Your browser doesn't support javascript.
loading
Oxidized Low-Density Lipoprotein Accumulation Suppresses Glycolysis and Attenuates the Macrophage Inflammatory Response by Diverting Transcription from the HIF-1α to the Nrf2 Pathway.
Ting, Kenneth K Y; Yu, Pei; Dow, Riley; Floro, Eric; Ibrahim, Hisham; Scipione, Corey A; Hyduk, Sharon J; Polenz, Chanele K; Zaslaver, Olga; Karmaus, Peer W F; Fessler, Michael B; Röst, Hannes L; Ohh, Michael; Tsai, Sue; Winer, Daniel A; Woo, Minna; Rocheleau, Jonathan; Jongstra-Bilen, Jenny; Cybulsky, Myron I.
  • Ting KKY; Department of Immunology, University of Toronto, Toronto, Ontario, Canada.
  • Yu P; Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.
  • Dow R; Department of Immunology, University of Toronto, Toronto, Ontario, Canada.
  • Floro E; Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.
  • Ibrahim H; Department of Immunology, University of Toronto, Toronto, Ontario, Canada.
  • Scipione CA; Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.
  • Hyduk SJ; Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.
  • Polenz CK; Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.
  • Zaslaver O; Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.
  • Karmaus PWF; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
  • Fessler MB; Department of Immunology, University of Toronto, Toronto, Ontario, Canada.
  • Röst HL; Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.
  • Ohh M; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
  • Tsai S; Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.
  • Winer DA; Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.
  • Woo M; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
  • Rocheleau J; Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
  • Jongstra-Bilen J; Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC.
  • Cybulsky MI; Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC.
J Immunol ; 211(10): 1561-1577, 2023 11 15.
Article en En | MEDLINE | ID: mdl-37756544
ABSTRACT
Lipid accumulation in macrophages (Mφs) is a hallmark of atherosclerosis, yet how lipid accumulation affects inflammatory responses through rewiring of Mφ metabolism is poorly understood. We modeled lipid accumulation in cultured wild-type mouse thioglycolate-elicited peritoneal Mφs and bone marrow-derived Mφs with conditional (Lyz2-Cre) or complete genetic deficiency of Vhl, Hif1a, Nos2, and Nfe2l2. Transfection studies employed RAW264.7 cells. Mφs were cultured for 24 h with oxidized low-density lipoprotein (oxLDL) or cholesterol and then were stimulated with LPS. Transcriptomics revealed that oxLDL accumulation in Mφs downregulated inflammatory, hypoxia, and cholesterol metabolism pathways, whereas the antioxidant pathway, fatty acid oxidation, and ABC family proteins were upregulated. Metabolomics and extracellular metabolic flux assays showed that oxLDL accumulation suppressed LPS-induced glycolysis. Intracellular lipid accumulation in Mφs impaired LPS-induced inflammation by reducing both hypoxia-inducible factor 1-α (HIF-1α) stability and transactivation capacity; thus, the phenotype was not rescued in Vhl-/- Mφs. Intracellular lipid accumulation in Mφs also enhanced LPS-induced NF erythroid 2-related factor 2 (Nrf2)-mediated antioxidative defense that destabilizes HIF-1α, and Nrf2-deficient Mφs resisted the inhibitory effects of lipid accumulation on glycolysis and inflammatory gene expression. Furthermore, oxLDL shifted NADPH consumption from HIF-1α- to Nrf2-regulated apoenzymes. Thus, we postulate that repurposing NADPH consumption from HIF-1α to Nrf2 transcriptional pathways is critical in modulating inflammatory responses in Mφs with accumulated intracellular lipid. The relevance of our in vitro models was established by comparative transcriptomic analyses, which revealed that Mφs cultured with oxLDL and stimulated with LPS shared similar inflammatory and metabolic profiles with foamy Mφs derived from the atherosclerotic mouse and human aorta.
Asunto(s)

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Aterosclerosis / Hipercolesterolemia 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: Aterosclerosis / Hipercolesterolemia Tipo de estudio: Prognostic_studies Límite: Animals / Humans Idioma: En Año: 2023 Tipo del documento: Article