1
|
Allen-Brady K, Clayton F, Hazel MW, Stevens J, Pyne AL, Pletneva MA, Cessna M, Stubben C, Robson J, Fang J, Peterson KA. Overlap of Genomic and Transcriptomic Genes Identified in Familial Eosinophilic Esophagitis. Gastroenterology 2025; 168:1101-1113.e18. [PMID: 39914776 PMCID: PMC12104003 DOI: 10.1053/j.gastro.2025.01.235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 12/27/2024] [Accepted: 01/10/2025] [Indexed: 02/26/2025]
Abstract
BACKGROUND & AIMS Evidence for a genetic contribution to eosinophilic esophagitis (EoE) exists from family and genome-wide association studies. Extensive investigation into rare variants contributing to EoE has not been performed. The study's aim was to evaluate families with multiple cases of EoE by genomic and transcriptomic sequencing to identify genes predisposing to EoE. METHODS Distant relative pairs (eg, cousins) in extended EoE families and other affected relatives were whole exome sequenced to identify rare, shared, potentially pathologic variants. RNA sequencing was performed in nuclear families with multiple EoE cases. We compared the overlap of genes from DNA and RNA sequencing for relevance to disease manifestations. RESULTS Whole exome sequencing was performed in 50 familial cases in 21 EoE extended pedigrees. We observed 189 rare candidate predisposition variants in 181 genes with complete sharing among all affected family members within each pedigree. RNA sequencing was performed for 43 EoE cases in 18 nuclear families, including 6 relatives without EoE. We observed 698 total differentially expressed genes compared with controls. We identified 3 genes (MUC16, ADGRE1, and TENM3) with evidence of rare variant sharing among all affected family members and differential gene expression. We identified 36 other genes with partial sharing of rare variants among some affected family members and with differential gene expression. Several genes identified as prominent in EoE were also differentially expressed in unaffected relatives. CONCLUSIONS Genes related to immune response, barrier dysfunction, and cell adhesion were identified in familial EoE cases and unaffected family members, supporting a genetic familial predisposition and a possible multihit background to disease pathophysiology.
Collapse
Affiliation(s)
- Kristina Allen-Brady
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah.
| | - Frederic Clayton
- Department of Pathology, University of Utah, Salt Lake City, Utah
| | - Mark W Hazel
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Jeff Stevens
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Ashley L Pyne
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Maria A Pletneva
- Department of Pathology, University of Utah, Salt Lake City, Utah
| | - Melissa Cessna
- Department of Pathology, Intermountain Healthcare, Murray, Utah
| | - Chris Stubben
- Cancer Bioinformatics, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Jacob Robson
- Pediatric Gastroenterology, Intermountain Healthcare, Murray, Utah
| | - John Fang
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Kathryn A Peterson
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| |
Collapse
|
2
|
Dickerson A, Dellon ES, Aceves SS. Future of therapy and monitoring for eosinophilic esophagitis and eosinophilic gastrointestinal diseases. Ann Allergy Asthma Immunol 2025:S1081-1206(25)00242-X. [PMID: 40393554 DOI: 10.1016/j.anai.2025.05.016] [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: 03/14/2025] [Revised: 05/12/2025] [Accepted: 05/14/2025] [Indexed: 05/22/2025]
Abstract
Eosinophilic gastrointestinal disorders, including eosinophilic esophagitis, are chronic Th2 mediated diseases. Establishing a diagnosis and initiating treatment is crucial to limit disease progression that may lead to tissue remodeling and the development of strictures that significantly impact patient quality of life. Expert consensus guidelines provide a framework for treating eosinophilic esophagitis with diet elimination, proton pump inhibitors, swallowed topical steroids, or dupilumab and for monitoring with sedated endoscopy for gross and histologic evaluation. While this provides an established algorithm for treating and monitoring eosinophilic esophagitis, there is less established for the rarer eosinophilic gastrointestinal disorders (eosinophilic gastritis, enteritis, and colitis). Research advancements continue to emerge at a rapid pace, identifying potential biomarkers, therapeutic targets, and monitoring strategies. In this article, we review the current accepted methods for treating and monitoring eosinophilic gastrointestinal disorders with a focus on eosinophilic esophagitis, assess what is currently under investigation, and provide an aspirational vision for future disease management with a streamlined algorithm of personalized medicine and less invasive monitoring.
Collapse
Affiliation(s)
- Andrew Dickerson
- Division of Allergy, Immunology, Department of Pediatrics, University of California, San Diego and Rady Children's Hospital, San Diego, California; Division of Gastroenterology, Department of Pediatrics, University of California, San Diego and Rady Children's Hospital, San Diego, California
| | - Evan S Dellon
- Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology. Department of Medicine, University of North Carolina, Chapel Hill, NC
| | - Seema S Aceves
- Division of Allergy, Immunology, Department of Pediatrics, University of California, San Diego and Rady Children's Hospital, San Diego, California; Department of Medicine, University of California, San Diego.
| |
Collapse
|
3
|
Zhao N, Wang G, Long S, Lv X, Ran X, Wang J, Su Y, Wang T. The antiprotease Spink7 promotes inflammation resolution by modulating multiple proteases activities during wound healing. Clin Transl Med 2025; 15:e70291. [PMID: 40147022 PMCID: PMC11949503 DOI: 10.1002/ctm2.70291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/25/2024] [Accepted: 12/20/2024] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Effective control of inflammation is crucial for the healing of cutaneous wounds, but the molecular mechanisms governing inflammation resolution during wound closure are still not yet clear. Here, we describe a homeostatic mechanism that facilitates the inflammation resolution by timely regulating the targeted proteases activities through antiprotease Spink7 (serine peptidase inhibitor, kazal type 7). METHODS The expression pattern of Spink7 was investigated by quantitative RT-PCR, immunohistochemistry (IHC) and in situ hybridization. In both Spink7 knockdown and knockout models, quantitative comparisons were made between the healing rate of wounds and histopathological morphometric analysis. Microarrays, multiple chemokine assays, IHC, immunofluorescence, protease activity measurement were performed to explore the underlying mechanisms of Spink7 knockout in impaired wound healing. Radiation-wound combined injury (R-W-CI) model was employed to evaluate the therapeutic effects of Spink7 manipulation. RESULTS Our study demonstrates that Spink7 is significantly upregulated in the differentiated epidermal granular keratinocytes of proliferative phase during murine wound closure. Both local knockdown of Spink7 levels in wounds using siRNA gel and systemic knockout of Spink7 using KO mice resulted in delayed wound closure with sustained neutrophil infiltration. Loss of Spink7 leads to augmented inflammatory responses, increased production of multiple chemokines/cytokines, and impaired M2 polarization of macrophages in wound healing. Furthermore, loss of Spink7 results in elevated proteolytic activities of uPA, MMP2/9 and KLK5/7 in proliferative phase. However, inhibiting KLK5/7 downstream PAR2 activation exacerbates the phenotype of KO mice. In R-W-CI model, further significant induction of Spink7 is observed in wounds with insufficient inflammatory response. Local suppression of Spink7 promotes wound healing in the R-W-CI model by augmenting inflammation. CONCLUSIONS Maintaining an endogenous balance between Spink7 and its target proteases is a crucial checkpoint for regulating inflammation resolution during healing. Therefore, manipulating levels of Spink7 might be an effective treatment for impaired wounds caused by inflammatory dysregulation.
Collapse
Affiliation(s)
- Na Zhao
- Institute of Combined InjuryChongqing Engineering Research Center for NanomedicineSchool of Preventive Military MedicineArmy Medical University (Third Military Medical University)ChongqingChina
- Institute of Materia Medica and Department of PharmaceuticsCollege of PharmacyArmy Medical University (Third Military Medical University)ChongqingChina
| | - Guojian Wang
- Institute of Combined InjuryChongqing Engineering Research Center for NanomedicineSchool of Preventive Military MedicineArmy Medical University (Third Military Medical University)ChongqingChina
| | - Shuang Long
- Institute of Combined InjuryChongqing Engineering Research Center for NanomedicineSchool of Preventive Military MedicineArmy Medical University (Third Military Medical University)ChongqingChina
| | - Xiaofan Lv
- Institute of Combined InjuryChongqing Engineering Research Center for NanomedicineSchool of Preventive Military MedicineArmy Medical University (Third Military Medical University)ChongqingChina
| | - Xinze Ran
- Institute of Combined InjuryChongqing Engineering Research Center for NanomedicineSchool of Preventive Military MedicineArmy Medical University (Third Military Medical University)ChongqingChina
| | - Junping Wang
- Institute of Combined InjuryChongqing Engineering Research Center for NanomedicineSchool of Preventive Military MedicineArmy Medical University (Third Military Medical University)ChongqingChina
- State Key Laboratory of Trauma and Chemical PoisoningChongqingChina
| | - Yongping Su
- Institute of Combined InjuryChongqing Engineering Research Center for NanomedicineSchool of Preventive Military MedicineArmy Medical University (Third Military Medical University)ChongqingChina
| | - Tao Wang
- Institute of Combined InjuryChongqing Engineering Research Center for NanomedicineSchool of Preventive Military MedicineArmy Medical University (Third Military Medical University)ChongqingChina
| |
Collapse
|
4
|
Dinardo G, Fiocchi A, Artesani MC, De Angelis P, Rea F, Tambucci R, Dahdah L, Fierro V, Valluzzi RL, Arasi S, Pecora V, Cafarotti A, Mazzuca C, Indolfi C, Miraglia del Giudice M, Urbani S. Eosinophilic Esophagitis and Cow's Milk: Mechanisms, Challenges, and Treatment Perspectives. Nutrients 2025; 17:265. [PMID: 39861395 PMCID: PMC11767713 DOI: 10.3390/nu17020265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/08/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Eosinophilic esophagitis is a chronic, antigen-driven, immune-mediated disease characterized by esophageal dysfunction and significant eosinophilic infiltration. Its rising incidence and prevalence over recent decades reflect both increased clinical awareness and the influence of environmental factors such as dietary patterns and allergen exposure. Among food allergens, cow's milk proteins are the most commonly implicated triggers, contributing to esophageal inflammation through complex immunological pathways involving both IgE-mediated and non-IgE-mediated mechanisms. Dietary elimination of cow's milk has been shown to induce histologic remission in over 60% of pediatric patients, underscoring its pivotal role in eosinophilic esophagitis management. Despite these promising results, challenges persist, including variability in individual responses, the burden of adherence to restrictive diets, and gaps in understanding the molecular mechanisms driving cow's milk-induced esophageal inflammation. This review examines the complex relationship between eosinophilic esophagitis and cow's milk, focusing on its role in disease pathogenesis and management, offering insights into its therapeutic implications. Understanding the interplay between eosinophilic esophagitis and dietary allergens, particularly cow's milk, may inform the development of targeted interventions and improve clinical outcomes for affected patients.
Collapse
Affiliation(s)
- Giulio Dinardo
- Department of Woman, Child and of General and Specialized Surgery, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (C.I.); (M.M.d.G.)
| | - Alessandro Fiocchi
- Allergy Diseases Research Area, Pediatric Allergology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.A.); (L.D.); (V.F.); (R.L.V.); (S.A.); (V.P.); (A.C.); (C.M.); (S.U.)
| | - Maria Cristina Artesani
- Allergy Diseases Research Area, Pediatric Allergology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.A.); (L.D.); (V.F.); (R.L.V.); (S.A.); (V.P.); (A.C.); (C.M.); (S.U.)
| | - Paola De Angelis
- Digestive Endoscopy Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (P.D.A.); (F.R.); (R.T.)
| | - Francesca Rea
- Digestive Endoscopy Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (P.D.A.); (F.R.); (R.T.)
| | - Renato Tambucci
- Digestive Endoscopy Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (P.D.A.); (F.R.); (R.T.)
| | - Lamia Dahdah
- Allergy Diseases Research Area, Pediatric Allergology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.A.); (L.D.); (V.F.); (R.L.V.); (S.A.); (V.P.); (A.C.); (C.M.); (S.U.)
| | - Vincenzo Fierro
- Allergy Diseases Research Area, Pediatric Allergology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.A.); (L.D.); (V.F.); (R.L.V.); (S.A.); (V.P.); (A.C.); (C.M.); (S.U.)
| | - Rocco Luigi Valluzzi
- Allergy Diseases Research Area, Pediatric Allergology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.A.); (L.D.); (V.F.); (R.L.V.); (S.A.); (V.P.); (A.C.); (C.M.); (S.U.)
| | - Stefania Arasi
- Allergy Diseases Research Area, Pediatric Allergology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.A.); (L.D.); (V.F.); (R.L.V.); (S.A.); (V.P.); (A.C.); (C.M.); (S.U.)
| | - Valentina Pecora
- Allergy Diseases Research Area, Pediatric Allergology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.A.); (L.D.); (V.F.); (R.L.V.); (S.A.); (V.P.); (A.C.); (C.M.); (S.U.)
| | - Arianna Cafarotti
- Allergy Diseases Research Area, Pediatric Allergology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.A.); (L.D.); (V.F.); (R.L.V.); (S.A.); (V.P.); (A.C.); (C.M.); (S.U.)
| | - Carmen Mazzuca
- Allergy Diseases Research Area, Pediatric Allergology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.A.); (L.D.); (V.F.); (R.L.V.); (S.A.); (V.P.); (A.C.); (C.M.); (S.U.)
| | - Cristiana Indolfi
- Department of Woman, Child and of General and Specialized Surgery, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (C.I.); (M.M.d.G.)
| | - Michele Miraglia del Giudice
- Department of Woman, Child and of General and Specialized Surgery, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (C.I.); (M.M.d.G.)
| | - Sara Urbani
- Allergy Diseases Research Area, Pediatric Allergology Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.A.); (L.D.); (V.F.); (R.L.V.); (S.A.); (V.P.); (A.C.); (C.M.); (S.U.)
| |
Collapse
|
5
|
Rodríguez-López EM, Hill DA. Eosinophilic gastrointestinal disorders and the role for the epithelium in pathogenesis and treatment. Curr Opin Pediatr 2024; 36:668-673. [PMID: 39319691 DOI: 10.1097/mop.0000000000001406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
PURPOSE OF REVIEW This review aims to provide an overview of the current understanding of eosinophilic gastrointestinal disorders (EGIDs) and the role of the epithelium in influencing disease pathogenesis to inform and devise future therapeutic strategies. RECENT FINDINGS Changes in epithelial cell structure, functions, and integrity are observed in EGIDs. In eosinophilic esophagitis (EoE), the esophageal epithelium has been shown to play key roles in perpetuating the inflammatory response in EoE through the expression of pro-inflammatory cytokines and immunological cell-surface proteins. Similar mechanisms appear to exist in the other EGIDs, including eosinophilic gastritis (EoG), eosinophilic enteritis (EoN), and eosinophilic colitis (EoC). Because of the increasing rarity of each non-EoE EGID, research focusing on how the epithelium is modulating disease in each lower gastrointestinal compartment is still in its rudimentary stages. SUMMARY While there has been significant progress in understanding the role of the epithelium in EoE, further research is needed to obtain a better understanding of the mechanisms mediating epithelial-immune crosstalk in non-EoE EGIDs. Using EoE-epithelial cell research to inform future EGID investigations could lead to the development of new therapeutic interventions, such as targeted therapies to restore epithelial barrier function and reduce inflammation, to improve rare disease-patient quality of life.
Collapse
Affiliation(s)
- Eric M Rodríguez-López
- Institute for Immunology and Immune Health
- Immunology Graduate Group, Perelman School of Medicine, University of Pennsylvania
- Division of Allergy and Clinical Immunology, Children's Hospital of Philadelphia
| | - David A Hill
- Institute for Immunology and Immune Health
- Immunology Graduate Group, Perelman School of Medicine, University of Pennsylvania
- Division of Allergy and Clinical Immunology, Children's Hospital of Philadelphia
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
6
|
Luengas‐Martinez A, Ismail D, Paus R, Young HS. Vascular endothelial growth factor A inhibition remodels the transcriptional signature of lipid metabolism in psoriasis non-lesional skin in 12 h ex vivo culture. SKIN HEALTH AND DISEASE 2024; 4:e471. [PMID: 39624732 PMCID: PMC11608907 DOI: 10.1002/ski2.471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/09/2024] [Accepted: 10/11/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Vascular endothelial growth factor A (VEGF-A)-mediated angiogenesis is involved in the pathogenesis of psoriasis. VEGF-A inhibitors are widely used to treat oncological and ophthalmological diseases but have not been used in psoriasis management. The molecular mechanisms underlying the effects of VEGF-A inhibition in psoriatic skin remain unknown. OBJECTIVES To identify the genes and canonical pathways affected by VEGF-A inhibition in non-lesional and plaque skin ex vivo. METHODS Total RNA sequencing was performed on skin biopsies from patients with psoriasis (n = 6; plaque and non-lesional skin) and healthy controls (n = 6) incubated with anti-VEGF-A monoclonal antibody (bevacizumab, Avastin®) or human IgG1 isotype control for 12 h in serum-free organ culture. Differentially expressed genes between paired control and treated samples with adjusted p-values <0.1 were considered significant. Gene ontology and ingenuity pathway analysis was used to identify enriched biological processes, canonical pathways and upstream regulators. RESULTS VEGF-A inhibition upregulated the expression of genes involved in lipid metabolism. Pathway enrichment analysis identified the activation of pathways involved in fatty acids and lipid biosynthesis and degradation in non-lesional skin and ferroptosis in plaque skin. VEGF-A inhibition downregulated endothelial cell apoptosis in non-lesional psoriasis skin and members of the interferon family were identified as potential regulators of the effects of VEGF-A inhibition in non-lesional skin. CONCLUSION Early response to VEGF-A inhibition is associated with changes in lipid metabolism in non-lesional psoriasis skin and cellular stress in psoriasis plaque. More investigation is needed to validate these findings.
Collapse
Affiliation(s)
- Andrea Luengas‐Martinez
- Centre for Dermatology Research and Manchester Academic Health Science CentreThe University of ManchesterManchesterUK
| | - Dina Ismail
- Centre for Dermatology Research and Manchester Academic Health Science CentreThe University of ManchesterManchesterUK
| | - Ralf Paus
- Centre for Dermatology Research and Manchester Academic Health Science CentreThe University of ManchesterManchesterUK
- Dr. Philip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Monasterium LaboratoryMuensterGermany
| | - Helen S. Young
- Centre for Dermatology Research and Manchester Academic Health Science CentreThe University of ManchesterManchesterUK
| |
Collapse
|
7
|
Matsuyama K, Yamada S, Sato H, Zhan J, Shoda T. Advances in omics data for eosinophilic esophagitis: moving towards multi-omics analyses. J Gastroenterol 2024; 59:963-978. [PMID: 39297956 PMCID: PMC11496339 DOI: 10.1007/s00535-024-02151-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/07/2024] [Indexed: 09/21/2024]
Abstract
Eosinophilic esophagitis (EoE) is a chronic, allergic inflammatory disease of the esophagus characterized by eosinophil accumulation and has a growing global prevalence. EoE significantly impairs quality of life and poses a substantial burden on healthcare resources. Currently, only two FDA-approved medications exist for EoE, highlighting the need for broader research into its management and prevention. Recent advancements in omics technologies, such as genomics, epigenetics, transcriptomics, proteomics, and others, offer new insights into the genetic and immunologic mechanisms underlying EoE. Genomic studies have identified genetic loci and mutations associated with EoE, revealing predispositions that vary by ancestry and indicating EoE's complex genetic basis. Epigenetic studies have uncovered changes in DNA methylation and chromatin structure that affect gene expression, influencing EoE pathology. Transcriptomic analyses have revealed a distinct gene expression profile in EoE, dominated by genes involved in activated type 2 immunity and epithelial barrier function. Proteomic approaches have furthered the understanding of EoE mechanisms, identifying potential new biomarkers and therapeutic targets. However, challenges in integrating diverse omics data persist, largely due to their complexity and the need for advanced computational methods. Machine learning is emerging as a valuable tool for analyzing extensive and intricate datasets, potentially revealing new aspects of EoE pathogenesis. The integration of multi-omics data through sophisticated computational approaches promises significant advancements in our understanding of EoE, improving diagnostics, and enhancing treatment effectiveness. This review synthesizes current omics research and explores future directions for comprehensively understanding the disease mechanisms in EoE.
Collapse
Affiliation(s)
- Kazuhiro Matsuyama
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, MLC 7028, Cincinnati, OH, 45229, USA
- Department of Computer Science, University of Cincinnati, Cincinnati, USA
| | - Shingo Yamada
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, MLC 7028, Cincinnati, OH, 45229, USA
| | - Hironori Sato
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, MLC 7028, Cincinnati, OH, 45229, USA
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Justin Zhan
- Department of Computer Science, University of Cincinnati, Cincinnati, USA
| | - Tetsuo Shoda
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, MLC 7028, Cincinnati, OH, 45229, USA.
| |
Collapse
|
8
|
Hiremath G, Choksi Y, Correa H, Jacobse J, Das SR, Ma S, Goettel JA, Rajagopala SV. Children with eosinophilic esophagitis non-responsive to combination therapy have distinct esophageal transcriptomic and microbiome profile. Allergy 2024; 79:2798-2811. [PMID: 38993131 PMCID: PMC11528550 DOI: 10.1111/all.16208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 04/22/2024] [Accepted: 06/03/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND A combination of proton-pump inhibitors (PPI) and topical steroids (TS) is used to treat children with eosinophilic esophagitis (EoE). However, a subset of children do not respond to this combination therapy. We aimed to identify the esophageal transcriptional, cell composition, and microbial differences between the non-responders (EoE-PPI-TSnr; n = 7) and responders (EoE-PPI-TSr; n = 7) to the combination therapy for EoE and controls (n = 9) using metatranscriptomics. METHODS Differential gene expression analysis was used to identify transcriptional differences, validated using the EoE diagnostic panel (EDP). Deconvolution analysis was performed to identify differences in their cell type composition. Microbiome analysis was conducted from esophageal biopsies RNAseq data, and microbial abundance was correlated with esophageal gene expression. RESULTS In all, 3164 upregulated and 3154 downregulated genes distinguished EoE-PPI-TSnr from EoE-PPI-TSr. Eosinophilic inflammatory response, cytokine signaling, and collagen formation pathways were significantly upregulated in EoE-PPI-TSnr. There was a 56% overlap in dysregulated genes between EoE-PPI-TSnr and EDP, with a perfect agreement in the directionality of modulation. Eosinophils, dendritic cells (DCs), immature DCs, megakaryocytic-erythroid progenitors, and T helper type 1 cells were significantly higher in EoE-PPI-TSnr. There was no significant difference in microbiome diversity. The relative abundance of Fusobacterium sp. and Acinetobacter sp. notably differed in EoE-PPI-TSnr and correlated with the key pathways. CONCLUSION Our results provide critical insights into the molecular, cellular, and microbial factors associated with the lack of response to PPI and TS combination therapy in children with EoE. This study advances our understanding of the pathobiology of EoE while guiding personalized treatment strategies.
Collapse
Affiliation(s)
- Girish Hiremath
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Vanderbilt Children's Hospital, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yash Choksi
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Tennesee Valley Health System, Veteran's Affairs, Nashville, Tennessee, USA
| | - Hernan Correa
- Division of Pathology, Vanderbilt Children's Hospital, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Justin Jacobse
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Willem-Alexander Children’s Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Suman R. Das
- Department of Medicine, Division of Infectious Disease, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Siyuan Ma
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jeremy A. Goettel
- Department of Pathology, Microbiology and Immunology, Division of Molecular Pathogenesis, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Seesandra V. Rajagopala
- Department of Medicine, Division of Infectious Disease, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
9
|
Abonia JP, Rudman Spergel AK, Hirano I, Shoda T, Zhang X, Martin LJ, Mukkada VA, Putnam PE, Blacklidge M, Neilson D, Collins MH, Yang GY, Capocelli KE, Foote H, Eby M, Dong S, Aceves SS, Rothenberg ME. Losartan Treatment Reduces Esophageal Eosinophilic Inflammation in a Subset of Eosinophilic Esophagitis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:2427-2438.e3. [PMID: 39059581 PMCID: PMC11552403 DOI: 10.1016/j.jaip.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) is a chronic, food antigen-driven esophageal disorder. Connective tissue disorders (CTDs) and esophageal connective tissue alterations are associated with EoE. Therefore, angiotensin II type 1 receptor blockade with losartan, an accepted CTD treatment, is a potential EoE treatment. OBJECTIVE We evaluated losartan's effects on esophageal pathology, symptoms, and safety in patients with EoE with and without a CTD in an open-label, non-placebo controlled multisite study. METHODS Fifteen participants with EoE, aged 5 to 23 years, underwent treatment with per-protocol titrated doses of losartan in an open-label, 16-week pilot trial. Losartan was added to standard of care therapy and 14 patients completed the study. Eosinophil counts served as the primary end point, whereas we also assessed the EoE Histology Scoring System, Endoscopic Reference Scores, EoE Diagnostic Panel, and patient-reported outcomes. RESULTS Esophageal eosinophilia was not reduced after losartan. The peak eosinophil count was not reduced for the proximal (median [interquartile range]: -3 [-22 to 3]; P = .49) and distal esophagus (median [interquartile range]: -18 [-39 to -1]; P = .23). There were no differences in losartan response in EoE with or without CTD (n = 7 and 8, respectively). Regardless, in a small subset of four participants esophageal eosinophilia was resolved with a concomitant reduction in EoE Histology Scoring System score and Endoscopic Reference Score. Across all subjects, the Pediatric EoE Symptom Score, Pediatric Quality of Life Inventory EoE Module, and EoE Diagnostic Panel improved after losartan (P < .05). CONCLUSIONS Losartan treatment was associated with improved patient-reported outcome scores and EoE Diagnostic Panel biomarkers although without a reduction in esophageal eosinophilia overall. A subset of patients demonstrated improved histopathologic and endoscopic features that could not be tied to a specific feature predicting response to treatment.
Collapse
Affiliation(s)
- J Pablo Abonia
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Amanda K Rudman Spergel
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, Md
| | - Ikuo Hirano
- Division of Gastroenterology and Hepatology, Northwestern University, Feinberg School of Medicine, Chicago, Ill
| | - Tetsuo Shoda
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Xue Zhang
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Lisa J Martin
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Vincent A Mukkada
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Philip E Putnam
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Melodie Blacklidge
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Derek Neilson
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Margaret H Collins
- Division of Pathology and Laboratory Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Guang-Yu Yang
- Division of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, Ill
| | | | - Heather Foote
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Mike Eby
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Stephanie Dong
- Division of Allergy Immunology, Rady Children's Hospital, University of California, San Diego, San Diego, Calif
| | - Seema S Aceves
- Division of Allergy Immunology, Rady Children's Hospital, University of California, San Diego, San Diego, Calif.
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
10
|
Melhem H, Niess JH. Eosinophilic Esophagitis and Inflammatory Bowel Disease: What Are the Differences? Int J Mol Sci 2024; 25:8534. [PMID: 39126102 PMCID: PMC11313654 DOI: 10.3390/ijms25158534] [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: 06/18/2024] [Revised: 07/19/2024] [Accepted: 07/20/2024] [Indexed: 08/12/2024] Open
Abstract
Eosinophilic esophagitis (EoE) and inflammatory bowel disease (IBD) are chronic inflammatory disorders of the gastrointestinal tract, with EoE predominantly provoked by food and aeroallergens, whereas IBD is driven by a broader spectrum of immunopathological and environmental triggers. This review presents a comprehensive comparison of the pathophysiological and therapeutic strategies for EoE and IBD. We examine the current understanding of their underlying mechanisms, particularly the interplay between environmental factors and genetic susceptibility. A crucial element in both diseases is the integrity of the epithelial barrier, whose disruption plays a central role in their pathogenesis. The involvement of eosinophils, mast cells, B cells, T cells, dendritic cells, macrophages, and their associated cytokines is examined, highlighting the importance of targeting cytokine signaling pathways to modulate immune-epithelial interactions. We propose that advances in computation tools will uncover the significance of G-protein coupled receptors (GPCRs) in connecting immune and epithelial cells, leading to novel therapies for EoE and IBD.
Collapse
Affiliation(s)
- Hassan Melhem
- Gastroenterology Group, Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Jan Hendrik Niess
- Gastroenterology Group, Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
- Department of Gastroenterology and Hepatology, University Digestive Healthcare Center, Clarunis, 4002 Basel, Switzerland
| |
Collapse
|
11
|
Kheshtchin N, Kanannejad Z, Ghahramani Z, Esmaeilzadeh H, Sepahi N. Balancing immune responses: regulatory cells in eosinophilic gastrointestinal disorders. Front Immunol 2024; 15:1372009. [PMID: 39136025 PMCID: PMC11317405 DOI: 10.3389/fimmu.2024.1372009] [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: 01/17/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024] Open
Abstract
Eosinophilic gastrointestinal disorders (EGIDs) are a group of conditions characterized by an abnormal accumulation of eosinophils in the gastrointestinal tract, leading to inflammation and tissue damage. Regulatory cells are a subset of immune cells that are crucial in maintaining the balance of the immune system and preventing the occurrence of autoimmune diseases. In EGIDs, regulatory cells are believed to play a key role in controlling the immune response and overseeing the growth and activation of eosinophils in the gastrointestinal tract. There is evidence indicating that regulatory T cells (Tregs) and regulatory eosinophils may play a role in suppressing the inflammatory response in EGIDs. Regulatory eosinophils are a subgroup of eosinophils that possess an anti-inflammatory role. Recent studies have shown that enhancing the number or effectiveness of regulatory eosinophils can reduce the severity of EGIDs. Regulatory eosinophils dampen inflammation through their regulatory mediators, such as galectin-10 and growth factor beta (TGF-β), which promote Treg expansion and inhibit effector T cell function. Further research on regulatory cells in EGIDs may have significant implications for the advancement of novel therapies for these uncommon and intricate disorders. The aim of this review is to provide complete view of the immune responses connected to EGIDs, examine the regulatory cells that control these responses, and evaluate their potential as therapeutic targets for EGID treatment.
Collapse
Affiliation(s)
- Nassim Kheshtchin
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Kanannejad
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Ghahramani
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Esmaeilzadeh
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pediatrics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Najmeh Sepahi
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
Patel H, Sheikh MS, Huang Y. ECRG2/SPINK7 Tumor Suppressor as Modulator of DNA Damage Response. Int J Mol Sci 2024; 25:5854. [PMID: 38892042 PMCID: PMC11172197 DOI: 10.3390/ijms25115854] [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: 04/29/2024] [Revised: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
Esophageal Cancer-Related Gene 2 (ECRG2), also known as Serine Peptidase Inhibitor Kazal type 7 (SPINK7), is a novel tumor suppressor gene from the SPINK family of genes that exhibits anticancer potential. ECRG2 was originally identified during efforts to discover genes involved in esophageal tumorigenesis. ECRG2 was one of those genes whose expression was absent or reduced in primary human esophageal cancers. Additionally, absent or reduced ECRG2 expression was also noted in several other types of human malignancies. ECRG2 missense mutations were identified in various primary human cancers. It was reported that a cancer-derived ECRG2 mutant (valine to glutamic acid at position 30) failed to induce cell death and caspase activation triggered by DNA-damaging anticancer drugs. Furthermore, ECRG2 suppressed cancer cell proliferation in cultured cells and grafted tumors in animals and inhibited cancer cell migration/invasion and metastasis. ECRG2 also was identified as a negative regulator of Hu-antigen R (HuR), an oncogenic RNA-binding protein that is known to regulate mRNA stability and the expression of transcripts corresponding to many cancer-related genes. ECRG2 function is important also for the regulation of inflammatory responses and the maintenance of epithelial barrier integrity in the esophagus. More recently, ECRG2 was discovered as one of the newest members of the pro-apoptotic transcriptional targets of p53. Two p53-binding sites (BS-1 and BS-2) were found within the proximal region of the ECRG2 gene promoter; the treatment of DNA-damaging agents in cancer cells significantly increased p53 binding to the ECRG2 promoter and triggered a strong ECRG2 promoter induction following DNA damage. Further, the genetic depletion of ECRG2 expression significantly impeded apoptotic cell death induced by DNA damage and wild-type p53 in cancer cells. These findings suggest that the loss of ECRG2 expression, commonly observed in human cancers, could play important roles in conferring anticancer drug resistance in human cancers. Thus, ECRG2 is a novel regulator in DNA damage-induced cell death that may also be a potential target for anticancer therapeutics.
Collapse
Affiliation(s)
| | - M. Saeed Sheikh
- Department of Pharmacology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA;
| | - Ying Huang
- Department of Pharmacology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA;
| |
Collapse
|
13
|
Baglivo I, Quaranta VN, Dragonieri S, Colantuono S, Menzella F, Selvaggio D, Carpagnano GE, Caruso C. The New Paradigm: The Role of Proteins and Triggers in the Evolution of Allergic Asthma. Int J Mol Sci 2024; 25:5747. [PMID: 38891935 PMCID: PMC11171572 DOI: 10.3390/ijms25115747] [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: 04/18/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Epithelial barrier damage plays a central role in the development and maintenance of allergic inflammation. Rises in the epithelial barrier permeability of airways alter tissue homeostasis and allow the penetration of allergens and other external agents. Different factors contribute to barrier impairment, such as eosinophilic infiltration and allergen protease action-eosinophilic cationic proteins' effects and allergens' proteolytic activity both contribute significantly to epithelial damage. In the airways, allergen proteases degrade the epithelial junctional proteins, allowing allergen penetration and its uptake by dendritic cells. This increase in allergen-immune system interaction induces the release of alarmins and the activation of type 2 inflammatory pathways, causing or worsening the main symptoms at the skin, bowel, and respiratory levels. We aim to highlight the molecular mechanisms underlying allergenic protease-induced epithelial barrier damage and the role of immune response in allergic asthma onset, maintenance, and progression. Moreover, we will explore potential clinical and radiological biomarkers of airway remodeling in allergic asthma patients.
Collapse
Affiliation(s)
- Ilaria Baglivo
- Centro Malattie Apparato Digerente (CEMAD) Digestive Disease Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Vitaliano Nicola Quaranta
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, Section of Respiratory Disease, University “Aldo Moro” of Bari, 70121 Bari, Italy (S.D.)
| | - Silvano Dragonieri
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, Section of Respiratory Disease, University “Aldo Moro” of Bari, 70121 Bari, Italy (S.D.)
| | - Stefania Colantuono
- Unità Operativa Semplice Dipartimentale Day Hospital (UOSD DH) Medicina Interna e Malattie dell’Apparato Digerente, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Francesco Menzella
- Pulmonology Unit, S. Valentino Hospital-AULSS2 Marca Trevigiana, 31100 Treviso, Italy
| | - David Selvaggio
- UOS di Malattie dell’Apparato Respiratorio Ospedale Cristo Re, 00167 Roma, Italy
| | - Giovanna Elisiana Carpagnano
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, Section of Respiratory Disease, University “Aldo Moro” of Bari, 70121 Bari, Italy (S.D.)
| | - Cristiano Caruso
- Unità Operativa Semplice Dipartimentale Day Hospital (UOSD DH) Medicina Interna e Malattie dell’Apparato Digerente, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| |
Collapse
|
14
|
Kennedy KV, Muir AB, Ruffner MA. Pathophysiology of Eosinophilic Esophagitis. Immunol Allergy Clin North Am 2024; 44:119-128. [PMID: 38575212 DOI: 10.1016/j.iac.2023.12.001] [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] [Indexed: 04/06/2024]
Abstract
Eosinophilic esophagitis (EoE) is a chronic, progressive immune-mediated disease associated with antigen-driven type 2 inflammation and symptoms of esophageal dysfunction. Research over the last 2 decades has dramatically furthered our understanding of the complex interplay between genetics, environmental exposures, and cellular and molecular interactions involved in EoE. This review provides an overview of our current understanding of EoE pathogenesis.
Collapse
Affiliation(s)
- Kanak V Kennedy
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, The Children's Hospital of Philadelphia, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Amanda B Muir
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Abramson Research Center 902E, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| | - Melanie A Ruffner
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Abramson Research Center 902E, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA; Division of Pediatric Allergy and Immunology, Department of Pediatrics, The Children's Hospital of Philadelphia
| |
Collapse
|
15
|
Ye Z, Jiang P, Zhu Q, Pei Z, Hu Y, Zhao G. Molecular stratification of the human fetal vaginal epithelium by spatial transcriptome analysis. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1521-1536. [PMID: 38666303 PMCID: PMC11612642 DOI: 10.3724/abbs.2024063] [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: 01/31/2024] [Accepted: 03/27/2024] [Indexed: 11/01/2024] Open
Abstract
The human vaginal epithelium is a crucial component of numerous reproductive processes and serves as a vital protective barrier against pathogenic invasion. Despite its significance, a comprehensive exploration of its molecular profiles, including molecular expression and distribution across its multiple layers, has not been performed. In this study, we perform a spatial transcriptomic analysis within the vaginal wall of human fetuses to fill this knowledge gap. We successfully categorize the vaginal epithelium into four distinct zones based on transcriptomic profiles and anatomical features. This approach reveals unique transcriptomic signatures within these regions, allowing us to identify differentially expressed genes and uncover novel markers for distinct regions of the vaginal epithelium. Additionally, our findings highlight the varied expressions of keratin ( KRT) genes across different zones of the vaginal epithelium, with a gradual shift in expression patterns observed from the basal layer to the surface/superficial layer. This suggests a potential differentiation trajectory of the human vaginal epithelium, shedding light on the dynamic nature of this tissue. Furthermore, abundant biological processes are found to be enriched in the basal zone by KEGG pathway analysis, indicating an active state of the basal zone cells. Subsequently, the expressions of latent stem cell markers in the basal zone are identified. In summary, our research provides a crucial understanding of human vaginal epithelial cells and the complex mechanisms of the vaginal mucosa, with potential applications in vaginal reconstruction and drug delivery, making this atlas a valuable tool for future research in women's health and reproductive medicine.
Collapse
Affiliation(s)
- Ziying Ye
- Department of Obstetrics and GynecologyAffiliated Drum Tower HospitalMedical SchoolNanjing UniversityNanjing210008China
| | - Peipei Jiang
- Department of Obstetrics and GynecologyAffiliated Drum Tower HospitalMedical SchoolNanjing UniversityNanjing210008China
| | - Qi Zhu
- Department of Obstetrics and GynecologyAffiliated Drum Tower HospitalMedical SchoolNanjing UniversityNanjing210008China
| | - Zhongrui Pei
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjing210008China
| | - Yali Hu
- Department of Obstetrics and GynecologyAffiliated Drum Tower HospitalMedical SchoolNanjing UniversityNanjing210008China
| | - Guangfeng Zhao
- Department of Obstetrics and GynecologyAffiliated Drum Tower HospitalMedical SchoolNanjing UniversityNanjing210008China
| |
Collapse
|
16
|
Lewis NE, Aceves SS. Eosinophilic esophagitis: Shifting immune complexity beyond the eosinophil. J Allergy Clin Immunol 2024; 153:669-671. [PMID: 38262501 PMCID: PMC11288620 DOI: 10.1016/j.jaci.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 01/25/2024]
Affiliation(s)
- Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, Calif; Department of Bioengineering, University of California, San Diego, Calif
| | - Seema S Aceves
- Department of Pediatrics, University of California, San Diego, Calif; Department of Medicine, University of California, San Diego, Calif; Rady Children's Hospital, San Diego, Calif.
| |
Collapse
|
17
|
Baglivo I, Colantuono S, Lumaca A, Papa A, Gasbarrini A, Caruso C. The last step to achieve barrier damage control. Front Immunol 2024; 15:1354556. [PMID: 38415254 PMCID: PMC10897052 DOI: 10.3389/fimmu.2024.1354556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/29/2024] [Indexed: 02/29/2024] Open
Abstract
Heterogeneity characterises inflammatory diseases and different phenotypes and endotypes have been identified. Both innate and adaptive immunity contribute to the immunopathological mechanism of these diseases and barrier damage plays a prominent role triggering type 2 inflammation through the alarmins system, such as anti-Thymic Stromal Lymphopoietin (TSLP). Treatment with anti-TSLP monoclonal antibodies showed efficacy in severe asthma and clinical trials for other eosinophilic diseases are ongoing. The aim of this perspective review is to analyse current advances and future applications of TSLP inhibition to control barrier damage.
Collapse
Affiliation(s)
- Ilaria Baglivo
- Centro Malattie Apparato Digerente (CEMAD) Digestive Disease Center, Fondazione Policlinico Universitario “A. Gemelli” Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Università Cattolica del Sacro Cuore, Roma, Italy
| | - Stefania Colantuono
- Unità Operativa Semplice Dipartimentale Day Hospital (UOSD DH) Medicina Interna e Malattie dell’ApparatoDigerente, Fondazione Policlinico Universitario “A. Gemelli” Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Università Cattolica del Sacro Cuore, Roma, Italy
| | - Arianna Lumaca
- Unità Operativa Semplice Dipartimentale (UOSD) di Allergologia, Ospedale Maria Santissima (SS) Dello Splendore, Teramo, Italy
| | - Alfredo Papa
- Unità Operativa Semplice Dipartimentale Day Hospital (UOSD DH) Medicina Interna e Malattie dell’ApparatoDigerente, Fondazione Policlinico Universitario “A. Gemelli” Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Università Cattolica del Sacro Cuore, Roma, Italy
| | - Antonio Gasbarrini
- Centro Malattie Apparato Digerente (CEMAD) Digestive Disease Center, Fondazione Policlinico Universitario “A. Gemelli” Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Università Cattolica del Sacro Cuore, Roma, Italy
| | - Cristiano Caruso
- Unità Operativa Semplice Dipartimentale Day Hospital (UOSD DH) Medicina Interna e Malattie dell’ApparatoDigerente, Fondazione Policlinico Universitario “A. Gemelli” Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
18
|
Markey GE, Ryan S, Furuta GT, Menard-Katcher C, McNamee EN, Masterson JC. Hypoxia-inducible microRNA-155 negatively regulates epithelial barrier in eosinophilic esophagitis by suppressing tight junction claudin-7. FASEB J 2024; 38:e23358. [PMID: 38050671 PMCID: PMC10699209 DOI: 10.1096/fj.202301934r] [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: 10/05/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/06/2023]
Abstract
MicroRNA (miRNA)-mediated mRNA regulation directs many homeostatic and pathological processes, but how miRNAs coordinate aberrant esophageal inflammation during eosinophilic esophagitis (EoE) is poorly understood. Here, we report a deregulatory axis where microRNA-155 (miR-155) regulates epithelial barrier dysfunction by selectively constraining tight junction CLDN7 (claudin-7). MiR-155 is elevated in the esophageal epithelium of biopsies from patients with active EoE and in cell culture models. MiR-155 localization using in situ hybridization (ISH) in patient biopsies and intra-epithelial compartmentalization of miR-155 show expression predominantly within the basal epithelia. Epithelial miR-155 activity was evident through diminished target gene expression in 3D organotypic cultures, particularly in relatively undifferentiated basal cell states. Mechanistically, generation of a novel cell line with enhanced epithelial miR-155 stable overexpression induced a functionally deficient epithelial barrier in 3D air-liquid interface epithelial cultures measured by transepithelial electrical resistance (TEER). Histological assessment of 3D esophageal organoid cultures overexpressing miR-155 showed notable dilated intra-epithelial spaces. Unbiased RNA-sequencing analysis and immunofluorescence determined a defect in epithelial barrier tight junctions and revealed a selective reduction in the expression of critical esophageal tight junction molecule, claudin-7. Together, our data reveal a previously unappreciated role for miR-155 in mediating epithelial barrier dysfunction in esophageal inflammation.
Collapse
Affiliation(s)
- Gary E Markey
- Allergy, Inflammation & Remodelling Research Laboratory, Kathleen Lonsdale Institute for Human Health Research, Department of Biology, National University of Ireland Maynooth, Co. Kildare, Ireland
| | - Sinéad Ryan
- Allergy, Inflammation & Remodelling Research Laboratory, Kathleen Lonsdale Institute for Human Health Research, Department of Biology, National University of Ireland Maynooth, Co. Kildare, Ireland
| | - Glenn T Furuta
- Gastrointestinal Eosinophilic Diseases Program, Digestive Health Institute, Children’s Hospital Colorado, Department of Pediatrics, University of Colorado School of Medicine, CO, USA
| | - Calies Menard-Katcher
- Gastrointestinal Eosinophilic Diseases Program, Digestive Health Institute, Children’s Hospital Colorado, Department of Pediatrics, University of Colorado School of Medicine, CO, USA
| | - Eoin N McNamee
- Mucosal Immunology Research Laboratory, Kathleen Lonsdale Institute for Human Health Research, Department of Biology, National University of Ireland Maynooth, Co. Kildare, Ireland
| | - Joanne C Masterson
- Allergy, Inflammation & Remodelling Research Laboratory, Kathleen Lonsdale Institute for Human Health Research, Department of Biology, National University of Ireland Maynooth, Co. Kildare, Ireland
- Gastrointestinal Eosinophilic Diseases Program, Digestive Health Institute, Children’s Hospital Colorado, Department of Pediatrics, University of Colorado School of Medicine, CO, USA
| |
Collapse
|
19
|
Li H, Liu P, Zhang B, Yuan Z, Guo M, Zou X, Qian Y, Deng S, Zhu L, Cao X, Tao T, Xia S, Bao X, Xu Y. Acute ischemia induces spatially and transcriptionally distinct microglial subclusters. Genome Med 2023; 15:109. [PMID: 38082331 PMCID: PMC10712107 DOI: 10.1186/s13073-023-01257-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Damage in the ischemic core and penumbra after stroke affects patient prognosis. Microglia immediately respond to ischemic insult and initiate immune inflammation, playing an important role in the cellular injury after stroke. However, the microglial heterogeneity and the mechanisms involved remain unclear. METHODS We first performed single-cell RNA-sequencing (scRNA-seq) and spatial transcriptomics (ST) on middle cerebral artery occlusion (MCAO) mice from three time points to determine stroke-associated microglial subclusters and their spatial distributions. Furthermore, the expression of microglial subcluster-specific marker genes and the localization of different microglial subclusters were verified on MCAO mice through RNAscope and immunofluorescence. Gene set variation analysis (GSVA) was performed to reveal functional characteristics of microglia sub-clusters. Additionally, ingenuity pathway analysis (IPA) was used to explore upstream regulators of microglial subclusters, which was confirmed by immunofluorescence, RT-qPCR, shRNA-mediated knockdown, and targeted metabolomics. Finally, the infarct size, neurological deficits, and neuronal apoptosis were evaluated in MCAO mice after manipulation of specific microglial subcluster. RESULTS We discovered stroke-associated microglial subclusters in the brains of MCAO mice. We also identified novel marker genes of these microglial subclusters and defined these cells as ischemic core-associated (ICAM) and ischemic penumbra-associated (IPAM) microglia, according to their spatial distribution. ICAM, induced by damage-associated molecular patterns, are probably fueled by glycolysis, and exhibit increased pro-inflammatory cytokines and chemokines production. BACH1 is a key transcription factor driving ICAM generation. In contrast, glucocorticoids, which are enriched in the penumbra, likely trigger IPAM formation, which are presumably powered by the citrate cycle and oxidative phosphorylation and are characterized by moderate pro-inflammatory responses, inflammation-alleviating metabolic features, and myelinotrophic properties. CONCLUSIONS ICAM could induce excessive neuroinflammation, aggravating brain injury, whereas IPAM probably exhibit neuroprotective features, which could be essential for the homeostasis and survival of cells in the penumbra. Our findings provide a biological basis for targeting specific microglial subclusters as a potential therapeutic strategy for ischemic stroke.
Collapse
Affiliation(s)
- Huiya Li
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Pinyi Liu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Bing Zhang
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Zengqiang Yuan
- The Brain Science Centre, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
- Centre of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Mengdi Guo
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Xinxin Zou
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Yi Qian
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Shiji Deng
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Liwen Zhu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Xiang Cao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Tao Tao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Shengnan Xia
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Xinyu Bao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Yun Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China.
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China.
- Nanjing Neurology Medical Centre, Nanjing, 210008, China.
| |
Collapse
|
20
|
Massironi S, Mulinacci G, Gallo C, Elvevi A, Danese S, Invernizzi P, Vespa E. Mechanistic Insights into Eosinophilic Esophagitis: Therapies Targeting Pathophysiological Mechanisms. Cells 2023; 12:2473. [PMID: 37887317 PMCID: PMC10605530 DOI: 10.3390/cells12202473] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023] Open
Abstract
Eosinophilic esophagitis (EoE) is a chronic inflammatory disease characterized by eosinophilic infiltration of the esophagus. It arises from a complex interplay of genetic predisposition (susceptibility loci), environmental triggers (allergens and dietary antigens), and a dysregulated immune response, mainly mediated by type 2 T helper cell (Th2)-released cytokines, such as interleukin (IL)-4, IL-5, and IL-13. These cytokines control eosinophil recruitment and activation as well as tissue remodeling, contributing to the characteristic features of EoE. The pathogenesis of EoE includes epithelial barrier dysfunction, mast cell activation, eosinophil degranulation, and fibrosis. Epithelial barrier dysfunction allows allergen penetration and promotes immune cell infiltration, thereby perpetuating the inflammatory response. Mast cells release proinflammatory mediators and promote eosinophil recruitment and the release of cytotoxic proteins and cytokines, causing tissue damage and remodeling. Prolonged inflammation can lead to fibrosis, resulting in long-term complications such as strictures and dysmotility. Current treatment options for EoE are limited and mainly focus on dietary changes, proton-pump inhibitors, and topical corticosteroids. Novel therapies targeting key inflammatory pathways, such as monoclonal antibodies against IL-4, IL-5, and IL-13, are emerging in clinical trials. A deeper understanding of the complex pathogenetic mechanisms behind EoE will contribute to the development of more effective and personalized therapeutic strategies.
Collapse
Affiliation(s)
- Sara Massironi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy; (G.M.); (C.G.); (A.E.)
- School of Medicine and Surgery, University of Milano-Bicocca, 20125 Milan, Italy
| | - Giacomo Mulinacci
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy; (G.M.); (C.G.); (A.E.)
- School of Medicine and Surgery, University of Milano-Bicocca, 20125 Milan, Italy
| | - Camilla Gallo
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy; (G.M.); (C.G.); (A.E.)
- School of Medicine and Surgery, University of Milano-Bicocca, 20125 Milan, Italy
| | - Alessandra Elvevi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy; (G.M.); (C.G.); (A.E.)
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Pietro Invernizzi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy; (G.M.); (C.G.); (A.E.)
- School of Medicine and Surgery, University of Milano-Bicocca, 20125 Milan, Italy
| | - Edoardo Vespa
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, 20132 Milan, Italy
| |
Collapse
|
21
|
Marasco G, Visaggi P, Vassallo M, Fiocca M, Cremon C, Barbaro MR, De Bortoli N, Bellini M, Stanghellini V, Savarino EV, Barbara G. Current and Novel Therapies for Eosinophilic Gastrointestinal Diseases. Int J Mol Sci 2023; 24:15165. [PMID: 37894846 PMCID: PMC10607071 DOI: 10.3390/ijms242015165] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/01/2023] [Accepted: 10/02/2023] [Indexed: 10/29/2023] Open
Abstract
Eosinophilic gastrointestinal diseases (EGIDs) are an emerging group of pathological entities characterized by an eosinophil-predominant infiltration of different tracts of the gut in the absence of secondary causes of eosinophilia. According to the specific tract of the gut involved, EGIDs can be classified into eosinophilic esophagitis (EoE), eosinophilic gastritis (EoG), eosinophilic enteritis (EoN), and eosinophilic colitis (EoC). The epidemiology of EGIDs is evolving rapidly. EoE, once considered a rare disease, now has an incidence and prevalence of 7.7 new cases per 100,000 inhabitants per years and 34.4 cases per 100,000 inhabitants per year, respectively. Fewer data are available regarding non-EoE EGIDs, whose prevalence are estimated to range between 2.1 and 17.6 in 100,000 individuals, depending on age, sex, and ethnicity. Diagnosis requires the presence of suggestive symptoms, endoscopic biopsies showing abnormal values of eosinophils infiltrating the gut, and exclusion of secondary causes of eosinophilia. EoE typically presents with dysphagia and episodes of food bolus impactions, while EoG, EoN, and EoC may all present with abdominal pain and diarrhea, with or without other non-specific symptoms. In addition, although different EGIDs are currently classified as different entities, there may be overlap between different diseases in the same patient. Despite EGIDs being relatively novel pathological entities, the research on possible treatments is rapidly growing. In this regard, several randomized controlled trials are currently ongoing to investigate novel molecules, including ad-hoc steroid formulations, immunosuppressants, and mostly monoclonal antibodies that target the specific molecular mediators of EGIDs. This narrative review provides an up-to-date overview of available and investigational drugs for different EGIDs.
Collapse
Affiliation(s)
- Giovanni Marasco
- IRCCS Azienda Ospedaliero, Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.M.); (M.V.); (M.F.); (C.C.); (M.R.B.); (V.S.)
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti, 9, 40138 Bologna, Italy
| | - Pierfrancesco Visaggi
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Risorgimento 36, 56126 Pisa, Italy; (P.V.); (N.D.B.); (M.B.)
| | - Mariagiulia Vassallo
- IRCCS Azienda Ospedaliero, Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.M.); (M.V.); (M.F.); (C.C.); (M.R.B.); (V.S.)
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti, 9, 40138 Bologna, Italy
| | - Miriam Fiocca
- IRCCS Azienda Ospedaliero, Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.M.); (M.V.); (M.F.); (C.C.); (M.R.B.); (V.S.)
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti, 9, 40138 Bologna, Italy
| | - Cesare Cremon
- IRCCS Azienda Ospedaliero, Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.M.); (M.V.); (M.F.); (C.C.); (M.R.B.); (V.S.)
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti, 9, 40138 Bologna, Italy
| | - Maria Raffaella Barbaro
- IRCCS Azienda Ospedaliero, Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.M.); (M.V.); (M.F.); (C.C.); (M.R.B.); (V.S.)
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti, 9, 40138 Bologna, Italy
| | - Nicola De Bortoli
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Risorgimento 36, 56126 Pisa, Italy; (P.V.); (N.D.B.); (M.B.)
| | - Massimo Bellini
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Risorgimento 36, 56126 Pisa, Italy; (P.V.); (N.D.B.); (M.B.)
| | - Vincenzo Stanghellini
- IRCCS Azienda Ospedaliero, Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.M.); (M.V.); (M.F.); (C.C.); (M.R.B.); (V.S.)
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti, 9, 40138 Bologna, Italy
| | - Edoardo Vincenzo Savarino
- Gastroenterology Unit, Azienda Ospedale Università of Padua, Via Giustiniani 2, 35128 Padua, Italy;
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Giustiniani 2, 35128 Padua, Italy
| | - Giovanni Barbara
- IRCCS Azienda Ospedaliero, Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.M.); (M.V.); (M.F.); (C.C.); (M.R.B.); (V.S.)
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti, 9, 40138 Bologna, Italy
| |
Collapse
|
22
|
Hill DA, Muir AB. The immune-epithelial interface in eosinophilic esophagitis: a conversation. FRONTIERS IN ALLERGY 2023; 4:1270581. [PMID: 37854541 PMCID: PMC10579787 DOI: 10.3389/falgy.2023.1270581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/19/2023] [Indexed: 10/20/2023] Open
Affiliation(s)
- David A. Hill
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics and Institute for Immunology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Amanda B. Muir
- Division of Gastroenterology, Children's Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
23
|
Kleuskens MTA, Bek MK, Al Halabi Y, Blokhuis BRJ, Diks MAP, Haasnoot ML, Garssen J, Bredenoord AJ, van Esch BCAM, Redegeld FA. Mast cells disrupt the function of the esophageal epithelial barrier. Mucosal Immunol 2023; 16:567-577. [PMID: 37302713 DOI: 10.1016/j.mucimm.2023.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/24/2023] [Accepted: 06/05/2023] [Indexed: 06/13/2023]
Abstract
Mast cells (MCs) accumulate in the epithelium of patients with eosinophilic esophagitis (EoE), an inflammatory disorder characterized by extensive esophageal eosinophilic infiltration. Esophageal barrier dysfunction plays an important role in the pathophysiology of EoE. We hypothesized that MCs contribute to the observed impaired esophageal epithelial barrier. Herein, we demonstrate that coculture of differentiated esophageal epithelial cells with immunoglobulin E-activated MCs significanly decreased epithelial resistance by 30% and increased permeability by 22% compared with non-activated MCs. These changes were associated with decreased messenger RNA expression of barrier proteins filaggrin, desmoglein-1 and involucrin, and antiprotease serine peptidase inhibitor kazal type 7. Using targeted proteomics, we detected various cytokines in coculture supernatants, most notably granulocyte-macrophage colony-stimulating factor and oncostatin M (OSM). OSM expression was increased by 12-fold in active EoE and associated with MC marker genes. Furthermore, OSM receptor-expressing esophageal epithelial cells were found in the esophageal tissue of patients with EoE, suggesting that the epithelial cells may respond to OSM. Stimulation of esophageal epithelial cells with OSM resulted in a dose-dependent decrease in barrier function and expression of filaggrin and desmoglein-1 and an increase in protease calpain-14. Taken together, these data suggest a role for MCs in decreasing esophageal epithelial barrier function in EoE, which may in part be mediated by OSM.
Collapse
Affiliation(s)
- Mirelle T A Kleuskens
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Marie K Bek
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Youmna Al Halabi
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Bart R J Blokhuis
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Mara A P Diks
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Maria L Haasnoot
- Department of Gastroenterology & Hepatology, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands; Danone Nutricia Research, Utrecht, The Netherlands
| | - Albert J Bredenoord
- Department of Gastroenterology & Hepatology, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - Betty C A M van Esch
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands; Danone Nutricia Research, Utrecht, The Netherlands
| | - Frank A Redegeld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
24
|
Janarthanam R, Kuang FL, Zalewski A, Amsden K, Wang MY, Ostilla L, Keeley K, Hirano I, Kagalwalla A, Wershil BK, Gonsalves N, Wechsler JB. Bulk T-cell receptor sequencing confirms clonality in pediatric eosinophilic esophagitis and identifies a food-specific repertoire. Allergy 2023; 78:2487-2496. [PMID: 37203302 PMCID: PMC10768854 DOI: 10.1111/all.15773] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 03/15/2023] [Accepted: 04/04/2023] [Indexed: 05/20/2023]
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) involves a chronic immune-mediated response to dietary antigens. Recent work identifies T-cell clonality in children with EoE, however, it is unknown whether this is true in adults or whether there is a restricted food-specific T-cell repertoire. We sought to confirm T-cell receptor (TCR) clonality in EoE and assess for differences with specific food triggers. METHODS Bulk TCR sequencing was performed on mRNA isolated from esophageal biopsies obtained from adults and children with EoE (n = 15) who had food triggers confirmed by endoscopic evaluation. Non-EoE adult and pediatric controls (n = 10) were included. Differences in TCR clonality by disease and treatment status were assessed. Shared and similar V-J-CDR3s were assessed based on specific food triggers. RESULTS Active EoE biopsies from children but not adults displayed decreased unique TCRα/β clonotypes and increased relative abundance of TCRs comprising >1% of the total compared to non-EoE controls and paired inactive EoE samples. Among patients in which baseline, post diet elimination, and food trigger reintroduction samples (n = 6) were obtained, we observed ~1% of TCRs were shared only between pre-diet elimination and trigger reintroduction. Patients with a shared EoE trigger (milk) had a greater degree of shared and similar TCRs compared to patients with differing triggers (seafood, wheat, egg, soy). CONCLUSION We confirmed relative clonality in children but not adults with active EoE and identified potential food-specific TCRs, particularly for milk-triggered EoE. Further studies are needed to better identify the broad TCR repertoire relevant to food triggers.
Collapse
Affiliation(s)
- Rethavathi Janarthanam
- Division of Gastroenterology, Hepatology & Nutrition, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg school of Medicine, Chicago, Illinois, USA
| | - Fei Li Kuang
- Division of Allergy-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Angelika Zalewski
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Katie Amsden
- Division of Gastroenterology, Hepatology & Nutrition, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg school of Medicine, Chicago, Illinois, USA
| | - Ming-Yu Wang
- Division of Gastroenterology, Hepatology & Nutrition, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg school of Medicine, Chicago, Illinois, USA
| | - Lorena Ostilla
- Division of Gastroenterology, Hepatology & Nutrition, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg school of Medicine, Chicago, Illinois, USA
| | - Kaitlyn Keeley
- Division of Gastroenterology, Hepatology & Nutrition, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg school of Medicine, Chicago, Illinois, USA
| | - Ikuo Hirano
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Amir Kagalwalla
- Department of Pediatrics, John H. Stroger Cook County Hospital of Chicago, Chicago, Illinois, USA
| | - Barry K Wershil
- Division of Gastroenterology, Hepatology & Nutrition, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg school of Medicine, Chicago, Illinois, USA
| | - Nirmala Gonsalves
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Joshua B Wechsler
- Division of Gastroenterology, Hepatology & Nutrition, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg school of Medicine, Chicago, Illinois, USA
- Division of Allergy-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
25
|
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
|
26
|
Soh WT, Zhang J, Hollenberg MD, Vliagoftis H, Rothenberg ME, Sokol CL, Robinson C, Jacquet A. Protease allergens as initiators-regulators of allergic inflammation. Allergy 2023; 78:1148-1168. [PMID: 36794967 PMCID: PMC10159943 DOI: 10.1111/all.15678] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 02/05/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023]
Abstract
Tremendous progress in the last few years has been made to explain how seemingly harmless environmental proteins from different origins can induce potent Th2-biased inflammatory responses. Convergent findings have shown the key roles of allergens displaying proteolytic activity in the initiation and progression of the allergic response. Through their propensity to activate IgE-independent inflammatory pathways, certain allergenic proteases are now considered as initiators for sensitization to themselves and to non-protease allergens. The protease allergens degrade junctional proteins of keratinocytes or airway epithelium to facilitate allergen delivery across the epithelial barrier and their subsequent uptake by antigen-presenting cells. Epithelial injuries mediated by these proteases together with their sensing by protease-activated receptors (PARs) elicit potent inflammatory responses resulting in the release of pro-Th2 cytokines (IL-6, IL-25, IL-1β, TSLP) and danger-associated molecular patterns (DAMPs; IL-33, ATP, uric acid). Recently, protease allergens were shown to cleave the protease sensor domain of IL-33 to produce a super-active form of the alarmin. At the same time, proteolytic cleavage of fibrinogen can trigger TLR4 signaling, and cleavage of various cell surface receptors further shape the Th2 polarization. Remarkably, the sensing of protease allergens by nociceptive neurons can represent a primary step in the development of the allergic response. The goal of this review is to highlight the multiple innate immune mechanisms triggered by protease allergens that converge to initiate the allergic response.
Collapse
Affiliation(s)
- Wai Tuck Soh
- Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Jihui Zhang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Morley D. Hollenberg
- Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Harissios Vliagoftis
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine & Dentistry, and Alberta Respiratory Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Marc E. Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Caroline L. Sokol
- Division of Rheumatology, Allergy and Immunology, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Clive Robinson
- Institute for Infection and Immunity, St George’s University of London, London, UK
| | - Alain Jacquet
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
27
|
Brusilovsky M, Rochman M, Shoda T, Kotliar M, Caldwell JM, Mack LE, Besse JA, Chen X, Weirauch MT, Barski A, Rothenberg ME. Vitamin D receptor and STAT6 interactome governs oesophageal epithelial barrier responses to IL-13 signalling. Gut 2023; 72:834-845. [PMID: 35918104 PMCID: PMC9892355 DOI: 10.1136/gutjnl-2022-327276] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 07/14/2022] [Indexed: 02/04/2023]
Abstract
OBJECTIVE The contribution of vitamin D (VD) deficiency to the pathogenesis of allergic diseases remains elusive. We aimed to define the impact of VD on oesophageal allergic inflammation. DESIGN We assessed the genomic distribution and function of VD receptor (VDR) and STAT6 using histology, molecular imaging, motif discovery and metagenomic analysis. We examined the role of VD supplementation in oesophageal epithelial cells, in a preclinical model of IL-13-induced oesophageal allergic inflammation and in human subjects with eosinophilic oesophagitis (EoE). RESULTS VDR response elements were enriched in oesophageal epithelium, suggesting enhanced VDR binding to functional gene enhancer and promoter regions. Metagenomic analysis showed that VD supplementation reversed dysregulation of up to 70% of the transcriptome and epigenetic modifications (H3K27Ac) induced by IL-13 in VD-deficient cells, including genes encoding the transcription factors HIF1A and SMAD3, endopeptidases (SERPINB3) and epithelial-mesenchymal transition mediators (TGFBR1, TIAM1, SRC, ROBO1, CDH1). Molecular imaging and chromatin immunoprecipitation showed VDR and STAT6 colocalisation within the regulatory regions of the affected genes, suggesting that VDR and STAT6 interactome governs epithelial tissue responses to IL-13 signalling. Indeed, VD supplementation reversed IL-13-induced epithelial hyperproliferation, reduced dilated intercellular spaces and barrier permeability, and improved differentiation marker expression (filaggrin, involucrin). In a preclinical model of IL-13-mediated oesophageal allergic inflammation and in human EoE, VD levels inversely associated with severity of oesophageal eosinophilia and epithelial histopathology. CONCLUSIONS Collectively, these findings identify VD as a natural IL-13 antagonist with capacity to regulate the oesophageal epithelial barrier functions, providing a novel therapeutic entry point for type 2 immunity-related diseases.
Collapse
Affiliation(s)
- Michael Brusilovsky
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Mark Rochman
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Tetsuo Shoda
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Michael Kotliar
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Julie M Caldwell
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Lydia E Mack
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - John A Besse
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Xiaoting Chen
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Matthew T Weirauch
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Divisions of Biomedical Informatics and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Artem Barski
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
28
|
Sato H, Osonoi K, Sharlin CS, Shoda T. Genetic and Molecular Contributors in Eosinophilic Esophagitis. Curr Allergy Asthma Rep 2023; 23:255-266. [PMID: 37084008 PMCID: PMC11136533 DOI: 10.1007/s11882-023-01075-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2023] [Indexed: 04/22/2023]
Abstract
PURPOSE OF REVIEW Eosinophilic esophagitis (EoE) is an allergic inflammatory esophageal disorder with a complex underlying genetic and molecular etiology. The interest of the scientific community in EoE has grown considerably over the past three decades, and the understanding of the genetic and molecular mechanisms involved in this disease has greatly increased. RECENT FINDINGS In this article, we aim to provide both historic aspects and updates on the recent genetic and molecular advances in the understanding of EoE. Although EoE is a relatively newly described disorder, much progress has been made toward identifying the genetic and molecular factors contributing to the disease pathogenesis by a variety of approaches with next-generation sequencing technologies, including genome-wide association study, whole exome sequencing, and bulk and single-cell RNA sequencing. This review highlights the multifaceted impacts of various findings that have shaped the current molecular and genetic landscape of EoE, providing insights that facilitate further understanding of the disease process.
Collapse
Affiliation(s)
- Hiroki Sato
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kasumi Osonoi
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Colby S Sharlin
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tetsuo Shoda
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, MLC 7028, 45229, Cincinnati, OH, USA.
| |
Collapse
|
29
|
Carta S, Cesarani A, Correddu F, Macciotta NPP. Understanding the phenotypic and genetic background of the lactose content in Sarda dairy sheep. J Dairy Sci 2023; 106:3312-3320. [PMID: 37028961 DOI: 10.3168/jds.2022-22579] [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: 07/26/2022] [Accepted: 12/17/2022] [Indexed: 04/09/2023]
Abstract
Lactose, the principal carbohydrate found in milk, plays an important role in the physiological processes of milk production because it is related to milk volume, and it is responsible for the osmotic equilibrium between blood and milk in the mammary gland. In this study, factors affecting lactose content (LC) in sheep milk are investigated. For this purpose, 2,358 test-day records were sampled from 509 ewes (3-7 records per animal). The LC and other main milk traits were analyzed using a mixed linear model that included days in milk (DIM) class, parity, lambing month, and type of lambing as fixed effects and animal, permanent environment, and flock test day as random effects. The pedigree-based approach was used to estimate the heritability and repeatability of LC. Moreover, the genomic background of LC was investigated through a GWAS. The LC was affected by all tested factors (i.e., DIM class, parity, lambing month, and type of lambing). Low heritability (0.10 ± 0.05) and moderate repeatability (0.42 ± 0.02) were estimated for LC. High negative genetic correlations were estimated between LC and NaCl (-0.99 ± 0.01) and between LC and somatic cell count (-0.94 ± 0.05). Only 2 markers passed the chromosome-wide Bonferroni threshold. Results of the present study, although obtained on a relatively small sample, suggest the possibility to include LC in the breeding programs, particularly because of its strong relationship with NaCl and somatic cell count.
Collapse
Affiliation(s)
- S Carta
- Dipartimento di Agraria, University of Sassari, 07100, Sassari, Italy
| | - A Cesarani
- Dipartimento di Agraria, University of Sassari, 07100, Sassari, Italy; Department of Animal and Dairy Science, University of Georgia, Athens 30602
| | - F Correddu
- Dipartimento di Agraria, University of Sassari, 07100, Sassari, Italy.
| | - N P P Macciotta
- Dipartimento di Agraria, University of Sassari, 07100, Sassari, Italy
| |
Collapse
|
30
|
Adel-Patient K, Campeotto F, Grauso M, Guillon B, Moroldo M, Venot E, Dietrich C, Machavoine F, Castelli FA, Fenaille F, Molina TJ, Barbet P, Delacourt C, Leite-de-Moraes M, Lezmi G. Assessment of local and systemic signature of eosinophilic esophagitis (EoE) in children through multi-omics approaches. Front Immunol 2023; 14:1108895. [PMID: 37006253 PMCID: PMC10050742 DOI: 10.3389/fimmu.2023.1108895] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 02/24/2023] [Indexed: 03/17/2023] Open
Abstract
BackgroundEosinophilic oesophagitis (EoE) is a chronic food allergic disorder limited to oesophageal mucosa whose pathogenesis is still only partially understood. Moreover, its diagnosis and follow-up need repeated endoscopies due to absence of non-invasive validated biomarkers. In the present study, we aimed to deeply describe local immunological and molecular components of EoE in well-phenotyped children, and to identify potential circulating EoE-biomarkers.MethodsBlood and oesophageal biopsies were collected simultaneously from French children with EoE (n=17) and from control subjects (n=15). Untargeted transcriptomics analysis was performed on mRNA extracted from biopsies using microarrays. In parallel, we performed a comprehensive analysis of immune components on both cellular and soluble extracts obtained from both biopsies and blood, using flow cytometry. Finally, we performed non-targeted plasma metabolomics using liquid chromatography coupled to high-resolution mass spectrometry (LC-HRMS). Uni/multivariate supervised and non-supervised statistical analyses were then conducted to identify significant and discriminant components associated with EoE within local and/or systemic transcriptomics, immunologic and metabolomics datasets. As a proof of concept, we conducted multi-omics data integration to identify a plasmatic signature of EoE.ResultsFrench children with EoE shared the same transcriptomic signature as US patients. Network visualization of differentially expressed (DE) genes highlighted the major dysregulation of innate and adaptive immune processes, but also of pathways involved in epithelial cells and barrier functions, and in perception of chemical stimuli. Immune analysis of biopsies highlighted EoE is associated with dysregulation of both type (T) 1, T2 and T3 innate and adaptive immunity, in a highly inflammatory milieu. Although an immune signature of EoE was found in blood, untargeted metabolomics more efficiently discriminated children with EoE from control subjects, with dysregulation of vitamin B6 and various amino acids metabolisms. Multi-blocks integration suggested that an EoE plasma signature may be identified by combining metabolomics and cytokines datasets.ConclusionsOur study strengthens the evidence that EoE results from alterations of the oesophageal epithelium associated with altered immune responses far beyond a simplistic T2 dysregulation. As a proof of concept, combining metabolomics and cytokines data may provide a set of potential plasma biomarkers for EoE diagnosis, which needs to be confirmed on a larger and independent cohort.
Collapse
Affiliation(s)
- Karine Adel-Patient
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), Gif-sur-Yvette, France
- *Correspondence: Karine Adel-Patient, ; Guillaume Lezmi,
| | - Florence Campeotto
- AP-HP, Hôpital Necker-Enfants Malades, Service de Gastro-Entérologie et Nutrition Pédiatriques, Paris, France
- Université de Paris Cité, INSERM UMR1139, Laboratoire de Microbiologie, Faculté de Pharmacie, Paris, France
| | - Marta Grauso
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), Gif-sur-Yvette, France
| | - Blanche Guillon
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), Gif-sur-Yvette, France
| | - Marco Moroldo
- Université Paris Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | - Eric Venot
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), Gif-sur-Yvette, France
| | - Céline Dietrich
- Université Paris Cité, CNRS UMR 8253, Inserm UMR 1151, Institut Necker Enfants Malades, Equipe Immunorégulation et Immunopathologie, Paris, France
| | - François Machavoine
- Université Paris Cité, CNRS UMR 8253, Inserm UMR 1151, Institut Necker Enfants Malades, Equipe Immunorégulation et Immunopathologie, Paris, France
| | - Florence A. Castelli
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), Gif-sur-Yvette, France
| | - François Fenaille
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), Gif-sur-Yvette, France
| | - Thierry Jo Molina
- Université de Paris, UMRS 1138, INSERM, Sorbonne Paris-Cité, Paris, France
- AP-HP, Centre-Université de Paris, hôpital Necker-Enfant-Malades, Service d'Anatomie et Cytologie Pathologiques, Paris, France
| | - Patrick Barbet
- Université de Paris, UMRS 1138, INSERM, Sorbonne Paris-Cité, Paris, France
- AP-HP, Centre-Université de Paris, hôpital Necker-Enfant-Malades, Service d'Anatomie et Cytologie Pathologiques, Paris, France
| | - Christophe Delacourt
- AP-HP, Hôpital Necker-Enfants Malades, Service de Pneumologie et Allergologie Pédiatriques, Paris, France
| | - Maria Leite-de-Moraes
- Université Paris Cité, CNRS UMR 8253, Inserm UMR 1151, Institut Necker Enfants Malades, Equipe Immunorégulation et Immunopathologie, Paris, France
| | - Guillaume Lezmi
- Université Paris Cité, CNRS UMR 8253, Inserm UMR 1151, Institut Necker Enfants Malades, Equipe Immunorégulation et Immunopathologie, Paris, France
- AP-HP, Hôpital Necker-Enfants Malades, Service de Pneumologie et Allergologie Pédiatriques, Paris, France
- *Correspondence: Karine Adel-Patient, ; Guillaume Lezmi,
| |
Collapse
|
31
|
Urbančič J, Košak Soklič T, Demšar Luzar A, Hočevar Boltežar I, Korošec P, Rijavec M. Transcriptomic Differentiation of Phenotypes in Chronic Rhinosinusitis and Its Implications for Understanding the Underlying Mechanisms. Int J Mol Sci 2023; 24:ijms24065541. [PMID: 36982612 PMCID: PMC10051401 DOI: 10.3390/ijms24065541] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/04/2023] [Accepted: 03/12/2023] [Indexed: 03/16/2023] Open
Abstract
Chronic rhinosinusitis (CRS) is a multifaceted disease with variable clinical courses and outcomes. We aimed to determine CRS-associated nasal-tissue transcriptome in clinically well-characterized and phenotyped individuals, to gain a novel insight into the biological pathways of the disease. RNA-sequencing of tissue samples of patients with CRS with polyps (CRSwNP), without polyps (CRSsNP), and controls were performed. Characterization of differently expressed genes (DEGs) and functional and pathway analysis was undertaken. We identified 782 common CRS-associated nasal-tissue DEGs, while 375 and 328 DEGs were CRSwNP- and CRSsNP-specific, respectively. Common key DEGs were found to be involved in dendritic cell maturation, the neuroinflammation pathway, and the inhibition of the matrix metalloproteinases. Distinct CRSwNP-specific DEGs were involved in NF-kβ canonical pathways, Toll-like receptor signaling, HIF1α regulation, and the Th2 pathway. CRSsNP involved the NFAT pathway and changes in the calcium pathway. Our findings offer new insights into the common and distinct molecular mechanisms underlying CRSwNP and CRSsNP, providing further understanding of the complex pathophysiology of the CRS, with future research directions for novel treatment strategies.
Collapse
Affiliation(s)
- Jure Urbančič
- Department of Otorhinolaryngology and Cervicofacial Surgery, University Medical Centre Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
- Correspondence:
| | - Tanja Košak Soklič
- Department of Otorhinolaryngology and Cervicofacial Surgery, University Medical Centre Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Ajda Demšar Luzar
- Laboratory for Clinical Immunology and Molecular Genetics, University Clinic of Respiratory and Allergic Diseases Golnik, Golnik 36, SI-4204 Golnik, Slovenia
| | - Irena Hočevar Boltežar
- Department of Otorhinolaryngology and Cervicofacial Surgery, University Medical Centre Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Peter Korošec
- Laboratory for Clinical Immunology and Molecular Genetics, University Clinic of Respiratory and Allergic Diseases Golnik, Golnik 36, SI-4204 Golnik, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, SI-1000 Ljubljana, Slovenia
| | - Matija Rijavec
- Laboratory for Clinical Immunology and Molecular Genetics, University Clinic of Respiratory and Allergic Diseases Golnik, Golnik 36, SI-4204 Golnik, Slovenia
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
32
|
Feng B, Zhou T, Guo Z, Jin J, Zhang S, Qiu J, Cao J, Li J, Peng X, Wang J, Xing Y, Ji R, Qiao L, Liang Y. Comprehensive analysis of immune-related genes for classification and immune microenvironment of asthma. Am J Transl Res 2023; 15:1052-1062. [PMID: 36915798 PMCID: PMC10006808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 02/14/2023] [Indexed: 03/16/2023]
Abstract
OBJECTIVES To determine the effects of immune-related genes (IRGs) and immune landscape of induced sputum, and develop novel, non-invasive diagnostic molecular therapeutic targets for asthma. METHODS GSE76262 datasets were used to identify differentially expressed IRGs in asthma. Key IRGs were detected using a protein-protein interaction network. Receiver operating characteristic (ROC) curves were analyzed to investigate the diagnostic value of key IRGs. Gene set enrichment analysis (GSEA) was performed with WebGestalt. Single-sample gene set enrichment analysis and CIBERSORT were used to investigate the immune landscape of induced sputum. RESULTS A total of 75 potential IRGs were associated with asthma, most of which were involved in the NF-kappa B signaling pathway. ROC analysis showed AUC values for the hub pathway ranging from 0.676-0.767, with moderate diagnostic value for asthma. We also identified IRGs-related cytokines (TNF-α, IL-1β, IL-8 and IL-6) in 76 asthma and 91 control serum samples to further explore diagnostic efficacy, showing a cumulative AUC of 0.998 for these four related cytokines. Analysis of immune cell infiltration levels showed that follicular helper T cells, activated dendritic cells, activated mast cells and eosinophils were significantly higher and macrophages M0 and macrophages M2 were significantly reduced in sputum from patients with asthma. CONCLUSIONS IRGs-related cytokines and immune infiltration may contribute to the diagnosis and immune classification of asthma.
Collapse
Affiliation(s)
- Bin Feng
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow University Suzhou 215006, Jiangsu, China
| | - Tong Zhou
- Medical College of Soochow University Suzhou 215006, Jiangsu, China
| | - Zhiyi Guo
- Medical College of Soochow University Suzhou 215006, Jiangsu, China
| | - Jieyu Jin
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow University Suzhou 215006, Jiangsu, China
| | - Sheng Zhang
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow University Suzhou 215006, Jiangsu, China
| | - Jun Qiu
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow University Suzhou 215006, Jiangsu, China
| | - Jun Cao
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow University Suzhou 215006, Jiangsu, China
| | - Jia Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine Shanghai 201209, China
| | - Xia Peng
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine Shanghai 201209, China
| | - Juan Wang
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine Shanghai 201209, China
| | - Yanru Xing
- Basecare Medical Device Co., Ltd. Suzhou 215006, China
| | - Renxin Ji
- The School of International Medical Technology of Shanghai Sanda University Shanghai 201209, China
| | - Longwei Qiao
- Center for Reproduction and Genetics, School of Gusu, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University Suzhou 215006, Jiangsu, China
| | - Yuting Liang
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow University Suzhou 215006, Jiangsu, China
| |
Collapse
|
33
|
Underwood B, Troutman TD, Schwartz JT. Breaking down the complex pathophysiology of eosinophilic esophagitis. Ann Allergy Asthma Immunol 2023; 130:28-39. [PMID: 36351516 PMCID: PMC10165615 DOI: 10.1016/j.anai.2022.10.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 11/08/2022]
Abstract
Eosinophilic esophagitis (EoE) is a chronic and progressive immune-mediated disease of the esophagus associated with antigen-driven type 2 inflammation and symptoms of esophageal dysfunction. Our understanding of EoE pathophysiology has evolved since its initial recognition more than 20 years ago and has translated into diagnostic and novel therapeutic approaches that are affecting patient care. The mechanisms underlying disease development and progression are influenced by diverse factors, such as genetics, age, allergic comorbidities, and allergen exposures. Central to EoE pathophysiology is a dysregulated feed-forward cycle that develops between the esophageal epithelium and the immune system. Allergen-induced, type 2-biased immune activation by the esophageal epithelium propagates a cycle of impaired mucosal barrier integrity and allergic inflammation, eventually leading to tissue remodeling and progressive organ dysfunction. Herein, we review the current understanding of fundamental pathophysiological mechanisms contributing to EoE pathogenesis.
Collapse
Affiliation(s)
- Brynne Underwood
- Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ty D Troutman
- Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Justin T Schwartz
- Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
34
|
Edwards-Salmon S, Moraczewski J, Offerle T, Sinclair EM, Xiang Y, Gillespie S, Kruszewski P. Comparing Eosinophilic Esophagitis in a Black and Non-Black Pediatric Cohort. J Pediatr Gastroenterol Nutr 2022; 75:485-490. [PMID: 35797567 DOI: 10.1097/mpg.0000000000003552] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES To compare presenting symptoms, comorbidities, disease, and treatment characteristics of a black pediatric eosinophilic esophagitis (EoE) group to a non-black pediatric EoE group. METHODS A retrospective chart review consisting of pediatric patients diagnosed with EoE between the years of 2010 and 2018 at a single urban pediatric hospital system comprising 143 black pediatric patients compared with 142 non-black pediatric patients with similar distribution of age and sex. RESULTS Both groups were majority male, and the median age of diagnosis between the black and non-black group was 5.1 and 6.7 years old, respectively. Comorbidities more commonly seen in the black group included food allergies, atopic dermatitis, asthma, and allergic rhinitis. Black patients were more likely to present with failure to thrive (FTT)/poor growth, whereas non-black patients were more likely to present with abdominal pain. There was no statistically significant difference between the groups in achieving remission using current therapies. The black group had higher rates of nonadherence to medical therapies. CONCLUSIONS This is the largest study to date comparing a black versus non-black pediatric EoE population. The black population had more atopic comorbidities and FTT at presentation and had significantly more issues with nonadherence. This new knowledge describing EoE in a minority population will hopefully improve awareness, diagnosis, and management of EoE in this population.
Collapse
Affiliation(s)
- Sofia Edwards-Salmon
- From the Emory School of Medicine, Atlanta, GA
- the Section of Allergy & Immunology, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA
| | | | | | - Elizabeth M Sinclair
- the Department of Pediatric Gastroenterology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA
| | - Yijin Xiang
- From the Emory School of Medicine, Atlanta, GA
- the Children's Healthcare of Atlanta, Atlanta, GA
| | - Scott Gillespie
- From the Emory School of Medicine, Atlanta, GA
- the Children's Healthcare of Atlanta, Atlanta, GA
| | - Patrice Kruszewski
- the Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
35
|
Khokhar D, Marella S, Idelman G, Chang JW, Chehade M, Hogan SP. Eosinophilic esophagitis: Immune mechanisms and therapeutic targets. Clin Exp Allergy 2022; 52:1142-1156. [PMID: 35778876 PMCID: PMC9547832 DOI: 10.1111/cea.14196] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 06/08/2022] [Accepted: 06/20/2022] [Indexed: 01/26/2023]
Abstract
Eosinophilic esophagitis (EoE) is an emerging chronic inflammatory disease of the oesophagus and is clinically characterized by upper gastrointestinal (GI) symptoms including dysphagia and esophageal food impaction. Histopathologic manifestations, which include intraepithelial eosinophilic inflammation and alterations of the esophageal squamous epithelium, such as basal zone hyperplasia (BZH) and dilated intercellular spaces (DIS), are thought to contribute to esophageal dysfunction and disease symptoms. Corroborative clinical and discovery science-based studies have established that EoE is characterized by an underlying allergic inflammatory response, in part, related to the IL-13/CCL26/eosinophil axis driving dysregulation of several key epithelial barrier and proliferative regulatory genes including kallikrein (KLK) serine proteases, calpain 14 (CAPN14) and anoctamin 1 (ANO1). The contribution of these inflammatory and proliferative processes to the clinical and histological manifestations of disease are not fully elucidated. Herein, we discuss the immune molecules and cells that are thought to underlie the clinical and pathologic manifestations of EoE and the emerging therapeutics targeting these processes for the treatment of EoE.
Collapse
Affiliation(s)
- Dilawar Khokhar
- Division of Allergy and ImmunologyUniversity of MichiganAnn ArborMichiganUSA
- Mary H Weiser Food Allergy CenterUniversity of MichiganAnn ArborMichiganUSA
| | - Sahiti Marella
- Mary H Weiser Food Allergy CenterUniversity of MichiganAnn ArborMichiganUSA
- Department of PathologyUniversity of MichiganAnn ArborMichiganUSA
| | - Gila Idelman
- Mary H Weiser Food Allergy CenterUniversity of MichiganAnn ArborMichiganUSA
| | - Joy W. Chang
- Division of Gastroenterology, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Mirna Chehade
- Mount Sinai Center for Eosinophilic DisordersIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Simon P. Hogan
- Mary H Weiser Food Allergy CenterUniversity of MichiganAnn ArborMichiganUSA
- Department of PathologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
36
|
Chen J, Oshima T, Huang X, Tomita T, Fukui H, Miwa H. Esophageal Mucosal Permeability as a Surrogate Measure of Cure in Eosinophilic Esophagitis. J Clin Med 2022; 11:jcm11144246. [PMID: 35888006 PMCID: PMC9319696 DOI: 10.3390/jcm11144246] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022] Open
Abstract
This study aimed to evaluate the relationship of esophageal epithelial permeability with mast cell infiltration and IgG4 deposits as well as chemokine levels in eosinophilic esophagitis (EoE) patients before and after treatment. Biopsies from controls and EoE patients before and after treatment were analyzed. Hematoxylin and eosin staining was used to show eosinophil infiltration. Paracellular permeability of the esophageal epithelium was assessed using surface biotinylation. Immunohistochemical staining was performed to examine mast cell infiltration and IgG4 deposits. Gene expression of chemokines was evaluated by qRT-PCR. Esophageal epithelial infiltration of mast cells, IgG4 deposits, and permeability were significantly increased in EoE patients. Levels of interleukin-13, calpain-14, and eotaxin-3 mRNAs were significantly upregulated, while filaggrin, serine peptidase inhibitor Kazal type 7 (SPINK7), and involucrin mRNAs were significantly downregulated in EoE patients. In patients achieving histologic remission diagnosed by eosinophil counts, a subset of EoE patients with unchanged permeability after treatment showed increases in mast cell infiltration, IgG4 deposits, and interleukin-13, calpain-14, filaggrin, and SPINK7 expression, with decreased eotaxin-3 and involucrin. Other EoE patients with decreased permeability displayed decreased eotaxin-3, involucrin, and mast cell infiltration, no IgG4 deposits, and increased IL-13, calpain-14, filaggrin, and SPINK7. Increased permeability of the esophagus in EoE patients without eosinophil infiltration after treatment was associated with mast cell infiltration and IgG4 deposits.
Collapse
|
37
|
Du P, Chai Y, Zong S, Yue J, Xiao H. Identification of a Prognostic Model Based on Fatty Acid Metabolism-Related Genes of Head and Neck Squamous Cell Carcinoma. Front Genet 2022; 13:888764. [PMID: 35846149 PMCID: PMC9280184 DOI: 10.3389/fgene.2022.888764] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/09/2022] [Indexed: 01/12/2023] Open
Abstract
The fatty acid metabolism (FAM) is known to impact tumorigenesis, tumor progression and treatment resistance via enhancing lipid synthesis, storage and catabolism. However, the role of FAM in head and neck squamous cell carcinoma (HNSCC) has remained elusive. In the present study, we obtained a total of 69 differentially expressed FAM-related genes between 502 HNSCC samples and 44 normal samples from The Cancer Genome Atlas (TCGA) database. The HNSCC samples were divided into 2 clusters according to 69 differentially expressed genes (DEGs) via cluster analysis. Then DEGs in the two clusters were found, and 137 prognostic DEGs were identified by univariate analysis. Subsequently, combined with the clinical information of 546 HNSCC patients from TCGA database, a 12-gene prognostic risk model was established (FEPHX3, SPINK7, FCRLA, MASP1, ZNF541, CD5, BEST2 and ZAP70 were down-regulation, ADPRHL1, DYNC1I1, KCNG1 and LINC00460 were up-regulation) using multivariate Cox regression and LASSO regression analysis. The risk scores of 546 HNSCC samples were calculated. According to the median risk score, 546 HNSCC patients were divided into the high- and low-risk (high- and low score) groups. The Kaplan-Meier survival analysis showed that the survival time of HNSCC patients was significantly shorter in the high-risk group than that in the low-risk group (p < 0.001). The same conclusion was obtained in the Gene Expression Omnibus (GEO) dataset. After that, the multivariate Cox regression analysis indicated that the risk score was an independent factor for patients with HNSCC in the TCGA cohort. In addition, single-sample gene set enrichment analysis (ssGSEA) indicated that the level of infiltrating immune cells was relatively low in the high-risk group compared with the low-risk group. In summary, FAM-related gene expression-based risk signature could predict the prognosis of HNSCC independently.
Collapse
Affiliation(s)
- Peiyu Du
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Chai
- Department of Medical Oncology, National Cancer Cente, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shimin Zong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianxin Yue
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jianxin Yue, ; Hongjun Xiao,
| | - Hongjun Xiao
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jianxin Yue, ; Hongjun Xiao,
| |
Collapse
|
38
|
Tanzer J, Meng D, Ohsaki A, Caldwell JM, Mingler MK, Rothenberg ME, Oyoshi MK. Laundry detergent promotes allergic skin inflammation and esophageal eosinophilia in mice. PLoS One 2022; 17:e0268651. [PMID: 35759448 PMCID: PMC9236249 DOI: 10.1371/journal.pone.0268651] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 05/04/2022] [Indexed: 11/24/2022] Open
Abstract
The prevalence of allergic diseases is on the rise, yet the environmental factors that contribute to this increase are still being elucidated. Laundry detergent (LD) that contains cytotoxic ingredients including microbial enzymes continuously comes into contact with the skin starting in infancy. An impaired skin barrier has been suggested as a route of allergic sensitization. We hypothesized that exposure of skin to LD damages the skin barrier resulting in systemic sensitization to allergens that enter through the impaired skin barrier. Mouse skin samples exposed in vitro to microbial proteases or LD exhibited physical damage, which was more pronounced in neonatal skin as compared to adult skin. Exposure of the skin to microbial proteases in vitro resulted in an increase in the levels of interleukin (IL)-33 and thymic stromal lymphopoietin (TSLP). BALB/c wild type mice epicutaneously exposed to LD and ovalbumin (OVA) showed an increase in levels of transepidermal water loss, serum OVA-specific immunoglobulin (Ig) G1 and IgE antibodies, and a local increase of Il33, Tslp, Il4 and Il13 compared with LD or OVA alone. Following intranasal challenge with OVA, mice epicutaneously exposed to LD showed an increase in allergen-induced esophageal eosinophilia compared with LD or OVA alone. Collectively, these results suggest that LD may be an important factor that impairs the skin barrier and leads to allergen sensitization in early life, and therefore may have a role in the increase in allergic disease.
Collapse
Affiliation(s)
- Jamie Tanzer
- Harvard College, Cambridge, MA, United States of America
| | - Di Meng
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA, United States of America
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States of America
| | - Asa Ohsaki
- Division of Immunology, Boston Children’s Hospital, Boston, MA, United States of America
| | - Julie M. Caldwell
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children’s Hospital, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Melissa K. Mingler
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children’s Hospital, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Marc E. Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children’s Hospital, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Michiko K. Oyoshi
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA, United States of America
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States of America
- Division of Immunology, Boston Children’s Hospital, Boston, MA, United States of America
| |
Collapse
|
39
|
Kaymak T, Kaya B, Wuggenig P, Nuciforo S, Göldi A, Oswald F, Roux J, Noti M, Melhem H, Hruz P, Niess JH. IL-20 subfamily cytokines impair the oesophageal epithelial barrier by diminishing filaggrin in eosinophilic oesophagitis. Gut 2022; 72:821-833. [PMID: 35613844 PMCID: PMC10086458 DOI: 10.1136/gutjnl-2022-327166] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/02/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Disruption of the epithelial barrier plays an essential role in developing eosinophilic oesophagitis (EoE), a disease defined by type 2 helper T cell (Th2)-mediated food-associated and aeroallergen-associated chronic inflammation. Although an increased expression of interleukin (IL)-20 subfamily members, IL-19, IL-20 and IL-24, in Th2-mediated diseases has been reported, their function in EoE remains unknown. DESIGN Combining transcriptomic, proteomic and functional analyses, we studied the importance of the IL-20 subfamily for EoE using patient-derived oesophageal three-dimensional models and an EoE mouse model. RESULTS Patients with active EoE have increased expression of IL-20 subfamily cytokines in the oesophagus and serum. In patient-derived oesophageal organoids stimulated with IL-20 cytokines, RNA sequencing and mass spectrometry revealed a downregulation of genes and proteins forming the cornified envelope, including filaggrins. On the contrary, abrogation of IL-20 subfamily signalling in Il20R2 -/- animals resulted in attenuated experimental EoE reflected by reduced eosinophil infiltration, lower Th2 cytokine expression and preserved expression of filaggrins in the oesophagus. Mechanistically, these observations were mediated by the mitogen-activated protein kinase (MAPK); extracellular-signal regulated kinases (ERK)1/2) pathway. Its blockade prevented epithelial barrier impairment in patient-derived air-liquid interface cultures stimulated with IL-20 cytokines and attenuated experimental EoE in mice. CONCLUSION Our findings reveal a previously unknown regulatory role of the IL-20 subfamily for oesophageal barrier function in the context of EoE. We propose that aberrant IL-20 subfamily signalling disturbs the oesophageal epithelial barrier integrity and promotes EoE development. Our study suggests that specific targeting of the IL-20 subfamily signalling pathway may present a novel strategy for the treatment of EoE.
Collapse
Affiliation(s)
- Tanay Kaymak
- Department of Biomedicine, Gastroenterology, University of Basel, Basel, Switzerland
| | - Berna Kaya
- Department of Biomedicine, Gastroenterology, University of Basel, Basel, Switzerland
| | - Philipp Wuggenig
- Department of Biomedicine, Gastroenterology, University of Basel, Basel, Switzerland
| | - Sandro Nuciforo
- Department of Biomedicine, Hepatology, University of Basel, Basel, Switzerland
| | - Andreas Göldi
- Department of Gastroenterology, Clarunis - University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | | | - Franz Oswald
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Julien Roux
- Department of Biomedicine, Gastroenterology, University of Basel, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Mario Noti
- Institute of Pathology, University of Bern, Bern, Switzerland, Current address: Nestlé SA, Nestlé Research, Nestlé Institute of Health Sciences, Department of Gastrointestinal Health Immunology, Vers-Chez-les-Blancs, Lausanne, Switzerland
| | - Hassan Melhem
- Department of Biomedicine, Gastroenterology, University of Basel, Basel, Switzerland
| | - Petr Hruz
- Department of Gastroenterology, Clarunis - University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Jan Hendrik Niess
- Department of Biomedicine, Gastroenterology, University of Basel, Basel, Switzerland .,Department of Gastroenterology, Clarunis - University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland.,Department of Clinical Research, University of Basel, Basel, Switzerland
| |
Collapse
|
40
|
Rochman M, Wen T, Kotliar M, Dexheimer PJ, Ben-Baruch Morgenstern N, Caldwell JM, Lim HW, Rothenberg ME. Single-cell RNA sequencing of human esophageal epithelium in homeostasis and allergic inflammation. JCI Insight 2022; 7:159093. [PMID: 35472002 PMCID: PMC9208762 DOI: 10.1172/jci.insight.159093] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/22/2022] [Indexed: 11/17/2022] Open
Abstract
Inflammation of the esophageal epithelium is a hallmark of eosinophilic esophagitis (EoE), an emerging chronic allergic disease. Herein, we probed human esophageal epithelial cells at single-cell resolution during homeostasis and EoE. During allergic inflammation, the epithelial differentiation program was blocked, leading to loss of KRT6high differentiated populations and expansion of TOP2high proliferating and DSPhigh, SERPINB3high transitioning populations; however, there was stability of the stem cell-enriched PDPNhigh basal epithelial compartment. This differentiation program blockade was associated with dysregulation of transcription factors, including nuclear receptor signalers, in the most differentiated epithelial cells and altered NOTCH-related cell-to-cell communication. Each epithelial population expressed genes with allergic disease risk variants, supporting their functional interplay. The esophageal epithelium differed notably between EoE in histologic remission and controls, indicating that remission is a transitory state poised to relapse. Collectively, our data uncover the dynamic nature of the inflamed human esophageal epithelium and provide a framework to better understand esophageal health and disease.
Collapse
Affiliation(s)
- Mark Rochman
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Ting Wen
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Michael Kotliar
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Phillip J Dexheimer
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Netali Ben-Baruch Morgenstern
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Julie M Caldwell
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Hee-Woong Lim
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| |
Collapse
|
41
|
Liao C, Wang Q, An J, Zhang M, Chen J, Li X, Xiao L, Wang J, Long Q, Liu J, Guan X. SPINKs in Tumors: Potential Therapeutic Targets. Front Oncol 2022; 12:833741. [PMID: 35223512 PMCID: PMC8873584 DOI: 10.3389/fonc.2022.833741] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 01/14/2022] [Indexed: 12/14/2022] Open
Abstract
The serine protease inhibitor Kazal type (SPINK) family includes SPINK1-14 and is the largest branch in the serine protease inhibitor family. SPINKs play an important role in pancreatic physiology and disease, sperm maturation and capacitation, Nager syndrome, inflammation and the skin barrier. Evidence shows that the unregulated expression of SPINK1, 2, 4, 5, 6, 7, and 13 is closely related to human tumors. Different SPINKs exhibit various regulatory modes in different tumors and can be used as tumor prognostic markers. This article reviews the role of SPINK1, 2, 4, 5, 6, 7, and 13 in different human cancer processes and helps to identify new cancer treatment targets.
Collapse
Affiliation(s)
- Chengcheng Liao
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Qian Wang
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Life Sciences Institute, Zunyi Medical University, Zunyi, China
| | - Jiaxing An
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Minglin Zhang
- Department of Gastroenterology, Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang, China
| | - Jie Chen
- Department of Urology, The Third Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaolan Li
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Life Sciences Institute, Zunyi Medical University, Zunyi, China
| | - Linlin Xiao
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Jiajia Wang
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Qian Long
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- *Correspondence: Qian Long, ; Xiaoyan Guan, ; Jianguo Liu,
| | - Jianguo Liu
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- *Correspondence: Qian Long, ; Xiaoyan Guan, ; Jianguo Liu,
| | - Xiaoyan Guan
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- *Correspondence: Qian Long, ; Xiaoyan Guan, ; Jianguo Liu,
| |
Collapse
|
42
|
Racca F, Pellegatta G, Cataldo G, Vespa E, Carlani E, Pelaia C, Paoletti G, Messina MR, Nappi E, Canonica GW, Repici A, Heffler E. Type 2 Inflammation in Eosinophilic Esophagitis: From Pathophysiology to Therapeutic Targets. Front Physiol 2022; 12:815842. [PMID: 35095572 PMCID: PMC8790151 DOI: 10.3389/fphys.2021.815842] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/09/2021] [Indexed: 12/11/2022] Open
Abstract
Eosinophilic esophagitis (EoE) is a chronic immune-mediated disease of the esophagus characterized clinically by symptoms related to esophageal dysfunction and histologically by eosinophil-predominant inflammation, whose incidence is rising. It significantly affects patients’ quality of life and, if left untreated, results in fibrotic complications. Although broad consensus has been achieved on first-line therapy, a subset of patients remains non-responder to standard therapy. The pathogenesis of EoE is multifactorial and results from the complex, still mostly undefined, interaction between genetics and intrinsic factors, environment, and antigenic stimuli. A deep understanding of the pathophysiology of this disease is pivotal for the development of new therapies. This review provides a comprehensive description of the pathophysiology of EoE, starting from major pathogenic mechanisms (genetics, type 2 inflammation, epithelial barrier dysfunction, gastroesophageal reflux, allergens, infections and microbiota) and subsequently focusing on the single protagonists of type 2 inflammation (involved cells, cytokines, soluble effectors, surface proteins and transcription factors) that could represent present and future therapeutic targets, while summarizing previous therapeutic approaches in literature.
Collapse
Affiliation(s)
- Francesca Racca
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- *Correspondence: Francesca Racca,
| | - Gaia Pellegatta
- Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Giuseppe Cataldo
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Edoardo Vespa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Elisa Carlani
- Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Corrado Pelaia
- Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Giovanni Paoletti
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Maria Rita Messina
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Emanuele Nappi
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Giorgio Walter Canonica
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Alessandro Repici
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Enrico Heffler
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| |
Collapse
|
43
|
Zhang S, Sicherer S, Berin MC, Agyemang A. Pathophysiology of Non-IgE-Mediated Food Allergy. Immunotargets Ther 2022; 10:431-446. [PMID: 35004389 PMCID: PMC8721028 DOI: 10.2147/itt.s284821] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022] Open
Abstract
Non-IgE-mediated food allergies are a group of disorders characterized by subacute or chronic inflammatory processes in the gut. Unlike IgE mediated food allergies that may result in multi-organ system anaphylaxis, the non-IgE mediated food allergies primarily affect the gastrointestinal tract. This review outlines the clinical manifestations, epidemiology, pathophysiology, and management of non-IgE-mediated food allergies. An updated literature search of selected non-IgE-mediated food allergies was conducted for this review using PubMed database to the current year (2021). Reviewed disorders include food protein-induced enterocolitis syndrome (FPIES), food-protein enteropathy (FPE), food protein-induced allergic proctocolitis (FPIAP), and eosinophilic gastrointestinal disorders (EGIDs) such as eosinophilic esophagitis (EoE). While extensive gains have been made in understanding FPIES, FPIAP, FPE, and EoE, more information is needed on the pathophysiology of these food allergies. Similarities among them include involvement of innate immunity, T-lymphocyte processes, alteration of the intestinal lumen at the cellular level with the appearance of inflammatory cells and associated histologic changes, and specific cytokine profiles suggesting food-specific, T-cell, and immune-mediated responses. While FPIES and FPIAP typically resolve in early childhood, EGIDs typically do not. Emerging new therapies for EoE offer promise of additional treatment options. Further studies identifying the immunopathogenesis, associated biomarkers, and mechanisms of tolerance are needed to inform prevention, diagnosis and management.
Collapse
Affiliation(s)
- Shouling Zhang
- Department of Pediatrics, Division of Allergy and Immunology, Icahn School of Medicine at Mount Sinai, Kravis Children's Hospital, The Elliot and Roslyn Jaffe Food Allergy Institute, New York, NY, USA
| | - Scott Sicherer
- Department of Pediatrics, Division of Allergy and Immunology, Icahn School of Medicine at Mount Sinai, Kravis Children's Hospital, The Elliot and Roslyn Jaffe Food Allergy Institute, New York, NY, USA
| | - M Cecilia Berin
- Department of Pediatrics, Division of Allergy and Immunology, Icahn School of Medicine at Mount Sinai, Kravis Children's Hospital, The Elliot and Roslyn Jaffe Food Allergy Institute, New York, NY, USA
| | - Amanda Agyemang
- Department of Pediatrics, Division of Allergy and Immunology, Icahn School of Medicine at Mount Sinai, Kravis Children's Hospital, The Elliot and Roslyn Jaffe Food Allergy Institute, New York, NY, USA
| |
Collapse
|
44
|
Williamson P, Proudfoot J, Gharibans A, Dohil L, Newbury R, Barsamian J, Hassan M, Rawson R, Katzka D, Kurten R, Dohil R, Mousa H, Aceves S. Plasminogen Activator Inhibitor-1 as a Marker of Esophageal Functional Changes in Pediatric Eosinophilic Esophagitis. Clin Gastroenterol Hepatol 2022; 20:57-64.e3. [PMID: 33007513 PMCID: PMC8007666 DOI: 10.1016/j.cgh.2020.09.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/15/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Esophageal remodeling in eosinophilic esophagitis (EoE) can lead to esophageal rigidity with eventual luminal compromise and stenoses. Gauging esophageal functional alterations in EoE is challenging. An epithelial marker of functional remodeling would impact EoE management. METHODS Esophageal biopsy specimens from children with and without EoE and primary human esophageal epithelial cells were used for PAI-1 immunohistochemistry, and cell proliferation experiments. PAI-1 immunostaining and basal cell hyperplasia were assessed in the context of concurrently obtained esophageal compliance measures on endoscopic functional lumen imaging probe (EndoFLIP). RESULTS EndoFLIPs were performed in 45 children (32 with and 13 without EoE). Epithelial PAI-1 was increased in patients with active EoE versus inactive or control patients (P < .01). Esophageal compliance was lower in EoE patients versus controls, particularly in the proximal esophagus (P < .001). Proximal compliance was the strongest predictor of EoE (AUROC 0.88, 95% CI 0.77, 0.98) with esophageal compliance of less than 2.6%mL/mmHg demonstrating 82% sensitivity and 84% specificity for EoE. PAI-1 inhibition significantly diminished esophageal epithelial cell proliferation, suggesting PAI-1 could trigger basal cell hyperplasia. A composite mid-esophageal BZH + PAI-1 score was the strongest predictor of altered compliance (P = .02, AUROC 0.89 (95% CI 0.80, 0.99). CONCLUSIONS PAI-1 is significantly elevated in pediatric EoE and distinguishes altered compliance in children. PAI-1 may be a novel disease marker and therapeutic target.
Collapse
Affiliation(s)
- Prerana Williamson
- Department of Pediatrics, University of California, San Diego, CA,Division of Pediatric Gastroenterology,Rady Children’s Hospital, San Diego, CA
| | - James Proudfoot
- Altman Clinical and Translational Research Institute, University of California, San Diego, CA
| | - Armen Gharibans
- Rady Children’s Hospital, San Diego, CA,Department of Surgery, University of Auckland, New Zealand,Auckland Bioengineering Institute, University of Auckland, New Zealand
| | - Lucas Dohil
- Department of Pediatrics, University of California, San Diego, CA,Division of Allergy and Immunology
| | - Robert Newbury
- Rady Children’s Hospital, San Diego, CA,Department of Pathology, University of California San Diego, CA
| | | | - Maheen Hassan
- Department of Pediatrics, University of California, San Diego, CA,Division of Pediatric Gastroenterology,Rady Children’s Hospital, San Diego, CA
| | - Renee Rawson
- Department of Pediatrics, University of California, San Diego, CA,Division of Allergy and Immunology
| | - David Katzka
- Department of Internal Medicine, Mayo Clinic, Rochester, MN,Division of Gastroenterology and Hepatology
| | - Richard Kurten
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AK
| | - Ranjan Dohil
- Department of Pediatrics, University of California, San Diego, CA,Division of Pediatric Gastroenterology,Rady Children’s Hospital, San Diego, CA
| | - Hayat Mousa
- Department of Pediatrics, University of California, San Diego, CA,Division of Pediatric Gastroenterology,Rady Children’s Hospital, San Diego, CA
| | - Seema Aceves
- Rady Children's Hospital, San Diego, California; Division of Allergy and Immunology, University of California San Diego, San Diego, California; Department of Medicine, University of California San Diego, San Diego, California.
| |
Collapse
|
45
|
Lee KE, Kang CM, Jeon M, Kim SO, Lee JH, Choi HJ. General gene expression patterns and stemness of the gingiva and dental pulp. J Dent Sci 2022; 17:284-292. [PMID: 35028049 PMCID: PMC8739237 DOI: 10.1016/j.jds.2021.02.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/26/2021] [Indexed: 12/01/2022] Open
Abstract
Background/purpose Due to the unique properties of healing processes and cellular differentiation, the gingiva and dental pulp have attracted attention as a potential source of mesenchymal stem cells (MSCs). The purpose of this study was to obtain molecular-level information on these tissues in terms of their function and differentiation processes and investigate stemness. Materials and methods Healthy gingival tissues were collected from patients (n = 9; aged 7–12 years) who underwent simple surgical procedures, and normal dental pulp tissues were obtained from patients (n = 25; aged 11–25 years) undergoing tooth extraction for orthodontic reasons. Complementary DNA microarray, qRT-qPCR, and immunohistochemical staining were performed to assess general and MSC gene expression patterns. Results In the gingival tissue, genes related to keratinization, the formation of epithelial cells and ectoderm, and immune and/or inflammatory responses were highly expressed. Meanwhile, in the dental pulp tissue, genes related to ion transport, neuronal development and axon guidance, bone and enamel mineralization, extracellular matrix organization, and angiogenesis were highly expressed. When focusing on the expression of MSC genes, induced pluripotent stem (iPS) cell genes, such as Sox2, c-Myc, and KLF4, were expressed at higher levels in the gingival tissue, whereas dental stem cell genes, such as NT5E and VCAM1, were expressed in dental pulp tissue. Conclusion We found different general and MSC gene expression patterns between the gingival and dental pulp tissue. These results have implications for future regenerative medicine, considering the application of gingival tissue as a potential source of iPS cells.
Collapse
Affiliation(s)
- Ko Eun Lee
- Department of Pediatric Dentistry, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Chung-Min Kang
- Department of Pediatric Dentistry, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Mijeong Jeon
- Oral Science Research Center, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| | - Seong-Oh Kim
- Department of Pediatric Dentistry, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Jae-Ho Lee
- Department of Pediatric Dentistry, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Hyung-Jun Choi
- Department of Pediatric Dentistry, Yonsei University College of Dentistry, Seoul, Republic of Korea
| |
Collapse
|
46
|
Liu H, Huang Q, Lv Y, Dong Y, Song D. CEBPB knockdown sensitizes nasopharyngeal carcinoma cells to cisplatin by promoting the expression of serine protease inhibitor Kazal-type 5. Anticancer Drugs 2022; 33:e327-e335. [PMID: 34387602 DOI: 10.1097/cad.0000000000001208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Serine protease inhibitor Kazal-type 5 (SPINK5) has been indicated to act as a prognostic predictor for patients with head and neck squamous cell carcinoma. However, its specific role in nasopharyngeal carcinoma (NPC), a malignancy that has a high propensity for chemoresistance, remains largely obscure. We, thus, sought to investigate the importance of SPINK5 expression in regulating chemoresistance in NPC. Differentially expressed genes in NPC were screened using the cancer genome atlas-head and neck squamous cell carcinoma database and microarray analysis. SPINK5 was downregulated in NPC tissues and cells. After SPINK5 upregulation, the cells treated with cisplatin showed reduced cell survival and the ability to migrate, invade and metastasize. Mechanistically, the transcription factors regulating SPINK5 were queried through the JASPAR website, followed by dual-luciferase and Chromatin immunoprecipitation assay validation. CCAAT enhancer-binding protein (CEBP) beta (CEBPB) bound to the SPINK5 promoter region in NPC cells. The silencing of CEBPB enhanced the expression of SPINK5. CEBPB overexpression reversed the inhibitory effects of cisplatin on NPC cell malignant phenotype in the presence of SPINK5 overexpression. In conclusion, CEBPB silencing promoted chemoresistance of NPC cells via activating SPINK5, signifying that targeting CEBPB was a new approach to enhance the chemotherapy efficacy in NPC.
Collapse
Affiliation(s)
- Hong Liu
- Department of Oncology, Binzhou Central Hospital, Binzhou
| | - Qingli Huang
- Department of Otolaryngology, Liaocheng People's Hospital
| | - Yunxia Lv
- Department of Otolaryngology, Yanggu People's Hospital, Liaocheng
| | - Youwei Dong
- Department of Otolaryngology, Juxian people's Hospital, Rizhao
| | - Daoliang Song
- Department of Otorhinolargology Head and Neck Surgery, Zibo Central Hospital, Zibo, Shandong, People's Republic of China
| |
Collapse
|
47
|
Dunn JLM, Rothenberg ME. 2021 year in review: Spotlight on eosinophils. J Allergy Clin Immunol 2021; 149:517-524. [PMID: 34838883 DOI: 10.1016/j.jaci.2021.11.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 02/06/2023]
Abstract
This review highlights recent advances in the understanding of eosinophils and eosinophilic diseases, particularly eosinophilic gastrointestinal diseases during the last year. The increasing incidence of diseases marked by eosinophilia has been documented and highlighted the need to understand eosinophil biology and eosinophilic contributions to disease. Significant insight into the nature of eosinophilic diseases has been achieved using next-generation sequencing technologies, proteomic analysis, and machine learning to analyze tissue biopsies. These technologies have elucidated mechanistic underpinnings of eosinophilic inflammation, delineated patient endotypes, and identified patient responses to therapeutic intervention. Importantly, recent clinical studies using mAbs that interfere with type 2 cytokine signaling or deplete eosinophils point to multiple and complex roles of eosinophils in tissues. Several studies identified distinct activation features of eosinophils in different tissues and disease states. The confluence of these studies supports a new paradigm of tissue-resident eosinophils that have pro- and anti-inflammatory immunomodulatory roles in allergic disease. Improved understanding of unique eosinophil activation states is now poised to identify novel therapeutic targets for eosinophilic diseases.
Collapse
Affiliation(s)
- Julia L M Dunn
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
48
|
Hsieh LY, Chiang AWT, Duong LD, Kuo CC, Dong SX, Dohil R, Kurten R, Lewis NE, Aceves SS. A unique esophageal extracellular matrix proteome alters normal fibroblast function in severe eosinophilic esophagitis. J Allergy Clin Immunol 2021; 148:486-494. [PMID: 33556465 PMCID: PMC8342625 DOI: 10.1016/j.jaci.2021.01.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 12/16/2020] [Accepted: 01/12/2021] [Indexed: 01/23/2023]
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) is a chronic TH2 disorder complicated by tissue fibrosis and loss of esophageal luminal patency. The fibrostenotic esophagus does not respond well to therapy, but profibrotic therapeutic targets are largely unclear. OBJECTIVE Our aim was to utilize proteomics and primary cells as a novel approach to determine relevant profibrotic factors. METHODS We utilized primary esophageal EoE and normal fibroblasts, their derivative extracellular matrixes (ECMs), an approach of fibroblast culture on autologous versus nonautologous ECM, and proteomics to elucidate EoE ECM proteins that dysregulate cellular function. RESULTS We cultured esophageal fibroblasts from normal esophagi and esophagi from patients with severe EoE on autologous versus nonautologous ECM. The EoE ECM proteome shifted normal esophageal fibroblast protein expression. Proteomic analysis demonstrated that thrombospondin-1 is detected only in the EoE ECM, is central in the EoE ECM protein-protein interactome, is found at significantly elevated levels in biopsy specimens from patients with active EoE, and induces fibroblast collagen I production. CONCLUSION Fibroblasts from patients with EoE secrete a unique ECM proteome that reflects their in vivo state and induces collagen I and α-smooth muscle actin protein expression from normal fibroblasts. Thrombospondin-1 is a previously unappreciated profibrotic molecule in EoE.
Collapse
Affiliation(s)
- Lance Y Hsieh
- Department of Pediatrics, University of California, San Diego, La Jolla, Calif; Division of Allergy Immunology, University of California, San Diego, La Jolla, Calif
| | - Austin W T Chiang
- Department of Pediatrics, University of California, San Diego, La Jolla, Calif; Department of Bioengineering, University of California, San Diego, La Jolla, Calif
| | - Loan D Duong
- Department of Pediatrics, University of California, San Diego, La Jolla, Calif; Division of Allergy Immunology, University of California, San Diego, La Jolla, Calif
| | - Chih-Chung Kuo
- Department of Bioengineering, University of California, San Diego, La Jolla, Calif
| | - Stephanie X Dong
- Department of Pediatrics, University of California, San Diego, La Jolla, Calif; Division of Allergy Immunology, University of California, San Diego, La Jolla, Calif
| | - Ranjan Dohil
- Department of Pediatrics, University of California, San Diego, La Jolla, Calif; Division of Gastroenterology, University of California, San Diego, La Jolla, Calif; Rady Children's Hospital San Diego, Calif, San Diego, Calif
| | - Richard Kurten
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, Ark
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, Calif; Department of Bioengineering, University of California, San Diego, La Jolla, Calif
| | - Seema S Aceves
- Department of Pediatrics, University of California, San Diego, La Jolla, Calif; Division of Allergy Immunology, University of California, San Diego, La Jolla, Calif; Rady Children's Hospital San Diego, Calif, San Diego, Calif; Department of Medicine, University of California, San Diego, La Jolla, Calif.
| |
Collapse
|
49
|
Erwin EA, Jaramillo LM, Smith B, Kruszewski PG, Kahwash B, Grayson MH, Mejias A, Ramilo O. Sex Differences in Blood Transcriptional Profiles and Clinical Phenotypes in Pediatric Patients with Eosinophilic Esophagitis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 9:3350-3358.e8. [PMID: 34265446 DOI: 10.1016/j.jaip.2021.06.043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) is an increasingly recognized, chronic inflammatory disease. Recent reports suggest clinical differences between males and females. OBJECTIVE To define the relevant molecular pathways that could be related to clinical phenotypes in children with EoE. METHODS We performed blood RNA expression analysis in children with newly diagnosed EoE and matched, healthy controls, and applied bioinformatics tools to define EoE host immune biosignatures. Questionnaires and medical records were used to characterize symptoms, esophagogastroduodenoscopy results, and treatment response. RESULTS Forty-one subjects (aged 2-17 years) were enrolled; the cohort consisted of 27 males and 14 females. Patients were randomly divided into a discovery cohort (21 EoE patients and 12 controls) that identified 544 significant differentially expressed transcripts (P ≤ .01; 1.25-fold change). Those 544 transcripts correctly classified most EoE patients in the validation cohort (n = 20) from healthy controls. Global transcriptional perturbation relative to healthy controls, Molecular Distance to Health scores were greater in EoE patients than controls (P = .003). When we analyzed subjects based on age and sex, males 13 years of age and older were more likely to have food impactions (P = .033) and to have higher endoscopic severity scores (P = .036). Separate group comparisons according to sex identified 294 differentially expressed transcripts in males and 643 transcripts in female EoE patients. Of those, 37 genes were shared and similarly expressed irrespective of sex. CONCLUSIONS Whole blood transcriptional analysis represents a promising noninvasive tool to assess activity of the immune/inflammatory response in children with EoE. Male and female EoE patients showed robust differences in gene expression suggesting distinct pathogenic endotypes.
Collapse
Affiliation(s)
- Elizabeth A Erwin
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children's Hospital and the Ohio State University College of Medicine, Columbus, Ohio.
| | - Lisa M Jaramillo
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Bennett Smith
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Patrice G Kruszewski
- Division of Gastroenterology, Hepatology and Nutrition, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, Ga
| | - Basil Kahwash
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children's Hospital and the Ohio State University College of Medicine, Columbus, Ohio
| | - Mitchell H Grayson
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children's Hospital and the Ohio State University College of Medicine, Columbus, Ohio; Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Asuncion Mejias
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio; Division of Infectious Diseases, Department of Pediatrics, Nationwide Children's Hospital and the Ohio State University College of Medicine, Columbus, Ohio
| | - Octavio Ramilo
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio; Division of Infectious Diseases, Department of Pediatrics, Nationwide Children's Hospital and the Ohio State University College of Medicine, Columbus, Ohio
| |
Collapse
|
50
|
Kaymak T, Hruz P, Niess JH. Immune system and microbiome in the esophagus: implications for understanding inflammatory diseases. FEBS J 2021; 289:4758-4772. [PMID: 34213831 PMCID: PMC9542113 DOI: 10.1111/febs.16103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 05/20/2021] [Accepted: 07/01/2021] [Indexed: 12/15/2022]
Abstract
The gastrointestinal tract is the largest compartment of the body's immune system exposed to microorganisms, structural components and metabolites, antigens derived from the diet, and pathogens. Most studies have focused on immune responses in the stomach, the small intestine, and the colon, but the esophagus has remained an understudied anatomic immune segment. Here, we discuss the esophagus' anatomical and physiological distinctions that may account for inflammatory esophageal diseases.
Collapse
Affiliation(s)
- Tanay Kaymak
- Department of Biomedicine, University of Basel, Switzerland
| | - Petr Hruz
- Clarunis - University Center for Gastrointestinal and Liver Diseases Basel, Switzerland
| | - Jan Hendrik Niess
- Department of Biomedicine, University of Basel, Switzerland.,Clarunis - University Center for Gastrointestinal and Liver Diseases Basel, Switzerland
| |
Collapse
|