期刊论文详细信息
Journal of Neuroinflammation
Acetate supplementation modulates brain adenosine metabolizing enzymes and adenosine A2A receptor levels in rats subjected to neuroinflammation
Thad A Rosenberger1  Jonathan D Geiger1  Dhaval P Bhatt1  Mark D Smith1 
[1] Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, 501 North Columbia Road, Grand Forks, North Dakota 58203, USA
关键词: neuroinflammation;    lipopolysaccharide;    glyceryl triacetate;    ecto-5’-nucleotidase;    brain;    adenosine A2A receptor;    adenosine kinase;    adenosine;    acetate;   
Others  :  803870
DOI  :  10.1186/1742-2094-11-99
 received in 2013-11-12, accepted in 2014-05-19,  发布年份 2014
PDF
【 摘 要 】

Background

Acetate supplementation reduces neuroglia activation and pro-inflammatory cytokine expression in rat models of neuroinflammation and Lyme neuroborreliosis. Because single-dose glyceryl triacetate (GTA) treatment increases brain phosphocreatine and reduces brain AMP levels, we postulate that GTA modulates adenosine metabolizing enzymes and receptors, which may be a possible mechanism to reduce neuroinflammation.

Methods

To test this hypothesis, we quantified the ability of GTA to alter brain levels of ecto-5’-nucleotidase (CD73), adenosine kinase (AK), and adenosine A2A receptor using western blot analysis and CD73 activity by measuring the rate of AMP hydrolysis. Neuroinflammation was induced by continuous bacterial lipopolysaccharide (LPS) infusion in the fourth ventricle of the brain for 14 and 28 days. Three treatment strategies were employed, one and two where rats received prophylactic GTA through oral gavage with LPS infusion for 14 or 28 days. In the third treatment regimen, an interventional strategy was used where rats were subjected to 28 days of neuroinflammation, and GTA treatment was started on day 14 following the start of the LPS infusion.

Results

We found that rats subjected to neuroinflammation for 28 days had a 28% reduction in CD73 levels and a 43% increase in AK levels that was reversed with prophylactic acetate supplementation. CD73 activity in these rats was increased by 46% with the 28-day GTA treatment compared to the water-treated rats. Rats subjected to neuroinflammation for 14 days showed a 50% increase in levels of the adenosine A2A receptor, which was prevented with prophylactic acetate supplementation. Interventional GTA therapy, beginning on day 14 following the induction of neuroinflammation, resulted in a 67% increase in CD73 levels and a 155% increase in adenosine A2A receptor levels.

Conclusion

These results support the hypothesis that acetate supplementation can modulate brain CD73, AK and adenosine A2A receptor levels, and possibly influence purinergic signaling.

【 授权许可】

   
2014 Smith et al.; licensee BioMed Central Ltd.

【 预 览 】
附件列表
Files Size Format View
20140708050722105.pdf 866KB PDF download
Figure 3. 75KB Image download
Figure 2. 90KB Image download
Figure 1. 88KB Image download
【 图 表 】

Figure 1.

Figure 2.

Figure 3.

【 参考文献 】
  • [1]Cunha RA: Neuroprotection by adenosine in the brain: From A (1) receptor activation to A (2A) receptor blockade. Purinergic Signal 2005, 1:111-134.
  • [2]Dunwiddie TV, Masino SA: The role and regulation of adenosine in the central nervous system. Annu Rev Neurosci 2001, 24:31-55.
  • [3]Shepel PN, Ramonet D, Stevens P, Geiger JD: Purine level regulation during energy depletion associated with graded excitatory stimulation in brain. Neurol Res 2005, 27:139-148.
  • [4]Newby AC: Adenosine and the concept of retaliatory metabolites. Trends Biochem Sci 1984, 9:42-44.
  • [5]Di Virgilio F, Ceruti S, Bramanti P, Abbracchio MP: Purinergic signalling in inflammation of the central nervous system. Trends Neurosci 2009, 32:79-87.
  • [6]Abbracchio MP, Burnstock G, Verkhratsky A, Zimmermann H: Purinergic signalling in the nervous system: an overview. Trends Neurosci 2009, 32:19-29.
  • [7]Masino SA, Kawamura M, Wasser CA, Pomeroy LT, Ruskin DN: Adenosine, ketogenic diet and epilepsy: the emerging therapeutic relationship between metabolism and brain activity. Curr Neuropharmacol 2009, 7:257-268.
  • [8]Boison D, Chen JF, Fredholm BB: Adenosine signaling and function in glial cells. Cell Death Differ 2010, 17:1071-1082.
  • [9]Chen JF, Pedata F: Modulation of ischemic brain injury and neuroinflammation by adenosine A2A receptors. Curr Pharm Des 2008, 14:1490-1499.
  • [10]Chen JF, Sonsalla PK, Pedata F, Melani A, Domenici MR, Popoli P, Geiger J, Lopes LV, de Mendonca A: Adenosine A2A receptors and brain injury: broad spectrum of neuroprotection, multifaceted actions and “fine tuning” modulation. Prog Neurobiol 2007, 83:310-331.
  • [11]Paterniti I, Melani A, Cipriani S, Corti F, Mello T, Mazzon E, Esposito E, Bramanti P, Cuzzocrea S, Pedata F: Selective adenosine A2A receptor agonists and antagonists protect against spinal cord injury through peripheral and central effects. J Neuroinflammation 2011, 8:31. BioMed Central Full Text
  • [12]Parkinson FE, Xiong W, Zamzow CR: Astrocytes and neurons: different roles in regulating adenosine levels. Neurol Res 2005, 27:153-160.
  • [13]Fredholm BB: Adenosine, an endogenous distress signal, modulates tissue damage and repair. Cell Death Differ 2007, 14:1315-1323.
  • [14]Thompson LF, Eltzschig HK, Ibla JC, Van De Wiele CJ, Resta R, Morote-Garcia JC, Colgan SP: Crucial role for ecto-5’-nucleotidase (CD73) in vascular leakage during hypoxia. J Exp Med 2004, 200:1395-1405.
  • [15]Boison D: Adenosine kinase, epilepsy and stroke: mechanisms and therapies. Trends Pharmacol Sci 2006, 27:652-658.
  • [16]Fredholm BB, Chen JF, Cunha RA, Svenningsson P, Vaugeois JM: Adenosine and brain function. Int Rev Neurobiol 2005, 63:191-270.
  • [17]Hauss-Wegrzyniak B, Dobrzanski P, Stoehr JD, Wenk GL: Chronic neuroinflammation in rats reproduces components of the neurobiology of Alzheimer’s disease. Brain Res 1998, 780:294-303.
  • [18]Fujihara M, Muroi M, Tanamoto K, Suzuki T, Azuma H, Ikeda H: Molecular mechanisms of macrophage activation and deactivation by lipopolysaccharide: roles of the receptor complex. Pharmacol Ther 2003, 100:171-194.
  • [19]Reisenauer CJ, Bhatt DP, Mitteness DJ, Slanczka ER, Gienger HM, Watt JA, Rosenberger TA: Acetate supplementation attenuates lipopolysaccharide-induced neuroinflammation. J Neurochem 2011, 117:264-274.
  • [20]Soliman ML, Smith MD, Houdek HM, Rosenberger TA: Acetate supplementation modulates brain histone acetylation and decreases interleukin-1beta expression in a rat model of neuroinflammation. J Neuroinflammation 2012, 9:51. BioMed Central Full Text
  • [21]Brissette CA, Houdek HM, Floden AM, Rosenberger TA: Acetate supplementation reduces microglia activation and brain interleukin-1beta levels in a rat model of Lyme neuroborreliosis. J Neuroinflammation 2012, 9:249. BioMed Central Full Text
  • [22]Arun P, Madhavarao CN, Moffett JR, Hamilton K, Grunberg NE, Ariyannur PS, Gahl WA, Anikster Y, Mog S, Hallows WC, Denu JM, Namboodiri AM: Metabolic acetate therapy improves phenotype in the tremor rat model of Canavan disease. J Inherit Metab Dis 2010, 33:195-210.
  • [23]Arun P, Ariyannur PS, Moffett JR, Xing G, Hamilton K, Grunberg NE, Ives JA, Namboodiri AM: Metabolic acetate therapy for the treatment of traumatic brain injury. J Neurotrauma 2010, 27:293-298.
  • [24]Tsen AR, Long PM, Driscoll HE, Davies MT, Teasdale BA, Penar PL, Pendlebury WW, Spees JL, Lawler SE, Viapiano MS, Jaworski DM: Triacetin-based acetate supplementation as a chemotherapeutic adjuvant therapy in glioma. Int J Cancer 2014, 134:1300-1310.
  • [25]Segel R, Anikster Y, Zevin S, Steinberg A, Gahl WA, Fisher D, Staretz-Chacham O, Zimran A, Altarescu G: A safety trial of high dose glyceryl triacetate for Canavan disease. Mol Genet Metab 2011, 103:203-206.
  • [26]Bhatt DP, Houdek HM, Watt JA, Rosenberger TA: Acetate supplementation increases brain phosphocreatine and reduces AMP levels with no effect on mitochondrial biogenesis. Neurochem Int 2013, 62:296-305.
  • [27]Soliman ML, Rosenberger TA: Acetate supplementation increases brain histone acetylation and inhibits histone deacetylase activity and expression. Mol Cell Biochem 2011, 352:173-180.
  • [28]Soliman ML, Combs CK, Rosenberger TA: Modulation of inflammatory cytokines and mitogen-activated protein kinases by acetate in primary astrocytes. J Neuroimmune Pharmacol 2013, 8:287-300.
  • [29]Soliman ML, Ohm JE, Rosenberger TA: Acetate reduces PGE2 release and modulates phospholipase and cyclooxygenase levels in neuroglia stimulated with lipopolysaccharide. Lipids 2013, 48:651-662.
  • [30]Soliman ML, Puig KL, Combs CK, Rosenberger TA: Acetate reduces microglia inflammatory signaling in vitro. J Neurochem 2012, 123:555-567.
  • [31]Lee H, Villacreses NE, Rapoport SI, Rosenberger TA: In vivo imaging detects a transient increase in brain arachidonic acid metabolism: a potential marker of neuroinflammation. J Neurochem 2004, 91:936-945.
  • [32]Rosenberger TA, Villacreses NE, Hovda JT, Bosetti F, Weerasinghe G, Wine RN, Harry GJ, Rapoport SI: Rat brain arachidonic acid metabolism is increased by a 6-day intracerebral ventricular infusion of bacterial lipopolysaccharide. J Neurochem 2004, 88:1168-1178.
  • [33]Bradford MM: A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 1976, 72:248-254.
  • [34]Bhatt DP, Chen X, Geiger JD, Rosenberger TA: A sensitive HPLC-based method to quantify adenine nucleotides in primary astrocyte cell cultures. J Chromatogr B Analyt Technol Biomed Life Sci 2012, 889–890:110-115.
  • [35]Brisevac D, Bjelobaba I, Bajic A, Clarner T, Stojiljkovic M, Beyer C, Andjus P, Kipp M, Nedeljkovic N: Regulation of ecto-5’-nucleotidase (CD73) in cultured cortical astrocytes by different inflammatory factors. Neurochem Int 2012, 61:681-688.
  • [36]Mathew R, Arun P, Madhavarao CN, Moffett JR, Namboodiri MA: Progress toward acetate supplementation therapy for Canavan disease: glyceryl triacetate administration increases acetate, but not N-acetylaspartate, levels in brain. J Pharmacol Exp Ther 2005, 315:297-303.
  • [37]Beal MF: Mitochondrial dysfunction and oxidative damage in Alzheimer’s and Parkinson’s diseases and coenzyme Q10 as a potential treatment. J Bioenerg Biomembr 2004, 36:381-386.
  • [38]Scafidi S, Racz J, Hazelton J, McKenna MC, Fiskum G: Neuroprotection by acetyl-L-carnitine after traumatic injury to the immature rat brain. Dev Neurosci 2010, 32:480-487.
  • [39]Bough KJ, Wetherington J, Hassel B, Pare JF, Gawryluk JW, Greene JG, Shaw R, Smith Y, Geiger JD, Dingledine RJ: Mitochondrial biogenesis in the anticonvulsant mechanism of the ketogenic diet. Ann Neurol 2006, 60:223-235.
  • [40]Ruskin DN, Masino SA: The nervous system and metabolic dysregulation: emerging evidence converges on ketogenic diet therapy. Front Neurosci 2012, 6:33.
  • [41]Masino SA, Kawamura M Jr, Ruskin DN, Gawryluk J, Chen X, Geiger JD: Purines and the Anti-Epileptic Actions of Ketogenic Diets. Open Neurosci J 2010, 4:58-63.
  • [42]Airas L, Niemela J, Yegutkin G, Jalkanen S: Mechanism of action of IFN-beta in the treatment of multiple sclerosis: a special reference to CD73 and adenosine. Ann N Y Acad Sci 2007, 1110:641-648.
  • [43]Niemela J, Ifergan I, Yegutkin GG, Jalkanen S, Prat A, Airas L: IFN-beta regulates CD73 and adenosine expression at the blood–brain barrier. Eur J Immunol 2008, 38:2718-2726.
  • [44]Bjelobaba I, Parabucki A, Lavrnja I, Stojkov D, Dacic S, Pekovic S, Rakic L, Stojiljkovic M, Nedeljkovic N: Dynamic changes in the expression pattern of ecto-5’-nucleotidase in the rat model of cortical stab injury. J Neurosci Res 2011, 89:862-873.
  • [45]Braun N, Lenz C, Gillardon F, Zimmermann M, Zimmermann H: Focal cerebral ischemia enhances glial expression of ecto-5’-nucleotidase. Brain Res 1997, 766:213-226.
  • [46]Nedeljkovic N, Bjelobaba I, Subasic S, Lavrnja I, Pekovic S, Stojkov D, Vjestica A, Rakic L, Stojiljkovic M: Up-regulation of ectonucleotidase activity after cortical stab injury in rats. Cell Biol Int 2006, 30:541-546.
  • [47]Cunha RA, Brendel P, Zimmermann H, Ribeiro JA: Immunologically distinct isoforms of ecto-5’-nucleotidase in nerve terminals of different areas of the rat hippocampus. J Neurochem 2000, 74:334-338.
  • [48]Arch JR, Newsholme EA: Activities and some properties of 5’-nucleotidase, adenosine kinase and adenosine deaminase in tissues from vertebrates and invertebrates in relation to the control of the concentration and the physiological role of adenosine. Biochem J 1978, 174:965-977.
  • [49]Aronica E, Zurolo E, Iyer A, de Groot M, Anink J, Carbonell C, van Vliet EA, Baayen JC, Boison D, Gorter JA: Upregulation of adenosine kinase in astrocytes in experimental and human temporal lobe epilepsy. Epilepsia 2011, 52:1645-1655.
  • [50]Fredholm BB, IJzerman AP, Jacobson KA, Klotz KN, Linden J: International union of pharmacology. XXV. Nomenclature and classification of adenosine receptors. Pharmacol Rev 2001, 53:527-552.
  • [51]Klotz KN: Adenosine receptors and their ligands. Naunyn Schmiedebergs Arch Pharmacol 2000, 362:382-391.
  • [52]Fredholm BB, Chen JF, Masino SA, Vaugeois JM: Actions of adenosine at its receptors in the CNS: insights from knockouts and drugs. Annu Rev Pharmacol Toxicol 2005, 45:385-412.
  • [53]Haselkorn ML, Shellington DK, Jackson EK, Vagni VA, Janesko-Feldman K, Dubey RK, Gillespie DG, Cheng D, Bell MJ, Jenkins LW, Homanics GE, Schnermann J, Kockanek PM: Adenosine A1 receptor activation as a brake on the microglial response after experimental traumatic brain injury in mice. J Neurotrauma 2010, 27:901-910.
  • [54]Rebola N, Simoes AP, Canas PM, Tome AR, Andrade GM, Barry CE, Agostinho PM, Lynch MA, Cunha RA: Adenosine A2A receptors control neuroinflammation and consequent hippocampal neuronal dysfunction. J Neurochem 2011, 117:100-111.
  • [55]Brothers HM, Marchalant Y, Wenk GL: Caffeine attenuates lipopolysaccharide-induced neuroinflammation. Neurosci Lett 2010, 480:97-100.
  • [56]Wei CJ, Li W, Chen JF: Normal and abnormal functions of adenosine receptors in the central nervous system revealed by genetic knockout studies. Biochim Biophys Acta 2011, 1808:1358-1379.
  • [57]Trincavelli ML, Costa B, Tuscano D, Lucacchini A, Martini C: Up-regulation of A (2A) adenosine receptors by proinflammatory cytokines in rat PC12 cells. Biochem Pharmacol 2002, 64:625-631.
  • [58]Simoes AP, Duarte JA, Agasse F, Canas PM, Tome AR, Agostinho P, Cunha RA: Blockade of adenosine A2A receptors prevents interleukin-1beta-induced exacerbation of neuronal toxicity through a p38 mitogen-activated protein kinase pathway. J Neuroinflammation 2012, 9:204. BioMed Central Full Text
  • [59]Dai SS, Zhou YG: Adenosine 2A receptor: a crucial neuromodulator with bidirectional effect in neuroinflammation and brain injury. Rev Neurosci 2011, 22:231-239.
  • [60]Li Y, Oskouian RJ, Day YJ, Rieger JM, Liu L, Kern JA, Linden J: Mouse spinal cord compression injury is reduced by either activation of the adenosine A2A receptor on bone marrow-derived cells or deletion of the A2A receptor on non-bone marrow-derived cells. Neuroscience 2006, 141:2029-2039.
  • [61]Adcock IM: HDAC inhibitors as anti-inflammatory agents. Br J Pharmacol 2007, 150:829-831.
  • [62]Strahl BD, Allis CD: The language of covalent histone modifications. Nature 2000, 403:41-45.
  文献评价指标  
  下载次数:9次 浏览次数:6次