Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.259
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(29): e2320709121, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-38985760

RESUMO

The Type-I interferon (IFN-I) response is the major outcome of stimulator of interferon genes (STING) activation in innate cells. STING is more abundantly expressed in adaptive T cells; nevertheless, its intrinsic function in T cells remains unclear. Intriguingly, we previously demonstrated that STING activation in T cells activates widespread IFN-independent activities, which stands in contrast to the well-known STING-mediated IFN response. Here, we have identified that STING activation induces regulatory T cells (Tregs) differentiation independently of IRF3 and IFN. Specifically, the translocation of STING from the endoplasmic reticulum to the Golgi activates mitogen-activated protein kinase (MAPK) activity, which subsequently triggers transcription factor cAMP response element-binding protein (CREB) activation. The activation of the STING-MAPK-CREB signaling pathway induces the expression of many cytokine genes, including interleukin-2 (IL-2) and transforming growth factor-beta 2 (TGF-ß2), to promote the Treg differentiation. Genetic knockdown of MAPK p38 or pharmacological inhibition of MAPK p38 or CREB markedly inhibits STING-mediated Treg differentiation. Administration of the STING agonist also promotes Treg differentiation in mice. In the Trex1-/- autoimmune disease mouse model, we demonstrate that intrinsic STING activation in CD4+ T cells can drive Treg differentiation, potentially counterbalancing the autoimmunity associated with Trex1 deficiency. Thus, STING-MAPK-CREB represents an IFN-independent signaling axis of STING that may have profound effects on T cell effector function and adaptive immunity.


Assuntos
Diferenciação Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Proteínas de Membrana , Linfócitos T Reguladores , Animais , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Camundongos , Transdução de Sinais , Sistema de Sinalização das MAP Quinases , Camundongos Endogâmicos C57BL , Transporte Proteico , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/genética , Camundongos Knockout , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
2.
Mol Cell ; 84(13): 2423-2435.e5, 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38917796

RESUMO

The innate immune cGAS-STING pathway is activated by cytosolic double-stranded DNA (dsDNA), a ubiquitous danger signal, to produce interferon, a potent anti-viral and anti-cancer cytokine. However, STING activation must be tightly controlled because aberrant interferon production leads to debilitating interferonopathies. Here, we discover PELI2 as a crucial negative regulator of STING. Mechanistically, PELI2 inhibits the transcription factor IRF3 by binding to phosphorylated Thr354 and Thr356 on the C-terminal tail of STING, leading to ubiquitination and inhibition of the kinase TBK1. PELI2 sets a threshold for STING activation that tolerates low levels of cytosolic dsDNA, such as that caused by silenced TREX1, RNASEH2B, BRCA1, or SETX. When this threshold is reached, such as during viral infection, STING-induced interferon production temporarily downregulates PELI2, creating a positive feedback loop allowing a robust immune response. Lupus patients have insufficient PELI2 levels and high basal interferon production, suggesting that PELI2 dysregulation may drive the onset of lupus and other interferonopathies.


Assuntos
Fator Regulador 3 de Interferon , Proteínas de Membrana , Proteínas Serina-Treonina Quinases , Transdução de Sinais , Ubiquitinação , Humanos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Fosforilação , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/genética , Animais , Células HEK293 , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Lúpus Eritematoso Sistêmico/virologia , Imunidade Inata , Interações Hospedeiro-Patógeno , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Camundongos , Interferons/metabolismo , Interferons/imunologia , Interferons/genética , Retroalimentação Fisiológica , Camundongos Endogâmicos C57BL , Exodesoxirribonucleases , Fosfoproteínas
3.
Vet Microbiol ; 295: 110148, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38851152

RESUMO

Water buffalo Hunnivirus (BufHuV) belongs to the family Picornaviridae and is a newly discovered member of the Hunnivirus A genus. It causes intestinal diseases in cattle, mainly lead to subclinical infections, thereby seriously threatening the health of cattle herds. In addition, it can also bring about various clinical disease syndromes which results in severe economic losses to the cattle industry. To date, there have been no reports worldwide on the study of Hunnivirus virus infecting host cells and causing innate immune responses. In this study, we found that interferon treatment effectively blocked BufHuV replication and infection with the virus weakened the host antiviral responses. Inhibiting the transcription of IFN-ß and ISGs induced by either Sendai virus (SeV) or poly(I:C) in MDBK and HCT-8 cells, were dependent on the IRF3 or NF-κB signaling pathways, and this inhibited the activation of IFN-ß promoter by TBK1 and its upstream molecules, RIGI and MDA5. By constructing and screening five BufHuV proteins, we found that VP2, 2 C, 3 C and 3D inhibited the activation of IFN-ß promoter induced by SeV. Subsequently, we showed that VP2 inhibited the activation of IRF3 induced by SeV or poly (I:C), and it inhibited IRF3 activation by inhibiting its phosphorylation and nuclear translocation. In addition, we confirmed that VP2 inhibited the activation of IFNß induced by signaling molecules, MDA5 and TBKI. In summary, these findings provide new insights into the pathogenesis of Hunnivirus and its mechanisms involved in evading host immune responses.


Assuntos
Fator Regulador 3 de Interferon , Interferon beta , Interferon beta/genética , Interferon beta/imunologia , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/genética , Animais , Humanos , Linhagem Celular , Transdução de Sinais/efeitos dos fármacos , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/metabolismo , Replicação Viral/efeitos dos fármacos , Imunidade Inata , Bovinos , Búfalos/virologia , NF-kappa B/metabolismo
4.
Biomolecules ; 14(6)2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38927097

RESUMO

MicroRNAs (miRNAs) are highly conserved endogenous single-stranded non-coding RNA molecules that play a crucial role in regulating gene expression to maintain normal physiological functions in fish. Nevertheless, the specific physiological role of miRNAs in lower vertebrates, particularly in comparison to mammals, remains elusive. Additionally, the mechanisms underlying the control of antiviral responses triggered by viral stimulation in fish are still not fully understood. In this study, we investigated the regulatory impact of miR-1388 on the signaling pathway mediated by IFN regulatory factor 3 (IRF3). Our findings revealed that following stimulation with the viral analog poly(I:C), the expression of miR-1388 was significantly upregulated in primary immune tissues and macrophages. Through a dual luciferase reporter assay, we corroborated a direct targeting relationship between miR-1388 and tumor necrosis factor receptor (TNFR)-associated factor 3 (TRAF3). Furthermore, our study demonstrated a distinct negative post-transcriptional correlation between miR-1388 and TRAF3. We observed a significant negative post-transcriptional regulatory association between miR-1388 and the levels of antiviral genes following poly(I:C) stimulation. Utilizing reporter plasmids, we elucidated the role of miR-1388 in the antiviral signaling pathway activated by TRAF3. By intervening with siRNA-TRAF3, we validated that miR-1388 regulates the expression of antiviral genes and the production of type I interferons (IFN-Is) through its interaction with TRAF3. Collectively, our experiments highlight the regulatory influence of miR-1388 on the IRF3-mediated signaling pathway by targeting TRAF3 post poly(I:C) stimulation. These findings provide compelling evidence for enhancing our understanding of the mechanisms through which fish miRNAs participate in immune responses.


Assuntos
Carpas , MicroRNAs , Poli I-C , Fator 3 Associado a Receptor de TNF , Animais , MicroRNAs/genética , MicroRNAs/metabolismo , Poli I-C/farmacologia , Carpas/genética , Carpas/metabolismo , Carpas/virologia , Fator 3 Associado a Receptor de TNF/genética , Fator 3 Associado a Receptor de TNF/metabolismo , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Transdução de Sinais
5.
Dev Comp Immunol ; 158: 105208, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38834141

RESUMO

Interferon regulatory factors (IRFs) are transcription factors involved in immune responses, such as pathogen response regulation, immune cell growth, and differentiation. IRFs are necessary for the synthesis of type I interferons through a signaling cascade when pathogen recognition receptors identify viral DNA or RNA. We discovered that irf3 is expressed in the early embryonic stages and in all immune organs of adult zebrafish. We demonstrated the antiviral immune mechanism of Irf3 against viral hemorrhagic septicemia virus (VHSV) using CRISPR/Cas9-mediated knockout zebrafish (irf3-KO). In this study, we used a truncated Irf3 protein, encoded by irf3 with a 10 bp deletion, for further investigation. Upon VHSV injection, irf3-KO zebrafish showed dose-dependent high and early mortality compared with zebrafish with the wild-type Irf3 protein (WT), confirming the antiviral activity of Irf3. Based on the results of expression analysis of downstream genes upon VHSV challenge, we inferred that Irf3 deficiency substantially affects the expression of ifnphi1 and ifnphi2. However, after 5 days post infection (dpi), ifnphi3 expression was not significantly altered in irf3-KO compared to that in WT, and irf7 transcription showed a considerable increase in irf3-KO after 5 dpi, indicating irf7's control over ifnphi3 expression. The significantly reduced expression of isg15, viperin, mxa, and mxb at 3 dpi also supported the effect of Irf3 deficiency on the antiviral activity in the early stage of infection. The higher mortality in irf3-KO zebrafish than in WT might be due to an increased inflammation and tissue damage that occurs in irf3-KO because of delayed immune response. Our results suggest that Irf3 plays a role in antiviral immunity of zebrafish by modulating critical immune signaling molecules and regulating antiviral immune genes.


Assuntos
Sistemas CRISPR-Cas , Técnicas de Inativação de Genes , Septicemia Hemorrágica Viral , Fator Regulador 3 de Interferon , Novirhabdovirus , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Peixe-Zebra/genética , Peixe-Zebra/imunologia , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Novirhabdovirus/fisiologia , Novirhabdovirus/imunologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Septicemia Hemorrágica Viral/imunologia , Septicemia Hemorrágica Viral/genética , Septicemia Hemorrágica Viral/virologia , Animais Geneticamente Modificados , Doenças dos Peixes/imunologia , Doenças dos Peixes/virologia , Doenças dos Peixes/genética , Imunidade Inata/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Infecções por Rhabdoviridae/imunologia , Infecções por Rhabdoviridae/virologia , Modelos Animais de Doenças , Interferons
6.
Adv Immunol ; 162: 1-21, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38866436

RESUMO

Double stranded DNA (dsDNA) in the cytoplasm triggers the cGAS-STING innate immune pathway to defend against pathogenic infections, tissue damage and malignant cells. Extensive structural and functional studies over the last couple of years have enabled the molecular understanding of dsDNA induced activation of the cGAS-STING signaling pathway. This review highlights recent advances in the structural characterization of key molecules in the cGAS-STING signaling axis by focusing on the mechanism of cGAS activation by dsDNA, the regulation of cGAS activity, the mechanism of STING activation by cGAMP, the molecular basis of TBK1 recruitment and activation by STING, the structural basis of IRF3 recruitment by STING, and the mechanism of IRF3 activation upon phosphorylation by TBK1. These comprehensive structural studies provide a detailed picture of the mechanism of the cGAS-STING signaling pathway, establishing a molecular framework for the development of novel therapeutic strategies targeting this pathway.


Assuntos
DNA , Imunidade Inata , Proteínas de Membrana , Nucleotidiltransferases , Transdução de Sinais , Humanos , Nucleotidiltransferases/metabolismo , DNA/metabolismo , DNA/imunologia , Proteínas de Membrana/metabolismo , Animais , Fator Regulador 3 de Interferon/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fosforilação
7.
Emerg Microbes Infect ; 13(1): 2372344, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38916407

RESUMO

The Orthopoxvirus (OPXV) genus of the Poxviridae includes human pathogens variola virus (VARV), monkeypox virus (MPXV), vaccinia virus (VACV), and a number of zoonotic viruses. A number of Bcl-2-like proteins of VACV are involved in escaping the host innate immunity. However, little work has been devoted to the evolution and function of their orthologues in other OPXVs. Here, we found that MPXV protein P2, encoded by the P2L gene, and P2 orthologues from other OPXVs, such as VACV protein N2, localize to the nucleus and antagonize interferon (IFN) production. Exceptions to this were the truncated P2 orthologues in camelpox virus (CMLV) and taterapox virus (TATV) that lacked the nuclear localization signal (NLS). Mechanistically, the NLS of MPXV P2 interacted with karyopherin α-2 (KPNA2) to facilitate P2 nuclear translocation, and competitively inhibited KPNA2-mediated IRF3 nuclear translocation and downstream IFN production. Deletion of the NLS in P2 or orthologues significantly enhanced IRF3 nuclear translocation and innate immune responses, thereby reducing viral replication. Moreover, deletion of NLS from N2 in VACV attenuated viral replication and virulence in mice. These data demonstrate that the NLS-mediated translocation of P2 is critical for P2-induced inhibition of innate immunity. Our findings contribute to an in-depth understanding of the mechanisms of OPXV P2 orthologue in innate immune evasion.


Assuntos
Imunidade Inata , Fator Regulador 3 de Interferon , Monkeypox virus , Sinais de Localização Nuclear , Proteínas Virais , Animais , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/genética , Camundongos , Humanos , Proteínas Virais/genética , Proteínas Virais/metabolismo , Proteínas Virais/imunologia , Sinais de Localização Nuclear/genética , Monkeypox virus/genética , Monkeypox virus/imunologia , Células HEK293 , alfa Carioferinas/genética , alfa Carioferinas/metabolismo , Evasão da Resposta Imune , Núcleo Celular/metabolismo , Interferons/genética , Interferons/imunologia , Interferons/metabolismo , Infecções por Poxviridae/imunologia , Infecções por Poxviridae/virologia , Infecções por Poxviridae/veterinária , Camundongos Endogâmicos C57BL
8.
Front Immunol ; 15: 1401086, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38903507

RESUMO

The mitochondrial anti-viral signaling (MAVS) protein is an intermediary adaptor protein of retinoic acid-inducible gene-1 (RIG-I) like receptor (RLR) signaling, which activates the transcription factor interferon (IFN) regulatory factor 3 (IRF3) and NF-kB to produce type I IFNs. MAVS expression has been reported in different fish species, but few studies have shown its functional role in anti-viral responses to fish viruses. In this study, we used the transcription activator-like effector nuclease (TALEN) as a gene editing tool to disrupt the function of MAVS in Chinook salmon (Oncorhynchus tshawytscha) embryonic cells (CHSE) to understand its role in induction of interferon I responses to infections with the (+) RNA virus salmonid alphavirus subtype 3 (SAV-3), and the dsRNA virus infectious pancreatic necrosis virus (IPNV) infection. A MAVS-disrupted CHSE clone with a 7-aa polypeptide (GVFVSRV) deletion mutation at the N-terminal of the CARD domain infected with SAV-3 resulted in significantly lower expression of IRF3, IFNa, and ISGs and increased viral titer (1.5 log10) compared to wild-type. In contrast, the IPNV titer in MAVS-disrupted cells was not different from the wild-type. Furthermore, overexpression of salmon MAVS in MAVS-disrupted CHSE cells rescued the impaired type I IFN-mediated anti-viral effect against SAV-3.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Infecções por Alphavirus , Alphavirus , Doenças dos Peixes , Vírus da Necrose Pancreática Infecciosa , Transdução de Sinais , Replicação Viral , Animais , Vírus da Necrose Pancreática Infecciosa/fisiologia , Vírus da Necrose Pancreática Infecciosa/imunologia , Alphavirus/imunologia , Alphavirus/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Doenças dos Peixes/imunologia , Doenças dos Peixes/virologia , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/virologia , Salmão/virologia , Salmão/imunologia , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Proteínas de Peixes/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/genética , Infecções por Birnaviridae/imunologia , Infecções por Birnaviridae/veterinária , Infecções por Birnaviridae/virologia
9.
Fish Shellfish Immunol ; 150: 109611, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38734119

RESUMO

During virus-host co-evolution, viruses have developed multiple strategies to dampen IFN response and prevent its antiviral activity in host cells. To date, the interactions between host IFN response and the immune evasion strategies exploited by fish iridoviruses still remain largely uncertain. Here, a potential immune evasion protein candidate of Singapore grouper iridovirus (SGIV), VP82 (encoded by SGIV ORF82) was screened and its roles during viral replication were investigated in detail. Firstly, VP82 overexpression dramatically decreased IFN or ISRE promoter activity and the transcription levels of IFN stimulated genes (ISGs) stimulated by grouper cyclic GMP-AMP synthase (EccGAS)/stimulator of interferon genes (EcSTING), TANK-binding kinase 1 (EcTBK1), IFN regulatory factor 3 (EcIRF3)and EcIRF7. Secondly, Co-IP assays indicated that VP82 interacted with EcIRF3 and EcIRF7, but not EcSTING and EcTBK1, which was consistent with the co-localization between VP82 and EcIRF3 or EcIRF7. Furthermore, VP82 promoted the degradation of EcIRF3 and EcIRF7 in a dose-dependent manner via the autophagy pathway. Finally, VP82 overexpression accelerated SGIV replication, evidenced by the increased transcriptions of viral core genes and viral production. Moreover, the antiviral action of EcIRF3 or EcIRF7 was significantly depressed in VP82 overexpressed cells. Together, VP82 was speculated to exert crucial roles for SGIV replication by inhibiting the IFN response via the degradation of IRF3 and IRF7. Our findings provided new insights into understanding the immune evasion strategies utilized by fish iridovirus through IFN regulation.


Assuntos
Infecções por Vírus de DNA , Doenças dos Peixes , Proteínas de Peixes , Fator Regulador 3 de Interferon , Fator Regulador 7 de Interferon , Ranavirus , Proteínas Virais , Animais , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/metabolismo , Fator Regulador 7 de Interferon/imunologia , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Proteínas de Peixes/metabolismo , Doenças dos Peixes/imunologia , Doenças dos Peixes/virologia , Infecções por Vírus de DNA/imunologia , Infecções por Vírus de DNA/veterinária , Ranavirus/fisiologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Imunidade Inata/genética , Interferons/genética , Interferons/imunologia , Interferons/metabolismo , Evasão da Resposta Imune , Bass/imunologia , Bass/genética , Replicação Viral , Proteínas de Peixe-Zebra , Fatores Reguladores de Interferon
10.
Nat Commun ; 15(1): 4099, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38816352

RESUMO

Chronic inflammation is a major cause of cancer worldwide. Interleukin 33 (IL-33) is a critical initiator of cancer-prone chronic inflammation; however, its induction mechanism by environmental causes of chronic inflammation is unknown. Herein, we demonstrate that Toll-like receptor (TLR)3/4-TBK1-IRF3 pathway activation links environmental insults to IL-33 induction in the skin and pancreas inflammation. An FDA-approved drug library screen identifies pitavastatin to effectively suppress IL-33 expression by blocking TBK1 membrane recruitment/activation through the mevalonate pathway inhibition. Accordingly, pitavastatin prevents chronic pancreatitis and its cancer sequela in an IL-33-dependent manner. The IRF3-IL-33 axis is highly active in chronic pancreatitis and its associated pancreatic cancer in humans. Interestingly, pitavastatin use correlates with a significantly reduced risk of chronic pancreatitis and pancreatic cancer in patients. Our findings demonstrate that blocking the TBK1-IRF3-IL-33 signaling axis suppresses cancer-prone chronic inflammation. Statins present a safe and effective prophylactic strategy to prevent chronic inflammation and its cancer sequela.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Fator Regulador 3 de Interferon , Interleucina-33 , Neoplasias Pancreáticas , Proteínas Serina-Treonina Quinases , Quinolinas , Transdução de Sinais , Interleucina-33/metabolismo , Animais , Fator Regulador 3 de Interferon/metabolismo , Humanos , Neoplasias Pancreáticas/prevenção & controle , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/genética , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Camundongos , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Quinolinas/farmacologia , Quinolinas/uso terapêutico , Inflamação/prevenção & controle , Inflamação/metabolismo , Pancreatite Crônica/prevenção & controle , Pancreatite Crônica/metabolismo , Receptor 3 Toll-Like/metabolismo , Camundongos Endogâmicos C57BL , Receptor 4 Toll-Like/metabolismo , Ácido Mevalônico/metabolismo , Masculino , Feminino , Camundongos Knockout
11.
Mol Immunol ; 170: 156-169, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38692097

RESUMO

Type-I and -III interferons play a central role in immune rejection of pathogens and tumors, thus promoting immunogenicity and suppressing tumor recurrence. Double strand RNA is an important ligand that stimulates tumor immunity via interferon responses. Differentiation of embryonic stem cells to pluripotent epithelial cells activates the interferon response during development, raising the question of whether epithelial vs. mesenchymal gene signatures in cancer potentially regulate the interferon pathway as well. Here, using genomics and signaling approaches, we show that Grainyhead-like-2 (GRHL2), a master programmer of epithelial cell identity, promotes type-I and -III interferon responses to double-strand RNA. GRHL2 enhanced the activation of IRF3 and relA/NF-kB and the expression of IRF1; a functional GRHL2 binding site in the IFNL1 promoter was also identified. Moreover, time to recurrence in breast cancer correlated positively with GRHL2 protein expression, indicating that GRHL2 is a tumor recurrence suppressor, consistent with its enhancement of interferon responses. These observations demonstrate that epithelial cell identity supports interferon responses in the context of cancer.


Assuntos
Neoplasias da Mama , Proteínas de Ligação a DNA , Fatores de Transcrição , Animais , Feminino , Humanos , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Regulação Neoplásica da Expressão Gênica , Fator Regulador 1 de Interferon/metabolismo , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/imunologia , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/genética , Interferons/metabolismo , Interferons/imunologia , Interferons/genética , Recidiva Local de Neoplasia/imunologia , RNA de Cadeia Dupla/imunologia , Transdução de Sinais/imunologia , Fator de Transcrição RelA/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia
12.
Nat Commun ; 15(1): 4646, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38821928

RESUMO

AgRP neurons in the arcuate nucleus of the hypothalamus (ARC) coordinate homeostatic changes in appetite associated with fluctuations in food availability and leptin signaling. Identifying the relevant transcriptional regulatory pathways in these neurons has been a priority, yet such attempts have been stymied due to their low abundance and the rich cellular diversity of the ARC. Here we generated AgRP neuron-specific transcriptomic and chromatin accessibility profiles from male mice during three distinct hunger states of satiety, fasting-induced hunger, and leptin-induced hunger suppression. Cis-regulatory analysis of these integrated datasets enabled the identification of 18 putative hunger-promoting and 29 putative hunger-suppressing transcriptional regulators in AgRP neurons, 16 of which were predicted to be transcriptional effectors of leptin. Within our dataset, Interferon regulatory factor 3 (IRF3) emerged as a leading candidate mediator of leptin-induced hunger-suppression. Measures of IRF3 activation in vitro and in vivo reveal an increase in IRF3 nuclear occupancy following leptin administration. Finally, gain- and loss-of-function experiments in vivo confirm the role of IRF3 in mediating the acute satiety-evoking effects of leptin in AgRP neurons. Thus, our findings identify IRF3 as a key mediator of the acute hunger-suppressing effects of leptin in AgRP neurons.


Assuntos
Fome , Fator Regulador 3 de Interferon , Leptina , Neurônios , Animais , Masculino , Camundongos , Proteína Relacionada com Agouti/metabolismo , Proteína Relacionada com Agouti/genética , Núcleo Arqueado do Hipotálamo/metabolismo , Cromatina , Epigênese Genética , Jejum , Regulação da Expressão Gênica , Fome/fisiologia , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/genética , Leptina/metabolismo , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Transdução de Sinais , Transcriptoma
13.
Sci Adv ; 10(22): eadk5011, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38809975

RESUMO

Healthy behavioral patterns could modulate organ functions to enhance the body's immunity. However, how exercise regulates antiviral innate immunity remains elusive. Here, we found that exercise promotes type I interferon (IFN-I) production in the liver and enhances IFN-I immune activity of the body. Despite the possibility that many exercise-induced factors could affect IFN-I production, we identified Gpld1 as a crucial molecule, and the liver as the major organ to promote IFN-I production after exercise. Exercise largely loses the efficiency to induce IFN-I in Gpld1-/- mice. Further studies demonstrated that exercise-produced 3-hydroxybutanoic acid (3-HB) critically induces Gpld1 expression in the liver. Gpld1 blocks the PP2A-IRF3 interaction, thus enhancing IRF3 activation and IFN-I production, and eventually improving the body's antiviral ability. This study reveals that exercise improves antiviral innate immunity by linking the liver metabolism to systemic IFN-I activity and uncovers an unknown function of liver cells in innate immunity.


Assuntos
Imunidade Inata , Fator Regulador 3 de Interferon , Interferon Tipo I , Fígado , Condicionamento Físico Animal , Animais , Masculino , Camundongos , Antivirais , Citocinas , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/metabolismo , Fígado/metabolismo , Fígado/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Ubiquitinas , Glicosilfosfatidilinositol Diacilglicerol-Liase/metabolismo
14.
Nat Immunol ; 25(6): 981-993, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38811816

RESUMO

Viral infection makes us feel sick as the immune system alters systemic metabolism to better fight the pathogen. The extent of these changes is relative to the severity of disease. Whether blood glucose is subject to infection-induced modulation is mostly unknown. Here we show that strong, nonlethal infection restricts systemic glucose availability, which promotes the antiviral type I interferon (IFN-I) response. Following viral infection, we find that IFNγ produced by γδ T cells stimulates pancreatic ß cells to increase glucose-induced insulin release. Subsequently, hyperinsulinemia lessens hepatic glucose output. Glucose restriction enhances IFN-I production by curtailing lactate-mediated inhibition of IRF3 and NF-κB signaling. Induced hyperglycemia constrained IFN-I production and increased mortality upon infection. Our findings identify glucose restriction as a physiological mechanism to bring the body into a heightened state of responsiveness to viral pathogens. This immune-endocrine circuit is disrupted in hyperglycemia, possibly explaining why patients with diabetes are more susceptible to viral infection.


Assuntos
Glicemia , Imunidade Inata , Interferon gama , Animais , Interferon gama/metabolismo , Interferon gama/imunologia , Camundongos , Glicemia/metabolismo , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , Camundongos Endogâmicos C57BL , Transdução de Sinais/imunologia , Insulina/metabolismo , Insulina/imunologia , Camundongos Knockout , Hiperglicemia/imunologia , Fator Regulador 3 de Interferon/metabolismo , NF-kappa B/metabolismo , Humanos , Fígado/imunologia , Fígado/virologia , Fígado/metabolismo , Masculino
15.
Immunobiology ; 229(3): 152810, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38772101

RESUMO

BACKGROUND AND AIMS: Activation of the cGAS-STING pathway induces the production of type I interferons, initiating the antiviral immune response, which contributes to the clearance of pathogens. Previous studies have shown that STING agonists promote hepatitis B virus (HBV) clearance; however, few studies have investigated the effect of activating the cGAS-STING pathway in macrophages on HBV. METHODS: The polarization status of HBV particle-stimulated RAW264.7 macrophages was analyzed. After stimulation with HBV particles, the analysis focused on determining whether the DNA sensors in RAW264.7 macrophages recognized the viral double-stranded DNA (dsDNA) and evaluating the activation of the cGAS-STING pathway. Coculture of mouse macrophages and hepatocytes harboring HBV was used to study the antiviral activity of HBV-stimulated RAW264.7 macrophages. RESULTS: After stimulation with HBV particles, HBV relaxed circular DNA (rcDNA) was detected in RAW264.7 macrophages, and the protein expression of phospho-STING, phospho-TBK1, and phospho-IRF3 in the STING pathway was increased, as shown by Western blot analysis, which revealed that M1 polarization of macrophages was caused by increased expression of CD86. RT-PCR analyses revealed elevated expression of M1 macrophage polarization-associated cytokines such as TNFα, IL-1ß, iNOS, and IFNα/ß. In the coculture experiment, both HBsAg and HBeAg expression levels were significantly decreased in AML12-HBV1.3 cells cocultured with the supernatants of HBV-stimulated RAW264.7 macrophages. CONCLUSION: The results suggest that macrophages can endocytose HBV particles. Additionally, viral dsDNA can be recognized by DNA pattern recognition receptors, which in turn activate the cGAS-STING pathway, promoting the M1 polarization of macrophages, while no significant M2 polarization is observed. Macrophages stimulated with HBV particles exhibit enhanced antiviral activity against HBV.


Assuntos
DNA Viral , Vírus da Hepatite B , Macrófagos , Proteínas de Membrana , Nucleotidiltransferases , Transdução de Sinais , Vírus da Hepatite B/fisiologia , Vírus da Hepatite B/imunologia , Animais , Nucleotidiltransferases/metabolismo , Camundongos , Macrófagos/imunologia , Macrófagos/virologia , Macrófagos/metabolismo , Proteínas de Membrana/metabolismo , Células RAW 264.7 , Hepatite B/imunologia , Hepatite B/virologia , Humanos , Ativação de Macrófagos/imunologia , Hepatócitos/virologia , Hepatócitos/imunologia , Hepatócitos/metabolismo , Fator Regulador 3 de Interferon/metabolismo
16.
Int Immunopharmacol ; 134: 112185, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38701540

RESUMO

Chronic ethanol consumption is a prevalent condition in contemporary society and exacerbates anxiety symptoms in healthy individuals. The activation of microglia, leading to neuroinflammatory responses, may serve as a significant precipitating factor; however, the precise molecular mechanisms underlying this phenomenon remain elusive. In this study, we initially confirmed that chronic ethanol exposure (CEE) induces anxiety-like behaviors in mice through open field test and elevated plus maze test. The cGAS/STING signaling pathway has been confirmed to exhibits a significant association with inflammatory signaling responses in both peripheral and central systems. Western blot analysis confirmed alterations in the cGAS/STING signaling pathway during CEE, including the upregulation of p-TBK1 and p-IRF3 proteins. Moreover, we observed microglial activation in the prefrontal cortex (PFC) of CEE mice, characterized by significant alterations in branching morphology and an increase in cell body size. Additionally, we observed that administration of CEE resulted in mitochondrial dysfunction within the PFC of mice, accompanied by a significant elevation in cytosolic mitochondrial DNA (mtDNA) levels. Furthermore, our findings revealed that the inhibition of STING by H-151 effectively alleviated anxiety-like behavior and suppressed microglial activation induced by CEE. Our study unveiled a significant association between anxiety-like behavior, microglial activation, inflammation, and mitochondria dysfunction during CEE.


Assuntos
Ansiedade , Etanol , Proteínas de Membrana , Camundongos Endogâmicos C57BL , Microglia , Nucleotidiltransferases , Córtex Pré-Frontal , Transdução de Sinais , Animais , Microglia/efeitos dos fármacos , Microglia/metabolismo , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/genética , Ansiedade/induzido quimicamente , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Etanol/toxicidade , Transdução de Sinais/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/patologia , Masculino , Camundongos , Comportamento Animal/efeitos dos fármacos , DNA Mitocondrial/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Modelos Animais de Doenças , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/genética , Proteínas Serina-Treonina Quinases
17.
Front Immunol ; 15: 1380220, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38799458

RESUMO

African swine fever (ASF) is an acute hemorrhagic and devastating infectious disease affecting domestic pigs and wild boars. It is caused by the African swine fever virus (ASFV), which is characterized by genetic diversity and sophisticated immune evasion strategies. To facilitate infection, ASFV encodes multiple proteins to antagonize host innate immune responses, thereby contributing to viral virulence and pathogenicity. The molecular mechanisms employed by ASFV-encoded proteins to modulate host antiviral responses have not been comprehensively elucidated. In this study, it was observed that the ASFV MGF505-6R protein, a member of the multigene family 505 (MGF505), effectively suppressed the activation of the interferon-beta (IFN-ß) promoter, leading to reduced mRNA levels of antiviral genes. Additional evidence has revealed that MGF505-6R antagonizes the cGAS-STING signaling pathway by interacting with the stimulator of interferon genes (STING) for degradation in the autophagy-lysosomal pathway. The domain mapping revealed that the N-terminal region (1-260aa) of MGF505-6R is the primary domain responsible for interacting with STING, while the CTT domain of STING is crucial for its interaction with MGF505-6R. Furthermore, MGF505-6R also inhibits the activation of STING by reducing the K63-linked polyubiquitination of STING, leading to the disruption of STING oligomerization and TANK binding kinase 1 (TBK1) recruitment, thereby impairing the phosphorylation and nuclear translocation of interferon regulatory factor 3 (IRF3). Collectively, our study elucidates a novel strategy developed by ASFV MGF505-6R to counteract host innate immune responses. This discovery may offer valuable insights for further exploration of ASFV immune evasion mechanisms and antiviral strategies.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Proteínas de Membrana , Proteínas Virais , Animais , Vírus da Febre Suína Africana/imunologia , Vírus da Febre Suína Africana/genética , Suínos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Febre Suína Africana/imunologia , Febre Suína Africana/virologia , Febre Suína Africana/metabolismo , Proteínas Virais/imunologia , Proteínas Virais/metabolismo , Proteínas Virais/genética , Humanos , Imunidade Inata , Interferon Tipo I/metabolismo , Interferon Tipo I/imunologia , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/imunologia , Transdução de Sinais , Proteólise , Células HEK293 , Interações Hospedeiro-Patógeno/imunologia , Evasão da Resposta Imune , Interferon beta/metabolismo , Interferon beta/imunologia , Interferon beta/genética
18.
Mol Cell ; 84(11): 2203-2213.e5, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38749421

RESUMO

The cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway plays a pivotal role in innate immune responses to viral infection and inhibition of autoimmunity. Recent studies have suggested that micronuclei formed by genotoxic stress can activate innate immune signaling via the cGAS-STING pathway. Here, we investigated cGAS localization, activation, and downstream signaling from micronuclei induced by ionizing radiation, replication stress, and chromosome segregation errors. Although cGAS localized to ruptured micronuclei via binding to self-DNA, we failed to observe cGAS activation; cGAMP production; downstream phosphorylation of STING, TBK1, or IRF3; nuclear accumulation of IRF3; or expression of interferon-stimulated genes. Failure to activate the cGAS-STING pathway was observed across primary and immortalized cell lines, which retained the ability to activate the cGAS-STING pathway in response to dsDNA or modified vaccinia virus infection. We provide evidence that micronuclei formed by genotoxic insults contain histone-bound self-DNA, which we show is inhibitory to cGAS activation in cells.


Assuntos
Segregação de Cromossomos , Proteínas de Membrana , Micronúcleos com Defeito Cromossômico , Nucleotídeos Cíclicos , Nucleotidiltransferases , Transdução de Sinais , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/genética , Humanos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Micronúcleos com Defeito Cromossômico/efeitos da radiação , Nucleotídeos Cíclicos/metabolismo , Fosforilação , Replicação do DNA/efeitos da radiação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/genética , Imunidade Inata/efeitos da radiação , Dano ao DNA , Células HEK293 , Animais , Radiação Ionizante , Células HeLa
19.
Arterioscler Thromb Vasc Biol ; 44(6): 1365-1378, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38695170

RESUMO

BACKGROUND: Macrophages play a crucial role in atherosclerotic plaque formation, and the death of macrophages is a vital factor in determining the fate of atherosclerosis. GSDMD (gasdermin D)-mediated pyroptosis is a programmed cell death, characterized by membrane pore formation and inflammatory factor release. METHODS: ApoE-/- and Gsdmd-/- ApoE-/- mice, bone marrow transplantation, and AAV (adeno-associated virus serotype 9)-F4/80-shGSDMD (shRNA-GSDMD) were used to examine the effect of macrophage-derived GSDMD on atherosclerosis. Single-cell RNA sequencing was used to investigate the changing profile of different cellular components and the cellular localization of GSDMD during atherosclerosis. RESULTS: First, we found that GSDMD is activated in human and mouse atherosclerotic plaques and Gsdmd-/- attenuates the atherosclerotic lesion area in high-fat diet-fed ApoE-/- mice. We performed single-cell RNA sequencing of ApoE-/- and Gsdmd-/- ApoE-/- mouse aortas and showed that GSDMD is principally expressed in atherosclerotic macrophages. Using bone marrow transplantation and AAV-F4/80-shGSDMD, we identified the potential role of macrophage-derived GSDMD in aortic pyroptosis and atherosclerotic injuries in vivo. Mechanistically, GSDMD contributes to mitochondrial perforation and mitochondrial DNA leakage and subsequently activates the STING (stimulator of interferon gene)-IRF3 (interferon regulatory factor 3)/NF-κB (nuclear factor kappa B) axis. Meanwhile, GSDMD regulates the STING pathway activation and macrophage migration via cytokine secretion. Inhibition of GSDMD with GSDMD-specific inhibitor GI-Y1 (GSDMD inhibitor Y1) can effectively alleviate the progression of atherosclerosis. CONCLUSIONS: Our study has provided a novel macrophage-derived GSDMD mechanism in the promotion of atherosclerosis and demonstrated that GSDMD can be a potential therapeutic target for atherosclerosis.


Assuntos
Aterosclerose , Modelos Animais de Doenças , Fator Regulador 3 de Interferon , Peptídeos e Proteínas de Sinalização Intracelular , Macrófagos , Proteínas de Membrana , Camundongos Endogâmicos C57BL , Mitocôndrias , NF-kappa B , Proteínas de Ligação a Fosfato , Piroptose , Transdução de Sinais , Animais , Aterosclerose/metabolismo , Aterosclerose/patologia , Aterosclerose/genética , Macrófagos/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteínas de Ligação a Fosfato/metabolismo , Proteínas de Ligação a Fosfato/genética , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/genética , Camundongos , NF-kappa B/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos Knockout para ApoE , Placa Aterosclerótica , Doenças da Aorta/patologia , Doenças da Aorta/metabolismo , Doenças da Aorta/genética , Doenças da Aorta/prevenção & controle , Gasderminas
20.
PLoS Pathog ; 20(5): e1012227, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38739631

RESUMO

IFN regulatory factor 3 (IRF3) is the transcription factor crucial for the production of type I IFN in viral defence and inflammatory responses. The activity of IRF3 is strictly modulated by post-translational modifications (PTMs) to effectively protect the host from infection while avoiding excessive immunopathology. Here, we report that zebrafish myosin-regulated light chain interacting protein b (mylipb) inhibits virus-induced type I IFN production via two synergistic mechanisms: induction of autophagic degradation of irf3 and reduction of irf3 phosphorylation. In vivo, mylipb-null zebrafish exhibit reduced lethality and viral mRNA levels compared to controls. At the cellular level, overexpression of mylipb significantly reduces cellular antiviral capacity, and promotes viral proliferation. Mechanistically, mylipb associates with irf3 and targets Lys 352 to increase K6-linked polyubiquitination, dependent on its E3 ubiquitin ligase activity, leading to autophagic degradation of irf3. Meanwhile, mylipb acts as a decoy substrate for the phosphokinase tbk1 to attenuate irf3 phosphorylation and cellular antiviral responses independent of its enzymatic activity. These findings support a critical role for zebrafish mylipb in the limitation of antiviral innate immunity through two synergistic mechanisms targeting irf3.


Assuntos
Imunidade Inata , Fator Regulador 3 de Interferon , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Fator Regulador 3 de Interferon/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Infecções por Rhabdoviridae/imunologia , Fosforilação , Ubiquitinação , Humanos , Autofagia/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA