Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 89(5): 2731-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25540366

RESUMEN

UNLABELLED: Previously we found that following intranasal (i.n.) infection with neurotropic vesicular stomatitis virus (VSV) type I interferon receptor (IFNAR) triggering of neuroectodermal cells was critically required to constrain intracerebral virus spread. To address whether locally active IFN-ß was induced proximally, we studied spatiotemporal conditions of VSV-mediated IFN-ß induction. To this end, we performed infection studies with IFN-ß reporter mice. One day after intravenous (i.v.) VSV infection, luciferase induction was detected in lymph nodes. Upon i.n. infection, luciferase induction was discovered at similar sites with delayed kinetics, whereas on days 3 and 4 postinfection enhanced luciferase expression additionally was detected in the foreheads of reporter mice. A detailed analysis of cell type-specific IFN-ß reporter mice revealed that within the olfactory bulb IFN-ß was expressed by neuroectodermal cells, primarily by astrocytes and to a lesser extent by neurons. Importantly, locally induced type I IFN triggered distal parts of the brain as indicated by the analysis of ISRE-eGFP mice which after i.n. VSV infection showed enhanced green fluorescent protein (eGFP) expression throughout the brain. Compared to wild-type mice, IFN-ß(-/-) mice showed increased mortality to i.n. VSV infection, whereas upon i.v. infection no such differences were detected highlighting the biological significance of intracerebrally expressed IFN-ß. In conclusion, upon i.n. VSV instillation, IFN-ß responses mounted by astrocytes within the olfactory bulb critically contribute to the antiviral defense by stimulating distal IFN-ß-negative brain areas and thus arresting virus spread. IMPORTANCE: The central nervous system has long been considered an immune privileged site. More recently, it became evident that specialized immune mechanisms are active within the brain to control pathogens. Previously, we showed that virus, which entered the brain via the olfactory route, was arrested within the olfactory bulb by a type I IFN-dependent mechanism. Since peripheral type I IFN would not readily cross the blood-brain barrier and within the brain thus far no abundant type I IFN responses have been detected, here we addressed from where locally active IFN originated from. We found that upon intranasal VSV instillation, primarily astrocytes, and to a lesser extent neurons, were stimulated within the olfactory bulb to mount IFN-ß responses that also activated and protected distal brain areas. Our results are surprising because in other infection models astrocytes have not yet been identified as major type I IFN producers.


Asunto(s)
Astrocitos/inmunología , Encefalitis Viral/inmunología , Interferón beta/metabolismo , Bulbo Olfatorio/inmunología , Infecciones por Rhabdoviridae/inmunología , Vesiculovirus/inmunología , Animales , Astrocitos/virología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Interferón beta/deficiencia , Luciferasas/análisis , Luciferasas/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/inmunología , Neuronas/virología , Bulbo Olfatorio/virología , Análisis de Supervivencia
2.
Proc Natl Acad Sci U S A ; 109(26): 10486-91, 2012 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-22689946

RESUMEN

The synthetic double-stranded RNA poly(I:C) is commonly used as an adjuvant to boost CD8 T-cell function; however, polyinosinic:polycytidylic acid [poly(I:C)] can also suppress autoimmune disease. The mechanism by which a single adjuvant achieves two distinct immunoregulatory roles is unknown. Although it is clear that coadministration of poly(I:C) with antigen elicits strong adjuvant effects in mice, we found that poly(I:C) injection before antigen substantially reduced antigen-dependent CD8 T-cell responses. Notably, CD8 T cells sensitized in poly(I:C)-pretreated mice failed to fully up-regulate IL-33R (ST2), which led to impaired T-cell receptor-independent responses to IL-33. In contrast, nonsensitized effector CD8 T cells responded robustly to IL-33 using a two-signal cytokine mechanism. During an acute lung response to Staphylococcus aureus enterotoxin, peripheral injection of poly(I:C) manifested a suppressive process by inhibiting the differentiation of both antigen- and IL-33-responsive CD8 effectors systemically. These findings highlight that early exposure to double-stranded RNA reverses its role as an adjuvant and, importantly, prevents IL-33R up-regulation on CD8 effector T cells to dampen inflammation.


Asunto(s)
Linfocitos T CD8-positivos/citología , Diferenciación Celular/fisiología , Interleucinas/fisiología , Receptor Toll-Like 3/metabolismo , Animales , Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Interleucina-33 , Ligandos , Activación de Linfocitos , Ratones , ARN Bicatenario/administración & dosificación
3.
Eur J Immunol ; 40(10): 2769-77, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20821729

RESUMEN

Virus-induced expansion of CD8(+) T cells may be promoted by type I IFN receptor (IFNAR)-triggering of T cells, depending on the pathogen tested. We studied modified vaccinia virus Ankara (MVA), a promising vaccine vector candidate, which was derived from conventional vaccinia virus (VACV) by more than 570 consecutive in vitro passages. In adoptive transfer experiments, we verified that VACV expressing the gp33 epitope of lymphocytic choriomeningitis virus (VACV(gp33)) induced largely IFNAR-independent expansion of gp33-specific T cells. On the contrary, MVA(gp33)-induced T-cell expansion was IFNAR dependent. Interestingly, under the latter conditions, T-cell activation was IFNAR independent, whereas T-cell apoptosis was enhanced in the absence of IFNAR. To address whether MVA-induced T-cell expansion was solely affected by IFNAR-triggering of T cells, expansion of endogenous T cells was studied in conditional mice with a T-cell- or DC-specific IFNAR deletion. Interestingly, both mouse strains showed moderately reduced T-cell expansion, whereas mice with a combined T-cell- and DC-specific IFNAR ablation showed massively reduced T-cell expansion similar to that of IFNAR(-/-) mice. These results are compatible with the model that IFN-inducing viruses such as MVA confer virus-specific CD8(+) T-cell expansion by concomitant IFNAR-triggering of DC and of T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Vectores Genéticos/inmunología , Interferón Tipo I/inmunología , Virus Vaccinia/inmunología , Traslado Adoptivo , Animales , Antígeno CD11c/inmunología , Epítopos de Linfocito T , Citometría de Flujo , Activación de Linfocitos/inmunología , Ratones , Transducción de Señal/inmunología
4.
J Virol ; 84(13): 6549-63, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20410285

RESUMEN

Type I interferons (IFNs) are considered to be important mediators of innate immunity due to their inherent antiviral activity, ability to drive the transcription of a number of genes involved in viral clearance, and their role in the initiation of innate and adaptive immune responses. Due to the central role of type I IFNs, we sought to determine their importance in the generation of immunity to a recombinant vaccine vector fowlpox virus (FPV). In analyzing the role of type I IFNs in immunity to FPV, we show that they are critical to the secretion of a number of innate and proinflammatory cytokines, including type I IFNs themselves as well as interleukin-12 (IL-12), tumor necrosis factor-alpha (TNF-alpha), IL-6, and IL-1beta, and that deficiency leads to enhanced virus-mediated antigen expression. Interestingly, however, type I IFNs were not required for adaptive immune responses to recombinant FPV even though plasmacytoid dendritic cells (pDCs), the primary producers of type I IFNs, have been shown to be requisite for this to occur. Furthermore, we provide evidence that the importance of pDCs may lie in their ability to capture and present virally derived antigen to T cells rather than in their capacity as professional type I IFN-producing cells.


Asunto(s)
Inmunidad Adaptativa , Citocinas/inmunología , Células Dendríticas/inmunología , Virus de la Viruela de las Aves de Corral/inmunología , Interferón Tipo I/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos Virales/inmunología , Ratones , Ratones Endogámicos C57BL , Linfocitos T/inmunología , Vacunas Sintéticas/inmunología , Vacunas Virales/inmunología
5.
J Immunol ; 182(9): 5217-24, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19380767

RESUMEN

Topical application of tumors with the TLR7 agonist imiquimod is an effective adjunct treatment for a range of primary dermatological cancers. However, for therapy to be effective against a broad range of solid tumor types, it must promote a strong systemic antitumor response that targets metastases in addition to primary tumor. We therefore investigated the potential of locally delivered imiquimod to stimulate an effective systemic antitumor response in a murine model of malignant mesothelioma (AB1-HA) with primary and distal tumors (dual tumor). Persistent delivery of imiquimod into primary tumor significantly retarded tumor growth in all treated mice compared with vehicle control. This local antitumor immune response required both CD8 T cells and NK cells, but not CD4 T cells, and was reliant on type I IFN induction. In vivo CTL studies and Ly6A/E staining of lymphocytes suggested that local imiquimod treatment had indeed induced a systemic, Ag-specific CD8 response. However, notably this response was not sufficient to retard the growth of an untreated distal tumor. Because local imiquimod treatment did not induce significant CD4 T cell responses, we investigated the efficacy of combining imiquimod with agonistic CD40 Ab (as a surrogate for CD4 T cell help). Combination of locally delivered imiquimod with systemic anti-CD40 immunotherapy not only significantly enhanced the local antitumor response, with 30% complete resolution, but it was also effective at significantly retarding growth of distal tumor. These results demonstrate that antitumor responses induced by locally delivered TLR7 agonists can be harnessed systemically for treating distal tumor.


Asunto(s)
Aminoquinolinas/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Antígenos CD40/inmunología , Glicoproteínas de Membrana/agonistas , Mesotelioma/inmunología , Mesotelioma/terapia , Receptor Toll-Like 7/agonistas , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/uso terapéutico , Aminoquinolinas/uso terapéutico , Animales , Anticuerpos Monoclonales/uso terapéutico , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Citotoxicidad Inmunológica , Quimioterapia Combinada , Femenino , Imiquimod , Interferón Tipo I/administración & dosificación , Interferón gamma/administración & dosificación , Células Asesinas Naturales/inmunología , Ligandos , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/uso terapéutico , Mesotelioma/prevención & control , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Desnudos , Ratones Transgénicos , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 7/uso terapéutico
6.
J Immunol ; 181(11): 7670-80, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19017955

RESUMEN

Poly(I:C) is an adjuvant used for antitumor treatment and vaccines because of its prominent effects on CD8 T cells and NK cells. Poly(I:C) binds TLR3 and this interaction is thought to be central for driving cell-mediated immune responses. We investigated the importance of TLR3 in poly(I:C)-mediated endogenous CD8 T cell responses using the pathogenic T cell stimulant Staphylococcus aureus enterotoxin A. While the responsive CD8 T cells expanded comparably in both wild-type and TLR3(-/-) mice, differentiation of effector CD8 T cells was enhanced by poly(I:C) in the TLR3(-/-) mice. A higher percentage of Ag-specific CD8 T cells became IFN-gamma and TNF-alpha producers in the absence of TLR3 signaling. Consistent with this boosted response was the observation that TLR3-deficient cells synthesized less IL-10 compared with TLR3-sufficient cells in response to poly(I:C). Ultimately, however, the fundamental mechanism of CD8 effector T cell differentiation through the TLR3-independent pathway was shown to be completely IFN-alpha/beta-dependent. Administration of IFN-alpha/beta-neutralizing Abs abolished the poly(I:C) effects in TLR3(-/-) mice. These findings reveal specific roles of how dsRNA receptors shape CD8 T cell responses, which should be considered as poly(I:C) is authenticated as a therapeutic adjuvant used in vaccines.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/efectos de los fármacos , Interferón-alfa/inmunología , Interferón beta/inmunología , Poli I-C/farmacología , Receptor Toll-Like 3/inmunología , Animales , Anticuerpos/farmacología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacología , Diferenciación Celular/inmunología , Enterotoxinas/inmunología , Enterotoxinas/farmacología , Interferón-alfa/antagonistas & inhibidores , Interferón-alfa/genética , Interferón beta/antagonistas & inhibidores , Interferón beta/genética , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Células Asesinas Naturales/inmunología , Ratones , Ratones Noqueados , Poli I-C/inmunología , ARN Bicatenario/inmunología , ARN Bicatenario/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Staphylococcus aureus/inmunología , Receptor Toll-Like 3/agonistas , Receptor Toll-Like 3/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
7.
J Infect Dis ; 200(10): 1548-55, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19821721

RESUMEN

Type II interferon (IFN), IFN-gamma, is important in innate immunity to the intestinal protozoan parasite Cryptosporidium species, which infects epithelial cells (enterocytes). This investigation is, to our knowledge, the first to characterize the role of type I IFN in innate immunity to this parasite. Pretreatment of human or murine enterocyte cell lines with IFN-alpha/beta inhibited parasite development, and we identified that a key mechanism of cytokine action was to prevent parasite invasion of enterocytes. IFN-alpha/beta was rapidly expressed by infected murine enterocytes and also by bone marrow-derived dendritic cells that were exposed to live parasites. Treatment of neonatal severe combined immunodeficiency mice with anti-IFN-alpha/beta neutralizing antibodies before infection increased oocyst reproduction, as measured at the peak of infection, and parasite numbers in gut epithelium were also increased 2 days after infection. The latter observation correlated with strong intestinal expression of both IFN-alpha and IFN-beta messenger RNA within 24 h after infection. Treatment with anti-IFN-alpha/beta, however, did not reduce early expression of IFN-gamma. These findings identify a novel early innate host response against Cryptosporidium parvum involving IFN-alpha/beta.


Asunto(s)
Criptosporidiosis/inmunología , Cryptosporidium parvum/inmunología , Enterocitos/inmunología , Inmunidad Innata , Interferón-alfa/inmunología , Interferón beta/inmunología , Animales , Células CACO-2 , Enterocitos/parasitología , Humanos , Interferón-alfa/genética , Interferón beta/genética , Ratones , ARN Mensajero
8.
Front Immunol ; 11: 515556, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33178180

RESUMEN

Highly sensitive reporter-gene assays have been developed that allow both the direct vascular endothelial growth factor (VEGF) neutralizing activity of bevacizumab and the ability of bevacizumab to activate antibody dependent cellular cytotoxicity (ADCC) to be quantified rapidly and in a highly specific manner. The use of these assays has shown that in 46 patients with ovarian cancer following four cycle of bevacizumab treatment, and in longitudinal samples from the two patients that respond to bevacizumab therapy from a small cohort of patients with glioblastoma, that there is a reasonably good correlation between bevacizumab drug levels determined by ELISA and bevacizumab activity, determined using either the VEGF-responsive reporter gene, or the ADCC assays. One of the two primary non-responders with glioblastoma exhibited high levels of ADCC activity suggesting reduced bevacizumab Fc engagement in vivo in contrast to the other primary non-responder, and the two secondary non-responders with a decreasing bevacizumab PK profile, determined by ELISA that exhibited low to undetectable ADCC activity. Drug levels were consistently higher than bevacizumab activity determined using the reporter gene assay in serial samples from one of the secondary non-responders and lower in some samples from the other secondary non-responder and ADCC activity was markedly lower in all samples from these patients suggesting that bevacizumab activity may be partially neutralized by anti-drug neutralizing antibodies (NAbs). These results suggest that ADCC activity may be correlated with the ability of some patients to respond to treatment with bevacizumab while the use of the VEGF-responsive reporter-gene assay may allow the appearance of anti-bevacizumab NAbs to be used as a surrogate maker of treatment failure prior to the clinical signs of disease progression.


Asunto(s)
Bevacizumab/administración & dosificación , Glioblastoma , Proteínas de Neoplasias/inmunología , Neoplasias Ováricas , Factor A de Crecimiento Endotelial Vascular/inmunología , Línea Celular Tumoral , Femenino , Glioblastoma/tratamiento farmacológico , Glioblastoma/inmunología , Glioblastoma/patología , Células HEK293 , Humanos , Estudios Longitudinales , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología
9.
Cell Signal ; 19(10): 2080-7, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17604604

RESUMEN

Interferons (IFNs) are pleiotropic cytokines involved in the regulation of physiological and pathological processes. Upon interaction with their specific receptors, IFNs activate the Jak/STAT signalling pathway. Numerous studies suggest, however, that the classical Jak/STAT pathway cannot alone account for the wide range of IFN's biological effects. To better understand the role of alternative signalling pathways in the type I IFNs response, we analyzed novel tyrosine-phosphorylated proteins following IFN-alpha2 stimulation. We showed for the first time that the Grb2-associated binder 2 (Gab2) protein is differentially phosphorylated upon the IFN subtype employed and the cells stimulated. We demonstrated that IFNAR1 physically interacts with Gab2. Moreover, the cellular content of Gab2 varies as a function of IFN receptor chain expression levels, and in particular of the ratio of IFNAR1 to IFNAR2, suggesting that Gab2 and IFNAR2 compete for interaction with IFNAR1. Analysis of Gab2 deletion mutants indicates that IFNAR1 might interact with a Gab2 region containing p85-PI3'kinase binding sites. Our results shed new light on recent data involving both Gab2 and type I IFNs in osteoclastogenesis and oncogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Interferón Tipo I/farmacología , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Sitios de Unión , Línea Celular , Humanos , Ratones , Fosforilación/efectos de los fármacos , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal
10.
Eur Cytokine Netw ; 18(2): 108-14, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17594944

RESUMEN

Through the activation of Toll-like receptors (TLRs) or cytosolic RNA helicases, a large number of pathogenic or synthetic components can induce the transcription of genes coding for type I interferons (IFNs). This family of related cytokines includes notably, a single IFN-beta protein and 13 different IFN-alpha subtypes, whose biological activities are probably not the same. The aim of this study was to characterize the type I IFN subtypes produced in vitro by human peripheral blood mononuclear cells (PBMCs) in response to specific inducers. Thus, PBMCs obtained from a single donor, were exposed to various agents including Sendai virus, Herpes simplex virus-1 (HSV-1), poliovirus-IgG complexes and serum from a patient with systemic lupus erythematosus (SLE). Six hours later, mRNA was extracted and amplified by RT-PCR using primers which recognize IFN-B mRNA and the different IFN-A mRNA subtypes. IFN-A subtypes were identified by cloning and sequencing the amplification product. Antiviral activity was assayed in supernatant at 18 hours. Human PBMCs were found to express constitutively type I IFNs mRNA. Antiviral activity and expression of IFN-A and IFN-B mRNA increased with each inducing agent. Although almost all the IFN-A subtypes were detected, their relative abundance appeared to be dependent upon the inducing agent. Incubation of PBMCs with a neutralizing monoclonal antibody directed against the type I IFN receptor (IFNAR) did not affect the level of antiviral activity in the supernatant of induced PBMCs. Our results suggest that the level of IFN-alpha expressed by PBMCs cells is independent of IFNAR feedback signalling and that the nature of the inducing agent modifies the pattern of IFN-A subtypes preferentially expressed by these cells.


Asunto(s)
Interferón Tipo I/metabolismo , Leucocitos Mononucleares/metabolismo , Lupus Eritematoso Sistémico/sangre , Antivirales/metabolismo , Células Cultivadas , Clonación Molecular , Citosol/metabolismo , Herpesvirus Humano 1/metabolismo , Humanos , Inmunoglobulina G/metabolismo , Interferón-alfa/metabolismo , Poliovirus/metabolismo , ARN Helicasas/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Virus Sendai/metabolismo
11.
J Immunol Res ; 2017: 3908289, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29104875

RESUMEN

Novel ADCC effector cells expressing the V-variant or F-variant of FcγRIIIa (CD16a) and firefly luciferase under the control of a chimeric promoter incorporating recognition sequences for the principal transcription factors involved in FcγRIIIa signal transduction, together with novel target cells overexpressing a constant high level of the specific antigen recognized by rituximab, trastuzumab, cetuximab, infliximab, adalimumab, or etanercept, confer improved sensitivity, specificity, and dynamic range in an ADCC assay relative to effector cells expressing a NFAT-regulated reporter gene and wild-type target cells. The effector cells also contain a normalization gene rendering ADCC assays independent of cell number or serum matrix effects. The novel effector and target cells in a frozen thaw-and-use format exhibit low vial-to-vial and lot-to-lot variation in their performance characteristics reflected by CVs of 10% or less. Homologous control target cells in which the specific target gene has been invalidated by genome editing providing an ideal control and a means of correcting for nonspecific effects were observed with certain samples of human serum. The novel effector cells and target cells expressing noncleavable membrane-bound TNFα have been used to quantify ADCC activity in serum from patients with Crohn's disease treated with infliximab and to relate ADCC activity to drug levels.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos CD20/genética , Enfermedad de Crohn/inmunología , Receptores ErbB/genética , Técnicas Inmunológicas/métodos , Factores de Transcripción NFATC/genética , Receptor ErbB-2/genética , Receptores de IgG/genética , Linfocitos T/fisiología , Factor de Necrosis Tumoral alfa/genética , Antígenos CD20/inmunología , Cetuximab/metabolismo , Receptores ErbB/inmunología , Etanercept/metabolismo , Genes Reporteros/genética , Células HEK293 , Humanos , Infliximab/metabolismo , Células Jurkat , Receptor ErbB-2/inmunología , Receptores de IgG/inmunología , Rituximab/metabolismo , Transducción de Señal , Transgenes/genética , Trastuzumab/metabolismo , Factor de Necrosis Tumoral alfa/inmunología
12.
Oncogene ; 24(4): 605-15, 2005 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-15580300

RESUMEN

Type I interferon (IFN) enhances the transcription of the tumor suppressor gene p53. To elucidate the molecular mechanism mediating IFN-induced apoptosis, we analysed programmed cell death in response to type I (IFNalpha) or type II (IFNgamma) treatment in relation to p53 status. In two cell lines (MCF-7, SKNSH), IFNalpha, but not IFNgamma, enhanced apoptosis in a p53-dependent manner. Furthermore, only IFNalpha upregulated p53 as well as p53 target genes (Noxa, Mdm2 and CD95). The apoptotic response to IFNalpha decreased in the presence of ZB4, an anti-CD95 antibody, suggesting that CD95 is involved in this process. When p53 was inactivated by the E6 viral protein or the expression of a p53 mutant, IFNalpha-induced apoptosis and p53 target genes upregulation were abrogated. Altogether these results demonstrate that p53 plays a pivotal role in the IFNalpha-induced apoptotic response. IFNalpha-induced PML was unable to recruit p53 into nuclear bodies and its downregulation by siRNA did not alter CD95 expression. In contrast, IFNgamma-induced apoptosis is p53-independent. CD95 and IFN-regulatory factor 1 (IRF1) are directly upregulated by this cytokine. Apoptotic response to IFNgamma is decreased in the presence of ZB4 and strongly diminished by IRF1 siRNA, implicating both CD95 and IRF1 in IFNgamma-induced apoptotic response. Taken together, these results show that in two different cell lines, IFNalpha and IFNgamma, induce p53-dependent -independent apoptosis, respectively.


Asunto(s)
Apoptosis/efectos de los fármacos , Interferón-alfa/farmacología , Interferón gamma/farmacología , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Factor 1 Regulador del Interferón , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteína de la Leucemia Promielocítica , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-mdm2 , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor , Regulación hacia Arriba/efectos de los fármacos , Receptor fas/genética , Receptor fas/metabolismo
13.
Neurosci Lett ; 405(3): 181-5, 2006 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-16884849

RESUMEN

The Hsp104 chaperone induces thermo-tolerance in yeast and rescues proteins trapped in aggregates. In this study, we showed that xenogenic expression of Hsp104 dramatically increased the viability of the neuronal mouse CAD cell line after exposure to heat shock. These results indicate that the Hsp104 protein confers thermo-resistance to mammalian neuronal cells, the canonical property of Hsp104 in yeast. Hsp104 also determines the prion state of prion-like proteins in yeast and to investigate whether Hsp104 expression may modify mammalian prion infection in vivo, transgenic mice with specific expression of Hsp104 in neurons were generated. Mice develop and reproduce normally, they show no detectable physical defect and may constitute valuable model for the study of aggregation-prone neuropathological disorders. Hsp104 transgenic and control littermates were infected intracerebrally with the ME7 strain of scrapie. No differences in the incubation time of the disease or in PrP(Sc) accumulation were observed between transgenic and control mice. These results suggest that the heat-shock protein Hsp104 is not efficient to modulate the multiplication of mammalian prions and/or to counteract neurodegeneration in the brain of scrapie-infected mice.


Asunto(s)
Proteínas Fúngicas/metabolismo , Proteínas de Choque Térmico/metabolismo , Neuronas/metabolismo , Priones/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Animales , Western Blotting/métodos , Encéfalo/citología , Encéfalo/metabolismo , Encéfalo/patología , Supervivencia Celular/fisiología , Técnica del Anticuerpo Fluorescente/métodos , Proteínas Fúngicas/genética , Expresión Génica , Células HeLa , Proteínas de Choque Térmico/genética , Humanos , Infecciones , Ratones , Ratones Transgénicos , Neuronas/patología , Proteínas PrPSc/genética , Proteínas PrPSc/metabolismo , Proteínas PrPSc/patogenicidad , Priones/genética , Priones/patogenicidad , Proteínas de Saccharomyces cerevisiae/genética , Scrapie/metabolismo , Temperatura
15.
J Interferon Cytokine Res ; 24(12): 717-28, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15684739

RESUMEN

Lymphocytes from aged autoimmune MRL/lpr mice overexpress Fas ligand (FasL), and are cytotoxic against Fas+ target cells. This cytotoxic potential is only partly due to FasL, as wild-type MRL+/+ lymphocytes are not able to kill Fas+ targets after induction of FasL. In addition, serum levels of interferon-alpha (IFN-alpha) increase in parallel with the Fas-dependent cytotoxic potential of lymphocytes from MRL/lpr mice as they age. To understand the mechanisms underlying these observations, combined suppression subtractive hybridization (SSH) and RT-PCR were used to study differential gene expression in splenocytes from MRL/lpr mice compared with splenocytes from MRL+/+ mice. Twenty-two genes were upregulated transcriptionally in MRL/lpr splenocytes compared with their MRL+/+ counterparts. Furthermore, 9 of these genes were also upregulated after treatment of MRL/lpr splenocytes with IFN-alpha, and 4 were strongly downregulated. MRL/lpr lymphocytes were also found to be hyperresponsive to IFN-alpha. Thus, MRL/lpr lymphocytes overexpressed mRNA for the IFN-alpha receptor (IFNAR-1 and IFNAR-2) chains of the IFN-alpha/beta receptor and exhibited high endogenous levels of both Stat1 and phosphorylated Stat1 proteins. Lymphocytes from young MRL/lpr mice, with low Fas-dependent cytotoxic activity, were found to become highly cytotoxic against Fas+ targets after treatment with IFN-alpha. These data suggest that IFN-alpha plays an important role in the physiopathology of the systemic lupus erythematosus (SLE)-like syndrome that occurs in MRL/lpr mice.


Asunto(s)
Citotoxicidad Inmunológica , Regulación de la Expresión Génica , Interferón-alfa/fisiología , Lupus Eritematoso Sistémico/inmunología , Linfocitos/inmunología , Glicoproteínas de Membrana/genética , Animales , Células Cultivadas , Citotoxicidad Inmunológica/genética , ADN Complementario/análisis , Proteínas de Unión al ADN/metabolismo , Proteína Ligando Fas , Interferón-alfa/sangre , Interferón-alfa/farmacología , Lupus Eritematoso Sistémico/genética , Linfocitos/efectos de los fármacos , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos MRL lpr , Hibridación de Ácido Nucleico , Fosforilación , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Receptor de Interferón alfa y beta , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1 , Bazo/citología , Bazo/metabolismo , Transactivadores/metabolismo , Transcripción Genética/efectos de los fármacos , Activación Transcripcional
16.
Bioanalysis ; 4(17): 2179-90, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23013400

RESUMEN

Biopharmaceuticals are used extensively for the treatment of a number of chronic debilitating and fatal diseases such as cancer and inflammatory or autoimmune diseases. Although biopharmaceuticals are in general well tolerated, the development of anti-drug antibodies can impair their safety and efficacy. Assessment of immunogenicity is essential for a more effective and rational use of biopharmaceuticals, and is dependent upon the establishment of efficient standardized assays that allow direct comparison of immunogenicity data with clinical outcome. Although regulatory authorities recommend the use of cell-based assays that reflect the mechanism of action of the drug for the detection of neutralizing anti-drug antibodies, conventional cell-based assays are difficult to standardize and often give variable results. A number of strategies have been adopted to improve the performance of cell-based assays, including quantification of drug-induced proteins using either real-time RT-PCR or branched DNA to detect mRNA, or ELISAs to detect protein, bridging assays using immobilized cells and the use of reporter gene assays. The relative merits and limitations of each of these methods is reviewed herein.


Asunto(s)
Anticuerpos Neutralizantes/análisis , Biofarmacia/normas , Anticuerpos Neutralizantes/inmunología , Biofarmacia/tendencias , Línea Celular Tumoral , Humanos
17.
Ther Adv Drug Saf ; 2(3): 113-28, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25083207

RESUMEN

Biopharmaceuticals are used widely for the treatment of cancer, chronic viral hepatitis, inflammatory, and autoimmune diseases. Biopharmaceuticals such as interferons are well tolerated for the most part with the most common adverse events observed being 'flu-like' symptoms that resolve rapidly after initial treatment. Prolonged treatment is associated, however, with more serious adverse events including leucopenia, thrombocytopenia, and neuropsychiatric effects, which may necessitate dose reduction or even cessation of treatment in some patients. Recombinant growth factors, such as erythropoietin (EPO), granulocyte colony-stimulating factor, or granulocyte macrophage colony-stimulating factor, are for the most part well tolerated, although severe complications have been reported in patients with cancer or chronic kidney disease treated with EPO. Similarly, treatment of patients with cancer with high doses of interleukin-2 is associated with significant toxicity. Treatment of chronic inflammatory diseases, such as rheumatoid arthritis, psoriasis, and Crohn's disease, with antitumor necrosis factor-alpha monoclonal antibodies is associated with an increased risk of granulomatous infections and, in particular, tuberculosis. The monoclonal antibody, natalizumab, that targets alpha4 integrins is effective in the treatment of multiple sclerosis but is associated with the activation of JC virus and development of progressive multifocal leukoencephalopathy. Repeated administration of recombinant proteins can cause a break in immune tolerance in some patients resulting in the production of a polyclonal antibody response that can adversely affect pharmacokinetics and clinical response. In addition, neutralizing antibodies that cross react with nonredundant essential proteins such as EPO can cause severe autoimmune reactions.

18.
Expert Rev Clin Pharmacol ; 4(5): 623-31, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22114889

RESUMEN

The safety and efficacy of biopharmaceuticals can be severely impaired by their immunogenicity. A risk-based strategy should be used to assess immunogenicity on a case-by-case basis using standardized methods to correlate anti-drug antibody levels with clinical outcome. In silico and in vitro techniques allow putative T-cell epitopes to be identified and eliminated in candidate molecules while maintaining structure and function. Putative T-cell epitopes can be studied in the context of the HLA allotypes representative of the target population in vitro and in transgenic mice that express human HLA genes. Mice immune tolerant to human proteins allow the study of the effect of factors such as aggregation on the loss of immune tolerance. However, significant challenges remain in order to be able predict the immunogenicity of a therapeutic protein in a particular individual.


Asunto(s)
Productos Biológicos/inmunología , Productos Biológicos/farmacología , Fenómenos Inmunogenéticos/efectos de los fármacos , Fenómenos Inmunogenéticos/inmunología , Animales , Epítopos de Linfocito T/efectos adversos , Epítopos de Linfocito T/inmunología , Humanos , Tolerancia Inmunológica
19.
J Immunol Methods ; 373(1-2): 229-39, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21910993

RESUMEN

A cell-based assay has been developed for the quantification of the activity of TNFα antagonists based on human erythroleukemic K562 cells transfected with a NFκB regulated firefly luciferase reporter-gene construct. Both drug activity and anti-drug neutralizing antibodies can be quantified with a high degree of precision within 2h, and without interference from cytokines and other factors known to activate NFκB. The assay cells also contain the Renilla luciferase reporter gene under the control of a constitutive promoter that allows TNFα-induced firefly luciferase activity to be normalized relative to Renilla luciferase expression. Thus, results are independent of cell number or differences in cell viability, resulting in intra and inter assay coefficients of variation of 10% or less. Normalization of results relative to the expression of an internal standard also provides a means for correcting for serum matrix effects and allows residual drug levels or anti-drug neutralizing antibodies to be quantified even in serum samples with a relatively high degree of cytotoxicity.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/análisis , Luciferasas/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Adalimumab , Animales , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados/sangre , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Artritis Reumatoide/sangre , Artritis Reumatoide/tratamiento farmacológico , Enfermedad de Crohn/sangre , Enfermedad de Crohn/tratamiento farmacológico , Medios de Cultivo/química , Medios de Cultivo/farmacología , Relación Dosis-Respuesta a Droga , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Infliximab , Células K562 , Luciferasas/genética , Luciferasas de Luciérnaga/genética , Luciferasas de Luciérnaga/metabolismo , Luciferasas de Renilla/genética , Luciferasas de Renilla/metabolismo , FN-kappa B/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Suero/química , Suero/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/farmacología
20.
Methods Mol Biol ; 626: 287-309, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20099135

RESUMEN

The activity of several potent adjuvants, including incomplete Freund's adjuvant, CpG oligodeoxynucleotides, and alum, has been shown to be due at least in part to the induction of cytokines, including type I interferons (IFNs), IFN-gamma, interleukin-2 (IL-2), and IL-12, that play key roles in the regulation of innate and adaptive immunity. The relatively short half-life of recombinant homologues of cytokines has limited their use as vaccine adjuvants. These difficulties have been overcome by encapsulation into liposomes and the use of cytokine expression vectors co-administered with DNA vaccines. Although a number of cytokines including IFN-alpha, IFN-gamma, IL-2, IL-12, IL-15, IL-18, IL-21, GM-CSF, and Flt-3 ligand have been shown to potentiate the immune response to vaccination in various experimental models, the full potential of cytokines as vaccine adjuvants remains to be established.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Citocinas/farmacología , Vacunación/métodos , Inmunidad Adaptativa/efectos de los fármacos , Animales , Humanos , Inmunidad Innata/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA