Cell-Type-Specific Effects of the Ovarian Cancer G-Protein Coupled Receptor (OGR1) on Inflammation and Fibrosis; Potential Implications for Idiopathic Pulmonary Fibrosis

Idiopathic pulmonary fibrosis (IPF) is a disease characterized by irreversible lung scarring. The pathophysiology is not fully understood, but the working hypothesis postulates that a combination of epithelial injury and myofibroblast differentiation drives progressive pulmonary fibrosis. We previou...

Full description

Bibliographic Details
Main Authors: David J. Nagel, Ashley R. Rackow, Wei-Yao Ku, Tyler J. Bell, Patricia J. Sime, Robert Matthew Kottmann
Format: Article
Language:English
Published: MDPI AG 2022-08-01
Series:Cells
Subjects:
Online Access:https://www.mdpi.com/2073-4409/11/16/2540
_version_ 1797432285535928320
author David J. Nagel
Ashley R. Rackow
Wei-Yao Ku
Tyler J. Bell
Patricia J. Sime
Robert Matthew Kottmann
author_facet David J. Nagel
Ashley R. Rackow
Wei-Yao Ku
Tyler J. Bell
Patricia J. Sime
Robert Matthew Kottmann
author_sort David J. Nagel
collection DOAJ
description Idiopathic pulmonary fibrosis (IPF) is a disease characterized by irreversible lung scarring. The pathophysiology is not fully understood, but the working hypothesis postulates that a combination of epithelial injury and myofibroblast differentiation drives progressive pulmonary fibrosis. We previously demonstrated that a reduction in extracellular pH activates latent TGF-β1, and that TGF-β1 then drives its own activation, creating a feed-forward mechanism that propagates myofibroblast differentiation. Given the important roles of extracellular pH in the progression of pulmonary fibrosis, we sought to identify whether pH mediates other cellular phenotypes independent of TGF-β1. Proton-sensing G-protein coupled receptors are activated by acidic environments, but their role in fibrosis has not been studied. Here, we report that the Ovarian Cancer G-Protein Coupled Receptor1 (OGR1 or GPR68) has dual roles in both promoting and mitigating pulmonary fibrosis. We demonstrate that OGR1 protein expression is significantly reduced in lung tissue from patients with IPF and that TGF-β1 decreases OGR1 expression. In fibroblasts, OGR1 inhibits myofibroblast differentiation and does not contribute to inflammation. However, in epithelial cells, OGR1 promotes epithelial to mesenchymal transition (EMT) and inflammation. We then demonstrate that sub-cellular localization and alternative signaling pathways may be responsible for the differential effect of OGR1 in each cell type. Our results suggest that strategies to selectively target OGR1 expression may represent a novel therapeutic strategy for pulmonary fibrosis.
first_indexed 2024-03-09T09:59:12Z
format Article
id doaj.art-3e6e3950c7e34610a09513be7ac00a77
institution Directory Open Access Journal
issn 2073-4409
language English
last_indexed 2024-03-09T09:59:12Z
publishDate 2022-08-01
publisher MDPI AG
record_format Article
series Cells
spelling doaj.art-3e6e3950c7e34610a09513be7ac00a772023-12-01T23:33:28ZengMDPI AGCells2073-44092022-08-011116254010.3390/cells11162540Cell-Type-Specific Effects of the Ovarian Cancer G-Protein Coupled Receptor (OGR1) on Inflammation and Fibrosis; Potential Implications for Idiopathic Pulmonary FibrosisDavid J. Nagel0Ashley R. Rackow1Wei-Yao Ku2Tyler J. Bell3Patricia J. Sime4Robert Matthew Kottmann5Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Rochester Medical Center, Rochester, NY 14642, USALaboratory Medicine, Department of Pathology, Division of Clinical Chemistry, Johns Hopkins University, Baltimore, MD 21287, USABMW of North America, Woodcliff Lake, NJ 07675, USADepartment of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USADepartment of Medicine, Virginia Commonwealth University Health System, Richmond, VA 23298, USADepartment of Medicine, Division of Pulmonary and Critical Care Medicine, University of Rochester Medical Center, Rochester, NY 14642, USAIdiopathic pulmonary fibrosis (IPF) is a disease characterized by irreversible lung scarring. The pathophysiology is not fully understood, but the working hypothesis postulates that a combination of epithelial injury and myofibroblast differentiation drives progressive pulmonary fibrosis. We previously demonstrated that a reduction in extracellular pH activates latent TGF-β1, and that TGF-β1 then drives its own activation, creating a feed-forward mechanism that propagates myofibroblast differentiation. Given the important roles of extracellular pH in the progression of pulmonary fibrosis, we sought to identify whether pH mediates other cellular phenotypes independent of TGF-β1. Proton-sensing G-protein coupled receptors are activated by acidic environments, but their role in fibrosis has not been studied. Here, we report that the Ovarian Cancer G-Protein Coupled Receptor1 (OGR1 or GPR68) has dual roles in both promoting and mitigating pulmonary fibrosis. We demonstrate that OGR1 protein expression is significantly reduced in lung tissue from patients with IPF and that TGF-β1 decreases OGR1 expression. In fibroblasts, OGR1 inhibits myofibroblast differentiation and does not contribute to inflammation. However, in epithelial cells, OGR1 promotes epithelial to mesenchymal transition (EMT) and inflammation. We then demonstrate that sub-cellular localization and alternative signaling pathways may be responsible for the differential effect of OGR1 in each cell type. Our results suggest that strategies to selectively target OGR1 expression may represent a novel therapeutic strategy for pulmonary fibrosis.https://www.mdpi.com/2073-4409/11/16/2540pulmonary fibrosisOGR1 (GPR68)fibroblastepithelial cellGPCRTGFβ signaling
spellingShingle David J. Nagel
Ashley R. Rackow
Wei-Yao Ku
Tyler J. Bell
Patricia J. Sime
Robert Matthew Kottmann
Cell-Type-Specific Effects of the Ovarian Cancer G-Protein Coupled Receptor (OGR1) on Inflammation and Fibrosis; Potential Implications for Idiopathic Pulmonary Fibrosis
Cells
pulmonary fibrosis
OGR1 (GPR68)
fibroblast
epithelial cell
GPCR
TGFβ signaling
title Cell-Type-Specific Effects of the Ovarian Cancer G-Protein Coupled Receptor (OGR1) on Inflammation and Fibrosis; Potential Implications for Idiopathic Pulmonary Fibrosis
title_full Cell-Type-Specific Effects of the Ovarian Cancer G-Protein Coupled Receptor (OGR1) on Inflammation and Fibrosis; Potential Implications for Idiopathic Pulmonary Fibrosis
title_fullStr Cell-Type-Specific Effects of the Ovarian Cancer G-Protein Coupled Receptor (OGR1) on Inflammation and Fibrosis; Potential Implications for Idiopathic Pulmonary Fibrosis
title_full_unstemmed Cell-Type-Specific Effects of the Ovarian Cancer G-Protein Coupled Receptor (OGR1) on Inflammation and Fibrosis; Potential Implications for Idiopathic Pulmonary Fibrosis
title_short Cell-Type-Specific Effects of the Ovarian Cancer G-Protein Coupled Receptor (OGR1) on Inflammation and Fibrosis; Potential Implications for Idiopathic Pulmonary Fibrosis
title_sort cell type specific effects of the ovarian cancer g protein coupled receptor ogr1 on inflammation and fibrosis potential implications for idiopathic pulmonary fibrosis
topic pulmonary fibrosis
OGR1 (GPR68)
fibroblast
epithelial cell
GPCR
TGFβ signaling
url https://www.mdpi.com/2073-4409/11/16/2540
work_keys_str_mv AT davidjnagel celltypespecificeffectsoftheovariancancergproteincoupledreceptorogr1oninflammationandfibrosispotentialimplicationsforidiopathicpulmonaryfibrosis
AT ashleyrrackow celltypespecificeffectsoftheovariancancergproteincoupledreceptorogr1oninflammationandfibrosispotentialimplicationsforidiopathicpulmonaryfibrosis
AT weiyaoku celltypespecificeffectsoftheovariancancergproteincoupledreceptorogr1oninflammationandfibrosispotentialimplicationsforidiopathicpulmonaryfibrosis
AT tylerjbell celltypespecificeffectsoftheovariancancergproteincoupledreceptorogr1oninflammationandfibrosispotentialimplicationsforidiopathicpulmonaryfibrosis
AT patriciajsime celltypespecificeffectsoftheovariancancergproteincoupledreceptorogr1oninflammationandfibrosispotentialimplicationsforidiopathicpulmonaryfibrosis
AT robertmatthewkottmann celltypespecificeffectsoftheovariancancergproteincoupledreceptorogr1oninflammationandfibrosispotentialimplicationsforidiopathicpulmonaryfibrosis