1
|
Sasahira M, Matsumoto H, Go TT, Yo S, Monden S, Ninomiya T, Oosawa M, Handa O, Umegaki E, Inoue R, Shiotani A. The Relationship Between Bacterial Flora in Saliva and Esophageal Mucus and Endoscopic Severity in Patients with Eosinophilic Esophagitis. Int J Mol Sci 2025; 26:3026. [PMID: 40243638 PMCID: PMC11989152 DOI: 10.3390/ijms26073026] [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: 02/05/2025] [Revised: 03/15/2025] [Accepted: 03/22/2025] [Indexed: 04/18/2025] Open
Abstract
Eosinophilic esophagitis (EoE) is a chronic inflammatory disease characterized by esophageal dysfunction and eosinophilic inflammation of the esophageal mucosa. In this study, we investigated the bacterial flora in saliva and esophageal mucus in patients with EoE and examined the relationship between EoE disease activity and mucosal cytokine expression, involving patients with active and inactive EoE (A-EoE and I-EoE, respectively). A-EoE was defined as a peak eosinophil count > 15/high-power field, according to the 2025 consensus guidelines. Saliva samples were collected from patients before the endoscopic examination. Brushing samples were collected from the distal esophagus of patients with EoE during endoscopic procedures. The degree of EoE inflammation was assessed using the EoE endoscopic reference score (EREFS). The samples were profiled using the Illumina MiSeq platform. The V3-V4 regions of the 16S rRNA gene (460 bp) were amplified using tailed PCR. Fifty-nine patients were enrolled, including eight with I-EoE, seventeen with A-EoE, and twenty-eight non-EoE controls. Major bacterial genera such as Streptococcus, Prevotella, Veillonella, and Haemophilus were detected in both the oral cavity and esophagus. Compared with the control group, the active EoE group had significantly more Prevotella spp. in the saliva and esophageal mucosa. Conversely, significantly fewer Neisseria spp. were found in the saliva and Streptococcus spp. in the esophageal mucosa of patients with active EoE. The EREFS of EoE and Streptococcus were inversely correlated. This study elucidated the characteristics of bacterial flora in the saliva and esophageal mucosa of patients with EoE.
Collapse
Affiliation(s)
- Momoyo Sasahira
- Department of Gastroenterology, Kawasaki Medical School, Okayama 701-0192, Japan; (M.S.); (T.T.G.); (S.Y.); (S.M.); (T.N.); (M.O.); (O.H.); (E.U.); (A.S.)
| | - Hiroshi Matsumoto
- Department of Gastroenterology, Kawasaki Medical School, Okayama 701-0192, Japan; (M.S.); (T.T.G.); (S.Y.); (S.M.); (T.N.); (M.O.); (O.H.); (E.U.); (A.S.)
| | - Tei Tei Go
- Department of Gastroenterology, Kawasaki Medical School, Okayama 701-0192, Japan; (M.S.); (T.T.G.); (S.Y.); (S.M.); (T.N.); (M.O.); (O.H.); (E.U.); (A.S.)
| | - Shogen Yo
- Department of Gastroenterology, Kawasaki Medical School, Okayama 701-0192, Japan; (M.S.); (T.T.G.); (S.Y.); (S.M.); (T.N.); (M.O.); (O.H.); (E.U.); (A.S.)
| | - Shuzo Monden
- Department of Gastroenterology, Kawasaki Medical School, Okayama 701-0192, Japan; (M.S.); (T.T.G.); (S.Y.); (S.M.); (T.N.); (M.O.); (O.H.); (E.U.); (A.S.)
| | - Takahirao Ninomiya
- Department of Gastroenterology, Kawasaki Medical School, Okayama 701-0192, Japan; (M.S.); (T.T.G.); (S.Y.); (S.M.); (T.N.); (M.O.); (O.H.); (E.U.); (A.S.)
| | - Motoyasu Oosawa
- Department of Gastroenterology, Kawasaki Medical School, Okayama 701-0192, Japan; (M.S.); (T.T.G.); (S.Y.); (S.M.); (T.N.); (M.O.); (O.H.); (E.U.); (A.S.)
| | - Osamu Handa
- Department of Gastroenterology, Kawasaki Medical School, Okayama 701-0192, Japan; (M.S.); (T.T.G.); (S.Y.); (S.M.); (T.N.); (M.O.); (O.H.); (E.U.); (A.S.)
| | - Eiji Umegaki
- Department of Gastroenterology, Kawasaki Medical School, Okayama 701-0192, Japan; (M.S.); (T.T.G.); (S.Y.); (S.M.); (T.N.); (M.O.); (O.H.); (E.U.); (A.S.)
| | - Ryo Inoue
- Faculty of Agriculture, Setsunan University, Hirakata-City 573-0101, Japan;
| | - Akiko Shiotani
- Department of Gastroenterology, Kawasaki Medical School, Okayama 701-0192, Japan; (M.S.); (T.T.G.); (S.Y.); (S.M.); (T.N.); (M.O.); (O.H.); (E.U.); (A.S.)
| |
Collapse
|
2
|
Barchi A, Massimino L, Mandarino FV, Vespa E, Sinagra E, Almolla O, Passaretti S, Fasulo E, Parigi TL, Cagliani S, Spanò S, Ungaro F, Danese S. Microbiota profiling in esophageal diseases: Novel insights into molecular staining and clinical outcomes. Comput Struct Biotechnol J 2024; 23:626-637. [PMID: 38274997 PMCID: PMC10808859 DOI: 10.1016/j.csbj.2023.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 01/27/2024] Open
Abstract
Gut microbiota is recognized nowadays as one of the key players in the development of several gastro-intestinal diseases. The first studies focused mainly on healthy subjects with staining of main bacterial species via culture-based techniques. Subsequently, lots of studies tried to focus on principal esophageal disease enlarged the knowledge on esophageal microbial environment and its role in pathogenesis. Gastro Esophageal Reflux Disease (GERD), the most widespread esophageal condition, seems related to a certain degree of mucosal inflammation, via interleukin (IL) 8 potentially enhanced by bacterial components, lipopolysaccharide (LPS) above all. Gram- bacteria, producing LPS), such as Campylobacter genus, have been found associated with GERD. Barrett esophagus (BE) seems characterized by a Gram- and microaerophils-shaped microbiota. Esophageal cancer (EC) development leads to an overturn in the esophageal environment with the shift from an oral-like microbiome to a prevalently low-abundant and low-diverse Gram--shaped microbiome. Although underinvestigated, also changes in the esophageal microbiome are associated with rare chronic inflammatory or neuropathic disease pathogenesis. The paucity of knowledge about the microbiota-driven mechanisms in esophageal disease pathogenesis is mainly due to the scarce sensitivity of sequencing technology and culture methods applied so far to study commensals in the esophagus. However, the recent advances in molecular techniques, especially with the advent of non-culture-based genomic sequencing tools and the implementation of multi-omics approaches, have revolutionized the microbiome field, with promises of implementing the current knowledge, discovering more mechanisms underneath, and giving insights into the development of novel therapies aimed to re-establish the microbial equilibrium for ameliorating esophageal diseases..
Collapse
Affiliation(s)
- Alberto Barchi
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Luca Massimino
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Edoardo Vespa
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Emanuele Sinagra
- Gastroenterology & Endoscopy Unit, Fondazione Istituto G. Giglio, Cefalù, Italy
| | - Omar Almolla
- Università Vita-Salute San Raffaele, Faculty of Medicine, Milan, Italy
| | - Sandro Passaretti
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Ernesto Fasulo
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Tommaso Lorenzo Parigi
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Faculty of Medicine, Milan, Italy
| | - Stefania Cagliani
- Università Vita-Salute San Raffaele, Faculty of Medicine, Milan, Italy
| | - Salvatore Spanò
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Federica Ungaro
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Silvio Danese
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Faculty of Medicine, Milan, Italy
| |
Collapse
|
3
|
Plate J, Bove M, Larsson HM, Norder Grusell E, Chatterjee N, Johansson LE, Bergquist H. Expression of inducible nitric oxide synthase, nitrotyrosine, eosinophilic peroxidase, eotaxin-3, and galectin-3 in patients with gastroesophageal reflux disease, eosinophilic esophagitis, and in healthy controls: a semiquantitative image analysis of 3,3'-diaminobenzidine-stained esophageal biopsies. Dis Esophagus 2024; 37:doae034. [PMID: 38679488 PMCID: PMC11360979 DOI: 10.1093/dote/doae034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 04/08/2024] [Indexed: 05/01/2024]
Abstract
Eosinophilic esophagitis (EoE) and gastroesophageal reflux disease (GERD) share many histopathological features; therefore, markers for differentiation are of diagnostic interest and may add to the understanding of the underlying mechanisms. The nitrergic system is upregulated in GERD and probably also in EoE. Esophageal biopsies of patients with EoE (n = 20), GERD (n = 20), and healthy volunteers (HVs) (n = 15) were exposed to antibodies against inducible nitric oxide synthase (iNOS), nitrotyrosine, eosinophilic peroxidase, eotaxin-3, and galectin-3. The stained object glasses were randomized, digitized, and blindly analyzed regarding the expression of DAB (3,3'-diaminobenzidine) by a protocol developed in QuPath software. A statistically significant overexpression of iNOS was observed in patients with any of the two inflammatory diseases compared with that in HVs. Eotaxin-3 could differentiate HVs versus inflammatory states. Gastroesophageal reflux patients displayed the highest levels of nitrotyrosine. Neither iNOS nor nitrotyrosine alone were able to differentiate between the two diseases. For that purpose, eosinophil peroxidase was a better candidate, as the mean levels increased stepwise from HVs via GERD to EoE. iNOS and nitrotyrosine are significantly overexpressed in patients with EoE and GERD compared with healthy controls, but only eosinophil peroxidase could differentiate the two types of esophagitis. The implications of the finding of the highest levels of nitrotyrosine among gastroesophageal reflux patients are discussed.
Collapse
Affiliation(s)
- John Plate
- Department of Otorhinolaryngology, Head and Neck Surgery, NU-Hospital Group, Region Västra Götaland, Trollhättan, Sweden
- Department of Otorhinolaryngology, Head and Neck Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mogens Bove
- Department of Otorhinolaryngology, Head and Neck Surgery, NU-Hospital Group, Region Västra Götaland, Trollhättan, Sweden
- Department of Otorhinolaryngology, Head and Neck Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Helen M Larsson
- Department of Otorhinolaryngology, Head and Neck Surgery, NU-Hospital Group, Region Västra Götaland, Trollhättan, Sweden
- Department of Otorhinolaryngology, Head and Neck Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Elisabeth Norder Grusell
- Department of Otorhinolaryngology, Head and Neck Surgery, Kungsbacka Hospital, Region Halland, Kungsbacka, Sweden
| | - Nabanita Chatterjee
- Core Facilities, Centre for Cellular Imaging (CCI), University of Gothenburg, Gothenburg, Sweden
| | - Leif E Johansson
- Department of ENT, Head and Neck Surgery, Skövde County Hospital, Region Västra Götaland, Skövde, Sweden
| | - Henrik Bergquist
- Department of Otorhinolaryngology, Head and Neck Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
4
|
Migliorisi G, Mastrorocco E, Dal Buono A, Gabbiadini R, Pellegatta G, Spaggiari P, Racca F, Heffler E, Savarino EV, Bezzio C, Repici A, Armuzzi A. Eosinophils, Eosinophilic Gastrointestinal Diseases, and Inflammatory Bowel Disease: A Critical Review. J Clin Med 2024; 13:4119. [PMID: 39064159 PMCID: PMC11278413 DOI: 10.3390/jcm13144119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/07/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Inflammatory bowel disease (IBD) and eosinophilic gastrointestinal diseases (EGIDs) are complex, multifactorial chronic inflammatory disorders affecting the gastrointestinal tract. Their epidemiology, particularly for eosinophilic esophagitis (EoE), is increasing worldwide, with a rise in the co-diagnosis of IBD and EGIDs. Both disorders share common risk factors, such as early exposure to antibiotics or specific dietary habits. Moreover, from a molecular perspective, eosinophilic infiltration is crucial in the diagnosis of eosinophilic disorders, and it also plays a pivotal role in IBD histological diagnosis. Indeed, recent evidence highlights the significant role of eosinophils in the health of the intestinal mucosal barrier and as mediators between innate and acquired immunity, even indicating a potential role in IBD pathogenesis. This narrative review aims to summarize the current evidence regarding the common clinical and molecular aspects of EGIDs and IBD and the current state of knowledge regarding overlap conditions and their pathogenesis. METHODS Pubmed was searched until May 2023 to assess relevant studies describing the epidemiology, pathophysiology, and therapy of EGIDs in IBD. RESULTS The immune pathways and mechanisms underlying both EGIDs and IBD remain partially known. An improved understanding of the role of eosinophils in overlapping conditions could lead to enhanced diagnostic precision, the development of more effective future therapeutic strategies, and a more accurate prediction of patient response. Consequently, the identification of red flags indicative of an eosinophilic disorder in IBD patients is of paramount importance and must be evaluated on a case-by-case basis.
Collapse
Affiliation(s)
- Giulia Migliorisi
- IBD Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy; (G.M.); (E.M.); (A.D.B.); (R.G.); (C.B.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (G.P.); (F.R.); (E.H.); (A.R.)
| | - Elisabetta Mastrorocco
- IBD Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy; (G.M.); (E.M.); (A.D.B.); (R.G.); (C.B.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (G.P.); (F.R.); (E.H.); (A.R.)
| | - Arianna Dal Buono
- IBD Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy; (G.M.); (E.M.); (A.D.B.); (R.G.); (C.B.)
| | - Roberto Gabbiadini
- IBD Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy; (G.M.); (E.M.); (A.D.B.); (R.G.); (C.B.)
| | - Gaia Pellegatta
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (G.P.); (F.R.); (E.H.); (A.R.)
- Endoscopic Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Paola Spaggiari
- Department of Pathology, Humanitas Research Hospital, 20089 Rozzano, Italy;
| | - Francesca Racca
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (G.P.); (F.R.); (E.H.); (A.R.)
- Personalized Medicine, Asthma and Allergy, IRCCS—Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Enrico Heffler
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (G.P.); (F.R.); (E.H.); (A.R.)
- Personalized Medicine, Asthma and Allergy, IRCCS—Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Edoardo Vincenzo Savarino
- Department of Surgery, Oncology and Gastroenterology, Department of Medical and Surgical Specialties, University of Padua, 35122 Padova, Italy;
| | - Cristina Bezzio
- IBD Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy; (G.M.); (E.M.); (A.D.B.); (R.G.); (C.B.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (G.P.); (F.R.); (E.H.); (A.R.)
| | - Alessandro Repici
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (G.P.); (F.R.); (E.H.); (A.R.)
- Endoscopic Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Alessandro Armuzzi
- IBD Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy; (G.M.); (E.M.); (A.D.B.); (R.G.); (C.B.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (G.P.); (F.R.); (E.H.); (A.R.)
| |
Collapse
|
5
|
Wrześniewska M, Wołoszczak J, Świrkosz G, Szyller H, Gomułka K. The Role of the Microbiota in the Pathogenesis and Treatment of Atopic Dermatitis-A Literature Review. Int J Mol Sci 2024; 25:6539. [PMID: 38928245 PMCID: PMC11203945 DOI: 10.3390/ijms25126539] [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: 04/09/2024] [Revised: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin condition with a high prevalence worldwide. AD pathogenesis is complex and consists of immune system dysregulation and impaired skin barrier, influenced by genetic and environmental factors. The purpose of the review is to show the complex interplay between atopic dermatitis and the microbiota. Human microbiota plays an important role in AD pathogenesis and the course of the disease. Dysbiosis is an important factor contributing to the development of atopic diseases, including atopic dermatitis. The gut microbiota can influence the composition of the skin microbiota, strengthening the skin barrier and regulating the immune response via the involvement of bacterial metabolites, particularly short-chain fatty acids, in signaling pathways of the gut-skin axis. AD can be modulated by antibiotic intake, dietary adjustments, hygiene, and living conditions. One of the promising strategies for modulating the course of AD is probiotics. This review offers a summary of how the microbiota influences the development and treatment of AD, highlighting aspects that warrant additional investigation.
Collapse
Affiliation(s)
- Martyna Wrześniewska
- Student Scientific Group of Internal Medicine and Allergology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.W.); (J.W.); (G.Ś.); (H.S.)
| | - Julia Wołoszczak
- Student Scientific Group of Internal Medicine and Allergology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.W.); (J.W.); (G.Ś.); (H.S.)
| | - Gabriela Świrkosz
- Student Scientific Group of Internal Medicine and Allergology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.W.); (J.W.); (G.Ś.); (H.S.)
| | - Hubert Szyller
- Student Scientific Group of Internal Medicine and Allergology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.W.); (J.W.); (G.Ś.); (H.S.)
| | - Krzysztof Gomułka
- Clinical Department of Internal Medicine, Pneumology and Allergology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland
| |
Collapse
|
6
|
Laky K, Frischmeyer-Guerrerio PA. Development and dysfunction of structural cells in eosinophilic esophagitis. J Allergy Clin Immunol 2024; 153:1485-1499. [PMID: 38849184 PMCID: PMC11626564 DOI: 10.1016/j.jaci.2024.04.006] [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: 12/18/2023] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 06/09/2024]
Abstract
Eosinophilic esophagitis (EoE) is a disorder characterized by dysfunction and chronic local inflammation of the esophagus. The incidence and prevalence of EoE are increasing worldwide. The mechanisms responsible are poorly understood, and effective treatment options are limited. From the lumen outward, the esophagus comprises stratified squamous epithelium, lamina propria, and muscle. The tissue-specific nature of EoE strongly suggests that structural cells in the esophagus are involved in the EoE diathesis. Epithelial basal cell hyperplasia and dilated intercellular spaces are cardinal features of EoE. Some patients with EoE develop lamina propria fibrosis, strictures, or esophageal muscle dysmotility. Clinical symptoms of EoE are only weakly correlated with peak eosinophil count, implying that other cell types contribute to EoE pathogenesis. Epithelial, endothelial, muscle, and fibroblast cells can each initiate inflammation and repair, regulate tissue resident immune cells, recruit peripheral leukocytes, and tailor adaptive immune cell responses. A better understanding of how structural cells maintain tissue homeostasis, respond to cell-intrinsic and cell-extrinsic stressors, and exacerbate and/or resolve inflammatory responses in the esophagus is needed. This knowledge will facilitate the development of more efficacious treatment strategies for EoE that can restore homeostasis of both hematopoietic and structural elements in the esophagus.
Collapse
Affiliation(s)
- Karen Laky
- Food Allergy Research Section, Laboratory of Allergic Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| | - Pamela A Frischmeyer-Guerrerio
- Food Allergy Research Section, Laboratory of Allergic Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| |
Collapse
|
7
|
Oliva S, McGowan EC. Associations of Eosinophilic Gastrointestinal Disorders with Other Gastrointestinal and Allergic Diseases. Immunol Allergy Clin North Am 2024; 44:329-348. [PMID: 38575227 DOI: 10.1016/j.iac.2024.01.005] [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] [Indexed: 04/06/2024]
Abstract
Eosinophilic gastrointestinal disorders (EGIDs) are becoming more common causing significant suffering and reduced quality of life. These conditions can affect different parts of the digestive system, either individually or in combination. Recognition of their link to allergic disorders or other gastrointestinal (GI) diseases has raised questions about their shared underlying mechanisms, which has had implications for diagnosis and management. The authors critically examine the current understanding of the connection between EGIDs and allergic conditions (ie, atopic dermatitis, allergic rhinitis, asthma, and food allergy) and GI diseases (ie, inflammatory bowel disease, celiac disease, gastroesophageal reflux disease, and motility disorders).
Collapse
Affiliation(s)
- Salvatore Oliva
- Department of Maternal and Child Health, Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy.
| | - Emily Clarke McGowan
- Division of Allergy and Immunology, Departments of Internal Medicine and Pediatrics, University of Virginia School of Medicine, P.O. Box 801355, Charlottesville, VA 22908, USA
| |
Collapse
|
8
|
Ravi A, Marietta EV, Alexander JA, Murray JA, Katzka DA. H influenzae LPS colocalization with Toll-like receptor 4 in eosinophilic esophagitis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2023; 2:100151. [PMID: 38024851 PMCID: PMC10679775 DOI: 10.1016/j.jacig.2023.100151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 05/24/2023] [Accepted: 06/16/2023] [Indexed: 12/01/2023]
Abstract
Background Patients with eosinophilic esophagitis (EoE) have a unique esophageal microbiome with increased presence of Haemophilus influenzae, but its role in the disease is unclear. Objective Microbiome-derived bacterial LPS activation of Toll-like receptors (TLR) is a potential mechanism for inducing inflammation in other chronic inflammatory diseases, but it has not been studied in EoE. Our aim was therefore to study microbiome-derived bacterial LPS activation of TLRs in EoE. Methods We studied 10 patients with active EoE, 9 patients with inactive EoE, and 10 control patients. Esophageal biopsy samples from the controls, patients with active EoE (>15 eosinophils/hpf), and patients with inactive EoE were immunostained for the presence of H influenzae LPS, presence of TLR4, and colocalization of LPS and TLR4. Staining intensity was measured by using confocal laser microscopy and scored on a scale from 0 to 3 as the average score assigned by 2 blinded observers. Results H influenzae LPS was detected by positive staining in 20 of the 29 patients (69.0%), including 9 of the 10 patients with active EoE (90.0%), 8 of the 9 patients with inactive EoE (89.9%), and 3 of the 10 controls (30%); its level was greater in the patients with active EoE than in the controls (P = .063). TLR4 was detected by positive staining in 19 of the 29 patients (65.5%), including 9 of the 10 patients with active EoE (90.0%), 4 of the 9 patients with inactive EoE (44.4%), and 6 of the 10 controls (60.0%); its level was higher in the patients with active EoE than in those with inactive EoE (P = .096). The result of testing for colocalization of LPS and TLR4 was positive in 8 of 10 patients with active EoE (80.0%), 1 of 9 patients with inactive EoE (11.1%), and 1 of 10 control patients (10.0%), with greater colocalization of H influenzae LPS and TLR4 staining density in the samples from patients with active EoE than in the controls or the patients with inactive EoE (P = .009 and P = .018, respectively). Conclusion Esophageal microbiome-rich H influenzae LPS colocalizes to TLR4 in active EoE. These data lend further support to a role for the esophageal microbiome in modulating the activity of EoE.
Collapse
Affiliation(s)
- Anupama Ravi
- Division of Pediatric Allergy, Mayo Clinic, Rochester
| | - Eric V. Marietta
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester
| | | | - Joseph A. Murray
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester
| | - David A. Katzka
- Division of Digestive and Liver Disease, Columbia University, New York
| |
Collapse
|
9
|
Morrison HA, Hoyt KJ, Mounzer C, Ivester HM, Barnes BH, Sauer B, McGowan EC, Allen IC. Expression profiling identifies key genes and biological functions associated with eosinophilic esophagitis in human patients. FRONTIERS IN ALLERGY 2023; 4:1239273. [PMID: 37692891 PMCID: PMC10484407 DOI: 10.3389/falgy.2023.1239273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/10/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction Eosinophilic Esophagitis (EoE) is a chronic allergic disease characterized by progressive inflammation of the esophageal mucosa. This chronic inflammatory disorder affects up to 50 per 100,000 individuals in the United States and Europe yet is limited in treatment options. While the transcriptome of EoE has been reported, few studies have examined the genetics among a cohort including both adult and pediatric EoE populations. To identify potentially overlooked biomarkers in EoE esophageal biopsies that may be promising targets for diagnostic and therapeutic development. Methods We used microarray analysis to interrogate gene expression using esophageal biopsies from EoE and Control subjects with a wide age distribution. Analysis of differential gene expression (DEGs) and prediction of impaired pathways was compared using conventional transcriptome analysis (TAC) and artificial intelligence-based (ADVAITA) programs. Principal Components Analysis revealed samples cluster by disease status (EoE and Control) irrespective of clinical features like sex, age, and disease severity. Results Global transcriptomic analysis revealed differential expression of several genes previously reported in EoE (CCL26, CPA3, POSTN, CTSC, ANO1, CRISP3, SPINK7). In addition, we identified differential expression of several genes from the MUC and SPRR families, which have been limited in previous reports. Discussion Our findings suggest that there is epithelial dysregulation demonstrated by DEGs that may contribute to impaired barrier integrity and loss of epidermal cell differentiation in EoE patients. These findings present two new gene families, SPRR and MUC, that are differentially expressed in both adult and pediatric EoE patients, which presents an opportunity for a future therapeutic target that would be useful in a large demographic of patients.
Collapse
Affiliation(s)
- Holly A. Morrison
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Kacie J. Hoyt
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Christina Mounzer
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Hannah M. Ivester
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Roanoke, VA, United States
| | - Barrett H. Barnes
- Division of Pediatric Gastroenterology/Nutrition, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Bryan Sauer
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Emily C. McGowan
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Irving C. Allen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Roanoke, VA, United States
- Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
| |
Collapse
|
10
|
Zhang X, Zhang N, Wang Z. Eosinophilic esophagitis and esophageal microbiota. Front Cell Infect Microbiol 2023; 13:1206343. [PMID: 37600943 PMCID: PMC10434796 DOI: 10.3389/fcimb.2023.1206343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
Eosinophilic esophagitis (EoE) is an antigen-mediated chronic inflammatory disease of the esophagus, the prevalence of which has steadily increased in recent years. The pathogenesis of EoE is not yet well-defined; however, recent studies have demonstrated that the esophageal microbiota is an essential regulator of physiological and pathological processes of EoE. Currently, research on EoE and microbiota is an emerging field of study that is receiving increasing attention. Here, we review existing EoE-related esophageal microbiota studies to explore the potential mechanisms underlying esophageal microbiota-mediated EoE. The esophageal microbiome is altered in patients with EoE. Although α diversity is usually not significantly different, an increase in Haemophilus and a decrease in Firmicutes were observed in EoE patients. The role of microbiota in initiating and perpetuating inflammation is not fully understood. Current evidence suggests that the penetration of microbiota leads to the activation of epithelial cells as well as innate and adaptive immune cells, with the subsequent release of cytokines, leading to immune responses and inflammation. The involvement of toll-like receptors in EoE also supports the potential role of the microbiota in the progression of this disease. While EoE-induced inflammation can also lead to alterations in the local microbiome. Moreover, dietary modifications, proton pump inhibitors, and corticosteroids can modulate the esophageal microbiota; however, definitive conclusions about the alterations of microbes after treatment cannot be drawn. These findings provide promising avenues for future studies.
Collapse
Affiliation(s)
- Xiaohan Zhang
- Microbiota Division, Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School, Nankai University, Tianjin, China
| | - Nana Zhang
- Microbiota Division, Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zikai Wang
- Microbiota Division, Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
11
|
Chang JW, Jensen ET, Dellon ES. Nature with Nurture: the Role of Intrinsic Genetic and Extrinsic Environmental Factors on Eosinophilic Esophagitis. Curr Allergy Asthma Rep 2022; 22:163-170. [PMID: 36190688 PMCID: PMC10838151 DOI: 10.1007/s11882-022-01042-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW As the rising prevalence and incidence of eosinophilic esophagitis (EoE) has quickly outpaced the rate of esophageal biopsies, particularly in Westernized countries, several studies have suggested a link between intrinsic genetic and extrinsic environmental risk factors and the development, presentation, and diagnosis of EoE. This review aims to critically assess existing studies describing the role of the environment on the development, symptomatic presentation, and diagnosis of this recently recognized chronic immune-mediated disease. RECENT FINDINGS We present and critically evaluate the working hypotheses and supportive studies thus far on environmental factors on EoE, describe sources of potential bias in diagnosis due to socioeconomic factors and thus undermining studies of EoE etiology, and highlight opportunities for future research. As genetics alone do not explain the rapid rise of EoE, we must look to environmental, or extrinsic, factors both in the early-life period which shape the development of the gut microbiome, as well as later life contributing to diagnosis of this new disease. Future etiologic studies linking risk factors to EoE development in individual patients are needed.
Collapse
Affiliation(s)
- Joy W Chang
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Elizabeth T Jensen
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Evan S Dellon
- Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
12
|
Fang Y, Yan C, Zhao Q, Zhao B, Liao Y, Chen Y, Wang D, Tang D. The Association Between Gut Microbiota, Toll-Like Receptors, and Colorectal Cancer. Clin Med Insights Oncol 2022; 16:11795549221130549. [PMID: 36338264 PMCID: PMC9634190 DOI: 10.1177/11795549221130549] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 09/16/2022] [Indexed: 11/05/2022] Open
Abstract
The large number of microbes found in the gut are involved in various critical biological processes in the human body and have dynamic and complex interactions with the immune system. Disruptions in the host's gut microbiota and the metabolites produced during fermentation promote the development of intestinal inflammation and colorectal cancer (CRC). Toll-like receptors (TLRs) recognize specific microbial-associated molecular patterns specific to microorganisms whose signaling is involved in maintaining intestinal homeostasis or, under certain conditions, mediating dysbiosis-associated intestinal inflammation. The signaling pathways of TLRs are described first, followed by a discussion of the interrelationship between gut microbes and TLRs, including the activation of TLRs by gut microbes and the effect of TLRs on the distribution of gut microbiota, particularly the role of microbes in colorectal carcinogenesis via TLRs. Finally, we discuss the potential roles of various TLRs in colorectal cancer.
Collapse
Affiliation(s)
- Yongkun Fang
- Department of General Surgery,
Institute of General Surgery, Northern Jiangsu People’s Hospital, Clinical Medical
College, Yangzhou University, Yangzhou, China
| | - Cheng Yan
- Department of Clinical Medical College,
Dalian Medical University, Dalian, China
- The People’s Hospital Of QianNan,
Duyun, China
| | - Qi Zhao
- Department of Clinical Medicine,
Clinical Medical College, Yangzhou University, Yangzhou, China
- Changshu No.2 People’s Hospital,
Suzhou, China
| | - Bin Zhao
- Department of Clinical Medical College,
Dalian Medical University, Dalian, China
| | - Yiqun Liao
- Department of Clinical Medical College,
Dalian Medical University, Dalian, China
| | - Yuji Chen
- Department of Clinical Medicine,
Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Daorong Wang
- Department of General Surgery,
Institute of General Surgery, Northern Jiangsu People’s Hospital, Clinical Medical
College, Yangzhou University, Yangzhou, China
| | - Dong Tang
- Department of General Surgery,
Institute of General Surgery, Northern Jiangsu People’s Hospital, Clinical Medical
College, Yangzhou University, Yangzhou, China
| |
Collapse
|
13
|
Angerami Almeida K, de Queiroz Andrade E, Burns G, Hoedt EC, Mattes J, Keely S, Collison A. The microbiota in eosinophilic esophagitis: A systematic review. J Gastroenterol Hepatol 2022; 37:1673-1684. [PMID: 35730344 PMCID: PMC9544137 DOI: 10.1111/jgh.15921] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/14/2022] [Accepted: 06/14/2022] [Indexed: 12/12/2022]
Abstract
Eosinophilic esophagitis (EoE) is an atopic disease of the esophagus that has shown a significant increase in incidence and prevalence in the last 20 years. The etiology of EoE is unclear, and few studies explore the esophageal microbiota in EoE. The local microbiome has been implicated in the pathogenesis of several allergic and inflammatory diseases, such as asthma and eczema. In this study, we performed a systematic review to evaluate differences in the microbiota profile of patients with EoE compared with controls. MEDLINE, Embase, Cochrane Library, Scopus, and CINAHL (Cumulative Index to Nursing and Allied Health Literature) databases were searched to identify studies investigating the microbiota composition in EoE. Three reviewers screened the articles for eligibility and quality. Seven articles underwent full-text review, and a narrative synthesis was undertaken. The microbiota of the mouth and esophagus are correlated. Patients with active EoE present increased esophageal microbial load and increased abundance in particular species, such as Haemophilus and Aggregatibacter. On the other hand, EoE patients present a decrease in Firmicutes. High microbial load and abundance of Haemophilus are observed in EoE patients, but little evidence exists to demonstrate their influence on inflammation and disease. Understanding microbial signatures in EoE might contribute to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Kaylani Angerami Almeida
- School of Medicine and Public HealthUniversity of NewcastleNewcastleNew South WalesAustralia,Priority Research Centre GrowUpWell, Hunter Medical Research InstituteUniversity of NewcastleNewcastleNew South WalesAustralia,Viruses, Infection, Immunity, Vaccine and Asthma (VIVA) ProgramHunter Medical Research Institute (HMRI)New Lambton HeightsNew South WalesAustralia,NHMRC Centre of Research Excellence (CRE) in Digestive HealthThe University of NewcastleCallaghanNew South WalesAustralia
| | - Ediane de Queiroz Andrade
- School of Medicine and Public HealthUniversity of NewcastleNewcastleNew South WalesAustralia,Priority Research Centre GrowUpWell, Hunter Medical Research InstituteUniversity of NewcastleNewcastleNew South WalesAustralia,Viruses, Infection, Immunity, Vaccine and Asthma (VIVA) ProgramHunter Medical Research Institute (HMRI)New Lambton HeightsNew South WalesAustralia
| | - Grace Burns
- NHMRC Centre of Research Excellence (CRE) in Digestive HealthThe University of NewcastleCallaghanNew South WalesAustralia,School of Biomedical Sciences and Pharmacy, College of Health, Medicine and WellbeingThe University of NewcastleCallaghanNew South WalesAustralia
| | - Emily C Hoedt
- NHMRC Centre of Research Excellence (CRE) in Digestive HealthThe University of NewcastleCallaghanNew South WalesAustralia,School of Biomedical Sciences and Pharmacy, College of Health, Medicine and WellbeingThe University of NewcastleCallaghanNew South WalesAustralia
| | - Joerg Mattes
- School of Medicine and Public HealthUniversity of NewcastleNewcastleNew South WalesAustralia,Priority Research Centre GrowUpWell, Hunter Medical Research InstituteUniversity of NewcastleNewcastleNew South WalesAustralia,Viruses, Infection, Immunity, Vaccine and Asthma (VIVA) ProgramHunter Medical Research Institute (HMRI)New Lambton HeightsNew South WalesAustralia
| | - Simon Keely
- NHMRC Centre of Research Excellence (CRE) in Digestive HealthThe University of NewcastleCallaghanNew South WalesAustralia,School of Biomedical Sciences and Pharmacy, College of Health, Medicine and WellbeingThe University of NewcastleCallaghanNew South WalesAustralia
| | - Adam Collison
- School of Medicine and Public HealthUniversity of NewcastleNewcastleNew South WalesAustralia,Priority Research Centre GrowUpWell, Hunter Medical Research InstituteUniversity of NewcastleNewcastleNew South WalesAustralia,Viruses, Infection, Immunity, Vaccine and Asthma (VIVA) ProgramHunter Medical Research Institute (HMRI)New Lambton HeightsNew South WalesAustralia
| |
Collapse
|
14
|
Aloi M, D'Arcangelo G, Rossetti D, Bucherini S, Felici E, Romano C, Martinelli M, Dipasquale V, Lionetti P, Oliva S. Occurrence and Clinical Impact of Eosinophilic Esophagitis in a Large Cohort of Children With Inflammatory Bowel Disease. Inflamm Bowel Dis 2022:6658537. [PMID: 35942651 DOI: 10.1093/ibd/izac172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND Scarce data have investigated the association between pediatric inflammatory bowel disease (IBD) and eosinophilic esophagitis (EoE). We, therefore, aimed to describe the epidemiology and the possible peculiar phenotype and natural history of such an association. METHODS Case-control study is based on the Italian Society for Pediatric Gastroenterology (SIGENP) national registry. All children with a combined diagnosis of IBD and EoE were included. The overall prevalence and incidence in 2 periods, 2009 to 2015, and 2016 to 2021, were calculated. Cases were matched with IBD only and EoE only patients in a 1:3:3 ratio. Phenotype and outcomes (courses of steroids, risk of complications, surgery, treatment escalation, and hospitalization) were compared between groups. RESULTS Eleven patients (age 11.2 ± 2.8 years, Males 91%) with EoE-IBD out of 3090 patients with IBD were identified, resulting in an overall prevalence of 0.35% and an incidence of 0.18% for 2009 to 2015 and 0.45% for 2016 to 2021. Treatment escalation rates for IBD were significantly higher in patients with IBD compared with EoE-IBD at 12- and 24-month follow-up (0% vs 30%, P = .04; and 9% vs 45.5%, P = .03, respectively). Furthermore, patients with IBD were at a significantly higher risk of hospitalization than both EoE-IBD and EoE patients (log rank P < .001). We found no significant differences in major outcomes related to the EoE course in EoE-IBD patients compared with EoE ones. CONCLUSIONS The incidence and prevalence of EoE in children with IBD are low, although the incidence seems to be rising in recent years. Having EoE appears to be associated with a milder IBD disease course, whereas having IBD does not seem to affect the natural history of EoE. More data are needed to better define the phenotype of such association.
Collapse
Affiliation(s)
- Marina Aloi
- Sapienza University of Rome - Umberto I Hospital, Rome, Italy
| | | | - Danilo Rossetti
- Sapienza University of Rome - Umberto I Hospital, Rome, Italy
| | | | - Enrico Felici
- Sapienza University of Rome - Umberto I Hospital, Rome, Italy
| | - Claudio Romano
- Sapienza University of Rome - Umberto I Hospital, Rome, Italy
| | | | | | - Paolo Lionetti
- Sapienza University of Rome - Umberto I Hospital, Rome, Italy
| | - Salvatore Oliva
- Sapienza University of Rome - Umberto I Hospital, Rome, Italy
| |
Collapse
|
15
|
Ghisa M, Savarino V, Buda A, Katzka DA, Savarino E. Toward a potential association between eosinophilic esophagitis and Klinefelter syndrome: a case series and review of the literature. Therap Adv Gastroenterol 2022; 15:17562848221076888. [PMID: 35186122 PMCID: PMC8855470 DOI: 10.1177/17562848221076888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 12/27/2021] [Indexed: 02/04/2023] Open
Abstract
Klinefelter syndrome (KS) is a sex aneuploidy abnormality comprised by one additional X chromosome. It occurs in 1:500-1000 male births. As with women, an increased susceptibility to autoimmune diseases is present. We report three cases of coexisting EoE and KS for a prevalence of 2% in our EoE clinic. Possible changes in gene expression in KS are reviewed, some of which may be related to activation of genes located on the X chromosome. We postulate that these X-activated genes in patients with KS yield a greater likelihood of developing EoE because of their genetic predisposition to autoimmune diseases.
Collapse
Affiliation(s)
- Matteo Ghisa
- Gastroenterology Unit, Department of Surgery,
Oncology and Gastroenterology, University of Padua, Padua, Italy
- Gastroenterology Unit, Department of
Oncological Gastrointestinal Surgery, S. Maria del Prato Hospital, Feltre,
Italy
| | - Vincenzo Savarino
- Gastroenterology Unit, Department of Internal
Medicine, University of Genoa, IRCCS Ospedale Policlinico San Martino,
Genoa, Italy
| | - Andrea Buda
- Gastroenterology Unit, Department of
Oncological Gastrointestinal Surgery, S. Maria del Prato Hospital, Feltre,
Italy
| | - David A. Katzka
- Division of Gastroenterology and Hepatology,
Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
16
|
Lucendo AJ, Santander C, Savarino E, Guagnozzi D, Pérez-Martínez I, Perelló A, Guardiola-Arévalo A, Barrio J, Elena Betoré-Glaria M, Gutiérrez-Junquera C, Ciriza de los Ríos C, Racca F, Fernández-Fernández S, Blas-Jhon L, Lund Krarup A, de la Riva S, Naves JE, Carrión S, Rodríguez Oballe JA, García-Morales N, Tamarit-Sebastián S, Navarro P, Arias Á, Laserna-Mendieta EJ, Casabona-Francés S, Pérez-Fernández T, Llerena Castro R, Ghisa M, Manie D, Pellegatta G, Suárez A, Alcedo J, Gil Simón P, Teresa Palomeque M, Asensio T, Granja-Navacerrada A, de Mendoza Guena LH, Rodríguez Sánchez A, Masiques Mas L, Dainese R, Feo-Ortega S. EoE CONNECT, the European Registry of Clinical, Environmental, and Genetic Determinants in Eosinophilic Esophagitis: rationale, design, and study protocol of a large-scale epidemiological study in Europe. Therap Adv Gastroenterol 2022; 15:17562848221074204. [PMID: 35126668 PMCID: PMC8814964 DOI: 10.1177/17562848221074204] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/27/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The growing prevalence of eosinophilic esophagitis (EoE) represents a considerable burden to patients and health care systems. Optimizing cost-effective management and identifying mechanisms for disease onset and progression are required. However, the paucity of large patient cohorts and heterogeneity of practice hinder the defining of optimal management of EoE. METHODS EoE CONNECT is an ongoing, prospective registry study initiated in 2016 and currently managed by EUREOS, the European Consortium for Eosinophilic Diseases of the Gastrointestinal Tract. Patients are managed and treated by their responsible specialists independently. Data recorded using a web-based system include demographic and clinical variables; patient allergies; environmental, intrapartum, and early life exposures; and family background. Symptoms are structurally assessed at every visit; endoscopic features and histological findings are recorded for each examination. Prospective treatment data are registered sequentially, with new sequences created each time a different treatment (active principle, formulation, or dose) is administered to a patient. EoE CONNECT database is actively monitored to ensure the highest data accuracy and the highest scientific and ethical standards. RESULTS EoE CONNECT is currently being conducted at 39 centers in Europe and enrolls patients of all ages with EoE. In its aim to increase knowledge, to date EoE CONNECT has provided evidence on the effectiveness of first- and second-line therapies for EoE in clinical practice, the ability of proton pump inhibitors to induce disease remission, and factors associated with improved response. Drug effects to reverse fibrous remodeling and endoscopic features of fibrosis in EoE have also been assessed. CONCLUSION This prospective registry study will provide important information on the epidemiological and clinical aspects of EoE and evidence as to the real-world and long-term effectiveness and safety of therapy. These data will potentially be a vital benchmark for planning future EoE health care services in Europe.
Collapse
Affiliation(s)
| | - Cecilio Santander
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain,Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain,Department of Gastroenterology, Hospital Universitario La Princesa, Madrid, Spain
| | - Edoardo Savarino
- Department of Surgery, Oncology and Gastroenterology, Università di Padova, Padova, Italy
| | - Danila Guagnozzi
- Department of Gastroenterology, Hospital Universitario Vall d’Hebron, Barcelona, Spain,Digestive System Research Unit, Unitat de Fisiología I Fisiopatología Digestiva, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Isabel Pérez-Martínez
- Department of Gastroenterology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Antonia Perelló
- Department of Gastroenterology, Hospital de Viladecans, Barcelona, Spain
| | - Antonio Guardiola-Arévalo
- Department of Gastroenterology, Hospital Universitario de Fuenlabrada, Fuenlabrada, Spain; Instituto de Investigación Sanitaria La Paz (IdiPAZ), Madrid, Spain
| | - Jesús Barrio
- Department of Gastroenterology, Hospital Universitario Río Hortega, Valladolid, Spain
| | | | | | - Constanza Ciriza de los Ríos
- Department of Gastroenterology, Hospital Clínico San Carlos, Universidad Complutense, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Francesca Racca
- Personalized Medicine, Asthma and Allergy Clinic, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Sonia Fernández-Fernández
- Pediatric Gastroenterology, Hepatology and Nutrition, University Hospital Severo Ochoa, Leganés, Spain
| | - Leonardo Blas-Jhon
- Department of Gastroenterology, University Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Anne Lund Krarup
- Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark,Department of Acute Medicine and Trauma Care, Aalborg University Hospital, Aalborg, Denmark,Faculty of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Susana de la Riva
- Department of Gastroenterology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Juan E. Naves
- Gastroenterology Unit, Hospital Universitari Germans Trias i Pujol, CIBERehd, Badalona, Spain
| | - Silvia Carrión
- Gastrointestinal Motility Laboratory, Department of Surgery, Hospital de Mataró, Consorci Sanitari del Maresme, Mataró, Spain
| | - Juan Armando Rodríguez Oballe
- Department of Gastroenterology, University Hospital Santa María and University Hospital Arnau de Vilanova, Lleida, Spain
| | | | - Sonsoles Tamarit-Sebastián
- Department of Gastroenterology, Hospital General de Tomelloso, Tomelloso, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Spain
| | - Pilar Navarro
- Department of Gastroenterology, Hospital General de Tomelloso, Tomelloso, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Spain
| | - Ángel Arias
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain,Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain,Research Unit, Hospital General Mancha Centro, Alcázar de San Juan, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Spain
| | - Emilio J. Laserna-Mendieta
- Department of Gastroenterology, Hospital General de Tomelloso, Tomelloso, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain,Clinical Laboratory, Hospital Universitario de La Princesa, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Gergen AK, Jarrett MJ, Li A, Meng X, Pratap A, Fullerton DA, Weyant MJ. Toll-like Receptor 4 Mediates Reflux-Induced Inflammation in a Murine Reflux Model. Semin Thorac Cardiovasc Surg 2021; 34:1324-1335. [PMID: 34534678 DOI: 10.1053/j.semtcvs.2021.07.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/01/2021] [Indexed: 01/04/2023]
Abstract
Dysregulation of toll-like receptor (TLR) signaling within the gastrointestinal epithelium has been associated with uncontrolled inflammation and tumorigenesis. We sought to evaluate the role of TLR4 in the development of gastroesophageal reflux-mediated inflammation and mucosal changes of the distal esophagus. Verified human esophageal Barrett's cells with high grade dysplasia (CPB) and esophageal adenocarcinoma cells (OE33) were treated with deoxycholic acid for 24 hours. Cells were pretreated with a TLR4-specific inhibitor peptide 2 hours prior to deoxycholic acid treatment. Inflammatory markers were evaluated using immunoblotting and enzyme-linked immunosorbent assay. A surgical reflux mouse model was generated by performing a side-to-side anastomosis between the second portion of the duodenum and the gastroesophageal junction. Control animals underwent laparotomy with incision and closure of the esophagus superior to the gastroesophageal junction (sham procedure). Esophageal sections were evaluated using hematoxylin and eosin staining and immunohistochemistry. Deoxycholic acid increased expression of inflammatory markers including intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and interleukin 8. Pretreatment with a TLR4 inhibitor significantly decreased deoxycholic acid-induced inflammatory marker expression. C3H/HeNCrl mice demonstrated a significant increase in mucosal hyperplasia and proliferation following DGEA compared to sham procedure. TLR4 mutant mice (C3H/HeJ) undergoing DGEA demonstrated an attenuated hyperplastic and proliferative response compared to C3H/HeNCrl mice. Inhibition of TLR4 signaling attenuates reflux-induced inflammation in vivo. These findings identify TLR4 inhibition as a potential therapeutic target to halt the progression of pathologic esophageal changes developing in the setting of chronic gastroesophageal reflux disease.
Collapse
Affiliation(s)
- Anna K Gergen
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado.
| | - Michael J Jarrett
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Anqi Li
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Xianzhong Meng
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Akshay Pratap
- Department of Surgery, Division of GI, Trauma, and Endocrine Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - David A Fullerton
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Michael J Weyant
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
18
|
Nortunen M, Väkiparta N, Porvari K, Saarnio J, Karttunen TJ, Huhta H. Pathophysiology of reflux oesophagitis: role of Toll-like receptors 2 and 4 and Farnesoid X receptor. Virchows Arch 2021; 479:285-293. [PMID: 33686512 PMCID: PMC8364528 DOI: 10.1007/s00428-021-03066-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 01/15/2021] [Accepted: 02/18/2021] [Indexed: 11/28/2022]
Abstract
The pathogenesis of gastroesophageal reflux disease (GERD) is not fully understood. It involves the activation of mucosal immune-mediated and inflammatory responses. Toll-like receptors (TLR) 2 and TLR4 are pattern-recognition receptors of the innate immune system; they recognize microbial and endogenous ligands. Farnesoid X receptor (FXR) is a bile acid receptor that regulates the inflammatory response. We aimed to evaluate TLR2, TLR4 and FXR expression patterns in GERD. We re-evaluated 84 oesophageal biopsy samples according to the global severity (GS) score, including 26 cases with histologically normal oesophagus, 28 with histologically mild oesophagitis and 30 with severe oesophagitis. We used immunohistochemistry and in situ hybridization to assess the expression patterns of TLR2, TLR4 and FXR in oesophageal squamous cells. Immunohistochemistry showed that nuclear and cytoplasmic TLR2 was expressed predominantly in the basal layer of normal oesophageal epithelium. In oesophagitis, TLR2 expression increased throughout the epithelium, and the superficial expression was significantly more intensive compared to normal epithelium, p <0.01. Nuclear and cytoplasmic TLR4 was expressed throughout the thickness of squamous epithelium, with no change in oesophagitis. FXR was expressed in the nuclei of squamous cells, and the intensity of the expression increased significantly in oesophagitis (p <0.05). FXR expression correlated with basal TLR2. In situ hybridization confirmed the immunohistochemical expression patterns of TLR2 and TLR4. In GERD, TLR2, but not TLR4, expression was upregulated which indicates that innate immunity is activated according to a specific pattern in GERD. FXR expression was increased in GERD and might have a regulatory connection to TLR2.
Collapse
Affiliation(s)
- Minna Nortunen
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, 90014, Oulu, Finland.
- Research Unit of Surgery, Anesthesia and Intensive Care, University of Oulu, Oulu, Finland.
- Department of Surgery, Oulu University Hospital and Medical Research Center Oulu, Oulu, Finland.
| | - Nina Väkiparta
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, 90014, Oulu, Finland
| | - Katja Porvari
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, 90014, Oulu, Finland
| | - Juha Saarnio
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, 90014, Oulu, Finland
- Research Unit of Surgery, Anesthesia and Intensive Care, University of Oulu, Oulu, Finland
- Department of Surgery, Oulu University Hospital and Medical Research Center Oulu, Oulu, Finland
| | - Tuomo J Karttunen
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, 90014, Oulu, Finland
| | - Heikki Huhta
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, 90014, Oulu, Finland
- Research Unit of Surgery, Anesthesia and Intensive Care, University of Oulu, Oulu, Finland
- Department of Surgery, Oulu University Hospital and Medical Research Center Oulu, Oulu, Finland
| |
Collapse
|
19
|
Martinez-Carrasco R, Argüeso P, Fini ME. Membrane-associated mucins of the human ocular surface in health and disease. Ocul Surf 2021; 21:313-330. [PMID: 33775913 PMCID: PMC8328898 DOI: 10.1016/j.jtos.2021.03.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/02/2021] [Accepted: 03/08/2021] [Indexed: 02/06/2023]
Abstract
Mucins are a family of high molecular weight, heavily-glycosylated proteins produced by wet epithelial tissues, including the ocular surface epithelia. Densely-packed O-linked glycan chains added post-translationally confer the biophysical properties of hydration, lubrication, anti-adhesion and repulsion. Membrane-associated mucins (MAMs) are the distinguishing components of the mucosal glycocalyx. At the ocular surface, MAMs maintain wetness, lubricate the blink, stabilize the tear film, and create a physical barrier to the outside world. In addition, it is increasingly appreciated that MAMs function as cell surface receptors that transduce information from the outside to the inside of the cell. Recently, our team published a comprehensive review/perspectives article for molecular scientists on ocular surface MAMs, including previously unpublished data and analyses on two new genes MUC21 and MUC22, as well as new MAM functions and biological roles, comparing human and mouse (PMID: 31493487). The current article is a refocus for the audience of The Ocular Surface. First, we update the gene and protein information in a more concise form, and include a new section on glycosylation. Next, we discuss biological roles, with some new sections and further updating from our previous review. Finally, we provide a new chapter on MAM involvement in ocular surface disease. We end this with discussion of an emerging mechanism responsible for damage to the epithelia and their mucosal glycocalyces: the unfolded protein response (UPR). The UPR offers a novel target for therapeutic intervention.
Collapse
Affiliation(s)
- Rafael Martinez-Carrasco
- Department of Ophthalmology, Tufts University School of Medicine at New England Eye Center, Tufts Medical Center, Boston, MA, 02111, USA.
| | - Pablo Argüeso
- Department of Ophthalmology, Harvard Medical School at Schepens Eye Research Institute of Mass, Eye and Ear, Boston, MA, 02114, USA.
| | - M Elizabeth Fini
- Department of Ophthalmology, Tufts University School of Medicine at New England Eye Center, Tufts Medical Center: Program in Pharmacology & Drug Development, Graduate School of Biomedical Sciences, Tufts University, Boston, MA, O2111, USA.
| |
Collapse
|
20
|
Sekai I, Watanabe T, Yoshikawa K, Takada R, Hara A, Yoshikawa T, Kamata K, Minaga K, Kudo M. A case with eosinophilic gastroenteritis exhibiting enhanced TNF-α and IL-6 responses. Clin J Gastroenterol 2021; 14:511-516. [PMID: 33400188 DOI: 10.1007/s12328-020-01320-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 12/08/2020] [Indexed: 11/26/2022]
Abstract
Eosinophilic gastroenteritis (EGE) is a chronic allergic disorder characterized by infiltration of eosinophils in the gastrointestinal (GI) tract and hypereosinophilia. Although T helper type 2 (Th2) responses play pathogenic roles in EGE, roles of innate immunity cytokines including IL-6 and TNF-α have been poorly defined. Here, we describe a case of EGE exhibiting accumulation of eosinophils in the upper GI mucosa and hypereosinophilia. Induction of remission by prednisolone reduced expression levels not only of Th2 cytokines but also of IL-6 and TNF-α in the GI mucosa. Moreover, induction of remission was accompanied by a marked reduction in serum levels of chemokine C-C motif ligand 17 (CCL17, TARC), IL-6 and TNF-α, implicating that both Th2 and innate immune responses were involved in the development of EGE in this case. Collectively, this case suggests possible involvement of IL-6 and TNF-α in the development of EGE.
Collapse
Affiliation(s)
- Ikue Sekai
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Tomohiro Watanabe
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan.
| | - Keisuke Yoshikawa
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Ryutaro Takada
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Akane Hara
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Tomoe Yoshikawa
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Ken Kamata
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Kosuke Minaga
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| |
Collapse
|
21
|
Relationship of the Esophageal Microbiome and Tissue Gene Expression and Links to the Oral Microbiome: A Randomized Clinical Trial. Clin Transl Gastroenterol 2020; 11:e00235. [PMID: 33512805 PMCID: PMC7721221 DOI: 10.14309/ctg.0000000000000235] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Although the microbiome is altered in various esophageal diseases, there is no direct evidence for a link between the oral or esophageal microbiome and underlying esophageal tissue. Here, we aimed to address these gaps through use of an antimicrobial mouth rinse to modify the esophageal microbiome and tissue gene expression. METHODS In this randomized controlled trial, patients scheduled to undergo endoscopy for clinical indications used chlorhexidine mouth rinse or no treatment for 2 weeks before endoscopy. Oral swabs and saliva were collected at baseline and at follow-up, and the esophagus was sampled on the day of endoscopy. The microbiome was analyzed by 16S rRNA gene sequencing, and esophageal tissue gene expression was ascertained by RNA-Seq. RESULTS Twenty subjects were enrolled and included in the analyses. Within individuals, the oral and esophageal microbiome composition was significantly correlated. Chlorhexidine treatment associated with significant alterations to the relative abundance of several esophageal bacterial taxa, and to expression of genes in the esophagus including reductions in periostin, claudin-18, chemokines CXCL1 and CXCL13, and several members of the tumor necrosis factor receptor superfamily. A taxon in genus Haemophilus in the esophagus also associated with significant changes in tissue gene expression. DISCUSSION The oral and esophageal microbiomes are closely related within individuals, and esophageal microbiome alterations correlate with tissue gene expression changes. The esophageal microbiome may act as an important cofactor that influences pathogenesis and outcomes of diseases such as eosinophilic esophagitis, gastroesophageal reflux, and Barrett's esophagus.
Collapse
|
22
|
Doyle AD, Masuda MY, Kita H, Wright BL. Eosinophils in Eosinophilic Esophagitis: The Road to Fibrostenosis is Paved With Good Intentions. Front Immunol 2020; 11:603295. [PMID: 33335531 PMCID: PMC7736408 DOI: 10.3389/fimmu.2020.603295] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 11/03/2020] [Indexed: 12/15/2022] Open
Abstract
Eosinophilic esophagitis (EoE) is an antigen-driven disease associated with epithelial barrier dysfunction and chronic type 2 inflammation. Eosinophils are the defining feature of EoE histopathology but relatively little is known about their role in disease onset and progression. Classically defined as destructive, end-stage effector cells, eosinophils (a resident leukocyte in most of the GI tract) are increasingly understood to play roles in local immunity, tissue homeostasis, remodeling, and repair. Indeed, asymptomatic esophageal eosinophilia is observed in IgE-mediated food allergy. Interestingly, EoE is a potential complication of oral immunotherapy (OIT) for food allergy. However, we recently found that patients with peanut allergy may have asymptomatic esophageal eosinophilia at baseline and that peanut OIT induces transient esophageal eosinophilia in most subjects. This is seemingly at odds with multiple studies which have shown that EoE disease severity correlates with tissue eosinophilia. Herein, we review the potential role of eosinophils in EoE at different stages of disease pathogenesis. Based on current literature we suggest the following: (1) eosinophils are recruited to the esophagus as a homeostatic response to epithelial barrier disruption; (2) eosinophils mediate barrier-protective activities including local antibody production, mucus production and epithelial turnover; and (3) when type 2 inflammation persists, eosinophils promote fibrosis.
Collapse
Affiliation(s)
- Alfred D Doyle
- Division of Allergy, Asthma, and Clinical Immunology, Department of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, United States
| | - Mia Y Masuda
- Division of Allergy, Asthma, and Clinical Immunology, Department of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, United States
| | - Hirohito Kita
- Division of Allergy, Asthma, and Clinical Immunology, Department of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, United States.,Department of Immunology, Mayo Clinic Arizona, Scottsdale, AZ, United States
| | - Benjamin L Wright
- Division of Allergy, Asthma, and Clinical Immunology, Department of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, United States.,Division of Pulmonology, Phoenix Children's Hospital, Phoenix, AZ, United States
| |
Collapse
|
23
|
Chen X, Yang H, Jia J, Chen Y, Wang J, Chen H, Jiang C. Mulberry leaf polysaccharide supplementation contributes to enhancing the respiratory mucosal barrier immune response in Newcastle disease virus-vaccinated chicks. Poult Sci 2020; 100:592-602. [PMID: 33518112 PMCID: PMC7858170 DOI: 10.1016/j.psj.2020.11.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/16/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023] Open
Abstract
Despite high global vaccination coverage, Newcastle disease (ND) remains a constant threat to poultry producers owing to low antibody levels. Given the respiratory mucosa is the important site for Newcastle disease virus (NDV) vaccination, enhancing respiratory mucosal immunity may help control ND. Our previous study showed that mulberry leaf polysaccharide (MLP) is very promising in delivering a robust balanced immune response, but the effects of it on respiratory immunity in chicks are unknown. In this study, we evaluated the potential of MLP to activate respiratory mucosal immunity and revealed the possible mechanism of MLP as an immunopotentiator for ND vaccines. Chicks were randomly divided into 5 groups: blank control, vaccination control (VC), and low-, middle-, and high-dose MLP (MLP-L, MLP-M, and MLP-H) (n = 30). The serum results of humoral and cell-mediated immune responses showed significant increases in NDV hemagglutination inhibition antibody titer, IgG and IgA antibody levels, and the T-lymphocyte population in the MLP-M group compared with the VC group. Validation of results also indicated remarkable increases in tracheal antibody-mediated immunity and a mucosal immune response in the MLP-M group. Furthermore, the upregulation of TLR7 revealed a possible mechanism. Our findings provided evidence to consider MLP as a potential mucosal vaccine adjuvant candidate against ND in chickens.
Collapse
Affiliation(s)
- Xiaolan Chen
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu Province 225300, PR China.
| | - Haifeng Yang
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu Province 225300, PR China
| | - Jiping Jia
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu Province 225300, PR China
| | - Yu Chen
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu Province 225300, PR China
| | - Jing Wang
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu Province 225300, PR China
| | - Haifeng Chen
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu Province 225300, PR China
| | - Chunmao Jiang
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu Province 225300, PR China
| |
Collapse
|
24
|
Arias Á, Lucendo AJ. Epidemiology and risk factors for eosinophilic esophagitis: lessons for clinicians. Expert Rev Gastroenterol Hepatol 2020; 14:1069-1082. [PMID: 32749898 DOI: 10.1080/17474124.2020.1806054] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The rapid expansion in the epidemiology of eosinophilic esophagitis (EoE) is being documented, along with cumulative research assessing environmental exposures associated with EoE and susceptibility due to genetic variants. AREAS COVERED Incidence rates for EoE of 5-10 new cases per 100,000 inhabitants annually have shown an increase in recent reports of up to 20 in some countries; the highest prevalence being reported for Europe and North America, where EoE now affects more than 1 out of 1,000 people. EoE has been shown to be associated with several disorders, Th2-mediated atopies being the most common. Patients with EoE exhibit increased frequency of asthma, allergic rhinitis and eczema, and EoE has been considered as a late component of the atopic march. Risk variants in TSLP, CAPN14 and LRCC32 genes, among others, have all been related to EoE, and interact with prenatal and early life exposure potentially modifying abundance and composition of gut microbiome. Dysregulated interactions between bacteria and mucosal immunity emerge as leading causes of EoE. EXPERT OPINION The expanding epidemiology of EoE, the resources needed and subsequent increasing healthcare costs require additional effort to optimize cost-effective management and unveil mechanisms that enhance the development of future preventive strategies.
Collapse
Affiliation(s)
- Ángel Arias
- Research Unit, Hospital General Mancha Centro , Alcázar De San Juan, Spain.,Centro De Investigación Biomédica En Red De Enfermedades Hepáticas Y Digestivas (Ciberehd) , Madrid, Spain.,Instituto De Investigación Sanitaria La Princesa , Madrid, Spain
| | - Alfredo J Lucendo
- Centro De Investigación Biomédica En Red De Enfermedades Hepáticas Y Digestivas (Ciberehd) , Madrid, Spain.,Instituto De Investigación Sanitaria La Princesa , Madrid, Spain.,Department of Gastroenterology, Hospital General De Tomelloso , Ciudad Real, Spain
| |
Collapse
|
25
|
Doulberis M, Kountouras J, Rogler G. Reconsidering the "protective" hypothesis of Helicobacter pylori infection in eosinophilic esophagitis. Ann N Y Acad Sci 2020; 1481:59-71. [PMID: 32770542 DOI: 10.1111/nyas.14449] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/12/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023]
Abstract
Since its discovery, Helicobacter pylori (H. pylori) has attracted attention in the biomedical world with its numerous pathophysiologic implications, both gastrointestinal and systemic. Beyond its well-established carcinogenic properties, emerging evidence also supports "harmful" proinflammatory and neurodegenerative roles of H. pylori. On the other hand, H. pylori infection has been proposed to be "protective" against several diseases, such as asthma and gastroesophageal reflux disease. Eosinophilic esophagitis (EoE) is a relatively new, allergen/immune-mediated disease, which has also been linked to these considerations. Main arguments are a postulated shift of immune responses by H. pylori from T helper 2 (TH 2) to TH 1 polarization, as well as a potential decline of the H. pylori burden with the dramatic parallel rise of ΕοΕ: a series of observational studies reported an inverse association. In this review, we counter these arguments by providing further epidemiological data, which point out that this generalization might be rather incomplete. We also discuss the limitations of the existing studies evaluating a possible association. Furthermore, we provide current evidence on common pathogenetic components, which share both entities. In summary, the claim that H. pylori is protective against EoE is rather incomplete, and further mechanistic studies are necessary to elucidate a possible association.
Collapse
Affiliation(s)
- Michael Doulberis
- Department of Gastroenterology and Hepatology, University of Zurich, Zurich, Switzerland.,Second Medical Clinic, Faculty of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Jannis Kountouras
- Second Medical Clinic, Faculty of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
26
|
Zhang C, Wang N, Tan H, Guo W, Chen F, Zhong Z, Man K, Tsao SW, Lao L, Feng Y. Direct inhibition of the TLR4/MyD88 pathway by geniposide suppresses HIF-1α-independent VEGF expression and angiogenesis in hepatocellular carcinoma. Br J Pharmacol 2020; 177:3240-3257. [PMID: 32144747 PMCID: PMC7312435 DOI: 10.1111/bph.15046] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE As a typical hypervascular tumour, hepatocellular carcinoma (HCC) is predominantly grown through angiogenesis. Geniposide is a promising anti-inflammatory compound found in Gardenia jasminoides, but its effects on the progression of HCC remain untested. EXPERIMENTAL APPROACH The anti-HCC effects of geniposide was investigated in cellular models and orthotopic HCC mice. Transcriptional regulation of the VEGF promoter was measured by dual-luciferase reporter assay. The anti-angiogenic action of geniposide was measured by tube formation assay. Both surface plasmon resonance techniques and human phospho-kinase array analysis were utilized to validate the relationship between targets of geniposide and hepatocarcinogenesis. KEY RESULTS Geniposide exhibited significant disruption of HCC proliferation, invasion, angiogenesis and lung metastasis in orthotopic HCC mice. Geniposide inhibited secretion of VEGF by HCC and suppressed the migration of endothelial cells and the formation of intra-tumour blood vessels, without cytotoxicity and independently of the transcription factor HIF-1α. Direct inhibition of TLR4 by geniposide led to the shutdown of the TLR4/MyD88 pathway and STAT3/Sp1-dependent VEGF production. However, LPS, an agonist of TLR4, restored STAT3/Sp1-related VEGF production in geniposide-inhibited HCC angiogenesis. CONCLUSION AND IMPLICATIONS The direct inhibitory effect of geniposide on TLR4/MyD88 activation contributes to the suppression of STAT3/Sp1-dependent VEGF overexpression in HCC angiogenesis and pulmonary metastasis. This action of geniposide was not affected by stabilization of HIF-1α. Our study offers a novel anti-VEGF mechanism for the inhibition of HCC.
Collapse
Affiliation(s)
- Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.China
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.China
| | - Hor‐Yue Tan
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.China
| | - Wei Guo
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.China
| | - Feiyu Chen
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.China
| | - Zhangfeng Zhong
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.China
| | - Kwan Man
- Department of Surgery, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.China
| | - Sai Wah Tsao
- School of Biomedical Science, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.China
| | - Lixing Lao
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.China
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong S.A.R.China
| |
Collapse
|
27
|
Oliva S, Azouz NP, Stronati L, Rothenberg ME. Recent advances in potential targets for eosinophilic esophagitis treatments. Expert Rev Clin Immunol 2020; 16:421-428. [PMID: 32163308 DOI: 10.1080/1744666x.2020.1742110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Diagnostic and therapeutic strategies in eosinophilic esophagitis (EoE) are constantly evolving. Recently, the improved understanding of EoE pathogenesis has led to identification of a variety of other potential targets that have never been considered before.Areas covered: In September 2019, we performed structured literature searches in Medline and PubMed, Cochrane meta-analyses, and abstracts of international congresses to review new potential therapeutic approaches for EoE.Expert opinion: The advent of omics disciplines has been helping in finding new molecular targets in EoE pathogenesis and may provide future guidance for deep phenotyping of the disease and therefore facilitate the possibility of personalized medicine. Interestingly, these new treatments should be focused on the restoration of epithelial barrier dysfunction, downregulation of specific molecular pathways of eosinophilic inflammation, and finally, prevention of esophageal remodeling. In this review, we highlight the most recent insights in EoE pathogenesis, which open new pathways for developing new therapeutic targets for clinical practice.
Collapse
Affiliation(s)
- Salvatore Oliva
- Pediatric Gastroenterology and Liver Unit, Maternal and Child Health Department, Sapienza - University of Rome, Rome, Italy.,Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Nurit P Azouz
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Laura Stronati
- Department of Molecular Medicine, Sapienza-University of Rome, Rome, Italy
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
28
|
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: 28] [Impact Index Per Article: 4.7] [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
|
29
|
Limketkai BN, Shah SC, Hirano I, Bellaguarda E, Colombel JF. Epidemiology and implications of concurrent diagnosis of eosinophilic oesophagitis and IBD based on a prospective population-based analysis. Gut 2019; 68:2152-2160. [PMID: 30923072 DOI: 10.1136/gutjnl-2018-318074] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Eosinophilic oesophagitis (EoO) and IBD are immune-mediated diseases of the gastrointestinal tract with possible overlapping pathogenic mechanisms. Our aim was to define the epidemiology and clinical implications of concurrent EoO and IBD diagnoses. DESIGN We conducted a prospective cohort analysis using the Truven MarketScan database (2009-2016) to estimate the incidence and prevalence of EoO in patients with Crohn's disease (CD) or UC and vice versa. Cox proportional hazards and Kaplan-Meier methods were used to estimate the risk of EoO-related or IBD-related complications among patients with concurrent diagnoses. RESULTS Among 134 013 536 individuals, the incidence of EoO, CD and UC were 23.1, 51.2 and 55.2 per 100 000 person-years, respectively. The risk of EoO was higher among patients with CD (incidence rate ratio [IRR] 5.4, p<0.01; prevalence ratio (PR) 7.8, p<0.01) or UC (IRR 3.5, p<0.01; PR 5.0, p<0.01), while the risk of IBD was higher among patients with EoO (CD: IRR 5.7, p<0.01; PR 7.6, p<0.01; UC: IRR 3.4, p<0.01; PR 4.9, p<0.01) versus individuals without either diagnosis. Concurrent diagnosis of EoO and IBD was associated with greater composite risk of IBD-related complications (CD: adjusted HR (aHR) 1.09, p=0.01; UC: aHR 1.10, p=0.04) but lower composite risk of EoO-related complications (aHR 0.59; p<0.01). CONCLUSION Based on a population-based prospective cohort analysis, the risk of EoO is significantly higher among patients with IBD and vice versa. Concurrent diagnoses might modify the risk of IBD-related and EoO-related complications. Studies defining the mechanisms underlying these observations are needed.
Collapse
Affiliation(s)
- Berkeley N Limketkai
- Division of Gastroenterology, Stanford University School of Medicine, Stanford, California, USA
| | - Shailja C Shah
- Division of Gastroenterology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ikuo Hirano
- Division of Gastroenterology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Emanuelle Bellaguarda
- Division of Gastroenterology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jean-Frederic Colombel
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| |
Collapse
|
30
|
Camilleri M. Leaky gut: mechanisms, measurement and clinical implications in humans. Gut 2019; 68:1516-1526. [PMID: 31076401 PMCID: PMC6790068 DOI: 10.1136/gutjnl-2019-318427] [Citation(s) in RCA: 628] [Impact Index Per Article: 104.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022]
Abstract
The objectives of this review on 'leaky gut' for clinicians are to discuss the components of the intestinal barrier, the diverse measurements of intestinal permeability, their perturbation in non-inflammatory 'stressed states' and the impact of treatment with dietary factors. Information on 'healthy' or 'leaky' gut in the public domain requires confirmation before endorsing dietary exclusions, replacement with non-irritating foods (such as fermented foods) or use of supplements to repair the damage. The intestinal barrier includes surface mucus, epithelial layer and immune defences. Epithelial permeability results from increased paracellular transport, apoptosis or transcellular permeability. Barrier function can be tested in vivo using orally administered probe molecules or in vitro using mucosal biopsies from humans, exposing the colonic mucosa from rats or mice or cell layers to extracts of colonic mucosa or stool from human patients. Assessment of intestinal barrier requires measurements beyond the epithelial layer. 'Stress' disorders such as endurance exercise, non-steroidal anti-inflammatory drugs administration, pregnancy and surfactants (such as bile acids and dietary factors such as emulsifiers) increase permeability. Dietary factors can reverse intestinal leakiness and mucosal damage in the 'stress' disorders. Whereas inflammatory or ulcerating intestinal diseases result in leaky gut, no such disease can be cured by simply normalising intestinal barrier function. It is still unproven that restoring barrier function can ameliorate clinical manifestations in GI or systemic diseases. Clinicians should be aware of the potential of barrier dysfunction in GI diseases and of the barrier as a target for future therapy.
Collapse
Affiliation(s)
- Michael Camilleri
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
31
|
Ma S, Wang SY, Zhu LP, Chen X, Wang BM. Esophageal microbiota and esophageal diseases. Shijie Huaren Xiaohua Zazhi 2019; 27:767-772. [DOI: 10.11569/wcjd.v27.i12.767] [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
The microbiota of the esophagus has been the least systematically studied among the organs of the gastrointestinal tract. Esophageal microbiota has been proved to be complex with a high diversity until recently. Alterations of its composition are associated with the development and progression of esophageal diseases. Immunoinflammatory responses caused by dysbiosis may play a role in the pathogenesis of esophageal disorders. This review addresses the characteristics of esophageal microbiota in physiological and pathological conditions, aiming to provide new insights into esophageal microecology-based intervention strategies for esophageal diseases.
Collapse
Affiliation(s)
- Shuang Ma
- Department of Gastroenterology and Hepatology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Sai-Yu Wang
- Department of Gastroenterology and Hepatology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Lan-Ping Zhu
- Department of Gastroenterology and Hepatology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Bang-Mao Wang
- Department of Gastroenterology and Hepatology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| |
Collapse
|
32
|
Arias Á, Lucendo AJ. Molecular basis and cellular mechanisms of eosinophilic esophagitis for the clinical practice. Expert Rev Gastroenterol Hepatol 2019; 13:99-117. [PMID: 30791784 DOI: 10.1080/17474124.2019.1546120] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Eosinophilic esophagitis (EoE) is a chronic, allergen-driven inflammatory esophageal disease characterized by predominantly eosinophilic inflammation leading to esophageal dysfunction. Recent efforts to understand EoE have increased our knowledge of the disease. Areas covered: Multiple cells, molecules, and genes interplay with early life environmental factors in the pathophysiology of EoE to converge in the esophageal epithelium at the center of disease pathogenesis. Epithelial cells constitute a mayor cytokine source for TSLP and Calpain-14; an impaired epithelial barrier function allowing penetration of food and microbiota-derived antigens is involved in triggering and maintaining inflammation. Eosinophil and mast cell-derived products, including TGFβ, together with IL-1β and TNFα, promote epithelial mesenchymal transition in EoE, contributing to tissue remodeling by synthetizing and depositing extracellular matrix in subepithelial layers. This article aims to provide a state-of-the-art update on the pathophysiology of EoE applied to clinical practice, and latest research and developments with potential interest to improve the diagnosis and treatment of patients with EoE are revised. Expert commentary: Preliminary approaches have provided promising results toward incorporating minimally invasive methods for patient diagnosis and monitoring in clinical practice. Early diagnosis and optimized therapies will allow for personalized medicine in EoE.
Collapse
Affiliation(s)
- Ángel Arias
- a Research Unit , Hospital General La Mancha Centro , Alcázar de San Juan , Spain.,b Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd) , Madrid , Spain
| | - Alfredo J Lucendo
- b Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd) , Madrid , Spain.,c Department of Gastroenterology , Hospital General de Tomelloso , Ciudad Real , Spain
| |
Collapse
|
33
|
Carbohydrate-based adjuvants activate tumor-specific Th1 and CD8+ T-cell responses and reduce the immunosuppressive activity of MDSCs. Cancer Lett 2019; 440-441:94-105. [DOI: 10.1016/j.canlet.2018.10.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/24/2018] [Accepted: 10/10/2018] [Indexed: 01/09/2023]
|
34
|
Choksi Y, Vaezi MF. Preliminary esophageal microbiome studies prompt important scientific questions. Clin Transl Gastroenterol 2018; 9:156. [PMID: 29807991 PMCID: PMC5972153 DOI: 10.1038/s41424-018-0029-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 05/04/2018] [Indexed: 12/14/2022] Open
Abstract
Analysis of the esophageal microbiome remains a new field of research. Two hypothesis-generating papers published in the current issue of the Journal go beyond characterizing the esophageal microbiome in Barrett's esophagus or eosinophilic esophagitis (EoE). Snider et al. suggest that the salivary microbiome can be used as a screening tool for Barrett's esophagus, and Arias et al. demonstrates abnormal expression of Toll-like receptors and innate immune effector proteins in patients with active EoE. We discuss these findings, raise fundamental questions about microbiome studies, and offer ideas for future studies.
Collapse
Affiliation(s)
- Yash Choksi
- Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt Medical Center, Nashville, TN, USA
| | - Michael F Vaezi
- Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt Medical Center, Nashville, TN, USA.
| |
Collapse
|