Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Int Arch Allergy Immunol ; 185(5): 503-518, 2024.
Article in English | MEDLINE | ID: mdl-38408438

ABSTRACT

BACKGROUND: Allergy represents a major health problem of increasing prevalence worldwide with a high socioeconomic impact. Our knowledge on the molecular mechanisms underlying allergic diseases and their treatments has significantly improved over the last years. The generation of allergen-specific regulatory T cells (Tregs) is crucial in the induction of healthy immune responses to allergens, preventing the development and worsening of allergic diseases. SUMMARY: In the last decades, intensive research has focused on the study of the molecular mechanisms involved in Treg development and Treg-mediated suppression. These mechanisms are essential for the induction of sustained tolerance by allergen-specific immunotherapy (AIT) after treatment discontinuation. Compelling experimental evidence demonstrated altered suppressive capacity of Tregs in patients suffering from allergic rhinitis, allergic asthma, food allergy, or atopic dermatitis, as well as the restoration of their numbers and functionality after successful AIT. KEY MESSAGE: The better understanding of the molecular mechanisms involved in Treg generation during allergen tolerance induction might well contribute to the development of novel strategies for the prevention and treatment of allergic diseases.


Subject(s)
Desensitization, Immunologic , Hypersensitivity , Immune Tolerance , T-Lymphocytes, Regulatory , T-Lymphocytes, Regulatory/immunology , Humans , Hypersensitivity/immunology , Hypersensitivity/therapy , Animals , Desensitization, Immunologic/methods , Allergens/immunology
2.
Nat Commun ; 14(1): 2880, 2023 05 19.
Article in English | MEDLINE | ID: mdl-37208336

ABSTRACT

Regulation of cutaneous immunity is severely compromised in inflammatory skin disease. To investigate the molecular crosstalk underpinning tolerance versus inflammation in atopic dermatitis, we utilise a human in vivo allergen challenge study, exposing atopic dermatitis patients to house dust mite. Here we analyse transcriptional programmes at the population and single cell levels in parallel with immunophenotyping of cutaneous immunocytes revealed a distinct dichotomy in atopic dermatitis patient responsiveness to house dust mite challenge. Our study shows that reactivity to house dust mite was associated with high basal levels of TNF-expressing cutaneous Th17 T cells, and documents the presence of hub structures where Langerhans cells and T cells co-localised. Mechanistically, we identify expression of metallothioneins and transcriptional programmes encoding antioxidant defences across all skin cell types, that appear to protect against allergen-induced inflammation. Furthermore, single nucleotide polymorphisms in the MTIX gene are associated with patients who did not react to house dust mite, opening up possibilities for therapeutic interventions modulating metallothionein expression in atopic dermatitis.


Subject(s)
Dermatitis, Atopic , Animals , Humans , Dermatitis, Atopic/genetics , Allergens , Inflammation/genetics , Skin , Pyroglyphidae
3.
Br J Dermatol ; 188(3): 396-406, 2023 02 22.
Article in English | MEDLINE | ID: mdl-36637891

ABSTRACT

BACKGROUND: Acute cutaneous inflammation causes microbiome alterations as well as ultrastructural changes in epidermis stratification. However, the interactions between keratinocyte proliferation and differentiation status and the skin microbiome have not been fully explored. OBJECTIVES: Hypothesizing that the skin microbiome contributes to regulation of keratinocyte differentiation and can modify antimicrobial responses, we examined the effect of exposure to commensal (Staphylococcus epidermidis, SE) or pathogenic (Staphylococcus aureus, SA) challenge on epidermal models. METHODS: Explant biopsies were taken to investigate species-specific antimicrobial effects of host factors. Further investigations were performed in reconstituted epidermal models by bulk transcriptomic analysis alongside secreted protein profiling. Single-cell RNA sequencing analysis was performed to explore the keratinocyte populations responsible for SA inflammation. A dataset of 6391 keratinocytes from control (2044 cells), SE challenge (2028 cells) and SA challenge (2319 cells) was generated from reconstituted epidermal models. RESULTS: Bacterial lawns of SA, not SE, were inhibited by human skin explant samples, and microarray analysis of three-dimensional epidermis models showed that host antimicrobial peptide expression was induced by SE but not SA. Protein analysis of bacterial cocultured models showed that SA exposure induced inflammatory mediator expression, indicating keratinocyte activation of other epidermal immune populations. Single-cell DropSeq analysis of unchallenged naive, SE-challenged and SA-challenged epidermis models was undertaken to distinguish cells from basal, spinous and granular layers, and to interrogate them in relation to model exposure. In contrast to SE, SA specifically induced a subpopulation of spinous cells that highly expressed transcripts related to epidermal inflammation and antimicrobial response. Furthermore, SA, but not SE, specifically induced a basal population that highly expressed interleukin-1 alarmins. CONCLUSIONS: These findings suggest that SA-associated remodelling of the epidermis is compartmentalized to different keratinocyte populations. Elucidating the mechanisms regulating bacterial sensing-triggered inflammatory responses within tissues will enable further understanding of microbiome dysbiosis and inflammatory skin diseases, such as atopic eczema.


Subject(s)
Dermatitis, Atopic , Staphylococcal Infections , Humans , Staphylococcus aureus , Keratinocytes/metabolism , Epidermis/metabolism , Inflammation , Cell Differentiation , Staphylococcal Infections/pathology
4.
Front Immunol ; 13: 892254, 2022.
Article in English | MEDLINE | ID: mdl-36203560

ABSTRACT

Human epidermal Langerhans cells (LCs) maintain immune homeostasis in the skin. To examine transcriptional programming of human primary LCs during homeostasis, we performed scRNA-seq analysis of LCs before and after migration from the epidermis, coupled with functional assessment of their regulatory T cell priming capabilities. The analysis revealed that steady-state LCs exist in a continuum of maturation states and upregulate antigen presentation genes along with an immunoregulatory module including the genes IDO1, LGALS1, LAMTOR1, IL4I, upon their migration. The migration-induced transition in genomic state is accompanied by the ability of LCs to more efficiently prime regulatory T cell responses in co-culture assays. Computational analyses of the scRNAseq datasets using SCENIC and Partial Information Decomposition in Context identified a set of migration-induced transcription factors including IRF4, KLF6 and RelB as key nodes within a immunoregulatory gene regulatory network. These findings support a model in which efficient priming of immunoregulatory responses by LCs is dependent on coordinated upregulation of a migration-coupled maturation program with a immunoregulation-promoting genomic module.


Subject(s)
Galectin 1 , Langerhans Cells , Cell Movement/genetics , Epidermis , Humans , Skin
5.
Front Immunol ; 12: 665312, 2021.
Article in English | MEDLINE | ID: mdl-34211464

ABSTRACT

Langerhans cells (LCs) reside in the epidermis as a dense network of immune system sentinels, coordinating both immunogenic and tolerogenic immune responses. To determine molecular switches directing induction of LC immune activation, we performed mathematical modelling of gene regulatory networks identified by single cell RNA sequencing of LCs exposed to TNF-alpha, a key pro-inflammatory signal produced by the skin. Our approach delineated three programmes of LC phenotypic activation (immunogenic, tolerogenic or ambivalent), and confirmed that TNF-alpha enhanced LC immunogenic programming. Through regulon analysis followed by mutual information modelling, we identified IRF1 as the key transcription factor for the regulation of immunogenicity in LCs. Application of a mathematical toggle switch model, coupling IRF1 with tolerance-inducing transcription factors, determined the key set of transcription factors regulating the switch between tolerance and immunogenicity, and correctly predicted LC behaviour in LCs derived from different body sites. Our findings provide a mechanistic explanation of how combinatorial interactions between different transcription factors can coordinate specific transcriptional programmes in human LCs, interpreting the microenvironmental context of the local tissue microenvironments.


Subject(s)
Interferon Regulatory Factors/metabolism , Langerhans Cells/immunology , Langerhans Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism , Epidermis/metabolism , Gene Expression Regulation , Gene Regulatory Networks , Humans , Interferon Regulatory Factors/genetics , Signal Transduction , Transcription, Genetic , Tumor Necrosis Factor-alpha/genetics
6.
Nat Commun ; 11(1): 313, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31949143

ABSTRACT

Langerhans cells (LC) can prime tolerogenic as well as immunogenic responses in skin, but the genomic states and transcription factors (TF) regulating these context-specific responses are unclear. Bulk and single-cell transcriptional profiling demonstrates that human migratory LCs are robustly programmed for MHC-I and MHC-II antigen presentation. Chromatin analysis reveals enrichment of ETS-IRF and AP1-IRF composite regulatory elements in antigen-presentation genes, coinciding with expression of the TFs, PU.1, IRF4 and BATF3 but not IRF8. Migration of LCs from the epidermis is accompanied by upregulation of IRF4, antigen processing components and co-stimulatory molecules. TNF stimulation augments LC cross-presentation while attenuating IRF4 expression. CRISPR-mediated editing reveals IRF4 to positively regulate the LC activation programme, but repress NF2EL2 and NF-kB pathway genes that promote responsiveness to oxidative stress and inflammatory cytokines. Thus, IRF4-dependent genomic programming of human migratory LCs appears to enable LC maturation while attenuating excessive inflammatory and immunogenic responses in the epidermis.


Subject(s)
Genomics , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Langerhans Cells/metabolism , Antigen Presentation/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , CRISPR-Cas Systems , Cell Movement , Cytokines/metabolism , Gene Editing , Gene Expression Profiling , Histocompatibility Antigens Class I , Histocompatibility Antigens Class II , Humans , Langerhans Cells/immunology , NF-kappa B/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Transcriptional Activation , Up-Regulation
7.
Eur J Immunol ; 48(1): 180-193, 2018 01.
Article in English | MEDLINE | ID: mdl-28799230

ABSTRACT

Recurrent respiratory tract infections (RRTIs) are the first leading cause of community- and nosocomial-acquired infections. Antibiotics remain the mainstay of treatment, enhancing the potential to develop antibiotic resistances. Therefore, the development of new alternative approaches to prevent and treat RRTIs is highly demanded. Daily sublingual administration of the whole heat-inactivated polybacterial preparation (PBP) MV130 significantly reduced the rate of respiratory infections in RRTIs patients, however, the immunological mechanisms of action remain unknown. Herein, we study the capacity of MV130 to immunomodulate the function of human dendritic cells (DCs) as a potential mechanism that contribute to the clinical benefits. We demonstrate that DCs from RRTIs patients and healthy controls display similar ex vivo immunological responses to MV130. By combining systems biology and functional immunological approaches we show that MV130 promotes the generation of Th1/Th17 responses via receptor-interacting serine/threonine-protein kinase-2 (RIPK2)- and myeloid-differentiation primary-response gene-88 (MyD88)-mediated signalling pathways under the control of IL-10. In vivo BALB/c mice sublingually immunized with MV130 display potent systemic Th1/Th17 and IL-10 responses against related and unrelated antigens. We elucidate immunological mechanisms underlying the potential way of action of MV130, which might help to design alternative treatments in other clinical conditions with high risk of recurrent infections.


Subject(s)
Bacterial Vaccines/immunology , Dendritic Cells/immunology , Interleukin-10/immunology , Myeloid Differentiation Factor 88/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Respiratory Tract Infections/prevention & control , Th1 Cells/immunology , Th17 Cells/immunology , Adult , Aged , Animals , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Signal Transduction/immunology
8.
Front Immunol ; 8: 1676, 2017.
Article in English | MEDLINE | ID: mdl-29238347

ABSTRACT

Langerhans cells (LCs) reside in the epidermis as a dense network of immune system sentinels. These cells determine the appropriate adaptive immune response (inflammation or tolerance) by interpreting the microenvironmental context in which they encounter foreign substances. In a normal physiological, "non-dangerous" situation, LCs coordinate a continuous state of immune tolerance, preventing unnecessary and harmful immune activation. Conversely, when they sense a danger signal, for example during infection or when the physical integrity of skin has been compromised as a result of a trauma, they instruct T lymphocytes of the adaptive immune system to mount efficient effector responses. Recent advances investigating the molecular mechanisms underpinning the cross talk between LCs and the epidermal microenvironment reveal its importance for programming LC biology. This review summarizes the novel findings describing LC origin and function through the analysis of the transcriptomic programs and gene regulatory networks (GRNs). Review and meta-analysis of publicly available datasets clearly delineates LCs as distinct from both conventional dendritic cells (DCs) and macrophages, suggesting a primary role for the epidermal microenvironment in programming LC biology. This concept is further supported by the analysis of the effect of epidermal pro-inflammatory signals, regulating key GRNs in human and murine LCs. Applying whole transcriptome analyses and in silico analysis has advanced our understanding of how LCs receive, integrate, and process signals from the steady-state and diseased epidermis. Interestingly, in homeostasis and under immunological stress, the molecular network in LCs remains relatively stable, reflecting a key evolutionary need related to tissue localization. Importantly, to fulfill their key role in orchestrating antiviral adaptive immune responses, LC share specific transcriptomic modules with other DC types able to cross-present antigens to cytotoxic CD8+ T cells, pointing to a possible evolutionary convergence mechanism. With the development of more advanced technologies allowing delineation of the molecular networks at the level of chromatin organization, histone modifications, protein translation, and phosphorylation, future "omics" investigations will bring in-depth understanding of the complex molecular mechanisms underpinning human LC biology.

9.
J Allergy Clin Immunol ; 138(2): 558-567.e11, 2016 08.
Article in English | MEDLINE | ID: mdl-27177779

ABSTRACT

BACKGROUND: Allergen immunotherapy (AIT) is the only curative treatment for allergy. AIT faces pitfalls related to efficacy, security, duration, and patient compliance. Novel vaccines overcoming such inconveniences are in demand. OBJECTIVES: We sought to study the immunologic mechanisms of action for novel vaccines targeting dendritic cells (DCs) generated by coupling glutaraldehyde-polymerized grass pollen allergoids to nonoxidized mannan (PM) compared with glutaraldehyde-polymerized allergoids (P) or native grass pollen extracts (N). METHODS: Skin prick tests and basophil activation tests with N, P, or PM were performed in patients with grass pollen allergy. IgE-blocking experiments, flow cytometry, confocal microscopy, cocultures, suppression assays, real-time quantitative PCR, ELISAs, and ELISpot assays were performed to assess allergen capture by human DCs and T-cell responses. BALB/c mice were immunized with PM, N, or P. Antibody levels, cytokine production by splenocytes, and splenic forkhead box P3 (FOXP3)(+) regulatory T (Treg) cells were quantified. Experiments with oxidized PM were also performed. RESULTS: PM displays in vivo hypoallergenicity, induces potent blocking antibodies, and is captured by human DCs much more efficiently than N or P by mechanisms depending on mannose receptor- and dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin-mediated internalization. PM endorses human DCs to generate functional FOXP3(+) Treg cells through programmed death ligand 1. Immunization of mice with PM induces a shift to nonallergic responses and increases the frequency of splenic FOXP3(+) Treg cells. Mild oxidation impairs these effects in human subjects and mice, demonstrating the essential role of preserving the carbohydrate structure of mannan. CONCLUSIONS: Allergoids conjugated to nonoxidized mannan represent suitable vaccines for AIT. Our findings might also be of the utmost relevance to development of therapeutic interventions in other immune tolerance-related diseases.


Subject(s)
Allergens/immunology , B7-H1 Antigen/metabolism , Dendritic Cells/immunology , Mannans , Plant Extracts , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Vaccines/immunology , Adjuvants, Immunologic , Allergens/metabolism , Allergoids , Animals , Antibodies/immunology , Antibodies, Blocking/immunology , Cytokines/metabolism , Dendritic Cells/metabolism , Humans , Immune Tolerance/immunology , Mice , Poaceae/immunology , Pollen/immunology , Rhinitis, Allergic, Seasonal/immunology , Rhinitis, Allergic, Seasonal/metabolism
10.
Glycoconj J ; 33(1): 93-101, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26603537

ABSTRACT

Immunotherapy for treating IgE-mediated allergies requires high doses of the corresponding allergen. This may result in undesired side effects and, to avoid them, hypoallergenic allergens (allergoids) polymerized with glutaraldehyde are commonly used. Targeting allergoids to dendritic cells to enhance cell uptake may result in a more effective immunotherapy. Allergoids coupled to yeast mannan, as source of polymannoses, would be suitable for this purpose, since mannose-binding receptors are expressed on these cells. Conventional conjugation procedures of mannan to proteins use oxidized mannan to release reactive aldehydes able to bind to free amino groups in the protein; yet, allergoids lack these latter because their previous treatment with glutaraldehyde. The aim of this study was to obtain allergoids conjugated to mannan by an alternative approach based on just glutaraldehyde treatment, taking advantage of the mannoprotein bound to the polymannose backbone. Allergoid-mannan glycoconjugates were produced in a single step by treating with glutaraldehyde a defined mixture of allergens derived from Phleum pratense grass pollen and native mannan (non-oxidized) from Saccharomyces cerevisae. Analytical and structural studies, including 2D-DOSY and (1)H-(13)C HSQC nuclear magnetic resonance spectra, demonstrated the feasibility of such an approach. The glycoconjugates obtained were polymers of high molecular weight showing a higher stability than the native allergen or the conventional allergoid without mannan. The allergoid-mannan glycoconjugates were hypoallergenic as detected by the IgE reactivity with sera from grass allergic patients, even with lower reactivity than conventional allergoid without mannan. Thus, stable hypoallergenic allergoids conjugated to mannan suitable for using in immunotherapy can be achieved using glutaraldehyde. In contrast to mannan oxidation, the glutaraldehyde approach allows to preserve mannoses with their native geometry, which may be functionally important for its receptor-mediated recognition.


Subject(s)
Allergens/chemistry , Fungal Polysaccharides/chemistry , Pollen/chemistry , Allergens/immunology , Antigen-Antibody Reactions , Fungal Polysaccharides/immunology , Humans , Immunoglobulin E/immunology , Poaceae , Pollen/immunology , Saccharomyces cerevisiae/chemistry , Vaccines/immunology
12.
PLoS One ; 7(12): e50799, 2012.
Article in English | MEDLINE | ID: mdl-23272072

ABSTRACT

The study of cross-reactivity in allergy is key to both understanding. the allergic response of many patients and providing them with a rational treatment In the present study, protein microarrays and a co-sensitization graph approach were used in conjunction with an allergen microarray immunoassay. This enabled us to include a wide number of proteins and a large number of patients, and to study sensitization profiles among members of the LTP family. Fourteen LTPs from the most frequent plant food-induced allergies in the geographical area studied were printed into a microarray specifically designed for this research. 212 patients with fruit allergy and 117 food-tolerant pollen allergic subjects were recruited from seven regions of Spain with different pollen profiles, and their sera were tested with allergen microarray. This approach has proven itself to be a good tool to study cross-reactivity between members of LTP family, and could become a useful strategy to analyze other families of allergens.


Subject(s)
Allergens/immunology , Antigens, Plant/immunology , Carrier Proteins/immunology , Plant Proteins/immunology , Protein Array Analysis/methods , Allergens/chemistry , Cross Reactions/immunology , Electrophoresis, Polyacrylamide Gel , Epitopes/chemistry , Food , Food Hypersensitivity/immunology , Geography , Humans , Immunoassay/methods , Lipids/chemistry , Models, Statistical , Pollen , Recombinant Proteins/chemistry , Spain , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
13.
Clin Transl Allergy ; 2(1): 23, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23231956

ABSTRACT

BACKGROUND: The 11S globulin Sin a 2 is a marker to predict severity of symptoms in mustard allergic patients. The potential implication of Sin a 2 in cross-reactivity with tree nuts and peanut has not been investigated so far. In this work, we studied at the IgG and IgE level the involvement of the 11S globulin Sin a 2 in cross-reactivity among mustard, tree nuts and peanut. METHODS: Eleven well-characterized mustard-allergic patients sensitized to Sin a 2 were included in the study. A specific anti-Sin a 2 serum was obtained in rabbit. Skin prick tests (SPT), enzyme-linked immunosorbent assay (ELISA), immunoblotting and IgG or IgE-inhibition immunoblotting experiments using purified Sin a 2, Sin a 1, Sin a 3, mustard, almond, hazelnut, pistachio, walnut or peanut extracts were performed. RESULTS: The rabbit anti-Sin a 2 serum showed high affinity and specificity to Sin a 2, which allowed us to demonstrate that Sin a 2 shares IgG epitopes with allergenic 11S globulins from tree nuts (almond, hazelnut, pistachio and walnut) but not from peanut. All the patients included in the study had positive skin prick test to tree nuts and/or peanut and we subdivided them into two different groups according to their clinical symptoms after ingestion of such allergenic sources. We showed that 11S globulins contain conserved IgE epitopes involved in cross-reactivity among mustard, tree nuts and peanut as well as species-specific IgE epitopes. CONCLUSIONS: The allergenic 11S globulin Sin a 2 from mustard is involved in cross-reactivity at the IgE level with tree nuts and peanut. Although the clinical relevance of the cross-reactive IgE epitopes present in 11S globulins needs to be investigated in further detail, our results contribute to improve the diagnosis and management of mustard allergic patients sensitized to Sin a 2.

14.
J Agric Food Chem ; 60(23): 6011-8, 2012 Jun 13.
Article in English | MEDLINE | ID: mdl-22594937

ABSTRACT

This work investigates the resistance to proteolysis and heating of the yellow mustard (Sinapis alba L.) allergens Sin a 1 (2S albumin), Sin a 3 (nonspecific lipid transfer protein, LTP), and Sin a 4 (profilin) to explain their potential capability to induce primary sensitization at the gastrointestinal level. Sin a 1 and Sin a 3 resisted gastric digestion showing no reduction of the IgE reactivity. Intestinal digestion of Sin a 1 and Sin a 3 produced a limited proteolysis but retained significant IgE-binding reactivity. Sin a 1 was stable after heating, and although Sin a 3 was modified, most of its structure was recovered after cooling back. These two allergens would be therefore able to sensitize by ingestion. Sin a 4 was completely digested by gastric treatment and its conformational structure markedly modified at 85 °C. Thus, this allergen can be described as a nonsensitizing mustard allergen.


Subject(s)
Albumins/metabolism , Allergens/metabolism , Carrier Proteins/metabolism , Mustard Plant/chemistry , Profilins/metabolism , Seeds/chemistry , Albumins/immunology , Allergens/immunology , Antigens, Plant/immunology , Antigens, Plant/metabolism , Carrier Proteins/immunology , Circular Dichroism , Digestion/drug effects , Electrophoresis, Polyacrylamide Gel , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Hot Temperature , Humans , Immunoblotting , Immunoglobulin E/metabolism , Molecular Structure , Plant Proteins/immunology , Plant Proteins/metabolism , Profilins/immunology , Protein Conformation , Proteolysis/drug effects , Spices/analysis
15.
Int Arch Allergy Immunol ; 156(3): 291-6, 2011.
Article in English | MEDLINE | ID: mdl-21720174

ABSTRACT

BACKGROUND: Cross-reactivity among plant food allergens belonging to the nonspecific lipid transfer protein (LTP) family is well known. In contrast, the relationship among these allergens and their putative homologs from olive (Ole e 7) and Parietaria (Par j 1) pollen has not been clarified. METHODS: Sera with specific IgE to LTP allergens were obtained from peach-, mustard- and olive pollen-allergic patients. Purified LTP allergens from foods (peach, apple, mustard and wheat) and pollens (olive, mugwort and Parietaria) were tested by ELISA and ELISA-inhibition assays. RESULTS: Plant food LTP-allergic patients showed a significantly higher number of sera (89-100 vs. 33-64%) with specific IgE and mean specific IgE levels (0.30-1.56 vs. 0.21-0.34 OD units) to the 4 food LTP allergens tested than to olive Ole e 7 and Parietaria Par j 1 pollen. ELISA-inhibition assays indicated cross-inhibition between food LTP allergens but no cross-reactivity between these allergens and Ole e 7 and Par j 1, or, even more, between the LTP allergens from olive and Parietaria pollen. CONCLUSIONS: LTP allergens from olive and Parietaria pollen cross-react neither with allergenic LTPs from plant foods nor between themselves. Therefore, both pollens do not seem to be related with the LTP syndrome.


Subject(s)
Allergens/immunology , Antigens, Plant/immunology , Carrier Proteins/immunology , Cross Reactions , Parietaria/immunology , Plant Proteins/immunology , Pollen/immunology , Amino Acid Sequence , Food Hypersensitivity , Humans , Immunoglobulin E/blood , Immunoglobulin E/immunology , Olea/immunology , Sequence Alignment
16.
Ann Allergy Asthma Immunol ; 106(5): 429-35, 2011 May.
Article in English | MEDLINE | ID: mdl-21530876

ABSTRACT

BACKGROUND: Profilins are commonly involved in polysensitization of allergic patients; therefore, appropriate markers should be used in component-resolved diagnosis. OBJECTIVE: To evaluate the immunological equivalence between profilins from pollens and plant-derived foods, to be used in component-resolved diagnosis. METHODS: Specific immunoglobulin (Ig) G antibodies against pollen and fruit profilins, as well as sera from patients allergic to mustard, melon, or olive pollen, were used. Purified profilins from mustard seeds, fruit melon, and chenopod and birch pollen were assayed in immunoblotting, enzyme-linked immunosorbent assay (ELISA), and ELISA inhibition assays. RESULTS: Significant correlation was found in the response of purified profilins by ELISA and immunoblotting for both specific IgG and IgE. The highest levels of IgE binding were obtained for olive pollen-allergic patients, which could be related to the route of sensitization. The responses of individual patients to profilins were also similar and independent of the sensitizing source. The inhibition between pairs of allergens was generally higher than 70%, indicating that profilins share most of the IgE epitopes. Modeling of mimotopes in the conformational structure of the implicated profilins supports their strong cross-reactivity obtained experimentally. CONCLUSIONS: No correlation exists between the level of IgE response of individual patients to specific profilins and the corresponding theoretical sensitizing source, suggesting that the sensitization could be attributable to any profilin present in the environment of the patients. This would bear out the use of most profilins as a common marker for polysensitization in component-resolved diagnosis and for therapeutic approaches.


Subject(s)
Allergens/immunology , Antigen-Antibody Reactions/immunology , Immunoglobulin E/immunology , Plants, Edible/immunology , Pollen/immunology , Profilins/immunology , Allergens/chemistry , Allergens/genetics , Amino Acid Sequence , Animals , Antigens, Plant/chemistry , Antigens, Plant/genetics , Antigens, Plant/immunology , Binding, Competitive/immunology , Chenopodium/chemistry , Chenopodium/immunology , Cross Reactions/immunology , Enzyme-Linked Immunosorbent Assay , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Humans , Hypersensitivity/diagnosis , Hypersensitivity/immunology , Immune Sera/immunology , Immunoblotting , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Mustard Plant/chemistry , Mustard Plant/immunology , Plants, Edible/chemistry , Pollen/chemistry , Profilins/chemistry , Profilins/genetics , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sequence Homology, Amino Acid
18.
Ann Allergy Asthma Immunol ; 94(5): 586-92, 2005 May.
Article in English | MEDLINE | ID: mdl-15945563

ABSTRACT

BACKGROUND: Although mustard seed allergy has been largely reported during the preceding 20 years, currently only 2 allergens, Sin a 1 and Bra j 1, have been identified. OBJECTIVE: To improve the characterization of the allergenic profile of yellow mustard seeds by reporting the identification and biochemical characterization of an 11S globulin as a new major allergen. METHODS: Mustard seed proteins were separated using size exclusion and ion-exchange chromatographic columns, sodium dodecyl sulfate-polyacrylamide gel electrophoresis, and 2-dimensional polyacrylamide gel electrophoresis. Separation of different polypeptide chains was achieved by reverse-phase high-performance liquid chromatography. Mass spectrometry after tryptic digestion and Edman degradation were used to determine amino acid sequences of peptides. IgE binding assays were performed with 13 serum samples from mustard allergic patients in immunoblotting and enzyme-linked immunosorbent inhibition assays. RESULTS: A protein of 51 kDa was recognized as a major allergen by patients allergic to mustard and called Sin a 2. The allergen was dissociated in 2 chains of 36 and 23 kDa, which also bound IgE. N-terminal end and internal amino acid sequences allowed identification of the new allergen as a seed storage 11S globulin belonging to the Cupin super family. Purified allergen was able to inhibit the IgE binding of sera from allergic patients to mustard seeds extract in up to 55% of the responses. CONCLUSIONS: An 11S globulin storage protein has been isolated and identified as a novel major allergen of mustard seeds.


Subject(s)
Allergens/isolation & purification , Mustard Plant , Plant Proteins/isolation & purification , Adolescent , Adult , Allergens/adverse effects , Allergens/immunology , Amino Acid Sequence , Antigen-Antibody Reactions , Antigens, Plant , Asthma/blood , Asthma/etiology , Electrophoresis, Polyacrylamide Gel , Female , Humans , Immune Sera/immunology , Immunoglobulin E/biosynthesis , Immunoglobulin E/immunology , Male , Middle Aged , Molecular Sequence Data , Molecular Weight , Mustard Plant/adverse effects , Plant Proteins/adverse effects , Plant Proteins/immunology , Rhinitis/blood , Rhinitis/etiology , Seed Storage Proteins , Seeds/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...