1
|
Ogulur I, Pat Y, Yazici D, Ardicli S, Ardicli O, Mitamura Y, Akdis M, Akdis CA. Epithelial barrier dysfunction, type 2 immune response, and the development of chronic inflammatory diseases. Curr Opin Immunol 2024; 91:102493. [PMID: 39321494 DOI: 10.1016/j.coi.2024.102493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 09/07/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
Abstract
The prevalence of many chronic noncommunicable diseases has been steadily rising over the past six decades. During this time, humans have been increasingly exposed to substances toxic for epithelial cells, including air pollutants, laundry and dishwashers, household chemicals, toothpaste, food additives, microplastics, and nanoparticles, introduced into our daily lives as part of industrialization, urbanization, and modernization. These substances disrupt the epithelial barriers and lead to microbial dysbiosis and cause immune response to allergens, opportunistic pathogens, bacterial toxins, and autoantigens followed by chronic inflammation due to epigenetic mechanisms. Recent evidence from studies on the mechanisms of epithelial barrier damage has demonstrated that even trace amounts of toxic substances can damage epithelial barriers and induce tissue inflammation. Further research in this field is essential for our understanding of the causal substances and molecular mechanisms involved in the initiation of leaky epithelial barriers that cascade into chronic inflammatory diseases.
Collapse
Affiliation(s)
- Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Ozge Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.
| |
Collapse
|
2
|
Sun N, Ogulur I, Mitamura Y, Yazici D, Pat Y, Bu X, Li M, Zhu X, Babayev H, Ardicli S, Ardicli O, D'Avino P, Kiykim A, Sokolowska M, van de Veen W, Weidmann L, Akdis D, Ozdemir BG, Brüggen MC, Biedermann L, Straumann A, Kreienbühl A, Guttman-Yassky E, Santos AF, Del Giacco S, Traidl-Hoffmann C, Jackson DJ, Wang DY, Lauerma A, Breiteneder H, Zhang L, O'Mahony L, Pfaar O, O'Hehir R, Eiwegger T, Fokkens WJ, Cabanillas B, Ozdemir C, Kistler W, Bayik M, Nadeau KC, Torres MJ, Akdis M, Jutel M, Agache I, Akdis CA. The epithelial barrier theory and its associated diseases. Allergy 2024; 79:3192-3237. [PMID: 39370939 DOI: 10.1111/all.16318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024]
Abstract
The prevalence of many chronic noncommunicable diseases has been steadily rising over the past six decades. During this time, over 350,000 new chemical substances have been introduced to the lives of humans. In recent years, the epithelial barrier theory came to light explaining the growing prevalence and exacerbations of these diseases worldwide. It attributes their onset to a functionally impaired epithelial barrier triggered by the toxicity of the exposed substances, associated with microbial dysbiosis, immune system activation, and inflammation. Diseases encompassed by the epithelial barrier theory share common features such as an increased prevalence after the 1960s or 2000s that cannot (solely) be accounted for by the emergence of improved diagnostic methods. Other common traits include epithelial barrier defects, microbial dysbiosis with loss of commensals and colonization of opportunistic pathogens, and circulating inflammatory cells and cytokines. In addition, practically unrelated diseases that fulfill these criteria have started to emerge as multimorbidities during the last decades. Here, we provide a comprehensive overview of diseases encompassed by the epithelial barrier theory and discuss evidence and similarities for their epidemiology, genetic susceptibility, epithelial barrier dysfunction, microbial dysbiosis, and tissue inflammation.
Collapse
Affiliation(s)
- Na Sun
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P. R. China
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Xiangting Bu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Manru Li
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Xueyi Zhu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Genetics, Faculty of Veterinary Medicine, Bursa Uludag University, Bursa, Turkey
| | - Ozge Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Division of Food Processing, Milk and Dairy Products Technology Program, Karacabey Vocational School, Bursa Uludag University, Bursa, Turkey
| | - Paolo D'Avino
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Ayca Kiykim
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Cerrahpasa School of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Lukas Weidmann
- Department of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Deniz Akdis
- Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | | | - Marie Charlotte Brüggen
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Luc Biedermann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Alex Straumann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Andrea Kreienbühl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Emma Guttman-Yassky
- Department of Dermatology, and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - 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
- Children's Allergy Service, Evelina London Children's Hospital, Guy's and St. Thomas' Hospital, London, UK
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Stefano Del Giacco
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | | | - David J Jackson
- Guy's Severe Asthma Centre, Guy's Hospital, Guy's & St Thomas' NHS Trust, London, UK
- School of Immunology & Microbial Sciences, King's College London, London, UK
| | - De-Yun Wang
- Department of Otolaryngology, Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore City, Singapore
| | - Antti Lauerma
- Department of Dermatology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Heimo Breiteneder
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Liam O'Mahony
- Department of Medicine and School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Robyn O'Hehir
- Allergy, Asthma & Clinical Immunology, The Alfred Hospital, Melbourne, Victoria, Australia
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Thomas Eiwegger
- Translational Medicine Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
- Department of Pediatric and Adolescent Medicine, University Hospital St. Pölten, St. Pölten, Austria
| | - Wytske J Fokkens
- Department of Otorhinolaryngology & Head and Neck Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Beatriz Cabanillas
- Department of Allergy, Instituto de Investigación Biosanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Cevdet Ozdemir
- Department of Pediatric Basic Sciences, Institute of Child Health, Istanbul University, Istanbul, Turkey
- Istanbul Faculty of Medicine, Department of Pediatrics, Division of Pediatric Allergy and Immunology, Istanbul University, Istanbul, Turkey
| | - Walter Kistler
- Department of Sports Medicine, Davos Hospital, Davos, Switzerland
- Swiss Research Institute for Sports Medicine (SRISM), Davos, Switzerland
- Medical Committee International Ice Hockey Federation (IIHF), Zurich, Switzerland
| | - Mahmut Bayik
- Department of Internal Medicine and Hematology, Marmara University, Istanbul, Turkey
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Maria J Torres
- Allergy Unit, IBIMA-Hospital Regional Universitario de Málaga-ARADyAL, UMA, Málaga, Spain
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Marek Jutel
- Department of Clinical Immunology, Wrocław Medical University, Wroclaw, Poland
| | - Ioana Agache
- Faculty of Medicine, Department of Allergy and Clinical Immunology, Transylvania University, Brasov, Romania
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| |
Collapse
|
3
|
Thulin H, Säfholm J, Lundahl J, Jovic V, Adner M, Nilsson C. Granzyme B is elevated in esophageal biopsies from children with eosinophilic esophagitis. J Pediatr Gastroenterol Nutr 2024; 78:313-319. [PMID: 38374566 DOI: 10.1002/jpn3.12084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/23/2023] [Accepted: 11/28/2023] [Indexed: 02/21/2024]
Abstract
OBJECTIVES Eosinophilic esophagitis (EoE) is an immune-mediated antigen-triggered inflammatory disease of the esophagus. Our aim was to investigate inflammatory responses by an ex vivo biopsy provocation-based method, stimulating biopsies with milk, wheat, and egg extracts. METHODS An experimental study was conducted on esophageal biopsies from children who underwent esophagogastroduodenoscopy. Supernatants were collected before and after stimulation of the biopsies with food extracts and analyzed for 45 different inflammatory markers. Biopsies were also stained for histological analyzes. RESULTS Study subjects included 13 controls, 9 active EoE, and 4 EoE in remission, median age 12 years. Of the 45 markers analyzed, three had significant differences between controls and patients with active EoE, Granzyme B, (GzmB), IL-1ra, and CXCL8 (p < .05). Levels of GzmB were higher, and levels of IL-1ra were lower in patients with active EoE compared with controls and EoE in remission both at baseline and after food extract stimulation. CXCL8 increased in active EoE compared with controls only after stimulation. The number of histologically detected GzmB-positive cells were significantly higher in patients with active EoE in contrast to control and EoE remission (p < .05). CONCLUSIONS The levels of the barrier-damaging protease GzmB were higher in the supernatant both before and after stimulation with food extract ex vivo in patients with active EoE. GzmB was also observed histologically in biopsies from patients with active EoE. The presence of elevated serine protease GzmB in esophageal mucosa of children with active EoE suggests a role in the pathogenesis of this disorder.
Collapse
Affiliation(s)
- Helena Thulin
- Department of Clinical Science and Education, Karolinska Institutet, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Division of Pediatric Gastroenterology, Hepatology and Nutrition, Karolinska University Hospital, Stockholm, Sweden
| | - Jesper Säfholm
- Experimental Asthma and Allergy Research, Department of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Joachim Lundahl
- Department of Clinical Science and Education, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Viktor Jovic
- Department of Clinical Pathology and Cytology, Karolinska University Laboratory, Stockholm, Sweden
| | - Mikael Adner
- Experimental Asthma and Allergy Research, Department of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Caroline Nilsson
- Department of Clinical Science and Education, Karolinska Institutet, Stockholm, Sweden
- Sachs Children and Youth Hospital, Department of Pediatric Allergology and Pulmonology, South Hospital, Stockholm, Sweden
| |
Collapse
|
4
|
Lal M, Burk CM, Gautam R, Mrozek Z, Trachsel T, Beers J, Carroll MC, Morgan DM, Muir AB, Shreffler WG, Ruffner MA. Interferon-γ signaling in eosinophilic esophagitis has implications for epithelial barrier function and programmed cell death. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.26.577407. [PMID: 38352458 PMCID: PMC10862711 DOI: 10.1101/2024.01.26.577407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Objective Eosinophilic esophagitis (EoE) is a chronic esophageal inflammatory disorder characterized by eosinophil-rich mucosal inflammation and tissue remodeling. Transcriptional profiling of esophageal biopsies has previously revealed upregulation of type I and II interferon (IFN) response genes. We aim to unravel interactions between immune and epithelial cells and examine functional significance in esophageal epithelial cells. Design We investigated epithelial gene expression from EoE patients using single-cell RNA sequencing and a confirmatory bulk RNA-sequencing experiment of isolated epithelial cells. The functional impact of interferon signaling on epithelial cells was investigated using in vitro organoid models. Results We observe upregulation of interferon response signature genes (ISGs) in the esophageal epithelium during active EoE compared to other cell types, single-cell data, and pathway analyses, identified upregulation in ISGs in epithelial cells isolated from EoE patients. Using an esophageal organoid and air-liquid interface models, we demonstrate that IFN-γ stimulation triggered disruption of esophageal epithelial differentiation, barrier integrity, and induced apoptosis via caspase upregulation. We show that an increase in cleaved caspase-3 is seen in EoE tissue and identify interferon gamma (IFNG) expression predominantly in a cluster of majority-CD8+ T cells with high expression of CD69 and FOS. Conclusion These findings offer insight into the interplay between immune and epithelial cells in EoE. Our data illustrate the relevance of several IFN-γ-mediated mechanisms on epithelial function in the esophagus, which have the potential to impact epithelial function during inflammatory conditions.
Collapse
Affiliation(s)
- Megha Lal
- Division of Allergy and Immunology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Caitlin M. Burk
- Food Allergy Center and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Ravi Gautam
- Division of Allergy and Immunology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Zoe Mrozek
- Division of Allergy and Immunology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tina Trachsel
- Division of Allergy and Immunology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Allergy, University Children’s Hospital Zurich, Zurich, Switzerland
- Division of Allergy, University Children’s Hospital Basel, Basel, Switzerland
| | - Jarad Beers
- Division of Allergy and Immunology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Margaret C. Carroll
- Division of Allergy and Immunology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Duncan M. Morgan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT); Department of Chemical Engineering, MIT, Cambridge, MA, USA
| | - Amanda B. Muir
- Divison of Gastroenterology, Hepatology and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania
| | - Wayne G. Shreffler
- Food Allergy Center and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Melanie A. Ruffner
- Division of Allergy and Immunology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania
| |
Collapse
|
5
|
Zysk W, Mesjasz A, Trzeciak M, Horvath A, Plata-Nazar K. Gastrointestinal Comorbidities Associated with Atopic Dermatitis-A Narrative Review. Int J Mol Sci 2024; 25:1194. [PMID: 38256267 PMCID: PMC10815992 DOI: 10.3390/ijms25021194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/14/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
The current understanding of atopic dermatitis (AD) seems to be extending beyond a skin-confined condition frequently associated with allergic comorbidities, as in a number of epidemiological studies, the prevalence rate of a range of illnesses has been determined to be greater in patients with AD, or inversely. In most cases, the reasons for this are vague. A subset of these conditions are gastrointestinal disorders, including food sensitization (FS) and food allergy (FA), eosinophilic esophagitis (EoE) (it is of mixed background, both IgE-dependent and independent), food protein-induced enterocolitis syndrome (FPIES) (it exemplifies an IgE-independent food allergy), Crohn's disease (CD), colitis ulcerosa (CU), celiac disease, irritable bowel syndrome (IBS), and gastroesophageal reflux disease (GERD). In this review, we performed a comprehensive search of the literature using the PubMed database. We addressed the epidemiology of the increased co-occurrence of these diseases with AD and discussed potential causes for this subject. Multiple gastroenterological comorbidities appear to be more common in patients with AD, according to our review. The mechanisms that underlie this phenomenon are largely unknown, highlighting the need for further study in this field.
Collapse
Affiliation(s)
- Weronika Zysk
- Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdansk, Poland;
| | - Alicja Mesjasz
- Dermatological Students Scientific Association, Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdansk, Poland;
| | - Magdalena Trzeciak
- Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdansk, Poland;
| | - Andrea Horvath
- Department of Paedistrics, Medical University of Warsaw, Żwirki I Wigury 63a, 02-091 Warszawa, Poland;
| | - Katarzyna Plata-Nazar
- Department of Paediatrics, Gastroenterology, Allergology and Paediatric Nutrition, Faculty of Medicine, Medical University of Gdańsk, Nowe Ogrody 1-6, 80-803 Gdańsk, Poland;
| |
Collapse
|
6
|
Rinaldi AO, Li M, Barletta E, D'Avino P, Yazici D, Pat Y, Ward S, Burla D, Tan G, Askary N, Larsson R, Bost J, Babayev H, Dhir R, Gaudenzio N, Akdis M, Nadeau K, Akdis CA, Mitamura Y. Household laundry detergents disrupt barrier integrity and induce inflammation in mouse and human skin. Allergy 2024; 79:128-141. [PMID: 37766519 DOI: 10.1111/all.15891] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/15/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Epithelial barrier impairment is associated with many skin and mucosal inflammatory disorders. Laundry detergents have been demonstrated to affect epithelial barrier function in vitro using air-liquid interface cultures of human epithelial cells. METHODS Back skin of C57BL/6 mice was treated with two household laundry detergents at several dilutions. Barrier function was assessed by electric impedance spectroscopy (EIS) and transepidermal water loss (TEWL) measurements after the 4 h of treatments with detergents. RNA sequencing (RNA-seq) and targeted multiplex proteomics analyses in skin biopsy samples were performed. The 6-h treatment effect of laundry detergent and sodium dodecyl sulfate (SDS) was investigated on ex vivo human skin. RESULTS Detergent-treated skin showed a significant EIS reduction and TEWL increase compared to untreated skin, with a relatively higher sensitivity and dose-response in EIS. The RNA-seq showed the reduction of the expression of several genes essential for skin barrier integrity, such as tight junctions and adherens junction proteins. In contrast, keratinization, lipid metabolic processes, and epidermal cell differentiation were upregulated. Proteomics analysis showed that the detergents treatment generally downregulated cell adhesion-related proteins, such as epithelial cell adhesion molecule and contactin-1, and upregulated proinflammatory proteins, such as interleukin 6 and interleukin 1 beta. Both detergent and SDS led to a significant decrease in EIS values in the ex vivo human skin model. CONCLUSION The present study demonstrated that laundry detergents and its main component, SDS impaired the epidermal barrier in vivo and ex vivo human skin. Daily detergent exposure may cause skin barrier disruption and may contribute to the development of atopic diseases.
Collapse
Affiliation(s)
- Arturo O Rinaldi
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Davos, Switzerland
| | - Manru Li
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Davos, Switzerland
| | - Elena Barletta
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Davos, Switzerland
| | - Paolo D'Avino
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Davos, Switzerland
| | - Siobhan Ward
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Davos, Switzerland
- Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Daniel Burla
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Davos, Switzerland
| | - Ge Tan
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Davos, Switzerland
| | | | | | | | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Davos, Switzerland
| | - Raja Dhir
- SEED, Inc, Co, Los Angeles, California, USA
| | - Nicolas Gaudenzio
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse, France
- Genoskin SAS, Toulouse, France
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Davos, Switzerland
| | - Kari Nadeau
- Department of Environmental Studies, Harvard T.H. Chan School of Public Health, Cambridge, Massachusetts, USA
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Davos, Switzerland
- Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Davos, Switzerland
| |
Collapse
|
7
|
Wakim El-Khoury J, Safroneeva E, Schoepfer AM. The Role of Esophageal Physiologic Tests in Eosinophilic Esophagitis. Inflamm Intest Dis 2024; 9:296-303. [PMID: 39659395 PMCID: PMC11631170 DOI: 10.1159/000542435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/09/2024] [Indexed: 12/12/2024] Open
Abstract
Background In patients with eosinophilic esophagitis (EoE), the correlation between symptoms of esophageal dysfunction and endoscopic and histologic disease activity is generally poor and probably related to multiple causes such as esophageal remodeling processes that might go undetected using endoscopy and histology as well as esophageal hypervigilance and symptom-specific anxiety. Hence, there is a need for a holistic management of patients that goes beyond the control of eosinophilia and symptoms. Summary and Key Messages Physiological esophageal testing using high-resolution manometry, functional lumen imaging probe, pH-impedance, wireless pH monitoring, and mucosal impedance may unveil the effects of chronic transmural fibro-inflammatory changes of the esophageal wall as well as esophageal hypervigilance, thereby assisting to phenotype patients, predict therapeutic response to therapy, and identify motility disorders that may need a specific targeted therapy to ameliorate patients' outcomes. This article discusses the role of functional esophageal examinations in the diagnosis and management of EoE.
Collapse
Affiliation(s)
- Jeanine Wakim El-Khoury
- Division of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Ekaterina Safroneeva
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Alain M. Schoepfer
- Division of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
8
|
McGowan EC, Singh R, Katzka DA. Barrier Dysfunction in Eosinophilic Esophagitis. Curr Gastroenterol Rep 2023; 25:380-389. [PMID: 37950816 DOI: 10.1007/s11894-023-00904-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2023] [Indexed: 11/13/2023]
Abstract
PURPOSE OF REVIEW Compelling evidence over the past decade supports the central role of epithelial barrier dysfunction in the pathophysiology of eosinophilic esophagitis (EoE). The purpose of this review is to summarize the genetic, environmental, and immunologic factors driving epithelial barrier dysfunction, and how this impaired barrier can further promote the inflammatory response in EoE. RECENT FINDINGS Common environmental exposures, such as detergents, may have a direct impact on the esophageal epithelial barrier. In addition, the effects of IL-13 on barrier dysfunction may be reduced by 17β-estradiol, Vitamin D, and the short chain fatty acids butyrate and propionate, suggesting novel therapeutic targets. There are many genetic, environmental, and immunologic factors that contribute to epithelial barrier dysfunction in EoE. This leads to further skewing of the immune response to a "Th2" phenotype, alterations in the esophageal microbiome, and penetration of relevant antigens into the esophageal mucosa, which are central to the pathophysiology of EoE.
Collapse
Affiliation(s)
- Emily C McGowan
- Division of Allergy and Immunology, University of Virginia School of Medicine, PO Box 801355, Charlottesville, VA, 22908, USA.
| | - Roopesh Singh
- Division of Allergy and Immunology, University of Virginia School of Medicine, PO Box 801355, Charlottesville, VA, 22908, USA
| | - David A Katzka
- Division of Digestive and Liver Disease, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| |
Collapse
|
9
|
Appanna R, Gargano D, Caputo A, De Bartolomeis F, Ricciardi L, Santonicola A, Stefanelli B, Caiazza L, Guarciariello M, D'Antonio A, D'Auria R, Conti V, Casolaro V, Iovino P. Changes in mucosal IgG4 +- and IL-10 +-cell frequencies in adults with eosinophilic esophagitis on a two-food elimination diet. Clin Immunol 2023; 257:109853. [PMID: 38013163 DOI: 10.1016/j.clim.2023.109853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/12/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2023]
Abstract
Eosinophilic esophagitis (EoE) is increasingly diagnosed in patients with dysphagia. Type-2 immunity can induce EoE histopathology via non-IgE-dependent mechanisms, possibly involving IgG4 and IL-10. To elucidate the contribution of this response to EoE pathogenesis, we examined its association with clinical and histologic endpoints in adult EoE patients given a two-food elimination diet. IgG4- and IL-10-expressing cells were counted in esophageal biopsies and serum food-specific IgG4 measured at baseline and follow-up. Variables were correlated with histologic measures of disease activity. Patients exhibited significant reduction in esophageal eosinophilia and overall histology. A significant decrease in IL-10+-cell frequencies correlated with histologic changes. In contrast, a decline in serum and esophageal IgG4, while substantial, did not correlate with IL-10+-cell frequencies or histologic parameters. These results suggest a critical role of IL-10 in EoE pathogenesis. Conversely, IgG4 expression, while reflecting exposure to food antigens, is not obviously related to EoE histopathology or IL-10 expression.
Collapse
Affiliation(s)
- Ramapraba Appanna
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
| | | | - Alessandro Caputo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy; San Giovanni di Dio e Ruggi d'Aragona University Hospital, Salerno, Italy
| | | | - Luca Ricciardi
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
| | - Antonella Santonicola
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy; San Giovanni di Dio e Ruggi d'Aragona University Hospital, Salerno, Italy
| | - Berenice Stefanelli
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
| | - Laura Caiazza
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
| | | | - Antonio D'Antonio
- San Giovanni di Dio e Ruggi d'Aragona University Hospital, Salerno, Italy
| | - Raffaella D'Auria
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
| | - Valeria Conti
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy; San Giovanni di Dio e Ruggi d'Aragona University Hospital, Salerno, Italy
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy; San Giovanni di Dio e Ruggi d'Aragona University Hospital, Salerno, Italy.
| | - Paola Iovino
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy; San Giovanni di Dio e Ruggi d'Aragona University Hospital, Salerno, Italy.
| |
Collapse
|
10
|
Pat Y, Ogulur I, Yazici D, Mitamura Y, Cevhertas L, Küçükkase OC, Mesisser SS, Akdis M, Nadeau K, Akdis CA. Effect of altered human exposome on the skin and mucosal epithelial barrier integrity. Tissue Barriers 2023; 11:2133877. [PMID: 36262078 PMCID: PMC10606824 DOI: 10.1080/21688370.2022.2133877] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/23/2022] [Accepted: 08/27/2022] [Indexed: 10/24/2022] Open
Abstract
Pollution in the world and exposure of humans and nature to toxic substances is continuously worsening at a rapid pace. In the last 60 years, human and domestic animal health has been challenged by continuous exposure to toxic substances and pollutants because of uncontrolled growth, modernization, and industrialization. More than 350,000 new chemicals have been introduced to our lives, mostly without any reasonable control of their health effects and toxicity. A plethora of studies show exposure to these harmful substances during this period with their implications on the skin and mucosal epithelial barrier and increasing prevalence of allergic and autoimmune diseases in the context of the "epithelial barrier hypothesis". Exposure to these substances causes an epithelial injury with peri-epithelial inflammation, microbial dysbiosis and bacterial translocation to sub-epithelial areas, and immune response to dysbiotic bacteria. Here, we provide scientific evidence on the altered human exposome and its impact on epithelial barriers.
Collapse
Affiliation(s)
- Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Medical Microbiology, Faculty of Medicine, Aydin Menderes University, Turkey
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Lacin Cevhertas
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Medical Immunology, Institute of Health Sciences, Bursa Uludag University, Turkey
| | - Ozan C Küçükkase
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sanne S Mesisser
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Kari Nadeau
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University and Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, Stanford, CA, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| |
Collapse
|
11
|
Salvador Nunes VS, Straumann A, Salvador Nunes L, Schoepfer AM, Greuter T. Eosinophilic Esophagitis beyond Eosinophils - an Emerging Phenomenon Overlapping with Eosinophilic Esophagitis: Collegium Internationale Allergologicum (CIA) Update 2023. Int Arch Allergy Immunol 2023; 184:411-420. [PMID: 36972571 PMCID: PMC10337666 DOI: 10.1159/000529910] [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: 01/02/2023] [Accepted: 02/17/2023] [Indexed: 03/29/2023] Open
Abstract
Having long been considered the mainstay in eosinophilic esophagitis (EoE) diagnosis and pathogenesis, the role of eosinophils has been questioned and might be less important than previously thought. It is well known now that EoE is a Th2-mediated disease with many more disease features than eosinophilic infiltration. With more knowledge on EoE, less pronounced phenotypes or nuances of the disease have become apparent. In fact, EoE might be only the tip of the iceberg (and the most extreme phenotype) with several variant forms, at least three, lying on a disease spectrum. Although a common (food induced) pathogenesis has yet to be confirmed, gastroenterologists and allergologists should be aware of these new phenomena in order to further characterize these patients. In the following review, we discuss the pathogenesis of EoE, particularly those mechanisms beyond eosinophilic infiltration of the esophageal mucosa, non-eosinophilic inflammatory cell populations, the new disease entity EoE-like disease, variant forms of EoE, and the recently coined term mast cell esophagitis.
Collapse
Affiliation(s)
| | - Alex Straumann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Luis Salvador Nunes
- Division of Gastroenterology and Hepatology, University Hospital Lausanne – CHUV, Lausanne, Switzerland
| | - Alain M. Schoepfer
- Division of Gastroenterology and Hepatology, University Hospital Lausanne – CHUV, Lausanne, Switzerland
| | - Thomas Greuter
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
- Division of Gastroenterology and Hepatology, University Hospital Lausanne – CHUV, Lausanne, Switzerland
- Department of Internal Medicine, GZO – Zurich Regional Health Center, Wetzikon, Switzerland
| |
Collapse
|
12
|
Ogulur I, Pat Y, Aydin T, Yazici D, Rückert B, Peng Y, Kim J, Radzikowska U, Westermann P, Sokolowska M, Dhir R, Akdis M, Nadeau K, Akdis CA. Gut epithelial barrier damage caused by dishwasher detergents and rinse aids. J Allergy Clin Immunol 2023; 151:469-484. [PMID: 36464527 DOI: 10.1016/j.jaci.2022.10.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND The increased prevalence of many chronic inflammatory diseases linked to gut epithelial barrier leakiness has prompted us to investigate the role of extensive use of dishwasher detergents, among other factors. OBJECTIVE We sought to investigate the effects of professional and household dishwashers, and rinse agents, on cytotoxicity, barrier function, transcriptome, and protein expression in gastrointestinal epithelial cells. METHODS Enterocytic liquid-liquid interfaces were established on permeable supports, and direct cellular cytotoxicity, transepithelial electrical resistance, paracellular flux, immunofluorescence staining, RNA-sequencing transcriptome, and targeted proteomics were performed. RESULTS The observed detergent toxicity was attributed to exposure to rinse aid in a dose-dependent manner up to 1:20,000 v/v dilution. A disrupted epithelial barrier, particularly by rinse aid, was observed in liquid-liquid interface cultures, organoids, and gut-on-a-chip, demonstrating decreased transepithelial electrical resistance, increased paracellular flux, and irregular and heterogeneous tight junction immunostaining. When individual components of the rinse aid were investigated separately, alcohol ethoxylates elicited a strong toxic and barrier-damaging effect. RNA-sequencing transcriptome and proteomics data revealed upregulation in cell death, signaling and communication, development, metabolism, proliferation, and immune and inflammatory responses of epithelial cells. Interestingly, detergent residue from professional dishwashers demonstrated the remnant of a significant amount of cytotoxic and epithelial barrier-damaging rinse aid remaining on washed and ready-to-use dishware. CONCLUSIONS The expression of genes involved in cell survival, epithelial barrier, cytokine signaling, and metabolism was altered by rinse aid in concentrations used in professional dishwashers. The alcohol ethoxylates present in the rinse aid were identified as the culprit component causing the epithelial inflammation and barrier damage.
Collapse
Affiliation(s)
- Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Department of Medical Microbiology, Faculty of Medicine, Aydin Adnan Menderes University, Aydin
| | - Tamer Aydin
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Beate Rückert
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yaqi Peng
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Juno Kim
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Patrick Westermann
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | | | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Kari Nadeau
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| |
Collapse
|
13
|
Avlas S, Shani G, Rhone N, Itan M, Dolitzky A, Hazut I, Tal SG, Gordon Y, Shoda T, Ballaban A, Baruch NMB, Rochman M, Diesendruck Y, Nahary L, Bitton A, Halpern Z, Benhar I, Varol C, Rothenberg ME, Munitz A. Epithelial cell-expressed type II IL-4 receptor mediates eosinophilic esophagitis. Allergy 2023; 78:464-476. [PMID: 36070083 PMCID: PMC9892241 DOI: 10.1111/all.15510] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 07/28/2022] [Accepted: 08/17/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) is a chronic, food-driven allergic disease, characterized by eosinophil-rich inflammation in the esophagus. The histopathological and clinical features of EoE have been attributed to overproduction of the type 2 cytokines IL-4 and IL-13, which mediate profound alterations in the esophageal epithelium and neutralizing of their shared receptor component (IL-4Rα) with a human antibody drug (dupilumab) demonstrates clinical efficacy. Yet, the relative contribution of IL-4 and IL-13 and whether the type II IL-4 receptor (comprised of the IL-4Rα chain in association with IL-13Rα1) mediates this effect has not been determined. METHODS Experimental EoE was induced in WT, Il13ra1-/- , and Krt14Cre /Il13ra1fl/fl mice by skin-sensitized using 4-ethoxymethylene-2-phenyl-2-oxazolin (OXA) followed by intraesophageal challenges. Esophageal histopathology was determined histologically. RNA was extracted and sequenced for transcriptome analysis and compared with human EoE RNAseq data. RESULTS Induction of experimental EoE in mice lacking Il13ra1 and in vivo IL-13 antibody-based neutralization experiments blocked antigen-induced esophageal epithelial and lamina propria thickening, basal cell proliferation, eosinophilia, and tissue remodeling. In vivo targeted deletion of Il13ra1 in esophageal epithelial cells rendered mice protected from experimental EoE. Single-cell RNA sequencing analysis of human EoE biopsies revealed predominant expression of IL-13Rα1 in epithelial cells and that EoE signature genes correlated with IL-13 expression compared with IL-4. CONCLUSIONS We demonstrate a definitive role for IL-13 signaling via IL-13Rα1 in EoE. These data provide mechanistic insights into the mode of action of current therapies in EoE and highlight the type II IL-4R as a future therapeutic target.
Collapse
Affiliation(s)
- Shmulik Avlas
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Guy Shani
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Natalie Rhone
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal Itan
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Avishay Dolitzky
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Inbal Hazut
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sharon Grisaru- Tal
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yaara Gordon
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tetsuo Shoda
- Division of Allergy/Immunology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Adina Ballaban
- Division of Allergy/Immunology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Netali Morgenstern Ben- Baruch
- Division of Allergy/Immunology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Mark Rochman
- Division of Allergy/Immunology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Yael Diesendruck
- The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Limor Nahary
- The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Almog Bitton
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel,The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Zamir Halpern
- Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center, Tel Aviv, Israel
| | - Itai Benhar
- The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Chen Varol
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel,Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center, Tel Aviv, Israel
| | - Marc E. Rothenberg
- Division of Allergy/Immunology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Ariel Munitz
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
14
|
Ilia S, Monica V, Daniel S. Mean nocturnal baseline impedance and endoscopic mucosal impedance measurements in patients with eosinophilic esophagitis: a new tool for follow up and management? Updates Surg 2023; 75:389-393. [PMID: 35840790 PMCID: PMC9852170 DOI: 10.1007/s13304-022-01331-4] [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: 06/25/2022] [Accepted: 07/05/2022] [Indexed: 01/24/2023]
Abstract
Eosinophilic esophagitis (EoE) is the second most common cause of chronic esophageal inflammation after GERD, with increasing incidence and prevalence across all age groups. Since current diagnosis and follow up of EoE require endoscopy with biopsies, there is an increased interest in non or less invasive tests for its diagnosis and follow up. Baseline mucosal impedance measurement allows evaluation of mucosal barrier properties and is widely accepted as an adjunct method in GERD diagnosis. As EoE is associated with increased mucosal permeability, the use of baseline impedance to evaluate mucosal integrity has been investigated in several studies. It was found that baseline mucosal impedance, measured either during 24 h reflux monitoring or during endoscopy, was significantly lower in all parts of the esophagus in EoE patients. Impedance measurement correlated with eosinophil counts on biopsies, offering a tool to monitor treatment response. Additionally, baseline impedance patterns differed between those responding to proton pump inhibitor (PPI) treatment and those resistant to PPI, potentially allowing to tailor future treatment to the individual patient. In summary, baseline impedance measurement offers a potential tool for diagnosis and follow up in EoE. Its exact place in EoE treatment is yet to be determined and requires further future studies.
Collapse
Affiliation(s)
- Sergeev Ilia
- Barts and the London School of Medicine and Dentistry, Wingate Institute of Neurogastroenterology, Queen Mary University of London, London, UK
| | - Velosa Monica
- Barts and the London School of Medicine and Dentistry, Wingate Institute of Neurogastroenterology, Queen Mary University of London, London, UK
| | - Sifrim Daniel
- Barts and the London School of Medicine and Dentistry, Wingate Institute of Neurogastroenterology, Queen Mary University of London, London, UK
| |
Collapse
|
15
|
Wagner AS, Vogel AK, Lumsdaine SW, Phillips EK, Willems HME, Peters BM, Reynolds TB. Mucosal Infection with Unmasked Candida albicans Cells Impacts Disease Progression in a Host Niche-Specific Manner. Infect Immun 2022; 90:e0034222. [PMID: 36374100 PMCID: PMC9753624 DOI: 10.1128/iai.00342-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/26/2022] [Indexed: 11/16/2022] Open
Abstract
Shielding the immunogenic cell wall epitope β(1, 3)-glucan under an outer layer of mannosylated glycoproteins is an essential virulence factor deployed by Candida albicans during systemic infection. Accordingly, mutants with increased β(1, 3)-glucan exposure (unmasking) display increased immunostimulatory capabilities in vitro and attenuated virulence during systemic infection in mice. However, little work has been done to assess the impact of increased unmasking during the two most common manifestations of candidiasis, namely, oropharyngeal candidiasis (OPC) and vulvovaginal candidiasis (VVC). We have shown previously that the expression of a single hyperactive allele of the MAP3K STE11ΔN467 induces unmasking via the Cek1 MAPK pathway, attenuates fungal burden, and prolongs survival during systemic infection in mice. Here, we expand on these findings and show that infection with an unmasked STE11ΔN467 mutant also impacts disease progression during OPC and VVC murine infection models. Male mice sublingually infected with the STE11ΔN467 mutant showed a significant reduction in tongue fungal burden at 2 days postinfection and a modest reduction at 5 days postinfection. However, we find that selection for STE11ΔN467 suppressor mutants that no longer display increased unmasking occurs within the oral cavity and is likely responsible for the restoration of fungal burden trends to wild-type levels later in the infection. In the VVC infection model, no attenuation in fungal burden was observed. However, polymorphonuclear cell recruitment and interleukin-1β (IL-1β) levels within the vaginal lumen, markers of immunopathogenesis, were increased in mice infected with unmasked STE11ΔN467 cells. Thus, our data suggest a niche-specific impact for unmasking on disease progression.
Collapse
Affiliation(s)
- Andrew S. Wagner
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Amanda K. Vogel
- Integrated Program in Biomedical Sciences, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | | | - Elise K. Phillips
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Hubertine M. E. Willems
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Brian M. Peters
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Todd B. Reynolds
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW This review will present what is known from recent research on the involvement of mast cells in eosinophilic esophagitis and identify questions requiring further investigation. RECENT FINDINGS In the adults and children with eosinophilic esophagitis, there is increasing evidence that mastocytosis can persist, despite resolution of eosinophilia and is associated with persistent mucosal abnormalities and symptoms. Despite, treatment mast cells have an activated transcriptome. Mast cells likely contribute to epithelial barrier dysfunction, smooth muscle hypertrophy and contraction, and subepithelial fibrosis. It remains unclear whether targeting MCs alone has therapeutic efficacy to improve tissue damage. SUMMARY Mast cells appear to play a key role in eosinophilic esophagitis and serve as a biomarker of mucosal healing in conjunction with eosinophils. Excessive mast cell activation likely contributes to tissue damage in eosinophilic esophagitis and need to be considered as a target of therapy along with eosinophils.
Collapse
|
17
|
Celebi Sozener Z, Özbey Yücel Ü, Altiner S, Ozdel Oztürk B, Cerci P, Türk M, Gorgülü Akin B, Akdis M, Yilmaz I, Ozdemir C, Mungan D, Akdis CA. The External Exposome and Allergies: From the Perspective of the Epithelial Barrier Hypothesis. FRONTIERS IN ALLERGY 2022; 3:887672. [PMID: 35873598 PMCID: PMC9304993 DOI: 10.3389/falgy.2022.887672] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/29/2022] [Indexed: 01/04/2023] Open
Abstract
Introduction In the last decades, we have seen a rapid increase in the prevalence of allergic diseases such as asthma, allergic rhinitis, atopic dermatitis, and food allergies. The environmental changes caused by industrialization, urbanization and modernization, including dramatic increases in air pollutants such as particulate matter (PM), diesel exhaust, nitrogen dioxide (NO2), ozone (O3), alarming effects of global warming, change and loss of biodiversity, affect both human health and the entire ecosystem. Objective In this review, we aimed to discuss the effects of the external exposome on epithelial barriers and its relationship with the development of allergic diseases by considering the changes in all stakeholders of the outer exposome together, in the light of the recently proposed epithelial barrier hypothesis. Method To reach current, prominent, and comprehensive studies on the subject, PubMed databases were searched. We included the more resounding articles with reliable and strong results. Results Exposure to altered environmental factors such as increased pollution, microplastics, nanoparticles, tobacco smoke, food emulsifiers, detergents, and household cleaners, and climate change, loss and change in microbial biodiversity, modifications in the consumption of dietary fatty acids, the use of emulsifiers, preservatives and the decrease in the antioxidant content of the widely consumed western diet may disrupt the epithelial barriers of the skin, respiratory and gastrointestinal tracts, making us more vulnerable to exogeneous allergens and microbes. Epithelial cell activation, microbial dysbiosis and bacterial translocation disrupt the immune balance and a chronic Th2 inflammation ensues. Conclusion Dramatic increases in air pollution, worrisome effects of global warming, dysbiosis, changing dietary habits and the complex interactions of all these factors affect the epithelial barriers and local and systemic inflammation. We want to draw attention to the emerging health effects of environmental changes and to motivate the public to influence government policies for the well-being of humans and the nature of the earth and the well-being of future generations.
Collapse
Affiliation(s)
- Zeynep Celebi Sozener
- Clinic of Immunology and Allergic Diseases, Ankara City Hospital, Ankara, Turkey
- *Correspondence: Zeynep Celebi Sozener ; orcid.org/0000-0003-4188-0959
| | - Ümüs Özbey Yücel
- Department of Nutrition and Diet, Ankara University, Ankara, Turkey
| | - Seda Altiner
- Division of Immunology and Allergic Diseases, Department of Internal Medicine, School of Medicine, Ankara University, Ankara, Turkey
| | - Betül Ozdel Oztürk
- Division of Immunology and Allergic Diseases, Department of Chest Diseases, School of Medicine, Ankara University, Ankara, Turkey
| | - Pamir Cerci
- Clinic of Immunology and Allergic Diseases, Eskisehir City Hospital, Eskisehir, Turkey
| | - Murat Türk
- Clinic of Immunology and Allergic Diseases, Kayseri City Hospital, Kayseri, Turkey
| | - Begüm Gorgülü Akin
- Clinic of Immunology and Allergic Diseases, Ankara City Hospital, Ankara, Turkey
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Insu Yilmaz
- Division of Immunology and Allergic Diseases, Department of Chest Diseases, Erciyes University, Kayseri, Turkey
| | - Cevdet Ozdemir
- Department of Pediatric Basic Sciences, Institute of Child Health, Istanbul University, Istanbul, Turkey
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Dilsad Mungan
- Division of Immunology and Allergic Diseases, Department of Chest Diseases, School of Medicine, Ankara University, Ankara, Turkey
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
- Cezmi A. Akdis ; orcid.org/0000-0001-8020-019X
| |
Collapse
|
18
|
Zhou S, Liu S, Tian G, Zhao L, Wang H, Li Y, Shen Y, Han L. KLK5 is associated with the radioresistance, aggression, and progression of cervical cancer. Gynecol Oncol 2022; 166:138-147. [PMID: 35595569 DOI: 10.1016/j.ygyno.2022.05.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The role of kallikrein-related peptidase 5 (KLK5) has been studied in several diseases, including skin and ovarian cancers. However, its role in cervical cancer remains unclear, particularly in regulating the radiation resistance and growth of cervical cancer cells. Radiation resistance of cervical cancer is associated with local recurrence, distant metastasis, and reduced survival. METHODS We first analyzed radiotherapy-naive samples and relevant clinical data from patients with cervical cancer who received radiotherapy without surgery or other antitumor treatment from 2014 to 2016. Subsequently, biopsied tissues, in vitro cells, and transplanted tumors in nude mice were investigated. RESULTS Gene sequencing and clinical data analysis showed that KLK5 overexpression was associated with a poor prognosis post-radiotherapy. In in vitro cell and tumor transplantation experiments, KLK5 overexpression significantly increased radiation resistance. However, downregulating KLK5 expression increased radiosensitivity. CONCLUSION Our results confirm that KLK5 is vital to the radioresistance of cervical cancer, and provide a new target and marker for the treatment of radioresistance in cervical cancer.
Collapse
Affiliation(s)
- Shunqing Zhou
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130022, China.
| | - Shuyan Liu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130022, China.
| | - Geng Tian
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130022, China.
| | - Lin Zhao
- Department of Obstetrics and Gynecology, The People's Hospital of LIAONING PROVINCE, Shenyang 110000, China
| | - Haichen Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China.
| | - Ying Li
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130022, China
| | - Yannan Shen
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China.
| | - Liying Han
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130022, China.
| |
Collapse
|
19
|
Xia Y, Cao H, Zheng J, Chen L. Claudin-1 Mediated Tight Junction Dysfunction as a Contributor to Atopic March. Front Immunol 2022; 13:927465. [PMID: 35844593 PMCID: PMC9277052 DOI: 10.3389/fimmu.2022.927465] [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: 05/03/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022] Open
Abstract
Atopic march refers to the phenomenon wherein the occurrence of asthma and food allergy tends to increase after atopic dermatitis. The mechanism underlying the progression of allergic inflammation from the skin to gastrointestinal (GI) tract and airways has still remained elusive. Impaired skin barrier was proposed as a risk factor for allergic sensitization. Claudin-1 protein forms tight junctions and is highly expressed in the epithelium of the skin, airways, and GI tract, thus, the downregulation of claudin-1 expression level caused by CLDN-1 gene polymorphism can mediate common dysregulation of epithelial barrier function in these organs, potentially leading to allergic sensitization at various sites. Importantly, in patients with atopic dermatitis, asthma, and food allergy, claudin-1 expression level was significantly downregulated in the skin, bronchial and intestinal epithelium, respectively. Knockdown of claudin-1 expression level in mouse models of atopic dermatitis and allergic asthma exacerbated allergic inflammation, proving that downregulation of claudin-1 expression level contributes to the pathogenesis of allergic diseases. Therefore, we hypothesized that the tight junction dysfunction mediated by downregulation of claudin-1 expression level contributes to atopic march. Further validation with clinical data from patients with atopic march or mouse models of atopic march is needed. If this hypothesis can be fully confirmed, impaired claudin-1 expression level may be a risk factor and likely a diagnostic marker for atopic march. Claudin-1 may serve as a valuable target to slowdown or block the progression of atopic march.
Collapse
|
20
|
Kaymak T, Kaya B, Wuggenig P, Nuciforo S, Göldi A, Oswald F, Roux J, Noti M, Melhem H, Hruz P, Niess JH. IL-20 subfamily cytokines impair the oesophageal epithelial barrier by diminishing filaggrin in eosinophilic oesophagitis. Gut 2022; 72:821-833. [PMID: 35613844 PMCID: PMC10086458 DOI: 10.1136/gutjnl-2022-327166] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/02/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Disruption of the epithelial barrier plays an essential role in developing eosinophilic oesophagitis (EoE), a disease defined by type 2 helper T cell (Th2)-mediated food-associated and aeroallergen-associated chronic inflammation. Although an increased expression of interleukin (IL)-20 subfamily members, IL-19, IL-20 and IL-24, in Th2-mediated diseases has been reported, their function in EoE remains unknown. DESIGN Combining transcriptomic, proteomic and functional analyses, we studied the importance of the IL-20 subfamily for EoE using patient-derived oesophageal three-dimensional models and an EoE mouse model. RESULTS Patients with active EoE have increased expression of IL-20 subfamily cytokines in the oesophagus and serum. In patient-derived oesophageal organoids stimulated with IL-20 cytokines, RNA sequencing and mass spectrometry revealed a downregulation of genes and proteins forming the cornified envelope, including filaggrins. On the contrary, abrogation of IL-20 subfamily signalling in Il20R2 -/- animals resulted in attenuated experimental EoE reflected by reduced eosinophil infiltration, lower Th2 cytokine expression and preserved expression of filaggrins in the oesophagus. Mechanistically, these observations were mediated by the mitogen-activated protein kinase (MAPK); extracellular-signal regulated kinases (ERK)1/2) pathway. Its blockade prevented epithelial barrier impairment in patient-derived air-liquid interface cultures stimulated with IL-20 cytokines and attenuated experimental EoE in mice. CONCLUSION Our findings reveal a previously unknown regulatory role of the IL-20 subfamily for oesophageal barrier function in the context of EoE. We propose that aberrant IL-20 subfamily signalling disturbs the oesophageal epithelial barrier integrity and promotes EoE development. Our study suggests that specific targeting of the IL-20 subfamily signalling pathway may present a novel strategy for the treatment of EoE.
Collapse
Affiliation(s)
- Tanay Kaymak
- Department of Biomedicine, Gastroenterology, University of Basel, Basel, Switzerland
| | - Berna Kaya
- Department of Biomedicine, Gastroenterology, University of Basel, Basel, Switzerland
| | - Philipp Wuggenig
- Department of Biomedicine, Gastroenterology, University of Basel, Basel, Switzerland
| | - Sandro Nuciforo
- Department of Biomedicine, Hepatology, University of Basel, Basel, Switzerland
| | - Andreas Göldi
- Department of Gastroenterology, Clarunis - University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | | | - Franz Oswald
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Julien Roux
- Department of Biomedicine, Gastroenterology, University of Basel, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Mario Noti
- Institute of Pathology, University of Bern, Bern, Switzerland, Current address: Nestlé SA, Nestlé Research, Nestlé Institute of Health Sciences, Department of Gastrointestinal Health Immunology, Vers-Chez-les-Blancs, Lausanne, Switzerland
| | - Hassan Melhem
- Department of Biomedicine, Gastroenterology, University of Basel, Basel, Switzerland
| | - Petr Hruz
- Department of Gastroenterology, Clarunis - University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Jan Hendrik Niess
- Department of Biomedicine, Gastroenterology, University of Basel, Basel, Switzerland .,Department of Gastroenterology, Clarunis - University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland.,Department of Clinical Research, University of Basel, Basel, Switzerland
| |
Collapse
|
21
|
Kleuskens MT, Haasnoot ML, Herpers BM, Ampting MTJV, Bredenoord AJ, Garssen J, Redegeld FA, van Esch BC. Butyrate and propionate restore interleukin 13-compromised esophageal epithelial barrier function. Allergy 2022; 77:1510-1521. [PMID: 34458999 PMCID: PMC9293003 DOI: 10.1111/all.15069] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 08/15/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) is a food allergen driven disease that is accompanied by interleukin (IL) 13 overexpression and esophageal barrier dysfunction allowing transepithelial food allergen permeation. Nutraceuticals, such as short-chain fatty acids (SCFAs) that restore barrier function and increase immune fitness may be a promising tool in the management of EoE. Here, we investigated the effects of the SCFAs acetate, propionate, and butyrate on an IL-13-compromised human esophageal epithelial barrier, including the mechanisms involved. METHODS An air-liquid interface culture model of differentiated human EPC2-hTERT (EPC2) was used to study whether SCFAs could restore barrier function after IL-13-induced impairment. Esophageal epithelial barrier function was monitored by transepithelial electrical resistance (TEER) and FITC-dextran paracellular flux, and was further examined by qPCR and immunohistochemical analysis. G protein-coupled receptor (GPR) GPR41, GPR43, GPR109a, or histone deacetylase (HDAC) (ant)agonists were used to assess mechanisms of action of SCFAs. RESULTS IL-13 stimulation decreased TEER and increased FITC flux, which was counteracted by butyrate and propionate, but not acetate treatment. Barrier proteins FLG and DSG1 mRNA expression was upregulated following butyrate and propionate treatment, whereas expression of eosinophil chemoattractant CCL26 and protease CAPN14 was downregulated. Similarly, butyrate and propionate restored FLG and DSG1 protein expression. Similar effects were observed with an HDAC antagonist but not with GPR agonists. CONCLUSION Nutraceuticals butyrate and propionate restore the barrier function of esophageal epithelial cells after an inflammatory insult and may be of therapeutic benefit in the management of EoE.
Collapse
Affiliation(s)
- Mirelle T.A. Kleuskens
- Division of Pharmacology Utrecht Institute for Pharmaceutical Sciences Faculty of Science Utrecht University Utrecht The Netherlands
| | - Maria L. Haasnoot
- Department of Gastroenterology & Hepatology Amsterdam UMC, location AMC Amsterdam The Netherlands
| | - Bart M. Herpers
- Division of Pharmacology Utrecht Institute for Pharmaceutical Sciences Faculty of Science Utrecht University Utrecht The Netherlands
| | | | - Albert J. Bredenoord
- Department of Gastroenterology & Hepatology Amsterdam UMC, location AMC Amsterdam The Netherlands
| | - Johan Garssen
- Division of Pharmacology Utrecht Institute for Pharmaceutical Sciences Faculty of Science Utrecht University Utrecht The Netherlands
- Danone Nutricia Research Utrecht The Netherlands
| | - Frank A. Redegeld
- Division of Pharmacology Utrecht Institute for Pharmaceutical Sciences Faculty of Science Utrecht University Utrecht The Netherlands
| | - Betty C.A.M. van Esch
- Division of Pharmacology Utrecht Institute for Pharmaceutical Sciences Faculty of Science Utrecht University Utrecht The Netherlands
- Danone Nutricia Research Utrecht The Netherlands
| |
Collapse
|
22
|
Celebi Sozener Z, Ozdel Ozturk B, Cerci P, Turk M, Gorgulu Akin B, Akdis M, Altiner S, Ozbey U, Ogulur I, Mitamura Y, Yilmaz I, Nadeau K, Ozdemir C, Mungan D, Akdis CA. Epithelial barrier hypothesis: Effect of the external exposome on the microbiome and epithelial barriers in allergic disease. Allergy 2022; 77:1418-1449. [PMID: 35108405 PMCID: PMC9306534 DOI: 10.1111/all.15240] [Citation(s) in RCA: 174] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/25/2022] [Accepted: 01/29/2022] [Indexed: 12/11/2022]
Abstract
Environmental exposure plays a major role in the development of allergic diseases. The exposome can be classified into internal (e.g., aging, hormones, and metabolic processes), specific external (e.g., chemical pollutants or lifestyle factors), and general external (e.g., broader socioeconomic and psychological contexts) domains, all of which are interrelated. All the factors we are exposed to, from the moment of conception to death, are part of the external exposome. Several hundreds of thousands of new chemicals have been introduced in modern life without our having a full understanding of their toxic health effects and ways to mitigate these effects. Climate change, air pollution, microplastics, tobacco smoke, changes and loss of biodiversity, alterations in dietary habits, and the microbiome due to modernization, urbanization, and globalization constitute our surrounding environment and external exposome. Some of these factors disrupt the epithelial barriers of the skin and mucosal surfaces, and these disruptions have been linked in the last few decades to the increasing prevalence and severity of allergic and inflammatory diseases such as atopic dermatitis, food allergy, allergic rhinitis, chronic rhinosinusitis, eosinophilic esophagitis, and asthma. The epithelial barrier hypothesis provides a mechanistic explanation of how these factors can explain the rapid increase in allergic and autoimmune diseases. In this review, we discuss factors affecting the planet's health in the context of the 'epithelial barrier hypothesis,' including climate change, pollution, changes and loss of biodiversity, and emphasize the changes in the external exposome in the last few decades and their effects on allergic diseases. In addition, the roles of increased dietary fatty acid consumption and environmental substances (detergents, airborne pollen, ozone, microplastics, nanoparticles, and tobacco) affecting epithelial barriers are discussed. Considering the emerging data from recent studies, we suggest stringent governmental regulations, global policy adjustments, patient education, and the establishment of individualized control measures to mitigate environmental threats and decrease allergic disease.
Collapse
Affiliation(s)
| | - Betul Ozdel Ozturk
- School of MedicineDepartment of Chest DiseasesDivision of Immunology and Allergic DiseasesAnkara UniversityAnkaraTurkey
| | - Pamir Cerci
- Clinic of Immunology and Allergic DiseasesEskisehir City HospitalEskisehirTurkey
| | - Murat Turk
- Clinic of Immunology and Allergic DiseasesKayseri City HospitalKayseriTurkey
| | - Begum Gorgulu Akin
- Clinic of Immunology and Allergic DiseasesAnkara City HospitalAnkaraTurkey
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
| | - Seda Altiner
- Clinic of Internal Medicine Division of Immunology and Allergic DiseasesKahramanmaras Necip Fazil City HospitalKahramanmarasTurkey
| | - Umus Ozbey
- Department of Nutrition and DietAnkara UniversityAnkaraTurkey
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
| | - Insu Yilmaz
- Department of Chest DiseasesDivision of Immunology and Allergic DiseasesErciyes UniversityKayseriTurkey
| | - Kari Nadeau
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University School of MedicineDivision of Pulmonary and Critical Care MedicineDepartment of MedicineStanford UniversityStanfordCaliforniaUSA
| | - Cevdet Ozdemir
- Institute of Child HealthDepartment of Pediatric Basic SciencesIstanbul UniversityIstanbulTurkey
- Istanbul Faculty of MedicineDepartment of PediatricsDivision of Pediatric Allergy and ImmunologyIstanbul UniversityIstanbulTurkey
| | - Dilsad Mungan
- School of MedicineDepartment of Chest DiseasesDivision of Immunology and Allergic DiseasesAnkara UniversityAnkaraTurkey
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
- Christine Kühne‐Center for Allergy Research and Education (CK‐CARE)DavosSwitzerland
| |
Collapse
|
23
|
Greuter T, Straumann A, Fernandez‐Marrero Y, Germic N, Hosseini A, Yousefi S, Simon D, Collins MH, Bussmann C, Chehade M, Dellon ES, Furuta GT, Gonsalves N, Hirano I, Moawad FJ, Biedermann L, Safroneeva E, Schoepfer AM, Simon H. Characterization of eosinophilic esophagitis variants by clinical, histological, and molecular analyses: A cross-sectional multi-center study. Allergy 2022; 77:2520-2533. [PMID: 35094416 PMCID: PMC9545458 DOI: 10.1111/all.15233] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/29/2021] [Accepted: 12/31/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Physicians are increasingly confronted with patients presenting with symptoms of esophageal dysfunction resembling eosinophilic esophagitis (EoE), but absence of significant esophageal eosinophilia. The purpose of this study was to characterize and classify this group of EoE variants. DESIGN Patients from six EoE-centers with symptoms of esophageal dysfunction, but peak eosinophil counts of <60/mm2 (<15/hpf) in esophageal biopsies and absence of gastro-esophageal reflux disease (GERD) were included. Clinical, endoscopic, (immuno)-histological, and molecular features were determined and compared with EoE, GERD, and healthy controls. RESULTS We included 69 patients with EoE variants. Endoscopic abnormalities were found in 53.6%. We identified three histological subtypes: EoE-like esophagitis (36/69, 52.2%), lymphocytic esophagitis (14/69, 20.3%), and non-specific esophagitis (19/69, 27.5%). Immunohistochemistry revealed-in contrast to EoE-no significant increase in inflammatory cell infiltrates compared with GERD and healthy controls, except for lymphocytes in lymphocytic esophagitis. EoE-typical Th2-response was absent in all EoE variants. However, considerable structural changes were detected based on histology and protein expression. Using next generation mRNA sequencing, we found the three EoE variants to have distinct molecular fingerprints partially sharing pronounced traits of EoE. Hierarchical sample clustering of RNA sequencing data confirmed the presence of an EoE-like (characterized by eotaxin-3 expression), non-specific, and lymphocytic variant cluster (characterized by CD3 cells and TSLP expression). CONCLUSION All EoE variants are clinically and histologically active conditions despite the absence of esophageal eosinophilia. EoE variants appear to be part of a disease spectrum, where classical EoE represents the most common and apparent phenotype.
Collapse
Affiliation(s)
- Thomas Greuter
- Department of Gastroenterology and Hepatology University Hospital Zurich Zurich Switzerland
- Division of Gastroenterology and Hepatology University Hospital Lausanne – Centre Hopitalier Universitaire Vaudois (CHUV) Lausanne Switzerland
| | - Alex Straumann
- Department of Gastroenterology and Hepatology University Hospital Zurich Zurich Switzerland
| | | | - Nina Germic
- Institute of Pharmacology University of Bern Bern Switzerland
| | - Aref Hosseini
- Institute of Pharmacology University of Bern Bern Switzerland
| | - Shida Yousefi
- Institute of Pharmacology University of Bern Bern Switzerland
| | - Dagmar Simon
- Department of Dermatology, Inselspital Bern University Hospital University of Bern Bern Switzerland
| | - Margaret H. Collins
- Division of Pathology Cincinnati Children`s Hospital Medical Center Cincinnati Ohio USA
| | | | - Mirna Chehade
- Mount Sinai Center for Eosinophilic Disorders Icahn School of Medicine at Mount Sinai New York New York USA
| | - Evan S. Dellon
- Division of Gastroenterology and Hepatology UNC Hospital Chapel Hill North Carolina USA
| | - Glenn T. Furuta
- Department of Pediatrics Gastrointestinal Eosinophilic Diseases Program Digestive Health Institute Children’s Hospital Colorado University of Colorado School of Medicine Aurora Colorado USA
| | - Nirmala Gonsalves
- Division of Gastroenterology and Hepatology Northwestern University Chicago Illinois USA
| | - Ikuo Hirano
- Division of Gastroenterology and Hepatology Northwestern University Chicago Illinois USA
| | - Fouad J. Moawad
- Division of Gastroenterology Scripps Clinic La Jolla California USA
| | - Luc Biedermann
- Department of Gastroenterology and Hepatology University Hospital Zurich Zurich Switzerland
| | | | - Alain M. Schoepfer
- Division of Gastroenterology and Hepatology University Hospital Lausanne – Centre Hopitalier Universitaire Vaudois (CHUV) Lausanne Switzerland
| | - Hans‐Uwe Simon
- Institute of Pharmacology University of Bern Bern Switzerland
- Institute of Biochemistry Brandenburg Medical School Neuruppin Germany
- Department of Clinical Immunology and Allergology Sechenov University Moscow Russia
- Laboratory of Molecular Immunology Institute of Fundamental Medicine and Biology Kazan Federal University Kazan Russia
| |
Collapse
|
24
|
Zhernov YV, Vysochanskaya SO, Sukhov VA, Zaostrovtseva OK, Gorshenin DS, Sidorova EA, Mitrokhin OV. Molecular Mechanisms of Eosinophilic Esophagitis. Int J Mol Sci 2021; 22:ijms222413183. [PMID: 34947981 PMCID: PMC8703627 DOI: 10.3390/ijms222413183] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/04/2021] [Accepted: 12/05/2021] [Indexed: 12/19/2022] Open
Abstract
Food hypersensitivity is a group of diseases arising from a specific immune response that reproduces on exposure to a given food. The current understanding of molecular mechanisms and immunopathology of non-IgE-mediated/mixed food hypersensitivity, e.g., eosinophilic esophagitis, contains many gaps in knowledge. This review aims to provide a modern classification and identify the primary diseases of non-IgE-mediated/mixed food hypersensitivity reactions, delineate the distinctive molecular features, and discuss recent findings in the immunopathology of eosinophilic esophagitis that may become a basis to develop valid biomarkers and novel therapies for this disease. Eosinophilic esophagitis is a recently recognized allergic-mediated disease with eosinophil-predominant esophagus inflammation. Its pathogenesis is a complicated network of interactions and signaling between epithelial, mesenchymal, and immune cells on molecular and intercellular levels. Alterations produced by overactivation of some cytokine signaling pathways, e.g., IL-13 or thymic stromal lymphopoietin (TSLP), were evolved and observed in this review from the viewpoints of molecular, genetic, epigenetic, and transcriptomic changes. Despite substantial experimental data, the reliable and representative mechanism of eosinophilic esophagitis pathogenesis has yet to show itself. So, the place of esophagitis between mixed and non-IgE-mediated allergic disorders and between eosinophilic gastrointestinal disorders currently seems vague and unclear.
Collapse
Affiliation(s)
- Yury V. Zhernov
- Department of General Hygiene, F. Erismann Institute of Public Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (S.O.V.); (V.A.S.); (O.K.Z.); (D.S.G.); (E.A.S.); (O.V.M.)
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
- Correspondence: ; Tel.: +7-(915)-1552000
| | - Sonya O. Vysochanskaya
- Department of General Hygiene, F. Erismann Institute of Public Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (S.O.V.); (V.A.S.); (O.K.Z.); (D.S.G.); (E.A.S.); (O.V.M.)
| | - Vitaly A. Sukhov
- Department of General Hygiene, F. Erismann Institute of Public Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (S.O.V.); (V.A.S.); (O.K.Z.); (D.S.G.); (E.A.S.); (O.V.M.)
| | - Olga K. Zaostrovtseva
- Department of General Hygiene, F. Erismann Institute of Public Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (S.O.V.); (V.A.S.); (O.K.Z.); (D.S.G.); (E.A.S.); (O.V.M.)
| | - Denis S. Gorshenin
- Department of General Hygiene, F. Erismann Institute of Public Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (S.O.V.); (V.A.S.); (O.K.Z.); (D.S.G.); (E.A.S.); (O.V.M.)
| | - Ekaterina A. Sidorova
- Department of General Hygiene, F. Erismann Institute of Public Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (S.O.V.); (V.A.S.); (O.K.Z.); (D.S.G.); (E.A.S.); (O.V.M.)
| | - Oleg V. Mitrokhin
- Department of General Hygiene, F. Erismann Institute of Public Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (S.O.V.); (V.A.S.); (O.K.Z.); (D.S.G.); (E.A.S.); (O.V.M.)
| |
Collapse
|
25
|
Does the epithelial barrier hypothesis explain the increase in allergy, autoimmunity and other chronic conditions? Nat Rev Immunol 2021; 21:739-751. [PMID: 33846604 DOI: 10.1038/s41577-021-00538-7] [Citation(s) in RCA: 486] [Impact Index Per Article: 121.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2021] [Indexed: 02/07/2023]
Abstract
There has been a steep increase in allergic and autoimmune diseases, reaching epidemic proportions and now affecting more than one billion people worldwide. These diseases are more common in industrialized countries, and their prevalence continues to rise in developing countries in parallel to urbanization and industrialization. Intact skin and mucosal barriers are crucial for the maintenance of tissue homeostasis as they protect host tissues from infections, environmental toxins, pollutants and allergens. A defective epithelial barrier has been demonstrated in allergic and autoimmune conditions such as asthma, atopic dermatitis, allergic rhinitis, chronic rhinosinusitis, eosinophilic esophagitis, coeliac disease and inflammatory bowel disease. In addition, leakiness of the gut epithelium is also implicated in systemic autoimmune and metabolic conditions such as diabetes, obesity, multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, ankylosing spondylitis and autoimmune hepatitis. Finally, distant inflammatory responses due to a 'leaky gut' and microbiome changes are suspected in Alzheimer disease, Parkinson disease, chronic depression and autism spectrum disorders. This article introduces an extended 'epithelial barrier hypothesis', which proposes that the increase in epithelial barrier-damaging agents linked to industrialization, urbanization and modern life underlies the rise in allergic, autoimmune and other chronic conditions. Furthermore, it discusses how the immune responses to dysbiotic microbiota that cross the damaged barrier may be involved in the development of these diseases.
Collapse
|
26
|
Mitamura Y, Ogulur I, Pat Y, Rinaldi AO, Ardicli O, Cevhertas L, Brüggen MC, Traidl-Hoffmann C, Akdis M, Akdis CA. Dysregulation of the epithelial barrier by environmental and other exogenous factors. Contact Dermatitis 2021; 85:615-626. [PMID: 34420214 PMCID: PMC9293165 DOI: 10.1111/cod.13959] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 12/26/2022]
Abstract
The “epithelial barrier hypothesis” proposes that the exposure to various epithelial barrier–damaging agents linked to industrialization and urbanization underlies the increase in allergic diseases. The epithelial barrier constitutes the first line of physical, chemical, and immunological defense against environmental factors. Recent reports have shown that industrial products disrupt the epithelial barriers. Innate and adaptive immune responses play an important role in epithelial barrier damage. In addition, recent studies suggest that epithelial barrier dysfunction plays an essential role in the pathogenesis of the atopic march by allergen sensitization through the transcutaneous route. It is evident that external factors interact with the immune system, triggering a cascade of complex reactions that damage the epithelial barrier. Epigenetic and microbiome changes modulate the integrity of the epithelial barrier. Robust and simple measurements of the skin barrier dysfunction at the point‐of‐care are of significant value as a biomarker, as recently reported using electrical impedance spectroscopy to directly measure barrier defects. Understanding epithelial barrier dysfunction and its mechanism is key to developing novel strategies for the prevention and treatment of allergic diseases. The aim of this review is to summarize recent studies on the pathophysiological mechanisms triggered by environmental factors that contribute to the dysregulation of epithelial barrier function.
Collapse
Affiliation(s)
- Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos.,Division of Pediatric Allergy and Immunology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos.,Department of Medical Microbiology, Faculty of Medicine, Aydin Menderes University, Aydin, Turkey
| | - Arturo O Rinaldi
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos
| | - Ozge Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos.,Department of Microbiology, Faculty of Veterinary Medicine, Bursa Uludag University, Bursa, Turkey
| | - Lacin Cevhertas
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos.,Department of Medical Immunology, Institute of Health Sciences, Bursa Uludag University, Bursa, Turkey
| | - Marie-Charlotte Brüggen
- Christine Kühne-Center for Allergy Research and Education, Davos.,Department of Dermatology, University Hospital Zurich, Zurich, Switzerland.,Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Claudia Traidl-Hoffmann
- Christine Kühne-Center for Allergy Research and Education, Davos.,Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos.,Christine Kühne-Center for Allergy Research and Education, Davos
| |
Collapse
|
27
|
Kaymak T, Hruz P, Niess JH. Immune system and microbiome in the esophagus: implications for understanding inflammatory diseases. FEBS J 2021; 289:4758-4772. [PMID: 34213831 PMCID: PMC9542113 DOI: 10.1111/febs.16103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 05/20/2021] [Accepted: 07/01/2021] [Indexed: 12/15/2022]
Abstract
The gastrointestinal tract is the largest compartment of the body's immune system exposed to microorganisms, structural components and metabolites, antigens derived from the diet, and pathogens. Most studies have focused on immune responses in the stomach, the small intestine, and the colon, but the esophagus has remained an understudied anatomic immune segment. Here, we discuss the esophagus' anatomical and physiological distinctions that may account for inflammatory esophageal diseases.
Collapse
Affiliation(s)
- Tanay Kaymak
- Department of Biomedicine, University of Basel, Switzerland
| | - Petr Hruz
- Clarunis - University Center for Gastrointestinal and Liver Diseases Basel, Switzerland
| | - Jan Hendrik Niess
- Department of Biomedicine, University of Basel, Switzerland.,Clarunis - University Center for Gastrointestinal and Liver Diseases Basel, Switzerland
| |
Collapse
|
28
|
Bertschi NL, Bazzini C, Schlapbach C. The Concept of Pathogenic TH2 Cells: Collegium Internationale Allergologicum Update 2021. Int Arch Allergy Immunol 2021; 182:365-380. [PMID: 33845475 DOI: 10.1159/000515144] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/09/2021] [Indexed: 11/19/2022] Open
Abstract
T helper (TH) cells have evolved into distinct subsets that mediate specific immune responses to protect the host against a myriad of infectious and noninfectious challenges. However, if dysregulated, TH-cell subsets can cause inflammatory disease. Emerging evidence now suggests that human allergic disease is caused by a distinct subpopulation of pathogenic TH2 cells. Pathogenic TH2 cells from different type-2-driven diseases share a core phenotype and show overlapping functional attributes. The unique differentiation requirements, activating signals, and metabolic characteristics of pathogenic TH2 cells are just being discovered. A better knowledge of this particular TH2 cell population will enable the specific targeting of disease-driving pathways in allergy. In this review, we introduce a rational for classifying TH cells into distinct subsets, discuss the current knowledge on pathogenic TH2 cells, and summarize their involvement in allergic diseases.
Collapse
Affiliation(s)
- Nicole L Bertschi
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Cecilia Bazzini
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Christoph Schlapbach
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
29
|
Jensen ET, Langefeld CD, Zimmerman KD, Howard TD, Dellon ES. Epigenetic methylation in Eosinophilic Esophagitis: Molecular ageing and novel biomarkers for treatment response. Clin Exp Allergy 2020; 50:1372-1380. [PMID: 32986922 DOI: 10.1111/cea.13748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 09/09/2020] [Accepted: 09/20/2020] [Indexed: 01/16/2023]
Abstract
BACKGROUND Treatment failure in eosinophilic esophagitis (EoE) is common. We hypothesize that DNA methylation differs between patients by treatment response to topical steroids (oral viscous budesonide), thus offering the potential to inform targeting therapies. OBJECTIVE We sought to identify differentially methylated sites and affiliated genes in pre-treatment oesophageal cells between responders and non-responders and test for accelerated epigenetic ageing in oesophageal cells of EoE patients. METHODS DNA was extracted from prospectively collected and biobanked oesophageal biopsies from 36 Caucasian treatment naïve EoE patients at diagnosis. Methylation assays were completed using the Infinium HumanMethylation450 BeadChip. Normalized β values for each CpG site were tested (t test) for differential methylation. Further, 353 CpG probes were used to estimate epigenetic age for each patient and a linear regression model tested whether chronologic age and epigenetic age differed. Epigenetic age results were confirmed in an independent cohort of healthy controls. RESULTS Eighteen CpG sites were differentially methylated by treatment response (P < .00001). The mean epigenetic age and chronological age were 56.1 ± 11.1 and 36.7 ± 12.3 years, a mean age difference of 19.3 ± 5.2 years (P < .0001); accelerated ageing was not observed in the oesophageal cells of healthy controls. CONCLUSIONS AND CLINICAL RELEVANCE EoE patients that respond versus do not respond to treatment have differences in their methylation profile, including enrichment of genes in pathways consistent with cellular injury and repair due to environmental stress and cell adhesion and barrier integrity. EoE also appears to accelerate cellular ageing. Whether treatment can arrest or reverse accelerated epigenetic ageing and the implications for long-term disease progression is important areas for future research.
Collapse
Affiliation(s)
- Elizabeth T Jensen
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Department of Internal Medicine, Section on Gastroenterology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Carl D Langefeld
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Kip D Zimmerman
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Timothy D Howard
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Evan S Dellon
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
30
|
Breiteneder H, Peng Y, Agache I, Diamant Z, Eiwegger T, Fokkens WJ, Traidl‐Hoffmann C, Nadeau K, O'Hehir RE, O'Mahony L, Pfaar O, Torres MJ, Wang D, Zhang L, Akdis CA. Biomarkers for diagnosis and prediction of therapy responses in allergic diseases and asthma. Allergy 2020; 75:3039-3068. [PMID: 32893900 PMCID: PMC7756301 DOI: 10.1111/all.14582] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/25/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023]
Abstract
Modern health care requires a proactive and individualized response to diseases, combining precision diagnosis and personalized treatment. Accordingly, the approach to patients with allergic diseases encompasses novel developments in the area of personalized medicine, disease phenotyping and endotyping, and the development and application of reliable biomarkers. A detailed clinical history and physical examination followed by the detection of IgE immunoreactivity against specific allergens still represents the state of the art. However, nowadays, further emphasis focuses on the optimization of diagnostic and therapeutic standards and a large number of studies have been investigating the biomarkers of allergic diseases, including asthma, atopic dermatitis, allergic rhinitis, food allergy, urticaria and anaphylaxis. Various biomarkers have been developed by omics technologies, some of which lead to a better classification of distinct phenotypes or endotypes. The introduction of biologicals to clinical practice increases the need for biomarkers for patient selection, prediction of outcomes and monitoring, to allow for an adequate choice of the duration of these costly and long‐lasting therapies. Escalating healthcare costs together with questions about the efficacy of the current management of allergic diseases require further development of a biomarker‐driven approach. Here, we review biomarkers in diagnosis and treatment of asthma, atopic dermatitis, allergic rhinitis, viral infections, chronic rhinosinusitis, food allergy, drug hypersensitivity and allergen immunotherapy with a special emphasis on specific IgE, the microbiome and the epithelial barrier. In addition, EAACI guidelines on biologicals are discussed within the perspective of biomarkers.
Collapse
Affiliation(s)
- Heimo Breiteneder
- Institute of Pathophysiology and Allergy Research Medical University of Vienna Vienna Austria
| | - Ya‐Qi Peng
- Swiss Institute of Allergy and Asthma Research (SIAF) University Zurich Davos Switzerland
- CK CARE Christine Kühne Center for Allergy Research and Education Davos Switzerland
- Otorhinolaryngology Hospital The First Affiliated Hospital Sun Yat‐Sen University Guangzhou China
| | - Ioana Agache
- Department of Allergy and Clinical Immunology Faculty of Medicine Transylvania University of Brasov Brasov Romania
| | - Zuzana Diamant
- Department of Respiratory Medicine & Allergology Institute for Clinical Science Skane University Hospital Lund University Lund Sweden
- Department of Respiratory Medicine First Faculty of Medicine Charles University and Thomayer Hospital Prague Czech Republic
- Department of Clinical Pharmacy & Pharmacology University of GroningenUniversity Medical Center Groningen Groningen Netherlands
| | - Thomas Eiwegger
- Translational Medicine Program, Research Institute Hospital for Sick Children Toronto ON Canada
- Department of Immunology University of Toronto Toronto ON Canada
- Division of Immunology and Allergy Food Allergy and Anaphylaxis Program The Hospital for Sick Children Departments of Paediatrics and Immunology University of Toronto Toronto ON Canada
| | - Wytske J. Fokkens
- Department of Otorhinolaryngology Amsterdam University Medical Centres Amsterdam The Netherlands
| | - Claudia Traidl‐Hoffmann
- CK CARE Christine Kühne Center for Allergy Research and Education Davos Switzerland
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
- ZIEL ‐ Institute for Food & Health Technical University of Munich Freising‐Weihenstephan Germany
| | - Kari Nadeau
- Sean N. Parker Center for Allergy & Asthma Research Stanford University Stanford CA USA
| | - Robyn E. O'Hehir
- Department of Allergy, immunology and Respiratory Medicine Central Clinical School Monash University Melbourne Vic. Australia
- Allergy, Asthma and Clinical Immunology Service Alfred Health Melbourne Vic. Australia
| | - Liam O'Mahony
- Departments of Medicine and Microbiology APC Microbiome Ireland National University of Ireland Cork Ireland
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery Section of Rhinology and Allergy University Hospital MarburgPhilipps‐Universität Marburg Marburg Germany
| | - Maria J. Torres
- Allergy Unit Regional University Hospital of Malaga‐IBIMA‐UMA‐ARADyAL Malaga Spain
| | - De‐Yun Wang
- Department of Otolaryngology Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery and Department of Allergy Beijing TongRen Hospital Beijing China
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University Zurich Davos Switzerland
- CK CARE Christine Kühne Center for Allergy Research and Education Davos Switzerland
| |
Collapse
|
31
|
Zdanowicz K, Kucharska M, Reszec J, Lebensztejn DM, Daniluk U. Immunohistochemical markers for eosinophilic esophagitis. Scand J Gastroenterol 2020; 55:1277-1283. [PMID: 33032462 DOI: 10.1080/00365521.2020.1831053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIMS Eosinophilic esophagitis (EoE) is a chronic, local immune-mediated esophageal disease with eosinophil-dominated inflammation. The incidence of the disease is rapidly increasing in both children and adults. The pathogenesis of the disease is still not well understood. We present a review of the literature devoted to the EoE immunopathology, in particular the markers of inflammation and epithelial integrity, and their usefulness in disease monitoring and therapy. METHODS We performed a systematic search of the MEDLINE/PubMed databases for studies to examine the use of immunohistochemistry as a diagnostic tool for EoE. RESULTS The gold standard of EoE diagnosis requires multiple endoscopies with biopsies for histological assessment. The minimum number of eosinophils evaluated in hematoxylin-eosin staining to diagnose EoE is 15 per high-power field in at least one esophageal mucosa biopsy. However, in some cases, the count of eosinophils is not specific and insufficient as the only indicator. Recent works confirm the usefulness of assessment of some biomarkers in establishing the diagnosis and monitoring the treatment effects. CONCLUSIONS Immunohistochemistry seems to be a promising option not only in clinical recognition, but also in the selection and monitoring of treatment effects. However, these methods have not yet recommended for routine clinical use.
Collapse
Affiliation(s)
- Katarzyna Zdanowicz
- Department of Pediatrics, Gastroenterology, Hepatology, Nutrition and Allergology, Medical University of Bialystok, Bialystok, Poland
| | - Magdalena Kucharska
- Department of Pediatrics, Gastroenterology, Hepatology, Nutrition and Allergology, Medical University of Bialystok, Bialystok, Poland
| | - Joanna Reszec
- Department of Medical Pathomorphology, Medical University of Bialystok, Bialystok, Poland
| | - Dariusz Marek Lebensztejn
- Department of Pediatrics, Gastroenterology, Hepatology, Nutrition and Allergology, Medical University of Bialystok, Bialystok, Poland
| | - Urszula Daniluk
- Department of Pediatrics, Gastroenterology, Hepatology, Nutrition and Allergology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
32
|
Braun C, Vocanson M, Nicolas JF, Nosbaum A. Physiopathologie de la dermatite atopique et des autres maladies atopiques : une approche globale est-elle possible ? Ann Dermatol Venereol 2020; 147:11S4-11S11. [PMID: 33250137 DOI: 10.1016/s0151-9638(20)31082-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Atopy is defined by the propensity to develop an exaggerated type-2 inflammatory response to environmental molecules. Clinically, atopy is diagnosed when atopic disease occurs: atopic dermatitis, food allergy, atopic asthma and allergic rhinitis and conjunctivitis. Whereas the classical "atopic march" is increasingly challenged through epidemiological studies, type-2 cellular inflammation is a characteristic shared by the atopic diseases. This inflammation can be innate (non-specific: eosinophils, mast cells, dendritic cells, innate lymphoid cells [ILC]), or adaptive (antigen-specific, involving T cells). Interleukins (IL-)4, 5 and 13 are major actors of type-2 inflammation and are mainly produced by ILC and T cells. The efficacy of treatments targeting these type-2 cytokines highlight the importance of type-2 inflammation in atopic diseases. However, several patients do not respond to type-2 targeting treatments, highlighting the presence of other actors in pathophysiology of atopic diseases: alteration of epithelial barrier, IgE-mediated allergic responses, type-17 inflammation. Thus, the term "endotype" can illustrate this diversity in pathophysiology. Finally, a global approach of atopic diseases, as type-2 inflammatory diseases, is fundamental, but not sufficient. An approach by endotype is advisable, in a personalized medicine perspective. © 2020 Elsevier Masson SAS. All rights reserved.
Collapse
Affiliation(s)
- C Braun
- CIRI - Centre international de recherche en infectiologie (International Center for Infectiology Research), INSERM U1111, CNRS UMR 5308, 21, avenue Tony-Garnier, 69007 Lyon, France; Hospices civils de Lyon, Hôpital Femme-Mère-Enfant, service de pneumologie et allergologie pédiatriques, Bron, France.
| | - M Vocanson
- CIRI - Centre international de recherche en infectiologie (International Center for Infectiology Research), INSERM U1111, CNRS UMR 5308, 21, avenue Tony-Garnier, 69007 Lyon, France
| | - J F Nicolas
- CIRI - Centre international de recherche en infectiologie (International Center for Infectiology Research), INSERM U1111, CNRS UMR 5308, 21, avenue Tony-Garnier, 69007 Lyon, France; Hospices civils de Lyon, Centre hospitalier Lyon-Sud, service d'allergologie et immunologie clinique, Pierre-Bénite, France
| | - A Nosbaum
- CIRI - Centre international de recherche en infectiologie (International Center for Infectiology Research), INSERM U1111, CNRS UMR 5308, 21, avenue Tony-Garnier, 69007 Lyon, France; Hospices civils de Lyon, Centre hospitalier Lyon-Sud, service d'allergologie et immunologie clinique, Pierre-Bénite, France
| |
Collapse
|
33
|
Sugita K, Soyka MB, Wawrzyniak P, Rinaldi AO, Mitamura Y, Akdis M, Akdis CA. Outside-in hypothesis revisited: The role of microbial, epithelial, and immune interactions. Ann Allergy Asthma Immunol 2020; 125:517-527. [PMID: 32454094 DOI: 10.1016/j.anai.2020.05.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/14/2020] [Accepted: 05/17/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Our understanding of the origin of allergic diseases has increased in recent years, highlighting the importance of microbial dysbiosis and epithelial barrier dysfunction in affected tissues. Exploring the microbial-epithelial-immune crosstalk underlying the mechanisms of allergic diseases will allow the development of novel prevention and treatment strategies for allergic diseases. DATA SOURCES This review summarizes the recent advances in microbial, epithelial, and immune interactions in atopic dermatitis, allergic rhinitis, chronic rhinosinusitis, and asthma. STUDY SELECTIONS We performed a literature search, identifying relevant recent primary articles and review articles. RESULTS Dynamic crosstalk between the environmental factors and microbial, epithelial, and immune cells in the development of atopic dermatitis, allergic rhinitis, chronic rhinosinusitis, and asthma underlies the pathogenesis of these diseases. There is substantial evidence in the literature suggesting that environmental factors directly affect barrier function of the epithelium. In addition, T-helper 2 (TH2) cells, type 2 innate lymphoid cells, and their cytokine interleukin 13 (IL-13) damage skin and lung barriers. The effects of environmental factors may at least in part be mediated by epigenetic mechanisms. Histone deacetylase activation by type 2 immune response has a major effect on leaky barriers and blocking of histone deacetylase activity corrects the defective barrier in human air-liquid interface cultures and mouse models of allergic asthma with rhinitis. We also present and discuss a novel device to detect and monitor skin barrier dysfunction, which provides an opportunity to rapidly and robustly assess disease severity. CONCLUSION A complex interplay between environmental factors, epithelium, and the immune system is involved in the development of systemic allergic diseases.
Collapse
Affiliation(s)
- Kazunari Sugita
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland; Division of Dermatology, Department of Medicine of Sensory and Motor Organs, Faculty of Medicine, Tottori University, Yonago, Japan.
| | - Michael B Soyka
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Department of Otorhinolaryngology, Head and Neck Surgery, University and University Hospital of Zurich, Zurich, Switzerland
| | - Paulina Wawrzyniak
- Division of Clinical Chemistry and Biochemistry and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Arturo O Rinaldi
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| |
Collapse
|
34
|
Abstract
Eosinophilic esophagitis (EoE) is a relatively new disease that has reached an incidence similar to that of Crohn disease and ulcerative colitis. With this increased presence, greater recognition is essential. This applies both to children with nonspecific but potentially debilitating symptoms and to adults who have spent years behaviorally compensating for narrow esophageal strictures. The pathogenesis of EoE is rapidly being unraveled and is based on initiation of a type II allergic response to specific food antigens, leading to dense esophageal eosinophilia, chronic inflammation, and esophageal fibrosis. With greater familiarity and understanding of EoE, treatments are evolving, including identification and avoidance of food antigens; broad applications of topical steroids; and, eventually, pathway-specific biologic therapy.
Collapse
|
35
|
Sugita K, Kabashima K. Tight junctions in the development of asthma, chronic rhinosinusitis, atopic dermatitis, eosinophilic esophagitis, and inflammatory bowel diseases. J Leukoc Biol 2020; 107:749-762. [PMID: 32108379 DOI: 10.1002/jlb.5mr0120-230r] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 01/12/2020] [Accepted: 01/26/2020] [Indexed: 02/06/2023] Open
Abstract
This review focuses on recent developments related to asthma, chronic rhinosinusitis, atopic dermatitis (AD), eosinophilic esophagitis, and inflammatory bowel diseases (IBD), with a particular focus on tight junctions (TJs) and their role in the pathogenetic mechanisms of these diseases. Lung, skin, and intestinal surfaces are lined by epithelial cells that interact with environmental factors and immune cells. Therefore, together with the cellular immune system, the epithelium performs a pivotal role as the first line physical barrier against external antigens. Paracellular space is almost exclusively sealed by TJs and is maintained by complex protein-protein interactions. Thus, TJ dysfunction increases paracellular permeability, resulting in enhanced flux across TJs. Epithelial TJ dysfunction also causes immune cell activation and contributes to the pathogenesis of chronic lung, skin, and intestinal inflammation. Characterization of TJ protein alteration is one of the key factors for enhancing our understanding of allergic diseases as well as IBDs. Furthermore, TJ-based epithelial disturbance can promote immune cell behaviors, such as those in dendritic cells, Th2 cells, Th17 cells, and innate lymphoid cells (ILCs), thereby offering new insights into TJ-based targets. The purpose of this review is to illustrate how TJ dysfunction can lead to the disruption of the immune homeostasis in barrier tissues and subsequent inflammation. This review also highlights the various TJ barrier dysfunctions across different organ sites, which would help to develop future drugs to target allergic diseases and IBD.
Collapse
Affiliation(s)
- Kazunari Sugita
- Division of Dermatology, Department of Medicine of Sensory and Motor Organs, Tottori University Faculty of Medicine, Yonago, Japan
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
36
|
Duong LD, Rawson R, Bezryadina A, Manresa MC, Newbury RO, Dohil R, Liu Z, Barrett K, Kurten R, Aceves SS. TGFβ1 single-nucleotide polymorphism C-509T alters mucosal cell function in pediatric eosinophilic esophagitis. Mucosal Immunol 2020; 13:110-117. [PMID: 31636346 PMCID: PMC6917872 DOI: 10.1038/s41385-019-0214-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 09/18/2019] [Accepted: 09/24/2019] [Indexed: 02/04/2023]
Abstract
Eosinophilic esophagitis (EoE) is a chronic Th2 antigen-driven disorder associated with tissue remodeling. Inflammation and remodeling lead to esophageal rigidity, strictures, and dysphagia. TGFβ1 drives esophageal remodeling including epithelial barrier dysfunction and subepithelial fibrosis. A functional SNP in the TGFβ1 gene that increases its transcription (C-509T) is associated with elevated numbers of esophageal TGFβ1-expressing cells. We utilized esophageal biopsies and fibroblasts from TT-genotype EoE children to understand if TGFβ1 influenced fibroblast and epithelial cell function in vivo. Genotype TT EoE esophageal fibroblasts had higher baseline TGFβ1, collagen1α1, periostin, and MMP2 (p < 0.05) gene expression and distinct contractile properties compared with CC genotype (n = 6 subjects per genotype). In vitro TGFβ1 exposure caused greater induction of target gene expression in genotype CC fibroblasts (p < 0.05). Esophageal biopsies from TT-genotype subjects had significantly less epithelial membrane-bound E-cadherin (p < 0.01) and wider cluster distribution at nanometer resolution. TGFβ1 treatment of stratified primary human esophageal epithelial cells and spheroids disrupted transepithelial resistance (p < 0.001) and E-cadherin localization (p < 0.0001). A TGFβ1-receptor-I inhibitor improved TGFβ1-mediated E-cadherin mislocalization. These data suggest that EoE severity can depend on genotypic differences that increase in vivo exposure to TGFβ1. TGFβ1 inhibition may be a useful therapy in subsets of EoE patients.
Collapse
Affiliation(s)
- L D Duong
- Division of Allergy & Immunology, University of California, San Diego, San Diego, CA, USA
- Department of Pediatrics, University of California, San Diego and Rady Children's Hospital San Diego, San Diego, CA, USA
| | - R Rawson
- Division of Allergy & Immunology, University of California, San Diego, San Diego, CA, USA
- Department of Pediatrics, University of California, San Diego and Rady Children's Hospital San Diego, San Diego, CA, USA
| | - A Bezryadina
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - M C Manresa
- Division of Allergy & Immunology, University of California, San Diego, San Diego, CA, USA
- Department of Pediatrics, University of California, San Diego and Rady Children's Hospital San Diego, San Diego, CA, USA
| | - R O Newbury
- Department of Pathology, University of California, San Diego, Rady Children's Hospital, San Diego, CA, USA
| | - R Dohil
- Department of Pediatrics, University of California, San Diego and Rady Children's Hospital San Diego, San Diego, CA, USA
- Division of Gastroenterology, San Diego, CA, USA
| | - Z Liu
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - K Barrett
- Division of Gastroenterology, San Diego, CA, USA
- Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - R Kurten
- Arkansas Children's Research Institute and University of Arkansas for Medical Sciences, Little Rock, AK, USA
| | - S S Aceves
- Division of Allergy & Immunology, University of California, San Diego, San Diego, CA, USA.
- Department of Pediatrics, University of California, San Diego and Rady Children's Hospital San Diego, San Diego, CA, USA.
- Department of Medicine, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
37
|
Plundrich NJ, Smith AR, Borst LB, Snider DB, Käser T, Dellon ES, Blikslager AT, Odle J, Lila MA, Laster SM. Oesophageal eosinophilia accompanies food allergy to hen egg white protein in young pigs. Clin Exp Allergy 2020; 50:95-104. [PMID: 31702085 PMCID: PMC6930966 DOI: 10.1111/cea.13527] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 11/01/2019] [Accepted: 11/04/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Esophagitis with eosinophilia, inflammation, and fibrosis represent a chronic condition in humans with food allergies. OBJECTIVE In this investigation, we asked whether esophagitis with an eosinophilic component is observed in young pigs rendered allergic to hen egg white protein (HEWP). METHODS Food allergy was induced in young pigs using two protocols. In one protocol, sensitized pigs were challenged by gavage with a single dose of HEWP. Clinical signs were monitored for 24 hours, and then, gastrointestinal (GI) tissues were collected for histological examination. The phenotype of circulating, ovalbumin (OVA)-specific T cells also was examined in HEWP challenged animals. In the second protocol, sensitized animals were fed HEWP for 28 days. Animals were then examined by endoscopy and gastrointestinal tissues collected for histological examination. RESULTS In pigs challenged by gavage with HEWP, clinical signs were noted in 5/6 pigs including diarrhoea, emesis, and skin rash. Clinical signs were not seen in any control group. Histological analysis revealed significant levels of oesophageal eosinophilic infiltration (P < .05) in 4/6 of these animals, with two also displaying eosinophilic infiltration in the stomach. Eosinophils were not increased in ileum or colon samples. Increased numbers of circulating, OVA-specific CD4+ T cells also were observed in pigs that received HEWP by gavage. In the group of animals fed HEWP, endoscopy revealed clinical signs of esophagitis including oedema, granularity, white spots, and furrowing, while histology revealed oedema, immune cell infiltration, and basal zone hyperplasia. CONCLUSIONS AND CLINICAL RELEVANCE Food allergy in the pig can be associated with esophagitis based on histological and endoscopic findings, including eosinophilic infiltration. The young pig may, therefore, be a useful large animal model for the study of eosinophilic esophagitis in humans.
Collapse
Affiliation(s)
- Nathalie J. Plundrich
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Andrew R. Smith
- Laboratory of Developmental Nutrition, Department of Animal Science, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Luke B. Borst
- Department of Population Health and Pathobiology, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Douglas B. Snider
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine and Translational Research Training Program, Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Tobias Käser
- Department of Population Health and Pathobiology, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Evan S. Dellon
- Center for Esophageal Diseases and Swallowing, Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Anthony T. Blikslager
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Jack Odle
- Laboratory of Developmental Nutrition, Department of Animal Science, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Mary Ann Lila
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Scott M. Laster
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
38
|
Ruffner MA, Song L, Maurer K, Shi L, Carroll MC, Wang JX, Muir AB, Spergel JM, Sullivan KE. Toll-like receptor 2 stimulation augments esophageal barrier integrity. Allergy 2019; 74:2449-2460. [PMID: 31267532 PMCID: PMC7083217 DOI: 10.1111/all.13968] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 04/26/2019] [Accepted: 05/13/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Germline-encoded innate immune pattern recognition receptors (PRR) are expressed at epithelial surfaces and modulate epithelial defenses. Evidence suggests that stimulation of the Toll-like receptor (TLR) family of PRR may regulate epithelial barrier integrity by upregulating tight junction (TJ) complex protein expression, but it is not known whether this mechanism is utilized in esophageal epithelial cells. TJ complex proteins maintain intact barrier function and are dysregulated in atopic disorders including eosinophilic esophagitis. METHODS Pattern recognition receptors expression was assessed in EoE and control primary esophageal epithelial cells, demonstrating robust expression of TLR2 and TLR3. The three-dimensional air-liquid interface culture (ALI) model was used to test whether TLR2 or TLR3 stimulation alters epithelial barrier function using an in vitro model of human epithelium. Transepithelial electrical resistance (TEER) and FITC-Dextran permeability were evaluated to assess membrane permeability. ALI cultures were evaluated by histology, immunohistochemistry, Western blotting, and chromatin immunoprecipitation (ChIP). RESULTS TLR3 stimulation did not change TEER in the ALI model. TLR2 stimulation increased TEER (1.28- to 1.31-fold) and decreased paracellular permeability to FITC-Dextran, and this effect was abolished by treatment with anti-TLR2 blocking antibody. TJ complex proteins claudin-1 and zonula occludens-1 were upregulated following TLR2 stimulation, and ChIP assay demonstrated altered histone 4 acetyl binding at the TJP1 enhancer and CLDN1 enhancer and promoter following zymosan treatment, implying the occurrence of durable chromatin changes. CONCLUSIONS Our findings implicate the TLR2 pathway as a potential regulator of esophageal epithelial barrier function and suggest that downstream chromatin modifications are associated with this effect.
Collapse
Affiliation(s)
- Melanie A Ruffner
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, USA
| | - Li Song
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kelly Maurer
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lihua Shi
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Margaret C Carroll
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Joshua X Wang
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Amanda B Muir
- Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jonathan M Spergel
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, USA
| | - Kathleen E Sullivan
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
39
|
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: 18] [Impact Index Per Article: 3.0] [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
|
40
|
Simon D, Simon HU. Relationship of skin barrier breakdown and eosinophilic esophagitis. J Allergy Clin Immunol 2019; 145:90-92.e1. [PMID: 31738992 DOI: 10.1016/j.jaci.2019.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/06/2019] [Accepted: 11/12/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Dagmar Simon
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland; Department of Clinical Immunology and Allergology, Sechenov University, Moscow, Russia
| |
Collapse
|
41
|
Rinaldi AO, Morita H, Wawrzyniak P, Dreher A, Grant S, Svedenhag P, Akdis CA. Direct assessment of skin epithelial barrier by electrical impedance spectroscopy. Allergy 2019; 74:1934-1944. [PMID: 30989659 DOI: 10.1111/all.13824] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 03/12/2019] [Accepted: 03/26/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Many skin and mucosal inflammatory disorders, such as atopic dermatitis, have been associated with an impaired epithelial barrier function, which allows allergens, pollutants, or microbes to enter the tissue and activate the immune response. The aim of this study was to establish a method to directly assess in vivo the epidermal barrier function by electrical impedance (EI) spectroscopy. METHODS Mice epidermal barrier was damaged by epicutaneous application of proteases and cholera toxin and by tape stripping. EI and transepidermal water loss (TEWL) were measured before and after the application. Additionally, histological analysis, immunofluorescence staining, and RT-PCR were performed on skin biopsies to evaluate the epithelial barrier. RESULTS A few hours after papain application, a dose-dependent reduction of EI was detected, reflecting the decreased barrier function. At the same time, an increase of TEWL was observed, with a significant negative correlation with EI, demonstrating that EI changes were directly linked to barrier defects. Twenty-four and 48 hours after the treatment, EI starts to increase to background levels, indicating tissue healing and restoration of skin barrier. Barrier disruption was confirmed by histological analysis showing an impaired stratum corneum and higher cellular infiltration after papain application. In addition, immunofluorescence staining and RT-PCR showed downregulation of molecules involved in the barrier function, such as filaggrin, occludin, and claudin-1, and mRNA levels of filaggrin, loricrin, and involucrin. Comparable results were observed after tape stripping and cholera toxin treatment. CONCLUSION Electrical impedance spectroscopy is a rapid and reliable diagnostic tool to detect skin barrier defects.
Collapse
Affiliation(s)
- Arturo O. Rinaldi
- Swiss Institute of Allergy and Asthma Research (SIAF) Davos Switzerland
- Christine Kühne – Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
| | - Hideaki Morita
- Swiss Institute of Allergy and Asthma Research (SIAF) Davos Switzerland
- Christine Kühne – Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
| | - Paulina Wawrzyniak
- Swiss Institute of Allergy and Asthma Research (SIAF) Davos Switzerland
- Christine Kühne – Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
| | - Anita Dreher
- Swiss Institute of Allergy and Asthma Research (SIAF) Davos Switzerland
- Christine Kühne – Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
| | | | | | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) Davos Switzerland
- Christine Kühne – Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
| |
Collapse
|
42
|
Rochman M, Azouz NP, Rothenberg ME. Epithelial origin of eosinophilic esophagitis. J Allergy Clin Immunol 2019; 142:10-23. [PMID: 29980278 DOI: 10.1016/j.jaci.2018.05.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/18/2018] [Accepted: 05/21/2018] [Indexed: 02/06/2023]
Abstract
Eosinophilic esophagitis (EoE) is a chronic, allergen-driven inflammatory disease of the esophagus characterized predominantly by eosinophilic inflammation, leading to esophageal dysfunction. Converging data have placed the esophageal epithelium at the center of disease pathogenesis. In particular, the main EoE disease susceptibility loci at 2p23 and 5p22 encode for gene products that are produced by the esophageal epithelium: the intracellular protease calpain 14 and thymic stromal lymphopoietin, respectively. Furthermore, genetic and functional data establish a primary role for impaired epithelial barrier function in disease susceptibility and pathoetiology. Additionally, the EoE transcriptome, a set of genes dysregulated in the esophagi of patients with EoE, is enriched in genes that encode for proteins involved in esophageal epithelial cell differentiation. This transcriptome has a high proportion of esophagus-specific epithelial genes that are notable for the unexpected enrichment in genes encoding for proteases and protease inhibitors, as well as in IL-1 family genes, demonstrating a previously unappreciated role for innate immunity responses in the esophagus under homeostatic conditions. Among these pathways, basal production of the serine protease inhibitor, Kazal-type 7 (SPINK7) has been demonstrated to be part of the normal differentiation program of esophageal epithelium. Profound lost expression of SPINK7 occurs in patients with EoE and is sufficient for unleashing increased proteolytic activity (including urokinase plasminogen activator), impaired barrier function, and production of large quantities of proinflammatory and proallergic cytokines, including thymic stromal lymphopoietin. Collectively, we put forth a model in which the esophagus is normally equipped as an anti-inflammatory sensing organ and that defects in this pathway, mediated by epithelial protease/protease inhibitor imbalances, unleash inflammatory responses resulting in disorders, such as EoE.
Collapse
Affiliation(s)
- Mark Rochman
- Division of Allergy and Immunology, Department of Pediatrics Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Nurit P Azouz
- Division of Allergy and Immunology, Department of Pediatrics Cincinnati Children's Hospital Medical Center, 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, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
43
|
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: 9.5] [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
|
44
|
Zheng P, Tan HY. New developments in diagnosis and treatment of eosinophilic esophagitis. Shijie Huaren Xiaohua Zazhi 2019; 27:828-834. [DOI: 10.11569/wcjd.v27.i13.828] [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] [Indexed: 02/06/2023] Open
Abstract
Eosinophilic esophagitis (EoE) is a chronic, allergen-driven, immune mediated disease of the esophagus that progresses to esophageal fibrostenosis if not well treated. The aim of this review is to provide an update on recent clinical advances in the development of diagnostic and therapeutic approaches for EoE. At present, the diagnosis and disease monitoring for EoE rely on repetitive endoscopic evaluations and esophageal tissue acquisition for histopathologic analysis. Recent advancements in EoE diagnosis include transnasal endoscopy, endoscopic functional lumen imaging probe, and the emergence of non-invasive diagnostic tools including cytosponge, esophageal string test, and mucosal impedance probe. Biomarkers for EoE have not yet proven their clinical utility. The current treatment modalities for EoE are topical corticosteroid, proton-pump inhibitors, elimination diet, and dilation. Promising results from clinical trials are emerging for biologic agents that target the interleukin (IL)-13 and the IL-4/IL-13 receptor, specifically, RPC4046 and dupilumab. Especially, dupilumab is promising to become the orphan drug of EoE.
Collapse
Affiliation(s)
- Pu Zheng
- Department of Emergency Medicine, Beijing Hospital, Beijing 100005, China
| | - Huang-Ying Tan
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
45
|
Masterson JC, Biette KA, Hammer JA, Nguyen N, Capocelli KE, Saeedi BJ, Harris RF, Fernando SD, Hosford LB, Kelly CJ, Campbell EL, Ehrentraut SF, Ahmed FN, Nakagawa H, Lee JJ, McNamee EN, Glover LE, Colgan SP, Furuta GT. Epithelial HIF-1α/claudin-1 axis regulates barrier dysfunction in eosinophilic esophagitis. J Clin Invest 2019; 129:3224-3235. [PMID: 31264974 PMCID: PMC6668670 DOI: 10.1172/jci126744] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 05/16/2019] [Indexed: 12/19/2022] Open
Abstract
Epithelial barrier dysfunction is a significant factor in many allergic diseases, including eosinophilic esophagitis (EoE). Infiltrating leukocytes and tissue adaptations increase metabolic demands and decrease oxygen availability at barrier surfaces. Understanding of how these processes impact barrier is limited, particularly in allergy. Here, we identified a regulatory axis whereby the oxygen-sensing transcription factor HIF-1α orchestrated epithelial barrier integrity, selectively controlling tight junction CLDN1 (claudin-1). Prolonged experimental hypoxia or HIF1A knockdown suppressed HIF-1α-dependent claudin-1 expression and epithelial barrier function, as documented in 3D organotypic epithelial cultures. L2-IL5OXA mice with EoE-relevant allergic inflammation displayed localized eosinophil oxygen metabolism, tissue hypoxia, and impaired claudin-1 barrier via repression of HIF-1α/claudin-1 signaling, which was restored by transgenic expression of esophageal epithelial-targeted stabilized HIF-1α. EoE patient biopsy analysis identified a repressed HIF-1α/claudin-1 axis, which was restored via pharmacologic HIF-1α stabilization ex vivo. Collectively, these studies reveal HIF-1α's critical role in maintaining barrier and highlight the HIF-1α/claudin-1 axis as a potential therapeutic target for EoE.
Collapse
Affiliation(s)
- Joanne C. Masterson
- Allergy, Inflammation and Remodeling Research Laboratory, Human Health Research Institute, Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
- Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics, University of Colorado School of Medicine; Digestive Health Institute, Children’s Hospital Colorado; Aurora, Colorado, USA
- Mucosal Inflammation Program, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Kathryn A. Biette
- Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics, University of Colorado School of Medicine; Digestive Health Institute, Children’s Hospital Colorado; Aurora, Colorado, USA
- Mucosal Inflammation Program, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Juliet A. Hammer
- Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics, University of Colorado School of Medicine; Digestive Health Institute, Children’s Hospital Colorado; Aurora, Colorado, USA
- Mucosal Inflammation Program, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Nathalie Nguyen
- Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics, University of Colorado School of Medicine; Digestive Health Institute, Children’s Hospital Colorado; Aurora, Colorado, USA
- Mucosal Inflammation Program, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Kelley E. Capocelli
- Department of Pathology, Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Bejan J. Saeedi
- Mucosal Inflammation Program, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Rachel F. Harris
- Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics, University of Colorado School of Medicine; Digestive Health Institute, Children’s Hospital Colorado; Aurora, Colorado, USA
- Mucosal Inflammation Program, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Shahan D. Fernando
- Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics, University of Colorado School of Medicine; Digestive Health Institute, Children’s Hospital Colorado; Aurora, Colorado, USA
- Mucosal Inflammation Program, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Lindsay B. Hosford
- Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics, University of Colorado School of Medicine; Digestive Health Institute, Children’s Hospital Colorado; Aurora, Colorado, USA
- Mucosal Inflammation Program, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Caleb J. Kelly
- Mucosal Inflammation Program, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Eric L. Campbell
- Mucosal Inflammation Program, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Stefan F. Ehrentraut
- Mucosal Inflammation Program, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Faria N. Ahmed
- Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics, University of Colorado School of Medicine; Digestive Health Institute, Children’s Hospital Colorado; Aurora, Colorado, USA
| | - Hiroshi Nakagawa
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Eóin N. McNamee
- Allergy, Inflammation and Remodeling Research Laboratory, Human Health Research Institute, Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
- Mucosal Inflammation Program, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Louise E. Glover
- Mucosal Inflammation Program, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Sean P. Colgan
- Mucosal Inflammation Program, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Glenn T. Furuta
- Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics, University of Colorado School of Medicine; Digestive Health Institute, Children’s Hospital Colorado; Aurora, Colorado, USA
- Mucosal Inflammation Program, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
46
|
Affiliation(s)
- Seema S Aceves
- Departments of Pediatrics and Medicine, Division of Allergy, Immunology, University of California, San Diego, La Jolla, California.
| |
Collapse
|
47
|
Comparison of mucosal impedance measurements throughout the esophagus and mucosal eosinophil counts in endoscopic biopsy specimens in eosinophilic esophagitis. Gastrointest Endosc 2019; 89:693-700.e1. [PMID: 30145316 DOI: 10.1016/j.gie.2018.08.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 08/17/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Assessing eosinophilic esophagitis (EoE) activity from limited esophageal mucosal biopsy samples has been questioned. Here our aim was to compare mucosal impedance (MI) throughout the esophagus and eosinophil counts in endoscopic biopsy samples in EoE. METHODS We compared 20-site MI using a balloon catheter in the esophagus and eosinophils per high-power field (eos/HPF) in esophageal mucosal biopsy samples. Data are summarized as median (interquartile range) comparing control subjects and EoE using Mann-Whitney rank sum test and between endoscopic reference score and MI (minimal and average) using rank Spearman correlation. RESULTS Ten adult control patients (ages 38-70) and 23 EoE patients (ages 21-80, 18 active) were studied. The mean (range) pan-esophageal MI in control subjects was significantly higher (6435 ohms [4546-7301]) compared with EoE patients (2004 ohms [1437-2546], P < .001). In control patients 172 of 180 (95.6%) individual impedance measurements (18 per patient) were normal when compared with 126 of 432 (29.2%) measurements in EoE. No EoE patient had uniformly normal MI. MI varied widely, with 19 of 23 patients having values above and below 2300 ohms (normal) regardless of EoE activity. Correlation of maximim eos/HPF with minimum and average MI per patient was r = -.243, P = .072 and r = -.358, P = .086, respectively. Of 5 patients with inactive EoE, 3 had >50% abnormal MI segments. Correlation coefficients of the endoscopic reference score with minimum and average MI were r = -.154, P = .47 and r = -.27, P = .20, respectively. The procedure was <5 minutes without adverse events. CONCLUSIONS MI is lower in the esophagus of EoE patients compared with control subjects with poor correlation between peak esophageal eosinophil counts, EoE activity, and MI. Segmental esophageal MI provides a unique marker of esophageal dysfunction in EoE. (Clinical trial registration number: NCT02995395.).
Collapse
|
48
|
Abstract
Gastrointestinal (GI) allergic disease is an umbrella term used to describe a variety of adverse, food antigen-driven, immune-mediated diseases. Although these diseases vary mechanistically, common elements include a breakdown of immunologic tolerance, a biased type 2 immune response, and an impaired mucosal barrier. These pathways are influenced by diverse factors such as diet, infections, exposure to antibiotics and chemicals, GI microbiome composition, and genetic and epigenetic elements. Early childhood has emerged as a critical period when these factors have a dramatic impact on shaping the immune system and therefore triggering or protecting against the onset of GI allergic diseases. In this Review, we will discuss the latest findings on the molecular and cellular mechanisms that govern GI allergic diseases and how these findings have set the stage for emerging preventative and treatment strategies.
Collapse
|
49
|
Nhu QM, Moawad FJ. New Developments in the Diagnosis and Treatment of Eosinophilic Esophagitis. CURRENT TREATMENT OPTIONS IN GASTROENTEROLOGY 2019; 17:48-62. [PMID: 30707411 PMCID: PMC6519728 DOI: 10.1007/s11938-019-00216-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW Eosinophilic esophagitis (EoE) is a chronic, allergen-driven, immune-mediated disease of the esophagus that progresses to esophageal fibrostenosis if left untreated. The aim of this review is to provide a concise update on recent clinically relevant advances in the development of diagnostic and therapeutic approaches for EoE. RECENT FINDINGS Current diagnostic and disease monitoring protocols for EoE rely on repetitive endoscopic evaluations and esophageal tissue acquisition for histopathologic analysis. Recent advancements in EoE diagnosis include endoscopic functional lumen imaging probe (FLIP), transnasal endoscopy (TNE), and the emergence of non-invasive diagnostic tools including cytosponge, esophageal string test, and mucosal impedance probe. Biomarkers for EoE have not yet proven their clinical utility. No Food and Drug Administration (FDA)-approved drugs currently exist for the treatment of EoE. Topical corticosteroid, proton-pump inhibitors (PPI), elimination diet, and dilation are the current treatment modalities for confirmed EoE. Promising results from clinical trials are emerging for biologic agents that target the interleukin (IL)-13 and the IL-4/IL-13 receptor, specifically, RPC4046 and dupilumab, respectively. New diagnostic algorithms, non-invasive diagnostic strategies, and treatment modalities for EoE are emerging. Patients with EoE continue to require a multimodal and multi-disciplinary management approach.
Collapse
Affiliation(s)
- Quan M Nhu
- Division of Gastroenterology & Hepatology, Scripps Clinic, 10666 N. Torrey Pines Road, Suite 203N, La Jolla, CA, 92037, USA.
- Scripps Research Translational Institute, The Scripps Research Institute, La Jolla, CA, USA.
| | - Fouad J Moawad
- Division of Gastroenterology & Hepatology, Scripps Clinic, 10666 N. Torrey Pines Road, Suite 203N, La Jolla, CA, 92037, USA.
| |
Collapse
|
50
|
Doshi A, Khamishon R, Rawson R, Duong L, Dohil L, Myers SJ, Bell B, Dohil R, Newbury RO, Barrett KE, Kurten RC, Aceves SS. Interleukin 9 Alters Epithelial Barrier and E-cadherin in Eosinophilic Esophagitis. J Pediatr Gastroenterol Nutr 2019; 68:225-231. [PMID: 30211842 PMCID: PMC6344288 DOI: 10.1097/mpg.0000000000002144] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) is a chronic TH2-assocated inflammatory condition accompanied by substantial impairments in epithelial barrier function and increased numbers of interleukin 9 (IL-9) expressing inflammatory cells. While IL-9 is known to affect barrier function in the intestine, the functional effects of IL-9 on the esophagus are unclear. Herein we aimed to understand the expression of the IL-9 receptor and effects of IL-9 on the epithelium in EoE. METHODS We used esophageal biopsies from pediatric EoE patients with active and inactive disease to analyze the expression of the IL-9 receptor, the adherens junction protein E-cadherin and the tight junction protein claudin-1. We treated primary human esophageal epithelial cells with IL-9 to understand its effects on E-cadherin expression and function. RESULTS Active EoE subjects had increased epithelial expression of IL-9 receptor mRNA and protein (P < 0.05) and decreased membrane bound E-cadherin (P < 0.01) and claudin-1 (P < 0.05) expression. IL-9 receptor expression and mislocalized claudin-1 positively correlated and while membrane bound E-cadherin expression negatively correlated with the degree of histologic epithelial remodeling (P < 0.05). IL-9 decreased epithelial resistance in stratified primary human esophageal epithelial cells (P < 0.01) and membrane bound E-cadherin in epithelial cell monolayers (P < 0.01). CONCLUSIONS These data suggest that IL-9, its receptor, and its effects on E-cadherin may be important mechanisms for epithelial barrier disruption in EoE.
Collapse
Affiliation(s)
- Ashmi Doshi
- Division of Allergy, Immunology, University of California, San Diego and Rady Children’s Hospital, San Diego
- Department of Pediatrics, University of California, San Diego and Rady Children’s Hospital, San Diego
- Department of Medicine, University of California, San Diego and Rady Children’s Hospital, San Diego
| | - Rebecca Khamishon
- Division of Allergy, Immunology, University of California, San Diego and Rady Children’s Hospital, San Diego
- Department of Pediatrics, University of California, San Diego and Rady Children’s Hospital, San Diego
- Department of Medicine, University of California, San Diego and Rady Children’s Hospital, San Diego
| | - Renee Rawson
- Division of Allergy, Immunology, University of California, San Diego and Rady Children’s Hospital, San Diego
- Department of Pediatrics, University of California, San Diego and Rady Children’s Hospital, San Diego
| | - Loan Duong
- Division of Allergy, Immunology, University of California, San Diego and Rady Children’s Hospital, San Diego
- Department of Pediatrics, University of California, San Diego and Rady Children’s Hospital, San Diego
| | - Lucas Dohil
- Division of Allergy, Immunology, University of California, San Diego and Rady Children’s Hospital, San Diego
- Department of Pediatrics, University of California, San Diego and Rady Children’s Hospital, San Diego
| | - Stephen J. Myers
- Division of Allergy, Immunology, University of California, San Diego and Rady Children’s Hospital, San Diego
- Department of Pediatrics, University of California, San Diego and Rady Children’s Hospital, San Diego
- Department of Medicine, University of California, San Diego and Rady Children’s Hospital, San Diego
| | - Braxton Bell
- Division of Allergy, Immunology, University of California, San Diego and Rady Children’s Hospital, San Diego
- Department of Pediatrics, University of California, San Diego and Rady Children’s Hospital, San Diego
| | - Ranjan Dohil
- Division of Gastroenterology, University of California, San Diego and Rady Children’s Hospital, San Diego
- Department of Pediatrics, University of California, San Diego and Rady Children’s Hospital, San Diego
| | - Robert O. Newbury
- Department of Pediatrics, University of California, San Diego and Rady Children’s Hospital, San Diego
- Department of Pathology, University of California, San Diego and Rady Children’s Hospital, San Diego
| | - Kim E. Barrett
- Division of Allergy, Immunology, University of California, San Diego and Rady Children’s Hospital, San Diego
- Division of Gastroenterology, University of California, San Diego and Rady Children’s Hospital, San Diego
- Department of Medicine, University of California, San Diego and Rady Children’s Hospital, San Diego
| | - Richard C. Kurten
- Department of Physiology & Biophysics, Arkansas Children’s Research Institute, University of Arkansas for Medical Sciences
| | - Seema S. Aceves
- Division of Allergy, Immunology, University of California, San Diego and Rady Children’s Hospital, San Diego
- Department of Pediatrics, University of California, San Diego and Rady Children’s Hospital, San Diego
- Department of Medicine, University of California, San Diego and Rady Children’s Hospital, San Diego
| |
Collapse
|