International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Fluid and blood transfusion-based resuscitation after hemorrhagic shock (HSR) can result in damage to multiple organs (1,2). Acute lung injury (ALI) is common and severe (3,4). However, the treatment options for managing lung injury are currently limited and primarily involve ventilation volume and positional changes (5,6). Thus, effective pharmacological treatments are limited (6–8). Our previous research demonstrated an increase in inflammatory cytokines and exacerbated apoptosis in the HSR-ALI model (9,10). In patients with HSR, tissue hypoxia induces an early inflammatory response characterized by the release of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α). This promotes neutrophil infiltration and vascular endothelial disruption. During reperfusion, the reintroduction of oxygen leads to the generation of reactive oxygen and nitrogen species (ROS/RNS), which further exacerbate inflammation and apoptosis (11). These findings suggest that targeting inflammation, apoptosis, and ROS/RNS might offer potential therapeutic strategies for mitigating HSR-induced lung injury.
After hemorrhagic shock (HS), patients are exposed to a hyper-catecholaminergic state characterized by enhanced sympathetic nervous system activity (12). In particular, β1 adrenergic receptor activation leads to increased heart rate and myocardial contractility, which consequently elevates metabolic demand and causes an imbalance between oxygen supply and demand. This imbalance may exacerbate ischemia-reperfusion injury and trigger systemic inflammation (13). β1-adrenergic receptor stimulation has been reported to enhance the secretion of pro-inflammatory cytokines in human monocytes and induce apoptosis when stimulated by β1-selective agonists (14,15). Therefore, β1-blockade is considered a promising strategy for suppressing these detrimental responses. Notably, the β2 receptor pathway is preserved even with the use of β1 blockers. The β2 receptor pathway includes the 5′ adenosine monophosphate-activated protein kinase (AMPK) pathway, which contributes to sustaining homeostasis; reducing oxidative stress, mitigating mitochondrial dysfunction, suppressing inflammatory responses, and activating autophagy to maintain normal bodily functions (13,16,17).
Landiolol hydrochloride, a highly selective β1 blocker, has been employed to manage tachycardia and reduce the risk of atrial fibrillation during the perioperative phase and cardiac decompensation. The β1/β2 receptor selectivity ratio of landiolol (255) is substantially higher than those of commonly used β-blockers such as esmolol (33), atenolol (4.3), metoprolol (2.3), and propranolol (0.68), as shown in Table I (18,19). It also exhibits approximately eightfold greater β1-selectivity than does esmolol, with minimal activity on β2 receptors (18). Owing to this pharmacological profile, landiolol was selected in this study to evaluate its effects on lung injury following HSR. Although primarily used as an anti-arrhythmic drug, recent studies have reported that β1 blockers offer organ protection, including lung protection (20–25). These findings have been primarily observed in sepsis and ischemia-reperfusion models. In the lipopolysaccharide (LPS) model, landiolol treatment has been shown to enhance serum levels of TNF-α, interleukin (IL)-6, and high mobility group box 1 (HMGB1), along with reducing TNF-α expression in the liver (20,24). Conversely, some studies have reported no alterations in serum levels of TNF-α, IL-1β, and IL-6, or in TNF-α and IL-6 expression in the lungs, resulting in inconsistent findings (21,25).
In this study, landiolol was administered following HSR. To our knowledge, no prior research has examined the effects of landiolol on inflammatory cytokines and lung apoptosis in the HSR-ALI model. The study aimed to explore the potential of landiolol treatment after HSR for protecting lung tissue by decreasing inflammatory responses and apoptosis.
This research received approval from the Department of Animal Resources at the Advanced Science Research Center, Okayama University (OKU-2021247 on April 1, 2021 and OKU-2023436 on April 24, 2023; Okayama, Japan) and adhered to the Guidelines for the Care and Use of Laboratory Animals based on ARRIVE (26) and the 2020 AVMA euthanasia guidelines (27). Male Sprague-Dawley rats, weighing 350–430 g (Clea Japan, Inc., Tokyo, Japan), were kept in temperature-regulated rooms (25°C) with a 12-h light/dark cycle and had unrestricted access to water and food prior to the experiments. A total of sixty-nine rats were used in this study. Five rats were assigned to a control group without undergoing any procedures. Since there was no statistically significant difference between the control and sham groups, data of the control group were excluded from statistical analysis. Twenty-four rats were excluded from the analysis. Of these twenty-four rats, nineteen were excluded due to technical issues encountered during the HS procedure. These issues included failure to monitor blood pressure due to thrombosis, inability to achieve the target hypotensive state due to insufficient bleeding, or catheter dislodgement that prevented continuation of the experiment. All of these complications occurred during the HS procedure, and animals that could not proceed with the study were promptly euthanized. The other five rats died naturally from worsening hypotension during the HS procedure, despite attempts at resuscitation. The remaining forty rats successfully completed the experimental protocol and their data were included in the final analysis. These were divided equally into two groups: twenty in the 3-h model (n=5 each, four groups) and twenty in the 24-h model (n=5 each, four groups). All animals that successfully underwent the HSR procedure survived. The sample size was decided based on previous studies (28,29).
All rats were numbered sequentially upon arrival and housed under identical conditions in uniformly sized cages placed in the same location. All experimental procedures were consistently performed under the same environmental conditions and followed a predetermined order. Group assignment and drug preparation were carried out by H. Shimizu. R. Sakamoto was responsible for performing the HSR and sham procedures. Both R. Sakamoto and H. Shimizu jointly conducted sampling and data analysis. Information on group assignments was disclosed to R. Sakamoto only after sample collection completion. The rats were euthanized for sampling under 2–3% isoflurane anesthesia by performing a laparotomy and exsanguination via the abdominal aorta.
All rats included were confirmed to be healthy and alive before starting the experiment. Isoflurane was used throughout all procedures to provide adequate sedation and analgesia. Humane endpoints were predefined based on criteria such as ≥20% body weight loss or a marked decrease in physical activity. However, none of the animals that successfully completed the HSR procedure met these exclusion criteria during the study course. No special housing conditions were required.
Prior to administration, landiolol (Landiolol Hydrochloride, Ono Pharmaceutical Co., Ltd., Osaka, Japan) was dissolved in saline and tailored to the correct dosage (100 µg/kg/min per body weight in 2 ml of saline). The dosage of landiolol used in this study was determined with reference to previously published studies (20,21,24,25).
During the experiments, the rats were anesthetized with isoflurane (0.8–2%) and underwent either sham or HSR surgery. Inhalational anesthesia was performed in a chamber containing isoflurane (4–5%) for induction. After confirming loss of both the righting reflex and the tail pinch reflex, anesthesia was maintained at a dosage of 0.8–2% via a face mask, in accordance with the approved animal experiment protocol (OKU2023436), with the most common concentrations being 5% for induction and 1.3 or 1% for maintenance, during which anesthetic depth was assessed by the absence of spontaneous movements. The vaporizer setting was reduced to below 1% only when marked bradycardia (approximately half of the normal heart rate) was observed. In the five rats that eventually died, this bradycardia was followed within several minutes by persistent hypotension, cardiac arrest and respiratory arrest. During these events, the isoflurane concentration was initially lowered and blood reinfusion was subsequently initiated in an attempt to stabilize the hemodynamic condition. Although euthanasia was also considered in these critical situations, it was difficult to perform because the progression from bradycardia to cardiac and respiratory arrest occurred within a very short period of time. The concentration of isoflurane was verified using a vaporizer. The HSR model was created as previously described (9,10,30,31). The left femoral artery and vein were dissected using aseptic procedures, and 22- and 24-gauge catheters were inserted, respectively. The left femoral artery catheter was used to measure blood pressure, whereas the left femoral vein catheter was used for inducing HSR. After measuring the baseline blood pressure, hemorrhage was initiated for over 15 min, aiming to maintain a mean arterial blood pressure of 30 mmHg, which was achieved by bleeding into a heparinized syringe (10 units/ml). The animals were maintained at this blood pressure (30±5 mmHg) for 45 min through additional blood withdrawal or shed blood infusion. Subsequently, resuscitation was performed for 15 min by administering all the shed blood until the pressure was restored to baseline levels. The rats were kept under anesthesia for 2 h, during which only the blood pressure and pulse rate were monitored (Fig. 1). The sham group underwent identical procedures, except for bleeding. All rats maintained spontaneous breathing throughout the experiment. All procedures were performed on a heating pad, with continuous monitoring and regulation to maintain the rectal temperature within the physiological range.
To examine the effects of landiolol administration on HSR-induced lung injury, the rats were randomly divided into the following four groups: sham/saline (n=10), sham/landiolol (n=10), HSR/saline (n=10), and HSR/landiolol (n=10). Landiolol (100 µg/kg/min) or vehicle (saline 2 ml/h) was injected into the tail vein after HSR induction or sham surgery (Fig. 1). Continuous infusion was initiated 15 min after starting resuscitation and continued for 2 h and 45 min. The duration of administration was the same in both the 3 and 24-h models. At specific time points (3 h or 24 h) after resuscitation, the animals were euthanized by phlebotomy under isoflurane inhalation (2–3%). In our previous study, we found that TNF-α mRNA expression peaked at 3 h after resuscitation in the rat HSR-ALI model, supporting this time point for evaluating early inflammatory gene expression (31). Meanwhile, previous studies have shown that histological lung injury, lung wet/dry ratio, and apoptosis markers such as cleaved caspase-3 are most prominent at 24 h post-insult in related ischemia-reperfusion injury models (32,33). Therefore, the 24-h time point was selected for assessing lung tissue damage and apoptosis. The left lung was removed to determine the pulmonary wet-to-dry weight ratio. The right upper lung was excised, quickly and gently rinsed with saline, fixed in formalin, and stained for histological analysis. Regarding the preparation of RNA and proteins, the right middle and lower lungs were frozen immediately in liquid nitrogen and stored at −80°C until further use.
To record the arterial pressure, an arterial catheter was connected to a pressure transducer (Meritrans DTSPlus® SCK-7874, Merit Medical Japan K.K, Shinjuku-ku, Tokyo, Japan) attached to a bridge amplifier (LifeScopeI® BSM-2303, Nihon Kohden Corporation, Shinjuku-ku, Tokyo, Japan). The blood pressure and heart rate were continuously recorded. The data were measured every 5 min and saved.
After 24 h of resuscitation, the rats were euthanized using the aforementioned method. Subsequently, the right upper lobe of the lungs was excised and fixed in 10% neutral buffered formalin, followed by paraffin embedding and sectioning at a thickness of 5 µm for histological examinations. Following deparaffinization and dehydration, the sections were stained with hematoxylin and eosin (H&E) staining. The lung histological changes were evaluated using blinded evaluation by five observers using a light microscope in accordance with previously described methods (34–36). In each rat, ten regions of lung parenchyma were assessed on a scale from 0 (normal) to 3 (severe) for four parameters: intravascular congestion, pulmonary edema, inflammatory cell infiltration, and intra-alveolar hemorrhage. The final results were reported as the median total score across these parameters, with a maximum score of 30 for each parameter and 120 for the combined lung injury score.
Neutrophils in the lungs were stained using a naphthol AS-D chloroacetate esterase staining kit (Sigma Diagnostics, St. Louis, MO, USA) on sections adjacent to those used for the histopathological analysis (30). An observer, blinded to the groups, counted the positively stained cells in five nonconsecutive sections per rat at ×400 magnification.
Transferase-mediated dUTP-fluorescein isothiocyanate (FITC) nick-end labeling (TUNEL) staining was conducted using the MEBSTAIN Apoptosis TUNEL Kit Direct (No. 8445; MBL, Nagano, Japan) following the manufacturer's instructions. The sections were briefly incubated with terminal deoxynucleotidyl transferase and FITC-labeled dUTP, then counterstained with 0.5 µg/ml propidium iodide. TUNEL-positive cells were counted in five non-consecutive sections per rat at ×400 magnification by a blinded observer using a Zeiss LSM510 confocal laser scanning microscope (Zeiss, Jena, Germany).
The left lung tissue samples were collected 24 h after resuscitation, weighed for their wet weight, and then dried at 110°C for 24 h to obtain the dry weight. The wet-to-dry weight ratio, calculated by dividing the wet weight by the dry weight, was used as an indicator of pulmonary edema (9,31,37).
The total RNA was isolated from the lung tissues at 3 h after HSR using TRI REAGENT® (Molecular Research Center, Inc., Cincinnati, OH, USA) following the manufacturer's instructions. The total RNA was purified using the RNeasy® Mini kit (Qiagen Sciences, Germantown, MD, USA). After removing potentially contaminating DNA with DNase I (RNase-Free DNase set; Qiagen GmbH, Hilden, Germany), reverse transcription of the total RNA was performed using a QuantiTect® Reverse Transcription Kit (Qiagen GmbH) to generate first-strand cDNA. The PCR mixture was prepared using the SYBR® Premix Ex Taq™ (Takara Bio Inc., Shiga, Japan). PCR was performed using StepOnePlus (Thermo Fisher Scientific Inc., Waltham, MA, USA) according to the manufacturer's protocol. The primer sequences for TNF-α, inducible nitric oxide synthase (iNOS), and β-actin were as follows: 5′-GCCCTGGTATGAGCCCATGTA-3′ and 5′-CCTCACAGAGCAATGACTCCAAAG-3′ for TNF-α; 5′-CAAACTGTGTGCCTGGAGGTTC-3′ and 5′-AAGTAGGTGAGGGCTTGCCTGA-3′ for iNOS; and 5′-AACCCTAAGGCCAACCGTGAA-3′ and 5′-CAGGGACAACACAGCCTGGA-3′ for β-actin, respectively. PCR specificity was confirmed using melting curve analysis and DNA sequencing. Quantification of gene expression was performed using a standard curve method, as previously described (38,39). The mRNA levels of TNF-α and iNOS were normalized to the mRNA level of β-actin.
The total proteins were extracted from a portion of the right lung lobe collected 24 h after establishing the HSR model, using Tissue Protein Extraction Reagent (T-PER) (Thermo Fisher Scientific Inc.), according to the manufacturer's protocol. Lung tissue was briefly homogenized in T-PER containing 5 mM dithiothreitol, 5 mM ethylenediaminetetraacetic acid, a protease inhibitor (cOmplete; Roche Diagnostics GmbH, Sigma-Aldrich, St. Louis, MO, USA), and phosphatase inhibitors (PhosSTOP; Roche Diagnostics GmbH, Sigma-Aldrich, St. Louis, MO, USA). The sample was centrifuged at 10,000 × g for 30 min at 4°C, after which the supernatants were collected and stored for later analysis. Protein concentrations in the lung homogenates were measured using the Pierce BCA™ Protein Assay Kit (Pierce, Rockford, IL, USA) following the manufacturer's instructions, with readings taken on a Nivo 5 Multimode Microplate Reader (PerkinElmer, Shelton, CT, USA).
Samples with approximately 50 µg of protein were loaded onto sodium dodecyl sulfate-polyacrylamide gels (10, 12, or 15% concentration) for electrophoresis, followed by the transfer of proteins onto Amersham Hybond-PVDF membranes (GE Healthcare Life Sciences, Chicago, IL, USA). The membranes were blocked at 25°C for 1 h using 4% (w/v) BlockAce (DS Pharma Biomedical Co., Ltd., Osaka, Japan) or Blocking One-P (NACALAI TESQUE, Inc., Kyoto, Japan). Subsequently, the membranes were incubated overnight at 4°C with primary antibodies: cleaved caspase-3 [rabbit anti-cleaved caspase-3 (Asp175), Cell Signaling, #9661, 1:500], Caspase-3 (rabbit anti-caspase-3, Cell Signaling, #9662, 1:1,000), GAPDH [rabbit anti-GAPDH (FL 335), Santa Cruz, sc-25778, 1:5,000], pAMPKα [rabbit anti-phosphorylated-AMPKα (Thr172)(40H9), Cell Signaling, #2535, 1:1,000], and AMPKα (rabbit anti-AMPKα, Cell Signaling, #2532 1:1,000). After washing in Tris-buffered saline with Tween 20, the membranes were incubated with secondary antibodies (goat anti-rabbit IgG-HRP, Abcam, ab6721, 1:20,000 and anti-rabbit IgG-HRP conjugate, Promega, W401B, 1:20,000) for 1 h at 25°C. The membranes were then treated with Clarity Western ECL Substrate (Bio-Rad, Hercules, CA, USA) following the manufacturer's guidelines. Imaging was done with the ChemiDoc XRS Plus System (Bio-Rad), with automatic exposure settings, and densitometry analysis was conducted using Image Lab Version 5.0 software (Bio-Rad).
The data are presented as the mean ± standard error of the mean (SEM) or as the median (interquartile range), as appropriate. Statistical analysis was performed using unpaired Student's t-test, one-way analysis of variance (ANOVA) with Tukey-Kramer multiple comparisons, or Kruskal-Wallis test followed by Dunn's post hoc test for non-parametric data, as appropriate. A two-sided P-value of <0.05 was considered statistically significant. All analyses were performed using GraphPad Prism 10 (GraphPad Software Inc., San Diego, CA, USA).
During the HS period, the mean arterial blood pressure in the HSR groups was maintained within the target range (30±5 mmHg) (Fig. 1). During the subsequent resuscitation period, the heart rate and mean blood pressure did not differ between the sham and HSR groups (Fig. 2A and B). However, during the observation period after resuscitation, the HSR/saline group exhibited a tendency toward decreased mean blood pressure compared with that by the sham/saline group. In contrast, the HSR/landiolol group showed no significant difference or tendency toward lower blood pressure compared with that in the HSR/saline group. This observation suggests that the HSR procedure could cause hypotension after the procedure; however, landiolol administration did not induce harmful blood pressure reduction. Regarding the heart rate, no significant differences were observed among the four groups after landiolol administration. No significant difference in blood loss was observed between the HSR/saline and HSR/landiolol groups (Fig. S1).
In this study, we examined the effect of landiolol treatment on lung injury induced by HSR following HS. Histopathological analysis showed that lung sections from the sham group appeared approximately normal. In contrast, the HSR/saline group displayed interstitial edema, marked by noticeable alveolar septal thickening and inflammatory cell infiltration, 24 h after HSR (Fig. 3A). Although there was no statistically significant difference between the HSR/saline and HSR/landiolol groups, the histopathological score in the HSR/landiolol group was reduced to a level comparable with that of the sham/saline group. Landiolol treatment after HSR tended to attenuate these pathological changes, such as edema, inflammation, and hemorrhage (Fig. 3A and B). The impact of landiolol was further validated through histopathological scoring by an independent, blinded researcher, showing a decrease in the histopathological score (Fig. 3C).
To provide additional visual and quantitative evidence of lung inflammation, neutrophils in the lung tissue were stained and counted (Fig. 4A). Neutrophil infiltration remained mild in the sham group, without notable difference between the sham/saline and sham/landiolol groups. However, in the HSR/saline group, the number of infiltrating neutrophils was significantly higher 24 h after HSR compared with the sham/saline and sham/landiolol groups. In contrast, neutrophil infiltration was substantially lower in the HSR/landiolol group than in the HSR/saline group (Fig. 4B). These neutrophil staining results indicated that landiolol administration alleviated tissue injury and decreased neutrophil infiltration.
The lung wet/dry ratio, a parameter of lung edema, was assessed in the 24 h models. Compared with those in the sham groups, no differences were observed; the wet/dry ratio in the sham/saline group was 5.01±0.03 and 4.97±0.04 in the sham/landiolol group. The lung wet/dry ratio in the HSR/saline group significantly increased compared with that in the sham groups (5.27±0.04). However, landiolol administration significantly attenuated HSR-induced lung edema, and the wet/dry ratio of HSR/landiolol was statistically comparable to the sham groups (5.05±0.02) (Fig. 5). These results suggest that landiolol administration could reduce lung edema in HSR model rats.
To assess the anti-inflammatory effects of landiolol, we analyzed the mRNA expression of TNF-α and iNOS in the lungs 3 h after HSR using RT-qPCR. There were no statistically significant differences in the levels of these inflammatory markers between the sham groups. However, the HSR/saline group demonstrated a significant increase in the TNF-α and iNOS expression compared with that in the sham group. In contrast, landiolol administration reduced the mRNA levels of TNF-α and iNOS by roughly 50 and 30%, respectively, bringing them close to the levels observed in the sham groups (Fig. 6A and B). These findings indicate that landiolol administration lowered inflammatory mRNA expression in the lungs of HSR model rats.
To assess apoptosis, we analyzed the TUNEL-positive cells and cleaved caspase-3 expression in the lungs 24 h after HSR using western blotting. The lung sections from the sham group had very few TUNEL-positive cells, but their numbers increased following the HSR procedure. In contrast, landiolol treatment significantly reduced the number of TUNEL-positive cells compared with the HSR/saline group (Fig. 7A and B). Similarly, cleaved caspase-3 expression was minimally detectable in the sham group but increased in the HSR group. However, landiolol treatment effectively inhibited the increase in cleaved caspase-3 (Fig. 7C and D).
We examined pAMPKα via western blotting. pAMPKα is a protein involved in mitochondrial biogenesis, autophagy, and related processes. Regarding pAMPKα, we found no significant differences between the sham/saline and sham/landiolol groups. The HSR/saline group also showed no differences compared with the sham group. However, landiolol treatment was found to increase pAMPKα expression compared with the other three groups (Fig. 8A and B).
This study showed that intravenous administration of landiolol at a dose of 100 µg/kg/min following the HSR procedure substantially alleviated lung injury induced by HSR. This finding was supported by a trend toward reductions in histological alterations, and was further substantiated by significant reductions in neutrophil infiltration and lung edema. Furthermore, landiolol administration markedly lowered the mRNA expression of inflammatory mediators, including TNF-α and iNOS. Additionally, landiolol administration reduced apoptotic cell death, as shown by a decrease in the TUNEL-positive cells and cleaved caspase-3 expression. We investigated the pAMPKα expression as a mechanism for the anti-inflammatory and anti-apoptotic effects of landiolol and found by western blotting that pAMPKα expression was increased when landiolol was administered after HSR procedures. In addition, landiolol administration following the HSR procedure had no discernible effect on the mean arterial blood pressure.
TNF-α promotes neutrophil infiltration and increases vascular permeability, leading to endothelial injury. Neutrophils act on vascular endothelial cells and produce superoxides and other ROS, which cause cellular injury and edema (40). This endothelial disruption induces the expression of iNOS, which generates reactive nitrogen species such as nitric oxide, thereby exacerbating oxidative injury during reperfusion (41). Furthermore, endothelial damage results in vascular leakage, which exacerbates tissue edema in the lungs (11). Targeting upstream inflammatory mediators such as TNF-α, iNOS-induced vascular injury, and neutrophil-mediated permeability enhancement may help attenuate lung damage. For this reason, these factors were selected as therapeutic targets in our study.
Landiolol is a drug used for treating arrhythmia; however, previous studies have shown that landiolol treatment has lung-protective effects. In a rat LPS model, Hagiwara et al reported that landiolol administration reduced HMGB1 expression in the lungs and serum and alleviated histological lung injury (20). Matsuishi et al reported that landiolol ameliorated histological findings and PaO2 levels in a rat ALI model of early sepsis by suppressing elevated levels of pulmonary endothelin-1 (25). Notably, our research is the first to suggest that landiolol administration may play a protective role against ALI in a non-septic HSR model. Considering our findings and those of other studies (20,25), landiolol appears to be highly effective in protecting the lungs from ALI.
When investigating the mechanisms underlying the protective effects of landiolol on ALI following HSR, we observed that landiolol administration suppressed HSR-induced expression of inflammatory genes. This finding aligns with those of previous reports. Hagiwara et al showed that landiolol administration reduces TNF-α and IL-6 levels in the serum of the LPS rat model (20). Yoshino showed that landiolol administration to the LPS rat model improves TNF-α expression levels in the liver (24). Furthermore, Ackland et al reported that the β1 adrenergic receptor blocker metoprolol suppresses the expression of IFN-γ, IL-1β, IL-6, TNF-α, IL-18, and MCP-1 in the liver when administered to the LPS rat model (42). The findings of our study also indicated that landiolol administration decreased the expression of inflammatory genes in the HSR-induced ALI model. Thus, landiolol exhibits anti-inflammatory effects, representing a key mechanism in its protective action against lung injury.
Our study demonstrated that landiolol inhibited apoptosis in HSR-induced lung injury. Previous reports have also shown that β1-blockers may have anti-apoptotic effects. Zaugg et al reported that a β1 blocker atenolol suppressed TUNEL-positive cells and increased the expression of the anti-apoptotic protein Bcl2 in catecholamine-induced apoptosis (15). Hashemi et al reported that atenolol administration increases MEF2 transcriptional activity, which is involved in the anti-apoptotic effects of catecholamine-induced apoptosis in rat cardiomyocytes (43). Additionally, Taha et al reported that the administration of atenolol suppressed the gene expression of the pro-apoptosis protein caspase 1 and increases the expression of Bcl2 in a rat intestinal ischemia-reperfusion model (44). These reports and our experimental results suggest that β1-blocker has anti-apoptotic effects. Although the precise mechanism underlying the anti-apoptotic effects of landiolol could not be fully elucidated in this study, increased p-AMPKα expression observed in the HSR/Landiolol group suggests a potential role of AMPK pathway activation. AMPK is known to suppress oxidative stress, promote autophagy, and preserve mitochondrial function, all of which may contribute to the observed reduction in apoptosis (16,17). In addition, the AMPK/Nrf2 signaling pathway has been reported as a potential therapeutic target for treating ALI. Activation of this pathway may upregulate downstream effectors such as heme oxygenase-1 and modulate macrophage activity, thereby attenuating inflammation (45). Further investigation into the upstream and downstream signaling pathways of AMPK is warranted in future studies. Additionally, in vitro studies have reported that landiolol can scavenge multiple types of free radicals, which may also contribute to its anti-apoptotic properties (46).
Our study had some limitations. First, our model does not comply with the Berlin definition regarding the disuse of positive end-expiratory pressure and image evaluations, oxygenation index, pulmonary function indicator (47). However, conforming to this definition for animal experiments is challenging. Therefore, we performed the experiments according to the ALI definition for animal experiments (48). The severity of ALI for the animals was approximated using H&E staining, neutrophil staining, and the wet/dry ratio. Second, this HSR model is difficult to establish, with a mortality rate of approximately 11% during the HS procedure, and several animals were excluded due to complications. Third, we lacked survival rate data. Although the mortality during the HS procedure was relatively high, all rats that successfully underwent HSR treatment survived, indicating that this represents a mild lung injury model. Forth, the primary aim of this study was to assess the lung-protective effects of landiolol administration; however, the detailed mechanisms underlying these effects were not fully elucidated. Previous studies have demonstrated the organ-protective role of AMPK using Compound C, a specific AMPK inhibitor (49–51). In the future, we plan to investigate whether the AMPK pathway contributes to the protective effects observed in this study by conducting validation experiments using Compound C. This study was conducted using only male rats to ensure consistency with previous experiments (9,10,28–31). The potential influence of sex differences on the outcomes was not assessed and remains a subject for future investigation.
In conclusion, landiolol administration after HSR improved HSR-induced ALI through its anti-inflammatory and anti-apoptotic effects, at least in part, mediated by the activation of the AMPK pathway without severe hypotension. Further research is required to elucidate the exact mechanisms and pharmacological characteristics of landiolol. Thus, landiolol administration may be a therapeutic approach for acute lung injury following HSR.
The StepOnePlus and Zeiss LSM510 devices used in this study were obtained from the Central Research Laboratory, Okayama University Medical School (Okayama, Japan). The authors would also like to thank Mr. Kosuke Iguchi, Ms. Misako Yanagita and Ms. Shukuko Wani (medical students at Okayama University, Okayama, Japan) for their technical support.
This work was supported by a Japan Society for the Promotion of Science Grant-in-Aid for Scientific Research (grant nos. JP19K09381 and JP23K08360).
The data generated in the present study may be requested from the corresponding author.
RS, HS and TT designed the study. RS wrote the first draft of the manuscript. HS and HM critically revised the manuscript for intellectual content. RS, HS, YLu and EO performed the experiments. RS and HS confirm the authenticity of all the raw data. RS, HS, YLi, RN and EO performed histological scoring, which was conducted in a blinded manner. RS, HS, TT and HM analyzed and interpreted the data. RS and HS performed the statistical analyses. All authors read and approved the final manuscript.
The animal experimental procedures in the present study were approved by the Department of Animal Resources, Advanced Science Research Center, Okayama University (approval no. OKU-2021247 on April 1, 2021 and approval no. OKU-2023436 on April 24, 2023).
Not applicable.
The authors declare that they have no competing interests.
During the preparation of this work, AI tools (ChatGPT and Consensus) were used to improve the readability and language of the manuscript, and subsequently, the authors revised and edited the content produced by the AI tools as necessary, taking full responsibility for the ultimate content of the present manuscript.
|
ALI |
acute lung injury |
|
AMPK |
5′ adenosine monophosphate-activated protein kinase |
|
ANOVA |
analysis of variance |
|
iNOS |
inducible nitric oxide synthase |
|
H&E |
hematoxylin and eosin |
|
HSR |
hemorrhagic shock and resuscitation |
|
MAP |
mean arterial blood pressure |
|
RT-qPCR |
reverse transcription-quantitative polymerase chain reaction |
|
SEM |
standard error of the mean |
|
TNF-α |
tumor necrosis factor-α |
|
IL |
interleukin |
|
TUNEL |
terminal deoxynucleotidyl transferase dUTP nick-end labeling |
|
FITC |
fluorescein isothiocyanate |
|
HMGB1 |
high mobility group box 1 |
|
LPS |
lipopolysaccharide |
|
Dewar D, Moore FA, Moore EE and Balogh Z: Postinjury multiple organ failure. Injury. 40:912–918. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Ciesla DJ, Moore EE, Johnson JL, Cothren CC, Banerjee A, Burch JM and Sauaia A: Decreased progression of postinjury lung dysfunction to the acute respiratory distress syndrome and multiple organ failure. Surgery. 140:640–648. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Ware LB: Pathophysiology of acute lung injury and the acute respiratory distress syndrome. Semin Respir Crit Care Med. 27:337–349. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
ARDS Definition Task Force, . Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND, Caldwell E, Fan E, Camporota L and Slutsky AS: Acute respiratory distress syndrome: The Berlin definition. JAMA. 307:2526–2533. 2012.PubMed/NCBI | |
|
Acute Respiratory Distress Syndrome Network, . Brower RG, Matthay MA, Morris A, Schoenfeld D, Thompson BT and Wheeler A: Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. N Engl J Med. 342:1301–1308. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Tonelli AR, Zein J, Adams J and Ioannidis JPA: Effects of interventions on survival in acute respiratory distress syndrome: An umbrella review of 159 published randomized trials and 29 meta-analyses. Intensive Care Med. 40:769–787. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Adhikari N, Burns KEA and Meade MO: Pharmacologic therapies for adults with acute lung injury and acute respiratory distress syndrome. Cochrane Database Syst Rev. 2004:CD0044772004.PubMed/NCBI | |
|
Peter JV, John P, Graham PL, Moran JL, George IA and Bersten A: Corticosteroids in the prevention and treatment of acute respiratory distress syndrome (ARDS) in adults: Meta-analysis. BMJ. 336:1006–1009. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Kanagawa F, Takahashi T, Inoue K, Shimizu H, Omori E, Morimatsu H, Maeda S, Katayama H, Nakao A and Morita K: Protective effect of carbon monoxide inhalation on lung injury after hemorrhagic shock/resuscitation in rats. J Trauma. 69:185–194. 2010.PubMed/NCBI | |
|
Kumada Y, Takahashi T, Shimizu H, Nakamura R, Omori E, Inoue K and Morimatsu H: Therapeutic effect of carbon monoxide-releasing molecule-3 on acute lung injury after hemorrhagic shock and resuscitation. Exp Ther Med. 17:3429–3440. 2019.PubMed/NCBI | |
|
Esiobu P and Childs EW: A rat model of hemorrhagic shock for studying vascular hyperpermeability. Methods Mol Biol. 1717:53–60. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Peitzman AB, Billiar TR, Harbrecht BG, Kelly E, Udekwu AO and Simmons RL: Hemorrhagic shock. Curr Probl Surg. 32:925–1002. 1995. View Article : Google Scholar : PubMed/NCBI | |
|
Pérez-Schindler J, Philp A and Hernandez-Cascales J: Pathophysiological relevance of the cardiac β2-adrenergic receptor and its potential as a therapeutic target to improve cardiac function. Eur J Pharmacol. 698:39–47. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Grisanti LA, Evanson J, Marchus E, Jorissen H, Woster AP, DeKrey W, Sauter ER, Combs CK and Porter JE: Pro-inflammatory responses in human monocytes are beta1-adrenergic receptor subtype dependent. Mol Immunol. 47:1244–1254. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Zaugg M, Xu W, Lucchinetti E, Shafiq SA, Jamali NZ and Siddiqui MA: Beta-adrenergic receptor subtypes differentially affect apoptosis in adult rat ventricular myocytes. Circulation. 102:344–350. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Aslam M and Ladilov Y: Emerging role of cAMP/AMPK signaling. Cells. 11:3082022. View Article : Google Scholar : PubMed/NCBI | |
|
Ding R, Wu W, Sun Z and Li Z: AMP-activated protein kinase: An attractive therapeutic target for ischemia-reperfusion injury. Eur J Pharmacol. 888:1734842020. View Article : Google Scholar : PubMed/NCBI | |
|
Plosker GL: Landiolol: A review of its use in intraoperative and postoperative tachyarrhythmias. Drugs. 73:959–977. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Bennett M, Chang CL, Tatley M, Savage R and Hancox RJ: The safety of cardioselective β1-blockers in asthma: Literature review and search of global pharmacovigilance safety reports. ERJ Open Res. 7:00801–2020. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Hagiwara S, Iwasaka H, Maeda H and Noguchi T: Landiolol, an ultrashort-acting beta1-adrenoceptor antagonist, has protective effects in an LPS-induced systemic inflammation model. Shock. 31:515–520. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Kiyonaga N, Moriyama T and Kanmura Y: Effects of landiolol in lipopolysaccharide-induced acute kidney injury in rats and in vitro. Shock. 52:e117–e123. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Goyagi T, Kimura T, Nishikawa T, Tobe Y and Masaki Y: Beta-adrenoreceptor antagonists attenuate brain injury after transient focal ischemia in rats. Anesth Analg. 103:658–663. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Iwata M, Inoue S, Kawaguchi M, Nakamura M, Konishi N and Furuya H: Posttreatment but not pretreatment with selective beta-adrenoreceptor 1 antagonists provides neuroprotection in the hippocampus in rats subjected to transient forebrain ischemia. Anesth Analg. 110:1126–1132. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Yoshino Y, Jesmin S, Islam M, Shimojo N, Sakuramoto H, Oki M, Khatun T, Suda M, Kawano S and Mizutani T: Landiolol hydrochloride ameliorates liver injury in a rat sepsis model by down regulating hepatic TNF-A. J Vasc Med Surg. 3:10001942015. | |
|
Matsuishi Y, Jesmin S, Kawano S, Hideaki S, Shimojo N, Mowa CN, Akhtar S, Zaedi S, Khatun T, Tsunoda Y, et al: Landiolol hydrochloride ameliorates acute lung injury in a rat model of early sepsis through the suppression of elevated levels of pulmonary endothelin-1. Life Sci. 166:27–33. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Percie du Sert N, Hurst V, Ahluwalia A, Alam S, Avey MT, Baker M, Browne WJ, Clark A, Cuthill IC, Dirnagl U, et al: The ARRIVE guidelines 2.0: Updated guidelines for reporting animal research. J Cereb Blood Flow Metab. 40:1769–1777. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Leary S, Underwood W, Anthony R, Cartner S, Grandin T, Greenacre C, Gwaltney-Brant S, McCrackin MA, Meyer R, et al: AVMA Guidelines for the Euthanasia of Animals: 2020 Edition. American Veterinary Medical Association; Schaumburg, IL, USA: pp. 1–121. 2020, https://www.avma.org/resources-tools/avma-policies/avma-guidelines-euthanasia-animalsMarch 1–2021 | |
|
Kosaka J, Morimatsu H, Takahashi T, Shimizu H, Kawanishi S, Omori E, Endo Y, Tamaki N, Morita M and Morita K: Effects of biliverdin administration on acute lung injury induced by hemorrhagic shock and resuscitation in rats. PLoS One. 8:e636062013. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Shimizu H, Nakamura R, Lu Y, Sakamoto R, Omori E, Takahashi T and Morimatsu H: The protective effect of carbamazepine on acute lung injury induced by hemorrhagic shock and resuscitation in rats. PLoS One. 19:e03096222024. View Article : Google Scholar : PubMed/NCBI | |
|
Umeda K, Takahashi T, Inoue K, Shimizu H, Maeda S, Morimatsu H, Omori E, Akagi R, Katayama H and Morita K: Prevention of hemorrhagic shock-induced intestinal tissue injury by glutamine via heme oxygenase-1 induction. Shock. 31:40–49. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Maeshima K, Takahashi T, Uehara K, Shimizu H, Omori E, Yokoyama M, Tani T, Akagi R and Morita K: Prevention of hemorrhagic shock-induced lung injury by heme arginate treatment in rats. Biochem Pharmacol. 69:1667–1680. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Yang Z, Zhang XR, Zhao Q, Wang SL, Xiong LL, Zhang P, Yuan B, Zhang ZB, Fan SY, Wang TH and Zhang YH: Knockdown of TNF-α alleviates acute lung injury in rats with intestinal ischemia and reperfusion injury by upregulating IL-10 expression. Int J Mol Med. 42:926–934. 2018.PubMed/NCBI | |
|
Hassoun HT, Lie ML, Grigoryev DN, Liu M, Tuder RM and Rabb H: Kidney ischemia-reperfusion injury induces caspase-dependent pulmonary apoptosis. Am J Physiol Renal Physiol. 297:F125–F137. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Murakami K, McGuire R, Cox RA, Jodoin JM, Bjertnaes LJ, Katahira J, Traber LD, Schmalstieg FC, Hawkins HK, Herndon DN and Traber DL: Heparin nebulization attenuates acute lung injury in sepsis following smoke inhalation in sheep. Shock. 18:236–241. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Zegdi R, Fabre O, Cambillau M, Fornès P, Tazi KA, Shen M, Hervé P, Carpentier A and Fabiani JN: Exhaled nitric oxide and acute lung injury in a rat model of extracorporeal circulation. Shock. 20:569–574. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang H, Meng F, Li W, Tong L, Qiao H and Sun X: Splenectomy ameliorates acute multiple organ damage induced by liver warm ischemia reperfusion in rats. Surgery. 141:32–40. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Stephens KE, Ishizaka A, Larrick JW and Raffin TA: Tumor necrosis factor causes increased pulmonary permeability and edema. Comparison to septic acute lung injury. Am Rev Respir Dis. 137:1364–1370. 1988. View Article : Google Scholar : PubMed/NCBI | |
|
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 | |
|
Nakatsuka K, Matsuoka Y, Kurita M, Wang R, Tsuboi C, Sue N, Kaku R and Morimatsu H: Intrathecal administration of the α1 adrenergic antagonist phentolamine upregulates spinal GLT-1 and improves mirror image pain in SNI model rats. Acta Med Okayama. 76:255–263. 2022.PubMed/NCBI | |
|
McMillen MA, Huribal M and Sumpio B: Common pathway of endothelial-leukocyte interaction in shock, ischemia, and reperfusion. Am J Surg. 166:557–562. 1993. View Article : Google Scholar : PubMed/NCBI | |
|
Cinelli MA, Do HT, Miley GP and Silverman RB: Inducible nitric oxide synthase: Regulation, structure, and inhibition. Med Res Rev. 40:158–189. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Ackland GL, Yao ST, Rudiger A, Dyson A, Stidwill R, Poputnikov D, Singer M and Gourine AV: Cardioprotection, attenuated systemic inflammation, and survival benefit of beta1-adrenoceptor blockade in severe sepsis in rats. Crit Care Med. 38:388–394. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Hashemi S, Salma J, Wales S and McDermott JC: Pro-survival function of MEF2 in cardiomyocytes is enhanced by β-blockers. Cell Death Discov. 1:150192015. View Article : Google Scholar : PubMed/NCBI | |
|
Taha MO, Silva TDMAE, Ota KS, Vilela WJ, Simões RS, Starzewski A and Fagundes DJ: The role of atenolol in the modulation of the expression of genes encoding pro-(caspase-1) and anti-(Bcl2L1) apoptotic proteins in endothelial cells exposed to intestinal ischemia and reperfusion in rats. Acta Cir Bras. 33:1061–1066. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Huang Q, Ren YX, Yuan P, Huang M, Liu G, Shi Y, Jia G and Chen M: Targeting the AMPK/Nrf2 pathway: A novel therapeutic approach for acute lung injury. J Inflamm Res. 17:4683–4700. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Matsumoto S, Tokumaru O, Ogata K, Kuribayashi Y, Oyama Y, Shingu C, Yokoi I and Kitano T: Dose-dependent scavenging activity of the ultra-short-acting β1-blocker landiolol against specific free radicals. J Clin Biochem Nutr. 71:185–190. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ferguson ND, Fan E, Camporota L, Antonelli M, Anzueto A, Beale R, Brochard L, Brower R, Esteban A, Gattinoni L, et al: The Berlin definition of ARDS: An expanded rationale, justification, and supplementary material. Intensive Care Med. 38:1573–1582. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Matute-Bello G, Downey G, Moore BB, Groshong SD, Matthay MA, Slutsky AS and Kuebler WM; Acute Lung Injury in Animals Study Group, : An official American Thoracic Society workshop report: Features and measurements of experimental acute lung injury in animals. Am J Respir Cell Mol Biol. 44:725–738. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Escobar DA, Botero-Quintero AM, Kautza BC, Luciano J, Loughran P, Darwiche S, Rosengart MR, Zuckerbraun BS and Gomez H: Adenosine monophosphate-activated protein kinase activation protects against sepsis-induced organ injury and inflammation. J Surg Res. 194:262–272. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang WL, Zhao KC, Yuan W, Zhou F, Song HY, Liu GL, Huang J, Zou JJ, Zhao B and Xie SP: MicroRNA-31-5p exacerbates lipopolysaccharide-induced acute lung injury via inactivating Cab39/AMPKα pathway. Oxid Med Cell Longev. 2020:88223612020. View Article : Google Scholar : PubMed/NCBI | |
|
Chen R, Cao C, Liu H, Jiang W, Pan R, He H, Ding K and Meng Q: Macrophage Sprouty4 deficiency diminishes sepsis-induced acute lung injury in mice. Redox Biol. 58:1025132022. View Article : Google Scholar : PubMed/NCBI |