Your browser doesn't support javascript.
loading
Targeting Pulmonary Fibrosis by SLC1A5-Dependent Glutamine Transport Blockade.
Choudhury, Malay; Schaefbauer, Kyle J; Kottom, Theodore J; Yi, Eunhee S; Tschumperlin, Daniel J; Limper, Andrew H.
Afiliación
  • Choudhury M; Thoracic Disease Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Biochemistry and Molecular Biology, and.
  • Schaefbauer KJ; Thoracic Disease Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Biochemistry and Molecular Biology, and.
  • Kottom TJ; Thoracic Disease Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Biochemistry and Molecular Biology, and.
  • Yi ES; Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota.
  • Tschumperlin DJ; Department of Physiology and Biomedical Engineering, College of Medicine and Science, and.
  • Limper AH; Thoracic Disease Research Unit, Division of Pulmonary and Critical Care Medicine, Department of Biochemistry and Molecular Biology, and.
Am J Respir Cell Mol Biol ; 69(4): 441-455, 2023 10.
Article en En | MEDLINE | ID: mdl-37459644
ABSTRACT
The neutral amino acid glutamine plays a central role in TGF-ß (transforming growth factor-ß)-induced myofibroblast activation and differentiation. Cells take up glutamine mainly through a transporter expressed on the cell surface known as solute carrier SLC1A5 (solute carrier transporter 1A5). In the present work, we demonstrated that profibrotic actions of TGF-ß are mediated, at least in part, through a metabolic maladaptation of SLC1A5 and that targeting SLC1A5 abrogates multiple facets of fibroblast activation. This approach could thus represent a novel therapeutic strategy to treat patients with fibroproliferative diseases. We found that SLC1A5 was highly expressed in fibrotic lung fibroblasts and fibroblasts isolated from idiopathic pulmonary fibrosis lungs. The expression of profibrotic targets, cell migration, and anchorage-independent growth by TGF-ß required the activity of SLC1A5. Loss or inhibition of SLC1A5 function enhanced fibroblast susceptibility to autophagy; suppressed mTOR, HIF (hypoxia-inducible factor), and Myc signaling; and impaired mitochondrial function, ATP production, and glycolysis. Pharmacological inhibition of SLC1A5 by the small-molecule inhibitor V-9302 shifted fibroblast transcriptional profiles from profibrotic to fibrosis resolving and attenuated fibrosis in a bleomycin-treated mouse model of lung fibrosis. This is the first study, to our knowledge, to demonstrate the utility of a pharmacological inhibitor of glutamine transport in fibrosis, providing a framework for new paradigm-shifting therapies targeting cellular metabolism for this devastating disease.
Asunto(s)
Palabras clave

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Fibrosis Pulmonar Idiopática / Glutamina / Pulmón Tipo de estudio: Prognostic_studies Límite: Animals / Humans Idioma: En Año: 2023 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Fibrosis Pulmonar Idiopática / Glutamina / Pulmón Tipo de estudio: Prognostic_studies Límite: Animals / Humans Idioma: En Año: 2023 Tipo del documento: Article