Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Front Immunol ; 15: 1324026, 2024.
Article in English | MEDLINE | ID: mdl-38533495

ABSTRACT

Background: Imidazole propionate (IMP) is a histidine metabolite produced by some gut microorganisms in the human colon. Increased levels of IMP are associated with intestinal inflammation and the development and progression of cardiovascular disease and diabetes. However, the anti-inflammatory activity of IMP has not been investigated. This study aimed to elucidate the role of IMP in treating atopic dermatitis (AD). Methods: To understand how IMP mediates immunosuppression in AD, IMP was intraperitoneally injected into a Dermatophagoides farinae extract (DFE)/1-chloro-2,4 dinitrochlorobenzene (DNCB)-induced AD-like skin lesions mouse model. We also characterized the anti-inflammatory mechanism of IMP by inducing an AD response in keratinocytes through TNF-α/IFN-γ or IL-4 stimulation. Results: Contrary to the prevailing view that IMP is an unhealthy microbial metabolite, we found that IMP-treated AD-like skin lesions mice showed significant improvement in their clinical symptoms, including ear thickness, epidermal and dermal thickness, and IgE levels. Furthermore, IMP antagonized the expansion of myeloid (neutrophils, macrophages, eosinophils, and mast cells) and Th cells (Th1, Th2, and Th17) in mouse skin and prevented mitochondrial reactive oxygen species production by inhibiting mitochondrial energy production. Interestingly, we found that IMP inhibited AD by reducing glucose uptake in cells to suppress proinflammatory cytokines and chemokines in an AD-like in vitro model, sequentially downregulating the PI3K and mTORC2 signaling pathways centered on Akt, and upregulating DDIT4 and AMPK. Discussion: Our results suggest that IMP exerts anti-inflammatory effects through the metabolic reprogramming of skin inflammation, making it a promising therapeutic candidate for AD and related skin diseases.


Subject(s)
Dermatitis, Atopic , Imidazoles , Humans , Animals , Mice , Dermatitis, Atopic/pathology , Skin/pathology , Reactive Oxygen Species , Immunoglobulin E/adverse effects , Anti-Inflammatory Agents/pharmacology , Inflammation/pathology
2.
Genes Genomics ; 46(3): 333-340, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37837514

ABSTRACT

OBJECTIVE: Atopic dermatitis (AD) is an inflammatory skin disease. Naringenin (Nar) possesses an anti-inflammatory property. This paper attempts to discuss the functional mechanism of Nar in AD mice through the Janus kinase 2 (JAK2)/signal transducer and activation of transcription 3 (STAT3) pathway. METHODS: Mouse models of DNFB-induced AD were established and treated with Nar, followed by intraperitoneal injection with the JAK2/STAT3 pathway activator Coumermycin A1. Dermatitis severity was scored and the thickness of right ear was measured. The pathological changes in dorsal skin tissues were observed by HE staining. The number of infiltrated mast cells and eosinophilic granulocytes was counted by TB staining. The serum IgE level and levels of TNF-α, IL-6, IFN-γ, IL-12, and IL-5 in dorsal skin tissues were measured by ELISA. The levels of p-JAK2, JAK2, p-STAT3, and STAT3 were determined by Western blot. RESULTS: Nar decreased dermatitis scores and right ear thickness, alleviated skin lesions, and reduced the number of infiltrated mast cells and eosinophilic granulocytes in AD mice. The serum IgE level and levels of TNF-α, IL-6, IFN-γ, IL-12, and IL-5 in dorsal skin tissues of AD mice were diminished after Nar treatment in a dose-dependent manner. Nar inhibited the activation of the JAK2/STAT3 pathway. The activation of the JAK2/STAT3 pathway partially nullified the therapeutic function of Nar on AD mice. CONCLUSION: Nar protects mice from AD by inhibiting inflammation and promoting immune responses through the inhibition of the JAK2/STAT3 pathway.


Subject(s)
Dermatitis, Atopic , Flavanones , Mice , Animals , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/pathology , Interleukin-6 , Tumor Necrosis Factor-alpha , Janus Kinase 2/metabolism , Interleukin-5/adverse effects , Cytokines , Inflammation/drug therapy , Immunoglobulin E/adverse effects , Interleukin-12/adverse effects
3.
Cells Dev ; 175: 203864, 2023 09.
Article in English | MEDLINE | ID: mdl-37321350

ABSTRACT

The incidence of allergic asthma has been increasing worldwide in recent decades. Also, an increasing number of women are suffering from poor pregnancy outcome. However, the causal relationship between allergic asthma and embryonic growth in terms of cell morphogenesis has not been well elucidated. Here, we investigated the impact of allergic asthma on the morphogenesis of preimplantation embryos. Twenty-four female BALB/c were randomly divided into control (PBS), 50-µg (OVA1), 100-µg (OVA2) and 150-µg (OVA3). On Days-0 and -14, mice were induced intraperitoneally (i.p) with ovalbumin (OVA). On Days-21 until -23, mice were challenged with OVA via intranasal instillation (i.n). Control animals were sensitized and challenged with PBS. At the end of treatment (Day-25), 2-cell embryos were retrieved and cultured in vitro until the blastocysts hatched. Results showed reduced number of preimplantation embryos at all developing stages in all treated groups (p ≤ 0.0001). Uneven blastomere size, partial compaction- and cavitation-activity, low formation of trophectoderm (TE), as well as cell fragmentation were noted in all the treated groups. Maternal serum interleukin (IL)-4, immunoglobulin (Ig)-E and 8-hydroxydeoxyguanosine (8-OHdG) were notably high (p ≤ 0.0001, p ≤ 0.01) in contrast with low total antioxidant capacity (TAOC) (p ≤ 0.0001). Our findings indicated that OVA-induced allergic asthma had compromised cell morphogenesis through reduced blastomere cleavage division, partial compaction and cavitation-activity, impairment of TE production, and cell fragmentation leading to embryonic cell death via OS mechanism.


Subject(s)
Asthma , Female , Animals , Mice , Pregnancy , Asthma/chemically induced , Asthma/drug therapy , Asthma/metabolism , Inflammation , Administration, Intranasal , Immunoglobulin E/adverse effects , Immunoglobulin E/metabolism , Oxidative Stress , Morphogenesis
4.
PLoS One ; 18(1): e0279893, 2023.
Article in English | MEDLINE | ID: mdl-36598904

ABSTRACT

Arsenic is a potent environmental toxicant and human carcinogen. Skin lesions are the most common manifestations of chronic exposure to arsenic. Advanced-stage skin lesions, particularly hyperkeratosis have been recognized as precancerous diseases. However, the underlying mechanism of arsenic-induced skin lesions remains unknown. Periostin, a matricellular protein, is implicated in the pathogenesis of many forms of skin lesions. The objective of this study was to examine whether periostin is associated with arsenic-induced skin lesions. A total of 442 individuals from low- (n = 123) and high-arsenic exposure areas (n = 319) in rural Bangladesh were evaluated for the presence of arsenic-induced skin lesions (Yes/No). Participants with skin lesions were further categorized into two groups: early-stage skin lesions (melanosis and keratosis) and advanced-stage skin lesions (hyperkeratosis). Drinking water, hair, and nail arsenic concentrations were considered as the participants' exposure levels. The higher levels of arsenic and serum periostin were significantly associated with skin lesions. Causal mediation analysis revealed the significant effect of arsenic on skin lesions through the mediator, periostin, suggesting that periostin contributes to the development of skin lesions. When skin lesion was used as a three-category outcome (none, early-stage, and advanced-stage skin lesions), higher serum periostin levels were significantly associated with both early-stage and advanced-stage skin lesions. Median (IQR) periostin levels were progressively increased with the increasing severity of skin lesions. Furthermore, there were general trends in increasing serum type 2 cytokines (IL-4, IL-5, IL-13, and eotaxin) and immunoglobulin E (IgE) levels with the progression of the disease. The median (IQR) of IL-4, IL-5, IL-13, eotaxin, and IgE levels were significantly higher in the early-and advanced-stage skin lesions compared to the group of participants without skin lesions. The results of this study suggest that periostin is implicated in the pathogenesis and progression of arsenic-induced skin lesions through the dysregulation of type 2 immune response.


Subject(s)
Arsenic , Keratosis, Actinic , Skin Diseases , Humans , Arsenic/toxicity , Arsenic/analysis , Interleukin-13 , Interleukin-4 , Interleukin-5 , Environmental Exposure , Water Supply , Skin Diseases/chemically induced , Immunoglobulin E/adverse effects
5.
BMC Cancer ; 23(1): 32, 2023 Jan 09.
Article in English | MEDLINE | ID: mdl-36624467

ABSTRACT

BACKGROUND: The link between immediate hypersensitivity reactions (HSR) following the first cetuximab infusion and the IgE sensitization against anti-galactose-α-1,3-galactose (α-Gal) is now well-established. An automated Fluoroenzyme-Immunoassay (FEIA) is available and may facilitate the screening of patients with anti-α-Gal IgE before treatment. METHODS: This study aimed to evaluate its performances as compared to a previously validated anti-cetuximab IgE ELISA, using 185 samples from two previously studied cohorts. RESULTS: Despite 21.1% of discrepancies between the two techniques, FEIA discriminated better positive patients and similarly negative ones with a ≥ 0.525 kUA/L threshold. Sensitivity was 87.5% for both tests, specificity was better for FEIA (96.3% vs ELISA: 82.1%). FEIA had a higher positive likelihood ratio (23.9 vs ELISA: 4.89) and a similar negative likelihood ratio (0.13 vs ELISA: 0.15). In our population, the risk of severe HSR following a positive test was higher with FEIA (56.7% vs ELISA: 19.6%) and similar following a negative test (0.7% vs ELISA: 0.8%). CONCLUSION: Although the predictive value of the IgE screening before cetuximab infusion remains discussed, this automated commercial test can identify high-risk patients and is suitable for routine use in laboratories. It could help avoiding cetuximab-induced HSR by a systematic anti-α-Gal IgE screening before treatment.


Subject(s)
Anaphylaxis , Food Hypersensitivity , Humans , Anaphylaxis/chemically induced , Anaphylaxis/diagnosis , Cetuximab/adverse effects , Food Hypersensitivity/diagnosis , Galactose/adverse effects , Immunoglobulin E/adverse effects , Enzyme-Linked Immunosorbent Assay
6.
Eur J Anaesthesiol ; 40(2): 95-104, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36301083

ABSTRACT

Acute hypersensitivity reactions (AHRs) occurring in present-day anaesthesia can have severe, sometimes fatal, consequences and their incidence is increasing. The most frequent allergens responsible for AHR during anaesthesia are neuromuscular blocking agents (NMBAs) (70% of the cases) followed by antibiotics (18%), patent blue dye and methylene blue dye (5%), and latex (5%). Following an AHR, strategies for subsequent anaesthetic procedures (especially the choice of an NMBA) may be difficult to formulate due to inconclusive diagnostic analysis in up to 30% of AHRs. Current diagnosis of AHR relies on the detection of mast cell degranulation products and drug-specific type E immunoglobulins (IgE) in order to document an IgE-mediated anaphylaxis (IgE endotype). Nonetheless, other IgE-independent pathways can be involved in AHR, but their detection is not currently available in standard situations. The different mechanisms (endotypes) involved in peri-operative AHR may contribute to the inconclusive diagnostic work-up and this generates uncertainty concerning the culpable drug and strategy for subsequent anaesthetic procedures. This review provides details on the IgE endotype; an update on non-IgE related endotypes and the novel diagnostic tools that could characterise them. This detailed update is intended to provide explicit clinical reasoning tools to the anaesthesiologist faced with an incomplete AHR diagnostic work-up and to facilitate the decision-making process regarding anaesthetic procedures following an AHR to NMBAs.


Subject(s)
Anaphylaxis , Anesthesia , Neuromuscular Blocking Agents , Humans , Immunoglobulin E/adverse effects , Anaphylaxis/chemically induced , Anaphylaxis/diagnosis , Neuromuscular Blocking Agents/adverse effects , Anesthesia/adverse effects , Allergens/adverse effects , Skin Tests/adverse effects , Skin Tests/methods
7.
Sangyo Eiseigaku Zasshi ; 65(1): 1-8, 2023 Jan 25.
Article in Japanese | MEDLINE | ID: mdl-35569932

ABSTRACT

Genetic and environmental factors and their interactions cause diseases and deteriorate health (Genetic and Environmental Interaction). Exposure to environmental factors plays a major role in the deterioration of health in the workplace.Occupational asthma (OA) is a common disorder in the workplace. Approaches to OA are well described and discussed in "Japanese Guideline for Diagnosis and Management of Occupational Allergic Diseases" by the Japanese Society of Occupational and Environmental Allergy. According to the guideline, OA and work-aggravated asthma comprise work-related asthma, and OA can be further divided into two disease entities: sensitizer-induced OA and irritant-induced OA. The guidelines also describe diagnostic and therapeutic strategies for OA. Since a definitive diagnosis of OA requires a comprehensive decision based on a detailed interview on clinical symptoms related to employment status and clinical tests, including inhalation tests of suspected substances as needed, the possibility of OA should be considered as the first step toward diagnosis of the patient. Otherwise, OA may not be diagnosed. Therapeutic strategies include exposure avoidance, environmental arrangements in the workplace, utilization of social resources for workers, and conventional pharmacotherapy for asthma.Artificially synthesized small compounds are used in various industries and can cause allergies. For example, isocyanates are small compounds in the -NCO group, which have been toxicologically studied. It was later shown that isocyanate could cause various nontoxic adverse health effects, including allergic reactions. Since small agents with low molecular weights bind to proteins, detecting their specific immunoglobulin E (IgE) antibodies targeting small compounds is generally difficult. In contrast, isocyanate-specific IgE antibodies are detectable in individuals with isocyanate allergies.Suspecting OA is essential in cases exposed to newly synthesized compounds, or to those that are already known but applied to new uses, which can be better understood and predicted by studying the health effects of isocyanates.Academic interest in various issues related to allergies, immunology, and toxicology in the workplace includes clinical medicine, epidemiology, and epigenetics related to environmental exposure. Further advanced research in these areas is necessary and promising.


Subject(s)
Asthma, Occupational , Clinical Medicine , Occupational Diseases , Occupational Exposure , Humans , Occupational Exposure/adverse effects , Occupational Exposure/prevention & control , Asthma, Occupational/chemically induced , Asthma, Occupational/diagnosis , Asthma, Occupational/prevention & control , Occupational Diseases/chemically induced , Occupational Diseases/diagnosis , Occupational Diseases/prevention & control , Isocyanates/adverse effects , Immunoglobulin E/adverse effects
8.
Animal Model Exp Med ; 6(3): 255-265, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36131559

ABSTRACT

BACKGROUND: Amarogentin (AMA) is a secoiridoid glycoside extracted from Swertia and Gentiana roots and exhibits many biological effects such as antioxidative, anti-inflammatory, and antitumor activities. Atopic dermatitis (AD) is a chronic inflammatory skin disease caused by disorders in the regulation of multiple inflammatory cytokines. No effective cure has been found for AD now. METHODS: We constructed the HaCat and splenocyte model and tested the inhibitory effect of AMA on IL-4, IL-6, and IL-13 secretions using enzyme-linked immunosorbent assay (ELISA). The AD mouse model was constructed and treated with AMA, the severity of skin lesions was observed, epidermal tissue was collected, and epidermal thickness and mast cell infiltration were observed using hematoxylin and eosin and toluidine blue staining, respectively. The expression of kallikrein-related peptidase 7 (KLK7) and filaggrin (FLG) was detected using immunostaining and Western blot analysis. The mRNA expression of KLK7 and FLG was detected using quantitative polymerase chain reaction (qPCR). Blood immunoglobulin E (IgE) secretion was detected. RESULTS: AMA inhibited IL-6 secreted by tumor necrosis factor (TNF)-α-induced HaCaT cells and reduced IL-4 and IL-13 secreted by phytohemagglutinin (PHA)-induced primary cells in the mice spleen. It was found that the treatment of AMA with 2,4-dinitrochlorobenzene-induced AD-like mice could promote the recovery of dermatitis, reduce the score of dermatitis severity and the scratching frequency, treat the skin lesions, reduce the epidermal thickness, decrease the infiltration of mast cells, reduce the IgE level in serum, decrease the expression levels of AD-related cytokines, increase protein and mRNA expression of FLG, and reduce the protein and mRNA expression of KLK7 in the skin tissues of AD-like mice. CONCLUSION: In conclusion, AMA inhibits inflammatory response at the cellular level, and AMA reduces the validation response of specific dermatitis mice, relieves pruritus, and repairs the damaged skin barrier.


Subject(s)
Dermatitis, Atopic , Animals , Mice , Humans , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/metabolism , Dinitrochlorobenzene/adverse effects , Interleukin-13/adverse effects , Interleukin-6/adverse effects , HaCaT Cells/metabolism , HaCaT Cells/pathology , Interleukin-4/adverse effects , Cytokines/genetics , Cytokines/metabolism , Tumor Necrosis Factor-alpha/metabolism , Anti-Inflammatory Agents/adverse effects , Immunoglobulin E/adverse effects , RNA, Messenger/adverse effects
9.
Front Immunol ; 14: 1241851, 2023.
Article in English | MEDLINE | ID: mdl-38274796

ABSTRACT

Background: Perioperative anaphylaxis is a rare and acute systemic manifestation of drug-induced hypersensitivity reactions that occurs following anesthesia induction; the two main classes of drugs responsible for these reactions being neuromuscular blocking agents (NMBA) and antibiotics. The sensitization mechanisms to the drugs are not precisely known, and few risk factors have been described. A growing body of evidence underlines a link between occurrence of allergy and microbiota composition. However, no data exist on microbiota in perioperative anaphylaxis. The aim of this study was to compare circulating microbiota richness and composition between perioperative anaphylaxis patients and matched controls. Methods: Circulating 16s rDNA was quantified and sequenced in serum samples from 20 individuals with fully characterized IgE-mediated NMBA-related anaphylaxis and 20 controls matched on sex, age, NMBA received, type of surgery and infectious status. Microbiota composition was analyzed with a published bioinformatic pipeline and links with patients clinical and biological data investigated. Results: Analysis of microbiota diversity showed that anaphylaxis patients seem to have a richer circulating microbiota than controls, but no major differences of composition could be detected with global diversity indexes. Pairwise comparison showed a difference in relative abundance between patients and controls for Saprospiraceae, Enterobacteriaceae, Veillonellaceae, Escherichia-Shigella, Pseudarcicella, Rhodoferax, and Lewinella. Some taxa were associated with concentrations of mast cell tryptase and specific IgE. Conclusion: We did not find a global difference in terms of microbiota composition between anaphylaxis patient and controls. However, several taxa were associated with anaphylaxis patients and with their biological data. These findings must be further confirmed in different settings to broaden our understanding of drug anaphylaxis pathophysiology and identify predisposition markers.


Subject(s)
Anaphylaxis , Drug Hypersensitivity , Neuromuscular Blocking Agents , Humans , Anaphylaxis/etiology , Tryptases , Risk Factors , Neuromuscular Blocking Agents/adverse effects , Immunoglobulin E/adverse effects
10.
J Med Chem ; 65(15): 10626-10637, 2022 08 11.
Article in English | MEDLINE | ID: mdl-35876064

ABSTRACT

Allergic diseases are a group of allergen-induced unfavorable immune responses initiating various symptoms in different organs. Mas-related G protein-coupled receptor X2 (MRGPRX2) on mast cells has been reported to be responsible for immunoglobulin E (IgE)-independent immune diseases and allergic drug reactions and has therefore been a crucial drug target for the development of anti-pseudo-allergic agents. Considering the active structural features of MRGPRX2, we designed and synthesized a series of diaryl ureas (DPUs). DPUs exert promising potency for inhibiting ß-hexosaminidase release in LAD2 cells with half-maximal inhibitory concentrations (IC50) values of 2.51-0.62 µM, as well as favorable antilocal and systemic anaphylaxis in mice at a dosage of 10 mg/kg. MRGPRX2 is further revealed to participate in the anti-pseudo-allergic activity of DPUs by binding with electrophilic urea and trifluoromethyl substituents. In brief, these results highlight entities with powerful electrophilic substituents as a prospective therapeutic strategy for the treatment of IgE-independent disorders.


Subject(s)
Anaphylaxis , Anti-Allergic Agents , Anaphylaxis/chemically induced , Anaphylaxis/metabolism , Animals , Anti-Allergic Agents/pharmacology , Anti-Allergic Agents/therapeutic use , Cell Degranulation , Immunoglobulin E/adverse effects , Immunoglobulin E/metabolism , Mast Cells , Mice , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Urea/pharmacology , Urea/therapeutic use
11.
J Food Biochem ; 46(7): e14135, 2022 07.
Article in English | MEDLINE | ID: mdl-35301731

ABSTRACT

Pinus koraiensis needles (PKN) and cones (PKC) have been shown to protect against inflammation and pathogenic bacteria. We investigated the efficacies and action mechanisms of topical applications of 1,3-butylene glycol (BG) extracts and oral administration of their water extracts on atopic dermatitis (AD) symptoms. After exposing HaCaT cells and Nc/Nga mice dorsal skins to 2,4-dinitrochlorobenzene (DNCB) to induce atopic dermatitis models, they were topically applied BG (AD-control), 30% PKNX, or 30% PKCX to the skin lesions and fed water extracts (0.5%) in high-fat diets for 5 weeks. Normal-control mice had no DNCB exposure. Serum immunoglobulin E (IgE), IL-4, and TNF-α levels and gene expressions of TNF-α, IL-4, IL-6, and IFN-γ in the dorsal skin and HaCaT cells were measured. The AD-control mice elevated TNF-α and IL-6 mRNA levels in HaCaT cells. Both extracts attenuated clinical AD symptoms in AD-induced Nc/Nga mice: PKNX improved hemorrhage, erythema, and lichenification of dorsal skin better than PKCX while both similarly alleviated erythema, edema, excoriation, and itching behavior. PKCX reduced IgE contents and increased filaggrin mRNA expression better than PKNX, but PKNX reduced lipid peroxides and mRNA levels of TNF-α and IL-4 in the dorsal skin. In the histological analysis of the dorsal skin, the administration of both extracts significantly decreased mast cell numbers, immune cell infiltration, gaps between the epidermis and dermis, and abnormal cell and nucleus shapes. In conclusion, both PKCX and PKNX treatment alleviated the DNCB-induced clinical symptoms of AD by alleviating immune-related symptoms and inflammation in partially different pathways. Therefore, PKNX and PKCX may be effective for AD therapy. PRACTICAL APPLICATIONS: Atopic dermatitis (AD) is related to an overly activated immune response, and it has steadily increased last 3 decades. However, no optimal sustainable treatments are available. Pinus koraiensis needles and cones extracts have been used for anti-inflammatory and antimicrobial treatment. The present study demonstrated that their intake and topical administration onto the AD lesion alleviated clinical AD symptoms associated with reduced proinflammatory cytokines, mast cell numbers, and immune cell infiltrates to maintain dermal structure with maintaining filaggrin expression in AD-induced HaCaT cells and Nc/Nga mice. These results suggested that Pinus koraiensis needles and cones extracts can be developed and applied as beneficial alternative therapies for AD.


Subject(s)
Dermatitis, Atopic , Pinus , Animals , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/drug therapy , Dinitrochlorobenzene/adverse effects , HaCaT Cells , Humans , Immunoglobulin E/adverse effects , Inflammation , Interleukin-4 , Interleukin-6 , Mice , RNA, Messenger , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Water
12.
Int J Mol Sci ; 22(4)2021 Feb 19.
Article in English | MEDLINE | ID: mdl-33669849

ABSTRACT

The development of food allergy has been reported to be related with the changes in the gut microbiome, however the specific microbe associated with the pathogenesis of food allergy remains elusive. This study aimed to comprehensively characterize the gut microbiome and identify individual or group gut microbes relating to food-allergy using 16S rRNA gene sequencing with network analysis. Faecal samples were collected from children with IgE-mediated food allergies (n = 33) and without food allergy (n = 27). Gut microbiome was profiled by 16S rRNA gene sequencing. OTUs obtained from 16S rRNA gene sequencing were then used to construct a co-abundance network using Weighted Gene Co-expression Network Analysis (WGCNA) and mapped onto Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. We identified a co-abundance network module to be positively correlated with IgE-mediated food allergy and this module was characterized by a hub taxon, namely Ruminococcaceae UCG-002 (phylum Firmicutes). Functional pathway analysis of all the gut microbiome showed enrichment of methane metabolism and glycerolipid metabolism in the gut microbiome of food-allergic children and enrichment of ubiquinone and other terpenoid-quinone biosynthesis in the gut microbiome of non-food allergic children. We concluded that Ruminococcaceae UCG-002 may play determinant roles in gut microbial community structure and function leading to the development of IgE-mediated food allergy.


Subject(s)
Food Hypersensitivity/microbiology , Gastrointestinal Microbiome , Immunoglobulin E/adverse effects , Biodiversity , Child , Discriminant Analysis , Female , Humans , Male , Phylogeny
13.
Nat Rev Dis Primers ; 6(1): 95, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33273461

ABSTRACT

Allergic rhinitis (AR) is caused by immunoglobulin E (IgE)-mediated reactions to inhaled allergens and is one of the most common chronic conditions globally. AR often co-occurs with asthma and conjunctivitis and is a global health problem causing major burden and disability worldwide. Risk factors include inhalant and occupational allergens, as well as genetic factors. AR impairs quality of life, affects social life, school and work, and is associated with substantial economic costs. The Allergic Rhinitis and its Impact on Asthma (ARIA) initiative classified AR into intermittent or persistent and mild or moderate/severe. The diagnosis is based on the clinical history and, if needed in patients with uncontrolled rhinitis despite medications or with long-lasting symptoms, on skin tests or the presence of serum-specific IgE antibodies to allergens. The most frequently used pharmacological treatments include oral, intranasal or ocular H1-antihistamines, intranasal corticosteroids or a fixed combination of intranasal H1-antihistamines and corticosteroids. Allergen immunotherapy prescribed by a specialist using high-quality extracts in stratified patients is effective in patients with persistent symptoms. Real-world data obtained by mobile technology offer new insights into AR phenotypes and management. The outlook for AR includes a better understanding of novel multimorbid phenotypes, health technology assessment and patient-centred shared decision-making.


Subject(s)
Rhinitis, Allergic/diagnosis , Rhinitis, Allergic/therapy , Adrenal Cortex Hormones/therapeutic use , Histamine Antagonists/therapeutic use , Humans , Immunoglobulin E/adverse effects , Immunoglobulin E/immunology , Immunotherapy/methods , Immunotherapy/trends , Rhinitis, Allergic/epidemiology
15.
Phytomedicine ; 79: 153346, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33002828

ABSTRACT

BACKGROUND: Immunoglobulin E (IgE)-mediated mast cell (MC) activation is crucial in multiple allergic diseases. Parkinson disease protein 7 (DJ-1) and Lyn kinase were reported as the receptor-proximal events in IgE receptor (FcεRI) signals in human MC. Kaempferol, a natural flavonol mainly derived from the rhizome of traditional Chinese herb Kaempferia galanga L. (Zingiberaceae), has been known to inhibit allergic reactions, but it was limited to the receptor-distal signals on rat basophilic leukemia cells. A thorough investigation of the inhibitory effects of kaempferol on human MC has not been done. PURPOSE: To investigate the inhibitory effects of kaempferol on IgE-mediated anaphylaxis in vivo and in human MCs, as well as the mechanism underlying its effects, especially the receptor-proximal signals. METHODS: IgE-mediated passive cutaneous anaphylaxis and systemic anaphylaxis model were applied to elucidate the antiallergic activity of kaempferol in vivo. The degranulation assay, calcium imaging, the release of cytokines and chemokines on the laboratory of allergic disease 2 (LAD2) cells were used to evaluate the antiallergic effect of kaempferol in vitro. Western blot analysis was performed to investigate the DJ-1/Lyn signaling pathway and downstream molecules. Kinase activity assay, immunofluorescence, and molecular docking were conducted to confirm the influence of kaempferol on DJ-1/Lyn molecules. RESULTS: Kaempferol dose-dependently attenuated ovalbumin/IgE-induced mice paw swelling, primary MC activation from paw skin, as well as rehabilitated the hypothermia, and reduced the serum concentrations of histamine, tumor necrosis factor-alpha, interleukin-8, and monocyte chemo-attractant protein-1. Additionally, kaempferol suppressed IgE-mediated LAD2 cell degranulation and calcium fluctuation. Remarkably, kaempferol was found to bind with DJ-1 protein, and initially prevented DJ-1 from translocating to the plasma membrane, thereby inhibited full activation of Lyn, and eventually restrained those receptor-distal signaling molecules, involved Syk, Btk, PLCγ, IP3R, PKC, MAPKs, Akt and NF-κB. CONCLUSION: Kaempferol could be used as a DJ-1 modulator for preventing MC-mediated allergic disorders through attenuating Lyn activation.


Subject(s)
Anaphylaxis/drug therapy , Anti-Allergic Agents/pharmacology , Kaempferols/pharmacology , Mast Cells/drug effects , Anaphylaxis/immunology , Animals , Cell Degranulation/drug effects , Cell Line , Dose-Response Relationship, Drug , Humans , Immunoglobulin E/adverse effects , Immunoglobulin E/metabolism , Kaempferols/chemistry , Male , Mast Cells/immunology , Mast Cells/metabolism , Mice, Inbred C57BL , Molecular Docking Simulation , Ovalbumin/toxicity , Passive Cutaneous Anaphylaxis/drug effects , Phospholipase C gamma/metabolism , Protein Deglycase DJ-1/metabolism , Receptors, IgE/metabolism , Signal Transduction/drug effects , src-Family Kinases/metabolism
16.
Curr Allergy Asthma Rep ; 20(11): 65, 2020 08 24.
Article in English | MEDLINE | ID: mdl-32830295

ABSTRACT

PURPOSE OF REVIEW: Over the last 13 years, the genetic etiologies have been determined for multiple conditions causing elevated serum IgE, infection susceptibilities, and variable other features. In this review, we discuss the clinical presentation, laboratory features, and genetics of these diseases caused by mutations in STAT3, DOCK8, PGM3, IL6ST, ZNF341, IL6R, IL6ST, CARD11, and CARD14, with particular focus given to STAT3LOF and DOCK8 deficiency. RECENT FINDINGS: Defining the phenotype of each of these syndromes with high IgE and infection susceptibility shows that some have a pronounced connective tissue phenotype such as STAT3LOF and IL6ST deficiency, some have worse viral susceptibility such as DOCK8 deficiency and heterozygous LOF CARD11, and some have more severe allergy and eczema such as LOF CARD14. Studying these distinct but overlapping monogenic diseases will allow a better understanding of more common disease processes such as allergy, eczema, infection susceptibility, scoliosis, and aneurysm.


Subject(s)
Disease Susceptibility/immunology , Immunoglobulin E/adverse effects , Infections/immunology , Child , Female , Humans , Immunoglobulin E/blood , Male
17.
Nature ; 582(7811): 265-270, 2020 06.
Article in English | MEDLINE | ID: mdl-32499653

ABSTRACT

Approximately one-third of the world's population suffers from allergies1. Exposure to allergens crosslinks immunoglobulin E (IgE) antibodies that are bound to mast cells and basophils, triggering the release of inflammatory mediators, including histamine2. Although IgE is absolutely required for allergies, it is not understood why total and allergen-specific IgE concentrations do not reproducibly correlate with allergic disease3-5. It is well-established that glycosylation of IgG dictates its effector function and has disease-specific patterns. However, whether IgE glycans differ in disease states or affect biological activity is completely unknown6. Here we perform an unbiased examination of glycosylation patterns of total IgE from individuals with a peanut allergy and from non-atopic individuals without allergies. Our analysis reveals an increase in sialic acid content on total IgE from individuals with a peanut allergy compared with non-atopic individuals. Removal of sialic acid from IgE attenuates effector-cell degranulation and anaphylaxis in several functional models of allergic disease. Therapeutic interventions-including removing sialic acid from cell-bound IgE with a neuraminidase enzyme targeted towards the IgE receptor FcεRI, and administering asialylated IgE-markedly reduce anaphylaxis. Together, these results establish IgE glycosylation, and specifically sialylation, as an important regulator of allergic disease.


Subject(s)
Immunoglobulin E/chemistry , Immunoglobulin E/immunology , N-Acetylneuraminic Acid/analysis , Peanut Hypersensitivity/immunology , Peanut Hypersensitivity/pathology , Adolescent , Adult , Aged , Allergens/immunology , Anaphylaxis/immunology , Animals , Case-Control Studies , Cell Degranulation/immunology , Child , Child, Preschool , Female , Glycosylation , Humans , Immunoglobulin E/adverse effects , Immunoglobulin E/pharmacology , Infant , Infant, Newborn , Male , Mice , Middle Aged , Models, Immunological , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/metabolism , Neuraminidase/metabolism , Receptors, IgE/metabolism , Young Adult
19.
Front Immunol ; 11: 50, 2020.
Article in English | MEDLINE | ID: mdl-32063904

ABSTRACT

Background: As the number of allergic disease increases, studies to identify new treatments take on new urgency. Epigallocatechin gallate (EGCG), a major component of green tea, has been shown to possess a wide range of pharmacological properties, including anti-inflammation and anti-viral infection. In previous study, gallic acid (GA), a part of EGCG, has shown anti-allergic inflammatory effect. To improve on preliminary evidence that GA has allergy mitigating effect, we designed SG-SP1 based on GA, and aimed to assess the effects of SG-SP1 on mast cell-mediated allergic inflammation using various animal and in vitro models. Methods: For in vitro experiments, various types of IgE-stimulated mast cells (RBL-2H3: mast cell-like basophilic leukemia cells, and primary cultured peritoneal and bone marrow-derived mast cells) were used to determine the role of SG-SP1 (0.1-1 nM). Immunoglobulin (Ig) E-induced passive cutaneous anaphylaxis and ovalbumin-induced systemic anaphylaxis, standard animal models for immediate-type hypersensitivity were also used. Results: For in vitro, SG-SP1 reduced degranulation of mast cells by down-regulating intracellular calcium levels in a concentration-dependent manner. SG-SP1 decreased expression and secretion of inflammatory cytokines in activated mast cells. This suppressive effect was associated with inhibition of the phosphorylation of Lyn, Syk and Akt, and the nuclear translocation of nuclear factor-κB. Due to the strong inhibitory effect of SG-SP1 on Lyn, the known upstream signaling to FcεRI-dependent pathway, we confirmed the direct binding of SG-SP1 to FcεRI, a high affinity IgE receptor by surface plasmon resonance experiment. Oral administration of SG-SP1 hindered allergic symptoms of both anaphylaxis models evidenced by reduction of hypothermia, serum IgE, ear thickness, and tissue pigmentation. This inhibition was mediated by the reductions in serum histamine and interleukin-4. Conclusions: We determined that SG-SP1 directly interacts with FcεRI and propose SG-SP1 as a therapeutic candidate for mast cell-mediated allergic inflammatory disorders via inhibition of FcεRI signaling.


Subject(s)
Anaphylaxis/drug therapy , Anaphylaxis/metabolism , Anti-Inflammatory Agents/administration & dosage , Gallic Acid/analogs & derivatives , Gallic Acid/administration & dosage , Mast Cells/metabolism , Passive Cutaneous Anaphylaxis/drug effects , Receptors, IgE/antagonists & inhibitors , Anaphylaxis/chemically induced , Animals , Anti-Inflammatory Agents/metabolism , Calcium/metabolism , Calcium Signaling/drug effects , Cell Degranulation/drug effects , Cell Survival/drug effects , Cells, Cultured , Gallic Acid/metabolism , Immunoglobulin E/adverse effects , Inflammation/immunology , Inflammation/metabolism , Male , Mast Cells/drug effects , Mice , Mice, Inbred ICR , Ovalbumin/adverse effects , Rats , Rats, Sprague-Dawley , Receptors, IgE/metabolism
20.
MAbs ; 12(1): 1685349, 2020.
Article in English | MEDLINE | ID: mdl-31769737

ABSTRACT

IgE monoclonal antibodies hold great potential for cancer therapy. Preclinical in vivo systems, particularly those in which the antibody recognizes the host species target antigen and binds to cognate Fc receptors, are often the closest approximation to human exposure and represent a key challenge for evaluating the safety of antibody-based therapies. We sought to develop an immunocompetent rat system to assess the safety of a rodent anti-tumor IgE, as a surrogate for the human therapeutic candidate. We generated a rat IgE against the human tumor-associated antigen chondroitin sulfate proteoglycan 4 (CSPG4) and cross-reactive for the rat antigen. We analyzed CSPG4 distribution in normal rat and human tissues and investigated the in vivo safety of the antibody by monitoring clinical signs and molecular biomarkers after systemic administration to immunocompetent rats. Human and rat CSPG4 expression in normal tissues were comparable. Animals receiving antibody exhibited transient mild to moderate adverse events accompanied by mild elevation of serum tryptase, but not of angiotensin II or cytokines implicated in allergic reactions or cytokine storm. In the long term, repeated antibody administration was well tolerated, with no changes in animal body weight, liver and kidney functions or blood cell counts. This model provides preclinical support for the safety profiling of IgE therapeutic antibodies. Due to the comparable antigen tissue distribution in human and rat, this model may also comprise an appropriate tool for proof-of-concept safety evaluations of different treatment approaches targeting CSPG4.


Subject(s)
Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/administration & dosage , Chondroitin Sulfate Proteoglycans/immunology , Immunoglobulin E/administration & dosage , Membrane Proteins/immunology , Recombinant Fusion Proteins/administration & dosage , Animals , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents, Immunological/adverse effects , Cell Line, Tumor , Cross Reactions , Female , Humans , Immunization, Secondary , Immunocompetence , Immunoglobulin E/adverse effects , Mice , Rats , Recombinant Fusion Proteins/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...