Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nature ; 620(7976): 1063-1070, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37587335

RESUMEN

High-grade serous ovarian cancers have low survival rates because of their late presentation with extensive peritoneal metastases and frequent chemoresistance1, and require new treatments guided by novel insights into pathogenesis. Here we describe the intrinsic tumour-suppressive activities of interferon-ε (IFNε). IFNε is constitutively expressed in epithelial cells of the fallopian tube, the cell of origin of high-grade serous ovarian cancers, and is then lost during development of these tumours. We characterize its anti-tumour activity in several preclinical models: ovarian cancer patient-derived xenografts, orthotopic and disseminated syngeneic models, and tumour cell lines with or without mutations in Trp53 and Brca genes. We use manipulation of the IFNε receptor IFNAR1 in different cell compartments, differential exposure status to IFNε and global measures of IFN signalling to show that the mechanism of the anti-tumour activity of IFNε involves direct action on tumour cells and, crucially, activation of anti-tumour immunity. IFNε activated anti-tumour T and natural killer cells and prevented the accumulation and activation of myeloid-derived suppressor cells and regulatory T cells. Thus, we demonstrate that IFNε is an intrinsic tumour suppressor in the female reproductive tract whose activities in models of established and advanced ovarian cancer, distinct from other type I IFNs, are compelling indications of potential new therapeutic approaches for ovarian cancer.


Asunto(s)
Interferón Tipo I , Neoplasias Ováricas , Proteínas Supresoras de Tumor , Animales , Femenino , Humanos , Línea Celular Tumoral , Células Epiteliales/metabolismo , Trompas Uterinas/metabolismo , Genes BRCA1 , Genes BRCA2 , Genes p53 , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Células Asesinas Naturales/inmunología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Linfocitos T/inmunología , Linfocitos T Reguladores , Proteínas Supresoras de Tumor/inmunología , Proteínas Supresoras de Tumor/metabolismo
2.
J Interferon Cytokine Res ; 43(9): 403-413, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37499093

RESUMEN

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.


Asunto(s)
Interferones , Receptores de Interferón , Humanos , Receptores de Interferón/genética , Interferón lambda , Proteínas de la Membrana , Anticuerpos Monoclonales , Citocinas
3.
PLoS Pathog ; 19(3): e1010843, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36897927

RESUMEN

The immunological surveillance factors controlling vulnerability of the female reproductive tract (FRT) to sexually transmitted viral infections are not well understood. Interferon-epsilon (IFNɛ) is a distinct, immunoregulatory type-I IFN that is constitutively expressed by FRT epithelium and is not induced by pathogens like other antiviral IFNs α, ß and λ. We show the necessity of IFNɛ for Zika Virus (ZIKV) protection by: increased susceptibility of IFNɛ-/- mice; their "rescue" by intravaginal recombinant IFNɛ treatment and blockade of protective endogenous IFNɛ by neutralising antibody. Complementary studies in human FRT cell lines showed IFNɛ had potent anti-ZIKV activity, associated with transcriptome responses similar to IFNλ but lacking the proinflammatory gene signature of IFNα. IFNɛ activated STAT1/2 pathways similar to IFNα and λ that were inhibited by ZIKV-encoded non-structural (NS) proteins, but not if IFNε exposure preceded infection. This scenario is provided by the constitutive expression of endogenous IFNε. However, the IFNɛ expression was not inhibited by ZIKV NS proteins despite their ability to antagonise the expression of IFNß or λ. Thus, the constitutive expression of IFNɛ provides cellular resistance to viral strategies of antagonism and maximises the antiviral activity of the FRT. These results show that the unique spatiotemporal properties of IFNε provides an innate immune surveillance network in the FRT that is a significant barrier to viral infection with important implications for prevention and therapy.


Asunto(s)
Infección por el Virus Zika , Virus Zika , Animales , Femenino , Humanos , Ratones , Antivirales/farmacología , Genitales Femeninos , Factores Inmunológicos , Interferón-alfa/farmacología , Virus Zika/genética
4.
JCI Insight ; 7(18)2022 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-35862222

RESUMEN

Although published studies have demonstrated that IFN-ε has a crucial role in regulating protective immunity in the mouse female reproductive tract, expression and regulation of IFN-ε in the human female reproductive tract (hFRT) have not been characterized to our knowledge. We obtained hFRT samples from a well-characterized cohort of women to enable us to comprehensively assess ex vivo IFN-ε expression in the hFRT at various stages of the menstrual cycle. We found that among the various types of IFNs, IFN-ε was uniquely, selectively, and constitutively expressed in the hFRT epithelium. It had distinct expression patterns in the surface and glandular epithelia of the upper hFRT compared with basal layers of the stratified squamous epithelia of the lower hFRT. There was cyclical variation of IFN-ε expression in the endometrial epithelium of the upper hFRT and not in the distal FRT, consistent with selective endometrial expression of the progesterone receptor and regulation of the IFNE promoter by progesterone. Because we showed IFN-ε stimulated important protective IFN-regulated genes in FRT epithelium, this characterization is a key element in understanding the mechanisms of hormonal control of mucosal immunity.


Asunto(s)
Endometrio , Inmunidad Innata , Interferones , Animales , Endometrio/inmunología , Epitelio/inmunología , Femenino , Regulación de la Expresión Génica , Humanos , Inmunidad Innata/genética , Interferones/genética , Interferones/metabolismo , Ratones , Progesterona/metabolismo , Regiones Promotoras Genéticas , Receptores de Progesterona/metabolismo
5.
J Leukoc Biol ; 108(3): 909-924, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-33448473

RESUMEN

The type I IFNs activate an array of signaling pathways, which are initiated after IFNs bind their cognate receptors, IFNα/ß receptor (IFNAR)1 and IFNAR2. These signals contribute to many aspects of human health including defense against pathogens, cancer immunosurveillance, and regulation of inflammation. How these cytokines interact with their receptors influences the quality of these signals. As such, the integrity of receptor structure is pivotal to maintaining human health and the response to immune stimuli. This review brings together genome wide association studies and clinical reports describing the association of nonsynonymous IFNAR1 and IFNAR2 polymorphisms with clinical disease, including altered susceptibility to viral and bacterial pathogens, autoimmune diseases, cancer, and adverse reactions to live-attenuated vaccines. We describe the amino acid substitutions or truncations induced by these polymorphisms and, using the knowledge of IFNAR conformational changes, IFNAR-IFN interfaces and overall structure-function relationship of the signaling complexes, we hypothesize the effect of these polymorphisms on receptor structure. That these predicted changes to IFNAR structure are associated with clinical manifestations of human disease, highlights the importance of IFNAR structural integrity to maintaining functional quality of these receptor-mediated responses. Type I IFNs are pivotal to innate immune responses and ultimately, to human health. Understanding the consequences of altered structure on the actions of these clinically significant cell receptors provides important information on the roles of IFNARs in health and disease.


Asunto(s)
Polimorfismo de Nucleótido Simple , Receptor de Interferón alfa y beta/genética , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Codón sin Sentido/genética , Cristalografía por Rayos X , Susceptibilidad a Enfermedades , Humanos , Inmunidad Innata , Inmunogenicidad Vacunal , Ligandos , Macrófagos/inmunología , Mamíferos/genética , Ratones , Modelos Moleculares , Unión Proteica , Conformación Proteica , Dominios Proteicos , Receptor de Interferón alfa y beta/química , Receptor de Interferón alfa y beta/fisiología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal , Relación Estructura-Actividad , Tuberculosis/inmunología
6.
Semin Immunol ; 43: 101328, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31734130

RESUMEN

Interferon epsilon (IFNε) is a type I IFN with unusual patterns of expression and therefore, function. It is constitutively expressed by reproductive tract epithelium and regulated by hormones during estrus cycle, reproduction, and menopause and by exogenous hormones. The IFNe protein is encoded by a gene in the type I IFN locus, binds to IFNAR1 and 2 which are required for signaling via the JAK STAT pathway. Its affinity for binding receptors and transducing signals is less potent than IFNα or ß subtypes in vitro. Nevertheless, in vivo experiments indicate its efficacy in regulating mucosal immune responses and protecting from bacterial and viral infections. These studies demonstrate a different mechanism of action to type I IFNs. In this organ system with dynamic fluxes in cellularity, requirement to tolerate an implanted fetus, and be protected from disease, there is co-option of a special IFN from a family of effective immunoregulators, with unique controls and modified potency to make it a safe and effective constitutive reproductive tract cytokine.


Asunto(s)
Inmunidad Mucosa , Infecciones/inmunología , Interferones/metabolismo , Animales , Implantación del Embrión , Femenino , Humanos , Inmunomodulación , Interferón Tipo I/genética , Interferones/genética , Quinasas Janus/metabolismo , Ciclo Menstrual , Embarazo , Reproducción , Factores de Transcripción STAT/metabolismo , Transducción de Señal
7.
J Biol Chem ; 292(18): 7554-7565, 2017 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-28289093

RESUMEN

The interaction of IFN-ß with its receptor IFNAR1 (interferon α/ß receptor subunit 1) is vital for host-protective anti-viral and anti-proliferative responses, but signaling via this interaction can be detrimental if dysregulated. Whereas it is established that IFNAR1 is an essential component of the IFNAR signaling complex, the key residues underpinning the IFN-ß-IFNAR1 interaction are unknown. Guided by the crystal structure of the IFN-ß-IFNAR1 complex, we used truncation variants and site-directed mutagenesis to investigate domains and residues enabling complexation of IFN-ß to IFNAR1. We have identified an interface on IFNAR1-subdomain-3 that is differentially utilized by IFN-ß and IFN-α for signal transduction. We used surface plasmon resonance and cell-based assays to investigate this important IFN-ß binding interface that is centered on IFNAR1 residues Tyr240 and Tyr274 binding the C and N termini of the B and C helices of IFN-ß, respectively. Using IFNAR1 and IFN-ß variants, we show that this interface contributes significantly to the affinity of IFN-ß for IFNAR1, its ability to activate STAT1, the expression of interferon stimulated genes, and ultimately to the anti-viral and anti-proliferative properties of IFN-ß. These results identify a key interface created by IFNAR1 residues Tyr240 and Tyr274 interacting with IFN-ß residues Phe63, Leu64, Glu77, Thr78, Val81, and Arg82 that underlie IFN-ß-IFNAR1-mediated signaling and biological processes.


Asunto(s)
Interferón beta/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal/fisiología , Sustitución de Aminoácidos , Animales , Línea Celular , Interferón beta/genética , Ratones , Ratones Noqueados , Mutación Missense , Dominios Proteicos , Receptor de Interferón alfa y beta/genética
8.
Immunol Cell Biol ; 95(5): 478-483, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28045025

RESUMEN

Interferon epsilon (IFNɛ) is a type I IFN that is expressed constitutively in the female reproductive tract (FRT), and contributes to protection in models of sexually transmitted infections. Using multiple cell systems, including reporter cell lines and activated peripheral blood lymphocytes (PBLs), we show that recombinant IFNɛ impairs HIV infection at stage(s) post HIV entry and up to the translation of viral proteins. Consistent with this, IFNɛ upregulated a number of host cell restriction factors that block HIV at these stages of the replication cycle. The potency of IFNɛ induction of these HIV restriction factors was comparable to conventional type I IFNs, namely IFNα and IFNß. IFNɛ also significantly reduced the infectivity of progeny virion particles likely by inducing expression of HIV restriction factors, such as IFITM3, which act at that stage of infection. Thus, our data demonstrate that human IFNɛ suppresses HIV replication at multiple stages of infection.


Asunto(s)
Infecciones por VIH/virología , VIH-1/fisiología , Interferones/metabolismo , Replicación Viral , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Células Epiteliales/virología , Infecciones por VIH/patología , Células HeLa , Humanos , Interferón-alfa/metabolismo , Fitohemaglutininas/farmacología , Linfocitos T/efectos de los fármacos , Linfocitos T/patología , Linfocitos T/virología , Virión/efectos de los fármacos , Virión/metabolismo , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA