Your browser doesn't support javascript.
loading
Dysfunctional lactate metabolism in human alveolar type II cells from idiopathic pulmonary fibrosis lung explant tissue.
Newton, Danforth A; Lottes, Robyn G; Ryan, Rita M; Spyropoulos, Demetri D; Baatz, John E.
Affiliation
  • Newton DA; Department of Pediatrics, Medical University of South Carolina, Charleston, SC, 29425, USA.
  • Lottes RG; Department of Pediatrics, Medical University of South Carolina, Charleston, SC, 29425, USA.
  • Ryan RM; Department of Pediatrics, Case Western Reserve University, UH Rainbow Babies and Children's Hospital, Cleveland, OH, 44106, USA.
  • Spyropoulos DD; Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA.
  • Baatz JE; Department of Pediatrics, Medical University of South Carolina, Charleston, SC, 29425, USA. baatzje@musc.edu.
Respir Res ; 22(1): 278, 2021 Oct 28.
Article in En | MEDLINE | ID: mdl-34711218
ABSTRACT

BACKGROUND:

Idiopathic Pulmonary Fibrosis (IPF) is the most common and progressive form of the interstitial lung diseases, leading most patients to require lung transplants to survive. Despite the relatively well-defined role of the fibroblast in the progression of IPF, it is the alveolar type II epithelial cell (AEC2) that is now considered the initiation site of damage, driver of disease, and the most efficacious therapeutic target for long-term resolution. Based on our previous studies, we hypothesize that altered lactate metabolism in AEC2 plays a pivotal role in IPF development and progression, affecting key cellular and molecular interactions within the pulmonary microenvironment.

METHODS:

AEC2s isolated from human patient specimens of non-fibrotic and IPF lungs were used for metabolic measurements, lactate dehydrogenase (LDH) analyses and siRNA-mediated knockdown experiments.

RESULTS:

AEC2s isolated from human IPF lung explant tissues had lower rates of oxidative metabolism and were more glycolytic lactate-producing cells than were AEC2 from control, non-fibrotic lung explant tissues. Consistent with this shift in metabolism, patient-derived IPF AEC2s exhibited LDH tetramers that have higher ratios of LDHALDHB (i.e., favoring pyruvate to lactate conversion) than control AEC2s. Experimental manipulation of LDHA subunit expression in IPF AEC2s restored the bioenergetic profile characteristic of AEC2 from non-fibrotic lungs.

CONCLUSIONS:

These results are consistent with the concept that altered lactate metabolism may be an underlying feature of AEC2 dysfunction in IPF and may be a novel and important target for therapeutic treatment.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Lactic Acid / Energy Metabolism / Idiopathic Pulmonary Fibrosis / Alveolar Epithelial Cells Type of study: Observational_studies Limits: Humans Language: En Journal: Respir Res Year: 2021 Document type: Article Affiliation country:

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Lactic Acid / Energy Metabolism / Idiopathic Pulmonary Fibrosis / Alveolar Epithelial Cells Type of study: Observational_studies Limits: Humans Language: En Journal: Respir Res Year: 2021 Document type: Article Affiliation country: