Metabolic regulation of the circadian clock in classically and alternatively activated macrophages

. 2023 May ; 101 (5) : 428-443. [epub] 20230403

Jazyk angličtina Země Spojené státy americké Médium print-electronic

Typ dokumentu časopisecké články, práce podpořená grantem

Perzistentní odkaz   https://www.medvik.cz/link/pmid36918728

Macrophages exhibit a range of functional pro- and anti-inflammatory states that induce changes in their cellular metabolism. We aimed to elucidate whether these changes affect the molecular properties of their circadian clock focusing on their anti-inflammatory phenotype. Primary cell cultures of bone marrow-derived macrophages (BMDMs; nonpolarized M0 BMDM) from PER2::LUC (fusion protein of PERIOD2 and LUCIFERASE) mice were polarized into the M1 (proinflammatory) or M2 (anti-inflammatory) phenotype, and PER2-driven bioluminescence was recorded in real-time at the cell-population and single-cell levels. Viability, clock gene expression profiles, polarization plasticity and peroxisome proliferator-activated receptor γ (PPARγ) protein levels were analyzed. The effects of pharmacological activation/inhibition of PPARγ (rosiglitazone/GW9662) and inhibition of fatty acid oxidation (FAO) by etomoxir in M2 BMDM cell cultures were examined. The parameters of PER2-driven bioluminescence rhythms differed between M0, M1 and M2 BMDM cultures at cell-population and single-cell levels. Compared with M0, polarization to M2 did not change the period but increased amplitude, mean bioluminescence level and rhythm persistence. Polarization to M1 shortened the period but had no effect on the amplitude of the rhythm. The same period changes were observed after a bidirectional switch between M1- and M2-polarized states in the same culture. Both PPARγ activation/inhibition and FAO inhibition modulated the clock in M2 BMDMs, suggesting metabolic regulation of the M2 clock. Our results indicate that bidirectional changes in the properties of BMDM circadian clocks in response to their actual polarization are mediated via changes in their metabolic state. They provide new information on the interrelationship between the BMDM polarization, their circadian clock and cellular metabolism.

Zobrazit více v PubMed

Scheiermann C, Kunisaki Y, Frenette PS. Circadian control of the immune system. Nat Rev Immunol 2013; 13: 190-198.

Born J, Lange T, Hansen K, Mölle M, Fehm HL. Effects of sleep and circadian rhythm on human circulating immune cells. J Immunol 1997; 158: 4454-4464.

Scheiermann C, Kunisaki Y, Lucas D, et al. Adrenergic nerves govern circadian leukocyte recruitment to tissues. Immunity 2012; 37: 290-301.

Nguyen KD, Fentress SJ, Qiu Y, Yun K, Cox JS, Chawla A. Circadian gene Bmal1 regulates diurnal oscillations of Ly6Chi inflammatory monocytes. Science (80-) 2013; 341: 1483-1488.

Halberg F, Johnson EA, Brown BW, Bittner JJ. Susceptibility rhythm to E. coli endotoxin and bioassay. Proc Soc Exp Biol Med 1960; 103: 142-144.

Keller M, Mazuch J, Abraham U, et al. A circadian clock in macrophages controls inflammatory immune responses. Proc Natl Acad Sci USA 2009; 106: 21407-21412.

Partch CL, Green CB, Takahashi JS. Molecular architecture of the mammalian circadian clock. Trends Cell Biol 2014; 24: 90-99.

Gachon F, Nagoshi E, Brown SA, Ripperger J, Schibler U. The mammalian circadian timing system: from gene expression to physiology. Chromosoma 2004; 113: 103-112.

Scheiermann C, Gibbs J, Ince L, Loudon A. Clocking in to immunity. Nat Rev Immunol 2018; 18: 423-437.

Honzlová P, Semenovykh K, Sumová A. The circadian clock of polarized microglia and its interaction with mouse brain oscillators. Cell Mol Neurobiol 2022; 43: 1319-1333.

Silver AC, Arjona A, Hughes ME, Nitabach MN, Fikrig E. Circadian expression of clock genes in mouse macrophages, dendritic cells, and B cells. Brain Behav Immun 2012; 26: 407-413.

Hayashi M, Shimba S, Tezuka M. Characterization of the molecular clock in mouse peritoneal macrophages. Biol Pharm Bull 2007; 30: 621-626.

Fonken LK, Frank MG, Kitt MM, Barrientos RM, Watkins LR, Maier SF. Microglia inflammatory responses are controlled by an intrinsic circadian clock. Brain Behav Immun 2015; 45: 171-179.

Gibbs JE, Blaikley J, Beesley S, et al. The nuclear receptor REV-ERBα mediates circadian regulation of innate immunity through selective regulation of inflammatory cytokines. Proc Natl Acad Sci USA 2012; 109: 582-587.

Petrovsky N, Mcnair P, Harrison LC. Diurnal rhythms of pro-inflammatory cytokines: regulation by plasma cortisol and therapeutic implications. Cytokine 1998; 10: 307-312.

Viola A, Munari F, Sánchez-Rodríguez R, Scolaro T, Castegna A. The metabolic signature of macrophage responses. Front Immunol 2019; 10: 1462.

Canton M, Sánchez-Rodríguez R, Spera I, et al. Reactive oxygen species in macrophages: sources and targets. Front Immunol 2021; 12: 734229.

Wang F, Zhang S, Vuckovic I, et al. Glycolytic stimulation is not a requirement for M2 macrophage differentiation. Cell Metab 2018; 28: 463-475.e4.

Van den Bossche J, Baardman J, de Winther MPJ. Metabolic characterization of polarized M1 and M2 bone marrow-derived macrophages using real-time extracellular flux analysis. J Vis Exp 2015; 105: 53424.

Gautier EL, Chow A, Spanbroek R, et al. Systemic analysis of PPARγ in mouse macrophage populations reveals marked diversity in expression with critical roles in resolution of inflammation and airway immunity. J Immunol 2012; 189: 2614-2624.

Li H, Ruan XZ, Powis SH, et al. EPA and DHA reduce LPS-induced inflammation responses in HK-2 cells: evidence for a PPAR-γ-dependent mechanism. Kidney Int 2005; 67: 867-874.

Chang HY, Lee HN, Kim W, Surh YJ. Docosahexaenoic acid induces M2 macrophage polarization through peroxisome proliferator-activated receptor γ activation. Life Sci 2015; 120: 39-47.

Fontaine C, Dubois G, Duguay Y, et al. The orphan nuclear receptor rev-Erbα is a peroxisome proliferator-activated receptor (PPAR) γ target gene and Promotes PPARγ-induced adipocyte differentiation. J Biol Chem 2003; 278: 37672-37680.

Wang N, Yang G, Jia Z, et al. Vascular PPARγ controls circadian variation in blood pressure and heart rate through Bmal1. Cell Metab 2008; 8: 482-491.

Chen L, Yang G. PPARs integrate the mammalian clock and energy metabolism. PPAR Res 2014; 2014: e653017.

Timmons GA, O'Siorain JR, Kennedy OD, Curtis AM, Early JO. Innate rhythms: clocks at the Center of Monocyte and Macrophage Function. Front Immunol 2020; 11: 1743.

Carter SJ, Durrington HJ, Gibbs JE, et al. A matter of time: study of circadian clocks and their role in inflammation. J Leukoc Biol 2016; 99: 549-560.

Najafi M, Hashemi Goradel N, Farhood B, et al. Macrophage polarity in cancer: a review. J Cell Biochem 2019; 120: 2756-2765.

Mantovani A, Biswas SK, Galdiero MR, Sica A, Locati M. Macrophage plasticity and polarization in tissue repair and remodelling. J Pathol 2013; 229: 176-185.

Chen S, Fuller KK, Dunlap JC, Loros JJ. A pro- and anti-inflammatory Axis modulates the macrophage circadian clock. Front Immunol 2020; 11: 867.

Lellupitiyage Don SS, Mas-Rosario JA, Lin H-H, Nguyen EM, Taylor SR, Farkas ME. Macrophage circadian rhythms are differentially affected based on stimuli. Integr Biol 2022; 14: 62-75.

Casella G, Garzetti L, Gatta AT, et al. IL4 induces IL6-producing M2 macrophages associated to inhibition of neuroinflammation in vitro and in vivo. J Neuroinflammation 2016; 13: 139.

Pajarinen J, Tamaki Y, Antonios JK, et al. Modulation of mouse macrophage polarization in vitro using IL-4 delivery by osmotic pumps. J Biomed Mater Res Part A 2015; 103: 1339-1345.

Varga T, Czimmerer Z, Nagy L. PPARs are a unique set of fatty acid regulated transcription factors controlling both lipid metabolism and inflammation. Biochim Biophys Acta - Mol Basis Dis 2011; 1812: 1007-1022.

Nomura M, Liu J, Rovira II, et al. Fatty acid oxidation in macrophage polarization. Nat Immunol 2016; 17: 216-217.

Divakaruni AS, Hsieh WY, Minarrieta L, et al. Etomoxir inhibits macrophage polarization by disrupting CoA homeostasis. Cell Metab 2018; 28: 490-503.e7.

Raulien N, Friedrich K, Strobel S, et al. Fatty acid oxidation compensates for lipopolysaccharide-induced Warburg effect in glucose-deprived monocytes. Front Immunol 2017; 8: 609.

Wang Y, Pati P, Xu Y, et al. Endotoxin disrupts circadian rhythms in macrophages via reactive oxygen species. PLoS One 2016; 11: e0155075.

Zhuang JC, Wogan GN. Growth and viability of macrophages continuously stimulated to produce nitric oxide. Proc Natl Acad Sci USA 1997; 94: 11875-11880.

Albina JE, Cui S, Mateo RB, Reichner JS. Nitric oxide-mediated apoptosis in murine peritoneal macrophages. J Immunol 1993; 150: 5080-5085.

Biswas SK, Chittezhath M, Shalova IN, Lim J-Y. Macrophage polarization and plasticity in health and disease. Immunol Res 2012; 53: 11-24.

Pena OM, Pistolic J, Raj D, Fjell CD, Hancock REW. Endotoxin tolerance represents a distinctive state of alternative polarization (M2) in human mononuclear cells. J Immunol 2011; 186: 7243-7254.

Everett LJ, Lazar MA. Nuclear receptor rev-erbα: up, down, and all around. Trends Endocrinol Metab 2014; 25: 586-592.

Grygiel-Górniak B. Peroxisome proliferator-activated receptors and their ligands: nutritional and clinical implications - a review. Nutr J 2014; 13: 17.

Yao C-H, Liu G-Y, Wang R, Moon SH, Gross RW, Patti GJ. Identifying off-target effects of etomoxir reveals that carnitine palmitoyltransferase I is essential for cancer cell proliferation independent of β-oxidation. PLoS Biol 2018; 16: e2003782.

Nakahata Y, Kaluzova M, Grimaldi B, et al. The NAD+-dependent deacetylase SIRT1 modulates CLOCK-mediated chromatin remodeling and circadian control. Cell 2008; 134: 329-340.

Yoo S-H, Yamazaki S, Lowrey PL, et al. PERIOD2::LUCIFERASE real-time reporting of circadian dynamics reveals persistent circadian oscillations in mouse peripheral tissues. Proc Natl Acad Sci USA 2004; 101: 5339-5346.

Tomida M, Yamamoto-Yamaguchi Y, Hozumi M. Purification of a factor inducing differentiation of mouse myeloid leukemic M1 cells from conditioned medium of mouse fibroblast L929 cells. J Biol Chem 1984; 259: 10978-10982.

Li Y, Shan Y, Desai RV, Cox KH, Weinberger LS, Takahashi JS. Noise-driven cellular heterogeneity in circadian periodicity. Proc Natl Acad Sci U S A 2020; 117: 10350-10356.

Ruf T. The Lomb-Scargle periodogram in biological rhythm research: analysis of incomplete and unequally spaced time-series. Biol Rhythm Res 1999; 30: 178-201.

Evans MS, Chaurette JP, Adams ST, et al. A synthetic luciferin improves bioluminescence imaging in live mice. Nat Methods 2014; 11: 393-395.

Tinevez J-Y, Perry N, Schindelin J, et al. TrackMate: an open and extensible platform for single-particle tracking. Methods 2017; 115: 80-90.

Schindelin J, Arganda-Carreras I, Frise E, et al. Fiji: an open-source platform for biological-image analysis. Nat Methods 2012; 9: 676-682.

Rampersad SN. Multiple applications of Alamar blue as an indicator of metabolic function and cellular health in cell viability bioassays. Sensors 2012; 12: 12347-12360.

Pfaffl MW. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res 2001; 29: e45.

Holm S. A simple sequentially Rejective multiple test procedure. Scand J Stat 1979; 6: 65-70.

Sládek M, Polidarová L, Nováková M, Parkanová D, Sumová A. Early Chronotype and tissue-specific alterations of circadian clock function in spontaneously hypertensive rats. PLoS One 2012; 7: e46951.

Honzlová P, Novosadová Z, Houdek P, Sládek M, Sumová A. Misaligned feeding schedule elicits divergent circadian reorganizations in endo- and exocrine pancreas clocks. Cell Mol Life Sci 2022; 79: 318.

Najít záznam

Citační ukazatele

Nahrávání dat...

Možnosti archivace

Nahrávání dat...