Open Access

Isoflurane reduces septic neuron injury by HO‑1‑mediated abatement of inflammation and apoptosis

  • Authors:
    • Lina Zhang
    • Xuece Zhang
    • Ting Wu
    • Xu Pan
    • Zhi Wang
  • View Affiliations

  • Published online on: December 22, 2020     https://doi.org/10.3892/mmr.2020.11794
  • Article Number: 155
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Sepsis‑associated encephalopathy (SAE) frequently occurs in critically ill patients with severe systemic infections. Subanesthetic isoflurane (0.7% ISO) possesses anti‑inflammatory, antioxidant and anti‑apoptotic properties against a number of human diseases, including brain injury. The activation of heme oxygenase‑1 (HO‑1) impedes inflammation, oxidation and apoptosis, thus alleviating sepsis‑induced brain damage. However, whether 0.7% ISO affords protection against septic neuronal injury involving HO‑1 activation is unclear. The present study aimed to investigate the neuroprotective effects of 0.7% ISO and its potential underlying mechanisms in SAE using a mouse model established by cecal ligation and puncture (CLP). The results indicated that the expression and activity of HO‑1 in the mouse hippocampus were increased by CLP, and further enhanced by ISO. ISO reduced the death rate, brain water content and blood‑brain barrier disruption, but improved the learning and memory functions of CLP‑treated mice. ISO significantly decreased the production of pro‑inflammatory cytokines and the levels of oxidative indictors in the serum and hippocampus, as well as the number of apoptotic neurons and the expression of pro‑apoptotic proteins in the hippocampus. Inversely, anti‑inflammatory factors, antioxidative enzymes and anti‑apoptotic proteins were markedly increased by ISO administration. However, the neuroprotective effects of ISO were abolished by a HO‑1 inhibitor. Overall, these findings suggested that 0.7% ISO alleviated SAE via its anti‑inflammatory, antioxidative and anti‑apoptotic properties, which involved the activated form of HO‑1.

Introduction

Sepsis is a severe systemic inflammatory response syndrome caused by a dysregulated host response to infection (1), and is the leading cause of morbidity and mortality in critically ill patients worldwide (2). Multiple complications occur during sepsis, such as acute lung injury (3), kidney lesions (4), cardiac dysfunction (5), liver disorders (6) and brain damage (7). Among these manifestations, sepsis-associated encephalopathy (SAE) appears earlier and more frequently than other complications (8). Despite advancements in the treatment of patients in the intensive care unit (ICU), individuals with SAE exhibit longer ICU stays and higher 28-day mortality rates (9), with survivors experiencing a substantial cognitive deficit (10). Learning and memory impairments have also been observed in septic mammalian models (11,12). Thus, identifying the pathogenic mechanisms of SAE and developing more effective therapeutic strategies are critical.

Inflammatory lesions, oxidative damage and cell death are well associated with SAE (13,14). Inflammatory triggers, such as lipopolysaccharide (LPS), can bind to Toll-like receptor 4 (TLR4) and subsequent activate downstream signals of TLR4, such as mitogen-activated protein kinases (MAPKs) and nuclear factor-κB (15,16), and untimely induce the production of adhesion molecules and proinflammatory factors, including tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6 in vascular endothelial cells (17), which can result in endothelial abnormalities, leakage of the blood-brain barrier (BBB), immune cell migration, neuronal death, and ultimately brain damage (18,19). In addition, intracellular calcium is involved in LPS-stimulated cytokine production; calcium influx due to calcium store depletion is referred to as store-operated calcium entry (20). Calcium signaling involves activation of tyrosine kinases and leads to activation of MAPKs in endothelial cells (21). Reportedly, oxidative stress triggers neuronal injury and cerebral dysfunction during sepsis (22,23). Therefore, the alleviation of inflammation, oxidative stress and apoptosis would be an efficacious remedy for SAE. Heme oxygenases (HOs) are the rate-limiting enzymes that convert heme into biliverdin, ferrous iron and carbon monoxide. Heme oxygenase-1 (HO-1), the inducible isoform of HO, confers cytoprotection by exerting anti-inflammatory, antioxidative and anti-apoptotic functions (2426). HO-1 upregulation and activation ameliorates sepsis-induced multiorgan injury, including lung, kidney, liver and brain damage (2730). Therefore, HO-1-targeted treatments are a promising avenue for neuroprotection during sepsis.

Isoflurane (ISO) is a widely used inhalational anesthetic during surgery. Mounting evidence suggests that besides its use as an anesthetic, ISO possesses non-anesthetic physiologic properties against inflammation, oxidation and apoptosis (31,32). ISO affords neuroprotection against hypoxic ischemia-induced cerebral damage (33,34), and ISO post-treatment improves pulmonary vascular permeability in septic rats by upregulating HO-1 (27). ISO elicits its anti-inflammatory activity by activating the HO-1 pathway in LPS-stimulated macrophages (35). ISO also protects against oxygen glucose deprivation-induced neuron injury, which relies on the upregulation of HO-1 (36). However, 1.2–2.5% ISO has adverse effects for patients in the ICU who cannot tolerate its hemodynamic effects. ISO at <1% has a weak effect on hemodynamics, which is more beneficial for ICU patients (37). The previous literature revealed that 0.7% ISO inhalation for 1 h at 1 and 6 h post-zymosan insult reduced zymosan-induced inflammation and apoptosis in vitro and in vivo (3840). Nevertheless, the roles of HO-1 in the neuroprotective effects of 0.7% ISO during sepsis remain largely unknown.

In the present study, the protective effects of 0.7% ISO were evaluated using an SAE mouse model created by cecal ligation and puncture (CLP), and the beneficial effects of 0.7% ISO were investigated for their potential association with HO-1 augmentation and/or activation.

Materials and methods

Animals and ethical guidelines

A total of 150 male C57BL/6 mice (age, 8 weeks; weight, 20–25 g) were purchased from the Animal Center of Xi'an Jiaotong University (Xi'an, China). The mice were maintained in specific pathogen-free conditions at 22±2°C with 60% relative humidity, 12-h dark/light cycles, and free access to food and water. This study was conducted in accordance with the ARRIVE guidelines for the Care and Use of Laboratory Animals (41), and the experimental procedures were approved by the Ethics Committee of Xi'an Jiaotong University (approval no. XAJU.No20180730A10120). All efforts were made to minimize suffering. The mice were anaesthetized with 50 mg/kg of pentobarbital sodium though intraperitoneal injection. At the end of the experiments, the mice were administrated with 30% volume displacement rate of CO2 for euthanasia. No heartbeat and mydriasis were used to confirm animal death.

CLP-induced SAE mouse model

A CLP-induced SAE mouse model was established as previously described with slight modifications (42). After anesthesia with pentobarbital sodium (50 mg/kg), the mice were immobilized on an aseptic operating table and received a 1-cm abdominal midline incision to expose the cecum. The distal 20% of the cecum (below the ileocecal valve and 1 cm from the tip) was ligated with a 6-0 suture and punctured twice with a 20-gauge needle. The cecum was gently squeezed to extrude a small amount of feces from the perforation sites, and then returned to the abdomen. The incision was subsequently sutured with sterile 6-0 silk. In the sham group, the mice underwent a similar procedure without ligation and puncture of the cecum. All animals received fluid resuscitation by subcutaneous injection of 1 ml pre-warmed 0.9% saline solution using a 25-gauge needle.

ISO administration

As previously reported, 0.7% ISO inhalation for 1 h was conducted at 1 and 6 h post-challenge in mice (40,43). The mice were placed in an air-tight plastic chamber (Billups-Rothenberg, Inc.) with an inflow and an outflow. ISO (Sigma-Aldrich; Merck KGaA) was delivered into the chamber by air at 2 l/min through a tube, and Baralyme (Allied Healthcare Products, Inc.) was used to remove CO2 from the chamber gases. The concentration (0.7%) of ISO in the outflow hose was continuously monitored using an anesthetic gas analyzer (Smart Anesthesia Multi-gas Module; Cytiva), and the temperature of the room and chamber was maintained at 20–24°C.

Experimental groups

A total of 150 mice were randomly assigned to the following five groups (n=30 per group): i) Control group, mice received sham surgery and no drug treatments; ii) ISO group, mice received sham surgery and ISO exposure; iii) CLP group, mice received CLP surgery and no drug treatments; iv) CLP + ISO group, mice received CLP surgery and ISO exposure; and v) CLP + ISO + zinc protoporphyrin IX (ZnPP; Sigma-Aldrich; Merck KGaA) group, mice received CLP surgery and both ISO and ZnPP treatments. 0.7% ISO inhalation for 1 h was conducted at 1 and 6 h post-CLP surgery, and ZnPP (10 mg/kg) was peritoneally injected into the mice 1 h before CLP surgery.

Evaluation of survival rate

Following CLP or sham treatment, 10 mice from each group were returned to their cages and the survival rate was calculated up to 7 days post-surgery.

Morris water maze (MWM) test

To evaluate the spatial learning and memory abilities of the mice, the MWM test was performed as previously described (44). The apparatus consisted of a circular pool (diameter, 150 cm; height, 60 cm) filled with water (22±2°C) containing food-grade titanium dioxide (Shanghai Jianghu Industrial Co., Ltd.). A removable platform (diameter, 10 cm) was placed in one quadrant of the pool and submerged 1 cm below the surface of the water. The pool was divided into four quadrants and the swimming paths of the mice were captured using a video camera above the center of the pool. From 4–7 days post-treatment, CLP or sham-treated mice (n=10) were individually placed into the pool at each of the four quadrants (facing the sidewalls) and allowed to circumnavigate in search of the platform for four trials per day (60 sec per trial). If a mouse failed to find the platform within 60 sec, it was guided by the investigator and permitted to search for an extra 10 sec. The escape latency, swimming distance and time spent in the target quadrant were recorded at each trial. The probe test comprised a single trial with the platform removed. The mice were released into the water and allowed to locate the original platform for 90 sec, and the time spent in the target quadrant was recorded. On the 7th day, the mice were subjected to the probe trial with the platform removed, 2 h after the last training trial. Each mouse was monitored for 60 sec to observe the swimming path, time spent in the target quadrant of the platform, and the number of times that the platform was crossed.

Brain water content

Brain water content was measured using the standard wet-dry method as previously reported (45). The mice were anesthetized and sacrificed 48 h post-CLP or sham surgery, and the brains were immediately removed and weighed to obtain the wet weight. Subsequently, the brain samples were dried in an oven at 100°C for 48 h, and the dry weight was acquired. Brain water content was calculated as follows: (Wet weight-dry weight/wet weight) ×100%.

BBB permeability assay

BBB permeability was evaluated by extravasation of Evans blue (EB) as previously reported (30). The mice were injected with 2% EB (3 ml/kg; Sigma-Aldrich; Merck KGaA) via the tail vein at 48 h post-CLP or sham treatment. After 2 h, the mice were anesthetized and transcardially perfused with normal saline. The brain samples were harvested, weighted, homogenized in formamide (10 ml/g), and incubated at 37°C for 48 h. The supernatants were collected after centrifugation at 10,000 × g for 30 min at 4°C, and the optical density (OD) at 625 nm was determined using a microplate reader (Molecular Devices, LLC). Standard formamide was used as the blank control, and the results were expressed as the relative amount of EB (µg/g wet weight).

Blood collection and hippocampus tissue preparation

At 48 h after CLP or sham surgery, the mice were anesthetized and blood samples were collected from the cervical artery, which were centrifuged at 1,000 × g (4°C) for 30 min. The mice were then euthanized, and the hippocampus tissues were immediately harvested. The tissues were homogenized and centrifuged at 10,000 × g and 4°C for 10 min. The sera and supernatants were stored at −80°C for the measurement of inflammatory mediators and oxidative parameters.

Enzyme-linked immunosorbent assay (ELISA)

The levels of TNF-α (cat. no. RTA00), IL-1β (cat. no. RLB00), IL-6 (cat. no. R6000B) and IL-10 (cat. no. R1000) in the serum and hippocampus supernatants were determined using commercially available ELISA kits (R&D Systems, Inc.), according to the manufacturer's instructions. The serum and hippocampus supernatants were collected and the OD at 450 nm was measured on an ELISA plate reader (Molecular Devices, LLC) and the results are expressed as pg/ml serum or pg/mg protein.

Reactive oxygen species (ROS) assay

ROS production was assessed using a 2′,7′-dichlorodihydrofluorescein diacetate (DCFH-DA) kit (Nanjing Jiancheng Bioengineering Institute) as per the manufacturer's protocol. Briefly, 50 µl serum or hippocampal supernatant were added to a 96-well plate and incubated with 100 µM DCFH-DA solution (200 µl) for 30 min (37°C) in the dark. DCFH-DA can be hydrolyzed by cellular esterases to form DCFH, which is oxidized by ROS to produce the fluorescent compound dichlorofluorescein (DCF). DCF oxidation was fluorometrically measured using a fluorescence microplate reader (Molecular Devices, LLC) at 485 nm excitation and 525 nm emission wavelengths. The data are represented as nmol/ml serum or nmol/mg protein.

Malondialdehyde (MDA), superoxide dismutase (SOD) and catalase (CAT) production

The levels of MDA (cat. no. A003-1-2), SOD (cat. no. A001-3-2) and CAT (cat. no. A007-1-1) in the serum and hippocampal supernatants were analyzed using commercial kits (Nanjing Jiancheng Bioengineering Institute) following the manufacturer's protocols. Briefly, the serum and supernatants were collected and the absorbance at 530, 450 and 405 nm was measured using a microplate reader (Bio-Rad Laboratories, Inc.) to determine MDA content and SOD and CAT levels, respectively. The MDA content is displayed as nmol/ml serum or nmol/mg protein, and the SOD and CAT activities were calculated as U/ml serum or U/mg protein.

Terminal deoxynucleotidyl transferase (TDT) dUTP nick end labeling (TUNEL) assay

TUNEL assay was carried out to detect neuronal apoptosis using the Cell Death Detection kit (Roche Diagnostics) as per the manufacturer's protocol. Hippocampal tissues were homogenized and centrifuged at 500 × g (4°C) for 5 min. The supernatants were discarded and the cell pellets were resuspended in PBS; 1×105 cells were seeded into 6-well plates. Cells were fixed with 4% paraformaldehyde for 1 h at 25°C. The TDT Enzyme (EMD Millipore) was added to the wells, which were then incubated for 1 h at 37°C in a dark humidified chamber. Stop/Wash buffer (EMD Millipore) was added for 10 min at room temperature, and the wells were rinsed with PBS before anti-digoxigenin fluorescein (EMD Millipore) was added. The cells were then incubated for 30 min at room temperature in a dark humidified chamber. In the negative control group, TDT was omitted. The cells were then incubated with DAPI for 15 min at 37°C in the dark. The slides were mounted with neutral balsam (Sangon Biotech Co., Ltd.). Stained cells were observed in five randomly selected fields of view using a fluorescence microscope (Olympus Corporation). The apoptotic index is expressed as the ratio of TUNEL+ (green) to DAPI+ cells (blue) ×100%.

Flow cytometric assessment of apoptosis

Hippocampal neuron apoptosis was detected via flow cytometry using the propidium iodide (PI)-Annexin V-FITC Apoptosis Detection kit (BD Biosciences). Hippocampal tissues were homogenized and centrifuged at 500 × g (4°C) for 5 min; the supernatants were discarded and the cell pellets were resuspended in 500 µl 1X Annexin V Binding Buffer. Subsequently, 5 µl Annexin V-FITC and 5 µl PI were added and the cells were incubated for 10 min in the dark at room temperature. The cells were immediately analyzed using a FACSCalibur™ flow cytometer (BD Biosciences) and the percentage of apoptotic cells (early + late apoptosis) was calculated by FlowJo software (version 7.6.2; FlowJo LLC).

Caspase-3 activity assay

Caspase-3 activity was measured using the Caspase-3/CPP32 Colorimetric Assay kit (BioVision, Inc.). Hippocampal tissues were homogenized and centrifuged at 500 × g (4°C) for 5 min. The supernatants were discarded and the cell pellets were resuspended in PBS; 1×105 cells were incubated on ice for 10 min with 50 µl chilled lysis buffer. The supernatant was then centrifuged at 12,000 × g for 5 min at 4°C, and 100 µg protein (in a total volume of 40 µl) was added to 40 µl 2X reaction buffer containing 5 µl N-Acetyl-Asp-Glu-Val-Asp-pNA substrate (150 µM final concentration). After incubation at 37°C for 2 h, N-Acetyl-Asp-Glu-Val-Asp-pNA cleavage was monitored by detecting the enzyme-catalyzed release of pNA using a microplate reader (BioTek Instruments, Inc.) at 405 nm.

Reverse transcription-quantitative (RT-q) PCR analysis

Total RNA was isolated from the hippocampal tissues using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) as per the manufacturer's instructions. cDNA was synthesized using the PrimeScript™ Reverse Transcription Reagent kit (Takara Biotechnology Co., Ltd.) with the following conditions: 37°C for 15 min, 85°C for 5 sec and maintained at 4°C. The qPCR assay was performed using the SYBR® Advantage® qPCR Premix (Takara Biotechnology Co., Ltd.) and conducted on a 7500 Fast Real-Time System (Applied Biosystems; Thermo Fisher Scientific, Inc.). The following thermocycling conditions were used for qPCR: 95°C for 10 min; followed by 40 cycles of 95°C for 10 sec and 60°C for 60 sec. Relative mRNA levels were calculated using the 2−ΔΔCq method (46). The primers sequences were as follows: HO-1 forward, 5′-TTCAGAAGGGTCAGGTCC-3′ and reverse, 5′-CAGTGAGGCCCATACCAGAA-3′; and GAPDH forward, 5′-ACCCAGAAGACTGTGGATGG-3′ and reverse, 5′-CACATTGGGGGTAGGAACAC-3′.

Western blot analysis

Hippocampal tissues were homogenized in RIPA lysis buffer (Beyotime Institute of Biotechnology) containing a protease inhibitor cocktail (Sigma-Aldrich; Merck KGaA). Total protein was quantified using a BCA protein assay kit (Beyotime Institute of Biotechnology). Equal amounts of protein sample (30 µg/lane) were separated via 10% SDS-PAGE, and then transferred onto polyvinylidene fluoride membranes (BD Biosciences). After blocking with 10% non-fat milk in Tris-buffered saline containing Tween-20 (50 mM Tris, pH 7.4, 250 mM NaCl and 0.1% Tween-20) at room temperature for 1 h, the membranes were incubated with primary antibodies against HO-1 (1:1,000; cat. no. ab189491; Abcam), Bax (1:1,000; cat. no. 5023s; Cell Signaling Technology, Inc.), Bcl-2 (1:1,000; cat. no. 3498s; Cell Signaling Technology, Inc.), caspase-3 (1:1,000; cat. no. 14220s; Cell Signaling Technology, Inc.), cleaved (cl)-caspase-3 (1:1,000; cat. no. 9664s; Cell Signaling Technology, Inc.) and β-actin (1:1,000; cat. no. 4967s; Cell Signaling Technology, Inc.) overnight at 4°C. Following treatment with the appropriate horseradish peroxidase-conjugated anti-rabbit IgG (1:2,000; cat. no. 7074s; Cell Signaling Technology, Inc.), the protein bands were visualized using an enhanced chemiluminescence detection kit (Amersham; Cytiva) and Kodak radiography film (FUJIFILM Wako Pure Chemical Corporation). The Western blot results were normalized to those of the internal control β-actin. The proteins were visualized using Image Quant™ LAS 4000 (GE Healthcare) and semi-quantified using ImageJ Software (version 1.46; National Institutes of Health).

Statistical analysis

All data are expressed as the mean ± standard deviation (SD). Survival rates were assessed using the Kaplan-Meier method and compared using the log-rank test. The latency, distance and time during water maze training were analyzed using two-way ANOVA followed by Bonferroni's post hoc test. Other comparisons among multiple groups were made using one-way ANOVA followed by Tukey's multiple comparisons test. The statistical analyses were performed using SPSS 18.0 software (SPSS, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

0.7% ISO increases the expression and activity of HO-1 in the hippocampus of CLP-induced septic mice

RT-qPCR and western blot assays were performed to investigate the expression of HO-1 in the hippocampus of CLP-challenged mice. As shown in Fig. 1A-C, CLP resulted in an increase in HO-1 mRNA and protein expression in the hippocampus, which was further enhanced by 0.7% ISO. However, the elevation of HO-1 was attenuated by treatment with the HO-1 inhibitor ZnPP. Similarly, HO-1 activity increased substantially in the hippocampus of mice subjected to CLP, compared with those in the control group. 0.7% ISO exposure resulted in further elevation of HO-1 activity compared with the CLP group, while ZnPP treatment reduced this augmented HO-1 activity (Fig. 1D). These results suggested that 0.7% ISO increases the expression and activity of HO-1 in the hippocampus of CLP-induced septic mice.

0.7% ISO reduces the mortality, brain water content and BBB disruption in CLP-induced septic mice

Next, the effects of 0.7% ISO on the survival rate of CLP-treated mice were investigated. During the 7-day observation period, no mortalities were observed in the control mice or those treated with ISO alone. However, the survival rate significantly decreased following CLP surgery, compared with that of the control group. 0.7% ISO significantly prolonged the survival time of CLP-challenged mice, which was subsequently abolished by ZnPP administration (Fig. 2A). As depicted in Fig. 2B and C, mice in the CLP group exhibited an increase in brain water content and a decrease in BBB integrity. 0.7% ISO exposure attenuated these effects in the CLP + ISO group compared with the CLP group. However, these beneficial effects were reversed by ZnPP treatment. These findings indicated that 0.7% ISO could decrease mortality rates and brain edema, and increase BBB integrity in CLP-induced mice, which partly depends on HO-1 activation.

0.7% ISO improves the memory and learning capacity of CLP-induced septic mice

A MWM training assay was conducted to investigate the effects of 0.7% ISO on the learning abilities of CLP-induced mice. Compared with the control group, CLP-treated mice showed markedly prolonged escape latency from the 5th day post-surgery. CLP-challenged mice exposed to 0.7% ISO displayed considerably shorter escape latency compared with mice that underwent CLP surgery only. Furthermore, ZnPP administration attenuated the effects of 0.7% ISO on escape latency (Fig. 3A). Of note, there were no significant differences in the distance and time spent in the target quadrant among all five groups (Fig. 3B and C). Following water maze training, the platform was removed to analyze the role of 0.7% ISO on the memory of CLP-challenged mice, including the distance, time spent in the target quadrant, and the frequency at which the platform was crossed. As depicted in Fig. 3D, the travelled distance was similar among the five groups. However, the CLP-treated mice spent a significantly shorter time in the target quadrant than the control mice, which was significantly prolonged by 0.7% ISO; the time spent at the quadrant was subsequently decreased by ZnPP treatment (Fig. 3E). Similarly, the frequency of platform crossing was significantly reduced in the CLP group compared with the control mice, which was significantly increased by 0.7% ISO, and decreased by ZnPP administration (Fig. 3F). These data demonstrated that 0.7% ISO ameliorates the learning and memory functions of CLP-induced septic mice, which is indicated to involve activated HO-1.

0.7% ISO inhibits CLP-induced inflammation in the serum and hippocampus of mice

To identify the effects of 0.7% ISO on inflammatory responses in CLP-induced septic mice, ELISA assays were conducted to evaluate the levels of pro- and anti-inflammatory factors in the serum and hippocampus. CLP was found to significantly increase the levels of pro-inflammatory cytokines, including TNF-α (Fig. 4A), IL-1β (Fig. 4B) and IL-6 (Fig. 4C), and the anti-inflammatory cytokine IL-10 (Fig. 4D) in the serum. 0.7% ISO resulted in a significant reduction of TNF-α, IL-1β and IL-6, and a further elevation of IL-10 in the sera of CLP-treated mice. Whereas, ZnPP reversed the changes in TNF-α, IL-1β, IL-6 and IL-10 expression (Fig. 4A-D). Consistently, 0.7% ISO exposure significantly reduced the CLP-induced increase in the levels of TNF-α, IL-1β and IL-6, and further enhanced the production of IL-10 in the hippocampus, which were significantly counteracted by ZnPP (Fig. 4E-H). These results indicated that HO-1 is highly important to the anti-inflammatory effects of 0.7% ISO in CLP-induced septic mice.

0.7% ISO represses CLP-induced oxidative stress in the serum and hippocampus of mice

To further elucidate the effects of 0.7% ISO on sepsis-induced oxidative stress, the ROS and MDA contents, and anti-oxidative SOD and CAT activities were determined in the serum and hippocampal tissues of the mice. Compared with the control group, the production of ROS (Fig. 5A) and MDA (Fig. 5B) significantly increased, and the SOD (Fig. 5C) and CAT (Fig. 5D) activities significantly decreased in the sera of CLP-challenged mice. 0.7% ISO exposure significantly reduced the levels of ROS and MDA, and enhanced the activities of SOD and CAT, which were reversed by ZnPP administration (Fig. 5A-D). Similarly, 0.7% ISO reduced CLP-induced ROS and MDA, and enhanced CLP-reduced SOD and CAT activity in the hippocampus, and ZnPP reversed these alterations (Fig. 5E-H). These findings illustrated that the antioxidative effects of 0.7% ISO on CLP-induced septic mice are reliant on the activation of HO-1.

0.7% ISO suppresses CLP-induced neuronal apoptosis in the mouse hippocampus

Next, a TUNEL assay was performed to assess the impacts of 0.7% ISO on CLP-induced neuronal apoptosis. As shown in Fig. 6A and B, an increased number of apoptotic neurons were observed in the hippocampus of CLP-challenged mice than those of the control group. 0.7% ISO significantly decreased the number of TUNEL+ cells in the hippocampus, while ZnPP abrogated these protective effects. The flow cytometry results also demonstrated that 0.7% ISO decreased CLP-induced neuronal apoptosis (Fig. 6C and D) in the hippocampus, which was reversed by ZnPP administration. Furthermore, 0.7% ISO decreased CLP-induced caspase-3 activation (Fig. 6E) in the hippocampus, whereas ZnPP administration significantly reversed this effect. On a molecular level, 0.7% ISO resulted in a significant upregulation in anti-apoptotic Bcl-2 and downregulation in pro-apoptotic Bax and cl-caspase-3 in the hippocampus of CLP-treated mice. ZnPP reversed the expression changes in these proteins (Fig. 6F-I). There were no significance differences in the expression of caspase-3 among the five groups (Fig. 6F and J). These data indicated that HO-1 is implicated in the anti-apoptotic functions of 0.7% ISO in CLP-induced septic mice.

Discussion

The present study demonstrated that 0.7% ISO exerted neuroprotective effects on CLP-induced septic mice, which involved HO-1 signaling. The following key findings were observed in septic mice: i) ISO enhanced the expression and activity of HO-1 in the hippocampus; ii) ISO improved the survival rate and reduced brain water content and BBB disruption; iii) ISO ameliorated the memory and learning ability; iv) ISO reduced the production of pro-inflammatory cytokines (TNF-α, IL-1β and IL-6) and increased the release of the anti-inflammatory cytokine IL-10 in the serum and hippocampus; v) ISO resulted in decreased oxidative products (ROS and MDA) and a simultaneous increase in antioxidant enzyme (SOD and CAT) activity in the serum and hippocampus; vi) ISO inhibited neuronal apoptosis by upregulating Bcl-2 and downregulating Bax and cl-caspase-3 in the hippocampus; and vii) the neuroprotective effects of ISO were mediated by HO-1 activation. Overall, 0.7% ISO prevented CLP-associated neuronal injury by reducing inflammation, oxidative stress and apoptosis, which was dependent on HO-1 activation.

A previous study reported that the use of ISO during surgery is considered optimal for neuroprotection (47). ISO post-conditioning notably decreases the infarct volume and improves the neurobehavioral performance of rats subjected to middle cerebral artery occlusion, thereby providing increased ischemic tolerance in the brain (48). ISO increases the weight ratio and neuronal density of neonatal rats, and ameliorates the memory and learning ability of adolescent rats following brain hypoxia and ischemia (49). ISO exerts a protective effect on hypoxia-induced neuronal injury in the hippocampus by suppressing apoptosis (50). The present study highlighted that ISO exerted neuroprotection against CLP-induced SAE in mice by increasing overall survival, and reducing brain water content and BBB disruption, as well as increasing the memory capacity and learning ability.

Systemic and neuroinflammation can induce endothelial cell damage, microglial activation and neuronal death (51). As a critical structure of learning and memory, the hippocampus is vulnerable to systemic inflammatory factors (52,53), and pro-inflammatory cytokines, such as TNF-α, IL-1β and IL-6, can readily promote endothelial dysfunction, BBB disruption and brain edema (54,55). IL-10 is a potent anti-inflammatory cytokine that alleviates such injuries (56). Reportedly, ISO represses neuroinflammation, coinciding with a decrease in TNF-α and IL-1β in the mouse brain following subarachnoid hemorrhage (57). In the current study, 0.7% ISO decreased the production of TNF-α, IL-1β and IL-6, and increased IL-10 generation in the serum and hippocampi of CLP-induced septic mice. Previous studies have suggested that ISO induces HO-1 expression in Hep3B cells (58) and hippocampal neurons (36). HO-1 signaling mediates the anti-inflammatory effects of ISO in CLP-induced lung injury (27) and ischemia/reperfusion-induced liver injury (59), as well as in LPS-challenged macrophages (35). Consistently, the present study revealed that 0.7% ISO induced the expression and activity of HO-1 in the hippocampi of CLP-treated mice, and that ZnPP reversed the anti-inflammatory abilities of 0.7% ISO, suggesting that the protective effects of 0.7% ISO against CLP-induced sepsis partially rely on activated HO-1 signaling.

Oxidative stress is one of the primary causes of SAE during sepsis (60). Excessive ROS generation induces lipid peroxidation, disrupts cellular and mitochondrial membranes, and ultimately results in the apoptosis and necrosis of cells (61). MDA is a direct product of lipid peroxidation and is recognized as an indicator of ROS-mediated injury (62). Antioxidant enzymes, such as SOD and CAT, are key ROS scavengers that specifically eliminate superoxide radicals and prevent ROS-induced brain damage during sepsis (24). A published review summarizes the anti-oxidative aspects of ISO in acute brain injury (63). HO-1 is a key participant in sepsis (2729), and has been reported to hamper oxidative stress during sepsis-induced acute lung injury (64). ISO enhanced HO-1 expression in the lungs of CLP-treated rats (27). Herein, the present findings demonstrated that 0.7% ISO decreased the ROS and MDA content, and enhanced SOD and CAT activity in the serum and hippocampi of CLP-induced septic mice. Moreover, ZnPP administration reversed these effects, indicating that HO-1 signaling is implicated in the anti-oxidative effects of 0.7% ISO in septic mice.

Due to its close association with cognitive dysfunction, neuronal apoptosis in the hippocampus is a major causative factor of SAE (65). The balance between anti- and pro-apoptotic Bcl-2 family proteins dominates the progression of neuron apoptosis (66). Pro-apoptotic Bax translocates to the outer mitochondrial membrane, disrupting mitochondrial transmembrane potential and ultimately promoting cell death. On the contrary, anti-apoptotic Bcl-2 inhibits Bax activation (67). Caspase-3, the final apoptotic executor, plays a crucial role in neuronal apoptosis (14). ISO has been confirmed to increase Bcl-2 expression and reduce cytochrome c release from mitochondria in the ischemic penumbra of the rat brain (68). Bedirli et al (69) reported that ISO upregulates Bcl-2 and downregulates Bax in the rat brain following focal cerebral ischemia. ISO reduces oxygen and glucose deprivation-induced hippocampal neuron apoptosis by inactivating caspase-3 (50). HO-1 signaling mediates the inhibitory effects of 0.7% ISO on zymosan-induced pulmonary cell apoptosis (70). In line with previous findings, it was confirmed in the present study that 0.7% ISO repressed neuronal apoptosis in the hippocampus of CLP-induced septic mice, which was accompanied by reduced expression of Bax and cl-caspase-3, and increased Bcl-2 expression. Treatment with ZnPP, a HO-1 inhibitor, neutralized the anti-apoptotic effects of 0.7% ISO, implying that HO-1 signaling is involved in the 0.7% ISO-reduced neuronal apoptosis in CLP-induced septic mice.

In conclusion, the present study demonstrated that 0.7% ISO plays crucial neuroprotective roles in a CLP-established SAE mouse model, largely involving the activation of HO-1. The upregulation and activation of HO-1 mediates the anti-inflammatory, antioxidative and anti-apoptotic effects of 0.7% ISO on CLP-induced neuronal damage in the brains of septic mice. These findings provided strong evidence for the potential application of 0.7% ISO for the treatment of patients with SAE.

Acknowledgements

Not applicable.

Funding

This study was supported in part by the National Natural Science Foundation of China (grant no. 81401138).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

ZW contributed to the conception and design of the present study. LZ and XZ conducted all the experiments. ZW and LZ drafted the manuscript and revised it critically for important intellectual content. TW and XP interpreted and analyzed the data. ZW provided final approval of the version to be published. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All animal experimental protocols were approved by the Animal Care and Use Committee of Xi'an Jiaotong University (approval no. XAJU.No20180730A10120) and were conducted in accordance with the National Institutes of Health guidelines for the Care and Use of Laboratory Animals.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Delano MJ and Ward PA: Sepsis-induced immune dysfunction: Can immune therapies reduce mortality? J Clin Invest. 126:23–31. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Dellinger RP, Levy MM, Rhodes A, Annane D, Gerlach H, Opal SM, Sevransky JE, Sprung CL, Douglas IS, Jaeschke R, et al: Surviving sepsis campaign: International guidelines for management of severe sepsis and septic shock: 2012. Crit Care Med. 41:580–637. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Gong Y, Lan H, Yu Z, Wang M, Wang S, Chen Y, Rao H, Li J, Sheng Z and Shao J: Blockage of glycolysis by targeting PFKFB3 alleviates sepsis-related acute lung injury via suppressing inflammation and apoptosis of alveolar epithelial cells. Biochem Biophys Res Commun. 491:522–529. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Alobaidi R, Basu RK, Goldstein SL and Bagshaw SM: Sepsis-associated acute kidney injury. Semin Nephrol. 35:2–11. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Martin L, Derwall M, Al Zoubi S, Zechendorf E, A Reuter D, Thiemermann C and Schuerholz T: The septic heart: Current understanding of molecular mechanisms and clinical implications. Chest. 155:427–437. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Yan J and Li S and Li S: The role of the liver in sepsis. Int Rev Immunol. 33:498–510. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Zhu Y, Wang K, Ma Z, Liu D, Yang Y, Sun M, Wen A, Hao Y, Ma S, Ren F, et al: SIRT1 activation by butein attenuates sepsis-induced brain injury in mice subjected to cecal ligation and puncture via alleviating inflammatory and oxidative stress. Toxicol Appl Pharmacol. 363:34–46. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Gofton TE and Young GB: Sepsis-associated encephalopathy. Nat Rev Neurol. 8:557–566. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Zhang LN, Wang XT, Ai YH, Guo QL, Huang L, Liu ZY and Yao B: Epidemiological features and risk factors of sepsis-associated encephalopathy in intensive care unit patients: 2008–2011. Chin Med J (Engl). 125:828–831. 2012.PubMed/NCBI

10 

Iwashyna TJ, Ely EW, Smith DM and Langa KM: Long-term cognitive impairment and functional disability among survivors of severe sepsis. JAMA. 304:1787–1794. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Barichello T, Martins MR, Reinke A, Feier G, Ritter C, Quevedo J and Dal-Pizzol F: Cognitive impairment in sepsis survivors from cecal ligation and perforation. Crit Care Med. 33:221–223, 262-223. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Semmler A, Frisch C, Debeir T, Ramanathan M, Okulla T, Klockgether T and Heneka MT: Long-term cognitive impairment, neuronal loss and reduced cortical cholinergic innervation after recovery from sepsis in a rodent model. Exp Neurol. 204:733–740. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Schwalm MT, Pasquali M, Miguel SP, JPAD Santos, Vuolo F, Comim CM, Petronilho F, Quevedo J, Gelain DP, Moreira JCF, et al: Acute brain inflammation and oxidative damage are related to long-term cognitive deficits and markers of neurodegeneration in sepsis-survivor rats. Mol Neurobiol. 49:380–385. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Comim CM, Barichello T, Grandgirard D, Dal-Pizzol F, Quevedo J and Leib SL: Caspase-3 mediates in part hippocampal apoptosis in sepsis. Mol Neurobiol. 47:394–398. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Heine H, Rietschel ET and Ulmer AJ: The biology of endotoxin. Mol Biotechnol. 19:279–296. 2001. View Article : Google Scholar : PubMed/NCBI

16 

Vaure C and Liu Y: A comparative review of toll-like receptor 4 expression and functionality in different animal species. Front Immunol. 5:3162014. View Article : Google Scholar : PubMed/NCBI

17 

Shukla P, Rao GM, Pandey G, Sharma S, Mittapelly N, Shegokar R and Mishra PR: Therapeutic interventions in sepsis: Current and anticipated pharmacological agents. Br J Pharmacol. 171:5011–5031. 2014.PubMed/NCBI

18 

Sonneville R, Verdonk F, Rauturier C, Klein IF, Wolff M, Annane D, Chretien F and Sharshar T: Understanding brain dysfunction in sepsis. Ann Intensive Care. 3:152013. View Article : Google Scholar : PubMed/NCBI

19 

Danielski LG, Giustina AD, Badawy M, Barichello T, Quevedo J, Dal-Pizzol F and Petronilho F: Brain barrier breakdown as a cause and consequence of neuroinflammation in sepsis. Mol Neurobiol. 55:1045–1053. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Abramowitz J and Birnbaumer L: Physiology and pathophysiology of canonical transient receptor potential channels. FASEB J. 23:297–328. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Fleming I, Fisslthaler B and Busse R: Calcium signaling in endothelial cells involves activation of tyrosine kinases and leads to activation of mitogen-activated protein kinases. Circ Res. 76:522–529. 1995. View Article : Google Scholar : PubMed/NCBI

22 

Barichello T, Fortunato JJ, Vitali AM, Feier G, Reinke A, Moreira JCF, Quevedo J and Dal-Pizzol F: Oxidative variables in the rat brain after sepsis induced by cecal ligation and perforation. Crit Care Med. 34:886–889. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Dal-Pizzol F, Ritter C, Cassol OJ Jr, Rezin GT, Petronilho F, Zugno AI, Quevedo J and Streck EL: Oxidative mechanisms of brain dysfunction during sepsis. Neurochem Res. 35:1–12. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Hoetzel A and Schmidt R: Regulatory role of anesthetics on heme oxygenase-1. Curr Drug Targets. 11:1495–1503. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Alcaraz MJ, Fernandez P and Guillen MI: Anti-inflammatory actions of the heme oxygenase-1 pathway. Curr Pharm Des. 9:2541–2551. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Chen K, Gunter K and Maines MD: Neurons overexpressing heme oxygenase-1 resist oxidative stress-mediated cell death. J Neurochem. 75:304–313. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Dong X, Hu R, Sun Y, Li Q and Jiang H: Isoflurane post-treatment improves pulmonary vascular permeability via upregulation of heme oxygenase-1. Exp Lung Res. 39:295–303. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Yu JB, Zhou F, Yao SL, Tang ZH, Wang M and Chen HR: Effect of heme oxygenase-1 on the kidney during septic shock in rats. Transl Res. 153:283–287. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Park JS, Choi HS, Yim SY and Lee SM: Heme oxygenase-1 protects the liver from septic injury by modulating TLR4-mediated mitochondrial quality control in mice. Shock. 50:209–218. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Liu L, Xie K, Chen H, Dong X, Li Y and Yu Y, Wang G and Yu Y: Inhalation of hydrogen gas attenuates brain injury in mice with cecal ligation and puncture via inhibiting neuroinflammation, oxidative stress and neuronal apoptosis. Brain Res. 1589:78–92. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Flondor M, Hofstetter C, Boost KA, Betz C, Homann M and Zwissler B: Isoflurane inhalation after induction of endotoxemia in rats attenuates the systemic cytokine response. Eur Surg Res. 40:1–6. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Jamnicki-Abegg M, Weihrauch D, Pagel PS, Kersten JR, Bosnjak ZJ, Warltier DC and Bienengraeber MW: Isoflurane inhibits cardiac myocyte apoptosis during oxidative and inflammatory stress by activating Akt and enhancing Bcl-2 expression. Anesthesiology. 103:1006–1014. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Head BP and Patel P: Anesthetics and brain protection. Curr Opin Anaesthesiol. 20:395–399. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Sakai H, Sheng H, Yates RB, Ishida K, Pearlstein RD and Warner DS: Isoflurane provides long-term protection against focal cerebral ischemia in the rat. Anesthesiology. 106:92–99; discussion 98–10. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Li QF, Zhu YS, Jiang H, Xu H and Sun Y: Heme oxygenase-1 mediates the anti-inflammatory effect of isoflurane preconditioning in LPS-stimulated macrophages. Acta Pharmacol Sin. 30:228–234. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Li Q, Zhu Y, Jiang H, Xu H and Liu H: Up-regulation of heme oxygenase-1 by isoflurane preconditioning during tolerance against neuronal injury induced by oxygen glucose deprivation. Acta Biochim Biophys Sin (Shanghai). 40:803–810. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Sackey PV, Martling CR, Carlswärd C, Sundin O and Radell PJ: Short- and long-term follow-up of intensive care unit patients after sedation with isoflurane and midazolam-a pilot study. Crit Care Med. 36:801–806. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Li JT, Wang H, Li W, Wang LF, Hou LC, Mu JL, Liu X, Chen HJ, Xie KL, Li NL and Gao CF: Anesthetic isoflurane posttreatment attenuates experimental lung injury by inhibiting inflammation and apoptosis. Mediators Inflamm. 2013:1089282013. View Article : Google Scholar : PubMed/NCBI

39 

Wang H, Fan J, Li NL, Li JT, Yuan SF, Yi J, Wang L, Chen JH, Lv YG, Yao Q, et al: A subanesthetic dose of isoflurane during postconditioning ameliorates zymosan-induced neutrophil inflammation lung injury and mortality in mice. Mediators Inflamm. 2013:4796282013. View Article : Google Scholar : PubMed/NCBI

40 

Wang H, Wang L, Li NL, Li JT, Yu F, Zhao YL, Wang L, Yi J, Wang L, Bian JF, et al: Subanesthetic isoflurane reduces zymosan-induced inflammation in murine Kupffer cells by inhibiting ROS-activated p38 MAPK/NF-κB signaling. Oxid Med Cell Longev. 2014:8516922014. View Article : Google Scholar : PubMed/NCBI

41 

Kilkenny C, Browne W, Cuthill IC, Emerson M and Altman DG; National Centre for the Replacement, Refinement and Reduction of Amimals in Research, : Animal research: Reporting in vivo experiments-the ARRIVE guidelines. J Cereb Blood Flow Metab. 31:991–993. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Toscano MG, Ganea D and Gamero AM: Cecal ligation puncture procedure. J Vis Exp. 7:28602011.

43 

Mu J, Xie K, Hou L, Peng D, Shang L, Ji G, Li J, Lu Y and Xiong L: Subanesthetic dose of isoflurane protects against zymosan-induced generalized inflammation and its associated acute lung injury in mice. Shock. 34:183–189. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Sui DM, Xie Q, Yi WJ, Gupta S, Yu XY, Li JB, Wang J, Wang JF and Deng XM: Resveratrol protects against sepsis-associated encephalopathy and inhibits the NLRP3/IL-1β axis in microglia. Mediators Inflamm. 2016:10456572016. View Article : Google Scholar : PubMed/NCBI

45 

Hatashita S, Hoff JT and Salamat SM: Ischemic brain edema and the osmotic gradient between blood and brain. J Cereb Blood Flow Metab. 8:552–559. 1988. View Article : Google Scholar : PubMed/NCBI

46 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

47 

Jiang M, Sun L, Feng DX, Yu ZQ, Gao R, Sun YZ and Chen G: Neuroprotection provided by isoflurane pre-conditioning and post-conditioning. Med Gas Res. 7:48–55. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Wang S, Yin J, Ge M, Dai Z, Li Y, Si J, Ma K, Li L and Yao S: Transforming growth-beta 1 contributes to isoflurane postconditioning against cerebral ischemia-reperfusion injury by regulating the c-Jun N-terminal kinase signaling pathway. Biomed Pharmacother. 78:280–290. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Xu Y, Xue H, Zhao P, Yang Y, Ji G, Yu W, Han G, Ding M and Wang F: Isoflurane postconditioning induces concentration- and timing-dependent neuroprotection partly mediated by the GluR2 AMPA receptor in neonatal rats after brain hypoxia-ischemia. J Anesth. 30:427–436. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Zhao DA, Bi LY, Huang Q, Zhang FM and Han ZM: Isoflurane provides neuroprotection in neonatal hypoxic ischemic brain injury by suppressing apoptosis. Braz J Anesthesiol. 66:613–621. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Matsuda A, Jacob A, Wu R, Aziz M, Yang WL, Matsutani T, Suzuki H, Furukawa K, Uchida E and Wang P: Novel therapeutic targets for sepsis: Regulation of exaggerated inflammatory responses. J Nippon Med Sch. 79:4–18. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Calabresi P, Castrioto A, Di Filippo M and Picconi B: New experimental and clinical links between the hippocampus and the dopaminergic system in Parkinson's disease. Lancet Neurol. 12:811–821. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Alexander JJ, Jacob A, Cunningham P, Hensley L and Quigg RJ: TNF is a key mediator of septic encephalopathy acting through its receptor, TNF receptor-1. Neurochem Int. 52:447–456. 2008. View Article : Google Scholar : PubMed/NCBI

54 

Rothwell NJ and Hopkins SJ: Cytokines and the nervous system II: Actions and mechanisms of action. Trends Neurosci. 18:130–136. 1995. View Article : Google Scholar : PubMed/NCBI

55 

Sharief MK and Thompson EJ: In vivo relationship of tumor necrosis factor-alpha to blood-brain barrier damage in patients with active multiple sclerosis. J Neuroimmunol. 38:27–33. 1992. View Article : Google Scholar : PubMed/NCBI

56 

Williams LM, Ricchetti G, Sarma U, Smallie T and Foxwell BM: Interleukin-10 suppression of myeloid cell activation-a continuing puzzle. Immunology. 113:281–292. 2004. View Article : Google Scholar : PubMed/NCBI

57 

Altay O, Suzuki H, Hasegawa Y, Ostrowski RP, Tang J and Zhang JH: Isoflurane on brain inflammation. Neurobiol Dis. 62:365–371. 2014. View Article : Google Scholar : PubMed/NCBI

58 

Li QF, Wang XR, Yang YW and Su DS: Up-regulation of hypoxia inducible factor 1alpha by isoflurane in Hep3B cells. Anesthesiology. 105:1211–1219. 2006. View Article : Google Scholar : PubMed/NCBI

59 

Schmidt R, Tritschler E, Hoetzel A, Loop T, Humar M, Halverscheid L, Geiger KK and Pannen BH: Heme oxygenase-1 induction by the clinically used anesthetic isoflurane protects rat livers from ischemia/reperfusion injury. Ann Surg. 245:931–942. 2007. View Article : Google Scholar : PubMed/NCBI

60 

Hopkins RO: Sepsis, oxidative stress, and brain injury. Crit Care Med. 35:2233–2234. 2007. View Article : Google Scholar : PubMed/NCBI

61 

Chandra J, Samali A and Orrenius S: Triggering and modulation of apoptosis by oxidative stress. Free Radic Biol Med. 29:323–333. 2000. View Article : Google Scholar : PubMed/NCBI

62 

Requena JR, Fu MX, Ahmed MU, Jenkins AJ, Lyons TJ and Thorpe SR: Lipoxidation products as biomarkers of oxidative damage to proteins during lipid peroxidation reactions. Nephrol Dial Transplant. 11 (Suppl 5):48–53. 1996. View Article : Google Scholar : PubMed/NCBI

63 

Yang T, Sun Y and Zhang F: Anti-oxidative aspect of inhaled anesthetic gases against acute brain injury. Med Gas Res. 6:223–226. 2016. View Article : Google Scholar : PubMed/NCBI

64 

Yu J, Wang Y, Li Z, Dong S, Wang D, Gong L, Shi J, Zhang Y, Liu D and Mu R: Effect of heme oxygenase-1 on mitofusin-1 protein in LPS-induced ALI/ARDS in rats. Sci Rep. 6:365302016. View Article : Google Scholar : PubMed/NCBI

65 

Semmler A, Okulla T, Sastre M, Dumitrescu-Ozimek L and Heneka MT: Systemic inflammation induces apoptosis with variable vulnerability of different brain regions. J Chem Neuroanat. 30:144–157. 2005. View Article : Google Scholar : PubMed/NCBI

66 

Yao XL, Liu J, Lee E, Ling GS and McCabe JT: Progesterone differentially regulates pro- and anti-apoptotic gene expression in cerebral cortex following traumatic brain injury in rats. J Neurotrauma. 22:656–668. 2005. View Article : Google Scholar : PubMed/NCBI

67 

Renault TT, Teijido O, Antonsson B, Dejean LM and Manon S: Regulation of Bax mitochondrial localization by Bcl-2 and Bcl-x(L): Keep your friends close but your enemies closer. Int J Biochem Cell Biol. 45:64–67. 2013. View Article : Google Scholar : PubMed/NCBI

68 

Li L, Peng L and Zuo Z: Isoflurane preconditioning increases B-cell lymphoma-2 expression and reduces cytochrome c release from the mitochondria in the ischemic penumbra of rat brain. Eur J Pharmacol. 586:106–113. 2008. View Article : Google Scholar : PubMed/NCBI

69 

Bedirli N, Bagriacik EU, Emmez H, Yilmaz G, Unal Y and Ozkose Z: Sevoflurane and isoflurane preconditioning provides neuroprotection by inhibition of apoptosis-related mRNA expression in a rat model of focal cerebral ischemia. J Neurosurg Anesthesiol. 24:336–344. 2012. View Article : Google Scholar : PubMed/NCBI

70 

Wang L, Zhao YL, Liu NN, Zhu XS, Liu QQ, Mei HY, Wang LF, Yang AG, Gao CF and Li JT: Epithelial HO-1/STAT3 affords the protection of subanesthetic isoflurane against zymosan-induced lung injury in mice. Oncotarget. 8:54889–54903. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2021
Volume 23 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang L, Zhang X, Wu T, Pan X and Wang Z: Isoflurane reduces septic neuron injury by HO‑1‑mediated abatement of inflammation and apoptosis. Mol Med Rep 23: 155, 2021
APA
Zhang, L., Zhang, X., Wu, T., Pan, X., & Wang, Z. (2021). Isoflurane reduces septic neuron injury by HO‑1‑mediated abatement of inflammation and apoptosis. Molecular Medicine Reports, 23, 155. https://doi.org/10.3892/mmr.2020.11794
MLA
Zhang, L., Zhang, X., Wu, T., Pan, X., Wang, Z."Isoflurane reduces septic neuron injury by HO‑1‑mediated abatement of inflammation and apoptosis". Molecular Medicine Reports 23.2 (2021): 155.
Chicago
Zhang, L., Zhang, X., Wu, T., Pan, X., Wang, Z."Isoflurane reduces septic neuron injury by HO‑1‑mediated abatement of inflammation and apoptosis". Molecular Medicine Reports 23, no. 2 (2021): 155. https://doi.org/10.3892/mmr.2020.11794