Skip to main content
Top
Published in: Neurocritical Care 3/2019

01-06-2019 | Original Article

Effect of Electrical Vagus Nerve Stimulation on Cerebral Blood Flow and Neurological Outcome in Asphyxial Cardiac Arrest Model of Rats

Authors: Byunghyun Kim, Inwon Park, Jae Hyuk Lee, Seonghye Kim, Min Ji Lee, You Hwan Jo

Published in: Neurocritical Care | Issue 3/2019

Login to get access

Abstract

Background

Vagus nerve stimulation (VNS) during post-resuscitation may increase recovery of cerebral blood flow (CBF) and reduce neurological injury.

Objective

This study was designed to investigate the effect of electrical VNS on neurological outcomes following cardiac arrest (CA).

Methods

Male Sprague–Dawley rats (n = 48) were subjected to the asphyxial CA model and blindly allocated to the VN isolation (CA + VN isolation) or VNS group (CA + VNS group). Cardiopulmonary resuscitation was initiated 450 s after pulseless electrical arrest, and the left cervical vagus nerve was electrically stimulated (0.05 mA, 1 Hz) for 3 h in the CA + VNS group. The neurological deficit score (NDS) and overall performance category (OPC) were assessed at 24 h after resuscitation, and histological injury of the hippocampus was evaluated. Independent experiments were performed to evaluate the effect of VNS on global cortical CBF after resuscitation using laser speckle Doppler imaging through a thinned skull window from pre-arrest to 6 h after resuscitation.

Results

The baseline characteristics were not significantly different between the two groups. The NDS was significantly higher, and the OPC was substantially lower in the CA + VNS group (p = 0.022 and p = 0.049, respectively) supported by decrease in histological injury of the hippocampal CA1 region. CBF in the early period of post-return of spontaneous circulation (ROSC) was significantly higher in the CA + VNS group (p < 0.05 at post-ROSC 2 h and 4 h), and 4-hydroxynonenal was significantly lower in the CA + VNS group (p = 0.026).

Conclusions

VNS improved cerebral perfusion and neurological outcomes at 24 h after ROSC in an asphyxial CA model of rats.
Appendix
Available only for authorised users
Literature
1.
go back to reference Nolan JP, Neumar RW, Adrie C, et al. Post-cardiac arrest syndrome: epidemiology, pathophysiology, treatment, and prognostication. A Scientific Statement from the International Liaison Committee on Resuscitation; the American Heart Association Emergency Cardiovascular Care Committee; the Council on Cardiovascular Surgery and Anesthesia; the Council on Cardiopulmonary, Perioperative, and Critical Care; the Council on Clinical Cardiology; the Council on Stroke. Resuscitation. 2008;79(3):350–79.CrossRefPubMed Nolan JP, Neumar RW, Adrie C, et al. Post-cardiac arrest syndrome: epidemiology, pathophysiology, treatment, and prognostication. A Scientific Statement from the International Liaison Committee on Resuscitation; the American Heart Association Emergency Cardiovascular Care Committee; the Council on Cardiovascular Surgery and Anesthesia; the Council on Cardiopulmonary, Perioperative, and Critical Care; the Council on Clinical Cardiology; the Council on Stroke. Resuscitation. 2008;79(3):350–79.CrossRefPubMed
2.
go back to reference Mangus DB, Huang L, Applegate PM, et al. A systematic review of neuroprotective strategies after cardiac arrest: from bench to bedside (part I—protection via specific pathways). Med Gas Res. 2014;4:9.CrossRefPubMedPubMedCentral Mangus DB, Huang L, Applegate PM, et al. A systematic review of neuroprotective strategies after cardiac arrest: from bench to bedside (part I—protection via specific pathways). Med Gas Res. 2014;4:9.CrossRefPubMedPubMedCentral
3.
go back to reference Huang L, Applegate PM, Gatling JW, et al. A systematic review of neuroprotective strategies after cardiac arrest: from bench to bedside (part II—comprehensive protection). Med Gas Res. 2014;4:10.CrossRefPubMedPubMedCentral Huang L, Applegate PM, Gatling JW, et al. A systematic review of neuroprotective strategies after cardiac arrest: from bench to bedside (part II—comprehensive protection). Med Gas Res. 2014;4:10.CrossRefPubMedPubMedCentral
4.
go back to reference Nielsen N, Wetterslev J, Cronberg T, et al. Targeted temperature management at 33 degrees C versus 36 degrees C after cardiac arrest. N Engl J Med. 2013;369(23):2197–206.CrossRef Nielsen N, Wetterslev J, Cronberg T, et al. Targeted temperature management at 33 degrees C versus 36 degrees C after cardiac arrest. N Engl J Med. 2013;369(23):2197–206.CrossRef
5.
go back to reference Bernard SA, Gray TW, Buist MD, et al. Treatment of comatose survivors of out-of-hospital cardiac arrest with induced hypothermia. N Engl J Med. 2002;346(8):557–63.CrossRef Bernard SA, Gray TW, Buist MD, et al. Treatment of comatose survivors of out-of-hospital cardiac arrest with induced hypothermia. N Engl J Med. 2002;346(8):557–63.CrossRef
6.
go back to reference Hypothermia after Cardiac Arrest Study Group. Mild therapeutic hypothermia to improve the neurologic outcome after cardiac arrest. N Engl J Med. 2002;346(8):549–56.CrossRef Hypothermia after Cardiac Arrest Study Group. Mild therapeutic hypothermia to improve the neurologic outcome after cardiac arrest. N Engl J Med. 2002;346(8):549–56.CrossRef
7.
8.
go back to reference van den Brule JM, Vinke EJ, van Loon LM, van der Hoeven JG, Hoedemaekers CW. Low spontaneous variability in cerebral blood flow velocity in non-survivors after cardiac arrest. Resuscitation. 2017;111:110–5.CrossRefPubMed van den Brule JM, Vinke EJ, van Loon LM, van der Hoeven JG, Hoedemaekers CW. Low spontaneous variability in cerebral blood flow velocity in non-survivors after cardiac arrest. Resuscitation. 2017;111:110–5.CrossRefPubMed
9.
go back to reference Chen WL, Tsai TH, Huang CC, Chen JH, Kuo CD. Heart rate variability predicts short-term outcome for successfully resuscitated patients with out-of-hospital cardiac arrest. Resuscitation. 2009;80(10):1114–8.CrossRefPubMed Chen WL, Tsai TH, Huang CC, Chen JH, Kuo CD. Heart rate variability predicts short-term outcome for successfully resuscitated patients with out-of-hospital cardiac arrest. Resuscitation. 2009;80(10):1114–8.CrossRefPubMed
10.
go back to reference Borovikova LV, Ivanova S, Zhang M, et al. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature. 2000;405(6785):458–62.CrossRefPubMed Borovikova LV, Ivanova S, Zhang M, et al. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature. 2000;405(6785):458–62.CrossRefPubMed
11.
go back to reference Jiang Y, Li L, Liu B, et al. Vagus nerve stimulation attenuates cerebral ischemia and reperfusion injury via endogenous cholinergic pathway in rat. PLoS ONE. 2014;9(7):e102342.CrossRefPubMedPubMedCentral Jiang Y, Li L, Liu B, et al. Vagus nerve stimulation attenuates cerebral ischemia and reperfusion injury via endogenous cholinergic pathway in rat. PLoS ONE. 2014;9(7):e102342.CrossRefPubMedPubMedCentral
12.
go back to reference Ay I, Nasser R, Simon B, Ay H. Transcutaneous cervical vagus nerve stimulation ameliorates acute ischemic injury in rats. Brain Stimul. 2016;9(2):166–73.CrossRef Ay I, Nasser R, Simon B, Ay H. Transcutaneous cervical vagus nerve stimulation ameliorates acute ischemic injury in rats. Brain Stimul. 2016;9(2):166–73.CrossRef
13.
go back to reference Jiang Y, Li L, Ma J, et al. Auricular vagus nerve stimulation promotes functional recovery and enhances the post-ischemic angiogenic response in an ischemia/reperfusion rat model. Neurochem Int. 2016;97:73–82.CrossRefPubMed Jiang Y, Li L, Ma J, et al. Auricular vagus nerve stimulation promotes functional recovery and enhances the post-ischemic angiogenic response in an ischemia/reperfusion rat model. Neurochem Int. 2016;97:73–82.CrossRefPubMed
14.
go back to reference Zhang L, Ma J, Jin X, et al. L-PGDS mediates vagus nerve stimulation-induced neuroprotection in a rat model of ischemic stroke by suppressing the apoptotic response. Neurochem Res. 2017;42(2):644–55.CrossRefPubMed Zhang L, Ma J, Jin X, et al. L-PGDS mediates vagus nerve stimulation-induced neuroprotection in a rat model of ischemic stroke by suppressing the apoptotic response. Neurochem Res. 2017;42(2):644–55.CrossRefPubMed
15.
go back to reference Sun P, Wang J, Zhao S, et al. Improved outcomes of cardiopulmonary resuscitation in rats treated with vagus nerve stimulation and its potential mechanism. Shock. 2018;49(6):698–703.CrossRefPubMed Sun P, Wang J, Zhao S, et al. Improved outcomes of cardiopulmonary resuscitation in rats treated with vagus nerve stimulation and its potential mechanism. Shock. 2018;49(6):698–703.CrossRefPubMed
16.
go back to reference Zobel A, Joe A, Freymann N, et al. Changes in regional cerebral blood flow by therapeutic vagus nerve stimulation in depression: an exploratory approach. Psychiatry Res. 2005;139(3):165–79.CrossRefPubMed Zobel A, Joe A, Freymann N, et al. Changes in regional cerebral blood flow by therapeutic vagus nerve stimulation in depression: an exploratory approach. Psychiatry Res. 2005;139(3):165–79.CrossRefPubMed
18.
go back to reference National Research Council (US) Committee for the Update of the Guide for the Care and Use of Laboratory Animals. Guide for the Care and Use of Laboratory Animals. 8th ed. Washington, DC: National Academies Press; 2011. National Research Council (US) Committee for the Update of the Guide for the Care and Use of Laboratory Animals. Guide for the Care and Use of Laboratory Animals. 8th ed. Washington, DC: National Academies Press; 2011.
19.
go back to reference Lee JH, Kim K, Jo YH, et al. Effect of valproic acid on survival and neurologic outcomes in an asphyxial cardiac arrest model of rats. Resuscitation. 2013;84(10):1443–9.CrossRefPubMed Lee JH, Kim K, Jo YH, et al. Effect of valproic acid on survival and neurologic outcomes in an asphyxial cardiac arrest model of rats. Resuscitation. 2013;84(10):1443–9.CrossRefPubMed
20.
go back to reference Jia X, Koenig MA, Shin HC, et al. Improving neurological outcomes post-cardiac arrest in a rat model: immediate hypothermia and quantitative EEG monitoring. Resuscitation. 2008;76(3):431–42.CrossRefPubMed Jia X, Koenig MA, Shin HC, et al. Improving neurological outcomes post-cardiac arrest in a rat model: immediate hypothermia and quantitative EEG monitoring. Resuscitation. 2008;76(3):431–42.CrossRefPubMed
21.
go back to reference Winship IR. Laser speckle contrast imaging to measure changes in cerebral blood flow. Methods Mol Biol. 2014;1135:223–35.CrossRefPubMed Winship IR. Laser speckle contrast imaging to measure changes in cerebral blood flow. Methods Mol Biol. 2014;1135:223–35.CrossRefPubMed
22.
go back to reference Buunk G, van der Hoeven JG, Meinders AE. Cerebral blood flow after cardiac arrest. Neth J Med. 2000;57(3):106–12.CrossRefPubMed Buunk G, van der Hoeven JG, Meinders AE. Cerebral blood flow after cardiac arrest. Neth J Med. 2000;57(3):106–12.CrossRefPubMed
23.
go back to reference He J, Lu H, Young L, et al. Real-time quantitative monitoring of cerebral blood flow by laser speckle contrast imaging after cardiac arrest with targeted temperature management. J Cereb Blood Flow Metab. 2017;2017:271678X17748787. He J, Lu H, Young L, et al. Real-time quantitative monitoring of cerebral blood flow by laser speckle contrast imaging after cardiac arrest with targeted temperature management. J Cereb Blood Flow Metab. 2017;2017:271678X17748787.
24.
go back to reference Manole MD, Kochanek PM, Bayir H, et al. Brain tissue oxygen monitoring identifies cortical hypoxia and thalamic hyperoxia after experimental cardiac arrest in rats. Pediatr Res. 2014;75(2):295–301.CrossRefPubMed Manole MD, Kochanek PM, Bayir H, et al. Brain tissue oxygen monitoring identifies cortical hypoxia and thalamic hyperoxia after experimental cardiac arrest in rats. Pediatr Res. 2014;75(2):295–301.CrossRefPubMed
25.
go back to reference Henry TR, Votaw JR, Pennell PB, et al. Acute blood flow changes and efficacy of vagus nerve stimulation in partial epilepsy. Neurology. 1999;52(6):1166–73.CrossRefPubMed Henry TR, Votaw JR, Pennell PB, et al. Acute blood flow changes and efficacy of vagus nerve stimulation in partial epilepsy. Neurology. 1999;52(6):1166–73.CrossRefPubMed
26.
go back to reference Pardo JV, Sheikh SA, Schwindt GC, et al. Chronic vagus nerve stimulation for treatment-resistant depression decreases resting ventromedial prefrontal glucose metabolism. Neuroimage. 2008;42(2):879–89.CrossRefPubMedPubMedCentral Pardo JV, Sheikh SA, Schwindt GC, et al. Chronic vagus nerve stimulation for treatment-resistant depression decreases resting ventromedial prefrontal glucose metabolism. Neuroimage. 2008;42(2):879–89.CrossRefPubMedPubMedCentral
27.
go back to reference Rutecki P. Anatomical, physiological, and theoretical basis for the antiepileptic effect of vagus nerve stimulation. Epilepsia. 1990;31(Suppl 2):S1–6.CrossRefPubMed Rutecki P. Anatomical, physiological, and theoretical basis for the antiepileptic effect of vagus nerve stimulation. Epilepsia. 1990;31(Suppl 2):S1–6.CrossRefPubMed
28.
go back to reference Parada E, Egea J, Buendia I, et al. The microglial alpha7-acetylcholine nicotinic receptor is a key element in promoting neuroprotection by inducing heme oxygenase-1 via nuclear factor erythroid-2-related factor 2. Antioxid Redox Signal. 2013;19(11):1135–48.CrossRefPubMedPubMedCentral Parada E, Egea J, Buendia I, et al. The microglial alpha7-acetylcholine nicotinic receptor is a key element in promoting neuroprotection by inducing heme oxygenase-1 via nuclear factor erythroid-2-related factor 2. Antioxid Redox Signal. 2013;19(11):1135–48.CrossRefPubMedPubMedCentral
29.
go back to reference Miyamoto O, Pang J, Sumitani K, et al. Mechanisms of the anti-ischemic effect of vagus nerve stimulation in the gerbil hippocampus. NeuroReport. 2003;14(15):1971–4.CrossRefPubMed Miyamoto O, Pang J, Sumitani K, et al. Mechanisms of the anti-ischemic effect of vagus nerve stimulation in the gerbil hippocampus. NeuroReport. 2003;14(15):1971–4.CrossRefPubMed
30.
go back to reference Follesa P, Biggio F, Gorini G, et al. Vagus nerve stimulation increases norepinephrine concentration and the gene expression of BDNF and bFGF in the rat brain. Brain Res. 2007;1179:28–34.CrossRefPubMed Follesa P, Biggio F, Gorini G, et al. Vagus nerve stimulation increases norepinephrine concentration and the gene expression of BDNF and bFGF in the rat brain. Brain Res. 2007;1179:28–34.CrossRefPubMed
31.
go back to reference Kim C, Fahrenbruch CE, Cobb LA, Eisenberg MS. Out-of-hospital cardiac arrest in men and women. Circulation. 2001;104(22):2699–703.CrossRefPubMed Kim C, Fahrenbruch CE, Cobb LA, Eisenberg MS. Out-of-hospital cardiac arrest in men and women. Circulation. 2001;104(22):2699–703.CrossRefPubMed
Metadata
Title
Effect of Electrical Vagus Nerve Stimulation on Cerebral Blood Flow and Neurological Outcome in Asphyxial Cardiac Arrest Model of Rats
Authors
Byunghyun Kim
Inwon Park
Jae Hyuk Lee
Seonghye Kim
Min Ji Lee
You Hwan Jo
Publication date
01-06-2019
Publisher
Springer US
Published in
Neurocritical Care / Issue 3/2019
Print ISSN: 1541-6933
Electronic ISSN: 1556-0961
DOI
https://doi.org/10.1007/s12028-018-0640-7

Other articles of this Issue 3/2019

Neurocritical Care 3/2019 Go to the issue

Neurocritical Care Through History

Stomaching Acute Brain Injury