Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 38(4): 110302, 2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35081346

RESUMEN

It is well known that interferon (IFN)-α/-ß activates the JAK/STAT signaling pathway and suppresses viral replication through the induction of IFN stimulated genes (ISGs). Here, we report that knockout of HDAC3 from macrophages results in the decreased expression of STAT1 and STAT2, leading to defective antiviral immunity in cells and mice. Further studies show that HDAC3 interacts with a conserved transcription factor Forkhead Box K1 (FOXK1), co-localizes with FOXK1 at the promoter of STAT1 and STAT2, and is required for protecting FOXK1 from lysosomal system-mediated degradation. FOXK1-deficient macrophages also show low STAT1 and STAT2 expression with defective responses to viruses. Thus, our studies uncover the biological importance of HDAC3 in regulating the antiviral immunity of macrophages through interacting with FOXK1 to regulate the expression of STAT1 and STAT2.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Histona Desacetilasas/inmunología , Inmunidad Innata/inmunología , Macrófagos/inmunología , Virosis/inmunología , Animales , Factores de Transcripción Forkhead/inmunología , Ratones , Factor de Transcripción STAT1/biosíntesis , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT2/biosíntesis , Factor de Transcripción STAT2/inmunología , Transcripción Genética
2.
J Clin Immunol ; 41(7): 1446-1456, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34448086

RESUMEN

STAT2 is distinguished from other STAT family members by its exclusive involvement in type I and III interferon (IFN-I/III) signaling pathways, and its unique behavior as both positive and negative regulator of IFN-I signaling. The clinical relevance of these opposing STAT2 functions is exemplified by monogenic diseases of STAT2. Autosomal recessive STAT2 deficiency results in heightened susceptibility to severe and/or recurrent viral disease, whereas homozygous missense substitution of the STAT2-R148 residue is associated with severe type I interferonopathy due to loss of STAT2 negative regulation. Here we review the clinical presentation, pathogenesis, and management of these disorders of STAT2.


Asunto(s)
Enfermedades Genéticas Congénitas/genética , Enfermedades del Sistema Inmune/genética , Interferón Tipo I/inmunología , Factor de Transcripción STAT2/genética , Virosis/genética , Animales , Mutación con Ganancia de Función , Enfermedades Genéticas Congénitas/inmunología , Predisposición Genética a la Enfermedad , Humanos , Enfermedades del Sistema Inmune/inmunología , Mutación con Pérdida de Función , Fenotipo , Factor de Transcripción STAT2/química , Factor de Transcripción STAT2/inmunología , Virosis/inmunología
3.
J Immunol ; 206(11): 2682-2691, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34021047

RESUMEN

Type I/III IFNs induce expression of hundreds of IFN-stimulated genes through the JAK/STAT pathway to combat viral infections. Although JAK/STAT signaling is seemingly straightforward, it is nevertheless subjected to complex cellular regulation. In this study, we show that an ubiquitination regulatory X (UBX) domain-containing protein, UBXN6, positively regulates JAK-STAT1/2 signaling. Overexpression of UBXN6 enhanced type I/III IFNs-induced expression of IFN-stimulated genes, whereas deletion of UBXN6 inhibited their expression. RNA viral replication was increased in human UBXN6-deficient cells, accompanied by a reduction in both type I/III IFN expression, when compared with UBXN6-sufficient cells. Mechanistically, UBXN6 interacted with tyrosine kinase 2 (TYK2) and inhibited IFN-ß-induced degradation of both TYK2 and type I IFNR. These results suggest that UBXN6 maintains normal JAK-STAT1/2 signaling by stabilizing key signaling components during viral infection.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/inmunología , Proteínas Relacionadas con la Autofagia/inmunología , Quinasas Janus/inmunología , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT2/inmunología , Animales , Células Cultivadas , Chlorocebus aethiops , Humanos , Transducción de Señal/inmunología
4.
Eur J Immunol ; 51(2): 393-407, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33029793

RESUMEN

Cytomegalovirus (CMV)-based vaccines show promising effects against chronic infections in nonhuman primates. Therefore, we examined the potential of hepatitis B virus (HBV) vaccines based on mouse CMV (MCMV) vectors expressing the small HBsAg. Immunological consequences of vaccine virus attenuation were addressed by either replacing the dispensable gene m157 ("MCMV-HBsȍ) or the gene M27 ("ΔM27-HBs"), the latter encodes a potent IFN antagonist targeting the transcription factor STAT2. M27 was chosen, since human CMV encodes an analogous gene product, which also induced proteasomal STAT2 degradation by exploiting Cullin RING ubiquitin ligases. Vaccinated mice were challenged with HBV through hydrodynamic injection. MCMV-HBs and ΔM27-HBs vaccination achieved accelerated HBV clearance in serum and liver as well as robust HBV-specific CD8+ T-cell responses. When we explored the therapeutic potential of MCMV-based vaccines, especially the combination of ΔM27-HBs prime and DNA boost vaccination resulted in increased intrahepatic HBs-specific CD8+ T-cell responses and HBV clearance in persistently infected mice. Our results demonstrated that vaccines based on a replication competent MCMV attenuated through the deletion of an IFN antagonist targeting STAT2 elicit robust anti-HBV immune responses and mediate HBV clearance in mice in prophylactic and therapeutic immunization regimes.


Asunto(s)
Antígenos de Superficie de la Hepatitis B/inmunología , Vacunas contra Hepatitis B/inmunología , Virus de la Hepatitis B/inmunología , Hepatitis B Crónica/inmunología , Muromegalovirus/inmunología , Animales , Antivirales/inmunología , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Femenino , Hepatitis B Crónica/virología , Inmunización/métodos , Interferones/inmunología , Hígado/inmunología , Hígado/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción STAT2/inmunología , Vacunación/métodos , Replicación Viral/inmunología
5.
J Virol ; 94(21)2020 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-32847859

RESUMEN

Interferon (IFN) family cytokines stimulate genes (interferon-stimulated genes [ISGs]) that are integral to antiviral host defense. Type I IFNs act systemically, whereas type III IFNs act preferentially at epithelial barriers. Among barrier cells, intestinal epithelial cells (IECs) are particularly dependent on type III IFN for the control and clearance of virus infection, but the physiological basis of this selective IFN response is not well understood. Here, we confirm that type III IFN treatment elicits robust and uniform ISG expression in neonatal mouse IECs and inhibits the replication of IEC-tropic rotavirus. In contrast, type I IFN elicits a marginal ISG response in neonatal mouse IECs and does not inhibit rotavirus replication. In vitro treatment of IEC organoids with type III IFN results in ISG expression that mirrors the in vivo type III IFN response. However, IEC organoids have increased expression of the type I IFN receptor relative to neonate IECs, and the response of IEC organoids to type I IFN is strikingly increased in magnitude and scope relative to type III IFN. The expanded type I IFN-specific response includes proapoptotic genes and potentiates toxicity triggered by tumor necrosis factor alpha (TNF-α). The ISGs stimulated in common by type I and III IFNs have strong interferon-stimulated response element (ISRE) promoter motifs, whereas the expanded set of type I IFN-specific ISGs, including proapoptotic genes, have weak ISRE motifs. Thus, the preferential responsiveness of IECs to type III IFN in vivo enables selective ISG expression during infection that confers antiviral protection but minimizes disruption of intestinal homeostasis.IMPORTANCE Enteric viral infections are a major cause of gastroenteritis worldwide and have the potential to trigger or exacerbate intestinal inflammatory diseases. Prior studies have identified specialized innate immune responses that are active in the intestinal epithelium following viral infection, but our understanding of the benefits of such an epithelium-specific response is incomplete. Here, we show that the intestinal epithelial antiviral response is programmed to enable protection while minimizing epithelial cytotoxicity that can often accompany an inflammatory response. Our findings offer new insight into the benefits of a tailored innate immune response at the intestinal barrier and suggest how dysregulation of this response could promote inflammatory disease.


Asunto(s)
Citocinas/inmunología , Mucosa Intestinal/inmunología , Infecciones por Rotavirus/inmunología , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT2/inmunología , Factor de Necrosis Tumoral alfa/toxicidad , Animales , Animales Recién Nacidos , Citocinas/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/virología , Regulación de la Expresión Génica , Humanos , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/inmunología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Organoides/efectos de los fármacos , Organoides/inmunología , Organoides/virología , Elementos de Respuesta , Rotavirus/efectos de los fármacos , Rotavirus/crecimiento & desarrollo , Rotavirus/patogenicidad , Infecciones por Rotavirus/genética , Infecciones por Rotavirus/patología , Infecciones por Rotavirus/virología , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT2/genética , Transducción de Señal , Replicación Viral
6.
Viruses ; 12(7)2020 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-32635475

RESUMEN

The host tropism of viral infection is determined by a variety of factors, from cell surface receptors to innate immune signaling. Many viruses encode proteins that interfere with host innate immune recognition in order to promote infection. STAT2 is divergent between species and therefore has a role in species restriction of some viruses. To understand the role of STAT2 in human metapneumovirus (HMPV) infection of human and murine tissues, we first infected STAT2-/- mice and found that HMPV could be serially passaged in STAT2-/-, but not WT, mice. We then used in vitro methods to show that HMPV inhibits expression of both STAT1 and STAT2 in human and primate cells, but not in mouse cells. Transfection of the murine form of STAT2 into STAT2-deficient human cells conferred resistance to STAT2 inhibition. Finally, we sought to understand the in vivo role of STAT2 by infecting hSTAT2 knock-in mice with HMPV, and found that mice had increased weight loss, inhibition of type I interferon signaling, and a Th2-polarized cytokine profile compared to WT mice. These results indicate that STAT2 is a target of HMPV in human infection, while the murine version of STAT2 restricts tropism of HMPV for murine cells and tissue.


Asunto(s)
Metapneumovirus/fisiología , Infecciones por Paramyxoviridae/inmunología , Factor de Transcripción STAT2/inmunología , Animales , Femenino , Especificidad del Huésped , Humanos , Inmunidad Innata , Interferones/genética , Interferones/inmunología , Masculino , Metapneumovirus/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/virología , Factor de Transcripción STAT2/genética , Células Th2
7.
J Immunol ; 205(5): 1281-1292, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32699158

RESUMEN

Type I IFNs play an important role in innate immunity against viral infections by inducing the expression of IFN-stimulated genes (ISGs), which encode effectors with various antiviral functions. We and others previously reported that HSV type 2 (HSV-2) inhibits the synthesis of type I IFNs, but how HSV-2 suppresses IFN-mediated signaling is less understood. In the current study, after the demonstration of HSV-2 replication resistance to IFN-ß treatment in human epithelial cells, we reveal that HSV-2 and the viral protein ICP22 significantly decrease the expression of ISG54 at both mRNA and protein levels. Likewise, us1 del HSV-2 (ICP22-deficient HSV-2) replication is more sensitive to IFN-ß treatment, indicating that ICP22 is a vital viral protein responsible for the inhibition of type I IFN-mediated signaling. In addition, overexpression of HSV-2 ICP22 inhibits the expression of STAT1, STAT2, and IFN regulatory factor 9 (IRF9), resulting in the blockade of ISG factor 3 (ISGF3) nuclear translocation, and mechanistically, this is due to ICP22-induced ubiquitination of STAT1, STAT2, and IRF9. HSV-2 ICP22 appears to interact with STAT1, STAT2, IRF9, and several other ubiquitinated proteins. Following further biochemical study, we show that HSV-2 ICP22 functions as an E3 ubiquitin protein ligase to induce the formation of polyubiquitin chains. Taken together, we demonstrate that HSV-2 interferes with type I IFN-mediated signaling by degrading the proteins of ISGF3, and we identify HSV-2 ICP22 as a novel E3 ubiquitin protein ligase to induce the degradation of ISGF3. Findings in this study highlight a new mechanism by which HSV-2 circumvents the host antiviral responses through a viral E3 ubiquitin protein ligase.


Asunto(s)
Herpes Genital/inmunología , Herpesvirus Humano 2/inmunología , Proteínas Inmediatas-Precoces/inmunología , Interferón beta/inmunología , Transducción de Señal/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Proteínas Virales/inmunología , Antivirales/inmunología , Línea Celular , Línea Celular Tumoral , Células HEK293 , Células HeLa , Herpesvirus Humano 1/inmunología , Humanos , Inmunidad Innata/inmunología , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT2/inmunología , Ubiquitinación/inmunología
8.
Front Immunol ; 11: 624415, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33679716

RESUMEN

STAT2 is a transcription factor that plays an essential role in antiviral immunity by mediating the activity of type I and III interferons (IFN-I and IFN-III). It also has a recently established function in the negative regulation of IFN-I signaling. Homozygous STAT2 deficiency is an ultra-rare inborn error of immunity which provides unique insight into the pathologic consequence of STAT2 dysfunction. We report here a novel genetic cause of homozygous STAT2 deficiency with several notable clinical features. The proband presented aged 12 months with hemophagocytic lymphohistiocytosis (HLH) closely followed by clinical varicella, both occurring within three weeks of measles, mumps, and rubella (MMR) and varicella vaccinations. There was a history of life-threatening influenza A virus (IAV) disease 2 months previously. Genetic investigation uncovered homozygosity for a novel nonsense variant in STAT2 (c. 1999C>T, p. Arg667Ter) that abrogated STAT2 protein expression. Compatible with STAT2 deficiency, dermal fibroblasts from the child demonstrated a defect of interferon-stimulated gene expression and a failure to mount an antiviral state in response to treatment with IFN-I, a phenotype that was rescued by lentiviral complementation by wild type STAT2. This case significantly expands the phenotypic spectrum of STAT2 deficiency. The occurrence of life-threatening influenza, which has not previously been reported in this condition, adds STAT2 to the list of monogenetic causes of this phenotype and underscores the critical importance of IFN-I and IFN-III to influenza immunity. The development of probable vaccine-strain varicella is also a novel occurrence in STAT2 deficiency, implying a role for IFN-I/III immunity in control of attenuated varicella zoster virus in vivo and reinforcing the susceptibility to pathologic effects of live-attenuated viral vaccines in disorders of IFN-I immunity. Finally, the occurrence of HLH in this case reinforces emerging links to hyperinflammation in patients with STAT2 deficiency and other related defects of IFN-I signaling-highlighting an important avenue for further scientific enquiry.


Asunto(s)
Vacuna contra la Varicela/efectos adversos , Codón sin Sentido , Homocigoto , Gripe Humana , Factor de Transcripción STAT2/deficiencia , Vacuna contra la Varicela/inmunología , Niño , Humanos , Gripe Humana/genética , Gripe Humana/inmunología , Gripe Humana/patología , Linfohistiocitosis Hemofagocítica/genética , Linfohistiocitosis Hemofagocítica/inmunología , Linfohistiocitosis Hemofagocítica/patología , Masculino , Factor de Transcripción STAT2/inmunología
9.
Virus Res ; 275: 197804, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31697988

RESUMEN

Porcine circovirus 3 (PCV3) is a novel circovirus that is associated with porcine dermatitis and nephropathy syndrome, reproductive failure, and multi-systemic inflammation. The type I Interferon (IFN) signaling pathway is an important innate immune signaling pathway for defense against viral infection. Many mammalian viruses inhibit host innate immune signaling through diverse strategies. Here, we found that the PCV3 capsid protein (Cap) significantly inhibited IFN-ß-stimulated response element (ISRE) promoter activity, suggesting that Cap suppresses IFN signaling. However, Cap did not affect expression and phosphorylation levels of STAT1 and STAT2 and did not interrupt the heterodimerization of pSTAT1 and pSTAT2. Although Cap interacted with KPAN1, it did not block the interaction between KPNA1 and pSTAT1 or the nuclear translocation of pSTAT1 and pSTAT2. Interestingly, we found that Cap inhibited the activation of ISRE promoter induced by IRF9-S2C. Mechanistically, Cap interacted with the transactivation domain of STAT2, a key protein in type I IFN signaling. In addition, we found that Cap bound to ISRE and prevented ISRE binding of IRF9-S2C. This work is the first to describe the mechanism of inhibition of IFN signaling by PCV3 Cap.


Asunto(s)
Proteínas de la Cápside/inmunología , Circovirus/inmunología , Interferón Tipo I/antagonistas & inhibidores , Factor de Transcripción STAT2/inmunología , Transducción de Señal/inmunología , Proteínas de la Cápside/genética , Circovirus/clasificación , Células HEK293 , Humanos , Inmunidad Innata , Interferón Tipo I/inmunología , Interferón beta/antagonistas & inhibidores , Interferón beta/inmunología , Fosforilación , Factor de Transcripción STAT1/inmunología
10.
Vet Immunol Immunopathol ; 219: 109971, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31739157

RESUMEN

Equine herpesvirus type 4 (EHV-4) is mildly pathogenic but is a common cause of respiratory disease in horses worldwide. We previously demonstrated that unlike EHV-1, EHV-4 is not a potent inducer of type-I IFN and does not suppress that IFN response, especially during late infection, when compared to EHV-1 infection in equine endothelial cells (EECs). Here, we investigated the impact of EHV-4 infection in EECs on type-I IFN signaling molecules at 3, 6, and 12 hpi. Findings from our study revealed that EHV-4 did not induce nor suppress TLR3 and TLR4 expression in EECs at all the studied time points. EHV-4 was able to induce variable amounts of IRF7 and IRF9 in EECs with no evidence of suppressive effect on these important transcription factors of IFN-α/ß induction. Intriguingly, EHV-4 did interfere with the phosphorylation of STAT1/STAT2 at 3 hpi and 6 hpi, less so at 12 hpi. An active EHV-4 viral gene expression was required for the suppressive effect of EHV-4 on STAT1/STAT2 phosphorylation during early infection. One or more early viral genes of EHV-4 are involved in the suppression of STAT1/STAT2 phosphorylation observed during early time points in EHV-4-infected EECs. The inability of EHV-4 to significantly down-regulate key molecules of type-I IFN signaling may be related to the lower severity of pathogenesis when compared with EHV-1. Harnessing this knowledge may prove useful in controlling future outbreaks of the disease.


Asunto(s)
Células Endoteliales/inmunología , Herpesvirus Équido 4/inmunología , Interacciones Microbiota-Huesped/inmunología , Inmunidad Innata , Interferón Tipo I/inmunología , Animales , Células Cultivadas , Células Endoteliales/virología , Herpesvirus Équido 4/patogenicidad , Enfermedades de los Caballos/inmunología , Enfermedades de los Caballos/virología , Caballos , Factor 3 de Genes Estimulados por el Interferón/inmunología , Interferón-alfa/inmunología , Interferón beta/inmunología , Fosforilación , Arteria Pulmonar/citología , Factor de Transcripción STAT2/inmunología , Transducción de Señal/inmunología , Receptores Toll-Like/inmunología
11.
PLoS One ; 14(12): e0225651, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31790472

RESUMEN

Systemic lupus erythematosus (SLE) is a chronic, remitting, and relapsing, inflammatory disease involving multiple organs, which exhibits abnormalities of both the innate and adaptive immune responses. A limited number of transcriptomic studies have characterized the gene pathways involved in SLE in an attempt to identify the key pathogenic drivers of the disease. In order to further advance our understanding of the pathogenesis of SLE, we used a novel Bayesian network algorithm to hybridize knowledge- and data-driven methods, and then applied the algorithm to build an SLE gene network using transcriptomic data from 1,760 SLE patients' RNA from the two tabalumab Phase III trials (ILLUMINATE-I & -II), the largest SLE RNA dataset to date. Further, based on the gene network, we carried out hub- and key driver-gene analyses for gene prioritization. Our analyses identified that the JAK-STAT pathway genes, including JAK2, STAT1, and STAT2, played essential roles in SLE pathogenesis, and reaffirmed the recent discovery of pathogenic relevance of JAK-STAT signaling in SLE. Additionally, we showed that other genes, such as IRF1, IRF7, PDIA4, FAM72C, TNFSF10, DHX58, SIGLEC1, and PML, may be also important in SLE and serve as potential therapeutic targets for SLE. In summary, using a hybridized network construction approach, we systematically investigated gene-gene interactions based on their transcriptomic profiles, prioritized genes based on their importance in the network structure, and revealed new insights into SLE activity.


Asunto(s)
Redes Reguladoras de Genes/inmunología , Lupus Eritematoso Sistémico/genética , Modelos Genéticos , Transducción de Señal/genética , Algoritmos , Teorema de Bayes , Ensayos Clínicos Fase III como Asunto , Simulación por Computador , Minería de Datos , Conjuntos de Datos como Asunto , Perfilación de la Expresión Génica , Humanos , Janus Quinasa 2/inmunología , Janus Quinasa 2/metabolismo , Modelos Lineales , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Ensayos Clínicos Controlados Aleatorios como Asunto , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/inmunología , Factor de Transcripción STAT2/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
12.
Nat Commun ; 10(1): 4700, 2019 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-31619669

RESUMEN

The functions of the IL-36 cytokines remain poorly understood. We report a previously unrecognized mechanism whereby IL-36 promotes innate antiviral immunity in mouse and human models of herpes simplex virus-1 (HSV-1) infections. HSV-1 actively suppresses production of type I interferon (IFN); our data reveal that IL-36 overcomes this immune evasion strategy by increasing cellular sensitivity to IFN. IL-36ß deficient mice display impaired IFN responses and poorly restrict viral replication in skin keratinocytes. In mouse and human keratinocytes IL-36 elicits an antiviral state driven by STAT1 and STAT2 via enhanced expression of IFNAR1 and IFNAR2 subunits of the type I IFN receptor. The degree of IFN regulatory factor 1 (IRF1) involvement is species dependent, with IRF1 playing a more prominent role in human cells. Similar mechanisms are activated by IL-1. Overall, IL-36 acts as an antiviral cytokine by potentiating type I IFN signaling and thereby upholds immune responses to viruses that limit the production of IFNs.


Asunto(s)
Herpes Simple/inmunología , Interferón Tipo I/inmunología , Interleucina-1/inmunología , Receptor de Interferón alfa y beta/genética , Animales , Modelos Animales de Enfermedad , Herpesvirus Humano 1 , Humanos , Evasión Inmune , Factor 1 Regulador del Interferón/inmunología , Interleucina-1/genética , Queratinocitos , Ratones , Ratones Noqueados , Receptor de Interferón alfa y beta/metabolismo , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT2/inmunología , Regulación hacia Arriba
14.
J Immunol ; 202(8): 2266-2275, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30842274

RESUMEN

It is not clear how hepatitis B virus (HBV) modulates host immunity during chronic infection. In addition to the key mediators of inflammatory response in viral infection, monocytes also express a high-level IFN-stimulated gene, CH25H, upon response to IFN-α exerting an antiviral effect. In this study, the mechanism by which HBV manipulates IFN signaling in human monocytes was investigated. We observed that monocytes from chronic hepatitis B patients express lower levels of IFN signaling/stimulated genes and higher levels of inflammatory cytokines compared with healthy donors. HBV induces monocyte production of inflammatory cytokines via TLR2/MyD88/NF-κB signaling and STAT1-Ser727 phosphorylation and inhibits IFN-α-induced stat1, stat2, and ch25h expression through the inhibition of STAT1-Tyr701 phosphorylation and in an IL-10-dependent, partially autocrine manner. Further, we found that enhancement of STAT1 activity with a small molecule (2-NP) rescued HBV-mediated inhibition of IFN signaling and counteracted the induction of inflammatory cytokines. In conclusion, HBV contributes to the monocyte inflammatory response but inhibits their IFN-α/ß responsiveness to impair antiviral innate immunity. These effects are mediated via differential phosphorylation of Tyr701 and Ser727 of STAT1.


Asunto(s)
Virus de la Hepatitis B/inmunología , Hepatitis B/inmunología , Inmunidad Innata , Monocitos/inmunología , Factor de Transcripción STAT1/inmunología , Transducción de Señal/inmunología , Células Hep G2 , Hepatitis B/patología , Humanos , Interleucina-10/inmunología , Monocitos/patología , Factor 88 de Diferenciación Mieloide/inmunología , FN-kappa B/inmunología , Fosforilación/inmunología , Factor de Transcripción STAT2/inmunología , Receptor Toll-Like 2/inmunología
15.
Front Immunol ; 9: 2151, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30337919

RESUMEN

Influenza is a common respiratory virus that infects between 5 and 20% of the US population and results in 30,000 deaths annually. A primary cause of influenza-associated death is secondary bacterial pneumonia. We have previously shown that influenza induces type I interferon (IFN)-mediated inhibition of Type 17 immune responses, resulting in exacerbation of bacterial burden during influenza and Staphylococcus aureus super-infection. In this study, we investigated the role of STAT2 signaling during influenza and influenza-bacterial super-infection in mice. Influenza-infected STAT2-/- mice had increased morbidity, viral burden, and inflammation when compared to wild-type mice. Despite an exaggerated inflammatory response to influenza infection, we found increased bacterial control and survival in STAT2 deficient mice during influenza-MRSA super-infection compared to controls. Further, we found that increased bacterial clearance during influenza-MRSA super-infection is not due to rescue of Type 17 immunity. Absence of STAT2 was associated with increased accumulation of M1, M2 and M1/M2 co-expressing macrophages during influenza-bacterial super-infection. Neutralization of IFNγ (M1) and/or Arginase 1 (M2) impaired bacterial clearance in Stat2-/- mice during super-infection, demonstrating that pulmonary macrophages expressing a mixed M1/M2 phenotype promote bacterial control during influenza-bacterial super-infection. Together, these results suggest that the STAT2 signaling is involved in suppressing macrophage activation and bacterial control during influenza-bacterial super-infection. Further, these studies reveal novel mechanistic insight into the roles of macrophage subpopulations in pulmonary host defense.


Asunto(s)
Gripe Humana/inmunología , Macrófagos Alveolares/inmunología , Neumonía Estafilocócica/inmunología , Factor de Transcripción STAT2/metabolismo , Sobreinfección/inmunología , Animales , Trasplante de Médula Ósea , Embrión de Pollo , Modelos Animales de Enfermedad , Femenino , Humanos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/diagnóstico , Gripe Humana/microbiología , Gripe Humana/mortalidad , Activación de Macrófagos/inmunología , Macrófagos Alveolares/metabolismo , Masculino , Células Madre Mesenquimatosas , Staphylococcus aureus Resistente a Meticilina/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía Estafilocócica/diagnóstico , Neumonía Estafilocócica/microbiología , Neumonía Estafilocócica/mortalidad , Cultivo Primario de Células , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT2/inmunología , Índice de Severidad de la Enfermedad , Transducción de Señal/inmunología , Sobreinfección/diagnóstico , Sobreinfección/microbiología , Sobreinfección/mortalidad , Quimera por Trasplante
16.
Proc Natl Acad Sci U S A ; 115(39): E9172-E9181, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30206152

RESUMEN

Genome-wide investigations of host-pathogen interactions are often limited by analyses of mixed populations of infected and uninfected cells, which lower sensitivity and accuracy. To overcome these obstacles and identify key mechanisms by which Zika virus (ZIKV) manipulates host responses, we developed a system that enables simultaneous characterization of genome-wide transcriptional and epigenetic changes in ZIKV-infected and neighboring uninfected primary human macrophages. We demonstrate that transcriptional responses in ZIKV-infected macrophages differed radically from those in uninfected neighbors and that studying the cell population as a whole produces misleading results. Notably, the uninfected population of macrophages exhibits the most rapid and extensive changes in gene expression, related to type I IFN signaling. In contrast, infected macrophages exhibit a delayed and attenuated transcriptional response distinguished by preferential expression of IFNB1 at late time points. Biochemical and genomic studies of infected macrophages indicate that ZIKV infection causes both a targeted defect in the type I IFN response due to degradation of STAT2 and reduces RNA polymerase II protein levels and DNA occupancy, particularly at genes required for macrophage identity. Simultaneous evaluation of transcriptomic and epigenetic features of infected and uninfected macrophages thereby reveals the coincident evolution of dominant proviral or antiviral mechanisms, respectively, that determine the outcome of ZIKV exposure.


Asunto(s)
Inmunidad Innata , Macrófagos/inmunología , Infección por el Virus Zika/inmunología , Virus Zika/inmunología , Efecto Espectador , Femenino , Humanos , Interferón beta/genética , Interferón beta/inmunología , Macrófagos/patología , Masculino , Proteolisis , ARN Polimerasa II/genética , ARN Polimerasa II/inmunología , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT2/inmunología , Infección por el Virus Zika/patología
17.
J Virol ; 92(21)2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30089701

RESUMEN

Both type I and type II interferons (IFNs) have been implicated in the host defense against varicella-zoster virus (VZV), a common human herpesvirus that causes varicella and zoster. The purpose of this study was to compare their contributions to the control of VZV replication, to identify the signaling pathways that are critical for mediating their antiviral activity, and to define the mechanisms by which the virus counteracts their effects. Gamma interferon (IFN-γ) was much more potent than IFN-α in blocking VZV infection, which was associated with a differential induction of the interferon regulatory factor (IRF) proteins IRF1 and IRF9, respectively. These observations account for the clinical experience that while the formation of VZV skin lesions is initially controlled by local immunity, adaptive virus-specific T cell responses are required to prevent life-threatening VZV infections.IMPORTANCE While both type I and type II IFNs are involved in the control of herpesvirus infections in the human host, to our knowledge, their relative contributions to the restriction of viral replication and spread have not been assessed. We report that IFN-γ has more potent activity than IFN-α against VZV. Findings from this comparative analysis show that the IFN-α-IRF9 axis functions as a first line of defense to delay the onset of viral replication and spread, whereas the IFN-γ-IRF1 axis has the capacity to block the infectious process. Our findings underscore the importance of IRFs in IFN regulation of herpesvirus infection and account for the clinical experience of the initial control of VZV skin infection attributable to IFN-α production, together with the requirement for induction of adaptive IFN-γ-producing VZV-specific T cells to resolve the infection.


Asunto(s)
Herpesvirus Humano 3/inmunología , Factor 1 Regulador del Interferón/inmunología , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/inmunología , Interferón-alfa/inmunología , Interferón gamma/inmunología , Infección por el Virus de la Varicela-Zóster/inmunología , Línea Celular Tumoral , Células HEK293 , Humanos , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT2/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Infección por el Virus de la Varicela-Zóster/virología , Replicación Viral/inmunología
18.
J Clin Invest ; 127(12): 4415-4420, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29106381

RESUMEN

Primary immunodeficiencies are often monogenic disorders characterized by vulnerability to specific infectious pathogens. Here, we performed whole-exome sequencing of a patient with disseminated Mycobacterium abscessus, Streptococcus viridians bacteremia, and cytomegalovirus (CMV) viremia and identified mutations in 2 genes that regulate distinct IFN pathways. The patient had a homozygous frameshift deletion in IFNGR2, which encodes the signal transducing chain of the IFN-γ receptor, that resulted in minimal protein expression and abolished downstream signaling. The patient also harbored a homozygous deletion in IFNAR1 (IFNAR1*557Gluext*46), which encodes the IFN-α receptor signaling subunit. The IFNAR1*557Gluext*46 resulted in replacement of the stop codon with 46 additional codons at the C-terminus. The level of IFNAR1*557Gluext*46 mutant protein expressed in patient fibroblasts was comparable to levels of WT IFNAR1 in control fibroblasts. IFN-α-induced signaling was impaired in the patient fibroblasts, as evidenced by decreased STAT1/STAT2 phosphorylation, nuclear translocation of STAT1, and expression of IFN-α-stimulated genes critical for CMV immunity. Pretreatment with IFN-α failed to suppress CMV protein expression in patient fibroblasts, whereas expression of WT IFNAR1 restored IFN-α-mediated suppression of CMV. This study identifies a human IFNAR1 mutation and describes a digenic immunodeficiency specific to type I and type II IFNs.


Asunto(s)
Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/inmunología , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Mutación , Receptor de Interferón alfa y beta , Receptores de Interferón , Bacteriemia/genética , Bacteriemia/inmunología , Bacteriemia/microbiología , Citomegalovirus/inmunología , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/inmunología , Femenino , Fibroblastos/inmunología , Fibroblastos/microbiología , Fibroblastos/virología , Enfermedades Genéticas Congénitas/microbiología , Enfermedades Genéticas Congénitas/virología , Humanos , Síndromes de Inmunodeficiencia/microbiología , Síndromes de Inmunodeficiencia/virología , Masculino , Infecciones por Mycobacterium no Tuberculosas/genética , Infecciones por Mycobacterium no Tuberculosas/inmunología , Mycobacterium abscessus/inmunología , Fosforilación/genética , Fosforilación/inmunología , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT2/inmunología , Infecciones Estreptocócicas/genética , Infecciones Estreptocócicas/inmunología , Viremia/genética , Viremia/inmunología , Viremia/virología , Estreptococos Viridans/inmunología
19.
Virology ; 507: 161-169, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28441586

RESUMEN

Emerged porcine kobuvirus (PKV) has adversely affected the global swine industry since 2008, but the etiological biology of PKV is unclear. Screening PKV-encoded structural and non-structural proteins with a type I IFN-responsive luciferase reporter showed that PKV VP3 protein inhibited the IFN-ß-triggered signaling pathway, resulting in the decrease of VSV-GFP replication. QPCR data showed that IFN-ß downstream cytokine genes were suppressed without cell-type specificity as well. The results from biochemical experiments indicated that PKV VP3 associated with STAT2 and IRF9, and interfered with the formation of the STAT2-IRF9 and STAT2-STAT2 complex, impairing nuclear translocation of STAT2 and IRF9. Taken together, these data reveal a new mechanism for immune evasion of PKV.


Asunto(s)
Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/inmunología , Interferón beta/inmunología , Kobuvirus/inmunología , Infecciones por Picornaviridae/inmunología , Factor de Transcripción STAT2/inmunología , Proteínas Virales/inmunología , Animales , Línea Celular , Dimerización , Humanos , Evasión Inmune , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Interferón beta/genética , Kobuvirus/genética , Ratones , Infecciones por Picornaviridae/genética , Infecciones por Picornaviridae/virología , Factor de Transcripción STAT2/química , Factor de Transcripción STAT2/genética , Transducción de Señal , Proteínas Virales/genética
20.
Infect Genet Evol ; 51: 143-152, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28365387

RESUMEN

The Zika virus outbreak in 2015-2016 is the largest of its kind for which WHO declared a Public Health Emergency of International Concerns. No FDA approved drug is available for the treatment of the viral infection. The interaction of flavivirus NS5 protein with SIAH2 ubiquitin ligase has been previously known. NS5 of Zika virus has been implicated in the degradation of STAT2 protein, which activates interferon-stimulated antiviral activity. Based on our proposition that NS5 utilizes SIAH2-mediated proteasomal degradation of STAT2, an in-silico study was carried out to characterize the protein-protein interactions between NS5, SIAH2 and STAT2 proteins. The aim of our study was to identify the amino acid residues of NS5 involved in IFN antagonism as well as to find the association between NS5, SIAH2 and STAT2 to predict the interaction pattern of these proteins. Analysis proposed that NS5 recruits SIAH2 for the ubiquitination-dependent degradation of STAT2. NS5 residues involved in interaction with SIAH2 and/or STAT2 were found to be mostly conserved across related flaviviruses. These are novel findings regarding the Zika virus and require confirmation through experimental approaches.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Proteínas Nucleares/química , Factor de Transcripción STAT2/química , Ubiquitina-Proteína Ligasas/química , Proteínas no Estructurales Virales/química , Virus Zika/química , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Expresión Génica , Humanos , Interferones/genética , Interferones/inmunología , Simulación del Acoplamiento Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteolisis , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT2/inmunología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología , Ubiquitinación , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Virus Zika/enzimología , Virus Zika/inmunología , Infección por el Virus Zika/genética , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...