Skip to main content
Top
Published in: BMC Anesthesiology 1/2023

Open Access 01-12-2023 | Research

Dexmedetomidine attenuates postoperative spatial memory impairment after surgery by reducing cytochrome C

Authors: Lina Sun, Kun Niu, Jian Guo, Jingru Tu, Baofeng Ma, Jianxiong An

Published in: BMC Anesthesiology | Issue 1/2023

Login to get access

Abstract

Background

Anesthesia and surgery can induce perioperative neurocognitive disorders (PND). Mitochondrial dysfunction has been proposed to be one of the earliest triggering events in surgery-induced neuronal damage. Dexmedetomidine has been demonstrated to attenuate the impairment of cognition in aged rats induced by surgery in our previous study.

Methods

Male Sprague-Dawley rats underwent hepatic apex resection under anesthesia with propofol to clinically mimic human abdominal surgery. The rats were divided into three groups: Control group, Model group and Dexmedetomidine (Dex) group. Cognitive function was evaluated with the Morris water maze (MWM), Open Field Test (OFT)and Novel object recognition task (NOR). Ultrastructural change in neuronal mitochondria was measured by transmission electron microscopy. Mitochondrial function was measured by mitochondrial membrane potential and activities of mitochondrial complexes. Neuronal morphology was observed with H&E staining and the activation of glial cells was observed by immunohistochemistry in the hippocampus. Protein levels were measured by Western blot (WB) and immunofluorescence at 3 and 7 days after surgery.

Results

Surgery-induced cognitive decline lasts three days, but not seven days after surgery in the model group. Transmission electron microscope showed the mitochondrial structure damage in the model group, similar changes were not induced in the Dex group. Dexmedetomidine may reverse the decrease in mitochondrial membrane potential and mitochondrial complex activity. Compared with the Control group, the expression of cytochrome c was significantly increased in model group by Western blot and immunofluorescence on days 3, but not day 7. Rats from the Model group expressed significantly greater levels of Iba-1 and GFAP compared with the Control group and the Dex group.

Conclusion

Dexmedetomidine appears to reverse surgery-induced behavior, mitigate the higher density of Iba-1 and GFAP, reduce the damage of mitochondrial structure and function by alleviating oxidative stress and protect mitochondrial respiratory chain, thus increasing cytochrome c oxidase (COX) expression and downregulate the expression of cytochrome c protein in the hippocampus of rats.
Appendix
Available only for authorised users
Literature
1.
go back to reference Norkiene I, Samalavicius R, Misiuriene I, Paulauskiene K, Budrys V, Ivaskevicius J. Incidence and risk factors for early postoperative cognitive decline after coronary artery bypass grafting. Med (Kaunas). 2010;46(7):460–4. Norkiene I, Samalavicius R, Misiuriene I, Paulauskiene K, Budrys V, Ivaskevicius J. Incidence and risk factors for early postoperative cognitive decline after coronary artery bypass grafting. Med (Kaunas). 2010;46(7):460–4.
3.
go back to reference Deiner S, Luo X, Lin HM, Sessler DI, Saager L, Sieber FE, Lee HB, Sano M, Jankowski C, Bergese SD, et al. Intraoperative infusion of Dexmedetomidine for Prevention of Postoperative Delirium and Cognitive Dysfunction in Elderly Patients undergoing major elective noncardiac surgery: a Randomized Clinical Trial. JAMA Surg. 2017;152(8):e171505.CrossRefPubMedPubMedCentral Deiner S, Luo X, Lin HM, Sessler DI, Saager L, Sieber FE, Lee HB, Sano M, Jankowski C, Bergese SD, et al. Intraoperative infusion of Dexmedetomidine for Prevention of Postoperative Delirium and Cognitive Dysfunction in Elderly Patients undergoing major elective noncardiac surgery: a Randomized Clinical Trial. JAMA Surg. 2017;152(8):e171505.CrossRefPubMedPubMedCentral
4.
go back to reference Skvarc DR, Berk M, Byrne LK, Dean OM, Dodd S, Lewis M, Marriott A, Moore EM, Morris G, Page RS, et al. Post-operative cognitive dysfunction: an exploration of the inflammatory hypothesis and novel therapies. Neurosci Biobehav Rev. 2018;84:116–33.CrossRefPubMed Skvarc DR, Berk M, Byrne LK, Dean OM, Dodd S, Lewis M, Marriott A, Moore EM, Morris G, Page RS, et al. Post-operative cognitive dysfunction: an exploration of the inflammatory hypothesis and novel therapies. Neurosci Biobehav Rev. 2018;84:116–33.CrossRefPubMed
5.
go back to reference He K, Zhang J, Zhang W, Wang S, Li D, Ma X, Wu X, Chai X, Liu Q. Hippocampus-based mitochondrial respiratory function decline is responsible for Perioperative Neurocognitive Disorders. Front Aging Neurosci. 2022;14:772066.CrossRefPubMedPubMedCentral He K, Zhang J, Zhang W, Wang S, Li D, Ma X, Wu X, Chai X, Liu Q. Hippocampus-based mitochondrial respiratory function decline is responsible for Perioperative Neurocognitive Disorders. Front Aging Neurosci. 2022;14:772066.CrossRefPubMedPubMedCentral
6.
go back to reference Smaili SS, Hsu YT, Carvalho AC, Rosenstock TR, Sharpe JC, Youle RJ. Mitochondria, calcium and pro-apoptotic proteins as mediators in cell death signaling. Braz J Med Biol Res. 2003;36(2):183–90.CrossRefPubMed Smaili SS, Hsu YT, Carvalho AC, Rosenstock TR, Sharpe JC, Youle RJ. Mitochondria, calcium and pro-apoptotic proteins as mediators in cell death signaling. Braz J Med Biol Res. 2003;36(2):183–90.CrossRefPubMed
7.
go back to reference Bhatia V, Sharma S. Role of mitochondrial dysfunction, oxidative stress and autophagy in progression of Alzheimer’s disease. J Neurol Sci. 2021;421:117253.CrossRefPubMed Bhatia V, Sharma S. Role of mitochondrial dysfunction, oxidative stress and autophagy in progression of Alzheimer’s disease. J Neurol Sci. 2021;421:117253.CrossRefPubMed
8.
go back to reference An J, Fang Q, Huang C, Qian X, Fan T, Lin Y, Guo Q. Deeper total intravenous anesthesia reduced the incidence of early postoperative cognitive dysfunction after microvascular decompression for facial spasm. J Neurosurg Anesthesiol. 2011;23(1):12–7.CrossRefPubMed An J, Fang Q, Huang C, Qian X, Fan T, Lin Y, Guo Q. Deeper total intravenous anesthesia reduced the incidence of early postoperative cognitive dysfunction after microvascular decompression for facial spasm. J Neurosurg Anesthesiol. 2011;23(1):12–7.CrossRefPubMed
9.
go back to reference Farag E, Chelune GJ, Schubert A, Mascha EJ. Is depth of anesthesia, as assessed by the Bispectral Index, related to postoperative cognitive dysfunction and recovery? Anesth Analg. 2006;103(3):633–40.CrossRefPubMed Farag E, Chelune GJ, Schubert A, Mascha EJ. Is depth of anesthesia, as assessed by the Bispectral Index, related to postoperative cognitive dysfunction and recovery? Anesth Analg. 2006;103(3):633–40.CrossRefPubMed
10.
go back to reference Quan C, Chen J, Luo Y, Zhou L, He X, Liao Y, Chou J, Guo Q, Chen AF, Wen O. BIS-guided deep anesthesia decreases short-term postoperative cognitive dysfunction and peripheral inflammation in elderly patients undergoing abdominal surgery. Brain and behavior. 2019;9(4):e01238.CrossRefPubMedPubMedCentral Quan C, Chen J, Luo Y, Zhou L, He X, Liao Y, Chou J, Guo Q, Chen AF, Wen O. BIS-guided deep anesthesia decreases short-term postoperative cognitive dysfunction and peripheral inflammation in elderly patients undergoing abdominal surgery. Brain and behavior. 2019;9(4):e01238.CrossRefPubMedPubMedCentral
11.
go back to reference Ma J, Williams J, Eastwood D, Lin S, Qian X, Fang Q, Cope D, Yuan Z, Cao L, An J. High-dose Propofol Anesthesia reduces the occurrence of postoperative cognitive dysfunction via maintaining Cytoskeleton. Neuroscience. 2019;421:136–43.CrossRefPubMed Ma J, Williams J, Eastwood D, Lin S, Qian X, Fang Q, Cope D, Yuan Z, Cao L, An J. High-dose Propofol Anesthesia reduces the occurrence of postoperative cognitive dysfunction via maintaining Cytoskeleton. Neuroscience. 2019;421:136–43.CrossRefPubMed
12.
go back to reference Niu K, Qin JL, Lu GF, Guo J, Williams JP, An JX. Dexmedetomidine reverses postoperative spatial memory deficit by targeting Surf1 and cytochrome c. Neuroscience. 2021;466:148–61.CrossRefPubMed Niu K, Qin JL, Lu GF, Guo J, Williams JP, An JX. Dexmedetomidine reverses postoperative spatial memory deficit by targeting Surf1 and cytochrome c. Neuroscience. 2021;466:148–61.CrossRefPubMed
13.
go back to reference Unchiti K, Leurcharusmee P, Samerchua A, Pipanmekaporn T, Chattipakorn N, Chattipakorn SC. The potential role of dexmedetomidine on neuroprotection and its possible mechanisms: evidence from in vitro and in vivo studies. Eur J Neurosci. 2021;54(9):7006–47.CrossRefPubMed Unchiti K, Leurcharusmee P, Samerchua A, Pipanmekaporn T, Chattipakorn N, Chattipakorn SC. The potential role of dexmedetomidine on neuroprotection and its possible mechanisms: evidence from in vitro and in vivo studies. Eur J Neurosci. 2021;54(9):7006–47.CrossRefPubMed
14.
go back to reference Chen J, Shen N, Duan X, Guo Y. An investigation of the mechanism of dexmedetomidine in improving postoperative cognitive dysfunction from the perspectives of alleviating neuronal mitochondrial membrane oxidative stress and electrophysiological dysfunction. Exp Ther Med. 2018;15(2):2037–43.PubMed Chen J, Shen N, Duan X, Guo Y. An investigation of the mechanism of dexmedetomidine in improving postoperative cognitive dysfunction from the perspectives of alleviating neuronal mitochondrial membrane oxidative stress and electrophysiological dysfunction. Exp Ther Med. 2018;15(2):2037–43.PubMed
15.
go back to reference Ghaffary S, Ghaeli P, Talasaz AH, Karimi A, Noroozian M, Salehiomran A, Jalali A. Effect of memantine on post-operative cognitive dysfunction after cardiac surgeries: a randomized clinical trial. Daru. 2017;25(1):24.CrossRefPubMedPubMedCentral Ghaffary S, Ghaeli P, Talasaz AH, Karimi A, Noroozian M, Salehiomran A, Jalali A. Effect of memantine on post-operative cognitive dysfunction after cardiac surgeries: a randomized clinical trial. Daru. 2017;25(1):24.CrossRefPubMedPubMedCentral
16.
go back to reference Andersen SL. Trajectories of brain development: point of vulnerability or window of opportunity? Neurosci Biobehav Rev. 2003;27(1–2):3–18.CrossRefPubMed Andersen SL. Trajectories of brain development: point of vulnerability or window of opportunity? Neurosci Biobehav Rev. 2003;27(1–2):3–18.CrossRefPubMed
17.
go back to reference Hu J, Vacas S, Feng X, Lutrin D, Uchida Y, Lai IK, Maze M. Dexmedetomidine prevents Cognitive decline by enhancing resolution of high mobility Group Box 1 protein-induced inflammation through a Vagomimetic Action in mice. Anesthesiology. 2018;128(5):921–31.CrossRefPubMed Hu J, Vacas S, Feng X, Lutrin D, Uchida Y, Lai IK, Maze M. Dexmedetomidine prevents Cognitive decline by enhancing resolution of high mobility Group Box 1 protein-induced inflammation through a Vagomimetic Action in mice. Anesthesiology. 2018;128(5):921–31.CrossRefPubMed
18.
go back to reference Othman MZ, Hassan Z, Che Has AT. Morris water maze: a versatile and pertinent tool for assessing spatial learning and memory. Exp Anim. 2022;71(3):264–80. Othman MZ, Hassan Z, Che Has AT. Morris water maze: a versatile and pertinent tool for assessing spatial learning and memory. Exp Anim. 2022;71(3):264–80.
19.
go back to reference Li J, Guo M, Liu Y, Wu G, Miao L, Zhang J, Zuo Z, Li Y. Both GSK-3beta/CRMP2 and CDK5/CRMP2 pathways participate in the protection of dexmedetomidine against propofol-induced learning and memory impairment in neonatal rats. Toxicol Sci. 2019 Jun 10:kfz135. Li J, Guo M, Liu Y, Wu G, Miao L, Zhang J, Zuo Z, Li Y. Both GSK-3beta/CRMP2 and CDK5/CRMP2 pathways participate in the protection of dexmedetomidine against propofol-induced learning and memory impairment in neonatal rats. Toxicol Sci. 2019 Jun 10:kfz135.
20.
go back to reference Mathiasen JR, DiCamillo A. Novel object recognition in the rat: a facile assay for cognitive function. Curr Protoc Pharmacol 2010, Chap. 5:Unit 5 59. Mathiasen JR, DiCamillo A. Novel object recognition in the rat: a facile assay for cognitive function. Curr Protoc Pharmacol 2010, Chap. 5:Unit 5 59.
21.
go back to reference Zheng T, Zheng C, Gao F, Huang F, Hu B, Zheng X. Dexmedetomidine suppresses bupivacaine-induced parthanatos in human SH-SY5Y cells via the miR-7-5p/PARP1 axis-mediated ROS. Naunyn Schmiedebergs Arch Pharmacol. 2021;394(4):783–96.CrossRefPubMed Zheng T, Zheng C, Gao F, Huang F, Hu B, Zheng X. Dexmedetomidine suppresses bupivacaine-induced parthanatos in human SH-SY5Y cells via the miR-7-5p/PARP1 axis-mediated ROS. Naunyn Schmiedebergs Arch Pharmacol. 2021;394(4):783–96.CrossRefPubMed
22.
go back to reference O’Toole JF, Patel HV, Naples CJ, Fujioka H, Hoppel CL. Decreased cytochrome c mediates an age-related decline of oxidative phosphorylation in rat kidney mitochondria. Biochem J. 2010;427(1):105–12.CrossRefPubMed O’Toole JF, Patel HV, Naples CJ, Fujioka H, Hoppel CL. Decreased cytochrome c mediates an age-related decline of oxidative phosphorylation in rat kidney mitochondria. Biochem J. 2010;427(1):105–12.CrossRefPubMed
23.
go back to reference Liu Y, Yu S, Xing X, Qiao J, Yin Y, Wang J, Liu M, Zhang W. Ginsenoside Rh2 stimulates the production of mitochondrial reactive oxygen species and induces apoptosis of cervical cancer cells by inhibiting mitochondrial electron transfer chain complex.Mol Med Rep2021, 24(6). Liu Y, Yu S, Xing X, Qiao J, Yin Y, Wang J, Liu M, Zhang W. Ginsenoside Rh2 stimulates the production of mitochondrial reactive oxygen species and induces apoptosis of cervical cancer cells by inhibiting mitochondrial electron transfer chain complex.Mol Med Rep2021, 24(6).
24.
go back to reference Norden DM, Trojanowski PJ, Villanueva E, Navarro E, Godbout JP. Sequential activation of microglia and astrocyte cytokine expression precedes increased Iba-1 or GFAP immunoreactivity following systemic immune challenge. Glia. 2016;64(2):300–16.CrossRefPubMed Norden DM, Trojanowski PJ, Villanueva E, Navarro E, Godbout JP. Sequential activation of microglia and astrocyte cytokine expression precedes increased Iba-1 or GFAP immunoreactivity following systemic immune challenge. Glia. 2016;64(2):300–16.CrossRefPubMed
25.
go back to reference Xiong B, Shi Q, Fang H. Dexmedetomidine alleviates postoperative cognitive dysfunction by inhibiting neuron excitation in aged rats. Am J Transl Res. 2016;8(1):70–80.PubMedPubMedCentral Xiong B, Shi Q, Fang H. Dexmedetomidine alleviates postoperative cognitive dysfunction by inhibiting neuron excitation in aged rats. Am J Transl Res. 2016;8(1):70–80.PubMedPubMedCentral
27.
go back to reference Johnson JA, Ogbi M. Targeting the F1Fo ATP synthase: modulation of the body’s powerhouse and its implications for human disease. Curr Med Chem. 2011;18(30):4684–714.CrossRefPubMed Johnson JA, Ogbi M. Targeting the F1Fo ATP synthase: modulation of the body’s powerhouse and its implications for human disease. Curr Med Chem. 2011;18(30):4684–714.CrossRefPubMed
28.
go back to reference Li J, Zhu X, Yang S, Xu H, Guo M, Yao Y, Huang Z, Lin D. Lidocaine attenuates cognitive impairment after isoflurane anesthesia by reducing mitochondrial damage. Neurochem Res. 2019;44(7):1703–14.CrossRefPubMed Li J, Zhu X, Yang S, Xu H, Guo M, Yao Y, Huang Z, Lin D. Lidocaine attenuates cognitive impairment after isoflurane anesthesia by reducing mitochondrial damage. Neurochem Res. 2019;44(7):1703–14.CrossRefPubMed
29.
go back to reference Reddy PH, Beal MF. Amyloid beta, mitochondrial dysfunction and synaptic damage: implications for cognitive decline in aging and Alzheimer’s disease. Trends Mol Med. 2008;14(2):45–53.CrossRefPubMedPubMedCentral Reddy PH, Beal MF. Amyloid beta, mitochondrial dysfunction and synaptic damage: implications for cognitive decline in aging and Alzheimer’s disease. Trends Mol Med. 2008;14(2):45–53.CrossRefPubMedPubMedCentral
30.
go back to reference Chen Y, Zhang P, Lin X, Zhang H, Miao J, Zhou Y, Chen G. Mitophagy impairment is involved in sevoflurane-induced cognitive dysfunction in aged rats. Aging (Albany NY). 2020;12(17):17235–56. Chen Y, Zhang P, Lin X, Zhang H, Miao J, Zhou Y, Chen G. Mitophagy impairment is involved in sevoflurane-induced cognitive dysfunction in aged rats. Aging (Albany NY). 2020;12(17):17235–56.
31.
go back to reference Bosetti F, Brizzi F, Barogi S, Mancuso M, Siciliano G, Tendi EA, Murri L, Rapoport SI, Solaini G. Cytochrome c oxidase and mitochondrial F1F0-ATPase (ATP synthase) activities in platelets and brain from patients with Alzheimer’s disease. Neurobiol Aging. 2002;23(3):371–6.CrossRefPubMed Bosetti F, Brizzi F, Barogi S, Mancuso M, Siciliano G, Tendi EA, Murri L, Rapoport SI, Solaini G. Cytochrome c oxidase and mitochondrial F1F0-ATPase (ATP synthase) activities in platelets and brain from patients with Alzheimer’s disease. Neurobiol Aging. 2002;23(3):371–6.CrossRefPubMed
32.
go back to reference Esparza-Molto PB, Romero-Carraminana I, Nunez de Arenas C, Pereira MP, Blanco N, Pardo B, Bates GR, Sanchez-Castillo C, Artuch R, Murphy MP, et al. Generation of mitochondrial reactive oxygen species is controlled by ATPase inhibitory factor 1 and regulates cognition. PLoS Biol. 2021;19(5):e3001252.CrossRefPubMedPubMedCentral Esparza-Molto PB, Romero-Carraminana I, Nunez de Arenas C, Pereira MP, Blanco N, Pardo B, Bates GR, Sanchez-Castillo C, Artuch R, Murphy MP, et al. Generation of mitochondrial reactive oxygen species is controlled by ATPase inhibitory factor 1 and regulates cognition. PLoS Biol. 2021;19(5):e3001252.CrossRefPubMedPubMedCentral
33.
go back to reference Terni B, Boada J, Portero-Otin M, Pamplona R, Ferrer I. Mitochondrial ATP-synthase in the entorhinal cortex is a target of oxidative stress at stages I/II of Alzheimer’s disease pathology. Brain Pathol. 2010;20(1):222–33.CrossRefPubMed Terni B, Boada J, Portero-Otin M, Pamplona R, Ferrer I. Mitochondrial ATP-synthase in the entorhinal cortex is a target of oxidative stress at stages I/II of Alzheimer’s disease pathology. Brain Pathol. 2010;20(1):222–33.CrossRefPubMed
34.
go back to reference Li Y, Wen S, Li D, Xie J, Wei X, Li X, Liu Y, Fang H, Yang Y, Lyu J. SURF1 mutations in chinese patients with Leigh syndrome: novel mutations, mutation spectrum, and the functional consequences. Gene. 2018;674:15–24.CrossRefPubMed Li Y, Wen S, Li D, Xie J, Wei X, Li X, Liu Y, Fang H, Yang Y, Lyu J. SURF1 mutations in chinese patients with Leigh syndrome: novel mutations, mutation spectrum, and the functional consequences. Gene. 2018;674:15–24.CrossRefPubMed
35.
go back to reference Cadonic C, Sabbir MG, Albensi BC. Mechanisms of mitochondrial dysfunction in Alzheimer’s Disease. Mol Neurobiol. 2016;53(9):6078–90.CrossRefPubMed Cadonic C, Sabbir MG, Albensi BC. Mechanisms of mitochondrial dysfunction in Alzheimer’s Disease. Mol Neurobiol. 2016;53(9):6078–90.CrossRefPubMed
36.
go back to reference Chandrasekaran K, Hatanpaa K, Brady DR, Stoll J, Rapoport SI. Downregulation of oxidative phosphorylation in Alzheimer disease: loss of cytochrome oxidase subunit mRNA in the hippocampus and entorhinal cortex. Brain Res. 1998;796(1–2):13–9.CrossRefPubMed Chandrasekaran K, Hatanpaa K, Brady DR, Stoll J, Rapoport SI. Downregulation of oxidative phosphorylation in Alzheimer disease: loss of cytochrome oxidase subunit mRNA in the hippocampus and entorhinal cortex. Brain Res. 1998;796(1–2):13–9.CrossRefPubMed
37.
go back to reference Douiev L, Abu-Libdeh B, Saada A. Cytochrome c oxidase deficiency, oxidative stress, possible antioxidant therapy and link to nuclear DNA damage. Eur J Hum Genet. 2018;26(4):579–81.CrossRefPubMedPubMedCentral Douiev L, Abu-Libdeh B, Saada A. Cytochrome c oxidase deficiency, oxidative stress, possible antioxidant therapy and link to nuclear DNA damage. Eur J Hum Genet. 2018;26(4):579–81.CrossRefPubMedPubMedCentral
38.
go back to reference Bhat AH, Dar KB, Anees S, Zargar MA, Masood A, Sofi MA, Ganie SA. Oxidative stress, mitochondrial dysfunction and neurodegenerative diseases; a mechanistic insight. Biomed Pharmacother. 2015;74:101–10.CrossRefPubMed Bhat AH, Dar KB, Anees S, Zargar MA, Masood A, Sofi MA, Ganie SA. Oxidative stress, mitochondrial dysfunction and neurodegenerative diseases; a mechanistic insight. Biomed Pharmacother. 2015;74:101–10.CrossRefPubMed
39.
go back to reference Giridharan VV, Collodel A, Generoso JS, Scaini G, Wassather R, Selvaraj S, Hasbun R, Dal-Pizzol F, Petronilho F, Barichello T. Neuroinflammation trajectories precede cognitive impairment after experimental meningitis-evidence from an in vivo PET study. J Neuroinflammation. 2020;17(1):5.CrossRefPubMedPubMedCentral Giridharan VV, Collodel A, Generoso JS, Scaini G, Wassather R, Selvaraj S, Hasbun R, Dal-Pizzol F, Petronilho F, Barichello T. Neuroinflammation trajectories precede cognitive impairment after experimental meningitis-evidence from an in vivo PET study. J Neuroinflammation. 2020;17(1):5.CrossRefPubMedPubMedCentral
40.
go back to reference Ghosh C, Mukherjee S, Dey SG. Direct electron transfer between Cyt c and heme-abeta relevant to Alzheimer’s disease. Chem Commun (Camb). 2013;49(51):5754–6.CrossRefPubMed Ghosh C, Mukherjee S, Dey SG. Direct electron transfer between Cyt c and heme-abeta relevant to Alzheimer’s disease. Chem Commun (Camb). 2013;49(51):5754–6.CrossRefPubMed
41.
go back to reference Gouveia A, Bajwa E, Klegeris A. Extracellular cytochrome c as an intercellular signaling molecule regulating microglial functions. Biochim Biophys Acta Gen Subj. 2017;1861(9):2274–81.CrossRefPubMed Gouveia A, Bajwa E, Klegeris A. Extracellular cytochrome c as an intercellular signaling molecule regulating microglial functions. Biochim Biophys Acta Gen Subj. 2017;1861(9):2274–81.CrossRefPubMed
42.
go back to reference Hu Z, Yu F, Gong P, Qiu Y, Zhou W, Cui Y, Li J, Chen H. Subneurotoxic copper(II)-induced NF-kappaB-dependent microglial activation is associated with mitochondrial ROS. Toxicol Appl Pharmacol. 2014;276(2):95–103.CrossRefPubMed Hu Z, Yu F, Gong P, Qiu Y, Zhou W, Cui Y, Li J, Chen H. Subneurotoxic copper(II)-induced NF-kappaB-dependent microglial activation is associated with mitochondrial ROS. Toxicol Appl Pharmacol. 2014;276(2):95–103.CrossRefPubMed
43.
go back to reference Bader V, Winklhofer KF. Mitochondria at the interface between neurodegeneration and neuroinflammation. Semin Cell Dev Biol. 2020;99:163–71.CrossRefPubMed Bader V, Winklhofer KF. Mitochondria at the interface between neurodegeneration and neuroinflammation. Semin Cell Dev Biol. 2020;99:163–71.CrossRefPubMed
Metadata
Title
Dexmedetomidine attenuates postoperative spatial memory impairment after surgery by reducing cytochrome C
Authors
Lina Sun
Kun Niu
Jian Guo
Jingru Tu
Baofeng Ma
Jianxiong An
Publication date
01-12-2023
Publisher
BioMed Central
Published in
BMC Anesthesiology / Issue 1/2023
Electronic ISSN: 1471-2253
DOI
https://doi.org/10.1186/s12871-023-02035-x

Other articles of this Issue 1/2023

BMC Anesthesiology 1/2023 Go to the issue