Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.228
Filtrar
1.
Sci Data ; 11(1): 328, 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38565538

RESUMO

Human infections caused by viral pathogens trigger a complex gamut of host responses that limit disease, resolve infection, generate immunity, and contribute to severe disease or death. Here, we present experimental methods and multi-omics data capture approaches representing the global host response to infection generated from 45 individual experiments involving human viruses from the Orthomyxoviridae, Filoviridae, Flaviviridae, and Coronaviridae families. Analogous experimental designs were implemented across human or mouse host model systems, longitudinal samples were collected over defined time courses, and global multi-omics data (transcriptomics, proteomics, metabolomics, and lipidomics) were acquired by microarray, RNA sequencing, or mass spectrometry analyses. For comparison, we have included transcriptomics datasets from cells treated with type I and type II human interferon. Raw multi-omics data and metadata were deposited in public repositories, and we provide a central location linking the raw data with experimental metadata and ready-to-use, quality-controlled, statistically processed multi-omics datasets not previously available in any public repository. This compendium of infection-induced host response data for reuse will be useful for those endeavouring to understand viral disease pathophysiology and network biology.


Assuntos
Multiômica , Viroses , Vírus , Animais , Humanos , Camundongos , Perfilação da Expressão Gênica/métodos , Metabolômica , Proteômica/métodos , Viroses/imunologia , Interações Hospedeiro-Patógeno
2.
BMC Infect Dis ; 24(1): 431, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38654203

RESUMO

BACKGROUND: Vaccination is effective in preventing viral respiratory infectious diseases through protective antibodies and the gut microbiome has been proven to regulate human immunity. This study explores the causal correlations between gut microbial features and serum-specific antiviral immunoglobulin G (IgG) levels. METHODS: We conduct a two-sample bidirectional Mendelian randomization (MR) analysis using genome-wide association study (GWAS) summary data to explore the causal relationships between 412 gut microbial features and four antiviral IgG (for influenza A, measles, rubella, and mumps) levels. To make the results more reliable, we used four robust methods and performed comprehensive sensitivity analyses. RESULTS: The MR analyses revealed 26, 13, 20, and 18 causal associations of the gut microbial features influencing four IgG levels separately. ​Interestingly, ten microbial features, like genus Collinsella, species Bifidobacterium longum, and the biosynthesis of L-alanine have shown the capacity to regulate multiple IgG levels with consistent direction (rise or fall). The ​reverse MR analysis suggested several potential causal associations of IgG levels affecting microbial features. CONCLUSIONS: The human immune response against viral respiratory infectious diseases could be modulated by changing the abundance of gut microbes, which provided new approaches for the intervention of viral respiratory infections.


Assuntos
Microbioma Gastrointestinal , Imunoglobulina G , Análise da Randomização Mendeliana , Infecções Respiratórias , Humanos , Imunoglobulina G/sangue , Infecções Respiratórias/imunologia , Infecções Respiratórias/prevenção & controle , Infecções Respiratórias/microbiologia , Estudo de Associação Genômica Ampla , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Vacinação , Viroses/imunologia , Viroses/prevenção & controle
4.
J Virol ; 98(3): e0008824, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38386781

RESUMO

Type I and type II IFNs are important immune modulators in both innate and adaptive immunity. They transmit signaling by activating JAK-STAT pathways. Sirtuin 1 (SIRT1), a class III NAD+-dependent deacetylase, has multiple functions in a variety of physiological processes. Here, we characterized the novel functions of SIRT1 in the regulation of type I and type II IFN-induced signaling. Overexpression of SIRT1 inhibited type I and type II IFN-induced interferon-stimulated response element activation. In contrast, knockout of SIRT1 promoted type I and type II IFN-induced expression of ISGs and inhibited viral replication. Treatment with SIRT1 inhibitor EX527 had similar positive effects. SIRT1 physically associated with STAT1 or STAT3, and this interaction was enhanced by IFN stimulation or viral infection. By deacetylating STAT1 at K673 and STAT3 at K679/K685/K707/K709, SIRT1 downregulated the phosphorylation of STAT1 (Y701) and STAT3 (Y705). Sirt1+/- primary peritoneal macrophages and Sirt1+/- mice exhibited enhanced IFN-induced signaling and antiviral activity. Thus, SIRT1 is a novel negative regulator of type I and type II IFN-induced signaling through its deacetylase activity.IMPORTANCESIRT1 has been reported in the precise regulation of antiviral (RNA and DNA) immunity. However, its functions in type I and type II IFN-induced signaling are still unclear. In this study, we deciphered the important functions of SIRT1 in both type I and type II IFN-induced JAK-STAT signaling and explored the potential acting mechanisms. It is helpful for understanding the regulatory roles of SIRT1 at different levels of IFN signaling. It also consolidates the notion that SIRT1 is an important target for intervention in viral infection, inflammatory diseases, or even interferon-related therapies.


Assuntos
Interferon Tipo I , Sirtuína 1 , Viroses , Animais , Camundongos , Imunidade Inata , Interferon Tipo I/metabolismo , Interferon gama , Fosforilação , Transdução de Sinais , Sirtuína 1/genética , Sirtuína 1/metabolismo , Fator de Transcrição STAT1/metabolismo , Viroses/imunologia
5.
Cell Death Differ ; 31(3): 280-291, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38383887

RESUMO

Detection of cytosolic nucleic acids by pattern recognition receptors, including STING and RIG-I, leads to the activation of multiple signalling pathways that culminate in the production of type I interferons (IFNs) which are vital for host survival during virus infection. In addition to protective immune modulatory functions, type I IFNs are also associated with autoimmune diseases. Hence, it is important to elucidate the mechanisms that govern their expression. In this study, we identified a critical regulatory function of the DUSP4 phosphatase in innate immune signalling. We found that DUSP4 regulates the activation of TBK1 and ERK1/2 in a signalling complex containing DUSP4, TBK1, ERK1/2 and IRF3 to regulate the production of type I IFNs. Mice deficient in DUSP4 were more resistant to infections by both RNA and DNA viruses but more susceptible to malaria parasites. Therefore, our study establishes DUSP4 as a regulator of nucleic acid sensor signalling and sheds light on an important facet of the type I IFN regulatory system.


Assuntos
Interferon Tipo I , Proteínas de Membrana , Proteínas Tirosina Fosfatases , Receptores de Superfície Celular , 60696 , Viroses , Animais , Camundongos , Imunidade Inata , Interferon Tipo I/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Viroses/imunologia , Viroses/metabolismo , Proteínas de Membrana/metabolismo , 60696/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Receptores de Superfície Celular/metabolismo
6.
Nature ; 624(7992): 645-652, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38093014

RESUMO

People with diabetes feature a life-risking susceptibility to respiratory viral infection, including influenza and SARS-CoV-2 (ref. 1), whose mechanism remains unknown. In acquired and genetic mouse models of diabetes, induced with an acute pulmonary viral infection, we demonstrate that hyperglycaemia leads to impaired costimulatory molecule expression, antigen transport and T cell priming in distinct lung dendritic cell (DC) subsets, driving a defective antiviral adaptive immune response, delayed viral clearance and enhanced mortality. Mechanistically, hyperglycaemia induces an altered metabolic DC circuitry characterized by increased glucose-to-acetyl-CoA shunting and downstream histone acetylation, leading to global chromatin alterations. These, in turn, drive impaired expression of key DC effectors including central antigen presentation-related genes. Either glucose-lowering treatment or pharmacological modulation of histone acetylation rescues DC function and antiviral immunity. Collectively, we highlight a hyperglycaemia-driven metabolic-immune axis orchestrating DC dysfunction during pulmonary viral infection and identify metabolic checkpoints that may be therapeutically exploited in mitigating exacerbated disease in infected diabetics.


Assuntos
Células Dendríticas , Complicações do Diabetes , Diabetes Mellitus , Suscetibilidade a Doenças , Hiperglicemia , Pulmão , Viroses , Animais , Camundongos , Acetilcoenzima A/metabolismo , Acetilação , Cromatina/genética , Cromatina/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Complicações do Diabetes/imunologia , Complicações do Diabetes/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/imunologia , Diabetes Mellitus/metabolismo , Glucose/metabolismo , Histonas/metabolismo , Hiperglicemia/complicações , Hiperglicemia/imunologia , Hiperglicemia/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/virologia , Linfócitos T/imunologia , Viroses/complicações , Viroses/imunologia , Viroses/mortalidade , Vírus/imunologia , Modelos Animais de Doenças , Humanos
7.
J Virol ; 97(12): e0150123, 2023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-37982618

RESUMO

IMPORTANCE: The type-I interferon (IFN-I) signaling pathway is the first line of antiviral innate immunity. It must be precisely regulated against virus-induced damage. The tightly regulated mechanisms of action of host genes in the antiviral innate immune signaling pathway are still worth studying. Here, we report a novel role of DLG1 in positively regulating the IκB kinase epsilon (IKKε)-mediated IFN-I signaling response against negative-stranded RNA virus replication, whereas the RNA virus inhibits the expression of DLG1 for immune escape. Importantly, the E3 ligase March2 interacts with and promotes K27-linked polyubiquitination of IKKε, and p62 is a cargo receptor that recognizes ubiquitinated IKKε for eventual autophagic degradation. Together, the current findings elucidate the role of DLG1 in the antiviral IFN-I signaling pathway and viral infection repression.


Assuntos
Autofagia , Proteína 1 Homóloga a Discs-Large , Quinase I-kappa B , Imunidade Inata , Vírus de RNA de Sentido Negativo , Proteína Sequestossoma-1 , Viroses , Humanos , Proteína 1 Homóloga a Discs-Large/metabolismo , Quinase I-kappa B/metabolismo , Imunidade Inata/imunologia , Vírus de RNA de Sentido Negativo/crescimento & desenvolvimento , Vírus de RNA de Sentido Negativo/imunologia , Poliubiquitina/metabolismo , Proteína Sequestossoma-1/antagonistas & inibidores , Transdução de Sinais , Viroses/imunologia , Animais , Linhagem Celular
8.
J Virol ; 97(11): e0088623, 2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-37843373

RESUMO

IMPORTANCE: The expression of circVPS13D was upregulated with SCRV invasion, which proved that circVPS13D was involved in the regulation of the antiviral immune response. Our study revealed that the existence of circVPS13D promoted the replication of SCRV. Functionally, circVPS13D negatively regulates the antiviral responses of fish. Mechanistically, we confirmed that circVPS13D inhibited RLRs antiviral signaling pathway via the encoded protein VPS13D-170aa by targeting MAVS. Our study provided novel insights into the roles of protein-coding circRNAs and supported VPS13D-170aa as a negative regulator in the antiviral immune responses of teleost fish.


Assuntos
Proteínas de Peixes , RNA Circular , Proteínas de Transporte Vesicular , Viroses , Animais , Proteínas de Peixes/genética , Peixes/imunologia , Peixes/virologia , Imunidade Inata , RNA Circular/metabolismo , Transdução de Sinais , Proteínas de Transporte Vesicular/genética , Viroses/imunologia , Viroses/metabolismo , Viroses/veterinária , Viroses/virologia , Doenças dos Peixes/imunologia , Doenças dos Peixes/metabolismo , Doenças dos Peixes/virologia
9.
J Virol ; 97(9): e0065823, 2023 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-37656959

RESUMO

Epigenetic modifications, such as DNA hypermethylation, histone acetylation/methylation, or nucleosome positioning, result in differential gene expression. These modifications can have an impact on various pathways, including host antiviral immune responses. In this review, we summarize the current understanding of epigenetic modifications induced by viruses to counteract host antiviral immune responses, which are crucial for establishing and maintaining infection of viruses. Finally, we provide insights into the potential use of epigenetic modulators in combating viral infections and virus-induced diseases.


Assuntos
Epigênese Genética , Viroses , Vírus , Humanos , Metilação de DNA , Imunidade , Viroses/genética , Viroses/imunologia , Vírus/genética , Interações Hospedeiro-Patógeno
10.
Front Cell Infect Microbiol ; 13: 1256882, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37719674

RESUMO

TRIM7 has been demonstrated to have significant roles in promoting host defense against viral infections and regulating immune signaling pathways. As an E3 ubiquitin ligase, it catalyzes the ubiquitination of various substrates, including adaptor proteins (MAVS and STING) and transcription factors (NF-κB and IRF3), thereby exerting positive or negative regulation on immune signal pathways. However, viruses have developed immune evasion mechanisms to counteract TRIM7. Some viruses can inhibit TRIM7 function by targeting it for degradation or sequestering it away from its targets. Moreover, TRIM7 may even facilitate viral infection by ubiquitinating viral proteins, including envelope proteins that are critical for tissue and species tropism. A comprehensive understanding of the interaction between TRIM7 and antiviral immunity is crucial for the development of innovative treatments for viral diseases.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases , Viroses , Evasão da Resposta Imune , NF-kappa B , Ubiquitina-Proteína Ligases/imunologia , Viroses/imunologia , Proteínas com Motivo Tripartido/imunologia
11.
Life Sci Alliance ; 6(10)2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37558422

RESUMO

RIG-I recognizes viral dsRNA and activates a cell-autonomous antiviral response. Upon stimulation, it triggers a signaling cascade leading to the production of type I and III IFNs. IFNs are secreted and signal to elicit the expression of IFN-stimulated genes, establishing an antiviral state of the cell. The topology of this pathway has been studied intensively, however, its exact dynamics are less understood. Here, we employed electroporation to synchronously activate RIG-I, enabling us to characterize cell-intrinsic innate immune signaling at a high temporal resolution. Employing IFNAR1/IFNLR-deficient cells, we could differentiate primary RIG-I signaling from secondary signaling downstream of the IFN receptors. Based on these data, we developed a comprehensive mathematical model capable of simulating signaling downstream of dsRNA recognition by RIG-I and the feedback and signal amplification by IFN. We further investigated the impact of viral antagonists on signaling dynamics. Our work provides a comprehensive insight into the signaling events that occur early upon virus infection and opens new avenues to study and disentangle the complexity of the host-virus interface.


Assuntos
Proteína DEAD-box 58 , Receptores Imunológicos , Transdução de Sinais , Viroses , Linhagem Celular , Receptores Imunológicos/imunologia , Proteína DEAD-box 58/imunologia , Viroses/imunologia
12.
Nature ; 621(7977): 179-187, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37648857

RESUMO

Tissue resident memory CD8+ T (TRM) cells offer rapid and long-term protection at sites of reinfection1. Tumour-infiltrating lymphocytes with characteristics of TRM cells maintain enhanced effector functions, predict responses to immunotherapy and accompany better prognoses2,3. Thus, an improved understanding of the metabolic strategies that enable tissue residency by T cells could inform new approaches to empower immune responses in tissues and solid tumours. Here, to systematically define the basis for the metabolic reprogramming supporting TRM cell differentiation, survival and function, we leveraged in vivo functional genomics, untargeted metabolomics and transcriptomics of virus-specific memory CD8+ T cell populations. We found that memory CD8+ T cells deployed a range of adaptations to tissue residency, including reliance on non-steroidal products of the mevalonate-cholesterol pathway, such as coenzyme Q, driven by increased activity of the transcription factor SREBP2. This metabolic adaptation was most pronounced in the small intestine, where TRM cells interface with dietary cholesterol and maintain a heightened state of activation4, and was shared by functional tumour-infiltrating lymphocytes in diverse tumour types in mice and humans. Enforcing synthesis of coenzyme Q through deletion of Fdft1 or overexpression of PDSS2 promoted mitochondrial respiration, memory T cell formation following viral infection and enhanced antitumour immunity. In sum, through a systematic exploration of TRM cell metabolism, we reveal how these programs can be leveraged to fuel memory CD8+ T cell formation in the context of acute infections and enhance antitumour immunity.


Assuntos
Linfócitos T CD8-Positivos , Linfócitos do Interstício Tumoral , Neoplasias , Animais , Humanos , Camundongos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Respiração Celular , Colesterol/metabolismo , Colesterol/farmacologia , Memória Imunológica , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Metabolômica , Ácido Mevalônico/metabolismo , Neoplasias/imunologia , Ubiquinona/metabolismo , Viroses/imunologia , Vírus/imunologia , Mitocôndrias/metabolismo
14.
Elife ; 122023 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-37566453

RESUMO

Monocytes are heterogeneous innate effector leukocytes generated in the bone marrow and released into circulation in a CCR2-dependent manner. During infection or inflammation, myelopoiesis is modulated to rapidly meet the demand for more effector cells. Danger signals from peripheral tissues can influence this process. Herein we demonstrate that repetitive TLR7 stimulation via the epithelial barriers drove a potent emergency bone marrow monocyte response in mice. This process was unique to TLR7 activation and occurred independently of the canonical CCR2 and CX3CR1 axes or prototypical cytokines. The monocytes egressing the bone marrow had an immature Ly6C-high profile and differentiated into vascular Ly6C-low monocytes and tissue macrophages in multiple organs. They displayed a blunted cytokine response to further TLR7 stimulation and reduced lung viral load after RSV and influenza virus infection. These data provide insights into the emergency myelopoiesis likely to occur in response to the encounter of single-stranded RNA viruses at barrier sites.


Assuntos
Mielopoese , Receptor 7 Toll-Like , Viroses , Animais , Camundongos , Citocinas , Pulmão , Camundongos Endogâmicos C57BL , Monócitos , Receptor 7 Toll-Like/genética , Viroses/imunologia
15.
Front Cell Infect Microbiol ; 13: 1173505, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37465759

RESUMO

The inflammasome is a multiprotein complex that further regulates cell pyroptosis and inflammation by activating caspase-1. The assembly and activation of inflammasome are associated with a variety of diseases. Accumulative studies have shown that inflammasome is a key modulator of the host's defense response to viral infection. Indeed, it has been established that activation of inflammasome occurs during viral infection. At the same time, the host has evolved a variety of corresponding mechanisms to inhibit unnecessary inflammasome activation. Therefore, here, we review and summarize the latest research progress on the interaction between inflammosomes and viruses, highlight the assembly and activation of inflammosome in related cells after viral infection, as well as the corresponding molecular regulatory mechanisms, and elucidate the effects of this activation on virus immune escape and host innate and adaptive immune defenses. Finally, we also discuss the potential therapeutic strategies to prevent and/or ameliorate viral infection-related diseases via targeting inflammasomes and its products.


Assuntos
Interações entre Hospedeiro e Microrganismos , Inflamassomos , Viroses , Vírus , Humanos , Inflamassomos/imunologia , Viroses/imunologia , Viroses/terapia , Vírus/imunologia , Interações entre Hospedeiro e Microrganismos/imunologia , Animais
16.
Nat Commun ; 14(1): 3928, 2023 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-37402742

RESUMO

Tissue-resident memory (TRM) CD8+ T cells are largely derived from recently activated effector T cells, but the mechanisms that control the extent of TRM differentiation within tissue microenvironments remain unresolved. Here, using an IFNγ-YFP reporter system to identify CD8+ T cells executing antigen-dependent effector functions, we define the transcriptional consequences and functional mechanisms controlled by TCR-signaling strength that occur within the skin during viral infection to promote TRM differentiation. TCR-signaling both enhances CXCR6-mediated migration and suppresses migration toward sphingosine-1-phosphate, indicating the programming of a 'chemotactic switch' following secondary antigen encounter within non-lymphoid tissues. Blimp1 was identified as the critical target of TCR re-stimulation that is necessary to establish this chemotactic switch and for TRM differentiation to efficiently occur. Collectively, our findings show that access to antigen presentation and strength of TCR-signaling required for Blimp1 expression establishes the chemotactic properties of effector CD8+ T cells to promote residency within non-lymphoid tissues.


Assuntos
Linfócitos T CD8-Positivos , Memória Imunológica , Receptores de Antígenos de Linfócitos T , Pele , Viroses , Linfócitos T CD8-Positivos/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Pele/imunologia , Pele/virologia , Viroses/imunologia , Movimento Celular , Feminino , Animais , Camundongos , Camundongos Endogâmicos C57BL , Interferon gama/metabolismo , Fator 1 de Ligação ao Domínio I Regulador Positivo/metabolismo , Receptores CXCR6/metabolismo
17.
Virulence ; 14(1): 2223394, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37332205

RESUMO

As a member of the pattern recognition receptors (PRRs) involving in the innate immune system, Toll-like receptors (TLRs) can sense a wide range of microbial pathogens and combat infections by producing antimicrobial products, inflammatory cytokines, and chemokines. All TLRs, with the exception of TLR3, activate a signalling cascade via the myeloid differentiation primary response gene 88 (MyD88). Therefore, the activation of MyD88-dependent signalling pathway must be finely controlled. Herein, we identified that cyclin-dependent kinase 5 (CDK5) negatively regulated TLR-MyD88 signalling pathway by targeting MyD88. Overexpression of CDK5 reduced the production of interferons (IFNs), while a deficiency in CDK5 increased the expression of IFNs in response to vesicular stomatitis virus (VSV) infection. Mechanistically, CDK5 suppressed the formation of MyD88 homodimers, resulting in the attenuated production of IFNs induced by VSV infection. Surprisingly, its kinase activity does not play a role in this process. Therefore, CDK5 can act as an internal regulator to prevent excessive production of IFNs by restricting TLR-MyD88-induced activation of antiviral innate immunity in A549 cells.


Assuntos
Fator 88 de Diferenciação Mieloide , Viroses , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Diferenciação Celular , Quinase 5 Dependente de Ciclina/metabolismo , Imunidade Inata , Fator 88 de Diferenciação Mieloide/genética , Receptores Toll-Like , Viroses/imunologia
18.
Sci Adv ; 9(25): eadg2339, 2023 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-37352355

RESUMO

Stringent control of type I interferon (IFN-I) signaling is critical to potent innate immune responses against viral infection, yet the underlying molecular mechanisms are still elusive. Here, we found that Van Gogh-like 2 (VANGL2) acts as an IFN-inducible negative feedback regulator to suppress IFN-I signaling during vesicular stomatitis virus (VSV) infection. Mechanistically, VANGL2 interacted with TBK1 and promoted the selective autophagic degradation of TBK1 via K48-linked polyubiquitination at Lys372 by the E3 ligase TRIP, which serves as a recognition signal for the cargo receptor OPTN. Furthermore, myeloid-specific deletion of VANGL2 in mice showed enhanced IFN-I production against VSV infection and improved survival. In general, these findings revealed a negative feedback loop of IFN-I signaling through the VANGL2-TRIP-TBK1-OPTN axis and highlighted the cross-talk between IFN-I and autophagy in preventing viral infection. VANGL2 could be a potential clinical therapeutic target for viral infectious diseases, including COVID-19.


Assuntos
Interferon Tipo I , Proteínas Serina-Treonina Quinases , Viroses , Animais , Camundongos , Autofagia , Polaridade Celular , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição , Viroses/imunologia , Interferon Tipo I/imunologia
19.
Science ; 380(6644): 478-484, 2023 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-37141353

RESUMO

Although all multicellular organisms have germ line-encoded innate receptors to sense pathogen-associated molecular patterns, vertebrates also evolved adaptive immunity based on somatically generated antigen receptors on B and T cells. Because randomly generated antigen receptors may also react with self-antigens, tolerance checkpoints operate to limit but not completely prevent autoimmunity. These two systems are intricately linked, with innate immunity playing an instrumental role in the induction of adaptive antiviral immunity. In this work, we review how inborn errors of innate immunity can instigate B cell autoimmunity. Increased nucleic acid sensing, often resulting from defects in metabolizing pathways or retroelement control, can break B cell tolerance and converge into TLR7-, cGAS-STING-, or MAVS-dominant signaling pathways. The resulting syndromes span a spectrum that ranges from chilblain and systemic lupus to severe interferonopathies.


Assuntos
Autoimunidade , Linfócitos B , Interações Hospedeiro-Patógeno , Imunidade Inata , Viroses , Vírus , Animais , Imunidade Adaptativa , Autoimunidade/genética , Linfócitos B/imunologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata/genética , Transdução de Sinais , Viroses/imunologia , Vírus/imunologia , Humanos
20.
Cell ; 186(10): 2144-2159.e22, 2023 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-37172565

RESUMO

Bats are special in their ability to live long and host many emerging viruses. Our previous studies showed that bats have altered inflammasomes, which are central players in aging and infection. However, the role of inflammasome signaling in combating inflammatory diseases remains poorly understood. Here, we report bat ASC2 as a potent negative regulator of inflammasomes. Bat ASC2 is highly expressed at both the mRNA and protein levels and is highly potent in inhibiting human and mouse inflammasomes. Transgenic expression of bat ASC2 in mice reduced the severity of peritonitis induced by gout crystals and ASC particles. Bat ASC2 also dampened inflammation induced by multiple viruses and reduced mortality of influenza A virus infection. Importantly, it also suppressed SARS-CoV-2-immune-complex-induced inflammasome activation. Four key residues were identified for the gain of function of bat ASC2. Our results demonstrate that bat ASC2 is an important negative regulator of inflammasomes with therapeutic potential in inflammatory diseases.


Assuntos
Proteínas Reguladoras de Apoptose , Quirópteros , Inflamassomos , Ribonucleoproteínas , Viroses , Animais , Humanos , Camundongos , Proteínas Reguladoras de Apoptose/metabolismo , Quirópteros/imunologia , COVID-19 , Inflamassomos/imunologia , Ribonucleoproteínas/metabolismo , SARS-CoV-2 , Viroses/imunologia , Fenômenos Fisiológicos Virais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA