Activated Lymphocyte-Derived DNA Drives Glucose Metabolic Adaptation for Inducing Macrophage Inflammatory Response in Systemic Lupus Erythematosus

Cells. 2023 Aug 18;12(16):2093. doi: 10.3390/cells12162093.

Abstract

Activated lymphocyte-derived DNA (ALD-DNA) has been reported to drive the polarization of macrophages toward M2b, producing inflammatory cytokines and inducing inflammation, correspondingly playing an essential role in the development of systemic lupus erythematosus (SLE). Recently, accumulating evidence has pinpointed metabolic adaptation as the crucial cell-intrinsic determinant for inflammatory response, in which glucose metabolism is the key event. However, whether and how glucose metabolism was involved in ALD-DNA-induced macrophage inflammatory response and SLE development remains unclear. Herein, we performed glucose metabolomic analyses of ALD-DNA-stimulated macrophages and uncovered increased glycolysis and diminished pentose phosphate pathway (PPP), as well as enhanced glycogenesis. In ALD-DNA-stimulated macrophages, increased glycolysis resulted in higher lactate production, whereas diminished PPP efficiently led to lower levels of nicotinamide adenine dinucleotide phosphate (NADPH) with higher levels of reactive oxygen species (ROS). While blockade of lactate generation exerted no significant effect on macrophage inflammation in response to ALD-DNA, scavenging ROS fundamentally inhibited the inflammatory response of ALD-DNA-stimulated macrophages. Further, cyclic adenosine monophosphate (cAMP), a master for regulating glycogen metabolism, was downregulated by ALD-DNA in macrophages, which subsequently imbalanced glycogen metabolism toward glycogenesis but not glycogenolysis. Administration of cAMP effectively restored glycogenolysis and enhanced PPP, which correspondingly reduced ROS levels and inhibited the inflammatory response of ALD-DNA-stimulated macrophages. Finally, blocking glucose metabolism using 2-deoxy-D-glucose (2-DG) efficiently restricted macrophage inflammatory response and alleviated ALD-DNA-induced lupus disease. Together, our findings demonstrate that ALD-DNA drives the adaptation of glucose metabolism for inducing macrophage inflammatory response in SLE, which might further our understanding of disease pathogenesis and provide clues for interventive explorations.

Keywords: ALD-DNA; SLE; cAMP; glucose metabolism; macrophage inflammatory response.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Cyclic AMP*
  • DNA
  • Glucose
  • Glycogen
  • Humans
  • Inflammation
  • Lactic Acid
  • Lupus Erythematosus, Systemic*
  • Lymphocytes
  • Macrophages
  • Reactive Oxygen Species

Substances

  • Reactive Oxygen Species
  • Cyclic AMP
  • DNA
  • Glucose
  • Lactic Acid
  • Glycogen

Grants and funding

This research and the APC were funded by the National Natural Science Foundation of China [31970844, 32170927, 82071826, 82271841], Natural Science Foundation of Jiangsu Province [BK20211542, BK20201407], Jiangsu Specially Appointed Professor Program, Suzhou Municipal Science and Technology Bureau [ZXL2022460], Major Project of Natural Science Research in Jiangsu Higher Education Institutions [22KJA310005], Jiangsu Provincial Innovative Research Team, and Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD).