Critical role of NLRP3-caspase-1 pathway in age-dependent isoflurane-induced microglial inflammatory response and cognitive impairment

Zhi Wang, Shiyu Meng, Lin Cao, Ying Chen, Zhiyi Zuo, Shuling Peng, Zhi Wang, Shiyu Meng, Lin Cao, Ying Chen, Zhiyi Zuo, Shuling Peng

Abstract

Background: Elderly patients are more likely to suffer from postoperative cognitive dysfunction (POCD) after surgery and anesthesia. Except for declined organ function, the particular pathogenesis of POCD in elderly patients remains unknown. This study is carried out to determine the critical role of the NOD-like receptor protein 3 (NLRP3)-caspase-1 pathway in isoflurane-induced cognitive impairment.

Methods: Young (6-8 months old) and aged (14 months old) healthy male C57BL/6 mice were exposed to 1.5% isoflurane for 2 h. Some mice received intraperitoneal injection of Ac-YVAD-cmk (8 mg/kg), a specific inhibitor of caspase-1, 30 min before the isoflurane exposure. Morris water maze test was carried out 1 week after the isoflurane anesthesia. Brain tissues were harvested 24 h after the isoflurane anesthesia. Western blotting was carried out to detect the expression of NLRP3, interleukin (IL)-1β, and IL-18 in the hippocampus. Mouse microglial cell line BV-2 and primary microglial cultures were primed by lipopolysaccharide for 30 min before being exposed to isoflurane. NLRP3 was downregulated by RNA interference.

Results: Compared to young mice, aged mice had an increased expression of NLRP3 in the hippocampus. Isoflurane induced cognitive impairment and hippocampal inflammation in aged mice but not in young mice. These effects were attenuated by Ac-YVAD-cmk pretreatment (P < 0.05). Isoflurane activated NLRP3-caspase-1 pathway and increased the secretion of IL-18 and IL-1β in cells pretreated with lipopolysaccharide but not in cells without pretreatment. Downregulation of NLRP3 attenuated the activation of NLRP3 inflammasome by isoflurane.

Conclusions: NLRP3 priming status in aged mouse brain may be involved in isoflurane-induced hippocampal inflammation and cognitive impairment.

Keywords: Aging; Isoflurane; NOD-like receptor protein 3 inflammasome; Neuroinflammation; Postoperative cognitive dysfunction.

Conflict of interest statement

Ethics approval

All animal experiments were performed in accordance with current Chinese regulations and standards regarding the use of laboratory animals, and approved by the animal ethics committee of Sun Yat-Sen University.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests with the material presented in the paper.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Isoflurane induced age-related cognitive impairment. Young (6–8 months old) and aged (14 monts old) healthy male C57BL/6 mice were exposed to 1.5% isoflurane for 2 h. Some aged mice received intraperitoneal injection of 8 mg/kg Ac-YVAD-cmk 30 min before the isoflurane exposure. YC = blank control of young mice; Y-ISO = young mice exposed to isoflurane; AC = blank control of aged mice; A-ISO = aged mice exposed to isoflurane; A-ISO-cmk = Ac-YVAD-cmk administered before aged mice exposed to isoflurane; A-ISO-PBS = solvent of Ac-YVAD-cmk (PBS+ less than 1% DMSO) administered before aged mice exposed to isoflurane. Morris water maze test was carried out 1 week after the isoflurane anesthesia. a, b, and c Escape latency to reach the platform. d Target quadrant traveling time. Normalization of the target quadrant traveling time of group YC and Y-ISO was by the mean of group YC, and group AC, A-ISO, A-ISO-cmk, and A-ISO-PBS by the mean of group AC. e Platform-crossing times. f Average swimming speed on the sixth day. All results are mean ± SD (n = 15). #P < 0.05 and ##P < 0.01 compared with the corresponding data of group AC. *P < 0.05 compared with the corresponding data of group A-ISO
Fig. 2
Fig. 2
Contribution of NLRP3 inflammasome activation to isoflurane-induced age-related neuroinflammation. Hippocampus was harvested at 24 h after the anesthesia. a Representative western blot images of NLRP3, caspase-1 P45, caspase-1 P20, ASC, IL-18, IL-1β, cleaved caspase-3, and Iba-1. bi Graphic presentation of abundance of each protein. All values are expressed as fold changes over the mean values of group YC and are presented as mean ± SD (n ≥ 3). *P < 0.05 compared with the corresponding data of group YC. #P < 0.05 and ##P < 0.05 compared with the corresponding data of group AC
Fig. 3
Fig. 3
NLRP3 priming was induced by LPS stimulation. Different doses of LPS were added to the cell culture media of BV-2 cells or primary microglial cultures for 30 min. a The mRNA of NLRP3 was quantified by real-time qPCR immediately after LPS stimulation. Values are expressed as fold changes over the mean values of blank control. b IL-1β concentration in the supernatant was measured 6 h later. All results are presented as mean ± SD (n ≥ 3). *P < 0.05 and **P < 0.01 compared with the corresponding data of blank control
Fig. 4
Fig. 4
Isoflurane-induced NLRP3 inflammasome activation after priming. BV-2 microglia were exposed to 4% isoflurane for 6 h in the cells primed with or without 1 μg/mL LPS. Primary microglial cultures were exposed to 2% isoflurane for 6 h in the cells primed with or without 5 ng/mL LPS. Control = blank control; ISO = Isoflurane exposure; NLRP3-primed = LPS stimulation; NLRP3-primed+ ISO = NLRP3-primed + isoflurane exposure. a Western blot images of NLRP3, caspase-1 P45, caspase-1 P20, ASC, IL-18, and IL-1β from BV-2 cells. bg Graphic presentation of abundance of each protein in BV-2 cells. h Western blot images of NLRP3, ASC, caspase-1 P45, and caspase-1 P20, IL-18, and IL-1β. in Graphic presentation of abundance of each protein in primary microglia. Values are expressed as fold changes over the mean values of control and are presented as mean ± SD (n ≥ 3). *P < 0.05 and **P < 0.01 compared with the corresponding data of group control. #P < 0.05 and ##P < 0.01 compared with the corresponding data of group NLRP3-primed
Fig. 5
Fig. 5
NLRP3 priming was necessary in isoflurane-induced IL-1β production. BV-2 cells primed with or without 1 μg/mL LPS were exposed to 4% isoflurane for 6 h. Primary microglial cultures primed with or without 5 ng/mL LPS were exposed to 2% isoflurane for 6 h. Control = blank control; ISO = isoflurane exposure; NLRP3-primed = LPS stimulation; NLRP3-primed + ISO = NLRP3-primed + isoflurane exposure. a The mRNA of NLRP3 in BV-2 cells. Values are expressed as fold changes over the mean values of blank control and are presented as mean ± SD (n = 6). b IL-1β concentration in the supernatant of BV-2 cells. c Viability of NLRP3-primed cells at 0 and 12 h after isoflurane exposure. Values are expressed as fold changes over the mean values of NLRP3-primed cells and are presented as mean ± SD (n = 3). d The mRNA of NLRP3 in primary microglial cultures. Values are expressed as fold changes over the mean values of blank control and are presented as mean ± SD (n = 3). e IL-1β concentration in the supernatant of primary microglial cultures. Values are expressed as fold changes over the mean values of control and are presented as mean ± SD (n = 3). *P < 0.05 and **P < 0.01 compared with the corresponding data of group control. #P < 0.05 and ##P < 0.01 compared with the corresponding data of group NLRP3-primed cells
Fig. 6
Fig. 6
NLRP3 knock-down reduced isoflurane-induced NLRP3 inflammasome activation. Cells were transfected with NLRP3 siRNA or negative control siRNA 24 h before NLRP3 priming and exposure to 4% isoflurane for 6 h. NC-siRNA + ISO = negative control of siRNA before NLRP3 priming and isoflurane exposure; NLRP3-siRNA + ISO = NLRP3 siRNA before NLRP3 priming and isoflurane exposure. a Western blot images of NLRP3, caspase-1 P45, caspase-1 P20, ASC, IL-18, and IL-1β. bg The graphic presentation of each protein abundance of Fig. 6a. h Whole cell protein samples from BV-2 cells were harvested immediately after isoflurane treatment. Immunoprecipitation was performed by using an anti-NLRP3 antibody and was immunoblotted for NLRP3, ASC, and pro-caspase-1. Values are expressed as fold changes over the mean values of NLRP3-primed cells and are presented as mean ± SD (n = 6). *P < 0.05 compared with the corresponding data of group control. #P < 0.05 compared with the corresponding data of NLRP3-primed + ISO

References

    1. Akbaraly TN, Hamer M, Ferrie JE, Lowe G, Batty GD, Hagger-Johnson G, Singh-Manoux A, Shipley MJ, Kivimaki M. Chronic inflammation as a determinant of future aging phenotypes. CMAJ. 2013;185:E763. doi: 10.1503/cmaj.122072.
    1. Bittner EA, Yue Y, Xie Z. Brief review: anesthetic neurotoxicity in the elderly, cognitive dysfunction and Alzheimer’s disease. Can J Anaesth. 2011;58:216. doi: 10.1007/s12630-010-9418-x.
    1. Androsova G, Krause R, Winterer G, Schneider R. Biomarkers of postoperative delirium and cognitive dysfunction. Front Aging Neurosci. 2015;7:112. doi: 10.3389/fnagi.2015.00112.
    1. Terrando N, Eriksson LI, Kyu Ryu J, Yang T, Monaco C, Feldmann M, Jonsson Fagerlund M, Charo IF, Akassoglou K, Maze M. Resolving postoperative neuroinflammation and cognitive decline. Ann Neurol. 2011;70:986. doi: 10.1002/ana.22664.
    1. Zuo Z. Postoperative cognitive effects in newborns: the role of inflammatory processes. Anesthesiology. 2013;118:481. doi: 10.1097/ALN.0b013e3182835276.
    1. Zhang Z, Li X, Li F, An L. Berberine alleviates postoperative cognitive dysfunction by suppressing neuroinflammation in aged mice. Int Immunopharmacol. 2016;38:426. doi: 10.1016/j.intimp.2016.06.031.
    1. Hovens IB, van Leeuwen BL, Nyakas C, Heineman E, van der Zee EA, Schoemaker RG. Postoperative cognitive dysfunction and microglial activation in associated brain regions in old rats. Neurobiol Learn Mem. 2015;118:74. doi: 10.1016/j.nlm.2014.11.009.
    1. Franceschi C, Bonafe M, Valensin S, Olivieri F, De Luca M, Ottaviani E, De Benedictis G. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci. 2000;908:244. doi: 10.1111/j.1749-6632.2000.tb06651.x.
    1. Goto M. Inflammaging (inflammation + aging): a driving force for human aging based on an evolutionarily antagonistic pleiotropy theory? Biosci Trends. 2008;2:218.
    1. Koellhoffer EC, McCullough LD, Ritzel RM. Old maids: aging and its impact on microglia function. Int J Mol Sci. 2017;18:e769
    1. Harry GJ. Microglia during development and aging. Pharmacol Ther. 2013;139:313. doi: 10.1016/j.pharmthera.2013.04.013.
    1. Li W. Phagocyte dysfunction, tissue aging and degeneration. Ageing Res Rev. 2013;12:1005. doi: 10.1016/j.arr.2013.05.006.
    1. Cao L, Li L, Lin D, Zuo Z. Isoflurane induces learning impairment that is mediated by interleukin 1beta in rodents. PLoS One. 2012;7:e51431. doi: 10.1371/journal.pone.0051431.
    1. Ji M, Dong L, Jia M, Liu W, Zhang M, Ju L, Yang J, Xie Z, Yang J. Epigenetic enhancement of brain-derived neurotrophic factor signaling pathway improves cognitive impairments induced by isoflurane exposure in aged rats. Mol Neurobiol. 2014;50:937. doi: 10.1007/s12035-014-8659-z.
    1. Lin D, Cao L, Wang Z, Li J, Washington JM, Zuo Z. Lidocaine attenuates cognitive impairment after isoflurane anesthesia in old rats. Behav Brain Res. 2012;228:319. doi: 10.1016/j.bbr.2011.12.010.
    1. Li X, Zhou M, Wang X, Ji M, Zhou Z, Yang J. Resveratrol pretreatment attenuates the isoflurane-induced cognitive impairment through its anti-inflammation and -apoptosis actions in aged mice. J Mol Neurosci. 2014;52:286. doi: 10.1007/s12031-013-0141-2.
    1. Lamkanfi M, Dixit VM. Mechanisms and functions of inflammasomes. Cell. 2014;157:1013. doi: 10.1016/j.cell.2014.04.007.
    1. Heneka MT, Kummer MP, Latz E. Innate immune activation in neurodegenerative disease. Nat Rev Immunol. 2014;14:463. doi: 10.1038/nri3705.
    1. Sutterwala FS, Haasken S, Cassel SL. Mechanism of NLRP3 inflammasome activation. Ann N Y Acad Sci. 2014;1319:82. doi: 10.1111/nyas.12458.
    1. Youm Y, Grant RW, McCabe LR, Albarado DC, Nguyen KY, Ravussin A, Pistell P, Newman S, Carter R, Laque A, Münzberg H, Rosen CJ, Ingram DK, Salbaum JM, Dixit VD. Canonical Nlrp3 inflammasome links systemic low-grade inflammation to functional decline in aging. Cell Metab. 2013;18:519. doi: 10.1016/j.cmet.2013.09.010.
    1. Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, Griep A, Axt D, Remus A, Tzeng T, Gelpi E, Halle A, Korte M, Latz E, Golenbock DT. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature. 2012;493:674. doi: 10.1038/nature11729.
    1. Goldmann T, Tay TL, Prinz M. Love and death: microglia, NLRP3 and the Alzheimer's brain. Cell Res. 2013;23:595. doi: 10.1038/cr.2013.24.
    1. Zhuang J, Wen X, Zhang YQ, Shan Q, Zhang ZF, Zheng GH, Fan SH, Li MQ, Wu DM, Hu B, Lu J, Zheng YL. TDP-43 upregulation mediated by the NLRP3 inflammasome induces cognitive impairment in 2 2′,4,4′-tetrabromodiphenyl ether (BDE-47)-treated mice. Brain Behav Immun. 2017;65:99. doi: 10.1016/j.bbi.2017.05.014.
    1. Latz E. The inflammasomes: mechanisms of activation and function. Curr Opin Immunol. 2010;22:28. doi: 10.1016/j.coi.2009.12.004.
    1. Guo H, Callaway JB, Ting JP. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med. 2015;21:677. doi: 10.1038/nm.3893.
    1. Abais JM, Xia M, Zhang Y, Boini KM, Li P. Redox regulation of NLRP3 inflammasomes: ROS as trigger or effector? Antioxid Redox Sign. 2015;22:1111.
    1. Zhang F, Wang L, Wang J, Luo P, Wang X, Xia Z. The caspase-1 inhibitor AC-YVAD-CMK attenuates acute gastric injury in mice: involvement of silencing NLRP3 inflammasome activities. Sci Rep. 2016;6:24166.
    1. Pan K, Li X, Chen Y, Zhu D, Li Y, Tao G, Zuo Z. Deferoxamine pre-treatment protects against postoperative cognitive dysfunction of aged rats by depressing microglial activation via ameliorating iron accumulation in hippocampus. Neuropharmacology. 2016;111:180. doi: 10.1016/j.neuropharm.2016.09.004.
    1. Johnson T, Monk T, Rasmussen LS, Abildstrom H, Houx P, Korttila K, Kuipers HM, Hanning CD, Siersma VD, Kristensen D, Canet J, Ibanaz MT, Moller JT. Postoperative cognitive dysfunction in middle-aged patients. Anesthesiology. 2002;96:1351. doi: 10.1097/00000542-200206000-00014.
    1. Cibelli M, Fidalgo AR, Terrando N, Ma D, Monaco C, Feldmann M, Takata M, Lever IJ, Nanchahal J, Fanselow MS, Maze M. Role of interleukin-1beta in postoperative cognitive dysfunction. Ann Neurol. 2010;68:360. doi: 10.1002/ana.22082.
    1. Ye X, Lian Q, Eckenhoff MF, Eckenhoff RG, Pan JZ. Differential general anesthetic effects on microglial cytokine expression. PLoS One. 2013;8:e52887. doi: 10.1371/journal.pone.0052887.
    1. Zhang L, Zhang J, Yang L, Dong Y, Zhang Y, Xie Z. Isoflurane and sevoflurane increase interleukin-6 levels through the nuclear factor-kappa B pathway in neuroglioma cells. Brit J Anaesth. 2013;110:i82. doi: 10.1093/bja/aet115.
    1. Schallner N, Ulbrich F, Engelstaedter H, Biermann J, Auwaerter V, Loop T, Goebel U. Isoflurane but not sevoflurane or desflurane aggravates injury to neurons in vitro and in vivo via p75NTR-NF-kB activation. Anesth Analg. 2014;119:1429. doi: 10.1213/ANE.0000000000000488.
    1. Eckel B, Richtsfeld M, Starker L, Blobner M. Transgenic Alzheimer mice have a larger minimum alveolar anesthetic concentration of isoflurane than their nontransgenic littermates. Anesth Analg. 2010;110:438. doi: 10.1213/ANE.0b013e3181b76383.
    1. Wu J, Li H, Sun X, Zhang H, Hao S, Ji M, Yang J, Li K. A mitochondrion-targeted antioxidant ameliorates isoflurane-induced cognitive deficits in aging mice. PLoS One. 2015;10:e138256.
    1. Tanino M, Kobayashi M, Sasaki T, Takata K, Takeda Y, Mizobuchi S, Morita K, Nagai T, Morimatsu H. Isoflurane induces transient impairment of retention of spatial working memory in rats. Acta Med Okayama. 2016;70:455.
    1. Wu X, Lu Y, Dong Y, Zhang G, Zhang Y, Xu Z, Culley DJ, Crosby G, Marcantonio ER, Tanzi RE, Xie Z. The inhalation anesthetic isoflurane increases levels of proinflammatory TNF-α, IL-6, and IL-1β. Neurobiol Aging. 2012;33:1364. doi: 10.1016/j.neurobiolaging.2010.11.002.
    1. Fougere B, Boulanger E, Nourhashemi F, Guyonnet S, Cesari M. Chronic inflammation: accelerator of biological aging. J Gerontol A Biol Sci Med Sci. 2016;72:1218.
    1. Fernandes A, Miller-Fleming L, Pais TF. Microglia and inflammation: conspiracy, controversy or control? Cell Mol Life Sci. 2014;71:3969. doi: 10.1007/s00018-014-1670-8.
    1. Norden DM, Godbout JP. Review: microglia of the aged brain: primed to be activated and resistant to regulation. Neuropath Appl Neuro. 2013;39:19. doi: 10.1111/j.1365-2990.2012.01306.x.
    1. Tang Y, Le W. Differential roles of M1 and M2 microglia in neurodegenerative diseases. Mol Neurobiol. 2015;53:1181.
    1. Gustin A, Kirchmeyer M, Koncina E, Felten P, Losciuto S, Heurtaux T, Tardivel A, Heuschling P, Dostert C. NLRP3 Inflammasome is expressed and functional in mouse brain microglia but not in astrocytes. PLoS One. 2015;10:e130624.
    1. Heneka MT. Inflammasome activation and innate immunity in Alzheimer’s disease. Brain Pathol. 2017;27:220. doi: 10.1111/bpa.12483.
    1. Trott DW, Henson GD, Ho MH, Allison SA, Lesniewski LA, Donato AJ. Age-related arterial immune cell infiltration in mice is attenuated by caloric restriction or voluntary exercise. Exp Gerontol. 2016;(16)30348-5.
    1. Butovsky O, Jedrychowski MP, Moore CS, Cialic R, Lanser AJ, Gabriely G, Koeglsperger T, Dake B, Wu PM, Doykan CE, Fanek Z, Liu L, Chen Z, Rothstein JD, Ransohoff RM, Gygi SP, Antel JP, Weiner HL. Identification of a unique TGF-β–dependent molecular and functional signature in microglia. Nat Neurosci. 2013;17:131. doi: 10.1038/nn.3599.
    1. Weber MD, Frank MG, Tracey KJ, Watkins LR, Maier SF. Stress induces the danger-associated molecular pattern HMGB-1 in the Hippocampus of male Sprague Dawley rats: a priming stimulus of microglia and the NLRP3 Inflammasome. J Neurosci. 2015;35:316. doi: 10.1523/JNEUROSCI.3561-14.2015.
    1. You T, Cheng Y, Zhong J, Bi B, Zeng B, Zheng W, Wang H, Xu J. Roflupram, a phosphodiesterase 4 inhibitior, suppresses inflammasome activation through autophagy in microglial cells. ACS Chem Neurosci. 2017;8:2381.
    1. Yang H, Chen Y, Yu L, Xu Y. Esculentoside a exerts anti-inflammatory activity in microglial cells. Int Immunopharmacol. 2017;51:148. doi: 10.1016/j.intimp.2017.08.014.
    1. Kaushal V, Dye R, Pakavathkumar P, Foveau B, Flores J, Hyman B, Ghetti B, Koller BH, LeBlanc AC. Neuronal NLRP1 inflammasome activation of Caspase-1 coordinately regulates inflammatory interleukin-1-beta production and axonal degeneration-associated Caspase-6 activation. Cell Death Differ. 2015;22:1676. doi: 10.1038/cdd.2015.16.

Source: PubMed

3
Tilaa