Your browser doesn't support javascript.
loading
Myeloid Slc2a1-Deficient Murine Model Revealed Macrophage Activation and Metabolic Phenotype Are Fueled by GLUT1.
Freemerman, Alex J; Zhao, Liyang; Pingili, Ajeeth K; Teng, Bin; Cozzo, Alyssa J; Fuller, Ashley M; Johnson, Amy R; Milner, J Justin; Lim, Maili F; Galanko, Joseph A; Beck, Melinda A; Bear, James E; Rotty, Jeremy D; Bezavada, Lavanya; Smallwood, Heather S; Puchowicz, Michelle A; Liu, Juan; Locasale, Jason W; Lee, Douglas P; Bennett, Brian J; Abel, E Dale; Rathmell, Jeff C; Makowski, Liza.
Afiliação
  • Freemerman AJ; Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799.
  • Zhao L; Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799.
  • Pingili AK; Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163.
  • Teng B; Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163.
  • Cozzo AJ; Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799.
  • Fuller AM; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.
  • Johnson AR; Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799.
  • Milner JJ; Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799.
  • Lim MF; Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799.
  • Galanko JA; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.
  • Beck MA; Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799.
  • Bear JE; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.
  • Rotty JD; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.
  • Bezavada L; Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163.
  • Smallwood HS; Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163.
  • Puchowicz MA; Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163.
  • Liu J; Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Duke University, Durham, NC 27710.
  • Locasale JW; Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Duke University, Durham, NC 27710.
  • Lee DP; Omic Insight, Inc., Durham, NC 27713.
  • Bennett BJ; U.S. Department of Agriculture Western Human Nutrition Research Center, Davis, CA 95616.
  • Abel ED; Division of Endocrinology and Metabolism, University of Iowa Carver College of Medicine, Iowa City, IA 52242.
  • Rathmell JC; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242; and.
  • Makowski L; Department of Pathology, Microbiology, and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt University, Nashville, TN 37232.
J Immunol ; 202(4): 1265-1286, 2019 02 15.
Article em En | MEDLINE | ID: mdl-30659108
ABSTRACT
Macrophages (MΦs) are heterogeneous and metabolically flexible, with metabolism strongly affecting immune activation. A classic response to proinflammatory activation is increased flux through glycolysis with a downregulation of oxidative metabolism, whereas alternative activation is primarily oxidative, which begs the question of whether targeting glucose metabolism is a viable approach to control MΦ activation. We created a murine model of myeloid-specific glucose transporter GLUT1 (Slc2a1) deletion. Bone marrow-derived MΦs (BMDM) from Slc2a1M-/- mice failed to uptake glucose and demonstrated reduced glycolysis and pentose phosphate pathway activity. Activated BMDMs displayed elevated metabolism of oleate and glutamine, yet maximal respiratory capacity was blunted in MΦ lacking GLUT1, demonstrating an incomplete metabolic reprogramming. Slc2a1M-/- BMDMs displayed a mixed inflammatory phenotype with reductions of the classically activated pro- and anti-inflammatory markers, yet less oxidative stress. Slc2a1M-/- BMDMs had reduced proinflammatory metabolites, whereas metabolites indicative of alternative activation-such as ornithine and polyamines-were greatly elevated in the absence of GLUT1. Adipose tissue MΦs of lean Slc2a1M-/- mice had increased alternative M2-like activation marker mannose receptor CD206, yet lack of GLUT1 was not a critical mediator in the development of obesity-associated metabolic dysregulation. However, Ldlr-/- mice lacking myeloid GLUT1 developed unstable atherosclerotic lesions. Defective phagocytic capacity in Slc2a1M-/- BMDMs may have contributed to unstable atheroma formation. Together, our findings suggest that although lack of GLUT1 blunted glycolysis and the pentose phosphate pathway, MΦ were metabolically flexible enough that inflammatory cytokine release was not dramatically regulated, yet phagocytic defects hindered MΦ function in chronic diseases.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Modelos Animais de Doenças / Transportador de Glucose Tipo 1 / Macrófagos Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Ano de publicação: 2019 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Modelos Animais de Doenças / Transportador de Glucose Tipo 1 / Macrófagos Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Ano de publicação: 2019 Tipo de documento: Article