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










Base de dados
Intervalo de ano de publicação
1.
Sci Transl Med ; 13(623): eabi7964, 2021 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-34878823

RESUMO

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.


Assuntos
Endorreduplicação , Cardiopatias , Animais , Ciclo Celular , Divisão Celular , Replicação do DNA , Camundongos
2.
J Pharmacol Toxicol Methods ; 108: 106956, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33609731

RESUMO

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.


Assuntos
Mutação com Ganho de Função , Genômica , Animais , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Humanos , Suínos/genética , Porco Miniatura
3.
Amino Acids ; 52(3): 411-424, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32008091

RESUMO

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.


Assuntos
Asma/terapia , Biolística/métodos , Hiper-Reatividade Brônquica/terapia , Células Dendríticas/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Vacinação/métodos , Animais , Antígenos/genética , Antígenos/imunologia , Asma/imunologia , Hiper-Reatividade Brônquica/imunologia , Proteínas de Transporte/genética , Vetores Genéticos , Camundongos , Camundongos Endogâmicos BALB C , Proteínas dos Microfilamentos/genética , Células NIH 3T3 , Infiltração de Neutrófilos , Linfócitos T Citotóxicos/imunologia , Células Th1/imunologia , Transgenes , beta-Galactosidase/genética , beta-Galactosidase/imunologia
4.
Clin Exp Allergy ; 50(1): 41-50, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31573731

RESUMO

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.


Assuntos
Acroleína/análogos & derivados , Linfócitos T CD4-Positivos/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cinnamomum zeylanicum , Cimenos/farmacologia , Células Dendríticas/efeitos dos fármacos , Extratos Vegetais/farmacologia , Rinite Alérgica Sazonal/imunologia , Acroleína/farmacologia , Animais , Basófilos/efeitos dos fármacos , Basófilos/imunologia , Betula , Linfócitos T CD4-Positivos/imunologia , Técnicas de Cocultura , Citocinas/efeitos dos fármacos , Citocinas/imunologia , Células Dendríticas/imunologia , Dermatite Atópica/imunologia , Modelos Animais de Doenças , Humanos , Hipersensibilidade Imediata/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina , Pletismografia Total , Poaceae , Pólen , Hipersensibilidade Respiratória/imunologia
5.
Immunobiology ; 225(1): 151867, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31761474

RESUMO

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.


Assuntos
Aspergillus fumigatus/fisiologia , Armadilhas Extracelulares/metabolismo , Aspergilose Pulmonar Invasiva/metabolismo , Pulmão/metabolismo , Neutrófilos/imunologia , Animais , Apoptose , Citrulinação/genética , Modelos Animais de Doenças , Humanos , Imunidade Inata , Aspergilose Pulmonar Invasiva/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína-Arginina Desiminase do Tipo 4/genética
6.
PLoS Pathog ; 15(3): e1007595, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30845208

RESUMO

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.


Assuntos
Alérgenos/metabolismo , Citomegalovirus/metabolismo , Células Dendríticas/imunologia , Alérgenos/efeitos adversos , Animais , Apresentação de Antígeno/imunologia , Antígenos CD11/imunologia , Citomegalovirus/patogenicidade , Células Dendríticas/microbiologia , Modelos Animais de Doenças , Feminino , Hipersensibilidade , Inflamação , Pulmão/fisiopatologia , Pulmão/virologia , Pneumopatias/etiologia , Pneumopatias/virologia , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina , Células Th2 , Ativação Viral/imunologia
7.
J Allergy Clin Immunol ; 143(1): 201-212.e4, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29574077

RESUMO

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.


Assuntos
Asma/imunologia , Linfócitos T CD4-Positivos/imunologia , Imunidade Inata/efeitos dos fármacos , Proteínas de Plantas/farmacologia , Triticum/química , Inibidores da Tripsina/farmacologia , Amilases/antagonistas & inibidores , Animais , Asma/dietoterapia , Asma/patologia , Linfócitos T CD4-Positivos/patologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Proteínas de Plantas/química , Células THP-1 , Inibidores da Tripsina/química
8.
Eur J Nutr ; 58(4): 1507-1514, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29600329

RESUMO

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.


Assuntos
Amilases/antagonistas & inibidores , Hipersensibilidade Respiratória/dietoterapia , Hipersensibilidade Respiratória/imunologia , Triticum/imunologia , Inibidores da Tripsina/imunologia , Inibidores da Tripsina/farmacologia , Amilases/imunologia , Animais , Líquido da Lavagem Broncoalveolar/imunologia , Modelos Animais de Doenças , Inflamação/dietoterapia , Inflamação/imunologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Triticum/química , Inibidores da Tripsina/química
9.
J Allergy Clin Immunol ; 142(5): 1558-1570, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29382591

RESUMO

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.


Assuntos
Anafilaxia/imunologia , Linfócitos B/imunologia , Hipersensibilidade/imunologia , Nanopartículas/administração & dosagem , Coroa de Proteína/imunologia , Animais , Antígenos/administração & dosagem , Dextranos/administração & dosagem , Portadores de Fármacos/administração & dosagem , Feminino , Compostos Ferrosos/administração & dosagem , Lectinas/imunologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oligodesoxirribonucleotídeos/administração & dosagem , Ovalbumina/administração & dosagem , Linfócitos T/imunologia , Vacinas/administração & dosagem
10.
Cell Immunol ; 308: 27-34, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27372382

RESUMO

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.


Assuntos
Asma/genética , Linfócitos T CD4-Positivos/fisiologia , Eosinófilos/fisiologia , Hipersensibilidade/genética , Síndromes Neoplásicas Hereditárias/genética , Neoplasias Cutâneas/genética , Proteínas Supressoras de Tumor/metabolismo , Animais , Enzima Desubiquitinante CYLD , Humanos , Interleucina-9/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Muco/metabolismo , Mutação/genética , Proteínas Supressoras de Tumor/genética
11.
J Immunol ; 194(3): 887-97, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25527785

RESUMO

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.


Assuntos
Asma/imunologia , Asma/metabolismo , Interleucina-10/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Remodelação das Vias Aéreas , Animais , Asma/genética , Asma/patologia , Asma/terapia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Dessensibilização Imunológica , Modelos Animais de Doenças , Feminino , Células Caliciformes/patologia , Imunoglobulina E/imunologia , Imunomodulação , Imunofenotipagem , Interleucina-10/biossíntese , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Metaplasia , Camundongos , Camundongos Knockout , Ovalbumina/efeitos adversos , Hipersensibilidade Respiratória/genética , Hipersensibilidade Respiratória/imunologia , Hipersensibilidade Respiratória/metabolismo , Hipersensibilidade Respiratória/patologia , Hipersensibilidade Respiratória/terapia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
12.
Immunobiology ; 220(4): 490-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25468560

RESUMO

The mechanisms of tolerance induction occurring in the course of allergen-specific immunotherapy have not been elucidated in full detail. Our study aimed to characterize high zone tolerance in mouse models of type I allergy and of allergic airway inflammation induced by subcutaneous sensitization of mice with high doses of the model allergen ovalbumin (OVA) without the use of adjuvant. Mice were immunized by subcutaneous injection of high doses (HD) of OVA or, for comparison, low doses (LD) of OVA in saline. HD-mice showed lower specific IgE, but augmented IgG in sera than LD-mice. Pre-treatment of mice with HD-OVA antigen-specifically inhibited IgE production subsequently induced by LD-OVA. OVA-restimulated splenocytes from HD-mice revealed hypoproliferation and impaired production of Th2-associated cytokines. HD-mice exhibited lower airway reactivity, goblet cell hyperplasia and mucus production, as well as IL-5 and IL-13 production in the lungs than LD-mice following local provocation. Recruitment of inflammatory cells into the airways was comparable, while the number of eosinophils in the bronchoalveolar lavage was substantially higher in HD-mice. Adoptive transfer of dnTC from HD-mice into naïve mice, which were subsequently sensitized with LD-OVA, suppressed IgE production in the recipients. The number of dnTC was higher in the spleens of HD-mice than LD-mice. In conclusion, our study demonstrates that subcutaneous sensitization of mice with high doses of allergen in the absence of adjuvant results in attenuated airway reactivity as compared with LD-sensitization and induces CD4(-)CD8(-) dnTC with regulatory function on IgE production.


Assuntos
Formação de Anticorpos/imunologia , Imunoglobulina E/biossíntese , Imunoglobulina E/imunologia , Imunomodulação , Hipersensibilidade Respiratória/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Especificidade de Anticorpos/imunologia , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/metabolismo , Feminino , Expressão Gênica , Imunização , Imunofenotipagem , Ativação Linfocitária , Camundongos , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/genética , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Hipersensibilidade Respiratória/metabolismo , Hipersensibilidade Respiratória/patologia , Hipersensibilidade Respiratória/fisiopatologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/metabolismo
13.
Proc Natl Acad Sci U S A ; 111(32): 11810-5, 2014 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-25074917

RESUMO

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.


Assuntos
Asma/microbiologia , Asma/terapia , Fatores de Transcrição de Zíper de Leucina Básica/imunologia , Células Dendríticas/imunologia , Helicobacter pylori/imunologia , Helicobacter pylori/patogenicidade , Fatores Imunológicos/uso terapêutico , Interleucina-10/imunologia , Proteínas Repressoras/imunologia , Alérgenos/administração & dosagem , Animais , Antígenos de Bactérias/administração & dosagem , Asma/imunologia , Proteínas de Bactérias/imunologia , Fatores de Transcrição de Zíper de Leucina Básica/deficiência , Fatores de Transcrição de Zíper de Leucina Básica/genética , Modelos Animais de Doenças , Humanos , Tolerância Imunológica , Interleucina-18/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Linfócitos T Reguladores/imunologia , gama-Glutamiltransferase/imunologia
14.
Proc Natl Acad Sci U S A ; 110(8): 3047-52, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23382221

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Helicobacter pylori/metabolismo , Tolerância Imunológica , Estômago/microbiologia , gama-Glutamiltransferase/metabolismo , Animais , Técnicas de Cocultura , Helicobacter pylori/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
15.
Methods Mol Biol ; 940: 357-70, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23104354

RESUMO

Gene gun-mediated delivery of allergen-encoding plasmid DNA has been in focus for many years now as being a needle-free alternative to the protein-based desensitization regimen used in specific immunotherapy. Biolistic immunization with the Helios gene gun has proven to be potent in the induction of antigen-specific CD4(+) and CD8(+) T cells. Here we describe biolistic vaccination in experimental mouse models of IgE-mediated type I allergy as well as allergen-induced airway inflammation.


Assuntos
Biolística/instrumentação , Hipersensibilidade/prevenção & controle , Vacinação/instrumentação , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia , Alérgenos/imunologia , Animais , Especificidade de Anticorpos , Lavagem Broncoalveolar , Feminino , Hipersensibilidade/genética , Hipersensibilidade/imunologia , Hipersensibilidade/fisiopatologia , Imunoglobulina E/sangue , Imunoglobulina E/imunologia , Pulmão/imunologia , Pulmão/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Doenças Respiratórias/genética , Doenças Respiratórias/imunologia , Doenças Respiratórias/fisiopatologia , Doenças Respiratórias/prevenção & controle , Transfecção/instrumentação , Vacinas de DNA/genética , beta-Galactosidase/imunologia
16.
J Allergy Clin Immunol ; 130(6): 1384-93, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23102918

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Quimiocinas CC/imunologia , Células Dendríticas/imunologia , Hipersensibilidade Respiratória/imunologia , Linfócitos T Reguladores/imunologia , Animais , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/transplante , Modelos Animais de Doenças , Humanos , Tolerância Imunológica , Interleucina-10/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Análise em Microsséries , Transcriptoma
17.
J Clin Invest ; 122(3): 1082-96, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22307326

RESUMO

Persistent colonization with the gastric bacterial pathogen Helicobacter pylori causes gastritis and predisposes infected individuals to gastric cancer. Conversely, it is also linked to protection from allergic, chronic inflammatory, and autoimmune diseases. We demonstrate here that H. pylori inhibits LPS-induced maturation of DCs and reprograms DCs toward a tolerance-promoting phenotype. Our results showed that DCs exposed to H. pylori in vitro or in vivo failed to induce T cell effector functions. Instead, they efficiently induced expression of the forkhead transcription factor FoxP3, the master regulator of Tregs, in naive T cells. Depletion of DCs in mice infected with H. pylori during the neonatal period was sufficient to break H. pylori-specific tolerance. DC depletion resulted in improved control of the infection but also aggravated T cell-driven immunopathology. Consistent with the mouse data, DCs infiltrating the gastric mucosa of human H. pylori carriers exhibited a semimature DC-SIGN(+)HLA-DR(hi)CD80(lo)CD86(lo) phenotype. Mechanistically, the tolerogenic activity of H. pylori-experienced DCs was shown to require IL-18 in vitro and in vivo; DC-derived IL-18 acted directly on T cells to drive their conversion to Tregs. CD4(+)CD25(+) Tregs from infected wild-type mice but not Il18(-/-) or Il18r1(-/-) mice prevented airway inflammation and hyperresponsiveness in an experimental model of asthma. Taken together, our results indicate that tolerogenic reprogramming of DCs ensures the persistence of H. pylori and protects against allergic asthma in a process that requires IL-18.


Assuntos
Asma/prevenção & controle , Helicobacter pylori/metabolismo , Interleucina-18/metabolismo , Linfócitos T Reguladores/citologia , Adulto , Animais , Linfócitos T CD4-Positivos/microbiologia , Diferenciação Celular , Técnicas de Cocultura , Feminino , Mucosa Gástrica/microbiologia , Infecções por Helicobacter/microbiologia , Humanos , Tolerância Imunológica , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Ligantes , Lipopolissacarídeos/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Fenótipo
18.
J Allergy Clin Immunol ; 128(1): 168-177.e8, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21459426

RESUMO

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.


Assuntos
Hiper-Reatividade Brônquica/imunologia , Ativação Linfocitária/imunologia , Pneumonia/imunologia , Células Th17/imunologia , Transferência Adotiva , Animais , Hiper-Reatividade Brônquica/metabolismo , Hiper-Reatividade Brônquica/patologia , Separação Celular , Citometria de Fluxo , Inalação , Interleucina-17/imunologia , Interleucina-17/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Pneumonia/metabolismo , Pneumonia/patologia
19.
Int Arch Allergy Immunol ; 152(4): 303-12, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20185922

RESUMO

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.


Assuntos
Asma/imunologia , Hipersensibilidade Tardia/imunologia , Interleucina-1alfa/metabolismo , Compostos de Alúmen/administração & dosagem , Animais , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Contagem de Células , Eosinófilos/patologia , Feminino , Hemocianinas/administração & dosagem , Hemocianinas/imunologia , Imunoglobulina E/metabolismo , Interferon gama/metabolismo , Interleucina-1alfa/imunologia , Interleucina-5/análise , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/patologia , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Células Th2/imunologia
20.
Cancer Res ; 69(7): 3069-76, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19318584

RESUMO

The Ca(2+)-regulated calcineurin/nuclear factor of activated T cells (NFAT) cascade controls alternative pathways of T-cell activation and peripheral tolerance. Here, we describe reduction of NFATc2 mRNA expression in the lungs of patients with bronchial adenocarcinoma. In a murine model of bronchoalveolar adenocarcinoma, mice lacking NFATc2 developed more and larger solid tumors than wild-type littermates. The extent of central tumor necrosis was decreased in the tumors in NFATc2((-/-)) mice, and this finding was associated with reduced tumor necrosis factor-alpha and interleukin-2 (IL-2) production by CD8(+) T cells. Adoptive transfer of CD8(+) T cells of NFATc2((-/-)) mice induced transforming growth factor-beta(1) in the airways of recipient mice, thus supporting CD4(+)CD25(+)Foxp-3(+)glucocorticoid-induced tumor necrosis factor receptor (GITR)(+) regulatory T (T(reg)) cell survival. Finally, engagement of GITR in NFATc2((-/-)) mice induced IFN-gamma levels in the airways, reversed the suppression by T(reg) cells, and costimulated effector CD4(+)CD25(+) (IL-2Ralpha) and memory CD4(+)CD127(+) (IL-7Ralpha) T cells, resulting in abrogation of carcinoma progression. Agonistic signaling through GITR, in the absence of NFATc2, thus emerges as a novel possible strategy for the treatment of human bronchial adenocarcinoma in the absence of NFATc2 by enhancing IL-2Ralpha(+) effector and IL-7Ralpha(+) memory-expressing T cells.


Assuntos
Adenocarcinoma/genética , Adenocarcinoma/imunologia , Neoplasias Brônquicas/genética , Neoplasias Brônquicas/imunologia , Linfócitos T CD8-Positivos/imunologia , Fatores de Transcrição NFATC/biossíntese , Adenocarcinoma/metabolismo , Animais , Neoplasias Brônquicas/metabolismo , Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/imunologia , Proteína Relacionada a TNFR Induzida por Glucocorticoide , Humanos , Interferon gama , Interleucina-2/biossíntese , Interleucina-2/imunologia , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Subunidade alfa de Receptor de Interleucina-2/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Fatores de Transcrição NFATC/deficiência , Fatores de Transcrição NFATC/genética , Receptores de Interleucina-7/biossíntese , Receptores de Interleucina-7/imunologia , Receptores de Fator de Crescimento Neural/biossíntese , Receptores de Fator de Crescimento Neural/imunologia , Receptores do Fator de Necrose Tumoral/biossíntese , Receptores do Fator de Necrose Tumoral/imunologia , Transcrição Gênica , Fator de Crescimento Transformador beta1/biossíntese , Fator de Crescimento Transformador beta1/imunologia , Transplante Heterólogo , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...