J Korean Neurosurg Soc.  2020 Nov;63(6):689-697. 10.3340/jkns.2020.0056.

Effect of Pioglitazone on Perihematomal Edema in Intracerebral Hemorrhage Mouse Model by Regulating NLRP3 Expression and Energy Metabolism

Affiliations
  • 1Department of Neurosurgery, Bucheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
  • 2Department of Neurosurgery, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
  • 3Biomedical Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
  • 4Department of Neurosurgery, Cell Death Disease Research Center, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea

Abstract


Objective
: Cerebral edema is the predominant mechanism of secondary inflammation after intracerebral hemorrhage (ICH). Pioglitazone, peroxisome proliferator-activated receptor gamma agonist has been shown to play a role in regulation of central nervous system inflammation. Here, we examined the pharmacological effects of pioglitazone in an ICH mouse model and investigated its regulation on NLRP3 inflammasome and glucose metabolism.
Methods
: The ICH model was established in C57 BL/6 mice by the stereotactical inoculation of blood (30 µL) into the right frontal lobe. The treatment group was administered i.p. pioglitazone (20 mg/kg) for 1, 3, and 6 days. The control group was administered i.p. phosphate-buffered saline for 1, 3, and 6 days. We investigated brain water contents, NLRP3 expression, and changes in the metabolites in the ICH model using liquid chromatography-tandem mass spectrometry.
Results
: On day 3, brain edema in the mice treated with pioglitazone was decreased more than that in the control group. Expression levels of NLRP3 in the ICH model treated with pioglitazone were decreased more than those of the control mice on days 3 and 7. The pioglitazone group showed higher levels of glycolytic metabolites than those in the ICH mice. Lactate production was increased in the ICH mice treated with pioglitazone.
Conclusion
: Our results demonstrated less brain swelling following ICH in mice treated with pioglitazone. Pioglitazone decreased NLRP3-related brain edema and increased anaerobic glycolysis, resulting in the production of lactate in the ICH mice model. NLRP3 might be a therapeutic target for ICH recovery.

Keyword

Cerebral hemorrhage; Brain edema; Inflammasomes; Pioglitazone; Lactates

Figure

  • Fig. 1. Establishment of intracranial hemorrhage (IcH) models (hematoxylin and eosin, H&E). coronal section of the whole brain (A, scale bar=3 mm) and magnification of the hematoma (B, ×200).

  • Fig. 2. Treatment schedule.

  • Fig. 3. comparison of brain water contents. The water contents of both hemispheres in IcH mice treated with pioglitazone were lower than those in IcH mice on days 3 and 7. In the treatment group, the water content of the ipsilateral hemispheres on day 7 was decreased more than on day 3 (n = 7, each group). *p<0.05. IcH : intracranial hemorrhage.

  • Fig. 4. Western blot of NLRP3 in the control mice, IcH mice, and IcH mice treated with pioglitazone (A). The expression level of NLRP3 in the IcH mice treated with pioglitazone was decreased more than those in the IcH mice on day 7 (B). *p<0.05. IcH : intracranial hemorrhage.

  • Fig. 5. Liquid chromatography-tandem mass spectrometry analysis. In the glycolysis pathway, the production of glucose-6-phosphate, fructose-1,6- biphosphate, pyruvate, and lactate were increased in IcH mice brains treated with pioglitazone compared to those IcH mice (A). In the TcA cycle, the production of nicotinamide adenine dinucleotide was decreased in IcH mice treated with pioglitazone more than that of IcH mice. Differences in other metabolites were not noted (B). In the PPP, there were no significant differences in PPP metabolites between the two groups (c). *p<0.05. IcH : intracranial hemorrhage, TcA : tricarboxylic acid, PPP : pentose phosphate pathway


Reference

References

1. Alano CC, Garnier P, Ying W, Higashi Y, Kauppinen TM, Swanson RA. NAD+ depletion is necessary and sufficient for poly(ADP-ribose) polymerase-1-mediated neuronal death. J Neurosci. 30:2967–2978. 2010.
Article
2. Alessandri B, Schwandt E, Kamada Y, Nagata M, Heimann A, Kempski O. The neuroprotective effect of lactate is not due to improved glutamate uptake after controlled cortical impact in rats. J Neurotrauma. 29:2181–2191. 2012.
Article
3. Aronowski J, Zhao X. Molecular pathophysiology of cerebral hemorrhage: secondary brain injury. Stroke. 42:1781–1786. 2011.
4. Au A. Metabolomics and lipidomics of ischemic stroke. Adv Clin Chem. 85:31–69. 2018.
Article
5. Becatti M, Taddei N, Cecchi C, Nassi N, Nassi PA, Fiorillo C. SIRT1 modulates MAPK pathways in ischemic-reperfused cardiomyocytes. Cell Mol Life Sci. 69:2245–2260. 2012.
Article
6. Berthet C, Castillo X, Magistretti PJ, Hirt L. New evidence of neuroprotection by lactate after transient focal cerebral ischaemia: extended benefit after intracerebroventricular injection and efficacy of intravenous administration. Cerebrovasc Dis. 34:329–335. 2012.
Article
7. Castro MA, Beltrán FA, Brauchi S, Concha II. A metabolic switch in brain: glucose and lactate metabolism modulation by ascorbic acid. J Neurochem. 110:423–440. 2009.
Article
8. Chinopoulos C. Which way does the citric acid cycle turn during hypoxia? The critical role of α-ketoglutarate dehydrogenase complex. J Neurosci Res. 91:1030–1043. 2013.
Article
9. Esen F, Erdem T, Aktan D, Kalayci R, Cakar N, Kaya M, et al. Effects of magnesium administration on brain edema and blood-brain barrier breakdown after experimental traumatic brain injury in rats. J Neurosurg Anesthesiol. 15:119–125. 2003.
Article
10. Fann DY, Lee SY, Manzanero S, Chunduri P, Sobey CG, Arumugam TV. Pathogenesis of acute stroke and the role of inflammasomes. Ageing Res Rev. 12:941–966. 2013.
Article
11. Gallagher CN, Carpenter KL, Grice P, Howe DJ, Mason A, Timofeev I, et al. The human brain utilizes lactate via the tricarboxylic acid cycle: a 13C-labelled microdialysis and high-resolution nuclear magnetic resonance study. Brain. 132(Pt 10):2839–2849. 2009.
Article
12. Helmy MM, Helmy MW, El-Mas MM. Additive renoprotection by pioglitazone and fenofibrate against inflammatory, oxidative and apoptotic manifestations of cisplatin nephrotoxicity: modulation by PPARs. PLoS One. 10:e0142303. 2015.
Article
13. Huang Q, Sun M, Li M, Zhang D, Han F, Wu JC, et al. Combination of NAD+ and NADPH offers greater neuroprotection in ischemic stroke models by relieving metabolic stress. Mol Neurobiol. 55:6063–6075. 2018.
Article
14. Hsu CP, Oka S, Shao D, Hariharan N, Sadoshima J. Nicotinamide phosphoribosyltransferase regulates cell survival through NAD+ synthesis in cardiac myocytes. Circ Res. 105:481–491. 2009.
Article
15. Irrera N, Pizzino G, Calò M, Pallio G, Mannino F, Famà F, et al. Lack of the Nlrp3 inflammasome improves mice recovery following traumatic brain injury. Front Pharmacol. 8:459. 2017.
Article
16. Izawa Y, Takahashi S, Suzuki N. Pioglitazone enhances pyruvate and lactate oxidation in cultured neurons but not in cultured astroglia. Brain Res. 1305:64–73. 2009.
Article
17. Katsuki H. Exploring neuroprotective drug therapies for intracerebral hemorrhage. J Pharmacol Sci. 114:366–378. 2010.
Article
18. Keep RF, Hua Y, Xi G. Intracerebral haemorrhage: mechanisms of injury and therapeutic targets. Lancet Neurol. 11:720–731. 2012.
Article
19. Kim SJ, Kim SH, Kim JH, Hwang S, Yoo HJ. Understanding metabolomics in biomedical research. Endocrinol Metab (Seoul). 31:7–16. 2016.
Article
20. Lebeaupin C, Proics E, de Bieville CH, Rousseau D, Bonnafous S, Patouraux S, et al. ER stress induces NLRP3 inflammasome activation and hepatocyte death. Cell Death Dis. 6:e1879. 2015.
Article
21. Li Y, Yang J, Chen MH, Wang Q, Qin MJ, Zhang T, et al. Ilexgenin A inhibits endoplasmic reticulum stress and ameliorates endothelial dysfunction via suppression of TXNIP/NLRP3 inflammasome activation in an AMPK dependent manner. Pharmacol Res. 99:101–115. 2015.
Article
22. Lu A, Tang Y, Ran R, Ardizzone TL, Wagner KR, Sharp FR. Brain genomics of intracerebral hemorrhage. J Cereb Blood Flow Metab. 26:230–252. 2006.
Article
23. Mayer SA, Rincon F. Treatment of intracerebral haemorrhage. Lancet Neurol. 4:662–672. 2005.
Article
24. Ribo M, Grotta JC. Latest advances in intracerebral hemorrhage. Curr Neurol Neurosci Rep. 6:17–22. 2006.
Article
25. Rice AC, Zsoldos R, Chen T, Wilson MS, Alessandri B, Hamm RJ, et al. Lactate administration attenuates cognitive deficits following traumatic brain injury. Brain Res. 928:156–159. 2002.
Article
26. Ricote M, Li AC, Willson TM, Kelly CJ, Glass CK. The peroxisome proliferator-activated receptor-γ is a negative regulator of macrophage activation. Nature. 391:79–82. 1998.
Article
27. Rolland WB 2nd, Manaenko A, Lekic T, Hasegawa Y, Ostrowski R, Tang J, et al. FTY720 is neuroprotective and improves functional outcomes after intracerebral hemorrhage in mice. Acta Neurochir Suppl. 111:213–217. 2011.
Article
28. Rynkowski MA, Kim GH, Komotar RJ, Otten ML, Ducruet AF, Zacharia BE, et al. A mouse model of intracerebral hemorrhage using autologous blood infusion. Nat Protoc. 3:122–128. 2008.
Article
29. Sahuquillo J, Merino MA, Sánchez-Guerrero A, Arikan F, Vidal-Jorge M, Martínez-Valverde T, et al. Lactate and the lactate-to-pyruvate molar ratio cannot be used as independent biomarkers for monitoring brain energetic metabolism: a microdialysis study in patients with traumatic brain injuries. PLoS One. 9:e102540. 2014.
Article
30. Sánchez-Abarca LI, Tabernero A, Medina JM. Oligodendrocytes use lactate as a source of energy and as a precursor of lipids. Glia. 36:321–329. 2001.
Article
31. Schroder K, Zhou R, Tschopp J. The NLRP3 inflammasome: a sensor for metabolic danger? Science. 327:296–300. 2010.
Article
32. Song N, Liu ZS, Xue W, Bai ZF, Wang QY, Dai J, et al. NLRP3 phosphorylation is an essential priming event for inflammasome activation. Mol Cell. 68:185–197.e6. 2017.
Article
33. Strbian D, Kovanen PT, Karjalainen-Lindsberg ML, Tatlisumak T, Lindsberg PJ. An emerging role of mast cells in cerebral ischemia and hemorrhage. Ann Med. 41:438–450. 2009.
Article
34. Stutz A, Kolbe CC, Stahl R, Horvath GL, Franklin BS, van Ray O, et al. NLRP3 inflammasome assembly is regulated by phosphorylation of the pyrin domain. J Exp Med. 214:1725–1736. 2017.
Article
35. Timofeev I, Carpenter KL, Nortje J, Al-Rawi PG, O’Connell MT, Czosnyka M, et al. Cerebral extracellular chemistry and outcome following traumatic brain injury: a microdialysis study of 223 patients. Brain. 134(Pt 2):484–494. 2011.
Article
36. Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 324:1029–1033. 2009.
Article
37. Wang J. Preclinical and clinical research on inflammation after intracerebral hemorrhage. Prog Neurobiol. 92:463–477. 2010.
Article
38. Wang J, Doré S. Inflammation after intracerebral hemorrhage. J Cereb Blood Flow Metab. 27:894–908. 2007.
Article
39. Wang J, Song MY, Lee JY, Kwon KS, Park BH. The NLRP3 inflammasome is dispensable for ER stress-induced pancreatic β-cell damage in Akita mice. Biochem Biophys Res Commun. 466:300–305. 2015.
Article
40. Wang Y, Yu B, Wang L, Yang M, Xia Z, Wei W, et al. Pioglitazone ameliorates glomerular NLRP3 inflammasome activation in apolipoprotein E knockout mice with diabetes mellitus. PLoS One. 12:e0181248. 2017.
Article
41. Yao ST, Cao F, Chen JL, Chen W, Fan RM, Li G, et al. NLRP3 is required for complement-mediated caspase-1 and IL-1beta activation in ICH. J Mol Neurosci. 61:385–395. 2017.
Article
42. Ying W, Wei G, Wang D, Wang Q, Tang X, Shi J, et al. Intranasal administration with NAD+ profoundly decreases brain injury in a rat model of transient focal ischemia. Front Biosci. 12:2728–2734. 2007.
Article
43. Zhao Y, Li Q, Zhao W, Li J, Sun Y, Liu K, et al. Astragaloside IV and cycloastragenol are equally effective in inhibition of endoplasmic reticulum stress-associated TXNIP/NLRP3 inflammasome activation in the endothelium. J Ethnopharmacol. 169:210–218. 2015.
Article
44. Zhu X, Tao L, Tejima-Mandeville E, Qiu J, Park J, Garber K, et al. Plasmalemma permeability and necrotic cell death phenotypes after intracerebral hemorrhage in mice. Stroke. 43:524–531. 2012.
Article
Full Text Links
  • JKNS
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr