Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Cytotherapy ; 20(3): 407-419, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29306566

RESUMEN

BACKGROUND AIMS: Chimeric antigen receptors (CARs) offer great potential toward a functional cure of human immunodeficiency virus (HIV) infection. To achieve the necessary long-term virus suppression, we believe that CARs must be designed for optimal potency and anti-HIV specificity, and also for minimal probability of virus escape and CAR immunogenicity. CARs containing antibody-based motifs are problematic in the latter regard due to epitope mutation and anti-idiotypic immune responses against the variable regions. METHODS: We designed bispecific CARs, each containing a segment of human CD4 linked to the carbohydrate recognition domain of a human C-type lectin. These CARs target two independent regions on HIV-1 gp120 that presumably must be conserved on clinically significant virus variants (i.e., the primary receptor binding site and the dense oligomannose patch). Functionality and specificity of these bispecific CARs were analyzed in assays of CAR-T cell activation and spreading HIV-1 suppression. RESULTS: T cells expressing a CD4-dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DCSIGN) CAR displayed robust stimulation upon encounter with Env-expressing targets, but negligible activity against intercellular adhesion molecule (ICAM)-2 and ICAM-3, the natural dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin ligands. Moreover, the presence of the lectin moiety prevented the CD4 from acting as an entry receptor on CCR5-expressing cells, including CD8+ T cells. However, in HIV suppression assays, the CD4-DCSIGN CAR and the related CD4-liver/lymph node-specific intercellular adhesion molecule-3-grabbing non-integrin CAR displayed only minimally increased potency compared with the CD4 CAR against some HIV-1 isolates and reduced potency against others. By contrast, the CD4-langerin and CD4-mannose binding lectin (MBL) CARs uniformly displayed enhanced potency compared with the CD4 CAR against all the genetically diverse HIV-1 isolates examined. Further experimental data, coupled with known biological features, suggest particular advantages of the CD4-MBL CAR. DISCUSSION: These studies highlight features of bispecific CD4-lectin CARs that achieve potency enhancement by targeting two distinct highly conserved Env determinants while lacking immunogenicity-prone antibody-based motifs.


Asunto(s)
Antígenos CD4/metabolismo , Proteína gp120 de Envoltorio del VIH/metabolismo , Infecciones por VIH/prevención & control , Receptores Quiméricos de Antígenos/metabolismo , Antígenos CD/metabolismo , Sitios de Unión , Linfocitos T CD8-positivos/metabolismo , Moléculas de Adhesión Celular/metabolismo , Técnicas de Cocultivo , Proteína gp120 de Envoltorio del VIH/química , Infecciones por VIH/terapia , VIH-1/fisiología , Humanos , Lectinas Tipo C/metabolismo , Manosa , Polisacáridos/química , Polisacáridos/metabolismo , Ingeniería de Proteínas/métodos , Receptores de Superficie Celular/metabolismo , Receptores Quiméricos de Antígenos/genética , Transducción Genética
2.
Immunity ; 29(1): 79-89, 2008 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-18571443

RESUMEN

DR3 (TRAMP, LARD, WSL-1, TNFRSF25) is a death-domain-containing tumor necrosis factor (TNF)-family receptor primarily expressed on T cells. TL1A, the TNF-family ligand for DR3, can costimulate T cells, but the physiological function of TL1A-DR3 interactions in immune responses is not known. Using DR3-deficient mice, we identified DR3 as the receptor responsible for TL1A-induced T cell costimulation and dendritic cells as the likely source for TL1A during T cell activation. Despite its role in costimulation, DR3 was not required for in vivo T cell priming, for polarization into T helper 1 (Th1), Th2, or Th17 effector cell subtypes, or for effective control of infection with Toxoplasma gondii. Instead, DR3 expression was required on T cells for immunopathology, local T cell accumulation, and cytokine production in Experimental Autoimmune Encephalomyelitis (EAE) and allergic lung inflammation, disease models that depend on distinct effector T cell subsets. DR3 could be an attractive therapeutic target for T cell-mediated autoimmune and allergic diseases.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Inflamación/inmunología , Activación de Linfocitos/inmunología , Miembro 25 de Receptores de Factores de Necrosis Tumoral/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular , Encefalomielitis Autoinmune Experimental/inmunología , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miembro 25 de Receptores de Factores de Necrosis Tumoral/metabolismo , Hipersensibilidad Respiratoria/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/metabolismo , Toxoplasmosis/inmunología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
3.
J Virol ; 89(13): 6685-94, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25878112

RESUMEN

UNLABELLED: Adoptive transfer of CD8 T cells genetically engineered to express "chimeric antigen receptors" (CARs) represents a potential approach toward an HIV infection "functional cure" whereby durable virologic suppression is sustained after discontinuation of antiretroviral therapy. We describe a novel bispecific CAR in which a CD4 segment is linked to a single-chain variable fragment of the 17b human monoclonal antibody recognizing a highly conserved CD4-induced epitope on gp120 involved in coreceptor binding. We compared a standard CD4 CAR with CD4-17b CARs where the polypeptide linker between the CD4 and 17b moieties is sufficiently long (CD4-35-17b CAR) versus too short (CD4-10-17b) to permit simultaneous binding of the two moieties to a single gp120 subunit. When transduced into a peripheral blood mononuclear cell (PBMC) or T cells thereof, all three CD4-based CARs displayed specific functional activities against HIV-1 Env-expressing target cells, including stimulation of gamma interferon (IFN-γ) release, specific target cell killing, and suppression of HIV-1 pseudovirus production. In assays of spreading infection of PBMCs with genetically diverse HIV-1 primary isolates, the CD4-10-17b CAR displayed enhanced potency compared to the CD4 CAR whereas the CD4-35-17b CAR displayed diminished potency. Importantly, both CD4-17b CARs were devoid of a major undesired activity observed with the CD4 CAR, namely, rendering the transduced CD8(+) T cells susceptible to HIV-1 infection. Likely mechanisms for the superior potency of the CD4-10-17b CAR over the CD4-35-17b CAR include the greater potential of the former to engage in the serial antigen binding required for efficient T cell activation and the ability of two CD4-10-17b molecules to simultaneously bind a single gp120 subunit. IMPORTANCE: HIV research has been energized by prospects for a cure for HIV infection or, at least, for a "functional cure" whereby antiretroviral therapy can be discontinued without virus rebound. This report describes a novel CD4-based "chimeric antigen receptor" (CAR) which, when genetically engineered into T cells, gives them the capability to selectively respond to and kill HIV-infected cells. This CAR displays enhanced features compared to previously described CD4-based CARs, namely, increased potency and avoidance of the undesired rendering of the genetically modified CD8 T cells susceptible to HIV infection. When adoptively transferred back to the individual, the genetically modified T cells will hopefully provide durable killing of infected cells and sustained virus suppression without continued antiretroviral therapy, i.e., a functional cure.


Asunto(s)
Fármacos Anti-VIH/metabolismo , VIH-1/inmunología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/virología , Receptores de Antígenos/metabolismo , Receptores del VIH/metabolismo , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Antígenos CD4/genética , Antígenos CD4/metabolismo , Anticuerpos Anti-VIH/genética , Anticuerpos Anti-VIH/metabolismo , Proteína gp120 de Envoltorio del VIH/metabolismo , Humanos , Unión Proteica , Receptores de Antígenos/genética , Receptores del VIH/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/metabolismo , Transducción Genética
4.
Proc Natl Acad Sci U S A ; 107(12): 5652-7, 2010 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-20231466

RESUMEN

Bone marrow stromal cells [BMSCs; also known as mesenchymal stem cells (MSCs)] effectively suppress inflammatory responses in acute graft-versus-host disease in humans and in a number of disease models in mice. Many of the studies concluded that BMSC-driven immunomodulation is mediated by the suppression of proinflammatory Th1 responses while rebalancing the Th1/Th2 ratio toward Th2. In this study, using a ragweed induced mouse asthma model, we studied if BMSCs could be beneficial in an allergic, Th2-dominant environment. When BMSCs were injected i.v. at the time of the antigen challenge, they protected the animals from the majority of asthma-specific pathological changes, including inhibition of eosinophil infiltration and excess mucus production in the lung, decreased levels of Th2 cytokines (IL-4, IL-5, and IL-13) in bronchial lavage, and lowered serum levels of Th2 immunoglobulins (IgG1 and IgE). To explore the mechanism of the effect we used BMSCs isolated from a variety of knockout mice, performed in vivo blocking of cytokines and studied the effect of asthmatic serum and bronchoalveolar lavage from ragweed challenged animals on the BMSCs in vitro. Our results suggest that IL-4 and/or IL-13 activate the STAT6 pathway in the BMSCs resulting in an increase of their TGF-beta production, which seems to mediate the beneficial effect, either alone, or together with regulatory T cells, some of which might be recruited by the BMSCs. These data suggest that, in addition to focusing on graft-versus-host disease and autoimmune diseases, allergic conditions--specifically therapy resistant asthma--might also be a likely target of the recently discovered cellular therapy approach using BMSCs.


Asunto(s)
Asma/inmunología , Células Madre Mesenquimatosas/inmunología , Factor de Crecimiento Transformador beta/inmunología , Ambrosia/efectos adversos , Ambrosia/inmunología , Animales , Asma/etiología , Asma/patología , Asma/terapia , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Citocinas/deficiencia , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Terapia de Inmunosupresión , Técnicas In Vitro , Pulmón/inmunología , Pulmón/patología , Trasplante de Células Madre Mesenquimatosas , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Células Th2/inmunología , Trasplante Homólogo , Trasplante Isogénico
5.
Retrovirology ; 7: 11, 2010 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-20158904

RESUMEN

BACKGROUND: We previously described a potent recombinant HIV-1 neutralizing protein, sCD4-17b, composed of soluble CD4 attached via a flexible polypeptide linker to an SCFv of the 17b human monoclonal antibody directed against the highly conserved CD4-induced bridging sheet of gp120 involved in coreceptor binding. The sCD4 moiety of the bifunctional protein binds to gp120 on free virions, thereby enabling the 17b SCFv moiety to bind and block the gp120/coreceptor interaction required for entry. The previous studies using the MAGI-CCR5 assay system indicated that sCD4-17b (in concentrated cell culture medium, or partially purified) potently neutralized several genetically diverse HIIV-1 primary isolates; however, at the concentrations tested it was ineffective against several other strains despite the conservation of binding sites for both CD4 and 17b. To address this puzzle, we designed variants of sCD4-17b with different linker lengths, and tested the neutralizing activities of the immunoaffinity purified proteins over a broader concentration range against a large number of genetically diverse HIV-1 primary isolates, using the TZM-bl Env pseudotype assay system. We also examined the sCD4-17b sensitivities of isogenic viruses generated from different producer cell types. RESULTS: We observed that immunoaffinity purified sCD4-17b effectively neutralized HIV-1 pseudotypes, including those from HIV-1 isolates previously found to be relatively insensitive in the MAGI-CCR5 assay. The potencies were equivalent for the original construct and a variant with a longer linker, as observed with both pseudotype particles and infectious virions; by contrast, a construct with a linker too short to enable simultaneous binding of the sCD4 and 17b SCFv moieties was much less effective. sCD4-17b displayed potent neutralizing activity against 100% of nearly 4 dozen HIV-1 primary isolates from diverse genetic subtypes (clades A, B, C, D, F, and circulating recombinant forms AE and AG). The neutralization breadth and potency were superior to what have been reported for the broadly neutralizing monoclonal antibodies IgG b12, 2G12, 2F5, and 4E10. The activity of sCD4-17b was found to be similar against isogenic virus particles from infectious molecular clones derived either directly from the transfected producer cell line or after a single passage through PBMCs; this contrasted with the monoclonal antibodies, which were less potent against the PMBC-passaged viruses. CONCLUSIONS: The results highlight the extremely potent and broad neutralizing activity of sCD4-17b against genetically diverse HIV-1 primary isolates. The bifunctional protein has potential applications for antiviral approaches to combat HIV infection.


Asunto(s)
Fármacos Anti-VIH/farmacología , Anticuerpos Monoclonales/farmacología , Productos Biológicos/genética , Productos Biológicos/farmacología , Antígenos CD4/farmacología , Anticuerpos Anti-VIH/farmacología , VIH-1/efectos de los fármacos , Anticuerpos Monoclonales/genética , Antígenos CD4/genética , Anticuerpos Anti-VIH/genética , Humanos , Concentración 50 Inhibidora , Pruebas de Neutralización , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Recombinación Genética
6.
Artículo en Inglés | MEDLINE | ID: mdl-32523897

RESUMEN

Anti-HIV chimeric antigen receptors (CARs) promote direct killing of infected cells, thus offering a therapeutic approach aimed at durable suppression of infection emerging from viral reservoirs. CD4-based CARs represent a favored option, since they target the essential conserved primary receptor binding site on the HIV envelope glycoprotein (Env). We have previously shown that adding a second Env-binding moiety, such as the carbohydrate recognition domain of human mannose-binding lectin (MBL) that recognizes the highly conserved oligomannose patch on gp120, increases CAR potency in an in vitro HIV suppression assay; moreover it reduces the undesired capacity for the CD4 of the CAR molecule to act as an entry receptor, thereby rendering CAR-expressing CD8+ T cells susceptible to infection. Here, we further improve the bispecific CD4-MBL CAR by adding a third targeting moiety against a distinct conserved Env determinant, i.e. a polypeptide sequence derived from the N-terminus of the HIV coreceptor CCR5. The trispecific CD4-MBL-R5Nt CAR displays enhanced in vitro anti-HIV potency compared to the CD4-MBL CAR, as well as undetectable HIV entry receptor activity. The high anti-HIV potency of the CD4-MBL-R5Nt CAR, coupled with its all-human composition and absence of immunogenic variable regions associated with antibody-based CARs, offer promise for the trispecific construct in therapeutic approaches seeking durable drug-free HIV remission.


Asunto(s)
Infecciones por VIH , Inmunoterapia Adoptiva , Receptores Quiméricos de Antígenos , Linfocitos T CD8-positivos/metabolismo , Infecciones por VIH/terapia , VIH-1 , Humanos , Unión Proteica , Receptores CCR5/metabolismo , Receptores Quiméricos de Antígenos/metabolismo
7.
J Leukoc Biol ; 82(6): 1531-41, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17761953

RESUMEN

gp49B, an Ig-like receptor, negatively regulates the activity of mast cells and neutrophils through cytoplasmic immunoreceptor tyrosine-based inhibition motifs. To characterize the role of gp49B further in vivo, gp49B-deficient mice were tested in two allergic models. Responses to ragweed (RW) challenge in the lung and conjunctiva were assessed in models of allergic inflammation and during an infection with parasitic larvae of the nematode Ascaris suum. Infiltration by inflammatory cells into the lung during allergic responses was under negative control of the inhibitory receptor gp49B. Furthermore, an increase in conjunctival inflammation with a predominance of eosinophils, neutrophils, and degranulated mast cells was observed in RW-sensitized, gp49B-deficient mice, which had been challenged in the eye, as compared with C57BL/6 wild-type (WT) controls. Finally, an increase in allergic inflammation in the lungs of A. suum-infected, RW-sensitized mice was observed upon RW challenge, as compared with C57BL/6 WT controls. The observed influx of eosinophils into mucus membranes is characteristic of allergic asthma and allergic conjunctivitis and may contribute to airway hyper-responsiveness, airway remodeling, and mucus production. Expression of gp49B was detected on peripheral eosinophils of control mice and on eosinophils from lungs of mice treated with RW, suggesting a role for gp49B on eosinophils in dampening allergic inflammatory responses.


Asunto(s)
Eosinófilos/inmunología , Hipersensibilidad/inmunología , Inflamación/inmunología , Glicoproteínas de Membrana/inmunología , Receptores Inmunológicos/inmunología , Alérgenos , Ambrosia , Animales , Ascaris suum/inmunología , Ascaris suum/fisiología , Pruebas de Provocación Bronquial , Degranulación de la Célula , Separación Celular , Conjuntivitis Alérgica/inmunología , Citocinas/metabolismo , Eosinofilia/inmunología , Citometría de Flujo , Hipersensibilidad/parasitología , Isotipos de Inmunoglobulinas/sangre , Inflamación/parasitología , Pulmón/inmunología , Pulmón/parasitología , Pulmón/patología , Mastocitos/citología , Mastocitos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Parásitos/inmunología , Parásitos/fisiología
8.
Invest Ophthalmol Vis Sci ; 46(8): 2772-80, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16043850

RESUMEN

PURPOSE: To assess alterations in allergic ocular responses to nonparasite antigens in an experimental system in which mice were skewed toward a Th2 cytokine profile by helminth infection. METHODS: Mice were inoculated with Ascaris suum (A. suum) eggs concurrent with ragweed (RW) sensitization (RW/acute) or by repeated inoculation before RW sensitization (RW/chronic). Control subjects were divided into RW, A. suum, and sham-sensitized groups. Animals were RW-challenged in the eye and examined for changes in ocular responses, inflammatory cell infiltrates, and in vitro assessment of cytokines after antigen restimulation. In subsequent experiments, CD4(+)/CD25+ T regulatory and CD4(+)/CD25- control T cells were adoptively transferred into mice before ocular challenge. RESULTS: RW sensitization and challenge increased ocular symptoms and eosinophil infiltration into the conjunctiva over PBS control eyes. Acute A. suum infection significantly increased RW-induced clinical symptoms and eosinophil infiltrates in the conjunctiva (P = 0.0001) and resulted in the development of anterior uveitis. In contrast, RW/chronic infection provided protection from allergic responses to RW with significantly fewer eosinophils in the eye and reduced eotaxin levels. Transfer of CD4(+)/CD25+ T cells from RW/chronic mice into RW/acute animals also decreased disease intensity, suggesting that T regulatory cells may contribute to protection from allergic eye disease. CONCLUSIONS: The current studies suggest acute parasitic infections exacerbate allergic symptoms, whereas chronic infections offer protection and provide possible explanations for the role of parasitic infection in susceptibility and resistance to nonparasite allergens.


Asunto(s)
Ascariasis/inmunología , Ascaris suum/inmunología , Blefaritis/inmunología , Conjuntivitis Alérgica/inmunología , Hipersensibilidad/inmunología , Uveítis Anterior/inmunología , Enfermedad Aguda , Traslado Adoptivo , Alérgenos/inmunología , Ambrosia/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Quimiocina CCL11 , Quimiocinas CC/sangre , Enfermedad Crónica , Ensayo de Inmunoadsorción Enzimática , Eosinofilia/inmunología , Eosinófilos/inmunología , Inmunoglobulina E/sangre , Interleucina-6/biosíntesis , Mastocitos/inmunología , Ratones , Ratones Endogámicos A , Ratones Endogámicos BALB C , Ratones Noqueados , Células Th2/inmunología
9.
Curr Opin Allergy Clin Immunol ; 3(5): 375-9, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14501438

RESUMEN

PURPOSE OF REVIEW: This review focuses on reports on animal models of ocular allergy published within the past year. A number of animal models are currently being used to clarify the pathophysiology of ocular allergy and to improve the therapeutic interventions for this disease. RECENT FINDINGS: Published literature examined the role of cytokines and other effector molecules which drive the immunopathology of ocular allergies in several animal models. Animal models were also used to compare the safety and efficacy of currently available drugs, and were utilized in initial trials of novel therapeutic agents. Novel therapeutic options being studied include DNA immunizations and recombinant peptides that block enzymes involved in the inflammatory processes. SUMMARY: Several animal models are currently being used in the study of ocular allergy. These include different strains within the mouse, rat, guinea pig, rabbit and dog species. Continuing investigations are needed to elucidate the complex molecular and cellular processes involved in the pathogenesis of ocular allergies. A better understanding of the interplay of effector cells, cytokines, adhesion molecules and a number of other inflammatory mediators will broaden our knowledge of the pathophysiology of ocular allergy and allow improved therapeutic options for this disease.


Asunto(s)
Oftalmopatías/inmunología , Hipersensibilidad/complicaciones , Animales , Perros , Oftalmopatías/tratamiento farmacológico , Oftalmopatías/fisiopatología , Cobayas , Humanos , Hipersensibilidad/tratamiento farmacológico , Hipersensibilidad/fisiopatología , Ratones , Modelos Animales , Conejos , Ratas
10.
Exp Eye Res ; 85(4): 575-9, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17765892

RESUMEN

IL-10 is a regulatory cytokine known to inhibit allergic and inflammatory events. Mast cells (MC) are effector cells which when stimulated release histamine, chemokines and cytokines that initiate the allergic inflammatory response. Recent studies have shown that IL-10 regulates MC function by affecting cytokine production and expression of FcvarepsilonR1 in in vitro assays. Using IL-10 knockout (IL10KO) mice, we examined the effect of its absence on MC susceptibility to degranulation by the basic secretagogue, Compound 48/80 (C48/80). C48/80 is a receptor mimetic that directly activates G proteins and stimulates vigorous MC degranulation. For these studies we stimulated conjunctival MC with C48/80 and found that conjunctival MC of IL10KO mice exhibit increased degranulation compared with wild type mice. Reconstitution of IL10KO mice by adding rIL-10 24h prior to challenge with C48/80 conferred increased resistance of MC to the degranulatory effects of C48/80. The protective effect therefore appears to be due to the presence of IL-10. This is the first in vivo rodent study which reports a novel role of IL-10 in stabilizing mast cells from degranulation by a secretagogue, by as of yet an unknown mechanism.


Asunto(s)
Degranulación de la Célula , Conjuntivitis Alérgica/patología , Interleucina-10/fisiología , Mastocitos/fisiología , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
11.
Infect Immun ; 74(12): 6632-41, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16966410

RESUMEN

We have previously demonstrated that protection from allergic inflammation by Ascaris suum infection was characterized by a global increase in interleukin-10 (IL-10) and the development of protective CD4(+)/CD25(+) T cells (L. Schopf, S. Luccioli, V. Bundoc, P. Justice, C. C. Chan, B. J. Wetzel, H. H. Norris, J. F. Urban, Jr., and A. Keane-Myers, Investig. Ophthalmol. Vis. Sci. 46:2772-2780, 2005). Here, we used A. suum pseudocoelomic fluid (PCF) in lieu of infection to define molecular mechanisms of allergic protection in a mouse model of allergic inflammation. Mice were sensitized with ragweed (RW) and PCF (RW/PCF), PCF alone, or RW alone and then challenged intratracheally, intranasally, and supraocularly with RW. Histological examination of the eyes and lungs, analysis of the bronchoalveolar lavage fluid (BALF), and characterization of ex vivo cytokine responses were performed to determine allergic inflammatory responses. RW/PCF-treated mice had suppressed allergic immune responses compared to mice given RW alone. To investigate whether IL-10 was involved in PCF-mediated allergic protection, similar experiments were performed using mice genetically deficient for IL-10. Persistent protection from allergic disease was observed in the absence of IL-10, indicating the primary mechanism of PCF protection is IL-10 independent. Ex vivo and in vitro analysis of PCF-treated dendritic cells (DC) demonstrated reduced activation receptor expression and cytokine production in response to either RW or lipopolysaccharide stimulation. These findings extend previous studies that showed infection with A. suum alters expression of allergic disease and suggest that PCF can contribute to this effect by interference with DC function.


Asunto(s)
Ascaris suum/inmunología , Asma/prevención & control , Conjuntivitis Alérgica/prevención & control , Células Dendríticas/inmunología , Interleucina-10/fisiología , Hipersensibilidad Respiratoria/prevención & control , Ambrosia/inmunología , Animales , Antígenos de Plantas/inmunología , Asma/patología , Líquido del Lavado Bronquioalveolar/inmunología , Conjuntivitis Alérgica/patología , Citocinas/metabolismo , Inflamación/prevención & control , Interleucina-10/genética , Lipopolisacáridos/inmunología , Ratones , Ratones Mutantes , Eosinofilia Pulmonar/prevención & control , Hipersensibilidad Respiratoria/patología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Células Th2/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA