Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.410
Filtrar
1.
Nat Commun ; 15(1): 5442, 2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38937436

RESUMO

Although patients benefit from immune checkpoint inhibition (ICI) therapy in a broad variety of tumors, resistance may arise from immune suppressive tumor microenvironments (TME), which is particularly true of hepatocellular carcinoma (HCC). Since oncolytic viruses (OV) can generate a highly immune-infiltrated, inflammatory TME, OVs could potentially restore ICI responsiveness via recruitment, priming, and activation of anti-tumor T cells. Here we find that on the contrary, an oncolytic vesicular stomatitis virus, expressing interferon-ß (VSV-IFNß), antagonizes the effect of anti-PD-L1 therapy in a partially anti-PD-L1-responsive model of HCC. Cytometry by Time of Flight shows that VSV-IFNß expands dominant anti-viral effector CD8 T cells with concomitant relative disappearance of anti-tumor T cell populations, which are the target of anti-PD-L1. However, by expressing a range of HCC tumor antigens within VSV, combination OV and anti-PD-L1 therapeutic benefit could be restored. Our data provide a cautionary message for the use of highly immunogenic viruses as tumor-specific immune-therapeutics by showing that dominant anti-viral T cell responses can inhibit sub-dominant anti-tumor T cell responses. However, through encoding tumor antigens within the virus, oncolytic virotherapy can generate anti-tumor T cell populations upon which immune checkpoint blockade can effectively work.


Assuntos
Antígenos de Neoplasias , Antígeno B7-H1 , Linfócitos T CD8-Positivos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Terapia Viral Oncolítica , Vírus Oncolíticos , Microambiente Tumoral , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Animais , Terapia Viral Oncolítica/métodos , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/imunologia , Microambiente Tumoral/imunologia , Camundongos , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Humanos , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/imunologia , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Interferon beta/metabolismo , Interferon beta/imunologia , Camundongos Endogâmicos C57BL , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/farmacologia , Linfócitos T/imunologia , Feminino , Vesiculovirus/imunologia , Vesiculovirus/genética
2.
Virol Sin ; 39(3): 501-512, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38789039

RESUMO

The infection caused by porcine epidemic diarrhea virus (PEDV) is associated with high mortality in piglets worldwide. Host factors involved in the efficient replication of PEDV, however, remain largely unknown. Our recent proteomic study in the virus-host interaction network revealed a significant increase in the accumulation of CALML5 (EF-hand protein calmodulin-like 5) following PEDV infection. A further study unveiled a biphasic increase of CALML5 in 2 and 12 â€‹h after viral infection. Similar trends were observed in the intestines of piglets in the early and late stages of the PEDV challenge. Moreover, CALML5 depletion reduced PEDV mRNA and protein levels, leading to a one-order-of-magnitude decrease in virus titer. At the early stage of PEDV infection, CALML5 affected the endosomal trafficking pathway by regulating the expression of endosomal sorting complex related cellular proteins. CALML5 depletion also suppressed IFN-ß and IL-6 production in the PEDV-infected cells, thereby indicating its involvement in negatively regulating the innate immune response. Our study reveals the biological function of CALML5 in the virology field and offers new insights into the PEDV-host cell interaction.


Assuntos
Calmodulina , Endossomos , Imunidade Inata , Vírus da Diarreia Epidêmica Suína , Replicação Viral , Animais , Vírus da Diarreia Epidêmica Suína/imunologia , Vírus da Diarreia Epidêmica Suína/fisiologia , Suínos , Calmodulina/metabolismo , Calmodulina/genética , Endossomos/metabolismo , Endossomos/virologia , Interações Hospedeiro-Patógeno/imunologia , Doenças dos Suínos/virologia , Doenças dos Suínos/imunologia , Células Vero , Chlorocebus aethiops , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Infecções por Coronavirus/veterinária , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-6/imunologia , Interferon beta/genética , Interferon beta/imunologia , Interferon beta/metabolismo
3.
PLoS Pathog ; 20(5): e1012125, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38696536

RESUMO

Major 5'-terminally deleted (5'TD) RNA forms of group-B coxsackievirus (CVB-5'TD) has been associated with myocarditis in both mice and humans. Although it is known that interferon-ß (IFN-ß) signaling is critical for an efficient innate immune response against CVB-induced myocarditis, the link between CVB-5'TD RNA forms and type I IFN signaling in cardiomyocytes remains to be explored. In a mouse model of CVB3/28-induced myocarditis, major early-emerging forms of CVB-5'TD RNA have been characterized as replicative viral populations that impair IFN-ß production in the heart. Synthetic CVB3/28 RNA forms mimicking each of these major 5'TD virus populations were transfected in mice and have been shown to modulate innate immune responses in the heart and to induce myocarditis in mice. Remarkably, transfection of synthetic viral RNA with deletions in the secondary structures of the 5'-terminal CVB3 RNA domain I, modifying stem-loops "b", "c" or "d", were found to impair IFN-ß production in human cardiomyocytes. In addition, the activation of innate immune response by Poly(I:C), was found to restore IFN-ß production and to reduce the burden of CVB-5'TD RNA-forms in cardiac tissues, thereby reducing the mortality rate of infected mice. Overall, our results indicate that major early-emerging CVB3 populations deleted in the domain I of genomic RNA, in the 5' noncoding region, modulate the activation of the type I IFN pathway in cardiomyocytes and induce myocarditis in mice. These findings shed new light on the role of replicative CVB-5'TD RNA forms as key pathophysiological factors in CVB-induced human myocarditis.


Assuntos
Infecções por Coxsackievirus , Enterovirus Humano B , Interferon Tipo I , Miocardite , Miócitos Cardíacos , RNA Viral , Miocardite/virologia , Miocardite/imunologia , Miocardite/genética , Animais , Miócitos Cardíacos/virologia , Miócitos Cardíacos/metabolismo , Camundongos , Enterovirus Humano B/imunologia , Infecções por Coxsackievirus/imunologia , Infecções por Coxsackievirus/virologia , Infecções por Coxsackievirus/genética , Interferon Tipo I/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Humanos , Imunidade Inata , Transdução de Sinais , Interferon beta/metabolismo , Interferon beta/genética , Interferon beta/imunologia , Masculino , Regiões 5' não Traduzidas
4.
Exp Mol Pathol ; 137: 104897, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38691979

RESUMO

BACKGROUND: Signaling by toll-like receptors (TLRs) initiates important immune responses against viral infection. The role of TLRs in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is not well elucidated. Thus, we investigated the interaction of TLRs agonists and SARS-COV-2 antigens with immune cells in vitro. MATERIAL & METHODS: 30 coronavirus disease 2019 (COVID-19) patients (15 severe and 15 moderate) and 10 age and sex-matched healthy control (HC) were enrolled. Peripheral blood mononuclear cells (PBMCs) were isolated and activated with TLR3, 7, 8, and 9 agonists, the spike protein (SP) of SARS-CoV-2, and the receptor binding domain (RBD) of SP. Frequencies of CD3+IFN-ß+ T cells, and CD3+IFN-γ+ T cells were evaluated by flow cytometry. Interferon (IFN)-ß gene expression was assessed by qRT-PCR. RESULTS: The frequency of CD3+IFN-ß+ T cells was higher in PBMCs from moderate (p < 0.0001) and severe (p = 0.009) patients at baseline in comparison with HCs. The highest increase in the frequency of CD3+IFN-ß+ T cells in cell from moderate patients was induced by TLR8 agonist and SP (p < 0.0001 for both) when compared to HC, while, the highest increase of the frequency of CD3+IFN-ß+ T cells in sample of severe patients was seen with TLR8 and TLR7 agonists (both p = 0.002). The frequency of CD3+IFN-γ+ T cells was significantly increased upon stimulation with TLR agonists in cell from patients with moderate and severe COVID-19, compared with HC (all p < 0.01), except with TLR7 and TLR8 agonists. The TLR8 agonist did not significantly increase the frequency of CD3+IFN-γ+ T cells in PBMCs of severe patients, but did so in cells from patients with moderate disease (p = 0.01). Moreover, IFN-ß gene expression was significantly upregulated in CD3+T cells from moderate (p < 0.0001) and severe (p = 0.002) COVID-19 patients, compared to HC after stimulation with the TLR8 agonist, while, stimulation of T cells with SP, significantly up-regulated IFN-ß mRNA expression in cells from patients with moderate (p = 0.0003), but not severe disease. CONCLUSION: Stimulation of PBMCs from COVID-19 patients, especially patients with moderate disease, with TLR8 agonist and SP increased the frequency of IFN-ß-producing T cells and IFN-ß gene expression.


Assuntos
Complexo CD3 , COVID-19 , SARS-CoV-2 , Linfócitos T , Receptores Toll-Like , Humanos , COVID-19/imunologia , COVID-19/virologia , SARS-CoV-2/imunologia , Masculino , Feminino , Pessoa de Meia-Idade , Receptores Toll-Like/agonistas , Receptores Toll-Like/genética , Complexo CD3/imunologia , Complexo CD3/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/efeitos dos fármacos , Adulto , Interferon gama/metabolismo , Interferon gama/genética , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/genética , Interferon beta/genética , Interferon beta/imunologia , Idoso , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Agonistas do Receptor Semelhante a Toll
5.
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
6.
Adv Sci (Weinh) ; 11(21): e2400888, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38638003

RESUMO

Circulating tumor cells (CTCs) shed from primary tumors must overcome the cytotoxicity of immune cells, particularly natural killer (NK) cells, to cause metastasis. The tumor microenvironment (TME) protects tumor cells from the cytotoxicity of immune cells, which is partially executed by cancer-associated mesenchymal stromal cells (MSCs). However, the mechanisms by which MSCs influence the NK resistance of CTCs remain poorly understood. This study demonstrates that MSCs enhance the NK resistance of cancer cells in a gap junction-dependent manner, thereby promoting the survival and metastatic seeding of CTCs in immunocompromised mice. Tumor cells crosstalk with MSCs through an intercellular cGAS-cGAMP-STING signaling loop, leading to increased production of interferon-ß (IFNß) by MSCs. IFNß reversely enhances the type I IFN (IFN-I) signaling in tumor cells and hence the expression of human leukocyte antigen class I (HLA-I) on the cell surface, protecting the tumor cells from NK cytotoxicity. Disruption of this loop reverses NK sensitivity in tumor cells and decreases tumor metastasis. Moreover, there are positive correlations between IFN-I signaling, HLA-I expression, and NK tolerance in human tumor samples. Thus, the NK-resistant signaling loop between tumor cells and MSCs may serve as a novel therapeutic target.


Assuntos
Interferon beta , Células Matadoras Naturais , Células-Tronco Mesenquimais , Células Neoplásicas Circulantes , Nucleotidiltransferases , Transdução de Sinais , Microambiente Tumoral , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Animais , Células Matadoras Naturais/imunologia , Camundongos , Interferon beta/metabolismo , Interferon beta/imunologia , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/genética , Humanos , Células Neoplásicas Circulantes/imunologia , Células Neoplásicas Circulantes/metabolismo , Microambiente Tumoral/imunologia , Proteínas de Membrana/metabolismo , Modelos Animais de Doenças , Linhagem Celular Tumoral
7.
J Virol ; 98(5): e0192523, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38624230

RESUMO

Recurrent respiratory papillomatosis (RRP) is a rare benign tumor caused mainly by the infection of the respiratory tract epithelial cells by the human papillomavirus (HPV) type 6/11. However, the specific mechanisms underlying the inhibition of the host's innate immune response by HPV remain unclear. For this purpose, we employed single-cell RNA sequencing to analyze the states of various immune cells in RRP samples post-HPV infection and utilized a cellular model of HPV infection to elucidate the mechanisms by which HPV evades the innate immune system in RRP. The results revealed distinct immune cell heterogeneity in RRP and demonstrated that HPV11 E7 can inhibit the phosphorylation of the stimulator of interferon genes protein, thereby circumventing the body's antiviral response. In vitro co-culture experiments demonstrated that stimulation of macrophages to produce interferon-beta induced the death of HPV-infected epithelial cells, also reducing HPV viral levels. In summary, our study preliminarily identifies the potential mechanisms by which HPV evades the host's antiviral immune response, as well as the latent antiviral functions exhibited by activated macrophages. This research serves as an initial exploration of antiviral immune evasion in RRP, laying a solid foundation for investigating immunotherapeutic approaches for the disease.IMPORTANCESurgical tumor reduction is the most common treatment for recurrent respiratory papillomatosis (RRP). One of the characteristics of RRP is its persistent recurrence, and multiple surgeries are usually required to control the symptoms. Recently, some adjuvant therapies have shown effectiveness, but none of them can completely clear human papillomavirus (HPV) infection, and thus, a localized antiviral immune response is significant for disease control; after all, HPV infection is limited to the epithelium. Inhibition of interferon-beta (IFN-ß) secretion by HPV11 E7 viral proteins in epithelial cells by affecting stimulator of interferon genes phosphorylation may account for the persistence of low-risk HPV replication in the RRP. Moreover, suppression of the IFN-I pathway in RRP cell types might provide clues regarding the hyporeactive function of local immune cells. However, activation of macrophage groups to produce IFN-ß can still destroy HPV-infected cells.


Assuntos
Papillomavirus Humano 11 , Proteínas E7 de Papillomavirus , Infecções por Papillomavirus , Infecções Respiratórias , Adulto , Feminino , Humanos , Masculino , Células Epiteliais/virologia , Células Epiteliais/imunologia , Papillomavirus Humano 11/genética , Papillomavirus Humano 11/imunologia , Evasão da Resposta Imune , Imunidade Inata , Interferon beta/metabolismo , Interferon beta/imunologia , Interferon beta/genética , Macrófagos/imunologia , Macrófagos/virologia , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteínas E7 de Papillomavirus/metabolismo , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/imunologia , Infecções por Papillomavirus/imunologia , Infecções por Papillomavirus/virologia , Infecções Respiratórias/virologia , Infecções Respiratórias/imunologia
8.
Mol Immunol ; 170: 131-143, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38663254

RESUMO

Mammalian reovirus (MRV) is a non-enveloped, gene segmented double-stranded RNA (dsRNA) virus. It is an important zoonotic pathogen that infects many mammals and vertebrates that act as natural hosts and causes respiratory and digestive tract diseases. Studies have reported that RIG-I and MDA5 in the innate immune cytoplasmic RNA-sensing RIG-like receptor (RLR) signaling pathway can recognize dsRNA from MRV and promote antiviral type I interferon (IFN) responses. However, the mechanism by which many MRV-encoded proteins evade the host innate immune response remains unclear. Here, we show that exogenous µ1 protein promoted the proliferation of MRV in vitro, while knockdown of MRV µ1 protein expression by shRNA could impair MRV proliferation. Specifically, µ1 protein inhibited MRV or poly(I:C)-induced IFN-ß expression, and attenuated RIG-I/MDA5-mediated signaling axis transduction during MRV infection. Importantly, we found that µ1 protein significantly decreased IFN-ß mRNA expression induced by MDA5, RIG-I, MAVS, TBK1, IRF3(5D), and degraded the protein expression of exogenous MDA5, RIG-I, MAVS, TBK1 and IRF3 via the proteasomal and lysosomal pathways. Additionally, we show that µ1 protein can physically interact with MDA5, RIG-I, MAVS, TBK1, and IRF3 and attenuate the RIG-I/MDA5-mediated signaling cascades by blocking the phosphorylation and nuclear translocation of IRF3. In conclusion, our findings reveal that MRV outer capsid protein µ1 is a key factor in antagonizing RLRs signaling cascades and provide new strategies for effective prevention and treatment of MRV infection.


Assuntos
Proteínas do Capsídeo , Proteína DEAD-box 58 , Fator Regulador 3 de Interferon , Helicase IFIH1 Induzida por Interferon , Orthoreovirus de Mamíferos , Receptores Imunológicos , Transdução de Sinais , Animais , Humanos , Transporte Ativo do Núcleo Celular , Núcleo Celular/metabolismo , Proteína DEAD-box 58/metabolismo , Células HEK293 , Imunidade Inata/imunologia , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/metabolismo , Interferon beta/imunologia , Helicase IFIH1 Induzida por Interferon/metabolismo , Helicase IFIH1 Induzida por Interferon/genética , Orthoreovirus de Mamíferos/imunologia , Orthoreovirus de Mamíferos/fisiologia , Fosforilação , Proteínas Serina-Treonina Quinases , Infecções por Reoviridae/imunologia , Transdução de Sinais/imunologia , Proteínas Virais/metabolismo , Proteínas do Capsídeo/metabolismo
9.
J Virol ; 97(10): e0095923, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37772825

RESUMO

IMPORTANCE: Viral encephalomyelitis outcome is dependent on host responses to neuronal infection. Interferon (IFN) is an important component of the innate response, and IFN regulatory factor (IRF) 7 is an inducible transcription factor for the synthesis of IFN-α. IRF7-deficient mice develop fatal paralysis after CNS infection with Sindbis virus, while wild-type mice recover. Irf7 -/- mice produce low levels of IFN-α but high levels of IFN-ß with induction of IFN-stimulated genes, so the reason for this difference is not understood. The current study shows that Irf7 -/- mice developed inflammation earlier but failed to clear virus from motor neuron-rich regions of the brainstem and spinal cord. Levels of IFN-γ and virus-specific antibody were comparable, indicating that IRF7 deficiency does not impair expression of these known viral clearance factors. Therefore, IRF7 is either necessary for the neuronal response to currently identified mediators of clearance or enables the production of additional antiviral factor(s) needed for clearance.


Assuntos
Infecções por Alphavirus , Encefalomielite , Fator Regulador 7 de Interferon , Sindbis virus , Animais , Camundongos , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/virologia , Tronco Encefálico/virologia , Encefalomielite/imunologia , Encefalomielite/virologia , Inflamação/virologia , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/metabolismo , Interferon beta/imunologia , Interferon beta/metabolismo , Neurônios Motores/virologia , Sindbis virus/imunologia , Medula Espinal/virologia
10.
J Immunol ; 211(3): 474-485, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37326494

RESUMO

Herpetic stromal keratitis (HSK) is a painful and vision-impairing disease caused by recurrent HSV-1 infection of the cornea. The virus replication in the corneal epithelium and associated inflammation play a dominant role in HSK progression. Current HSK treatments targeting inflammation or virus replication are partially effective and promote HSV-1 latency, and long-term use can cause side effects. Thus, understanding molecular and cellular events that control HSV-1 replication and inflammation is crucial for developing novel HSK therapies. In this study, we report that ocular HSV-1 infection induces the expression of IL-27, a pleiotropic immunoregulatory cytokine. Our data indicate that HSV-1 infection stimulates IL-27 production by macrophages. Using a primary corneal HSV-1 infection mouse model and IL-27 receptor knockout mice, we show that IL-27 plays a critical role in controlling HSV-1 shedding from the cornea, the optimum induction of effector CD4+ T cell responses, and limiting HSK progression. Using in vitro bone marrow-derived macrophages, we show that IL-27 plays an antiviral role by regulating macrophage-mediated HSV-1 killing, IFN-ß production, and IFN-stimulated gene expression after HSV-1 infection. Furthermore, we report that IL-27 is critical for macrophage survival, Ag uptake, and the expression of costimulatory molecules involved in the optimum induction of effector T cell responses. Our results indicate that IL-27 promotes endogenous antiviral and anti-inflammatory responses and represents a promising target for suppressing HSK progression.


Assuntos
Córnea , Interleucinas , Ceratite Herpética , Animais , Feminino , Masculino , Camundongos , Córnea/imunologia , Córnea/virologia , Herpesvirus Humano 1 , Interferon beta/imunologia , Interleucinas/imunologia , Ceratite Herpética/imunologia , Macrófagos/imunologia , Camundongos Knockout , Eliminação de Partículas Virais , Células Th1/imunologia , Imunidade Inata
11.
Nature ; 615(7952): 490-498, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36890227

RESUMO

Metabolic rewiring underlies the effector functions of macrophages1-3, but the mechanisms involved remain incompletely defined. Here, using unbiased metabolomics and stable isotope-assisted tracing, we show that an inflammatory aspartate-argininosuccinate shunt is induced following lipopolysaccharide stimulation. The shunt, supported by increased argininosuccinate synthase (ASS1) expression, also leads to increased cytosolic fumarate levels and fumarate-mediated protein succination. Pharmacological inhibition and genetic ablation of the tricarboxylic acid cycle enzyme fumarate hydratase (FH) further increases intracellular fumarate levels. Mitochondrial respiration is also suppressed and mitochondrial membrane potential increased. RNA sequencing and proteomics analyses demonstrate that there are strong inflammatory effects resulting from FH inhibition. Notably, acute FH inhibition suppresses interleukin-10 expression, which leads to increased tumour necrosis factor secretion, an effect recapitulated by fumarate esters. Moreover, FH inhibition, but not fumarate esters, increases interferon-ß production through mechanisms that are driven by mitochondrial RNA (mtRNA) release and activation of the RNA sensors TLR7, RIG-I and MDA5. This effect is recapitulated endogenously when FH is suppressed following prolonged lipopolysaccharide stimulation. Furthermore, cells from patients with systemic lupus erythematosus also exhibit FH suppression, which indicates a potential pathogenic role for this process in human disease. We therefore identify a protective role for FH in maintaining appropriate macrophage cytokine and interferon responses.


Assuntos
Fumarato Hidratase , Interferon beta , Macrófagos , Mitocôndrias , RNA Mitocondrial , Humanos , Argininossuccinato Sintase/metabolismo , Ácido Argininossuccínico/metabolismo , Ácido Aspártico/metabolismo , Respiração Celular , Citosol/metabolismo , Fumarato Hidratase/antagonistas & inibidores , Fumarato Hidratase/genética , Fumarato Hidratase/metabolismo , Fumaratos/metabolismo , Interferon beta/biossíntese , Interferon beta/imunologia , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/metabolismo , Lúpus Eritematoso Sistêmico/enzimologia , Macrófagos/enzimologia , Macrófagos/imunologia , Macrófagos/metabolismo , Potencial da Membrana Mitocondrial , Metabolômica , Mitocôndrias/genética , Mitocôndrias/metabolismo , RNA Mitocondrial/metabolismo
12.
J Immunol ; 210(3): 283-296, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36548461

RESUMO

Foot-and-mouth disease virus (FMDV) is the causative agent of foot-and-mouth disease, one of the most highly infectious animal viruses throughout the world. The JAK-STAT signaling pathway is a highly conserved pathway for IFN-ß-induced antiviral gene expression. Previous studies have shown that FMDV can strongly suppress the innate immune response. Moreover, although STAT1 and STAT2 (STAT1/2) have been well established in JAK-STAT signaling-induced antiviral gene expression, whether FMDV proteins inhibit IFN-ß-induced JAK-STAT signaling remains poorly understood. In this study, we described the Lb leader protease (Lbpro) of FMDV as a candidate for inhibiting IFN-ß-induced signaling transduction via directly interacting with STAT1/2. We further showed that Lbpro colocalized with STAT1/2 to inhibit their nuclear translocation. Importantly, Lbpro cleaved STAT1/2 to inhibit IFN-ß-induced signal transduction, whereas the catalytically inactive mutant of LC51A (Lbpro with cysteine substituted with alanine at amino acid residue 51) had no effect on the stability of STAT1/2 proteins. The cleavage of the STAT1/2 proteins was also determined during FMDV infection in vitro. Lbpro could cleave the residues between 252 and 502 aa for STAT1 and the site spanning residues 140 - 150 aa (QQHEIESRIL) for STAT2. The in vivo results showed that Lbpro can cleave STAT1/2 in pigs. Overall, our findings suggest that FMDV Lbpro-mediated targeting of STAT1/2 may reveal a novel mechanism for viral immune evasion.


Assuntos
Endopeptidases , Vírus da Febre Aftosa , Interferon beta , Fator de Transcrição STAT1 , Fator de Transcrição STAT2 , Animais , Vírus da Febre Aftosa/enzimologia , Imunidade Inata , Peptídeo Hidrolases , Transdução de Sinais , Suínos , Interferon beta/imunologia
13.
Vet Microbiol ; 275: 109582, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36306554

RESUMO

Pseudorabies virus (PRV) is a member of the genus Varicellovirus, family Herpesviridae and causes Aujeszky's disease to lead to huge economic losses in the global pig industry. The Non-POU domain-containing octamer-binding protein (NONO), as a Drosophila behavior/human splicing (DBHS) protein, plays a key role in multiple biological functions in cells, including transcriptional regulation, RNA splicing, DNA repair and so on. However, whether swine NONO (sNONO) inhibits PRV infection is less understood. In this study, we showed that sNONO was a crucial host factor for antagonizing PRV infection and positive regulated transcription levels of ISGs. After PRV infection, sNONO enhanced the activation of IFN-ß promoter and IFN-ß expression. Furthermore, knockout of sNONO in PAM-KNU cells impaired activation of type I IFN pathway and increased PRV propagation. Taken together, we have first elucidated the anti-PRV function and mechanism of sNONO, which may provide a new strategy for preventing DNA virus infection.


Assuntos
Proteínas de Ligação a DNA , Pseudorraiva , Proteínas de Ligação a RNA , Doenças dos Suínos , Animais , Proteínas de Ligação a DNA/genética , Herpesvirus Suídeo 1 , Interferon beta/imunologia , Pseudorraiva/imunologia , Proteínas de Ligação a RNA/genética , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/virologia , Fatores de Transcrição
14.
Proc Natl Acad Sci U S A ; 119(36): e2206327119, 2022 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-36037380

RESUMO

Cerebral malaria (CM) is a life-threatening form of Plasmodium falciparum infection caused by brain inflammation. Brain endothelium dysfunction is a hallmark of CM pathology, which is also associated with the activation of the type I interferon (IFN) inflammatory pathway. The molecular triggers and sensors eliciting brain type I IFN cellular responses during CM remain largely unknown. We herein identified the stimulator of interferon response cGAMP interactor 1 (STING1) as the key innate immune sensor that induces Ifnß1 transcription in the brain of mice infected with Plasmodium berghei ANKA (Pba). This STING1/IFNß-mediated response increases brain CXCL10 governing the extent of brain leukocyte infiltration and blood-brain barrier (BBB) breakdown, and determining CM lethality. The critical role of brain endothelial cells (BECs) in fueling type I IFN-driven brain inflammation was demonstrated in brain endothelial-specific IFNß-reporter and STING1-deficient Pba-infected mice, which were significantly protected from CM lethality. Moreover, extracellular particles (EPs) released from Pba-infected erythrocytes activated the STING1-dependent type I IFN response in BECs, a response requiring intracellular acidification. Fractionation of the EPs enabled us to identify a defined fraction carrying hemoglobin degradation remnants that activates STING1/IFNß in the brain endothelium, a process correlated with heme content. Notably, stimulation of STING1-deficient BECs with heme, docking experiments, and in vitro binding assays unveiled that heme is a putative STING1 ligand. This work shows that heme resultant from the parasite heterotrophic activity operates as an alarmin, triggering brain endothelial inflammatory responses via the STING1/IFNß/CXCL10 axis crucial to CM pathogenesis and lethality.


Assuntos
Encéfalo , Heme , Interferon beta , Malária Cerebral , Proteínas de Membrana , Animais , Encéfalo/parasitologia , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Células Endoteliais/parasitologia , Endotélio/imunologia , Endotélio/parasitologia , Heme/metabolismo , Interferon beta/imunologia , Malária Cerebral/imunologia , Malária Cerebral/parasitologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Plasmodium berghei/metabolismo , Ativação Transcricional/imunologia
15.
Proc Natl Acad Sci U S A ; 119(31): e2201146119, 2022 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-35878041

RESUMO

Aberrant immune responses, including hyperresponsiveness to Toll-like receptor (TLR) ligands, underlie acute respiratory distress syndrome (ARDS). Type I interferons confer antiviral activities and could also regulate the inflammatory response, whereas little is known about their actions to resolve aberrant inflammation. Here we report that interferon-ß (IFN-ß) exerts partially overlapping, but also cooperative actions with aspirin-triggered 15-epi-lipoxin A4 (15-epi-LXA4) and 17-epi-resolvin D1 to counter TLR9-generated cues to regulate neutrophil apoptosis and phagocytosis in human neutrophils. In mice, TLR9 activation impairs bacterial clearance, prolongs Escherichia coli-evoked lung injury, and suppresses production of IFN-ß and the proresolving lipid mediators 15-epi-LXA4 and resolvin D1 (RvD1) in the lung. Neutralization of endogenous IFN-ß delays pulmonary clearance of E. coli and aggravates mucosal injury. Conversely, treatment of mice with IFN-ß accelerates clearance of bacteria, restores neutrophil phagocytosis, promotes neutrophil apoptosis and efferocytosis, and accelerates resolution of airway inflammation with concomitant increases in 15-epi-LXA4 and RvD1 production in the lungs. Pharmacological blockade of the lipoxin receptor ALX/FPR2 partially prevents IFN-ß-mediated resolution. These findings point to a pivotal role of IFN-ß in orchestrating timely resolution of neutrophil and TLR9 activation-driven airway inflammation and uncover an IFN-ß-initiated resolution program, activation of an ALX/FPR2-centered, proresolving lipids-mediated circuit, for ARDS.


Assuntos
Interferon beta , Lipoxinas , Síndrome do Desconforto Respiratório , Animais , Ácidos Docosa-Hexaenoicos/farmacologia , Ácidos Docosa-Hexaenoicos/uso terapêutico , Escherichia coli , Infecções por Escherichia coli/imunologia , Humanos , Inflamação/tratamento farmacológico , Interferon beta/imunologia , Interferon beta/farmacologia , Lipoxinas/farmacologia , Camundongos , Receptores de Formil Peptídeo/antagonistas & inibidores , Síndrome do Desconforto Respiratório/tratamento farmacológico , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/imunologia , Ativação Transcricional/efeitos dos fármacos
16.
Cell Rep ; 39(13): 110989, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35767946

RESUMO

The interleukin-12 (IL-12) family comprises the only heterodimeric cytokines mediating diverse functional effects. We previously reported a striking bimodal IL-12p70 response to lipopolysaccharide (LPS) stimulation in healthy donors. Herein, we demonstrate that interferon ß (IFNß) is a major upstream determinant of IL-12p70 production, which is also associated with numbers and activation of circulating monocytes. Integrative modeling of proteomic, genetic, epigenomic, and cellular data confirms IFNß as key for LPS-induced IL-12p70 and allowed us to compare the relative effects of each of these parameters on variable cytokine responses. Clinical relevance of our findings is supported by reduced IFNß-IL-12p70 responses in patients hospitalized with acute severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or chronically infected with hepatitis C (HCV). Importantly, these responses are resolved after viral clearance. Our systems immunology approach defines a better understanding of IL-12p70 and IFNß in healthy and infected persons, providing insights into how common genetic and epigenetic variation may impact immune responses to bacterial infection.


Assuntos
Interferon beta , Interleucina-12 , Receptor 4 Toll-Like , COVID-19/imunologia , COVID-19/metabolismo , COVID-19/virologia , Citocinas/imunologia , Citocinas/metabolismo , Humanos , Interferon beta/imunologia , Interferon beta/metabolismo , Interleucina-12/imunologia , Interleucina-12/metabolismo , Lipopolissacarídeos/farmacologia , Proteômica , SARS-CoV-2/imunologia
17.
Mol Cell Proteomics ; 21(7): 100247, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35594991

RESUMO

Since the discovery of oncogenes, there has been tremendous interest to understand their mechanistic basis and to develop broadly actionable therapeutics. Some of the most frequently activated oncogenes driving diverse cancers are c-MYC, EGFR, HER2, AKT, KRAS, BRAF, and MEK. Using a reductionist approach, we explored how cellular proteomes are remodeled in isogenic cell lines engineered with or without these driver oncogenes. The most striking discovery for all oncogenic models was the systematic downregulation of scores of antiviral proteins regulated by type 1 interferon. These findings extended to cancer cell lines and patient-derived xenograft models of highly refractory pancreatic cancer and osteosarcoma driven by KRAS and MYC oncogenes. The oncogenes reduced basal expression of and autocrine stimulation by type 1 interferon causing remarkable convergence on common phenotypic and functional profiles. In particular, there was dramatically lower expression of dsRNA sensors including DDX58 (RIG-I) and OAS proteins, which resulted in attenuated functional responses when the oncogenic cells were treated with the dsRNA mimetic, polyI:C, and increased susceptibility to infection with an RNA virus shown using SARS-CoV-2. Our reductionist approach provides molecular and functional insights connected to immune evasion hallmarks in cancers and suggests therapeutic opportunities.


Assuntos
COVID-19 , Interferon beta , Oncogenes , Proteômica , Animais , Fatores de Restrição Antivirais , COVID-19/imunologia , Carcinogênese , Linhagem Celular Tumoral , Humanos , Interferon beta/imunologia , Proteínas Proto-Oncogênicas p21(ras)/genética , SARS-CoV-2
18.
Microbiol Spectr ; 10(1): e0188321, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35196784

RESUMO

Virus infection triggers intricate signal cascade reactions to activate the host innate immunity, which leads to the production of type I interferon (IFN-I). Herpes simplex virus 1 (HSV-1), a human-restricted pathogen, is capable of encoding over 80 viral proteins, and several of them are involved in immune evasion to resist the host antiviral response through the IFN-I signaling pathway. Here, we determined that HSV-1 UL31, which is associated with nuclear matrix and is essential for the formation of viral nuclear egress complex, could inhibit retinoic acid-inducible gene I (RIG-I)-like receptor pathway-mediated interferon beta (IFN-ß)-luciferase (Luc) and (PRDIII-I)4-Luc (an expression plasmid of IFN-ß positive regulatory elements III and I) promoter activation, as well as the mRNA transcription of IFN-ß and downstream interferon-stimulated genes (ISGs), such as ISG15, ISG54, ISG56, etc., to promote viral infection. UL31 was shown to restrain IFN-ß activation at the interferon regulatory factor 3 (IRF3)/IRF7 level. Mechanically, UL31 was demonstrated to interact with TANK binding kinase 1 (TBK1), inducible IκB kinase (IKKi), and IRF3 to impede the formation of the IKKi-IRF3 complex but not the formation of the IRF7-related complex. UL31 could constrain the dimerization and nuclear translocation of IRF3. Although UL31 was associated with the CREB binding protein (CBP)/p300 coactivators, it could not efficiently hamper the formation of the CBP/p300-IRF3 complex. In addition, UL31 could facilitate the degradation of IKKi and IRF3 by mediating their K48-linked polyubiquitination. Taken together, these results illustrated that UL31 was able to suppress IFN-ß activity by inhibiting the activation of IKKi and IRF3, which may contribute to the knowledge of a new immune evasion mechanism during HSV-1 infection. IMPORTANCE The innate immune system is the first line of host defense against the invasion of pathogens. Among its mechanisms, IFN-I is an essential cytokine in the antiviral response, which can help the host eliminate a virus. HSV-1 is a double-stranded DNA virus that can cause herpes and establish a lifelong latent infection, due to its possession of multiple mechanisms to escape host innate immunity. In this study, we illustrate for the first time that the HSV-1-encoded UL31 protein has a negative regulatory effect on IFN-ß production by blocking the dimerization and nuclear translocation of IRF3, as well as promoting the K48-linked polyubiquitination and degradation of both IKKi and IRF3. This study may be helpful for fully understanding the pathogenesis of HSV-1.


Assuntos
Herpesvirus Humano 1/genética , Herpesvirus Humano 1/imunologia , Interferon beta/genética , Interferon beta/imunologia , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Animais , Chlorocebus aethiops , Citocinas , Proteína DEAD-box 58 , Células HEK293 , Células HeLa , Herpes Simples , Interações Hospedeiro-Patógeno , Humanos , Evasão da Resposta Imune , Imunidade Inata , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon , Interferon Tipo I , Interferon beta/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases , Receptores Imunológicos , Transdução de Sinais , Células Vero , Proteínas Virais/metabolismo
19.
Virology ; 567: 77-86, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35032866

RESUMO

Type-I interferon (IFN-I) signals exert a critical role in disease progression during viral infections. However, the immunomodulatory mechanisms by which IFN-I dictates disease outcomes remain to be fully defined. Here we report that IFN-I signals mediate thymic atrophy in viral infections, with more severe and prolonged loss of thymic output and unique kinetics and subtypes of IFN-α/ß expression in chronic infection compared to acute infection. Loss of thymic output was linked to inhibition of early stages of thymopoiesis (DN1-DN2 transition, and DN3 proliferation) and pronounced apoptosis during the late DP stage. Notably, infection-associated thymic defects were largely abrogated upon ablation of IFNαßR and partially mitigated in the absence of CD8 T cells, thus implicating direct as well as indirect effects of IFN-I on thymocytes. These findings provide mechanistic underpinnings for immunotherapeutic strategies targeting IFN-1 signals to manipulate disease outcomes during chronic infections and cancers.


Assuntos
Atrofia/virologia , Interferon-alfa/imunologia , Interferon beta/imunologia , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/imunologia , Timócitos/virologia , Timo/virologia , Animais , Atrofia/genética , Atrofia/imunologia , Atrofia/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Doença Crônica , Feminino , Regulação da Expressão Gênica , Humanos , Memória Imunológica , Interferon-alfa/genética , Interferon beta/genética , Linfonodos/imunologia , Linfonodos/patologia , Linfonodos/virologia , Depleção Linfocítica , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/patologia , Vírus da Coriomeningite Linfocítica/patogenicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/imunologia , Transdução de Sinais/imunologia , Análise de Célula Única , Timócitos/imunologia , Timócitos/patologia , Timo/imunologia , Timo/patologia
20.
Nat Commun ; 13(1): 105, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013224

RESUMO

Zika virus (ZIKV) infection can be associated with neurological pathologies, such as microcephaly in newborns and Guillain-Barre syndrome in adults. Effective therapeutics are currently not available. As such, a comprehensive understanding of virus-host interactions may guide the development of medications for ZIKV. Here we report a human genome-wide overexpression screen to identify host factors that regulate ZIKV infection and find TMEM120A as a ZIKV restriction factor. TMEM120A overexpression significantly inhibits ZIKV replication, while TMEM120A knockdown increases ZIKV infection in cell lines. Moreover, Tmem120a knockout in mice facilitates ZIKV infection in primary mouse embryonic fibroblasts (MEF) cells. Mechanistically, the antiviral activity of TMEM120A is dependent on STING, as TMEM120A interacts with STING, promotes the translocation of STING from the endoplasmic reticulum (ER) to ER-Golgi intermediate compartment (ERGIC) and enhances the phosphorylation of downstream TBK1 and IRF3, resulting in the expression of multiple antiviral cytokines and interferon-stimulated genes. In summary, our gain-of-function screening identifies TMEM120A as a key activator of the antiviral signaling of STING.


Assuntos
Interações Hospedeiro-Patógeno/genética , Canais Iônicos/genética , Proteínas de Membrana/genética , Infecção por Zika virus/genética , Zika virus/genética , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Linhagem Celular Tumoral , Retículo Endoplasmático/genética , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/virologia , Feminino , Regulação da Expressão Gênica , Complexo de Golgi/genética , Complexo de Golgi/imunologia , Complexo de Golgi/virologia , Hepatócitos/imunologia , Hepatócitos/virologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Interferon beta/genética , Interferon beta/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Canais Iônicos/deficiência , Canais Iônicos/imunologia , Proteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Fosforilação , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/imunologia , Transdução de Sinais , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Zika virus/crescimento & desenvolvimento , Zika virus/patogenicidade , Infecção por Zika virus/imunologia , Infecção por Zika virus/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...