AICAR ameliorates high-fat diet-associated pathophysiology in mouse and ex vivo models, independent of adiponectin

Emma Börgeson, Ville Wallenius, Gulam H Syed, Manjula Darshi, Juan Lantero Rodriguez, Christina Biörserud, Malin Ragnmark Ek, Per Björklund, Marianne Quiding-Järbrink, Lars Fändriks, Catherine Godson, Kumar Sharma, Emma Börgeson, Ville Wallenius, Gulam H Syed, Manjula Darshi, Juan Lantero Rodriguez, Christina Biörserud, Malin Ragnmark Ek, Per Björklund, Marianne Quiding-Järbrink, Lars Fändriks, Catherine Godson, Kumar Sharma

Abstract

Aims/hypothesis: In this study, we aimed to evaluate the therapeutic potential of 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), an activator of AMP-activated protein kinase, for ameliorating high-fat diet (HFD)-induced pathophysiology in mice. We also aimed to determine whether the beneficial effects of AICAR were dependent on adiponectin. Furthermore, human adipose tissue was used to examine the effect of AICAR ex vivo.

Methods: Six-week-old male C57BL/6J wild-type and Adipoq -/- mice were fed a standard-fat diet (10% fat) or an HFD (60% fat) for 12 weeks and given vehicle or AICAR (500 μg/g) three times/week from weeks 4-12. Diet-induced pathophysiology was examined in mice after 11 weeks by IPGTT and after 12 weeks by flow cytometry and western blotting. Human adipose tissue biopsies from obese (BMI 35-50 kg/m2) individuals were incubated with vehicle or AICAR (1 mmol/l) for 6 h at 37°C, after which inflammation was characterised by ELISA (TNF-α) and flow cytometry.

Results: AICAR attenuated adipose inflammation in mice fed an HFD, promoting an M1-to-M2 macrophage phenotype switch, while reducing infiltration of CD8+ T cells. AICAR treatment of mice fed an HFD partially restored glucose tolerance and attenuated hepatic steatosis and kidney disease, as evidenced by reduced albuminuria (p < 0.05), urinary H2O2 (p < 0.05) and renal superoxide levels (p < 0.01) in both wild-type and Adipoq -/- mice. AICAR-mediated protection occurred independently of adiponectin, as similar protection was observed in wild-type and Adipoq -/- mice. In addition, AICAR promoted an M1-to-M2 macrophage phenotype switch and reduced TNF-α production in tissue explants from obese human patients.

Conclusions/interpretation: AICAR may promote metabolic health and protect against obesity-induced systemic diseases in an adiponectin-independent manner. Furthermore, AICAR reduced inflammation in human adipose tissue explants, suggesting by proof-of-principle that the drug may reduce obesity-induced complications in humans.

Trial registration: ClinicalTrials.gov NCT02322073.

Keywords: AICAR; Adiponectin; Inflammation; Kidney disease; Liver disease; Macrophages; Obesity.

Conflict of interest statement

Data availability

Data supporting the results reported in the article can be found in the Sharma laboratory (kumarsharma@ucsd.edu).

Duality of interest

The authors declare that there is no duality of interest associated with this manuscript.

Contribution statement

EB, KS, and CG conceived and designed the study; EB, JLR, GHS, MD, VW, MRE, PB and CB contributed to data acquisition; EB, VW, LF and MQ-J analysed the data; all authors interpreted the data, drafted the article, revised it critically for important intellectual content and approved the final version to be published. KS is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

Figures

Fig. 1
Fig. 1
AICAR attenuates adipose inflammation independent of adiponectin in obese mice. Wild-type and Adipoq−/− mice fed a 12 week SFD (10% fat) or HFD (60% fat) received vehicle or AICAR (500 μg/g) during weeks 4–12. WAT leucocytes were characterised by flow cytometry; (a)–(d) Macrophages were characterised as pan-macrophages (F4/80+), proinflammatory M1-macrophages (CD11c+) or anti-inflammatory M2-macrophages (CD206+) (n = 5). (e)–(h) Lymphocytes were characterised as T killer (CD8+) vs T helper (CD4+) cells (n = 5). (i), (j) AMPK activation and adiponectin levels were analysed by western blot. AMPK blots were cut as indicated (full blots and details of cutting are presented in ESM Fig. 4) (n = 3). Data are presented as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001; ANOVA with Bonferroni correction; †p < 0.05, ††p < 0.01, †††p < 0.001, Adipoq−/− vs wild-type mice for respective conditions. N/A, not applicable
Fig. 2
Fig. 2
AICAR promotes AMPK activation and an M1-to-M2 phenotype switch in cultured macrophages. J774 macrophages were incubated with vehicle or AICAR (1 mmol/l) for 16 h (n = 3). Cells were characterised as (a) proinflammatory M1 (CD11c++) or (b) anti-inflammatory M2 (CD206+) by flow cytometry and (ce) p-AMPK/AMPK was assessed by western blot. Data are presented as mean ± SEM. *p < 0.05, **p < 0.01, paired Student’s t test
Fig. 3
Fig. 3
AICAR partially restores glucose tolerance independent of adiponectin in obese mice. Wild-type and Adipoq−/− mice fed a 12 week SFD (10% fat) or HFD (60% fat) received vehicle or AICAR (500 μg/g) during weeks 4–12 of feeding. Glucose tolerance was tested at week 11 by an IPGTT in (a) wild-type (n = 4 for all groups) and (b) Adipoq−/− (n = 3 for SFD and HFD, n = 7 for HFD + AICAR) mice. Circles, SFD; squares, HFD; triangles, HFD + AICAR. (c) Graph of the AUC of IPGTT curves. Data are presented as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ANOVA with Bonferroni correction; †p < 0.05, ††p < 0.01, Adipoq−/− vs wild-type mice for respective conditions
Fig. 4
Fig. 4
AICAR attenuates hepatic steatosis independent of adiponectin in obese mice. Wild-type and Adipoq−/− mice fed a 12 week SFD (10% fat) or HFD (60% fat) received vehicle or AICAR (500 μg/g) during weeks 4–12. (a) Representative images of hepatic haematoxylin and eosin staining (magnification × 40). Hepatic (b) triacylglycerol and (c) cholesterol (n = 3). Data are presented as mean ± SEM. *p < 0.05, **p < 0.01, ANOVA with Bonferroni correction
Fig. 5
Fig. 5
AICAR attenuates kidney disease independent of adiponectin in obese mice. Wild-type and Adipoq−/− mice fed a 12 week SFD (10% fat) or HFD (60% fat) received vehicle or AICAR (500 μg/g) during weeks 4–12. (a) Micro-albuminuria, (b) urine H2O2 and (c) renal hypertrophy were assessed (n = 7). (d) DHE was injected 16 h prior to killing the mice and renal DHE oxidation was quantified as a measurement of superoxide production (magnification × 100; n = 4). (e) Renal pan-macrophages (F4/80+) and (f) proinflammatory M1 macrophages (CD11c+) were characterised and quantified by flow cytometry (n = 5). (g–h) Flow cytometry histogram showing number of CD11c+ positive cells in (g) wild-type and (h) Adipoq−/− mice, incubated with vehicle (dotted line), fed an HFD without AICAR (solid line), or fed an HFD with AICAR (dashed line). Data are presented as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ANOVA with Bonferroni correction; †p < 0.05, ††p < 0.01, †††p < 0.001, Adipoq−/− vs wild-type mice for respective conditions
Fig. 6
Fig. 6
AICAR reduces inflammation in adipose explants from obese individuals. Omental WAT explants from obese individuals (BMI 35–50 kg/m2; n = 4 individuals) were incubated with vehicle or AICAR (1 mmol/l) for 6 h. (a) Tissue macrophages were characterised as M1 (CD11c+), M1/M2b (CD86+), M2a (CD206+) or M2a/M2c (CD163+) and (b) quantified. (c) T cells were characterised as CD4+ and CD8+ and (d) quantified. (e) Levels of TNF-α in the supernatant fraction were determined by ELISA. Data are presented as mean ± SEM. *p < 0.05, ***p < 0.001, Student’s t tests
Fig. 7
Fig. 7
Schematic illustration of proposed AICAR-mediated effects in obesity. (a) In obese mice, AICAR treatment attenuates HFD-induced adipose inflammation, promoting an M1-to-M2 macrophage phenotype switch by reducing CD8+ T cell infiltration, while increasing p-AMPK levels. This results in reduced liver and kidney disease and enhanced glucose tolerance. All of these effects are independent of adiponectin. (b) AICAR mediates a similar M1-to-M2 macrophage phenotype switch in adipose explants isolated from obese individuals undergoing bariatric surgery. MΦ, macrophage

References

    1. Borgeson E, Sharma K. Obesity, immunomodulation and chronic kidney disease. Curr Opin Pharmacol. 2013;13:618–624. doi: 10.1016/j.coph.2013.05.011.
    1. Mathew AV, Okada S, Sharma K. Obesity related kidney disease. Curr Diabetes Rev. 2011;7:41–49. doi: 10.2174/157339911794273928.
    1. NCD Risk Factor Collaboration Trends in adult body-mass index in 200 countries from 1975 to 2014: a pooled analysis of 1698 population-based measurement studies with 19.2 million participants. Lancet. 2016;387:1377–1396. doi: 10.1016/S0140-6736(16)30054-X.
    1. Viollet B, Guigas B, Leclerc J, et al. AMP-activated protein kinase in the regulation of hepatic energy metabolism: from physiology to therapeutic perspectives. Acta Physiol. 2009;196:81–98. doi: 10.1111/j.1748-1716.2009.01970.x.
    1. Misra P, Chakrabarti R. The role of AMP kinase in diabetes. Indian J Med Res. 2007;125:389–398.
    1. Aschenbach WG, Hirshman MF, Fujii N, Sakamoto K, Howlett KF, Goodyear LJ. Effect of AICAR treatment on glycogen metabolism in skeletal muscle. Diabetes. 2002;51:567–573. doi: 10.2337/diabetes.51.3.567.
    1. Gaidhu MP, Frontini A, Hung S, Pistor K, Cinti S, Ceddia RB. Chronic AMP-kinase activation with AICAR reduces adiposity by remodeling adipocyte metabolism and increasing leptin sensitivity. J Lipid Res. 2011;52:1702–1711. doi: 10.1194/jlr.M015354.
    1. Narkar VA, Downes M, Yu RT, et al. AMPK and PPARδ agonists are exercise mimetics. Cell. 2008;134:405–415. doi: 10.1016/j.cell.2008.06.051.
    1. Henriksen BS, Curtis ME, Fillmore N, Cardon BR, Thomson DM, Hancock CR. The effects of chronic AMPK activation on hepatic triglyceride accumulation and glycerol 3-phosphate acyltransferase activity with high fat feeding. Diabetol Metab Syndr. 2013;5:29. doi: 10.1186/1758-5996-5-29.
    1. Liu S, Jing F, Yu C, Gao L, Qin Y, Zhao J. AICAR-induced activation of AMPK inhibits TSH/SREBP-2/HMGCR pathway in liver. PLoS One. 2015;10:e0124951. doi: 10.1371/journal.pone.0124951.
    1. Lv Q, Zhen Q, Liu L, et al. AMP-kinase pathway is involved in tumor necrosis factor alpha-induced lipid accumulation in human hepatoma cells. Life Sci. 2015;131:23–29. doi: 10.1016/j.lfs.2015.03.003.
    1. Decleves AE, Zolkipli Z, Satriano J, et al. Regulation of lipid accumulation by AMP-activated kinase in high fat diet-induced kidney injury. Kidney Int. 2014;85:611–623. doi: 10.1038/ki.2013.462.
    1. Decleves AE, Mathew AV, Cunard R, Sharma K. AMPK mediates the initiation of kidney disease induced by a high-fat diet. J Am Soc Nephrol. 2011;22:1846–1855. doi: 10.1681/ASN.2011010026.
    1. Dugan LL, You YH, Ali SS, et al. AMPK dysregulation promotes diabetes-related reduction of superoxide and mitochondrial function. J Clin Invest. 2013;123:4888–4899. doi: 10.1172/JCI66218.
    1. Lee MJ, Feliers D, Mariappan MM, et al. A role for AMP-activated protein kinase in diabetes-induced renal hypertrophy. Am J Physiol Ren Physiol. 2007;292:F617–F627. doi: 10.1152/ajprenal.00278.2006.
    1. Kadowaki T, Yamauchi T. Adiponectin and adiponectin receptors. Endocr Rev. 2005;26:439–451. doi: 10.1210/er.2005-0005.
    1. Ortega Moreno L, Lamacchia O, Salvemini L, et al. The paradoxical association of adiponectin with mortality rate in patients with type 2 diabetes: evidence of synergism with kidney function. Atherosclerosis. 2016;245:222–227. doi: 10.1016/j.atherosclerosis.2015.12.026.
    1. Sweiss N, Sharma K. Adiponectin effects on the kidney. Best Pract Res Clin Endocrinol Metab. 2014;28:71–79. doi: 10.1016/j.beem.2013.08.002.
    1. Saraheimo M, Forsblom C, Fagerudd J, et al. Serum adiponectin is increased in type 1 diabetic patients with nephropathy. Diabetes Care. 2005;28:1410–1414. doi: 10.2337/diacare.28.6.1410.
    1. Saraheimo M, Forsblom C, Thorn L, et al. Serum adiponectin and progression of diabetic nephropathy in patients with type 1 diabetes. Diabetes Care. 2008;31:1165–1169. doi: 10.2337/dc07-2306.
    1. Moller KF, Dieterman C, Herich L, Klaassen IA, Kemper MJ, Muller-Wiefel DE. High serum adiponectin concentration in children with chronic kidney disease. Pediatr Nephrol. 2012;27:243–249. doi: 10.1007/s00467-011-1971-1.
    1. Kim HY, Bae EH, Ma SK, et al. Association of serum adiponectin level with albuminuria in chronic kidney disease patients. Clin Exp Nephrol. 2016;20:443–449. doi: 10.1007/s10157-015-1173-4.
    1. Nanayakkara PW, Le Poole CY, Fouque D, et al. Plasma adiponectin concentration has an inverse and a non linear association with estimated glomerular filtration rate in patients with K/DOQI 3–5 chronic kidney disease. Clin Nephrol. 2009;72:21–30. doi: 10.5414/CNP72021.
    1. Menon V, Li L, Wang X, et al. Adiponectin and mortality in patients with chronic kidney disease. J Am Soc Nephrol. 2006;17:2599–2606. doi: 10.1681/ASN.2006040331.
    1. Börgeson E. The role of lipoxins in cardiometabolic physiology and disease. Cardiovasc Endocrinol. 2016;5:4–13. doi: 10.1097/XCE.0000000000000068.
    1. McNelis JC, Olefsky JM. Macrophages, immunity, and metabolic disease. Immunity. 2014;41:36–48. doi: 10.1016/j.immuni.2014.05.010.
    1. Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annu Rev Physiol. 2010;72:219–246. doi: 10.1146/annurev-physiol-021909-135846.
    1. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW., Jr Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112:1796–1808. doi: 10.1172/JCI200319246.
    1. Borgeson E, Johnson AM, Lee YS, et al. Lipoxin A attenuates obesity-induced adipose inflammation and associated liver and kidney disease. Cell Metab. 2015;22:125–137. doi: 10.1016/j.cmet.2015.05.003.
    1. Andrikopoulos S, Blair AR, Deluca N, Fam BC, Proietto J. Evaluating the glucose tolerance test in mice. Am J Physiol Endocrinol Metab. 2008;295:E1323–E1332. doi: 10.1152/ajpendo.90617.2008.
    1. Borgeson E, McGillicuddy FC, Harford KA, et al. Lipoxin A4 attenuates adipose inflammation. FASEB J. 2012;26:4287–4294. doi: 10.1096/fj.12-208249.
    1. Teteris SA, Hochheiser K, Kurts C. Isolation of functional dendritic cells from murine kidneys for immunological characterization. Nephrology. 2012;17:364–371. doi: 10.1111/j.1440-1797.2012.01581.x.
    1. Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization. Front Biosci. 2008;13:453–461. doi: 10.2741/2692.
    1. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007;117:175–184. doi: 10.1172/JCI29881.
    1. Gonzalez-Periz A, Claria J. Resolution of adipose tissue inflammation. Sci World J. 2010;10:832–856. doi: 10.1100/tsw.2010.77.
    1. Zhu YP, Brown JR, Sag D, Zhang L, Suttles J. Adenosine 5'-monophosphate-activated protein kinase regulates IL-10-mediated anti-inflammatory signaling pathways in macrophages. J Immunol. 2015;194:584–594. doi: 10.4049/jimmunol.1401024.
    1. Koscso B, Csoka B, Kokai E, et al. Adenosine augments IL-10-induced STAT3 signaling in M2c macrophages. J Leukoc Biol. 2013;94:1309–1315. doi: 10.1189/jlb.0113043.
    1. Sag D, Carling D, Stout RD, Suttles J. Adenosine 5'-monophosphate-activated protein kinase promotes macrophage polarization to an anti-inflammatory functional phenotype. J Immunol. 2008;181:8633–8641. doi: 10.4049/jimmunol.181.12.8633.
    1. Nishimura S, Manabe I, Nagasaki M, et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med. 2009;15:914–920. doi: 10.1038/nm.1964.
    1. Spite M, Claria J, Serhan CN. Resolvins, specialized proresolving lipid mediators, and their potential roles in metabolic diseases. Cell Metab. 2014;19:21–36. doi: 10.1016/j.cmet.2013.10.006.
    1. Fabbrini E, Magkos F. Hepatic steatosis as a marker of metabolic dysfunction. Nutrients. 2015;7:4995–5019. doi: 10.3390/nu7064995.
    1. Ohashi K, Iwatani H, Kihara S, et al. Exacerbation of albuminuria and renal fibrosis in subtotal renal ablation model of adiponectin-knockout mice. Arterioscler Thromb Vasc Biol. 2007;27:1910–1917. doi: 10.1161/ATVBAHA.107.147645.
    1. Rutkowski JM, Wang ZV, Park AS, et al. Adiponectin promotes functional recovery after podocyte ablation. J Am Soc Nephrol. 2013;24:268–282. doi: 10.1681/ASN.2012040414.
    1. Sharma K, Ramachandrarao S, Qiu G, et al. Adiponectin regulates albuminuria and podocyte function in mice. J Clin Invest. 2008;118:1645–1656.

Source: PubMed

3
Abonnieren