Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2022

Open Access 01-12-2022 | Intracranial Aneurysm | Research

An imbalance between RAGE/MR/HMGB1 and ATP1α3 is associated with inflammatory changes in rat brain harboring cerebral aneurysms prone to rupture

Authors: Eiji Shikata, Takeshi Miyamoto, Tadashi Yamaguchi, Izumi Yamaguchi, Hiroshi Kagusa, Daiki Gotoh, Kenji Shimada, Yoshiteru Tada, Kenji Yagi, Keiko T. Kitazato, Yasuhisa Kanematsu, Yasushi Takagi

Published in: Journal of Neuroinflammation | Issue 1/2022

Login to get access

Abstract

Background and purpose

An aneurysmal subarachnoid hemorrhage is a devastating event. To establish an effective therapeutic strategy, its pathogenesis must be clarified, particularly the pathophysiology of brain harboring intracranial aneurysms (IAs). To elucidate the pathology in brain harboring IAs, we examined the significance of the receptor for advanced glycation end-products (RAGE)/mineralocorticoid receptor (MR) pathway and Na+/K+-ATPase (ATP1α3).

Methods

Ten-week-old female rats were subjected to oophorectomy as well as hypertension and hemodynamic changes to induce IAs, and were fed a high-salt diet. Brain damage in these rats was assessed by inflammatory changes in comparison to sham-operated rats fed a standard diet.

Results

Six weeks after IA induction (n = 30), irregular morphological changes, i.e., an enlarged vessel diameter and vascular wall, were observed in all of the left posterior cerebral arteries (Lt PCAs) prone to rupture. Approximately 20% of rats had ruptured IAs within 6 weeks. In brain harboring unruptured IAs at the PCA, the mRNA levels of RAGE and MR were higher, and that of ATP1α3 was lower than those in the sham-operated rats (p < 0.05, each). Immunohistochemically, elevated expression of RAGE and MR, and decreased expression of ATP1α3 were observed in the brain parenchyma adjacent to the Lt PCA, resulting in increased Iba-1 and S100B expression that reflected the inflammatory changes. There was no difference between the unruptured and ruptured aneurysm rat groups. Treatment with the MR antagonist esaxerenone abrogated these changes, and led to cerebral and vascular normalization and prolonged subarachnoid hemorrhage-free survival (p < 0.05).

Conclusions

Regulation of the imbalance between the RAGE/MR pathway and ATP1α3 may help attenuate the damage in brain harboring IAs, and further studies are warranted to clarify the significance of the down-regulation of the MR/RAGE pathway and the up-regulation of ATP1α3 for attenuating the pathological changes in brain harboring IAs.
Literature
1.
go back to reference Macdonald RL, Schweizer TA. Spontaneous subarachnoid haemorrhage. Lancet. 2017;389(10069):655–66.CrossRef Macdonald RL, Schweizer TA. Spontaneous subarachnoid haemorrhage. Lancet. 2017;389(10069):655–66.CrossRef
2.
go back to reference Miyamoto T, Kung DK, Kitazato KT, Yagi K, Shimada K, Tada Y, et al. Site-specific elevation of interleukin-1beta and matrix metalloproteinase-9 in the Willis circle by hemodynamic changes is associated with rupture in a novel rat cerebral aneurysm model. J Cereb Blood Flow Metab. 2017;37(8):2795–805.CrossRef Miyamoto T, Kung DK, Kitazato KT, Yagi K, Shimada K, Tada Y, et al. Site-specific elevation of interleukin-1beta and matrix metalloproteinase-9 in the Willis circle by hemodynamic changes is associated with rupture in a novel rat cerebral aneurysm model. J Cereb Blood Flow Metab. 2017;37(8):2795–805.CrossRef
3.
go back to reference Yamaguchi T, Miyamoto T, Kitazato KT, Shikata E, Yamaguchi I, Korai M, et al. Time-dependent and site-dependent morphological changes in rupture-prone arteries: ovariectomized rat intracranial aneurysm model. J Neurosurg. 2019:1–9. Yamaguchi T, Miyamoto T, Kitazato KT, Shikata E, Yamaguchi I, Korai M, et al. Time-dependent and site-dependent morphological changes in rupture-prone arteries: ovariectomized rat intracranial aneurysm model. J Neurosurg. 2019:1–9.
4.
go back to reference Matsushita N, Kitazato KT, Tada Y, Sumiyoshi M, Shimada K, Yagi K, et al. Increase in body Na+/water ratio is associated with cerebral aneurysm formation in oophorectomized rats. Hypertension. 2012;60(5):1309–15.CrossRef Matsushita N, Kitazato KT, Tada Y, Sumiyoshi M, Shimada K, Yagi K, et al. Increase in body Na+/water ratio is associated with cerebral aneurysm formation in oophorectomized rats. Hypertension. 2012;60(5):1309–15.CrossRef
5.
go back to reference Tada Y, Kitazato KT, Tamura T, Yagi K, Shimada K, Kinouchi T, et al. Role of mineralocorticoid receptor on experimental cerebral aneurysms in rats. Hypertension. 2009;54(3):552–7.CrossRef Tada Y, Kitazato KT, Tamura T, Yagi K, Shimada K, Kinouchi T, et al. Role of mineralocorticoid receptor on experimental cerebral aneurysms in rats. Hypertension. 2009;54(3):552–7.CrossRef
6.
go back to reference Nagahiro S, Tada Y, Satomi J, Kinouchi T, Kuwayama K, Yagi K, et al. Treatment of unruptured cerebral aneurysms with the mineralocorticoid receptor blocker eplerenone-pilot study. J Stroke Cerebrovasc Dis. 2018;27(8):2134–40.CrossRef Nagahiro S, Tada Y, Satomi J, Kinouchi T, Kuwayama K, Yagi K, et al. Treatment of unruptured cerebral aneurysms with the mineralocorticoid receptor blocker eplerenone-pilot study. J Stroke Cerebrovasc Dis. 2018;27(8):2134–40.CrossRef
7.
go back to reference Li H, Wu W, Sun Q, Liu M, Li W, Zhang XS, et al. Expression and cell distribution of receptor for advanced glycation end-products in the rat cortex following experimental subarachnoid hemorrhage. Brain Res. 2014;1543:315–23.CrossRef Li H, Wu W, Sun Q, Liu M, Li W, Zhang XS, et al. Expression and cell distribution of receptor for advanced glycation end-products in the rat cortex following experimental subarachnoid hemorrhage. Brain Res. 2014;1543:315–23.CrossRef
8.
go back to reference Hudson BI, Lippman ME. Targeting RAGE signaling in inflammatory disease. Annu Rev Med. 2018;69:349–64.CrossRef Hudson BI, Lippman ME. Targeting RAGE signaling in inflammatory disease. Annu Rev Med. 2018;69:349–64.CrossRef
9.
go back to reference Piras S, Furfaro AL, Domenicotti C, Traverso N, Marinari UM, Pronzato MA, et al. RAGE expression and ROS generation in neurons: differentiation versus damage. Oxid Med Cell Longev. 2016;2016:9348651.CrossRef Piras S, Furfaro AL, Domenicotti C, Traverso N, Marinari UM, Pronzato MA, et al. RAGE expression and ROS generation in neurons: differentiation versus damage. Oxid Med Cell Longev. 2016;2016:9348651.CrossRef
10.
go back to reference Richard SA. Elucidating the novel biomarker and therapeutic potentials of high-mobility group box 1 in subarachnoid hemorrhage: a review. AIMS Neurosci. 2019;6(4):316–32.CrossRef Richard SA. Elucidating the novel biomarker and therapeutic potentials of high-mobility group box 1 in subarachnoid hemorrhage: a review. AIMS Neurosci. 2019;6(4):316–32.CrossRef
11.
go back to reference Zhu XD, Chen JS, Zhou F, Liu QC, Chen G, Zhang JM. Relationship between plasma high mobility group box-1 protein levels and clinical outcomes of aneurysmal subarachnoid hemorrhage. J Neuroinflamm. 2012;9:194.CrossRef Zhu XD, Chen JS, Zhou F, Liu QC, Chen G, Zhang JM. Relationship between plasma high mobility group box-1 protein levels and clinical outcomes of aneurysmal subarachnoid hemorrhage. J Neuroinflamm. 2012;9:194.CrossRef
12.
go back to reference Muhammad S, Chaudhry SR, Kahlert UD, Lehecka M, Korja M, Niemela M, et al. Targeting high mobility group box 1 in subarachnoid hemorrhage: a systematic review. Int J Mol Sci. 2020;21(8):2709.CrossRef Muhammad S, Chaudhry SR, Kahlert UD, Lehecka M, Korja M, Niemela M, et al. Targeting high mobility group box 1 in subarachnoid hemorrhage: a systematic review. Int J Mol Sci. 2020;21(8):2709.CrossRef
13.
go back to reference Jeong J, Lee J, Lim J, Cho S, An S, Lee M, et al. Soluble RAGE attenuates AngII-induced endothelial hyperpermeability by disrupting HMGB1-mediated crosstalk between AT1R and RAGE. Exp Mol Med. 2019;51(9):1–15.CrossRef Jeong J, Lee J, Lim J, Cho S, An S, Lee M, et al. Soluble RAGE attenuates AngII-induced endothelial hyperpermeability by disrupting HMGB1-mediated crosstalk between AT1R and RAGE. Exp Mol Med. 2019;51(9):1–15.CrossRef
14.
go back to reference Wang HW, Huang BS, Chen A, Ahmad M, White RA, Leenen FH. Role of brain aldosterone and mineralocorticoid receptors in aldosterone-salt hypertension in rats. Neuroscience. 2016;314:90–105.CrossRef Wang HW, Huang BS, Chen A, Ahmad M, White RA, Leenen FH. Role of brain aldosterone and mineralocorticoid receptors in aldosterone-salt hypertension in rats. Neuroscience. 2016;314:90–105.CrossRef
15.
go back to reference Sumiyoshi M, Kitazato KT, Yagi K, Miyamoto T, Kurashiki Y, Matsushita N, et al. The accumulation of brain water-free sodium is associated with ischemic damage independent of the blood pressure in female rats. Brain Res. 2015;1616:37–44.CrossRef Sumiyoshi M, Kitazato KT, Yagi K, Miyamoto T, Kurashiki Y, Matsushita N, et al. The accumulation of brain water-free sodium is associated with ischemic damage independent of the blood pressure in female rats. Brain Res. 2015;1616:37–44.CrossRef
16.
go back to reference Vetro A, Nielsen HN, Holm R, Hevner RF, Parrini E, Powis Z, et al. ATP1A2- and ATP1A3-associated early profound epileptic encephalopathy and polymicrogyria. Brain. 2021;144(5):1435–50.CrossRef Vetro A, Nielsen HN, Holm R, Hevner RF, Parrini E, Powis Z, et al. ATP1A2- and ATP1A3-associated early profound epileptic encephalopathy and polymicrogyria. Brain. 2021;144(5):1435–50.CrossRef
17.
go back to reference Smith RS, Florio M, Akula SK, Neil JE, Wang Y, Hill RS, et al. Early role for a Na(+), K(+)-ATPase (ATP1A3) in brain development. Proc Natl Acad Sci U S A. 2021;118(25): e2023333118.CrossRef Smith RS, Florio M, Akula SK, Neil JE, Wang Y, Hill RS, et al. Early role for a Na(+), K(+)-ATPase (ATP1A3) in brain development. Proc Natl Acad Sci U S A. 2021;118(25): e2023333118.CrossRef
18.
go back to reference Ito S, Itoh H, Rakugi H, Okuda Y, Yoshimura M, Yamakawa S. Double-blind randomized phase 3 study comparing esaxerenone (CS-3150) and eplerenone in patients with essential hypertension (ESAX-HTN Study). Hypertension. 2020;75(1):51–8.CrossRef Ito S, Itoh H, Rakugi H, Okuda Y, Yoshimura M, Yamakawa S. Double-blind randomized phase 3 study comparing esaxerenone (CS-3150) and eplerenone in patients with essential hypertension (ESAX-HTN Study). Hypertension. 2020;75(1):51–8.CrossRef
19.
go back to reference Tóbon-Velasco JC, Cuevas E, Torres-Ramos MA. Receptor for AGEs (RAGE) as mediator of NF-kB pathway activation in neuroinflammation and oxidative stress. CNS Neurol Disord Drug Targets. 2014;13(9):1615–26.CrossRef Tóbon-Velasco JC, Cuevas E, Torres-Ramos MA. Receptor for AGEs (RAGE) as mediator of NF-kB pathway activation in neuroinflammation and oxidative stress. CNS Neurol Disord Drug Targets. 2014;13(9):1615–26.CrossRef
20.
go back to reference Chen Y, Yu Y, Qiao J, Zhu L, Xiao Z. Mineralocorticoid receptor excessive activation involved in glucocorticoid-related brain injury. Biomed Pharmacother. 2020;122: 109695.CrossRef Chen Y, Yu Y, Qiao J, Zhu L, Xiao Z. Mineralocorticoid receptor excessive activation involved in glucocorticoid-related brain injury. Biomed Pharmacother. 2020;122: 109695.CrossRef
21.
go back to reference Gasparotto J, Ribeiro CT, da Rosa-Silva HT, Bortolin RC, Rabelo TK, Peixoto DO, et al. Systemic inflammation changes the site of RAGE expression from endothelial cells to neurons in different brain areas. Mol Neurobiol. 2019;56(5):3079–89.CrossRef Gasparotto J, Ribeiro CT, da Rosa-Silva HT, Bortolin RC, Rabelo TK, Peixoto DO, et al. Systemic inflammation changes the site of RAGE expression from endothelial cells to neurons in different brain areas. Mol Neurobiol. 2019;56(5):3079–89.CrossRef
22.
go back to reference Heinzen EL, Arzimanoglou A, Brashear A, Clapcote SJ, Gurrieri F, Goldstein DB, et al. Distinct neurological disorders with ATP1A3 mutations. Lancet Neurol. 2014;13(5):503–14.CrossRef Heinzen EL, Arzimanoglou A, Brashear A, Clapcote SJ, Gurrieri F, Goldstein DB, et al. Distinct neurological disorders with ATP1A3 mutations. Lancet Neurol. 2014;13(5):503–14.CrossRef
23.
go back to reference Sumiyoshi M, Satomi J, Kitazato KT, Yagi K, Shimada K, Kurashiki Y, et al. PPARgamma-dependent and -independent inhibition of the HMGB1/TLR9 pathway by eicosapentaenoic acid attenuates ischemic brain damage in ovariectomized rats. J Stroke Cerebrovasc Dis. 2015;24(6):1187–95.CrossRef Sumiyoshi M, Satomi J, Kitazato KT, Yagi K, Shimada K, Kurashiki Y, et al. PPARgamma-dependent and -independent inhibition of the HMGB1/TLR9 pathway by eicosapentaenoic acid attenuates ischemic brain damage in ovariectomized rats. J Stroke Cerebrovasc Dis. 2015;24(6):1187–95.CrossRef
24.
go back to reference Wakayama K, Shimamura M, Suzuki JI, Watanabe R, Koriyama H, Akazawa H, et al. Angiotensin II peptide vaccine protects ischemic brain through reducing oxidative stress. Stroke. 2017;48(5):1362–8.CrossRef Wakayama K, Shimamura M, Suzuki JI, Watanabe R, Koriyama H, Akazawa H, et al. Angiotensin II peptide vaccine protects ischemic brain through reducing oxidative stress. Stroke. 2017;48(5):1362–8.CrossRef
25.
go back to reference Abiodun OA, Ola MS. Role of brain renin angiotensin system in neurodegeneration: an update. Saudi J Biol Sci. 2020;27(3):905–12.CrossRef Abiodun OA, Ola MS. Role of brain renin angiotensin system in neurodegeneration: an update. Saudi J Biol Sci. 2020;27(3):905–12.CrossRef
26.
go back to reference Farmer DG, Kennedy S. RAGE, vascular tone and vascular disease. Pharmacol Ther. 2009;124(2):185–94.CrossRef Farmer DG, Kennedy S. RAGE, vascular tone and vascular disease. Pharmacol Ther. 2009;124(2):185–94.CrossRef
27.
go back to reference Park S, Yoon S-J, Tae H-J, Shim CY. RAGE and cardiovascular disease. FBL. 2011;16(2):486–97.PubMed Park S, Yoon S-J, Tae H-J, Shim CY. RAGE and cardiovascular disease. FBL. 2011;16(2):486–97.PubMed
28.
go back to reference Dinh QN, Arumugam TV, Young MJ, Drummond GR, Sobey CG, Chrissobolis S. Aldosterone and the mineralocorticoid receptor in the cerebral circulation and stroke. Exp Transl Stroke Med. 2012;4(1):21.CrossRef Dinh QN, Arumugam TV, Young MJ, Drummond GR, Sobey CG, Chrissobolis S. Aldosterone and the mineralocorticoid receptor in the cerebral circulation and stroke. Exp Transl Stroke Med. 2012;4(1):21.CrossRef
29.
go back to reference Pena Silva RA, Chu Y, Miller JD, Mitchell IJ, Penninger JM, Faraci FM, et al. Impact of ACE2 deficiency and oxidative stress on cerebrovascular function with aging. Stroke. 2012;43(12):3358–63.CrossRef Pena Silva RA, Chu Y, Miller JD, Mitchell IJ, Penninger JM, Faraci FM, et al. Impact of ACE2 deficiency and oxidative stress on cerebrovascular function with aging. Stroke. 2012;43(12):3358–63.CrossRef
30.
go back to reference Taguchi K, Yamagishi SI, Yokoro M, Ito S, Kodama G, Kaida Y, et al. RAGE-aptamer attenuates deoxycorticosterone acetate/salt-induced renal injury in mice. Sci Rep. 2018;8(1):2686.CrossRef Taguchi K, Yamagishi SI, Yokoro M, Ito S, Kodama G, Kaida Y, et al. RAGE-aptamer attenuates deoxycorticosterone acetate/salt-induced renal injury in mice. Sci Rep. 2018;8(1):2686.CrossRef
31.
go back to reference Shikata E, Tamura T, Shinno K, Okayama Y, Shinohara N, Shimada K, et al. Importance of managing the water-electrolyte balance by delivering the optimal minimum amount of water and sodium after subarachnoid hemorrhage. World Neurosurg. 2019;129:e352–60.CrossRef Shikata E, Tamura T, Shinno K, Okayama Y, Shinohara N, Shimada K, et al. Importance of managing the water-electrolyte balance by delivering the optimal minimum amount of water and sodium after subarachnoid hemorrhage. World Neurosurg. 2019;129:e352–60.CrossRef
Metadata
Title
An imbalance between RAGE/MR/HMGB1 and ATP1α3 is associated with inflammatory changes in rat brain harboring cerebral aneurysms prone to rupture
Authors
Eiji Shikata
Takeshi Miyamoto
Tadashi Yamaguchi
Izumi Yamaguchi
Hiroshi Kagusa
Daiki Gotoh
Kenji Shimada
Yoshiteru Tada
Kenji Yagi
Keiko T. Kitazato
Yasuhisa Kanematsu
Yasushi Takagi
Publication date
01-12-2022
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2022
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-022-02526-7

Other articles of this Issue 1/2022

Journal of Neuroinflammation 1/2022 Go to the issue