β-Hydroxybutyrate suppresses inflammasome formation by ameliorating endoplasmic reticulum stress via AMPK activation

Ha Ram Bae, Dae Hyun Kim, Min Hi Park, Bonggi Lee, Min Jo Kim, Eun Kyeong Lee, Ki Wung Chung, Seong Min Kim, Dong Soon Im, Hae Young Chung, Ha Ram Bae, Dae Hyun Kim, Min Hi Park, Bonggi Lee, Min Jo Kim, Eun Kyeong Lee, Ki Wung Chung, Seong Min Kim, Dong Soon Im, Hae Young Chung

Abstract

β-Hydroxybutyrate, a ketone body that is used as an energy source in organs such as the brain, muscle, and heart when blood glucose is low, is produced by fatty acid oxidation in the liver under the fasting state. Endoplasmic reticulum (ER) stress is linked with the generation of intracellular reactive oxygen species and the accumulation of misfolded protein in the ER. ER stress is known to induce the NOD-like receptor protein 3 inflammasome, which mediates activation of the proinflammatory cytokine interleukin-1β, whose maturation is caspase-1-dependent. We investigated whether β-hydroxybutyrate modulates ER stress, inflammasome formation, and insulin signaling. Sprague Dawley rats (6 and 24 months of age) that were starved for 3 d and rats treated with β-hydroxybutyrate (200 mg·kg-1·d-1 i.p., for 5 d) were used for in vivo investigations, whereas human hepatoma HepG2 cells were used for in vitro studies. Overexpression of AMPK in cultured cells was performed to elucidate the molecular mechanism. The starvation resulted in increased serum β-hydroxybutyrate levels with decreased ER stress (PERK, IRE1, and ATF6α) and inflammasome (ASC, caspase-1, and NLRP3) formation compared with non-fasted 24-month-old rats. In addition, β-hydroxybutyrate suppressed the increase of ER stress- and inflammasome-related marker proteins. Furthermore, β-hydroxybutyrate treatment increased the expression of manganese superoxide dismutase and catalase via the AMP-activated protein kinase-forkhead box protein O3α transcription factor pathway both in vivo and in vitro. The significance of the current study was the discovery of the potential therapeutic role of β-hydroxybutyrate in suppressing ER-stress-induced inflammasome formation.

Keywords: AMPK; Gerotarget; aging; endoplasmic reticulum; inflammasome; β-hydroxybutyrate.

Conflict of interest statement

CONFLICTS OF INTEREST

The authors declare no conflict of interest.

Figures

Figure 1. Modulation of ER-stress-induced inflammasome formation…
Figure 1. Modulation of ER-stress-induced inflammasome formation in acute starvation rat liver
A. IL-1β and IL-6 levels were determined in the serum. Results of one-factor ANOVA: *p < 0.05 vs. Ad libitum. B. Western blot analysis was performed to determine the inflammasome levels in the cytoplasmic extracts. As shown, the protein levels of the inflammasome were decreased by acute starvation. C. Western blot analysis was performed to determine the protein levels of UPR markers. As shown, the levels were significantly decreased in the starvation model. One representative blot of each protein is shown from three experiments that yielded similar results. D. Western blot analysis was performed to detect the presence of insulin signaling in the cytoplasmic extracts. The protein levels of serine phosphorylation of IRS-1 and phosphorylation of Akt were decreased by starvation. In contrast, tyrosine phosphorylation of IRS-1 was increased. E. IRS-2 interaction of Akt and its modulation by starvation. cytosol extracts were prepared from young and old rat livers. Immunoprecipitated IRS-2 was determined to be physically associated with p-seine, p-tyrosine, p-IRS-1/2 (t632), p-Akt, and Akt by Western blotting. F. Constituents were determined in the serum. Results of one-factor ANOVA: *p < 0.05 vs. Ad libitum.
Figure 2. Modulation of β-hydroxybutyrate on ER-stress-induced…
Figure 2. Modulation of β-hydroxybutyrate on ER-stress-induced inflammasome formation and the AMPK pathway
A. Western blot analysis was performed to detect the presence of caspase-1, ASC, NLRP3, β-actin, IL-18, and IL-1β in liver homogenates from β-hydroxybutyrate-treated rats (200 mg·kg−1·day−1 for 5 d). B. To determine ER stress marker changes by β-hydroxybutyrate treatment, the expressions of p-PERK, p-IRE1, and ATF6α were examined. C. Western blot analysis was performed to detect the presence of phosphorylated IRS-1 at Ser 307 and Tyr 632, phosphorylated Akt, and phosphorylated JNK in liver homogenates. D. Western blot analysis was performed to detect the presence of p-AMPK, catalase, and SOD-2 (MnSOD) in liver homogenates.
Figure 3. Effect of β-hydroxybutyrate on AMPK…
Figure 3. Effect of β-hydroxybutyrate on AMPK activation and ER-stress-induced inflammasome formation in HepG2 cells
A. HepG2 cells were treated with β-hydroxybutyrate (1 and 10 mM). Cells were lysed after 2 h of treatment, and western blot analysis was performed to determine the change of phosphorylation of AMPK in the cytosolic fraction. B. HepG2 cells were treated with 1 mM β-hydroxybutyrate and incubated. After 2 h, the cells were treated with 250 μM palmitate. Levels of p-PERK, p-IRE1, ATF6α, NLRP3, ASC, p-AMPK, and AMPK were then measured by western blot analysis using their specific antibodies. β-Actin and TFIIB blot analyses are shown, to clarify the same amount of proteins loaded in the cytosolic and nuclear fractions, respectively.
Figure 4. Effect of β-hydroxybutyrate on ER-stress-induced…
Figure 4. Effect of β-hydroxybutyrate on ER-stress-induced inflammasome formation via the AMPK pathway in vitro
A. HepG2 cells were transfected with lentiviral particle wild-type AMPK. After 12 h, cells were treated with 1 mM β-hydroxybutyrate, followed by 250 μM palmitate for 1 h. Western blot analysis was performed to determine the levels of p-PERK, p-IRE1, p-JNK, and ATF6α. β-Actin and histone H1 blots are shown, to clarify the same amount of protein loaded in the cytosolic and nuclear fractions, respectively. B., C. HepG2 cells were transfected with lentiviral particle wild-type AMPK. After 12 h, cells were treated with 1 mM β-hydroxybutyrate, followed by 250 μM palmitate for 1 h. D. Reactive oxygen species generation was measured by dichlorofluorescein formation with the fluorescent probe 2′,7′-dichlorofluorescein diacetate. Results of one-factor ANOVA: ***p < 0.001 vs. control; ###p < 0.001 vs. palmitate. HB, β-hydroxybutyrate.

References

    1. Veech RL, Chance B, Kashiwaya Y, Lardy HA, Cahill GF., Jr Ketone bodies, potential therapeutic uses. IUBMB Life. 2001;51:241–247.
    1. Cahill GF, Jr, Herrera MG, Morgan AP, Soeldner JS, Steinke J, Levy PL, Reichard GA, Jr, Kipnis DM. Hormone-fuel interrelationships during fasting. J Clin Invest. 1996;45:1751–1769.
    1. Laffel L. Ketone bodies: a review of physiology, pathophysiology and application of monitoring to diabetes. Diabetes Metab Res Rev. 1999;15:412–426.
    1. Hegardt FG. Mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase : a control enzyme in ketogenesis. Biochem J. 1999;338:569–582.
    1. Owen OE, Felig P, Morgan AP, Wahren J, Cahill GF., Jr Liver and kidney metabolism during prolonged starvation. J Clin Invest. 1969;48:574–583.
    1. Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, Le Moan N, Grueter CA, Lim H, Saunders LR, Stevens RD, Newgard CB, Farese RV, Jr, de Cabo R, Ulrich S, Akassoglou K, Verdin E. Suppression of oxidative stress by β-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science. 2013;339:211–214.
    1. Maalouf M, Rho JM, Mattson MP. The neuroprotective properties of calorie restriction, the ketogenic diet, and ketone bodies. Brain Res Rev. 2009;59:293–315.
    1. Cahill GF., Jr Fuel metabolism in starvation. Annu Rev Nutr. 2006;26:1–22.
    1. Nirinjini N. ER and aging-Protein folding and the ER stress response. Ageing Res Rev. 2009;8:150–159.
    1. Finkel T, Holbrook NJ. Oxidants, oxidative stress and the biology of ageing. Nature. 2000;408:239–247.
    1. Salminen A, Kaarniranta K. Regulation of the aging process by autophagy. Trends Mol Med. 2009;15:217–224.
    1. Menu P, Mayor A, Zhou R, Tardivel A, Ichijo H, Mori K, Tschopp J. ER stress activates the NLRP3 inflammasome via an UPR-independent pathway. Cell Death and Dis. 2012;26:e261.
    1. Greer EL, Dowlatshahi D, Banko MR, Villen J, Hoang K, Blanchard D, Gygi SP, Brunet A. An AMPK-FOXO pathway mediates longevity induced by a novel method of dietary restriction in C. elegans. Curr Biol. 2007;17:1646–1656.
    1. Greer EL, Oskoui PR, Banko MR, Maniar JM, Gygi MP, Gygi SP, Brunet A. The energy sensor AMP-activated protein kinase directly regulates the mammalian FOXO3 transcription factor. J Biol Chem. 2007;282:30107–30119.
    1. Kimura I, Inoue D, Maeda T, Hara T, Ichimura A, Miyauchi S, Kobayashi M, Hirasawa A, Tsujimoto G. Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41) Proc Natl Acad Sci USA. 2011;108:8030–8035.
    1. Ozcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi NN, Ozdelen E, Tuncman G, Görgün C, Glimcher LH, Hotamisligil GS. Endoplasmic Reticulum Stress Links Obesity, Insulin Action, and Type 2 Diabetes. Science. 2004;306:457–461.
    1. Kim DS, Jeong SK, Kim HR, Kim DS, Chae SW, Chae HJ. Metformin regulates palmitate-induced apoptosis and ER stress response in HepG2 liver cells. Immunopharmacol Immunotoxicol. 2010;32:251–257.
    1. Edwards C, Canfield J, Copes N, Rehan M, Lipps D, Bradshaw PC. D-beta-hydroxybutyrate extends lifespan in C.elegans. Aging (Albany NY) 2014;6:621–644. doi: 10.18632/aging.100683.
    1. Karaskov E, Scott C, Zhang L, Teodoro T, Ravazzola M, Volchuk A. Chronic palmitate but not oleate exposure induces endoplasmic reticulum stress, which may contribute to INS-1 pancreatic beta-cell apoptosis. Endocrinology. 2006;147:3398–3407.
    1. Hardie DG. Roles of the AMP-activated/SNF1 protein kinase family in the response to cellular stress. Biochem Soc Symp. 1999;64:13–27.
    1. Dong Y, Zhang M, Wang S, Liang B, Zhao Z, Liu C, Wu M, Choi HC, Lyons TJ, Zou MH. Activation of AMP-Activated Protein Kinase Inhibits Oxidized LDL-Triggered Endoplasmic Reticulum Stress in Vivo. Diabetes. 2010;59:1386–1396.
    1. Choi AM, Nakahira K. Dampening insulin signaling by an NLRP3 ‘meta-flammasome'. Nat Immunol. 2011;12:379–380.
    1. Salminen A, Kaarniranta K, Kauppinen A. Inflammaging: disturbed interplay between autophagy and inflammasomes. Aging (Albany NY) 2012;4:166–175. doi: 10.18632/aging.100444.
    1. Heitmann RN, Dawes DJ, Sensenig SC. Hepatic ketogenesis and peripheral ketone body utilization in the ruminant. J Nutr. 1987;177:1174–1780.
    1. Kitabchi AE, Wall BM. Diabetic ketoacidosis. Med Clin North Am. 1995;79:33–37.
    1. Balietti M, Casoli T, Di Stefano G, Giorgetti B, Aicardi G, Fattoretti P. Ketogenic diets: an historical antiepileptic therapy with promising potentialities for the aging brain. Ageing Res Rev. 2010;9:273–279.
    1. Youm YH, Nguyen KY, Grant RW, Goldberg EL, Bodogai M, Kim D, D'Agostino D, Planavsky N, Lupfer C, Kanneganti TD, Kang S, Horvath TL, Fahmy TM, Crawford PA, Biragyn A, Alnemri E, Dixit VD. The ketone metabolite β-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med. 2015;21:263–269.
    1. Haynes CM, Titus EA, Cooper AA. Degradation of misfolded proteins prevents ER-derived oxidative stress and cell death. Mol Cell. 2004;15:767–776.
    1. Menu P, Mayor A, Zhou R, Tardivel A, Ichijo H, Mori K, Tschopp J. ER stress activates the NLRP3 inflammasome via an UPR-independent pathway. Cell Death and Dis. 2012;26:e261.
    1. Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome Activation. Nature. 2011;469:221–225.
    1. Malozowski S, Sahlroot JT. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med. 2007;357:302–303.
    1. Henao-Mejia J, Elinav E, Jin C, Hao L, Mehal WZ, Strowig T, Thaiss CA, Kau AL, Eisenbarth SC, Jurczak MJ, Camporez JP, Shulman GI, Gordon JI, Hoffman HM, Flavell RA. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature. 2012;482:179–185.
    1. Flamment M, Hajduch E, Ferré P, Foufelle F. New insights into ER stress induced insulin resistance. Trends Endocrinol Metab. 2012;23:381–390.
    1. Gu Y, Sun XX, Ye JM, He L, Yan SS, Zhang HH, Hu LH, Yuan JY, Yu Q. Arctigenin alleviates ER stress via activating AMPK. Acta Pharmacol Sin. 2012;7:941–952.
    1. Kops GJ, Dansen TB, Polderman PE, Saarloos I, Wirtz KW, Coffer PJ, Huang TT, Bos JL, Medema RH, Burgering BM. Forkhead transcription factor From acute ER stress to physiological roles of the Unfolded Protein Response FOXO3a protects quiescent cells from oxidative stress. Nature. 2002;419:316–321.
    1. Sengupta A, Molkentin JD, Paik JH, DePinho RA, Yutzey KE. FoxO transcription factors promote cardiomyocyte survival upon induction of oxidative stress. J Biol Chem. 2011;286:7468–7478.
    1. Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140:821–832.

Source: PubMed

3
Suscribir