Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 15(3): e1007595, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30845208

RESUMEN

Despite a broad cell-type tropism, cytomegalovirus (CMV) is an evidentially pulmonary pathogen. Predilection for the lungs is of medical relevance in immunocompromised recipients of hematopoietic cell transplantation, in whom interstitial CMV pneumonia is a frequent and, if left untreated, fatal clinical manifestation of human CMV infection. A conceivable contribution of CMV to airway diseases of other etiology is an issue that so far attracted little medical attention. As the route of primary CMV infection upon host-to-host transmission in early childhood involves airway mucosa, coincidence of CMV airway infection and exposure to airborne environmental antigens is almost unavoidable. For investigating possible consequences of such a coincidence, we established a mouse model of airway co-exposure to CMV and ovalbumin (OVA) representing a protein antigen of an inherently low allergenic potential. Accordingly, intratracheal OVA exposure alone failed to sensitize for allergic airway disease (AAD) upon OVA aerosol challenge. In contrast, airway infection at the time of OVA sensitization predisposed for AAD that was characterized by airway inflammation, IgE secretion, thickening of airway epithelia, and goblet cell hyperplasia. This AAD histopathology was associated with a T helper type 2 (Th2) transcription profile in the lungs, including IL-4, IL-5, IL-9, and IL-25, known inducers of Th2-driven AAD. These symptoms were all prevented by a pre-challenge depletion of CD4+ T cells, but not of CD8+ T cells. As to the underlying mechanism, murine CMV activated migratory CD11b+ as well as CD103+ conventional dendritic cells (cDCs), which have been associated with Th2 cytokine-driven AAD and with antigen cross-presentation, respectively. This resulted in an enhanced OVA uptake and recruitment of the OVA-laden cDCs selectively to the draining tracheal lymph nodes for antigen presentation. We thus propose that CMV, through activation of migratory cDCs in the airway mucosa, can enhance the allergenic potential of otherwise poorly allergenic environmental protein antigens.


Asunto(s)
Alérgenos/metabolismo , Citomegalovirus/metabolismo , Células Dendríticas/inmunología , Alérgenos/efectos adversos , Animales , Presentación de Antígeno/inmunología , Antígenos CD11/inmunología , Citomegalovirus/patogenicidad , Células Dendríticas/microbiología , Modelos Animales de Enfermedad , Femenino , Hipersensibilidad , Inflamación , Pulmón/fisiopatología , Pulmón/virología , Enfermedades Pulmonares/etiología , Enfermedades Pulmonares/virología , Ratones , Ratones Endogámicos C57BL , Ovalbúmina , Células Th2 , Activación Viral/inmunología
2.
Clin Exp Allergy ; 50(1): 41-50, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31573731

RESUMEN

BACKGROUND: Ceylon cinnamon has been shown to possess anti-inflammatory properties in many diseases including allergic inflammation. OBJECTIVE: The aim of this study was to analyse in more detail the effects of cinnamon extract (CE) and its major compounds p-cymene and trans-cinnamaldehyde (CA) on allergen-specific immune responses in vitro and in vivo. METHODS: Therefore, monocyte-derived mature dendritic cells (DC) from grass or birch pollen allergic donors were pulsed with the respective allergen in the presence or absence of CE, p-cymene, CA or the solvent ethanol and co-cultured with autologous CD4+ T cells. Furthermore, basophil activation test was performed with or without CE or ethanol treatment. For the in vivo experiments, BALB/c mice were immunized with ovalbumin (OVA) and orally treated with CE or ethanol. RESULTS: Addition of CE, p-cymene or CA, but not ethanol significantly inhibited DC maturation and subsequent allergen-specific T cell proliferation as well as Th1 and Th2 cytokine production. Sulphidoleukotriene release and CD63 expression by basophils were also significantly diminished after addition of CE. In vivo, treatment of OVA-sensitized mice with CE led to a significant shift from OVA-specific IgE towards IgG2a production and to a strong inhibition of OVA-specific proliferation. Moreover, airway inflammation as well as anaphylaxis after intranasal or systemic allergen challenge was significantly reduced in CE-treated mice. Furthermore, topical application of CE prevented calcipotriol-induced atopic dermatitis-like inflammation in these mice. CONCLUSIONS AND CLINICAL RELEVANCE: Taken together, our data indicate that the anti-inflammatory effect of cinnamon might be exploited for treatment of allergic inflammation, which needs to be further investigated.


Asunto(s)
Acroleína/análogos & derivados , Linfocitos T CD4-Positivos/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cinnamomum zeylanicum , Cimenos/farmacología , Células Dendríticas/efectos de los fármacos , Extractos Vegetales/farmacología , Rinitis Alérgica Estacional/inmunología , Acroleína/farmacología , Animales , Basófilos/efectos de los fármacos , Basófilos/inmunología , Betula , Linfocitos T CD4-Positivos/inmunología , Técnicas de Cocultivo , Citocinas/efectos de los fármacos , Citocinas/inmunología , Células Dendríticas/inmunología , Dermatitis Atópica/inmunología , Modelos Animales de Enfermedad , Humanos , Hipersensibilidad Inmediata/inmunología , Ratones , Ratones Endogámicos BALB C , Ovalbúmina , Pletismografía Total , Poaceae , Polen , Hipersensibilidad Respiratoria/inmunología
3.
Amino Acids ; 52(3): 411-424, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32008091

RESUMEN

We have previously shown that particle-mediated epidermal delivery (PMED) of plasmids encoding ß-galactosidase (ßGal) under control of the fascin-1 promoter (pFascin-ßGal) yielded selective production of the protein in skin dendritic cells (DCs), and suppressed Th2 responses in a mouse model of type I allergy by inducing Th1/Tc1 cells. However, intranasal challenge of mice immunized with pFascin-ßGal induced airway hyperreactivity (AHR) and neutrophilic inflammation in the lung. The tryptophan-catabolizing enzyme indoleamine 2,3-dioxygenase (IDO) has been implicated in immune suppression and tolerance induction. Here we investigated the consequences of co-application of an IDO-encoding vector on the modulatory effect of DNA vaccination by PMED using pFascin-ßGal in models of eosinophilic allergic and non-eosinophilic intrinsic airway inflammation. IDO-encoding plasmids and pFascin-ßGal or pCMV-ßGal were co-applied to abdominal skin of BALB/c mice without, before or after sensitization with ßGal protein. Immune responses in the lung were analysed after intranasal provocation and airway reactivity was determined by whole body plethysmography. Co-application of pCMV-IDO with pFascin-ßGal, but not pCMV-ßGal inhibited the Th1/Tc1 immune response after PMED. Moreover, AHR in those mice was attenuated following intranasal challenge. Therapeutic vaccination of ßGal-sensitized mice with pFascin-ßGal plus pCMV-IDO slightly suppressed airway inflammation and AHR after provocation with ßGal protein, while prophylactic vaccination was not effective. Altogether, our data suggest that only the combination of DC-restricted antigen and ubiquitous IDO expression attenuated asthma responses in mice, most probably by forming a tryptophan-depleted and kynurenine-enriched micromilieu known to affect neutrophils and T cells.


Asunto(s)
Asma/terapia , Biolística/métodos , Hiperreactividad Bronquial/terapia , Células Dendríticas/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Vacunación/métodos , Animales , Antígenos/genética , Antígenos/inmunología , Asma/inmunología , Hiperreactividad Bronquial/inmunología , Proteínas Portadoras/genética , Vectores Genéticos , Ratones , Ratones Endogámicos BALB C , Proteínas de Microfilamentos/genética , Células 3T3 NIH , Infiltración Neutrófila , Linfocitos T Citotóxicos/inmunología , Células TH1/inmunología , Transgenes , beta-Galactosidasa/genética , beta-Galactosidasa/inmunología
4.
J Allergy Clin Immunol ; 143(1): 201-212.e4, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29574077

RESUMEN

BACKGROUND: Amylase-trypsin inhibitors (ATIs) in wheat and related cereals are potent activators of myeloid innate immune cells via engagement of TLR4. Furthermore, ATIs have been shown to serve as adjuvants in experimental intestinal inflammatory diseases. OBJECTIVE: The aim of this study was to analyze whether ATIs are also modifiers of allergic inflammation. METHODS: Therefore, CD4+ T cells from donors sensitized to grass or birch pollen were stimulated with autologous allergen-pulsed dendritic cells in the presence or absence of ATIs or the control storage protein zein from corn. To analyze allergen-induced gut and lung inflammation, immunodeficient mice were engrafted with PBMCs from these allergic donors plus the respective allergen, and fed with selected diets. Three weeks later, inflammation was induced by rectal or intranasal allergen challenge and monitored by mini endoscopy or airway hyperreactivity, respectively. RESULTS: Allergen-specific T-cell proliferation and cytokine production was significantly exacerbated by ATIs and not by zein. In vivo, allergen-specific human IgE level was strongly elevated in sera of mice receiving an ATI-containing diet compared with mice that were fed gluten-free and thus ATI-free diet. Importantly, allergen-induced IgE-dependent colitis and airway hyperreactivity were also enhanced in ATI-fed mice. Gut inflammation was further increased in mice receiving an additional ATI injection and even detectable in the absence of the aeroallergen, whereas zein had no such effect. Injection of anti-human TLR4 mAbs or the anti-human IgE mAb omalizumab completely abolished ATI-induced allergic inflammation. CONCLUSIONS: These results underline that wheat ATIs are important nutritional activators and adjuvants of allergy, which might be exploited for nutritional therapeutic strategies.


Asunto(s)
Asma/inmunología , Linfocitos T CD4-Positivos/inmunología , Inmunidad Innata/efectos de los fármacos , Proteínas de Plantas/farmacología , Triticum/química , Inhibidores de Tripsina/farmacología , Amilasas/antagonistas & inhibidores , Animales , Asma/dietoterapia , Asma/patología , Linfocitos T CD4-Positivos/patología , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Proteínas de Plantas/química , Células THP-1 , Inhibidores de Tripsina/química
5.
Eur J Nutr ; 58(4): 1507-1514, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29600329

RESUMEN

BACKGROUND: Wheat amylase trypsin inhibitors (ATI) are dietary non-gluten proteins that activate the toll-like receptor 4 on myeloid cells, promoting intestinal inflammation. AIM OF THE STUDY: We investigated the effects of dietary ATI on experimental allergic airway inflammation. METHODS: Mice on a gluten and ATI-free diet (GAFD), sensitized with PBS or ovalbumin (OVA) and challenged with OVA, were compared to mice on a commercial standard chow, a gluten diet naturally containing ~ 0.75% of protein as ATI (G+AD), a gluten diet containing ~ 0.19% of protein as ATI (G-AD) and a GAFD with 1% of protein as ATI (AD). Airway hyperreactivity (AHR), inflammation in bronchoalveolar lavage (BAL) and pulmonary tissue sections were analyzed. Allergic sensitization was assessed ex vivo via proliferation of OVA-stimulated splenocytes. RESULTS: Mice on a GAFD sensitized with PBS did not develop AHR after local provocation with methacholine. Mice on a GAFD or on a G-AD and sensitized with OVA developed milder AHR compared to mice fed a G+AD or an AD. The increased AHR was paralleled by increased BAL eosinophils, IL-5 and IL-13 production, and an enhanced ex vivo splenocyte activation in the ATI-fed groups. CONCLUSIONS: Dietary ATI enhance allergic airway inflammation in OVA-challenged mice, while an ATI-free or ATI-reduced diet has a protective effect on AHR. Nutritional wheat ATI, activators of intestinal myeloid cells, may be clinically relevant adjuvants to allergic airway inflammation.


Asunto(s)
Amilasas/antagonistas & inhibidores , Hipersensibilidad Respiratoria/dietoterapia , Hipersensibilidad Respiratoria/inmunología , Triticum/inmunología , Inhibidores de Tripsina/inmunología , Inhibidores de Tripsina/farmacología , Amilasas/inmunología , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Modelos Animales de Enfermedad , Inflamación/dietoterapia , Inflamación/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Triticum/química , Inhibidores de Tripsina/química
6.
J Allergy Clin Immunol ; 142(5): 1558-1570, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29382591

RESUMEN

BACKGROUND: Nanoparticle (NP)-based vaccines are attractive immunotherapy tools because of their capability to codeliver antigen and adjuvant to antigen-presenting cells. Their cellular distribution and serum protein interaction ("protein corona") after systemic administration and their effect on the functional properties of NPs is poorly understood. OBJECTIVES: We analyzed the relevance of the protein corona on cell type-selective uptake of dextran-coated NPs and determined the outcome of vaccination with NPs that codeliver antigen and adjuvant in disease models of allergy. METHODS: The role of protein corona constituents for cellular binding/uptake of dextran-coated ferrous nanoparticles (DEX-NPs) was analyzed both in vitro and in vivo. DEX-NPs conjugated with the model antigen ovalbumin (OVA) and immunostimulatory CpG-rich oligodeoxynucleotides were administered to monitor the induction of cellular and humoral immune responses. Therapeutic effects of this DEX-NP vaccine in mouse models of OVA-induced anaphylaxis and allergic asthma were assessed. RESULTS: DEX-NPs triggered lectin-induced complement activation, yielding deposition of activated complement factor 3 on the DEX-NP surface. In the spleen DEX-NPs targeted predominantly B cells through complement receptors 1 and 2. The DEX-NP vaccine elicited much stronger OVA-specific IgG2a production than coadministered soluble OVA plus CpG oligodeoxynucleotides. B-cell binding of the DEX-NP vaccine was critical for IgG2a production. Treatment of OVA-sensitized mice with the DEX-NP vaccine prevented induction of anaphylactic shock and allergic asthma accompanied by IgE inhibition. CONCLUSIONS: Opsonization of lectin-coated NPs by activated complement components results in selective B-cell targeting. The intrinsic B-cell targeting property of lectin-coated NPs can be exploited for treatment of allergic immune responses.


Asunto(s)
Anafilaxia/inmunología , Linfocitos B/inmunología , Hipersensibilidad/inmunología , Nanopartículas/administración & dosificación , Corona de Proteínas/inmunología , Animales , Antígenos/administración & dosificación , Dextranos/administración & dosificación , Portadores de Fármacos/administración & dosificación , Femenino , Compuestos Ferrosos/administración & dosificación , Lectinas/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Oligodesoxirribonucleótidos/administración & dosificación , Ovalbúmina/administración & dosificación , Linfocitos T/inmunología , Vacunas/administración & dosificación
7.
J Immunol ; 194(3): 887-97, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25527785

RESUMEN

Human studies demonstrated that allergen-specific immunotherapy (IT) represents an effective treatment for allergic diseases. IT involves repeated administration of the sensitizing allergen, indicating a crucial contribution of T cells to its medicinal benefit. However, the underlying mechanisms of IT, especially in a chronic disease, are far from being definitive. In the current study, we sought to elucidate the suppressive mechanisms of IT in a mouse model of chronic allergic asthma. OVA-sensitized mice were challenged with OVA or PBS for 4 wk. After development of chronic airway inflammation, mice received OVA-specific IT or placebo alternately to airway challenge for 3 wk. To analyze the T cell-mediated mechanisms underlying IT in vivo, we elaborated the role of T-bet-expressing Th1 cells, T cell-derived IL-10, and Ag-specific thymic as well as peripherally induced Foxp3(+) regulatory T (Treg) cells. IT ameliorated airway hyperresponsiveness and airway inflammation in a chronic asthma model. Of note, IT even resulted in a regression of structural changes in the airways following chronic inhaled allergen exposure. Concomitantly, IT induced Th1 cells, Foxp3(+), and IL-10-producing Treg cells. Detailed analyses revealed that thymic Treg cells crucially contribute to the effectiveness of IT by promoting IL-10 production in Foxp3-negative T cells. Together with the peripherally induced Ag-specific Foxp3(+) Treg cells, thymic Foxp3(+) Treg cells orchestrate the curative mechanisms of IT. Taken together, we demonstrate that IT is effective in a chronic allergic disease and dependent on IL-10 and thymic as well as peripherally induced Ag-specific Treg cells.


Asunto(s)
Asma/inmunología , Asma/metabolismo , Interleucina-10/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Asma/genética , Asma/patología , Asma/terapia , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Desensibilización Inmunológica , Modelos Animales de Enfermedad , Femenino , Células Caliciformes/patología , Inmunoglobulina E/inmunología , Inmunomodulación , Inmunofenotipificación , Interleucina-10/biosíntesis , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Metaplasia , Ratones , Ratones Noqueados , Ovalbúmina/efectos adversos , Hipersensibilidad Respiratoria/genética , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/metabolismo , Hipersensibilidad Respiratoria/patología , Hipersensibilidad Respiratoria/terapia , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
8.
Proc Natl Acad Sci U S A ; 111(32): 11810-5, 2014 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-25074917

RESUMEN

The prevalence of allergic asthma and other atopic diseases has reached epidemic proportions in large parts of the developed world. The gradual loss of the human indigenous microbiota has been held responsible for this trend. The bacterial pathogen Helicobacter pylori is a constituent of the normal gastric microbiota whose presence has been inversely linked to allergy and asthma in humans and experimental models. Here we show that oral or i.p. tolerization with H. pylori extract prevents the airway hyperresponsiveness, bronchoalveolar eosinophilia, pulmonary inflammation, and Th2 cytokine production that are hallmarks of allergen-induced asthma in mice. Asthma protection is not conferred by extracts from other enteropathogens and requires a heat-sensitive H. pylori component and the DC-intrinsic production of IL-10. The basic leucine zipper ATF-like 3 (BATF3)-dependent CD103(+)CD11b(-) dendritic cell lineage is enriched in the lungs of protected mice and strictly required for protection. Two H. pylori persistence determinants, the γ-glutamyl-transpeptidase GGT and the vacuolating cytotoxin VacA, are required and sufficient for asthma protection and can be administered in purified form to prevent asthma. In conclusion, we provide preclinical evidence for the concept that the immunomodulatory properties of H. pylori can be exploited for tolerization strategies aiming to prevent allergen-induced asthma.


Asunto(s)
Asma/microbiología , Asma/terapia , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/inmunología , Células Dendríticas/inmunología , Helicobacter pylori/inmunología , Helicobacter pylori/patogenicidad , Factores Inmunológicos/uso terapéutico , Interleucina-10/inmunología , Proteínas Represoras/inmunología , Alérgenos/administración & dosificación , Animales , Antígenos Bacterianos/administración & dosificación , Asma/inmunología , Proteínas Bacterianas/inmunología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/deficiencia , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Modelos Animales de Enfermedad , Humanos , Tolerancia Inmunológica , Interleucina-18/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Linfocitos T Reguladores/inmunología , gamma-Glutamiltransferasa/inmunología
9.
Cell Immunol ; 308: 27-34, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27372382

RESUMEN

Cylindromatosis (CYLD) is a ubiquitously expressed deubiquitinating enzyme which removes activating ubiquitin residues from important signaling molecules of the NF-κB pathway. In CYLDex7/8 transgenic mice, a naturally occurring short isoform (sCYLD) is overexpressed in the absence of full length CYLD, leading to excessive NF-κB activity. Herein, we investigated the impact of the CYLDex7/8 mutation selectively in T cells on the development of experimental allergic airway disease induced by sensitization and challenge with ovalbumin. Compared with their wildtype littermates, mice bearing the T cell-specific mutation (CD4+CYLDex7/8) display stronger eosinophilia and mucus production in the lungs and higher IgE serum levels. The reason for these observations is excessive production of T cell-derived IL-9, a cytokine to whom allergy-promoting properties were ascribed. Consequently, blockade of IL-9 in CD4+CYLDex7/8 mice alleviates the development of disease symptoms. Thus, by polarization of the T cell cytokine response, sCYLD can favor the development of allergic airway disease.


Asunto(s)
Asma/genética , Linfocitos T CD4-Positivos/fisiología , Eosinófilos/fisiología , Hipersensibilidad/genética , Síndromes Neoplásicos Hereditarios/genética , Neoplasias Cutáneas/genética , Proteínas Supresoras de Tumor/metabolismo , Animales , Enzima Desubiquitinante CYLD , Humanos , Interleucina-9/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Moco/metabolismo , Mutación/genética , Proteínas Supresoras de Tumor/genética
10.
Proc Natl Acad Sci U S A ; 110(8): 3047-52, 2013 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-23382221

RESUMEN

Infection with the gastric bacterial pathogen Helicobacter pylori is typically contracted in early childhood and often persists for decades. The immunomodulatory properties of H. pylori that allow it to colonize humans persistently are believed to also account for H. pylori's protective effects against allergic and chronic inflammatory diseases. H. pylori infection efficiently reprograms dendritic cells (DCs) toward a tolerogenic phenotype and induces regulatory T cells (Tregs) with highly suppressive activity in models of allergen-induced asthma. We show here that two H. pylori virulence determinants, the γ-glutamyl transpeptidase GGT and the vacuolating cytotoxin VacA, contribute critically and nonredundantly to H. pylori's tolerizing effects on murine DCs in vitro and in vivo. The tolerance-promoting effects of both factors are independent of their described suppressive activity on T cells. Isogenic H. pylori mutants lacking either GGT or VacA are incapable of preventing LPS-induced DC maturation and fail to drive DC tolerization as assessed by induction of Treg properties in cocultured naive T cells. The Δggt and ΔvacA mutants colonize mice at significantly reduced levels, induce stronger T-helper 1 (Th1) and T-helper 17 (Th17) responses, and/or trigger more severe gastric pathology. Both factors promote the efficient induction of Tregs in vivo, and VacA is required to prevent allergen-induced asthma. The defects of the Δggt mutant in vitro and in vivo are phenocopied by pharmacological inhibition of the transpeptidase activity of GGT in all readouts. In conclusion, our results reveal the molecular players and mechanistic basis for H. pylori-induced immunomodulation, promoting persistent infection and conferring protection against allergic asthma.


Asunto(s)
Proteínas Bacterianas/metabolismo , Helicobacter pylori/metabolismo , Tolerancia Inmunológica , Estómago/microbiología , gamma-Glutamiltransferasa/metabolismo , Animales , Técnicas de Cocultivo , Helicobacter pylori/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
11.
J Allergy Clin Immunol ; 130(6): 1384-93, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23102918

RESUMEN

BACKGROUND: IL-10-treated dendritic cells (DCs) have been shown to inhibit T-cell responses through induction of anergy and regulatory T cells in various model systems, including allergic inflammation, but the factors being involved in this inhibition are still unclear. OBJECTIVE: This study set out to analyze such factors produced or induced by IL-10-treated DCs by using gene expression profiling and to explore their function. METHODS: CD4(+) T cells from allergic donors were stimulated with autologous monocyte-derived allergen-pulsed mature DCs or IL-10-treated DCs. After 24 hours, the transcriptional profile was analyzed by using Affymetrix technology. Results were validated by using quantitative real-time PCR, protein expression, and functional in vitro and in vivo studies. RESULTS: In CD4(+) T-cell/IL-10-treated DC cocultures the expression of several known genes, such as IL13, IL5 and OX40, was suppressed. Interestingly, there was only one factor that was strongly upregulated: the DC-derived chemokine CCL18. In vitro addition of CCL18 to cocultures of CD4(+) T cells and allergen-pulsed DCs resulted in a similar inhibition of T(H)2 cytokine production as induced by allergen-pulsed IL-10-treated DCs without exogenous CCL18, whereas T(H)1 cytokine production, IL-10 production, and proliferation were not affected. Furthermore, in a humanized mouse model of allergy using PBMC-engrafted NOD-scid-γc(-/-) mice, CCL18, but not another T(H)2-associated chemokine, CCL17, inhibited airway reactivity and lung inflammation. Chemotaxis assays revealed that CCL18 preferentially attracted regulatory T cells and, less efficiently, T(H)2 cells. CONCLUSION: These data demonstrate that CCL18 might represent a molecule of significant importance in immunoregulation and might be a therapeutic target in patients with allergic airway diseases.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Quimiocinas CC/inmunología , Células Dendríticas/inmunología , Hipersensibilidad Respiratoria/inmunología , Linfocitos T Reguladores/inmunología , Animales , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/trasplante , Modelos Animales de Enfermedad , Humanos , Tolerancia Inmunológica , Interleucina-10/inmunología , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Análisis por Micromatrices , Transcriptoma
12.
J Allergy Clin Immunol ; 128(1): 168-177.e8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21459426

RESUMEN

BACKGROUND: Our laboratory has shown that inhalational sensitization to new antigens is facilitated through an ongoing T(H)2-polarized inflammation of the lung, a phenomenon we call "collateral priming." OBJECTIVE: We were interested to analyze whether a T(H)1-polarized pulmonary inflammation also facilitates priming toward new antigens and which cytokine or cytokines are involved. METHODS: T(H)1-polarized T cells were generated in vitro and transferred into congenic mice. Mice were challenged initially with cognate antigen and an unrelated antigen; consecutively, they received cognate antigen or the secondary antigen. Airway inflammation, antigen-specific IgG2a levels, and airway hyperresponsiveness were assessed to determine the inflammatory phenotype, with antibody blocking studies used to determine cytokine requirements for T(H)1 collateral priming. RESULTS: Our experiments revealed that ongoing inflammation of the lung induced by the transfer of T(H)1-polarized cells also facilitates priming toward new antigens, which results in lymphocytic inflammation of the lung. Interestingly, blocking studies identified IL-17A as a major contributor to this pathology. Accordingly, we could demonstrate for the first time that T(H)17-polarized cells alone can facilitate priming toward new antigens, inducing lymphocytic airway inflammation and strong airway hyperresponsiveness. Flow cytometric analysis revealed priming of endogenous T cells for IL-17A secretion with a distinct memory/effector phenotype compared to T(H)1 cells, thus presenting an exciting model to further elucidate differentiation of T(H)17 cells. CONCLUSIONS: We show that airway inflammation mediated by T(H)17 cells facilitates sensitization to new antigens and confers increased airway responsiveness in a murine model of polysensitization, suggesting a mechanism involving IL-17A behind the increased risk for allergic sensitization in polysensitized subjects.


Asunto(s)
Hiperreactividad Bronquial/inmunología , Activación de Linfocitos/inmunología , Neumonía/inmunología , Células Th17/inmunología , Traslado Adoptivo , Animales , Hiperreactividad Bronquial/metabolismo , Hiperreactividad Bronquial/patología , Separación Celular , Citometría de Flujo , Inhalación , Interleucina-17/inmunología , Interleucina-17/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Neumonía/metabolismo , Neumonía/patología
13.
J Pharmacol Toxicol Methods ; 108: 106956, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33609731

RESUMEN

Göttingen Minipigs show several anatomical, physiological, and pathogenetical similarities to humans and serve an important role in translational studies for example as large animal models of disease. In recent years, the number of transgenic Göttingen Minipigs models has increased, as advanced genetic techniques simplify the generation of animals with precisely tailored modifications. These modifications are designed to replicate genetic alterations responsible for human disease. In addition to serving as valuable large animal disease models, transgenic Göttingen Minipigs are also considered promising donors for xenotransplantation. Current technologies for generation of transgenic minipigs demand a long development and production time of typically 2-3 years. To overcome this limitation and expand the use of Göttingen Minipigs for disease modelling and drug testing, we developed the GENISYST (Genomics Integrated Systems Transgenesis) technology platform for rapid and efficient generation of minipigs based transgenic disease models. As proof of concept, we report the successful generation of transgenic minipigs expressing green fluorescent protein (GFP) in multiple disease-relevant tissues including liver, heart, kidney, lungs, and the central nervous system (CNS). Our data demonstrates the feasibility, efficiency, and utility of GENISYST for rapid one-step generation of transgenic minipigs for human disease modelling in drug discovery and development.


Asunto(s)
Mutación con Ganancia de Función , Genómica , Animales , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Humanos , Porcinos/genética , Porcinos Enanos
14.
Sci Transl Med ; 13(623): eabi7964, 2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34878823

RESUMEN

Endoreplication, duplication of the nuclear genome without cell division, occurs in disease to drive morphologic growth, cell fate, and function. Despite its criticality, the metabolic underpinnings of disease-induced endoreplication and its link to morphologic growth are unknown. Heart disease is characterized by endoreplication preceding cardiac hypertrophy. We identify ATP synthase as a central control node and determinant of cardiac endoreplication and hypertrophy by rechanneling free mitochondrial ADP to methylenetetrahydrofolate dehydrogenase 1 L (MTHFD1L), a mitochondrial localized rate-limiting enzyme of formate and de novo nucleotide biosynthesis. Concomitant activation of the adenosine monophosphate­activated protein kinase (AMPK)­retinoblastoma protein (Rb)-E2F axis co-opts metabolic products of MTHFD1L function to support DNA endoreplication and pathologic growth. Gain- and loss-of-function studies in genetic and surgical mouse heart disease models and correlation in individuals confirm direct coupling of deregulated energetics with endoreplication and pathologic overgrowth. Together, we identify cardiometabolic endoreplication as a hitherto unknown mechanism dictating pathologic growth progression in the failing myocardium.


Asunto(s)
Endorreduplicación , Cardiopatías , Animales , Ciclo Celular , División Celular , Replicación del ADN , Ratones
15.
Int Arch Allergy Immunol ; 152(4): 303-12, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20185922

RESUMEN

BACKGROUND: Using a T helper (Th)1/Th2 disease model, we previously showed that genetically determined Th development depends on dendritic cell-derived interleukin (IL)-1alpha. In Leishmania major infections, Th1 immunity develops if IL-1alpha is present during T cell priming, whereas at later time points, IL-1alpha worsens disease outcome. In the present study, we determined the role of IL-1alpha in other Th2-mediated diseases. METHODS: BALB/c mice were subjected to delayed-type hypersensitivity (DTH) or ovalbumin (OVA)/alum-induced allergic asthma in the presence or absence of IL-1alpha. RESULTS: In DTH, mice treated with IL-1alpha during sensitization with keyhole limpet hemocyanin (KLH)/alum developed decreased footpad swelling associated with elevated KLH-specific interferon-gamma levels. In asthma, significantly decreased airway hypersensitivity responses (AHRs) were detected upon treatment with IL-1alpha during T cell priming. In contrast to control mice, IL-1alpha-treated mice showed reduced peribronchial inflammatory infiltrates. The bronchoalveolar lavage (BAL) fluid contained significantly decreased eosinophil numbers (approximately 50%), but 4 times more neutrophils. The BAL fluid of IL-1alpha-treated BALB/c exhibited reduced amounts of IL-5 and OVA-specific IgE serum levels. In contrast, IL-1alpha treatment at later time points after sensitization or during allergen challenge worsened AHR, had no effect on lung inflammation and BAL fluid cell composition. Furthermore, cytokine levels (IL-5, IL-13) and antigen-specific IgE were increased or unaltered under these conditions. CONCLUSION: Similarly to leishmaniasis, IL-1alpha administration during sensitization of Th2-mediated allergic reactions suppresses the course of disease by shifting the immune response towards Th1, whereas later treatments worsen disease outcome. Future studies will elucidate the therapeutic value of IL-1alpha in asthmatic patients.


Asunto(s)
Asma/inmunología , Hipersensibilidad Tardía/inmunología , Interleucina-1alfa/metabolismo , Compuestos de Alumbre/administración & dosificación , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Recuento de Células , Eosinófilos/patología , Femenino , Hemocianinas/administración & dosificación , Hemocianinas/inmunología , Inmunoglobulina E/metabolismo , Interferón gamma/metabolismo , Interleucina-1alfa/inmunología , Interleucina-5/análisis , Ratones , Ratones Endogámicos BALB C , Neutrófilos/patología , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Células Th2/inmunología
16.
J Immunol ; 181(9): 6148-57, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18941205

RESUMEN

EBV-induced gene 3 (EBI-3) codes for a soluble type I receptor homologous to the p40 subunit of IL-12 that is expressed by APCs following activation. In this study, we assessed the role of EBI-3 in a model of lung melanoma metastasis. Intravenous injection of the B16-F10 cell line resulted in a significant reduction of lung tumor metastasis in EBI-3(-/-) recipient mice compared with wild-type mice. The immunological finding accompanying this effect was the expansion of a newly described cell subset called IFN-gamma producing killer dendritic cells associated with CD8(+) T cell responses in the lung of EBI-3(-/-) mice including IFN-gamma release and TNF-alpha-induced programmed tumor cell death. Depletion of CD8(+) T cells as well as targeting T-bet abrogated the protective effects of EBI-3 deficiency on lung melanoma metastases. Finally, adoptive transfer of EBI-3(-/-) CD8(+) T cells into tumor bearing wild-type mice inhibited lung metastasis in recipient mice. Taken together, these data demonstrate that targeting EBI-3 leads to a T-bet-mediated antitumor CD8(+) T cell responses in the lung.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Melanoma Experimental/inmunología , Melanoma Experimental/secundario , Receptores de Citocinas/deficiencia , Receptores de Citocinas/genética , Animales , Línea Celular Transformada , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/patología , Citotoxicidad Inmunológica/genética , Técnicas de Inactivación de Genes , Vigilancia Inmunológica/genética , Inyecciones Intravenosas , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/terapia , Melanoma Experimental/metabolismo , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Antígenos de Histocompatibilidad Menor , Trasplante de Neoplasias , Receptores de Citocinas/fisiología , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/fisiología
17.
J Immunol ; 181(11): 7751-8, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19017964

RESUMEN

In allergic airway disease, Treg may play an important role in the modulation of airway hyperreactivity (AHR) and inflammation. We therefore investigated the therapeutic potential of Treg in an Ag-dependent murine asthma model. We here describe that AHR can be completely suppressed by adoptive transfer of Treg overexpressing active TGF-beta1. Using mice with impaired TGF-beta signaling in T cells, we could demonstrate that TGF-beta signaling in recipient effector T cells or transferred Treg themselves is not required for the protective effects on AHR. However, the expression of IL-10 by Treg was found to be essential for the suppression of AHR, since Treg overexpressing active TGF-beta1 but deficient in IL-10 lacked protective effects. Airway inflammation could not be significantly suppressed by wild-type or transgenic Treg. In conclusion, modulation of cytokine expression by Treg may have therapeutic potential for the treatment of AHR in asthma. The mechanisms of the effects of Treg on airway inflammation require further clarification.


Asunto(s)
Traslado Adoptivo , Interleucina-10/inmunología , Hipersensibilidad Respiratoria/terapia , Linfocitos T Reguladores/trasplante , Factor de Crecimiento Transformador beta1/inmunología , Animales , Modelos Animales de Enfermedad , Expresión Génica , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/terapia , Interleucina-10/biosíntesis , Interleucina-10/genética , Ratones , Ratones Transgénicos , Hipersensibilidad Respiratoria/genética , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/genética
18.
J Immunol ; 181(3): 1917-26, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18641329

RESUMEN

IL-2 influences both survival and differentiation of CD4(+) T effector and regulatory T cells. We studied the effect of i.n. administration of Abs against the alpha- and the beta-chains of the IL-2R in a murine model of allergic asthma. Blockade of the beta- but not the alpha-chain of the IL-2R after allergen challenge led to a significant reduction of airway hyperresponsiveness. Although both treatments led to reduction of lung inflammation, IL-2 signaling, STAT-5 phosphorylation, and Th2-type cytokine production (IL-4 and IL-5) by lung T cells, IL-13 production and CD4(+) T cell survival were solely inhibited by the blockade of the IL-2R beta-chain. Moreover, local blockade of the common IL-2R/IL-15R beta-chain reduced NK cell number and IL-2 production by lung CD4(+)CD25(+) and CD4(+)CD25(-) T cells while inducing IL-10- and TGF-beta-producing CD4(+) T cells in the lung. This cytokine milieu was associated with reduced CD4(+) T cell proliferation in the draining lymph nodes. Thus, local blockade of the beta-chain of the IL-2R restored an immunosuppressive cytokine milieu in the lung that ameliorated both inflammation and airway hyperresponsiveness in experimental allergic asthma. These findings provide novel insights into the functional role of IL-2 signaling in experimental asthma and suggest that blockade of the IL-2R beta-chain might be useful for therapy of allergic asthma in humans.


Asunto(s)
Asma/inmunología , Linfocitos T CD4-Positivos/inmunología , Hipersensibilidad/inmunología , Tolerancia Inmunológica/inmunología , Subunidad beta del Receptor de Interleucina-2/inmunología , Ganglios Linfáticos/inmunología , Transducción de Señal/inmunología , Alérgenos/inmunología , Animales , Anticuerpos/inmunología , Apoptosis , Asma/genética , Asma/metabolismo , Asma/patología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular , Citocinas/biosíntesis , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Hipersensibilidad/genética , Hipersensibilidad/metabolismo , Hipersensibilidad/patología , Subunidad alfa del Receptor de Interleucina-2/genética , Subunidad alfa del Receptor de Interleucina-2/inmunología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Subunidad beta del Receptor de Interleucina-2/genética , Subunidad beta del Receptor de Interleucina-2/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ganglios Linfáticos/citología , Ratones , Ratones Endogámicos BALB C
19.
Immunobiology ; 225(1): 151867, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31761474

RESUMEN

Neutrophil extracellular traps (NETs) are formed by polymorphonuclear neutrophils (PMN) and contribute to the innate host defense by binding and killing bacterial and fungal pathogens. Because NET formation depends on histone hypercitrullination by peptidylarginine deiminase 4 (PAD4), we used PAD4 gene deficient (Pad4-/-) mice in a mouse model of invasive pulmonary aspergillosis (IPA) to address the contribution of NETs to the innate host defense in vivo. After the induction (24 h) of IPA by i.t. infection with Aspergillus fumigatus conidia, Pad4-/- mice revealed lower fungal burden in the lungs, accompanied by less acute lung injury, TNFα and citH3 compared to wildtype controls. These findings suggest that release of NETs contributes to tissue damage and limits control of fungal outgrowth. Thus inhibition of NETosis might be a useful strategy to maintain neutrophil function and avoid lung damage in patients suffering from IPA, especially in those suffering from preexisting pulmonary disease.


Asunto(s)
Aspergillus fumigatus/fisiología , Trampas Extracelulares/metabolismo , Aspergilosis Pulmonar Invasiva/metabolismo , Pulmón/metabolismo , Neutrófilos/inmunología , Animales , Apoptosis , Citrulinación/genética , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata , Aspergilosis Pulmonar Invasiva/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Arginina Deiminasa Proteína-Tipo 4/genética
20.
J Allergy Clin Immunol ; 121(4): 992-9.e6, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18329088

RESUMEN

BACKGROUND: The transcriptional regulation of cytokines released and controlled by memory T cells is not well understood. Defective IFN-gamma production in allergic asthma correlates in human beings with the risk of wheezing in childhood. OBJECTIVE: To understand the role of the transcription factor nuclear factor of activated T cells 2 (NFATc2) in memory and effector T cells in the airways in experimental allergic asthma. METHODS: We used murine models of allergic asthma and adoptive cell transfer of fluorescence-activated sorted cells in a disease model. RESULTS: Mice lacking NFATc2 developed an increase in airwayhyperresponsiveness (AHR), remodeling, and serum IgE levelson ovalbumin sensitization. This phenotype was associated withCD81CD1222 T cells deficient in IFN-g production in theairways. The origin of this phenotype in NFATc2(2/2) mice wasrelated to an expanded population of lung CD81CD1221(IL-2Rb chain) CD127hi (IL-7 receptor [R] a chain1) long-livedmemory cells. Adoptive transfer of ovalbumin-specific CD81NFATc2(2/2) T cells enhanced the AHR generated byNFATc2(2/2) CD41 T cells in immunodeficient mice, increasedIL-17, and reduced IFN-g production in the reconstituted mice. Depletion of the memory CD81CD1221IL-7Rhigh T-cellpopulation corrected the defect in IFN-g production by lungNFATc2(2/2) CD81CD1222 cells and abrogated the increasedAHR observed in NFATc2(2/2) CD81 T-cell-reconstituted micewith a severe combined immunodeficiency disorder. CONCLUSION: Taken together, our results suggest that NFATc2 expression in long-lived memory CD8+ T cells controls IL-2 and IFN-gamma production in lung CD8+ T cells, which then limits TH17 and TH2 development in the airways during allergen challenge.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Hipersensibilidad/inmunología , Hipersensibilidad/prevención & control , Memoria Inmunológica , Factores de Transcripción NFATC/fisiología , Subgrupos de Linfocitos T/inmunología , Traslado Adoptivo , Animales , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/metabolismo , Hiperreactividad Bronquial/prevención & control , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/trasplante , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Femenino , Inhibidores de Crecimiento/deficiencia , Inhibidores de Crecimiento/genética , Inhibidores de Crecimiento/fisiología , Hipersensibilidad/metabolismo , Interferón gamma/antagonistas & inhibidores , Interferón gamma/deficiencia , Interferón gamma/genética , Interleucina-17/biosíntesis , Subunidad beta del Receptor de Interleucina-2/biosíntesis , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones SCID , Factores de Transcripción NFATC/deficiencia , Factores de Transcripción NFATC/genética , Receptores de Interleucina-7/biosíntesis , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/trasplante , Regulación hacia Arriba/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA