Astragalin and dihydromyricetin as adjuncts to histidine‑tryptophan‑ketoglutarate cardioplegia enhances protection during cardioplegic arrest

  • Authors:
    • Dong Wang
    • Xinjie Zhang
    • Daoxu Qu
    • Jichun Han
    • Fanqing Meng
    • Minglei Xu
    • Qiusheng Zheng
  • View Affiliations

  • Published online on: July 5, 2018     https://doi.org/10.3892/mmr.2018.9254
  • Pages: 2929-2936
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study used an in vitro model of cold cardioplegia in isolated working rat hearts to evaluate the possible effects of two flavonoids, astragalin and dihydromyricetin, as adjuncts to histidine‑tryptophan‑ketoglutarate (HTK) cardioplegia. The following three groups of male Sprague Dawley rats were evaluated: The HTK group, treated with HTK alone; the HTK‑A group, treated with 10 µmol/l astragalin; and the HTK‑D group, treated with 10 µmol/l dihydromyricetin. Isolated rat hearts were perfused with Krebs‑Henseleit buffer for 30 min and incubated with the respective cardioplegic solution for 6 h at 4˚C. Subsequently, astragalin or dihydromyricetin was added to the cardioplegic solutions. Following 30 min of reperfusion, the left ventricular developed pressure (LVDP), maximum up/down rate of left ventricular pressure (±dp/dtmax) and heart rate were documented as indices of myocardial function using a physiological recorder. Myocardial infarct size (IS) was estimated using 2,3,5‑triphenyltetrazolium chloride staining. Lactate dehydrogenase (LDH) and creatine kinase (CK) levels were also determined to assess the degree of cardiac injury. Cardiomyocyte apoptosis analysis was performed using an in situ cell death detection kit. In addition, malondialdehyde (MDA), superoxide dismutase (SOD), interleukin‑6 (IL‑6), tumor necrosis factor‑α (TNF‑α), C‑reactive protein (CRP) levels, as well as the glutathione/glutathione disulfide (GSH/GSSG) ratio were determined and analyzed using ELISA kits. The protein levels of caspase‑9 and B‑cell lymphoma‑2 (Bcl‑2) were determined using western blot analysis. The results demonstrated that exposure to astragalin or dihydromyricetin significantly improved the recovery of LVDP (P<0.05 and P<0.01, respectively), the +dP/dtmax (P<0.05 for dihydromyricetin only) and the ‑dP/dtmax (P<0.05 and P<0.01, respectively), increased SOD levels (P<0.05 and P<0.01, respectively) and GSH/GSSG ratios (P<0.05), reduced myocardial IS (P<0.05 and P<0.01, respectively), decreased CK, LDH, IL‑6 (all P<0.05 and P<0.01, respectively), MDA (P<0.05), CRP (P<0.05) and TNF‑α levels (P<0.05 and P<0.01, respectively), increased Bcl‑2 levels (P<0.01) and decreased caspase‑9 levels (P<0.01). The results indicated that the addition of either flavonoid (particularly dihydromyricetin) to HTK enhances protection during ischemia, decreases myocardial dysfunction by enhancing anti‑inflammatory activities, attenuates myocardial oxidative injury and prevents apoptosis during ischemia/reperfusion.

Introduction

At present, heart transplantation is considered the critical standard for end-stage heart failure treatment (1). Following retrieval, the donor heart is maintained in cold (4°C) cardioplegic solution, and then introduced to the recipient (2). Therefore, a period of cold ischemia and ischemia/reperfusion (I/R) are inevitable due to tissue matching and transportation. However, inadequate myocardial protection associated with prolonged cold ischemia and I/R injury will result in postoperative myocardial dysfunction (2,3). Therefore, minimization of ischemia and I/R injury during transport is essential to reduce the risk of primary graft failure and improve short- and long-term outcomes (4,5).

Histidine-tryptophan-ketoglutarate (HTK) solution is clinically used for effective organ preservation (6). A previous study revealed that the HTK solution is a valid alternative solution for organ preservation due its superior role in compensating for cellular acidosis and prolonging anaerobic glycolysis (7). However, studies have demonstrated that reperfusion following resuscitation from cardiac arrest increases reactive oxygen species (ROS) production, leading to oxidative stress (8,9). Oxidative stress is a factor involved in epithelial apoptosis (10). Previous research has also demonstrated that the inflammatory response may be induced during myocardial I/R, and that the cold ischemic donor organ is a major source of pro-inflammatory mediators (11,12). However, it is known that the HTK solution is unable to effectively reduce hypothermia-induced oxidative stress (13,14).

Astragalin (kaempferol-3-O-glucoside, a flavonoid that is extracted from leaves of persimmon, rosa agrestis, or green tea seeds) and dihydromyricetin (a flavonoid that is extracted from ampelopsis grossedentata) flavonoids, present a wide range of pharmacological activities, including anti-oxidative (15,16) and anti-inflammatory effects (10,17), and they have been demonstrated to ameliorate apoptosis (18,19). In addition, astragalin and dihydromyricetin may protect cells from hydrogen peroxide-induced oxidative stress damage (16,20). Furthermore, supplementation with dihydromyricetin improves glucose and lipid metabolism, and decreases the serum level of tumor necrosis factor-α (TNF-α) in nonalcoholic fatty liver disease patients (17). Previous studies have revealed that astragalin or dihydromyricetin may protect endothelial cells from oxidative stress damage via anti-oxidative, anti-inflammatory and anti-apoptotic signaling pathways in vitro (10,16). However, whether administration of astragalin or dihydromyricetin as an additive to the HTK solution during the hypothermic storage stage, exerts a significant protective effect on isolated cardiac grafts subject to cold ischemia, remains unclear.

Based on the pharmacological potential of astragalin and dihydromyricetin, the authors of the present study hypothesized that the addition of astragalin or dihydromyricetin to HTK may reduce cold ischemia and I/R induced injuries, thereby improving cardioprotective potential. To investigate this hypothesis, an isolated perfused heart model was generated in the present study, whereby rat hearts were perfused in the working state following preservation in the respective cardioplegic solution for 6 h at 4°C. To the best of our knowledge, the present study is the first to investigate the cardioprotective effects of cardioplegic solution containing astragalin or dihydromyricetin. In addition, the current study compared the effects of astragalin and dihydromyricetin on myocardial preservation.

Materials and methods

Chemicals

Astragalin and dihydromyricetin (purity, ≥98%) were purchased from Chengdu Must Bio-Technology Co., Ltd. (Sichuan, China). Astragalin and dihydromyricetin were dissolved in dimethyl sulfoxide (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) prior to use. The dimethyl sulfoxide concentration in the working solutions was <0.1%, which was found to exert no effects on the rats/cardiomyocytes in preliminary experiments. HTK and was obtained from Beijing Witton Economic and Trade Co., Ltd. (Beijing, China).

Animals

A total of 24 adult male Sprague Dawley rats (weight, 260±22 g; age, 2–3 months) were obtained from Jinan Pengyue Experimental Animal Breeding Co., Ltd. [license number, SCXK (Lu) 2014–0007; Jinan, China] and housed in a specific environment (temperature, 22–25°C; relative humidity, 50–60%; 12-h light/dark cycle; and free access to standard rodent food and water). All experimental protocols were performed in accordance with the regulations of the Guide for the Care and Use of Laboratory Animals and approved by the Ethics Committee of Animal Laboratory and Experimental Management Center of Shandong University [Shandong, China; license number SYXK (Lu) 20130001, revised 2013].

Study groups

Astragalin and dihydromyricetin doses used in the present study were determined by preliminary experiments; 5, 10 and 20 µmol/l of these agents were selected for preliminary analysis. Measurement of heart hemodynamic parameters revealed that 10 µmol/l of each agent significantly improved functional recovery during early reperfusion (data not shown). This is consistent with the results of some studies indicating that this moderate dose may demonstrate protective effects (21,22). Therefore, 10 µmol/l was selected for the experiments performed in the current study.

The hearts from rats subjected to 6 h ischemia followed by 30 min perfusion were divided into the following three groups according to the cardioplegic solution used: The HTK group, treated with HTK alone; the HTK-A group, treated with HTK containing 10 µmol/l astragalin; and the HTK-D group, treated with HTK containing 10 µmol/l dihydromyricetin.

Model of the isolated and perfused working rat heart

Ischemia was performed according to procedures described previously (5). Rats were anesthetized with 10% chloral hydrate (300 mg/kg body weight) through intraperitoneal injection, and then administered with 250 U/kg heparin (Solebo Biotechnology Co., Ltd., Beijing, China) through sublingual venous injection to prevent coagulation. The chest was then opened using a bilateral sternocostal triangle. The hearts were immediately excised and placed into 40 ml of respective cardioplegic solution. The hearts were carefully washed for 10 sec to remove blood from the coronaries and to avoid the formation of blood clots. Subsequently, the hearts were placed in a conical tube with 10 ml ice-cold cardioplegic solution at 4°C for 6 h. A Langendorff perfusion system (Powerlab, Australia; www.adinstruments.com/) was then used to perform heart perfusion. Hearts were perfused via their aortas at a constant pressure of 75 mmHg using Krebs-Henseleit buffer (120 mM NaCl, 1.2 mM KH2PO4, 1.2 mM CaCl2, 1.2 mM MgSO4, 25 mM sodium acetate and 11 mM glucose, pH 7.4. All chemicals and reagents were purchased from Solebo Biotechnology Co., Ltd.) at 37°C for 30 min. A water-filled latex balloon connected to a pressure transducer was inserted into the left ventricle through an incision in the left atrium and through the mitral valve, and the volume was adjusted to achieve a stable end-diastolic pressure (8–12 mmHg). A number of cardiac parameters, including heart rate (HR), left ventricular developed pressure (LVDP) and maximum up/down rate of left ventricular pressure (±dp/dtmax), were continuously monitored and recorded using the 4S AD Instruments biology polygraph data acquisition system and Chart5 software (both Powerlab). Following 30 min reperfusion, the hearts were stored at −80°C until downstream analysis.

Measurement of cellular injury

The myocardium contains an abundance of diagnostic marker enzymes that can be used to detect myocardial infarction. Following metabolic damage to the myocardium, intracellular contents are released into the extracellular fluid (23). Therefore, the levels of these marker enzymes reflect alterations in membrane integrity and/or permeability. In the present study lactate dehydrogenase (LDH) and creatine kinase (CK) levels were measured to evaluate the degree of cardiac injury. LDH ELISA kits (Nanjing Jiancheng Biological Products, Co., Ltd., Nanjing, China. Cat. no. A02020150421) and CK ELISA kits (Nanjing Jiancheng Biological Products, Co., Ltd., Nanjing, China. Cat. no. A03220150501) were used to measure LDH and CK levels according to the manufacturer's protocol. Samples were collected from the respective cardioplegic solutions prior to reperfusion and from the coronary effluent following 30 min of reperfusion.

Evaluation of myocardial infarct size (IS)

Myocardial IS was evaluated using triphenyltetrazolium chloride (TTC) staining, as previously described (24). At the end of the experiments, the hearts were removed from the cardioplegic solutions, washed in phosphate-buffered saline and stored at −20°C for 30 min. Subsequently, the hearts were sliced perpendicularly into 1 mm-thick sections along the long axis from apex to base and incubated in 1% TTC at 37°C for 15 min. Slices were imaged using a digital camera following fixation in 10% formaldehyde solution at 25°C for 24 h. Image-Pro Plus 7.0 software (Media Cybernetics, Inc., Rockville, MD, USA) was used to measure the IS area. Red regions indicated non-ischemic areas, whereas white regions indicated ischemic areas. The IS (%) was calculated using the following equation: Infarct volume (%)=(infarct volume/total volume of slice) ×100.

Analysis of oxidative stress in heart tissue homogenates

Following 30 min perfusion, the hearts were harvested and maintained at −80°C for subsequent analysis. The frozen ventricles were crushed to a powder using liquid nitrogen-chilled tissue pulverizer. For tissue analysis, weighed quantities of frozen tissues (100 mg) were homogenized in a stroke-physiological saline solution using a microcentrifuge tube homogenizer. Superoxide dismutase (SOD) activity and the glutathione/glutathione disulfide (GSH/GSSG) ratio are typically used to represent the antioxidant molecules of antioxidant systems (24). In addition, malondialdehyde (MDA) is the end-product of lipid peroxidation metabolism, and its content directly reflects the rate and extent of lipid peroxidation (25,26). Therefore, the MDA level, SOD activity and GSH/GSSG ratio in heart homogenates were measured using MDA kit (cat. no. A00320150624), SOD kit (cat. no. A00120150221), GSH kit (cat. no. A00620150214) and GSSG kit (cat. no. A06120150207), according to the manufacturer's protocol.

Analysis of inflammation in heart tissue homogenates

TNF-α, C-reactive protein (CRP) and interleukin-6 (IL-6) were analyzed using ELISA TNF-α kit (cat. no. Ml002126), CRP kit (cat. no. Ml105432) and IL-6 kit (cat. no. ml012815) following the manufacturer's protocol (Enzyme-linked Biotechnology Co., Ltd., Shanghai, China). The concentrations of the cytokines were quantified by referencing standard curves.

Evaluation of apoptosis

Terminal deoxynucleotidyl-transferase-mediated dUTP nick end labelling (TUNEL) assay was performed according to the manufacturer's instructions using the In Situ Cell Death Detection kit (Roche Diagnostics GmbH, Mannheim, Germany) according to a previously described method (27). Following deparaffinization and rehydration, the sections were treated with 10 mM protease K at 37°C for 15 min. Slides were immersed in TUNEL reaction mixture for 60 min at 37°C in a humidified atmosphere in the dark and converter-peroxide was used to incubate the slides at 37°C for 30 min to reveal blue nuclear staining. The slides were subsequently analyzed using light microscopy. The TUNEL index (%) was obtained as the ratio of the number of TUNEL-positive cells divided by the total number of cells and used to evaluate the apoptotic index of TUNEL-stained heart tissues. For each sample, eight randomly selected areas of TUNEL-stained slices were counted and the mean value was calculated.

Western blot analysis

Protein expression levels of caspase-9 and B-cell lymphoma-2 (Bcl-2) were determined using western blot analysis. Following 30 min of perfusion with the Langendorff apparatus, the ventricular apical of the rats was cut, homogenized in appropriate buffer (50 mM Tris-HCl, pH 7.6, 0.5% Triton X-100, 20% glycerol. All chemicals and reagents were purchased from Solebo Biotechnology Co., Ltd.) and centrifuged at 15,000 g for 15 min at 4°C. Supernatant was extracted and boiled for 15 min to promote protein denaturation. Protein extracts (the protein concentration was determined by bicinchoninic method) were separated using 12% SDS-PAGE. Proteins were transferred to nylon membranes using an electrophoretic transfer system. The membranes were blocked with 5% skimmed milk blocking buffer at 25°C for 1 h and then incubated with primary antibodies overnight (18 h) at 4°C. The membranes were subsequently washed with Tris-buffered saline with Tween-20 (3 times, 5 min each) at 25°C and the corresponding secondary antibodies were used to identify primary antibody binding at 25°C for 60 min. Finally, the blots were visualized with enhanced chemiluminescence-plus reagent (Beijing Solarbio Biological Products, Co., Ltd.) to visualize protein bands, and imaged using the Bio-Rad Gel Doc 2000 imaging system.

Statistical analysis

Data are presented as mean ± or + standard deviation as indicated (n=8 for each condition). A Student's t-test and two-way analysis of variance, followed by Tukey's test were used to analyze the results. Statistical analysis was performed using SPSS 17.0 (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Addition of dihydromyricetin improves the recovery of cardiac function

Cardiac mechanical performance was evaluated by measuring LVDP, ±dp/dtmax and HR Table I indicates the LVDP, ±dp/dtmax and HR obtained during reperfusion of donor hearts. LVDP (P<0.01), +dp/dtmax (P<0.05), -dp/dtmax (P<0.01) and HR (P<0.05) were significantly increased in the HTK-D group when compared with the HTK group (Table I). Hearts in the HTK-A group also demonstrated a significant increase in LVDP, -dp/dtmax and HR compared with those in the HTK group (P<0.05; Table I).

Table I.

Hemodynamic variables among different groups following 6 h of storage and 30 min of reperfusion.

Table I.

Hemodynamic variables among different groups following 6 h of storage and 30 min of reperfusion.

Physical indexHTK groupHTK-A groupHTK-D group
LVDP (mmHg)64.8±0.38 77.90±3.21a 91.51±5.21b
+dp/dtmax (mmHg/sec) 1,234.89±100.131,409.38±90.13 1,613.84±126.13a
dp/dtmax (mmHg/sec)−806.21±70.13 −868.01±60.13a −1,040.09±101.13b
HR (beats/min)238.43±43.34 266.80±40.13a 274.70±60.13a

a P<0.05

b P<0.01 vs. HTK group. Values are presented as the mean ± standard deviation (n=8). LVDP, left ventricular developed pressure; ±dp/dtmax, maximum up/down rate of left ventricular pressure; HR, heart rate; HTK, histidine-tryptophan-ketoglutarate; HTK-A, histidine-tryptophan-ketoglutarate + astragalin; HTK-D, histidine-tryptophan-ketoglutarate + dihydromyricetin.

Addition of astragalin or dihydromyricetin attenuates I/R-induced enzyme release

Necrosis was assessed by the release of LDH and CK into the coronary effluent among the three groups. As indicated in Table II, LDH and CK levels released in the three cardioplegic solutions were not significantly different among the groups during cold storage. However, significantly increased LDH and CK levels were observed in the HTK-A (P<0.05) and HTK-D (P<0.01) groups compared with the respective HTK groups following 30 min reperfusion. These results reflect the cell death of myocytes during reperfusion. The present findings suggested that the addition of astragalin to HTK significantly reduced the release of LDH and CK following reperfusion. In particular, administration of dihydromyricetin significantly prevented the release of LDH and CK to a greater extent when compared with astragalin.

Table II.

Effect of astragalin or dihydromyricetin on levels of LDH and CK in cardioplegic solution prior to reperfusion and in the coronary effluent following reperfusion.

Table II.

Effect of astragalin or dihydromyricetin on levels of LDH and CK in cardioplegic solution prior to reperfusion and in the coronary effluent following reperfusion.

Physical indexIn cardioplegic solutionAt 30 min following reperfusion
CK (U/l)
  HTK group3.90±0.1357.24±6.13
  HTK-A group4.50±0.24 48.97±4.13a
  HTK-D group4.23±0.19 38.67±5.13b
LDH (U/l)
  HTK group3.16±0.2365.31±5.13
  HTK-A group2.98±0.16 50.12±4.32a
  HTK-A group3.36±0.21 41.12±3.62b

a P<0.05

b P<0.01 vs. HTK group. Values are presented as the mean ± standard deviation (n=8). LFH, lactate dehydrogenase; CK, creatine kinase; HTK, histidine-tryptophan-ketoglutarate; HTK-A, histidine-tryptophan-ketoglutarate + astragalin; HTK-D, histidine-tryptophan-ketoglutarate + dihydromyricetin.

Addition of astragalin or dihydromyricetin to HTK-induced IS

Myocardial IS may be used as an indicator of myocardial injury (24). Representative images of heart sections from rats stained with TTC are indicated in Fig. 1. The addition of astragalin significantly reduced I/R-induced myocardial IS by 26.67±1.98% (P<0.05), whereas the addition of dihydromyricetin significantly reduced I/R-induced myocardial IS by 18.36±1.67% (P<0.01) compared with the HTK group (Fig. 1).

Addition of astragalin or dihydromyricetin to HTK alleviates oxidative stress

As indicated in Fig. 2, the addition of astragalin (P<0.05) or dihydromyricetin (P<0.01) to HTK significantly decreased MDA levels (P<0.05; Fig. 2A), and significantly increased SOD activities (Fig. 2B) and GSH/GGSG ratios (P<0.05; Fig. 2C) compared with the HTK group. These results indicated that the addition of astragalin or dihydromyricetin may increase the antioxidant ability of HTK.

Addition of astragalin or dihydromyricetin to HTK weakens cardiomyocyte apoptosis

To test whether the addition of astragalin or dihydromyricetin enhanced the protection against apoptosis during storage and reperfusion, TUNEL assays were performed. Under an optical microscope, TUNEL staining revealed a large number of apoptosis (42.58±4.29%) in the HTK group (Fig. 3A). The number of apoptotic cells was significantly decreased in the HTK-A (36.51±3.63%) and HTK-D (28.13±3.36%) groups (Fig. 3B). The HTK-D group exhibited the greatest reduction in the number of apoptotic cells (Fig. 3B).

Addition of astragalin or dihydromyricetin to HTK reduces the inflammatory response

Inflammation is an important mechanism underlying myocardial injury. The presence of inflammatory cytokines, including CRP, IL-6 and TNF-α, is associated with myocardial injury (24). Therefore, the present study determined the levels of these inflammatory cytokines in myocardial tissues to identify the possible mechanisms underlying the cardioprotective activity of astragalin or dihydromyricetin. As demonstrated in Fig. 4A, the content of TNF-α in the HTK-A group (708.59±51.15 ng/l) and HTK-D group (623.93±51.15 ng/l) were significantly decreased (P<0.05 and P<0.01, respectively) compared with that in the HTK group (772.10±47.53 pg/ml; Fig. 4A). Secretion of CRP (Fig. 4B) was also significantly decreased following astragalin (P<0.05) or dihydromyricetin (P<0.05) treatment compared with the HTK group. In addition, secretion of IL-6 (Fig. 4C) was significantly decreased following astragalin (P<0.05) or dihydromyricetin (P<0.01) treatment compared with the HTK group.

Effect of astragalin and dihydromyricetin preconditioning on the protein expression levels of caspase-9 and Bcl-2

The expression levels of caspase-9 and Bcl-2 proteins determined by western blot analysis from the left ventricular cavity of the rats was determined (Fig. 5A). Quantification of the western blot results indicated that astragalin and dihydromyricetin preconditioning inhibited the increase of proapoptotic protein caspase-9 (P<0.01; Fig. 5B). Furthermore, astragalin and dihydromyricetin preconditioning significantly increased the protein expression levels of antiapoptotic Bcl-2 compared with the HTK group (P<0.01; Fig. 5C). The results indicated that the effect of dihydromyricetin on the expression levels of the two proteins is superior compared with astragalin.

Discussion

The present study demonstrated that donor heart preservation solution containing astragalin or dihydromyricetin improves cardio protection compared with HTK alone during the first 6 h of cold static storage and 30 min of reperfusion. Improved cardio protection was indicated by normalized cardiac function, reduced intracellular oxidation status and inflammatory response, and inhibited myocardial apoptosis. Furthermore, the present results suggested that the effect of dihydromyricetin may exceed that of an equivalent dosage of astragalin as an adjunct to the HTK solution. However, further research is required to investigate this phenomenon in larger and longer-term studies.

Developments in science and technology have significantly improved the methods of graft preservation, and the availability of donor hearts has also increased (28). However, the composition of the cardioplegic solution is one of the key factors influencing the success of cold static preservation, and the methods of minimizing graft dysfunction caused by IR injury remain one of the primary objectives for myocardium protection (2,29).

The coupled effects of ischemia and hypothermia during organ preservation result in inevitable deterioration prior to organ transplantation (2,3). Previous studies have demonstrated that cold ischemia alone may induce the marked generation of ROS, which are decisive mediators of cold storage-induced injury (2,30). When the amount of ROS available exceeds the capacity of the enzymes (such as GSH and SOD) and cannot be diminished during reperfusion, oxidative stress occurs (26). Oxidative stress-associated alterations in several intracellular signaling pathways have been implicated in the pathophysiology of severe donor heart damage during reperfusion (31). In line with these findings, preventing or at least controlling the increase in the oxidative stress levels of donor hearts during cold storage seems to be a crucial part of heart transplantation in cardiac surgery. Thus, the capability of astragalin or dihydromyricetin to alleviate oxidative injury was assessed by investigating the myocardial levels of MDA, GSH/GSSG ratio and SOD activity in the present study.

The present results demonstrated the improved antioxidant status of astragalin or dihydromyricetin-rich HTK preserved grafts, which was consistent with the results of the number of released myocardial enzymes following reperfusion. Although the quantity of enzymes released was not significantly different during cold storage, levels were significantly increased following reperfusion. This finding may demonstrate that the period immediately following reperfusion is the most crucial stage for generating ROS (32). Studies have also indicated that the histidine component in HTK exerts antioxidant effects (33). Thus, enhancement of antioxidant activity and inhibition of free radical peroxidation in the myocardium may be partially involved in the cardioprotective mechanisms of HTK combined with astragalin or dihydromyricetin. Previous studies have demonstrated that the biological markers of hemodynamic parameters, including LVDP, ±dp/dtmax and HR and IS, are major determinants of myocardial function and viability (5,34). In the present study, HTK combined with dihydromyricetin or astragalin significantly improved the recovery of hemodynamic parameters and attenuated IS.

Inflammation and apoptosis, particularly the former, are typical consequences of myocardial damage during I/R, and inflammation may result in activation of the innate immune response (5,26,35). An under-explored area that may hold great potential for improving transplantation outcomes is the design of novel strategies to apply to organs specifically to reduce intra-graft inflammation (36). To investigate the association between the anti-inflammatory effects and the cardioprotective effects of the astragalin or dihydromyricetin flavonoids, an experiment was performed to explore whether they can affect changes in CRP, IL-6, and TNF-α induced by I/R.

In the present study, that addition of astragalin or dihydromyricetin significantly reduced the concentrations of CRP, IL-8, IL-6 and TNF-α compared with HTK alone. A significant decrease in TUNEL-positive nuclear staining in the sectioned left ventricular myocardium was observed in the HTK-D group and HTK-A group when compared with the group treated with HTK alone. ROS have been reported to be potent inducers of various cytokines, including TNF-α and IL-6, and promote cell apoptosis (2,10). Notably, the present study indicated that astragalin and dihydromyricetin demonstrate antiapoptotic effects. However, further investigation is required to investigate whether the observed anti-inflammatory and anti-apoptotic effects of astragalin and dihydromyricetin are direct or only secondary to their anti-oxidative effects.

In conclusion, the results of the present study indicated that the addition of astragalin or dihydromyricetin to HTK significantly reduces myocardial injury, decreases oxidative stress, prevents the apoptotic process, reduces inflammatory response and enhances cardiac performance. In particular, the present study provided preliminary evidence that HTK combined with dihydromyricetin may have potential clinical applications in cardiac transplantation. However, as heart transplantation using donor hearts stored in HTK combined with adjuncts has not yet been performed, other cardiac parameters, including echocardiograms, should also be tested. Studies investigating the biological activities of astragalin and dihydromyricetin are in the initial stages, and further studies are necessary to fully understand the molecular mechanisms involved in the protective effects of astragalin or dihydromyricetin combined with cardioplegic solutions.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Science and Technology Development Planning of Shandong Province (grant. no. 2014GSF118090) to Dong Wang (Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China).

Availability of data and materials

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

Authors' contributions

DW and XZ designed the experiments. DW, XZ, DQ, JH, FM, MX performed the research. DW and QZ analyzed the data and wrote the paper, which was revised by DW and XZ.

Ethics approval and consent to participate

All experimental protocols were performed in accordance with the regulations of the Guide for the Care and Use of Laboratory Animals and approved by the Ethics Committee of Animal Laboratory and Experimental Management Center of Shandong University [Shandong, China; license no. SYXK (Lu) 20130001, revised 2013].

Competing interests

The authors report they have no competing interests.

References

1 

Smit FE and Dohmen PM: Bio-artificial heart as ultimate treatment of end-stage heart failure. Med Sci Monit Basic Res. 20:161–163. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Tan M, Sun X, Guo L, Su C, Sun X and Xu Z: Hydrogen as additive of HTK solution fortifies myocardial preservation in grafts with prolonged cold ischemia. Int J Cardiol. 167:383–390. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Nakao A, Kaczorowski DJ, Wang Y, Cardinal JS, Buchholz BM, Sugimoto R, Tobita K, Lee S, Toyoda Y, Billiar TR and McCurry KR: Amelioration of rat cardiac cold ischemia/reperfusion injury with inhaled hydrogen or carbon monoxide, or both. J Heart Lung Transplant. 29:544–553. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Taylor MJ and Baicu SC: Current state of hypothermic machine perfusion preservation of organs: The clinical perspective. Cryobiology. 60 3 Suppl:S20–S35. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Alves MG, Soares AF, Carvalho RA and Oliveira PJ: Sodium hydrosulfide improves the protective potential of the cardioplegic histidine buffer solution. Eur J Pharmacol. 654:60–67. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Fridell JA, Mangus RS and Tector AJ: Clinical experience with histidine-tryptophan-ketoglutarate solution in abdominal organ preservation: A review of recent literature. Clin Transplant. 23:305–312. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Guibert EE, Petrenko AY, Balaban CL, Somov AY, Rodriguez JV and Fuller BJ: Organ preservation: Current concepts and new strategies for the next decade. Transfus Med Hemother. 38:125–142. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Hackenhaar FS, Fumagalli F, Li Volti G, Sorrenti V, Russo I, Staszewsky L, Masson S, Latini R and Ristagno G: Relationship between post-cardiac arrest myocardial oxidative stress and myocardial dysfunction in the rat. J Biomed Sci. 21:702014. View Article : Google Scholar : PubMed/NCBI

9 

Koch A, Loganathan S, Radovits T, Sack FU, Karck M and Szabó GB: Deferoxamine, the newly developed iron chelator LK-614 and N-alpha-acetyl-histidine in myocardial protection. Interact Cardiovasc Thorac Surg. 10:181–184. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Cho IH, Gong JH, Kang MK, Lee EJ, Park JH, Park SJ and Kang YH: Astragalin inhibits airway eotaxin-1 induction and epithelial apoptosis through modulating oxidative stress-responsive MAPK signaling. BMC Pulm Med. 14:1222014. View Article : Google Scholar : PubMed/NCBI

11 

Collard CD and Gelman S: Pathophysiology, clinical manifestations, and prevention of ischemia-reperfusion injury. Anesthesiology. 94:1133–1138. 2001. View Article : Google Scholar : PubMed/NCBI

12 

Chhabra P, Linden J, Lobo P, Okusa MD and Brayman KL: The immunosuppressive role of adenosine A2A receptors in ischemia reperfusion injury and islet transplantation. Curr Diabetes Rev. 8:419–433. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Semmelmann A, Neeff H, Sommer O, Thomusch O, Hopt UT and von Dobschuetz E: Evaluation of preservation solutions by ESR-spectroscopy: Superior effects of university of wisconsin over histidine-tryptophan-ketoglutarate in reducing renal reactive oxygen species. Kidney Int. 71:875–881. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Rauen U, Klempt S and de Groot H: Histidine-induced injury to cultured liver cells, effects of histidine derivatives and of iron chelators. Cell Mol Life Sci. 64:192–205. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Choi J, Kang HJ, Kim SZ, Kwon TO, Jeong SI and Jang SI: Antioxidant effect of astragalin isolated from the leaves of morus alba L. against free radical-induced oxidative hemolysis of human red blood cells. Arch Pharm Res. 36:912–917. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Hou X, Tong Q, Wang W, Xiong W, Shi C and Fang J: Dihydromyricetin protects endothelial cells from hydrogen peroxide-induced oxidative stress damage by regulating mitochondrial pathways. Life Sci. 130:38–46. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Chen S, Zhao X, Wan J, Ran L, Qin Y, Wang X, Gao Y, Shu F, Zhang Y, Liu P, et al: Dihydromyricetin improves glucose and lipid metabolism and exerts anti-inflammatory effects in nonalcoholic fatty liver disease: A randomized controlled trial. Pharmacol Res. 99:74–81. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Burmistrova O, Quintana J, Díaz JG and Estévez F: Astragalin heptaacetate-induced cell death in human leukemia cells is dependent on caspases and activates the MAPK pathway. Cancer Lett. 309:71–77. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Liu J, Shu Y, Zhang Q, Liu B, Xia J, Qiu M, Miao H, Li M and Zhu R: Dihydromyricetin induces apoptosis and inhibits proliferation in hepatocellular carcinoma cells. Oncol Lett. 8:1645–1651. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Cho IH, Choi YJ, Gong JH, Shin D, Kang MK and Kang YH: Astragalin inhibits autophagy-associated airway epithelial fibrosis. Respir Res. 16:512015. View Article : Google Scholar : PubMed/NCBI

21 

Allahyari S, Delazar A and Najafi M: Evaluation of general toxicity, anti-oxidant activity and effects of ficus carica leaves extract on ischemia/reperfusion injuries in isolated heart of rat. Adv Pharm Bull. 4 Suppl 2:S577–S582. 2014.

22 

Qu D, Han J, Ren H, Yang W, Zhang X, Zheng Q and Wang D: Cardioprotective effects of astragalin against myocardial ischemia/reperfusion injury in isolated rat heart. Oxid Med Cell Longev. 2016:81946902016. View Article : Google Scholar : PubMed/NCBI

23 

Suchalatha S and Devi Shyamala CS: Protective effect of terminalia chebula against experimental myocardial injury induced by isoproterenol. Indian J Exp Biol. 42:174–178. 2004.PubMed/NCBI

24 

Han J, Wang D, Yu B, Wang Y, Ren H, Zhang B, Wang Y and Zheng Q: Cardioprotection against ischemia/reperfusion by licochalcone B in isolated rat hearts. Oxid Med Cell Longev. 2014:1348622014. View Article : Google Scholar : PubMed/NCBI

25 

Michel F, Bonnefont-Rousselot D, Mas E, Drai J and Thérond P: Biomarkers of lipid peroxidation: Analytical aspects. Ann Biol Clin (Paris). 66:605–620. 2008.(In French). PubMed/NCBI

26 

Toth A, Halmosi R, Kovacs K, Deres P, Kalai T, Hideg K, Toth K and Sumegi B: Akt activation induced by an antioxidant compound during ischemia-reperfusion. Free Radic Biol Med. 35:1051–1063. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Zhou M, Liu L, Wang W, Han J, Ren H, Zheng Q and Wang D: Role of licochalcone C in cardioprotection against ischemia/reperfusion injury of isolated rat heart via antioxidant, anti-inflammatory, and anti-apoptotic activities. Life Sci. 132:27–33. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Kuang J, Sun Y, Wang W, Ke H and Ye H: Myocardial apoptosis and injury of donor hearts kept in completely beating status with normothermic blood perfusion for transplants. Int J Clin Exp Med. 8:5767–5773. 2015.PubMed/NCBI

29 

Minasian SM, Galagudza MM, Dmitriev YV, Karpov AA and Vlasov TD: Preservation of the donor heart: From basic science to clinical studies. Interact Cardiovasc Thorac Surg. 20:510–519. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Rauen U and de Groot H: New insights into the cellular and molecular mechanisms of cold storage injury. J Investig Med. 52:299–309. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Ozcinar E, Okatan EN, Tuncay E, Eryilmaz S and Turan B: Improvement of functional recovery of donor heart following cold static storage with doxycycline cardioplegia. Cardiovasc Toxicol. 14:64–73. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Bolli R, Patel BS, Jeroudi MO, Lai EK and McCay PB: Demonstration of free radical generation in ‘stunned’ myocardium of intact dogs with the use of the spin trap-alpha-phenyl N-tert-butyl nitrone. J Clin Invest. 82:476–485. 1988. View Article : Google Scholar : PubMed/NCBI

33 

Akbulut S, Sevmis S, Karakayali H, Bayraktar N, Unlukaplan M, Oksuz E1 and Dagdeviren A: Amifostine enhances the antioxidant and hepatoprotective effects of UW and HTK preservation solutions. World J Gastroenterol. 20:12292–12300. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Hou X, Han J, Yuan C, Ren H, Zhang Y, Zhang T, Xu L, Zheng Q and Chen W: Cardioprotective effects of total flavonoids extracted from xinjiang sprig rosa rugosa against acute ischemia/reperfusion-induced myocardial injury in isolated rat heart. Cardiovasc Toxicol. 16:54–66. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Timmers L, Pasterkamp G, de Hoog VC, Arslan F, Appelman Y and de Kleijn DP: The innate immune response in reperfused myocardium. Cardiovasc Res. 94:276–283. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Solhjou Z, Athar H, Xu Q and Abdi R: Emerging therapies targeting intra-organ inflammation in transplantation. Am J Transplant. 15:305–311. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2018
Volume 18 Issue 3

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang D, Zhang X, Qu D, Han J, Meng F, Xu M and Zheng Q: Astragalin and dihydromyricetin as adjuncts to histidine‑tryptophan‑ketoglutarate cardioplegia enhances protection during cardioplegic arrest. Mol Med Rep 18: 2929-2936, 2018
APA
Wang, D., Zhang, X., Qu, D., Han, J., Meng, F., Xu, M., & Zheng, Q. (2018). Astragalin and dihydromyricetin as adjuncts to histidine‑tryptophan‑ketoglutarate cardioplegia enhances protection during cardioplegic arrest. Molecular Medicine Reports, 18, 2929-2936. https://doi.org/10.3892/mmr.2018.9254
MLA
Wang, D., Zhang, X., Qu, D., Han, J., Meng, F., Xu, M., Zheng, Q."Astragalin and dihydromyricetin as adjuncts to histidine‑tryptophan‑ketoglutarate cardioplegia enhances protection during cardioplegic arrest". Molecular Medicine Reports 18.3 (2018): 2929-2936.
Chicago
Wang, D., Zhang, X., Qu, D., Han, J., Meng, F., Xu, M., Zheng, Q."Astragalin and dihydromyricetin as adjuncts to histidine‑tryptophan‑ketoglutarate cardioplegia enhances protection during cardioplegic arrest". Molecular Medicine Reports 18, no. 3 (2018): 2929-2936. https://doi.org/10.3892/mmr.2018.9254