Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.246
Filtrar
1.
Int J Mol Sci ; 24(19)2023 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-37834019

RESUMO

Janus tyrosine kinase (JAK) variants are known drivers for hematological disorders. With the full-length structure of mouse JAK1 being recently resolved, new observations on the localization of variants within closed, open, and dimerized JAK structures are possible. Full-length homology models of human wild-type JAK family members were developed using the Glassman et al. reported mouse JAK1 containing the V658F structure as a template. Many mutational sites related to proliferative hematological disorders reside in the JH2 pseudokinase domains facing the region important in dimerization of JAKs in both closed and open states. More than half of all JAK gain of function (GoF) variants are changes in polarity, while only 1.2% are associated with a change in charge. Within a JAK1-JAK3 homodimer model, IFNLR1 (PDB ID7T6F) and the IL-2 common gamma chain subunit (IL2Rγc) were aligned with the respective dimer implementing SWISS-MODEL coupled with ChimeraX. JAK3 variants were observed to encircle the catalytic site of the kinase domain, while mutations in the pseudokinase domain align along the JAK-JAK dimerization axis. FERM domains of JAK1 and JAK3 are identified as a hot spot for hematologic malignancies. Herein, we propose new allosteric surfaces for targeting hyperactive JAK dimers.


Assuntos
Neoplasias Hematológicas , Janus Quinases , Animais , Humanos , Camundongos , Janus Quinases/genética , Tirosina/genética , Janus Quinase 1/genética , Neoplasias Hematológicas/tratamento farmacológico , Neoplasias Hematológicas/genética , Mutação , Desenvolvimento de Medicamentos , Janus Quinase 2/genética , Receptores de Interferon/genética
2.
Sci Signal ; 16(806): eadf5494, 2023 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-37816090

RESUMO

Interferons (IFNs) play crucial roles in antiviral defenses. Despite using the same Janus-activated kinase (JAK)-signal transducer and activator of transcription (STAT) signaling cascade, type I and III IFN receptors differ in the magnitude and dynamics of their signaling in terms of STAT phosphorylation, gene transcription, and antiviral responses. These differences are not due to ligand-binding affinity and receptor abundance. Here, we investigated the ability of the intracellular domains (ICDs) of IFN receptors to differentiate between type I and III IFN signaling. We engineered synthetic, heterodimeric type I and III IFN receptors that were stably expressed at similar amounts in human cells and responded to a common ligand. We found that our synthetic type I IFN receptors stimulated STAT phosphorylation and gene expression to greater extents than did the corresponding type III IFN receptors. Furthermore, we identified short "box motifs" within ICDs that bind to JAK1 that were sufficient to encode differences between the type I and III IFN receptors. Together, our results indicate that specific regions within the ICDs of IFN receptor subunits encode different downstream signaling strengths that enable type I and III IFN receptors to produce distinct signaling outcomes.


Assuntos
Interferon Tipo I , Receptores de Interferon , Humanos , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Ligantes , Interferons/metabolismo , Transdução de Sinais , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Janus Quinases/metabolismo , Fosforilação , Antivirais/farmacologia , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo
3.
J Interferon Cytokine Res ; 43(9): 427-434, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37725010

RESUMO

Biliary atresia (BA) is a life-threatening cholangiopathy occurring in infancy, the most common indication for pediatric liver transplantation. The etiology of BA remains unknown; however, a viral etiology has been proposed as multiple viruses have been detected in explants of infants afflicted with BA. In the murine model of BA, Rhesus rotavirus (RRV) infection of newborn BALB/c pups results in a cholangiopathy that mirrors human BA. Infected BALB/c pups experience 100% symptomatology and mortality, while C57BL/6 mice are asymptomatic. Interferon-λ (IFN-λ) is an epithelial cytokine that provides protection against viral infection. We demonstrated that IFN-λ is highly expressed in C57BL/6, leading to reduced RRV replication. RRV-infection of C57BL/6 IFN-λ receptor knockout (C57BL/6 IFN-λR KO) pups resulted in 90% developing obstructive symptoms and 45% mortality with a higher viral titer in bile ducts and profound periportal inflammation compared to C57BL/6. Histology revealed complete biliary obstruction in symptomatic C57BL/6 IFN-λR KO pups, while C57BL/6 ducts were patent. These findings suggest that IFN-λ is critical in preventing RRV replication. Deficiency in IFN-λ permits RRV infection, which triggers the inflammatory cascade causing biliary obstruction. Further IFN-λ study is warranted as it may play an important role in infant susceptibility to BA.


Assuntos
Atresia Biliar , Colestase , Receptores de Interferon , Animais , Camundongos , Atresia Biliar/genética , Modelos Animais de Doenças , Interferon lambda/metabolismo , Interferons , Camundongos Endogâmicos C57BL , Receptores de Interferon/genética , Receptores de Interferon/metabolismo
4.
J Interferon Cytokine Res ; 43(9): 403-413, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37499093

RESUMO

Type III interferons (IFN-lambdas, IFN-λs) are important antiviral cytokines that can also modulate immune responses by acting through a heterodimeric receptor composed of the specific and limited expressed IFN-λR1 chain and the ubiquitous IL-10R2 chain, which is shared with IL-10 family cytokines. Conflicting data have been reported regarding which cells express the IFN-λR1 subunit and directly respond to IFN-λs. This is, in part, owing to transcript levels of the IFN-λR1 gene, IFNLR1, not always correlating with cell surface protein levels. In this study, we tested a panel of novel monoclonal antibodies (mAbs) that specifically recognize human IFN-λR1. Initially, antigen specificity was confirmed by enzyme-linked immunosorbent assay (ELISA), from which a subset of antibodies was selected for additional flow cytometry and neutralization assays. We further characterized two antibodies based on their strong ELISA binding activity (HLR1 and HLR14) and found only HLR14 could reliably detect cell surface IFN-λR1 protein on a variety of cell lines by flow cytometry. HLR14 could also detect IFN-λR1 protein on certain primary human blood cells, including plasmacytoid dendritic cells and B cells from peripheral blood. Availability of the HLR14 mAb will enable the quantification of IFN-λR1 protein levels on cells and better characterization of the cell specificity of the IFN-λ response.


Assuntos
Interferons , Receptores de Interferon , Humanos , Receptores de Interferon/genética , Interferon lambda , Proteínas de Membrana , Anticorpos Monoclonais , Citocinas
5.
Nat Genet ; 55(6): 1034-1047, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37277650

RESUMO

Down syndrome (DS), the genetic condition caused by trisomy 21, is characterized by variable cognitive impairment, immune dysregulation, dysmorphogenesis and increased prevalence of diverse co-occurring conditions. The mechanisms by which trisomy 21 causes these effects remain largely unknown. We demonstrate that triplication of the interferon receptor (IFNR) gene cluster on chromosome 21 is necessary for multiple phenotypes in a mouse model of DS. Whole-blood transcriptome analysis demonstrated that IFNR overexpression associates with chronic interferon hyperactivity and inflammation in people with DS. To define the contribution of this locus to DS phenotypes, we used genome editing to correct its copy number in a mouse model of DS, which normalized antiviral responses, prevented heart malformations, ameliorated developmental delays, improved cognition and attenuated craniofacial anomalies. Triplication of the Ifnr locus modulates hallmarks of DS in mice, suggesting that trisomy 21 elicits an interferonopathy potentially amenable to therapeutic intervention.


Assuntos
Síndrome de Down , Cardiopatias Congênitas , Animais , Camundongos , Síndrome de Down/genética , Receptores de Interferon/genética , Interferons , Fenótipo , Modelos Animais de Doenças
6.
BMC Pediatr ; 23(1): 278, 2023 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-37277724

RESUMO

BACKGROUND: Interferon-gamma receptor deficiency is a heterogeneous spectrum of disease which involves mutations in IFNGR1, IFNGR2 genes, and the downstream signaling proteins such as STAT1. These mutations are associated with immunodeficiency 27 A and 27B, making the patient prone to mycobacterial infections. Patients with this condition are also at increased risk for affliction with viral and bacterial infections, such as with the Herpesviridae family, Listeria, and Salmonella. Moreover, SH2B3 mutation is associated with autoimmune and lymphoproliferative conditions. CASE PRESENTATION: the patient was a 19-month-old infant girl who presented with a two-week history of fever. She had near-normal flowcytometry with high IgM and IgE. She had pneumonic infiltration in her chest and right hilar and para-aortic lymphadenopathy. PCR of whole blood for Aspergillus fumigatus came back positive. In her Whole Exome Sequencing she had IFNGR1 and SH2B3 mutations. CONCLUSION: systemic fungal infections such as Aspergillosis can occur in patients with interferon-gamma receptor one deficiency. This type of immunodeficiency should be considered in treating patients with systemic Aspergillosis.


Assuntos
Aspergilose , Síndromes de Imunodeficiência , Lactente , Feminino , Humanos , Interferon gama/genética , Aspergilose/diagnóstico , Aspergilose/genética , Receptores de Interferon/genética , Síndromes de Imunodeficiência/complicações , Síndromes de Imunodeficiência/genética
7.
Cytokine ; 169: 156274, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37327531

RESUMO

Interferon-gamma (IFN-γ) is a pro-inflammatory cytokine playing essential role in immunity against tuberculosis (TB). IFN-γ performs function by binding to its receptor complex, consisting of two polypeptide chains viz. IFN-γ receptor 1 (IFN-γR1) and IFN-γ receptor 2 (IFN-γR2). Structural and functional deficiencies in IFN-γR1 can make individual vulnerable to even weak mycobacterial infections. Studies from different populations of the world have reported the association of single nucleotide polymorphisms (SNPs) present in IFNGR1 gene with TB, however, there are no such studies from India. Thus, the present study was designed to analyse the association of rs2234711 (C/T), rs7749390 (C/T) and rs1327475 (C/T) SNPs of IFNGR1 with TB in the population of North India. For the present study, 263 TB patients (at zero day of anti-tuberculosis therapy) and 256 healthy controls (HCs) were recruited. The genotyping of selected SNPs was done by high-resolution melting (HRM) curve analysis. The mRNA and surface expression data of IFNGR1 was extracted from our previous study and was grouped according to the genotypes of studied SNPs. The genotype 'TT' and 'T' allele of SNP rs2234711 (C/T) were found to be associated with TB in studied population ['T' vs 'C': OR (CI) = 1.79 (1.39-2.29); p-value < 0.0001]. The haplotypes 'C-C-C' of rs2234711-rs7749390-rs1327475 confers protection, while haplotype 'T-C-C' is a risk factor for TB in studied population. It was also found that 'TT' genotype of rs2234711 in HCs is associated with lower surface expression of IFNGR1 (p-value = 0.0078). In conclusion, 'TT' genotype is associated with lower surface expression of IFNGR1 and is increasing the susceptibility to TB in North Indian population.


Assuntos
Predisposição Genética para Doença , Polimorfismo de Nucleotídeo Único , Receptores de Interferon , Tuberculose Pulmonar , Humanos , Estudos de Casos e Controles , Genótipo , Interferon gama/genética , Polimorfismo de Nucleotídeo Único/genética , Receptores de Interferon/genética , Tuberculose Pulmonar/genética
8.
Viruses ; 15(3)2023 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-36992341

RESUMO

Interferon lambdas (IFNLs) are innate immune cytokines that induce antiviral cellular responses by signaling through a heterodimer composed of IL10RB and the interferon lambda receptor 1 (IFNLR1). Multiple IFNLR1 transcriptional variants are expressed in vivo and are predicted to encode distinct protein isoforms whose function is not fully established. IFNLR1 isoform 1 has the highest relative transcriptional expression and encodes the full-length functional form that supports canonical IFNL signaling. IFNLR1 isoforms 2 and 3 have lower relative expression and are predicted to encode signaling-defective proteins. To gain insight into IFNLR1 function and regulation, we explored how altering relative expression of IFNLR1 isoforms influenced the cellular response to IFNLs. To achieve this, we generated and functionally characterized stable HEK293T clones expressing doxycycline-inducible FLAG-tagged IFNLR1 isoforms. Minimal FLAG-IFNLR1 isoform 1 overexpression markedly increased IFNL3-dependent expression of antiviral and pro-inflammatory genes, a phenotype that could not be further augmented by expressing higher levels of FLAG-IFNLR1 isoform 1. Expression of low levels of FLAG-IFNLR1 isoform 2 led to partial induction of antiviral genes, but not pro-inflammatory genes, after IFNL3 treatment, a phenotype that was largely abrogated at higher FLAG-IFNLR1 isoform 2 expression levels. Expression of FLAG-IFNLR1 isoform 3 partially augmented antiviral gene expression after IFNL3 treatment. In addition, FLAG-IFNLR1 isoform 1 significantly reduced cellular sensitivity to the type-I IFN IFNA2 when overexpressed. These results identify a unique influence of canonical and non-canonical IFNLR1 isoforms on mediating the cellular response to interferons and provide insight into possible pathway regulation in vivo.


Assuntos
Interferon lambda , Receptores de Interferon , Humanos , Células HEK293 , Interferon lambda/metabolismo , Interferons , Isoformas de Proteínas/genética , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Fatores de Restrição Antivirais
9.
Mikrobiyol Bul ; 57(1): 83-96, 2023 01.
Artigo em Turco | MEDLINE | ID: mdl-36636848

RESUMO

Mendelian susceptibility to mycobacterial diseases (MSMD) is a rare primary immune deficiency (PID). IL-12Rß1 deficiency is the most frequently observed of more than 16 genetic defects that have been identified for MSMD. Genetic and immunological tests are remarkable in the diagnosis of PID. In this study, it was aimed to determine the expression of IFN-γR1 and IL-12Rß1 in patients with MSMD, their relatives, and healthy individuals and to evaluate the importance of flow cytometry as a fast and reliable method in the diagnosis of MSMD. IFN-γR1 and IL-12Rß1 expression levels were analyzed in 32 volunteers including six patients, six relatives, and 20 healthy individuals. The normal range of IFN-γR1 and IL-12Rß1 levels among healthy individuals were determined. IL-12Rß1 expression level in lymphocytes was found to be low in one patient's relative, and less than 1% in three patients and in one patient's relative. It was observed that the IL-12Rß1 expression levels of the patient with STAT1 deficiency were increased compared to the healthy individuals. No difference was found in the expression levels of IFN-γR1 and IL-12Rß1 in one patient, but IFN-γR1 expression was decreased in one patient compared to healthy individuals. Our results show that the determination of IL-12Rß1 and IFN-γR1 deficiencies by flow cytometry can be used as a rapid and reliable method for the diagnosis of MSMD. The use of this method as a screening test will enable early diagnosis especially in patients whose genetic diagnosis has not been confirmed and clinically compatible with MSMD. In addition, it is thought that IL-12Rß1 and IFN-γR1 range data obtained from healthy individuals will be considered as a reference source in routine and research studies to be conducted with MSMD.


Assuntos
Predisposição Genética para Doença , Infecções por Mycobacterium , Receptores de Interferon , Receptores de Interleucina-12 , Humanos , Citometria de Fluxo , Mutação , Infecções por Mycobacterium/diagnóstico , Infecções por Mycobacterium/genética , Receptores de Interleucina-12/genética , Receptores de Interferon/genética
10.
J Clin Invest ; 133(3)2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36445781

RESUMO

The absence of IFN-γ receptor (IFN-γR) or STAT1 signaling in donor cells has been shown to result in reduced induction of acute graft-versus-host disease (GVHD). In this study, we unexpectedly observed increased activation and expansion of donor lymphocytes in both lymphohematopoietic organs and GVHD target tissues of IFN-γR/STAT1-deficient recipient mice, leading to rapid mortality following the induction of GVHD. LPS-matured, BM-derived Ifngr1-/- Stat1-/- DCs (BMDCs) were more potent allogeneic stimulators and expressed increased levels of MHC II and costimulatory molecules. Similar effects were observed in human antigen-presenting cells (APCs) with knockdown of Stat1 by CRISPR/Cas9 and treatment with a JAK1/2 inhibitor. Furthermore, we demonstrated that the absence of IFN-γR/STAT1 signaling in hematopoietic APCs impaired the presentation of exogenous antigens, while promoting the presentation of endogenous antigens. Thus, the indirect presentation of host antigens to donor lymphocytes was defective in IFN-γR/STAT1-deficient, donor-derived APCs in fully donor chimeric mice. The differential effects of IFN-γR/STAT1 signaling on endogenous and exogenous antigen presentation could provide further insight into the roles of the IFN-γ/STAT1 signaling pathway in the pathogenesis of GVHD, organ rejection, and autoimmune diseases.


Assuntos
Células Apresentadoras de Antígenos , Doença Enxerto-Hospedeiro , Camundongos , Humanos , Animais , Receptores de Interferon/genética , Doença Enxerto-Hospedeiro/genética , Transdução de Sinais , Transplante de Medula Óssea/efeitos adversos , Camundongos Endogâmicos C57BL , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo
11.
Eur Rev Med Pharmacol Sci ; 26(23): 8777-8787, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36524496

RESUMO

OBJECTIVE: Previous studies suggested that single-nucleotide polymorphisms (SNPs) of interferon gamma (IFNG) and its receptor IFNGR1 may be involved in the pathogenesis of tuberculosis (TB). We aimed to examine the association of IFNG gene polymorphisms with TB in the Tibetan population and use the machine learning method to establish a clinical prediction model of TB. PATIENTS AND METHODS: A total of 613 TB patients and 603 healthy controls were selected for the study. Associations between SNPs and TB were analyzed using logistic regression, adjusted for sex and age. Clinical data and SNPs were integrated to construct a TB prediction model using random forest (RF) machine learning. RESULTS: For IFNG, rs1861494 CT was a protective factor against TB compared with TT genotype (p = 0.010). The rs1861494 C allele was a protective factor for TB (p = 0.010). For IFNGR1, the rs3799488 C allele reduced the risk of TB by 30% (p < 0.001). rs9376267 CT (p = 0.005) and TT (p = 0.001) genotypes were protective factors for TB. Compared with the rs1327475 GG genotype, the frequency of the GA genotype in the case group significantly differed from the controls (p = 0.013). rs2234711 GA (p < 0.001), AA (p < 0.001) genotype and A (p < 0.001) alleles were also associated with TB. Finally, five markers are identified using the RF model. The area under the curve (AUC) reaches 0.6 in the training set and 0.59 in the test set. CONCLUSIONS: Our study found that IFNG and IFNGR1 gene polymorphisms were associated with TB in a Tibetan population. The results also demonstrate the potential of clinical-SNPs as diagnostic tools for TB.


Assuntos
Interferon gama , Receptores de Interferon , Tuberculose , Humanos , Estudos de Casos e Controles , Predisposição Genética para Doença , Genótipo , Interferon gama/genética , Modelos Estatísticos , Polimorfismo de Nucleotídeo Único , Prognóstico , Tuberculose/genética , Receptores de Interferon/genética
12.
Mol Cell ; 82(20): 3901-3918.e7, 2022 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-36206767

RESUMO

How cancer-associated chromatin abnormalities shape tumor-immune interaction remains incompletely understood. Recent studies have linked DNA hypomethylation and de-repression of retrotransposons to anti-tumor immunity through the induction of interferon response. Here, we report that inactivation of the histone H3K36 methyltransferase NSD1, which is frequently found in squamous cell carcinomas (SCCs) and induces DNA hypomethylation, unexpectedly results in diminished tumor immune infiltration. In syngeneic and genetically engineered mouse models of head and neck SCCs, NSD1-deficient tumors exhibit immune exclusion and reduced interferon response despite high retrotransposon expression. Mechanistically, NSD1 loss results in silencing of innate immunity genes, including the type III interferon receptor IFNLR1, through depletion of H3K36 di-methylation (H3K36me2) and gain of H3K27 tri-methylation (H3K27me3). Inhibition of EZH2 restores immune infiltration and impairs the growth of Nsd1-mutant tumors. Thus, our work uncovers a druggable chromatin cross talk that regulates the viral mimicry response and enables immune evasion of DNA hypomethylated tumors.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Histona Metiltransferases , Evasão Tumoral , Animais , Camundongos , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Cromatina , Metilação de DNA , Neoplasias de Cabeça e Pescoço/genética , Histona Metiltransferases/genética , Histona Metiltransferases/metabolismo , Histonas/genética , Histonas/metabolismo , Interferons/genética , Proteínas Nucleares/metabolismo , Receptores de Interferon/genética , Retroelementos , Evasão Tumoral/genética
13.
Front Public Health ; 10: 976221, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36148347

RESUMO

The association of IFN-γ receptor 1 (IFNGR1) gene polymorphisms with tuberculosis (TB) susceptibility has not been systematically studied. We therefore conducted a meta-analysis to assess their association. Literature search was conducted in PubMed, EMBASE, Web of Science, and the Cochrane Library. Odds ratio (OR) and 95% confidence interval (CI) was pooled by the random-effect model. Statistical analyses were performed using STATA 12.0 software. Fourteen studies involved 7,699 TB cases and 8,289 controls were included in this meta-analysis. A significant association was found between the IFNGR1 rs2234711 polymorphism and TB susceptibility among Africans in dominant model (OR = 1.24, 95%CI:1.01-1.52), and among Asians in allele model (OR = 0.89, 95%CI: 0.79-0.99), homozygote model (OR = 0.82, 95%CI: 0.70-0.98) and additive model (OR = 0.90, 95%CI: 0.83-0.97). In addition, a significant association was observed between the IFNGR1 rs7749390 polymorphism and TB susceptibility among Africans in allele model (OR = 0.89, 95%CI: 0.82-0.98). No significant association was found between the IFNGR1 rs1327474 polymorphism and TB susceptibility. In summary, IFNGR1 rs2234711 polymorphism was associated with increased TB susceptibility in Africans and decreased TB susceptibility in Asians, while IFNGR1 rs7749390 polymorphism was associated with decreased TB susceptibility in Africans.


Assuntos
Predisposição Genética para Doença , Tuberculose , Humanos , Polimorfismo Genético , Receptores de Interferon/genética , Tuberculose/genética
14.
Biomed Pharmacother ; 155: 113683, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36095965

RESUMO

IFN-γ, a soluble cytokine being produced by T lymphocytes, macrophages, mucosal epithelial cells, or natural killer cells, is able to bind to the IFN-γ receptor (IFNγR) and in turn activate the Janus kinase (JAK)-signal transducer and transcription protein (STAT) pathway and induce expression of IFN-γ-stimulated genes. IFN-γ is critical for innate and adaptive immunity and aberrant IFN-γ expression and functions have been associated with different human diseases. However, the IFN-γ/IFNγR signaling could be a double-edged sword in cancer development because the tissue microenvironments could determine its anti- or pro-tumorigenic activities. The IFNγR protein consists of two IFNγR1 and IFNγR2 chains, subunits of which play different roles under certain conditions. This review assessed IFNγR polymorphisms, expression and functions in development and progression of various human diseases in an IFN-γ-dependent or independent manner. This review also discussed tumor microenvironment, microbial infection, and vital molecules in the IFN-γ upstream signaling that might regulate IFNγR expression, drug resistance, and druggable strategy, to provide evidence for further application of IFNγR.


Assuntos
Interferon gama , Neoplasias , Humanos , Interferon gama/metabolismo , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Janus Quinases/metabolismo , Neoplasias/genética , Fator de Transcrição STAT1/metabolismo , Microambiente Tumoral
15.
Virus Res ; 319: 198884, 2022 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-35931226

RESUMO

Japanese encephalitis virus (JEV) is a single-stranded positive-sense RNA virus belonging to the Flaviviridae family. The JEV is the leading cause of viral encephalitis in children and the elderly which is spread by mosquitoes. JEV infection has been established in different animal models such as mouse, hamster, guinea pig, swine, rat, monkey, rabbit by using the different routes of inoculations. Here, we have shown that the alpha/beta and gamma -receptor deficient AG129 mouse induces fatal encephalitis in both young and aged old mice, when challenged with high titer JEV Indian clinical isolate by both intraperitoneal and intradermal route. The JEV infected AG129 mouse have shown neurological symptoms, JEV-induced pathological features and supported high level viral replication. Additionally, administration of JEV in AG129 mice resulted in the induction of severe peripheral vascular permeability, which is a major hall mark of Dengue infection but not shown in JEV. Taken together, our results demonstrate interferon α/ß and γ receptors knock out AG129 mouse does not need adaptation of JEV clinical isolates and could be is a promising JEV challenge mouse model by mimicking the natural intradermal route of administration for rapid screening of novel antivirals and vaccines.


Assuntos
Vírus da Encefalite Japonesa (Espécie) , Vírus da Encefalite Japonesa (Subgrupo) , Encefalite Japonesa , Animais , Vírus da Encefalite Japonesa (Espécie)/genética , Cobaias , Camundongos , Camundongos Knockout , Coelhos , Ratos , Receptores de Interferon/genética , Vasodilatação
16.
Hum Immunol ; 83(8-9): 656-661, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35792002

RESUMO

Interferon-γ (IFN-γ) is an essential pro-inflammatory cytokine against tuberculosis (TB). To initiate immune response, IFN-γ binds to its receptor complex which consists of two subunits IFN-γ receptor 1 (IFN-γR1) and IFN-γ receptor 2 (IFN-γR2). The deficiency in either receptor subunit can alter IFN-γ signalling thus influencing host susceptibility to TB. In the present study IFN-γ receptor expression at transcriptional and translational level was analysed in pulmonary TB patients from North India. A total of 46 pulmonary TB patients (at 0 day of anti-tuberculosis therapy) and 48 healthy controls (HCs) were recruited. It was found that the mRNA expression of IFN-γR1 was decreased in male TB patients (p = 0.003). The surface expression of IFN-γR1 (p = 0.0005) and IFN-γR2 (p = 0.024) was also found to be decreased in male TB patients. In conclusion, we found sex-dependent regulation of IFN-γR1 and IFN-γR2 expression in pulmonary TB patients of studied population.


Assuntos
Receptores de Interferon , Fatores Sexuais , Tuberculose Pulmonar , Membrana Celular/metabolismo , Feminino , Humanos , Interferon gama/genética , Interferon gama/metabolismo , Masculino , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Tuberculose Pulmonar/genética
17.
J Exp Med ; 219(9)2022 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-35833912

RESUMO

Anti-interferon (IFN)-γ autoantibodies (AIGAs) are a pathogenic factor in late-onset immunodeficiency with disseminated mycobacterial and other opportunistic infections. AIGAs block IFN-γ function, but their effects on IFN-γ signaling are unknown. Using a single-cell capture method, we isolated 19 IFN-γ-reactive monoclonal antibodies (mAbs) from patients with AIGAs. All displayed high-affinity (KD < 10-9 M) binding to IFN-γ, but only eight neutralized IFN-γ-STAT1 signaling and HLA-DR expression. Signal blockade and binding affinity were correlated and attributed to somatic hypermutations. Cross-competition assays identified three nonoverlapping binding sites (I-III) for AIGAs on IFN-γ. We found that site I mAb neutralized IFN-γ by blocking its binding to IFN-γR1. Site II and III mAbs bound the receptor-bound IFN-γ on the cell surface, abolishing IFN-γR1-IFN-γR2 heterodimerization and preventing downstream signaling. Site III mAbs mediated antibody-dependent cellular cytotoxicity, probably through antibody-IFN-γ complexes on cells. Pathogenic AIGAs underlie mycobacterial infections by the dual blockade of IFN-γ signaling and by eliminating IFN-γ-responsive cells.


Assuntos
Infecções por Mycobacterium , Receptores de Interferon , Anticorpos Monoclonais , Autoanticorpos , Impedância Elétrica , Humanos , Interferon gama , Infecções por Mycobacterium/genética , Infecções por Mycobacterium/microbiologia , Receptores de Interferon/genética
19.
mBio ; 13(3): e0385721, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35471083

RESUMO

Interferon lambda (IFN-λ) (type III IFN) is constitutively secreted from human placental cells in culture and reduces Zika virus (ZIKV) transplacental transmission in mice. However, the roles of IFN-λ during healthy pregnancy and in restricting congenital infection remain unclear. Here, we used mice lacking the IFN-λ receptor (Ifnlr1-/-) to generate pregnancies lacking either maternal or fetal IFN-λ responsiveness and found that the antiviral effect of IFN-λ resulted from signaling exclusively in maternal tissues. This protective effect depended on gestational stage, as infection earlier in pregnancy (E7 rather than E9) resulted in enhanced transplacental transmission of ZIKV. In Ifnar1-/- dams, which sustain robust ZIKV infection, maternal IFN-λ signaling caused fetal resorption and intrauterine growth restriction. Pregnancy pathology elicited by poly(I·C) treatment also was mediated by maternal IFN-λ signaling, specifically in maternal leukocytes, and also occurred in a gestational stage-dependent manner. These findings identify an unexpected effect of IFN-λ signaling, specifically in maternal (rather than placental or fetal) tissues, which is distinct from the pathogenic effects of IFN-αß (type I IFN) during pregnancy. These results highlight the complexity of immune signaling at the maternal-fetal interface, where disparate outcomes can result from signaling at different gestational stages. IMPORTANCE Pregnancy is an immunologically complex situation, which must balance protecting the fetus from maternal pathogens with preventing maternal immune rejection of non-self fetal and placental tissue. Cytokines, such as interferon lambda (IFN-λ), contribute to antiviral immunity at the maternal-fetal interface. We found in a mouse model of congenital Zika virus infection that IFN-λ can have either a protective antiviral effect or cause immune-mediated pathology, depending on the stage of gestation when IFN-λ signaling occurs. Remarkably, both the protective and pathogenic effects of IFN-λ occurred through signaling exclusively in maternal immune cells rather than in fetal or placental tissues or in other maternal cell types, identifying a new role for IFN-λ at the maternal-fetal interface.


Assuntos
Doenças Fetais , Complicações Infecciosas na Gravidez , Receptores de Interferon , Infecção por Zika virus , Animais , Feminino , Doenças Fetais/tratamento farmacológico , Doenças Fetais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Placenta , Gravidez , Complicações Infecciosas na Gravidez/virologia , Receptores de Interferon/genética , Zika virus , Infecção por Zika virus/prevenção & controle
20.
Transbound Emerg Dis ; 69(5): e2164-e2174, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35403352

RESUMO

Bluetongue virus (BTV), a member of the genus Orbivirus in the family Reoviridae, is transmitted by biting midges and causes severe disease in domestic and wild ruminants. In the present study, a BTV strain, BTV-20/GX015/China/2013 (GX015), was isolated from sentinel cattle in Guangxi, China. Virus neutralization tests and phylogenetic analyses based on genomic segments 2 (S2) and 6 (S6) indicated that GX015 belongs to BTV serotype 20 (BTV-20) and represents a new topotype within BTV-20 strains, which makes GX015 the first BTV-20 strain isolated in China. Genomic analyses suggested that the 10 genomic segments of GX015 originated from a reassortment event, in which S2 and S6 are derived from exotic BTV-20 strains (South Africa or Australia), whereas the remaining eight genomic segments are apparently of Chinese origin and most likely share the same ancestor with a Taiwanese BTV-12 strain. Importantly, we evaluated the infectivity and pathogenicity of the BTV-20 strain in mice lacking the interferon receptor (IFNAR-/- mice, a good animal model for studying the pathogenesis, virulence and transmission of BTVs) and sheep for the first time, and found that GX015 causes severe disease and death in IFNAR-/- mice and clinical signs and viraemia in the natural host sheep. These results improve our understanding of the genetic characteristics, diversity and pathogenicity of BTVs, which is important for developing diagnostic methods and vaccines for the surveillance and prevention of bluetongue disease.


Assuntos
Vírus Bluetongue , Bluetongue , Doenças dos Bovinos , Doenças dos Roedores , Doenças dos Ovinos , Animais , Bluetongue/epidemiologia , Bovinos , Doenças dos Bovinos/epidemiologia , China/epidemiologia , Camundongos , Filogenia , Receptores de Interferon/genética , Sorogrupo , Ovinos , Doenças dos Ovinos/epidemiologia , Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...