Galectin-1 ameliorates perioperative neurocognitive disorders in aged mice

DOI: https://doi.org/10.21203/rs.3.rs-80016/v1

Abstract

Background

The incidence of perioperative neurocognitive disorders (PND) is higher in the elderly patients undergoing surgery. Microglia activation-mediated neuroinflammation is one of the hallmarks of PND. Galectin-1 has been identified as pivotal modulator in central nervous system (CNS), while the role of galectin-1 in PND induced by microglia-mediated neuroinflammation is still undetermined.

Methods

An exploratory laparotomy model anesthetized with isoflurane was employed to investigate the role of galectin-1 on PND in aged mice. Open field test and Morris water maze were used to test the cognitive function 3 or 7 days after surgery. Immunofluorescence staining was employed to detect the activation of microglia and neuronal integrity in the hippocampus of aged mice. Enzyme-linked immunosorbent assay (ELISA), reverse transcription quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot were used to elucidate the underlying mechanisms.

Results

Pretreatment with galectin-1 attenuated the cognitive dysfunction induced by surgery in aged mice and inhibited microglial activity. Moreover, galectin-1 decreased the expression level of inflammatory proteins (interleukin-1β, interleukin-6 and tumor necrosis factor-α), and disrupted neuronal integrity in hippocampus. Galectin-1 inhibited the inflammation of BV2 microglial cells induced by lipopolysaccharide via decreasing the translocation of c-Jun and NF-κB p65, while this kind of inhibition was rescued when overexpressing IRAK1.

Conclusions

Our findings provide evidence that galectin-1 may inhibit IRAK1 expression, thus suppressing inflammatory response, inhibiting neuroinflammation, and improving ensuing cognitive dysfunction. Collectively, these findings unveil that galectin-1 may elicit protective effect on surgery-induced neuroinflammation and neurocognitive disorders.

1. Introduction

Perioperative neurocognitive disorders (PND) are common postoperative complications, characterized by cognitive dysfunctions in the aged patient undergoing surgery and anesthesia. The occurrence of PND not only affects the quality of life and increases the burden of medical treatment, but also leads to increasing morbidity and mortality[1, 2, 3]. PND are accompanied by obvious neuroinflammation characterized by microgliosis, reactive astrocytosis and increased levels of proinflammatory cytokines[4, 5]. Despite multiple pathological events have been proposed to be associated with PND[6, 7], no effective clinical therapeutics treating PND has been developed.

As resident inflammatory cells in the central nervous system (CNS), microglia play a crucial role in neuroinflammation. Depending upon the nature of the original stimulation, the activated-microglia exert multifarious role that could be either detrimental or beneficial. Microglia usually stay at a resting condition, but when exposed to exogenous antigens such as viruses or bacteria, microglia are rapidly activated and become neurotoxic[8]. In addition, the activation of microglial may augment the transcriptional activity of c-Jun and NF-κB p65 and accelerate their translocation from the cytoplasm to the nucleus, leading to the increased level of inflammatory cytokines. Furthermore, activating microglia may promote to generate inflammatory proteins, such as interleukin-1β (IL-1β), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), giving rise to neuroinflammation and the ensuing cognitive deficits[9, 10]. Consequently, the inactivation of microglial may attenuate neurocognitive dysfunction through attenuating the translocation of c-Jun and NF-κB p65 and neuroinflammation.

As a β-galactoside-binding protein, galectin-1 has various biological activities and acts as a critical regulator in neuroinflammation. It has been shown that the expression of galectin-1 is altered in neurological disorders and is associated with neuroinflammation[11, 12]. In vitro, galectin-1 modulates the function of microglia by reducing the LPS-stimulated pro-inflammatory cytokines ( nitric oxide and TNF-α) and contributing to the increased secretion of anti-inflammatory cytokines (TGF-β and IL-10) [13]. Whether galectin-1 attenuates perioperative neurocognitive disorders and the underlying mechanism are still undetermined.

In the present study, we provided evidence that galectin-1 treatment attenuated surgery-induced microglia activation and disruption of neuronal integrity in aged mice. This issue indicates how galectin-1 exerted protective effect on PND, and demonstrates that galectin-1 may be therapeutic by inhibiting IRAK1-mediated translocation of c-Jun and NF-κB p65 and inflammatory response, which may retard the development of neuroinflammation and the ensuing neurocognitive dysfunction effectively.

2. Method And Materials

2.1 Animals

C57BL/6 mice (26–38 g, 12 months old) were purchased from Guangdong Medical Laboratory Animal Research Institute. All animals were housed in polypropylene cages and the relative humidity was maintained at 50 ± 10%. The room temperature was maintained at 22 °C with a 12 h light-dark cycle. All of the experiment in this study were performed according to the National Institutes of Health Guidelines for experimental animals and all experimental protocols and procedures were approved by the animal care committee at Sun Yat-sen University.

2.2 Surgery

Anesthetized with 1.5% isflurane, a modified abdominal exploratory laparotomy was performed[14]. In an induction chamber with 1.5% isoflurane and oxygen at 2 L/min, mice were first anesthetized. And after the mouse was exposed to isoflurane for 30 min, the abdominal region was shaved and sterilized thoroughly using 0.5% iodine. Then, a 1 cm median abdominal incision was made, so that the peritoneal cavity was penetrated. Next, the intestine, musculature and viscera were explored. Using sterile 4 − 0 chromic gut sutures, the peritoneal lining and skin were sutured. The entire surgical procedure was done under isoflurane anesthesia and lasted for 30 min. Neither surgery nor anesthesia was performed for the mice in the control group.

2.3 Open-field test (OFT)

The OFT was performed as we described before[15]. In brief, OFT was performed in the center of open field device (50 cm × 50 cm × 37 cm), which consists of black floor and Plexiglas wall. By employing a video camera and the EthoVision™ XT 7.0 software (Noldus, Wageningen, Netherlands), the behaviors of experimental animals were recorded and analyzed. The chamber was cleaned thoroughly with 75% ethanol after each trial to avoid olfactory cues. The total traveling distance and speed were recorded and analyzed. All of the OFT were performed on POD 3.

2.4 Morris water maze (MWM)

The spatial memory performance of mice was assessed by MWM as described previously[16]. Briefly, mice were first placed in the water maze pool to adapt to the environment for 2 days. During the training stage, mice were released from three different quadrants and trained to find the hidden platform for five consecutive days and the time taken (latency) to find the hidden platform was recorded. On the sixth day of MWM test, the hidden platform was removed, and mice were set free to swim at the starting point for 60 s to test their memory (target quadrant traveling time and platform-crossing times were recorded). The MWM was performed on POD 3 and POD 7.

2.5 Experimental protocol

The mice were randomized into one of four experimental groups: (1) normal control group; (2) galectin-1 group; (3) surgery group and (4) galectin-1 + surgery group. The mice in the galectin-1 and galectin-1 + surgery groups received an intraperitoneal injection of galectin-1 (0.5 mg/kg, PeproTech, USA) diluted in 0.1 ml of sterile saline 30 min prior the surgery. The dose (0.5 mg/kg) of galectin-1 in vivo was chosen based on previous studies[17, 18]. We harvested peripheral blood (by heart puncture) and hippocampus 6 h after surgery to assess the acute inflammatory response. Moreover, on POD 1 and POD 7, we also obtained hippocampus for analyses of immunofluorescence and Western blot .

2.6 Culture of primary microglial cell

Primary microglial cells were isolated from newborn mice (postnatal day 0–2). In brief, mice were euthanized and the brains were isolated in HBSS (H9269, Sigma), supplemented with 1% DNase (EN0521, Thermo) and 2.5% trypsin (15090-046, Gibco) for 15 min at 37 °C. Cells were centrifuged at 1000 × g for 5 min. The suspensions were incubated in the poly-L-lysine-pretreated flasks at a density of 1 × 106 and incubated until 90% confluent. The flask was shaken for 5 h at 180 rpm and centrifuged, so that we can collect the floating cells. Then, we suspended the pelleted cells and cultured them in DMEM/F12, which contained 1 mM of sodium pyruvate and 10% FBS in a 6-cm dish precoated with poly l-lysine ( P8920, Sigma-Aldrich) for 3 days to obtain the primary microglia.

2.7 BV2 cells

The murine microglial BV2 cell line was purchased from National Infrastructure of Cell Line Resource (China) and was cultured in Dulbecco’s modified Eagle’s medium (DMEM, ThermoFisher, USA) supplemented with 10% fetal bovine serum (FBS), 0.1% penicillin/streptomycin at 37 °C in a 5% CO2 humidified incubator. The BV2 cells were treated with LPS (100 ng/mL, Sigma, China) with or without galectin-1 (5 µg/ml). The expression levels of inflammatory proteins and signaling molecules (c-Jun and p65) in BV2 cells 24 h after LPS treatment were detected by ELISA and western blotting. Galectin-1 was administrated 30 min before LPS stimulation for all in vitro experiments.

2.8 Enzyme-linked Immunosorbent assay (ELISA)

The level of IL-1β, IL-6 and TNF-α in peripheral blood, hippocampus and cell supernatants were measured by ELISA kits (R&D Systems, MN) in strict accordance with manufacturer’s instructions.

2.9 Quantitative real-time PCR (qRT-PCR)

Total RNA was extracted from cultured cells in accordance with the manufacture’s protocol (Invitrogen, USA). By employing Superscript First-Strand cDNA Synthesis Kit (Invitrogen, USA), total RNA (1 µg ) was reverse transcribed into cDNAs. SYBR Premix Ex Taq II kit (TAKARA, Japan) on LightCycler 480 System (Roche, Switzerland) was used to performed qRT-PCR. The oligonucleotide sequences of the qRT-PCR primers are listed in Table S1.

2.10 Plasmid and Transfection

For overexpression of the IRAK1, BV2 cells were transfected with the plasmid pcDNA-3.1-IRAK1 (Hanheng Biotechnology, China) using Lipofectamine 2000 (Invitrogen, USA) according to the manufacturer’s instructions. Plasmid pcDNA-3.1 was used as a control. In brief, BV2 cells were seeded in DMEM (10% FBS) and cultured until them until achieved 80% confluence. We then replaced the culture medium with low-serum media (0.5% FBS), and then BV2 cells were transfected with 10 µg/well of plasmid. Cells were collected for further study at 48 h post-transfection. The efficiency of IRAK1 overexpression was measured qRT-PCR as indicated in Fig. 8E.

2.11 Nitrite assay

Griess assay was employed to detect the accumulation of nitrite (NO2) in cultured supernatant. Microglial cells were first planted into 96-well plates at the density of 5 × 104 cells/well, and then the cells were treated with LPS (100 ng/ml) for 24 h. Next, 50 µl Griess reagent and 50 µl culture supernatant fluids were mixed together for 15 min at 37 °C, and the absorbance was determined at 540 nm.

2.12 Luciferase reporter assay

Luciferase reporter assay was conducted as previously described. In brief, by employing Lipo-fectamine 3000 reagent (Invitrogen), BV2 cells were cotransfected with Renilla luciferase plasmid (pRL-SV40-C) and AP-1 luciferase reporter plasmid or NF-κB luciferase reporter plasmid for 48 h. Then, the BV2 cells were treated with LPS (100 ng/mL) for 24 h with or without galectin-1 (5 µg/ml). Reporter activity was detected using Dual Luciferase Reporter Gene Assay Kit (Beyotime, China) according to the manufacturer’s instructions.

2.13 MTT cell viability assay

MTT assay was used to measure the cell viability in strict accordance with the manufacturer’s instruction. In brief, cells administered with LPS for 6 h with or without galectin-1, were seeded at the density of 1 × 104 cells/well in 96-well plates and cultured for 24 h at 37 °C. MTT (Sigma-Aldrich, M2128) was added to the cells at a final concentration of 0.5 mg/mL, and the cells were incubated at 37℃ for another 4 h. Finally, cells were washed with PBS for three times and formazan crystals were dissolved in 100 mL DMSO. The absorbance was read at 490 nM using a microplate reader (PerkinElmer, USA) .

2.14 Western blot

Proteins in cell culture media and hippocampal tissues were lysed in RIPA lysis buffer with protease inhibitor cocktail. Protein samples were subjected to 10% SDS-PAGE and transferred to PVDF membranes. The membranes were subsequently incubated with antibodies iNOS (Abcam #ab178945, USA, 1:500), IL-1β (Abcam #ab234437, 1:500), Il-6 (Abcam #ab233706, 1:500), TNF-α (Abcam #ab215188, USA, 1:500), c-Jun (CST #9165, USA, 1:500), p65(CST #8242, USA, 1:500), IRAK1 (CST #4504, USA, 1:500), β-actin (CST #3700, USA, 1:2000). Then the membranes were incubated with HRP-conjugated secondary antibodies for 1 h at room temperature and detected with enhanced chemiluminescence. The results were analyzed by using ImageJ software (National Institutes of Health, USA).

2.15 Immunofluorescence

The mice were sacrificed, and their brains were fixed in 4% paraformaldehyde at 4 °C for 24 h and then embedded in paraffin. The brain tissues were cut into 10-µm-thick slices. The slices were blocked with 10% donkey serum for 1 h, incubated with anti-Iba-1 (Abcam, #ab178847, 1:200) or anti-NeuN (CST #24307, USA, 1:200) antibodies overnight at 4 °C. After washing with PBS solution for three times, sections were incubated with secondary antibodies at room temperature for 1 h. After being washed, the sections were incubated with DAPI for nuclear staining. By using ImageJ software (National Institutes of Health, USA), Iba-1-positive cells and NeuN-positive cells were counted with a DAPI counterstain.

2.16 Statistical analysis

The sample size was calculated by using the SPSS 16.0 software (SPSS, Chicago, USA) to achieve an 80% power at a significance level of 0.05. Two‒way ANOVA was employed to evaluate latency in the MWM test and student's t test was used to assess the differences between two groups. Whether the data were normally distributed is detected by the Shapiro-Wilk normality test. The GraphPad Prism software (version 7.0, CA, USA) was employed to perform the statistical analyses. In all cases, statistical significance was accepted at P < 0.05.

3. Results

3.1 Galectin-1 ameliorated hippocampus-dependent cognitive dysfunction induced by surgery

To investigate the physiological role of galectin-1 on cognitive dysfunction induced by surgery, OFT was first performed to test the locomotor activity on postoperative day 3 (POD 3). As shown by the results of OFT, no significant change was observed in duration in center (Fig. 1A, P > 0.05), distance travel (Fig. 1B, P > 0.05) and travelling speed (Fig. 1C, P > 0.05) among the four groups, indicating that no spontaneous locomotor activity decline was caused by the surgery.

Then, on POD 3 and POD 7, the hippocampus-dependent memory was evaluated by WWM. As show by the results of training session in WWM, no remarkable difference was observed in escape lantency (Fig. 1E, P > 0.05) and average swimming speed (Fig. 1F, P > 0.05) among the four groups, suggesting that the mice in the study have learned to find the hidden platform after training for five consecutive days. During the probe session, the time spent in the target quardrant (Fig. 1G, P < 0.05) and the number of platform crossing (Fig. 1H, P < 0.05) significantly decreased in surgery group on both POD 3 and POD 7, when compared to control group, indicating surgery had induced cognitive dysfunction. However, when pretreated with galectin-1, the decline in cognitive dysfunction induced by surgery (Fig. 1G&1H, P < 0.05) on both POD 3 and POD 7 was rescued, demonstrating that galectin-1 may elicit protective effect on the cognitive impairment induced by surgery.

3.2 Galectin-1 mitigated systemic inflammation and neuroinflammation induced by surgery

After surgery, we then collected the peripheral blood and the hippocampus sample to test the acute inflammatory response. In peripheral blood, the expression levels of IL-1β, IL-6 and TNF-α significantly increase 6 h after surgery, while pretreated with galectin-1 remarkably decreased the surgery-induce elevation of these inflammatory proteins (Fig. 2A-2C, P < 0.05). In term of neuroinflammation in hippocampus, analogical results were observed: galectin-1 significantly rescued the upregulated inflammatory factors (including IL-1β, IL-6 and TNF-α) induced by surgery in the hippocampus 6 h after surgery (Fig. 2D-2F, P < 0.05). The expression levels of inflammatory cytokine in hippocampus remarkably elevated in the surgery group in comparison with the normal group. Nevertheless, on POD 1, pretreatment with galectin-1 attenuated the expression levels of IL-1β, IL-6 and TNF-α in hippocampus (Fig. 2D-2F, P < 0.05). On POD 7, no significant difference was observed in the expression levels of inflammatory cytokine among the four groups (Fig. 2D-2F, P > 0.05), indicating that the inflammation in the aged mice might have abated as time goes on. All of these results suggested that galectin-1 might inhibit surgery-induced systemic inflammation and neuroinflammation.

3.3 Pretreatment with galectin-1 suppressed microglia activation in the hippocampus

Since microglia activation may promote neuroinflammation in the process of PND, we measured the microglial activation through immunofluorescence marked by ionized calcium-binding adaptor molecule 1 (Iba-1) on POD 7. The average number of Iba1-positive cells per field in CA1 region (Fig. 3A&3C, P < 0.05) and DG region (Fig. 3B&3D, P < 0.05) was significantly increased after surgical trauma. However, pretrement with galectin-1 remarkably reversed these elevation (Fig. 3A-3D, P < 0.05), suggesting that galectin-1 may suppress the activation of microglial and attenuate the ensuing inflammatory response.

3.4 Galectin-1 protects neurons from inflammatory cytokines induced by surgery

Since neuroinflammation exerts detrimental effect on neurons, we then tested the neuronal integrity among the four groups using a NeuN antibody, which indicates the neuronal integrity. As shown by the results of immunofluorescence, the number of NeuN-positive cells in CA1 region (Fig. 4A&4B, P < 0.05) and DG region (Fig. 4C, P < 0.05) remarkably decreased after treating with LPS, while pretreatment with galectin-1 significantly rescued the reduction caused by LPS, compared with the control group in CA1 region and DG region. Therefore, we suggested that galectin-1 protected neurons from LPS-induced inflammatory cytokines in vivo.

3.5 Galectin-1 reduced NO release induced by LPS through elevating the expression of iNOS.

To further explore the protective effect of galectin-1 in PND, we then examine the role of galectin-1 in vitro. BV2 cell treated with LPS (100 ng/mL) was used as a neuroinflammation model, which is a common approach studying PND[19, 20]. By employing nitrite assays, the effect of galectin-1 on microglial activation was determined. There is no significant difference among the four groups on cell viability (Fig. 5A, P > 0.05). Compared with the control group, the production of nitric oxide (NO) significantly increased in BV2 cells treated with LPS (Fig. 5B, P < 0.05). However, galectin-1 treatment reversed this elevation induced by LPS (Fig. 5B, P < 0.05). Inducible nitric oxide synthase (iNOS), induced in microglia by inflammatory mediators such as LPS and cytokines, contribute to the release of NO [21]. Consequently, we used iNOS production to evaluate whether galectin-1 affects the release of NO. As shown by the results, galectin-1 significantly decreased the mRNA (Fig. 5C, P < 0.05) and protein (Fig. 5D&5E, P < 0.05) expression levels of iNOS, when compared with the LPS group, suggesting that galectin-1 mitigated LPS-induced NO release through reducing the expression of iNOS.

3.6 Galectin-1 decreased LPS-induced inflammatory cytokines production.

We then assessed the effect of galectin-1 on proinflammatory cytokines to explore the influence of galectin-1 on the inflammatory response. The expression levels of the pro-inflammatory cytokines (IL-1β, IL-6 and TNF-α) were measured in BV2 cells. When exposed to LPS, the mRNA production of IL-1β, IL-6 and TNF-α show remarkable increase, but galectin-1 administration significantly reduced the elevated mRNA levels of IL-1β, IL-6 and TNF-α (Fig. 6A-6C, P < 0.05). Moreover, we tested the protein expression of IL-1β, IL-6 and TNF-α 24 h after LPS treatment. When administrated with LPS, the protein levels of IL-1β, IL-6 and TNF-α remarkably elevated among the four groups, but galectin-1 treatment reduced the LPS-induced increased protein level of IL-1β, IL-6 and TNF-α (Fig. 6D-6G, P < 0.05). In addition, the secreted protein levels of IL-1β and TNF-α were assayed 24 h after administration with LPS by ELISA assay, and the results are in accord with the results presented above. After LPS administration, the expression levels of secreted IL-1β and TNF-α remarkably increased, while galectin-1 significantly decreased the elevation induced by LPS (Fig. 6H&6I, P < 0.05).

3.7 Galectin-1 attenuated the transcriptional activity and translocation of c-Jun and NF-κB p65

The proteins of the MAPK and NF-κB pathways are the main modulator in pro-inflammatory cytokine production and secretion[22, 23]. According to the results of NO release and cytokine production, whether galectin-1 affects cytokine production controlled by MAPK-dependent AP-1 transcription factors and NF-κB p65 was explored. The nuclei of BV2 cells were separated from the cytoplasm after exposing to LPS for 30 min. Stimulated by LPS, the translocation of c-Jun and NF-κB p65 from the cytoplasm to the nucleus showed remarkable increase in comparison with the control group (Fig. 7A-7F, P < 0.05). However, pretreatment with galectin-1 rescued the elevation induced by LPS (Fig. 7A-7F, P < 0.05). Furthermore, we then tested the transcriptional activity of c-Jun and NF-κB p65 in BV2 cells. Prereatment with galectin-1 significantly attenuated the increased transcriptional activity of c-Jun (Fig. 7G, P < 0.05) and NF-κB p65 (Fig. 7H, P < 0.05) following LPS stimulation. Our finding suggest that galectin-1 decreases the release of NO and inflammatory cytokine by attenuating the transcriptional activity and translocation of c-Jun and NF-κB p65.

3.8 Overexpressing IRAK1 rescued the protective effect on the production of inflammatory factors exerted by galectin-1

We then explored the mechanism by which galectin-1 attenuated the transcriptional activity and translocation of c-Jun and NF-κB p65. As shown by our results, the protein expression level of IRAK1 showed significant reduction in BV2 cells when treated with galectin-1 (Fig. 8A&8B, P < 0.05). Moreover, the mRNA expression of IRAK1 in BV2 cells (Fig. 8C, P < 0.05) and primary microglia (Fig. 8D, P < 0.05) also showed significant reduction when pretreated with galectin-1.

Next, pcDNA-3.1-IRAK1 (Fig. 8E) were transfected in BV cells for 24 h to further determine whether galectin-1 decreased cytokine production through IRAK1. We separated the nuclei from the cytoplasm after the BV2 cells were treated with LPS for 30 min. As indicated by our results, the translocation of c-Jun and NF-κB p65 from the cytoplasm to the nucleus significantly suppressed when pretreated with galectin-1, while this kind of protective effect was rescued when transfected with pcDNA-3.1-IRAK1 (Fig. 8F-8K, P < 0.05). Then we detected whether overexpressing IRAK1 could rescue the galectin-1-mediated ameliorating effect on inflammatory factors production. After the BV2 cells were transfected with pcDNA-3.1-IRAK1 for 24 h followed by stimulation with LPS for 30 min, the supernatants were collected and analyzed by ELISA. The galectin-1induced decreased pro-inflammatory cytokines of IL-1β (Fig. 8L, P < 0.05) and TNF-α (Fig. 8M, P < 0.05) was rescued when overexpressing IRAK1. These results indicated that galectin-1 might attenuate the inflammatory response via inhibiting IRAK1 expression.

4. Discussion

Emerging emphasis has been put on the role of neuroinflammation in the progression of PND[24, 25]. Characterized by producing inflammatory mediators that facilitate the development and severity of PND, microglia play a crucial role in the progression of PND[26, 27]. In a mouse model of PND, we investigate the correlation between galectin-1 treatment and neurocognitive dysfunction. Our findings suggest that treatment with galectin-1 attenuates the surgery-induced neuroinflammation and hippocampus-dependent cognitive dysfunction. By suppressing the activation of microglial effectively, galectin-1 decreased the translocation of c-Jun and NF-κB p65, inhibited the inflammatory response, and thus ameliorating the ensuing neurocognitive dysfunction. This protective effects of galectin-1 seemed to depend on inhibiting IRAK1 expression.

PND induced by surgery are relatively common, which occurs in 10%-54% of patients underwent surgery in the first few weeks[28]. However, the postoperative neurocognitive complications may cause chronic cognitive dysfunction and higher risk of mortality, which not only happen in the acute phase after surgery[29]. The occurrence of PND is affected by various factors, including education level, age, gender, etc. Advanced age is an independent risk factor among these factors suggested by clinical studies[30]. When exposed to systemic stress source such as surgery, many organ systems gradually deteriorate in the process of aging, including the brain[31]. Consequently, in the present study, we chose aged mice to imitate the clinical condition of PND.

Exploratory laparotomy under the anesthesia of isoflurane is a common approach to induce PND in aged mice[32, 33], which simulates clinical status closely. Therefore, this approach was chosen to establish the animal model of PND. The surgical trauma caused remarkable neurocognitive dysfunction as indicated by our results of OFT and MWM test. It has been reported that isoflurane alone may induce cognitive dysfunction [34, 35]. The underlying neurotoxicity of isoflurane include elevating ROS production, inducing neuroinflammation[36], and disrupting calcium homeostasis [37].

Microglia is one of the critical facilitators in inducing neuroinflammation, a hallmark of PND. Peripheral inflammation induced by the surgical trauma may activate the microglia staying at a resting condition, contributing to the production of inflammatory cytokines into the CNS and thus leading to neuroinflammation. It has been reported that suppressing the microglia activation inhibited the expression of pro-inflammatory cytokines, such as TNF-α and IL-1β, thereby mitigating neuroinflammation and neurocognitive impairment[38]. In our study, galectin-1 inhibited the neuroinflammation in hippocampus, and administrated with galectin-1 not only decreased the peripheral inflammatory cytokines and the protein expression of inflammatory cytokines, but also suppressed microglia activation and disruption of neuronal integrity after surgery in the hippocampus.

In the present study, galectin-1 attenuated the expression of pro-inflammatory cytokine induced by LPS in BV2 cells. The expression of IL-1β, IL-6, TNF-α, and iNOS is harmfully correlated with the development of neurodegenerative diseases[39, 40]. Suggested by previous studies, using small molecule inhibitors to modulate TNF-α has the potential for preventing and treating Alzheimer’s disease[41], and the increased expression level of iNOS in activated microglia contributes to neurodegeneration[42]. These findings revel that galectin-1 might exert protective effects on neuroinflammation in neurocognitive disorders.

Suggested by mounting evidence, IRAKs are important mediators in metabolic and inflammatory diseases[43, 44]. IRAK1 deficiency inhibits early pro-inflammatory cytokine production following polymicrobial sepsis and affects multiple TLR-dependent pathways[45], demonstrating that inhibiting IRAK1 expression may be therapeutic. IRAK1 plays a key role in signal cascade caused by ligand binding to TLRs and IL-1R[46, 47]. In the present study, we showed that galectin-1 decreased the protein expression of IRAK1, augmenting the NF-κB activity, and the expression level of its target genes, including TNFα and IL-1β [48, 49].

Since IL-1β and TNF-α get involved in neuronal survival, their production induced by surgical trauma might lead to neuronal death or morphological abnormalities in the hippocampus, and disrupt cognitive function in aged mice. Thus, the effect of galectin-1 exerting on the hippocampal neurons following neuroinflammation caused by surgery was explored. As suggested by the results in the study, the number of NeuN-positive cells significantly reduced after surgery[50]. Galectin-1 administration significantly alleviated the disruption of neuronal integrity induced by surgical trauma. This result may account for the phenomenon that galectin-1 improved memory deficits after surgery. Furthermore, the therapeutic potential of retarding the translocation of c-Jun and NF-κB p65 from cytoplasm to nucleus is emphasized in our study, which attenuated the inflammatory response. Collectively, inflammatory mediators, which derived from the activation of AP-1 and NF-κB, might mediate in neuronal integrity and neurocognitive dysfunction. The crucial role of the galectin-1 in surgery-induced PND demonstrates that treatment blocking the microglia activity, neuronal integrity and surgery-mediated inflammatory cytokines could retard the progression of neurocognitive dysfunction.

5. Conclusions

In summary, galectin-1 administration attenuated surgery-induced neuroinflammation and cognitive dysfunction by decreasing IRAK1 expression. Conversely, overexpressing IRAK1 contributed to increased transcriptional activity and translocation of c-Jun and NF-κB p65 in microglia BV2 cells. All together, our findings demonstrate that galectin-1 alleviates inflammatory response and exerts an ameliorating effect in surgery-induced neurocognitive disorders, which might be a potential therapeutic for decelerating the progression of PND.

Declarations

6.1 Ethics Approval and consent to participate

No applicable

6.2 Consent for Publication

No applicable

6.3 Availability of data and materials

All data and models generated or used during the study appear in the submitted article.

6.4 Competing interest

The authors declare that they have no competing interests

6.5 Funding

This work was supported by the Natural Science Foundation of Guangdong Province, China (No.2016A030313827, No.2018A0303130272); the Science and Technology Planning Project of Guangzhou, China (No. 201707010207).

6.6 Authors’ contributions

Yi Wu, Wen Song and Zhiwen Shen: Investigation, Methodology, Writing-Original draft preparation. Yaowei Zhi: Conceptualization, Data curation. Guiyun Wu: Visualization, Software, Supervision. Yujuan Li and Junhua Li: Writing-Reviewing and Editing.

6.7 Acknowledgement

Not applicable

References

  1. Evered L, Silbert B, Knopman DS, Scott DA, DeKosky ST, Rasmussen LS, Oh ES, Crosby G, Berger M and Eckenhoff RG. Recommendations for the Nomenclature of Cognitive Change Associated with Anaesthesia and Surgery-20181. J Alzheimers Dis. 2018; 66(1):1-10.10.3233/JAD-189004 PMID:30347621
  2. Deo H, West G, Butcher C and Lewis P. The prevalence of cognitive dysfunction after conventional and computer-assisted total knee replacement. Knee. 2011; 18(2):117-120.10.1016/j.knee.2010.03.006 PMID:20615709
  3. Skvarc DR, Berk M, Byrne LK, Dean OM, Dodd S, Lewis M, Marriott A, Moore EM, Morris G, Page RS and Gray L. Post-Operative Cognitive Dysfunction: An exploration of the inflammatory hypothesis and novel therapies. Neurosci Biobehav Rev. 2018; 84:116-133.10.1016/j.neubiorev.2017.11.011 PMID:29180259
  4. Li D, Chen M, Meng T and Fei J. Hippocampal microglial activation triggers a neurotoxic-specific astrocyte response and mediates etomidate-induced long-term synaptic inhibition. J Neuroinflammation. 2020; 17(1):109.10.1186/s12974-020-01799-0 PMID:32264970
  5. Wen C, Xie T, Pan K, Deng Y, Zhao Z, Li N, Bian J, Deng X and Zha Y. Acetate attenuates perioperative neurocognitive disorders in aged mice. Aging (Albany NY). 2020; 12(4):3862-3879.10.18632/aging.102856 PMID:32139660
  6. Zhang ZJ, Zheng XX, Zhang XY, Zhang Y, Huang BY and Luo T. Aging alters Hv1-mediated microglial polarization and enhances neuroinflammation after peripheral surgery. Cns Neurosci Ther. 2020; 26(3):374-384.10.1111/cns.13271 PMID:31774629
  7. Ni P, Dong H, Wang Y, Zhou Q, Xu M, Qian Y and Sun J. IL-17A contributes to perioperative neurocognitive disorders through blood-brain barrier disruption in aged mice. J Neuroinflammation. 2018; 15(1):332.10.1186/s12974-018-1374-3 PMID:30501622
  8. Burm SM, Zuiderwijk-Sick EA, T JA, van der Putten C, Veth J, Kondova I and Bajramovic JJ. Inflammasome-induced IL-1β secretion in microglia is characterized by delayed kinetics and is only partially dependent on inflammatory caspases. J Neurosci. 2015; 35(2):678-687.10.1523/JNEUROSCI.2510-14.2015 PMID:25589762
  9. Zheng JW, Meng B, Li XY, Lu B, Wu GR and Chen JP. NF-κB/P65 signaling pathway: a potential therapeutic target in postoperative cognitive dysfunction after sevoflurane anesthesia. Eur Rev Med Pharmacol Sci. 2017; 21(2):394-407 PMID:28165545
  10. Zhang X, Dong H, Li N, Zhang S, Sun J, Zhang S and Qian Y. Activated brain mast cells contribute to postoperative cognitive dysfunction by evoking microglia activation and neuronal apoptosis. J Neuroinflammation. 2016; 13(1):127.10.1186/s12974-016-0592-9 PMID:27245661
  11. Wada M, Ono S, Kadoya T, Kawanami T, Kurita K and Kato T. Decreased galectin-1 immunoreactivity of the skin in amyotrophic lateral sclerosis. J Neurol Sci. 2003; 208(1-2):67-70.10.1016/s0022-510x(02)00424-0 PMID:12639727
  12. McGraw J, Gaudet AD, Oschipok LW, Kadoya T, Horie H, Steeves JD, Tetzlaff W and Ramer MS. Regulation of neuronal and glial galectin-1 expression by peripheral and central axotomy of rat primary afferent neurons. Exp Neurol. 2005; 195(1):103-114.10.1016/j.expneurol.2005.04.004 PMID:15893752
  13. Almkvist J and Karlsson A. Galectins as inflammatory mediators. Glycoconj J. 2002; 19(7-9):575-581.10.1023/B:GLYC.0000014088.21242.e0 PMID:14758082
  14. Barrientos RM, Hein AM, Frank MG, Watkins LR and Maier SF. Intracisternal interleukin-1 receptor antagonist prevents postoperative cognitive decline and neuroinflammatory response in aged rats. J Neurosci. 2012; 32(42):14641-14648.10.1523/JNEUROSCI.2173-12.2012 PMID:23077050
  15. Li J, Guo M, Liu Y, Wu G, Miao L, Zhang J, Zuo Z and Li Y. Both GSK-3β/CRMP2 and CDK5/CRMP2 pathways participate in the protection of dexmedetomidine against propofol-induced learning and memory impairment in neonatal rats. Toxicol Sci. 2019.10.1093/toxsci/kfz135 PMID:31187143
  16. Vorhees CV and Williams MT. Morris water maze: procedures for assessing spatial and related forms of learning and memory. Nat Protoc. 2006; 1(2):848-858.10.1038/nprot.2006.116
  17. Starossom SC, Mascanfroni ID, Imitola J, Cao L, Raddassi K, Hernandez SF, Bassil R, Croci DO, Cerliani JP, Delacour D, Wang Y, Elyaman W and Khoury SJ, et al. Galectin-1 deactivates classically activated microglia and protects from inflammation-induced neurodegeneration. Immunity. 2012; 37(2):249-263.10.1016/j.immuni.2012.05.023 PMID:22884314
  18. Li Y, Chen N, Wu C, Lu Y, Gao G, Duan C, Yang H and Lu L. Galectin-1 attenuates neurodegeneration in Parkinson's disease model by modulating microglial MAPK/IκB/NFκB axis through its carbohydrate-recognition domain. Brain Behav Immun. 2020; 83:214-225.10.1016/j.bbi.2019.10.015 PMID:31669519
  19. Zhang D, Li N, Wang Y, Lu W, Zhang Y, Chen Y, Deng X and Yu X. Methane ameliorates post-operative cognitive dysfunction by inhibiting microglia NF-κB/MAPKs pathway and promoting IL-10 expression in aged mice. Int Immunopharmacol. 2019; 71:52-60.10.1016/j.intimp.2019.03.003 PMID:30877874
  20. Lee JH, Kam EH, Kim SY, Cheon SY, Kim EJ, Chung S, Jeong JH and Koo BN. Erythropoietin Attenuates Postoperative Cognitive Dysfunction by Shifting Macrophage Activation toward the M2 Phenotype. Front Pharmacol. 2017; 8:839.10.3389/fphar.2017.00839 PMID:29201007
  21. Nam HY, Nam JH, Yoon G, Lee JY, Nam Y, Kang HJ, Cho HJ, Kim J and Hoe HS. Ibrutinib suppresses LPS-induced neuroinflammatory responses in BV2 microglial cells and wild-type mice. J Neuroinflammation. 2018; 15(1):271.10.1186/s12974-018-1308-0 PMID:30231870
  22. Wang T, Jin X, Liao Y, Sun Q, Luo C, Wang G, Zhao F and Jin Y. Association of NF-κB and AP-1 with MMP-9 Overexpression in 2-Chloroethanol Exposed Rat Astrocytes. Cells-Basel. 2018; 7(8).10.3390/cells7080096 PMID:30087244
  23. Deng Z, Sui G, Rosa PM and Zhao W. Radiation-induced c-Jun activation depends on MEK1-ERK1/2 signaling pathway in microglial cells. Plos One. 2012; 7(5):e36739.10.1371/journal.pone.0036739 PMID:22606284
  24. Subramaniyan S and Terrando N. Neuroinflammation and Perioperative Neurocognitive Disorders. Anesth Analg. 2019; 128(4):781-788.10.1213/ANE.0000000000004053 PMID:30883423
  25. Saxena S, Lai IK, Li R and Maze M. Neuroinflammation is a putative target for the prevention and treatment of perioperative neurocognitive disorders. Br Med Bull. 2019; 130(1):125-135.10.1093/bmb/ldz010 PMID:31049563
  26. Wang DS, Terrando N and Orser BA. Targeting microglia to mitigate perioperative neurocognitive disorders. Br J Anaesth. 2020.10.1016/j.bja.2020.06.027 PMID:32654743
  27. Chen YN, Sha HH, Wang YW, Zhou Q, Bhuiyan P, Li NN, Qian YN and Dong HQ. Histamine 2/3 receptor agonists alleviate perioperative neurocognitive disorders by inhibiting microglia activation through the PI3K/AKT/FoxO1 pathway in aged rats. J Neuroinflammation. 2020; 17(1):217.10.1186/s12974-020-01886-2 PMID:32698899
  28. Androsova G, Krause R, Winterer G and Schneider R. Biomarkers of postoperative delirium and cognitive dysfunction. Front Aging Neurosci. 2015; 7:112.10.3389/fnagi.2015.00112 PMID:26106326
  29. Alam A, Hana Z, Jin Z, Suen KC and Ma D. Surgery, neuroinflammation and cognitive impairment. Ebiomedicine. 2018; 37:547-556.10.1016/j.ebiom.2018.10.021 PMID:30348620
  30. Tasbihgou SR and Absalom AR. Postoperative neurocognitive disorders. Korean J Anesthesiol. 2020.10.4097/kja.20294 PMID:32623846
  31. Cunningham C and Maclullich AM. At the extreme end of the psychoneuroimmunological spectrum: delirium as a maladaptive sickness behaviour response. Brain Behav Immun. 2013; 28:1-13.10.1016/j.bbi.2012.07.012 PMID:22884900
  32. Fu Q, Li J, Qiu L, Ruan J, Mao M, Li S and Mao Q. Inhibiting NLRP3 inflammasome with MCC950 ameliorates perioperative neurocognitive disorders, suppressing neuroinflammation in the hippocampus in aged mice. Int Immunopharmacol. 2020; 82:106317.10.1016/j.intimp.2020.106317 PMID:32087497
  33. Qiu LL, Luo D, Zhang H, Shi YS, Li YJ, Wu D, Chen J, Ji MH and Yang JJ. Nox-2-Mediated Phenotype Loss of Hippocampal Parvalbumin Interneurons Might Contribute to Postoperative Cognitive Decline in Aging Mice. Front Aging Neurosci. 2016; 8:234.10.3389/fnagi.2016.00234 PMID:27790135
  34. Cui H, Xu Z and Qu C. Tetramethylpyrazine ameliorates isoflurane-induced cognitive dysfunction by inhibiting neuroinflammation via miR-150 in rats. Exp Ther Med. 2020; 20(4):3878-3887.10.3892/etm.2020.9110 PMID:32855738
  35. Que YY, Zhu T, Zhang FX and Peng J. Neuroprotective effect of DUSP14 overexpression against isoflurane-induced inflammatory response, pyroptosis and cognitive impairment in aged rats through inhibiting the NLRP3 inflammasome. Eur Rev Med Pharmacol Sci. 2020; 24(12):7101-7113.10.26355/eurrev_202006_21704 PMID:32633405
  36. Matta JA, Cornett PM, Miyares RL, Abe K, Sahibzada N and Ahern GP. General anesthetics activate a nociceptive ion channel to enhance pain and inflammation. Proc Natl Acad Sci U S A. 2008; 105(25):8784-8789.10.1073/pnas.0711038105 PMID:18574153
  37. Tong BC, Wu AJ, Li M and Cheung KH. Calcium signaling in Alzheimer's disease & therapies. Biochim Biophys Acta Mol Cell Res. 2018; 1865(11 Pt B):1745-1760.10.1016/j.bbamcr.2018.07.018 PMID:30059692
  38. Feng X, Valdearcos M, Uchida Y, Lutrin D, Maze M and Koliwad SK. Microglia mediate postoperative hippocampal inflammation and cognitive decline in mice. JCI Insight. 2017; 2(7):e91229.10.1172/jci.insight.91229 PMID:28405620
  39. Smith JA, Das A, Ray SK and Banik NL. Role of pro-inflammatory cytokines released from microglia in neurodegenerative diseases. Brain Res Bull. 2012; 87(1):10-20.10.1016/j.brainresbull.2011.10.004 PMID:22024597
  40. Voet S, Srinivasan S, Lamkanfi M and van Loo G. Inflammasomes in neuroinflammatory and neurodegenerative diseases. Embo Mol Med. 2019; 11(6).10.15252/emmm.201810248 PMID:31015277
  41. Gabbita SP, Srivastava MK, Eslami P, Johnson MF, Kobritz NK, Tweedie D, Greig NH, Zemlan FP, Sharma SP and Harris-White ME. Early intervention with a small molecule inhibitor for tumor necrosis factor-α prevents cognitive deficits in a triple transgenic mouse model of Alzheimer's disease. J Neuroinflammation. 2012; 9:99.10.1186/1742-2094-9-99 PMID:22632257
  42. Verma M, Beaulieu-Abdelahad D, Ait-Ghezala G, Li R, Crawford F, Mullan M and Paris D. Chronic Anatabine Treatment Reduces Alzheimer's Disease (AD)-Like Pathology and Improves Socio-Behavioral Deficits in a Transgenic Mouse Model of AD. Plos One. 2015; 10(5):e128224.10.1371/journal.pone.0128224 PMID:26010758
  43. Kinsella S, König HG and Prehn JH. Bid Promotes K63-Linked Polyubiquitination of Tumor Necrosis Factor Receptor Associated Factor 6 (TRAF6) and Sensitizes to Mutant SOD1-Induced Proinflammatory Signaling in Microglia. eNeuro. 2016; 3(2).10.1523/ENEURO.0099-15.2016 PMID:27257617
  44. Mendonca P, Taka E, Bauer D, Cobourne-Duval M and Soliman KF. The attenuating effects of 1,2,3,4,6 penta-O-galloyl-β-d-glucose on inflammatory cytokines release from activated BV-2 microglial cells. J Neuroimmunol. 2017; 305:9-15.10.1016/j.jneuroim.2017.01.011 PMID:28284353
  45. Chandra R, Federici S, Bishwas T, Németh ZH, Deitch EA, Thomas JA and Spolarics Z. IRAK1-dependent signaling mediates mortality in polymicrobial sepsis. Inflammation. 2013; 36(6):1503-1512.10.1007/s10753-013-9692-1 PMID:23856940
  46. Cohen P and Strickson S. The role of hybrid ubiquitin chains in the MyD88 and other innate immune signalling pathways. Cell Death Differ. 2017; 24(7):1153-1159.10.1038/cdd.2017.17 PMID:28475177
  47. Ghosh S and Dass J. Study of pathway cross-talk interactions with NF-κB leading to its activation via ubiquitination or phosphorylation: A brief review. Gene. 2016; 584(1):97-109.10.1016/j.gene.2016.03.008 PMID:26968890
  48. Gao M, Wang X, Zhang X, Ha T, Ma H, Liu L, Kalbfleisch JH, Gao X, Kao RL, Williams DL and Li C. Attenuation of Cardiac Dysfunction in Polymicrobial Sepsis by MicroRNA-146a Is Mediated via Targeting of IRAK1 and TRAF6 Expression. J Immunol. 2015; 195(2):672-682.10.4049/jimmunol.1403155 PMID:26048146
  49. Boldin MP, Taganov KD, Rao DS, Yang L, Zhao JL, Kalwani M, Garcia-Flores Y, Luong M, Devrekanli A, Xu J, Sun G, Tay J and Linsley PS, et al. miR-146a is a significant brake on autoimmunity, myeloproliferation, and cancer in mice. J Exp Med. 2011; 208(6):1189-1201.10.1084/jem.20101823 PMID:21555486
  50. Seong KJ, Lee HG, Kook MS, Ko HM, Jung JY and Kim WJ. Epigallocatechin-3-gallate rescues LPS-impaired adult hippocampal neurogenesis through suppressing the TLR4-NF-κB signaling pathway in mice. Korean J Physiol Pharmacol. 2016; 20(1):41-51.10.4196/kjpp.2016.20.1.41 PMID:26807022