Skip to main content
  • Research Articles
  • Open access
  • Published:

Protective effect of canagliflozin on post-resuscitation myocardial function in a rat model of cardiac arrest

Abstract

Background

Currently, most patients with cardiac arrest (CA) show reversible myocardial dysfunction, hemodynamic instability, systemic inflammation and other pathophysiological state in early stage of resuscitation, some patients may eventually progress to multiple organ failure. There is evidence that heart failure is the terminal stage in the development of various cardiovascular diseases. Although the cardio-protective effect of canagliflozin (CANA) has been confirmed in large clinical studies and recommended in domestic and international heart failure-related guidelines, the effectiveness of CANA after resuscitation remains unclear. In this study, we constructed a modified CA/CPR rat model to investigate whether CANA administered on post-resuscitation improves myocardial function.

Methods

Twenty-fourth healthy male Sprague–Dawley rats were randomized into four groups: (1) Sham + placebo group, (2) Sham + CANA group, (3) CPR + placebo group, and (4) CPR + CANA group. Ventricular fibrillation was induced by transcutaneous electrical stimulation on epicardium. After 6 min untreated ventricular fibrillation, chest compressions was initiated. The rats were received an injection of placebo or canagliflozin (3 ug/kg) randomly 15 min after restore of spontaneous circulation (ROSC). Electrocardiogram (ECG) and blood pressure were continuously detected in each group throughout the experiment. The rats were killed 6 h after ROSC to collected the arterial serum and myocardial tissue. Myocardial injury was estimated with concentrations of inflammatory factors, oxidative stress indexes and, apoptosis index, myocardial injury markers, echocardiography and myocardial pathological slices.

Results

After resuscitation, mean arterial pressure (MAP) were significantly increased after cardiopulmonary resuscitation in CANA group rats when compared with placebo group. Heart rate, body lactate returned and left ventricular ejection fraction (LVEF) to normal levels in a shorter time and the myocardial injury was obviously attenuated in CPR + CANA group. Inflammatory factors (IL-6, TNF-α) and oxidative stress indexes (MAD, SOD, CAT) were dramatically decreased with the administration of CANA. The expression of apoptosis index (BAX, caspase-3) were higher in CPR + placebo group and the expression of anti-apoptosis index (Bcl-2) was lower (P < 0.05).

Conclusions

The administration of CANA effectively reduces myocardial ischaemia/reperfusion (I/R) injury after cardiac arrest and cardiopulmonary resuscitation (CPR), and the underlying mechanism may be related to anti-inflammation, oxidative stress and apoptosis.

Cardiac arrest (CA) is a global health issue, accounting for approximately 1/5 of adult deaths in the United States each year [1]. Despite rapid advances in medical technology, up to 50% of resuscitated patients still die before discharge from the hospital [2]. More than 2/3 of patients with ventricular fibrillation-related cardiac arrest have hemodynamic disorders, considered as post-resuscitation myocardial dysfunction (PRMD), the leading cause of early death after recovery of spontaneous circulation (ROSC) [3,4,5].

PRMD occurs in approximately 68% of post-resuscitation patients which pathophysiological mechanism is complex [6, 7]. During cardiac arrest, the body's energy metabolism and oxygen transport are in a state of suspended animation. However, chest compressions provide only limited perfusion pressure, which cannot counteract the ischaemia caused by microthrombus and vasospasm [8]. The heart undergoes cardiac arrest and CPR leads to myocardial I/R injury. In pathological conditions, cardiomyocyte death or injury releases endogenous signals known as danger-associated molecular patterns (DAMPs) [9, 10], which in turn release a variety of pro-inflammatory chemokines and cytokines, initiate and regulate innate immune responses. Recently, the adaptive immune response has been highlighted as another significant factor in MIRI [11, 12]. However, uncontrolled or excessive inflammatory factors can aggravate tissue injury. In addition, a large number of oxygen-free radicals disrupt the dynamic balance between oxidative and antioxidant systems. Excessive reactive oxygen species (ROS) cause the damage to different membrane structural components and harm cellular physiological functions. For example, ROS lead to electrophysiological impairment and contractile dysfunction of cardiomyocytes by affecting the Na + /Ca2 + exchanger (NCX), the L-type calcium channel and the sarcoplasmic reticulum Ca2 + -ATPase [13]. On the other hand, inflammatory factors increase vascular permeability and induce neutrophil adhesion, which increases the expression of pro-apoptotic factors. Inflammation can also activate the death receptor pathway and induce cardiomyocyte apoptosis [14, 15].

Several large clinical trials (CANVAS, CREDENCE) have shown that CANA can significantly reduce cardiovascular events [16, 17]. International guidelines recommend that CANA are considered as first-line therapy in patients with type 2 diabetes mellitus (T2DM) at high cardiovascular risk [18, 19]. A post-hoc analysis of the CANVAS trial showed that CANA reduced plasma IL-6 levels in people at high cardiovascular risk [20], and Heerspink et al. also found the same anti-inflammatory properties of CANA [21]. Peyton et al. demonstrated that CANA inhibits the inflammatory response of human endothelial cells by regulating HO-1 expression. In addition to its anti-inflammatory properties, CANA also reduces mitochondrial ROS production, thereby preserving the functional integrity of endothelial cells [22]. Several studies have shown that SGLT2 inhibitors significantly reduce oxidative stress markers in myocardial tissue with and without diabetes [23, 24]. In addition, more and more studies have found the anti-apoptotic effect of CANA in many diseases. CANA can inhibit carfilzomib (CFZ)-induced endothelial cell apoptosis via the AMPK pathway for the adjuvant treatment of relapsed/refractory multiple myeloma [25]. It can also inhibit apoptosis by down-regulating CX3CL1 expression in the heart and kidney to treat cardiorenal syndrome [26]. There is increasing evidence that CANA has anti-inflammatory, anti-apoptotic, and antioxidant properties. Therefore, we hypothesize that CANA may improve post-resuscitation cardiac dysfunction in CA/CPR patients.

Studies on the effects of CANA in patients with cardiac arrest are still at an early stage, and the specific mechanisms are still unclear. We hypothesize that CANA is effective in attenuating myocardial I/R injury after cardiac arrest and resuscitation, and that the underlying mechanisms are related to anti-inflammation, oxidative stress and apoptosis.

Methods

Design

This was a randomized controlled study in rats to investigate the effect of intravenous CANA on cardiac function after cardiopulmonary resuscitation. The Animal Experiment Ethics Committee of Anhui Medical University approved this study. All experimental animals were treated according to the Guide for the Care and Use of Laboratory Animals (NIH Publication No. 85-23, revised 1996).

Anaesthesia and animal preparation

Twenty-fourth healthy male Sprague–Dawley rats, 8–10 months, weighing 400–450 g, were provided by the Laboratory Animal Center of Anhui Medical University. The rats were housed in a temperature-controlled room and maintained on a 12-h diurnal rhythm with adequate water and food. After inhalation of carbon dioxide (CO2) for approximately 30 s to induce anesthesia, the animals were injected intraperitoneally with pentobarbital at an initial dose of 45 mg/kg. A maintenance dose of 10 mg/kg was administrated as needed. The trachea of the animals was orally intubated with a 14G cannula mounted on a blunt needle (Abbocath-T; Abbott Hospital Products Division, United States) with a 145°angled tip. The rats were mechanically ventilated with a multichannel small-animal ventilator (KW-100-2, NJKEWBIO, China) was used for mechanical ventilation, set at a tidal volume of 0.6 mL/100 g body weight, a rate of 100 breaths/min, and an inspired oxygen fraction of 0.21. Two PE-50 catheters (PE50, Smith Medical, United Kingdom) were prepared for cannulation of the left femoral artery and vein, the left femoral artery catheter to the physiological monitor for the measurements of vital signs, and the collection of blood samples before sacrificing rats. The venous catheter is used to establish an intravenous infusion route for the administration of CANA or placebo. All catheters were primed with 2.5U/ml heparinized saline. ECG recording with lead II and heart rate monitoring throughout the experiment. During the experiment, the rectal temperature was maintained at 37 ± 0.5℃ with a heating lamp.

Experimental protocol

Twenty-fourth male rats were randomly divided into four groups: (1) CPR + CANA group (n = 6): canagliflozin was administered by intravenous injection (dose: 3 µg/kg) after 15 min of recovery of spontaneous circulation (ROSC). (2) CPR + placebo group (n = 6): equal volumes of placebo were administered by intravenous injection at the same timepoint. (3) Sham + CANA group (n = 6): the equal dose of canagliflozin was administered. (4) Sham + placebo group (n = 6): equal volumes of placebo were administered.

The experimental procedure is shown in Fig. 1. Baseline vital signs (heart rate and blood pressure) and arterial blood gases (RAPID Point500, SIEMENS, Germany) were recorded 15 min before induction of ventricular fibrillation (VF). VF was induced by acupuncture needles inserted into epicardial electrical stimulation. Point A is positioned at the most vital point of the cardiac apex beat of a rat; point B is set at approximately 2.0 cm horizontally to the right of point A. Points A and B are the needle entry points for epicardial electrical stimulation (Fig. 2). The needle insertion angle of point A is approximately 60°, while the needle insertion angle of point B is approximately 75°, and the insertion depth is approximately 1.5–2.0 cm. When the needle tip reaches the epicardium, the needle tail has a rapid swinging action consistent with the beating frequency of the rat's heart, and the rat's ECG would show a transient arrhythmia. There may also be a transient drop in blood pressure. Small animal cardiopulmonary resuscitation instruments (KW-XF, NJKEWBIO, China) were used for the instrument construction of CA/CPR model. The direct current was progressively increased from an initial 0.5 mA to a maximum of 1.5 mA. In addition, the stimulation was maintained for 3 min to prevent spontaneous cardioversion. Mechanical ventilation was stopped throughout the VF process. Following 6 min of untreated VF, chest compressions (compression rate of 200 times/min, compression depth of 1.0–1.3 cm) and mechanical ventilation (inspired oxygen fraction of 1.0) were initiated and maintained for 8 min. Defibrillation was performed with up to 4 J countershocks. Successful defibrillation is indicated by ROSC, defined as MAP ≥ 50 mmHg maintained for 5 min. If defibrillation fails, continue compressions for 30 s and then defibrillation again. Failure to achieve ROSC after more than 3 cycles is considered as a failure. After ROSC, an inspired oxygen fraction of 1.0 was maintained for 1 h, adjusted to 0.5 for another hour, and then to 0.21. Detailed ECG changes during the experiment are shown in Fig. 3.

Fig. 1
figure 1

Experimental procedure. 15 means 15 min before ventricular fibrillation was induced; BL baseline, VF ventricular fibrillation, CPR cardiopulmonary resuscitation, CANA canagliflozin

Fig. 2
figure 2

Sketch of point A/B

Fig. 3
figure 3

ECG during experiment. a Normal ECG, b ECG of needle insertion, c ECG during induction of VF, d ECG during CPR, e ECG of post-resuscitation arrhythmias, f normal ECG of post-resuscitation

Echocardiograms were obtained at baseline and at post-resuscitation 2 h, 4 h, and 6 h. At the end of 6 h after ROSC, animals were sacrificed with an intravenous administration of 150 mg/kg pentobarbital. Myocardial tissue and serum samples were collected and stored in a – 80 ℃ refrigerator for further analysis. In addition, each rat was necropsied to check for any severe injury due to improper handling.

Measurements

Basic vital signs

ECG, heart rate (HR), mean aortic pressure (MAP), body temperature were continuously recorded on a personal computer-based biological signal acquisition and processing system (Medlab-U/4C501H, NJKEWBIO, China). Left ventricular ejection fraction (LVEF) was measured by echocardiography (6LAB, VINNO, China) at baseline and at post-resuscitation 2 h, 4 h, and 6 h.

Myocardial histopathological damage

Myocardial tissue was fixed in 10% neutral formalin for 24 h. Paraffin was used for routine embedding, and the embedded specimens were sectioned at 4 μm thickness. After sectioning, the slices were baked in an oven for 12 min and then placed in an automatic hematoxylin and eosin (H&E) staining machine for HE staining. Myocardial pathology images were captured using an upright biological microscope (Axio Scope A1, Carl Zeiss, Germany) and imaging software at 400* magnification. The degree of myocardial edema, inflammatory cell infiltration, and myocardial fiber arrangement were observed to determine myocardial injury. Myocardial histopathological damage was graded independently by three independent observers.

Assessment of related protein expression

After ROSC 6 h, arterial blood and extracted supernatant of animals were collected for determining relevant inflammatory markers (IL-6, TNF-α), oxidative stress markers (SOD, MDA, CAT) and myocardial injury markers (NT-Pro BNP, cTnI). IL-6, TNF-α, NT-Pro BNP and cTnI were measured with commercial ELISA kits according to the manufacturer’s instructions (E-EL-R0015c/E-EL-R2856c/E-EL-R3023/E-EL-R1253c, Elabscience Biotechnology Co., Ltd, China), respectively. SOD, MDA and CAT were measured with commercial ELISA kits according to the manufacturer’s instructions (JYM0990Ra/JYM0266Ra/JYM1143Ra, Wuhan ColorfulGene Biological Technology Co., LTD, China), respectively.

An appropriate amount of myocardial tissue samples was weighed, ground. In addition, total proteins were extracted in grinding reagent with 500ul RIPA (P0013B, Beyotime Biotechnology, China) + 5 μl PMSF (ST505, Beyotime Biotechnology, China) per 100 mg of tissue. The concentration of extracted protein was determined by BCA protein assay kit. Then, 5 × loading buffer (P0015L, Beyotime Biotechnology, China) was added and boiled for 10 min. The protein homogenate was separated by sodium dodecyl sulfate (SDS)–polyacrylamide gel electrophoresis and then transferred to the PVDF membrane (IPVH00010, Merck Millipore, Germany) using a membrane transfer device. The membrane was blocked with 5% skim milk powder for 2 h, washed, and incubated with primary antibodies overnight at 4℃: BAX (1:1000; ab32503, Abcam, United Kingdom), Bcl-2 (1:500; GTX100064, Gene Tex, United States), Caspase-3 (1:2000; ab184787, Abcam, United Kingdom), Nrf2 (1:500; AF0639, Affinity Biosciences, United States), HO-1 (1:500; AF5393, Affinity Biosciences, United States), GAPDH (1:5000; AF0911; Affinity Biosciences, United States). After washing, they were incubated with secondary antibody (1:1 0000; E-AB-1034; Elabscience Biotechnology Co., Ltd, China) for 1 h at room temperature. The gray value of the target band was analyzed using Image J, and the expressions level of the target proteins were expressed as the ratio of the gray value of the target band to the gray value of the GAPDH band.

Statistics

SPSS version 19.0 statistical software was used to analyze all data. Measurement data conforming to normal distribution are expressed as mean ± SD. One-way ANOVA was used for intergroup comparisons, and the SNK-q test was used for pairwise comparisons. The Chi-squared test was used to compare enumeration data between groups. A value of P < 0.05 was considered statistically significant.

Results

A total of 24 rats were used and analyzed. There was no difference in baseline characteristics between the four groups of rats (Table 1). All rats that received epicardial fibrillation were successfully resuscitated and survived.

Table 1 Baseline characteristics

To investigate the cardioprotective effect of CANA, we established a CA/CPR rat model and CANA or an equivalent volume of placebo was intravenously administered 15 min after ROSC. Invasive arterial blood pressure is the most commonly used and basic index of hemodynamic measurement, and mean arterial pressure (MAP) was recorded every 30 min. We found that CANA can improve hemodynamics after resuscitation. All rats that received CPR had a significant decrease in blood pressure, which lasted for a period of time and gradually recovered at about 1 h after resuscitation. However, MAP in the control group showed a downward trend in the late stage of resuscitation, but CANA could prevent this trend and keep the blood pressure stable in the late stage of resuscitation. The difference in blood pressure between CPR + placebo and CPR + CANA groups was statistically significant (P < 0.05) at 3 h (Fig. 4).

Fig. 4
figure 4

Mean aortic pressure changes in mean arterial pressure during post-resuscitation in 4 groups (each group contains 6 rats); &P < 0.05, Sham + placebo vs. CPR + placebo; λP < 0.05, Sham + placebo vs. CPR + CANA; *P < 0.05, CPR + placebo vs. CPR + CANA

Figure 5 shows myocardial function in rats. Following resuscitation, model groups exhibited significant impairment in left ventricular ejection fraction (LVEF) at 2 h compared to baseline. As time progressed, cardiac function gradually improved in both groups that underwent CA/CPR procedures. Notably, animals treated with CANA demonstrated significantly less impaired cardiac function at 6 h after ROSC compared to the control group (P < 0.05).

Fig. 5
figure 5

Echocardiography. a Echocardiograms of rats in each group; b ejection fraction (EF); &P < 0.05, Sham + placebo vs. CPR + placebo; λP < 0.05, Sham + placebo vs. CPR + CANA; *P < 0.05, CPR + placebo vs. CPR + CANA

The concentrations of cTnI and NT-proBNP in each group were determined by the ELISA method. We found that the concentrations of cTnI and NT-proBNP were significantly increased in each model group after resuscitation, which was consistent with our previous experimental results. However, the concentrations of cTnI and NT-proBNP in the CPR + CANA group were significantly lower than those in the CPR + placebo group (P < 0.05) (Fig. 6). In addition, we further evaluated the myocardial histopathological changes after 6 h of resuscitation by HE staining. We can see that the rats in the sham-operated group had an interconnected network of myocardial fibers, neatly arranged, evenly stained nuclei, and visible striated; In the model group, the intercellular space of myocardial cells increased, the nuclear staining became light or even disappeared, the arrangement of myocardial fibers was disordered, and the transverse striations disappeared. In the CPR + CANA group, the myocardial cell texture was still clear, and the degree of interstitial swelling and cell destruction was reduced (Fig. 7). All these suggest that CANA can ameliorate the myocardial injury in CA/CPR.

Fig. 6
figure 6

Myocardial injury, inflammation and oxidative stress index. a NT-Pro BNP expression at 6 h after return of spontaneous circulation in four groups; b cTnI expression at 6 h after return of spontaneous circulation in four groups; c TNF-α expression at 6 h after return of spontaneous circulation in four groups; d IL-6 expression at 6 h after return of spontaneous circulation in four groups; e CAT expression at 6 h after return of spontaneous circulation in four groups; f SOD expression at 6 h after return of spontaneous circulation in four groups; g MDA expression at 6 h after return of spontaneous circulation in four groups; #P < 0.05 vs. the Sham + placebo group. *P < 0.05 vs. the CPR + placebo group

Fig. 7
figure 7

Myocardial pathological slices. Normal myocardial tissue morphological in Sham + placebo group (a) and Sham + CANA group (b), representative myocardial tissue morphological injuries in the CPR + placebo (c) and CPR + CANA group (d). Myocardial injury in the CPR + CANA group was reduced compared with the CPR + placebo group, as characterized by myocardial cell edema, transverse striations disappeared and cell arrangement disorder.

Using ELISA to detect indicators of myocardial oxidative stress injury, we found that the levels of SOD and CAT in the myocardial tissue of rats in the sham operation + CANA group were the highest. In the CPR + placebo group, MDA increased significantly (p < 0.05), while CAT and SOD decreased significantly (p < 0.05). We found that canagliflozin can alleviate this change and reduce the oxidative stress injury in the model group, which was manifested by a significant increase in myocardial tissue SOD and CAT content and a reduction in myocardial tissue MDA content (P < 0.05) (Fig. 6).

To determine the level of myocardial inflammation in rats after CPR, we detected the levels of TNF-α and IL-6 in the myocardial homogenate of rats in each group 6 h after CPR by ELISA. The results showed that the contents of TNF-α and IL-6 in the myocardium of the Sham + CANA group were the lowest. However, there was no significant difference between them and the Sham + placebo group. The model group up-regulated the level of myocardial inflammation, and the expression of TNF-α and IL-6 was significantly higher than that of the sham-operated group (P < 0.05). CANA significantly decreased the levels of TNF-α and IL-6 after CA/CPR (P < 0.05), suggesting that CANA can reduce the inflammatory response after cardiopulmonary resuscitation (Fig. 6).

To determine whether cardiac dysfunction after cardiopulmonary resuscitation is related to apoptosis, we detected the expression of apoptotic protein and anti-apoptotic protein in myocardial tissue homogenate of rats in each group by Western blotting 6 h after resuscitation. We found that the level of cardiomyocyte apoptosis in the CPR + placebo group was significantly higher than that in the Sham + placebo group, suggesting that apoptosis is involved in cardiac dysfunction after resuscitation. CANA could reduce CPR-induced apoptosis, and the expression of BAX and caspase-3 was significantly decreased, and the expression of Bcl-2 was significantly increased compared with the placebo group (P < 0.05) (Fig. 8).

Fig. 8
figure 8

Apoptosis index and Nrf2/HO-1 signaling pathway. a BAX expression at 6 h after return of spontaneous circulation in 4 groups; b Bcl-2 expression at 6 h after return of spontaneous circulation in four groups; c Caspase-3 expression at 6 h after return of spontaneous circulation in four groups; d Nrf2 expression at 6 h after return of spontaneous circulation in four groups; e HO-1 expression at 6 h after return of spontaneous circulation in four groups; #P < 0.05 vs. the Sham + placebo group. *P < 0.05 vs. the CPR + placebo group

More and more studies have shown that the Nrf2/HO-1 signaling pathway is important in oxidative stress and also closely related to inflammation and apoptosis. Therefore, we measured protein levels of key signaling pathway molecules, namely, Nrf2 and HO-1. We found that both CANA and I/R injury could up-regulate the expression of Nrf2 and HO-1 protein. However, the endogenous antioxidant effect regulated by CANA was not enough to resist the reperfusion-induced oxidative stress. The CPR + CANA group significantly increased the content of Nrf2 and HO-1 (P < 0.05), which increased this advantage, reduced oxidative stress injury in the model group, and even potentially regulated the inflammatory response and apoptosis. However, the specific regulatory mechanism needs to be further verified by cell experiments (Fig. 8).

Discussion

This study investigated whether intravenous injection of CANA during the initial resuscitation period can improve cardiac function after cardiac arrest in a CA/CPR rat model. The present study demonstrated that the administration of CANA significantly ameliorated PRMD induced by cardiac arrest and CPR with decreased myocardial injury score, increased MAP, decreased levels of cTnI and NT-ProBNP. We also found that CANA directly suppressed the expression of inflammatory cytokines (IL-6, TNF-α), reduced oxidative stress and apoptosis via a Nrf2/HO-1-dependent mechanism. In the rat model of cardiac arrest and CPR, CANA attenuated oxidative stress and inhibited pro-inflammatory and pro-apoptotic signaling pathways, providing a potentially novel mechanism underlying the protective effects against PRMD.

Cardiac arrest is a common clinical emergency. The heart stops beating and the blood supply is cut off, leading to acute organ failure throughout the whole body. Patients who do not receive effective medical care often die within this short period of time. CPR is a life-saving intervention that successfully restores circulation and cardiac function in patients with sudden cardiac arrest (SCA). Unfortunately, the outcomes of CPR are disappointing. Post-cardiac arrest syndrome (PCAS) is an independent risk factor that affects the survival rate of resuscitated patients and is closely related to prognosis [27]. Myocardial dysfunction is the most common manifestation of PCAS and the leading cause of early death in patients with cardiac arrest. More than 68% of patients develop PRMD [4, 6]. Therefore, it's of great value to identify novel therapeutic strategies to improve PRMD after CA/CPR.

In clinical studies, several sodium–glucose cotransporter 2 inhibitors (SGLT2i) have shown different cardiovascular advantages in individuals with heart failure, myocardial infarction, arrhythmia, and cardiomyopathy [28,29,30]. In addition, this cardioprotective effect was independent of glucose management. In other words, SGLT2 inhibitors showed cardioprotective benefits in non-diabetic subjects. The use of CANA in heart failure patients regardless of pre-existing diabetes is recommended in the CHIEF-HF clinical trial [31]. In recent EMMY study, empagliflozin significantly enhanced cardiac outcomes in patients with acute myocardial infarction following 72-h intervention [32]. In contrast to local I/R in myocardial infarction, in this study we investigated the cardioprotective effect of acute intravenous injection of CANA on systemic I/R injury after CA/CPR.

Currently, the most commonly used animal models of CA/CPR include asphyxia, electric shock ventricular fibrillation and the high potassium method [33]. Relevant studies have shown that rapid ventricular arrhythmias, including ventricular fibrillation and ventricular tachycardia, are the most common in sudden cardiac death (SCD) with complete ECG recordings [34]. Therefore, electric shock ventricular fibrillation seems more aligned with clinical practice than other methods. However, the traditional ventricular endocardium electric shock ventricular fibrillation method requires highly professional operation and has relatively significant trauma. In the present study, we used an improved trans epicardial electrical stimulation to establish a standard rat model of VF-induced cardiac arrest and resuscitation. In addition, we determined the dose of intravenous CANA based on available studies (3 ug/kg) [35]. Our study was the first study to investigate the effects of CANA on myocardial dysfunction after VF-induced cardiac arrest and CPR model in rats.

PRMD is a common complication in post-resuscitation patients, manifesting mainly as systolic and diastolic dysfunction, arrhythmias and recurrent cardiac arrest. This cardiac dysfunction is usually reversible and recovery is possible within 48–72 h. Approximately 2/3 of patients with VF-induced cardiac arrest develop circulatory dysfunction during the course of the disease, requiring pharmacological or mechanical intervention. However, failure to treat these disturbances promptly will result in an adverse outcome after cardiac arrest [36]. Myocardial tissue mild I/R injury can release cTnI and NT-ProBNP, which are routinely used in the clinic as indicators of early myocardial damage. Moreover, the concentrations of cTnI and NT-ProBNP indicate the severity of the myocardial injury [37, 38]. In our study, there was a significant increase in both cTnI and NT-ProBNP concentrations in the CA/CPR model group 6 h after resuscitation. The canagliflozin therapy intervention significantly reduced this rising trend, myocardial injury biomarkers in CPR + CANA group higher than sham surgery controls while significantly lower than CPR + placebo group. On echocardiography, LVEF showed similar changes. The left ventricular function decreased significantly after CA/CPR and improved gradually with time. CANA can accelerate the recovery of left ventricular function. The change suggests that CANA can ameliorate myocardial I/R injury. The same change was also confirmed in myocardial pathological sections. In the sham surgery group, the morphology and structure of myocardial cells were normal, and no inflammatory cell infiltration was observed. However, in the CPR + placebo group, edema of myocardial interstitial substance, light staining or even disappearance of the nucleus, and disorder of myocardial fibre arrangement were observed. Compared with the CPR + placebo group, CANA treatment significantly reduced the myocardial fibre swelling and distortion in CA/CPR rats.

Previous studies have shown that ROS can be generated immediately after ischaemia, and various ROS affect normal physiological functions of cells by damaging protein, lipid and even DNA structure [39, 40]. In addition, oxidative stress is a critical factor in the occurrence and development of cardiac I/R injury. Oxidative stress also links pro-inflammatory response and apoptosis to aggravate myocardial cell injury, except for direct toxic injury [41, 42]. In addition to glucose-lowering effects, antioxidant properties of SGLT inhibitors have been demonstrated [23, 43, 44]. As a highly selective inhibitor of SGLT-2, CANA reduces endothelial injury to slow coronary atherosclerosis by reducing ROS production and blocking oxidative stress signaling pathway (EGFR/Src/Rho-kinase) mediated by ROS [22]. Based on previous studies, we further verified that CANA can protect I/R cardiac by reducing oxidative stress. In this study, we found that the expression of oxidative markers was significantly increased in CA/CPR rat myocardium compared with the sham-operated group after 6 h of resuscitation. However, its expression in CANA + CA/CPR group showed a significant downward trend. We found that the expression of antioxidant enzymes in the myocardium of I/R rats was significantly upregulated after treatment with CANA.

During cardiac arrest, ischaemia, hypoxia and disruption of the energy chain drive injured cardiomyocytes to release a variety of DAMPs. After hearts pumping recovery, innate immune cells recognise DAMP and activate the TLR pathway to release excessive inflammatory factors and chemokines, causing inflammatory damage [45]. TNF-α and IL-6 are master cytokines of the inflammatory response. Under pathological conditions, myocardial cells and macrophages secrete TNF-α, which combines with membrane TNFR1 to form a complex. This complex participates in inflammation-mediated cell injury process, and also participates in pathophysiological reactions, such as the induction of cardiomyocyte apoptosis and the regulation of ventricular remodelling [46]. In addition, TNF-α can activate NF-KB, which further activates TNF-α, IL-6, and other inflammatory factors to form an inflammatory cascade response. IL-6 is a multi-effect secondary cytokine, often playing an anti-inflammatory role in the acute phase and gradually becoming pro-inflammatory after prolonged activation. It can mediate the release of oxygen-free radicals and proteolytic enzymes, regulate the excitation–contraction coupling of cardiomyocytes and promote myocardial fibrosis to damage the normal myocardial function [47]. Multiple meta-analyses have shown that IL-6 is significantly associated with all-cause mortality in multiple cardiovascular diseases [48, 49]. In this study, we found that the expression of relevant inflammatory factors (TNF-α, IL-6) in cardiac myocytes of CA/CPR rats was significantly increased after 6 h of resuscitation, consistent with previous studies [50]. In addition, CANA can significantly alleviate the expression of the above inflammatory factors. Previous studies have reported the anti-inflammatory performance of CANA in chronic kidney disease, fatty liver, atherosclerosis, myocarditis, and other diseases [51,52,53,54], showing the independent anti-inflammatory properties of CANA. Our results have shown that attenuation of the inflammatory response is also one of the mechanisms by which CANA alleviated myocardial injury after resuscitation.

Arrhythmias, myocardial stunning, no-reflow phenomena, and myocardial cell death due to reperfusion are the most common manifestations of myocardial I/R injury [55], whereas apoptosis is the most common form of reperfusion injury-induced cardiomyocyte death. Some studies have shown that apoptosis is not associated with myocardial dysfunction after resuscitation [56]. However, recent research has shown that prostaglandin E1 (PGE1) inhibits caspase-mediated apoptosis following resuscitation by reducing the opening of mitochondrial permeability transition pores (mPTP) in CA/CPR rat model and the H9C2 cells hypoxia/reoxygenation model [57]. Similarly, in an experimental study on porcine, caspase-3 expression and the number of TUNEL-stained positive cells in cardiac tissue 24 h after resuscitation were considerably greater in the CA/CPR group than in the sham group [58]. Apoptosis is a process in which cells die to adapt to their surroundings. It involves many signaling pathways, including the Fas/FasL pathway-mediated exogenous pathway and the mitochondria-mediated endogenous pathway, and caspase-3 is the final common pathway in most cases. In our investigation, we discovered that the expression of anti-apoptotic proteins BAX and caspase-3 was significantly decreased in the CPR + placebo group. In contrast, the expression of the apoptotic protein Bcl-2 was increased. However, canagliflozin could alleviate this apoptosis trend in I/R injury. We found that the degree of myocardial apoptosis in the CPR + CANA group was significantly decreased.

SGLT2 inhibitors have shown good cardiovascular benefits in many clinical trials. In diabetic patients, they can significantly improve early hemodynamics, direct myocardial protection (regulate myocardial energy metabolism, anti-myocardial fibrosis, etc.) and vascular protection (improve vascular endothelial function, regulate vascular remodeling, etc.) to improve cardiovascular event outcomes [23, 59,60,61,62]. As the cardiovascular protective effects independent of glucose-lowering effects are increasingly recognized, researchers redirect the focus of studies to non-diabetic patients, particularly in ischemic heart disease. Sabe et al. found that in a swine model of chronic myocardial ischaemia, CANA can reduce ischemic area and improve myocardial function through JAK/STAT pathway, AMPK pathway, and oxidative signaling pathways [63]. In a model of regional myocardial I/R, researchers also found that CANA can improve ischemic myocardium contractile function [64, 65]. However, most animal studies used CANA pretreatment with an intragastric administration time as long as several weeks, which was inconsistent with the actual situation in clinical emergency patients. Moreover, most of the experiments used the coronary artery ligation method, which could only induce local myocardial ischaemia but could not cause global I/R injury. Research about whether the acute administration of CANA alleviates myocardial dysfunction after whole-heart I/R is still at an early stage, and the underlying molecular mechanisms are unknown. The nuclear factor-erythroid 2-related factor 2 (Nrf2) signaling pathway plays an important role in I/R injury. Nrf2 and Keap1 form a complex in the cytoplasm under physiological circumstances. When oxidative stress products such as ROS are stimulated, Nrf2 dissociates from Keap1, increases nuclear translocation of Nrf2, binds to nuclear antioxidant response element AER, and induces downstream antioxidant stress gene transcription (such as HO-1), which initiates endogenous antioxidant pathways and maintains intracellular redox balance. Both I/R injury and CANA were observed to promote the expression of Nrf2 in our investigation. Endogenous antioxidant mechanisms, however, are unable to counteract the huge quantity of ROS produced during reperfusion. We discovered that intravenous use of CANA in the resuscitated rat significantly increased Nrf2 expression, activated the Nrf2/HO-1 signaling pathway, and ameliorated oxidative stress damage. Hasan et al. 's et al. also found that canagliflozin can reduce cardiac oxidative stress injury through the Nrf2/HO-1 pathway in the isoproterenol (ISO)-induced cardiac injury model [66]. Neutrophil infiltration and inflammatory cytokine release are critical steps in the development of IRI. Activation of the Nrf2/HO-1 pathway can alleviate I/R injury and inflammatory responses in tissues or organs. Some investigations have revealed that Nrf2 is a significant mediator linking the inflammatory response and oxidative stress, although its precise molecular mechanism remains unknown. Currently, relevant research focuses mostly on the regulation of Nrf2 and NLRP3 inflammasomes [67, 68]. For example, Hou et al. found that activation of the NLRP3 inflammatory complex plays an important role in inflammatory damage in brain I/R injury, and Nrf2 inhibits activation of the NLRP3 inflammatory complex by regulating the Trx1/TXNIP complex, thus playing a role in alleviating I/R injury [69]. The anti-inflammatory effect of the Nrf2 pathway may also be related to the NF-κB and MAPK pathways. Nrf2 has been shown to reduce the production of inflammatory factors and the occurrence of inflammatory responses by inhibiting the activation of NF-κB and MAPK. Furthermore, the downstream protein HO-1 promotes macrophage change from pro-inflammatory to anti-inflammatory [70, 71]. Its catalytic products have anti-inflammatory properties. Yan et al. discovered a more severe inflammatory response in both the I/R model of Nrf2−/−mice and the hypoxia/reoxygenation cell model with Nrf2 gene silencing in research on I/R-induced lung injury [72]. Reduced oxidative stress and inflammatory response can minimize cardiac cell damage and apoptosis, and the researchers have also found that activation of the Nrf2/HO-1 signaling pathway can reduce apoptosis following myocardial I/R[73, 74]. Lu et al. reported that Artesunate reduces apoptosis by up-regulating the expression of the anti-apoptotic transcription factor Nrf2 and down-regulating the ROS-dependent p38MAPK pathway [75]. Wang et al. found that neohesperidin can inhibit apoptosis and oxidative stress through activation of the Akt/Nrf2/HO-1 pathway, which can have a neuroprotective effect on cerebral IRI with neuroprotective effects [76]. Induced HO-1 expression in retransplanted hearts could significantly reduce the number of apoptotic cells. In addition, inhibition of HO-1 activity reversed this protective effect [77, 78]. Keap1/Nrf2/HO-1, SIRT1/Nrf2/HO-1 and others are important pathways to attenuate I/R injury [79,80,81]. As a result, we postulated that the anti-inflammatory, anti-oxidative stress and anti-apoptotic effects of CANA in myocardial I/R may be directly or indirectly regulated by the Nrf2/HO-1 signaling pathway.

Our study investigated the effects of acute administration of CANA after cardiac arrest on ischemic myocardium. We found that canagliflozin has a cardioprotective effect independent of its glucose-lowering effect, can improve the cardiac function of rats after resuscitation, and reduce the myocardial pathological changes induced by I/R. The specific mechanism may be related to the regulation of the Nrf2/HO-1 signaling pathway to improve myocardial antioxidant and anti-inflammatory capacity, and then reduce apoptosis. These results indicate that CANA is a potential drug the prevention and treatment of I/R injury and provide an experimental basis for the clinical application of CANA in ischemic cardiomyopathy.

Limitations

This study has several limitations. First, the experimental animals had no underlying disease, which is not consistent with the high-risk populations for cardiac arrest; second, CANA was administered at ROSC 15 min, which is not consistent with actual clinical administration; third, the CA + CANA + Nrf2 inhibitor group was not set up to verify the role of the Nrf2/HO-1 pathway; fourth, no corresponding cell experiments were performed; finally, the sample size of this study was relatively small.

Conclusions

We demonstrated for the first time that the acute use of canagliflozin alleviated post-resuscitation myocardial dysfunction after cardiac arrest and CPR in the rat model. The underlying mechanism could be through Nrf2 signaling pathway while suppressing proinflammatory and proapoptotic pathways. Our work describes canagliflozin, elucidates the potential mechanism of its cardioprotective effect, and in the future, it may be used to reduce post-resuscitation myocardial dysfunction in patients resuscitated by cardiac arrest and CPR.

Availability of data and materials

The data sets used and analysed during the current study are available from the corresponding author on reasonable request.

Abbreviations

CA:

Cardiac arrest

CANA:

Canagliflozin

ROSC:

Restore of spontaneous circulation

ECG:

Electrocardiogram

MAP:

Mean arterial pressure

LVEF:

Left ventricular ejection fraction

I/R:

Ischaemia/reperfusion

CPR:

Cardiopulmonary resuscitation

PRMD:

Post-resuscitation myocardial dysfunction

DAMPs:

Danger-associated molecular patterns

ROS:

Reactive oxygen species

T2DM:

Type 2 diabetes mellitus

VF:

Ventricular fibrillation

CO2:

Carbon dioxide

SCA:

Sudden cardiac arrest

PCAS:

Post-cardiac arrest syndrome

SGLT2i:

Sodium–glucose cotransporter 2 inhibitors

SCD:

Sudden cardiac death

PGE1:

Prostaglandin E1

References

  1. Zimmerman DS, Tan HL (2021) Epidemiology and risk factors of sudden cardiac arrest. Curr Opin Crit Care 27(6):613–616

    Article  PubMed  Google Scholar 

  2. Roberts BW, Kilgannon JH, Hunter BR et al (2019) Association between elevated mean arterial blood pressure and neurologic outcome after resuscitation from cardiac arrest: results from a multicenter prospective cohort study. Crit Care Med 47(1):93–100

    Article  PubMed  PubMed Central  Google Scholar 

  3. Oksanen T, Skrifvars M, Wilkman E et al (2014) Postresuscitation hemodynamics during therapeutic hypothermia after out-of-hospital cardiac arrest with ventricular fibrillation: a retrospective study. Resuscitation 85(8):1018–1024

    Article  PubMed  Google Scholar 

  4. Laurent I, Monchi M, Chiche JD et al (2002) Reversible myocardial dysfunction in survivors of out-of-hospital cardiac arrest. J Am Coll Cardiol 40(12):2110–2116

    Article  PubMed  Google Scholar 

  5. Lemiale V, Dumas F, Mongardon N et al (2013) Intensive care unit mortality after cardiac arrest: the relative contribution of shock and brain injury in a large cohort. Intensive Care Med 39(11):1972–1980

    Article  PubMed  Google Scholar 

  6. Ruiz-Bailén M, Aguayo de Hoyos E, Ruiz-Navarro S et al (2005) Reversible myocardial dysfunction after cardiopulmonary resuscitation. Resuscitation 66(2):175–181

    Article  PubMed  Google Scholar 

  7. Chalkias A, Xanthos T (2012) Pathophysiology and pathogenesis of post-resuscitation myocardial stunning. Heart Fail Rev 17(1):117–128

    Article  PubMed  Google Scholar 

  8. Niccoli G, Burzotta F, Galiuto L et al (2009) Myocardial no-reflow in humans. J Am Coll Cardiol 54(4):281–292

    Article  PubMed  Google Scholar 

  9. Saxena A, Russo I, Frangogiannis NG (2016) Inflammation as a therapeutic target in myocardial infarction: learning from past failures to meet future challenges. Transl Res 167(1):152–166

    Article  CAS  PubMed  Google Scholar 

  10. Ruparelia N, Chai JT, Fisher EA et al (2017) Inflammatory processes in cardiovascular disease: a route to targeted therapies. Nat Rev Cardiol 14(3):133–144

    Article  CAS  PubMed  Google Scholar 

  11. Boag SE, Andreano E, Spyridopoulos I (2017) Lymphocyte communication in myocardial ischemia/reperfusion injury. Antioxid Redox Signal 26(12):660–675

    Article  CAS  PubMed  Google Scholar 

  12. Hofmann U, Frantz S (2016) Role of T-cells in myocardial infarction. Eur Heart J 37(11):873–879

    Article  CAS  PubMed  Google Scholar 

  13. van der Pol A, van Gilst WH, Voors AA et al (2019) Treating oxidative stress in heart failure: past, present and future. Eur J Heart Fail 21(4):425–435

    Article  PubMed  Google Scholar 

  14. Kar S, Kambis TN, Mishra PK (2019) Hydrogen sulfide-mediated regulation of cell death signaling ameliorates adverse cardiac remodeling and diabetic cardiomyopathy. Am J Physiol Heart Circ Physiol 316(6):H1237–H1252

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ou D, Ni D, Li R et al (2022) Galectin-1 alleviates myocardial ischemia-reperfusion injury by reducing the inflammation and apoptosis of cardiomyocytes. Exp Ther Med 23(2):143

    Article  CAS  PubMed  Google Scholar 

  16. Neal B, Perkovic V, Mahaffey KW et al (2017) Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med 377(7):644–657

    Article  CAS  PubMed  Google Scholar 

  17. Sarraju A, Li J, Cannon CP et al (2021) Effects of canagliflozin on cardiovascular, renal, and safety outcomes in participants with type 2 diabetes and chronic kidney disease according to history of heart failure: results from the CREDENCE trial. Am Heart J 233:141–148

    Article  CAS  PubMed  Google Scholar 

  18. Correction to: 2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Circulation. 2022;146(13):e185.

  19. Arnett DK, Blumenthal RS, Albert MA et al (2019) 2019 ACC/AHA guideline on the primary prevention of cardiovascular disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol 74(10):e177–e232

    Article  PubMed  PubMed Central  Google Scholar 

  20. Koshino A, Schechter M, Sen T et al (2022) Interleukin-6 and cardiovascular and kidney outcomes in patients with type 2 diabetes: new insights from CANVAS. Diabetes Care 45(11):2644–2652

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Heerspink HJL, Perco P, Mulder S et al (2019) Canagliflozin reduces inflammation and fibrosis biomarkers: a potential mechanism of action for beneficial effects of SGLT2 inhibitors in diabetic kidney disease. Diabetologia 62(7):1154–1166

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. El-Daly M, Pulakazhi Venu VK, Saifeddine M et al (2018) Hyperglycaemic impairment of PAR2-mediated vasodilation: prevention by inhibition of aortic endothelial sodium-glucose-co-Transporter-2 and minimizing oxidative stress. Vascul Pharmacol 109:56–71

    Article  CAS  PubMed  Google Scholar 

  23. Li C, Zhang J, Xue M et al (2019) SGLT2 inhibition with empagliflozin attenuates myocardial oxidative stress and fibrosis in diabetic mice heart. Cardiovasc Diabetol 18(1):15

    Article  PubMed  PubMed Central  Google Scholar 

  24. Koizumi T, Watanabe M, Yokota T et al (2023) Empagliflozin suppresses mitochondrial reactive oxygen species generation and mitigates the inducibility of atrial fibrillation in diabetic rats. Front Cardiovasc Med 10:1005408

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Dabour MS, Abdelgawad IY, Grant MKO et al (2023) Canagliflozin mitigates carfilzomib-induced endothelial apoptosis via an AMPK-dependent pathway. Biomed Pharmacother 164:114907

    Article  CAS  PubMed  Google Scholar 

  26. Zheng C, Xuan W, Chen Z et al (2022) CX3CL1 worsens cardiorenal dysfunction and serves as a therapeutic target of canagliflozin for cardiorenal syndrome. Front Pharmacol 13:848310

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Yao Y, Johnson NJ, Perman SM et al (2018) Myocardial dysfunction after out-of-hospital cardiac arrest: predictors and prognostic implications. Intern Emerg Med 13(5):765–772

    Article  PubMed  Google Scholar 

  28. Wiviott SD, Raz I, Bonaca MP et al (2019) Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med 380(4):347–357

    Article  CAS  PubMed  Google Scholar 

  29. Cannon CP, Pratley R, Dagogo-Jack S et al (2020) Cardiovascular outcomes with Ertugliflozin in Type 2 diabetes. N Engl J Med 383(15):1425–1435

    Article  CAS  PubMed  Google Scholar 

  30. Bhatt DL, Szarek M, Steg PG et al (2021) Sotagliflozin in patients with diabetes and recent worsening heart failure. N Engl J Med 384(2):117–128

    Article  CAS  PubMed  Google Scholar 

  31. Spertus JA, Birmingham MC, Nassif M et al (2022) The SGLT2 inhibitor canagliflozin in heart failure: the CHIEF-HF remote, patient-centered randomized trial. Nat Med 28(4):809–813

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. von Lewinski D, Kolesnik E, Tripolt NJ et al (2022) Empagliflozin in acute myocardial infarction: the EMMY trial. Eur Heart J 43(41):4421–4432

    Article  Google Scholar 

  33. Vognsen M, Fabian-Jessing BK, Secher N et al (2017) Contemporary animal models of cardiac arrest: a systematic review. Resuscitation 113:115–123

    Article  PubMed  Google Scholar 

  34. McElwee SK, Velasco A, Doppalapudi H (2016) Mechanisms of sudden cardiac death. J Nucl Cardiol 23(6):1368–1379

    Article  PubMed  Google Scholar 

  35. Sayour AA, Korkmaz-Icöz S, Loganathan S et al (2019) Acute canagliflozin treatment protects against in vivo myocardial ischemia-reperfusion injury in non-diabetic male rats and enhances endothelium-dependent vasorelaxation. J Transl Med 17(1):127

    Article  PubMed  PubMed Central  Google Scholar 

  36. Jentzer JC, Chonde MD, Dezfulian C (2015) Myocardial dysfunction and shock after cardiac arrest. Biomed Res Int 2015:314796

    Article  PubMed  PubMed Central  Google Scholar 

  37. Yang M, Hu X, Lu X et al (2015) The effects of α- and β-adrenergic blocking agents on postresuscitation myocardial dysfunction and myocardial tissue injury in a rat model of cardiac arrest. Transl Res 165(5):589–598

    Article  CAS  PubMed  Google Scholar 

  38. Wang J, Lin J, Zhang M et al (2017) Hydrogen can alleviate post-cardiac arrest myocardium injury in rabbits. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue 29(10):911–915

    PubMed  Google Scholar 

  39. Zhu X, Zuo L (2013) Characterization of oxygen radical formation mechanism at early cardiac ischemia. Cell Death Dis 4(9):e787

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Xiang M, Lu Y, Xin L et al (2021) Role of oxidative stress in reperfusion following myocardial ischemia and its treatments. Oxid Med Cell Longev 2021:6614009

    Article  PubMed  PubMed Central  Google Scholar 

  41. Qiao SG, Sun Y, Sun B et al (2019) Sevoflurane postconditioning protects against myocardial ischemia/reperfusion injury by restoring autophagic flux via an NO-dependent mechanism. Acta Pharmacol Sin 40(1):35–45

    Article  CAS  PubMed  Google Scholar 

  42. Sundd P, Gladwin MT, Novelli EM (2019) Pathophysiology of sickle cell disease. Annu Rev Pathol 14:263–292

    Article  CAS  PubMed  Google Scholar 

  43. Kolijn D, Pabel S, Tian Y et al (2021) Empagliflozin improves endothelial and cardiomyocyte function in human heart failure with preserved ejection fraction via reduced pro-inflammatory-oxidative pathways and protein kinase Gα oxidation. Cardiovasc Res 117(2):495–507

    Article  CAS  PubMed  Google Scholar 

  44. Semo D, Obergassel J, Dorenkamp M et al (2023) The sodium-glucose co-transporter 2 (SGLT2) inhibitor empagliflozin reverses hyperglycemia-induced monocyte and endothelial dysfunction primarily through glucose transport-independent but redox-dependent mechanisms. J Clin Med. https://doi.org/10.3390/jcm12041356

    Article  PubMed  PubMed Central  Google Scholar 

  45. Hu S-Y, Yang J-E, Zhang F (2022) Signaling pathways of inflammation in myocardial ischemia/reperfusion injury. Cardiol Plus 7(1):29–38

    Article  CAS  Google Scholar 

  46. Galeone A, Grano M, Brunetti G (2023) Tumor necrosis factor family members and myocardial ischemia-reperfusion injury: state of the art and therapeutic implications. Int J Mol Sci. https://doi.org/10.3390/ijms24054606

    Article  PubMed  PubMed Central  Google Scholar 

  47. Liu Y, Zhang D, Yin D (2023) Pathophysiological effects of various interleukins on primary cell types in common heart disease. Int J Mol Sci. https://doi.org/10.3390/ijms24076497

    Article  PubMed  PubMed Central  Google Scholar 

  48. Aimo A, Vergaro G, Passino C et al (2017) Prognostic value of soluble suppression of tumorigenicity-2 in chronic heart failure: a meta-analysis. JACC Heart Fail 5(4):280–286

    Article  PubMed  Google Scholar 

  49. Li H, Liu W, Xie J (2017) Circulating interleukin-6 levels and cardiovascular and all-cause mortality in the elderly population: a meta-analysis. Arch Gerontol Geriatr 73:257–262

    Article  CAS  PubMed  Google Scholar 

  50. Braunstein M, Williamson M, Kusmenkov T et al (2017) Significant cytokine mRNA expression changes immediately after initiation of cardiopulmonary resuscitation. Mediators Inflamm 2017:8473171

    Article  PubMed  PubMed Central  Google Scholar 

  51. Bellanti F, Lo Buglio A, Dobrakowski M et al (2022) Impact of sodium glucose cotransporter-2 inhibitors on liver steatosis/fibrosis/inflammation and redox balance in non-alcoholic fatty liver disease. World J Gastroenterol 28(26):3243–3257

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Long Q, Li L, Yang H et al (2022) SGLT2 inhibitor, canagliflozin, ameliorates cardiac inflammation in experimental autoimmune myocarditis. Int Immunopharmacol 110:109024

    Article  CAS  PubMed  Google Scholar 

  53. Nasiri-Ansari Ν, Dimitriadis GK, Agrogiannis G et al (2018) Canagliflozin attenuates the progression of atherosclerosis and inflammation process in APOE knockout mice. Cardiovasc Diabetol 17(1):106

    Article  CAS  PubMed  Google Scholar 

  54. Solomon J, Festa MC, Chatzizisis YS et al (2023) Sodium-glucose co-transporter 2 inhibitors in patients with chronic kidney disease. Pharmacol Ther 242:108330

    Article  CAS  PubMed  Google Scholar 

  55. Vander Heide RS, Steenbergen C (2013) Cardioprotection and myocardial reperfusion: pitfalls to clinical application. Circ Res 113(4):464–477

    Article  CAS  PubMed  Google Scholar 

  56. Song F, Shan Y, Cappello F et al (2010) Apoptosis is not involved in the mechanism of myocardial dysfunction after resuscitation in a rat model of cardiac arrest and cardiopulmonary resuscitation. Crit Care Med 38(5):1329–1334

    Article  PubMed  Google Scholar 

  57. Su C, Fan X, Xu F et al (2021) Prostaglandin E1 attenuates post-cardiac arrest myocardial dysfunction through inhibition of mitochondria-mediated cardiomyocyte apoptosis. Mol Med Rep. https://doi.org/10.3892/mmr.2020.11749

    Article  PubMed  PubMed Central  Google Scholar 

  58. Liu S, Xu J, Gao Y et al (2018) Multi-organ protection of ulinastatin in traumatic cardiac arrest model. World J Emerg Surg 13:51

    Article  PubMed  PubMed Central  Google Scholar 

  59. Alshnbari AS, Millar SA, O’Sullivan SE et al (2020) Effect of sodium-glucose cotransporter-2 inhibitors on endothelial function: a systematic review of preclinical studies. Diabetes Ther 11(9):1947–1963

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Hallow KM, Helmlinger G, Greasley PJ et al (2018) Why do SGLT2 inhibitors reduce heart failure hospitalization? A differential volume regulation hypothesis. Diabetes Obes Metab 20(3):479–487

    Article  CAS  PubMed  Google Scholar 

  61. Takahashi H, Nomiyama T, Terawaki Y et al (2019) Combined treatment with DPP-4 inhibitor linagliptin and SGLT2 inhibitor empagliflozin attenuates neointima formation after vascular injury in diabetic mice. Biochem Biophys Rep 18:100640

    PubMed  PubMed Central  Google Scholar 

  62. Verma S, Rawat S, Ho KL et al (2018) Empagliflozin increases cardiac energy production in diabetes: novel translational insights into the heart failure benefits of SGLT2 inhibitors. JACC Basic Transl Sci 3(5):575–587

    Article  PubMed  PubMed Central  Google Scholar 

  63. Sabe SA, Xu CM, Sabra M et al (2023) Canagliflozin improves myocardial perfusion, fibrosis, and function in a swine model of chronic myocardial ischemia. J Am Heart Assoc 12(1):e028623

    Article  CAS  PubMed  Google Scholar 

  64. Baker HE, Kiel AM, Luebbe ST et al (2019) Inhibition of sodium-glucose cotransporter-2 preserves cardiac function during regional myocardial ischemia independent of alterations in myocardial substrate utilization. Basic Res Cardiol 114(3):25

    Article  PubMed  PubMed Central  Google Scholar 

  65. Baker HE, Tune JD, Mather KJ et al (2022) Acute SGLT-2i treatment improves cardiac efficiency during myocardial ischemia independent of Na(+)/H(+) exchanger-1. Int J Cardiol 363:138–148

    Article  PubMed  Google Scholar 

  66. Hasan R, Lasker S, Hasan A et al (2020) Canagliflozin attenuates isoprenaline-induced cardiac oxidative stress by stimulating multiple antioxidant and anti-inflammatory signaling pathways. Sci Rep 10(1):14459

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Fan ZX, Yang CJ, Li YH et al (2023) Ginsenoside Rh2 attenuates myocardial ischaemia-reperfusion injury by regulating the Nrf2/HO-1/NLRP3 signalling pathway. Exp Ther Med 25(1):35

    Article  CAS  PubMed  Google Scholar 

  68. Zeng J, Chen Y, Ding R et al (2017) Isoliquiritigenin alleviates early brain injury after experimental intracerebral hemorrhage via suppressing ROS- and/or NF-κB-mediated NLRP3 inflammasome activation by promoting Nrf2 antioxidant pathway. J Neuroinflamm 14(1):119

    Article  Google Scholar 

  69. Hou Y, Wang Y, He Q et al (2018) Nrf2 inhibits NLRP3 inflammasome activation through regulating Trx1/TXNIP complex in cerebral ischemia reperfusion injury. Behav Brain Res 336:32–39

    Article  CAS  PubMed  Google Scholar 

  70. Wenzel P, Rossmann H, Müller C et al (2015) Heme oxygenase-1 suppresses a pro-inflammatory phenotype in monocytes and determines endothelial function and arterial hypertension in mice and humans. Eur Heart J 36(48):3437–3446

    Article  CAS  PubMed  Google Scholar 

  71. Shen K, Jia Y, Wang X et al (2021) Exosomes from adipose-derived stem cells alleviate the inflammation and oxidative stress via regulating Nrf2/HO-1 axis in macrophages. Free Radic Biol Med 165:54–66

    Article  CAS  PubMed  Google Scholar 

  72. Yan J, Li J, Zhang L et al (2018) Nrf2 protects against acute lung injury and inflammation by modulating TLR4 and Akt signaling. Free Radic Biol Med 121:78–85

    Article  CAS  PubMed  Google Scholar 

  73. Zhang J, Pan W, Zhang Y et al (2022) Comprehensive overview of Nrf2-related epigenetic regulations involved in ischemia-reperfusion injury. Theranostics 12(15):6626–6645

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Hou W, Zhu X, Liu J et al (2020) Correction to: Inhibition of miR-153 ameliorates ischemia/reperfusion-induced cardiomyocytes apoptosis by regulating Nrf2/HO-1 signaling in rats. Biomed Eng Online 19(1):43

    Article  PubMed  PubMed Central  Google Scholar 

  75. Lu H, Wang B, Cui N et al (2018) Artesunate suppresses oxidative and inflammatory processes by activating Nrf2 and ROS-dependent p38 MAPK and protects against cerebral ischemia-reperfusion injury. Mol Med Rep 17(5):6639–6646

    CAS  PubMed  Google Scholar 

  76. Wang JJ, Cui P (2013) Neohesperidin attenuates cerebral ischemia-reperfusion injury via inhibiting the apoptotic pathway and activating the Akt/Nrf2/HO-1 pathway. J Asian Nat Prod Res 15(9):1023–1037

    Article  CAS  PubMed  Google Scholar 

  77. Akamatsu Y, Haga M, Tyagi S et al (2004) Heme oxygenase-1-derived carbon monoxide protects hearts from transplant associated ischemia reperfusion injury. FASEB J 18(6):771–772

    Article  CAS  PubMed  Google Scholar 

  78. Katori M, Buelow R, Ke B et al (2002) Heme oxygenase-1 overexpression protects rat hearts from cold ischemia/reperfusion injury via an antiapoptotic pathway. Transplantation 73(2):287–292

    Article  CAS  PubMed  Google Scholar 

  79. Hou Z, Yang F, Chen K et al (2023) hUC-MSC-EV-miR-24 enhances the protective effect of dexmedetomidine preconditioning against myocardial ischemia-reperfusion injury through the KEAP1/Nrf2/HO-1 signaling. Drug Deliv Transl Res. https://doi.org/10.1007/s13346-023-01388-7

    Article  PubMed  Google Scholar 

  80. Huo K, Xu J, Wei M et al (2023) Solasonine ameliorates cerebral ischemia-reperfusion injury via suppressing TLR4/MyD88/NF-κB pathway and activating AMPK/Nrf2/HO-1 pathway. Int Immunopharmacol 124(Pt A):110862

    Article  CAS  PubMed  Google Scholar 

  81. Lu C, Jiang B, Xu J et al (2023) Neferine protected cardiomyocytes against hypoxia/oxygenation injury through SIRT1/Nrf2/HO-1 signaling. J Biochem Mol Toxicol 37(8):e23398

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

All work was performed at Laboratory of Cardiopulmonary Resuscitation and Critical Care Medicine at the second hospital of Anhui medical university.

Funding

This study was supported by a research grant from the National Natural Science Foundation of China (No. 82072134), the Fund Project of Anhui Medical University (No. 9101001821), the Fund Project of Anhui Medical University (No. 9101001804), the Fund Project of Anhui Medical University (No.2019xkjT028), the Fund Project of Anhui Medical University (No. 2020GMFY05) and 2020 universities Talents Training Project of Anhui Provincial Education Department Grant (gxyq2020007).

Author information

Authors and Affiliations

Authors

Contributions

MW, YZ, WS, QH and LZ analysed and interpreted data. TH and YC drafted the manuscript. MY was in charge of manuscript revision. All authors have read and approved the final manuscript.

Corresponding author

Correspondence to Min Yang.

Ethics declarations

Ethics approval and consent to participate

The Animal Experiment Ethics Committee of Anhui Medical University approved this study. All experimental animals were treated according to the Guide for the Care and Use of Laboratory Animals (NIH Publication No. 85-23, revised 1996).

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hua, T., Chu, Y., Wang, M. et al. Protective effect of canagliflozin on post-resuscitation myocardial function in a rat model of cardiac arrest. ICMx 11, 78 (2023). https://doi.org/10.1186/s40635-023-00562-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40635-023-00562-y

Keywords