1
|
Mak HW, Au EY, Yeung MH, Chiang V, Lam K, Wong JC, Yeung HH, Chan EY, Lau CS, Li PH. Extending the role of tryptase in perioperative anaphylaxis: Predicting positive results in basophil activation tests. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2024; 3:100297. [PMID: 39176077 PMCID: PMC11339248 DOI: 10.1016/j.jacig.2024.100297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/02/2024] [Accepted: 04/05/2024] [Indexed: 08/24/2024]
Abstract
Background Basophil activation tests (BATs) are useful in identifying culprits of perioperative anaphylaxis (PA), but their utility remains limited due to technical limitations, cost, and availability. Being able to prioritize patients with likely higher yields for BAT would be useful in reducing costs and manpower. Objective We sought to investigate whether tryptase levels and clinical parameters may be useful for selecting patients for BATs. Methods We performed a 10-year retrospective study in Hong Kong to investigate the performance of BATs associated with tryptase levels (taking during PA) and other clinical parameters. Results Of 90 patients, 70 (77.8%) showed significant tryptase level elevation and 37 (41.1%) had a positive BAT result. BAT-positive patients presented with significantly higher absolute levels (15.9 μg/L vs 9.1 μg/L; P = .018), absolute elevation (12.8 μg/L vs 7.1 μg/L; P = .012), and fold elevation (5.6- vs 4.1-fold; P = .014) of acute tryptase than did BAT-negative patients. Among patients with positive BAT result, 94.6% (35 of 37) demonstrated elevated acute tryptase, significantly more than the BAT-negative group (66.0%; P < .001). In regression analysis, tryptase elevation was the sole significant factor correlated to BAT positivity (odds ratio, 10.14; 95% CI, 2.15-47.85; P = .003). Overall, elevated acute tryptase demonstrated a sensitivity of 94.7% and a negative predictive value of 90.0% in predicting positive results with BATs. Conclusions We observed that tryptase elevation is a very sensitive predictor of BAT positivity among patients with identified culprits of PA. Acute elevation of tryptase would not only aid in confirming anaphylaxis but may also help guide the decision toward selecting labor-intensive and costly in vitro tests such as BATs.
Collapse
Affiliation(s)
- Hugo W.F. Mak
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Queen Mary Hospital, Hong Kong
| | - Elaine Y.L. Au
- Division of Clinical Immunology, Department of Pathology, Queen Mary Hospital, Hong Kong
| | - Maegan H.Y. Yeung
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Queen Mary Hospital, Hong Kong
| | - Valerie Chiang
- Division of Clinical Immunology, Department of Pathology, Queen Mary Hospital, Hong Kong
| | - Ki Lam
- Division of Clinical Immunology, Department of Pathology, Queen Mary Hospital, Hong Kong
| | - Jane C.Y. Wong
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Queen Mary Hospital, Hong Kong
| | - Heather H.F. Yeung
- Division of Clinical Immunology, Department of Pathology, Queen Mary Hospital, Hong Kong
| | - Eric Y.T. Chan
- Division of Clinical Immunology, Department of Pathology, Queen Mary Hospital, Hong Kong
| | - Chak-sing Lau
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Queen Mary Hospital, Hong Kong
| | - Philip H. Li
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Queen Mary Hospital, Hong Kong
| |
Collapse
|
2
|
Koren A, Dejanović L, Rijavec M, Kopač P, Bizjak M, Zidarn M, Košnik M, Korošec P. Autoimmune Mast Cell Activation Test as a Diagnostic Tool in Chronic Spontaneous Urticaria. Int J Mol Sci 2024; 25:9281. [PMID: 39273229 PMCID: PMC11395619 DOI: 10.3390/ijms25179281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 09/15/2024] Open
Abstract
Chronic spontaneous urticaria (CSU) is associated with skin mast cell activation, and its triggering mechanisms are not completely elucidated. Evidence suggests an autoimmune component of CSU. Our aim was to assess the usefulness of an autoimmune mast cell activation test (aiMAT) for diagnosing and differentiating CSU into different subtypes. We enrolled 43 patients with active, uncontrolled CSU before starting treatment with omalizumab and 15 controls. Patients were evaluated based on omalizumab response. aiMATs were performed using non-IgE-sensitized (NS) or myeloma IgE-sensitized (S) LAD2 cells, which were then stimulated with CSU/control sera (25 µL and 10 µL). The expression of CD63 was assessed with flow cytometry. CD63 response on NS-LAD2 was significantly increased in CSU patients compared to controls after the stimulation with 25 µL CSU/control sera (p = 0.0007) and with 10 µL CSU/control sera (p = 0.0001). The ROC curve analysis demonstrated an area under the curve (AUC) of 0.82. The cutoff for autoimmune-non-IgE-sensitized-MAT was 40.3% CD63+ LAD2, which resulted in 73.3% sensitivity and 81.4% specificity. CD63 response on S-LAD2 was significantly increased in CSU patients compared to controls after the stimulation with 25 µL CSU/control sera (p = 0.03). The ROC curve analysis demonstrated an AUC of 0.66. The cutoff for the autoimmune-myeloma IgE-sensitized-MAT was 58.4% CD63+ cells, which resulted in 62.8% sensitivity and 66.7% specificity. Overall, 36 out of 43 (84%) patients responded to omalizumab, and 7 (16%) were nonresponders. We found no differences between LAD2 CD63 response and response to omalizumab. In conclusion, aiMAT could represent a new diagnostic tool in CSU. Additional studies are needed to evaluate the potential benefits during omalizumab therapy.
Collapse
Affiliation(s)
- Ana Koren
- University Clinic of Respiratory and Allergic Diseases Golnik, 4204 Golnik, Slovenia
| | - Luka Dejanović
- University Clinic of Respiratory and Allergic Diseases Golnik, 4204 Golnik, Slovenia
| | - Matija Rijavec
- University Clinic of Respiratory and Allergic Diseases Golnik, 4204 Golnik, Slovenia
- Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Peter Kopač
- University Clinic of Respiratory and Allergic Diseases Golnik, 4204 Golnik, Slovenia
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Mojca Bizjak
- University Clinic of Respiratory and Allergic Diseases Golnik, 4204 Golnik, Slovenia
| | - Mihaela Zidarn
- University Clinic of Respiratory and Allergic Diseases Golnik, 4204 Golnik, Slovenia
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Mitja Košnik
- University Clinic of Respiratory and Allergic Diseases Golnik, 4204 Golnik, Slovenia
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Peter Korošec
- University Clinic of Respiratory and Allergic Diseases Golnik, 4204 Golnik, Slovenia
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
3
|
Jordan J, Levy JH, Gonzalez-Estrada A. Perioperative anaphylaxis: updates on pathophysiology. Curr Opin Allergy Clin Immunol 2024; 24:183-188. [PMID: 38743470 DOI: 10.1097/aci.0000000000000994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
PURPOSE OF REVIEW Perioperative anaphylaxis has historically been attributed to IgE/FcεRI-mediated reactions; there is now recognition of allergic and nonallergic triggers encompassing various reactions beyond IgE-mediated responses. This review aims to present recent advancements in knowledge regarding the mechanisms and pathophysiology of perioperative anaphylaxis. RECENT FINDINGS Emerging evidence highlights the role of the mast-cell related G-coupled protein receptor X2 pathway in direct mast cell degranulation, shedding light on previously unknown mechanisms. This pathway, alongside traditional IgE/FcεRI-mediated reactions, contributes to the complex nature of anaphylactic reactions. Investigations into the microbiota-anaphylaxis connection are ongoing, with potential implications for future treatment strategies. While serum tryptase levels serve as mast cell activation indicators, identifying triggers remains challenging. A range of mediators have been associated with anaphylaxis, including vasoactive peptides, proteases, lipid molecules, cytokines, chemokines, interleukins, complement components, and coagulation factors. SUMMARY Further understanding of clinical endotypes and the microenvironment where anaphylactic reactions unfold is essential for standardizing mediator testing and characterization in perioperative anaphylaxis. Ongoing research aims to elucidate the mechanisms, pathways, and mediators involved across multiple organ systems, including the cardiovascular, respiratory, and integumentary systems, which will be crucial for improving patient outcomes.
Collapse
Affiliation(s)
- Justin Jordan
- TMC Health Medical Education Program, Tucson, Arizona
| | - Jerrold H Levy
- Departments of Anesthesiology, Critical Care, and Surgery, Duke University School of Medicine, Durham, North Carolina
| | | |
Collapse
|
4
|
Starshinova A, Borozinets A, Kulpina A, Sereda V, Rubinstein A, Kudryavtsev I, Kudlay D. Bronchial Asthma and COVID-19: Etiology, Pathological Triggers, and Therapeutic Considerations. PATHOPHYSIOLOGY 2024; 31:269-287. [PMID: 38921725 PMCID: PMC11206645 DOI: 10.3390/pathophysiology31020020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
Bronchial asthma (BA) continues to be a difficult disease to diagnose. Various factors have been described in the development of BA, but to date, there is no clear evidence for the etiology of this chronic disease. The emergence of COVID-19 has contributed to the pandemic course of asthma and immunologic features. However, there are no unambiguous data on asthma on the background and after COVID-19. There is correlation between various trigger factors that provoke the development of bronchial asthma. It is now obvious that the SARS-CoV-2 virus is one of the provoking factors. COVID-19 has affected the course of asthma. Currently, there is no clear understanding of whether asthma progresses during or after COVID-19 infection. According to the results of some studies, a significant difference was identified between the development of asthma in people after COVID-19. Mild asthma and moderate asthma do not increase the severity of COVID-19 infection. Nevertheless, oral steroid treatment and hospitalization for severe BA were associated with higher COVID-19 severity. The influence of SARS-CoV-2 infection is one of the protective factors. It causes the development of severe bronchial asthma. The accumulated experience with omalizumab in patients with severe asthma during COVID-19, who received omalizumab during the pandemic, has strongly suggested that continued treatment with omalizumab is safe and may help prevent the severe course of COVID-19. Targeted therapy for asthma with the use of omalizumab may also help to reduce severe asthma associated with COVID-19. However, further studies are needed to prove the effect of omalizumab. Data analysis should persist, based on the results of the course of asthma after COVID-19 with varying degrees of severity.
Collapse
Affiliation(s)
- Anna Starshinova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia;
| | - Anastasia Borozinets
- Medical Department, I.M. Sechenov First Moscow State Medical University, 197022 Moscow, Russia
| | - Anastasia Kulpina
- Medical Department, Saint Petersburg State Pediatric Medical University, 194100 St. Petersburg, Russia;
| | - Vitaliy Sereda
- Medical Department, Saint Petersburg State University, 199034 St. Petersburg, Russia;
| | - Artem Rubinstein
- Department of immunology, Institution of Experimental Medicine, 197376 St. Petersburg, Russia;
| | - Igor Kudryavtsev
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia;
- Department of immunology, Institution of Experimental Medicine, 197376 St. Petersburg, Russia;
| | - Dmitry Kudlay
- Institute of Immunology FMBA of Russia, 115478 Moscow, Russia;
- Department of Pharmacognosy and Industrial Pharmacy, Faculty of Fundamental Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| |
Collapse
|
5
|
Pałgan K, Tretyn A. Platelet-activating factor as an endogenous cofactor of food anaphylaxis. Biofactors 2023; 49:976-983. [PMID: 37203358 DOI: 10.1002/biof.1956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 03/02/2023] [Indexed: 05/20/2023]
Abstract
Anaphylaxis is a severe, acute, life-threatening generalized or systemic hypersensitivity reaction. The incidence of anaphylaxis is increasing worldwide, with medications and food contributing to most cases. Physical exercise, acute infections, drugs, alcohol, and menstruation are the external cofactors associated with more severe systemic reaction. The aim of this review is to show that platelet-activating factor contributes to the development of severe anaphylactic reaction, and even to anaphylactic shock.
Collapse
Affiliation(s)
- Krzysztof Pałgan
- Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Andrzej Tretyn
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Toruń, Poland
| |
Collapse
|
6
|
Lin L, Liang Y, Cao T, Huang Y, Li W, Li J, Wang J, Peng X, Ge Y, Li Y, Li L. Transcriptome profiling and ceRNA network of small extracellular vesicles from resting and degranulated mast cells. Epigenomics 2023; 15:845-862. [PMID: 37846550 DOI: 10.2217/epi-2023-0175] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023] Open
Abstract
Aim: This study aimed to investigate the transcriptomic characteristics and interactions between competitive endogenous RNAs (ceRNAs) within small extracellular vesicles (sEVs) derived from mast cells (MCs). Methods: Transcriptome sequencing analyzed lncRNA, circRNA and mRNA expression in resting and degranulated MC-derived sEVs. Constructed ceRNA regulatory network through correlation analysis and target gene prediction. Results: Differentially expressed 1673 mRNAs, 173 lncRNAs and 531 circRNAs were observed between resting and degranulated MCs-derived sEVs. Enrichment analysis revealed involvement of neurodegeneration, infection and tumor pathways. CeRNA networks included interactions between lncRNA-miRNA, circRNA-miRNA and miRNA-mRNA, targeting genes in the hippo and wnt signaling pathways linked to tumor immune regulation. Conclusion: This study provides valuable insights into MC-sEV molecular mechanisms, offering significant data resources for further investigations.
Collapse
Affiliation(s)
- Lihui Lin
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P.R. China
| | - Yuting Liang
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow University Suzhou, Jiangsu, 215006, P.R. China
| | - Tianyu Cao
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P.R. China
| | - Yuji Huang
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P.R. China
| | - Weize Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P.R. China
| | - Jia Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P.R. China
| | - Juan Wang
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P.R. China
| | - Xia Peng
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P.R. China
| | - Yiqin Ge
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P.R. China
| | - Yanning Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P.R. China
| | - Li Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P.R. China
| |
Collapse
|
7
|
Pajulas A, Fu Y, Cheung CCL, Chu M, Cannon A, Alakhras N, Zhang J, Ulrich BJ, Nelson AS, Zhou B, Kaplan MH. Interleukin-9 promotes mast cell progenitor proliferation and CCR2-dependent mast cell migration in allergic airway inflammation. Mucosal Immunol 2023; 16:432-445. [PMID: 37172907 PMCID: PMC10482122 DOI: 10.1016/j.mucimm.2023.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023]
Abstract
Allergic asthma is a chronic lung disease characterized by airway hyperresponsiveness and cellular infiltration that is exacerbated by immunoglobulin E-dependent mast cell (MC) activation. Interleukin-9 (IL-9) promotes MC expansion during allergic inflammation but precisely how IL-9 expands tissue MCs and promotes MC function is unclear. In this report, using multiple models of allergic airway inflammation, we show that both mature MCs (mMCs) and MC progenitors (MCp) express IL-9R and respond to IL-9 during allergic inflammation. IL-9 acts on MCp in the bone marrow and lungs to enhance proliferative capacity. Furthermore, IL-9 in the lung stimulates the mobilization of CCR2+ mMC from the bone marrow and recruitment to the allergic lung. Mixed bone marrow chimeras demonstrate that these are intrinsic effects in the MCp and mMC populations. IL-9-producing T cells are both necessary and sufficient to increase MC numbers in the lung in the context of allergic inflammation. Importantly, T cell IL-9-mediated MC expansion is required for the development of antigen-induced and MC-dependent airway hyperreactivity. Collectively, these data demonstrate that T cell IL-9 induces lung MC expansion and migration by direct effects on the proliferation of MCp and the migration of mMC to mediate airway hyperreactivity.
Collapse
Affiliation(s)
- Abigail Pajulas
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA
| | - Yongyao Fu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA
| | - Cherry C L Cheung
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA
| | - Michelle Chu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA
| | - Anthony Cannon
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA
| | - Nada Alakhras
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, USA
| | - Jilu Zhang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA
| | - Benjamin J Ulrich
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA
| | - Andrew S Nelson
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA
| | - Baohua Zhou
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, USA
| | - Mark H Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA.
| |
Collapse
|
8
|
Crespo JF, Cabanillas B. Recent advances in cellular and molecular mechanisms of IgE-mediated food allergy. Food Chem 2023; 411:135500. [PMID: 36682170 DOI: 10.1016/j.foodchem.2023.135500] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 01/12/2023] [Accepted: 01/14/2023] [Indexed: 01/19/2023]
Abstract
Food allergy is a public health issue the prevalence of which is steadily increasing. New discoveries have contributed to the understanding of the molecular and cellular mechanisms that lead to IgE-mediated food allergy. Novel scientific findings have defined roles for specific cell types, such as T follicular helper cells, in induction of high-affinity IgE by B cells. Also, not only mast cells and basophils contribute to food anaphylaxis, but also other cell types, such as neutrophils and macrophages. Elucidation of mechanisms involved in sensitization to food allergens through organs including the skin is key to deepening our understanding of the "dual exposure" hypothesis, which suggests that allergic sensitization is mainly acquired through inflamed skin while the oral route induces tolerance. This review considers the latest scientific knowledge about the molecular and cellular mechanisms of IgE-mediated food allergy. It reveals crucial components involved in the sensitization and elicitation phases and emerging approaches in anaphylaxis pathophysiology.
Collapse
Affiliation(s)
- Jesus F Crespo
- Department of Allergy, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Avenida de Córdoba s/n, 28041 Madrid, Spain
| | - Beatriz Cabanillas
- Department of Allergy, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Avenida de Córdoba s/n, 28041 Madrid, Spain.
| |
Collapse
|
9
|
Bartko EA, Blom LH, Elberling J, Poulsen LK, Jensen BM. Expression of CCR8 and CCX-CKR on Basophils in Chronic Urticaria Is Amplified by IgE-Mediated Activation. Biomedicines 2023; 11:1537. [PMID: 37371632 DOI: 10.3390/biomedicines11061537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Recruitment to the local tissue and alerted phenotype are the hallmarks of basophils in chronic urticaria (CU). Chemokine receptors such as chemokine (C-C motif) receptor 4 (CCR4) or CCR8 have been studied in skin diseases, e.g., atopic dermatitis, but not in CU. In this study, we aimed to define CU's basophil homing potential and receptor profile and the effect of Omalizumab treatment on these. Unstimulated and activated (anti-IgE, fMLP, C5a, and Substance P) whole blood basophils from 11 Omalizumab-treated CU patients and 10 healthy subjects were investigated with flow cytometry. Unstimulated basophils in CU showed higher expression of the skin-associated (CCR8) and scavenger (CCX-CKR) receptors and lower expression of the lung-associated (CCR3) receptor in contrast to healthy ones. IgE-mediated activation increased the percentage of CCR8 and CCX-CKR in CU compared to healthy group and elevated the expression of the lung-associated chemokine receptor, XCR1, in all groups. A trend of augmented expression of the coagulation cascade (CD87) and fMLP (FPR1) receptors was seen on basophils in CU, while a tendency of reduced expression was seen for itch (IL-31RA) and immunotolerance (CD109) receptors. fMLP and C5a increased the expression of CCR4, CCR8, CCX-CKR, and CD87 and decreased CCR2 and CCR3, though no changes between the groups were found. In conclusion, CU basophils exhibit skin-homing potential amplified by IgE-mediated stimulation.
Collapse
Affiliation(s)
- Ewa A Bartko
- Allergy Clinic, Department of Dermatology and Allergy, Copenhagen University Hospital at Gentofte, 2900 Hellerup, Denmark
| | - Lars H Blom
- Allergy Clinic, Department of Dermatology and Allergy, Copenhagen University Hospital at Gentofte, 2900 Hellerup, Denmark
| | - Jesper Elberling
- Allergy Clinic, Department of Dermatology and Allergy, Copenhagen University Hospital at Gentofte, 2900 Hellerup, Denmark
| | - Lars K Poulsen
- Allergy Clinic, Department of Dermatology and Allergy, Copenhagen University Hospital at Gentofte, 2900 Hellerup, Denmark
| | - Bettina M Jensen
- Allergy Clinic, Department of Dermatology and Allergy, Copenhagen University Hospital at Gentofte, 2900 Hellerup, Denmark
| |
Collapse
|
10
|
Basophils and anaphylaxis. REVUE FRANÇAISE D'ALLERGOLOGIE 2023. [DOI: 10.1016/j.reval.2023.103291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
11
|
Dramburg S, Hilger C, Santos AF, de Las Vecillas L, Aalberse RC, Acevedo N, Aglas L, Altmann F, Arruda KL, Asero R, Ballmer-Weber B, Barber D, Beyer K, Biedermann T, Bilo MB, Blank S, Bosshard PP, Breiteneder H, Brough HA, Bublin M, Campbell D, Caraballo L, Caubet JC, Celi G, Chapman MD, Chruszcz M, Custovic A, Czolk R, Davies J, Douladiris N, Eberlein B, Ebisawa M, Ehlers A, Eigenmann P, Gadermaier G, Giovannini M, Gomez F, Grohman R, Guillet C, Hafner C, Hamilton RG, Hauser M, Hawranek T, Hoffmann HJ, Holzhauser T, Iizuka T, Jacquet A, Jakob T, Janssen-Weets B, Jappe U, Jutel M, Kalic T, Kamath S, Kespohl S, Kleine-Tebbe J, Knol E, Knulst A, Konradsen JR, Korošec P, Kuehn A, Lack G, Le TM, Lopata A, Luengo O, Mäkelä M, Marra AM, Mills C, Morisset M, Muraro A, Nowak-Wegrzyn A, Nugraha R, Ollert M, Palosuo K, Pastorello EA, Patil SU, Platts-Mills T, Pomés A, Poncet P, Potapova E, Poulsen LK, Radauer C, Radulovic S, Raulf M, Rougé P, Sastre J, Sato S, Scala E, Schmid JM, Schmid-Grendelmeier P, Schrama D, Sénéchal H, Traidl-Hoffmann C, Valverde-Monge M, van Hage M, van Ree R, Verhoeckx K, Vieths S, Wickman M, Zakzuk J, Matricardi PM, Hoffmann-Sommergruber K. EAACI Molecular Allergology User's Guide 2.0. Pediatr Allergy Immunol 2023; 34 Suppl 28:e13854. [PMID: 37186333 DOI: 10.1111/pai.13854] [Citation(s) in RCA: 72] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/05/2022] [Indexed: 05/17/2023]
Abstract
Since the discovery of immunoglobulin E (IgE) as a mediator of allergic diseases in 1967, our knowledge about the immunological mechanisms of IgE-mediated allergies has remarkably increased. In addition to understanding the immune response and clinical symptoms, allergy diagnosis and management depend strongly on the precise identification of the elicitors of the IgE-mediated allergic reaction. In the past four decades, innovations in bioscience and technology have facilitated the identification and production of well-defined, highly pure molecules for component-resolved diagnosis (CRD), allowing a personalized diagnosis and management of the allergic disease for individual patients. The first edition of the "EAACI Molecular Allergology User's Guide" (MAUG) in 2016 rapidly became a key reference for clinicians, scientists, and interested readers with a background in allergology, immunology, biology, and medicine. Nevertheless, the field of molecular allergology is moving fast, and after 6 years, a new EAACI Taskforce was established to provide an updated document. The Molecular Allergology User's Guide 2.0 summarizes state-of-the-art information on allergen molecules, their clinical relevance, and their application in diagnostic algorithms for clinical practice. It is designed for both, clinicians and scientists, guiding health care professionals through the overwhelming list of different allergen molecules available for testing. Further, it provides diagnostic algorithms on the clinical relevance of allergenic molecules and gives an overview of their biology, the basic mechanisms of test formats, and the application of tests to measure allergen exposure.
Collapse
Affiliation(s)
- Stephanie Dramburg
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Christiane Hilger
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Alexandra F Santos
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
- Children's Allergy Service, Evelina London, Guy's and St Thomas' Hospital, London, United Kingdom
| | | | - Rob C Aalberse
- Sanquin Research, Dept Immunopathology, University of Amsterdam, Amsterdam, The Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Nathalie Acevedo
- Institute for Immunological Research, University of Cartagena, Cartagena de Indias, Colombia, Colombia
| | - Lorenz Aglas
- Department of Biosciences and Medical Biology, Paris Lodron University Salzburg, Salzburg, Austria
| | - Friedrich Altmann
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Karla L Arruda
- Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Sao Paulo, Brasil, Brazil
| | - Riccardo Asero
- Ambulatorio di Allergologia, Clinica San Carlo, Paderno Dugnano, Italy
| | - Barbara Ballmer-Weber
- Klinik für Dermatologie und Allergologie, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Domingo Barber
- Institute of Applied Molecular Medicine Nemesio Diez (IMMAND), Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
- RETIC ARADyAL and RICORS Enfermedades Inflamatorias (REI), Madrid, Spain
| | - Kirsten Beyer
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Tilo Biedermann
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University Munich, Munich, Germany
| | - Maria Beatrice Bilo
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
- Allergy Unit Department of Internal Medicine, University Hospital Ospedali Riuniti di Ancona, Torrette, Italy
| | - Simon Blank
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany
| | - Philipp P Bosshard
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Heimo Breiteneder
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Helen A Brough
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
- Children's Allergy Service, Evelina London, Guy's and St Thomas' Hospital, London, United Kingdom
| | - Merima Bublin
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Dianne Campbell
- Department of Allergy and Immunology, Children's Hospital at Westmead, Sydney Children's Hospitals Network, Sydney, New South Wales, Australia
- Child and Adolescent Health, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Luis Caraballo
- Institute for Immunological Research, University of Cartagena, Cartagena de Indias, Colombia, Colombia
| | - Jean Christoph Caubet
- Pediatric Allergy Unit, Department of Child and Adolescent, University Hospitals of Geneva, Geneva, Switzerland
| | - Giorgio Celi
- Centro DH Allergologia e Immunologia Clinica ASST- MANTOVA (MN), Mantova, Italy
| | | | - Maksymilian Chruszcz
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, USA
| | - Adnan Custovic
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Rebecca Czolk
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Janet Davies
- Queensland University of Technology, Centre for Immunology and Infection Control, School of Biomedical Sciences, Herston, Queensland, Australia
- Metro North Hospital and Health Service, Emergency Operations Centre, Herston, Queensland, Australia
| | - Nikolaos Douladiris
- Allergy Department, 2nd Paediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Bernadette Eberlein
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University Munich, Munich, Germany
| | - Motohiro Ebisawa
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization, Sagamihara National Hospital, Kanagawa, Japan
| | - Anna Ehlers
- Chemical Biology and Drug Discovery, Utrecht University, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Immunology and Dermatology/ Allergology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Philippe Eigenmann
- Pediatric Allergy Unit, Department of Child and Adolescent, University Hospitals of Geneva, Geneva, Switzerland
| | - Gabriele Gadermaier
- Department of Biosciences and Medical Biology, Paris Lodron University Salzburg, Salzburg, Austria
| | - Mattia Giovannini
- Allergy Unit, Department of Pediatrics, Meyer Children's University Hospital, Florence, Italy
| | - Francisca Gomez
- Allergy Unit IBIMA-Hospital Regional Universitario de Malaga, Malaga, Spain
- Spanish Network for Allergy research RETIC ARADyAL, Malaga, Spain
| | - Rebecca Grohman
- NYU Langone Health, Department of Internal Medicine, New York, New York, USA
| | - Carole Guillet
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Christine Hafner
- Department of Dermatology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, St. Poelten, Austria
| | - Robert G Hamilton
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael Hauser
- Department of Biosciences and Medical Biology, Paris Lodron University Salzburg, Salzburg, Austria
| | - Thomas Hawranek
- Department of Dermatology and Allergology, Paracelsus Private Medical University, Salzburg, Austria
| | - Hans Jürgen Hoffmann
- Institute for Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
- Department of Respiratory Diseases and Allergy, Aarhus University Hospital, Aarhus, Denmark
| | | | - Tomona Iizuka
- Laboratory of Protein Science, Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Alain Jacquet
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Thilo Jakob
- Department of Dermatology and Allergology, University Medical Center, Justus Liebig University Gießen, Gießen, Germany
| | - Bente Janssen-Weets
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Odense Research Center for Anaphylaxis, University of Southern Denmark, Odense, Denmark
| | - Uta Jappe
- Division of Clinical and Molecular Allergology, Priority Research Area Asthma and Allergy, Research Center Borstel, Borstel, Germany
- Leibniz Lung Center, Airway Research Center North (ARCN), Member of the German Center for Lung Research, Germany
- Interdisciplinary Allergy Outpatient Clinic, Dept. of Pneumology, University of Lübeck, Lübeck, Germany
| | - Marek Jutel
- Department of Clinical Immunology, Wroclaw Medical University, Wroclaw, Poland
| | - Tanja Kalic
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
- Department of Dermatology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, St. Poelten, Austria
| | - Sandip Kamath
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia
- Molecular Allergy Research Laboratory, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Queensland, Australia
| | - Sabine Kespohl
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr- Universität Bochum, Bochum, Germany
| | - Jörg Kleine-Tebbe
- Allergy & Asthma Center Westend, Outpatient Clinic and Clinical Research Center, Berlin, Germany
| | - Edward Knol
- Department of Immunology and Dermatology/ Allergology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - André Knulst
- Department of Immunology and Dermatology/ Allergology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jon R Konradsen
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Pediatric Allergy and Pulmonology Unit at Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Peter Korošec
- University Clinic of Respiratory and Allergic Diseases Golnik, Golnik, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Annette Kuehn
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Gideon Lack
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
- Children's Allergy Service, Evelina London, Guy's and St Thomas' Hospital, London, United Kingdom
| | - Thuy-My Le
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Immunology and Dermatology/ Allergology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Andreas Lopata
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia
- Molecular Allergy Research Laboratory, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Queensland, Australia
| | - Olga Luengo
- RETIC ARADyAL and RICORS Enfermedades Inflamatorias (REI), Madrid, Spain
- Allergy Section, Internal Medicine Department, Vall d'Hebron University Hospital, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mika Mäkelä
- Division of Allergy, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Pediatric Department, Skin and Allergy Hospital, Helsinki University Central Hospital, Helsinki, Finland
| | | | - Clare Mills
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | | | - Antonella Muraro
- Food Allergy Referral Centre, Department of Woman and Child Health, Padua University Hospital, Padua, Italy
| | - Anna Nowak-Wegrzyn
- Division of Pediatric Allergy and Immunology, NYU Grossman School of Medicine, Hassenfeld Children's Hospital, New York, New York, USA
- Department of Pediatrics, Gastroenterology and Nutrition, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | - Roni Nugraha
- Molecular Allergy Research Laboratory, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Queensland, Australia
- Department of Aquatic Product Technology, Faculty of Fisheries and Marine Science, IPB University, Bogor, Indonesia
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Odense Research Center for Anaphylaxis, University of Southern Denmark, Odense, Denmark
| | - Kati Palosuo
- Department of Allergology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | | | - Sarita Ulhas Patil
- Division of Rheumatology, Allergy and Immunology, Departments of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Division of Allergy and Immunology, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Thomas Platts-Mills
- Division of Allergy and Clinical Immunology, University of Virginia, Charlottesville, Virginia, USA
| | | | - Pascal Poncet
- Institut Pasteur, Immunology Department, Paris, France
- Allergy & Environment Research Team Armand Trousseau Children Hospital, APHP, Paris, France
| | - Ekaterina Potapova
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Lars K Poulsen
- Allergy Clinic, Department of Dermatology and Allergy, Copenhagen University Hospital-Herlev and Gentofte, Copenhagen, Denmark
| | - Christian Radauer
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Suzana Radulovic
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
- Children's Allergy Service, Evelina London, Guy's and St Thomas' Hospital, London, United Kingdom
| | - Monika Raulf
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr- Universität Bochum, Bochum, Germany
| | - Pierre Rougé
- UMR 152 PharmaDev, IRD, Université Paul Sabatier, Faculté de Pharmacie, Toulouse, France
| | - Joaquin Sastre
- Allergy Service, Fundación Jiménez Díaz; CIBER de Enfermedades Respiratorias (CIBERES); Faculty of Medicine, Universidad Autonoma de Madrid, Madrid, Spain
| | - Sakura Sato
- Allergy Department, 2nd Paediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Enrico Scala
- Clinical and Laboratory Molecular Allergy Unit - IDI- IRCCS, Fondazione L M Monti Rome, Rome, Italy
| | - Johannes M Schmid
- Department of Respiratory Diseases and Allergy, Aarhus University Hospital, Aarhus, Denmark
| | - Peter Schmid-Grendelmeier
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Christine Kühne Center for Allergy Research and Education CK-CARE, Davos, Switzerland
| | - Denise Schrama
- Centre of Marine Sciences (CCMAR), Universidade do Algarve, Faro, Portugal
| | - Hélène Sénéchal
- Allergy & Environment Research Team Armand Trousseau Children Hospital, APHP, Paris, France
| | - Claudia Traidl-Hoffmann
- Christine Kühne Center for Allergy Research and Education CK-CARE, Davos, Switzerland
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Marcela Valverde-Monge
- Allergy Service, Fundación Jiménez Díaz; CIBER de Enfermedades Respiratorias (CIBERES); Faculty of Medicine, Universidad Autonoma de Madrid, Madrid, Spain
| | - Marianne van Hage
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Ronald van Ree
- Department of Experimental Immunology and Department of Otorhinolaryngology, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Kitty Verhoeckx
- Department of Immunology and Dermatology/ Allergology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Stefan Vieths
- Division of Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Magnus Wickman
- Department of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Josefina Zakzuk
- Institute for Immunological Research, University of Cartagena, Cartagena de Indias, Colombia, Colombia
| | - Paolo M Matricardi
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
12
|
Alakhras NS, Shin J, Smith SA, Sinn AL, Zhang W, Hwang G, Sjoerdsma J, Bromley EK, Pollok KE, Bilgicer B, Kaplan MH. Peanut allergen inhibition prevents anaphylaxis in a humanized mouse model. Sci Transl Med 2023; 15:eadd6373. [PMID: 36753563 PMCID: PMC10205092 DOI: 10.1126/scitranslmed.add6373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 01/17/2023] [Indexed: 02/10/2023]
Abstract
Peanut-induced allergy is an immunoglobulin E (IgE)-mediated type I hypersensitivity reaction that manifests symptoms ranging from local edema to life-threatening anaphylaxis. Although there are treatments for symptoms in patients with allergies resulting from allergen exposure, there are few preventive therapies other than strict dietary avoidance or oral immunotherapy, neither of which are successful in all patients. We have previously designed a covalent heterobivalent inhibitor (cHBI) that binds in an allergen-specific manner as a preventive for allergic reactions. Building on previous in vitro testing, here, we developed a humanized mouse model to test cHBI efficacy in vivo. Nonobese diabetic-severe combined immunodeficient γc-deficient mice expressing transgenes for human stem cell factor, granulocyte-macrophage colony-stimulating factor, and interleukin-3 developed mature functional human mast cells in multiple tissues and displayed robust anaphylactic reactions when passively sensitized with patient-derived IgE monoclonal antibodies specific for peanut Arachis hypogaea 2 (Ara h 2). The allergic response in humanized mice was IgE dose dependent and was mediated by human mast cells. Using this humanized mouse model, we showed that cHBI prevented allergic reactions for more than 2 weeks when administered before allergen exposure. cHBI also prevented fatal anaphylaxis and attenuated allergic reactions when administered shortly after the onset of symptoms. cHBI impaired mast cell degranulation in vivo in an allergen-specific manner. cHBI rescued the mice from lethal anaphylactic responses during oral Ara h 2 allergen-induced anaphylaxis. Together, these findings suggest that cHBI has the potential to be an effective preventative for peanut-specific allergic responses in patients.
Collapse
Affiliation(s)
- Nada S. Alakhras
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Jaeho Shin
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556
| | - Scott A. Smith
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Anthony L. Sinn
- In Vivo Therapeutics Core, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, Indiana 46202
| | - Wenwu Zhang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Gyoyeon Hwang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556
| | - Jenna Sjoerdsma
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556
| | - Emily K. Bromley
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556
| | - Karen E. Pollok
- In Vivo Therapeutics Core, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, Indiana 46202
- Department of Pediatrics, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Basar Bilgicer
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556
| | - Mark H. Kaplan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Department of Pediatrics, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202
| |
Collapse
|
13
|
Blom LH, Bartko EA, Møller TKR, Poulsen LK, Jensen BM. FcεRI-activated basophils express CCR4, CCR8, CCR9, CCX-CKR and XCR1. Allergy 2023; 78:539-543. [PMID: 35986598 DOI: 10.1111/all.15488] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 08/01/2022] [Accepted: 08/15/2022] [Indexed: 02/01/2023]
Affiliation(s)
- Lars H Blom
- Allergy Clinic, Department of Dermatology and Allergy, Copenhagen University Hospital Herlev and Gentofte, Copenhagen, Denmark
| | - Ewa A Bartko
- Allergy Clinic, Department of Dermatology and Allergy, Copenhagen University Hospital Herlev and Gentofte, Copenhagen, Denmark
| | - Thea K R Møller
- Allergy Clinic, Department of Dermatology and Allergy, Copenhagen University Hospital Herlev and Gentofte, Copenhagen, Denmark
| | - Lars K Poulsen
- Allergy Clinic, Department of Dermatology and Allergy, Copenhagen University Hospital Herlev and Gentofte, Copenhagen, Denmark
| | - Bettina M Jensen
- Allergy Clinic, Department of Dermatology and Allergy, Copenhagen University Hospital Herlev and Gentofte, Copenhagen, Denmark
| |
Collapse
|
14
|
Dejoux A, de Chaisemartin L, Bruhns P, Longrois D, Gouel-Chéron A. Neuromuscular blocking agent induced hypersensitivity reaction exploration: an update. Ugeskr Laeger 2023; 40:95-104. [PMID: 36301083 DOI: 10.1097/eja.0000000000001765] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
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.
Collapse
Affiliation(s)
- Alice Dejoux
- From the Institut Pasteur, Université de Paris, Unit of Antibodies in Therapy and Pathology, Inserm UMR1222 (AD, LdC, PB, AGC), Immunology Department, DMU BIOGEM, Bichat Hospital, AP-HP (LdC), Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Châtenay-Malabry (LdC), Anaesthesiology and Critical Care Medicine Department, DMU PARABOL, Bichat Hospital, AP-HP (DL, AGC), Université de Paris, FHU PROMICE (DL), Anaesthesiology and Critical Care Medicine Department, DMU PARABOL, Bichat-Claude Bernard and Louis Mourier Hospitals, APHP (DL), INSERM1148, Paris, France (DL), and Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA (AGC)
| | | | | | | | | |
Collapse
|
15
|
Selnø ATH, Sumbayev VV, Gibbs BF. IgE-dependent human basophil responses are inversely associated with the sarcoplasmic reticulum Ca 2+-ATPase (SERCA). Front Immunol 2023; 13:1052290. [PMID: 36685580 PMCID: PMC9846818 DOI: 10.3389/fimmu.2022.1052290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023] Open
Abstract
Basophils crucially contribute to allergies and other Th2-driven diseases by rapidly releasing inflammatory and immunomodulatory mediators following high-affinity IgE-receptor crosslinking. Although these basophil-mediated responses depend on sensitization with antigen-specific IgE, this does not necessarily predict clinical symptom severity. It is thought that the balance of early stimulatory (e.g. SYK) and inhibitory (e.g. SHIP-1) intracellular signals are associated with basophil responsiveness, which is also critically dependent on calcium mobilization. Previous studies suggest that the sarcoplasmic reticulum Ca2+-ATPase (SERCA2), which regulates cytosolic calcium levels, may be inversely associated with airway smooth muscle reactivity in asthma. Since basophils are implicated in asthma severity, our aims were to address whether SERCA2 is implicated in human basophil responses, especially following IgE-mediated activation. Human basophils were obtained from buffy coats, following research ethics approval, and further purified by immunomagnetic cell sorting. Expressions of SERCA2, and other isoforms, were determined by Western blotting in parallel to measuring IgE-dependent histamine releases from the same donors. The effects of a SERCA-activator and inhibitor were also assessed on their abilities to modulate basophil histamine release. We observed an inverse correlation between basophil responsiveness to IgE-dependent stimulation and SERCA2 expression. Thapsigargin, a highly-specific SERCA inhibitor, stimulated basophil histamine release and potentiated IgE-dependent secretion of the amine. Conversely, disulfiram, a SERCA activator, inhibited IgE-dependent basophil activation. The results obtained from this exploratory study indicate that SERCA2 may be an additional regulator of basophil reactivity alongside early excitatory or inhibitory signal transduction pathways.
Collapse
Affiliation(s)
| | - Vadim V. Sumbayev
- School of Pharmacy, University of Kent, Chatham Maritime, United Kingdom
| | - Bernhard F. Gibbs
- School of Pharmacy, University of Kent, Chatham Maritime, United Kingdom,Department of Human Medicine, University of Oldenburg, Oldenburg, Germany,*Correspondence: Bernhard F. Gibbs,
| |
Collapse
|
16
|
Shibuya R, Kim BS. Skin-homing basophils and beyond. Front Immunol 2022; 13:1059098. [PMID: 36618424 PMCID: PMC9815541 DOI: 10.3389/fimmu.2022.1059098] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Basophils have been implicated in type 2 inflammation and numerous disorders in the skin such as helminth infection, atopic dermatitis, and urticaria. Although similar in form and function to tissue-resident mast cells, classical studies on basophils have centered on those from the hematopoietic compartment. However, increasing studies in tissues like the skin demonstrate that basophils may take on particular characteristics by responding to unique developmental, chemotactic, and activation cues. Herein, we highlight how recent studies in barrier immunology suggest the presence of skin-homing basophils that harbor a unique identity in terms of phenotype, function, and motility. These concepts may uniquely inform how basophils contribute to diseases at multiple epithelial surfaces and our ability to therapeutically target the innate immune system in disease.
Collapse
Affiliation(s)
- Rintaro Shibuya
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States,Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York City, NY, United States,Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Brian S. Kim
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States,Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York City, NY, United States,Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States,*Correspondence: Brian S. Kim,
| |
Collapse
|
17
|
Poto R, Gambardella AR, Marone G, Schroeder JT, Mattei F, Schiavoni G, Varricchi G. Basophils from allergy to cancer. Front Immunol 2022; 13:1056838. [PMID: 36578500 PMCID: PMC9791102 DOI: 10.3389/fimmu.2022.1056838] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/21/2022] [Indexed: 12/14/2022] Open
Abstract
Human basophils, first identified over 140 years ago, account for just 0.5-1% of circulating leukocytes. While this scarcity long hampered basophil studies, innovations during the past 30 years, beginning with their isolation and more recently in the development of mouse models, have markedly advanced our understanding of these cells. Although dissimilarities between human and mouse basophils persist, the overall findings highlight the growing importance of these cells in health and disease. Indeed, studies continue to support basophils as key participants in IgE-mediated reactions, where they infiltrate inflammatory lesions, release pro-inflammatory mediators (histamine, leukotriene C4: LTC4) and regulatory cytokines (IL-4, IL-13) central to the pathogenesis of allergic diseases. Studies now report basophils infiltrating various human cancers where they play diverse roles, either promoting or hampering tumorigenesis. Likewise, this activity bears remarkable similarity to the mounting evidence that basophils facilitate wound healing. In fact, both activities appear linked to the capacity of basophils to secrete IL-4/IL-13, with these cytokines polarizing macrophages toward the M2 phenotype. Basophils also secrete several angiogenic factors (vascular endothelial growth factor: VEGF-A, amphiregulin) consistent with these activities. In this review, we feature these newfound properties with the goal of unraveling the increasing importance of basophils in these diverse pathobiological processes.
Collapse
Affiliation(s)
- Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy,Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy,World Allergy Organization (WAO), Center of Excellence (CoE), Naples, Italy
| | - Adriana Rosa Gambardella
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy,Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy,World Allergy Organization (WAO), Center of Excellence (CoE), Naples, Italy,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy,Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (CNR), Naples, Italy
| | - John T. Schroeder
- Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins Asthma and Allergy Center, Johns Hopkins University, Baltimore, MD, United States
| | - Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy,*Correspondence: Gilda Varricchi, ; Giovanna Schiavoni,
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy,World Allergy Organization (WAO), Center of Excellence (CoE), Naples, Italy,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy,Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (CNR), Naples, Italy,*Correspondence: Gilda Varricchi, ; Giovanna Schiavoni,
| |
Collapse
|
18
|
Rijavec M, Maver A, Turner PJ, Hočevar K, Košnik M, Yamani A, Hogan S, Custovic A, Peterlin B, Korošec P. Integrative transcriptomic analysis in human and mouse model of anaphylaxis identifies gene signatures associated with cell movement, migration and neuroinflammatory signalling. Front Immunol 2022; 13:1016165. [PMID: 36569939 PMCID: PMC9772259 DOI: 10.3389/fimmu.2022.1016165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022] Open
Abstract
Background Anaphylaxis is an acute life-threatening allergic reaction and a concern at a global level; therefore, further progress in understanding the underlying mechanisms and more effective strategies for diagnosis, prevention and management are needed. Objective We sought to identify the global architecture of blood transcriptomic features of anaphylaxis by integrating expression data from human patients and mouse model of anaphylaxis. Methods Bulk RNA-sequencings of peripheral whole blood were performed in: i) 14 emergency department (ED) patients with acute anaphylaxis, predominantly to Hymenoptera venom, ii) 11 patients with peanut allergy undergoing double-blind, placebo-controlled food challenge (DBPCFC) to peanut, iii) murine model of IgE-mediated anaphylaxis. Integrative characterisation of differential gene expression, immune cell-type-specific gene expression profiles, and functional and pathway analysis was undertaken. Results 1023 genes were commonly and significantly dysregulated during anaphylaxis in ED and DBPCFC patients; of those genes, 29 were also dysregulated in the mouse model. Cell-type-specific gene expression profiles showed a rapid downregulation of blood basophil and upregulation of neutrophil signature in ED and DBPCFC patients and the mouse model, but no consistent and/or significant differences were found for other blood cells. Functional and pathway analysis demonstrated that human and mouse blood transcriptomic signatures of anaphylaxis follow trajectories of upregulation of cell movement, migration and neuroinflammatory signalling, and downregulation of lipid activating nuclear receptors signalling. Conclusion Our study highlights the matched and extensive blood transcriptomic changes and suggests the involvement of discrete cellular components and upregulation of migration and neuroinflammatory pathways during anaphylaxis.
Collapse
Affiliation(s)
- Matija Rijavec
- University Clinic of Respiratory and Allergic Diseases Golnik, Golnik, Slovenia
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Aleš Maver
- Clinical Institute of Medical Genetics, University Medical Centre, Ljubljana, Slovenia
| | - Paul J. Turner
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Keli Hočevar
- Clinical Institute of Medical Genetics, University Medical Centre, Ljubljana, Slovenia
| | - Mitja Košnik
- University Clinic of Respiratory and Allergic Diseases Golnik, Golnik, Slovenia
- Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Amnah Yamani
- Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, MI, United States
- Mary H. Weiser Food Allergy Center (MHWFAC), Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Simon P. Hogan
- Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, MI, United States
- Mary H. Weiser Food Allergy Center (MHWFAC), Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Adnan Custovic
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Borut Peterlin
- Clinical Institute of Medical Genetics, University Medical Centre, Ljubljana, Slovenia
| | - Peter Korošec
- University Clinic of Respiratory and Allergic Diseases Golnik, Golnik, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
19
|
Siddhuraj P, Nordström FU, Jogdand P, Elangovan G, Mori M, Bjermer L, Greiff L, Erjefält JS. Histology-based blood leukocyte profiling reveals parallel Th2 and Th17 signatures in seasonal allergic rhinitis. Acta Otolaryngol 2022; 142:696-704. [PMID: 36562632 DOI: 10.1080/00016489.2022.2155316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Allergic rhinitis (AR), a common condition in the westernized world, is suggested to be more immunologically complex than the archetypical 'Th2' inflammation. New approaches are needed to decode this complexity. AIMS/OBJECTIVES In this study, we explored a novel histology-based analysis for circulating blood leukocyte profiling in 16 patients with seasonal AR outside and during the pollen season. MATERIAL AND METHODS Leukocytes were purified with minimal ex-vivo artefacts, embedded into agarose-paraffin pellets for immunohistochemistry-based immune cell profiling. Blood leukocyte mapping was performed. RESULTS Samples collected during the pollen season had statistically increased eosinophils, neutrophils, monocytes, and CD8+ T-lymphocytes compared to the off-season baseline. In contrast, no change was observed for CD20+ B-lymphocytes and CD3+ T-lymphocytes. Subclassification of CD4+ T-helper cells demonstrated a parallel and significant expansion of Th2 and Th17-cells during the pollen season, while Th1-cells remained unchanged. Whereas absolute basophils numbers were unaltered, the basophil markers GATA2 and CPA3 increased during the pollen season. CONCLUSIONS AND SIGNIFICANCE This study introduces a novel and applicable method for systemic immune cell screening and provides further evidence of complex and parallel Th2 and Th17-immune signatures in seasonal AR. It also forwards GATA2 and CPA3 as potential biomarkers for ongoing allergic inflammation.
Collapse
Affiliation(s)
| | - Franziska U Nordström
- Department of ORL, Head and Neck Surgery, Skåne University Hospital, Lund, Sweden.,Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Prajakta Jogdand
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Gayathri Elangovan
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Michiko Mori
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Leif Bjermer
- Department of Respiratory Medicine and Allergology, Lund University Hospital, Lund, Sweden
| | - Lennart Greiff
- Department of ORL, Head and Neck Surgery, Skåne University Hospital, Lund, Sweden.,Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jonas S Erjefält
- Department of Experimental Medical Science, Lund University, Lund, Sweden.,Department of Respiratory Medicine and Allergology, Lund University Hospital, Lund, Sweden
| |
Collapse
|
20
|
Antibiotic Allergy De-Labeling: A Pathway against Antibiotic Resistance. Antibiotics (Basel) 2022; 11:antibiotics11081055. [PMID: 36009924 PMCID: PMC9404790 DOI: 10.3390/antibiotics11081055] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 12/10/2022] Open
Abstract
Antibiotics are one of the most frequently prescribed drugs. Unfortunately, they also are the most common cause for self-reported drug allergy, limiting the use of effective therapies. However, evidence shows that more than 90% of patients labeled as allergic to antibiotics are not allergic. Importantly, the label of antibiotic allergy, whether real or not, constitutes a major public health problem as it directly impacts antimicrobial stewardship: it has been associated with broad-spectrum antibiotic use, often resulting in the emergence of bacterial resistance. Therefore, an accurate diagnosis is crucial for de-labeling patients who claim to be allergic but are not really allergic. This review presents allergy methods for achieving successful antibiotic allergy de-labeling. Patient clinical history is often inaccurately reported, thus not being able to de-label most patients. In vitro testing offers a complementary approach but it shows limitations. Immunoassay for quantifying specific IgE is the most used one, although it gives low sensitivity and is limited to few betalactams. Basophil activation test is not validated and not available in all centers. Therefore, true de-labeling still relies on in vivo tests including drug provocation and/or skin tests, which are not risk-exempt and require specialized healthcare professionals for results interpretation and patient management. Moreover, differences on the pattern of antibiotic consumption cause differences in the diagnostic approach among different countries. A multidisciplinary approach is recommended to reduce the risks associated with the reported penicillin allergy label.
Collapse
|
21
|
Network Pharmacology and Molecular Docking Study of Yupingfeng Powder in the Treatment of Allergic Diseases. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1323744. [PMID: 35855823 PMCID: PMC9288288 DOI: 10.1155/2022/1323744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022]
Abstract
Objective To explore the potential mechanisms of Yupingfeng Powder (YPFP) in the treatment of allergic diseases by using network pharmacology and molecular docking technology. Methods The active components and targets of YPFP were screened by the TCMSP database. The targets associated with atopic dermatitis, asthma, allergic rhinitis, and food allergy were obtained from GeneCards and OMIM databases, respectively. The intersection of the above disease-related targets was identified as allergy-related targets. Then, allergy-related targets and YPFP-related targets were crossed to obtain the potential targets of YPFP for allergy treatment. A protein-protein-interaction (PPI) network and a drug-target-disease topology network were constructed to screen hub targets and key ingredients. Next, GO and KEGG pathway enrichment analyses were performed separately on the potential targets and hub targets to identify the biological processes and signaling pathways involved. Finally, molecular docking was conducted to verify the binding affinity between key ingredients and hub targets. Results In this study, 45 active ingredients were identified from YPFP, and 48 allergy-related targets were predicted by network pharmacology. IL6, TNF, IL1B, PTGS2, CXCL8, JUN, CCL2, IL10, IFNG, and IL4 were screened as hub targets by the PPI network. However, quercetin, kaempferol, wogonin, formononetin, and 7-O-methylisomucronulatol were identified as key ingredients by the drug-target-disease topological network. GO and KEGG pathway enrichment analysis indicated that the therapeutic effect of YPFP on allergy involved multiple biological processes and signaling pathways, including positive regulation of fever generation, positive regulation of neuroinflammatory response, vascular endothelial growth factor production, negative regulation of cytokine production involved in immune response, positive regulation of mononuclear cell migration, type 2 immune response, and negative regulation of lipid storage. Molecular docking verified that all the key ingredients had good binding affinity with hub targets. Conclusion This study revealed the key ingredients, hub targets, and potential mechanisms of YPFP antiallergy, and these data can provide some theoretical basis for subsequent allergy treatment and drug development.
Collapse
|
22
|
Morsy MA, Patel SS, Bakrania A, Kandeel M, Nair AB, Shah JN, Akrawi SH, El-Daly M. Ameliorative Effect of a Neoteric Regimen of Catechin plus Cetirizine on Ovalbumin-Induced Allergic Rhinitis in Rats. Life (Basel) 2022; 12:life12060820. [PMID: 35743851 PMCID: PMC9225010 DOI: 10.3390/life12060820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/25/2022] [Accepted: 05/28/2022] [Indexed: 11/30/2022] Open
Abstract
Allergic rhinitis (AR) affects 20–50% of the global population. Available treatments are limited by their adverse effects. We investigated the anti-allergic effects of catechin alone and combined with cetirizine against ovalbumin-induced AR. Rats were sensitized with ovalbumin and received catechin (14 days) and then challenged with aerosolized ovalbumin (1%) to determine AR clinical scores. Histamine, histamine release, and histidine decarboxylase (HDC) activity were determined in blood, peritoneal mast cells, and stomachs, respectively. Vascular permeability and safety were assessed using Evans blue leakage and barbiturate-induced sleeping-time assays, respectively. Catechin and cetirizine binding with HDC was investigated by docking and binding energy analyses. The clinical scores of the combination regimen were superior to either drug alone. All treatments reduced vascular leakage, with no effect on barbiturate-induced sleeping time. Only the catechin-treated rats showed reduced histamine levels and HDC activity. Docking studies revealed that catechin has a 1.34-fold higher extra-precision docking score than L-histidine. The binding energy scores for catechin-HDC, L-histidine-HDC, and histamine-HDC were −50.86, −37.64, and −32.27 kcal/mol, respectively. The binding pattern of catechin was comparable to the standard HDC inhibitor, histidine methyl ester, but with higher binding free energy. Catechin binds the catalytic residue S354, unlike cetirizine. The anti-allergic effects of catechin can be explained by HDC inhibition and possible antihistaminic activity.
Collapse
Affiliation(s)
- Mohamed A. Morsy
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (A.B.N.); (S.H.A.)
- Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia 61511, Egypt
- Correspondence: (M.A.M.); (S.S.P.)
| | - Snehal S. Patel
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujarat, India;
- Correspondence: (M.A.M.); (S.S.P.)
| | - Anita Bakrania
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujarat, India;
| | - Mahmoud Kandeel
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - Anroop B. Nair
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (A.B.N.); (S.H.A.)
| | - Jigar N. Shah
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujarat, India;
| | - Sabah H. Akrawi
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (A.B.N.); (S.H.A.)
| | - Mahmoud El-Daly
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Minia University, El-Minia 61511, Egypt;
| |
Collapse
|
23
|
Miyake K, Ito J, Karasuyama H. Role of Basophils in a Broad Spectrum of Disorders. Front Immunol 2022; 13:902494. [PMID: 35693800 PMCID: PMC9186123 DOI: 10.3389/fimmu.2022.902494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
Basophils are the rarest granulocytes and have long been overlooked in immunological research due to their rarity and similarities with tissue-resident mast cells. In the last two decades, non-redundant functions of basophils have been clarified or implicated in a broad spectrum of immune responses, particularly by virtue of the development of novel analytical tools for basophils. Basophils infiltrate inflamed tissues of patients with various disorders, even though they circulate in the bloodstream under homeostatic conditions. Depletion of basophils results in the amelioration or exaggeration of inflammation, depending on models of disease, indicating basophils can play either beneficial or deleterious roles in a context-dependent manner. In this review, we summarize the recent findings of basophil pathophysiology under various conditions in mice and humans, including allergy, autoimmunity, tumors, tissue repair, fibrosis, and COVID-19. Further mechanistic studies on basophil biology could lead to the identification of novel biomarkers or therapeutic targets in a broad range of diseases.
Collapse
|
24
|
Brandolini L, d'Angelo M, Novelli R, Castelli V, Giorgio C, Sirico A, Cocchiaro P, D'Egidio F, Benedetti E, Cristiano C, Bugatti A, Ruocco A, Amendola PG, Talarico C, Manelfi C, Iaconis D, Beccari A, Quadros AU, Cunha TM, Caruso A, Russo R, Cimini A, Aramini A, Allegretti M. Paclitaxel binds and activates C5aR1: A new potential therapeutic target for the prevention of chemotherapy-induced peripheral neuropathy and hypersensitivity reactions. Cell Death Dis 2022; 13:500. [PMID: 35614037 PMCID: PMC9130998 DOI: 10.1038/s41419-022-04964-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 05/10/2022] [Accepted: 05/17/2022] [Indexed: 12/14/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) and hypersensitivity reactions (HSRs) are among the most frequent and impairing side effects of the antineoplastic agent paclitaxel. Here, we demonstrated that paclitaxel can bind and activate complement component 5a receptor 1 (C5aR1) and that this binding is crucial in the etiology of paclitaxel-induced CIPN and anaphylaxis. Starting from our previous data demonstrating the role of interleukin (IL)-8 in paclitaxel-induced neuronal toxicity, we searched for proteins that activate IL-8 expression and, by using the Exscalate platform for molecular docking simulations, we predicted the high affinity of C5aR1 with paclitaxel. By in vitro studies, we confirmed the specific and competitive nature of the C5aR1-paclitaxel binding and found that it triggers intracellularly the NFkB/P38 pathway and c-Fos. In F11 neuronal cells and rat dorsal root ganglia, C5aR1 inhibition protected from paclitaxel-induced neuropathological effects, while in paclitaxel-treated mice, the absence (knock-out mice) or the inhibition of C5aR1 significantly ameliorated CIPN symptoms-in terms of cold and mechanical allodynia-and reduced the chronic pathological state in the paw. Finally, we found that C5aR1 inhibition can counteract paclitaxel-induced anaphylactic cytokine release in macrophages in vitro, as well as the onset of HSRs in mice. Altogether these data identified C5aR1 as a key mediator and a new potential pharmacological target for the prevention and treatment of CIPN and HSRs induced by paclitaxel.
Collapse
Affiliation(s)
- Laura Brandolini
- Dompé Farmaceutici SpA, Via Campo di Pile, 67100, L'Aquila, Italy
| | - Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Rubina Novelli
- Dompé Farmaceutici SpA, Via S. Lucia, 20122, Milan, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Cristina Giorgio
- Dompé Farmaceutici SpA, Via Tommaso De Amicis, 80131, Naples, Italy
| | - Anna Sirico
- Dompé Farmaceutici SpA, Via Tommaso De Amicis, 80131, Naples, Italy
| | | | - Francesco D'Egidio
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Claudia Cristiano
- Department of Pharmacy, University of Naples Federico II, 80131, Naples, Italy
| | - Antonella Bugatti
- Department of Molecular and Traslational Medicine, University of Brescia Medical School, 25123, Brescia, Italy
| | - Anna Ruocco
- Dompé Farmaceutici SpA, Via Tommaso De Amicis, 80131, Naples, Italy
| | | | - Carmine Talarico
- Dompé Farmaceutici SpA, Via Tommaso De Amicis, 80131, Naples, Italy
| | - Candida Manelfi
- Dompé Farmaceutici SpA, Via Tommaso De Amicis, 80131, Naples, Italy
| | - Daniela Iaconis
- Dompé Farmaceutici SpA, Via Tommaso De Amicis, 80131, Naples, Italy
| | - Andrea Beccari
- Dompé Farmaceutici SpA, Via Tommaso De Amicis, 80131, Naples, Italy
| | - Andreza U Quadros
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Thiago M Cunha
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Arnaldo Caruso
- Department of Molecular and Traslational Medicine, University of Brescia Medical School, 25123, Brescia, Italy
| | - Roberto Russo
- Department of Pharmacy, University of Naples Federico II, 80131, Naples, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100, L'Aquila, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA, 19122, USA
| | - Andrea Aramini
- Dompé Farmaceutici SpA, Via Campo di Pile, 67100, L'Aquila, Italy
| | | |
Collapse
|
25
|
Elst J, De Puysseleyr LP, Ebo DG, Faber MA, Van Gasse AL, van der Poorten MLM, Decuyper II, Bridts CH, Mertens C, Van Houdt M, Hagendorens MM, De Clerck LS, Verlinden A, Vermeulen K, Maes MB, Berneman ZN, Valent P, Sabato V. Overexpression of FcεRI on Bone Marrow Mast Cells, but Not MRGPRX2, in Clonal Mast Cell Disorders With Wasp Venom Anaphylaxis. Front Immunol 2022; 13:835618. [PMID: 35281031 PMCID: PMC8914951 DOI: 10.3389/fimmu.2022.835618] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/04/2022] [Indexed: 12/17/2022] Open
Abstract
Background Uncertainties remain about the molecular mechanisms governing clonal mast cell disorders (CMCD) and anaphylaxis. Objective This study aims at comparing the burden, phenotype and behavior of mast cells (MCs) and basophils in patients with CMCD with wasp venom anaphylaxis (CMCD/WVA+), CMCD patients without anaphylaxis (CMCD/ANA-), patients with an elevated baseline serum tryptase (EBST), patients with wasp venom anaphylaxis without CMCD (WVA+) and patients with a non-mast cell haematological pathology (NMHP). Methods This study included 20 patients with CMCD/WVA+, 24 with CMCD/ANA-, 19 with WVA+, 6 with EBST and 5 with NMHP. We immunophenotyped MCs and basophils and compared baseline serum tryptase (bST) and both total and venom specific IgE in the different groups. For basophil studies, 13 healthy controls were also included. Results Higher levels of bST were found in CMCD patients with wasp venom anaphylaxis, CMCD patients without anaphylaxis and EBST patients. Total IgE levels were highest in patients with wasp venom anaphylaxis with and without CMCD. Bone marrow MCs of patients with CMCD showed lower CD117 expression and higher expression of CD45, CD203c, CD63, CD300a and FcεRI. Within the CMCD population, patients with wasp venom anaphylaxis showed a higher expression of FcεRI as compared to patients without anaphylaxis. Expression of MRGPRX2 on MCs did not differ between the study populations. Basophils are phenotypically and functionally comparable between the different patient populations. Conclusion Patients with CMCD show an elevated burden of aberrant activated MCs with a significant overexpression of FcεRI in patients with a wasp venom anaphylaxis.
Collapse
Affiliation(s)
- Jessy Elst
- Department of Immunology, Allergology, Rheumatology and the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Immunology, Allergology, Rheumatology, Antwerp University Hospital, Antwerp, Belgium
| | - Leander P De Puysseleyr
- Department of Immunology, Allergology, Rheumatology and the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Immunology, Allergology, Rheumatology, Antwerp University Hospital, Antwerp, Belgium
| | - Didier G Ebo
- Department of Immunology, Allergology, Rheumatology and the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Immunology, Allergology, Rheumatology, Antwerp University Hospital, Antwerp, Belgium.,Department of Immunology and Allergology, AZ Jan Palfijn Gent, Ghent, Belgium
| | - Margaretha A Faber
- Department of Immunology, Allergology, Rheumatology and the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Immunology, Allergology, Rheumatology, Antwerp University Hospital, Antwerp, Belgium
| | - Athina L Van Gasse
- Department of Immunology, Allergology, Rheumatology and the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Immunology, Allergology, Rheumatology, Antwerp University Hospital, Antwerp, Belgium.,Department of Paediatrics and the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Paediatrics, Antwerp University Hospital, Antwerp, Belgium
| | - Marie-Line M van der Poorten
- Department of Immunology, Allergology, Rheumatology and the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Immunology, Allergology, Rheumatology, Antwerp University Hospital, Antwerp, Belgium.,Department of Paediatrics and the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Paediatrics, Antwerp University Hospital, Antwerp, Belgium
| | - Ine I Decuyper
- Department of Immunology, Allergology, Rheumatology and the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Immunology, Allergology, Rheumatology, Antwerp University Hospital, Antwerp, Belgium.,Department of Paediatrics and the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Paediatrics, Antwerp University Hospital, Antwerp, Belgium
| | - Chris H Bridts
- Department of Immunology, Allergology, Rheumatology and the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Immunology, Allergology, Rheumatology, Antwerp University Hospital, Antwerp, Belgium
| | - Christel Mertens
- Department of Immunology, Allergology, Rheumatology and the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Immunology, Allergology, Rheumatology, Antwerp University Hospital, Antwerp, Belgium
| | - Michel Van Houdt
- Department of Immunology, Allergology, Rheumatology and the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Immunology, Allergology, Rheumatology, Antwerp University Hospital, Antwerp, Belgium
| | - Margo M Hagendorens
- Department of Immunology, Allergology, Rheumatology and the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Immunology, Allergology, Rheumatology, Antwerp University Hospital, Antwerp, Belgium.,Department of Paediatrics and the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Paediatrics, Antwerp University Hospital, Antwerp, Belgium
| | - Luc S De Clerck
- Department of Immunology, Allergology, Rheumatology and the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Immunology, Allergology, Rheumatology, Antwerp University Hospital, Antwerp, Belgium
| | - Anke Verlinden
- Department of Haematology, Antwerp University Hospital, Antwerp, Belgium
| | - Katrien Vermeulen
- Department of Clinical Biology, Antwerp University Hospital, Antwerp, Belgium
| | - Marie-Berthe Maes
- Department of Clinical Biology, Antwerp University Hospital, Antwerp, Belgium
| | - Zwi N Berneman
- Department of Haematology, Antwerp University Hospital, Antwerp, Belgium
| | - Peter Valent
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Vito Sabato
- Department of Immunology, Allergology, Rheumatology and the Infla-Med Centre of Excellence, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Immunology, Allergology, Rheumatology, Antwerp University Hospital, Antwerp, Belgium.,Department of Immunology and Allergology, AZ Jan Palfijn Gent, Ghent, Belgium
| |
Collapse
|
26
|
Castaño N, Kim S, Martin AM, Galli SJ, Nadeau KC, Tang SKY. Exponential magnetophoretic gradient for the direct isolation of basophils from whole blood in a microfluidic system. LAB ON A CHIP 2022; 22:1690-1701. [PMID: 35438713 PMCID: PMC9080715 DOI: 10.1039/d2lc00154c] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Despite their rarity in peripheral blood, basophils play important roles in allergic disorders and other diseases including sepsis and COVID-19. Existing basophil isolation methods require many manual steps and suffer from significant variability in purity and recovery. We report an integrated basophil isolation device (i-BID) in microfluidics for negative immunomagnetic selection of basophils directly from 100 μL of whole blood within 10 minutes. We use a simulation-driven pipeline to design a magnetic separation module to apply an exponentially increasing magnetic force to capture magnetically tagged non-basophils flowing through a microtubing sandwiched between magnetic flux concentrators sweeping across a Halbach array. The exponential profile captures non-basophils effectively while preventing their excessive initial buildup causing clogging. The i-BID isolates basophils with a mean purity of 93.9% ± 3.6% and recovery of 95.6% ± 3.4% without causing basophil degradation or unintentional activation. Our i-BID has the potential to enable basophil-based point-of-care diagnostics such as rapid allergy assessment.
Collapse
Affiliation(s)
- Nicolas Castaño
- Department of Mechanical Engineering, Stanford University, USA.
| | - Sungu Kim
- Department of Mechanical Engineering, Stanford University, USA.
| | - Adrian M Martin
- Department of Mechanical Engineering, Stanford University, USA.
| | - Stephen J Galli
- Department of Pathology, Stanford University, USA.
- Department of Microbiology and Immunology, Stanford University, USA
| | - Kari C Nadeau
- Department of Medicine and Pediatrics, with courtesy in Otolaryngology and in Population Science and Epidemiology, Stanford University, USA.
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, USA
| | - Sindy K Y Tang
- Department of Mechanical Engineering, Stanford University, USA.
| |
Collapse
|
27
|
Nuñez-Borque E, Fernandez-Bravo S, Yuste-Montalvo A, Esteban V. Pathophysiological, Cellular, and Molecular Events of the Vascular System in Anaphylaxis. Front Immunol 2022; 13:836222. [PMID: 35371072 PMCID: PMC8965328 DOI: 10.3389/fimmu.2022.836222] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/07/2022] [Indexed: 01/10/2023] Open
Abstract
Anaphylaxis is a systemic hypersensitivity reaction that can be life threatening. Mechanistically, it results from the immune activation and release of a variety of mediators that give rise to the signs and symptoms of this pathological event. For years, most of the research in anaphylaxis has focused on the contribution of the immune component. However, approaches that shed light on the participation of other cellular and molecular agents are necessary. Among them, the vascular niche receives the various signals (e.g., histamine) that elicit the range of anaphylactic events. Cardiovascular manifestations such as increased vascular permeability, vasodilation, hypotension, vasoconstriction, and cardiac alterations are crucial in the pathophysiology of anaphylaxis and are highly involved to the development of the most severe cases. Specifically, the endothelium, vascular smooth muscle cells, and their molecular signaling outcomes play an essential role downstream of the immune reaction. Therefore, in this review, we synthesized the vascular changes observed during anaphylaxis as well as its cellular and molecular components. As the risk of anaphylaxis exists both in clinical procedures and in routine life, increasing our knowledge of the vascular physiology and their molecular mechanism will enable us to improve the clinical management and how to treat or prevent anaphylaxis. Key Message Anaphylaxis, the most severe allergic reaction, involves a variety of immune and non-immune molecular signals that give rise to its pathophysiological manifestations. Importantly, the vascular system is engaged in processes relevant to anaphylactic events such as increased vascular permeability, vasodilation, hypotension, vasoconstriction, and decreased cardiac output. The novelty of this review focuses on the fact that new studies will greatly improve the understanding of anaphylaxis when viewed from a vascular molecular angle and specifically from the endothelium. This knowledge will improve therapeutic options to treat or prevent anaphylaxis.
Collapse
Affiliation(s)
- Emilio Nuñez-Borque
- Department of Allergy and Immunology, Instituto en Investigación Sanitaria - Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Sergio Fernandez-Bravo
- Department of Allergy and Immunology, Instituto en Investigación Sanitaria - Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Alma Yuste-Montalvo
- Department of Allergy and Immunology, Instituto en Investigación Sanitaria - Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Vanesa Esteban
- Department of Allergy and Immunology, Instituto en Investigación Sanitaria - Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Faculty of Medicine and Biomedicine, Alfonso X El Sabio University, Madrid, Spain
| |
Collapse
|
28
|
López‐Sanz C, Jimenéz‐Saiz R, Ehlers AM. Local inflammation enables a basophil-neuronal circuITCH in atopic dermatitis. Allergy 2022; 77:708-710. [PMID: 34318483 DOI: 10.1111/all.15026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/14/2021] [Accepted: 07/21/2021] [Indexed: 12/01/2022]
Affiliation(s)
- Celia López‐Sanz
- Department of Immunology Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS‐IP) Madrid Spain
| | - Rodrigo Jimenéz‐Saiz
- Department of Immunology Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS‐IP) Madrid Spain
- Department of Immunology and Oncology Centro Nacional de Biotecnología (CNB)‐CSIC Madrid Spain
- Faculty of Experimental Sciences Universidad Francisco de Vitoria (UFV) Madrid Spain
- Department of Medicine McMaster Immunology Research Centre McMaster University Hamilton Ontario Canada
| | - Anna M. Ehlers
- Center for Translational Immunology, University Medical Center Utrecht Utrecht University Utrecht The Netherlands
- Department of Dermatology/Allergology University Medical Center Utrecht, Utrecht University Utrecht The Netherlands
| |
Collapse
|
29
|
State-of-the-Art on Biomarkers for Anaphylaxis in Obstetrics. Life (Basel) 2021; 11:life11090870. [PMID: 34575019 PMCID: PMC8467046 DOI: 10.3390/life11090870] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/21/2021] [Accepted: 08/23/2021] [Indexed: 12/25/2022] Open
Abstract
Anaphylaxis is an unpredictable systemic hypersensitivity reaction and constitutes a high risk of maternal and fetal morbidity and mortality when occurring during pregnancy. Currently, the acute management of anaphylaxis is based on clinical parameters. A total serum tryptase is only used to support an accurate diagnosis. There is a need to detect other biomarkers to further assess high-risk patients in obstetrics. Our objective is to present biomarkers in this complex interdisciplinary approach beyond obstetrician and anaesthetic management. Candidate biomarkers derive either from mediators involved in immunopathogenesis or upcoming molecules from systems biology and proteomics. Serum tryptase is determined by singleplex immunoassay method and is important in the evaluation of anaphylactic mast cell degranulation but also in the assessment of other risk factors for anaphylaxis such as systemic mastocytosis. Another category of biomarkers investigates the IgE-mediated sensitization to triggers potentially involved in the etiology of anaphylaxis in pregnant women, using singleplex or multiplex immunoassays. These in vitro tests with natural extracts from foods, venoms, latex or drugs, as well as with molecular allergen components, are useful because in vivo allergy tests cannot be performed on pregnant women in such a major medical emergency due to their additional potential risk of anaphylaxis.
Collapse
|
30
|
Santos AF, Alpan O, Hoffmann H. Basophil activation test: Mechanisms and considerations for use in clinical trials and clinical practice. Allergy 2021; 76:2420-2432. [PMID: 33475181 DOI: 10.1111/all.14747] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/15/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022]
Abstract
The basophil activation test (BAT) is a functional assay that measures the degree of degranulation following stimulation with allergen or controls by flow cytometry. It correlates directly with histamine release. From the dose-response curve resulting from BAT in allergic patients, basophil reactivity (%CD63+ basophils) and basophil sensitivity (EC50 or similar) are the main outcomes of the test. BAT takes into account all characteristics of IgE and allergen and thus can be more specific than sensitization tests in the diagnosis of allergic disease. BAT reduces the need for in vivo procedures, such as intradermal tests and allergen challenges, which can cause allergic reactions of unpredictable severity. As it closely reflects the patients' phenotype in most cases, it may be used to support the diagnosis of food, venom and drug allergies and chronic urticaria, to monitor the natural resolution of food allergies and to predict and monitor clinical the response to immunomodulatory treatments, such as allergen-specific immunotherapy and biologicals. Clinical application of BAT requires analytical validation, clinical validation, standardization of procedures and quality assurance to ensure reproducibility and reliability of results. Currently, efforts are ongoing to establish a platform that could be used by laboratories in Europe and in the USA for quality assurance and certification.
Collapse
Affiliation(s)
- Alexandra F. Santos
- Department of Women and Children's Health (Pediatric Allergy) School of Life Course Sciences Faculty of Life Sciences and Medicine King's College London London UK
- Peter Gorer Department of Immunobiology School of Immunology and Microbial Sciences King's College London London UK
- Asthma UK Centre in Allergic Mechanisms of Asthma London UK
- Children's Allergy ServiceEvelina London Children's HospitalGuy's and St Thomas' Hospital London UK
| | | | - Hans‐Jürgen Hoffmann
- Department of Clinical Medicine Aarhus University Aarhus Denmark
- Department of Respiratory Diseases and Allergy Aarhus University Hospital Aarhus Denmark
| |
Collapse
|
31
|
Callesen KT, Poulsen LK, Garvey LH, Jensen BM. Comparing baseline and reaction samples of perioperative anaphylaxis patients reveals IL-6 and CCL2 as potential biomarkers. Clin Exp Allergy 2021; 51:1250-1253. [PMID: 34143525 DOI: 10.1111/cea.13969] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/05/2021] [Accepted: 06/15/2021] [Indexed: 12/25/2022]
Affiliation(s)
- Katrine T Callesen
- Allergy Clinic, Copenhagen University Hospital at Herlev-Gentofte, Hellerup, Denmark
| | - Lars K Poulsen
- Allergy Clinic, Copenhagen University Hospital at Herlev-Gentofte, Hellerup, Denmark
| | - Lene H Garvey
- Allergy Clinic, Copenhagen University Hospital at Herlev-Gentofte, Hellerup, Denmark
| | - Bettina M Jensen
- Allergy Clinic, Copenhagen University Hospital at Herlev-Gentofte, Hellerup, Denmark
| |
Collapse
|
32
|
Miyake K, Shibata S, Yoshikawa S, Karasuyama H. Basophils and their effector molecules in allergic disorders. Allergy 2021; 76:1693-1706. [PMID: 33205439 DOI: 10.1111/all.14662] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/26/2020] [Accepted: 11/12/2020] [Indexed: 12/20/2022]
Abstract
Basophils are the rarest granulocytes which represent <1% of peripheral blood leukocytes. Basophils bear several phenotypic similarities to tissue-resident mast cells and therefore had been erroneously considered as blood-circulating mast cells. However, recent researches have revealed that basophils play nonredundant roles in allergic inflammation, protective immunity against parasitic infections and regulation of innate and acquired immunity. Basophils are recruited to inflamed tissues and activated in an IgE-dependent or IgE-independent manner to release a variety of effector molecules. Such molecules, including IL-4, act on various types of cells and play versatile roles, including the induction and termination of allergic inflammation and the regulation of immune responses. Recent development of novel therapeutic agents has enabled us to gain further insights into basophil biology in human disorders. In this review, we highlight the recent advances in the field of basophil biology with a particular focus on the role of basophils in allergic inflammation. Further studies on basophils and their effector molecules will help us identify novel therapeutic targets for treating allergic disorders.
Collapse
Affiliation(s)
- Kensuke Miyake
- Inflammation, Infection and Immunity Laboratory TMDU Advanced Research Institute Tokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Sho Shibata
- Department of Respiratory Medicine Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Soichiro Yoshikawa
- Department of Cell Physiology Graduate School of Medicine, Dentistry and Pharmaceutical Sciences Okayama University Okayama Japan
| | - Hajime Karasuyama
- Inflammation, Infection and Immunity Laboratory TMDU Advanced Research Institute Tokyo Medical and Dental University (TMDU) Tokyo Japan
| |
Collapse
|
33
|
Rijavec M, Košnik M, Koren A, Kopač P, Šelb J, Vantur R, Kogovšek Ž, Bizjak M, Bajrović N, Zidarn M, Korošec P. A very low number of circulating basophils is predictive of a poor response to omalizumab in chronic spontaneous urticaria. Allergy 2021; 76:1254-1257. [PMID: 32876979 DOI: 10.1111/all.14577] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 08/06/2020] [Accepted: 08/26/2020] [Indexed: 01/28/2023]
Affiliation(s)
- Matija Rijavec
- University Clinic of Respiratory and Allergic Diseases Golnik Golnik Slovenia
- Biotechnical Faculty University of Ljubljana Ljubljana Slovenia
| | - Mitja Košnik
- University Clinic of Respiratory and Allergic Diseases Golnik Golnik Slovenia
- Faculty of Medicine University of Ljubljana Ljubljana Slovenia
| | - Ana Koren
- University Clinic of Respiratory and Allergic Diseases Golnik Golnik Slovenia
| | - Peter Kopač
- University Clinic of Respiratory and Allergic Diseases Golnik Golnik Slovenia
- Faculty of Medicine University of Ljubljana Ljubljana Slovenia
| | - Julij Šelb
- University Clinic of Respiratory and Allergic Diseases Golnik Golnik Slovenia
| | - Romana Vantur
- University Clinic of Respiratory and Allergic Diseases Golnik Golnik Slovenia
| | - Žan Kogovšek
- University Clinic of Respiratory and Allergic Diseases Golnik Golnik Slovenia
| | - Mojca Bizjak
- University Clinic of Respiratory and Allergic Diseases Golnik Golnik Slovenia
| | - Nissera Bajrović
- University Clinic of Respiratory and Allergic Diseases Golnik Golnik Slovenia
| | - Mihaela Zidarn
- University Clinic of Respiratory and Allergic Diseases Golnik Golnik Slovenia
- Faculty of Medicine University of Ljubljana Ljubljana Slovenia
| | - Peter Korošec
- University Clinic of Respiratory and Allergic Diseases Golnik Golnik Slovenia
| |
Collapse
|
34
|
Dribin TE, Schnadower D, Spergel JM, Campbell RL, Shaker M, Neuman MI, Michelson KA, Capucilli PS, Camargo CA, Brousseau DC, Rudders SA, Assa'ad AH, Risma KA, Castells M, Schneider LC, Wang J, Lee J, Mistry RD, Vyles D, Pistiner M, Witry JK, Zhang Y, Sampson HA. Severity grading system for acute allergic reactions: A multidisciplinary Delphi study. J Allergy Clin Immunol 2021; 148:173-181. [PMID: 33476673 DOI: 10.1016/j.jaci.2021.01.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/29/2020] [Accepted: 01/08/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND There is no widely adopted severity grading system for acute allergic reactions, including anaphylactic and nonanaphylactic reactions, thus limiting the ability to optimize and standardize management practices and advance research. OBJECTIVE The aim of this study was to develop a severity grading system for acute allergic reactions for use in clinical care and research. METHODS From May to September 2020, we convened a 21-member multidisciplinary panel of allergy and emergency care experts; 9 members formed a writing group to critically appraise and assess the strengths and limitations of prior severity grading systems and develop the structure and content for an optimal severity grading system. The entire study panel then revised the grading system and sought consensus by utilizing Delphi methodology. RESULTS The writing group recommended that an optimal grading system encompass the severity of acute allergic reactions on a continuum from mild allergic reactions to anaphylactic shock. Additionally, the severity grading system must be able to discriminate between clinically important differences in reaction severity to be relevant in research while also being intuitive and straightforward to apply in clinical care. Consensus was reached for all elements of the proposed severity grading system. CONCLUSION We developed a consensus severity grading system for acute allergic reactions, including anaphylactic and nonanaphylactic reactions. Successful international validation, refinement, dissemination, and application of the grading system will improve communication among providers and patients about the severity of allergic reactions and will help advance future research.
Collapse
Affiliation(s)
- Timothy E Dribin
- Division of Emergency Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| | - David Schnadower
- Division of Emergency Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Jonathan M Spergel
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Perelman School of Medicine at University of Pennsylvania, Philadelphia, Pa
| | | | - Marcus Shaker
- Dartmouth Geisel School of Medicine, Hanover, NH; Dartmouth-Hitchcock Medical Center, Hanover, NH
| | - Mark I Neuman
- Division of Emergency Medicine, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Kenneth A Michelson
- Division of Emergency Medicine, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | | | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - David C Brousseau
- Section of Pediatric Emergency Medicine, Department of Pediatrics, Medical College of Wisconsin, Milwaukee
| | - Susan A Rudders
- Department of Pediatrics, Harvard Medical School, Boston, Mass; Division of Immunology, Boston Children's Hospital, Boston, Mass
| | - Amal H Assa'ad
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kimberly A Risma
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Mariana Castells
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Lynda C Schneider
- Department of Pediatrics, Harvard Medical School, Boston, Mass; Division of Immunology, Boston Children's Hospital, Boston, Mass
| | - Julie Wang
- Division of Allergy and Immunology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Juhee Lee
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Perelman School of Medicine at University of Pennsylvania, Philadelphia, Pa
| | - Rakesh D Mistry
- Section of Emergency Medicine, Department of Pediatrics, Children's Hospital Colorado, Aurora
| | - David Vyles
- Section of Pediatric Emergency Medicine, Department of Pediatrics, Medical College of Wisconsin, Milwaukee
| | - Michael Pistiner
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - John K Witry
- Division of Emergency Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Yin Zhang
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Hugh A Sampson
- Division of Allergy and Immunology, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
35
|
Vantur R, Rihar M, Koren A, Rijavec M, Kopac P, Bidovec-Stojkovic U, Erzen R, Korosec P. Chemokines during anaphylaxis: the importance of CCL2 and CCL2-dependent chemotactic activity for basophils. Clin Transl Allergy 2020; 10:63. [PMID: 33317619 PMCID: PMC7737350 DOI: 10.1186/s13601-020-00367-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/26/2020] [Indexed: 12/23/2022] Open
Abstract
Background The role of chemokines in anaphylaxis is unclear. Methods We prospectively recruited 49 patients presenting to the emergency department with an acute episode of anaphylaxis and 28 healthy subjects. We measured serum levels of the chemokines CCL2, CCL5, CCL7, CCL8, CCL11, CCL13, CCL17, CCL21, CCL22, CCL24, and CCL26, tryptase, the absolute number of circulating basophils, monocytes, lymphocytes, and PMNs, and whole blood FCER1A, CPA3 and HDC gene expression at two time points: during the anaphylactic episode and in convalescent samples collected approximately 3 months later. We then investigated the in vitro chemotactic activity of chemokines induced during anaphylaxis for the in vitro migration of the corresponding cells. Results Only CCL2 chemokine levels were significantly increased in anaphylaxis samples (median 514 pg/ml) compared to convalescent samples (284 pg/ml, P < 0.0001) and healthy subjects (279 pg/ml, P < 0.0001); there was no significant difference in any of the other chemokines. There was a significant positive correlation between the rates of increase of serum CCL2 (median [range]: 106.0% [− 44.7% to 557.4%]) and tryptase (133.8% [− 6.6% to 893.4%]; r = 0.68, P < 0.0001) and between the acute concentration of serum CCL2 and the acute concentration of serum tryptase (r = 0.77, P < 0.0001). The number of circulating basophils, but not other blood cells, significantly decreased during anaphylaxis (median 5.0 vs. 19.1 cells/µl in convalescent samples; P < 0.0001); a decrease in whole-blood gene expression of basophil markers (P ≤ 0.0018) confirmed these changes. Anaphylactic serum enhances the in vitro migration of basophils via CCL2-dependent chemotactic activity; in contrast, no CCL2-dependent chemotactic activity was observed for convalescent samples. Conclusions Our findings imply an important and specific role for CCL2-mediated chemotactic activity in the pathophysiology of human anaphylaxis.
Collapse
Affiliation(s)
- Romana Vantur
- University Clinic of Respiratory and Allergic Diseases Golnik, Golnik 36, 4204, Golnik, Slovenia
| | - Marusa Rihar
- University Clinic of Respiratory and Allergic Diseases Golnik, Golnik 36, 4204, Golnik, Slovenia
| | - Ana Koren
- University Clinic of Respiratory and Allergic Diseases Golnik, Golnik 36, 4204, Golnik, Slovenia
| | - Matija Rijavec
- University Clinic of Respiratory and Allergic Diseases Golnik, Golnik 36, 4204, Golnik, Slovenia.,Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Peter Kopac
- University Clinic of Respiratory and Allergic Diseases Golnik, Golnik 36, 4204, Golnik, Slovenia.,Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Urska Bidovec-Stojkovic
- University Clinic of Respiratory and Allergic Diseases Golnik, Golnik 36, 4204, Golnik, Slovenia
| | - Renato Erzen
- University Clinic of Respiratory and Allergic Diseases Golnik, Golnik 36, 4204, Golnik, Slovenia.,Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Peter Korosec
- University Clinic of Respiratory and Allergic Diseases Golnik, Golnik 36, 4204, Golnik, Slovenia.
| |
Collapse
|
36
|
The Challenge of Establishing the Burden of Anaphylaxis: Some Recent Trends. CURRENT TREATMENT OPTIONS IN ALLERGY 2020. [DOI: 10.1007/s40521-020-00257-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
37
|
|
38
|
Buyuktiryaki B, Santos AF. Food allergy severity predictions based on cellular in vitro tests. Expert Rev Mol Diagn 2020; 20:679-692. [PMID: 32536279 DOI: 10.1080/14737159.2020.1782192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Food allergy is increasing in prevalence and the severity of allergic reactions is unpredictable. Identifying food-allergic patients at high risk of severe reactions would allow us to offer a personalized and improved management for these patients. AREAS COVERED We review the evidence for using the levels of specific IgE, the nature of the allergen, and cellular tests to identify patients at high risk of developing severe allergic reactions to foods. EXPERT OPINION The evidence about whether the quantity of allergen-specific IgE reflects the severity of allergic reactions to foods is conflicting, with some positive and some negative studies. For some foods, specific IgE to individual components (e.g. Ara h 2 in peanut) can provide additional information. However, more important than the quantity of IgE is possibly the quality of IgE, which can be captured by individual measurements of affinity/avidity, diversity, and specific activity, but is best measured overall using the basophil and mast cell activation tests, which assess the function of IgE in its ability to induce cell activation, degranulation, and mediator release. Biomarkers look at a single aspect of the allergic response and should be interpreted in the broader clinical context for each individual patient assessed.
Collapse
Affiliation(s)
- Betul Buyuktiryaki
- Division of Pediatric Allergy, Koc University Hospital , İstanbul, Turkey.,Department of Paediatric Allergy, Evelina London, Guy's and ST Thomas' Hospital NHS Foundation Trust , London, UK
| | - Alexandra F Santos
- Department of Paediatric Allergy, Evelina London, Guy's and ST Thomas' Hospital NHS Foundation Trust , London, UK.,Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London , London, UK.,Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London , London, UK.,Asthma UK Centre in Allergic Mechanisms of Asthma , London, UK
| |
Collapse
|
39
|
Affiliation(s)
- Alexandra F Santos
- Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London , London, UK.,Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London , London, UK.,Children's Allergy Service, Evelina London, Guy's and St Thomas' Hospital , London, UK.,Asthma UK Centre in Allergic Mechanisms of Asthma , London, UK
| |
Collapse
|
40
|
Ariza A, Torres MJ, Moreno-Aguilar C, Fernández-Santamaría R, Fernández TD. Early Biomarkers for Severe Drug Hypersensitivity Reactions. Curr Pharm Des 2019; 25:3829-3839. [DOI: 10.2174/1381612825666191107105440] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 11/06/2019] [Indexed: 02/08/2023]
Abstract
Drug hypersensitivity reactions (DHRs) are typically classified into immediate and delayed reactions
based on the time interval between drug exposure and onset of symptoms. Clinical manifestations range from
mild to severe and life-threatening reactions. The most severe clinical entities are anaphylaxis and anaphylactic
shock for immediate reactions, and severe cutaneous adverse reactions such as Steven Johnson Syndrome and
Toxic Epidermal Necrolysis for delayed reactions. The diagnosis is complex and challenging, as drug provocation
tests and even skin tests can be very risky procedures, which makes them not recommended. Therefore, it is necessary
to search for useful early biomarkers to manage the diagnosis of these reactions. These biomarkers could
be useful to determine the clinical entity, but not to identify the culprit drug. Some of the currently available
biomarkers are few genetic associations of drug allergy with polymorphisms of human leukocyte antigen (HLA),
the detection of inflammatory and lipid mediators in serum, or the detection of cytokines, chemokines, and cytotoxic
markers in skin biopsies. In this literature review, it has been summarize the immunological mechanisms
involved in severe reactions, both immediate and delayed, and different early biomarkers: those currently used for
the diagnosis of these reactions as well as possible early biomarkers that could be useful with further studies to
standardize their clinical use.
Collapse
Affiliation(s)
- Adriana Ariza
- Allergy Research Group, Instituto de Investigacion Biomedica de Malaga-IBIMA, Malaga, Spain
| | - Maria J. Torres
- Allergy Research Group, Instituto de Investigacion Biomedica de Malaga-IBIMA, Malaga, Spain
| | - Carmen Moreno-Aguilar
- Immunology and Allergy Unit, IMIBICHospital Universitario Reina Sofía, Córdoba, Spain
| | | | - Tahia D. Fernández
- Allergy Research Group, Instituto de Investigacion Biomedica de Malaga-IBIMA, Malaga, Spain
| |
Collapse
|
41
|
Gunasekara P, Handunnetti SM, Premawansa S, Kaluarachchi P, Karunatilake C, Ratnayake IP, Dias RKS, Premakumara GAS, Dasanayake WMDK, Seneviratne SL, de Silva R. Diagnosis of Vespa affinis venom allergy: use of immunochemical methods and a passive basophil activation test. Allergy Asthma Clin Immunol 2019; 15:80. [PMID: 31827544 PMCID: PMC6894289 DOI: 10.1186/s13223-019-0394-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 11/26/2019] [Indexed: 11/10/2022] Open
Abstract
Background Allergy to Vespa affinis venom is common in the Asia Pacific region. Venom preparations for diagnosis are not commercially available for this species. Methods The prominent allergens in V. affinis venom were identifiedusing immunochemical methods. Use of ImmunoCAP of Vespula vulgaris crude venom/its components and a passive basophil activation test (BAT) in the diagnosis of patients who had anaphylaxis to V. affinis venom (n = 30) were also accessed. The IgE double-positivity rates (positive to both hornet and honeybee) in ImmunoCAP and the passive BAT were determined. Results High IgE reactivity was seen with the five allergens in V. affinis venom; 96% (29/30) for 34 and 24 kDa, 93% (28/30) for 45 kDa and 90% (27/30) reactivity for the 100 and 80 kDa respectively. IgE cross-reactivity was low with ImmunoCAP using V. vulgaris venom (43%; 13/30) and Ves v1 (3%; 1/30), but relatively high with Ves v5 (73%; 22/30). All patients (100%) were positive to V. affinis venom in passive BAT. In ImmunoCAP, a high double-positivity rate (76%; 23/30) was detected while no double-positivity was detected in passive BAT. Conclusions High IgE reactivity for five allergens of V. affinis points to the potential of using these allergens in component resolved diagnosis (CRD). The passive BAT has shown its importance as a promising diagnostic tool with high accuracy. It would be particularly useful in cases with doubtful double-positive results of other diagnostic tests.
Collapse
Affiliation(s)
- Peshala Gunasekara
- 1Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo 3, Sri Lanka
| | - S M Handunnetti
- 1Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo 3, Sri Lanka
| | - Sunil Premawansa
- 2Department of Zoology and Environment Sciences, Faculty of Science, University of Colombo, Colombo 3, Sri Lanka
| | - Pradeep Kaluarachchi
- Healthcare Division, A. Baur & Co. (Pvt.) Ltd., No. 62, Jethawana Road, Colombo 14, Sri Lanka
| | | | | | - R K S Dias
- 6Department of Zoology and Environmental Management, Faculty of Science, University of Kelaniya, Dalugama, Sri Lanka
| | - G A S Premakumara
- 7Department of Basic Sciences & Social Science, University of Colombo, Colombo, Sri Lanka
| | - W M D K Dasanayake
- 4Department of Immunology, Medical Research Institute, Colombo 8, Sri Lanka
| | - Suranjith L Seneviratne
- 8Institute of Immunity and Transplantation, Royal Free Hospital and University College London, London, UK.,9Department of Surgery, Faculty of Medicine, University of Colombo, Colombo 8, Sri Lanka
| | - Rajiva de Silva
- 4Department of Immunology, Medical Research Institute, Colombo 8, Sri Lanka
| |
Collapse
|
42
|
Roberts G, Allen K, Ballmer-Weber B, Clark A, Crevel R, Dunn Galvin A, Fernandez-Rivas M, Grimshaw KEC, Hourihane JO, Poulsen LK, van Ree R, Regent L, Remington B, Schnadt S, Turner PJ, Mills ENC. Identifying and managing patients at risk of severe allergic reactions to food: Report from two iFAAM workshops. Clin Exp Allergy 2019; 49:1558-1566. [PMID: 31631439 DOI: 10.1111/cea.13516] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 07/20/2019] [Accepted: 10/17/2019] [Indexed: 12/01/2022]
Abstract
Food allergy affects a small but important number of children and adults. Much of the morbidity associated with food allergy is driven by the fear of a severe reaction and fatalities continue to occur. Foods are the commonest cause of anaphylaxis. One of the aims of the European Union-funded Integrated Approaches to Food Allergen and Allergy Risk Management (iFAAM) project was to improve the identification and management of children and adults at risk of experiencing a severe reaction. A number of interconnected studies within the project have focused on quantifying the severity of allergic reactions; the impact of food matrix, immunological factors on severity of reactions; the impact of co-factors such as medications on the severity of reactions; utilizing single-dose challenges to understand threshold and severity of reactions; and community studies to understand the experience of patients suffering real-life allergic reactions to food. Associated studies have examined population thresholds and co-factors such as exercise and stress. This paper summarizes two workshops focused on the severity of allergic reactions to food. It outlines the related studies being undertaken in the project indicating how they are likely to impact on our ability to identify individuals at risk of severe reactions and improve their management.
Collapse
Affiliation(s)
- Graham Roberts
- University of Southampton Faculty of Medicine, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,The David Hide Asthma and Allergy Research Centre, St Mary's Hospital, Isle of Wight, UK
| | - Katie Allen
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Vic., Australia.,Department of Paediatrics, University of Melbourne, Parkville, Vic., Australia.,Institute of Inflammation and Repair, University of Manchester, Manchester, UK
| | - Barbara Ballmer-Weber
- Allergy Unit, Department of Dermatology, University Hospital, University of Zurich, Zurich, Switzerland.,Faculty of Medicine, University of Zürich, Zürich, Switzerland.,Clinic for Dermatology and Allergology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Andrew Clark
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Rene Crevel
- René Crevel Consulting Ltd, Bedford, UK.,Safety and Environmental Assurance Centre, Unilever, Colworth Science Park, Sharnbrook, Bedford, UK
| | - Audrey Dunn Galvin
- Applied Psychology and Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Montserrat Fernandez-Rivas
- Servicio de Alergia, Hospital Clınico San Carlos, IdISSC, ARADyAL, Universidad Complutense, Madrid, Spain
| | | | | | - Lars K Poulsen
- Allergy Clinic, Copenhagen University Hospital at Gentofte, Copenhagen, Denmark
| | - Ronald van Ree
- Departments of Experimental Immunology and of Otorhinolaryngology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | - Sabine Schnadt
- German Allergy and Asthma Association, Mönchengladbach, Germany
| | - Paul J Turner
- Section of Paediatrics (Allergy and Infectious Diseases), Imperial College London, London, UK
| | - E N Clare Mills
- Institute of Inflammation and Repair, University of Manchester, Manchester, UK
| |
Collapse
|
43
|
Matucci A, Nencini F, Maggi E, Vultaggio A. Hypersensitivity reactions to biologics used in rheumatology. Expert Rev Clin Immunol 2019; 15:1263-1271. [DOI: 10.1080/1744666x.2020.1684264] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Andrea Matucci
- Immunoallergology Unit, Department of Medicine and Geriatrics, Careggi University Hospital, Florence, Italy
| | - Francesca Nencini
- Immunoallergology Unit, Department of Medicine and Geriatrics, Careggi University Hospital, Florence, Italy
| | - Enrico Maggi
- Translational Immunology Unit, Immunology Area, Pediatric Hospital Bambino Gesù, I.R.C.C.S., Rome, Italy
| | - Alessandra Vultaggio
- Immunoallergology Unit, Department of Medicine and Geriatrics, Careggi University Hospital, Florence, Italy
| |
Collapse
|
44
|
Gibbs BF, Patsinakidis N, Raap U. Role of the Pruritic Cytokine IL-31 in Autoimmune Skin Diseases. Front Immunol 2019; 10:1383. [PMID: 31281316 PMCID: PMC6598004 DOI: 10.3389/fimmu.2019.01383] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/31/2019] [Indexed: 12/31/2022] Open
Abstract
Many autoimmune skin diseases, such as bullous pemphigoid (BP), psoriasis and certain types of chronic urticaria, are associated with intensive pruritus. While histamine and neuropeptides have previously been ascribed to play a role in itch that accompanies these diseases, recent evidence suggests that the pruritogenic cytokine interleukin (IL)-31 is a major driver of pruritic responses. IL-31 was originally shown to be produced by activated helper T cells, particularly Th2 cells, mast cells, macrophages and dendritic cells. However, more recent evidence demonstrated that eosinophils are a major source of this cytokine too, particularly in bullous pemphigoid. Basophils have also been shown to express the cytokine which, through autocrine action, strongly supports the production of other Th2-type cytokines from these cells. These investigations suggest that the dynamic recruitment of eosinophils and basophils in some autoimmune skin diseases could play an important role in the severity of IL-31-mediated itch. Furthermore, these studies suggest that IL-31, in addition to its pruritic actions, also has potential immunomodulatory roles in terms of supporting Th2-type immunity, which often underpins IgE-associated autoimmune diseases (such as bullous pemphigoid and urticaria) as well as allergies. While the role of IL-31 in psoriasis remains to be clarified, current evidence shows that this cytokine plays a major role in BP, chronic spontaneous urticaria and dermatomyositis. This suggests potential use of IL-31 receptor-blocking therapeutic approaches (e.g., Nemolizumab) for the treatment of IL-31-associated disorders.
Collapse
Affiliation(s)
- Bernhard F Gibbs
- Division of Experimental Allergy and Immunodermatology, University of Oldenburg, Oldenburg, Germany
| | - Nikolaos Patsinakidis
- Division of Experimental Allergy and Immunodermatology, University of Oldenburg, Oldenburg, Germany
| | - Ulrike Raap
- Division of Experimental Allergy and Immunodermatology, University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
45
|
Beck SC, Wilding T, Buka RJ, Baretto RL, Huissoon AP, Krishna MT. Biomarkers in Human Anaphylaxis: A Critical Appraisal of Current Evidence and Perspectives. Front Immunol 2019; 10:494. [PMID: 31024519 PMCID: PMC6459955 DOI: 10.3389/fimmu.2019.00494] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 02/25/2019] [Indexed: 12/12/2022] Open
Abstract
Anaphylaxis is a type I hypersensitivity reaction that is potentially fatal if not promptly treated. It is a clinical diagnosis, although measurement of serial serum total mast cell tryptase (MCT) is gold standard and may help differentiate anaphylaxis from its mimics. The performance characteristics of MCT assays in anaphylaxis has been variable in previous studies, due to multiple factors including differences in the definition of anaphylaxis, methods of MCT interpretation, clinical setting of anaphylaxis, causative agents, and timing of blood sample. An international consensus equation for MCT to interpret mast cell activation has been proposed and recently validated in the context of peri-operative anaphylaxis during general anesthesia. There has been an interest in the detection of newer biomarkers in anaphylaxis including platelet activation factor (PAF), chymase, carboxypeptidase A3, dipeptidyl peptidase I (DPPI), basogranulin, and CCL-2. The key determinants of an ideal biomarker in anaphylaxis are half-life, sample handling and processing requirements, and cost. There may be a role for metabolomics and systems biology in the exploration of novel biomarkers in anaphylaxis. Future studies applying these approaches might provide greater insight into factors determining severity, clinical risk stratification, identification of mast cell disorders and improving our understanding of this relatively complex acute immunological condition. Post mortem MCT evaluation is used in Forensic Medicine during autopsy for cases involving sudden death or suspected anaphylaxis. Interpretation of post mortem MCT is challenging since there is limited published evidence and the test is confounded by multiple variables largely linked to putrefaction and site of sampling. Thus, there is no international consensus on a reference range. In this state of the art review, we will focus on the practical challenges in the laboratory diagnosis of anaphylaxis and critically appraise (a) performance characteristics of MCT in anaphylaxis in different clinical scenarios (b) the role for novel biomarkers and (c) post mortem MCT and its role in fatal anaphylaxis.
Collapse
Affiliation(s)
- Sarah C Beck
- Department of Allergy and Immunology, Birmingham Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Thomas Wilding
- Department of Allergy and Immunology, Birmingham Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Richard J Buka
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Richard L Baretto
- Department of Allergy and Immunology, Birmingham Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Aarnoud P Huissoon
- Department of Allergy and Immunology, Birmingham Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Mamidipudi T Krishna
- Department of Allergy and Immunology, Birmingham Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom.,Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
46
|
Piliponsky AM, Shubin NJ, Lahiri AK, Truong P, Clauson M, Niino K, Tsuha AL, Nedospasov SA, Karasuyama H, Reber LL, Tsai M, Mukai K, Galli SJ. Basophil-derived tumor necrosis factor can enhance survival in a sepsis model in mice. Nat Immunol 2019; 20:129-140. [PMID: 30664762 PMCID: PMC6352314 DOI: 10.1038/s41590-018-0288-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 11/20/2018] [Indexed: 11/11/2022]
Abstract
Basophils are evolutionarily conserved in vertebrates, despite their small numbers and short lifespan, suggesting that basophils have beneficial roles in maintaining health. However, these roles are not fully defined. Here, we demonstrate that basophil-deficient mice exhibited reduced bacterial clearance, and increased morbidity and mortality, in the cecal ligation and puncture (CLP) model of sepsis. Among the several pro-inflammatory mediators we measured, tumor necrosis factor (TNF) was the only cytokine that was significantly reduced in basophil-deficient mice after CLP. In accordance with that observation, we found that mice with genetic ablation of Tnf in basophils exhibited reduced systemic TNF concentrations during endotoxemia. Moreover, during CLP, mice whose basophils could not produce TNF exhibited reduced neutrophil and macrophage TNF production and effector functions, reduced bacterial clearance, and increased mortality. Taken together, our studies show that basophils can enhance the innate immune response against bacterial infection and help prevent sepsis.
Collapse
Affiliation(s)
- Adrian M Piliponsky
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA. .,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA. .,Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA.
| | - Nicholas J Shubin
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Asha K Lahiri
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Phuong Truong
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Morgan Clauson
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Kerri Niino
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Avery L Tsuha
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Sergei A Nedospasov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Hajime Karasuyama
- Tokyo Medical and Dental University Graduate School of Medical and Dental Sciences, Tokyo, Japan
| | - Laurent L Reber
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mindy Tsai
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA, USA
| | - Kaori Mukai
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA, USA
| | - Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA, USA.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
47
|
Varricchi G, Raap U, Rivellese F, Marone G, Gibbs BF. Human mast cells and basophils-How are they similar how are they different? Immunol Rev 2019; 282:8-34. [PMID: 29431214 DOI: 10.1111/imr.12627] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mast cells and basophils are key contributors to allergies and other inflammatory diseases since they are the most prominent source of histamine as well as numerous additional inflammatory mediators which drive inflammatory responses. However, a closer understanding of their precise roles in allergies and other pathological conditions has been marred by the considerable heterogeneity that these cells display, not only between mast cells and basophils themselves but also across different tissue locations and species. While both cell types share the ability to rapidly degranulate and release histamine following high-affinity IgE receptor cross-linking, they differ markedly in their ability to either react to other stimuli, generate inflammatory eicosanoids or release immunomodulating cytokines and chemokines. Furthermore, these cells display considerable pharmacological heterogeneity which has stifled attempts to develop more effective anti-allergic therapies. Mast cell- and basophil-specific transcriptional profiling, at rest and after activation by innate and adaptive stimuli, may help to unravel the degree to which these cells differ and facilitate a clearer understanding of their biological functions and how these could be targeted by new therapies.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research, University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy
| | - Ulrike Raap
- Department of Dermatology and Allergology, University of Oldenburg, Oldenburg, Germany
| | - Felice Rivellese
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research, University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy.,Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gianni Marone
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research, University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy.,Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore" (IEOS), National Research Council (CNR), Naples, Italy
| | - Bernhard F Gibbs
- Department of Dermatology and Allergology, University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
48
|
Keet CA, Allen KJ. Advances in food allergy in 2017. J Allergy Clin Immunol 2018; 142:1719-1729. [DOI: 10.1016/j.jaci.2018.10.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/01/2018] [Accepted: 10/19/2018] [Indexed: 12/31/2022]
|
49
|
Multifaceted roles of basophils in health and disease. J Allergy Clin Immunol 2018; 142:370-380. [DOI: 10.1016/j.jaci.2017.10.042] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 09/19/2017] [Accepted: 10/10/2017] [Indexed: 01/10/2023]
|
50
|
Balbino B, Conde E, Marichal T, Starkl P, Reber LL. Approaches to target IgE antibodies in allergic diseases. Pharmacol Ther 2018; 191:50-64. [PMID: 29909239 DOI: 10.1016/j.pharmthera.2018.05.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 05/08/2018] [Indexed: 12/26/2022]
Abstract
IgE is the antibody isotype found at the lowest concentration in the circulation. However IgE can undeniably play an important role in mediating allergic reactions; best exemplified by the clinical benefits of anti-IgE monoclonal antibody (omalizumab) therapy for some allergic diseases. This review will describe our current understanding of the interactions between IgE and its main receptors FcεRI and CD23 (FcεRII). We will review the known and potential functions of IgE in health and disease: in particular, its detrimental roles in allergic diseases and chronic spontaneous urticaria, and its protective functions in host defense against parasites and venoms. Finally, we will present an overview of the drugs that are in clinical development or have therapeutic potential for IgE-mediated allergic diseases.
Collapse
Affiliation(s)
- Bianca Balbino
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France; INSERM, U1222, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Eva Conde
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France; INSERM, U1222, Paris, France; Université Pierre et Marie Curie, Paris, France; Neovacs SA, Paris, France
| | - Thomas Marichal
- GIGA-Research and Faculty of Veterinary Medicine, University of Liege, 4000, Liege, Belgium; Walloon Excellence in Life Sciences and Biotechnology, Wallonia, Belgium
| | - Philipp Starkl
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Austria; Department of Medicine I, Research Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Laurent L Reber
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France; INSERM, U1222, Paris, France.
| |
Collapse
|