1
|
Pyne AL, Uchida AM, Hazel MW, Pletneva MA, Allen-Brady K, Peterson KA. Differing Epithelial and Immunologic Activation Patterns Following Food Reintroduction Reveal Different Transcriptional Profiles in Active Eosinophilic Esophagitis. Gastroenterology 2025; 168:598-600.e3. [PMID: 39490770 DOI: 10.1053/j.gastro.2024.10.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/26/2024] [Accepted: 10/13/2024] [Indexed: 11/05/2024]
Affiliation(s)
- Ashley L Pyne
- Division of Gastroenterology, Hepatology & Nutrition, University of Utah School of Medicine, Salt Lake City, Utah.
| | - Amiko M Uchida
- Division of Gastroenterology, Hepatology & Nutrition, University of Utah School of Medicine, Salt Lake City, Utah
| | - Mark W Hazel
- Division of Gastroenterology, Hepatology & Nutrition, University of Utah School of Medicine, Salt Lake City, Utah
| | - Maria A Pletneva
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah
| | - Kristina Allen-Brady
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Kathryn A Peterson
- Division of Gastroenterology, Hepatology & Nutrition, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
2
|
Hoelz H, Faro T, Frank ML, Forné I, Kugelmann D, Jurk A, Buehler S, Siebert K, Matchado M, Straub T, Hering A, Piontek G, Mueller S, Koletzko S, List M, Steiger K, Rudelius M, Waschke J, Schwerd T. Persistent desmoglein-1 downregulation and periostin accumulation in histologic remission of eosinophilic esophagitis. J Allergy Clin Immunol 2025; 155:505-519. [PMID: 39343172 DOI: 10.1016/j.jaci.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/19/2024] [Accepted: 09/04/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Patients with eosinophilic esophagitis (EoE) require long-lasting resolution of inflammation to prevent fibrostenosis and dysphagia. However, the dissociation between symptoms and histologic improvement suggests persistent molecular drivers despite histologic remission. OBJECTIVE We characterized persisting molecular alterations in pediatric patients with EoE using tissue transcriptomics and proteomics. METHODS Esophageal biopsy samples (n = 247) collected prospectively during 189 endoscopies from pediatric patients with EoE (n = 36, up to 11 follow-up endoscopies) and pediatric controls (n = 44, single endoscopies) were subjected to bulk transcriptomics (n = 96) and proteomics (n = 151). Intercellular junctions (desmoglein-1/3, desmoplakin, E-cadherin) and epithelial-to-mesenchymal transition (vimentin:E-cadherin ratio) were assessed by immunofluorescence staining. RESULTS Active EoE (≥15 eosinophils per high-power field [eos/hpf]), inactive EoE (<15 eos/hpf), and deep-remission EoE (0 eos/hpf) were diagnosed in 107 of 185, 78 of 185, and 41 of 185 biopsy samples, respectively. Among the dysregulated genes (up-/downregulated 310/112) and proteins (up-/downregulated 68/16) between active EoE and controls, 17 genes, and 6 proteins remained dysregulated in inactive EoE. Using persistently upregulated genes (n = 9) and proteins (n = 3) only, such as ALOX15, CXCL1, CXCL6, CTSG, CDH26, PRRX1, CLC, EPX, and periostin (POSTN), was sufficient to separate inactive EoE and deep-remission biopsy samples from control tissue. While 32 differentially expressed genes persisted in deep-remission EoE compared to controls, the proteome normalized except for persistently upregulated POSTN. Epithelial-to-mesenchymal transition normalized in inactive EoE, whereas desmosome recovery remained impaired as a result of desmoglein-1 downregulation. CONCLUSION The analysis of molecular changes shows persistent EoE-associated esophageal dysregulation despite histologic remission. These data expand our understanding of inflammatory processes and possible mechanisms that underlie tissue remodeling in EoE.
Collapse
Affiliation(s)
- Hannes Hoelz
- Department of Pediatrics, Dr von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Tim Faro
- Department of Pediatrics, Dr von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Marie-Luise Frank
- Department of Pediatrics, Dr von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Ignasi Forné
- Protein Analysis Unit, Biomedical Center Munich, LMU Munich, Munich, Germany
| | - Daniela Kugelmann
- Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Anja Jurk
- Department of Pediatrics, Dr von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Simon Buehler
- Department of Pediatrics, Dr von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Kolja Siebert
- Department of Pediatrics, Dr von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Monica Matchado
- Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Tobias Straub
- Biomedical Center Munich, Bioinformatics Core Facility, LMU Munich, Munich, Germany
| | - Annett Hering
- Institute of Pathology, School of Medicine and Health, Technische Universität München, Munich, Germany; Comparative Experimental Pathology, School of Medicine and Health, Technische Universität München, Munich, Germany
| | - Guido Piontek
- Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Susanna Mueller
- Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Sibylle Koletzko
- Department of Pediatrics, Dr von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany; Department of Pediatrics, Gastroenterology and Nutrition, School of Medicine Collegium Medicum University of Warmia and Mazury, Olsztyn, Poland
| | - Markus List
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Freising, Germany; Munich Data Science Institute (MDSI), Technical University of Munich, Garching, Germany
| | - Katja Steiger
- Institute of Pathology, School of Medicine and Health, Technische Universität München, Munich, Germany; Comparative Experimental Pathology, School of Medicine and Health, Technische Universität München, Munich, Germany
| | - Martina Rudelius
- Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Jens Waschke
- Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Tobias Schwerd
- Department of Pediatrics, Dr von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
3
|
Lal M, Burk CM, Gautam R, Mrozek Z, Canziani KE, Trachsel T, Beers J, Carroll MC, Morgan DM, Muir AB, Shreffler WG, Ruffner MA. Interferon-γ Signaling in Eosinophilic Esophagitis Affects Epithelial Barrier Function and Programmed Cell Death. Cell Mol Gastroenterol Hepatol 2025; 19:101466. [PMID: 39884574 PMCID: PMC11964763 DOI: 10.1016/j.jcmgh.2025.101466] [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: 04/08/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND & AIMS Eosinophilic esophagitis (EoE) is a chronic esophageal inflammatory disorder characterized by eosinophil-rich mucosal inflammation and tissue remodeling. Prior research has revealed the upregulation of interferon (IFN) response signature genes (ISGs) in biopsy tissue from patients with EoE, but the specific cell types that contribute to this IFN response and the effect of interferons on the esophageal epithelium remain incompletely understood. Here, we use single-cell RNA sequencing (scRNA-seq) to examine the expression of IFN and ISGs during EoE and explore how IFN-α and IFN-γ treatments affect epithelial function. METHODS Epithelial gene expression from patients with EoE was examined using scRNA-seq and a confirmatory bulk RNA-seq experiment of isolated epithelial cells. The functional impact of IFN-α and IFN-γ on epithelial cells was investigated using organoid models. RESULTS Using scRNA-seq, the highest number of differentially regulated ISGs was found in the epithelial cells of patients with active EoE, and ISGs in transitional epithelial cells correlated significantly with eosinophil counts and endoscopic reference scores. IFN-γ and IFN-α treatments reduced organoid formation rate and size in a dose-dependent manner, with IFN-γ showing a more pronounced impact on measures of epithelial barrier formation and induction of caspase activity. We identify high IFNG expression in a cluster of majority CD8+ T cells with high expression of CD69 and FOS. CONCLUSIONS These findings reveal that interferon, especially IFN-γ, plays a central role in epithelial cell dysfunction, significantly affecting gene expression, cellular differentiation, and barrier integrity. Clarifying the contribution of varied cytokine signals in EoE may help explain the heterogeneity in patient presentation and therapeutic response.
Collapse
Affiliation(s)
- Megha Lal
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Caitlin M Burk
- Food Allergy Center and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Massachusetts
| | - Ravi Gautam
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Zoe Mrozek
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Karina E Canziani
- Food Allergy Center and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Massachusetts
| | - Tina Trachsel
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Division of Allergy, University Children's Hospital Zurich, Zurich, Switzerland; Division of Allergy, University Children's Hospital Basel, Basel, Switzerland
| | - Jarad Beers
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Margaret C Carroll
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Duncan M Morgan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT); Department of Chemical Engineering, MIT, Cambridge, Massachusetts
| | - Amanda B Muir
- Divison of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania, Philadelphia, Pennsylvania
| | - Wayne G Shreffler
- Food Allergy Center and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Massachusetts; Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Melanie A Ruffner
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
4
|
Choksi Y. Interferon Signaling Alters Epithelial Function in Eosinophilic Esophagitis. Cell Mol Gastroenterol Hepatol 2025; 19:101464. [PMID: 39870362 PMCID: PMC12009094 DOI: 10.1016/j.jcmgh.2025.101464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 01/09/2025] [Indexed: 01/29/2025]
Affiliation(s)
- Yash Choksi
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
5
|
Laserna‐Mendieta EJ, Casabona‐Francés S, Amorena E, Savarino EV, Pérez‐Martínez I, Blas‐Jhon L, Guardiola‐Arévalo A, Coletta M, Pellegatta G, Guagnozzi D, Barrio J, Perello A, Betoré E, Krarup AL, Votto M, Gutiérrez‐Junquera C, Naves JE, Oliva S, Teruel Sánchez‐Vegazo C, Carrión S, de la Riva S, Espina‐Cadenas S, Fernández‐Fernández S, Llorente‐Barrio M, Pascual‐Lopez I, Masiques‐Mas ML, Honrubia‐López R, Dainese R, García‐Morales N, Cobian J, Bisso‐Zein JK, Roales V, Juan‐Juan A, Rodríguez‐Sánchez A, Feo‐Ortega S, Martín‐Domínguez V, Nantes‐Castillejo Ó, Nicolay‐Maneru J, Ghisa M, Maniero D, Suarez A, Maray I, Álvarez‐García M, Granja‐Navacerrada A, Penagini R, Racca F, Llerena‐Castro R, Santander C, Arias Á, Lucendo AJ, EUREOS and EoE CONNECT research group. Sex-related differences in the presentation, management and response to treatment of eosinophilic esophagitis: Cross sectional analysis of EoE CONNECT registry. United European Gastroenterol J 2024; 12:1388-1398. [PMID: 39513462 PMCID: PMC11652331 DOI: 10.1002/ueg2.12699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/02/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) predominantly affects males across all ages; however, little is known about sex differences for other aspects of EoE. OBJECTIVE To investigate associations between sex and clinical presentation, endoscopic features, treatment choice and response in EoE patients in real-world practice. METHODS Cross-sectional analysis of the multicenter EoE CONNECT registry. The independent contribution of patients' sex and other relevant variables were statistically assessed by multivariate models. RESULTS A total of 2976 patients (76% male) were evaluated. Males were diagnosed at a younger age compared to females (32.7 ± 14.8 vs. 34.8 ± 15.6 years, respectively; p = 0.002) with similar diagnostic delay. EoE symptoms varied significantly between sexes, with food impaction predominating in males and dysphagia, heartburn, regurgitation and abdominal and epigastric pain in females. However, female sex contributed to higher symptom severity at diagnosis as measured with Dysphagia Symptom Score (R2 = 0.57; p = 0.013) and presented higher peak eosinophil count in esophageal biopsies (p = 0.005). Males showed increased risk of stricturing or mixed phenotypes (adjusted OR 1.43, 95%CI:1.05-1.96; p = 0.024). No association was found between patients' sex and first-line treatment modality: proton pump inhibitors (PPI) were preferred over topical corticosteroids in patients with inflammatory phenotypes instead of stricturing or mixed phenotypes, and in patients who did not present food impaction. Both topical corticosteroids and dietary interventions were preferred over PPI in pediatric patients regardless of sex. CONCLUSIONS Sex is associated with clinical and phenotypical presentation of EoE at diagnosis, with more fibrotic findings in males but higher symptom score in females.
Collapse
Affiliation(s)
- Emilio J. Laserna‐Mendieta
- Department of GastroenterologyHospital General de TomellosoTomellosoSpain
- Instituto de Investigación Sanitaria La PrincesaMadridSpain
- Instituto de Investigación Sanitaria de Castilla‐La Mancha (IDISCAM)MadridSpain
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y DigestivasMadridSpain
| | - Sergio Casabona‐Francés
- Instituto de Investigación Sanitaria La PrincesaMadridSpain
- Department of GastroenterologyHospital Universitario de La PrincesaMadridSpain
| | - Edurne Amorena
- Department of GastroenterologyComplejo Universitario de NavarraPamplonaSpain
| | - Edoardo V. Savarino
- Surgery, Oncology and Gastroenterology, Gastroenterology UnitAzienza Ospedaliera di PadovaPadovaItaly
| | - Isabel Pérez‐Martínez
- Department of GastroenterologyHospital Universitario Central de AsturiasOviedoSpain
- Diet, Microbiota and Health GroupInstituto de Investigación Sanitaria del Principado de Asturias (ISPA)OviedoSpain
| | | | | | - Marina Coletta
- Department of GastroenterologyFondazione IRCCS Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Gaia Pellegatta
- Endoscopy UnitDepartment of GastroenterologyIRCCS Humanitas Research HospitalMilanItaly
| | - Danila Guagnozzi
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y DigestivasMadridSpain
- Department of GastroenterologyHospital Universitario Vall d’HebrónBarcelonaSpain
| | - Jesús Barrio
- Department of GastroenterologyHospital Universitario Rio HortegaValladolidSpain
| | - Antonia Perello
- Department of GastroenterologyHospital Universitari Son EspasesPalma de MallorcaSpain
| | - Elena Betoré
- Department of GastroenterologyHospital Universitario Miguel ServetZaragozaSpain
| | - Anne Lund Krarup
- Department of Emergency Medicine and Trauma CenterDepartment of Gastroenterology and HepatologyAalborg University Hospital and Institute of Clinical Medicine Aalborg UniversityAalborgDenmark
| | - Martina Votto
- Pediatric UnitDepartment of Clinical, Surgical, Diagnostic and Pediatric SciencesUniversity of PaviaPaviaItaly
| | | | | | - Salvatore Oliva
- Department of Pediatric Digestive EndoscopyUniversity Hospital Umberto I & Sapienza University of RomeRomeItaly
| | | | - Silvia Carrión
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y DigestivasMadridSpain
- Department of GastroenterologyHospital de MataróMataróSpain
| | - Susana de la Riva
- Department of GastroenterologyClínica Universidad de NavarraPamplonaSpain
| | | | | | | | - Irene Pascual‐Lopez
- Department of GastroenterologyHospital Universitario Arnau de Vilanova & Hospital Universitario Santa MaríaLéridaSpain
| | | | - Raúl Honrubia‐López
- Department of GastroenterologyHospital Universitario Infanta SofíaSan Sebastián de los ReyesSpain
| | - Raffaella Dainese
- Department of GastroenterologyCentre Hospitalier d'Antibes Juan‐les‐PinsAntibesFrance
| | | | - Julyssa Cobian
- Department of GastroenterologyHospital DonostiaSan SebastiánSpain
| | | | - Valentín Roales
- Department of GastroenterologyHospital Clínico Universitario San CarlosMadridSpain
| | - Alba Juan‐Juan
- Department of GastroenterologyHospital Sant Joan Despí Moisès BroggiBarcelonaSpain
| | | | - Sara Feo‐Ortega
- Instituto de Investigación Sanitaria de Castilla‐La Mancha (IDISCAM)MadridSpain
- Department of PediatricsHospital General de TomellosoTomellosoSpain
| | - Verónica Martín‐Domínguez
- Instituto de Investigación Sanitaria La PrincesaMadridSpain
- Department of GastroenterologyHospital Universitario de La PrincesaMadridSpain
| | - Óscar Nantes‐Castillejo
- Department of GastroenterologyComplejo Universitario de NavarraPamplonaSpain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA)PamplonaSpain
| | | | - Matteo Ghisa
- Surgery, Oncology and Gastroenterology, Gastroenterology UnitAzienza Ospedaliera di PadovaPadovaItaly
| | - Daria Maniero
- Surgery, Oncology and Gastroenterology, Gastroenterology UnitAzienza Ospedaliera di PadovaPadovaItaly
| | - Adolfo Suarez
- Department of GastroenterologyHospital Universitario Central de AsturiasOviedoSpain
- Diet, Microbiota and Health GroupInstituto de Investigación Sanitaria del Principado de Asturias (ISPA)OviedoSpain
| | - Iván Maray
- Diet, Microbiota and Health GroupInstituto de Investigación Sanitaria del Principado de Asturias (ISPA)OviedoSpain
- Department of PharmacyHospital Universitario Central de AsturiasOviedoSpain
| | | | | | - Roberto Penagini
- Department of GastroenterologyFondazione IRCCS Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Francesca Racca
- Personalized Medicine, Asthma and Allergy ClinicIRCCS Humanitas Research HospitalRozzano ‐ MilanItaly
| | | | - Cecilio Santander
- Instituto de Investigación Sanitaria La PrincesaMadridSpain
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y DigestivasMadridSpain
- Department of GastroenterologyHospital Universitario de La PrincesaMadridSpain
| | - Ángel Arias
- Instituto de Investigación Sanitaria La PrincesaMadridSpain
- Instituto de Investigación Sanitaria de Castilla‐La Mancha (IDISCAM)MadridSpain
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y DigestivasMadridSpain
- Research Support UnitHospital General La Mancha CentroAlcázar de San JuanSpain
| | - Alfredo J. Lucendo
- Department of GastroenterologyHospital General de TomellosoTomellosoSpain
- Instituto de Investigación Sanitaria La PrincesaMadridSpain
- Instituto de Investigación Sanitaria de Castilla‐La Mancha (IDISCAM)MadridSpain
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y DigestivasMadridSpain
| | | |
Collapse
|
6
|
Ryan S, Crowe L, Almeida Cruz SN, Galbraith MD, O'Brien C, Hammer JA, Bergin R, Kellett SK, Markey GE, Benson TM, Fagan O, Espinosa JM, Conlon N, Donohoe CL, McKiernan S, Hogan AE, McNamee EN, Furuta GT, Menard-Katcher C, Masterson JC. Metabolic dysfunction mediated by HIF-1α contributes to epithelial differentiation defects in eosinophilic esophagitis. J Allergy Clin Immunol 2024; 154:1472-1488. [PMID: 39209164 DOI: 10.1016/j.jaci.2024.07.030] [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: 03/12/2024] [Revised: 07/10/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Investigating the contributory role that epithelial cell metabolism plays in allergic inflammation is a key factor to understanding what influences dysfunction and the pathogenesis of the allergic disease eosinophilic esophagitis (EoE). We previously highlighted that the absence of hypoxia signaling through hypoxia-inducible factor (HIF)-1α in EoE contributes to esophageal epithelial dysfunction. However, metabolic regulation by HIF-1α has not been explored in esophageal allergy. OBJECTIVES We sought to define the role of HIF-1α-mediated metabolic dysfunction in esophageal epithelial differentiation processes and barrier function in EoE. METHODS In RNA sequencing of EoE patient biopsy samples, we observed the expression pattern of key genes involved in mitochondrial metabolism/oxidative phosphorylation (OXPHOS) and glycolysis. Seahorse bioenergetics analysis was performed on EPC2-hTERT cells to decipher the metabolic processes involved in epithelial differentiation processes. In addition, air-liquid interface cultures were used to delineate metabolic dependency mechanisms required for epithelial differentiation. RESULTS Transcriptomic analysis identified an increase in genes associated with OXPHOS in patients with EoE. Epithelial origin of this signature was confirmed by complex V immunofluorescence of patient biopsy samples. Bioenergetic analysis in vitro revealed that differentiated epithelium was less reliant on OXPHOS compared with undifferentiated epithelium. Increased OXPHOS potential and reduced glycolytic capacity was mirrored in HIF1A-knockdown EPC2-hTERT cells that exhibited a significant absence of terminal markers of epithelial differentiation, including involucrin. Pharmacologic glucose transport inhibition phenocopied this, while rescue of the HIF-1α-deficient phenotype using the pan-prolyl hydroxylase inhibitor dimethyloxalylglycine resulted in restored expression of epithelial differentiation markers. CONCLUSIONS An OXPHOS-dominated metabolic pattern in EoE patients, brought about largely by the absence of HIF-1α-mediated glycolysis, is linked with the deficit in esophageal epithelial differentiation.
Collapse
Affiliation(s)
- Sinéad Ryan
- Allergy, Inflammation, and Remodeling Research Laboratory, Department of Biology, National University of Ireland, Maynooth, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Louise Crowe
- Allergy, Inflammation, and Remodeling Research Laboratory, Department of Biology, National University of Ireland, Maynooth, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Sofía N Almeida Cruz
- Allergy, Inflammation, and Remodeling Research Laboratory, Department of Biology, National University of Ireland, Maynooth, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Matthew D Galbraith
- Linda Crinc Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, Colo; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Carol O'Brien
- Allergy, Inflammation, and Remodeling Research Laboratory, Department of Biology, National University of Ireland, Maynooth, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Juliet A Hammer
- Gastrointestinal Eosinophilic Diseases Program, Digestive Health Institute, Children's Hospital Colorado, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colo
| | - Ronan Bergin
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Shauna K Kellett
- Allergy, Inflammation, and Remodeling Research Laboratory, Department of Biology, National University of Ireland, Maynooth, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Gary E Markey
- Allergy, Inflammation, and Remodeling Research Laboratory, Department of Biology, National University of Ireland, Maynooth, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Taylor M Benson
- Allergy, Inflammation, and Remodeling Research Laboratory, Department of Biology, National University of Ireland, Maynooth, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Olga Fagan
- Department of Gastroenterology, St James's Hospital, Dublin, Ireland
| | - Joaquin M Espinosa
- Linda Crinc Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Niall Conlon
- Department of Allergy and Immunology, St James's Hospital, Dublin, Ireland
| | - Claire L Donohoe
- National Centre for Oesophageal and Gastric Cancer, Trinity St James's Cancer Institute, St James's Hospital, Trinity College, Dublin, Ireland
| | - Susan McKiernan
- Department of Gastroenterology, St James's Hospital, Dublin, Ireland
| | - Andrew E Hogan
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland; Department of Biology, Obesity Immunology Research Group, Maynooth University, Maynooth, Ireland
| | - Eóin N McNamee
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland; Department of Biology, Mucosal Immunology Research Laboratory, National University of Ireland, Maynooth, Ireland
| | - Glenn T Furuta
- Gastrointestinal Eosinophilic Diseases Program, Digestive Health Institute, Children's Hospital Colorado, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colo
| | - Calies Menard-Katcher
- Gastrointestinal Eosinophilic Diseases Program, Digestive Health Institute, Children's Hospital Colorado, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colo
| | - Joanne C Masterson
- Allergy, Inflammation, and Remodeling Research Laboratory, Department of Biology, National University of Ireland, Maynooth, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland; Gastrointestinal Eosinophilic Diseases Program, Digestive Health Institute, Children's Hospital Colorado, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colo.
| |
Collapse
|
7
|
Shimonosono M, Morimoto M, Hirose W, Tomita Y, Matsuura N, Flashner S, Ebadi MS, Okayasu EH, Lee CY, Britton WR, Martin C, Wuertz BR, Parikh AS, Sachdeva UM, Ondrey FG, Atigadda VR, Elmets CA, Abrams JA, Muir AB, Klein-Szanto AJ, Weinberg KI, Momen-Heravi F, Nakagawa H. Modeling Epithelial Homeostasis and Perturbation in Three-Dimensional Human Esophageal Organoids. Biomolecules 2024; 14:1126. [PMID: 39334892 PMCID: PMC11430971 DOI: 10.3390/biom14091126] [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: 05/07/2024] [Revised: 08/01/2024] [Accepted: 08/19/2024] [Indexed: 09/30/2024] Open
Abstract
Background: Esophageal organoids from a variety of pathologies including cancer are grown in Advanced Dulbecco's Modified Eagle Medium-Nutrient Mixture F12 (hereafter ADF). However, the currently available ADF-based formulations are suboptimal for normal human esophageal organoids, limiting the ability to compare normal esophageal organoids with those representing a given disease state. Methods: We have utilized immortalized normal human esophageal epithelial cell (keratinocyte) lines EPC1 and EPC2 and endoscopic normal esophageal biopsies to generate three-dimensional (3D) organoids. To optimize the ADF-based medium, we evaluated the requirement of exogenous epidermal growth factor (EGF) and inhibition of transforming growth factor-(TGF)-β receptor-mediated signaling, both key regulators of the proliferation of human esophageal keratinocytes. We have modeled human esophageal epithelial pathology by stimulating esophageal 3D organoids with interleukin (IL)-13, an inflammatory cytokine, or UAB30, a novel pharmacological activator of retinoic acid signaling. Results: The formation of normal human esophageal 3D organoids was limited by excessive EGF and intrinsic TGFβ-receptor-mediated signaling. Optimized HOME0 improved normal human esophageal organoid formation. In the HOME0-grown organoids, IL-13 and UAB30 induced epithelial changes reminiscent of basal cell hyperplasia, a common histopathologic feature in broad esophageal disease conditions including eosinophilic esophagitis. Conclusions: HOME0 allows modeling of the homeostatic differentiation gradient and perturbation of the human esophageal epithelium while permitting a comparison of organoids from mice and other organs grown in ADF-based media.
Collapse
Affiliation(s)
- Masataka Shimonosono
- Herbert Irving Comprehensive Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (M.M.); (W.H.); (Y.T.); (N.M.); (S.F.); (M.S.E.); (E.H.O.); (C.Y.L.); (W.R.B.); (C.M.); (A.S.P.); (J.A.A.); (F.M.-H.)
| | - Masaki Morimoto
- Herbert Irving Comprehensive Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (M.M.); (W.H.); (Y.T.); (N.M.); (S.F.); (M.S.E.); (E.H.O.); (C.Y.L.); (W.R.B.); (C.M.); (A.S.P.); (J.A.A.); (F.M.-H.)
| | - Wataru Hirose
- Herbert Irving Comprehensive Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (M.M.); (W.H.); (Y.T.); (N.M.); (S.F.); (M.S.E.); (E.H.O.); (C.Y.L.); (W.R.B.); (C.M.); (A.S.P.); (J.A.A.); (F.M.-H.)
| | - Yasuto Tomita
- Herbert Irving Comprehensive Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (M.M.); (W.H.); (Y.T.); (N.M.); (S.F.); (M.S.E.); (E.H.O.); (C.Y.L.); (W.R.B.); (C.M.); (A.S.P.); (J.A.A.); (F.M.-H.)
| | - Norihiro Matsuura
- Herbert Irving Comprehensive Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (M.M.); (W.H.); (Y.T.); (N.M.); (S.F.); (M.S.E.); (E.H.O.); (C.Y.L.); (W.R.B.); (C.M.); (A.S.P.); (J.A.A.); (F.M.-H.)
| | - Samuel Flashner
- Herbert Irving Comprehensive Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (M.M.); (W.H.); (Y.T.); (N.M.); (S.F.); (M.S.E.); (E.H.O.); (C.Y.L.); (W.R.B.); (C.M.); (A.S.P.); (J.A.A.); (F.M.-H.)
| | - Mesra S. Ebadi
- Herbert Irving Comprehensive Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (M.M.); (W.H.); (Y.T.); (N.M.); (S.F.); (M.S.E.); (E.H.O.); (C.Y.L.); (W.R.B.); (C.M.); (A.S.P.); (J.A.A.); (F.M.-H.)
| | - Emilea H. Okayasu
- Herbert Irving Comprehensive Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (M.M.); (W.H.); (Y.T.); (N.M.); (S.F.); (M.S.E.); (E.H.O.); (C.Y.L.); (W.R.B.); (C.M.); (A.S.P.); (J.A.A.); (F.M.-H.)
| | - Christian Y. Lee
- Herbert Irving Comprehensive Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (M.M.); (W.H.); (Y.T.); (N.M.); (S.F.); (M.S.E.); (E.H.O.); (C.Y.L.); (W.R.B.); (C.M.); (A.S.P.); (J.A.A.); (F.M.-H.)
| | - William R. Britton
- Herbert Irving Comprehensive Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (M.M.); (W.H.); (Y.T.); (N.M.); (S.F.); (M.S.E.); (E.H.O.); (C.Y.L.); (W.R.B.); (C.M.); (A.S.P.); (J.A.A.); (F.M.-H.)
| | - Cecilia Martin
- Herbert Irving Comprehensive Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (M.M.); (W.H.); (Y.T.); (N.M.); (S.F.); (M.S.E.); (E.H.O.); (C.Y.L.); (W.R.B.); (C.M.); (A.S.P.); (J.A.A.); (F.M.-H.)
- Organoid & Cell Culture Core, Columbia University Digestive and Liver Diseases Research Center, New York, NY 10032, USA
| | - Beverly R. Wuertz
- Department of Otolaryngology, Head and Neck Surgery, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; (B.R.W.); (F.G.O.)
| | - Anuraag S. Parikh
- Herbert Irving Comprehensive Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (M.M.); (W.H.); (Y.T.); (N.M.); (S.F.); (M.S.E.); (E.H.O.); (C.Y.L.); (W.R.B.); (C.M.); (A.S.P.); (J.A.A.); (F.M.-H.)
- Department of Otolaryngology, Head and Neck Surgery, Columbia University, New York, NY 10032, USA
| | - Uma M. Sachdeva
- Division of Thoracic Surgery, Massachusetts General Hospital, Boston, MA 02114, USA;
| | - Frank G. Ondrey
- Department of Otolaryngology, Head and Neck Surgery, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; (B.R.W.); (F.G.O.)
| | - Venkatram R. Atigadda
- Department of Dermatology, University of Alabama, Birmingham, AL 35294, USA; (V.R.A.); (C.A.E.)
| | - Craig A. Elmets
- Department of Dermatology, University of Alabama, Birmingham, AL 35294, USA; (V.R.A.); (C.A.E.)
| | - Julian A. Abrams
- Herbert Irving Comprehensive Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (M.M.); (W.H.); (Y.T.); (N.M.); (S.F.); (M.S.E.); (E.H.O.); (C.Y.L.); (W.R.B.); (C.M.); (A.S.P.); (J.A.A.); (F.M.-H.)
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Amanda B. Muir
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA;
| | | | - Kenneth I. Weinberg
- Department of Pediatrics, Maternal & Child Health Research Institute, Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA;
| | - Fatemeh Momen-Heravi
- Herbert Irving Comprehensive Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (M.M.); (W.H.); (Y.T.); (N.M.); (S.F.); (M.S.E.); (E.H.O.); (C.Y.L.); (W.R.B.); (C.M.); (A.S.P.); (J.A.A.); (F.M.-H.)
- Cancer Biology and Immunology Laboratory, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Hiroshi Nakagawa
- Herbert Irving Comprehensive Cancer Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (M.M.); (W.H.); (Y.T.); (N.M.); (S.F.); (M.S.E.); (E.H.O.); (C.Y.L.); (W.R.B.); (C.M.); (A.S.P.); (J.A.A.); (F.M.-H.)
- Organoid & Cell Culture Core, Columbia University Digestive and Liver Diseases Research Center, New York, NY 10032, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
8
|
Laky K, Frischmeyer-Guerrerio PA. Development and dysfunction of structural cells in eosinophilic esophagitis. J Allergy Clin Immunol 2024; 153:1485-1499. [PMID: 38849184 PMCID: PMC11626564 DOI: 10.1016/j.jaci.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 06/09/2024]
Abstract
Eosinophilic esophagitis (EoE) is a disorder characterized by dysfunction and chronic local inflammation of the esophagus. The incidence and prevalence of EoE are increasing worldwide. The mechanisms responsible are poorly understood, and effective treatment options are limited. From the lumen outward, the esophagus comprises stratified squamous epithelium, lamina propria, and muscle. The tissue-specific nature of EoE strongly suggests that structural cells in the esophagus are involved in the EoE diathesis. Epithelial basal cell hyperplasia and dilated intercellular spaces are cardinal features of EoE. Some patients with EoE develop lamina propria fibrosis, strictures, or esophageal muscle dysmotility. Clinical symptoms of EoE are only weakly correlated with peak eosinophil count, implying that other cell types contribute to EoE pathogenesis. Epithelial, endothelial, muscle, and fibroblast cells can each initiate inflammation and repair, regulate tissue resident immune cells, recruit peripheral leukocytes, and tailor adaptive immune cell responses. A better understanding of how structural cells maintain tissue homeostasis, respond to cell-intrinsic and cell-extrinsic stressors, and exacerbate and/or resolve inflammatory responses in the esophagus is needed. This knowledge will facilitate the development of more efficacious treatment strategies for EoE that can restore homeostasis of both hematopoietic and structural elements in the esophagus.
Collapse
Affiliation(s)
- Karen Laky
- Food Allergy Research Section, Laboratory of Allergic Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| | - Pamela A Frischmeyer-Guerrerio
- Food Allergy Research Section, Laboratory of Allergic Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| |
Collapse
|
9
|
Gabryszewski SJ, Ruffner MA, Spergel JM. Pediatric and adult EoE: A spectrum or distinct diseases? J Allergy Clin Immunol 2024; 153:1533-1535. [PMID: 38555978 DOI: 10.1016/j.jaci.2024.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/02/2024]
Affiliation(s)
- Stanislaw J Gabryszewski
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Melanie A Ruffner
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa; Institute for Immunology and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Jonathan M Spergel
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pa; Institute for Immunology and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa.
| |
Collapse
|
10
|
Shimonosono M, Morimoto M, Hirose W, Tomita Y, Matsuura N, Flashner S, Ebadi MS, Okayasu EH, Lee CY, Britton WR, Martin C, Wuertz BR, Parikh AS, Sachdeva UM, Ondrey FG, Atigadda VR, Elmets CA, Abrams JA, Muir AB, Klein-Szanto AJ, Weinberg KI, Momen-Heravi F, Nakagawa H. Modeling epithelial homeostasis and perturbation in three-dimensional human esophageal organoids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.20.595023. [PMID: 38826379 PMCID: PMC11142071 DOI: 10.1101/2024.05.20.595023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Background Esophageal organoids from a variety of pathologies including cancer are grown in Advanced Dulbecco's Modified Eagle Medium-Nutrient Mixture F12 (hereafter ADF). However, the currently available ADF-based formulations are suboptimal for normal human esophageal organoids, limiting the ability to compare normal esophageal organoids with those representing a given disease state. Methods We have utilized immortalized normal human esophageal epithelial cell (keratinocyte) lines EPC1 and EPC2 and endoscopic normal esophageal biopsies to generate three-dimensional (3D) organoids. To optimize ADF-based medium, we evaluated the requirement of exogenous epidermal growth factor (EGF) and inhibition of transforming growth factor-(TGF)-β receptor-mediated signaling, both key regulators of proliferation of human esophageal keratinocytes. We have modeled human esophageal epithelial pathology by stimulating esophageal 3D organoids with interleukin (IL)-13, an inflammatory cytokine, or UAB30, a novel pharmacological activator of retinoic acid signaling. Results The formation of normal human esophageal 3D organoids was limited by excessive EGF and intrinsic TGFβ receptor-mediated signaling. In optimized HOME0, normal human esophageal organoid formation was improved, whereas IL-13 and UAB30 induced epithelial changes reminiscent of basal cell hyperplasia, a common histopathologic feature in broad esophageal disease conditions including eosinophilic esophagitis. Conclusions: HOME0 allows modeling of the homeostatic differentiation gradient and perturbation of the human esophageal epithelium while permitting a comparison of organoids from mice and other organs grown in ADF-based media.
Collapse
|
11
|
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
|
12
|
Jacobse J, Pilat JM, Li J, Brown RE, Kwag A, Buendia MA, Choksi YA, Washington MK, Williams CS, Markham NO, Short SP, Goettel JA. Distinct roles for interleukin-23 receptor signaling in regulatory T cells in sporadic and inflammation-associated carcinogenesis. Front Oncol 2024; 13:1276743. [PMID: 38375204 PMCID: PMC10876294 DOI: 10.3389/fonc.2023.1276743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 12/29/2023] [Indexed: 02/21/2024] Open
Abstract
Introduction The pro-inflammatory cytokine interleukin-23 (IL-23) has been implicated in colorectal cancer (CRC). Yet, the cell-specific contributions of IL-23 receptor (IL-23R) signaling in CRC remain unknown. One of the cell types that highly expresses IL-23R are colonic regulatory T cells (Treg cells). The aim of this study was to define the contribution of Treg cell-specific IL-23R signaling in sporadic and inflammation-associated CRC. Methods In mice, the role of IL-23R in Treg cells in colitis-associated cancer (CAC) was investigated using azoxymethane/dextran sodium sulphate in wild-type Treg cell reporter mice (WT, Foxp3 YFP-iCre), and mice harboring a Treg cell-specific deletion of IL-23 (Il23r ΔTreg). The role of IL-23R signaling in Treg cells in sporadic CRC was examined utilizing orthotopic injection of the syngeneic colon cancer cell line MC-38 submucosally into the colon/rectum of mice. The function of macrophages was studied using clodronate. Finally, single-cell RNA-seq of a previously published dataset in human sporadic cancer was reanalyzed to corroborate these findings. Results In CAC, Il23r ΔTreg mice had increased tumor size and increased dysplasia compared to WT mice that was associated with decreased tumor-infiltrating macrophages. In the sporadic cancer model, Il23r ΔTreg mice had increased survival and decreased tumor size compared to WT mice. Additionally, MC-38 tumors of Il23r ΔTreg mice exhibited a higher frequency of pro-inflammatory macrophages and IL-17 producing CD4+ T cells. The decreased tumor size in Il23r ΔTreg mice was macrophage-dependent. These data suggest that loss of IL-23R signaling in Treg cells permits IL-17 production by CD4+ T cells that in turn promotes pro-inflammatory macrophages to clear tumors. Finally, analysis of TCGA data and single-cell RNA-seq analysis of a previously published dataset in human sporadic cancer, revealed that IL23R was highly expressed in CRC compared to other cancers and specifically in tumor-associated Treg cells. Conclusion Inflammation in colorectal carcinogenesis differs with respect to the contribution of IL-23R signaling in regulatory T cells.
Collapse
Affiliation(s)
- Justin Jacobse
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pediatrics, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, Netherlands
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, United States
| | - Jennifer M. Pilat
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Jing Li
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Rachel E. Brown
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, United States
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Aaron Kwag
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Matthew A. Buendia
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Monroe Carell Jr. Children’s Hospital at Vanderbilt, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Yash A. Choksi
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, United States
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, United States
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, United States
| | - M. Kay Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Christopher S. Williams
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, United States
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, United States
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Nicholas O. Markham
- Department of Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, United States
| | - Sarah P. Short
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, United States
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jeremy A. Goettel
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, United States
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|