Your browser doesn't support javascript.
loading
Metabolic dysfunction mediated by HIF-1α contributes to epithelial differentiation defects in eosinophilic esophagitis.
Ryan, Sinéad; Crowe, Louise; Almeida Cruz, Sofía N; Galbraith, Matthew D; O'Brien, Carol; Hammer, Juliet A; Bergin, Ronan; Kellett, Shauna K; Markey, Gary E; Benson, Taylor M; Fagan, Olga; Espinosa, Joaquin M; Conlon, Niall; Donohoe, Claire L; McKiernan, Susan; Hogan, Andrew E; McNamee, Eóin N; Furuta, Glenn T; Menard-Katcher, Calies; Masterson, Joanne C.
Afiliación
  • Ryan S; 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.
  • Crowe L; 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.
  • Almeida Cruz SN; 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.
  • Galbraith MD; 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.
  • O'Brien C; 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.
  • Hammer JA; Gastrointestinal Eosinophilic Diseases Program, Digestive Health Institute, Children's Hospital Colorado, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colo.
  • Bergin R; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland.
  • Kellett SK; 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.
  • Markey GE; 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.
  • Benson TM; 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.
  • Fagan O; Department of Gastroenterology, St James's Hospital, Dublin, Ireland.
  • Espinosa JM; Linda Crinc Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, Colo.
  • Conlon N; Department of Allergy and Immunology, St James's Hospital, Dublin, Ireland.
  • Donohoe CL; National Centre for Oesophageal and Gastric Cancer, Trinity St James's Cancer Institute, St James's Hospital, Trinity College, Dublin, Ireland.
  • McKiernan S; Department of Gastroenterology, St James's Hospital, Dublin, Ireland.
  • Hogan AE; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland; Department of Biology, Obesity Immunology Research Group, Maynooth University, Maynooth, Ireland.
  • McNamee EN; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland; Department of Biology, Mucosal Immunology Research Laboratory, National University of Ireland, Maynooth, Ireland.
  • Furuta GT; Gastrointestinal Eosinophilic Diseases Program, Digestive Health Institute, Children's Hospital Colorado, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colo.
  • Menard-Katcher C; Gastrointestinal Eosinophilic Diseases Program, Digestive Health Institute, Children's Hospital Colorado, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colo.
  • Masterson JC; 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 Institu
Article en En | MEDLINE | ID: mdl-39209164
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.
Palabras clave

Texto completo: 1 Bases de datos: MEDLINE Idioma: En Revista: J Allergy Clin Immunol Año: 2024 Tipo del documento: Article País de afiliación: Irlanda

Texto completo: 1 Bases de datos: MEDLINE Idioma: En Revista: J Allergy Clin Immunol Año: 2024 Tipo del documento: Article País de afiliación: Irlanda