Home Propofol ameliorates ischemic brain injury by blocking TLR4 pathway in mice
Article Open Access

Propofol ameliorates ischemic brain injury by blocking TLR4 pathway in mice

  • Kazuha Mitsui , Masakazu Kotoda EMAIL logo , Sohei Hishiyama , Ayasa Takamino , Sho Morikawa , Tadahiko Ishiyama and Takashi Matsukawa
Published/Copyright: September 1, 2022
Become an author with De Gruyter Brill

Abstract

Ischemic brain injury is one of the most serious perioperative complications. However, effective preventative methods have not yet been established. This study aimed to investigate whether propofol has neuroprotective effects against ischemic brain injury, with a specific focus on Toll-like receptor 4 (TLR4). Focal brain ischemia was induced via a combination of left common carotid artery occlusion and distal left middle cerebral artery coagulation in mice. Either propofol (10 mg/kg) or vehicle was intravenously injected 10 min prior to the induction of brain ischemia in wild-type and TLR4 knockout mice. Infarct volume, pro-inflammatory cytokine expression, inflammatory cell infiltration, and neurobehavioral function were assessed. Propofol administration significantly reduced infarct volume in wild-type mice (26.9 ± 2.7 vs 15.7 ± 2.0 mm3 at day 7), but not in TLR4 knockout mice. Compared with the control mice, the propofol-treated wild-type mice exhibited lower levels of IL-6 (0.57 ± 0.23 vs 1.00 ± 0.39 at 24 h), and smaller numbers of TLR4-expressing microglia in the penumbra (11.7 ± 3.1 vs 25.1 ± 4.7 cells/0.1 mm2). In conclusion, propofol administration prior to ischemic brain insult attenuated brain injury by blocking the TLR4-dependent pathway and suppressing pro-inflammatory cytokine production.

1 Background

Ischemic brain injury is a serious disease that compromises people’s lives and health [1] and is one of the most concerning perioperative complications. There are various causes including hypotension, hypoxia, arrhythmia, systemic inflammation, and blood loss due to surgery/anesthesia that jeopardize cerebral circulation. Surprisingly, in recent studies using magnetic resonance imaging, the incidence of ischemic brain injury associated with noncardiac surgery was as high as 10% in patients with cardiovascular risk factors [2], and thrombotic stroke is the most common type of this complication [3]. The occurrence of ischemic brain injury largely impedes recovery from surgery and is associated with an eightfold increase in perioperative mortality [4]. Furthermore, a recent prospective cohort study indicates that even covert ischemic brain injuries significantly increase the risk of long-term cognitive decline and can impair quality of life [5].

However, promptly detecting acute ischemic brain injury is sometimes challenging due to the remaining anesthetic effects or the use of postoperative opioids and sedative drugs. Consequently, only a limited number of patients receive reperfusion therapy during the acute phase. Therefore, preventative strategies for minimizing ischemic brain injury are vital. In addition, the effects of commonly used anesthetic drugs on the ischemic brain and its underlying mechanisms are relevant to research targets. Given these considerations, continuous efforts have been made in recent decades to elucidate the effects of anesthetic drugs on ischemic brain injury, and volatile anesthetic drugs have been shown to exert preconditioning and neuroprotective effect via activation of mitochondrial adenosine triphosphate-sensitive potassium channels [6,7]. Some earlier studies using animal models of ischemic stroke or cell culture have also suggested neuroprotective effects of propofol [8,9,10]. However, the evidence is still limited and controversial [11], and the mechanisms underlying the possible neuroprotective effects of propofol remain unclear. Several recent studies suggest that propofol blocks Toll-like receptor 4 (TLR4) is the key mediator of inflammation and suppresses inflammatory cytokine production [12,13,14]. Given that inflammation is a crucial factor in secondary injury after brain ischemia and that TLR4 plays a pivotal role in the inflammatory response in the ischemic brain [15,16], the TLR4-dependent pathway may be involved in the possible neuroprotective effects of propofol against ischemic brain injury.

Thus, we tested the hypothesis that propofol attenuates ischemic brain injury via inhibition of the TLR4-dependent pathway and suppression of consequent inflammatory cytokine production. The primary outcome of this study was infarct volume; the secondary outcomes were pro-inflammatory cytokine mRNA expression levels, the number of TLR4-expressing microglia in the penumbra, and neurological function.

2 Materials and methods

2.1 Animals

Male C57BL/6 mice (8 weeks old) were purchased from Japan SLC (Tokyo, Japan). Two breeding pairs of TLR4 knockout mice with a C57BL/6 genetic background were purchased from Oriental Bio Service (Kyoto, Japan), and their male offspring mice (10–14 weeks old) were used in the study. All mice were group-housed at 23°C ± 2°C with free access to standard food and water and a 12-h light/dark cycle. All experiments were performed between 09:00 and 17:00 under normal room light and temperature (23°C ± 2°C) conditions.

  1. Ethical approval: All experiments were conducted in accordance with the National Institutes of Health guidelines for the care and use of laboratory animals. The experimental protocol was reviewed and approved by the University of Yamanashi Animal Care Committee.

2.2 Ischemic brain injury

Focal brain ischemia was induced via a combination of permanent left common carotid artery occlusion and distal left middle cerebral artery (MCA) coagulation [17]. Briefly, mice were anesthetized with 2–3% isoflurane and placed in a dorsal position. The left common carotid artery was isolated and ligated via ventral middle neck incision. Mice were then placed in the lateral position, and a 2 mm burr-hole craniectomy was performed with a microdrill (Ideal Microdrill; Bio Research, Nagoya, Japan) between the left orbit and the left ear. The distal left MCA was exposed and coagulated using a small vessel cauterizer (Fine Science Tools, Inc., CA, USA) followed by a transection of the artery. During the surgery, rectal temperature was maintained at 37°C ± 0.5°C with a thermostat-regulated heating pad. Brains were removed 24 h after the induction of ischemic brain injury for real-time polymerase chain reaction (PCR) or immunofluorescence or at 7 days for measurement of infarct volume.

2.3 Propofol treatment

Propofol (10 mg/kg, 1% Diprivan, Aspen Japan, Tokyo, Japan) diluted with fat emulsion by 10 times (10 µL/g, Intralipos, Otsuka Pharmaceutical, Tokyo, Japan) was administered via the tail vein 10 min prior to MCA occlusion. In control mice, an equal volume of the fat emulsion alone was administered. The dose was chosen based on an earlier study, in which propofol produced neuroprotection against ischemic brain injury in mice [18].

2.4 Measurement of infarct volume

Seven days after MCA occlusion and after hemodynamic measurements, mice were deeply anesthetized with 5% isoflurane and euthanized via cervical dislocation. Brains were removed and cut into 1-mm-thick coronal sections. The brain slices were immersed in 2% 2,3,5-triphenyltetrazolium chloride (Sigma Aldrich, St. Louis, MO, USA) at 37°C for 15 min in a dark room. The infarct area was traced and measured using image analysis software (ImageJ, National Institutes of Health, Bethesda, MD, USA) by an individual who was blinded to the grouping and study design. To correct for the contribution of edema, the infarct area was calculated as follows: total ipsilateral hemisphere – infarct region [19]. Total infarct volume was calculated as the sum of all infarct areas multiplied by section thickness.

2.5 Hemodynamic measurements

Heart rate and arterial blood pressure were measured non-invasively using a tail-cuff monitor (Softron, Tokyo, Japan) to evaluate the effects of propofol on hemodynamics. Values were recorded 1 h before MCA occlusion (baseline), 10 min after the injection of either propofol or the control solution, 1 h after MCA occlusion, and 7 days after MCA occlusion.

2.6 Blood gas analysis

Blood gas analysis was performed to evaluate pH, PCO2, PO2, and glucose levels (i-STAT 300 F, Abbot Co., Abbot Park, IL, USA). Blood samples were collected before induction of MCA occlusion (baseline), 1 h after the injection of either propofol or the control solution, and 7 days after MCA occlusion.

2.7 Real-time PCR

Real-time PCR was used to measure mRNA expression levels of interleukin (IL)-6, IL-1β, and tumor necrosis factor α (TNF-α). The brain was the target mRNAs from the brain slices of propofol-treated wild-type mice that were compared with those of control wild-type mice. The RNeasy Mini Kit (Qiagen, Hilden, Germany) was used for the extraction of total mRNA from brain slices. About 1µg of mRNA was reverse transcribed into complementary DNA with a QuantiTect Reverse Transcription Kit (Qiagen, Hilden, Germany). PCR was performed on a StepOneTM real-time PCR system (Life Technologies, Carlsbad, CA, USA) using the PowerSYBR® Green PCR Master Mix and corresponding primers to quantify target genes (Table S1). The relative changes were expressed as a ratio to glyceraldehyde-3-phosphate dehydrogenase (GAPDH) mRNAs of the same sample. The data were analyzed using the 2−ΔΔCT method. The 2−ΔΔCT value of the target transcript from each mouse was normalized with those of the control group mice as 1.0. The normalized 2−ΔΔCT values derived from the two groups were then compared.

2.8 Immunofluorescence

The mouse brains were fixed with 4% paraformaldehyde in phosphate-buffered saline (PBS) for 6 days and cut into 1mm-thick sections using a slicer (BD80 HS, Leica, Bensheim, Germany). Prior to immunofluorescence, CUBIC tissue clearing [20] was performed to identify ischemic core and surrounding penumbra. The brain sections were washed three times with PBS and then immersed in 50% CUBIC-L (T3740, Tokyo Chemical Industry, Tokyo, Japan) overnight with shaking at 40°C, followed by immersing in 100% CUBIC-L with shaking at 40°C for 3 days, replacing 100% CUBIC-L with new solution every day. Then, the brain sections were washed three times with PBS and stained with 0.5% TritonX-100 (12967-32, NACALAI TESQUE, INC., Kyoto, Japan) in PBS and the following primary antibodies for 5 days in the dark with shaking at room temperature: FITC-conjugated rat anti-TLR4 (1:25, sc-13591, Santa Cruz, CA, USA) and Alexa Fluor 594-conjugated rat anti-CD11b (1:100, 101254, BioLegend, CA, USA). The stained-brain sections were washed three times with PBS and then post-fixed with 1% formaldehyde in PBS overnight. After washing twice with PBS, to complete CUBIC tissue clearing, the brain sections were immersed in 50% CUBIC-R (T3741) overnight followed by 100% CUBIC-R overnight with shaking at room temperature. Finally, the sections were photographed using a confocal microscope (A1R HD25, Nikon, Tokyo, Japan).

TLR4 + CD11b + double-positive cells in the penumbra, defined as the region immediately adjacent to the infarct area, were counted in 400× magnification microscopic fields. For each animal, ten 0.1 mm2 areas within the cortex were randomly chosen, and the number of TLR4 + CD11b + double-positive cells was automatically counted using Image J software. Ten readouts per animal were averaged.

2.9 Neurological evaluation

Neurological function was assessed using the neurological deficit scores (0: no deficit; 1: flexion of the torso; 2: spontaneous circling; 3: longitudinal circling or leaning; 4: no spontaneous movement; 5: death), lateral push test, and body asymmetry test at baseline and 7 days after the induction of ischemic brain injury. The lateral push test and body asymmetry test were performed as previously described [21,22].

2.10 Statistical analysis

Statistical analysis was performed using Prism 8 software (GraphPad Software, San Diego, CA, USA). The collected data were assessed for the normal distribution and equal variance using the Shapiro–Wilk and F test, respectively. Infarct volumes, numbers of TLR4 + CD11b + double-positive cells, and body asymmetry test results were analyzed using two-tailed Student’s t test; cytokine expression levels and neurological deficit scores were analyzed using Mann–Whitney U test, based on the results of the Shapiro–Wilk and F tests. Heart rate, blood pressure, and blood gas parameters (pH, PCO2, PO2, and glucose) were analyzed using the two-way analysis of variance for repeated measures; the lateral push test results were analyzed using Chi-square test. For infarct volumes and cytokine expression levels, the sample size was calculated to detect significance with 95% confidence, assuming alpha of 0.05 and power of 0.8 (G*Power 3.1.9.3). Values are presented as mean ± standard error of mean for the infarct volumes and numbers of cells, as median ± quantile for cytokine mRNA expression levels, and as mean ± standard deviation for heart rate, blood pressure, and blood gas parameters. p-values of less than 0.05 were considered statistically significant.

3 Results

3.1 Propofol reduced cerebral infarct volume

To investigate the effects of propofol on ischemic brain injury, 8- to 9-week-old wild-type mice were treated with either propofol or 10% fat emulsion (control) 10 min before the induction of ischemic brain injury. Propofol-treated mice exhibited significantly smaller infarct volumes than control mice 7 days after ischemic brain injury (26.9 ± 2.7 vs 15.7 ± 2.0 mm3, n  =  10 each, p  <0.05, Figure 1). Propofol treatment did not affect the hemodynamic and blood gas parameters and neurological function (neurological deficit scores, baseline and day 7: 0.0 ± 0.0 vs 0.0 ± 0.0, p  > 0.99; lateral push test, baseline and day 7: 0.0 ± 0.0 vs 0.0 ± 0.0, p  > 0.99; body asymmetry test, baseline: 6.0 ± 5.4 vs 1.0 ± 6.4, p  = 0.56, day 7: 9.0 ± 12.5 vs 26.0 ± 9.1, p  = 0.29, all n  =  10 each, Table 1).

Figure 1 
                  Effect of propofol pretreatment on infarct volume: (a) Staining for 2,3,5-triphenyltetrazolium chloride in representative 1-mm-thick coronal sections. Arrowheads indicate the infarct area (white). (b) Infarct volumes 7 days after induction of ischemic brain injury. Mice in the propofol group exhibited smaller infarct volumes.
Figure 1

Effect of propofol pretreatment on infarct volume: (a) Staining for 2,3,5-triphenyltetrazolium chloride in representative 1-mm-thick coronal sections. Arrowheads indicate the infarct area (white). (b) Infarct volumes 7 days after induction of ischemic brain injury. Mice in the propofol group exhibited smaller infarct volumes.

Table 1

Results of hemodynamic measurements and blood gas analysis

    Wild-type + vehicle Wild-type + propofol TLR4KO + vehicle TLR4KO + propofol
Heart rate (bpm) Baseline 443 ± 54 423 ± 36 445 ± 64 465 ± 50
  10 min Before MCAO 428 ± 29 448 ± 63 388 ± 26 367 ± 40
  1 h After MCAO 442 ± 77 429 ± 84 375 ± 27 395 ± 20
  7 Days after MCAO 473 ± 74 468 ± 64 421 ± 37 454 ± 41
Mean blood pressure (mmHg) Baseline 77 ± 17 82 ± 9 79 ± 8 80 ± 8
  10 min Before MCAO 74 ± 12 77 ± 9 81 ± 8 74 ± 12
  1 h After MCAO 87 ± 14 76 ± 12 83 ± 4 80 ± 16
  7 Days after MCAO 86 ± 8 80 ± 10 85 ± 8 83 ± 16
pH Baseline 7.34 ± 0.06 7.28 ± 0.06 7.33 ± 0.05 7.33 ± 0.06
  1 h After MCAO 7.30 ± 0.01 7.31 ± 0.04 7.30 ± 0.04 7.31 ± 0.04
  7 Days after MCAO 7.30 ± 0.05 7.35 ± 0.02 7.34 ± 0.02 7.33 ± 0.03
PCO2 (mmHg) Baseline 34.8 ± 5.7 38.9 ± 4.7 34.6 ± 1.4 36.4 ± 4.6
  1 h After MCAO 45.4 ± 1.2 44.2 ± 3.2 45.0 ± 3.8 46.3 ± 9.2
  7 Days after MCAO 44.0 ± 3.3 39.6 ± 6.3 41.6 ± 3.8 38.8 ± 5.9
PO2 (mmHg) Baseline 59.4 ± 8.8 70.6 ± 10.9 64.0 ± 4.6 63.8 ± 5.5
  1 h After MCAO 64.0 ± 13.0 64.0 ± 6.7 70.4 ± 6.3 68.2 ± 5.4
  7 days After MCAO 53.6 ± 7.9 55.0 ± 6.4 64.2 ± 9.9 61.0 ± 5.1
Glucose (mg/dL) Baseline 252.6 ± 46.8 264.6 ± 35.1 257.8 ± 67.5 255.8 ± 15.0
  1 h After MCAO 251.0 ± 42.6 204.2 ± 40.5 246.2 ± 33.5 229.8 ± 19.8
  7 Days after MCAO 254.8 ± 31.3 227.2 ± 22.2 209.4 ± 62.4 191.2 ± 42.2

There were no significant differences in heart rate, blood pressure, and blood gas parameters among groups. (All parameters: p > 0.05.). Data were presented as mean ± SD. MCAO, middle cerebral artery occlusion; TLR4KO, toll-like receptor 4 knockout.

3.2 Propofol treatment reduced pro-inflammatory cytokine expressions after ischemic brain injury

To investigate the possible involvement of anti-inflammatory action of propofol in the neuroprotective effect observed, mRNA expression of pro-inflammatory cytokines was measured 24 h after ischemic brain injury. As shown in Figure 2, the propofol-treated mice exhibited lower mRNA expression levels of pro-inflammatory cytokines (IL-6: 0.57 ±  0.23 vs 1.00 ±  0.39, p  <  0.05, IL-1β: 0.53 ±  0.24 vs 1.00  ±  0.36, p = 0.087, n  =  15 each) compared with the control mice.

Figure 2 
                  Assessment of pro-inflammatory expression levels 24 h after the induction of ischemic brain injury. The 2−ΔΔCT value of the target transcript from each mouse was normalized with the median 2−ΔΔCT value from the control wild-type mice as 1.0. Mice in the propofol group exhibited lower levels of inflammatory cytokine expression.
Figure 2

Assessment of pro-inflammatory expression levels 24 h after the induction of ischemic brain injury. The 2−ΔΔCT value of the target transcript from each mouse was normalized with the median 2−ΔΔCT value from the control wild-type mice as 1.0. Mice in the propofol group exhibited lower levels of inflammatory cytokine expression.

3.3 Propofol decreased the number of TLR4-expressing microglia in the penumbra

Immunofluorescence targeting TLR4 and microglia was performed to assess whether the propofol treatment influenced TLR4 expression or infiltration of microglia in the penumbra. As shown in Figure 3, mice treated with propofol had smaller numbers of TLR4-expressing microglia in the penumbra, compared with the control mice (11.7 ± 3.1 vs 25.1 ± 4.7 cells/0.1 mm2, n = 10 each, p  <  0.05).

Figure 3 
                  Assessment of the number of TLR4-expressing microglia in the penumbra: (a) the representative image of the brain slice after CUBIC tissue cleaning. The red arrowheads indicate the penumbra, defined as the region immediately adjacent to the infarct area (encircled with white dashed line), used for immunofluorescence assay. (b) Immunofluorescence staining of DAPI (blue), TLR4 (green), CD11b-positive microglia (red), and merged images were presented. The white arrowheads indicate TLR4 + CD11b + double-positive cells in the merged images. (c) Compared with the control mice, the propofol-treated mice had significantly smaller numbers of TLR4-expressing microglia in the penumbra.
Figure 3

Assessment of the number of TLR4-expressing microglia in the penumbra: (a) the representative image of the brain slice after CUBIC tissue cleaning. The red arrowheads indicate the penumbra, defined as the region immediately adjacent to the infarct area (encircled with white dashed line), used for immunofluorescence assay. (b) Immunofluorescence staining of DAPI (blue), TLR4 (green), CD11b-positive microglia (red), and merged images were presented. The white arrowheads indicate TLR4 + CD11b + double-positive cells in the merged images. (c) Compared with the control mice, the propofol-treated mice had significantly smaller numbers of TLR4-expressing microglia in the penumbra.

3.4 Neuroprotective effects of propofol were abolished by depletion of the TLR4-dependent pathway

In experiments testing the effects of propofol treatment against ischemic brain injury in TLR4 knockout mice, there was no significant difference in the infarct volume between the propofol-treated TLR4 knockout mice and control-treated TLR4 knockout mice (25.6 ± 3.8 vs 26.5 ± 3.1 mm3, n = 10 each, p = 0.91, Figure 4). Similar to the experiments using wild-type mice, hemodynamic and blood gas parameters and neurological function were not significantly different between the groups (neurological deficit scores, baseline and day 7: 0.0 ± 0.0 vs 0.0 ± 0.0, p  > 0.99; lateral push test, baseline and day 7: 0.0 ± 0.0 vs 0.0 ± 0.0, p  > 0.99; body asymmetry test, baseline: 7.5 ± 3.1 vs 11.1 ± 8.9, p  = 0.72, 7 day after: −10.0 ± 6.5 vs −1.1 ± 2.0, p  = 0.19, all n = 10 each, Table 1).

Figure 4 
                  Effect of propofol on infarct volumes in Toll-like receptor-4 (TLR4) knockout mice. Infarct volumes were assessed 7 days after the induction of ischemic brain injury. There was no significant difference in infarct volumes between the propofol-treated TLR4 knockout mice and control-treated TLR4 knockout mice.
Figure 4

Effect of propofol on infarct volumes in Toll-like receptor-4 (TLR4) knockout mice. Infarct volumes were assessed 7 days after the induction of ischemic brain injury. There was no significant difference in infarct volumes between the propofol-treated TLR4 knockout mice and control-treated TLR4 knockout mice.

4 Discussion

In the present study, we found that propofol significantly reduced infarct volume after ischemic brain insult in wild-type mice but not in TLR4-knockout mice. The reduced infarct volume was associated with reduced pro-inflammatory cytokine expressions and TLR4-expresing microglia in the penumbra. These results indicated that propofol exerts neuroprotective effects via its anti-inflammatory capacity, specifically the inhibitory effect on TLR4. Since the hemodynamic and blood gas parameters were not affected by the propofol administration, the effects of propofol on cardiovascular and respiratory function and blood sugar levels were not likely to be involved in the neuroprotective effects of propofol observed in the present study.

The immune system is promptly activated once ischemic brain injury occurs [23]. Among various inflammatory mediators, TLR is considered crucial in innate immune system as a first-line defense and mediator of inflammation [24,25]. Exogenous/endogenous TLR ligands such as heat shock proteins, fibrinogen, and components of the extracellular matrix are upregulated by ischemic brain insult. Consequently, those ligands then activate TLR4 [26,27], the TLR shown to be the pivotal inflammatory mediator in the pathogenesis of ischemic brain injury [15,16]. It has been reported that while activation of TLR4 exacerbates cerebral infarction [15], inhibition of the receptor suppresses pro-inflammatory responses and attenuates brain injury [16].

In the current study, the neuroprotective effect of propofol against ischemic brain injury was abrogated by depletion of TLR4, indicating that the TLR4-dependent pathway was substantially involved in the mechanism underlying that neuroprotective effect. The results of the current study are concordant with recent in vivo studies in which propofol suppressed inflammatory cytokine production via inhibition of TLR4-dependent pathways in various disease models, including lung [12], liver [13], and gastric injuries [14], and asthma [28]. Several in vitro studies using lipopolysaccharide-treated microglia [29], macrophages [30], spinal astrocytes [31], and alveolar epithelial cells [32] also suggest that the anti-inflammatory action of propofol involves blocking the TLR4-dependent pathway and consequent pro-inflammatory cytokine production.

Our result showed significantly lower IL-6 mRNA expression levels in the brains of propofol-treated wild-type mice than in the brains of control mice, which is consistent with an earlier study in which there was a correlation between infarct volume and IL-6 mRNA expression in the brain [33] Among various pro-inflammatory cytokines, IL-6 plays pivotal roles in local inflammation and cytotoxicity after ischemic brain injury and is involved in the mechanism underlying the expansion of ischemic brain injury [34,35]. Blockade of IL-6 receptors has been shown to reduce infarct volume and improve cognitive function in an experimental model of ischemic stroke [36]. These earlier studies are concordant with the suppression of IL-6 by propofol observed in the current study after ischemic brain insult, as well as the reduction of infarct volume.

Our immunofluorescence study demonstrated that the pre-treatment with propofol reduced the number of TLR4-expressing microglia in the penumbra. Recent studies have reported that microglia are involved in the exacerbation of cerebral infarction [37,38,39]. TLR4-expressing microglia release pro-inflammatory cytokines [37], which causes inflammation in the penumbra and aggravates the brain injury. On the other hand, propofol has been shown to inhibit TLR4 upregulation in microglia [40]. Another previous study reported that propofol has neuroprotective effects against ischemic stroke by suppressing microglia [41]. Our real-time PCR and immunofluorescence results demonstrated that the propofol treatment decreased the expression level of IL-6 mRNA and the number of TLR4-expressing microglia after cerebral ischemia. Based on the collective results of the previous studies and the current study, it is reasonable to surmise that propofol exerts neuroprotection against ischemic brain injury by blocking TLR4 and suppressing consequent production of pro-inflammatory cytokines, particularly IL-6, from microglia in the penumbra.

If ischemic brain injury occurs during general anesthesia, the consequences are tragic. The clinical importance of the present study is that propofol administration prior to ischemic insult may have the potential to protect against ischemic brain injury, presumably by blocking the TLR4-dependent pathway in microglia in the penumbra. This understanding of the mechanism underlying the neuroprotective effect of propofol against ischemic brain injury may lead to a novel strategy to prevent exacerbation of ischemic brain injury during general anesthesia.

This study has some limitations. First, we did not find neurological improvement in the propofol-treated mice, possibly because of the relatively small infarct volumes and low sensitivities of the neurological assays we used in the current study. Although infarct volume is clinically relevant and was the primary outcome of this study, further studies with more sensitive neurobehavioral evaluation are needed to elucidate the effects of propofol on neurobehavioral function after ischemic brain injury. Second, we only used relatively young male mice. The neuroprotective effects of propofol should be tested in aged mice and female mice at different menopausal stages because those biological variables can affect the outcomes of ischemic brain injury. Third, we used only GAPDH as a reference gene for the real-time PCR analysis. Although this gene is widely used as a reference gene for evaluating inflammatory responses after ischemic stroke in mice, it is indicated that hypoxia upregulates GAPDH mRNA expression [42], which could have influenced the interpretation of the data. Lastly, we unexpectedly observed similar infarct sizes between TLR4 KO and wild-type mice. This observation is not consistent with the findings of previous studies that reported reduced infarct size by TLR4 gene knockout [43,44]. This may be explained by the compensatory upregulation of other related genes following a genetic depletion of a certain gene [45,46]. The TLR4 knockout could have induced the upregulation of other inflammatory pathways, such as TLR2 pathway, at least in our model. Another possible explanation would be that the first experiment (wild-type mice with/without propofol) and the second experiment (TLR4 knockout mice with/without propofol) were separately conducted. The ages of the mice used in the experiments were different (WT: 8 weeks old; TLR4 KO: 10–14 weeks old due to the breeding capacity). The wild-type animals were purchased, while the knockout mice were bred in our laboratory. Therefore, the infarct volume data from these two experiments might not be directly comparable. In this study, propofol pretreatment significantly attenuated brain infarction in the WT mice, but not in the TLR4 KO mice, indicating the involvement of the TLR4 pathway in the neuroprotective effects of propofol.

5 Conclusions

Propofol administration prior to ischemic brain insult attenuated brain injury by blocking the TRL4-dependent pathway and suppressing pro-inflammatory cytokine production. This understanding of the mechanism underlying the neuroprotective effect of propofol against ischemic brain injury may lead to a new strategy to prevent exacerbation of cerebral infarction during general anesthesia.

List of abbreviations

IL-1β

interleukin-1β

IL-6

interleukin-6

MCA

middle cerebral artery

PCR

polymerase chain reaction

TLR4

Toll-like receptor 4

TNF-α

tumor necrosis factor-α

Acknowledgment

We thank Masako Abe for her technical assistance.

  1. Funding information: This work was supported by Japan Society for the Promotion of Science (JSPS KAKENHI Grant Number 20K09238). The funding bodies had no role in the design of the study, the collection, analysis, and interpretation of data, or the writing of the manuscript.

  2. Author contributions: KM conceived the study, acquired, analyzed, and interpreted the data, and drafted the manuscript. MK conceived the study, performed experiments and statistical analysis, and drafted the manuscript. SH, AT, and SM performed experiments and helped prepare the manuscript. TI and TM performed statistical analysis and revised the manuscript for important intellectual content. All authors have read and approved the final manuscript.

  3. Conflict of interest: The authors state no conflict of interest.

  4. Data availability statement: The data sets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

References

[1] Chen L, Han Z, Gu J. Early path nursing on neurological function recovery of cerebral infarction. Transl Neurosci. 2019;10:160–3.10.1515/tnsci-2019-0028Search in Google Scholar

[2] Mashour GA, Moore LE, Lele AV, Robicsek SA, Gelb AW. Perioperative care of patients at high risk for stroke during or after non-cardiac, non-neurologic surgery: consensus statement from the society for neuroscience in anesthesiology and critical care*. J Neurosurg Anesthesiol. 2014;26:273–85.10.1097/ANA.0000000000000087Search in Google Scholar

[3] Ng JL, Chan MT, Gelb AW. Perioperative stroke in noncardiac, nonneurosurgical surgery. Anesthesiology. 2011;115:879–90.10.1097/ALN.0b013e31822e9499Search in Google Scholar

[4] Sanders RD, Jørgensen ME, Mashour GA. Perioperative stroke: a question of timing? Br J Anaesth. 2015;115:11–3.10.1093/bja/aev031Search in Google Scholar

[5] Neuro VI. Perioperative covert stroke in patients undergoing non-cardiac surgery (NeuroVISION): A prospective cohort study. Lancet. 2019;394:1022–9.10.1016/S0140-6736(19)31795-7Search in Google Scholar

[6] Kehl F, Payne RS, Roewer N, Schurr A. Sevoflurane-induced preconditioning of rat brain in vitro and the role of KATP channels. Brain Res. 2004;1021:76–81.10.1016/j.brainres.2004.06.038Search in Google Scholar PubMed

[7] Ren X, Wang Z, Ma H, Zuo Z. Sevoflurane postconditioning provides neuroprotection against brain hypoxia-ischemia in neonatal rats. Neurol Sci. 2014;35:1401–4.10.1007/s10072-014-1726-4Search in Google Scholar PubMed

[8] Xi H-J, Zhang T-H, Tao T, Song C-Y, Lu S-J, Cui X-G, et al. Propofol improved neurobehavioral outcome of cerebral ischemia-reperfusion rats by regulating Bcl-2 and Bax expression. Brain Res. 2011;1410:24–32.10.1016/j.brainres.2011.06.060Search in Google Scholar PubMed

[9] Wang H, Luo M, Li C, Wang G. Propofol post-conditioning induced long-term neuroprotection and reduced internalization of AMPAR GluR2 subunit in a rat model of focal cerebral ischemia/reperfusion. J Neurochem. 2011;119:210–9.10.1111/j.1471-4159.2011.07400.xSearch in Google Scholar PubMed

[10] Wang G, Yu Y, Wang Y. Effects of propofol on neuroblastoma cells via the HOTAIRM1/miR-519a-3p axis. Transl Neurosci. 2022;13:57–69.10.1515/tnsci-2022-0212Search in Google Scholar PubMed PubMed Central

[11] Zhang C-Y, Du J, Zhang R, Jin J, Qiao L-Y. Erythropoietin attenuates propofol-induced hippocampal neuronal cell injury in developing rats by inhibiting toll-like receptor 4 expression. Neurosci Lett. 2020;716:134647.10.1016/j.neulet.2019.134647Search in Google Scholar PubMed

[12] Wang X, Liu C, Wang G. Propofol protects rats and human alveolar epithelial cells against lipopolysaccharide-induced acute lung injury via inhibiting HMGB1 expression. Inflammation. 2016;39:1004–16.10.1007/s10753-016-0330-6Search in Google Scholar PubMed

[13] Wu G-J, Lin Y-W, Chuang C-Y, Tsai H-C, Chen R-M. Liver nitrosation and inflammation in septic rats were suppressed by propofol via downregulating TLR4/NF-κB-mediated iNOS and IL-6 gene expressions. Life Sci. 2018;195:25–32.10.1016/j.lfs.2018.01.005Search in Google Scholar PubMed

[14] Ye H-H, Wu K-J, Fei S-J, Zhang X-W, Liu H-X, Zhang J-L, et al. Propofol participates in gastric mucosal protection through inhibiting the toll-like receptor-4/nuclear factor kappa-B signaling pathway. Clin Res Hepatol Gastroenterol. 2013;37:e3–e15.10.1016/j.clinre.2012.03.004Search in Google Scholar PubMed

[15] Marsh BJ, Williams-Karnesky RL, Stenzel-Poore MP. Toll-like receptor signaling in endogenous neuroprotection and stroke. Neuroscience. 2009;158:1007–20.10.1016/j.neuroscience.2008.07.067Search in Google Scholar PubMed PubMed Central

[16] Andresen L, Theodorou K, Grünewald S, Czech-Zechmeister B, Könnecke B, Lühder F, et al. Evaluation of the therapeutic potential of Anti-TLR4-Antibody MTS510 in experimental stroke and significance of different routes of application. PLoS One. 2016;11:e0148428.10.1371/journal.pone.0148428Search in Google Scholar PubMed PubMed Central

[17] Munji RN, Soung AL, Weiner GA, Sohet F, Semple BD, Trivedi A, et al. Profiling the mouse brain endothelial transcriptome in health and disease models reveals a core blood-brain barrier dysfunction module. Nat Neurosci. 2019;22:1892–902.10.1038/s41593-019-0497-xSearch in Google Scholar PubMed PubMed Central

[18] Kotani Y, Nakajima Y, Hasegawa T, Satoh M, Nagase H, Shimazawa M, et al. Propofol exerts greater neuroprotection with disodium edetate than without it. J Cereb Blood Flow Metab. 2008;28:354–66.10.1038/sj.jcbfm.9600532Search in Google Scholar PubMed

[19] Llovera G, Roth S, Plesnila N, Veltkamp R, Liesz A. Modeling stroke in mice: permanent coagulation of the distal middle cerebral artery. J Visualized Exp. 2014;89:e51729.10.3791/51729Search in Google Scholar PubMed PubMed Central

[20] Susaki EA, Shimizu C, Kuno A, Tainaka K, Li X, Nishi K, et al. Versatile whole-organ/body staining and imaging based on electrolyte-gel properties of biological tissues. Nat Commun. 2020;11:1982.10.1038/s41467-020-15906-5Search in Google Scholar PubMed PubMed Central

[21] Rehni AK, N, Singh. Role of phosphoinositide 3-kinase in ischemic postconditioning-induced attenuation of cerebral ischemia-evoked behavioral deficits in mice. Pharmacol Rep. 2007;59:192–8.Search in Google Scholar

[22] Zhu W, Fan Y, Frenzel T, Gasmi M, Bartus RT, Young WL, et al. Insulin growth factor-1 gene transfer enhances neurovascular remodeling and improves long-term stroke outcome in mice. Stroke. 2008;39:1254–61.10.1161/STROKEAHA.107.500801Search in Google Scholar PubMed PubMed Central

[23] Lambertsen KL, Biber K, Finsen B. Inflammatory cytokines in experimental and human stroke. J Cereb Blood Flow Metab. 2012;32:1677–98.10.1038/jcbfm.2012.88Search in Google Scholar PubMed PubMed Central

[24] Fitzgerald KA, Kagan JC, Toll-like receptors and the control of immunity. Cell. 2020;180:1044–66.10.1016/j.cell.2020.02.041Search in Google Scholar PubMed PubMed Central

[25] Doyle KP, Simon RP, Stenzel-Poore MP. Mechanisms of ischemic brain damage. Neuropharmacology. 2008;55:310–8.10.1016/j.neuropharm.2008.01.005Search in Google Scholar PubMed PubMed Central

[26] Ziegler G, Harhausen D, Schepers C, Hoffmann O, Röhr C, Prinz V, et al. TLR2 has a detrimental role in mouse transient focal cerebral ischemia. Biochem Biophys Res Commun. 2007;359:574–9.10.1016/j.bbrc.2007.05.157Search in Google Scholar PubMed

[27] Cao C-X, Yang Q-W, Lv F-L, Cui J, Fu H-B, Wang J-Z. Reduced cerebral ischemia-reperfusion injury in Toll-like receptor 4 deficient mice. Biochem Biophys Res Commun. 2007;353:509–14.10.1016/j.bbrc.2006.12.057Search in Google Scholar PubMed

[28] Li H-Y, Meng J-X, Liu Z, Liu X-W, Huang Y-G, Zhao J. Propofol attenuates airway inflammation in a mast cell-dependent mouse model of allergic asthma by inhibiting the toll-like receptor 4/reactive oxygen species/nuclear factor κB signaling pathway. Inflammation. 2018;41:914–23.10.1007/s10753-018-0746-2Search in Google Scholar PubMed

[29] Gui B, Su M, Chen J, Jin L, Wan R, Qian Y. Neuroprotective effects of pretreatment with propofol in LPS-induced BV-2 microglia cells: role of TLR4 and GSK-3β. Inflammation. 2012;35:1632–40.10.1007/s10753-012-9478-xSearch in Google Scholar PubMed

[30] Wu G-J, Chen T-L, Chang C-C, Chen R-M. Propofol suppresses tumor necrosis factor-alpha biosynthesis in lipopolysaccharide-stimulated macrophages possibly through downregulation of nuclear factor-kappa B-mediated toll-like receptor 4 gene expression. Chem Biol Interact. 2009;180:465–71.10.1016/j.cbi.2009.05.003Search in Google Scholar PubMed

[31] Zhou C-H, Zhu Y-Z, Zhao P-P, Xu C-M, Zhang M-X, Huang H, et al. Propofol inhibits lipopolysaccharide-induced inflammatory responses in spinal astrocytes via the toll-like receptor 4/MyD88-dependent nuclear factor-κB, extracellular signal-regulated protein kinases 1/2, and p38 mitogen-activated protein kinase pathways. Anesth Analg. 2015;120:1361–8.10.1213/ANE.0000000000000645Search in Google Scholar PubMed

[32] Ma L, Wu X, Chen W, Fujino Y. Propofol has anti-inflammatory effects on alveolar type II epithelial cells. Acta Anaesthesiol Scand. 2010;54:362–9.10.1111/j.1399-6576.2009.02127.xSearch in Google Scholar

[33] Gredal H, Thomsen BB, Boza-Serrano A, Garosi L, Rusbridge C, Anthony D, et al. Interleukin-6 is increased in plasma and cerebrospinal fluid of community-dwelling domestic dogs with acute ischaemic stroke. Neuroreport. 2017;28:134–40.10.1097/WNR.0000000000000728Search in Google Scholar

[34] Armstead WM, Hekierski H, Pastor P, Yarovoi S, Higazi AA-R, Cines DB. Release of IL-6 after stroke contributes to impaired cerebral autoregulation and hippocampal neuronal necrosis through NMDA receptor activation and upregulation of ET-1 and JNK. Transl Stroke Res. 2019;10:104–11.10.1007/s12975-018-0617-zSearch in Google Scholar

[35] Cojocaru IM, Cojocaru M, Tănăsescu R, Iliescu I, Dumitrescu L, Silosi I. Expression of IL-6 activity in patients with acute ischemic stroke. Rom J Intern Med. 2009;47:393–6.Search in Google Scholar

[36] Wei J, Sun C, Liu C, Zhang Q. Effects of rat anti-mouse interleukin-6 receptor antibody on the recovery of cognitive function in stroke mice. Cell Mol Neurobiol. 2018;38:507–15.10.1007/s10571-017-0499-8Search in Google Scholar

[37] Lambertsen KL, Biber K, Finsen B. Inflammatory cytokines in experimental and human stroke. J Cereb Blood Flow Metab. 2012;32:1677–98.10.1038/jcbfm.2012.88Search in Google Scholar

[38] Sun M, Deng B, Zhao X, Gao C, Yang L, Zhao H, et al. Isoflurane preconditioning provides neuroprotection against stroke by regulating the expression of the TLR4 signalling pathway to alleviate microglial activation. Sci Rep. 2015;5:11445.10.1038/srep11445Search in Google Scholar

[39] Xu S, Lu J, Shao A, Zhang JH, Zhang J. Glial cells: Role of the immune response in ischemic stroke. Front Immunology. 2020;11:294.10.3389/fimmu.2020.00294Search in Google Scholar

[40] Qin X, Sun ZQ, Zhang XW, Dai XJ, Mao SS, Zhang YM. TLR4 signaling is involved in the protective effect of propofol in BV2 microglia against OGD/reoxygenation. J Physiol Biochem. 2013;69:707–18.10.1007/s13105-013-0247-6Search in Google Scholar

[41] Zhou R, Yang Z, Tang X, Tan Y, Wu X, Liu F. Propofol protects against focal cerebral ischemia via inhibition of microglia-mediated proinflammatory cytokines in a rat model of experimental stroke. PLoS One. 2013;8:e82729.10.1371/journal.pone.0082729Search in Google Scholar

[42] Yamaji R, Fujita K, Takahashi S, Yoneda H, Nagao K, Masuda W, et al. Hypoxia up-regulates glyceraldehyde-3-phosphate dehydrogenase in mouse brain capillary endothelial cells: involvement of Na+/Ca2+ exchanger. Biochim Biophys Acta. 2003;1593:269–76.10.1016/S0167-4889(02)00397-XSearch in Google Scholar

[43] Hyakkoku K, Hamanaka J, Tsuruma K, Shimazawa M, Tanaka H, Uematsu S, et al. Toll-like receptor 4 (TLR4), but not TLR3 or TLR9, knock-out mice have neuroprotective effects against focal cerebral ischemia. Neuroscience. 2010;171:258–67.10.1016/j.neuroscience.2010.08.054Search in Google Scholar PubMed

[44] Pushkov D, Nicholson JD, Michowiz S, Novitzky I, Weiss S, Ben Hemou M, et al. Relative neuroprotective effects hyperbaric oxygen treatment and TLR4 knockout in a mouse model of temporary middle cerebral artery occlusion. Int J Neurosci. 2016;126:174–81.10.3109/00207454.2014.1002609Search in Google Scholar PubMed

[45] Ma Z, Zhu P, Shi H, Guo L, Zhang Q, Chen Y, et al. PTC-bearing mRNA elicits a genetic compensation response via Upf3a and COMPASS components. Nature. 2019;568:259–63.10.1038/s41586-019-1057-ySearch in Google Scholar PubMed

[46] El-Brolosy MA, Kontarakis Z, Rossi A, Kuenne C, Günther S, Fukuda N, et al. Genetic compensation triggered by mutant mRNA degradation. Nature. 2019;568:193–7.10.1038/s41586-019-1064-zSearch in Google Scholar PubMed PubMed Central

Received: 2022-05-24
Revised: 2022-07-21
Accepted: 2022-08-01
Published Online: 2022-09-01

© 2022 Kazuha Mitsui et al., published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Articles in the same Issue

  1. Research Articles
  2. Mice can recognise water depths and will avoid entering deep water
  3. CLSPCOL rescues Alzheimer’s disease mouse models
  4. Ischemic stroke following STA–MCA double bypass
  5. Relationship between subclasses low-density lipoprotein and carotid plaque
  6. Construction and effect evaluation of different sciatic nerve injury models in rats
  7. Effects of propofol on neuroblastoma cells via the HOTAIRM1/miR-519a-3p axis
  8. Effects of Goldblatt hypertension on rats’ hippocampal cholinergic system
  9. Longitudinal PET studies of mGluR5 in FXS using an FMR1 knockout mouse model
  10. Molecular recognition of the interaction between ApoE and the TREM2 protein
  11. Remimazolam induced cognitive dysfunction in mice via glutamate excitotoxicity
  12. Hybrid 18F-florbetapir PET/MRI for assessing myelin recovery in GFAP-A patients
  13. Preliminary study on pathogenic mechanism of first Chinese family with PNKD
  14. Aged rats learn Morris Water maze using non-spatial search strategies evidenced by a parameter-based algorithm
  15. Embelin and levodopa combination therapy for improved Parkinson’s disease treatment
  16. The roles and diagnostic value of miRNA-1246 in the serum of patients with intracranial aneurysms
  17. Predictors of high dose of massed practice following stroke
  18. The effect of stimulation frequency on transcranial evoked potentials
  19. Increased levels of neurofilament light chain in suicide attempters’ serum
  20. Propofol ameliorates ischemic brain injury by blocking TLR4 pathway in mice
  21. Single-cell analysis of gene expression in the substantia nigra pars compacta of a pesticide-induced mouse model of Parkinson’s disease
  22. Psychobiotics improve propionic acid-induced neuroinflammation in juvenile rats, rodent model of autism
  23. Isoliquiritigenin attenuates neuroinflammation in mice model of Parkinson’s disease by promoting Nrf2/NQO-1 pathway
  24. Ketamine alleviating depressive-like behaviors is associated with regulation of nNOS–CAPON–Dexras1 complex in chronic unpredictable mild stress rats
  25. The association of anxiety and other clinical features with CACNA1C rs1006737 in patients with depression
  26. Outcomes of acute ischemic stroke in kidney transplant recipients: An analysis of US Nationwide inpatient sample
  27. Brain atrophy pattern in patients with mild cognitive impairment: MRI study
  28. Correlation of serum RBP4 level with oxidative stress and unstable carotid plaque in patients with cerebral infarction
  29. Prevalence of neurodegenerative/demyelinating disorders in patients with achalasia
  30. Protection against H2O2-evoked toxicity in HT22 hippocampal neuronal cells by geissoschizine methyl ether via inhibiting ERK pathway
  31. Alteration in NMDAR subunits in different brain regions of chronic unpredictable mild stress (CUMS) rat model
  32. Acacetin improves cognitive function of APP/PS1 Alzheimer’s disease model mice via the NLRP3 inflammasome signaling pathway
  33. Assessment of malformations, variations and diameters of vessels forming the circle of Willis – An autopsy study in a non-cerebrovascular cohort
  34. Singular versus combinatory glucose-sensitive signal control of metabolic sensor protein profiles in hypothalamic astrocyte cultures from each sex
  35. Mild acute stress prevents the memory impairment induced by long-term isoflurane anesthesia
  36. Esketamine improves propofol-induced brain injury and cognitive impairment in rats
  37. Correlations between EEG and intestinal electrical stimulation
  38. Factors influencing recovery of upper limb motor function during constraint-induced movement therapy for people with stroke
  39. Application of auditory mismatch negativity in tinnitus patients based on high-resolution electroencephalogram signals
  40. The value of transcranial Doppler monitoring of cerebral blood flow changes during carotid endarterectomy performed under regional anesthesia – A case series
  41. Acacetin alleviates neuroinflammation and oxidative stress injury via the Nrf2/HO-1 pathway in a mouse model of spinal cord injury
  42. Pre-ischemic exercise prevents inflammation and apoptosis by inhibiting MAPK pathway in ischemic stroke
  43. Fronto-parietal single-trial brain connectivity benefits successful memory recognition
  44. Review Articles
  45. Machine learning approaches for diagnosing depression using EEG: A review
  46. Perinatal diet and offspring anxiety: A scoping review
  47. Brain and gut microbiota disorders in the psychopathology of anorexia nervosa
  48. Nanotechnology-based drug delivery for the treatment of CNS disorders
  49. Case Reports
  50. A rare giant intracranial arachnoid cyst confused the diagnosis and treatment of Wilson disease
  51. Cerebral infarction following bee stings: Case report and literature review
  52. Neurosarcoidosis presenting as longitudinally extensive myelitis: Diagnostic assessment, differential diagnosis, and therapeutic approach
  53. Long-acting paliperidone in Ekbom’s syndrome in Lewy body dementia: A case report
  54. Patients with post-COVID-19 vaccination facial palsy: To boost or not to boost?
  55. Autosomal dominant sleep-related hypermotor epilepsy associated with a novel mutation of KCNT1
  56. A rare brainstem anaplastic astrocytoma
  57. A singular case of hyposmia and transient audiovestibular post-vaccine disorders: case report and literature review
  58. A rare lymphoplasmacyte-rich meningioma involving the dura of the skull base and cervical spinal cord: A case report
  59. Communication
  60. The system’s genetics of depression and its somatic and mental comorbidities
  61. Commentary
  62. The possible association between neurodegenerative/demyelinating neurological disorders in achalasia patients
  63. Letter to the Editor
  64. A patient with hereditary transthyretin amyloidosis involving multiple cranial nerves due to a rare p.(Phe84Ser) variant
  65. Erratum
  66. Erratum to “Spinocerebellar ataxia type 40: A case report and literature review”
  67. Erratum to “Construction and effect evaluation of different sciatic nerve injury models in rats”
  68. Retraction
  69. Nervonic acid amends motor disorder in a mouse model of Parkinson’s disease
  70. Retraction of “Isovitexin modulates autophagy in Alzheimer’s disease via miR-107 signalling”
Downloaded on 3.11.2025 from https://www.degruyterbrill.com/document/doi/10.1515/tnsci-2022-0238/html
Scroll to top button