PFKFB2-mediated glycolysis promotes lactate-driven continual efferocytosis by macrophages

 Back to publications

2023

Nat Metab. 2023 Feb 16. doi: 10.1038/s42255-023-00736-8. Online ahead of print.

PFKFB2-mediated glycolysis promotes lactate-driven continual efferocytosis by macrophages

Maaike Schilperoort, David Ngai, Marina Katerelos, David A Power, Ira Tabas

Kidney Laboratory, Department of Nephrology, Austin Health, Heidelberg, Victoria, Australia. Department of Medicine, The University of Melbourne, Heidelberg, Victoria, Australia. The Institute for Breathing and Sleep (IBAS), Austin Health, Heidelberg, Victoria, Australia. Columbia University Irving Medical Center, New York, NY, USA.

Service type: Knock-in mice

Abstract

Resolving-type macrophages prevent chronic inflammation by clearing apoptotic cells through efferocytosis. These macrophages are thought to rely mainly on oxidative phosphorylation, but emerging evidence suggests a possible link between efferocytosis and glycolysis. To gain further insight into this issue, we investigated molecular-cellular mechanisms involved in efferocytosis-induced macrophage glycolysis and its consequences. We found that efferocytosis promotes a transient increase in macrophage glycolysis that is dependent on rapid activation of the enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 2 (PFKFB2), which distinguishes this process from glycolysis in pro-inflammatory macrophages. Mice transplanted with activation-defective PFKFB2 bone marrow and then subjected to dexamethasone-induced thymocyte apoptosis exhibit impaired thymic efferocytosis, increased thymic necrosis, and lower expression of the efferocytosis receptors MerTK and LRP1 on thymic macrophages compared with wild-type control mice. In vitro mechanistic studies revealed that glycolysis stimulated by the uptake of a first apoptotic cell promotes continual efferocytosis through lactate-mediated upregulation of MerTK and LRP1. Thus, efferocytosis-induced macrophage glycolysis represents a unique metabolic process that sustains continual efferocytosis in a lactate-dependent manner. The differentiation of this process from inflammatory macrophage glycolysis raises the possibility that it could be therapeutically enhanced to promote efferocytosis and resolution in chronic inflammatory diseases.

View Publication