Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.406
Filtrar
1.
PLoS Pathog ; 20(5): e1012125, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38696536

RESUMEN

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.


Asunto(s)
Infecciones por Coxsackievirus , Enterovirus Humano B , Interferón Tipo I , Miocarditis , Miocitos Cardíacos , ARN Viral , Miocarditis/virología , Miocarditis/inmunología , Miocarditis/genética , Animales , Miocitos Cardíacos/virología , Miocitos Cardíacos/metabolismo , Ratones , Enterovirus Humano B/inmunología , Infecciones por Coxsackievirus/inmunología , Infecciones por Coxsackievirus/virología , Infecciones por Coxsackievirus/genética , Interferón Tipo I/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Humanos , Inmunidad Innata , Transducción de Señal , Interferón beta/metabolismo , Interferón beta/genética , Interferón beta/inmunología , Masculino , Regiones no Traducidas 5'
2.
Front Immunol ; 15: 1380220, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38799458

RESUMEN

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.


Asunto(s)
Virus de la Fiebre Porcina Africana , Fiebre Porcina Africana , Proteínas de la Membrana , Proteínas Virales , Animales , Virus de la Fiebre Porcina Africana/inmunología , Virus de la Fiebre Porcina Africana/genética , Porcinos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Fiebre Porcina Africana/inmunología , Fiebre Porcina Africana/virología , Fiebre Porcina Africana/metabolismo , Proteínas Virales/inmunología , Proteínas Virales/metabolismo , Proteínas Virales/genética , Humanos , Inmunidad Innata , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/inmunología , Transducción de Señal , Proteolisis , Células HEK293 , Interacciones Huésped-Patógeno/inmunología , Evasión Inmune , Interferón beta/metabolismo , Interferón beta/inmunología , Interferón beta/genética
3.
J Virol ; 98(5): e0192523, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38624230

RESUMEN

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.


Asunto(s)
Papillomavirus Humano 11 , Inmunidad Innata , Interferón beta , Macrófagos , Proteínas de la Membrana , Infecciones por Papillomavirus , Infecciones del Sistema Respiratorio , Interferón beta/metabolismo , Interferón beta/inmunología , Interferón beta/genética , Humanos , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/virología , Papillomavirus Humano 11/genética , Papillomavirus Humano 11/inmunología , Infecciones del Sistema Respiratorio/virología , Infecciones del Sistema Respiratorio/inmunología , Macrófagos/inmunología , Macrófagos/virología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Femenino , Células Epiteliales/virología , Células Epiteliales/inmunología , Evasión Inmune , Proteínas E7 de Papillomavirus/metabolismo , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/inmunología , Masculino , Adulto
4.
Mol Immunol ; 170: 131-143, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38663254

RESUMEN

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.


Asunto(s)
Proteína 58 DEAD Box , Factor 3 Regulador del Interferón , Helicasa Inducida por Interferón IFIH1 , Orthoreovirus de los Mamíferos , Receptores Inmunológicos , Transducción de Señal , Helicasa Inducida por Interferón IFIH1/metabolismo , Helicasa Inducida por Interferón IFIH1/genética , Factor 3 Regulador del Interferón/metabolismo , Proteína 58 DEAD Box/metabolismo , Transducción de Señal/inmunología , Humanos , Fosforilación , Orthoreovirus de los Mamíferos/inmunología , Orthoreovirus de los Mamíferos/fisiología , Células HEK293 , Interferón beta/metabolismo , Interferón beta/inmunología , Animales , Núcleo Celular/metabolismo , Infecciones por Reoviridae/inmunología , Proteínas Virales/metabolismo , Transporte Activo de Núcleo Celular , Inmunidad Innata/inmunología , Proteínas Serina-Treonina Quinasas
5.
J Virol ; 97(10): e0095923, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37772825

RESUMEN

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.


Asunto(s)
Infecciones por Alphavirus , Encefalomielitis , Factor 7 Regulador del Interferón , Virus Sindbis , Animales , Ratones , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/virología , Tronco Encefálico/virología , Encefalomielitis/inmunología , Encefalomielitis/virología , Inflamación/virología , Factor 7 Regulador del Interferón/deficiencia , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Interferón beta/inmunología , Interferón beta/metabolismo , Neuronas Motoras/virología , Virus Sindbis/inmunología , Médula Espinal/virología
6.
J Immunol ; 211(3): 474-485, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37326494

RESUMEN

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.


Asunto(s)
Córnea , Interleucinas , Queratitis Herpética , Animales , Femenino , Masculino , Ratones , Córnea/inmunología , Córnea/virología , Herpesvirus Humano 1 , Interferón beta/inmunología , Interleucinas/inmunología , Queratitis Herpética/inmunología , Macrófagos/inmunología , Ratones Noqueados , Esparcimiento de Virus , Células TH1/inmunología , Inmunidad Innata
7.
Nature ; 615(7952): 490-498, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36890227

RESUMEN

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.


Asunto(s)
Fumarato Hidratasa , Interferón beta , Macrófagos , Mitocondrias , ARN Mitocondrial , Humanos , Argininosuccinato Sintasa/metabolismo , Ácido Argininosuccínico/metabolismo , Ácido Aspártico/metabolismo , Respiración de la Célula , Citosol/metabolismo , Fumarato Hidratasa/antagonistas & inhibidores , Fumarato Hidratasa/genética , Fumarato Hidratasa/metabolismo , Fumaratos/metabolismo , Interferón beta/biosíntesis , Interferón beta/inmunología , Lipopolisacáridos/farmacología , Lipopolisacáridos/metabolismo , Lupus Eritematoso Sistémico/enzimología , Macrófagos/enzimología , Macrófagos/inmunología , Macrófagos/metabolismo , Potencial de la Membrana Mitocondrial , Metabolómica , Mitocondrias/genética , Mitocondrias/metabolismo , ARN Mitocondrial/metabolismo
8.
J Immunol ; 210(3): 283-296, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36548461

RESUMEN

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.


Asunto(s)
Endopeptidasas , Virus de la Fiebre Aftosa , Interferón beta , Factor de Transcripción STAT1 , Factor de Transcripción STAT2 , Animales , Virus de la Fiebre Aftosa/enzimología , Inmunidad Innata , Péptido Hidrolasas , Transducción de Señal , Porcinos , Interferón beta/inmunología
9.
Vet Microbiol ; 275: 109582, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36306554

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN , Seudorrabia , Proteínas de Unión al ARN , Enfermedades de los Porcinos , Animales , Proteínas de Unión al ADN/genética , Herpesvirus Suido 1 , Interferón beta/inmunología , Seudorrabia/inmunología , Proteínas de Unión al ARN/genética , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/virología , Factores de Transcripción
10.
Proc Natl Acad Sci U S A ; 119(36): e2206327119, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-36037380

RESUMEN

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.


Asunto(s)
Encéfalo , Hemo , Interferón beta , Malaria Cerebral , Proteínas de la Membrana , Animales , Encéfalo/parasitología , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Células Endoteliales/parasitología , Endotelio/inmunología , Endotelio/parasitología , Hemo/metabolismo , Interferón beta/inmunología , Malaria Cerebral/inmunología , Malaria Cerebral/parasitología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Plasmodium berghei/metabolismo , Activación Transcripcional/inmunología
11.
Proc Natl Acad Sci U S A ; 119(31): e2201146119, 2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35878041

RESUMEN

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.


Asunto(s)
Interferón beta , Lipoxinas , Síndrome de Dificultad Respiratoria , Animales , Ácidos Docosahexaenoicos/farmacología , Ácidos Docosahexaenoicos/uso terapéutico , Escherichia coli , Infecciones por Escherichia coli/inmunología , Humanos , Inflamación/tratamiento farmacológico , Interferón beta/inmunología , Interferón beta/farmacología , Lipoxinas/farmacología , Ratones , Receptores de Formil Péptido/antagonistas & inhibidores , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología , Activación Transcripcional/efectos de los fármacos
12.
Cell Rep ; 39(13): 110989, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35767946

RESUMEN

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.


Asunto(s)
Interferón beta , Interleucina-12 , Receptor Toll-Like 4 , COVID-19/inmunología , COVID-19/metabolismo , COVID-19/virología , Citocinas/inmunología , Citocinas/metabolismo , Humanos , Interferón beta/inmunología , Interferón beta/metabolismo , Interleucina-12/inmunología , Interleucina-12/metabolismo , Lipopolisacáridos/farmacología , Proteómica , SARS-CoV-2/inmunología
13.
Mol Cell Proteomics ; 21(7): 100247, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35594991

RESUMEN

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.


Asunto(s)
COVID-19 , Interferón beta , Oncogenes , Proteómica , Animales , Factores de Restricción Antivirales , COVID-19/inmunología , Carcinogénesis , Línea Celular Tumoral , Humanos , Interferón beta/inmunología , Proteínas Proto-Oncogénicas p21(ras)/genética , SARS-CoV-2
14.
Microbiol Spectr ; 10(1): e0188321, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35196784

RESUMEN

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.


Asunto(s)
Herpesvirus Humano 1/genética , Herpesvirus Humano 1/inmunología , Interferón beta/genética , Interferón beta/inmunología , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología , Animales , Chlorocebus aethiops , Citocinas , Proteína 58 DEAD Box , Células HEK293 , Células HeLa , Herpes Simple , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune , Inmunidad Innata , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón , Interferón Tipo I , Interferón beta/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas , Receptores Inmunológicos , Transducción de Señal , Células Vero , Proteínas Virales/metabolismo
15.
Virology ; 567: 77-86, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35032866

RESUMEN

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.


Asunto(s)
Atrofia/virología , Interferón-alfa/inmunología , Interferón beta/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Timocitos/virología , Timo/virología , Animales , Atrofia/genética , Atrofia/inmunología , Atrofia/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Enfermedad Crónica , Femenino , Regulación de la Expresión Génica , Humanos , Memoria Inmunológica , Interferón-alfa/genética , Interferón beta/genética , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ganglios Linfáticos/virología , Depleción Linfocítica , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Transducción de Señal/inmunología , Análisis de la Célula Individual , Timocitos/inmunología , Timocitos/patología , Timo/inmunología , Timo/patología
16.
Nat Commun ; 13(1): 105, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013224

RESUMEN

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.


Asunto(s)
Interacciones Huésped-Patógeno/genética , Canales Iónicos/genética , Proteínas de la Membrana/genética , Infección por el Virus Zika/genética , Virus Zika/genética , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Línea Celular Tumoral , Retículo Endoplásmico/genética , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/virología , Femenino , Regulación de la Expresión Génica , Aparato de Golgi/genética , Aparato de Golgi/inmunología , Aparato de Golgi/virología , Hepatocitos/inmunología , Hepatocitos/virología , Interacciones Huésped-Patógeno/inmunología , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Interferón beta/genética , Interferón beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Canales Iónicos/deficiencia , Canales Iónicos/inmunología , Proteínas de la Membrana/inmunología , Ratones , Ratones Noqueados , Fosforilación , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Transducción de Señal , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Virus Zika/crecimiento & desarrollo , Virus Zika/patogenicidad , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología
17.
Mol Immunol ; 143: 7-16, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34990938

RESUMEN

DDX43 is one of the members of the DExD/H-box protein family, and emerging data suggest that it may play an important role in antiviral immunity across mammals. However, little is known about DDX43 in the fish immune response. In this study, we isolated the cDNA sequence of ddx43 in Nile tilapia (Oreochromis niloticus). The ddx43 gene was 2338 bp in length, contained an open reading frame (ORF) of 2064 bp and encoded a polypeptide of 687 amino acids. The predicted protein of OnDDX43 has three conserved domains, including the RNA binding domain KH, DEAD-like helicase superfamily DEXDc and C-terminal HELICc domain. In healthy Nile tilapia, the Onddx43 transcript was broadly expressed in all examined tissues, with the highest expression levels in the muscle and brain and the lowest in the liver. After challenge with Streptococcus agalactiae, lipopolysaccharides (LPS) and polyinosinic polycytidylic acid (Poly I:C), the expression level of Onddx43 mRNA was upregulated or downregulated in all of the tissues tested. Overexpression of OnDDX43 in 293 T cells showed that it has a positive regulatory effect on IFN-ß. The subcellular localization showed that OnDDX43 was expressed in the cytoplasm. We performed further pull-down assays and found that OnDDX43 interacted with both interferon-ß promoter stimulator1 (IPS-1) and TIR domain-containing adaptor inducing interferon-ß (TRIF).


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Cíclidos/inmunología , ARN Helicasas DEAD-box/inmunología , Enfermedades de los Peces/inmunología , Proteínas de Peces/inmunología , Interferón beta/inmunología , Transducción de Señal/inmunología , Animales , Cíclidos/microbiología
18.
J Immunol ; 208(2): 338-346, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34893528

RESUMEN

IL-15 exhibits pleiotropic effects on NK and CD8+ T cells and contributes to host protection or immunopathology during infection. Although both type I IFNs and IFN-γ upregulate IL-15 expression, their effects on IL-15 upregulation and underlying mechanisms have not been compared comprehensively. In addition, little is known about trans-presentation of IL-15 by epithelial cells to lymphocytes. In this study, we analyzed the expression of IL-15 and IL-15Rα in the human hepatocyte-derived Huh-7 cell line after stimulation with IFN-α, IFN-ß, or IFN-γ using RT-PCR, flow cytometry, and confocal microscopy. We also performed knockdown experiments to investigate the signaling pathway involved in IL-15 upregulation. IFN-γ more potently upregulated IL-15 expression in Huh-7 cells than IFN-α and IFN-ß. Knockdown experiments revealed that IFN-γ- and IFN-ß-induced IL-15 expression relied on IFN regulatory factor 1 (IRF1), which is upregulated by STAT1 and IFN-stimulated gene factor 3, respectively. Inhibitor of κB kinase α/ß was also involved in IFN-γ-induced upregulation of IL-15. Furthermore, human NK cells were activated by coculture with IFN-γ-treated Huh-7 cells, which was abrogated by knocking down IL-15Rα in IFN-γ-treated Huh-7 cells, indicating that IFN-γ-induced IL-15 on Huh-7 cells activates NK cells via trans-presentation. In summary, our data demonstrate that IFN-γ potently elicits IL-15 trans-presentation by epithelial cells via IRF1. These data also suggest that the IFN-γ-IRF1-IL-15 axis may be a regulatory target for the treatment of diseases with IL-15 dysregulation.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Factor 1 Regulador del Interferón/metabolismo , Interferón gamma/inmunología , Interleucina-15/metabolismo , Células Asesinas Naturales/inmunología , Células A549 , Línea Celular Tumoral , Células Epiteliales/metabolismo , Células HeLa , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferón-alfa/inmunología , Interferón beta/inmunología , Activación de Linfocitos/inmunología , Receptores de Interleucina-15/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/fisiología , Activación Transcripcional/genética , Regulación hacia Arriba/genética
19.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34969857

RESUMEN

Type I interferons (IFNs) are the first frontline of the host innate immune response against invading pathogens. Herein, we characterized an unknown protein encoded by phospholipase A2 inhibitor and LY6/PLAUR domain-containing (PINLYP) gene that interacted with TBK1 and induced type I IFN in a TBK1- and IRF3-dependent manner. Loss of PINLYP impaired the activation of IRF3 and production of IFN-ß induced by DNA virus, RNA virus, and various Toll-like receptor ligands in multiple cell types. Because PINLYP deficiency in mice engendered an early embryonic lethality in mice, we generated a conditional mouse in which PINLYP was depleted in dendritic cells. Mice lacking PINLYP in dendritic cells were defective in type I IFN induction and more susceptible to lethal virus infection. Thus, PINLYP is a positive regulator of type I IFN innate immunity and important for effective host defense against viral infection.


Asunto(s)
Células Dendríticas/inmunología , Inhibidores Enzimáticos/inmunología , Inmunidad Innata , Interferón beta/inmunología , Animales , Línea Celular , Infecciones por Virus ADN/genética , Infecciones por Virus ADN/inmunología , Virus ADN/genética , Virus ADN/inmunología , Humanos , Interferón beta/genética , Ratones , Ratones Noqueados , Infecciones por Virus ARN/genética , Infecciones por Virus ARN/inmunología , Virus ARN/genética , Virus ARN/inmunología
20.
mBio ; 12(6): e0226721, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34903048

RESUMEN

The cGAS/STING/TBK1 (cyclic guanine monophosphate-AMP synthase/stimulator of interferon genes/Tank-binding kinase 1) innate immunity pathway is activated during human cytomegalovirus (HCMV) productive (lytic) replication in fully differentiated cells and during latency within incompletely differentiated myeloid cells. While multiple lytic-phase HCMV proteins neutralize steps along this pathway, none of them are expressed during latency. Here, we show that the latency-associated protein UL138 inhibits the cGAS/STING/TBK1 innate immunity pathway during transfections and infections, in fully differentiated cells and incompletely differentiated myeloid cells, and with loss of function and restoration of function approaches. UL138 inhibits the pathway downstream of STING but upstream of interferon regulatory factor 3 (IRF3) phosphorylation and NF-κB function and reduces the accumulation of interferon beta mRNA during both lytic and latent infections. IMPORTANCE While a cellular restriction versus viral countermeasure arms race between innate immunity and viral latency is expected, few examples have been documented. Our identification of the first HCMV latency protein that inactivates the cGAS/STING/TBK1 innate immune pathway opens the door to understanding how innate immunity, or its neutralization, impacts long-term persistence by HCMV and other latent viruses.


Asunto(s)
Infecciones por Citomegalovirus , Citomegalovirus , Interferón beta , Proteínas de la Membrana , Latencia del Virus , Humanos , Citomegalovirus/genética , Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/fisiopatología , Infecciones por Citomegalovirus/virología , Interacciones Huésped-Patógeno , Inmunidad Innata , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Interferón beta/genética , Interferón beta/inmunología , Infección Latente/genética , Infección Latente/inmunología , Infección Latente/virología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , FN-kappa B/genética , FN-kappa B/inmunología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA