Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.329
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 42(1): 401-425, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38360544

ABSTRACT

IgE-mediated food allergy (IgE-FA) occurs due to a breakdown in immune tolerance that leads to a detrimental type 2 helper T cell (TH2) adaptive immune response. While the processes governing this loss of tolerance are incompletely understood, several host-related and environmental factors impacting the risk of IgE-FA development have been identified. Mounting evidence supports the role of an impaired epithelial barrier in the development of IgE-FA, with exposure of allergens through damaged skin and gut epithelium leading to the aberrant production of alarmins and activation of TH2-type allergic inflammation. The treatment of IgE-FA has historically been avoidance with acute management of allergic reactions, but advances in allergen-specific immunotherapy and the development of biologics and other novel therapeutics are rapidly changing the landscape of food allergy treatment. Here, we discuss the pathogenesis and immunobiology of IgE-FA in addition to its diagnosis, prognosis, and treatment.


Subject(s)
Allergens , Food Hypersensitivity , Immunoglobulin E , Humans , Food Hypersensitivity/therapy , Food Hypersensitivity/immunology , Animals , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Allergens/immunology , Desensitization, Immunologic/methods , Th2 Cells/immunology , Immune Tolerance , Disease Susceptibility
2.
Annu Rev Immunol ; 37: 377-403, 2019 04 26.
Article in English | MEDLINE | ID: mdl-31026410

ABSTRACT

The gut-associated lymphoid tissue (GALT) faces a considerable challenge. It encounters antigens derived from an estimated 1014 commensal microbes and greater than 30 kg of food proteins yearly. It must distinguish these harmless antigens from potential pathogens and mount the appropriate host immune response. Local and systemic hyporesponsiveness to dietary antigens, classically referred to as oral tolerance, comprises a distinct complement of adaptive cellular and humoral immune responses. It is increasingly evident that a functional epithelial barrier engaged in intimate interplay with innate immune cells and the resident microbiota is critical to establishing and maintaining oral tolerance. Moreover, innate immune cells serve as a bridge between the microbiota, epithelium, and the adaptive immune system, parlaying tonic microbial stimulation into signals critical for mucosal homeostasis. Dysregulation of gut homeostasis and the subsequent disruption of tolerance therefore have clinically significant consequences for the development of food allergy.


Subject(s)
Dysbiosis/immunology , Food Hypersensitivity/immunology , Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Administration, Oral , Allergens/immunology , Animals , Food , Food Hypersensitivity/microbiology , Homeostasis , Humans , Immune Tolerance , Immunity, Innate , Intestinal Mucosa/microbiology
3.
Annu Rev Cell Dev Biol ; 36: 511-528, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32634325

ABSTRACT

Pediatric allergic disease is a significant health concern worldwide, and the prevalence of childhood eczema, asthma, allergic rhinitis, and food allergy continues to increase. Evidence to support specific interventions for the prevention of eczema, asthma, and allergic rhinitis is limited, and no consensus on prevention strategies has been reached. Randomized controlled trials investigating the prevention of food allergy via oral tolerance induction and the early introduction of allergenic foods have been successful in reducing peanut and egg allergy prevalence. Infant weaning guidelines in the United Sates were recently amended to actively encourage the introduction of peanut for prevention of peanut allergy.


Subject(s)
Food Hypersensitivity/immunology , Immune Tolerance , Animals , Child , Humans , Immunotherapy , Models, Biological , Peanut Hypersensitivity/immunology , Practice Guidelines as Topic
4.
Immunity ; 57(10): 2269-2279, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39383844

ABSTRACT

The immune system recognizes a multitude of innocuous antigens from food and intestinal commensal microbes toward which it orchestrates appropriate, non-inflammatory responses. This process requires antigen-presenting cells (APCs) that induce T cells with either regulatory or effector functions. Compromised APC function disrupts the T cell balance, leading to inflammation and dysbiosis. Although their precise identities continue to be debated, it has become clear that multiple APC lineages direct the differentiation of distinct microbiota-specific CD4+ T cell programs. Here, we review how unique APC subsets instruct T cell differentiation and function in response to microbiota and dietary antigens. These discoveries provide new opportunities to investigate T cell-APC regulatory networks controlling immune homeostasis and perturbations associated with inflammatory and allergic diseases.


Subject(s)
Antigen-Presenting Cells , Humans , Antigen-Presenting Cells/immunology , Animals , Cell Differentiation/immunology , Intestines/immunology , Homeostasis/immunology , Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Inflammation/immunology , T-Lymphocytes/immunology
5.
Immunity ; 53(6): 1202-1214.e6, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33086036

ABSTRACT

The mechanisms by which regulatory T (Treg) cells differentially control allergic and autoimmune responses remain unclear. We show that Treg cells in food allergy (FA) had decreased expression of transforming growth factor beta 1 (TGF-ß1) because of interleukin-4 (IL-4)- and signal transducer and activator of transciription-6 (STAT6)-dependent inhibition of Tgfb1 transcription. These changes were modeled by Treg cell-specific Tgfb1 monoallelic inactivation, which induced allergic dysregulation by impairing microbiota-dependent retinoic acid receptor-related orphan receptor gamma t (ROR-γt)+ Treg cell differentiation. This dysregulation was rescued by treatment with Clostridiales species, which upregulated Tgfb1 expression in Treg cells. Biallelic deficiency precipitated fatal autoimmunity with intense autoantibody production and dysregulated T follicular helper and B cell responses. These results identify a privileged role of Treg cell-derived TGF-ß1 in regulating allergy and autoimmunity at distinct checkpoints in a Tgfb1 gene dose- and microbiota-dependent manner.


Subject(s)
Autoimmunity/immunology , Hypersensitivity/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1/immunology , Adolescent , Animals , Autoimmunity/genetics , B-Lymphocytes/immunology , Cell Differentiation , Child , Child, Preschool , Food Hypersensitivity/immunology , Gene Dosage , Humans , Hypersensitivity/genetics , Immunoglobulin G/immunology , Infant , Mast Cells/immunology , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T Follicular Helper Cells/immunology , T-Lymphocytes, Regulatory/metabolism , Transcription, Genetic , Transforming Growth Factor beta1/genetics , Young Adult
6.
Immunity ; 50(5): 1262-1275.e4, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31027995

ABSTRACT

Mast cell (MC) mediator release after crosslinking of surface-bound IgE antibody by ingested antigen underlies food allergy. However, IgE antibodies are not uniformly associated with food allergy, and intestinal MC load is an important determinant. Atopic dermatitis (AD), characterized by pruritis and cutaneous sensitization to allergens, including foods, is strongly associated with food allergy. Tape stripping mouse skin, a surrogate for scratching, caused expansion and activation of small intestinal MCs, increased intestinal permeability, and promoted food anaphylaxis in sensitized mice. Tape stripping caused keratinocytes to systemically release interleukin-33 (IL-33), which synergized with intestinal tuft-cell-derived IL-25 to drive the expansion and activation of intestinal type-2 innate lymphoid cells (ILC2s). These provided IL-4, which targeted MCs to expand in the intestine. Duodenal MCs were expanded in AD. In addition to promoting cutaneous sensitization to foods, scratching may promote food anaphylaxis in AD by expanding and activating intestinal MCs.


Subject(s)
Dermatitis, Atopic/immunology , Food Hypersensitivity/immunology , Intestinal Mucosa/immunology , Lymphocytes/immunology , Mast Cells/immunology , Adolescent , Anaphylaxis/immunology , Animals , Cell Proliferation , Child , Child, Preschool , Female , Humans , Immunoglobulin E/immunology , Interleukin-13/metabolism , Interleukin-33/metabolism , Interleukin-4/metabolism , Interleukins/metabolism , Intestinal Mucosa/cytology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction/immunology , Skin/immunology , Skin/injuries
7.
Immunol Rev ; 326(1): 173-190, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39295093

ABSTRACT

The critical importance of the immunoregulatory mechanisms, which prevent adverse responses to dietary proteins is demonstrated by the consequences of their failure in two common but distinct human pathological conditions, food allergy and celiac disease. The mechanisms of tolerance to dietary proteins have been extensively studied in mouse models but the extent to which the results in mice can be extrapolated to humans remains unclear. Here, after summarizing the mechanisms known to control oral tolerance in mouse models, we discuss how the monogenic immune disorders associated with food allergy on the one hand, and celiac disease, on the other hand, represent model diseases to gain insight into the key immunoregulatory pathways that control immune responses to food antigens in humans. The spectrum of monogenic disorders, in which the dysfunction of a single gene, is strongly associated with TH2-mediated food allergy suggests an important overlap between the mechanisms that regulate TH2 and IgE responses to food antigens in humans and mice. In contrast, celiac disease provides a unique example of the link between autoimmunity and loss of tolerance to a food antigen.


Subject(s)
Celiac Disease , Dietary Proteins , Disease Models, Animal , Food Hypersensitivity , Immune Tolerance , Animals , Humans , Mice , Food Hypersensitivity/immunology , Celiac Disease/immunology , Celiac Disease/etiology , Celiac Disease/metabolism , Dietary Proteins/immunology , Dietary Proteins/metabolism , Th2 Cells/immunology , Autoimmunity , Immunoglobulin E/immunology , Immunoglobulin E/metabolism
8.
Immunol Rev ; 326(1): 35-47, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39054597

ABSTRACT

Food allergies occur due to a lack of tolerance to the proteins found in foods. While IgE- and non-IgE-mediated food allergies have different clinical manifestations, epidemiology, pathophysiology, and management, they share dysregulated T cell responses. Recent studies have shed light on the contributions of different T cell subsets to the development and persistence of different food allergic diseases. This review discusses the role of T cells in both IgE- and non-IgE-mediated food allergies and considers the potential future investigations in this context.


Subject(s)
Food Hypersensitivity , Immunoglobulin E , T-Lymphocyte Subsets , Food Hypersensitivity/immunology , Food Hypersensitivity/therapy , Humans , Animals , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Allergens/immunology , Immune Tolerance
9.
Immunol Rev ; 326(1): 8-16, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39054615

ABSTRACT

Immune tolerance to foods develops in the intestine upon food ingestion and is essential to prevent IgE-mediated food allergy and gut inflammation. In homeostasis, the intestine is a tolerogenic environment that favors the formation of food-specific Foxp3+ regulatory T cells. A tolerogenic intestinal environment depends on colonization by diverse microbiota and exposure to solid foods at a critical period in early life. These early immune responses lead to the induction of antigen-specific Foxp3+ regulatory T cells in draining mesenteric lymph nodes. These peripherally induced regulatory cells circulate and seed the lamina propria of the gut, exerting suppressive function systemically and locally in the intestine. Successful establishment of a tolerogenic intestinal environment in early life sets the stage for oral tolerance to new antigens in adult life.


Subject(s)
Food Hypersensitivity , Immune Tolerance , T-Lymphocytes, Regulatory , Animals , Humans , Antigens/immunology , Food Hypersensitivity/immunology , Forkhead Transcription Factors/metabolism , Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestines/immunology , T-Lymphocytes, Regulatory/immunology
10.
Immunol Rev ; 326(1): 66-82, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39046160

ABSTRACT

The increasing prevalence of food allergy and related pathologies in recent years has underscored the need to understand the factors affecting adverse reactions to food. Food allergy is caused when food-specific IgE triggers the release of histamine from mast cells. However, other food-specific antibody isotypes exist as well, including IgG and IgA. IgA is the main antibody isotype in the gut and mediates noninflammatory reactions to toxins, commensal bacteria, and food antigens. It has also been thought to induce tolerance to food, thus antagonizing the role of food-specific IgE. However, this has remained unclear as food-specific IgA generation is poorly understood. Particularly, the location of IgA induction, the role of T cell help, and the fates of food-specific B cells remain elusive. In this review, we outline what is known about food-specific IgA induction and highlight areas requiring further study. We also explore how knowledge of food-specific IgA induction can be informed by and subsequently contribute to our overall knowledge of gut immunity.


Subject(s)
Food Hypersensitivity , Immunoglobulin A , Humans , Animals , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Food Hypersensitivity/immunology , B-Lymphocytes/immunology , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Immunity, Mucosal , T-Lymphocytes/immunology , Immune Tolerance , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Gastrointestinal Tract/immunology , Gastrointestinal Microbiome/immunology , Allergens/immunology
11.
Immunol Rev ; 326(1): 191-202, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39046826

ABSTRACT

Cow milk protein allergy (CMPA) is one of the most common food allergies in the pediatric age worldwide. Prevalence, persistence, and severity of this condition are on the rise, with a negative impact on the health-related quality of life of the patients and families and on the costs related to its management. Another relevant issue is that CMPA in early life may be the first stage of the "allergic march," leading to the occurrence of other atopic manifestations later in life, especially asthma, atopic eczema, urticaria, and rhinoconjunctivitis. Thus, "disease modification" options that are able to modulate the disease course of pediatric patients affected by CMPA would be very welcomed by affected families and healthcare systems. In this review, we report the most relevant progress on this topic.


Subject(s)
Milk Hypersensitivity , Milk Proteins , Humans , Milk Hypersensitivity/immunology , Milk Hypersensitivity/therapy , Animals , Milk Proteins/immunology , Cattle , Allergens/immunology , Child , Quality of Life
12.
Immunol Rev ; 326(1): 162-172, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39034662

ABSTRACT

The last few decades have seen striking changes in the field of food allergy. The prevalence of the disease has risen dramatically in many parts of the globe, and management of the condition has undergone major revision. While delayed introduction of common allergenic foods during infancy was advised for many years, the learning early about peanut allergy (LEAP) trial and other studies led to a major shift in infant feeding practices, with deliberate early introduction of these foods now recommended. Additionally, the Food and Drug Administration approved the first treatment for food allergy in 2020-a peanut oral immunotherapy (OIT) product that likely represents just the beginning of new immunotherapy-based and other treatments for food allergy. Our knowledge of the environmental and genetic factors contributing to the pathogenesis of food allergy has also undergone transformational advances. Here, we will discuss our efforts to improve the clinical care of patients with food allergy and our understanding of the immunological mechanisms contributing to this common disease.


Subject(s)
Allergens , Food Hypersensitivity , Humans , Food Hypersensitivity/therapy , Food Hypersensitivity/immunology , Food Hypersensitivity/genetics , Allergens/immunology , Animals , Desensitization, Immunologic/methods , Genetic Predisposition to Disease , Peanut Hypersensitivity/therapy , Peanut Hypersensitivity/immunology , Peanut Hypersensitivity/genetics
13.
Immunol Rev ; 326(1): 130-150, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39275992

ABSTRACT

The prevalence of allergies has been globally escalating. While allergies could appear at any age, they often develop in early life. However, the significant knowledge gap in the field is the mechanisms by which allergies affect certain people but not others. Investigating early factors and events in neonatal life that have a lasting impact on determining the susceptibilities of children to develop allergies is a significant area of the investigation as it promotes the understanding of neonatal immune system that mediates tolerance versus allergies. This review focuses on the research over the recent 10 years regarding the potential maternal factors that influence offspring allergies with a view to food allergy, a potentially life-threatening cause of anaphylaxis. The role of breast milk, maternal diet, maternal antibodies, and microbiota that have been suggested as key maternal factors regulating offspring allergies are discussed here. We also suggest future research area to expand our knowledge of maternal-offspring interactions on the pathogenesis of food allergy.


Subject(s)
Food Hypersensitivity , Humans , Food Hypersensitivity/immunology , Female , Pregnancy , Animals , Milk, Human/immunology , Prenatal Exposure Delayed Effects/immunology , Immune Tolerance , Microbiota/immunology , Anaphylaxis/immunology , Anaphylaxis/etiology , Maternal Exposure/adverse effects , Infant, Newborn , Allergens/immunology
14.
Immunol Rev ; 326(1): 203-218, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39285525

ABSTRACT

The increasing prevalence of immune-mediated non-communicable chronic diseases, such as food allergies, has prompted a deeper investigation into the role of the gut microbiome in modulating immune responses. Here, we explore the complex interactions between commensal microbes and the host immune system, highlighting the critical role of gut bacteria in maintaining immune homeostasis. We examine how modern lifestyle practices and environmental factors have disrupted co-evolved host-microbe interactions and discuss how changes in microbiome composition impact epithelial barrier function, responses to food allergens, and susceptibility to allergic diseases. Finally, we examine the potential of bioengineered microbiome-based therapies, and live biotherapeutic products, for reestablishing immune homeostasis to prevent or treat food allergies.


Subject(s)
Food Hypersensitivity , Gastrointestinal Microbiome , Symbiosis , Humans , Animals , Gastrointestinal Microbiome/immunology , Food Hypersensitivity/immunology , Symbiosis/immunology , Homeostasis , Allergens/immunology , Food , Immunomodulation , Host Microbial Interactions/immunology , Probiotics/therapeutic use
15.
Immunol Rev ; 326(1): 83-101, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39092839

ABSTRACT

Food allergy is classically characterized by an inappropriate type-2 immune response to allergenic food antigens. However, how allergens are detected and how that detection leads to the initiation of allergic immunity is poorly understood. In addition to the gastrointestinal tract, the barrier epithelium of the skin may also act as a site of food allergen sensitization. These barrier epithelia are densely innervated by sensory neurons, which respond to diverse physical environmental stimuli. Recent findings suggest that sensory neurons can directly detect a broad array of immunogens, including allergens, triggering sensory responses and the release of neuropeptides that influence immune cell function. Reciprocally, immune mediators modulate the activation or responsiveness of sensory neurons, forming neuroimmune feedback loops that may impact allergic immune responses. By utilizing cutaneous allergen exposure as a model, this review explores the pivotal role of sensory neurons in allergen detection and their dynamic bidirectional communication with the immune system, which ultimately orchestrates the type-2 immune response. Furthermore, it sheds light on how peripheral signals are integrated within the central nervous system to coordinate hallmark features of allergic reactions. Drawing from this emerging evidence, we propose that atopy arises from a dysregulated neuroimmune circuit.


Subject(s)
Allergens , Food Hypersensitivity , Neuroimmunomodulation , Sensory Receptor Cells , Humans , Food Hypersensitivity/immunology , Animals , Sensory Receptor Cells/immunology , Sensory Receptor Cells/metabolism , Allergens/immunology , Skin/immunology
16.
Immunol Rev ; 326(1): 219-226, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39285835

ABSTRACT

The early development of the neonatal immune system is profoundly influenced by exposure to dietary and microbial antigens, which shapes mucosal tolerance. Successful oral tolerance induction is crucially dependent on microbially imprinted immune cells, most notably the RORγt+ regulatory T (Treg) and antigen presenting cells and is essential for preventing food allergy (FA). The development of FA can be envisioned to result from disruptions at key checkpoints (CKPTs) that govern oral tolerance induction. These include gut epithelial sensory and effector circuits that when dysregulated promote pro-allergic gut dysbiosis. They also include microbially imprinted immune regulatory circuits that are disrupted by dysbiosis and pro-allergic immune responses unleashed by the dysregulation of the aforementioned cascades. Understanding these checkpoints is essential for developing therapeutic strategies to restore immune homeostasis in FA.


Subject(s)
Food Hypersensitivity , Gastrointestinal Microbiome , Immune Tolerance , T-Lymphocytes, Regulatory , Humans , Food Hypersensitivity/immunology , Animals , Gastrointestinal Microbiome/immunology , T-Lymphocytes, Regulatory/immunology , Dysbiosis/immunology , Allergens/immunology
17.
Trends Immunol ; 44(9): 665-667, 2023 09.
Article in English | MEDLINE | ID: mdl-37586904

ABSTRACT

Growing evidence suggests that food allergies are regulated by neuroimmune interactions. However, the underlying molecular mechanisms remain unclear. Plum et al. and Florsheim et al. identify IgE-mediated mast cell activation, leukotrienes, and growth differentiating factor 15 (GDF15) as key regulators of the avoidance response to food allergens in mice.


Subject(s)
Antigen Presentation , Avoidance Learning , Animals , Mice , Neuroimmunomodulation
18.
Int Immunol ; 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39271155

ABSTRACT

Atopic diseases, including atopic dermatitis (AD), food allergy (FA), asthma, and allergic rhinitis (AR) are closely related to inflammatory diseases involving different body sites (i.e. the skin, airway, and digestive tract) with characteristic features including specific IgE to allergens (so-called 'atopy') and Th2 cell-mediated inflammation. It has been recognized that AD often precedes the development of other atopic diseases. The progression from AD during infancy to FA or asthma/AR in later childhood is referred as the 'atopic march' (AM). Clinical, genetic and experimental studies have provided evidence that allergen sensitization occurring through AD skin could be the origin of the AM. Here, we provide an updated review focusing on the role of the skin in the AM, from genetic mutations and environmental factors associated with epidermal barrier dysfunction in AD and the AM, to immunological mechanisms for skin sensitization, particularly recent progress on the function of key cytokines produced by epidermal keratinocytes or by immune cells infiltrating the skin during AD. We also highlight the importance of developing strategies that target AD skin to prevent and attenuate the AM.

19.
J Allergy Clin Immunol ; 154(1): 157-167, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38522626

ABSTRACT

BACKGROUND: A substantial proportion of sensitized individuals tolerate suspected foods without developing allergic symptoms; this phenomenon is known as sensitized tolerance. The immunogenic and metabolic features underlying the sensitized-tolerant phenotype remain largely unknown. OBJECTIVE: We aimed to uncover the metabolic signatures associated with clinical milk allergy (MA) and sensitized tolerance using metabolomics. METHODS: We characterized the serum metabolic and immunologic profiles of children with clinical IgE-mediated MA (n = 30) or milk-sensitized tolerance (n = 20) and healthy controls (n = 21). A comparative analysis was performed to identify dysregulated pathways associated with the clinical manifestations of food allergy. We also analyzed specific biomarkers indicative of different sensitization phenotypes in children with MA. The candidate metabolites were validated in an independent quantification cohort (n = 41). RESULTS: Metabolomic profiling confirmed the presence of a distinct metabolic signature that discriminated children with MA from those with milk-sensitized tolerance. Amino acid metabolites generated via arginine, proline, and glutathione metabolism were uniquely altered in children with sensitized tolerance. Arginine depletion and metabolism through the polyamine pathway to fuel glutamate synthesis were closely associated with suppression of clinical symptoms in the presence of allergen-specific IgE. In children with MA, the polysensitized state was characterized by disturbances in tryptophan metabolism. CONCLUSIONS: By combining untargeted metabolomics with targeted validation in an independent quantification cohort, we identified candidate metabolites as phenotypic and diagnostic biomarkers of food allergy. Our results provide insights into the pathologic mechanisms underlying childhood allergy and suggest potential therapeutic targets.


Subject(s)
Amino Acids , Biomarkers , Immune Tolerance , Metabolomics , Milk Hypersensitivity , Humans , Milk Hypersensitivity/immunology , Milk Hypersensitivity/blood , Male , Female , Amino Acids/metabolism , Child , Child, Preschool , Immunoglobulin E/blood , Immunoglobulin E/immunology , Phenotype , Infant , Animals , Allergens/immunology
20.
J Allergy Clin Immunol ; 154(3): 511-522, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38971539

ABSTRACT

Food allergy is a growing problem with limited treatment options. It is important to understand the mechanisms of food tolerance and allergy to promote the development of directed therapies. Dendritic cells (DCs) are specialized antigen-presenting cells (APCs) that prime adaptive immune responses, such as those involved in the development of oral tolerance and food allergies. The DC subsets in the gut and skin are defined by their surface markers and function. The default response to an ingested innocuous antigen is oral tolerance, which requires either gut DCs or a subset of newly identified RORγt+ APCs to induce the development of gut peripheral regulatory T cells. However, DCs in the skin, gut, and lung can also promote allergic sensitization when they are activated under certain inflammatory conditions, such as with alarmin release or gut dysbiosis. DCs also play a role in the responses to the various modalities of food immunotherapy. Langerhans cells in the skin appear to be necessary for the response to epicutaneous immunotherapy. It will be important to determine which real-world stimuli activate the DCs that prime allergic sensitization and discover methods to selectively initiate a tolerogenic program in APCs.


Subject(s)
Dendritic Cells , Food Hypersensitivity , Immune Tolerance , Humans , Food Hypersensitivity/immunology , Food Hypersensitivity/therapy , Dendritic Cells/immunology , Animals , Desensitization, Immunologic/methods , T-Lymphocytes, Regulatory/immunology , Allergens/immunology
SELECTION OF CITATIONS
SEARCH DETAIL