Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.743
1.
Vopr Pitan ; 93(2): 31-40, 2024.
Article Ru | MEDLINE | ID: mdl-38809797

The development of innovative ingredients of specialized formula for children with intolerance to cow's milk proteins requires accounting the influence of the protein component on the allergic sensitivity. The aim of the research was to study the effect of lactoferrin (LF) from cow colostrum, cow's milk protein hydrolysate (CMPH) and mare's milk protein hydrolysate (MMPH) on the severity of the systemic anaphylaxis reaction, the levels of specific IgG antibodies and cytokines in rats parenterally sensitized with ovalbumin (OVA). Material and methods. The experiment was carried out on 4 groups of 26 male Wistar rats, which were sensitized intraperitoneally with chicken egg OVA and a systemic anaphylaxis reaction was induced on the day 29 by intravenous administration of a challenge dose of the antigen (6 mg per kg body weight). LF, CMPH and MMPH were introduced into the diet in doses of 1.4-2 g/kg body weight per day (on an average 1.59±0.04, 1.53±0.05 and 1.48±0.05 g per kg body weight respectively). The content of IgG antibodies in the blood serum was determined by an indirect ELISA; the levels of cytokines IL-1α, IL-1ß, IL-2, IL-4, IL-5, IL-6, IL-10, IL-12(p70), IL-13, GM-CSF, IFN-γ and TNF-α were detected by multiplex immunoassay. Results. Dietary LF did not have a significant effect on the severity of active anaphylactic shock (AAS), concentrations of antibodies and cytokines in the blood of animals. As a result of CMPH consumption, there were no significant changes in AAS severity and IgG antibodies to OVA but significant increase in TNF-α level was observed as well as a significant decrease in IL-1α (p<0.05). In animals receiving MMPH, there were also no significant changes in the severity of AAS, but a 1.9-fold decrease in the level of IgG antibodies to OVA (p<0.001) was noticed along with a significant increase in IL-12(p70) (p<0.05) and IL-10 (p<0.10) level. Conclusion. Cosumption of LF by sensitized rats didn't significantly affect their anaphylactic sensitivity and cytokine profile, while CMPH intake induced some signs of pro-inflammatory processes. Consumption of MMPH was accompanied by the formation of an anti-inflammatory cytokine profile, which corresponds to a decrease in the intensity of the humoral immune response to the model allergen. Differences in the effects of two hydrolisates, which have similar degrees of hydrolysis, may be associated with the specific composition of glycopeptides formed during the enzymatic cleavage of milk protein produced by these two species of dairy animals.


Anaphylaxis , Cytokines , Lactoferrin , Protein Hydrolysates , Rats, Wistar , Animals , Rats , Anaphylaxis/immunology , Anaphylaxis/chemically induced , Male , Cytokines/blood , Cytokines/metabolism , Lactoferrin/pharmacology , Cattle , Protein Hydrolysates/pharmacology , Horses , Milk/chemistry , Immunoglobulin G/blood , Milk Hypersensitivity/immunology , Milk Hypersensitivity/blood , Female
2.
Vopr Pitan ; 93(2): 41-51, 2024.
Article Ru | MEDLINE | ID: mdl-38809798

The improvement of the novel foods' safety assessment algorithms is currently one of the food hygiene significant areas. Within the studying of Hermetia illucens insects' effect, the standard in vivo allergological research integrated in the protocol of medical and biological evaluation of genetically modified food has been used. The protocol was supplemented with cytokine profile indicators and pathomorphologic characteristics of immunocompetent organs' lymphoid tissue. The purpose of the research was to study the effect of black soldier fly (Hermetia illucens) larvae biomass on the rats' immune status in the experiment on the induced anaphylactic shock model. Material and methods. The effect of black soldier fly (Hermetia illucens) larvae biomass was studied in a 29-day experiment on growing (43-72 days of life) male Wistar rats fed with Hermetia illucens biomass - main group (n=29) and semi-synthetic casein diet - control group (n=29). The complex assessment of allergenic potential of Hermetia illucens biomass was carried out in the experiment on the induced anaphylactic shock model in Wistar rats. An expanded pool of immune status indicators was studied including active anaphylactic shock severity (lethality, number of severe anaphylaxis reactions, anaphylactic index); cytokine profile (content of proinflammatory and anti-inflammatory cytokines, as well as regulators of cellular and humoral immune response); IgG1 and IgG4 level before and after administration of ovalbumin permissive dose (4 mg/kg b.w.). In addition to this pathomorphologic characteristics of lymphoid tissue of the main immunocompetent organs (thymus, spleen, Payer's patches) have been obtained. Results. The significant systemic anaphylaxis reaction decrease in the main group has been shown. Comparative assessment of the serum cytokines (GM-CSF, IFN-γ, IL-10, IL-12(p70), IL-13, IL-1α, IL-1ß, IL-2, IL-4, IL-5, IL-6, TNF-α) as well as the level of immunoglobulins of the IgG1, IgG4 class before and after administration of ovalbumin permissive dose did not reveal significant differences in rats of the control and main groups. In the main group, there was a decrease in blood serum proallergic cytokines: the level of IL-4 reduced by 1.3 fold, IL-10 - 1.1 and IL-13 - 1.2 fold (p>0.05), and in animals with mild anaphylactic reaction - by 1.8, 1.4 and 1.4 times, respectively (p>0.05). The morphologic studies of the immune system organs showed no intergroup differences. Conclusion. Thus, allergological studies of black soldier fly (Hermetia illucens) larvae in the experiment with the use of systemic anaphylaxis rat model and determination of immune status indicators (anaphylactic shock severity, cytokine profile, IgG1 and IgG4 level, morphologic structure of immunocompetent organs) did not reveal any allergenic effect of the studied product.


Anaphylaxis , Cytokines , Larva , Rats, Wistar , Animals , Rats , Male , Larva/immunology , Anaphylaxis/immunology , Anaphylaxis/chemically induced , Cytokines/metabolism , Cytokines/immunology , Biomass , Simuliidae/immunology , Diptera/immunology , Diptera/growth & development
3.
Allergol Immunopathol (Madr) ; 52(3): 73-77, 2024.
Article En | MEDLINE | ID: mdl-38721958

For the first time 15 years ago, tablet allergen immunotherapy (T-AIT) formulations were approved by regulatory agencies for treating allergic rhinitis caused by grass pollen in adults and children aged >5 years. Extensive evidences existed about effectiveness and safety of AIT. However, the safety profile is particularly compelling in children. Generally, T-AIT causes local reactions, mostly in the oral cavity, that are usually mild-to-moderate and often self-resolving. However, systemic allergic reactions are also observed with T-AIT, anaphylaxis representing the most fearsome adverse event, considering that it occurs in subjects treated for allergic rhinitis. Therefore, we conducted a literature search of patients reporting anaphylaxis because of T-AIT. Nine cases of anaphylactic reactions were reported in literature. Notably, no death was reported using T-AIT. This outcome was very important as it underscored the substantial safety of T-AIT. However, T-AIT deserves careful attention, mainly in the pediatric population. In this regard, after the first report of anaphylactic reaction at the first administration of T-AIT, manufacturers recommended that the first dose should be administered in a medical facility in the presence of staff with experience in managing anaphylaxis and the patient should be observed for at least 30 min. Interestingly, reported anaphylactic reactions were due to grass pollen extracts, with no report concerning other allergen extracts. However, it is relevant to note that anaphylactic reactions because of T-AIT are not reported in recent years.


Allergens , Anaphylaxis , Desensitization, Immunologic , Tablets , Humans , Anaphylaxis/therapy , Anaphylaxis/etiology , Anaphylaxis/immunology , Desensitization, Immunologic/methods , Desensitization, Immunologic/adverse effects , Allergens/immunology , Allergens/administration & dosage , Allergens/adverse effects , Child , Pollen/immunology , Pollen/adverse effects , Poaceae/immunology , Poaceae/adverse effects , Rhinitis, Allergic, Seasonal/therapy , Rhinitis, Allergic, Seasonal/immunology , Adult , Rhinitis, Allergic/therapy , Rhinitis, Allergic/immunology , Child, Preschool
4.
Front Immunol ; 15: 1392099, 2024.
Article En | MEDLINE | ID: mdl-38686384

Background: Asparaginase (ASNase) is a crucial part of acute leukemia treatment, but immune responses to the agent can reduce its effectiveness and increase the risk of relapse. Currently, no reliable and validated biomarker predicts ASNase-induced hypersensitivity reactions during therapy. We aimed to identify predictive biomarkers and determine immune cells responsible for anaphylaxis using a murine model of ASNase hypersensitivity. Methods: Our preclinical study uses a murine model to investigate predictive biomarkers of ASNase anaphylaxis, including anti-ASNase antibody responses, immune complex (IC) levels, ASNase-specific binding to leukocytes or basophils, and basophil activation. Results: Our results indicate that mice immunized to ASNase exhibited dynamic IgM, IgG, and IgE antibody responses. The severity of ASNase-induced anaphylaxis was found to be correlated with levels of IgG and IgE, but not IgM. Basophils from immunized mice were able to recognize and activate in response to ASNase ex vivo, and the extent of recognition and activation also correlated with the severity of anaphylaxis observed. Using a multivariable model that included all biomarkers significantly associated with anaphylaxis, independent predictors of ASNase-induced hypersensitivity reactions were found to be ASNase IC levels and ASNase-specific binding to leukocytes or basophils. Consistent with our multivariable analysis, we found that basophil depletion significantly protected mice from ASNase-induced hypersensitivity reactions, supporting that basophils are essential and can be used as a predictive marker of ASNase-induced anaphylaxis. Conclusions: Our study demonstrates the need for using tools that can detect both IC- and IgE-mediated hypersensitivity reactions to mitigate the risk of ASNase-induced hypersensitivity reactions during treatment.


Anaphylaxis , Asparaginase , Basophils , Drug Hypersensitivity , Immunoglobulin E , Animals , Asparaginase/adverse effects , Asparaginase/immunology , Basophils/immunology , Basophils/metabolism , Mice , Drug Hypersensitivity/immunology , Drug Hypersensitivity/diagnosis , Anaphylaxis/immunology , Anaphylaxis/chemically induced , Immunoglobulin E/immunology , Immunoglobulin E/blood , Female , Disease Models, Animal , Biomarkers , Immunoglobulin G/immunology , Immunoglobulin G/blood , Antineoplastic Agents/adverse effects
5.
J Food Sci ; 89(5): 3037-3047, 2024 May.
Article En | MEDLINE | ID: mdl-38563099

Bovine casein is a major allergen present in cow milk to induce anaphylaxis. In this study, the potential allergenicity of enzymatically hydrolyzed casein (HC) was evaluated based on in vitro and in vivo. The results showed that Alcalase and Protamex treatment (AT, PT) reduced the potential allergenicity of CN, with the greatest reductions of 68.25% and 50.75%, respectively. In addition, in vivo results showed that HC effectively alleviated allergic response symptoms of Balb/c mice; a significant tendency toward decreased serum IgG1 and mast cell tryptase levels was observed, accompanied by a decrease of Th2-associated IL-4, IL-5, and IL-13 and an increase of IFN-γ levels in spleen. Moreover, the inflammation of the lung, jejunum, and ileum was remarkably ameliorated. The findings indicated that HC induced a shift toward Th1 response and maintained the Th1/Th2 immune balance. Importantly, our results provide the basis for the production of hypoallergenic dairy products.


Allergens , Caseins , Mice, Inbred BALB C , Th2 Cells , Animals , Mice , Caseins/immunology , Allergens/immunology , Female , Th2 Cells/immunology , Hydrolysis , Immunoglobulin G/blood , Disease Models, Animal , Cattle , Spleen/immunology , Milk Hypersensitivity/immunology , Interferon-gamma/metabolism , Th1 Cells/immunology , Interleukin-4/metabolism , Tryptases/metabolism , Cytokines/metabolism , Jejunum/immunology , Milk/immunology , Milk/chemistry , Interleukin-13/immunology , Interleukin-13/metabolism , Anaphylaxis/immunology , Anaphylaxis/chemically induced , Anaphylaxis/prevention & control , Interleukin-5/immunology
6.
Curr Opin Allergy Clin Immunol ; 24(3): 153-159, 2024 06 01.
Article En | MEDLINE | ID: mdl-38538146

PURPOSE OF REVIEW: Food allergy is a growing health problem that affects both patients and society in multiple ways. Despite the emergence of novel diagnostic tools, such as component-resolved diagnostics (CRD) and basophil activation tests (BAT), oral food challenge (OFC) still plays an indispensable role in the management of food allergies. This review aimed to highlight the indications and safety concerns of conducting an OFC and to provide insights into post-OFC management based on recent findings. RECENT FINDINGS: Standardized OFC protocols have regional diversification, especially in Japan and Western countries. Recent studies suggested that the interval between doses should be at least more than an hour. Furthermore, applying a stepwise method tailored to the patient's specific immunoglobulin E level and history of anaphylaxis seems to mitigate these risks. Recent surveys have shown that, following a positive OFC, options other than strict avoidance are also selected. SUMMARY: OFC serves diverse purposes, yet the risks it carries warrant caution. The stepwise protocol appears promising for its safety. Subthreshold consumption following OFC shows potential; however, further research on its efficacy and safety is required. Management following OFC should be tailored and well discussed between clinicians and patients.


Allergens , Food Hypersensitivity , Humans , Food Hypersensitivity/immunology , Food Hypersensitivity/therapy , Food Hypersensitivity/diagnosis , Administration, Oral , Allergens/immunology , Allergens/administration & dosage , Anaphylaxis/immunology , Anaphylaxis/diagnosis , Anaphylaxis/prevention & control , Anaphylaxis/therapy , Immunoglobulin E/immunology , Immunoglobulin E/blood , Food/adverse effects
7.
Int Arch Allergy Immunol ; 185(5): 456-459, 2024.
Article En | MEDLINE | ID: mdl-38412847

INTRODUCTION: Changes in the cytokine profile from type 2 to type 1 together with the induction of regulatory cells are expected during hymenoptera venom immunotherapy (VIT). The present study was aimed to investigate the changes in type 1, type 2, and regulatory cytokines induced by a Vespula spp. VIT in patients with anaphylaxis to Vespa velutina. METHODS: Twenty consecutive patients with anaphylaxis due to Vespa velutina were treated with Vespula spp. VIT. Serum cytokines (IL-4, IL-5, IL-10, IL-13, and IFN-É£) were measured at baseline, 6, and 12 months after starting VIT. RESULTS: A significant increase in serum IFN-y was detected after 6 and 12 months of VIT. An increase in serum IL-10 and a decrease in IL-5 were observed after 12 months. IL-4 was undetectable all along the study, and an unexpected increase of IL-13 was present at 12 months of treatment. CONCLUSION: Vespula spp. VIT seems to be able to induce a shift to type 1 cytokine production measured through IFN-y levels and IL-10 production after, at least, 6 and 12 months of VIT, respectively.


Anaphylaxis , Cytokines , Desensitization, Immunologic , Wasp Venoms , Wasps , Humans , Anaphylaxis/immunology , Anaphylaxis/therapy , Anaphylaxis/etiology , Cytokines/metabolism , Cytokines/blood , Male , Female , Adult , Animals , Desensitization, Immunologic/methods , Wasp Venoms/immunology , Wasps/immunology , Middle Aged , Insect Bites and Stings/immunology , Insect Bites and Stings/therapy , Young Adult , Allergens/immunology
8.
Allergy ; 79(6): 1440-1454, 2024 Jun.
Article En | MEDLINE | ID: mdl-38193233

The recent recognition of a syndrome of tick-acquired mammalian meat allergy has transformed the previously held view that mammalian meat is an uncommon allergen. The syndrome, mediated by IgE antibodies against the oligosaccharide galactose-alpha-1,3-galactose (alpha-gal), can also involve reactions to visceral organs, dairy, gelatin and other products, including medications sourced from non-primate mammals. Thus, fittingly, this allergic disorder is now called the alpha-gal syndrome (AGS). The syndrome is strikingly regional, reflecting the important role of tick bites in sensitization, and is more common in demographic groups at risk of tick exposure. Reactions in AGS are delayed, often by 2-6 h after ingestion of mammalian meat. In addition to classic allergic symptomatology such as urticaria and anaphylaxis, AGS is increasingly recognized as a cause of isolated gastrointestinal morbidity and alpha-gal sensitization has also been linked with cardiovascular disease. The unusual link with tick bites may be explained by the fact that allergic cells and mediators are mobilized to the site of tick bites and play a role in resistance against ticks and tick-borne infections. IgE directed to alpha-gal is likely an incidental consequence of what is otherwise an adaptive immune strategy for host defense against endo- and ectoparasites, including ticks.


Anaphylaxis , Food Hypersensitivity , Immunoglobulin E , Tick Bites , Tick-Borne Diseases , Urticaria , Animals , Humans , Allergens/immunology , Anaphylaxis/immunology , Anaphylaxis/etiology , Anaphylaxis/diagnosis , Disaccharides/immunology , Food Hypersensitivity/immunology , Food Hypersensitivity/etiology , Immunoglobulin E/immunology , Mammals/immunology , Meat/adverse effects , Syndrome , Tick Bites/immunology , Tick Bites/complications , Ticks/immunology , Urticaria/immunology , Urticaria/etiology , Tick-Borne Diseases/immunology
9.
Hum Vaccin Immunother ; 19(2): 2246542, 2023 08 01.
Article En | MEDLINE | ID: mdl-37614152

A good safety and immunogenicity profile was reported in Phase I and II clinical trials of inactivated SARS-CoV-2 vaccines. Here, we report two cases associated with vaccine-associated adverse events, including one patient with fever and another with anaphylactic shock resulting from inactivated SARS-CoV-2 vaccination. Cell sub-types and the importance of genetic characteristics were assessed using single-cell mRNA sequencing and machine learning. Overall, the patient with fever showed a significant increase in the numbers of cytotoxic CD8 T cells and MKI67high CD8 T cells. A potential concurrent infection with the Epstein-Barr virus enhanced interferon type I responses to vaccination against the virus. STAT1, E2F1, YBX1, and E2F7 played a key role in the transcription regulation of MKI67high CD8 T cells. In contrast, the patient with allergic shock displayed predominant increases in the numbers of S100A9high monocytes, activated CD4 T cells, and PPBPhigh megakaryocytes. The decision tree showed that LYZ and S100A8 in S100A9high monocytes contributed to the degranulation of neutrophils and activation of neutrophils involved in allergic shock. PPBP and PF4 were major contributors to platelet degranulation. These findings highlight the diversity of adverse reactions following inactivated SARS-CoV-2 vaccination and show the emerging role of cellular subtypes and central genes in vaccine-associated adverse reactions.


The identification of cell sub-types may help in the diagnosis of COVID-19 vaccine-related adverse events.COVID-19 vaccination-related acute pulmonary edema may induce a higher risk of thrombosis.The long-term fever after vaccination may attribute to the excessive type I interferon responses.


COVID-19 Vaccines , Humans , Male , Female , Adult , COVID-19 Vaccines/adverse effects , Fever/immunology , Fever/pathology , Pulmonary Edema/immunology , Pulmonary Edema/pathology , CD8-Positive T-Lymphocytes/cytology , Cell Proliferation , Megakaryocytes/pathology , Single-Cell Gene Expression Analysis , B-Lymphocytes/cytology , Monocytes/cytology , Anaphylaxis/immunology , Anaphylaxis/pathology
10.
Pediatr Allergy Immunol Pulmonol ; 36(3): 90-93, 2023 09.
Article En | MEDLINE | ID: mdl-37433203

Objective: Cases of cow's milk allergy (CMA) who reacted to measles or measles, mumps, and rubella (MMR) vaccines containing alpha-lactalbumin have been reported. The purpose of this study was to assess patients with CMA who received measles or MMR vaccines containing alpha-lactalbumin, as well as the characteristics of those who developed reactions to these vaccines. Study Design: Patients followed up in the allergy clinic for CMA and who received measles or MMR vaccines containing alpha-lactalbumin at 9 or 12 months of age were included in the study, and their characteristics were analyzed retrospectively from the hospital registry system. Results: Forty-nine patients were included in the study. Six patients received the measles vaccine, whereas 43 patients received the MMR vaccine containing alpha-lactalbumin. Vaccine skin tests were performed on these 6 patients. One patient had a positive intradermal test, so an alternative vaccine not containing alpha-lactalbumin was administered. The other 5 patients were vaccinated, and no reaction was observed. Anaphylaxis was observed in 3 of 43 patients who received the MMR vaccine containing alpha-lactalbumin. In all of these patients, the first reaction to dairy products was anaphylaxis. In 2 of those patients, cow's milk-specific IgE (spIgE) levels were >100 kU/L, and alpha-lactalbumin-spIgE levels were also high at 97 and 90 kU/L. The third patient's cow's milk-spIgE level was 15.9 kU/L, whereas the alpha-lactalbumin-spIgE level was 0.04 kU/L. Conclusion: Especially in patients with an initial reaction of anaphylaxis to dairy products and high cow's milk-spIgE levels, the risk of reaction is high with the MMR vaccine.


Anaphylaxis , Lactalbumin , Measles-Mumps-Rubella Vaccine , Milk Hypersensitivity , Milk , Animals , Cattle , Female , Anaphylaxis/immunology , Immunoglobulin E , Lactalbumin/adverse effects , Lactalbumin/immunology , Measles-Mumps-Rubella Vaccine/adverse effects , Measles-Mumps-Rubella Vaccine/immunology , Milk Hypersensitivity/immunology , Retrospective Studies , Humans , Milk/adverse effects , Milk/immunology
11.
Allergol. immunopatol ; 51(3): 8-14, 01 mayo 2023. tab
Article En | IBECS | ID: ibc-219808

Background: Food allergy (FA), hence the incidence of food anaphylaxis, is a public health problem that has increased in recent years. There are still no biomarkers for patients with FA to predict severe allergic reactions such as anaphylaxis. Objective: There is limited information on whether regulatory T (Treg) cell levels are a biomarker that predicts clinical severity in cases presenting with FA, and which patients are at a greater risk for anaphylaxis. Methods: A total of 70 children were included in the study: 25 who had IgE-mediated cow’s milk protein allergy (CMPA) and presented with non-anaphylactic symptoms (FA/A−), 16 who had IgE-mediated CMPA and presented with anaphylaxis (FA/A+) (a total of 41 FA cases), and a control group consisting of 29 children without FA. The study was conducted by performing CD4+CD25+CD127loFOXP3+ cell flow cytometric analysis during resting at least 2 weeks after the elimination diet to FA subjects. Results: When the FA group was compared with healthy control subjects, CD4+CD25+CD127loFOXP3+ cell rates were found to be significantly lower in the FA group (p < 0.001). When the FA/A− and FA/A+ groups and the control group were compared in terms of CD4+CD25+CD127loFOXP3+ cell ratios, they were significantly lower in the FA/A− and FA/A+ groups compared to the control group (p < 0.001). Conclusions: Although there was no significant difference between the FA/A+ group and the FA/A− group in terms of CD4+CD25+CD127loFOXP3+ cells, our study is important, as it is the first pediatric study we know to investigate whether CD4+CD25+CD127loFOXP3+ cells in FA predict anaphylaxis (AU)


Humans , Male , Female , Infant , Child, Preschool , Forkhead Box Protein O3/blood , Milk Hypersensitivity/diagnosis , Milk Hypersensitivity/immunology , Anaphylaxis/etiology , Anaphylaxis/immunology , /enzymology , Milk Hypersensitivity/complications , Biomarkers/blood , CD4 Antigens/immunology , Interleukin-2 Receptor alpha Subunit/immunology
12.
Proc Natl Acad Sci U S A ; 119(28): e2123212119, 2022 07 12.
Article En | MEDLINE | ID: mdl-35867757

Humans lack the capacity to produce the Galα1-3Galß1-4GlcNAc (α-gal) glycan, and produce anti-α-gal antibodies upon exposure to the carbohydrate on a diverse set of immunogens, including commensal gut bacteria, malaria parasites, cetuximab, and tick proteins. Here we use X-ray crystallographic analysis of antibodies from α-gal knockout mice and humans in complex with the glycan to reveal a common binding motif, centered on a germline-encoded tryptophan residue at Kabat position 33 (W33) of the complementarity-determining region of the variable heavy chain (CDRH1). Immunoglobulin sequencing of anti-α-gal B cells in healthy humans and tick-induced mammalian meat anaphylaxis patients revealed preferential use of heavy chain germline IGHV3-7, encoding W33, among an otherwise highly polyclonal antibody response. Antigen binding was critically dependent on the presence of the germline-encoded W33 residue for all of the analyzed antibodies; moreover, introduction of the W33 motif into naive IGHV3-23 antibody phage libraries enabled the rapid selection of α-gal binders. Our results outline structural and genetic factors that shape the human anti-α-galactosyl antibody response, and provide a framework for future therapeutics development.


Anaphylaxis , Antibodies , Food Hypersensitivity , Immunoglobulin Heavy Chains , Immunoglobulin Variable Region , Tick-Borne Diseases , Trisaccharides , Anaphylaxis/immunology , Animals , Antibodies/chemistry , Antibodies/genetics , Antibody Formation/genetics , Antigen-Antibody Complex/chemistry , Crystallography, X-Ray , Food Hypersensitivity/immunology , Humans , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/immunology , Mice , Mice, Knockout , Peptide Library , Protein Conformation , Tick-Borne Diseases/immunology , Trisaccharides/genetics , Trisaccharides/immunology
14.
Sci Rep ; 12(1): 2797, 2022 02 18.
Article En | MEDLINE | ID: mdl-35181694

To investigate food allergy-tolerance mechanisms induced through allergen-specific immunotherapy we used RNA-Sequencing to measure gene expression in lymph-node-derived dendritic cells from Pru p 3-anaphylactic mice after immunotherapy with glycodendropeptides at 2 nM and 5 nM, leading to permanent tolerance and short-term desensitization, respectively. Gene expression was also measured in mice receiving no immunotherapy (anaphylaxis); and in which anaphylaxis could never occur (antigen-only). Compared to anaphylaxis, the antigen-only group showed the greatest number of expression-changes (411), followed by tolerant (186) and desensitized (119). Only 29 genes changed in all groups, including Il12b, Cebpb and Ifngr1. The desensitized group showed enrichment for genes related to chronic inflammatory response, secretory granule, and regulation of interleukin-12 production; the tolerant group showed genes related to cytokine receptor activity and glucocorticoid receptor binding, suggesting distinct pathways for similar outcomes. We identified genes and processes potentially involved in the restoration of long-term tolerance via allergen-specific immunotherapy, representing potential prognostic biomarkers.


CCAAT-Enhancer-Binding Protein-beta/genetics , Desensitization, Immunologic , Immune Tolerance/genetics , Interleukin-12 Subunit p40/genetics , Receptors, Interferon/genetics , Allergens/immunology , Allergens/pharmacology , Anaphylaxis/genetics , Anaphylaxis/immunology , Animals , Antigens, Plant/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Food Hypersensitivity/genetics , Food Hypersensitivity/immunology , Gene Expression Regulation/drug effects , Glycopeptides/pharmacology , Humans , Interleukin-12/genetics , Lymph Nodes/immunology , Mice , Plant Proteins/pharmacology , RNA-Seq , Interferon gamma Receptor
17.
J Allergy Clin Immunol ; 149(1): 156-167.e7, 2022 01.
Article En | MEDLINE | ID: mdl-34051221

BACKGROUND: Binding IgE to a cognate allergen causes aggregation of Fcε receptor I (FcεRI) in mast cells, resulting in activation of receptor-associated Src family tyrosine kinases, including Lyn and Syk. Protein tyrosine phosphatase, receptor type C (PTPRC), also known as CD45, has emerged as a positive regulator of FcεRI signaling by dephosphorylation of the inhibitory tyrosine of Lyn. OBJECTIVE: Sirtuin 6 (Sirt6), a NAD+-dependent deacetylase, exhibits an anti-inflammatory property. It remains to be determined, however, whether Sirt6 attenuates mast cell-associated diseases, including anaphylaxis. METHODS: FcεRI signaling and mast cell degranulation were measured after IgE cross-linking in murine bone marrow-derived mast cells (BMMCs) and human cord blood-derived mast cells. To investigate the function of Sirt6 in mast cell activation in vivo, we used mast cell-dependent animal models of passive systemic anaphylaxis (PSA) and passive cutaneous anaphylaxis (PCA). RESULTS: Sirt6-deficient BMMCs augmented IgE-FcεRI-mediated signaling and degranulation compared to wild-type BMMCs. Reconstitution of mast cell-deficient KitW-sh/W-sh mice with BMMCs received from Sirt6 knockout mice developed more severe PSA and PCA compared to mice engrafted with wild-type BMMCs. Similarly, genetic overexpression or pharmacologic activation of Sirt6 suppressed mast cell degranulation and blunted responses to PCA. Mechanistically, Sirt6 deficiency increased PTPRC transcription via acetylating histone H3, leading to enhanced aggregation of FcεRI in BMMCs. Finally, we recapitulated the Sirt6 regulation of PTPRC and FcεRI signaling in human mast cells. CONCLUSIONS: Sirt6 acts as a negative regulator of FcεRI signaling cascade in mast cells by suppressing PTPRC transcription. Activation of Sirt6 may therefore represent a promising and novel therapeutic strategy for anaphylaxis.


Anaphylaxis/immunology , Mast Cells/immunology , Receptors, IgE/immunology , Sirtuins/immunology , Animals , Bone Marrow Cells/cytology , Fetal Blood/cytology , Humans , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Sirtuins/genetics
18.
Front Immunol ; 12: 688930, 2021.
Article En | MEDLINE | ID: mdl-34867939

Acute anaphylaxis to small molecule drugs is largely considered to be antibody-mediated with immunogloblin E (IgE) and mast cell activation being key. More recently, a role for drug-reactive immunoglobulin G (IgG) with neutrophil activation has also been suggested, at least in reactions to neuromuscular blocking agents (NMBAs). However, the mast cell receptor MRGPRX2 has also been highlighted as a possible triggering mechanism in acute anaphylaxis to many clinically used drugs. Significantly, MRGPRX2 activation is not dependent upon the presence of drug-recognising antibody. Given the reasonable assumption that MRGPRX2 is expressed in all individuals, the corollary of this is that in theory, anybody could respond detrimentally to triggering drugs (recently suggested to be around 20% of a drug-like compound library). But this clearly is not the case, as the incidence of acute drug-induced anaphylaxis is very low. In this mini-review we consider antibody-dependent and -independent mechanisms of mast cell activation by small molecule drugs with a focus on the MRGPRX2 pathway. Moreover, as a juxtaposition to these adverse drug actions, we consider how increased understanding of the role of MRGPRX2 in anaphylaxis is important for future drug development and can complement exploration of this receptor as a drug target in broader clinical settings.


Anaphylaxis/immunology , Nerve Tissue Proteins/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/immunology , Anaphylaxis/etiology , Anaphylaxis/therapy , Drug Hypersensitivity/etiology , Drug Hypersensitivity/immunology , Drug Hypersensitivity/therapy , Drug-Related Side Effects and Adverse Reactions/etiology , Drug-Related Side Effects and Adverse Reactions/immunology , Drug-Related Side Effects and Adverse Reactions/therapy , Gene Expression , Humans , Mast Cell Activation Disorders/etiology , Mast Cell Activation Disorders/immunology , Mast Cell Activation Disorders/therapy , Mast Cells/drug effects , Mast Cells/immunology , Models, Immunological , Nerve Tissue Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/genetics
19.
Anal Biochem ; 633: 114392, 2021 11 15.
Article En | MEDLINE | ID: mdl-34597615

Mas-related G-protein-coupled receptor X2 (MRGPRX2) has recently been reported to be associated with anaphylaxis. Detection of MRGPRX2 levels in human peripheral blood might serve as a powerful tool for predicting the predisposition of patients to anaphylactic reactions. For rapid measurement of MRGPRX2, we established a paper-based double-antibody sandwich enzyme-linked immunosorbent assay (ELISA) using mouse monoclonal antibody and horseradish peroxidase (HRP)-labelled rabbit polyclonal antibody as capture antibody and detection antibody, respectively. We avoided chemical functionalization of the cellulose paper by introducing bovine serum albumin (BSA) to provide COOH and NH2 groups for covalent immobilization of the capture antibody. Through amide condensation, a two-layer immobilization strategy was applied with BSA-BSA and BSA-capture antibody networks as the first and second layers, respectively. This strategy improved the quantity, activity and stability of the immobilized antibody. We then established a paper-based ELISA to detect MRGPRX2 in human peripheral blood. Our method is less laborious, easier to implement, and more cost-effective than conventional ELISA, while offering similar sensitivity, specificity, and accuracy. Therefore, it could serve as an innovative clinical point-of-care diagnostic tool, especially in areas that lack advanced clinical equipment.


Anaphylaxis/blood , Enzyme-Linked Immunosorbent Assay , Nerve Tissue Proteins/blood , Paper , Receptors, G-Protein-Coupled/blood , Receptors, Neuropeptide/blood , Anaphylaxis/immunology , Humans , Nerve Tissue Proteins/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, Neuropeptide/immunology
20.
Front Immunol ; 12: 668962, 2021.
Article En | MEDLINE | ID: mdl-34385999

Neuromuscular blocking agents (NMBAs) like atracurium and rocuronium as well as fluoroquinolones (FQs) cause mast cell-mediated anaphylaxis by activating Mas-related G protein-coupled receptor X2 (MRGPRX2), but many questions remain unanswered. Here, we address three of them, namely whether primary human mast cells show similar activation by these drugs as murine mast cells and mast cell lines, how sugammadex protects from atracurium-induced MRGPRX2-mediated mast cell activation, and why some but not all patients treated with rocuronium develop anaphylaxis. We used peripheral blood-derived cultured mast cells from healthy donors and patients, assessed mast cell activation and degranulation by quantifying intracellular calcium and CD63 expression, respectively, and made use of MRGPRX2-silencing, via electroporation with Dicer-substrate small interfering RNAs, and single cell flow cytometric analyses. Atracurium, ciprofloxacin, and levofloxacin activated and degranulated primary human mast cells, but only MRGPRX2-positive and not MRGPRX2-negative or -silenced mast cells. Sugammadex attenuated the atracurium-induced and MRGPRX2-mediated activation and degranulation of human mast cells by reducing free atracurium levels. The mast cells of patients with IgE-independent anaphylaxis to rocuronium were similar, in their MRGPRX2 expression and function, to those of patients with IgE-mediated anaphylaxis. These findings further improve our understanding of the role and relevance of MRGPRX2-driven mast cell activation in anaphylactic reactions to NMBAs and FQs and may help to improve their prediction, prevention, and treatment.


Anaphylaxis/chemically induced , Anti-Bacterial Agents/toxicity , Cell Degranulation/drug effects , Drug Hypersensitivity/etiology , Mast Cells/drug effects , Nerve Tissue Proteins/metabolism , Neuromuscular Nondepolarizing Agents/toxicity , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Anaphylaxis/immunology , Anaphylaxis/metabolism , Atracurium/toxicity , Calcium Signaling/drug effects , Cells, Cultured , Ciprofloxacin/toxicity , Drug Hypersensitivity/immunology , Drug Hypersensitivity/metabolism , Humans , Immunoglobulin E/immunology , Levofloxacin/toxicity , Mast Cells/immunology , Mast Cells/metabolism , Nerve Tissue Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/genetics , Rocuronium/toxicity , Time Factors
...