Skip to main content

Adenosine 2 receptor regulates autophagy and apoptosis to alleviate ischemia reperfusion injury in type 2 diabetes via IRE-1 signaling

Abstract

Purpose

This study aimed to determine the effect and mechanism of action of adenosine 2 receptor (A2R) activation on myocardial ischemia reperfusion injury (MIRI) under diabetic conditions.

Methods

MIRI type 2 diabetic rats and H9C2 cardiomyocytes were treated with A2R agonist and then subjected to hypoxia for 6 h and reoxygenation for 18 h. Myocardial damage, and infarct size were determined by cardiac ultrasound. Indicators of cardiomyocyte injury, creatine kinase-MB and cardiac troponin I were detected by Enzyme Linked Immunosorbent Assay. Endoplasmic reticulum stress (ERS) was determined through measuring the expression levels of ERS related genes GRP78, p-IRE1/IRE1, and p-JNKJNK. The mechanism of A2R cardio protection in MIRI through regulating ERS induced autophagy was determined by investigating the ER resident protein IRE-1. The ER-stress inducer Tunicamycin, and the IRE-1 inhibitor STF in combination with the A2R agonist NECA were used, and the cellular responses were assessed through autophagy proteins expression Beclin-1, p62, LC3 and apoptosis.

Results

NECA improved left ventricular function post MIRI, limited myocardial infarct size, reduced myocardial damage, decreased cardiomyocytes apoptosis, and attenuated ERS induced autophagy through regulating the IRE-XBP1s-CHOP pathway. These actions resulted into overall protection of the myocardium against MIRI.

Conclusion

In summary, A2R activation by NECA prior to ischemia attenuates apoptosis, reduces ERS induced autophagy and restores left ventricular function. This protective effect occurs through regulating the IRE1-XBPs-CHOP related mechanisms. NECA is thus a potential target for the treatment of MIRI in patient with type 2 diabetes.

Peer Review reports

Introduction

Myocardial infarction (MI) is an ischemic heart condition whose risk remains very high among type 2 diabetic (T2DM) patients [1, 2]. Indeed, T2DM patients are twice as likely to suffer ischemic heart disease, and have a significantly raised post MI mortality rate compared to non-diabetics [3, 4]. First line treatment for acute myocardial ischemia to save the ischemic myocardium is reperfusion. However, this treatment is often associated with tissue injury in a condition called “myocardial ischemia reperfusion injury” (MIRI). Various factors are responsible for MIRI including cellular autophagy, which decreases the number of cardiomyocytes needed to repair cardiac systolic function [5]. Moreover, T2DM has been shown to exacerbate endoplasmic reticulum stress mediated autophagy in the pancreas [6]. Therefore, inhibition of autophagy could be a way of alleviating MIRI.

During MI, the extracellular concentration of the purine nucleoside molecule, Adenosine, has been found to dramatically increase, indicating a possible function in MI [7]. Studies have shown that binding of adenosine to its receptors (A1, A2a, A2b and A3); that are extensively expressed on the mammalian myocardium, has beneficial effects on MIRI [8]. Adenosine 2a receptor in particular is cardioprotective during reperfusion and this cardio protection is associated with downregulated autophagy [9]. A recent study demonstrated that activation of A2R in diabetic rats protects against MIRI by preventing apoptosis [10]. Whether the endoplasmic reticulum stress induced autophagy is involved is not yet clear.

Endoplasmic reticulum stress (ERS) activates a process called autophagy, where nutrients are recycled through lysosomal degradation of unfolded or misfolded proteins and worn out cellular organelles [11]. Under extreme stress such as those induced by MIRI, autophagy is activated to encourage cellular survival and energy conservation [12]. However, autophagy is a double edged sword; enhanced autophagy following MIRI can lead to significant reduction of viable cardiomyocytes which impairs recovery of ventricular functions and exacerbate the condition. In normal cells, activation of A2R promotes cardio-protection through regulating autophagy [9]. Whether the ER is involved has not been elucidated. Previous studies have demonstrated that the endoplasmic reticulum membrane protein, IRE-1 regulates autophagy in various cells in response to ER- stress [13,14,15]. In this study, we examined whether A2R activation regulates ERS induced autophagy in diabetic rats to offer cardio protection during MIRI, and the mechanism involved.

Materials and methods

Animals

Fifty Sprague Dawley (SD) adult rats (250–280 g) were offered by Hubei Experimental Animal Research Center for this experiment (License No.: SCXK (E) 2015-0018). All animal experiments were approved by the Animal Experiment Committee of Wuhan University (Ethical approval number: WP2020-01108), and strictly conducted in compliance with the study protocol, the ARRIVE guidelines, and the Guide for the Care and Use of Laboratory animals (No. 85-23, revised 1996) published by National Institutes of Health (NIH).

H9C2 myocardial cells

The rat embryonic cells H9C2 cells were provided by Procell (Wuhan, China) and maintained in Dulbecco’s Modified Eagle Medium (DMEM, Thermofischer scientific, USA) supplemented with 10% fetal bovine serum (FBS, Life technologies, Carlsbad, Ca, USA) and 1% Streptomycin-Penicillin antibiotics (FBS, Life technologies, Carlsbad, Ca, USA). Cells were cultured at 37 °C in 5% CO2.

Establishment of ischemia reperfusion injury model in vitro

After 72 h of culture, the neonatal rat cardiomyocytes (NRCM) were subjected to hypoxia reperfusion (H/R) to induce ischemia reperfusion injury. The cells were cultured for 6 h in a glucose-free, serum-free medium in the presence of 95% N2 and 5% CO2. Cells were then re-oxygenated for 18 h in fresh DMEM/F12 supplemented with 10% FBS, 1% penicillin–streptomycin, 1% BrdU an incubated at 37 °C and 5% CO2 [29].

Measurement of autophagic flux

One day prior to ischemia reperfusion (I/R), the H9C2 cells were transfected with RFP-GFP-LC3 and LAMP-2 for 24 h to observe co-localization. Immediately after reoxygenation, cells were fixed with 4% paraformaldehyde, and then stained with DAPI. Confocal microscope (LEICA TCS SP8, Germany) was used to observe the fluorescence signal under multiple visions; yellow spots representing autophagosomes, and red spots representing autolysosomes. Images were analyzed using image J software.

Measurement of endoplasmic reticulum stress

Cultured in a six plate well were removed, culture medium discarded and the cells washed 3 times with PBS for 5 min each. Cells were then fixed for 20 min with 4% paraformaldehyde, and then washed again 3 times with PBS for 5 min each. A histochemical pen was used to draw a circle to prevent the incubation solution from flowing away in the following process. Cells were then incubated overnight at 4 °C in primary antibodies (GRP8, dilution: 1: 1000; p-IRE1, dilution: 1: 500; XBP1, dilution: 1: 500) diluted with 5% BSA. The following day, they were washed 3 times with PBS for 5 min each and re-incubated in the dark in corresponding secondary antibodies (HRP-Goat anti Rabbit, and HRP-Goat anti mouse, dilutions: 1: 10,000 each) at 37 °C for 40 min, later washed with PBS and then stained with DAPI, incubated in the dark for 20 min at room temperature. They were washed with PBS and the film sealed with an anti-fluorescence quenching solution, then observed under the microscope.

Detection of cell survival rate by CCK-8

Cells were cultured for 16 h at 9.5 × 105/cm2 density in a 48-well plate, 10% CCK-8 solution was then added to the culture medium and further incubated for 2 h. A cell free control group was created and CCK-8 solution added as well. Absorbance was then measured at 450 nm (PerkinElmer EnSpire Microplate Reader, USA) and the optical density (OD) determined to calculate cell viability.

RNA extraction, reverse transcription and qRT-PCR

Total RNA was extracted from the cardiomyocytes using TRIpure Total RNA Extraction kit (Cat: EP013) form ELK Biotechnology Co. Ltd., China according to the manufacturer’s instructions. Reverse transcription to make cDNA was conducted using M-MLV Reverse Transcriptase kit (Cat: EQ002) also from ELK Biotechnology Co. Ltd., China. Quantitative real-time PCR was performed using StepOne™ Real-Time PCR system (Life Technologies, CA, USA) using QuFast SYBR Green PCR Master Mix (Cat: EQ001, ELK Biotechnology Co.Ltd., China. A 10-μl total reaction volume was used, and the reaction were as follows: 95 °C for 1 min; 40 cycles of 95 for 15 s, 58 °C for 20 s and 72 °C for 45 s, melting curve 60 °C → 95 °C, with 1 °C temperature increase every 20 s. Beta-actin was used as the endogenous control for data normalization and the double delta method used to calculate gene expression. Ct values used for calculations were averages of three independent repeats. All primer information is provided in Additional file 1: Table S1.

Establishment of diabetic rats’ model

To establish the diabetic model, SD rats were fed on a high fat diet containing 60% fats, 20% carbohydrates and 20% proteins for 30 days. After the 30 days, they were allowed to fast for 12 h, weighed and then injected with l% Streptozotocin citrate buffer at a 45 mg/kg dose. They continued to be fed on the high fat diet, and after 72 h, blood glucose from the tail veins were measured. The glucose levels were all greater than 16.7 mmol/L, indicating successful development of type 2 diabetes mellitus. The rats continued to be fed on high fat diet to maintain chronic type II diabetes (Additional file 2, Additional file 3, Additional file 4, Additional file 5 and Additional file 6).

Myocardial ischemia-reperfusion injury model

To develop myocardial ischemia reperfusion models, each rat was weighed, and anesthetized by injection with 1% sodium pentobarbital intraperitoneally at 50 mg/Kg. A cut was then made along the midline of the neck, muscles and tissues separated, the trachea exposed and an inverted T-shaped incision made along the cartilage ring of the trachea to intubate it. Parameters of the ventilator were set as: VT = 30–50ML/; frequency = 50–70 times/min; and breathing ratio = 1:1. The left thorax was opened to expose the heart. A 5–0 suture was used to ligate the left anterior descending coronary artery (LAD) at about 2 mm under the junction between the pulmonary artery cone and the left atrial appendage. To ensure reversible LAD occlusion, an openable knot was created by passing a ploythene tube through it just before it was tightened. In the sham group, no ligation was conducted. Ischemia was confirmed when transient drop in blood pressure was noticed, and the surface of the myocardium turned blue, indicating cyanosis. Reperfusion recovery was demonstrated by the rapid disappearance of cyanosis and hyperemic responses in the epicardium. Occlusion was conducted for 30 min and reperfusion 120 min then the rats euthanized by anesthesia overdose.

Hemodynamic measurements

Following anesthesia, while in the supine position, a puncture was made in the right internal carotid artery and an electrocardiogram connected. (BL-420, Taimeng Informatization Biological Signal Acquisition and Analysis System, China). 100U/Kg heparin anticoagulant was then injected and a catheter inserted into the right internal carotid artery to monitor the arterial blood pressure of the rat.

Echocardiography

The GE Vivid 7 (GE Health Medical, USA), was used to conduct transthoracic echocardiography. It was fitted with an 11 MHz image transducer to generate images. The ultrasound personnel was blinded to the study. Data collected were on: structure of the left ventricle, systolic ventricular septal thickness (IVSs), diastolic ventricular septal thickness (IVSd), left ventricular end-systolic diameter (LVIDs), left ventricular end-diastolic diameter (LVIDd), left ventricular diastolic posterior wall thickness (LVPWd), and left posterior wall thickness (LVPWs). These were all collected by M-mode ultrasound, while left ventricular end-systolic volume (LVESV), Left ventricular end-diastolic volume (LVEDV), left ventricular ejection fraction (LVEF), stroke volume (SV) and left ventricular fractional shortening (FS) were calculated automatically using alogarithms in the computer.

Measurement of myocardial infarction area (Evans blue staining)

Once reperfusion was complete, the reversible LAD occlusion was fully ligated. Large amounts of pentobarbital sodium was administered via the tail vain to euthanize the rats, while Evans blue dye was injected using the femoral vein. The hearts were removed, washed with phosphate buffered saline 3 times and then quickly frozen for 10 min at − 80 °C. The ventricular part was then sliced from the top into small equal parts. They were then incubated in 1% triphenyl tetrazolium chloride (TTC) for 15 min at 37 °C and allowed to slowly stain. The sections were then converted into digital photographs, and measurements of the myocardial infarction area (pale), ischemic risk area (red) and the non-infarct area (blue) carried out using the Image Pro plus 6.0 software.

Experimental procedures

The diabetic rats were randomly divided into sham operation group (Sham group), myocardial ischemia–reperfusion group (I/R group), myocardial ischemia–reperfusion + Adenosine receptor A2 agonist group (I/R + NECA group), myocardial ischemia–reperfusion + NECA + ERS inducer group (I/R + NECA + TM group), myocardial ischemia–reperfusion + adenosine receptor A2 agonist + ERS inducer group + IRE1 inhibitor group (I/R + NECA + TM + STF group). NECA was administered 10 min before ischemia.

The cardiomyocytes HC9c2 after high glucose treatments were randomly divided into sham operation group (HC9c2 + high glucose), Hypoxia-reperfusion group (HC9c2 + high glucose + H/R), myocardial ischemia–reperfusion + Adenosine receptor A2 agonist group (HC9c2 + high glucose + NECA + H/R), myocardial ischemia–reperfusion + NECA + ERS inducer group (HC9c2 + high glucose + NECA + Tunicamycin + H/R), myocardial ischemia–reperfusion + adenosine receptor A2 agonist + ERS inducer group + IRE1 inhibitor group (HC9c2 + high glucose + NECA + Tunicamycin + STF + H/R). NECA was administered 10 min before hypoxia.

Protein extraction and western blot

Radio immunoprecipitation assay (RIPA) lysis buffer (Beyotime Biotechnology, Shanghai, China), mixed with protease inhibitor, phenylmethylsulfonyl fluoride (PMSF) (Thermofischer scientific, USA) was used to extract total protein from cells and tissues. Final protein concentration was measured using Bicinchoninic acid (BCA) assay kit (Beyotime Biotechnology, Shanghai, China). Appropriate concentrations were determined and the proteins loaded onto 10% SDS PAGE gels and separated by electrophoresis. The proteins were then transferred on to polyvinylidene difluoride (PVDF) membranes, (Sigma-Aldrich, Beijing, China) incubated in primary antibodies overnight at 4 °C, then incubated in appropriate secondary antibodies for 1 h at room temperature and finally target bands visualized using the ECL imaging system (model 5200, Tianneng, China).

Detection of cardiac troponin and creatine kinase-MB levels

Enzyme linked immunosorbent assay (ELISA) was used to detect indicators of cardiomyocyte injury, CK-MB (creatine kinase-MB) and cTnI (cardiac troponin I), using kits obtained from CUSABIO BIOTECH Co., Ltd., Wuhan, China following the corresponding manufacturer’s instruction. 4 ml of blood samples were tapped into EDTA anticoagulant tubes from the femoral vein 2 h post reperfusion, centrifuged at 2000 rpm for 20 min at 4 °C and ELISA conducted.

Hematoxylin and Eosin staining

Heart tissues quickly harvested after reperfusion were washed 3 times in sterile PBS, and fixed using formalin for 24 h. The fixed tissues were then successively dehydrated in ethanol of increasing concentration; 70%, 80%, 90%, 95%, and 100%, then washed in xylene before paraffin embedding. 5 μm paraffin tissue blocks were prepared, and the tissue sections progressively dewaxed in xylene. They were then dehydrated in decreasing concentration of ethanol; 100%, 95%, 80%, and 75%), and then stained using hematoxylin and eosin. The tissues were then dehydrated, and sealed with neutral resins, and observed under the microscope.

Tunel assay

To further demonstrate apoptosis, Tunnel assay was conducted. Briefly, cells were grown on small glass slides inside a six well plate. The cells were fixed with 4% formaldehyde, washed 3 times with PBS, incubated in 70% ethanol for 30 min at 4 °C, again washed again 3 times with PBS, then incubated in a mixture of TdT enzyme and dUTP for 60 min at 37 °C. The cells were then rinsed with PBS, and incubated with DAPI counter stain for 10 min at room temperature and kept in the dark. They were then washed 3 times in PBS and re incubated with 7-AAD / RNase A solution and incubated for 30 min at room temperature then observed under a fluorescence microscope and images collected.

Electron microscopy

Following reoxygenation, cells were fixed in 2.5% glutaraldehyde for 4 h at 4 °C, rinsed 3 times using 0.1 M phosphoric acid buffer then fixed for 2 h in 1% osmic acid. Gradual dehydration in increasing concentration of ethanol was conducted; (50%, 70%, 90%, 100%), then permeated, and embedded in epoxy resin for 48 h at 60 °C. An ultrathin slicer (Leica, EM UC7, German) was used to make 80 nm slices of the embedded tissues and then stained with 2% uranyl acetate and lead citrate. Cellular morphology and autophagic flux were observed under an electron transmission microscope (Hitachi TEM system, HT7800, Japan).

Statistical analysis

All data were expressed as mean ± standard error of the mean unless explicitly specified, and analyzed statistically using GraphPad Prism 8.0 (GraphPad Software, Inc., La Jolla, CA). Normality tests were conducted and data normalized to the sham/control group. The unpaired Student's t test was used to compare the differences between two groups, and one-way ANOVA used to verify differences among more than two groups with Bonferroni or Dunnett post-hoc conducted, unless otherwise stated. P value of < 0.05 was considered significant.

Results

Activation of A2R facilitated myocardial function recovery after MI/RI in diabetic rats

To evaluate the effect of adenosine A2a receptor activation on myocardial reperfusion injury in diabetic rats, an in vivo diabetic rat model of MIRI was established, and NECA, a potent, non-selective A2R agonist was administered to activate A2R prior to reperfusion. Successful establishment of the model was confirmed by using an electrocardiogram (ECG), as indicated by the presence of ST segment elevation on the ECG following left anterior descending coronary artery (LAD) ligation of the rats. After 120 min of reperfusion, there was an obvious inverted Q wave and a prolonged QT interval in the ECG of the I/R group of rats. In contrast, QT interval prolongation was relatively improved in the I/R + NECA group (Fig. 1A). The changes in heart rate and mean arterial pressure during I/R were not statistically significant among the groups, (Fig. 1B, C). These results suggested that activation of adenosine A2a receptor prior to reperfusion, facilitated quick ventricular repolarization after ischemia of the myocardium.

Fig. 1
figure 1

Dynamic electrocardiogram and hemodynamic monitoring of the heart. AECG performance at baseline, ischemia, and reperfusion for each group. B Changes in heart rate at each time point expressed as (X ± SD); C Changes in mean arterial pressure in each group (X ± SD). D Echocardiographic manifestations of each group; E, F Left ventricular ejection fraction (LVEF) and left ventricular shortening fraction (FS) of rats in each group compared with control group (sham) and IR group (IR), ** P < 0.01

About five minutes to the end of reperfusion, cardiac ultrasound was conducted to observe changes in cardiac functions of the rats, (Fig. 1D). Echocardiography revealed that the left ventricular ejection function (LVEF) and shortening fraction (lvfs) in I/R group were significantly lower than those in the sham group (Fig. 1E, F; P < 0.05), indicating that I/R significantly damaged the systolic function of the heart. Compared with IR group, LVEF and lvfs in I/R + NECA group were significantly improved (Fig. 1E, F; P < 0.05). This indicates that the activation of adenosine A2a receptor significantly facilitated recovery of cardiac systolic function.

Activation of adenosine A2R attenuated MIRI in diabetic rats

To determine the effects of A2R activation on myocardial damage following ischemia, Serum levels of the myocardial enzymes creatinine kinase MB (CK-MB) and cardiac troponin (cTnI) were determined. CK-MB was significantly increased following ischemia, from 731.45 ± 49.61 in the sham group to 1329.30 ± 44.23 in the IR group. This was dramatically reduced to 656.91 ± 7.02 in I/R + NECA group. Similarly, cTnI increased following ischemia from 117.26 ± 27.53 in the sham group to 215.98 ± 42.97 in I/R group. This was also significantly reduced to 165.55 ± 30.36 in I/R + NECA group, demonstrating that A2R activation by NECA attenuated ischemic myocardial damage (Fig. 2A, B). Hematoxylin and eosin (H&E) staining and electron microcopy were then conducted on the myocardial tissues obtained before and after reperfusion. The results revealed that the myocardial damage seen in the I/R group was almost completely reversed after A2R activation prior to reperfusion in the I/R + NECA group (Fig. 2C–E). To determine the size of myocardial infarction area, Evans Blue staining was used. Compared to the IR group, the myocardial infarction area in the I/R + NECA group decreased by about 41% (Fig. 2F, G). The ischemic area of the two groups did not differ significantly signifying consistency in the LAD ligation sites of the models. Taken together, these result demonstrated that activation of adenosine A2a receptor using NECA prior to reperfusion attenuated myocardial ischemia reperfusion injury in the diabetic rats.

Fig. 2
figure 2

Adenosine A2 receptors activation attenuated MI/RI in the rats. A, B Plasma levels of cTnI and CK-MB in the different groups of rats. Data were presented as mean ± SD and analyzed by one-way ANOVA with uncorrected Fisher’s LSD post hoc test, ***P < 0.001, and ****P < 0.0001 respectively. C–E H&E staining and electron microscopy of the cardiac tissues. F, G Comparative percentage of myocardial ischemic area in each group. Infarct size was significantly reduced in the IR + NECA group versus IR group, * P < 0.05

Activation of A2R protected the myocardium through regulating endoplasmic reticulum stress-induced autophagy and apoptosis

To explore the roles of the endoplasmic reticulum stress (ERS) induced autophagy in MIRI in the diabetic rats in response to A2R activation, its agonist NECA was administered 1 h prior to reoxygenation. NECA administration in the I/R + NECA group significantly decreased the expression of the autophagy promoting protein, Beclin-1 compared to the I/R and the sham groups. On the other hand, its administration attenuated the decrease in the autophagosome substrate p62. (Fig. 3A–C). These results indicated that A2R activation had an anti-autophagy effect following ERS induction. ERS was determined by measuring the expression levels of ERS related genes GRP78, p-IRE1/IRE1, and p-JNK/JNK and their corresponding protein levels. Increase in GRP78, p-IRE1/IRE1, and p-JNK/JNK in the IR group of rats indicated increase in ERS induced apoptosis via the IRE-1/JNK pathway, which was significantly attenuated by A2R activation in the IR + NECA group (Fig. 3D–G). The above results demonstrated that A2R activation could have protected the myocardium during MIRI through regulating ERS induced autophagy.

Fig. 3
figure 3

Effects of A2R activation on autophagy and ERS. A–C Expression of autophagy proteins Beclin and p62. Beclin is decreased while p62 decline is attenuated by A2R activation in the IR + NECA group compared to sham group and the IR group, * P < 0.05, ** P < 0.01. D–G ERS proteins (GRP78, p-IRE-1/IRE1, p-JNK / JNK) are increased in IR group but administration of NECA significantly reduced their expression in the IR + NECA group, **P < 0.01

To explore the mechanism by which A2R activation influences the endoplasmic reticulum stress induced autophagy to confer cardio protection in MIRI, the ER resident protein IRE-1 was modulated. Previous studies have shown that IRE-1 regulates ER-stress induced autophagy in various cells [13,14,15]. The ER-stress inducer Tunicamycin (TM), and the IRE-1 inhibitor STF in combination with the A2R agonist NECA, were used, and the cellular responses measured using expression of the autophagy proteins Beclin-1, p62 and LC3. H&E staining of the I/R + NECA + TM group revealed broken muscle fibers, interrupted and discontinuous Z-line, and shifted & vacuolated mitochondria indicating induction of ER stress, (Fig. 4A–C). These were however all reversed following administration of the IRE-1 inhibitor in the I/R + NECA + TM + STF group; the myocardial fibers were continuous, Z-line was continuous, and mitochondrial edema and vacuolation were significantly reduced. Similar results were observed with the electron microscopy, indicating that inhibition of IRE-1 alleviated ER-stress, (Fig. 4D–F). In the western blot analysis, the administration of IRE-1 inhibitor prior to induction of ERS by TM, significantly reduced the expression levels of Beclin-1, prevented the conversion of LC3I to II and attenuated the reduction of p62, indicating attenuation of ERS-induced autophagy (Fig. 4G–J). Moreover, the protein expression levels of the IRE1 signaling pathway-related proteins GRP78, IRE1, and p-JNK were substantially raised during I/R injury yet significantly decreased following treatment with STF These results indicated that inhibition of IRE-1attenuated ERS-induced autophagy in the diabetic rats (Fig. 3D–G).

Fig. 4
figure 4

H&E staining, A In the Sham group, the myocardial fibers are complete; the Z-line is clear, continuous and straight, and the structural position is normal. B After ERS induction with TM, the heart muscle fibers were broken, Z-line was interrupted and discontinuous, and the mitochondria shifted and vacuolation increased. C After administration of IRE-1 inhibitor before reperfusion, the myocardial fibers were again continuous, Z-line was continuous, and mitochondrial edema and vacuolation were reduced. Electron microscopy: D In the Sham group, the myocardial fibers are complete; the mitochondria normal, and the cristae are clear. E After ERS induction using TM, muscle fibers were broken, Z-line was interrupted and discontinuous, mitochondria shifted and vacuolation increased; F After administration of IRE-1 inhibitor prior to reperfusion, the, myocardial fibers were continuous again, Z-line was continuous, and mitochondrial edema and vacuolation were reduced. GJ Administration of IRE-1 inhibitor significantly reduced the expression levels of Beclin-1, prevented the conversion of LC3I to II and attenuated the reduction of p62

A2R activation attenuated ERS induced apoptosis under hypoxia/reperfusion

To further assess the regulatory effects of adenosine A2 receptor agonists on endoplasmic reticulum stress induced autophagy and apoptosis in MIRI and the interaction between the two mechanisms, an in vitro ischemia reperfusion injury model in diabetic cardiomyocytes was developed. H9C2 cardiomyocytes cell line was used. Exposing the H9C2 cells to hypoxia induced significant cell death, which was alleviated by administration of the adenosine A2R agonist NECA (Fig. 5A). Apoptosis related proteins CHOP, and Caspase 12 were significantly increased in the H9C2 + high glucose + H/R group compared to Sham group, and sharply decreased following administration of the adenosine A2R agonist, NECA in the H9C2 + high glucose + H/R + NECA group (Fig. 5B–D). However, the total amount of JNK did not change (Fig. 5B, E), while its activated form that is known to induce apoptosis [16], significantly increased in the I/R group following reperfusion and was attenuated by A2R activation in the IR + NECA group (Fig. 3D, F). Similarly, tunnel staining indicated increased apoptosis in the H9C2 + high glucose + H/R group, which was alleviated in the H9C2 + Glu + high glucose + NECA group (Fig. 5F).

Fig. 5
figure 5

Effect of A2R activation on autophagy. A Cell viability was detected by CCK-8. BE Western blot analysis of apoptosis related proteins Caspase-12, CHOP, and JNK analyzed by Image J. All data presented as mean ± SEM and analyzed by one-way ANOVA and Dunnett’s post hoc test; n = 3. **P < 0.01, ***P < 0.001 and *P < 0.05 respectively. F TUNEL staining demonstrating apoptosis

Cardio-protection effect of A2R activation in MIRI is by regulating endoplasmic reticulum stress induced autophagy through IRE-1 protein

To investigate the protective role of A2R in H/R, its effect on endoplasmic reticulum stress, and autophagy was evaluated. NECA was used to activate A2R while Tunicamycin was used to induce endoplasmic reticulum stress (ERS) and the levels of ERS related proteins GRP78, p-ire1and XBP1s determined. In addition, p-IRE-1 inhibitor STF was used to evaluate the possible involvement of p-IRE-1 protein in regulating autophagy in response to ERS. As expected, the levels of ERS proteins GRP78, p-IRE1and XBP1 were all increased in response to Tunicamycin treatment (Fig. 6A–D), while the levels of autophagy related proteins Beclin-1 and LC3 were increased and the autophagosome substrate protein, p62 decreased indicating increased autophagy (Fig. 6E–H). Similarly, increased co-localization of LC3 and LAMP-2 was observed in the ERS induced group using immunofluorescence staining (Fig. 6I) indicating that ERS induction promoted autophagosomes and lysosomes fusion. These were however all reversed following treatment with p-IRE-1 inhibitor STF, indicating that p-IRE-1 could be involved in the regulation of ERS induced autophagy.

Fig. 6
figure 6

Effect of A2R activation on autophagy and endoplasmic reticulum stress. AD Expression levels ERS proteins GRP78, p-IRE-1 and XBP1s evaluated by western blot and analyzed by image J. *P < 0.05, **P < 0.01, ***P < 0.001. EH Expression levels of autophagy proteins, p62, Beclin-1, LC3I and LC3 II evaluated by western blot and analyzed by image J. ***P < 0.001, ****P < 0.0001. All data presented as mean ± SEM, analyzed by one way ANOVA and Tukey’s post hoc test. I LC3-LAMP-2 co-localization viewed by fluorescence microscopy

Discussion

MIRI remains a major cause of death of cardiomyocytes in patients with coronary heart disease [17]. Mechanisms responsible for cardiomyocytes death during MIRI include; autophagy, apoptosis, ferroptosis and pyroptosis [18]. Autophagy is a highly conserved cellular process where nutrients are catabolized and recycled through lysosomal degradation of unfolded or misfolded proteins and worn out cellular organelles [11]. It releases amino acids, fatty acids, ATP, and other high energy molecules that are essential for cell survival and normal tissue function during ischemia or nutrient deficiency [11]. This is particularly important for non-proliferative cells such as cardiomyocytes as it ensures cell survival and maintains structural and functional stability of the heart. The endoplasmic reticulum (ER) is one of the major cellular organelles that regulates autophagy [19]. Under extreme external stress, protein folding in the ER is disrupted and so misfolded and unfolded proteins accumulate in the ER lumen and the cytoplasm. These proteins interact with p62, which then recruits LC3II [20, 21], and together form autophagosomes. In the presence of LAMP2, these autophagosomes fuse with lysosomes to form autolysosomes leading to lysis and recycling of the contents [22, 23], and eventual survival of the cell. This process is called endoplasmic reticulum stress (ERS) induced autophagy. However, autophagy is a double edged sword; enhanced autophagy following MIRI can lead to significant reduction of viable cardiomyocytes which impairs recovery of ventricular functions and exacerbate the condition [24,25,26,27].

Given the upregulation of CHOP and caspase 12 in our experiment, we investigated ERS involvement in the resultant autophagy and apoptosis using the ERS inducer Tunicamycin. Induction of ERS resulted into increased expression of BiP (GRP78), p-IRE1 and XBP1s. The response of CHOP to ERS is often regulated by three factors; protein kinase RNA-like endoplasmic reticulum kinase (PERK), activating transcription factor 6 (ATF6), and inositol requiring protein 1 (IRE1) [28]. The dramatic increase in the expression of IRE1 here suggested that it was the likely pathway in this experiment. IRE1 is kept in its inactive state by GRP78 interaction. Upon accumulation of unfolded proteins, GRP78, having more affinity for unfolded proteins, disintegrates from IRE1 thus activating it [29]. In its active form, IRE1 cleaves its substrate precursor XBP1 to its active form XBP1s [30, 31]. XBP1s then upregulates the expression of CHOP and many other genes involved in UPR to restore protein homeostasis [32,33,34]. However, when ERS is prolonged or severe, IRE1 stimulates the activation of the apoptotic-signaling kinase-1 (ASK1), which in turn activates JNK and p38 MAK hence inducing apoptosis [35]. To confirm the involvement of IRE1, the cardiomyocytes were treated with the IRE1 inhibitor STF. This significantly inhibited the autophagy promoter Beclin-1 and resulted into accumulation of p62 indicating reduced or inhibited autophagy.

In this experiment, subjecting the cardiomyocytes to hypoxia and reoxygenation (H/R) induced significant apoptosis as demonstrated by the CCK-8 assay and the dramatic increase in apoptosis related proteins CHOP and caspase-12. CHOP and caspase-12 are particularly associated with endoplasmic reticulum stress (ERS) induced apoptosis [36, 37]. Since JNK levels remained constant, this apoptosis was most likely independent of the JNK pathway. ERS-induced apoptosis follows three major pathway; the IRE1/ASK1/JNK pathway, the caspase-12 kinase pathway, and the C/EBP homologous protein (CHOP)/GADD153 pathway [28, 30], so it was likely that the later pathway was involved. Activation of A2R using its agonist NECA swiftly reversed apoptosis. This suggested that NECA could be a very useful therapeutic for early stage MIRI management in diabetics. The finding are consistent with our previous results [9, 38] which demonstrated that A2aR activation protects the myocardium against MIRI through downregulating autophagy and regulating autophagy flux and apoptosis in non-diabetic cardiomyocytes. In our most recent work, [10], we demonstrated that NECA attenuated MIRI in type-2 diabetic rats through A2aR/PCK/miR-15a mechanism. However, we speculated that the autophagic flux seen by Xia et al. [38] in nondiabetic rats could also occur in type-2 diabetic rats, and decided to use NECA, a non-selective activator of adenosine receptors to demonstrate ERS involvement in this cardio protection. This results has thus demonstrated that indeed A2R activation in type-2 diabetic rats by NECA could also be cardioprotective via ERS inhibition mechanisms.

In the animal experiment, myocardial ischemia (MI) resulted into leakage of cTnI and CK-MB into the blood stream and infarction of the myocardium. These myocardial damages were promptly reversed by activation of A2R by NECA. Similarly, the ventricular systolic function that was impaired by MI also resolved following A2R activation. MI also impaired the LVEF and LVFS indicating impaired ventricular systolic function. However, A2R activation ameliorated both conditions, in effect restoring left ventricular systolic function. Studies have shown that recovery of ventricular systolic function following MIRI is dependent on A2R inducing the elevation of cAMP and PKA, which then triggers multiple cAMP-PKA dependent calcium ion channels, heightening the maximum peak transient outward current thus enabling post ischemia repolarization of the ventricles [39,40,41]. Therefore, cAMP mediated influx of Ca2 + promotes reestablishment of myocardial contractility.

On the electrocardiogram, MI elevated the ST segment and caused prolongation of the QT interval. This was possibly caused by the imbalance in the in and out flows of intracellular and extracellular ion (Ca2 + , K + , H + , and Na +), due to the interrupted blood and oxygen supply following ischemia, hence changing the repolarization current (42). This current change in the ischemic area thus prolonged the QT wave and elevated the ST-segment. Activation of A2R during ischemic post conditioning has an antiarrhythmic effect associated with action potential shortening [44, 45].

This study had the following limitations: (1) we have previously demonstrated that A2aR activation protects the myocardium against MIRI through regulating autophagy flux and apoptosis in non-diabetic cardiomyocytes, and that NECA attenuates MIRI in type-2 diabetic rats through A2aR/PCK/miR-15a mechanism [10, 38]. Here, to investigate ERS involvement in MIRI in diabetic rats we used NECA, a nonselective A2R activator. Our result here is therefore, not specific to which adenosine receptor subtype is responsible for this ERS involved cardio protection in type-2 diabetic rats. In a future study, this will be resolved. (2) According to the IMproving Preclinical Assessment of Cardioprotective Therapies (IMPACT) criteria by the European Union-CARDIOPROTECTION COST Action [46], in small animal models of acute ischemia reperfusion injury (IRI), it is desirable to demonstrate benefit of the intervention after at least 28 days post intervention. However, this was not the case in this study, therefore these results should be interpreted accordingly.

Conclusions

In summary, activation of A2R prior to reperfusion following myocardial ischemia effectively attenuated apoptosis, reduced ERS induced autophagy and re-established impaired left ventricular function, hence reducing the damaging effects of MIRI on diabetic cardiomyocytes. This protective effect occurs through regulating the IRE1-XBPs-CHOP related mechanisms.

Availability of data and materials

All the supporting data are available with the corresponding author and shall be made available upon request.

References

  1. Li H, Yao W, Liu Z, Xu A, Huang Y, Ma XL, et al. Hyperglycemia abrogates ischemic postconditioning cardioprotection by impairing AdipoR1/Caveolin-3/STAT3 signaling in diabetic rats. Diabetes. 2016;65(4):942–55.

    Article  CAS  PubMed  Google Scholar 

  2. Bouwman RA, Musters RJP, van Beek-Harmsen BJ, de Lange JJ, Lamberts RR, Loer SA, et al. Sevoflurane-induced cardioprotection depends on PKC-alpha activation via production of reactive oxygen species. Br J Anaesth. 2007;99(5):639–45.

    Article  CAS  PubMed  Google Scholar 

  3. Buddeke J, Bots ML, van Dis I, Visseren FL, Hollander M, Schellevis FG, et al. Comorbidity in patients with cardiovascular disease in primary care: a cohort study with routine healthcare data. Br J Gen Pract. 2019;69(683):e398-406.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Das A, Salloum FN, Filippone SM, Durrant DE, Rokosh G, Bolli R, et al. Inhibition of mammalian target of rapamycin protects against reperfusion injury in diabetic heart through STAT3 signaling. Basic Res Cardiol. 2015;110(3):31.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Ma S, Wang Y, Chen Y, Cao F. The role of the autophagy in myocardial ischemia/reperfusion injury. Biochim Biophys Acta. 2015;1852(2):271–6.

    Article  CAS  PubMed  Google Scholar 

  6. Bhattacharya D, Mukhopadhyay M, Bhattacharyya M, Karmakar P. Is autophagy associated with diabetes mellitus and its complications? A review. EXCLI J [Internet]. 2018 Jul 24 [cited 2022 May 16];17:709–20. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6123605/

  7. Cohen MV, Downey JM. Adenosine: trigger and mediator of cardioprotection. Basic Res Cardiol. 2008;103(3):203–15.

    Article  CAS  PubMed  Google Scholar 

  8. McIntosh VJ, Lasley RD. Adenosine receptor-mediated cardioprotection: are all 4 subtypes required or redundant? J Cardiovasc Pharmacol Ther. 2012;17(1):21–33.

    Article  CAS  PubMed  Google Scholar 

  9. Ke J, Yao B, Li T, Cui S, Ding H. A2 adenosine receptor-mediated cardioprotection against reperfusion injury in rat hearts is associated with autophagy downregulation. J Cardiovasc Pharmacol [Internet]. 2015 Jul [cited 2022 Mar 20];66(1). Available from: https://pubmed.ncbi.nlm.nih.gov/25706370/

  10. Chao C, JianJuan K, Huang D, Chengjun H, Wang Y. 5′-N-Ethylcarboxamido adenosine attenuates myocardial ischemia/reperfusion injury in type 2 diabetic rats through A2AR/PKCα/miR-15a signaling. 2021 Sep 15.

  11. Gatica D, Chiong M, Lavandero S, Klionsky DJ. Molecular mechanisms of autophagy in the cardiovascular system. Circ Res [Internet]. 2015 Jan 30 [cited 2022 Mar 21];116(3). Available from: https://pubmed.ncbi.nlm.nih.gov/25634969/

  12. Salabei JK, Conklin DJ. Cardiovascular autophagy: crossroads of pathology, pharmacology and toxicology. Cardiovasc Toxicol. 2013;13(3):220–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Lee H, Noh JY, Oh Y, Kim Y, Chang JW, Chung CW, et al. IRE1 plays an essential role in ER stress-mediated aggregation of mutant huntingtin via the inhibition of autophagy flux. Hum Mol Genet. 2012;21(1):101–14.

    Article  PubMed  Google Scholar 

  14. Li Z, Huang Z, Zhang H, Lu J, Wei Y, Yang Y, et al. IRE1-mTOR-PERK Axis Coordinates Autophagy and ER Stress-Apoptosis Induced by P2X7-Mediated Ca2+ Influx in Osteoarthritis. Frontiers in Cell and Developmental Biology [Internet]. 2021 [cited 2022 Apr 28];9. Available from: https://www.frontiersin.org/article/https://doi.org/10.3389/fcell.2021.695041

  15. Sozen E, Yazgan B, Tok OE, Demirel T, Ercan F, Proto JD, et al. Cholesterol induced autophagy via IRE1/JNK pathway promotes autophagic cell death in heart tissue. Metabolism [Internet]. 2020 May 1 [cited 2022 Apr 28];106:154205. Available from: https://www.sciencedirect.com/science/article/pii/S002604952030069X

  16. Dhanasekaran DN, Reddy EP. JNK Signaling in Apoptosis. Oncogene [Internet]. 2008 Oct 20 [cited 2023 Jan 17];27(48):6245–51. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3063296/

  17. Patel P, Karch J. Regulation of cell death in the cardiovascular system. Int Rev Cell Mol Biol. 2020;353:153–209.

    Article  CAS  PubMed  Google Scholar 

  18. Del Re DP, Amgalan D, Linkermann A, Liu Q, Kitsis RN. fundamental mechanisms of regulated cell death and implications for heart disease. Physiol Rev [Internet]. 2019 Oct 1 [cited 2022 Aug 3];99(4). Available from: https://pubmed.ncbi.nlm.nih.gov/31364924/

  19. Qi Z, Chen L. Endoplasmic reticulum stress and autophagy. In: Qin ZH, editor. Autophagy: biology and diseases: basic science [Internet]. Singapore: Springer; 2019 [cited 2022 Mar 4]. p. 167–77. (Advances in experimental medicine and biology). Available from: https://doi.org/10.1007/978-981-15-0602-4_8

  20. Cherra SJ, Kulich SM, Uechi G, Balasubramani M, Mountzouris J, Day BW, et al. Regulation of the autophagy protein LC3 by phosphorylation. J Cell Biol. 2010;190(4):533–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen H, et al. p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem. 2007;282(33):24131–45.

    Article  CAS  PubMed  Google Scholar 

  22. Hariharan N, Zhai P, Sadoshima J. Oxidative stress stimulates autophagic flux during ischemia/reperfusion. Antioxid Redox Signal. 2011;14(11):2179–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Ma X, Liu H, Foyil SR, Godar RJ, Weinheimer CJ, Hill JA, et al. Impaired autophagosome clearance contributes to cardiomyocyte death in ischemia/reperfusion injury. Circulation. 2012;125(25):3170–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Gurusamy N, Das DK. Is autophagy a double-edged sword for the heart? Acta Physiol Hung. 2009;96(3):267–76.

    Article  CAS  PubMed  Google Scholar 

  25. Mokhtari B, Badalzadeh R. The potentials of distinct functions of autophagy to be targeted for attenuation of myocardial ischemia/reperfusion injury in preclinical studies: an up-to-date review. J Physiol Biochem [Internet]. 2021 Aug 1 [cited 2022 Mar 3];77(3):377–404. Available from: https://doi.org/10.1007/s13105-021-00824-x

  26. Mokhtari B, Badalzadeh R. Protective and deleterious effects of autophagy in the setting of myocardial ischemia/reperfusion injury: an overview. Mol Biol Rep. 2022;49(11):11081–99.

    Article  CAS  PubMed  Google Scholar 

  27. Schiattarella GG, Hill JA. Therapeutic targeting of autophagy in cardiovascular disease. J Mol Cell Cardiol. 2016;95:86–93.

    Article  CAS  PubMed  Google Scholar 

  28. Oyadomari S, Mori M. Roles of CHOP/GADD153 in endoplasmic reticulum stress. Cell Death Differ [Internet]. 2004 Apr [cited 2022 Aug 4];11(4):381–9. Available from: https://www.nature.com/articles/4401373

  29. Amin-Wetzel N, Saunders RA, Kamphuis MJ, Rato C, Preissler S, Harding HP, et al. A J-protein co-chaperone recruits BiP to monomerize IRE1 and repress the unfolded protein response. Cell. 2017;171(7):1625-1637.e13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Hetz C. The unfolded protein response: controlling cell fate decisions under ER stress and beyond. Nat Rev Mol Cell Biol. 2012;13(2):89–102.

    Article  CAS  PubMed  Google Scholar 

  31. Yoshida H, Matsui T, Yamamoto A, Okada T, Mori K. XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell [Internet]. 2001 Dec 28 [cited 2022 Aug 4];107(7):881–91. Available from: https://www.cell.com/cell/abstract/S0092-8674(01)00611-0

  32. Kim I, Xu W, Reed JC. Cell death and endoplasmic reticulum stress: disease relevance and therapeutic opportunities. Nat Rev Drug Discov [Internet]. 2008 Dec [cited 2022 Aug 4];7(12):1013–30. Available from: https://www.nature.com/articles/nrd2755

  33. Yang Y, Liu L, Naik I, Braunstein Z, Zhong J, Ren B. Transcription factor C/EBP homologous protein in health and diseases. Front Immunol [Internet]. 2017 [cited 2022 Aug 4];8. Available from: https://www.frontiersin.org/articles/https://doi.org/10.3389/fimmu.2017.01612

  34. Madhusudhan T, Wang H, Dong W, Ghosh S, Bock F, Thangapandi VR, et al. Defective podocyte insulin signalling through p85-XBP1 promotes ATF6-dependent maladaptive ER-stress response in diabetic nephropathy. Nat Commun [Internet]. 2015 Mar 10 [cited 2022 Aug 4];6(1):6496. Available from: https://www.nature.com/articles/ncomms7496

  35. Ron D, Hubbard SR. How IRE1 Reacts to ER Stress. Cell [Internet]. 2008 Jan 11 [cited 2022 Aug 4];132(1):24–6. Available from: https://www.cell.com/cell/abstract/S0092-8674(07)01672-8

  36. Hu H, Tian M, Ding C, Yu S. The C/EBP homologous protein (CHOP) transcription factor functions in endoplasmic reticulum stress-induced apoptosis and microbial infection. Front Immunolo [Internet]. 2019 [cited 2022 Aug 4];9. Available from: https://www.frontiersin.org/articles/https://doi.org/10.3389/fimmu.2018.03083

  37. Szegezdi E, Fitzgerald U, Samali A. Caspase-12 and ER-stress-mediated apoptosis: the story so far. Ann N Y Acad Sci. 2003;1010:186–94.

    Article  CAS  PubMed  Google Scholar 

  38. Xia Y, He F, Moukeila Yacouba MB, Zhou H, Li J, Xiong Y, et al. Adenosine A2a receptor regulates autophagy flux and apoptosis to alleviate ischemia-reperfusion injury via the cAMP/PKA signaling pathway. Front Cardiovasc Med. 2022;9:755619.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Hou Y, Wang J, Feng J. The neuroprotective effects of curcumin are associated with the regulation of the reciprocal function between autophagy and HIF-1α in cerebral ischemia-reperfusion injury. Drug Des Devel Ther [Internet]. 2019 Apr 9 [cited 2022 Aug 4];13:1135–44. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6461000/

  40. Batchu SN, Law E, Brocks DR, Falck JR, Seubert JM. Epoxyeicosatrienoic acid prevents postischemic electrocardiogram abnormalities in an isolated heart model. J Mol Cell Cardiol. 2009;46(1):67–74.

    Article  CAS  PubMed  Google Scholar 

  41. Sanada S, Asanuma H, Tsukamoto O, Minamino T, Node K, Takashima S, et al. Protein Kinase A as another mediator of ischemic preconditioning independent of protein Kinase C. Circulation [Internet]. 2004 Jul 6 [cited 2022 Aug 4];110(1):51–7. Available from: https://www.ahajournals.org/doi/https://doi.org/10.1161/01.cir.0000133390.12306.c7

  42. Baumert M, Porta A, Vos MA, Malik M, Couderc JP, Laguna P, et al. QT interval variability in body surface ECG: measurement, physiological basis, and clinical value: position statement and consensus guidance endorsed by the European Heart Rhythm Association jointly with the ESC Working Group on Cardiac Cellular Electrophysiology. Europace. 2016;18(6):925–44.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Liang BT, Morley JF. A New Cyclic AMP-independent, Gs-mediated Stimulatory Mechanism via the Adenosine A2a Receptor in the Intact Cardiac Cell *. Journal of Biological Chemistry [Internet]. 1996 Aug 2 [cited 2022 Aug 4];271(31):18678–85. Available from: https://www.jbc.org/article/S0021-9258(18)32034-9/abstract

  44. Diez ER, Sánchez JA, Prado NJ, Ponce Zumino AZ, García-Dorado D, Miatello RM, et al. Ischemic Postconditioning Reduces Reperfusion Arrhythmias by Adenosine Receptors and Protein Kinase C Activation but Is Independent of KATP Channels or Connexin 43. Int J Mol Sci [Internet]. 2019 Nov 25 [cited 2022 Mar 20];20(23):5927. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6928819/

  45. Ma H, Guo R, Yu L, Zhang Y, Ren J. Aldehyde dehydrogenase 2 (ALDH2) rescues myocardial ischaemia/reperfusion injury: role of autophagy paradox and toxic aldehyde. Eur Heart J. 2011;32(8):1025–38.

    Article  CAS  PubMed  Google Scholar 

  46. Lecour S, Andreadou I, Bøtker HE, Davidson SM, Heusch G, Ruiz-Meana M, et al. IMproving Preclinical Assessment of Cardioprotective Therapies (IMPACT) criteria: guidelines of the EU-CARDIOPROTECTION COST action. Basic Res Cardiol [Internet]. 2021 Sep 13 [cited 2023 Jan 17];116(1):52. Available from: https://doi.org/10.1007/s00395-021-00893-5

Download references

Acknowledgements

Not applicable.

Funding

This research was funded by the National Natural Science Foundation of China (81871553).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, MBMY, JK, ZZ and YW; methodology, MBMY, JK, ET, CCB, FH, YX and FW. software, ET and CCB.; validation, ET and CCB.; formal analysis, MBMY, ET and CCB; investigation, MBMY, ET, CCB, FH,YX and FW.; resources, JK, ZZ, YW.; data curation, CCB.; writing—original draft preparation, MBMY.; writing—review and editing, ET, CCB, AFG, HM, SK, BO,HAO,DD, JK, ZZ and YW; visualization, MBMY, ET; supervision, JK, ZZ, YW; project administration, JK and ZZ; funding acquisition, ZZ and YW. All authors have read and agreed to the published version of the manuscript.

Corresponding authors

Correspondence to Jianjuan Ke, Zongze Zhang or Yanlin Wang.

Ethics declarations

Ethics approval and consent to participate

The animal study protocol was approved by the Institutional Review Board of Wuhan University China, (Approval No. WP2020–01108).

Consent for publication

Not applicable.

Competing interests

All authors declare no competing interest.

Additional information

Publisher's Note

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

Supplementary Information

Additional file 1

. Table S1. Information on all the primers used for qPCR in the study.

Additional file 2

. Figure F1. Original blots for the effects of A2R activation on autophagy and ERS. The autophagy proteins Beclin 1 and P62 were assessed. The 60Kda protein is Beclin 1, 62Kda protein is P62 and the 36Kda protein is GAPDH; the internal control protein.

Additional file 3

. Figure F2. Original blots for the effects of A2R activation on autophagy and ERS. The ERS proteins Grp78, P-JKN, and P-IRE1 were assessed. The 78Kda protein is Grp78, 54Kda and 46Kda proteins are isoforms of P-JKN, and the 110Kda protein is P-IRE1. The 36Kda protein is GAPDH; the internal control protein.

Additional file 4

. Figure F3. Original blots of autophagy proteins measured in response to P-IRE1 inhibition. The 16Kda protein is LC3, the 62Kda protein is P62 and the 60Kda protein is Beclin 1. The 36Kda protein is GAPDH as the internal control protein.

Additional file 5

. Figure F4. Original blots of apoptosis related proteins in H9C2 cells following hypoxia. The proteins assessed were CHOP, JKN, and Caspase 12. The 27Kda protein is CHOP, the 54Kda and 46Kda proteins are isoforms of JKN and the 46Kda protein is Caspase 12. The 43Kda protein is Beta-actin; the internal control protein.

Additional file 6

. Figure F5. Original blots of autophagy and ERS proteins in H9C2 cells following activation of A2R. The autophagy proteins assessed were Beclin 1, p62, and LC3, while the ERS proteins were Grp78, P-IRE1 and XBP1s. Beta actin was the internal control protein.

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/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yacouba Moukeila, M.B., Thokerunga, E., He, F. et al. Adenosine 2 receptor regulates autophagy and apoptosis to alleviate ischemia reperfusion injury in type 2 diabetes via IRE-1 signaling. BMC Cardiovasc Disord 23, 154 (2023). https://doi.org/10.1186/s12872-023-03116-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12872-023-03116-y

Keywords