Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2023 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-36865100

RESUMEN

Interferons are essential for innate and adaptive immune responses against a wide variety of pathogens. Interferon lambda (IFN-λ) protects mucosal barriers during pathogen exposure. The intestinal epithelium is the first contact site for Toxoplasma gondii (T. gondii) with its hosts and the first defense line that limits parasite infection. Knowledge of very early T. gondii infection events in the gut tissue is limited and a possible contribution of IFN-λ has not been investigated so far. Here, we demonstrate with systemic interferon lambda receptor (IFNLR1) and conditional (Villin-Cre) knockout mouse models and bone marrow chimeras of oral T. gondii infection and mouse intestinal organoids a significant impact of IFN-λ signaling in intestinal epithelial cells and neutrophils to T. gondii control in the gastrointestinal tract. Our results expand the repertoire of interferons that contribute to the control of T. gondii and may lead to novel therapeutic approaches against this world-wide zoonotic pathogen.

2.
J Exp Med ; 219(12)2022 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-36129445

RESUMEN

Severity of COVID-19 shows an extraordinary correlation with increasing age. We generated a mouse model for severe COVID-19 and show that the age-dependent disease severity is caused by the disruption of a timely and well-coordinated innate and adaptive immune response due to impaired interferon (IFN) immunity. Aggravated disease in aged mice was characterized by a diminished IFN-γ response and excessive virus replication. Accordingly, adult IFN-γ receptor-deficient mice phenocopied the age-related disease severity, and supplementation of IFN-γ reversed the increased disease susceptibility of aged mice. Further, we show that therapeutic treatment with IFN-λ in adults and a combinatorial treatment with IFN-γ and IFN-λ in aged Ifnar1-/- mice was highly efficient in protecting against severe disease. Our findings provide an explanation for the age-dependent disease severity and clarify the nonredundant antiviral functions of type I, II, and III IFNs during SARS-CoV-2 infection in an age-dependent manner. Our data suggest that highly vulnerable individuals could benefit from immunotherapy combining IFN-γ and IFN-λ.


Asunto(s)
COVID-19 , Animales , Antivirales , Inmunidad , Interferones , Ratones , SARS-CoV-2
3.
EMBO Rep ; 23(2): e53865, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-34927793

RESUMEN

The ongoing COVID-19 pandemic and the emergence of new SARS-CoV-2 variants of concern (VOCs) requires continued development of effective therapeutics. Recently, we identified high-affinity neutralizing nanobodies (Nbs) specific for the receptor-binding domain (RBD) of SARS-CoV-2. Taking advantage of detailed epitope mapping, we generate two biparatopic Nbs (bipNbs) targeting a conserved epitope outside and two different epitopes inside the RBD:ACE2 interface. Both bipNbs bind all currently circulating VOCs with high affinities and are capable to neutralize cellular infection with VOC B.1.351 (Beta) and B.1.617.2 (Delta) in vitro. To assess if the bipNbs NM1267 and NM1268 confer protection against SARS-CoV-2 infection in vivo, human ACE2 transgenic mice are treated intranasally before infection with a lethal dose of SARS-CoV-2 B.1, B.1.351 (Beta) or B.1.617.2 (Delta). Nb-treated mice show significantly reduced disease progression and increased survival rates. Histopathological analyses further reveal a drastically reduced viral load and inflammatory response in lungs. These data suggest that both bipNbs are broadly active against a variety of emerging SARS-CoV-2 VOCs and represent easily applicable drug candidates.


Asunto(s)
COVID-19 , Anticuerpos de Dominio Único , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Humanos , Ratones , Ratones Transgénicos , Pandemias , SARS-CoV-2 , Anticuerpos de Dominio Único/genética , Glicoproteína de la Espiga del Coronavirus
4.
Sci Transl Med ; 13(621): eabi7826, 2021 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-34609205

RESUMEN

Broadly neutralizing antibodies are an important treatment for individuals with coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Antibody-based therapeutics are also essential for pandemic preparedness against future Sarbecovirus outbreaks. Camelid-derived single domain antibodies (VHHs) exhibit potent antimicrobial activity and are being developed as SARS-CoV-2­neutralizing antibody-like therapeutics. Here, we identified VHHs that neutralize both SARS-CoV-1 and SARS-CoV-2, including now circulating variants. We observed that the VHHs bound to a highly conserved epitope in the receptor binding domain of the viral spike protein that is difficult to access for human antibodies. Structure-guided molecular modeling, combined with rapid yeast-based prototyping, resulted in an affinity enhanced VHH-human immunoglobulin G1 Fc fusion molecule with subnanomolar neutralizing activity. This VHH-Fc fusion protein, produced in and purified from cultured Chinese hamster ovary cells, controlled SARS-CoV-2 replication in prophylactic and therapeutic settings in mice expressing human angiotensin converting enzyme 2 and in hamsters infected with SARS-CoV-2. These data led to affinity-enhanced selection of the VHH, XVR011, a stable anti­COVID-19 biologic that is now being evaluated in the clinic.


Asunto(s)
COVID-19 , Glicoproteína de la Espiga del Coronavirus , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Humanos , Modelos Animales , SARS-CoV-2
5.
Front Immunol ; 12: 749325, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34659250

RESUMEN

Previous work showed that interferon-λ (IFN-λ) can trigger the synthesis of thymic stromal lymphopoietin (TSLP) by specialized epithelial cells in the upper airways of mice, thereby improving the performance of intranasally administered influenza vaccines. Here we demonstrate that protein-only influenza vaccines containing either IFN-λ or TSLP boosted antigen-specific IgG1 and IgA responses and enhanced the resistance of mice to influenza virus challenge, irrespective of whether the vaccines were applied via the intranasal or the rectal route. TSLP receptor deficiency negatively influenced vaccine-induced antiviral immunity by impairing the migration of dendritic cells from the airways to the draining lymph nodes of immunized mice, thereby restraining follicular helper T cell and germinal center B cell responses. As previously observed during intranasal vaccination, the adjuvant effect of IFN-λ on a rectally administered influenza vaccine was no longer observed when TSLP receptor-deficient mice were used for immunization, highlighting the central role of the IFN-λ/TSLP axis for vaccine-induced antiviral immunity in the mucosa.


Asunto(s)
Citocinas/administración & dosificación , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Vacunas contra la Influenza/administración & dosificación , Interferones/administración & dosificación , Infecciones por Orthomyxoviridae/prevención & control , Vacunas de Subunidad/administración & dosificación , Administración Intranasal , Administración Rectal , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Líquido del Lavado Bronquioalveolar/inmunología , Femenino , Inmunoglobulinas/genética , Virus de la Influenza A , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Citocinas/genética , Linfopoyetina del Estroma Tímico
6.
PLoS One ; 16(8): e0247738, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34383769

RESUMEN

The commensal microbiota regulates susceptibility to enteric pathogens by fine-tuning mucosal innate immune responses, but how susceptibility to enteric viruses is shaped by the microbiota remains incompletely understood. Past reports have indicated that commensal bacteria may either promote or repress rotavirus replication in the small intestine of mice. We now report that rotavirus replicated more efficiently in the intestines of germ-free and antibiotic-treated mice compared to animals with an unmodified microbiota. Antibiotic treatment also facilitated rotavirus replication in type I and type III interferon (IFN) receptor-deficient mice, revealing IFN-independent proviral effects. Expression of interleukin-22 (IL-22) was strongly diminished in the intestine of antibiotic-treated mice. Treatment with exogenous IL-22 blocked rotavirus replication in microbiota-depleted wild-type and Stat1-/- mice, demonstrating that the antiviral effect of IL-22 in animals with altered microbiome is not dependent on IFN signaling. In antibiotic-treated animals, IL-22-induced a specific set of genes including Fut2, encoding fucosyl-transferase 2 that participates in the biosynthesis of fucosylated glycans which can mediate rotavirus binding. Interestingly, IL-22 also blocked rotavirus replication in antibiotic-treated Fut2-/- mice. Furthermore, IL-22 inhibited rotavirus replication in antibiotic-treated mice lacking key molecules of the necroptosis or pyroptosis pathways of programmed cell death. Taken together, our results demonstrate that IL-22 determines rotavirus susceptibility of antibiotic-treated mice, yet the IL-22-induced effector molecules conferring rotavirus resistance remain elusive.


Asunto(s)
Antibacterianos/efectos adversos , Interleucinas/metabolismo , Infecciones por Rotavirus/etiología , Animales , Antibacterianos/farmacología , Susceptibilidad a Enfermedades , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Perfilación de la Expresión Génica , Interleucinas/fisiología , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Rotavirus/fisiología , Interleucina-22
7.
Sci Immunol ; 6(59)2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33990378

RESUMEN

Inflammatory diseases are frequently treated with Janus kinase (JAK) inhibitors to diminish cytokine signaling. These treatments can lead to inadvertent immune suppression and may increase the risk of viral infection. Tyrosine kinase 2 (TYK2) is a JAK family member required for efficient type I interferon (IFN-α/ß) signaling. We report here that selective TYK2 inhibition preferentially blocked potentially detrimental type I IFN signaling, whereas IFN-λ-mediated responses were largely preserved. In contrast, the clinically used JAK1/2 inhibitor baricitinib was equally potent in blocking IFN-α/ß- or IFN-λ-driven responses. Mechanistically, we showed that epithelial cells did not require TYK2 for IFN-λ-mediated signaling or antiviral protection. TYK2 deficiency diminished IFN-α-induced protection against lethal influenza virus infection in mice but did not impair IFN-λ-mediated antiviral protection. Our findings suggest that selective TYK2 inhibitors used in place of broadly acting JAK1/2 inhibitors may represent a superior treatment option for type I interferonopathies to counteract inflammatory responses while preserving antiviral protection mediated by IFN-λ.


Asunto(s)
Virus de la Influenza A , Interferones/inmunología , Infecciones por Orthomyxoviridae/inmunología , TYK2 Quinasa/antagonistas & inhibidores , Animales , Azetidinas/farmacología , Células Cultivadas , Células Epiteliales/inmunología , Expresión Génica , Compuestos Heterocíclicos/farmacología , Humanos , Inhibidores de las Cinasas Janus/farmacología , Masculino , Ratones Noqueados , Neutrófilos/inmunología , Purinas/farmacología , Pirazoles/farmacología , Sulfonamidas/farmacología , TYK2 Quinasa/genética , TYK2 Quinasa/inmunología
8.
J Interferon Cytokine Res ; 40(6): 292-300, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32364818

RESUMEN

Interferon-λ (IFN-λ) plays an important role in mucosal immunity, but reliable information regarding the expression of the IFN-λ receptor in individual cells is still missing. One reason for this knowledge gap is the lack of antibodies that specifically recognize the unique IFNLR1 subunit of the dimeric IFN-λ receptor complex. In this study, we investigated whether a reporter mouse carrying a bacterial ß-galactosidase gene inserted into the Ifnlr1 locus could be used to visualize IFN-λ receptor-expressing cells in whole organs. First we confirmed that insertion of the reporter cassette inactivated the Ifnlr1 gene, and that gene function could be restored by removing the ß-galactosidase insert by site-specific recombination. When whole tissues were analyzed, prominent ß-galactosidase activity was confined to the intestinal tract of reporter mice. However, only the snout expressed ß-galactosidase at levels high enough for reliable detection in whole tissue extracts. Interestingly, individual epithelial cells in the upper airways expressed ß-galactosidase activity to variable degrees as determined by flow cytometry and histology, suggesting a remarkable heterogeneity in IFNLR1 expression levels. Taken together, our results demonstrate a surprisingly strong within- and cross-tissue heterogeneity of IFNLR1 expression that may have physiological implications.


Asunto(s)
Receptores de Interferón/genética , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptor de Interferón gamma
9.
J Virol ; 93(23)2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31511392

RESUMEN

Type I and type III interferons (IFNs) can promote adaptive immune responses in mice and improve vaccine-induced resistance to viral infections. The adjuvant effect of type III IFN (IFN-λ) specifically boosts mucosal immunity by an indirect mechanism, involving IFN-λ-induced production of thymic stromal lymphopoietin (TSLP), a cytokine that activates immune cells. To date, it remained unclear whether the previously described adjuvant effect of type I IFN (IFN-α/ß) would also depend on TSLP and whether type I IFN stimulates different antibody subtypes. Here, we show that after infection with a live attenuated influenza virus, mice lacking functional type I IFN receptors failed to produce normal amounts of virus-specific IgG2c and IgA antibodies. In contrast, mice lacking functional IFN-λ receptors contained normal levels of virus-specific IgG2c but had reduced IgG1 and IgA antibody levels. When applied together with protein antigen, IFN-α stimulated the production of antigen-specific IgA and IgG2c to a greater extent than IgG1, irrespective of whether the mice expressed functional TSLP receptors and irrespective of whether the vaccine was applied by the intranasal or the intraperitoneal route. Taken together, these results demonstrate that the adjuvant activities of type I and type III IFNs are mechanistically distinct.IMPORTANCE Interferons can shape antiviral immune responses, but it is not well understood how they influence vaccine efficacy. We find that type I IFN preferentially promotes the production of antigen-specific IgG2c and IgA antibodies after infection with a live attenuated influenza virus or after immunization with influenza subunit vaccines. In contrast, type III IFN specifically enhances influenza virus-specific IgG1 and IgA production. The adjuvant effect of type I IFN was not dependent on TSLP, which is essential for the adjuvant effect of type III IFN. Type I IFN boosted vaccine-induced antibody production after immunization by the intranasal or the intraperitoneal route, whereas type III IFN exhibited its adjuvant activity only when the vaccine was delivered by the mucosal route. Our findings demonstrate that type I and type III IFNs trigger distinct pathways to enhance the efficacy of vaccines. This knowledge might be used to design more efficient vaccines against infectious diseases.


Asunto(s)
Inmunidad Adaptativa/inmunología , Adyuvantes Inmunológicos , Vacunas contra la Influenza/inmunología , Interferones/inmunología , Animales , Formación de Anticuerpos/inmunología , Citocinas , Modelos Animales de Enfermedad , Femenino , Inmunidad Mucosa/inmunología , Inmunización , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulinas/genética , Interferón Tipo I , Interferones/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/virología , Receptores de Citocinas/genética , Vacunación , Interferón lambda , Linfopoyetina del Estroma Tímico
10.
J Gen Virol ; 97(9): 2096-2103, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27439314

RESUMEN

Avian bornaviruses are the causative agents of proventricular dilatation disease (PDD), a widely distributed and often fatal disease in captive psittacines. Because neither specific prevention measures nor therapies against PDD and bornavirus infections are currently available, new antiviral strategies are required to improve animal health. We show here that the nucleoside analogue ribavirin inhibited bornavirus activity in a polymerase reconstitution assay and reduced viral load in avian cell lines infected with two different parrot bornaviruses. Furthermore, we observed that ribavirin enhanced type I IFN signalling in avian cells. Combined treatment of avian bornavirus-infected cells with ribavirin and recombinant IFN-α strongly enhanced the antiviral efficiency compared to either drug alone. The combined use of ribavirin and type I IFN might represent a promising new strategy for therapeutic treatment of captive parrots persistently infected with avian bornaviruses.


Asunto(s)
Antivirales/farmacología , Bornaviridae/efectos de los fármacos , Interferón-alfa/farmacología , Ribavirina/farmacología , Animales , Línea Celular , Pruebas de Sensibilidad Microbiana , Loros , Carga Viral
11.
J Virol ; 87(3): 1912-5, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23192869

RESUMEN

Resistance of influenza A viruses to neuraminidase inhibitors can arise through mutations in the neuraminidase (NA) gene. We show here that a Q136K mutation in the NA of the 2009 pandemic H1N1 virus confers a high degree of resistance to zanamivir. Resistance is accompanied by reduced numbers of NA molecules in viral particles and reduced intrinsic enzymatic activity of mutant NA. Interestingly, the Q136K mutation strongly impairs viral fitness in the guinea pig transmission model.


Asunto(s)
Antivirales/farmacología , Farmacorresistencia Viral , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Neuraminidasa/genética , Proteínas Virales/genética , Factores de Virulencia/genética , Zanamivir/farmacología , Sustitución de Aminoácidos , Animales , Modelos Animales de Enfermedad , Cobayas , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Datos de Secuencia Molecular , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Missense , Neuraminidasa/metabolismo , Infecciones por Orthomyxoviridae/transmisión , Infecciones por Orthomyxoviridae/virología , ARN Viral/genética , Análisis de Secuencia de ADN , Proteínas Virales/metabolismo , Virulencia , Factores de Virulencia/metabolismo
12.
J Gen Virol ; 93(Pt 3): 555-559, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22170637

RESUMEN

Types I and III interferons (IFNs) elicit protective antiviral immune responses during influenza virus infection. Although many cell types can synthesize IFN in response to virus infection, it remains unclear which IFN sources contribute to antiviral protection in vivo. We found that mice carrying functional alleles of the Mx1 influenza virus resistance gene partially lost resistance to infection with a highly pathogenic H7N7 influenza A virus strain if Toll-like receptor 7 (TLR7) signalling was compromised. This effect was achieved by deleting either the TLR7 gene or the gene encoding the TLR7 adaptor molecule MyD88. A similar decrease of influenza virus resistance was observed when animals were deprived of plasmacytoid dendritic cells (pDCs) at day 1 post-infection. Our results provide in vivo proof that pDCs contribute to the protection of the lung against influenza A virus infections, presumably via signals from TLR7.


Asunto(s)
Células Dendríticas/inmunología , Subtipo H7N7 del Virus de la Influenza A/inmunología , Subtipo H7N7 del Virus de la Influenza A/patogenicidad , Glicoproteínas de Membrana/inmunología , Infecciones por Orthomyxoviridae/inmunología , Transducción de Señal , Receptor Toll-Like 7/inmunología , Animales , Resistencia a la Enfermedad/inmunología , Femenino , Eliminación de Gen , Masculino , Glicoproteínas de Membrana/genética , Ratones , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Receptor Toll-Like 7/genética
13.
J Interferon Cytokine Res ; 27(2): 97-101, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17316136

RESUMEN

Recognition of pathogens by toll-like receptors (TLRs) causes activation of signaling cascades that trigger cytokine secretion and, ultimately, innate immunity. Genes encoding proteins with substantial homology to mammalian TLR1, TLR2, TLR3, TLR4, TLR5, and TLR7 are present in the chicken genome, whereas orthologs of TLR8, TLR9, and TLR10 seem to be defective or missing. Except for chicken TLR2 (ChTLR2), which was previously shown to recognize lipopeptides and lipopolysaccharides (LPS), the ligand specificity of ChTLRs had not been determined. We found that polyI:C, LPS, R848, S-28463, and ODN2006, which are specifically recognized by TLR3, TLR4, TLR7/8, and TLR9 in mammals, induced substantial amounts of type I interferon (IFN) and interleukin-6 (IL-6) in freshly prepared chicken splenocytes. To determine the ligand specificity of ChTLR3 and ChTLR7, we used a standard reporter assay frequently employed for analysis of mammalian TLRs. Neither S-28463 nor any other TLR ligand induced reporter activity in human 293 cells expressing ChTLR7. However, human 293 cells expressing ChTLR3 strongly and specifically responded to polyI:C, demonstrating that this chicken receptor represents a true ortholog of mammalian TLR3.


Asunto(s)
Pollos/inmunología , Receptor Toll-Like 3/inmunología , Animales , Línea Celular , Pollos/genética , Pollos/metabolismo , Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Humanos , Inductores de Interferón/farmacología , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Interleucina-6/biosíntesis , Interleucina-6/inmunología , Lipopolisacáridos/farmacología , Poli I-C/farmacología , Bazo/citología , Bazo/inmunología , Receptor Toll-Like 3/biosíntesis , Receptor Toll-Like 3/genética , Receptores Toll-Like/biosíntesis , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología
14.
J Interferon Cytokine Res ; 24(3): 179-84, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15035851

RESUMEN

Transcripts of interferon-alpha (IFN-alpha) and IFN-beta genes are present in virus-infected chicken cells, but because of a lack of appropriate assays and reagents, it was unclear if biologically active IFN-beta is secreted. We have established a nonviral bioassay for the sensitive detection of chicken IFN (ChIFN). This assay is based on a quail cell line that carries a luciferase gene that is controlled by the IFN-responsive chicken Mx promoter. Luciferase activity was strongly stimulated when the indicator cells were incubated with ChIFN-alpha, ChIFN-beta, or ChIFN-gamma but not with chicken interleukin-1beta (ChIL-1beta). Unlike the classic antiviral assay that preferentially detects ChIFN-alpha, the Mx-luciferase assay detected ChIFN-alpha and ChIFN-beta with similar sensitivity. With the help of this novel assay and with rabbit antisera specific for either IFN-alpha or IFN-beta, we analyzed the composition of IFN in supernatants of virus-infected chicken embryo cells. Virtually all IFN produced in response to Newcastle disease virus (NDV) was IFN-alpha. However, IFN produced in response to influenza A or vaccinia virus (VV) was a mixture of usually more than 80% IFN-alpha and up to 20% IFN-beta. Thus, IFN-alpha and IFN-beta both contribute to the cytokine activity in supernatants of virus-infected chicken cells. Furthermore, the infecting virus appears to determine the IFN subtype composition.


Asunto(s)
Bioensayo/métodos , Embrión de Pollo/inmunología , Embrión de Pollo/virología , Sueros Inmunes , Interleucina-1/análisis , Interleucina-1/biosíntesis , Animales , Especificidad de Anticuerpos , Línea Celular , Proteínas de Unión al GTP/genética , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Sueros Inmunes/inmunología , Virus de la Influenza A/patogenicidad , Interleucina-1/inmunología , Luciferasas/análisis , Luciferasas/biosíntesis , Luciferasas/genética , Proteínas de Resistencia a Mixovirus , Virus de la Enfermedad de Newcastle/patogenicidad , Infecciones por Orthomyxoviridae/inmunología , Regiones Promotoras Genéticas , Codorniz , Virus Vaccinia/patogenicidad
15.
J Interferon Cytokine Res ; 23(5): 223-8, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12804064

RESUMEN

Chicken interleukin-1beta (ChIL-1beta) is synthesized as a precursor molecule that unlike its mammalian counterpart, lacks a typical caspase-1 cleavage site. Therefore, it was unclear if proteolytic cleavage of ChIL-1beta can occur and if cleavage might modulate the biologic activity of this cytokine. Using an avian indicator cell line that carries an NF-kappaB-regulated luciferase reporter gene, we established a sensitive and highly specific bioassay for ChIL-1beta. Experiments with a rabbit antiserum indicated that the NF-kappaB-stimulating activity in supernatants of lipopolysaccharide (LPS)-treated chicken HD-11 macrophages is largely due to IL-1beta and that proteolytic processing of natural and recombinant ChIL-1beta is not very efficient. Functional analyses further revealed that cDNAs for either full-length or N-terminally truncated chicken ChIL-1beta yielded active cytokine. A truncated molecule that closely resembled putative mature ChIL-1beta exhibited more than 100-fold enhanced biologic activity after expression in mammalian cells, indicating that precursor cleavage is indeed of critical importance for maximal activity.


Asunto(s)
Pollos/inmunología , Interleucina-1/metabolismo , Animales , Sitios de Unión , Bioensayo , Línea Celular , Pollos/genética , ADN Complementario/genética , Genes Reporteros , Humanos , Interleucina-1/química , Interleucina-1/genética , Luciferasas/genética , FN-kappa B/metabolismo , Procesamiento Proteico-Postraduccional , Codorniz , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
16.
J Immunol Methods ; 274(1-2): 229-32, 2003 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-12609548

RESUMEN

Conventional tests for measuring the biological activity of chicken interleukin (IL)-18 require primary chicken spleen cells. We now describe a sensitive bioassay that is based on interleukin-18-induced release of interferon (IFN)-gamma by a permanent chicken cell line. In B19-2D8 cells, cytoplasmically stored interferon-gamma is quickly secreted in response to interleukin-18 exposure.


Asunto(s)
Bioensayo , Pollos/inmunología , Interferón gamma/análisis , Interleucina-18/farmacología , Animales , Línea Celular Transformada , Relación Dosis-Respuesta a Droga , Interferón gamma/genética , Interferón gamma/metabolismo , Cinética , Sensibilidad y Especificidad , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...