1
|
Packi K, Matysiak J, Plewa S, Klupczyńska-Gabryszak A, Matuszewska E, Rzetecka N, Bręborowicz A, Matysiak J. Amino Acid Profiling Identifies Disease-Specific Signatures in IgE-Mediated and Non-IgE-Mediated Food Allergy in Pediatric Patients with Atopic Dermatitis. Biomedicines 2023; 11:1919. [PMID: 37509558 PMCID: PMC10377369 DOI: 10.3390/biomedicines11071919] [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: 06/02/2023] [Revised: 07/02/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
An IgE-mediated food allergy (FA) in atopic dermatitis (AD) children should be easily differentiated from other immune-mediated adverse effects related to food. Specific IgEs for particular protein components has provided additional diagnostic value. However, component-resolved diagnostics (CRD) has not solved all diagnostic problems either. We analysed the serum profile of 42 amino acids (AAs) in 76 AD children aged 2-60 months with an IgE-mediated FA (n = 36), with a non-IgE-mediated FA (n = 15) and without an FA (n = 25) using high-performance liquid chromatography coupled with mass spectrometry (LC-MS/MS) and an aTRAQ kit. We identified homocitrulline (Hcit), sarcosine (Sar) and L-tyrosine (Tyr) as features that differentiated the studied groups (one-way ANOVA with least significant difference post hoc test). The Hcit concentrations in the non-IgE-mediated FA group were significantly decreased compared with the IgE-mediated FA group (p = 0.018) and the control group (p = 0.008). In AD children with a non-IgE-mediated FA, the Tyr levels were also significantly reduced compared with the controls (p = 0.009). The mean concentration of Sar was the highest in the non-IgE-mediated FA group and the lowest in the IgE-mediated FA group (p = 0.047). Future studies should elucidate the involvement of these AAs in the molecular pathway of IgE- and non-IgE-mediated allergic responses.
Collapse
Affiliation(s)
- Kacper Packi
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60-806 Poznan, Poland
- AllerGen, Center of Personalized Medicine, 97-300 Piotrkow Trybunalski, Poland
| | - Joanna Matysiak
- Faculty of Health Sciences, Calisia University-Kalisz, 62-800 Kalisz, Poland
| | - Szymon Plewa
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | | | - Eliza Matuszewska
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Natalia Rzetecka
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Anna Bręborowicz
- Department of Pulmonology, Pediatric Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| | - Jan Matysiak
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| |
Collapse
|
2
|
Rossi CM, Lenti MV, Merli S, Licari A, Votto M, Marseglia GL, Di Sabatino A. Primary eosinophilic gastrointestinal disorders and allergy: Clinical and therapeutic implications. Clin Transl Allergy 2022; 12:e12146. [PMID: 35620572 PMCID: PMC9125508 DOI: 10.1002/clt2.12146] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 12/14/2022] Open
Abstract
Primary eosinophilic gastrointestinal disorders (EGID) are increasingly prevalent, immune‐mediated, chronic conditions which primarily affect pediatric and young adult patients, leading to substantial disease burden, and poor quality of life. EGID may either involve single portions of the gastrointestinal tract (i.e., esophagus, stomach, small bowel, and colon) or a combination. Their strong association with allergic disorders has been recently recognized, and although their shared pathophysiological basis remains partly elusive, this feature greatly impacts the diagnostic and treatment work‐up. We herein critically discuss the current knowledge on the association of EGID and allergic disorders, including atopic dermatitis, allergic rhinitis, allergic asthma, and food or drug allergy. In particular, we reviewed the literature focusing on their epidemiology, pathophysiological basis and mechanisms, and diagnostic strategies. Finally, we discuss the currently ongoing clinical trials targeting EGID and allergic diseases, including, among others the monoclonal antibodies dupilumab, mepolizumab, benralizumab, and lirentelimab.
Collapse
Affiliation(s)
- Carlo Maria Rossi
- First Department of Internal Medicine IRCCS Fondazione Policlinico San Matteo University of Pavia Pavia Italy
| | - Marco Vincenzo Lenti
- First Department of Internal Medicine IRCCS Fondazione Policlinico San Matteo University of Pavia Pavia Italy
| | - Stefania Merli
- First Department of Internal Medicine IRCCS Fondazione Policlinico San Matteo University of Pavia Pavia Italy
| | - Amelia Licari
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences University of Pavia Fondazione IRCCS Policlinico San Matteo Pavia Italy
| | - Martina Votto
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences University of Pavia Fondazione IRCCS Policlinico San Matteo Pavia Italy
| | - Gian Luigi Marseglia
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences University of Pavia Fondazione IRCCS Policlinico San Matteo Pavia Italy
| | - Antonio Di Sabatino
- First Department of Internal Medicine IRCCS Fondazione Policlinico San Matteo University of Pavia Pavia Italy
| |
Collapse
|
3
|
Zeng-Yun-Ou Z, Zhong-Yu J, Wei L. Bidirectional associations between eosinophils, basophils, and lymphocytes with atopic dermatitis: A multivariable Mendelian randomization study. Front Immunol 2022; 13:1001911. [PMID: 36569933 PMCID: PMC9780468 DOI: 10.3389/fimmu.2022.1001911] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
Background Despite being prone to reverse causation and having unmeasured confounding factors, many clinical observational studies have highlighted the critical association between basophils, eosinophils, and lymphocytes and atopic dermatitis (AD). Whether these cells play a causal role in AD development remains uncertain. Methods Data were obtained from the UK Biobank and the Blood Cell Consortium, from a large publicly available genome-wide association study (GWAS) with more than 500,000 subjects of European ancestry and for AD from three independent cohorts with more than 700,000 subjects of European ancestry. We performed single-variable Mendelian randomization (SVMR), followed by multivariable Mendelian randomization (MVMR) to assess the total and direct effects of immune cell counts on AD risk. Results SVMR estimates showed that genetically predicted higher eosinophil [odds ratio (OR): 1.23, 95% confidence interval (CI): 1.17-1.29, p = 5.85E-16] and basophil counts (OR: 1.11, 95% CI: 1.03-1.19, p = 0.004) had an adverse effect on the risk of AD, while a higher lymphocyte count (OR: 0.93, 95% CI: 0.89-0.98, p = 0.006) decreased the risk. Reverse MR analysis showed higher basophil (beta: 0.04, 95% CI: 0.01-0.07, p = 0.014) and lower lymphocyte counts (beta: -0.05, 95% CI: -0.09 to -0.01, p = 0.021) in patients with AD. In MVMR, the effects of eosinophils (OR: 1.19, 95% CI: 1.09-1.29, p = 8.98E-05), basophils (OR: 1.19, 95% CI: 1.14-1.24, p = 3.72E-15), and lymphocytes (OR: 0.93, 95% CI: 0.89-0.98, p = 0.006) were still significant. Discussion Mendelian randomization (MR) findings suggest that an increase in the eosinophil and basophil counts and a decrease in the lymphocyte counts are potential causal risk factors for AD. These risk factors are independent of each other.
Collapse
Affiliation(s)
- Zhang Zeng-Yun-Ou
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Zhong-Yu
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, China
| | - Li Wei
- Department of Dermatology and Rare Disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Li Wei,
| |
Collapse
|
4
|
Review of eosinophilic oesophagitis in children and young people. Eur J Pediatr 2021; 180:3471-3475. [PMID: 34173043 DOI: 10.1007/s00431-021-04174-0] [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: 04/19/2021] [Revised: 06/12/2021] [Accepted: 06/17/2021] [Indexed: 10/21/2022]
Abstract
Eosinophilic oesophagitis is a relapsing inflammatory disorder involving oesophagus identified over 30 years ago. Diagnosis is made by upper gastrointestinal endoscopy and oesophageal biopsies. There is huge variation in management practices across the globe. Therapeutic options include the use of proton pump inhibitors, topical steroids, and elimination diet. Biologics and immunomodulator drugs are being explored but not yet recommended in children. Long-term treatment may be required to control symptoms and to prevent complications such as fibrosis/stricture.Conclusion: Even though clinicians have better understanding of eosinophilic oesophagitis, further research is warranted in exploring the unmet needs of developing a highly sensitive non-invasive biomarker for its diagnosis and response to treatment along with a robust and easily deliverable therapeutic option. What is Known: •Incidence of eosinophilic oesophagitis has increased over the recent years. •Diagnostic confirmation requires upper gastrointestinal endoscopy and therapeutic options include elimination diet and/or topical steroids. What is New: •There is a lack of consensus ion management strategy with wide variation across the globe. •There is a need to develop a highly reliable and non-invasive biomarker to diagnose eosinophilic oesophagitis and to monitor the response to treatment.
Collapse
|
5
|
Atopy Patch Testing with Aeroallergens and Food Proteins. Contact Dermatitis 2021. [DOI: 10.1007/978-3-030-36335-2_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
6
|
Cafone J, Capucilli P, Hill DA, Spergel JM. Eosinophilic esophagitis during sublingual and oral allergen immunotherapy. Curr Opin Allergy Clin Immunol 2020; 19:350-357. [PMID: 31058677 DOI: 10.1097/aci.0000000000000537] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The aim of this review is to discuss the current evidence regarding the development of eosinophilic esophagitis (EoE) in individuals undergoing oral and sublingual immunotherapy (SLIT) for both food and environmental allergens. Cumulative incidence of EoE in patients on allergen immunotherapy for peanut, milk, and egg is estimated. RECENT FINDINGS De novo development of EoE in patients undergoing oral and SLIT has been demonstrated on the scale of case reports and prospective randomized trials. However, few individuals with EoE-like symptoms during immunotherapy undergo endoscopy, and the long-term outcomes of immunotherapy-associated EoE are unknown. SUMMARY Evidence exists to suggest that allergen immunotherapy could place individuals at risk for the development of EoE, the true incidence of which may vary depending on antigen exposure and methods used to define the condition.
Collapse
Affiliation(s)
- Joseph Cafone
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia
| | - Peter Capucilli
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia
| | - David A Hill
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia.,Department of Pediatrics, Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jonathan M Spergel
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia.,Department of Pediatrics, Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
Darsow U, Balzer C, Mahler V, Ring J. Atopy Patch Testing with Aeroallergens and Food Proteins. Contact Dermatitis 2020. [DOI: 10.1007/978-3-319-72451-5_25-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
8
|
Aceves S, Collins MH, Rothenberg ME, Furuta GT, Gonsalves N. Advancing patient care through the Consortium of Eosinophilic Gastrointestinal Disease Researchers (CEGIR). J Allergy Clin Immunol 2019; 145:28-37. [PMID: 31758958 PMCID: PMC6981250 DOI: 10.1016/j.jaci.2019.11.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/15/2019] [Accepted: 11/15/2019] [Indexed: 12/19/2022]
Abstract
Recent advances in rare disease research are accelerated by the work of consortia that have been supported by the National Institutes of Health. Development of such consortia rely on multidisciplinary relationships and engagement with patient advocacy groups, as well as the National Institutes of Health and industry and academic partners. In this rostrum we present the development of such a process that focuses on eosinophilic gastrointestinal diseases. Principal investigators, patient advocacy groups, research assistants, and trainees work together to perform natural history studies that promote clinical trial readiness tools, conduct clinical trials, train a new generation of investigators, and perform innovative pilot studies.
Collapse
Affiliation(s)
- Seema Aceves
- Division of Allergy Immunology, University of California, San Diego, and Rady Children's Hospital, San Diego, La Jolla, Calif
| | - Margaret H Collins
- Division of Pathology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Glenn T Furuta
- Digestive Health Institute, Children's Hospital Colorado, Gastrointestinal Eosinophilic Diseases Program, University of Colorado School of Medicine, Aurora, Colo.
| | - Nirmala Gonsalves
- Division of Gastroenterology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| |
Collapse
|
9
|
Capucilli P, Hill DA. Allergic Comorbidity in Eosinophilic Esophagitis: Mechanistic Relevance and Clinical Implications. Clin Rev Allergy Immunol 2019; 57:111-127. [PMID: 30903437 PMCID: PMC6626558 DOI: 10.1007/s12016-019-08733-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Allergic eosinophilic esophagitis (EoE) is a chronic, allergen-mediated inflammatory disease of the esophagus, and the most common cause of prolonged dysphagia in children and young adults in the developed world. While initially undistinguished from gastroesophageal reflux disease-associated esophageal eosinophilia, EoE is now recognized as a clinically distinct entity that shares fundamental inflammatory features of other allergic conditions and is similarly increasing in incidence and prevalence. The clinical and epidemiologic associations between EoE and other allergic manifestations are well established. In addition to exaggerated rates of atopic dermatitis, IgE-mediated food allergy, asthma, and allergic rhinitis in EoE patients, each of these allergic manifestations imparts individual and cumulative risk for subsequent EoE diagnosis. As such, EoE may be a member of the "allergic march"-the natural history of allergic manifestations during childhood. Several determinants likely contribute to the relationship between these conditions, including shared genetic, environmental, and immunologic factors. Herein, we present a comprehensive review of allergic comorbidity in EoE. We discuss areas of the genome associated with both EoE and other allergic diseases, including the well-studied variants encoding thymic stromal lymphopoietin and calpain 14, among other "atopic" regions. We summarize ways that environmental factors (such as microbiome-altering pressures and aeroallergen exposure) may predispose to multiple allergic conditions including EoE. Finally, we touch on some fundamental features of type 2 inflammation, and the resulting implications for the development of multiple allergic manifestations. We conclude with an analysis of the "type 2" biologics, and how mechanistic similarities between EoE and the other allergic manifestations have important implications for screening and treatment of the allergic patient.
Collapse
Affiliation(s)
- Peter Capucilli
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Abramson Research Building, 3615 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - David A Hill
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Abramson Research Building, 3615 Civic Center Blvd, Philadelphia, PA, 19104, USA.
- Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
10
|
Weerasekera K, Sim D, Coughlan F, Inns S. Eosinophilic esophagitis incidence in New Zealand: high but not increasing. Clin Exp Gastroenterol 2019; 12:367-374. [PMID: 31534357 PMCID: PMC6681430 DOI: 10.2147/ceg.s216126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/16/2019] [Indexed: 02/06/2023] Open
Abstract
Background and aim Eosinophilic esophagitis (EoE) is an immune-mediated inflammatory condition of the esophagus. Recent literature has shown an increasing incidence of the disease. However, no epidemiological data exist regarding New Zealand rates of EoE. The disease is associated with atopy, and New Zealand's high rate of atopic disease means the disease may be important in our population. We carried out a retrospective study to describe the incidence of EoE in the Wellington region of New Zealand, as well as key histological and clinical factors associated with the disease. Method A search was made of laboratory and endoscopic databases in the Wellington region to identify all diagnosed cases in the five years between January 1, 2011, and December 31, 2015. Case notes were examined to determine the key demographic and clinical parameters in the cases. Incidence rates were calculated for each year, and the effects of age group and sex on the incidence rates were analyzed. Result We found 152 cases of EoE in the Wellington region with an annual incidence of 6.95 per 100,000 person/years. We found no evidence of a significant difference in incidence rates by year in our study population. There was a significantly lower incidence rate in those aged <16 compared to those aged ≥16 (RR=0.26). Males had a higher incidence rate than females with an estimated rate ratio of 2.45 (p<0.05). Conclusion Our results are in contrast to previous reports of increasing incidence rates and may reflect a leveling off of incidence. Further research is needed to determine whether the low incidence in our pediatric age group is due to ascertainment bias or due to a real difference in the epidemiology of EoE in NZ compared to other countries.
Collapse
Affiliation(s)
| | - Dalice Sim
- Department of Public Health, University of Otago, Wellington, New Zealand
| | - Finbarr Coughlan
- Department of Pathology, Capital & Coast District Health Board, Wellington, New Zealand
| | - Stephen Inns
- Department of Medicine, University of Otago, Wellington, New Zealand
| |
Collapse
|
11
|
Carr S, Chan ES, Watson W. Correction to: Eosinophilic esophagitis. Allergy Asthma Clin Immunol 2019; 15:22. [PMID: 31007687 PMCID: PMC6456987 DOI: 10.1186/s13223-019-0336-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 03/19/2019] [Indexed: 12/12/2022] Open
Abstract
Eosinophilic esophagitis (EoE) is an atopic condition of the esophagus that has become increasingly recognized over the last 15 years. Diagnosis of the disorder is dependent on the patient’s clinical manifestations, and must be confirmed by histologic findings on esophageal mucosal biopsies. Patients with EoE should be referred to an allergist for optimal management, which may include dietary modifications and pharmacologic agents such as proton pump inhibitors (PPI) and corticosteroids, and for the diagnosis and management of comorbid atopic conditions. Mechanical dilation of the esophagus may also be necessary. The epidemiology, pathophysiology, diagnosis, treatment, and prognosis of EoE are discussed in this review.
Collapse
Affiliation(s)
- Stuart Carr
- 1Department of Pediatrics, University of Alberta, Edmonton, AB Canada
| | - Edmond S Chan
- 2Division of Allergy & Immunology, Department of Pediatrics, University of British Columbia, Vancouver, BC Canada.,3EoE Clinic, BC Children's Hospital, Vancouver, BC Canada
| | - Wade Watson
- 4Division of Allergy, Department of Pediatrics, IWK Health Centre, Dalhousie University, Halifax, NS Canada
| |
Collapse
|
12
|
Ramaswamy AT, No JS, Anderson L, Solomon A, Ciecierega T, Barfield E, Chien K, Schnoll‐Sussman F, Reisacher WR. Esophageal IgE, IgG4, and mucosal eosinophilia in individuals with dysphagia. Int Forum Allergy Rhinol 2019; 9:870-875. [DOI: 10.1002/alr.22339] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 02/25/2019] [Accepted: 03/12/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Apoorva T. Ramaswamy
- Department of Otolaryngology–Head and Neck SurgeryWeill Cornell Medical College New York NY
| | | | | | - Aliza Solomon
- Department of Pediatric GastroenterologyWeill Cornell Medical College New York NY
| | - Thomas Ciecierega
- Department of Pediatric GastroenterologyWeill Cornell Medical College New York NY
| | - Elaine Barfield
- Department of Pediatric GastroenterologyWeill Cornell Medical College New York NY
| | - Kimberly Chien
- Department of Pediatric GastroenterologyWeill Cornell Medical College New York NY
| | | | - William R. Reisacher
- Department of Otolaryngology–Head and Neck SurgeryWeill Cornell Medical College New York NY
| |
Collapse
|
13
|
Vinit C, Dieme A, Courbage S, Dehaine C, Dufeu C, Jacquemot S, Lajus M, Montigny L, Payen E, Yang D, Dupont C. Eosinophilic esophagitis: Pathophysiology, diagnosis, and management. Arch Pediatr 2019; 26:182-190. [DOI: 10.1016/j.arcped.2019.02.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 12/10/2018] [Accepted: 02/03/2019] [Indexed: 12/31/2022]
|
14
|
Mehr S, Brown-Whitehorn T. What do allergists in practice need to know about non-IgE-mediated food allergies. Ann Allergy Asthma Immunol 2019; 122:589-597. [PMID: 30935977 DOI: 10.1016/j.anai.2019.03.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/21/2019] [Accepted: 03/22/2019] [Indexed: 02/07/2023]
Affiliation(s)
- Sam Mehr
- Department of Allergy and Immunology, Royal Children's Hospital, Melbourne, Victoria, Australia; Department of Allergy and Immunology, Children's Hospital at Westmead, Sydney, New South Wales, Australia.
| | - Terri Brown-Whitehorn
- Division of Allergy and Immunology, Department of Pediatric, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
15
|
Iuliano S, Minelli R, Vincenzi F, Gaiani F, Ruberto C, Leandro G, Bizzarri B, Nouvenne A, Di Mario F, De'Angelis GL. Eosinophilic esophagitis in pediatric age, state of the art and review of the literature. ACTA BIO-MEDICA : ATENEI PARMENSIS 2018; 89:20-26. [PMID: 30561413 PMCID: PMC6502212 DOI: 10.23750/abm.v89i8-s.7866] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Indexed: 02/06/2023]
Abstract
Eosinophilic esophagitis (EoE) is a chronic immune-mediated relapsing disease caused by eosinophilic infiltration of the esophageal mucosa which is normally lacking these cells. EoE belongs to the group of the so called Eosinophilic Gastrointestinal Disorders (EGIDs). From a rare and unusual disease, EoE has become an emerging entity and in recent years its incidence and prevalence have increased all over the world, also in children. The pathogenesis is very complex and still not completely clear. Esophageal disfunction symptoms (e.g. dysphagia and food impaction) represent the typical manifestation of EoE and this condition could be difficult to recognize, more in pediatric age than in adults. Moreover, symptoms can often overlap with those of gastro-esophageal reflux disease (GERD), leading to a delayed diagnosis. EoE is often related to atopy and an allergological evaluation is recommended. Untreated EoE could provoke complications such as strictures, esophageal rings, narrowing of the esophagus. Diagnosis is confirmed by the demonstration in biopsy specimens obtained through upper endoscopy of eosinophilic inflammation (>15 for high powered field) of the esophageal mucosa and other histological features. Other tests could be useful not specifically for the diagnosis, but for the characterization of the subtype of EoE. Since EoE incidence and knowledge about physiopathology and natural history have increased, the goal of the review is to provide some helpful tools for the correct management in pediatric age together with an overview about epidemiology, pathogenesis, clinical, diagnosis and treatment of the disease.
Collapse
Affiliation(s)
- Silvia Iuliano
- Gastroenterology and Endoscopy Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
OBJECTIVES Eosinophilic esophagitis (EoE) is an inflammatory, atopic disease of the esophagus without a clear etiology. Our objective was to identify exposures and conditions in early infancy associated with the development of EoE. METHODS A case-control study was performed using the Military Health System Database. Subjects diagnosed with EoE from October 2008 to September 2015 were matched 1:2 on age and sex. Early infant risk factors from the first 6 months of life were investigated. RESULTS A total of 1410 cases with EoE were matched to 2820 controls. The median (interquartile range) age at diagnosis of EoE was 4.2 years (2.1-7.2) and 68.7% were boys. Proton pump inhibitors (adjusted odds ratio [aOR], 2.73; 95% confidence interval [CI] 1.93-3.88), histamine-2 receptor antagonists (aOR, 1.64; 95% CI 1.27-2.13), and antibiotics (aOR, 1.31; 95% CI 1.10-1.56) were associated with EoE. Prematurity (aOR, 1.46; 95% CI 1.12-1.89) and early manifestations of atopic disease such as milk protein allergy (aOR, 2.37; 95% CI 1.26-4.44) and eczema (aOR, 1.97; 95% CI 1.64-2.36) were related to increased odds for EoE. Erythema toxicum in infancy was strongly associated with a diagnosis of EoE (aOR 3.52; 95% CI 1.03-12.04). Infants with feeding difficulty (aOR, 1.45; 95% CI 1.18-1.77) and gastroesophageal reflux disease (aOR, 1.79; 96% CI 1.43-2.26) were also at increased risk for EoE. CONCLUSIONS Acid-blocking medications and antibiotics during infancy were associated with later diagnosis of EoE. Erythema toxicum neonatorum, an eosinophilic immune phenomenon, was strongly associated with EoE. Identifying early infant risk factors for EoE may help to risk stratify the need for endoscopy.
Collapse
|
17
|
Ferguson AE, Mukkada VA, Fulkerson PC. Pediatric Eosinophilic Esophagitis Endotypes: Are We Closer to Predicting Treatment Response? Clin Rev Allergy Immunol 2018; 55:43-55. [PMID: 29270819 DOI: 10.1007/s12016-017-8658-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Eosinophilic esophagitis (EoE) is a chronic, food antigen-driven gastrointestinal disease that is characterized by esophageal eosinophilia. Currently, there are no Food and Drug Administration (FDA)-approved treatments for EoE, but the two most commonly prescribed therapies include topical corticosteroids and food elimination diets. Clinical trials have revealed a significant proportion of cases that are resistant to topical corticosteroids, and although we define EoE as a food antigen-driven disease, not all patients with EoE respond to elimination diets or even elemental diets. The varied response to treatments highlights the heterogeneity of EoE and the need for new treatment strategies. Despite the clinical differences in treatment response, predicting the outcome remains difficult since factors including age, histologic severity at diagnosis, atopic history, and anthropometrics are not predictive of treatment response. In our practice at an academic pediatric referral center, we observe distinct clinical EoE phenotypes, including cases with atopy, connective tissue disorders, or responsiveness to a proton pump inhibitor. Similar to the work in progress with asthma, stratification of patients with EoE by clinical phenotypes and/or molecular endotypes will likely assist with therapy selection and prediction of natural history. Molecular analysis with gene expression panels also shows promise in helping us classify patients based on molecular endotypes. In additional to the clinical and molecular classifications, more accurate histologic diagnostic criteria for EoE may help us tease out small differences between patient cohorts. Despite the leaps in knowledge over the past decade regarding EoE pathogenesis, it remains a challenge to predict the response to treatment. Future studies focused on molecular, genetic, and immunologic analyses of larger patient cohorts are needed to assist in identifying EoE phenotypes and endotypes as we attempt to improve patient outcomes in pediatric EoE.
Collapse
Affiliation(s)
- Anna E Ferguson
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Vince A Mukkada
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Patricia C Fulkerson
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, ML7028, Cincinnati, OH, 45229, USA.
| |
Collapse
|
18
|
Abstract
Eosinophilic esophagitis (EoE) is an atopic condition of the esophagus that has become increasingly recognized over the last 15 years. Diagnosis of the disorder is dependent on the patient’s clinical manifestations, and must be confirmed by histologic findings on esophageal mucosal biopsies. Patients with EoE should be referred to an allergist for optimal management, which may include dietary modifications and pharmacologic agents such as corticosteroids, and for the diagnosis and management of comorbid atopic conditions. Mechanical dilation of the esophagus may also be necessary. The epidemiology, pathophysiology, diagnosis, treatment, and prognosis of EoE are discussed in this review.
Collapse
|
19
|
Schoepfer A, Blanchard C, Dawson H, Lucendo A, Mauro A, Ribi C, Safroneeva E, Savarino EV, Penagini R. Eosinophilic esophagitis: latest insights from diagnosis to therapy. Ann N Y Acad Sci 2018; 1434:84-93. [DOI: 10.1111/nyas.13731] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/06/2018] [Accepted: 03/21/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Alain Schoepfer
- Division of Gastroenterology and HepatologyCentre Hospitalier Universitaire Vaudois/CHUV and University of Lausanne (UNIL) Lausanne Switzerland
| | | | - Heather Dawson
- Institute of PathologyUniversity of Bern Bern Switzerland
| | - Alfredo Lucendo
- Department of GastoenterologyHospital General de Tomelloso Tomelloso Ciudad Real Spain
| | - Aurelio Mauro
- Division of Gastroenterology, Dipartimento di Fisiopatologia Medico‐Chirurgica e dei TrapiantiUniversità degli Studi of Milan, Fondazione IRCCS Cà Granda – Ospedale Maggiore Policlinico Milan Italy
| | - Camillo Ribi
- Division of Allergy and Clinical ImmunologyCentre Hospitalier Universitaire Vaudois Lausanne Switzerland
| | | | - Edoardo V. Savarino
- Division of Gastroenterology, Department of SurgeryOncology and Gastroenterology – DiSCOG Azienza Ospedaliera di Padova Padova Italy
| | - Roberto Penagini
- Division of Gastroenterology, Dipartimento di Fisiopatologia Medico‐Chirurgica e dei TrapiantiUniversità degli Studi of Milan, Fondazione IRCCS Cà Granda – Ospedale Maggiore Policlinico Milan Italy
| |
Collapse
|
20
|
Sallis BF, Erkert L, Moñino-Romero S, Acar U, Wu R, Konnikova L, Lexmond WS, Hamilton MJ, Dunn WA, Szepfalusi Z, Vanderhoof JA, Snapper SB, Turner JR, Goldsmith JD, Spencer LA, Nurko S, Fiebiger E. An algorithm for the classification of mRNA patterns in eosinophilic esophagitis: Integration of machine learning. J Allergy Clin Immunol 2018; 141:1354-1364.e9. [PMID: 29273402 PMCID: PMC6425755 DOI: 10.1016/j.jaci.2017.11.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 11/17/2017] [Accepted: 11/30/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Diagnostic evaluation of eosinophilic esophagitis (EoE) remains difficult, particularly the assessment of the patient's allergic status. OBJECTIVE This study sought to establish an automated medical algorithm to assist in the evaluation of EoE. METHODS Machine learning techniques were used to establish a diagnostic probability score for EoE, p(EoE), based on esophageal mRNA transcript patterns from biopsies of patients with EoE, gastroesophageal reflux disease and controls. Dimensionality reduction in the training set established weighted factors, which were confirmed by immunohistochemistry. Following weighted factor analysis, p(EoE) was determined by random forest classification. Accuracy was tested in an external test set, and predictive power was assessed with equivocal patients. Esophageal IgE production was quantified with epsilon germ line (IGHE) transcripts and correlated with serum IgE and the Th2-type mRNA profile to establish an IGHE score for tissue allergy. RESULTS In the primary analysis, a 3-class statistical model generated a p(EoE) score based on common characteristics of the inflammatory EoE profile. A p(EoE) ≥ 25 successfully identified EoE with high accuracy (sensitivity: 90.9%, specificity: 93.2%, area under the curve: 0.985) and improved diagnosis of equivocal cases by 84.6%. The p(EoE) changed in response to therapy. A secondary analysis loop in EoE patients defined an IGHE score of ≥37.5 for a patient subpopulation with increased esophageal allergic inflammation. CONCLUSIONS The development of intelligent data analysis from a machine learning perspective provides exciting opportunities to improve diagnostic precision and improve patient care in EoE. The p(EoE) and the IGHE score are steps toward the development of decision trees to define EoE subpopulations and, consequently, will facilitate individualized therapy.
Collapse
Affiliation(s)
- Benjamin F. Sallis
- Department of Pediatrics, Division of Gastroenterology,
Hepatology and Nutrition, Medical University of Vienna.,Department of Medicine, Harvard Medical School, Medical
University of Vienna
| | - Lena Erkert
- Department of Pediatrics, Division of Gastroenterology,
Hepatology and Nutrition, Medical University of Vienna
| | - Sherezade Moñino-Romero
- Department of Pediatrics, Division of Gastroenterology,
Hepatology and Nutrition, Medical University of Vienna.,Department of Pediatrics and Adolescent Medicine, Medical
University of Vienna
| | - Utkucan Acar
- Department of Pediatrics, Division of Gastroenterology,
Hepatology and Nutrition, Medical University of Vienna.,Department of Medicine, Harvard Medical School, Medical
University of Vienna
| | - Rina Wu
- Department of Pediatrics, Division of Gastroenterology,
Hepatology and Nutrition, Medical University of Vienna
| | - Liza Konnikova
- Department of Pediatrics, Division of Gastroenterology,
Hepatology and Nutrition, Medical University of Vienna.,Department of Medicine, Harvard Medical School, Medical
University of Vienna
| | - Willem S. Lexmond
- Department of Pediatrics, Division of Gastroenterology,
Hepatology and Nutrition, Medical University of Vienna.,Department of Medicine, Harvard Medical School, Medical
University of Vienna
| | - Matthew J. Hamilton
- Department of Medicine, Harvard Medical School, Medical
University of Vienna.,Department of Pathology, Medical University of
Vienna.,Division of Gastroenterology, Hepatology, and Endoscopy,
Brigham and Women’s Hospital; Medical University of Vienna
| | - W. Augustine Dunn
- Department of Pediatrics, Division of Gastroenterology,
Hepatology and Nutrition, Medical University of Vienna.,Department of Medicine, Harvard Medical School, Medical
University of Vienna
| | - Zsolt Szepfalusi
- Department of Pediatrics and Adolescent Medicine, Medical
University of Vienna
| | - Jon A. Vanderhoof
- Department of Pediatrics, Division of Gastroenterology,
Hepatology and Nutrition, Medical University of Vienna
| | - Scott B. Snapper
- Department of Pediatrics, Division of Gastroenterology,
Hepatology and Nutrition, Medical University of Vienna.,Department of Medicine, Harvard Medical School, Medical
University of Vienna
| | - Jerrold R. Turner
- Department of Pathology, Medical University of
Vienna.,Division of Gastroenterology, Hepatology, and Endoscopy,
Brigham and Women’s Hospital; Medical University of Vienna
| | - Jeffrey D. Goldsmith
- Department of Pathology, Boston Children’s Hospital,
Medical University of Vienna
| | - Lisa A. Spencer
- Department of Medicine, Harvard Medical School, Medical
University of Vienna.,Department of Medicine, Division of Allergy and
Inflammation, Beth Israel Deaconess Medical Center, Boston, Medical University of
Vienna
| | - Samuel Nurko
- Department of Pediatrics, Division of Gastroenterology,
Hepatology and Nutrition, Medical University of Vienna.,Department of Medicine, Harvard Medical School, Medical
University of Vienna
| | - Edda Fiebiger
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass.
| |
Collapse
|
21
|
Vimalathas P, Farris A, Letner D, Deshpande V, Yajnik V, Shreffler W, Garber J. Integrin αM activation and upregulation on esophageal eosinophils and periostin-mediated eosinophil survival in eosinophilic esophagitis. Immunol Cell Biol 2018; 96:426-438. [PMID: 29424023 DOI: 10.1111/imcb.12018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 02/01/2018] [Accepted: 02/02/2018] [Indexed: 12/12/2022]
Abstract
Eosinophilic esophagitis (EoE) is an increasingly recognized allergic disease associated with dysphagia and esophageal fibrosis. We aimed to determine expression patterns of specific eosinophil integrins that promote eosinophilic infiltration of the esophageal epithelium, and to determine how key EoE-related cytokines influence eosinophil activation and survival. Esophageal and peripheral eosinophils were isolated from 20 adult subjects with EoE for immunophenotyping and integrin profiling using multicolor flow cytometry and immunohistochemistry. Expression signatures of eosinophil integrins were further assessed by immunohistochemistry using serial sections of esophageal biopsy specimens. Purified eosinophils were used to assess the effect of EoE-relevant cytokines and recombinant periostin on expression of known eosinophil integrins and eosinophil survival and activation. We found that resting eosinophils express high levels of the β2-pairing integrins αL and αM, and lower levels of α4, α6 and α4β7. The migration of peripheral eosinophils to the esophagus is characterized by the specific induction of αM, and a significant increase in the proportion of αM in high-activity conformation. Periostin, a secreted extracellular matrix protein that is significantly overexpressed in EoE, enhances eosinophil survival, and this effect is mediated by αM interaction. Integrin αM is a specific marker of activated tissue eosinophils in EoE, and promotes eosinophil survival through interactions with periostin. The ability of αMβ2 to mediate eosinophil tissue residency via periostin represents a key mechanism for disease development and a potential therapeutic target in EoE.
Collapse
Affiliation(s)
| | - Alexandra Farris
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Dorothea Letner
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Vikram Deshpande
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA.,Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Vijay Yajnik
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Wayne Shreffler
- Food Allergy Center, Division of Allergy & Immunology, Massachusetts General Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - John Garber
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
22
|
Lee K, Furuta GT, Nguyen N. Eosinophilic Esophagitis Is an Underlying Cause for Gastrointestinal Concerns in Children. Front Pediatr 2018; 6:113. [PMID: 29780792 PMCID: PMC5946022 DOI: 10.3389/fped.2018.00113] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/05/2018] [Indexed: 12/19/2022] Open
Abstract
Eosinophilic esophagitis (EoE) is a chronic immune antigen-mediated disorder characterized by symptoms of esophageal dysfunction in combination with dense esophageal eosinophilia. The clinical presentation of EoE can vary depending on children's age and their ability to report symptoms, therefore a high index of suspicion for EoE is required because children and teenagers may develop coping strategies around eating. The development of symptoms measurement tools in EoE assists in not only assessing symptoms, but also coping strategies children may have developed. While the diagnosis of EoE requires endoscopic evaluation with histologic assessment of esophageal mucosal biopsy samples, several emerging methods to assess and survey the esophageal mucosa have been developed. Advances in the field to better understand the natural history, clinical and molecular features of phenotypes in EoE will be important in considering novel therapeutic options and assessing outcomes.
Collapse
Affiliation(s)
- Kunsong Lee
- Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States.,Department of Pediatrics, Dankook University College of Medicine, Cheonan, South Korea
| | - Glenn T Furuta
- Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States.,Section of Pediatric Gastroenterology, Hepatology and Nutrition, Digestive Health Institute, Children's Hospital Colorado, Aurora, CO, United States
| | - Nathalie Nguyen
- Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States.,Section of Pediatric Gastroenterology, Hepatology and Nutrition, Digestive Health Institute, Children's Hospital Colorado, Aurora, CO, United States
| |
Collapse
|
23
|
Elisa GT, Diaz YM, Lozano LM, Ortega AM, Sanchez-Migallon JR, Segade JM, Brito JFF, Rodríguez RG. Eosinophilic Esophagitis due to Gluten without Celiac Disease and Unusual Comorbidities. EXPLORATORY RESEARCH AND HYPOTHESIS IN MEDICINE 2017; 2:41-42. [DOI: 10.14218/erhm.2016.00011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
24
|
Pelletier T, Tamayev R, Iammatteo M, Nautsch D, Hudes G, Lukin D, Jerschow E. Eosinophilic esophagitis as possible complication of aspirin treatment in patient with aspirin-exacerbated respiratory disease. Ann Allergy Asthma Immunol 2016; 118:120-122. [PMID: 27865717 DOI: 10.1016/j.anai.2016.10.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/13/2016] [Accepted: 10/20/2016] [Indexed: 10/20/2022]
Affiliation(s)
- Teresa Pelletier
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Robert Tamayev
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Melissa Iammatteo
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Deborah Nautsch
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Golda Hudes
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Dana Lukin
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Elina Jerschow
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York.
| |
Collapse
|
25
|
Muir AB, Benitez AJ, Dods K, Spergel JM, Fillon SA. Microbiome and its impact on gastrointestinal atopy. Allergy 2016; 71:1256-63. [PMID: 27240281 DOI: 10.1111/all.12943] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2016] [Indexed: 01/01/2023]
Abstract
The prevalence of allergic conditions has continuously increased in the last few decades in Westernized countries. A dysbiotic gut microbiome may play an important role in the development of allergic diseases. Genetic, environmental, and dietary factors may alter the commensal microbiota leading to inflammatory dysregulation of homeostasis. Murine and human studies have begun to elucidate the role of the microbiota in the pathogenesis of atopic diseases including asthma, atopic dermatitis, and food allergies. However, the role of the microbiome in most eosinophilic gastrointestinal diseases (EGIDs) is not yet known. This review provides an overview of what is currently known about the development of tolerance from both molecular and clinical standpoints. We also look at the gut-specific microbiome and its role in atopic conditions with the hope of applying this knowledge to the understanding, prevention, and treatment of EGIDs, particularly EoE.
Collapse
Affiliation(s)
- A. B. Muir
- Division of Gastroenterology, Hepatology, and Nutrition; The Children's Hospital of Philadelphia; Philadelphia PA USA
- Department of Pediatrics; Perelman School of Medicine at the University of Pennsylvania; Philadelphia PA USA
| | - A. J. Benitez
- Division of Allergy and Immunology; The Children's Hospital of Philadelphia; Philadelphia PA USA
| | - K. Dods
- Division of Gastroenterology, Hepatology, and Nutrition; The Children's Hospital of Philadelphia; Philadelphia PA USA
| | - J. M. Spergel
- Department of Pediatrics; Perelman School of Medicine at the University of Pennsylvania; Philadelphia PA USA
- Division of Allergy and Immunology; The Children's Hospital of Philadelphia; Philadelphia PA USA
| | - S. A. Fillon
- University of Colorado School of Medicine; Aurora CO USA
- Digestive Health Institute; Section of Pediatric Gastroenterology, Hepatology and Nutrition; Gastrointestinal Eosinophilic Diseases Program; Mucosal Inflammation Program; Children's Hospital Colorado; Aurora CO USA
| |
Collapse
|
26
|
Mudde ACA, Lexmond WS, Blumberg RS, Nurko S, Fiebiger E. Eosinophilic esophagitis: published evidences for disease subtypes, indications for patient subpopulations, and how to translate patient observations to murine experimental models. World Allergy Organ J 2016; 9:23. [PMID: 27458501 PMCID: PMC4947322 DOI: 10.1186/s40413-016-0114-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 07/01/2016] [Indexed: 02/06/2023] Open
Abstract
Eosinophilic esophagitis (EoE) is a chronic inflammatory disorder of the esophagus and commonly classified as a Th2-type allergy. Major advances in our understanding of the EoE pathophysiology have recently been made, but clinicians struggle with highly unpredictable therapy responses indicative of phenotypic diversity within the patient population. Here, we summarize evidences for the existence of EoE subpopulations based on diverse inflammatory characteristics of the esophageal tissue in EoE. Additionally, clinical characteristics of EoE patients support the concept of disease subtypes. We conclude that clinical and experimental evidences indicate that EoE is an umbrella term for conditions that are unified by esophageal eosinophilia but that several disease subgroups with various inflammatory esophageal patterns and/or different clinical features exist. We further discuss strategies to study the pathophysiologic differences as observed in EoE patients in murine experimental EoE. Going forward, models of EoE that faithfully mimic EoE subentities as defined in humans will be essential because mechanistic studies on triggers which regulate the onset of diverse EoE subpopulations are not feasible in patients. Understanding how and why different EoE phenotypes develop will be a first and fundamental step to establish strategies that integrate individual variations of the EoE pathology into personalized therapy.
Collapse
Affiliation(s)
- Anne C A Mudde
- Department of Medicine, Harvard Medical School, and Division of Gastroenterology and Nutrition, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115 USA
| | - Willem S Lexmond
- Department of Medicine, Harvard Medical School, and Division of Gastroenterology and Nutrition, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115 USA
| | - Richard S Blumberg
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA USA
| | - Samuel Nurko
- Department of Medicine, Harvard Medical School, and Division of Gastroenterology and Nutrition, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115 USA ; Center for Motility and Functional Gastrointestinal Disorders, Boston, MA USA ; Eosinophilic Gastrointestinal Disease Center, Boston Children's Hospital, Boston, MA USA
| | - Edda Fiebiger
- Department of Medicine, Harvard Medical School, and Division of Gastroenterology and Nutrition, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115 USA
| |
Collapse
|