Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
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.
Nature ; 620(7973): 374-380, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37532932

RESUMO

Low-grade inflammation is a hallmark of old age and a central driver of ageing-associated impairment and disease1. Multiple factors can contribute to ageing-associated inflammation2; however, the molecular pathways that transduce aberrant inflammatory signalling and their impact in natural ageing remain unclear. Here we show that the cGAS-STING signalling pathway, which mediates immune sensing of DNA3, is a critical driver of chronic inflammation and functional decline during ageing. Blockade of STING suppresses the inflammatory phenotypes of senescent human cells and tissues, attenuates ageing-related inflammation in multiple peripheral organs and the brain in mice, and leads to an improvement in tissue function. Focusing on the ageing brain, we reveal that activation of STING triggers reactive microglial transcriptional states, neurodegeneration and cognitive decline. Cytosolic DNA released from perturbed mitochondria elicits cGAS activity in old microglia, defining a mechanism by which cGAS-STING signalling is engaged in the ageing brain. Single-nucleus RNA-sequencing analysis of microglia and hippocampi of a cGAS gain-of-function mouse model demonstrates that engagement of cGAS in microglia is sufficient to direct ageing-associated transcriptional microglial states leading to bystander cell inflammation, neurotoxicity and impaired memory capacity. Our findings establish the cGAS-STING pathway as a driver of ageing-related inflammation in peripheral organs and the brain, and reveal blockade of cGAS-STING signalling as a potential strategy to halt neurodegenerative processes during old age.


Assuntos
Envelhecimento , Encéfalo , Disfunção Cognitiva , Inflamação , Proteínas de Membrana , Doenças Neurodegenerativas , Nucleotidiltransferases , Animais , Humanos , Camundongos , Envelhecimento/metabolismo , Envelhecimento/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Efeito Espectador , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , DNA/imunologia , Inflamação/enzimologia , Inflamação/metabolismo , Proteínas de Membrana/metabolismo , Transtornos da Memória/enzimologia , Transtornos da Memória/metabolismo , Microglia/metabolismo , Mitocôndrias/metabolismo , Doenças Neurodegenerativas/enzimologia , Doenças Neurodegenerativas/metabolismo , Nucleotidiltransferases/metabolismo , Especificidade de Órgãos , Transdução de Sinais , Hipocampo/metabolismo , Hipocampo/patologia
3.
Nat Cancer ; 3(12): 1452-1463, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36510011

RESUMO

The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway has emerged as a critical innate immune pathway that, following engagement by DNA, promotes distinct immune effector responses that can impact virtually all aspects of tumorigenesis, from malignant cell transformation to metastasis. Here we address how natural tumor-associated processes and traditional cancer therapies are shaped by cGAS-STING signaling, and how this contributes to beneficial or detrimental outcomes of cancer. We consider current efforts to target the cGAS-STING axis in tumors and highlight new frontiers in cGAS-STING biology to inspire thinking about their connection to cancer.


Assuntos
Proteínas de Membrana , Neoplasias , Nucleotidiltransferases , Transdução de Sinais , Humanos , Carcinogênese , DNA/metabolismo , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo
4.
Nature ; 612(7939): 347-353, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36385525

RESUMO

Solid cancers exhibit a dynamic balance between cell death and proliferation ensuring continuous tumour maintenance and growth1,2. Increasing evidence links enhanced cancer cell apoptosis to paracrine activation of cells in the tumour microenvironment initiating tissue repair programs that support tumour growth3,4, yet the direct effects of dying cancer cells on neighbouring tumour epithelia and how this paracrine effect potentially contributes to therapy resistance are unclear. Here we demonstrate that chemotherapy-induced tumour cell death in patient-derived colorectal tumour organoids causes ATP release triggering P2X4 (also known as P2RX4) to mediate an mTOR-dependent pro-survival program in neighbouring cancer cells, which renders surviving tumour epithelia sensitive to mTOR inhibition. The induced mTOR addiction in persisting epithelial cells is due to elevated production of reactive oxygen species and subsequent increased DNA damage in response to the death of neighbouring cells. Accordingly, inhibition of the P2X4 receptor or direct mTOR blockade prevents induction of S6 phosphorylation and synergizes with chemotherapy to cause massive cell death induced by reactive oxygen species and marked tumour regression that is not seen when individually applied. Conversely, scavenging of reactive oxygen species prevents cancer cells from becoming reliant on mTOR activation. Collectively, our findings show that dying cancer cells establish a new dependency on anti-apoptotic programs in their surviving neighbours, thereby creating an opportunity for combination therapy in P2X4-expressing epithelial tumours.


Assuntos
Neoplasias do Colo , Organoides , Humanos , Espécies Reativas de Oxigênio , Causas de Morte , Morte Celular , Microambiente Tumoral , Serina-Treonina Quinases TOR
6.
Nature ; 610(7933): 761-767, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36261523

RESUMO

Stimulator of interferon genes (STING) functions downstream of cyclic GMP-AMP synthase in DNA sensing or as a direct receptor for bacterial cyclic dinucleotides and small molecules to activate immunity during infection, cancer and immunotherapy1-10. Precise regulation of STING is essential to ensure balanced immune responses and prevent detrimental autoinflammation11-16. After activation, STING, a transmembrane protein, traffics from the endoplasmic reticulum to the Golgi, where its phosphorylation by the protein kinase TBK1 enables signal transduction17-20. The mechanism that ends STING signalling at the Golgi remains unknown. Here we show that adaptor protein complex 1 (AP-1) controls the termination of STING-dependent immune activation. We find that AP-1 sorts phosphorylated STING into clathrin-coated transport vesicles for delivery to the endolysosomal system, where STING is degraded21. We identify a highly conserved dileucine motif in the cytosolic C-terminal tail (CTT) of STING that, together with TBK1-dependent CTT phosphorylation, dictates the AP-1 engagement of STING. A cryo-electron microscopy structure of AP-1 in complex with phosphorylated STING explains the enhanced recognition of TBK1-activated STING. We show that suppression of AP-1 exacerbates STING-induced immune responses. Our results reveal a structural mechanism of negative regulation of STING and establish that the initiation of signalling is inextricably associated with its termination to enable transient activation of immunity.


Assuntos
Complexo 1 de Proteínas Adaptadoras , Clatrina , Complexo 1 de Proteínas Adaptadoras/química , Complexo 1 de Proteínas Adaptadoras/metabolismo , Complexo 1 de Proteínas Adaptadoras/ultraestrutura , Clatrina/metabolismo , Microscopia Crioeletrônica , DNA/metabolismo , Imunidade Inata , Proteínas Serina-Treonina Quinases , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Proteínas de Membrana/ultraestrutura , Motivos de Aminoácidos , Endossomos/metabolismo , Lisossomos/metabolismo , Fosforilação
7.
J Exp Med ; 219(10)2022 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-35997679

RESUMO

Autoimmune vasculitis is a group of life-threatening diseases, whose underlying pathogenic mechanisms are incompletely understood, hampering development of targeted therapies. Here, we demonstrate that patients suffering from anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitis (AAV) showed increased levels of cGAMP and enhanced IFN-I signature. To identify disease mechanisms and potential therapeutic targets, we developed a mouse model for pulmonary AAV that mimics severe disease in patients. Immunogenic DNA accumulated during disease onset, triggering cGAS/STING/IRF3-dependent IFN-I release that promoted endothelial damage, pulmonary hemorrhages, and lung dysfunction. Macrophage subsets played dichotomic roles in disease. While recruited monocyte-derived macrophages were major disease drivers by producing most IFN-ß, resident alveolar macrophages contributed to tissue homeostasis by clearing red blood cells and limiting infiltration of IFN-ß-producing macrophages. Moreover, pharmacological inhibition of STING, IFNAR-I, or its downstream JAK/STAT signaling reduced disease severity and accelerated recovery. Our study unveils the importance of STING/IFN-I axis in promoting pulmonary AAV progression and identifies cellular and molecular targets to ameliorate disease outcomes.


Assuntos
Interferon Tipo I , Ácidos Nucleicos , Vasculite , Animais , Pulmão , Macrófagos , Proteínas de Membrana/metabolismo , Camundongos , Nucleotidiltransferases
8.
Curr Opin Immunol ; 74: 164-171, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35124516

RESUMO

The recognition of DNA through the cGAS-STING pathway plays a critical role in antiviral immunity, but can also drive maladaptive immune responses underlying several pathological conditions. Despite its importance, understanding the organization of cGAS-STING signaling axis in the 3D space of a cell is relatively limited. In this review, we discuss recent progress in understanding the spatial coordination of DNA-induced cGAS-STING activity and its relevance in balancing innate immune responses toward (self-) DNA. We also consider the importance of context-specific co-factors and end by suggesting interesting areas for further research into spatial aspects of DNA-induced innate immunity.


Assuntos
Proteínas de Membrana , Nucleotidiltransferases , Animais , DNA , Humanos , Imunidade Inata/genética , Proteínas de Membrana/metabolismo , Nucleotidiltransferases/metabolismo , Transdução de Sinais
9.
Nature ; 603(7899): 145-151, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35045565

RESUMO

COVID-19, which is caused by infection with SARS-CoV-2, is characterized by lung pathology and extrapulmonary complications1,2. Type I interferons (IFNs) have an essential role in the pathogenesis of COVID-19 (refs 3-5). Although rapid induction of type I IFNs limits virus propagation, a sustained increase in the levels of type I IFNs in the late phase of the infection is associated with aberrant inflammation and poor clinical outcome5-17. Here we show that the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, which controls immunity to cytosolic DNA, is a critical driver of aberrant type I IFN responses in COVID-19 (ref. 18). Profiling COVID-19 skin manifestations, we uncover a STING-dependent type I IFN signature that is primarily mediated by macrophages adjacent to areas of endothelial cell damage. Moreover, cGAS-STING activity was detected in lung samples from patients with COVID-19 with prominent tissue destruction, and was associated with type I IFN responses. A lung-on-chip model revealed that, in addition to macrophages, infection with SARS-CoV-2 activates cGAS-STING signalling in endothelial cells through mitochondrial DNA release, which leads to cell death and type I IFN production. In mice, pharmacological inhibition of STING reduces severe lung inflammation induced by SARS-CoV-2 and improves disease outcome. Collectively, our study establishes a mechanistic basis of pathological type I IFN responses in COVID-19 and reveals a principle for the development of host-directed therapeutics.


Assuntos
COVID-19/imunologia , COVID-19/patologia , Interferon Tipo I/imunologia , Proteínas de Membrana/metabolismo , Nucleotidiltransferases/metabolismo , SARS-CoV-2/imunologia , Animais , COVID-19/metabolismo , COVID-19/virologia , Células Cultivadas , DNA Mitocondrial/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Células Endoteliais/patologia , Feminino , Regulação da Expressão Gênica/imunologia , Humanos , Imunidade Inata , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Macrófagos/imunologia , Proteínas de Membrana/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia/imunologia , Pneumonia/metabolismo , Pneumonia/patologia , Pneumonia/virologia , SARS-CoV-2/patogenicidade , Transdução de Sinais , Pele/imunologia , Pele/metabolismo , Pele/patologia
10.
Nat Rev Immunol ; 21(10): 620, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34580458
11.
EMBO J ; 40(22): e108234, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34586646

RESUMO

DNA methylation is a fundamental epigenetic modification, important across biological processes. The maintenance methyltransferase DNMT1 is essential for lineage differentiation during development, but its functions in tissue homeostasis are incompletely understood. We show that epidermis-specific DNMT1 deletion severely disrupts epidermal structure and homeostasis, initiating a massive innate immune response and infiltration of immune cells. Mechanistically, DNA hypomethylation in keratinocytes triggered transposon derepression, mitotic defects, and formation of micronuclei. DNA release into the cytosol of DNMT1-deficient keratinocytes activated signaling through cGAS and STING, thus triggering inflammation. Our findings show that disruption of a key epigenetic mark directly impacts immune and tissue homeostasis, and potentially impacts our understanding of autoinflammatory diseases and cancer immunotherapy.


Assuntos
Metilação de DNA , Dermatite/genética , Epiderme/fisiopatologia , Nucleotidiltransferases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Aberrações Cromossômicas , Citosol/fisiologia , DNA (Citosina-5-)-Metiltransferase 1/genética , Dermatite/imunologia , Dermatite/patologia , Humanos , Imunidade Inata/genética , Helicase IFIH1 Induzida por Interferon/metabolismo , Queratinócitos/imunologia , Queratinócitos/metabolismo , Queratinócitos/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Transgênicos , Nucleotidiltransferases/genética
12.
Trends Biochem Sci ; 46(10): 822-831, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34215510

RESUMO

Recognition of DNA is an evolutionarily highly conserved mechanism of immunity. In mammals, the cGAS-STING pathway plays a central role in coupling DNA sensing to the execution of innate immune mechanisms, both in contexts of infection as well as in noninfectious settings of cellular stress and injury. The indiscriminate ability of double-stranded DNA (dsDNA) to activate cGAS challenges our understanding on how engagement of this pathway is prevented on genomic self-DNA under homeostatic conditions. Here, we review recent discoveries on the regulation of cGAS on chromatin and we discuss implications for cGAS-dependent inflammatory phenotypes. We close by highlighting emerging developments on the role of nuclear cGAS and related open questions for future research.


Assuntos
Cromatina , Proteínas de Membrana , Animais , DNA , Proteínas de Membrana/genética , Nucleotidiltransferases/genética
13.
Nat Rev Immunol ; 21(9): 548-569, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33833439

RESUMO

The cGAS-STING signalling pathway has emerged as a key mediator of inflammation in the settings of infection, cellular stress and tissue damage. Underlying this broad involvement of the cGAS-STING pathway is its capacity to sense and regulate the cellular response towards microbial and host-derived DNAs, which serve as ubiquitous danger-associated molecules. Insights into the structural and molecular biology of the cGAS-STING pathway have enabled the development of selective small-molecule inhibitors with the potential to target the cGAS-STING axis in a number of inflammatory diseases in humans. Here, we outline the principal elements of the cGAS-STING signalling cascade and discuss the general mechanisms underlying the association of cGAS-STING activity with various autoinflammatory, autoimmune and degenerative diseases. Finally, we outline the chemical nature of recently developed cGAS and STING antagonists and summarize their potential clinical applications.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Inflamação/terapia , Proteínas de Membrana/antagonistas & inibidores , Nucleotidiltransferases/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Autofagia , Morte Celular , Proliferação de Células , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Inibidores Enzimáticos/química , Inibidores Enzimáticos/uso terapêutico , Mutação com Ganho de Função , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Modelos Biológicos , Modelos Moleculares , Nucleotídeos Cíclicos/imunologia , Nucleotídeos Cíclicos/metabolismo , Nucleotidiltransferases/genética , Nucleotidiltransferases/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/imunologia
14.
Dev Cell ; 56(7): 876-877, 2021 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-33823133

RESUMO

Coupling DNA sensing to the initiation of immune responses necessitates auxiliary control mechanisms to avoid autoimmunity. A key factor is the exonuclease TREX1, which antagonizes DNA-mediated activation of cGAS. Two studies, by Mohr et al. (2021) and Zhou et al. (2021), uncover new aspects of this important control mechanism.


Assuntos
Nucleotidiltransferases , Fosfoproteínas , Autoimunidade , DNA/genética , Nucleotidiltransferases/genética
15.
Science ; 371(6535): 1204-1205, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33737475
16.
Open Biol ; 11(3): 210030, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33715389

RESUMO

Nucleic acid sensing through pattern recognition receptors is critical for immune recognition of microbial infections. Microbial DNA is frequently methylated at the N6 position of adenines (m6A), a modification that is rare in mammalian host DNA. We show here how that m6A methylation of 5'-GATC-3' motifs augments the immunogenicity of synthetic double-stranded (ds)DNA in murine macrophages and dendritic cells. Transfection with m6A-methylated DNA increased the expression of the activation markers CD69 and CD86, and of Ifnß, iNos and Cxcl10 mRNA. Similar to unmethylated cytosolic dsDNA, recognition of m6A DNA occurs independently of TLR and RIG-I signalling, but requires the two key mediators of cytosolic DNA sensing, STING and cGAS. Intriguingly, the response to m6A DNA is sequence-specific. m6A is immunostimulatory in some motifs, but immunosuppressive in others, a feature that is conserved between mouse and human macrophages. In conclusion, epigenetic alterations of DNA depend on the context of the sequence and are differentially perceived by innate cells, a feature that could potentially be used for the design of immune-modulating therapeutics.


Assuntos
Adenina/análogos & derivados , Metilação de DNA , Imunidade Inata , Oligodesoxirribonucleotídeos/imunologia , Adenina/metabolismo , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Antígeno B7-2/metabolismo , Células Cultivadas , Quimiocina CXCL10/metabolismo , Citoplasma/metabolismo , Células Dendríticas/imunologia , Humanos , Interferon beta/metabolismo , Lectinas Tipo C/metabolismo , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II/metabolismo , Oligodesoxirribonucleotídeos/química , Receptores Toll-Like/metabolismo
17.
J Exp Med ; 217(11)2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-32991673

RESUMO

Two studies published in this issue of JEM, by Lepelley et al. (https://doi.org/10.1084/jem.20200600) and Deng et al. (https://doi.org/10.1084/jem.20201045), and two additional manuscripts by Mukai et al. (https://doi.org/10.1101/2020.05.20.107664 Preprint v1) and Steiner et al. (https://doi.org/10.1101/2020.07.09.194399 Preprint v1) demonstrate that COPA syndrome-associated high interferon titers are linked to mutations in COPA preventing STING's retrieval from the Golgi back to the ER and thereby causing chronic immune activation.


Assuntos
Antivirais , Mordeduras e Picadas , Complexo I de Proteína do Envoltório , Humanos
18.
Nature ; 587(7835): 668-672, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32911482

RESUMO

The DNA sensor cyclic GMP-AMP synthase (cGAS) initiates innate immune responses following microbial infection, cellular stress and cancer1. Upon activation by double-stranded DNA, cytosolic cGAS produces 2'3' cGMP-AMP, which triggers the induction of inflammatory cytokines and type I interferons 2-7. cGAS is also present inside the cell nucleus, which is replete with genomic DNA8, where chromatin has been implicated in restricting its enzymatic activity9. However, the structural basis for inhibition of cGAS by chromatin remains unknown. Here we present the cryo-electron microscopy structure of human cGAS bound to nucleosomes. cGAS makes extensive contacts with both the acidic patch of the histone H2A-H2B heterodimer and nucleosomal DNA. The structural and complementary biochemical analysis also find cGAS engaged to a second nucleosome in trans. Mechanistically, binding of the nucleosome locks cGAS into a monomeric state, in which steric hindrance suppresses spurious activation by genomic DNA. We find that mutations to the cGAS-acidic patch interface are sufficient to abolish the inhibitory effect of nucleosomes in vitro and to unleash the activity of cGAS on genomic DNA in living cells. Our work uncovers the structural basis of the interaction between cGAS and chromatin and details a mechanism that permits self-non-self discrimination of genomic DNA by cGAS.


Assuntos
Microscopia Crioeletrônica , Nucleossomos/metabolismo , Nucleotidiltransferases/antagonistas & inibidores , Nucleotidiltransferases/química , Células HeLa , Histonas/metabolismo , Humanos , Modelos Moleculares , Mutação , Nucleossomos/química , Nucleossomos/ultraestrutura , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/ultraestrutura
19.
Science ; 369(6505): 823-828, 2020 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-32792394

RESUMO

The appearance of DNA in the cytosol is perceived as a danger signal that stimulates potent immune responses through cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS). How cells regulate the activity of cGAS toward self-DNA and guard against potentially damaging autoinflammatory responses is a fundamental biological question. Here, we identify barrier-to-autointegration factor 1 (BAF) as a natural opponent of cGAS activity on genomic self-DNA. We show that BAF dynamically outcompetes cGAS for DNA binding, hence prohibiting the formation of DNA-cGAS complexes that are essential for enzymatic activity. Upon acute loss of nuclear membrane integrity, BAF is necessary to restrict cGAS activity on exposed DNA. Our observations reveal a safeguard mechanism, distinct from physical separation, by which cells protect themselves against aberrant immune responses toward genomic DNA.


Assuntos
Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , DNA/imunologia , Imunidade Inata , Nucleotidiltransferases/metabolismo , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Membrana Nuclear/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA