Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(34): e2403235121, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39145933

RESUMEN

The ZIKA virus (ZIKV) evades the host immune response by degrading STAT2 through its NS5 protein, thereby inhibiting type I interferon (IFN)-mediated antiviral immunity. However, the molecular mechanism underlying this process has remained elusive. In this study, we performed a genome-wide CRISPR/Cas9 screen, revealing that ZSWIM8 as the substrate receptor of Cullin3-RING E3 ligase is required for NS5-mediated STAT2 degradation. Genetic depletion of ZSWIM8 and CUL3 substantially impeded NS5-mediated STAT2 degradation. Biochemical analysis illuminated that NS5 enhances the interaction between STAT2 and the ZSWIM8-CUL3 E3 ligase complex, thereby facilitating STAT2 ubiquitination. Moreover, ZSWIM8 knockout endowed A549 and Huh7 cells with partial resistance to ZIKV infection and protected cells from the cytopathic effects induced by ZIKV, which was attributed to the restoration of STAT2 levels and the activation of IFN signaling. Subsequent studies in a physiologically relevant model, utilizing human neural progenitor cells, demonstrated that ZSWIM8 depletion reduced ZIKV infection, resulting from enhanced IFN signaling attributed to the sustained levels of STAT2. Our findings shed light on the role of ZIKV NS5, serving as the scaffold protein, reprograms the ZSWIM8-CUL3 E3 ligase complex to orchestrate STAT2 proteasome-dependent degradation, thereby facilitating evasion of IFN antiviral signaling. Our study provides unique insights into ZIKV-host interactions and holds promise for the development of antivirals and prophylactic vaccines.


Asunto(s)
Proteínas Cullin , Interferón Tipo I , Proteolisis , Factor de Transcripción STAT2 , Transducción de Señal , Ubiquitina-Proteína Ligasas , Ubiquitinación , Proteínas no Estructurales Virales , Infección por el Virus Zika , Virus Zika , Humanos , Factor de Transcripción STAT2/metabolismo , Virus Zika/inmunología , Virus Zika/fisiología , Virus Zika/metabolismo , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Interferón Tipo I/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología , Proteínas Cullin/metabolismo , Células A549 , Células HEK293 , Sistemas CRISPR-Cas
2.
Fish Shellfish Immunol ; 152: 109776, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39019128

RESUMEN

Type I IFNs are a subset of cytokines exerting their antiviral effects mainly through the JAK-STAT signalling. Immunogenetic studies have shown that fish possess key components of IFN-JAK-STAT cascade, but the information about the distinct responses of STAT1 and STAT2 to different IFNs is rather limited in fish. Here, we identified and cloned STAT1 and STAT2 genes (named as On-STAT1 and On-STAT2) from tilapia, Oreochromis niloticus. On-STAT1 and On-STAT2 genes were detected in all orangs/tissues examined, and were rapidly induced in spleen, head kidney, and liver following the stimulation of poly(I:C). In addition, the stimulation of poly(I:C), poly(A:T), and different subgroups of recombinant IFNs could induce the expression of On-STAT1 and On-STAT2 in TA-02 cells with distinct induction levels. Importantly, On-STAT2 was rapidly phosphorylated by all three subgroups of IFNs, but the phosphorylation of On-STAT1 was only observed in IFNc- and IFNh-treated TA-02 cells, reflecting the distinct activation of STAT by different subgroups of fish IFNs. The present results thus contribute to better understanding of the JAK-STAT signalling mediated by different subgroups of IFNs in fish.


Asunto(s)
Proteínas de Peces , Factor de Transcripción STAT1 , Factor de Transcripción STAT2 , Animales , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT2/metabolismo , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Proteínas de Peces/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Fosforilación , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Cíclidos/inmunología , Cíclidos/genética , Secuencia de Aminoácidos , Regulación de la Expresión Génica/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Filogenia , Poli I-C/farmacología , Alineación de Secuencia/veterinaria , Transducción de Señal/efectos de los fármacos
3.
Nat Commun ; 15(1): 5426, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38926343

RESUMEN

Zika and dengue virus nonstructural protein 5 antagonism of STAT2, a critical interferon signaling transcription factor, to suppress the host interferon response is required for viremia and pathogenesis in a vertebrate host. This affects viral species tropism, as mouse STAT2 resistance renders only immunocompromised or humanized STAT2 mice infectable. Here, we explore how STAT2 evolution impacts antagonism. By measuring the susceptibility of 38 diverse STAT2 proteins, we demonstrate that resistance arose numerous times in mammalian evolution. In four species, resistance requires distinct sets of multiple amino acid changes that often individually disrupt STAT2 signaling. This reflects an evolutionary ridge where progressive resistance is balanced by the need to maintain STAT2 function. Furthermore, resistance may come with a fitness cost, as resistance that arose early in lemur evolution was subsequently lost in some lemur lineages. These findings underscore that while it is possible to evolve resistance to antagonism, complex evolutionary trajectories are required to avoid detrimental host fitness consequences.


Asunto(s)
Evolución Molecular , Factor de Transcripción STAT2 , Proteínas no Estructurales Virales , Factor de Transcripción STAT2/metabolismo , Factor de Transcripción STAT2/genética , Animales , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Humanos , Ratones , Virus del Dengue/genética , Virus del Dengue/fisiología , Virus Zika/genética , Flavivirus/genética , Flavivirus/fisiología , Filogenia , Interacciones Huésped-Patógeno/genética
4.
J Biol Chem ; 300(7): 107472, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38879005

RESUMEN

African swine fever virus (ASFV) causes severe disease in domestic pigs and wild boars, seriously threatening the development of the global pig industry. Type I interferon (IFN-I) is an important component of innate immunity, inducing the transcription and expression of antiviral cytokines by activating Janus-activated kinase-signal transducer and activator of transcription (STAT). However, the underlying molecular mechanisms by which ASFV antagonizes IFN-I signaling have not been fully elucidated. Therefore, using coimmunoprecipitation, confocal microscopy, and dual luciferase reporter assay methods, we investigated these mechanisms and identified a novel ASFV immunosuppressive protein, pB475L, which interacts with the C-terminal domain of STAT2. Consequently, pB475L inhibited IFN-I signaling by inhibiting STAT1 and STAT2 heterodimerization and nuclear translocation. Furthermore, we constructed an ASFV-B475L7PM mutant strain by homologous recombination, finding that ASFV-B475L7PM attenuated the inhibitory effects on IFN-I signaling compared to ASFV-WT. In summary, this study reveals a new mechanism by which ASFV impairs host innate immunity.


Asunto(s)
Virus de la Fiebre Porcina Africana , Inmunidad Innata , Interferón Tipo I , Factor de Transcripción STAT2 , Transducción de Señal , Proteínas Virales , Animales , Humanos , Fiebre Porcina Africana/inmunología , Fiebre Porcina Africana/virología , Fiebre Porcina Africana/metabolismo , Fiebre Porcina Africana/genética , Virus de la Fiebre Porcina Africana/inmunología , Virus de la Fiebre Porcina Africana/genética , Células HEK293 , Evasión Inmune , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT2/metabolismo , Factor de Transcripción STAT2/genética , Porcinos , Proteínas Virales/genética , Proteínas Virales/metabolismo , Proteínas Virales/inmunología
5.
mBio ; 15(8): e0103824, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-38940561

RESUMEN

Human adenoviruses (HAdVs) are small DNA viruses that generally cause mild disease. Certain strains, particularly those belonging to species B HAdVs, can cause severe pneumonia and have a relatively high mortality rate. Little is known about the molecular aspects of how these highly pathogenic species affect the infected cell and how they suppress innate immunity. The present study provides molecular insights into how species B adenoviruses suppress the interferon signaling pathway. Our study shows that these viruses, unlike HAdV-C2, are resistant to type I interferon. This resistance likely arises due to the highly efficient suppression of interferon-stimulated gene expression. Unlike in HAdV-C2, HAdV-B7 and B14 sequester STAT2 and RNA polymerase II from interferon-stimulated gene promoters in infected cells. This results in suppressed interferon- stimulated gene activation. In addition, we show that RuvBL1 and RuvBL2, cofactors important for RNA polymerase II recruitment to promoters and interferon-stimulated gene activation, are redirected to the cytoplasm forming high molecular weight complexes that, likely, are unable to associate with chromatin. Proteomic analysis also identified key differences in the way these viruses affect the host cell, providing insights into species B-associated high pathogenicity. Curiously, we observed that at the level of protein expression changes to the infected cell, HAdV-C2 and B7 were more similar than those of the same species, B7 and B14. Collectively, our study represents the first such study of innate immune suppression by the highly pathogenic HAdV-B7 and B14, laying an important foundation for future investigations.IMPORTANCEHuman adenoviruses form a large family of double-stranded DNA viruses known for a variety of usually mild diseases. Certain strains of human adenovirus cause severe pneumonia leading to much higher mortality and morbidity than most other strains. The reasons for this enhanced pathogenicity are unknown. Our study provides a molecular investigation of how these highly pathogenic strains might inactivate the interferon signaling pathway, highlighting the lack of sensitivity of these viruses to type I interferon in general while providing a global picture of how viral changes in cellular proteins drive worse disease outcomes.


Asunto(s)
Adenovirus Humanos , Interferón Tipo I , Humanos , Adenovirus Humanos/genética , Adenovirus Humanos/patogenicidad , Adenovirus Humanos/fisiología , Adenovirus Humanos/inmunología , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Interferón Tipo I/genética , Factor de Transcripción STAT2/metabolismo , Factor de Transcripción STAT2/genética , Inmunidad Innata , ARN Polimerasa II/metabolismo , ARN Polimerasa II/genética , Transducción de Señal , Infecciones por Adenovirus Humanos/virología , Infecciones por Adenovirus Humanos/inmunología , Virulencia , Interacciones Huésped-Patógeno/inmunología , Animales , Regiones Promotoras Genéticas , Evasión Inmune , Células A549
6.
J Virol ; 98(7): e0058524, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38869319

RESUMEN

Senecavirus A (SVA), a picornavirus, causes vesicular diseases and epidemic transient neonatal losses in swine, resulting in a multifaceted economic impact on the swine industry. SVA counteracts host antiviral response through multiple strategies facilitatng viral infection and transmission. However, the mechanism of how SVA modulates interferon (IFN) response remains elusive. Here, we demonstrate that SVA 3C protease (3Cpro) blocks the transduction of Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathway to antagonize type I IFN response. Mechanistically, 3Cpro selectively cleaves and degrades STAT1 and STAT2 while does not target JAK1, JAK2, and IRF9, through its protease activity. Notably, SVA 3Cpro cleaves human and porcine STAT1 on a Leucine (L)-Aspartic acid (D) motif, specifically L693/D694. In the case of STAT2, two cleavage sites were identified: glutamine (Q) 707 was identified in both human and porcine, while the second cleavage pattern differed, with residues 754-757 (Valine-Leucine-Glutamine-Serine motifs) in human STAT2 and Q758 in porcine STAT2. These cleavage patterns by SVA 3Cpro partially differ from previously reported classical motifs recognized by other picornaviral 3Cpro, highlighting the distinct characteristics of SVA 3Cpro. Together, these results reveal a mechanism by which SVA 3Cpro antagonizes IFN-induced antiviral response but also expands our knowledge about the substrate recognition patterns for picornaviral 3Cpro.IMPORTANCESenecavirus A (SVA), the only member in the Senecavirus genus within the Picornaviridae family, causes vesicular diseases in pigs that are clinically indistinguishable from foot-and-mouth disease (FMD), a highly contagious viral disease listed by the World Organization for Animal Health (WOAH). Interferon (IFN)-mediated antiviral response plays a pivotal role in restricting and controlling viral infection. Picornaviruses evolved numerous strategies to antagonize host antiviral response. However, how SVA modulates the JAK-STAT signaling pathway, influencing the type I IFN response, remains elusive. Here, we identify that 3Cpro, a protease of SVA, functions as an antagonist for the IFN response. 3Cpro utilizes its protease activity to cleave STAT1 and STAT2, thereby diminishing the host IFN response to promote SVA infection. Our findings underscore the significance of 3Cpro as a key virulence factor in the antagonism of the type I signaling pathway during SVA infection.


Asunto(s)
Cisteína Endopeptidasas , Infecciones por Picornaviridae , Picornaviridae , Factor de Transcripción STAT1 , Factor de Transcripción STAT2 , Transducción de Señal , Proteínas Virales , Animales , Porcinos , Factor de Transcripción STAT2/metabolismo , Humanos , Infecciones por Picornaviridae/virología , Infecciones por Picornaviridae/inmunología , Infecciones por Picornaviridae/metabolismo , Factor de Transcripción STAT1/metabolismo , Cisteína Endopeptidasas/metabolismo , Proteínas Virales/metabolismo , Proteasas Virales 3C , Enfermedades de los Porcinos/virología , Enfermedades de los Porcinos/inmunología , Células HEK293 , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/metabolismo , Línea Celular , Quinasas Janus/metabolismo , Quinasas Janus/antagonistas & inhibidores
7.
Nat Commun ; 15(1): 4484, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802340

RESUMEN

Deciphering the intricate dynamic events governing type I interferon (IFN) signaling is critical to unravel key regulatory mechanisms in host antiviral defense. Here, we leverage TurboID-based proximity labeling coupled with affinity purification-mass spectrometry to comprehensively map the proximal human proteomes of all seven canonical type I IFN signaling cascade members under basal and IFN-stimulated conditions. This uncovers a network of 103 high-confidence proteins in close proximity to the core members IFNAR1, IFNAR2, JAK1, TYK2, STAT1, STAT2, and IRF9, and validates several known constitutive protein assemblies, while also revealing novel stimulus-dependent and -independent associations between key signaling molecules. Functional screening further identifies PJA2 as a negative regulator of IFN signaling via its E3 ubiquitin ligase activity. Mechanistically, PJA2 interacts with TYK2 and JAK1, promotes their non-degradative ubiquitination, and limits the activating phosphorylation of TYK2 thereby restraining downstream STAT signaling. Our high-resolution proximal protein landscapes provide global insights into the type I IFN signaling network, and serve as a valuable resource for future exploration of its functional complexities.


Asunto(s)
Interferón Tipo I , Janus Quinasa 1 , Receptor de Interferón alfa y beta , Factor de Transcripción STAT2 , Transducción de Señal , TYK2 Quinasa , Ubiquitinación , Humanos , Células HEK293 , Interferón Tipo I/metabolismo , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Janus Quinasa 1/metabolismo , Fosforilación , Proteoma/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , TYK2 Quinasa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
8.
Curr Opin Immunol ; 87: 102427, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38781720

RESUMEN

The essential and redundant functions of human type I and II interferons (IFNs) have been delineated over the last three decades by studies of patients with inborn errors of immunity or their autoimmune phenocopies, but much less is known about type III IFNs. Patients with cells that do not respond to type III IFNs due to inherited IL10RB deficiency display no overt viral disease, and their inflammatory disease phenotypes can be explained by defective signaling via other interleukine10RB-dependent pathways. Moreover, patients with inherited deficiencies of interferon-stimulated gene factor 3 (ISGF-3) (STAT1, STAT2, IRF9) present viral diseases also seen in patients with inherited deficiencies of the type I IFN receptor (IFNAR1/2). Finally, patients with autoantibodies neutralizing type III IFNs have no obvious predisposition to viral disease. Current findings thus suggest that type III IFNs are largely redundant in humans. The essential functions of human type III IFNs, particularly in antiviral defenses, remain to be discovered.


Asunto(s)
Interferón lambda , Interferones , Virosis , Humanos , Interferones/metabolismo , Interferones/inmunología , Virosis/inmunología , Animales , Transducción de Señal/inmunología , Factor de Transcripción STAT2/metabolismo , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT2/inmunología , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/inmunología , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/inmunología , Subunidad beta del Receptor de Interleucina-10/genética , Subunidad beta del Receptor de Interleucina-10/inmunología , Subunidad beta del Receptor de Interleucina-10/metabolismo
9.
Fish Shellfish Immunol ; 150: 109656, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38801844

RESUMEN

Interferon regulatory factor 11 (IRF11), a fish specific member of IRF family, is a transcription factor known for its positive role in teleost antiviral defense by regulating IFN expression. Despite its recognized function, the precise mechanism of IRF11 in type I IFNs production remains largely unknown. In this study, we identified IRF11 in Japanese eel, Anguilla japonica, (AjIRF11) and determined its involvement in the later phase of fish IFN production. Our results demonstrate that IRF11-induced IFN production operates through ISRE binding. Mutations in each ISRE site within the promoter of AjIFN2 or AjIFN4 abolished IRF11-mediated activation of IFN promoters. In addition, the overexpression of AjIRF11 does not significantly impact the activation of AjIFN promoters induced by RLR-related signaling pathway proteins. Furthermore, IRF11-knockdown in ZFLs (zebrafish liver cells) has no effect on the RLRs-induced expression of zebrafish IFN-φ1 and IFN-φ3, indicating that IRF11 is not involved in the RLR-mediated IFN production. However, AjIRF11 can form transcription complexes with AjSTAT1 or AjSTAT2, or form homo- or heterodimers with AjIRF1 to stimulate the transcription of type I IFNs. Overall, it is shown in this study that IRF11 can act synergistically with STAT1 and/or STAT2 for the induction of IFN.


Asunto(s)
Anguilla , Proteínas de Peces , Interferón Tipo I , Factor de Transcripción STAT1 , Factor de Transcripción STAT2 , Animales , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT2/metabolismo , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Proteínas de Peces/metabolismo , Anguilla/genética , Anguilla/inmunología , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Factores Reguladores del Interferón/inmunología , Enfermedades de los Peces/inmunología , Inmunidad Innata/genética , Pez Cebra/genética , Pez Cebra/inmunología , Regulación de la Expresión Génica/inmunología
10.
J Biol Chem ; 300(6): 107376, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38762176

RESUMEN

Liver fibrosis/cirrhosis is a pathological state caused by excessive extracellular matrix deposition. Sustained activation of hepatic stellate cells (HSC) is the predominant cause of liver fibrosis, but the detailed mechanism is far from clear. In this study, we found that long noncoding RNA Fendrr is exclusively increased in hepatocytes in the murine model of CCl4- and bile duct ligation-induced liver fibrosis, as well as in the biopsies of liver cirrhosis patients. In vivo, ectopic expression of Fendrr aggravated the severity of CCl4-induced liver fibrosis in mice. In contrast, inhibiting Fendrr blockaded the activation of HSC and ameliorated CCl4-induced liver fibrosis. Our mechanistic study showed that Fendrr binds to STAT2 and enhances its enrichment in the nucleus, which then promote the expression of interleukin 6 (IL-6), and, ultimately, activates HSC in a paracrine manner. Accordingly, disrupting the interaction between Fendrr and STAT2 by ectopic expression of a STAT2 mutant attenuated the profibrotic response inspired by Fendrr in the CCl4-induced liver fibrosis. Notably, the increase of Fendrr in patient fibrotic liver is positively correlated with the severity of fibrosis and the expression of IL-6. Meanwhile, hepatic IL-6 positively correlates with the extent of liver fibrosis and HSC activation as well, thus suggesting a causative role of Fendrr in HSC activation and liver fibrosis. In conclusion, these observations identify an important regulatory cross talk between hepatocyte Fendrr and HSC activation in the progression of liver fibrosis, which might represent a potential strategy for therapeutic intervention.


Asunto(s)
Hepatocitos , Interleucina-6 , Cirrosis Hepática , ARN Largo no Codificante , Animales , Humanos , Masculino , Ratones , Tetracloruro de Carbono/toxicidad , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Hepatocitos/metabolismo , Hepatocitos/patología , Interleucina-6/metabolismo , Interleucina-6/genética , Cirrosis Hepática/metabolismo , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Ratones Endogámicos C57BL , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Factor de Transcripción STAT2/metabolismo , Factor de Transcripción STAT2/genética
11.
Am J Physiol Cell Physiol ; 326(6): C1697-C1709, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38586875

RESUMEN

Alzheimer's disease (AD) is the leading cause of dementia and presents a considerable disease burden. Its pathology involves substantial neuronal loss, primarily attributed to neuronal apoptosis. Although sirtuin 4 (SIRT4) has been implicated in regulating apoptosis in various diseases, the role of SIRT4 in AD pathology remains unclear. The study used APP/PS1 mice as an animal model of AD and amyloid-ß (Aß)1-42-treated HT-22 cells as an AD cell model. SIRT4 expression was determined by quantitative real-time polymerase chain reaction, Western blot, and immunofluorescence. A Sirt4 knockdown model was established by intracranial injection of lentivirus-packaged sh-SIRT4 and cellular lentivirus transfection. Immunohistochemistry and flow cytometry were used to examine Aß deposition in mice and apoptosis, respectively. Protein expression was assessed by Western blot analysis. The UCSC and JASPAR databases were used to predict upstream transcription factors of Sirt4. Subsequently, the binding of transcription factors to Sirt4 was analyzed using a dual-luciferase assay and chromatin immunoprecipitation. SIRT4 expression was upregulated in both APP/PS1 mice and Aß-treated HT-22 cells compared with their respective control groups. Sirt4 knockdown in animal and cellular models of AD resulted in reduced apoptosis, decreased Aß deposition, and amelioration of learning and memory impairments in mice. Mechanistically, SIRT4 modulates apoptosis via the mTOR pathway and is negatively regulated by the transcription factor signal transducer and activator of transcription 2 (STAT2). Our study findings suggest that targeting the STAT2-SIRT4-mTOR axis may offer a new treatment approach for AD.NEW & NOTEWORTHY The study reveals that in Alzheimer's disease models, SIRT4 expression increases, contributing to neuronal apoptosis and amyloid-ß deposition. Reducing SIRT4 lessens apoptosis and amyloid-ß accumulation, improving memory in mice. This process involves the mTOR pathway, regulated by STAT2 transcription factor. These findings suggest targeting the STAT2-SIRT4-mTOR axis as a potential Alzheimer's treatment strategy.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Apoptosis , Modelos Animales de Enfermedad , Ratones Transgénicos , Neuronas , Factor de Transcripción STAT2 , Transducción de Señal , Sirtuinas , Serina-Treonina Quinasas TOR , Animales , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Sirtuinas/metabolismo , Sirtuinas/genética , Serina-Treonina Quinasas TOR/metabolismo , Ratones , Neuronas/metabolismo , Neuronas/patología , Factor de Transcripción STAT2/metabolismo , Factor de Transcripción STAT2/genética , Péptidos beta-Amiloides/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Línea Celular , Proteínas Mitocondriales
12.
Fish Shellfish Immunol ; 148: 109510, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38521143

RESUMEN

The signal transducer and activator of transcription 2 (STAT2), a downstream factor of type I interferons (IFNs), is a key component of the cellular antiviral immunity response. However, the role of STAT2 in the upstream of IFN signaling, such as the regulation of pattern recognition receptors (PRRs), remains unknown. In this study, STAT2 homologue of black carp (Mylopharyngodon piceus) has been cloned and characterized. The open reading frame (ORF) of bcSTAT2 comprises 2523 nucleotides and encodes 841 amino acids, which presents the conserved structure to that of mammalian STAT2. The dual-luciferase reporter assay and the plaque assay showed that bcSTAT2 possessed certain IFN-inducing ability and antiviral ability against both spring viremia of carp virus (SVCV) and grass carp reovirus (GCRV). Interestingly, we detected the association between bcSTAT2 and bcRIG-I through co-immunoprecipitation (co-IP) assay. Moreover, when bcSTAT2 was co-expressed with bcRIG-I, bcSTAT2 obviously suppressed bcRIG-I-induced IFN expression and antiviral activity. The subsequent co-IP assay and immunoblotting (IB) assay further demonstrated that bcSTAT2 inhibited K63-linked polyubiquitination but not K48-linked polyubiquitination of bcRIG-I, however, did not affect the oligomerization of bcRIG-I. Thus, our data conclude that black carp STAT2 negatively regulates RIG-I through attenuates its K63-linked ubiquitination, which sheds a new light on the regulation of the antiviral innate immunity cascade in vertebrates.


Asunto(s)
Carpas , Enfermedades de los Peces , Infecciones por Reoviridae , Reoviridae , Infecciones por Rhabdoviridae , Animales , Carpas/genética , Carpas/metabolismo , Infecciones por Rhabdoviridae/veterinaria , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT2/metabolismo , Reoviridae/fisiología , Inmunidad Innata/genética , Proteínas de Peces , Mamíferos/metabolismo
13.
Immunology ; 172(3): 440-450, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38514903

RESUMEN

Analysis of genetically defined immunodeficient patients allows study of the effect of the absence of specific proteins on human immune function in real-world conditions. Here we have addressed the importance of type I interferon signalling for human NK cell development by studying the phenotype and function of circulating NK cells isolated from patients suffering primary immunodeficiency disease due to mutation of either the human interferon regulatory factor 9 (IRF9) or the signal transducer and activator of transcription 2 (STAT2) genes. IRF9, together with phosphorylated STAT1 and STAT2, form a heterotrimer called interferon stimulated gene factor 3 (ISGF3) which promotes the expression of hundreds of IFN-stimulated genes that mediate antiviral function triggered by exposure to type I interferons. IRF9- and STAT2-deficient patients are unable to respond efficiently to stimulation by type I interferons and so our experiments provide insights into the importance of type I interferon signalling and the consequences of its impairment on human NK cell biology. Surprisingly, the NK cells of these patients display essentially normal phenotype and function.


Asunto(s)
Interferón Tipo I , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón , Células Asesinas Naturales , Factor de Transcripción STAT2 , Transducción de Señal , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Factor de Transcripción STAT2/metabolismo , Factor de Transcripción STAT2/genética , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Interferón Tipo I/metabolismo , Mutación , Diferenciación Celular , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT1/genética , Células Cultivadas
14.
Dev Cell ; 59(8): 1028-1042.e5, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38452758

RESUMEN

The interferon signaling pathway is critical for host defense by serving diverse functions in both innate and adaptive immune responses. Here, we show that type I gamma phosphatidylinositol phosphate 5-kinase i5 (PIPKIγi5), an enzyme that synthesizes phosphatidylinositol-4,5-bisphosphate (PI4,5P2), controls the sensitivity to interferon in both human and mouse cells. PIPKIγi5 directly binds to the interferon-gamma (IFN-γ) downstream effector signal transducer and activator of transcription 1 (STAT1), which suppresses the STAT1 dimerization, IFN-γ-induced STAT1 nuclear translocation, and transcription of IFN-γ-responsive genes. Depletion of PIPKIγi5 significantly enhances IFN-γ signaling and strengthens an antiviral response. In addition, PIPKIγi5-synthesized PI4,5P2 can bind to STAT1 and promote the PIPKIγi5-STAT1 interaction. Similar to its interaction with STAT1, PIPKIγi5 is capable of interacting with other members of the STAT family, including STAT2 and STAT3, thereby suppressing the expression of genes mediated by these transcription factors. These findings identify the function of PIPKIγi5 in immune regulation.


Asunto(s)
Interferón gamma , Fosfotransferasas (Aceptor de Grupo Alcohol) , Transducción de Señal , Animales , Humanos , Ratones , Células HEK293 , Interferón gamma/metabolismo , Interferón gamma/farmacología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Unión Proteica , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética
15.
Front Immunol ; 15: 1331731, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38384473

RESUMEN

The establishment of a virus infection is the result of the pathogen's ability to replicate in a hostile environment generated by the host's immune system. Here, we found that ISG15 restricts Dengue and Zika viruses' replication through the stabilization of its binding partner USP18. ISG15 expression was necessary to control DV replication driven by both autocrine and paracrine type one interferon (IFN-I) signaling. Moreover, USP18 competes with NS5-mediated STAT2 degradation, a major mechanism for establishment of flavivirus infection. Strikingly, reconstitution of USP18 in ISG15-deficient cells was sufficient to restore the STAT2's stability and restrict virus growth, suggesting that the IFNAR-mediated ISG15 activity is also antiviral. Our results add a novel layer of complexity in the virus/host interaction interface and suggest that NS5 has a narrow window of opportunity to degrade STAT2, therefore suppressing host's IFN-I mediated response and promoting virus replication.


Asunto(s)
Dengue , Interferón Tipo I , Infección por el Virus Zika , Virus Zika , Humanos , Interferón Tipo I/metabolismo , Infección por el Virus Zika/genética , Replicación Viral , Dengue/genética , Ubiquitinas/metabolismo , Citocinas/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT2/metabolismo
16.
Commun Biol ; 7(1): 76, 2024 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-38195857

RESUMEN

Flaviviruses, including Zika virus (ZIKV) and Dengue virus (DENV), rely on their non-structural protein 5 (NS5) for both replication of viral genome and suppression of host IFN signaling. DENV and ZIKV NS5s were shown to facilitate proteosome-mediated protein degradation of human STAT2 (hSTAT2). However, how flavivirus NS5s have evolved for species-specific IFN-suppression remains unclear. Here we report structure-function characterization of the DENV serotype 2 (DENV2) NS5-hSTAT2 complex. The MTase and RdRP domains of DENV2 NS5 form an extended conformation to interact with the coiled-coil and N-terminal domains of hSTAT2, thereby promoting hSTAT2 degradation in cells. Disruption of the extended conformation of DENV2/ZIKV NS5, but not the alternative compact state, impaired their hSTAT2 binding. Our comparative structural analysis of flavivirus NS5s further reveals a conserved protein-interaction platform with subtle amino-acid variations likely underpinning diverse IFN-suppression mechanisms. Together, this study uncovers a conformational selection mechanism underlying species-specific hSTAT2 inhibition by flavivirus NS5.


Asunto(s)
Flavivirus , Factor de Transcripción STAT2 , Proteínas no Estructurales Virales , Infección por el Virus Zika , Virus Zika , Humanos , Proteolisis , Especificidad de la Especie , Factor de Transcripción STAT2/metabolismo , Proteínas no Estructurales Virales/metabolismo
18.
Int J Mol Sci ; 24(24)2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38139463

RESUMEN

In addition to the canonical ISGF3 and non-canonical STAT2/IRF9 complexes, evidence is emerging of the role of their unphosphorylated counterparts in IFN-dependent and -independent ISG transcription. To better understand the relation between ISGF3 and U-ISGF3 and STAT2/IRF9 and U-STAT2/IRF9 in IFN-I-stimulated transcriptional responses, we performed RNA-Seq and ChIP-Seq, in combination with phosphorylation inhibition and antiviral experiments. First, we identified a group of ISRE-containing ISGs that were commonly regulated in IFNα-treated WT and STAT1-KO cells. Thus, in 2fTGH and Huh7.5 WT cells, early and long-term IFNα-inducible transcription and antiviral activity relied on the DNA recruitment of the ISGF3 components STAT1, STAT2 and IRF9 in a phosphorylation- and time-dependent manner. Likewise, in ST2-U3C and Huh-STAT1KO cells lacking STAT1, delayed IFN responses correlated with DNA binding of phosphorylated STAT2/IRF9 but not U-STAT2/IRF9. In addition, comparative experiments in U3C (STAT1-KO) cells overexpressing all the ISGF3 components (ST1-ST2-IRF9-U3C) revealed U-ISGF3 (and possibly U-STAT2/IRF9) chromatin interactions to correlate with phosphorylation-independent ISG transcription and antiviral activity. Together, our data point to the dominant role of the canonical ISGF3 and non-canonical STAT2/IRF9, without a shift to U-ISGF3 or U-STAT2/IRF9, in the regulation of early and prolonged ISG expression and viral protection. At the same time, they suggest the threshold-dependent role of U-ISFG3, and potentially U-STAT2/IRF9, in the regulation of constitutive and possibly long-term IFNα-dependent responses.


Asunto(s)
Interferón Tipo I , Factor 3 de Genes Estimulados por el Interferón , Proteína 1 Similar al Receptor de Interleucina-1 , Factor de Transcripción STAT2 , Antivirales/farmacología , ADN/farmacología , Inmunoglobulinas/metabolismo , Interferón Tipo I/metabolismo , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Transducción de Señal , Factor de Transcripción STAT1/metabolismo , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Factor de Transcripción STAT2/metabolismo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...