Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunol Cell Biol ; 94(4): 322-33, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26437769

RESUMEN

The adenylate cyclase toxin-hemolysin (CyaA) of Bordetella pertussis is a bi-functional leukotoxin. It penetrates myeloid phagocytes expressing the complement receptor 3 and delivers into their cytosol its N-terminal adenylate cyclase enzyme domain (~400 residues). In parallel, ~1300 residue-long RTX hemolysin moiety of CyaA forms cation-selective pores and permeabilizes target cell membrane for efflux of cytosolic potassium ions. The non-enzymatic CyaA-AC(-) toxoid, has repeatedly been successfully exploited as an antigen delivery tool for stimulation of adaptive T-cell immune responses. We show that the pore-forming activity confers on the CyaA-AC(-) toxoid a capacity to trigger Toll-like receptor and inflammasome signaling-independent maturation of CD11b-expressing dendritic cells (DC). The DC maturation-inducing potency of mutant toxoid variants in vitro reflected their specifically enhanced or reduced pore-forming activity and K(+) efflux. The toxoid-induced in vitro phenotypic maturation of DC involved the activity of mitogen activated protein kinases p38 and JNK and comprised increased expression of maturation markers, interleukin 6, chemokines KC and LIX and granulocyte-colony-stimulating factor secretion, prostaglandin E2 production and enhancement of chemotactic migration of DC. Moreover, i.v. injected toxoids induced maturation of splenic DC in function of their cell-permeabilizing capacity. Similarly, the capacity of DC to stimulate CD8(+) and CD4(+) T-cell responses in vitro and in vivo was dependent on the pore-forming activity of CyaA-AC(-). This reveals a novel self-adjuvanting capacity of the CyaA-AC(-) toxoid that is currently under clinical evaluation as a tool for delivery of immunotherapeutic anti-cancer CD8(+) T-cell vaccines into DC.


Asunto(s)
Toxina de Adenilato Ciclasa/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Activación de Linfocitos , Proteínas Citotóxicas Formadoras de Poros/inmunología , Dominios Proteicos/inmunología , Toxina de Adenilato Ciclasa/genética , Adyuvantes Inmunológicos/genética , Animales , Vacunas contra el Cáncer/inmunología , Diferenciación Celular , Permeabilidad de la Membrana Celular , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/microbiología , Transporte Iónico , Ratones , Ratones Endogámicos C57BL , Proteínas Citotóxicas Formadoras de Poros/genética , Dominios Proteicos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
Gastroenterology ; 142(2): 316-25.e1-12, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22079593

RESUMEN

BACKGROUND & AIMS: Copolymers of hydroxyethyl methacrylate and styrene sulfonate complex with isolated gliadin (the toxic fraction of gluten) and prevent damage to the intestinal barrier in HLA-HCD4/DQ8 mice. We studied the activity toward gluten and hordein digestion and biologic effects of poly(hydroxyethyl methacrylate-co-styrene sulfonate (P(HEMA-co-SS)). We also investigated the effect of gliadin complex formation in intestinal biopsy specimens from patients with celiac disease. METHODS: We studied the ability of P(HEMA-co-SS) to reduce digestion of wheat gluten and barley hordein into immunotoxic peptides using liquid chromatography-mass spectrometry. The biodistribution and pharmacokinetic profile of orally administered P(HEMA-co-SS) was established in rodents using tritium-labeled polymer. We assessed the capacity of P(HEMA-co-SS) to prevent the immunologic and intestinal effects induced by a gluten-food mixture in gluten-sensitized HLA-HCD4/DQ8 mice after short-term and long-term administration. We measured the effects of gliadin complex formation on cytokine release ex vivo using intestinal biopsy specimens from patients with celiac disease. RESULTS: P(HEMA-co-SS) reduced digestion of wheat gluten and barley hordein in vitro, thereby decreasing formation of toxic peptides associated with celiac disease. After oral administration to rodents, P(HEMA-co-SS) was predominantly excreted in feces, even in the presence of low-grade mucosal inflammation and increased intestinal permeability. In gluten-sensitized mice, P(HEMA-co-SS) reduced paracellular permeability, normalized anti-gliadin immunoglobulin A in intestinal washes, and modulated the systemic immune response to gluten in a food mixture. Furthermore, incubation of P(HEMA-co-SS) with mucosal biopsy specimens from patients with celiac disease showed that secretion of tumor necrosis factor-α was reduced in the presence of partially digested gliadin. CONCLUSIONS: The copolymer P(HEMA-co-SS) reduced digestion of wheat gluten and barley hordein and attenuated the immune response to gluten in a food mixture in rodents. It might be developed to prevent or reduce gluten-induced disorders in humans.


Asunto(s)
Enfermedad Celíaca/metabolismo , Digestión/efectos de los fármacos , Glútenes/metabolismo , Mucosa Intestinal/efectos de los fármacos , Polihidroxietil Metacrilato/análogos & derivados , Poliestirenos/farmacología , Estirenos/farmacología , Animales , Enfermedad Celíaca/tratamiento farmacológico , Enfermedad Celíaca/inmunología , Cromatografía Liquida , Femenino , Gliadina/metabolismo , Gliadina/toxicidad , Glútenes/toxicidad , Humanos , Absorción Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Yeyuno/efectos de los fármacos , Yeyuno/inmunología , Yeyuno/patología , Masculino , Espectrometría de Masas , Ratones , Permeabilidad/efectos de los fármacos , Polihidroxietil Metacrilato/farmacocinética , Polihidroxietil Metacrilato/farmacología , Polihidroxietil Metacrilato/uso terapéutico , Poliestirenos/farmacocinética , Poliestirenos/uso terapéutico , Unión Proteica , Distribución Aleatoria , Ratas , Estirenos/farmacocinética , Estirenos/uso terapéutico
3.
Oncology ; 82(1): 48-55, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22310016

RESUMEN

OBJECTIVE: Calreticulin is a multicompartmental protein which regulates many important cellular responses. The aim of this study was to elucidate whether the intensity and location of calreticulin overexpression in tumor cells are related to the elevated humoral immunity to calreticulin in patients with benign or malignant breast disease. METHODS: This study involved 27 patients with benign and 58 patients with malignant breast tumors before surgical resection and 38 healthy volunteers. Cytoplasmatic or membranous calreticulin overexpression in malignant or benign cells in paraffin-embedded tissues was determined using immunohistochemistry. Levels of the serum anti-calreticulin autoantibodies were detected by ELISA. RESULTS: Statistically significant differences between serum levels of IgA of anti-calreticulin antibodies in controls and patients with breast tumors, and between controls and patients with nonmalignant breast diseases were found, but no statistically significant differences were found between levels of serum IgG anti-calreticulin antibodies. Humoral immunity to calreticulin developed against cytoplasmatic and co-localized membranous calreticulin was not correlated to the intensity of its overexpression and was present even in the absence of its membranous localization. CONCLUSIONS: The degree of calreticulin overexpression in lobular breast carcinoma is lower than in ductal breast carcinoma. Elevated concentrations of anti-calreticulin IgA antibodies were present more frequently in patients with metastasis in locoregional lymph nodes in comparison to anti-calreticulin IgG antibodies.


Asunto(s)
Neoplasias de la Mama/metabolismo , Calreticulina/biosíntesis , Regulación Neoplásica de la Expresión Génica , Calreticulina/metabolismo , Carcinoma/metabolismo , Estudios de Casos y Controles , Membrana Celular/metabolismo , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Humanos , Inmunidad Humoral , Inmunoglobulina A/química , Inmunoglobulina G/química , Inmunohistoquímica/métodos , Metástasis Linfática , Metástasis de la Neoplasia
4.
J Cell Mol Med ; 15(4): 938-48, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20406323

RESUMEN

In genetically predisposed individuals, ingestion of wheat gliadin provokes a T-cell-mediated enteropathy, celiac disease. Gliadin fragments were previously reported to induce phenotypic maturation and Th1 cytokine production by human dendritic cells (DCs) and to boost their capacity to stimulate allogeneic T cells. Here, we monitor the effects of gliadin on migratory capacities of DCs. Using transwell assays, we show that gliadin peptic digest stimulates migration of human DCs and their chemotactic responsiveness to the lymph node-homing chemokines CCL19 and CCL21. The gliadin-induced migration is accompanied by extensive alterations of the cytoskeletal organization, with dissolution of adhesion structures, podosomes, as well as up-regulation of the CC chemokine receptor (CCR) 7 on cell surface and induction of cyclooxygenase (COX)-2 enzyme that mediates prostaglandin E2 (PGE2) production. Blocking experiments confirmed that gliadin-induced migration is independent of the TLR4 signalling. Moreover, we showed that the α-gliadin-derived 31-43 peptide is an active migration-inducing component of the digest. The migration promoted by gliadin fragments or the 31-43 peptide required activation of p38 mitogen-activated protein kinase (MAPK). As revealed using p38 MAPK inhibitor SB203580, this was responsible for DC cytoskeletal transition, CCR7 up-regulation and PGE2 production in particular. Taken together, this study provides a new insight into pathogenic features of gliadin fragments by demonstrating their ability to promote DC migration, which is a prerequisite for efficient priming of naive T cells, contributing to celiac disease pathology.


Asunto(s)
Quimiotaxis/efectos de los fármacos , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Gliadina/farmacología , Fragmentos de Péptidos/farmacología , Quimiocina CCL19/farmacología , Quimiocina CCL21/farmacología , Ciclooxigenasa 2/metabolismo , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Células Dendríticas/enzimología , Dinoprostona/biosíntesis , Activación Enzimática/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Modelos Biológicos , Receptores CCR7/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Inflamm Res ; 58(8): 503-12, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19271150

RESUMEN

OBJECTIVE: It is an open question whether multifunctional galectin-3 can be a serum marker in inflammatory bowel disease. METHODS: Western blots and commercial ELISA detected and quantitated the lectin immunocytochemistry using double labeling localized it in tissue sections. RESULTS: Serum concentrations were significantly increased in specimen of patients with active and remission-stage ulcerative colitis and Crohn's disease, associated with emerging positivity of CD14(+) cells. CONCLUSION: Enhanced concentration of galectin-3 in serum reflects presence of disease and points to its involvement in the pathogenesis.


Asunto(s)
Galectina 3/sangre , Enfermedades Inflamatorias del Intestino/sangre , Animales , Biomarcadores , Western Blotting , Colitis/inducido químicamente , Colitis Ulcerosa/sangre , Colon/metabolismo , Enfermedad de Crohn/sangre , Sulfato de Dextran , Electroforesis en Gel de Poliacrilamida , Escherichia coli/metabolismo , Femenino , Fluoresceína-5-Isotiocianato , Colorantes Fluorescentes , Humanos , Inmunohistoquímica , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/diagnóstico , Lectinas/metabolismo , Receptores de Lipopolisacáridos/análisis , Receptores de Lipopolisacáridos/metabolismo , Ratones , Ratones Endogámicos BALB C
6.
Front Immunol ; 10: 205, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30809227

RESUMEN

Background: Mucosal mast cells (MC) are key players in IgE-mediated food allergy (FA). The evidence on the interaction between gut microbiota, MC and susceptibility to FA is contradictory. Objective: We tested the hypothesis that commensal bacteria are essential for MC migration to the gut and their maturation impacting the susceptibility to FA. Methods: The development and severity of FA symptoms was studied in sensitized germ-free (GF), conventional (CV), and mice mono-colonized with L. plantarum WCFS1 or co-housed with CV mice. MC were phenotypically and functionally characterized. Results: Systemic sensitization and oral challenge of GF mice with ovalbumin led to increased levels of specific IgE in serum compared to CV mice. Remarkably, despite the high levels of sensitization, GF mice did not develop diarrhea or anaphylactic hypothermia, common symptoms of FA. In the gut, GF mice expressed low levels of the MC tissue-homing markers CXCL1 and CXCL2, and harbored fewer MC which exhibited lower levels of MC protease-1 after challenge. Additionally, MC in GF mice were less mature as confirmed by flow-cytometry and their functionality was impaired as shown by reduced edema formation after injection of degranulation-provoking compound 48/80. Co-housing of GF mice with CV mice fully restored their susceptibility to develop FA. However, this did not occur when mice were mono-colonized with L. plantarum. Conclusion: Our results demonstrate that microbiota-induced maturation and gut-homing of MC is a critical step for the development of symptoms of experimental FA. This new mechanistic insight into microbiota-MC-FA axis can be exploited in the prevention and treatment of FA in humans.


Asunto(s)
Hipersensibilidad a los Alimentos/etiología , Hipersensibilidad a los Alimentos/metabolismo , Mastocitos/inmunología , Mastocitos/metabolismo , Microbiota , Animales , Biomarcadores , Degranulación de la Célula/genética , Degranulación de la Célula/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Hipersensibilidad a los Alimentos/patología , Microbioma Gastrointestinal , Vida Libre de Gérmenes , Metagenoma , Metagenómica/métodos , Ratones , Microbiota/inmunología , ARN Ribosómico 16S
7.
Proteomics ; 8(8): 1677-91, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18340628

RESUMEN

Wheat belongs to six major food allergens inducing IgE-mediated hypersensitivity reaction manifesting as cutaneous, gastrointestinal, and respiratory symptoms. Although cereals are a staple food item in most diets, only a few wheat proteins causing hypersensitivity have been identified. To characterize wheat allergens, salt-soluble wheat extracts were separated by 1-DE and 2-DE and IgE-binding proteins were detected by immunoblotting using sera of patients with allergy to ingested wheat. Proteins, frequently recognized by IgE on 2-DE were analyzed by MALDI-TOF and QTOF and their spectrum was completed by 1-DE and LCQ(DECA) nLC-MS/MS IT technique. Using all three techniques we identified 19 potential wheat allergens such as alpha-amylase inhibitors, beta-amylase, profilin, serpin, beta-D-glucan exohydrolase, and 27K protein. Employing newly developed ELISA, levels of IgE Abs against Sulamit wheat extract and alpha-amylase inhibitors type 1 and 3 were quantified and shown to be significantly elevated in sera of allergic patients compared to those of healthy controls. The level of IgE Abs against alpha-amylase inhibitor type 3 was lower, slightly above the cut-off value in the majority of patients' sera. Our findings contribute to the identification of wheat allergens aimed to increase the specificity of serum IgE and cell activation diagnostic assays.


Asunto(s)
Alérgenos/inmunología , Inmunoglobulina E/sangre , Proteínas de Plantas/metabolismo , Proteoma/análisis , Triticum/inmunología , Hipersensibilidad al Trigo/inmunología , Adolescente , Adulto , Niño , Electroforesis en Gel Bidimensional , Electroforesis en Gel de Poliacrilamida , Inhibidores Enzimáticos/inmunología , Inhibidores Enzimáticos/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Immunoblotting , Fragmentos de Péptidos/análisis , Fragmentos de Péptidos/metabolismo , Proteínas de Plantas/inmunología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Triticum/efectos adversos , Triticum/metabolismo , Hipersensibilidad al Trigo/diagnóstico , alfa-Amilasas/antagonistas & inhibidores
8.
Nutrients ; 10(7)2018 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-30002342

RESUMEN

Immunologically mediated liver diseases belong to the common extraintestinal manifestations of celiac disease. We have reviewed the current literature that addresses the association between celiac disease and liver disorders. We searched relevant articles on MEDLINE/PubMed up to 15 June 2018. The objective of the article is to provide a comprehensive and up-to-date review on the latest hypotheses explaining the pathogenetic relationship between celiac disease and liver injury. Besides the involvement of gut⁻liver axis, tissue transglutaminase antibodies, and impairment of intestinal barrier, we integrate the latest achievements made in elucidation of the role of gut microbiota in celiac disease and liver disorders, that has not yet been sufficiently discussed in the literature in this context. The further objective is to provide a complete clinical overview on the types of liver diseases frequently found in celiac disease. In conclusion, the review highlights the clinical implication, recommend a rational approach for managing elevated transaminases in celiac patients, and underscore the importance of screening for celiac disease in patients with associated liver disease.


Asunto(s)
Autoinmunidad , Enfermedad Celíaca/inmunología , Hepatitis Autoinmune/inmunología , Intestinos/inmunología , Hígado/inmunología , Enfermedad del Hígado Graso no Alcohólico/inmunología , Animales , Autoanticuerpos/inmunología , Enfermedad Celíaca/dietoterapia , Enfermedad Celíaca/epidemiología , Enfermedad Celíaca/microbiología , Dieta Sin Gluten , Disbiosis , Proteínas de Unión al GTP/inmunología , Microbioma Gastrointestinal , Hepatitis Autoinmune/dietoterapia , Hepatitis Autoinmune/epidemiología , Hepatitis Autoinmune/microbiología , Humanos , Mucosa Intestinal/metabolismo , Hígado/microbiología , Enfermedad del Hígado Graso no Alcohólico/dietoterapia , Enfermedad del Hígado Graso no Alcohólico/epidemiología , Enfermedad del Hígado Graso no Alcohólico/microbiología , Permeabilidad , Pronóstico , Proteína Glutamina Gamma Glutamiltransferasa 2 , Factores de Riesgo , Transglutaminasas/inmunología , Deficiencia de Vitamina D/epidemiología , Deficiencia de Vitamina D/inmunología
9.
Gene ; 397(1-2): 169-77, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17560741

RESUMEN

Calreticulin is a highly conserved calcium-binding protein affecting many cellular processes inside and outside of the endoplasmic reticulum (ER). It participates in the regulation of Ca(2+) homeostasis, acts as a chaperone and modulates gene transcription, integrin-mediated cell signalling as well as cell adhesion. Here we report on the sequence characterization of a calreticulin-coding cDNA of Eisenia fetida earthworms. The neighbor-joining phylogeny tree constructed based on the deduced amino acid sequence indicates a common origin of the E. fetida calreticulin molecule and that of mollusks. A polyclonal anti-calreticulin antibody used for immunocytochemistry and immunohistochemistry localized the protein in the mesenchymal lining of the coelomic cavity and in coelomocytes of E. fetida. In situ hybridization revealed high expression of E. fetida calreticulin in various cells and tissues, namely epidermis, neurons of the ventral nerve cord, intestine, sperms, body wall muscles and some coelomocytes. Real-time PCR confirmed the strong expression of calreticulin in the nervous system, particularly in cerebral ganglia, in body wall muscles and in seminal vesicles. Moreover, a high calreticulin expression was measured in the muscular pharynx.


Asunto(s)
Calreticulina/genética , Calreticulina/metabolismo , Oligoquetos/genética , Oligoquetos/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , ADN Complementario/genética , Femenino , Inmunohistoquímica , Hibridación in Situ , Masculino , Datos de Secuencia Molecular , Filogenia , Proteínas Recombinantes/genética , Distribución Tisular
10.
J Agric Food Chem ; 55(7): 2627-32, 2007 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-17335223

RESUMEN

A gluten-free diet (GFD) is the sole effective long-lasting treatment of celiac disease. Four monoclonal antibodies (Abs) were prepared by immunization of animals kept on GFD with gliadin. The specificity of these Abs to decapeptides of alpha- and gamma-gliadin and omega-secalin was analyzed by the PEPSCAN technique. Repetitive sequences of alpha- and gamma-gliadin and omega-secalin containing the motifs QPFPXQ (X = Q, L, P) were recognized by all Abs tested. These Abs also frequently reacted with peptides containing the sequences QQSFPQQ, QQTFPQP, and QPFRPQ. On the basis of PEPSCAN results two Abs--8D4 and 7C6--were selected for the construction of a new ELISA kit for the detection of gliadin in food. The comparison of data obtained using the newly developed ELISA kit and commercially available ones indicated that Abs selection on the basis of their fine specificity to gliadin is useful for sensitive detection of gliadin in foods.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Análisis de los Alimentos/métodos , Gliadina/análisis , Gliadina/inmunología , Glútenes/análisis , Secuencia de Aminoácidos , Animales , Especificidad de Anticuerpos , Enfermedad Celíaca/dietoterapia , Ensayo de Inmunoadsorción Enzimática/métodos , Gliadina/química , Ratones
11.
Autoimmunity ; 49(8): 554-562, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27689957

RESUMEN

Distinct cellular level of the Ca2+-binding chaperone calreticulin (CRT) is essential for correct embryonal cardiac development and postnatal function. However, CRT is also a potential autoantigen eliciting formation of antibodies (Ab), whose role is not yet clarified. Immunization with CRT leads to cardiac injury, while overexpression of CRT in cardiomyocytes induces dilated cardiomyopathy (DCM) in animals. Hence, we analysed levels of anti-CRT Ab and calreticulin in the sera of patients with idiopatic DCM and hypertrophic cardiomyopathy (HCM). ELISA and immunoblot using human recombinant CRT and Pepscan with synthetic, overlapping decapeptides of CRT were used to detect anti-CRT Ab. Serum CRT concentration was tested by ELISA. Significantly increased levels of anti-CRT Ab of isotypes IgA (p < 0.001) and IgG (p < 0.05) were found in patients with both DCM (12/34 seropositive for IgA, 7/34 for IgG) and HCM (13/38 seropositive for IgA, 11/38 for IgG) against healthy controls (2/79 for IgA, 1/79 for IgG). Titration analysis in seropositive DCM and HCM patients documented anti-CRT Ab detected at 1/1600 dilution for IgG and 1/800 for IgA (and IgA1) and at least at 1/200 dilution for IgA2, IgG1, IgG2 and IgG3. Pepscan identified immunogenic CRT epitopes recognized by IgA and IgG Ab of these patients. Significantly increased levels of CRT relative to healthy controls were found in sera of patients with HCM (p < 0.01, 5/19). These data extend the knowledge of seroprevalence of anti-CRT Ab and CRT, and suggest possible involvement of autoimmune mechanisms directed to CRT in some forms of cardiomyopathies, which are clinically heterogeneous.


Asunto(s)
Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Calreticulina/sangre , Calreticulina/inmunología , Cardiomiopatía Dilatada/sangre , Cardiomiopatía Dilatada/inmunología , Cardiomiopatía Hipertrófica/sangre , Cardiomiopatía Hipertrófica/inmunología , Adulto , Anciano , Autoantígenos/sangre , Autoantígenos/inmunología , Autoinmunidad , Biomarcadores , Cardiomiopatía Dilatada/diagnóstico , Cardiomiopatía Hipertrófica/diagnóstico , Estudios de Casos y Controles , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunoglobulina A/sangre , Inmunoglobulina A/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Masculino , Persona de Mediana Edad , Adulto Joven
12.
Cell Mol Immunol ; 13(2): 251-62, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25942514

RESUMEN

Increasing numbers of clinical trials and animal experiments have shown that probiotic bacteria are promising tools for allergy prevention. Here, we analyzed the immunomodulatory properties of three selected lactobacillus strains and the impact of their mixture on allergic sensitization to Bet v 1 using a gnotobiotic mouse model. We showed that Lactobacillus (L.) rhamnosus LOCK0900, L. rhamnosus LOCK0908 and L. casei LOCK0919 are recognized via Toll-like receptor 2 (TLR2) and nucleotide-binding oligomerization domain-containing protein 2 (NOD2) receptors and stimulate bone marrow-derived dendritic cells to produce cytokines in species- and strain-dependent manners. Colonization of germ-free (GF) mice with a mixture of all three strains (Lmix) improved the intestinal barrier by strengthening the apical junctional complexes of enterocytes and restoring the structures of microfilaments extending into the terminal web. Mice colonized with Lmix and sensitized to the Bet v 1 allergen showed significantly lower levels of allergen-specific IgE, IgG1 and IgG2a and an elevated total IgA level in the sera and intestinal lavages as well as an increased transforming growth factor (TGF)-ß level compared with the sensitized GF mice. Splenocytes and mesenteric lymph node cells from the Lmix-colonized mice showed the significant upregulation of TGF-ß after in vitro stimulation with Bet v 1. Our results show that Lmix colonization improved the gut epithelial barrier and reduced allergic sensitization to Bet v 1. Furthermore, these findings were accompanied by the increased production of circulating and secretory IgA and the regulatory cytokine TGF-ß. Thus, this mixture of three lactobacillus strains shows potential for use in the prevention of increased gut permeability and the onset of allergies in humans.


Asunto(s)
Vida Libre de Gérmenes/inmunología , Hipersensibilidad/prevención & control , Mucosa Intestinal/inmunología , Lacticaseibacillus casei/inmunología , Lacticaseibacillus rhamnosus/inmunología , Animales , Antígenos de Plantas/toxicidad , Células HEK293 , Humanos , Hipersensibilidad/inmunología , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos BALB C , Proteína Adaptadora de Señalización NOD2/inmunología , Receptor Toll-Like 2/inmunología
13.
Ann N Y Acad Sci ; 1051: 787-98, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16127016

RESUMEN

Initial events and effector mechanisms of most inflammatory and autoimmune diseases remain largely unknown. Dysfunction of the innate and adaptive immune systems associated with mucosae (the major interface between the organism and its environment, e.g., microbiota, food) can conceivably cause impairment of mucosal barrier function and development of localized or systemic inflammatory and autoimmune processes. Animal models help in elucidating the etiology and pathogenetic mechanisms of human diseases, such as the inflammatory bowel diseases, Crohn's disease and ulcerative colitis, severe chronic diseases affecting the gut. To study the role of innate immunity and gut microbiota in intestinal inflammation, colitis was induced by dextran sulfate sodium (DSS) in mice with severe combined immunodeficiency (SCID). Conventionally reared (microflora-colonized) SCID mice displayed severe inflammation like that seen in immunocompetent Balb/c mice, whereas only minor changes appeared in the intestinal mucosa of DSS-fed gnotobiotic germ-free SCID mice. The presence of microflora facilitates the inflammation in DSS-induced colitis that develops in immunodeficient SCID mice, that is, in the absence of T and B lymphocytes. Celiac disease, a chronic autoimmune small bowel disorder, afflicts genetically susceptible individuals with wheat gluten intolerance. We showed that, in contrast with any other food proteins, wheat gliadin and its peptic fragments activate mouse macrophages and human monocytes to produce proinflammatory cytokines through the nuclear factor-kappaB signaling pathway. Activation of innate immunity cells by food proteins or components from gut microbiota thus could participate in the impairment of intestinal mucosa and the development of intestinal and/or systemic inflammation.


Asunto(s)
Enfermedades Autoinmunes/etiología , Inmunidad Innata , Inflamación/etiología , Animales , Enfermedades Autoinmunes/inmunología , Enfermedad Celíaca/etiología , Modelos Animales de Enfermedad , Humanos , Inmunidad Mucosa , Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/inmunología , Ratones
14.
J Leukoc Biol ; 71(4): 625-31, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11927649

RESUMEN

Celiac disease, induced by dietary gluten, is characterized by mucosal atrophy and local inflammation associated with cell infiltration and activation. Unlike other food proteins, gluten and its proteolytic fragments, besides inducing a specific immune response, were shown to activate components of innate immunity and cause, e.g., direct stimulation of TNF-alpha and IL-10 and a significant rise in NO production by peritoneal macrophages. The identity of the active fragments was established by separating the peptic digest of gliadin by RP-HPLC chromatography. The purest fraction with the highest activity was analyzed by mass spectrometry, and the gliadin peptide sequence was identified as VSFQQPQQQYPSSQ. This peptide (T) and its N- and C-terminally shortened forms (A, B, C and D, E, F) were synthesized. Peptide B (FQQPQQQYPSSQ) elicited the highest TNF-alpha, IL-10, and RANTES secretion and increase in IFN-gamma-primed NO production by mouse macrophages. In contrast, C-terminally shortened peptides had a lower ability to stimulate macrophages than the native form.


Asunto(s)
Gliadina/farmacología , Activación de Macrófagos/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Animales , Quimiocina CCL5/biosíntesis , Femenino , Interleucina-10/biosíntesis , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Fragmentos de Péptidos/aislamiento & purificación , Factor de Necrosis Tumoral alfa/biosíntesis
15.
FEBS Lett ; 571(1-3): 81-5, 2004 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-15280021

RESUMEN

Wheat gliadin is the triggering agent in coeliac disease. In this study, we documented that proteolytic fragments of gliadin, in contrast to other food antigens, induced interleukin (IL)-8 and tumour necrosis factor-alpha (TNF-alpha) production and significantly increased interferon (IFN)-gamma-induced cytokine secretion in human monocytic line THP-1 cells. Stimulation with gliadin resulted in elevated phosphorylation of the IkappaBalpha molecule and increased NF-kappaB/DNA binding activity that was inhibited by sulfasalazine, l-1-tosylamido-2-phenylethyl chloromethyl ketone and pyrrolidine dithiocarbamate (PDTC). The activation pathway was shown to be independent of the CD14 molecule. Less mature U-937 monocytes responded to gliadin stimulation by low IL-8 secretion, TNF-alpha production was not detectable. We propose that gliadin-induced activation of monocytes/macrophages can participate in mechanisms leading to the impairment of intestinal mucosa in coeliac patients.


Asunto(s)
Gliadina/farmacología , Interleucina-8/biosíntesis , Monocitos/inmunología , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Secuencia de Aminoácidos , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Humanos , Cinética , Monocitos/efectos de los fármacos , Monocitos/fisiología , Ovalbúmina/farmacología , Fragmentos de Péptidos/farmacología , Proteínas de Soja/farmacología , Triticum , Células U937
16.
Immunol Lett ; 93(2-3): 97-108, 2004 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15158604

RESUMEN

Commensal microflora (normal microflora, indigenous microbiota) consists of those micro-organisms, which are present on body surfaces covered by epithelial cells and are exposed to the external environment (gastrointestinal and respiratory tract, vagina, skin, etc.). The number of bacteria colonising mucosal and skin surfaces exceeds the number of cells forming human body. Commensal bacteria co-evolved with their hosts, however, under specific conditions they are able to overcome protective host responses and exert pathologic effects. Resident bacteria form complex ecosystems, whose diversity is enormous. The most abundant microflora is present in the distal parts of the gut; the majority of the intestinal bacteria are Gram-negative anaerobes. More than 50% of intestinal bacteria cannot be cultured by conventional microbiological techniques. Molecular biological methods help in analysing the structural and functional complexity of the microflora and in identifying its components. Resident microflora contains a number of components able to activate innate and adaptive immunity. Unlimited immune activation in response to signals from commensal bacteria could pose the risk of inflammation; immune responses to mucosal microbiota therefore require a precise regulatory control. The mucosal immune system has developed specialised regulatory, anti-inflammatory mechanisms for eliminating or tolerating non-dangerous, food and airborne antigens and commensal micro-organisms (oral, mucosal tolerance). However, at the same time the mucosal immune system must provide local defense mechanisms against environmental threats (e.g. invading pathogens). This important requirement is fulfilled by several mechanisms of mucosal immunity: strongly developed innate defense mechanisms ensuring appropriate function of the mucosal barrier, existence of unique types of lymphocytes and their products, transport of polymeric immunoglobulins through epithelial cells into secretions (sIgA) and migration and homing of cells originating from the mucosal organised tissues in mucosae and exocrine glands. The important role of commensal bacteria in development of optimally functioning mucosal immune system was demonstrated in germ-free animals (using gnotobiological techniques). Involvement of commensal microflora and its components with strong immunoactivating properties (e.g. LPS, peptidoglycans, superantigens, bacterial DNA, Hsp) in etiopathogenetic mechanism of various complex, multifactorial and multigenic diseases, including inflammatory bowel diseases, periodontal disease, rheumatoid arthritis, atherosclerosis, allergy, multiorgan failure, colon cancer has been recently suggested. Animal models of human diseases reared in defined gnotobiotic conditions are helping to elucidate the aetiology of these frequent disorders. An improved understanding of commensal bacteria-host interactions employing germ-free animal models with selective colonisation strategies combined with modern molecular techniques could bring new insights into the mechanisms of mucosal immunity and also into pathogenetic mechanisms of several infectious, inflammatory, autoimmune and neoplastic diseases. Regulation of microflora composition (e.g. by probiotics and prebiotics) offers the possibility to influence the development of mucosal and systemic immunity but it can play a role also in prevention and treatment of some diseases.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Bacterias/inmunología , Inmunidad Mucosa/inmunología , Inflamación/inmunología , Membrana Mucosa/inmunología , Enfermedades Autoinmunes/etiología , Bacterias/crecimiento & desarrollo , Enfermedad Crónica , Islas de CpG/inmunología , Células Epiteliales/inmunología , Proteínas de Choque Térmico/inmunología , Humanos , Tolerancia Inmunológica/inmunología , Inmunidad Innata/inmunología , Inflamación/etiología , Lipopolisacáridos/inmunología , Tejido Linfoide/inmunología , Membrana Mucosa/microbiología , Peptidoglicano/inmunología , Piel/inmunología , Piel/microbiología , Superantígenos/inmunología
17.
J Mass Spectrom ; 37(5): 507-11, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12112756

RESUMEN

Celiac disease, a chronic disorder of the small intestine, is caused by dietary gluten and is characterized by villous atrophy and local inflammation associated with infiltration of B and T lymphocytes and/or macrophages into the intestinal wall. In genetically predisposed individuals, the infiltrating cells are activated by gluten, gliadin and their proteolytic fragments and produce chemokines, cytokines and reactive radicals. The sequence of one of the macrophage-stimulatory gliadin peptic fragment was determined by mass spectrometry (MS) as VSFQQPQQQYPSSQ. The role of tissue transglutaminase (tTG) in innate immunity stimulation was studied by mass spectrometric monitoring of sequence changes in this active peptide. Two sites of glutamine deamidation in this peptide were localized by high-resolution scanning in MS/MS mode in an ion trap. A single deamidation in the parent peptide led to the complete loss of its stimulatory effect on macrophages.


Asunto(s)
Enfermedad Celíaca/metabolismo , Gliadina/análisis , Oligopéptidos/análisis , Fragmentos de Péptidos/análisis , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Secuencia de Aminoácidos , Animales , Proteínas de Unión al GTP/metabolismo , Gliadina/farmacología , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Cobayas , Humanos , Interleucina-10/metabolismo , Activación de Macrófagos/efectos de los fármacos , Datos de Secuencia Molecular , Óxido Nítrico/metabolismo , Oligopéptidos/farmacología , Fragmentos de Péptidos/farmacología , Proteína Glutamina Gamma Glutamiltransferasa 2 , Procesamiento Proteico-Postraduccional , Análisis de Secuencia de Proteína , Transglutaminasas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
18.
PLoS One ; 9(8): e104064, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25084094

RESUMEN

Adenylate cyclase toxin (CyaA) is a key virulence factor of the whooping cough agent Bordetella pertussis. The toxin targets CD11b-expressing phagocytes and delivers into their cytosol an adenylyl cyclase (AC) enzyme that subverts cellular signaling by increasing cAMP levels. In the present study, we analyzed the modulatory effects of CyaA on adhesive, migratory and antigen presenting properties of Toll-like receptor (TLR)-activated murine and human dendritic cells (DCs). cAMP signaling of CyaA enhanced TLR-induced dissolution of cell adhesive contacts and migration of DCs towards the lymph node-homing chemokines CCL19 and CCL21 in vitro. Moreover, we examined in detail the capacity of toxin-treated DCs to induce CD4(+) and CD8(+) T cell responses. Exposure to CyaA decreased the capacity of LPS-stimulated DCs to present soluble protein antigen to CD4+ T cells independently of modulation of co-stimulatory molecules and cytokine production, and enhanced their capacity to promote CD4(+)CD25(+)Foxp3(+) T regulatory cells in vitro. In addition, CyaA decreased the capacity of LPS-stimulated DCs to induce CD8(+) T cell proliferation and limited the induction of IFN-γ producing CD8(+) T cells while enhancing IL-10 and IL-17-production. These results indicate that through activation of cAMP signaling, the CyaA may be mobilizing DCs impaired in T cell stimulatory capacity and arrival of such DCs into draining lymph nodes may than contribute to delay and subversion of host immune responses during B. pertussis infection.


Asunto(s)
Toxina de Adenilato Ciclasa/farmacología , Bordetella pertussis/química , Movimiento Celular/efectos de los fármacos , Células Dendríticas/citología , Células Dendríticas/inmunología , Activación de Linfocitos/efectos de los fármacos , Receptores Toll-Like/metabolismo , Animales , Antígenos CD/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Adhesión Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Humanos , Ratones Endogámicos C57BL , Solubilidad , Linfocitos T Reguladores/efectos de los fármacos
19.
J Agric Food Chem ; 61(37): 8851-60, 2013 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-24016103

RESUMEN

Similarity among food allergens is a great problem affecting the specificity of diagnosis and treatment of allergic patients. We have observed that 80% of patients with food (including wheat) and pollen allergies have increased IgE antibodies against rice proteins. By immunoblotting, we documented that boiling decreased solubility and IgE reactivity of PBS-extracted rice and wheat proteins, yet in SDS extracts this reactivity was only slightly changed. The sera of patients highly positive on the IgE immunoblot and positive in basophil activation and skin prick test with boiled rice components were used for characterizing the IgE-binding proteins separated by 1D or 2D electrophoresis. Using mass spectrometry, we identified 22 rice SDS soluble proteins. Six of them were new thermostable potential rice allergens: glutelin C precursor, granule-bound starch synthase 1 protein, disulfide isomerase-like 1-1 protein, hypothetical protein OsI_13867, putative acid phosphatase precursor 1, and a protein encoded by locus Os02g0453600. All of the identified rice proteins differed from known wheat allergens, except proteins belonging to the α-amylase/trypsin inhibitor family. Furthermore, we would suggest that in patients with high IgE reactivity to wheat and rice components, the IgE immunoblot and skin prick test with boiled rice proteins could be beneficial before diet recommendation.


Asunto(s)
Hipersensibilidad a los Alimentos/inmunología , Inmunoglobulina E/inmunología , Oryza/inmunología , Proteínas de Plantas/inmunología , Adolescente , Adulto , Anciano , Basófilos/inmunología , Niño , Preescolar , Femenino , Hipersensibilidad a los Alimentos/diagnóstico , Humanos , Immunoblotting , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Oryza/química , Proteínas de Plantas/química , Pruebas Cutáneas , Triticum/química , Triticum/inmunología , Adulto Joven
20.
PLoS One ; 8(4): e62426, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23658628

RESUMEN

Celiac disease (CD) is a gluten-responsive, chronic inflammatory enteropathy. IL-1 cytokine family members IL-1ß and IL-18 have been associated with the inflammatory conditions in CD patients. However, the mechanisms of IL-1 molecule activation in CD have not yet been elucidated. We show in this study that peripheral blood mononuclear cells (PBMC) and monocytes from celiac patients responded to pepsin digest of wheat gliadin fraction (PDWGF) by a robust secretion of IL-1ß and IL-1α and a slightly elevated production of IL-18. The analysis of the upstream mechanisms underlying PDWGF-induced IL-1ß production in celiac PBMC show that PDWGF-induced de novo pro-IL-1ß synthesis, followed by a caspase-1 dependent processing and the secretion of mature IL-1ß. This was promoted by K+ efflux and oxidative stress, and was independent of P2X7 receptor signaling. The PDWGF-induced IL-1ß release was dependent on Nod-like receptor family containing pyrin domain 3 (NLRP3) and apoptosis-associated speck like protein (ASC) as shown by stimulation of bone marrow derived dendritic cells (BMDC) from NLRP3(-/-) and ASC(-/-) knockout mice. Moreover, treatment of human PBMC as well as MyD88(-/-) and Toll-interleukin-1 receptor domain-containing adaptor-inducing interferon-ß (TRIF)(-/-) BMDC illustrated that prior to the activation of caspase-1, the PDWGF-triggered signal constitutes the activation of the MyD88/TRIF/MAPK/NF-κB pathway. Moreover, our results indicate that the combined action of TLR2 and TLR4 may be required for optimal induction of IL-1ß in response to PDWGF. Thus, innate immune pathways, such as TLR2/4/MyD88/TRIF/MAPK/NF-κB and an NLRP3 inflammasome activation are involved in wheat proteins signaling and may play an important role in the pathogenesis of CD.


Asunto(s)
Proteínas Portadoras/inmunología , Gliadina/inmunología , Inflamasomas/efectos de los fármacos , Interleucina-1beta/inmunología , Leucocitos Mononucleares/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Adulto , Animales , Proteínas Portadoras/genética , Enfermedad Celíaca , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Gliadina/química , Humanos , Inflamasomas/genética , Inflamasomas/inmunología , Interleucina-1beta/genética , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/patología , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/inmunología , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR , Pepsina A , Cultivo Primario de Células , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA