Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
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
2.
J Immunol ; 211(3): 389-402, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37272847

ABSTRACT

The impact of endemic parasitic infection on vaccine efficacy is an important consideration for vaccine development and deployment. We have examined whether intestinal infection with the natural murine helminth Heligmosomoides polygyrus bakeri alters Ag-specific Ab and cellular immune responses to oral and parenteral vaccination in mice. Oral vaccination of mice with a clinically relevant, live, attenuated, recombinant Salmonella vaccine expressing chicken egg OVA (Salmonella-OVA) induced the accumulation of activated, OVA-specific T effector cells rather than OVA-specific regulatory T cells in the GALT. Intestinal helminth infection significantly reduced Th1-skewed Ab responses to oral vaccination with Salmonella-OVA. Activated, adoptively transferred, OVA-specific CD4+ T cells accumulated in draining mesenteric lymph nodes of vaccinated mice, regardless of their helminth infection status. However, helminth infection increased the frequencies of adoptively transferred OVA-specific CD4+ T cells producing IL-4 and IL-10 in the mesenteric lymph node. Ab responses to the oral Salmonella-OVA vaccine were reduced in helminth-free mice adoptively transferred with OVA-specific CD4+ T cells harvested from mice with intestinal helminth infection. Intestinal helminth infection also significantly reduced Th2-skewed Ab responses to parenteral vaccination with OVA adsorbed to alum. These findings suggest that vaccine-specific CD4+ T cells induced in the context of helminth infection retain durable immunomodulatory properties and may promote blunted Ab responses to vaccination. They also underscore the potential need to treat parasitic infection before mass vaccination campaigns in helminth-endemic areas.


Subject(s)
Helminthiasis , Intestinal Diseases, Parasitic , Mice , Animals , Vaccine Efficacy , CD4-Positive T-Lymphocytes , Vaccines, Synthetic , Ovalbumin , Mice, Inbred BALB C
3.
J Immunol ; 208(2): 267-277, 2022 01 15.
Article in English | MEDLINE | ID: mdl-35017216

ABSTRACT

Alpha-gal syndrome (AGS) describes a collection of symptoms associated with IgE-mediated hypersensitivity responses to the glycan galactose-alpha-1,3-galactose (alpha-gal). Individuals with AGS develop delayed hypersensitivity reactions, with symptoms occurring >2 h after consuming mammalian ("red") meat and other mammal-derived food products. The mechanisms of pathogenesis driving this paradigm-breaking food allergy are not fully understood. We review the role of tick bites in the development of alpha-gal-specific IgE and highlight innate and adaptive immune cells possibly involved in alpha-gal sensitization. We discuss the impact of alpha-gal glycosylation on digestion and metabolism of alpha-gal glycolipids and glycoproteins, and the implications for basophil and mast cell activation and mediator release that generate allergic symptoms in AGS.


Subject(s)
Food Hypersensitivity/immunology , Food Hypersensitivity/physiopathology , Immunoglobulin E/immunology , Tick Bites/physiopathology , Animals , Bacteria/immunology , Disease Models, Animal , Glycolipids/metabolism , Glycoproteins/metabolism , Humans , Hypersensitivity, Immediate/immunology , Hypersensitivity, Immediate/pathology , Lymphocytes/immunology , Mice , Red Meat/adverse effects , Tick Bites/microbiology
4.
JAMA ; 331(6): 510-521, 2024 02 13.
Article in English | MEDLINE | ID: mdl-38349368

ABSTRACT

Importance: An estimated 7.6% of children and 10.8% of adults have IgE-mediated food-protein allergies in the US. IgE-mediated food allergies may cause anaphylaxis and death. A delayed, IgE-mediated allergic response to the food-carbohydrate galactose-α-1,3-galactose (alpha-gal) in mammalian meat affects an estimated 96 000 to 450 000 individuals in the US and is currently a leading cause of food-related anaphylaxis in adults. Observations: In the US, 9 foods account for more than 90% of IgE-mediated food allergies-crustacean shellfish, dairy, peanut, tree nuts, fin fish, egg, wheat, soy, and sesame. Peanut is the leading food-related cause of fatal and near-fatal anaphylaxis in the US, followed by tree nuts and shellfish. The fatality rate from anaphylaxis due to food in the US is estimated to be 0.04 per million per year. Alpha-gal syndrome, which is associated with tick bites, is a rising cause of IgE-mediated food anaphylaxis. The seroprevalence of sensitization to alpha-gal ranges from 20% to 31% in the southeastern US. Self-injectable epinephrine is the first-line treatment for food-related anaphylaxis. The cornerstone of IgE-food allergy management is avoidance of the culprit food allergen. There are emerging immunotherapies to desensitize to one or more foods, with one current US Food and Drug Administration-approved oral immunotherapy product for treatment of peanut allergy. Conclusions and Relevance: IgE-mediated food allergies, including delayed IgE-mediated allergic responses to red meat in alpha-gal syndrome, are common in the US, and may cause anaphylaxis and rarely, death. IgE-mediated anaphylaxis to food requires prompt treatment with epinephrine injection. Both food-protein allergy and alpha-gal syndrome management require avoiding allergenic foods, whereas alpha-gal syndrome also requires avoiding tick bites.


Subject(s)
Anaphylaxis , Food Hypersensitivity , Tick Bites , Adult , Child , Humans , Anaphylaxis/etiology , Anaphylaxis/therapy , Arachis , Epinephrine , Food Hypersensitivity/complications , Food Hypersensitivity/therapy , Galactose , Immunoglobulin E , Mammals , Meat , Seroepidemiologic Studies , United States/epidemiology
5.
Int J Mol Sci ; 22(6)2021 Mar 20.
Article in English | MEDLINE | ID: mdl-33804792

ABSTRACT

The mechanisms of pathogenesis driving alpha-gal syndrome (AGS) are not fully understood. Differences in immune gene expression between AGS individuals and non-allergic controls may illuminate molecular pathways and targets critical for AGS development. We performed immune expression profiling with RNA from the peripheral blood mononuclear cells (PBMCs) of seven controls, 15 AGS participants, and two participants sensitized but not allergic to alpha-gal using the NanoString nCounter PanCancer immune profiling panel, which includes 770 genes from 14 different cell types. The top differentially expressed genes (DEG) between AGS subjects and controls included transcription factors regulating immune gene expression, such as the NFκB pathway (NFKBIA, NFKB2, REL), antigen presentation molecules, type 2/allergic immune responses, itch, and allergic dermatitis. The differential expression of genes linked to T and B cell function was also identified, including transcription factor BCL-6, markers of antigen experience (CD44) and memory (CD27), chemokine receptors (CXCR3, CXCR6), and regulators of B-cell proliferation, cell cycle entry and immunoglobulin production (CD70). The PBMCs from AGS subjects also had increased TNF and IFN-gamma mRNA expression compared to controls. AGS is associated with a distinct gene expression profile in circulating PBMCs. DEGs related to antigen presentation, antigen-experienced T-cells, and type 2 immune responses may promote the development of alpha-gal specific IgE and the maintenance of AGS.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Food Hypersensitivity/diagnosis , Food Hypersensitivity/etiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcription, Genetic , Adolescent , Adult , Aged , Aged, 80 and over , Allergens/immunology , Case-Control Studies , Computational Biology/methods , Diagnosis, Differential , Female , Gene Expression Profiling , Humans , Immunization , Immunoglobulin E/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation , Male , Middle Aged , Red Meat/adverse effects , Young Adult
6.
Curr Opin Pediatr ; 32(6): 816-824, 2020 12.
Article in English | MEDLINE | ID: mdl-33009122

ABSTRACT

PURPOSE OF REVIEW: Alpha-gal syndrome encompasses a constellation of symptoms associated with immune-mediated hypersensitivity responses to galactose-alpha-1,3-galactose (alpha-gal). The purpose of this review is to discuss our current understanding of the etiology, clinical symptoms, natural history, epidemiology, and management of alpha-gal syndrome. RECENT FINDINGS: Sensitization to alpha-gal is associated with bites from ectoparasites like the lone star tick Amblyomma americanum. Allergic reactions in alpha-gal syndrome are often delayed and inconsistent. The magnitude of the allergic response depends on co-factors like exercise and alcohol consumption and the amount of alpha-gal and fat present in the food. Assaying alpha-gal-specific IgE in the serum is the primary diagnostic test used to confirm the allergy. Long-term management of the condition involves avoidance of both mammalian food products and tick bites. SUMMARY: Alpha-gal syndrome disrupts the current paradigm for understanding food allergy. Exposure to an ectoparasite is critical for the development of specific IgE antibodies underlying sensitization, and allergic reactions depend on the activation of mast cells and basophils sensitized with IgE against a carbohydrate rather than a protein. Research in this field may lead to the development of improved diagnostic and therapeutic tools that can revolutionize the management of patients with alpha-gal syndrome.


Subject(s)
Food Hypersensitivity , Food Hypersensitivity/diagnosis , Food Hypersensitivity/therapy , Humans
7.
Yale J Biol Med ; 93(5): 719-731, 2020 12.
Article in English | MEDLINE | ID: mdl-33380934

ABSTRACT

Mast cells are tissue resident allergic effector cells that drive IgE-mediated food allergies. There are several steps leading to mast cell activation in the context of allergic disease that can be targeted to prevent mast cell activation and degranulation. These include blocking IgE-FcεRI crosslinking and type 2 cytokine receptor activation; modulating cell-surface neural chemical receptors; stabilizing mast cell membranes to prevent co-localization of activating receptors; impeding intracellular signaling; and engaging cell surface inhibitory receptors. This review highlights several ITIM-containing inhibitory mast cell surface receptors that could serve as pharmaceutical targets to prevent mast cell activation and degranulation in the context of food allergy. When activated, these ITIM-containing inhibitory receptors recruit the phosphatases SHP-1, SHP-2, and/or SHIP to dephosphorylate the tyrosine kinases responsible for activation signals downstream of the IgE-FcεRI complex. We describe several members of the Ig and Ig-like inhibitory receptor and C-type lectin inhibitory receptor superfamilies. Fundamental studies exploring the behavior of these receptors within the context of experimental food allergy models are needed. A deeper understanding of how these receptors modulate mast cell-driven food allergic responses will shape future strategies to harness these inhibitory receptors to treat food allergy.


Subject(s)
Food Hypersensitivity , Mast Cells , Humans , Receptors, IgE , Signal Transduction
8.
Ann Allergy Asthma Immunol ; 122(6): 598-602, 2019 06.
Article in English | MEDLINE | ID: mdl-30922956

ABSTRACT

OBJECTIVE: To review published studies on galactose α-1,3-galactose (α-gal), a carbohydrate epitope found on proteins and lipids in nonprimate mammals and present in foods (particularly organ or fat-rich red meat) and medications, where it causes delayed-onset and immediate-onset anaphylaxis. DATA SOURCES: A literature search for the terms galactose α-1,3-galactose and α-gal using PubMed and Embase was performed. STUDY SELECTIONS: Studies on α-gal were included in this review. RESULTS: Several species of ticks contain α-gal epitopes and possibly salivary adjuvants that promote high titer sensitization and clinical reactivity. Risk factors for α-gal syndrome include exposure to ticks of particular species. Age and sex differences seen in various cohorts possibly reflect the prevalence of these exposures that vary according to setting. CONCLUSION: The reason and mechanisms for delayed onset of food-related anaphylaxis and the preponderance of abdominal reactions are not clear but may involve the kinetics of allergen digestion and processing or immunologic presentation via a different mechanism from usual immediate-type food allergy.


Subject(s)
Allergens/immunology , Anaphylaxis/immunology , Food Hypersensitivity/immunology , Galactose/immunology , Insect Proteins/immunology , Population Groups , Animals , Humans , Hypersensitivity, Delayed , Phenotype , Red Meat , Ticks/immunology
9.
Curr Gastroenterol Rep ; 20(5): 17, 2018 Apr 05.
Article in English | MEDLINE | ID: mdl-29623490

ABSTRACT

PURPOSE OF REVIEW: The goal of this review is to present an updated summary of the natural history of major childhood and adult food allergies and report recent advances in potential treatments for food allergy. RECENT FINDINGS: The most common childhood food allergies are typically outgrown by adolescence or adulthood. However, peanut/tree nut allergies appear to more commonly persist into adulthood. Adults can develop new IgE-mediated food allergies; the most common is oral allergy syndrome. There are multiple different approaches being tried as possible treatments for food allergy. The prevalence of food allergy appears to be increasing but the varied approaches to treatment are being actively pursued such that an approved modality may not be too far in the future.


Subject(s)
Food Hypersensitivity/therapy , Desensitization, Immunologic/methods , Food Hypersensitivity/diagnosis , Food Hypersensitivity/immunology , Humans , Immunoglobulin E/immunology , Immunotherapy/methods
11.
Curr Allergy Asthma Rep ; 16(5): 37, 2016 May.
Article in English | MEDLINE | ID: mdl-27041704

ABSTRACT

Food allergy is defined as an IgE-mediated hypersensitivity response to ingested food with allergic symptoms ranging from urticaria to life-threatening anaphylaxis. Food allergy is thought to develop because of (1) failed induction of tolerance upon initial exposure to food antigen or (2) breakdown of established tolerance to food antigen. We review current understanding of the pathogenesis, epidemiology, and natural history of food allergy, including the unconventional IgE-mediated food allergy to mammalian meat known as alpha-gal food allergy. We highlight emerging data on food allergy treatment and prevention, emphasizing the growing appeal of manipulating the gut microenvironment using probiotics and helminth products to blunt systemic allergic responses to food.


Subject(s)
Food Hypersensitivity/immunology , Animals , Food Hypersensitivity/etiology , Food Hypersensitivity/prevention & control , Food Hypersensitivity/therapy , Humans , Immunoglobulin E/immunology , Meat , Microbiota , Probiotics/therapeutic use
14.
J Clin Sleep Med ; 19(8): 1545-1552, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37082825

ABSTRACT

STUDY OBJECTIVES: The treatment of obstructive sleep apnea is often impeded by intolerance of positive airway pressure therapy, which is frequently attributed to the inability to breathe through the nose. Providers caring for patients with sleep apnea need a working knowledge of nasal passage disease and available treatments to better manage this common comorbidity. METHODS: This review examines the literature connecting rhinosinusitis to adverse sleep and sleep apnea outcomes. It explores the different types of nasal and sinus diseases a sleep apnea provider might encounter, focusing on the medications used to treat them and indications for referral to otolaryngology. RESULTS: Chronic rhinosinusitis can be either allergic or nonallergic. Both types can interfere with sleep and sleep apnea therapy. The successful management of chronic rhinosinusitis can improve positive airway pressure tolerance and adherence. A wide range of over-the-counter and prescription pharmacotherapy is available, with data supporting intranasal over oral treatment. Surgical treatment for chronic rhinosinusitis in obstructive sleep apnea addresses nasal obstruction, often with inferior turbinate reduction and septoplasty. CONCLUSIONS: Sleep specialists should have a working knowledge of the available options to treat chronic rhinosinusitis. These options are often safe, effective, and readily accessible. Otolaryngologists and allergists/immunologists provide additional treatment options for more complicated patients. Providing treatment for chronic rhinosinusitis should be included as part of comprehensive sleep apnea care. CITATION: Ali MM, Ellison M, Iweala OI, Spector AR. A sleep clinician's guide to runny noses: evaluation and management of chronic rhinosinusitis to improve sleep apnea care in adults. J Clin Sleep Med. 2023;19(8):1545-1552.


Subject(s)
Sinusitis , Sleep Apnea, Obstructive , Humans , Adult , Nose , Administration, Intranasal , Rhinorrhea , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/diagnosis , Sleep Apnea, Obstructive/therapy , Sinusitis/complications , Sinusitis/therapy , Sleep
15.
J Asthma Allergy ; 16: 661-665, 2023.
Article in English | MEDLINE | ID: mdl-37388705

ABSTRACT

Acute and chronic cough are common symptoms in patients with severe allergic asthma. Although asthma-related cough can be controlled by asthma-specific medications, both prescription and over-the-counter antitussives are often also necessary. The anti-immunoglobulin E monoclonal antibody omalizumab is an effective treatment for patients with moderate-to-severe asthma, but little is known about subsequent antitussive use patterns. This post hoc analysis examined data from the Phase 3 EXTRA study that included patients aged 12-75 years with inadequately controlled moderate-to-severe asthma. Baseline antitussive use was low overall (16/427, 3.7% for omalizumab and 18/421, 4.3% for placebo). Among patients with no baseline antitussive use (n = 411 omalizumab, n = 403 placebo), most patients (88.3% omalizumab, 83.4% placebo) reported not using antitussives during the 48-week treatment period. The percentage of patients using 1 antitussive was lower for patients treated with omalizumab than placebo (7.1% vs 13.2%), although the adjusted rate of antitussive use during the treatment period was similar for omalizumab and placebo (0.22 and 0.25). Non-narcotics were used more often than narcotics. In conclusion, this analysis found low use of antitussives in patients with severe asthma and suggests that omalizumab may have the potential to decrease antitussive use.

16.
Immunotargets Ther ; 11: 37-54, 2022.
Article in English | MEDLINE | ID: mdl-36134173

ABSTRACT

Alpha-gal syndrome is an unconventional food allergy, characterized by IgE-mediated hypersensitivity responses to the glycan galactose-alpha-1,3-galactose (alpha-gal) and not to a food-protein. In this review, we discuss how alpha-gal syndrome reframes our current conception of the mechanisms of pathogenesis of food allergy. The development of alpha-gal IgE is associated with tick bites though the possibility of other parasites promoting sensitization to alpha-gal remains. We review the immune cell populations involved in the sensitization and effector phases of alpha-gal syndrome and describe the current understanding of why allergic responses to ingested alpha-gal can be delayed by several hours. We review the foundation of management in alpha-gal syndrome, namely avoidance, but also discuss the use of antihistamines, mast cell stabilizers, and the emerging role of complementary and alternative therapies, biological products, and oral immunotherapy in the management of this condition. Alpha-gal syndrome influences the safety and tolerability of medications and medical devices containing or derived from mammalian products and impacts quality of life well beyond food choices.

17.
Front Immunol ; 13: 974374, 2022.
Article in English | MEDLINE | ID: mdl-36248809

ABSTRACT

Background: 10% of US residents have food allergies, including 2% with peanut allergy. Mast cell mediators released during the allergy effector phase drive allergic reactions. Therefore, targeting sensitized mast cells may prevent food allergy symptoms. Objective: We used novel, human, allergen-specific, IgE monoclonal antibodies (mAbs) created using human hybridoma techniques to design an in vitro system to evaluate potential therapeutics targeting sensitized effector cells. Methods: Two human IgE mAbs specific for peanut, generated through human hybridoma techniques, were used to sensitize rat basophilic leukemia (RBL) SX-38 cells expressing the human IgE receptor (FcϵRI). Beta-hexosaminidase release (a marker of degranulation), cytokine production, and phosphorylation of signal transduction proteins downstream of FcϵRI were measured after stimulation with peanut. Degranulation was also measured after engaging inhibitory receptors CD300a and Siglec-8. Results: Peanut-specific human IgE mAbs bound FcϵRI, triggering degranulation after stimulation with peanut in RBL SX-38 cells. Sensitized RBL SX-38 cells stimulated with peanut increased levels of phosphorylated SYK and ERK, signal transduction proteins downstream of FcϵRI. Engaging inhibitory cell surface receptors CD300a or Siglec-8 blunted peanut-specific activation. Conclusion: Allergen-specific human IgE mAbs, expressed from human hybridomas and specific for a clinically relevant food allergen, passively sensitize allergy effector cells central to the in vitro models of the effector phase of food allergy. Peanut reproducibly activates and induces degranulation of RBL SX-38 cells sensitized with peanut-specific human IgE mAbs. This system provides a unique screening tool to assess the efficacy of therapeutics that target allergy effector cells and inhibit food allergen-induced effector cell activation.


Subject(s)
Food Hypersensitivity , Peanut Hypersensitivity , Allergens , Animals , Antibodies, Monoclonal/pharmacology , Arachis , Cell Degranulation , Cytokines , Humans , Immunoglobulin E , Rats , Receptors, IgE/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins , beta-N-Acetylhexosaminidases
18.
J Immunol ; 183(4): 2252-60, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19620295

ABSTRACT

The requirement for TLR signaling in the initiation of an Ag-specific Ab response is controversial. In this report we show that a novel OVA-expressing recombinant Salmonella vaccine (Salmonella-OVA) elicits a Th1-biased cell-mediated and serum Ab response upon oral or i.p. immunization of C57BL/6 mice. In MyD88(-/-) mice, Th1-dependent Ab responses are greatly reduced while Th2-dependent Ab isotypes are elevated in response to oral and i.p., but not s.c. footpad, immunization. When the T effector response to oral vaccination is examined we find that activated, adoptively transferred Ag-specific CD4(+) T cells accumulate in the draining lymph nodes, but fail to produce IFN-gamma, in MyD88(-/-) mice. Moreover, CD1d tetramer staining shows that invariant NKT cells are activated in response to oral Salmonella-OVA vaccination in wild-type, but not MyD88(-/-), mice. Treatment with neutralizing Ab to CD1d reduces the OVA-specific Ab response only in MyD88-sufficient wild-type mice, suggesting that both Ag-specific CD4 T cell and invariant NKT cell effector responses to Salmonella-OVA vaccination are MyD88 dependent. Taken together, our data indicate that the type of adaptive immune response generated to this live attenuated vaccine is regulated by both the presence of MyD88-mediated signals and vaccination route, which may have important implications for future vaccine design.


Subject(s)
Antibodies, Bacterial/biosynthesis , CD4-Positive T-Lymphocytes/immunology , Immunoglobulin Isotypes/biosynthesis , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Natural Killer T-Cells/immunology , Salmonella Vaccines/administration & dosage , Administration, Oral , Animals , Antibodies, Bacterial/blood , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Female , Immunoglobulin Isotypes/blood , Injections, Intraperitoneal , Injections, Subcutaneous , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88/physiology , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/pathology , Salmonella Vaccines/genetics , Salmonella Vaccines/immunology , Toll-Like Receptors/physiology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
19.
Immun Inflamm Dis ; 9(3): 984-990, 2021 09.
Article in English | MEDLINE | ID: mdl-34034363

ABSTRACT

INTRODUCTION: Alpha-gal syndrome (AGS) is characterized by delayed hypersensitivity to non-primate mammalian meat in people having specific immunoglobulin E (sIgE) to the oligosaccharide galactose-alpha-1,3-galactose. AGS has been linked to tick bites from Amblyomma americanum (Aa) in the U.S. A small animal model of meat allergy is needed to study the mechanism of alpha-gal sensitization, the effector phase leading to delayed allergic responses and potential therapeutics to treat AGS. METHODS: Eight- to ten-weeks old mice with a targeted inactivation of alpha-1,3-galactosyltransferase (AGKO) were injected intradermally with 50 µg of Aa tick salivary gland extract (TSGE) on days 0, 7, 21, 28, 42, and 49. Total IgE and alpha-gal sIgE were quantitated on Day 56 by enzyme-linked immunosorbent assay. Mice were challenged orally with 400 mg of cooked pork kidney homogenate or pork fat. Reaction severity was assessed by measuring a drop in core body temperature and scoring allergic signs. RESULTS: Compared to control animals, mice treated with TSGE had 190-fold higher total IgE on Day 56 (0.60 ± 0.12 ng/ml vs. 113.2 ± 24.77 ng/ml; p < 0.001). Alpha-gal sIgE was also produced in AGKO mice following TSGE sensitization (undetected vs. 158.4 ± 72.43 pg/ml). Further, sensitized mice displayed moderate clinical allergic signs along with a drop in core body temperature of ≥2°C as an objective measure of a systemic allergic reaction. Interestingly, female mice had higher total IgE responses to TSGE treatment but male mice had larger declines in mean body temperature. CONCLUSION: TSGE-sensitized AGKO mice generate sIgE to alpha-gal and demonstrate characteristic allergic responses to pork fat and pork kidney. In keeping with the AGS responses documented in humans, mice reacted more rapidly to organ meat than to high fat pork challenge. This mouse model establishes the central role of tick bites in the development of AGS and provides a small animal model to mechanistically study mammalian meat allergy.


Subject(s)
Food Hypersensitivity , Ticks , Animals , Female , Male , Mice , Plant Extracts , Salivary Glands
20.
Gastroenterology ; 137(4): 1380-90.e1-3, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19596011

ABSTRACT

BACKGROUND & AIMS: The commensal microbiota is believed to have an important role in regulating immune responsiveness and preventing intestinal inflammation. Intestinal microbes produce signals that regulate inflammation via Toll-like receptor (TLR) signaling, but the mechanisms of this process are poorly understood. We investigated the role of the anti-inflammatory cytokine interleukin (IL)-10 in this signaling pathway using a mouse model of colitis. METHODS: Clinical, histopathologic, and functional parameters of intestinal inflammation were evaluated in TLR4(-/-), IL-10(-/-), and TLR4(-/-) x IL-10(-/-) mice that were free of specific pathogens and in TLR4(-/-) x IL-10(-/-) mice following eradication and reintroduction of Helicobacter hepaticus. Regulatory T-cell (Treg) function was evaluated by crossing each of the lines with transgenic mice that express green fluorescent protein under control of the endogenous regulatory elements of Foxp3. Apoptotic cells in the colonic lamina propria were detected by a TUNEL assay. RESULTS: TLR4-mediated signals have 2 interrelated roles in promoting inflammation in TLR4(-/-) x IL-10(-/-) mice. In the absence of TLR4-mediated signals, secretion of proinflammatory and immunoregulatory cytokines is dysregulated. Tregs (Foxp3(+)) that secrete interferon-gamma and IL-17 accumulate in the colonic lamina propria of TLR4(-/-) x IL-10(-/-) mice and do not prevent inflammation. Aberrant control of epithelial cell turnover results in the persistence of antigen-presenting cells that contain apoptotic epithelial fragments in the colonic lamina propria of Helicobacter-infected TLR4(-/-) mice. CONCLUSIONS: In mice that lack both IL-10- and TLR4-mediated signals, aberrant regulatory T-cell function and dysregulated control of epithelial homeostasis combine to exacerbate intestinal inflammation.


Subject(s)
Colitis/immunology , Epithelial Cells/immunology , Helicobacter Infections/microbiology , Helicobacter hepaticus/immunology , Inflammation Mediators/metabolism , Interleukin-10/deficiency , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptor 4/metabolism , Animals , Apoptosis , Colitis/microbiology , Colitis/pathology , Colitis/prevention & control , Disease Models, Animal , Epithelial Cells/microbiology , Epithelial Cells/pathology , Forkhead Transcription Factors/genetics , Genes, Reporter , Green Fluorescent Proteins/genetics , Helicobacter Infections/immunology , Helicobacter Infections/pathology , Interferon-gamma/metabolism , Interleukin-10/genetics , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Rectal Prolapse/immunology , Rectal Prolapse/microbiology , Spleen/immunology , Spleen/microbiology , T-Lymphocytes, Regulatory/microbiology , Th1 Cells/immunology , Th1 Cells/microbiology , Time Factors , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
SELECTION OF CITATIONS
SEARCH DETAIL