Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Mol Cell ; 71(5): 745-760.e5, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30193098

RESUMO

DNA damage can be sensed as a danger-associated molecular pattern by the innate immune system. Here we find that keratinocytes and other human cells mount an innate immune response within hours of etoposide-induced DNA damage, which involves the DNA sensing adaptor STING but is independent of the cytosolic DNA receptor cGAS. This non-canonical activation of STING is mediated by the DNA binding protein IFI16, together with the DNA damage response factors ATM and PARP-1, resulting in the assembly of an alternative STING signaling complex that includes the tumor suppressor p53 and the E3 ubiquitin ligase TRAF6. TRAF6 catalyzes the formation of K63-linked ubiquitin chains on STING, leading to the activation of the transcription factor NF-κB and the induction of an alternative STING-dependent gene expression program. We propose that STING acts as a signaling hub that coordinates a transcriptional response depending on its mode of activation.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/genética , Núcleo Celular/genética , Dano ao DNA/genética , Proteínas de Membrana/genética , NF-kappa B/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Transdução de Sinais/genética , Linhagem Celular , Citosol/metabolismo , DNA/genética , Células HEK293 , Humanos , Imunidade Inata/genética , Queratinócitos/fisiologia , Poli(ADP-Ribose) Polimerase-1/genética , Proteína Supressora de Tumor p53/genética , Ubiquitina/genética , Ubiquitina-Proteína Ligases/genética
2.
Immunology ; 157(2): 163-172, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30919991

RESUMO

It is well understood that the STING signalling pathway is critical for generating a robust innate immune response to pathogens. Human and mouse STING signalling pathways are not identical, however. For example, mice lack IFI16, which has been proven important for the human STING pathway. Therefore, we investigated whether humanized mice are an appropriate experimental platform for exploring the human STING signalling cascade in vivo. We found that NOG mice reconstituted with human cord blood haematopoietic stem cells (humanized NOG mice) exhibit human STING signalling responses to an analogue of the cyclic di-nucleotide cGAMP. There was an increase in the proportions of monocytes in the lungs of mice receiving cGAMP analogue. The most robust levels of STING expression and STING-induced responses were observed in mice exhibiting the highest levels of human chimerization. Notably, differential levels of STING in lung versus spleen following cGAMP analogue treatment suggest that there are tissue-specific kinetics of STING activation and/or degradation in effector versus inductive sites. We also examined the mouse innate immune response to cGAMP analogue treatment. We detected that mouse cells in the immunodeficient NOG mice responded to the cGAMP analogue and they do so with distinct kinetics from the human response. In conclusion, humanized NOG mice represent a valuable experimental model for examining in vivo human STING responses.


Assuntos
Proteínas de Membrana/imunologia , Nucleotídeos Cíclicos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Feminino , Humanos , Camundongos , Camundongos Endogâmicos NOD , Proteínas Nucleares/imunologia , Fosfoproteínas/imunologia
3.
EMBO Rep ; 18(10): 1707-1715, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28801534

RESUMO

Cytosolic DNA stimulates innate immune responses, including type I interferons (IFN), which have antiviral and immunomodulatory activities. Cyclic GMP-AMP synthase (cGAS) recognizes cytoplasmic DNA and signals via STING to induce IFN production. Despite the importance of DNA in innate immunity, the nature of the DNA that stimulates IFN production is not well described. Using low DNA concentrations, we show that dsDNA induces IFN in a length-dependent manner. This is observed over a wide length-span of DNA, ranging from the minimal stimulatory length to several kilobases, and is fully dependent on cGAS irrespective of DNA length. Importantly, in vitro studies reveal that long DNA activates recombinant human cGAS more efficiently than short DNA, showing that length-dependent DNA recognition is an intrinsic property of cGAS independent of accessory proteins. Collectively, this work identifies long DNA as the molecular entity stimulating the cGAS pathway upon cytosolic DNA challenge such as viral infections.


Assuntos
DNA/química , DNA/imunologia , Interferon Tipo I/biossíntese , Nucleotidiltransferases/metabolismo , Linhagem Celular , Citosol/imunologia , Citosol/metabolismo , DNA/genética , DNA/metabolismo , Ativação Enzimática , Humanos , Imunidade Inata , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Proteínas de Membrana/metabolismo , Nucleotidiltransferases/genética , Transdução de Sinais
4.
Antimicrob Agents Chemother ; 59(7): 3984-94, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25896701

RESUMO

Adjunct therapy with the histone deacetylase inhibitor (HDACi) romidepsin increases plasma viremia in HIV patients on combination antiretroviral therapy (cART). However, a potential concern is that reversing HIV latency with an HDACi may reactivate the virus in anatomical compartments with suboptimal cART concentrations, leading to de novo infection of susceptible cells in these sites. We tested physiologically relevant romidepsin concentrations known to reactivate latent HIV in order to definitively address this concern. We found that romidepsin significantly inhibited HIV infection in peripheral blood mononuclear cells and CD4(+) T cells but not in monocyte-derived macrophages. In addition, romidepsin impaired HIV spreading in CD4(+) T cell cultures. When we evaluated the impact of romidepsin on quantitative viral outgrowth assays with primary resting CD4(+) T cells, we found that resting CD4(+) T cells exposed to romidepsin exhibited reduced proliferation and viability. This significantly lowered assay sensitivity when measuring the efficacy of romidepsin as an HIV latency reversal agent. Altogether, our data indicate that romidepsin-based HIV eradication strategies are unlikely to reseed a latent T cell reservoir, even under suboptimal cART conditions, because romidepsin profoundly restricts de novo HIV infections.


Assuntos
Depsipeptídeos/uso terapêutico , Infecções por HIV/tratamento farmacológico , HIV-1 , Inibidores de Histona Desacetilases/uso terapêutico , Antivirais/farmacologia , Linfócitos T CD4-Positivos/virologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Infecções por HIV/virologia , Humanos , Interferon gama/farmacologia , Monócitos/virologia , Cultura Primária de Células , Latência Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
5.
J Extracell Vesicles ; 12(8): e12350, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37525396

RESUMO

A key phenomenon in cancer is the establishment of a highly immunosuppressive tumour microenvironment (TME). Despite advances in immunotherapy, where the purpose is to induce tumour recognition and hence hereof tumour eradication, the majority of patients applicable for such treatment still fail to respond. It has been suggested that high immunological activity in the tumour is essential for achieving effective response to immunotherapy, which therefore have led to exploration of strategies that triggers inflammatory pathways. Here activation of the stimulator of interferon genes (STING) signalling pathway has been considered an attractive target, as it is a potent trigger of pro-inflammatory cytokines and types I and III interferons. However, immunotherapy combined with targeted STING agonists has not yielded sustained clinical remission in humans. This suggests a need for exploring novel adjuvants to improve the innate immunological efficacy. Here, we demonstrate that extracellular vesicles (EVs), derived from activated CD4+ T cells (T-EVs), sensitizes macrophages to elevate STING activation, mediated by IFNγ carried on the T-EVs. Our work support that T-EVs can disrupt the immune suppressive environment in the tumour by reprogramming macrophages to a pro-inflammatory phenotype, and priming them for a robust immune response towards STING activation.


Assuntos
Vesículas Extracelulares , Neoplasias , Humanos , Linfócitos T , Vesículas Extracelulares/metabolismo , Interferons/genética , Interferons/metabolismo , Imunoterapia , Macrófagos/metabolismo , Neoplasias/metabolismo , Microambiente Tumoral
6.
J Interferon Cytokine Res ; 39(4): 191-204, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30855198

RESUMO

Incoming viruses challenge the cell with diverse foreign molecules, which need to be sensed quickly to initiate immune responses and to remove the viral components. In this study, we investigate the cellular requirements for sensing and degradation of incoming viral DNA and capsids during herpes simplex virus type 1 (HSV-1) infections. Using click chemistry labeling of the viral genome, we found that HSV-1 DNA was released from a subset of capsids into the cytosol early in infection. By next-generation sequencing of cyclic GMP-AMP (cGAMP) synthase (cGAS)-bound DNA from HSV-1-infected cells, we show that HSV-1 DNA was bound by the cytosolic DNA sensor cGAS. Activation of cGAS enzymatic activity by viral DNA did not require proteasomal activity, indicating that viral DNA release into the cytosol is not proteasome-dependent. However, induction of interferon (IFN)-ß expression was blocked by inhibition of the proteasome, suggesting a contribution of the proteasome to IFN-ß induction through the cGAS-stimulator of interferon genes pathway. Viral DNA was cleared from the cytosol within few hours, in a manner dependent on TREX1 and a cGAS-dependent process. Capsid material in the cytoplasm was also degraded rapidly. This was partially blocked by treatment with a proteasome inhibitor. This treatment led to accumulation of DNA-containing viral capsids near the nucleus and reduced nuclear entry of viral DNA. Thus, cells infected with HSV-1 use a panel of mechanisms to eliminate viral DNA and capsids. This represents a barrier for establishment of infection and potentially enables the host to gear the IFN-ß response to a level required for antiviral defense without causing immunopathology.


Assuntos
Capsídeo/imunologia , DNA Viral/imunologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/imunologia , Animais , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Células Vero , Replicação Viral/genética , Replicação Viral/imunologia
7.
Cell Host Microbe ; 25(6): 858-872.e13, 2019 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-31175045

RESUMO

The interferon γ-inducible protein 16 (IFI16) is known as immune sensor of retroviral DNA intermediates. We show that IFI16 restricts HIV-1 independently of immune sensing by binding and inhibiting the host transcription factor Sp1 that drives viral gene expression. This antiretroviral activity and ability to bind Sp1 require the N-terminal pyrin domain and nuclear localization of IFI16, but not the HIN domains involved in DNA binding. Highly prevalent clade C HIV-1 strains are more resistant to IFI16 and less dependent on Sp1 than other HIV-1 subtypes. Furthermore, inhibition of Sp1 by IFI16 or pharmacologically by Mithramycin A suppresses reactivation of latent HIV-1 in CD4+ T cells. Finally, IFI16 also inhibits retrotransposition of LINE-1, known to engage Sp1, and murine IFI16 homologs restrict Friend retrovirus replication in mice. Thus, IFI16 restricts retroviruses and retrotransposons by interfering with Sp1-dependent gene expression, and evasion from this restriction may facilitate spread of HIV-1 subtype C.


Assuntos
HIV-1/imunologia , Fatores Imunológicos/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Fator de Transcrição Sp1/antagonistas & inibidores , Transcrição Gênica , Ativação Viral , Latência Viral , Animais , Linfócitos T CD4-Positivos/virologia , Células Cultivadas , Genótipo , HIV-1/classificação , HIV-1/genética , HIV-1/crescimento & desenvolvimento , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA