly responsive to thymic IFNs, and IFNs were required for the activation and maturation of thymic type 1 conventional dendritic cells, macrophages, and B cells. Loss of IFN sensing led to reduced regulatory T cell selection, reduced T cell receptor (TCR) repertoire diversity, and enhanced autoreactive T cell responses to self-antigens expressed during peripheral IFN signaling. Thus, constitutive exposure to IFNs in the thymus is required for generating a tolerant and diverse TCR repertoire.


Assuntos
Interferons , Camundongos Endogâmicos C57BL , Timo , Animais , Timo/imunologia , Camundongos , Interferons/imunologia , Camundongos Knockout , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia
2.
Immunity ; 57(4): 718-730, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38599167

RESUMO

The cGAS-STING intracellular DNA-sensing pathway has emerged as a key element of innate antiviral immunity and a promising therapeutic target. The existence of an innate immune sensor that can be activated by any double-stranded DNA (dsDNA) of any origin raises fundamental questions about how cGAS is regulated and how it responds to "foreign" DNA while maintaining tolerance to ubiquitous self-DNA. In this review, we summarize recent evidence implicating important roles for cGAS in the detection of foreign and self-DNA. We describe two recent and surprising insights into cGAS-STING biology: that cGAS is tightly tethered to the nucleosome and that the cGAMP product of cGAS is an immunotransmitter acting at a distance to control innate immunity. We consider how these advances influence our understanding of the emerging roles of cGAS in the DNA damage response (DDR), senescence, aging, and cancer biology. Finally, we describe emerging approaches to harness cGAS-STING biology for therapeutic benefit.


Assuntos
Nucleotidiltransferases , Transdução de Sinais , Nucleotidiltransferases/metabolismo , Imunidade Inata , DNA
3.
Cell ; 186(25): 5536-5553.e22, 2023 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-38029747

RESUMO

Mycobacterium tuberculosis (Mtb) causes 1.6 million deaths annually. Active tuberculosis correlates with a neutrophil-driven type I interferon (IFN) signature, but the cellular mechanisms underlying tuberculosis pathogenesis remain poorly understood. We found that interstitial macrophages (IMs) and plasmacytoid dendritic cells (pDCs) are dominant producers of type I IFN during Mtb infection in mice and non-human primates, and pDCs localize near human Mtb granulomas. Depletion of pDCs reduces Mtb burdens, implicating pDCs in tuberculosis pathogenesis. During IFN-driven disease, we observe abundant DNA-containing neutrophil extracellular traps (NETs) described to activate pDCs. Cell-type-specific disruption of the type I IFN receptor suggests that IFNs act on IMs to inhibit Mtb control. Single-cell RNA sequencing (scRNA-seq) indicates that type I IFN-responsive cells are defective in their response to IFNγ, a cytokine critical for Mtb control. We propose that pDC-derived type I IFNs act on IMs to permit bacterial replication, driving further neutrophil recruitment and active tuberculosis disease.


Assuntos
Interferon Tipo I , Tuberculose , Humanos , Camundongos , Animais , Macrófagos/microbiologia , Citocinas , Neutrófilos , Células Dendríticas
4.
Nat Struct Mol Biol ; 30(1): 72-80, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36593311

RESUMO

Cyclic GMP-AMP synthase (cGAS) is a pattern recognition receptor critical for the innate immune response to intracellular pathogens, DNA damage, tumorigenesis and senescence. Binding to double-stranded DNA (dsDNA) induces conformational changes in cGAS that activate the enzyme to produce 2'-3' cyclic GMP-AMP (cGAMP), a second messenger that initiates a potent interferon (IFN) response through its receptor, STING. Here, we combined two-state computational design with informatics-guided design to create constitutively active, dsDNA ligand-independent cGAS (CA-cGAS). We identified CA-cGAS mutants with IFN-stimulating activity approaching that of dsDNA-stimulated wild-type cGAS. DNA-independent adoption of the active conformation was directly confirmed by X-ray crystallography. In vivo expression of CA-cGAS in tumor cells resulted in STING-dependent tumor regression, demonstrating that the designed proteins have therapeutically relevant biological activity. Our work provides a general framework for stabilizing active conformations of enzymes and provides CA-cGAS variants that could be useful as genetically encoded adjuvants and tools for understanding inflammatory diseases.


Assuntos
Imunidade Inata , Nucleotidiltransferases , Nucleotidiltransferases/metabolismo , DNA/química
5.
Immunity ; 55(10): 1799-1812.e4, 2022 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-36070769

RESUMO

The DNA sensor cyclic GMP-AMP synthase (cGAS) is important for antiviral and anti-tumor immunity. cGAS generates cyclic GMP-AMP (cGAMP), a diffusible cyclic dinucleotide that activates the antiviral response through the adaptor protein stimulator of interferon genes (STING). cGAMP cannot passively cross cell membranes, but recent advances have established a role for extracellular cGAMP as an "immunotransmitter" that can be imported into cells. However, the mechanism by which cGAMP exits cells remains unknown. Here, we identifed ABCC1 as a direct, ATP-dependent cGAMP exporter in mouse and human cells. We show that ABCC1 overexpression enhanced cGAMP export and limited STING signaling and that loss of ABCC1 reduced cGAMP export and potentiated STING signaling. We demonstrate that ABCC1 deficiency exacerbated cGAS-dependent autoimmunity in the Trex1-/- mouse model of Aicardi-Goutières syndrome. Thus, ABCC1-mediated cGAMP export is a key regulatory mechanism that limits cell-intrinsic activation of STING and ameliorates STING-dependent autoimmune disease.


Assuntos
Proteínas de Membrana Transportadoras , Proteínas Associadas à Resistência a Múltiplos Medicamentos , Nucleotídeos Cíclicos , Trifosfato de Adenosina , Animais , DNA/metabolismo , Humanos , Interferons/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Nucleotídeos Cíclicos/metabolismo , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo
6.
Nature ; 607(7920): 769-775, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35859177

RESUMO

The RNA-editing enzyme ADAR1 is essential for the suppression of innate immune activation and pathology caused by aberrant recognition of self-RNA, a role it carries out by disrupting the duplex structure of endogenous double-stranded RNA species1,2. A point mutation in the sequence encoding the Z-DNA-binding domain (ZBD) of ADAR1 is associated with severe autoinflammatory disease3-5. ZBP1 is the only other ZBD-containing mammalian protein6, and its activation can trigger both cell death and transcriptional responses through the kinases RIPK1 and RIPK3, and the protease caspase 8 (refs. 7-9). Here we show that the pathology caused by alteration of the ZBD of ADAR1 is driven by activation of ZBP1. We found that ablation of ZBP1 fully rescued the overt pathology caused by ADAR1 alteration, without fully reversing the underlying inflammatory program caused by this alteration. Whereas loss of RIPK3 partially phenocopied the protective effects of ZBP1 ablation, combined deletion of caspase 8 and RIPK3, or of caspase 8 and MLKL, unexpectedly exacerbated the pathogenic effects of ADAR1 alteration. These findings indicate that ADAR1 is a negative regulator of sterile ZBP1 activation, and that ZBP1-dependent signalling underlies the autoinflammatory pathology caused by alteration of ADAR1.


Assuntos
Adenosina Desaminase , Doenças do Sistema Imunitário , Inflamação , Mutação , Proteínas de Ligação a RNA , Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo , Animais , Caspase 8/genética , Caspase 8/metabolismo , Morte Celular , Deleção de Genes , Doenças do Sistema Imunitário/genética , Doenças do Sistema Imunitário/metabolismo , Doenças do Sistema Imunitário/patologia , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Mamíferos/genética , Proteínas Quinases/deficiência , Proteínas Quinases/genética , RNA de Cadeia Dupla/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/deficiência , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Transdução de Sinais
7.
Nat Rev Immunol ; 22(8): 471-483, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-34671122

RESUMO

As brutally demonstrated by the COVID-19 pandemic, an effective immune system is essential for survival. Developed over evolutionary time, viral nucleic acid detection is a central pillar in the defensive armamentarium used to combat foreign microbial invasion. To ensure cellular homeostasis, such a strategy necessitates the efficient discrimination of pathogen-derived DNA and RNA from that of the host. In 2011, it was suggested that an upregulation of type I interferon signalling might serve as a defining feature of a novel set of Mendelian inborn errors of immunity, where antiviral sensors are triggered by host nucleic acids due to a failure of self versus non-self discrimination. These rare disorders have played a surprisingly significant role in informing our understanding of innate immunity and the relevance of type I interferon signalling for human health and disease. Here we consider what we have learned in this time, and how the field may develop in the future.


Assuntos
COVID-19 , Interferon Tipo I , Ácidos Nucleicos , Humanos , Imunidade Inata , Interferon Tipo I/genética , Pandemias
8.
Elife ; 102021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34342578

RESUMO

Many host RNA sensors are positioned in the cytosol to detect viral RNA during infection. However, most positive-strand RNA viruses replicate within a modified organelle co-opted from intracellular membranes of the endomembrane system, which shields viral products from cellular innate immune sensors. Targeting innate RNA sensors to the endomembrane system may enhance their ability to sense RNA generated by viruses that use these compartments for replication. Here, we reveal that an isoform of oligoadenylate synthetase 1, OAS1 p46, is prenylated and targeted to the endomembrane system. Membrane localization of OAS1 p46 confers enhanced access to viral replication sites and results in increased antiviral activity against a subset of RNA viruses including flaviviruses, picornaviruses, and SARS-CoV-2. Finally, our human genetic analysis shows that the OAS1 splice-site SNP responsible for production of the OAS1 p46 isoform correlates with protection from severe COVID-19. This study highlights the importance of endomembrane targeting for the antiviral specificity of OAS1 and suggests that early control of SARS-CoV-2 replication through OAS1 p46 is an important determinant of COVID-19 severity.


Assuntos
2',5'-Oligoadenilato Sintetase/metabolismo , COVID-19/virologia , SARS-CoV-2/metabolismo , Animais , COVID-19/imunologia , Sistemas CRISPR-Cas , Linhagem Celular , Edição de Genes , Humanos , Polimorfismo de Nucleotídeo Único , SARS-CoV-2/isolamento & purificação
9.
Immunity ; 54(9): 1948-1960.e5, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34343497

RESUMO

The RNA deaminase ADAR1 is an essential negative regulator of the RNA sensor MDA5, and loss of ADAR1 function triggers inappropriate activation of MDA5 by self-RNAs. Mutations in ADAR, the gene that encodes ADAR1, cause human immune diseases, including Aicardi-Goutières syndrome (AGS). However, the mechanisms of MDA5-dependent disease pathogenesis in vivo remain unknown. Here we generated mice with a single amino acid change in ADAR1 that models the most common human ADAR AGS mutation. These Adar mutant mice developed lethal disease that required MDA5, the RIG-I-like receptor LGP2, type I interferons, and the eIF2α kinase PKR. A small-molecule inhibitor of the integrated stress response (ISR) that acts downstream of eIF2α phosphorylation prevented immunopathology and rescued the mice from mortality. These findings place PKR and the ISR as central components of immunopathology in vivo and identify therapeutic targets for treatment of human diseases associated with the ADAR1-MDA5 axis.


Assuntos
Adenosina Desaminase/metabolismo , Doenças Autoimunes do Sistema Nervoso/patologia , Malformações do Sistema Nervoso/patologia , Estresse Fisiológico/fisiologia , eIF-2 Quinase/metabolismo , Células A549 , Animais , Doenças Autoimunes do Sistema Nervoso/genética , Doenças Autoimunes do Sistema Nervoso/metabolismo , Modelos Animais de Doenças , Células HEK293 , Humanos , Camundongos , Camundongos Mutantes , Mutação , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/metabolismo
10.
Sci Immunol ; 5(43)2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31980485

RESUMO

Detection of intracellular DNA by the cGAS-STING pathway activates a type I interferon-mediated innate immune response that protects from virus infection. Whether there are additional DNA sensing pathways, and how such pathways might function, remains controversial. We show here that humans-but not laboratory mice-have a second, potent, STING-independent DNA sensing pathway (SIDSP). We identify human DNA-dependent protein kinase (DNA-PK) as the sensor of this pathway and demonstrate that DNA-PK activity drives a robust and broad antiviral response. We show that the E1A oncoprotein of human adenovirus 5 and the ICP0 protein of herpes simplex virus 1 block this response. We found heat shock protein HSPA8/HSC70 as a target for inducible phosphorylation in the DNA-PK antiviral pathway. Last, we demonstrate that DNA damage and detection of foreign DNA trigger distinct modalities of DNA-PK activity. These findings reveal the existence, sensor, a specific downstream target, and viral antagonists of a SIDSP in human cells.


Assuntos
Proteína Quinase Ativada por DNA/imunologia , Adenoviridae , Proteínas E1A de Adenovirus/imunologia , Animais , Linhagem Celular , Herpes Simples/imunologia , Herpesvirus Humano 1 , Humanos , Proteínas Imediatamente Precoces/imunologia , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Ubiquitina-Proteína Ligases/imunologia
11.
Elife ; 82019 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-31808743

RESUMO

cGAS is an intracellular innate immune sensor that detects double-stranded DNA. The presence of billions of base pairs of genomic DNA in all nucleated cells raises the question of how cGAS is not constitutively activated. A widely accepted explanation for this is the sequestration of cGAS in the cytosol, which is thought to prevent cGAS from accessing nuclear DNA. Here, we demonstrate that endogenous cGAS is predominantly a nuclear protein, regardless of cell cycle phase or cGAS activation status. We show that nuclear cGAS is tethered tightly by a salt-resistant interaction. This tight tethering is independent of the domains required for cGAS activation, and it requires intact nuclear chromatin. We identify the evolutionarily conserved tethering surface on cGAS and we show that mutation of single amino acids within this surface renders cGAS massively and constitutively active against self-DNA. Thus, tight nuclear tethering maintains the resting state of cGAS and prevents autoreactivity.


Assuntos
Núcleo Celular/metabolismo , Citosol/enzimologia , Clivagem do DNA , DNA/metabolismo , Proteínas Nucleares/metabolismo , Nucleotidiltransferases/metabolismo , Animais , Linhagem Celular Tumoral , Núcleo Celular/genética , Células Cultivadas , DNA/genética , Dano ao DNA , Células HeLa , Humanos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/genética , Nucleotidiltransferases/genética , Ligação Proteica
13.
Proc Natl Acad Sci U S A ; 115(26): 6798-6803, 2018 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-29891701

RESUMO

Detection of nucleic acids by innate immune sensors triggers the production of type I interferons (IFNs). While IFNs are essential for host defense against viral infection, dysregulated production of IFNs underlies numerous autoinflammatory diseases. We have found that the loss of sumoylation results in a potent, spontaneous IFN response. Vertebrates possess three small ubiquitin-like modifiers (SUMOs) that can be conjugated onto target proteins and alter protein function in diverse but still poorly characterized ways. We demonstrate that regulation of IFN by sumoylation is redundantly mediated by both SUMO2 and SUMO3, but not SUMO1, revealing a previously unknown function of SUMO2/3. Remarkably, this IFN response is independent of all known IFN-inducing pathways and does not require either of the canonical IFN-associated transcription factors IRF3 or IRF7. Taken together, our findings demonstrate that SUMO2 and SUMO3 are specific and essential negative regulators of a noncanonical mechanism of IFN induction.


Assuntos
Interferon Tipo I/metabolismo , Transdução de Sinais/fisiologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação/fisiologia , Ubiquitinas/metabolismo , Células HEK293 , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/metabolismo , Interferon Tipo I/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Células THP-1 , Ubiquitinas/genética
14.
J Immunol ; 200(8): 2748-2756, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29540580

RESUMO

The sensing of viral nucleic acids within the cytosol is essential for the induction of innate immune responses following infection. However, this sensing occurs within cells that have already been infected. The death of infected cells can be beneficial to the host by eliminating the virus's replicative niche and facilitating the release of inflammatory mediators. In this study, we show that sensing of intracellular DNA or RNA by cGAS-STING or RIG-I-MAVS, respectively, leads to activation of RIPK3 and necroptosis in bone marrow-derived macrophages. Notably, this requires signaling through both type I IFN and TNF receptors, revealing synergy between these pathways to induce cell death. Furthermore, we show that hyperactivation of STING in mice leads to a shock-like phenotype, the mortality of which requires activation of the necroptotic pathway and IFN and TNF cosignaling, demonstrating that necroptosis is one outcome of STING signaling in vivo.


Assuntos
Morte Celular/fisiologia , Interferon Tipo I/metabolismo , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , DNA Viral/imunologia , Imunidade Inata/imunologia , Interferon Tipo I/imunologia , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , RNA Viral/imunologia , Fator de Necrose Tumoral alfa/imunologia , Viroses/imunologia
15.
Nat Immunol ; 18(7): 744-752, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28553952

RESUMO

The single-nucleotide polymorphism rs1990760 in the gene encoding the cytosolic viral sensor IFIH1 results in an amino-acid change (A946T; IFIH1T946) that is associated with multiple autoimmune diseases. The effect of this polymorphism on both viral sensing and autoimmune pathogenesis remains poorly understood. Here we found that human peripheral blood mononuclear cells (PBMCs) and cell lines expressing the risk variant IFIH1T946 exhibited heightened basal and ligand-triggered production of type I interferons. Consistent with those findings, mice with a knock-in mutation encoding IFIH1T946 displayed enhanced basal expression of type I interferons, survived a lethal viral challenge and exhibited increased penetrance in autoimmune models, including a combinatorial effect with other risk variants. Furthermore, IFIH1T946 mice manifested an embryonic survival defect consistent with enhanced responsiveness to RNA self ligands. Together our data support a model wherein the production of type I interferons driven by an autoimmune risk variant and triggered by ligand functions to protect against viral challenge, which probably accounts for its selection within human populations but provides this advantage at the cost of modestly promoting the risk of autoimmunity.


Assuntos
Autoimunidade/genética , Infecções por Cardiovirus/genética , Interferon Tipo I/imunologia , Helicase IFIH1 Induzida por Interferon/genética , Adolescente , Adulto , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Autoimunidade/imunologia , Southern Blotting , Infecções por Cardiovirus/imunologia , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Vírus da Encefalomiocardite/imunologia , Feminino , Predisposição Genética para Doença , Células HEK293 , Humanos , Immunoblotting , Helicase IFIH1 Induzida por Interferon/imunologia , Masculino , Camundongos , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Viroses/genética , Viroses/imunologia , Adulto Jovem
16.
Annu Rev Immunol ; 35: 313-336, 2017 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-28142323

RESUMO

Protective immune responses to viral infection are initiated by innate immune sensors that survey extracellular and intracellular space for foreign nucleic acids. The existence of these sensors raises fundamental questions about self/nonself discrimination because of the abundance of self-DNA and self-RNA that occupy these same compartments. Recent advances have revealed that enzymes that metabolize or modify endogenous nucleic acids are essential for preventing inappropriate activation of the innate antiviral response. In this review, we discuss rare human diseases caused by dysregulated nucleic acid sensing, focusing primarily on intracellular sensors of nucleic acids. We summarize lessons learned from these disorders, we rationalize the existence of these diseases in the context of evolution, and we propose that this framework may also apply to a number of more common autoimmune diseases for which the underlying genetics and mechanisms are not yet fully understood.


Assuntos
Doenças Autoimunes do Sistema Nervoso/imunologia , Autoimunidade , Lúpus Eritematoso Sistêmico/imunologia , Malformações do Sistema Nervoso/imunologia , Ácidos Nucleicos/imunologia , Viroses/imunologia , Animais , Humanos , Imunidade Inata , Interferon Tipo I/metabolismo , Receptores Toll-Like/metabolismo
17.
Immunity ; 45(2): 255-66, 2016 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-27496731

RESUMO

Detection of intracellular DNA triggers activation of the STING-dependent interferon-stimulatory DNA (ISD) pathway, which is essential for antiviral responses. Multiple DNA sensors have been proposed to activate this pathway, including AIM2-like receptors (ALRs). Whether the ALRs are essential for activation of this pathway remains unknown. To rigorously explore the function of ALRs, we generated mice lacking all 13 ALR genes. We found that ALRs are dispensable for the type I interferon (IFN) response to transfected DNA ligands, DNA virus infection, and lentivirus infection. We also found that ALRs do not contribute to autoimmune disease in the Trex1(-/-) mouse model of Aicardi-Goutières Syndrome. Finally, CRISPR-mediated disruption of the human AIM2-like receptor IFI16 in primary fibroblasts revealed that IFI16 is not essential for the IFN response to human cytomegalovirus infection. Our findings indicate that ALRs are dispensable for the ISD response and suggest that alternative functions for these receptors should be explored.


Assuntos
Doenças Autoimunes do Sistema Nervoso/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Proteínas de Ligação a DNA/metabolismo , Infecções por Lentivirus/imunologia , Lentivirus/imunologia , Malformações do Sistema Nervoso/imunologia , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Animais , Linhagem Celular , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , DNA/imunologia , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Exodesoxirribonucleases/genética , Loci Gênicos/genética , Humanos , Interferon Tipo I/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/genética , Fosfoproteínas/genética
18.
Cell ; 163(7): 1716-29, 2015 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-26686653

RESUMO

Cellular lipid requirements are achieved through a combination of biosynthesis and import programs. Using isotope tracer analysis, we show that type I interferon (IFN) signaling shifts the balance of these programs by decreasing synthesis and increasing import of cholesterol and long chain fatty acids. Genetically enforcing this metabolic shift in macrophages is sufficient to render mice resistant to viral challenge, demonstrating the importance of reprogramming the balance of these two metabolic pathways in vivo. Unexpectedly, mechanistic studies reveal that limiting flux through the cholesterol biosynthetic pathway spontaneously engages a type I IFN response in a STING-dependent manner. The upregulation of type I IFNs was traced to a decrease in the pool size of synthesized cholesterol and could be inhibited by replenishing cells with free cholesterol. Taken together, these studies delineate a metabolic-inflammatory circuit that links perturbations in cholesterol biosynthesis with activation of innate immunity.


Assuntos
Colesterol/metabolismo , Imunidade Inata , Interferon gama/metabolismo , Transdução de Sinais , Animais , Linhagem Celular Tumoral , Humanos , Interferon beta-1b , Proteínas de Membrana/metabolismo , Ácido Mevalônico/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo
19.
Immunity ; 43(5): 933-44, 2015 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-26588779

RESUMO

Mutations in ADAR, which encodes the ADAR1 RNA-editing enzyme, cause Aicardi-Goutières syndrome (AGS), a severe autoimmune disease associated with an aberrant type I interferon response. How ADAR1 prevents autoimmunity remains incompletely defined. Here, we demonstrate that ADAR1 is a specific and essential negative regulator of the MDA5-MAVS RNA sensing pathway. Moreover, we uncovered a MDA5-MAVS-independent function for ADAR1 in the development of multiple organs. We showed that the p150 isoform of ADAR1 uniquely regulated the MDA5 pathway, whereas both the p150 and p110 isoforms contributed to development. Abrupt deletion of ADAR1 in adult mice revealed that both of these functions were required throughout life. Our findings delineate genetically separable roles for both ADAR1 isoforms in vivo, with implications for the human diseases caused by ADAR mutations.


Assuntos
Adenosina Desaminase/metabolismo , Autoimunidade/fisiologia , RNA Helicases DEAD-box/metabolismo , Isoformas de Proteínas/metabolismo , Edição de RNA/fisiologia , RNA/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Doenças Autoimunes do Sistema Nervoso/metabolismo , Células HEK293 , Humanos , Interferon Tipo I/metabolismo , Helicase IFIH1 Induzida por Interferon , Camundongos , Malformações do Sistema Nervoso/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais/fisiologia
20.
Science ; 350(6260): 568-71, 2015 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-26405230

RESUMO

Cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) detects intracellular DNA and signals through the adapter protein STING to initiate the antiviral response to DNA viruses. Whether DNA viruses can prevent activation of the cGAS-STING pathway remains largely unknown. Here, we identify the oncogenes of the DNA tumor viruses, including E7 from human papillomavirus (HPV) and E1A from adenovirus, as potent and specific inhibitors of the cGAS-STING pathway. We show that the LXCXE motif of these oncoproteins, which is essential for blockade of the retinoblastoma tumor suppressor, is also important for antagonizing DNA sensing. E1A and E7 bind to STING, and silencing of these oncogenes in human tumor cells restores the cGAS-STING pathway. Our findings reveal a host-virus conflict that may have shaped the evolution of viral oncogenes.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Vírus de DNA Tumorais/imunologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Nucleotídeos Cíclicos/antagonistas & inibidores , Proteínas Oncogênicas Virais/metabolismo , Evasão Tumoral , Proteínas E1A de Adenovirus/química , Proteínas E1A de Adenovirus/genética , Motivos de Aminoácidos , Sequência de Aminoácidos , DNA de Neoplasias/imunologia , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Evolução Molecular , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Redes e Vias Metabólicas , Dados de Sequência Molecular , Proteínas Oncogênicas Virais/química , Proteínas Oncogênicas Virais/genética , Proteína do Retinoblastoma/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA