Crocin inhibits KBTBD7 to prevent excessive inflammation and cardiac dysfunction following myocardial infarction

  • Authors:
    • Chunju Yuan
    • Zhongpu Chen
    • Qianxing Zhou
  • View Affiliations

  • Published online on: December 5, 2022     https://doi.org/10.3892/mmr.2022.12907
  • Article Number: 20
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Myocardial infarction (MI) refers to myocardial ischemic necrosis that is caused by coronary artery disease. Notably, crocin has protective effects on the heart. The present study aimed to i) investigate the protective effect of crocin, an active ingredient in Gardenia jasminoides Ellis and Crocus sativus L., on myocardial ischemia and ii) to verify the interaction between crocin and kelch repeat and BTB domain‑containing protein 7 (KBTBD7), which is a novel member of the BTB‑kelch protein family. In the present study, the left anterior descending coronary artery was ligated to establish a myocardial ischemia‑reperfusion injury (MIRI) model in rats and the protective effect of crocin on rat myocardial tissue was observed. The levels of the inflammatory cytokines, interleukin (IL)‑1β, IL‑6 and tumor necrosis factor α (TNFα), in the sham, MI model, MI + crocin (100 mg/kg) and MI + crocin (200 mg/kg) groups were compared in the rat myocardial tissue. The TUNEL assay was used to detect apoptosis of myocardial cells. In addition, RAW264.7 cells were stimulated with the inflammatory factors recombinant mouse high mobility group box 1 (rmHMGB1) and recombinant mouse heat shock protein 60 (rmHSP60). The inhibitory effect of crocin on inflammatory cytokine levels was observed using ELISA. Western blotting was used to detect the inhibitory effect of crocin on KBTBD7. The inhibitory effect of KBTBD7 knockdown on MAPK and nuclear factor (NF)‑κB signaling pathways was also analyzed. The expression levels of IL‑1β, IL‑6 and TNFα were significantly decreased in the crocin‑treated groups compared with in the model group. Crocin significantly reduced the apoptosis of myocardial cells and significantly inhibited the release of inflammatory cytokines induced by rmHMGB1 and rmHSP60. KBTBD7 was determined to be a target of crocin. Knockdown of KBTBD7 significantly inhibited p38 and NF‑κB signaling pathways. Furthermore, the results demonstrated that KBTBD7 knockdown significantly reduced the production of inflammatory cytokines induced by rmHMGB1 and rmHSP60. KBTBD7 knockdown also significantly reduced p38 and NF‑κB signaling in the rmHMGB1‑ and rmHSP60‑treated groups. The present study demonstrated the potential protective effect of crocin on MIRI in rats. The underlying mechanism may be through direct inhibition of KBTBD7, thereby inhibiting excessive inflammatory responses and myocardial cell apoptosis following myocardial infarction.

Introduction

Acute myocardial infarction (MI) leads to myocardial ischemia, and reperfusion of ischemic myocardium is the primary treatment strategy for acute MI (1). Reperfusion therapy is a treatment that reopens completely blocked blood vessels using thrombolytic drugs, interventional therapy, or surgery, improving outcome for patients with myocardial infarction (MI) (2). Although the ischemic myocardium can be restored to normal perfusion, reperfusion can increase the pathological process of patients with MI, resulting in tissue damage and cardiac dysfunction, called myocardial ischemia-reperfusion injury (MIRI) (3). Increased apoptosis of myocardial cells during MI is accentuated by the subsequent reperfusion-induced inflammatory response (4,5). MI caused by coronary artery obstruction leads to myocardial tissue damage and ischemic death, and is a serious threat to human health. A Sprague-Dawley rat model is often used to study the inflammatory response following MI. Macrophages are the main inflammatory cell type that trigger and regulate the inflammatory response following MI (6,7). Macrophages phagocytose apoptotic or necrotic cells and activate the anti-inflammatory process by releasing cytokines (8). The quantity of inflammatory cytokines released is proportional to cardiac function damage and the level of myocardial apoptosis following ischemia (9). Notably, inflammatory responses are activated during MI and reperfusion can significantly increase the myocardial inflammatory response; MIRI is common in clinical practice (10). An effective and feasible therapeutic treatment that can protect ischemic myocardium, limiting MI-associated damage and reducing MIRI, is therefore necessary. Over the last decade, drug preconditioning has received extensive attention in multiple diseases, including MI (1113).

Traditional Chinese medicines have been extensively investigated because of their potential curative effects (14,15), low toxicity, limited side effects and capacity to act on multiple molecular interactions associated with disease (16). Crocin is an active component found in Gardenia jasminoides Ellis and Crocus sativus L. (1719). Crocin reportedly exhibits cardioprotective (2022) and antitumor properties, and serves an important pharmacological role in the treatment of central nervous diseases (23). Xu et al (24) revealed that crocin suppresses the apoptosis of bovine endothelial cells through increasing the ratio of Bcl-2/Bax, leading to an anti-atherosclerotic role. In addition, Bcl-2/Bax mRNA expression has been reported to be increased in response to crocin, thus inhibiting cell apoptosis (25). A previous study investigating platelet apoptosis induced by Agkistrodon pallas venom demonstrated that crocin may inhibit the expression of caspase-3 and caspase-9 (26). Furthermore, crocin has been shown to effectively inhibit cardiovascular cell and platelet apoptosis (27,28). However, the pharmacological mechanism of crocin remains unclear.

The BTB-kelch protein family is involved in numerous cellular functions, including transcriptional regulation, cytoskeleton formation, ion channels, protein ubiquitination, angiogenesis and apoptosis (29). Kelch repeat and BTB domain-containing protein 7 (KBTBD7) is located on chromosome 13q14.11, has a cDNA length of 3,008 bases, an open reading frame of 2,052 bases and encodes a protein of 684 amino acid residues (30). The KBTBD7 protein contains a conserved BTB domain and a series of kelch repeats. Homologous proteins have been found in a range of species, from yeast to fruit flies to mice, displaying a high level of inter-species protein conservation. Fragment analysis has demonstrated that the BTB, BTB and C-terminal kelch, and kelch repeat domains exhibit transcriptional activation activity (31). However, few cardiovascular studies have reported on KBTBD7. Moreover, the role of KBTBD7 in MIRI and whether crocin functions through KBTBD7 remain unclear.

In the present study, a rat model of acute MI was established to investigate the protective effect of crocin on acute MI aggravated by reperfusion injuries. The interaction between crocin and KBTBD7 was also examined. The present study has provided the foundations for further MI research and the development of new therapeutics.

Materials and methods

Animal model of MI

Clean-grade Sprague Dawley rats (male; age, 8–10 weeks; weight, 200–220 g; n=40) were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd. Animals were housed at 21–25°C and 50–60% humidity, under a 12-h light/dark cycle, with free access to food and water. Sprague-Dawley rats were randomly divided into four groups (n=10): Sham operation group, MI model group, MI + crocin (Amyjet Scientific, Inc.) low-dose group (100 mg/kg) and MI + crocin high-dose group (200 mg/kg). Rats in the MI + crocin low-dose group and MI + crocin high-dose group were intraperitoneally injected with crocin once per day (volume 0.2 ml/100 g) for 14 days. Rats in the sham operation and MI groups were intraperitoneally injected with an equal volume of normal saline for 14 days. The rats were anesthetized 30 min after the last administration by intraperitoneal injection of 2% sodium pentobarbital (40 mg/kg). Following endotracheal intubation, an artificial respirator was connected (tidal volume, 1.5–2 ml/100 g; frequency, 48–54 times/min). The right common carotid artery was carefully separated and the left ventricle was intubated. An incision was made between ribs 3 and 4 on the left side of the chest of rats. The pericardium was excised to completely expose the heart. With the exception of the sham operation group, the left anterior descending coronary artery was ligated. After ligation for 30 min, the ligature was loosened for reperfusion for 120 min. Left ventricular end-diastolic pressure, left ventricular systolic pressure and cardiac function were all evaluated following ischemia and reperfusion in each group. At the end of the experiment, rats were euthanized via carbon dioxide; rats were placed within a box and 100% CO2 was used at a displacement rate of 30% vol/min. After 10 min, the rats were observed to no longer be breathing and had discolored eyes. Minimum CO2 flow was maintained for a further 2 min following respiratory arrest. Animal death was then determined. All animal experiments were approved by the Ethics Committee of Zhongda Hospital Southeast University (approval no. 190501017; Nanjing, China).

Cell culture

The mouse leukemia RAW264.7 cell line was purchased from the American Type Culture Collection and seeded in RPMI 1640 media (Gibco; Thermo Fisher Scientific, Inc.) containing 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.). The cells were cultured in an incubator at 37°C with 5% CO2. After 24 h, the culture medium was changed to serum-free medium overnight. An inflammatory response model was established using recombinant mouse high mobility group box 1 (rmHMGB1; 1 µg/ml; cat. no. ab255799; Abcam) and recombinant mouse heat shock protein 60 (rmHSP60; 1 mol/ml; cat. no. ab92364; Abcam) for 24 h at 37°C. Crocin (low dose, 25 µg/ml; high dose, 50 µg/ml) was used to pretreat RAW264.7 cells 1 h before establishment of the inflammatory response model. The cells in the control group were not treated with crocin, rmHMGB1 or rmHSP60.

Cell transfection

RAW264.7 cells (2×105 cells/well) were transfected with 50 nM small interfering RNA (si)-negative control (NC; 5′-UUCUCCGAACGUGUCACGU-3′) and 50 nM si-KBTBD7 (5′-GGAUUAAUAUAGGCACCAU-3′), synthesized by Shanghai GenePharma Co., Ltd. Lipofectamine® 2000 (Thermo Fisher Scientific, Inc.) reagent was used for transfection according to the manufacturer's protocol. The transfection was performed at 37°C for 24 h.

TUNEL assay

Cardiac tissue apoptosis was detected using the TUNEL assay (cat. no.; 40307ES50; Shanghai Yeasen Biotechnology Co., Ltd.) according to the manufacturer's protocol. Briefly, RAW264.7 cells were fixed with 4% paraformaldehyde at 4°C for 25 min and permeabilized using 0.2% Triton X-100 at room temperature for 5 min. After washing with PBS, the cells were stained with 50 µl TdT buffer at 37°C for 1 h. The nuclei were stained with 2 µg/ml DAPI at room temperature for 5 min in the dark. Images were captured using a confocal microscope and 10 randomly-selected high-magnification fields were observed. The apoptotic rate was calculated as follows: Apoptotic nuclei/total nuclei ×100.

Reverse transcription quantitative-PCR (RT-qPCR)

Total RNA was extracted from rat myocardium and RAW264.7 cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). An ultraviolet spectrophotometer was used to determine the concentration and purity of the RNA. Total RNA was reverse transcribed into cDNA using the following components: High-Fidelity Script reverse transcriptase, dNTPs, primers, RNA, RT buffer, dithiothreitol and RNase-free water (Invitrogen; Thermo Fisher Scientific, Inc.). RT was performed at 37°C for 15 min followed by 85°C for 5 sec. qPCR was subsequently performed using the UltraSYBR Mixture (CoWin Biosciences) according to the manufacturer's protocol. The following thermocycling conditions were used for qPCR: Pre-denaturation at 95°C for 10 min; 35 cycles of denaturation at 95°C for 15 sec, and annealing and elongation at 60°C for 1 min; followed by final extension at 95°C for 15 sec, 60°C for 1 min, 95°C for 15 sec and 60°C for 15 sec. The following primer pairs were used for qPCR: KBTBD7 forward (F), 5′-AGCTCAGTTGTATCGGTAGCA-3′ and reverse (R), 5′-CCGGAATAAGGGTCGTAACAGA-3′; interleukin (IL)-1β F, 5′-GGTGTGTGACGTTCCCATTAGAC-3′ and R, 5′-CATGGAGAATATCACTTGTTGGTTGA-3′; IL-6 F, 5′-TAGTCCTTCCTACCCCAATTTCC-3′ and R, 5′-TTGGTCCTTAGCCACTCCTTC-3′; tumor necrosis factor α (TNFα) F, 5′-AAGCCTGTAGCCCACGTCGTA-3′ and R, 5′-GGCACCACTAGTTGGTTGTCTTTG-3′; Bax F, 5′-AGTGTCTCAGGCGAATTGGC-3′ and R, 5′-CACGGAAGAAGACCTCTCGG-3′; Bcl-2 F, 5′-ACTGAGTACCTGAACCGGCATC-3′ and R, 5′-GGAGAAATCAAACAGAGGTCGC-3′; and GAPDH F, 5′-AGGTCGGTGTGAACGGATTTG-3′ and R, 5′-TGTAGACCATGTAGTTGAGGTCA-3′. mRNA expression levels were quantified using the 2−∆∆Cq method (32) and normalized to the internal reference gene GAPDH.

Western blotting

After 30 min of ischemia and 120 min of reperfusion, 100 mg myocardial tissue was immediately extracted from the ischemic area in the left ventricle of the rats. Tissue was subsequently washed with 1X PBS before tissue lysis solution was added. The tissue was then incubated with RIPA buffer (Beyotime Institute of Biotechnology) on ice for 30 min. Total protein content was determined by bicinchoninic acid (BCA) assay (Thermo Fisher Scientific, Inc.). In addition, RAW264.7 cells were collected, cells were cleared on ice, and total protein was extracted using RIPA buffer and was quantified using a BCA assay. Total protein (20 µg) was separated by SDS-PAGE on 12% gels. After blocking with 5% skim-milk at room temperature for 1 h, the PVDF membranes were incubated with primary antibodies against HMGB1 (1:1,000; cat. no. ab227526; Abcam), HSP60 (1:1,000; cat no. ab190828; Abcam), p38 (1:1,000: cat. no. 8690; Cell Signaling Technology, Inc.), phosphorylated (p)-p38 (1:1,000; cat. no. 4511; Cell Signaling Technology, Inc.), JNK (1:1,000; cat. no. 9252; Cell Signaling Technology, Inc.), p-JNK (1:1,000; cat. no. 9251; Cell Signaling Technology, Inc.), p65 (1:1,000; cat. no. 8242; Cell Signaling Technology, Inc.), p-p65 (1:1,000; cat. no. 3033; Cell Signaling Technology, Inc.) and GAPDH (1:1,000; cat. no. 5174; Cell Signaling Technology, Inc.) overnight at 4°C. Horseradish peroxidase-labeled secondary antibodies (1:3,000; cat. no. 7074; Cell Signaling Technology, Inc.) were then added to incubate with the membranes at room temperature for 2 h. Protein bands were visualized using ECL (Shanghai Yeasen Biotechnology Co., Ltd.). The ratio of gray values was used for semi-quantitative analysis with GAPDH as the loading control using ImageJ software (version 1.8.0; National Institutes of Health).

ELISA

The RAW 264.7 cell supernatant solution was collected. ELISA was performed according to the manufacturers' instructions. The levels of cytokines in each treatment group were determined using the standard curve and optical density. The following ELISA kits were used: IL-1β ELISA Kit [cat. no. EK201B/3; Hangzhou Multi Sciences (Lianke) Biotech, Co., Ltd.], TNFα ELISA Kit [cat. no. EK282/4; Hangzhou Multi Sciences (Lianke) Biotech, Co., Ltd.], IL-6 ELISA Kit [cat. no. EK206/3; Hangzhou Multi Sciences (Lianke) Biotech, Co., Ltd.], HSP60 ELISA Kit (cat. no. EM1136; FineTest), and HMGB1 ELISA Kit (cat. no. EM0382; FineTest).

Statistical analysis

Data were analyzed using SPSS 19.0 software (IBM Corporation) from three independent experiments. Data are presented as the mean ± standard deviation. Unpaired Student's t-test was used for comparisons between two groups. One-way ANOVA and Tukey's post-hoc test was used for comparisons between multiple groups (3335). P<0.05 was considered to indicate a statistically significant difference.

Results

Crocin inhibits the inflammatory response and myocardial apoptosis caused by MIRI

A rat model of MIRI was constructed to study the pharmacological effects of crocin, the chemical structure of which is shown in Fig. 1A. To assess the inflammatory response and the degree of myocardial injury, mRNA expression levels of IL-1β, IL-6 and TNFα in myocardial tissue were determined. The mRNA expression levels of IL-1β, IL-6 and TNFα were significantly increased in the MI group compared with those in the sham group. However, the mRNA expression levels of IL-1β, IL-6 and TNFα were significantly decreased following crocin treatment compared with in the MI group (Fig. 1B-D). Furthermore, the TUNEL assay was used to detect apoptosis. Apoptosis was observed following myocardial I/R in the MI group, this was significant compared with the sham group; however, the apoptotic rate in the low and high crocin treatment groups was significantly reduced compared with that in the MI group (Fig. 1E). The mRNA expression levels of the apoptosis-related protein Bax and antiapoptotic protein Bcl-2 were also detected. The mRNA expression levels of Bax in the myocardial tissue of the MI group were significantly increased compared with those in the sham group; however, Bax expression was significantly reduced following treatment with high and low doses of crocin compared with in the MI group (Fig. 1F). The mRNA expression levels of Bcl-2 were significantly downregulated in the MI group compared with those in the sham group, and were significantly upregulated following high and low levels of crocin treatment compared with in the MI group (Fig. 1G). These results therefore suggested that crocin could inhibit myocardial apoptosis caused by MIRI.

Crocin inhibits the production of damage-associated molecular pattern (DAMP)-triggered inflammatory cytokines

Changes in the levels of HMGB1 and HSP60 in myocardial tissue following MI were determined. HMGB1 and HSP60 levels, measured using ELISA, were significantly increased in the MI group compared with those in the sham group (Fig. 2A and B). Western blotting results also demonstrated that the protein expression levels of HMGB1 and HSP60 were significantly increased in the MI group compared with those in the sham group (Fig. 2C). Subsequently, rmHMGB1 and rmHSP60 were used to stimulate an inflammatory response in RAW264. 7 cells and the effect of crocin on these macrophages was observed. The results demonstrated that the levels of IL-1β, IL-6 and TNFα following rmHMGB1 treatment were significantly downregulated by the addition of crocin, in a dose-dependent manner, compared with the control (no crocin treatment) (Fig. 2D). These results were further reflected upon RAW 264. 7 cells treatment with rmHSP60 (Fig. 2E). These results indicated that crocin may inhibit the inflammatory response triggered by HMGB1 and HSP60.

Activation of MAPK and NF-κB signaling pathways is inhibited by crocin

Western blotting was used to detect the expression levels of proteins involved in the MAPK and NF-κB signaling pathways following treatment with rmHMGB1, rmHSP60 and crocin. The results demonstrated that the protein expression levels of p-p38, p-JNK and p-p65 were significantly increased following treatment with rmHMGB1 and rmHSP60 compared with those in the control group. However, the protein expression levels of p-p38, p-JNK and p-p65 were significantly reduced following high-dose crocin treatment compared with cells that received no crocin (Fig. 3A and B). Therefore, the results indicated that rmHMGB1 and rmHSP60 macrophage stimulation may activate the p38 and NF-κB signaling pathways. The results also suggested that crocin may suppress the production of inflammatory cytokines through inhibition of the MAPK and NF-κB signaling pathways.

Crocin targets and inhibits KBTBD7

KBTBD7 plays a crucial role in MI, especially in the response to inflammation and regulation of myocardial function (6). Therefore, the present study investigated whether crocin could function through regulating KBTBD7. qPCR detection demonstrated that KBTBD7 was significantly upregulated following MI compared with in the sham group. Furthermore, high-dose crocin treatment significantly downregulated KBTBD7 mRNA expression levels compared with those in the MI group (Fig. 4A). In RAW264.7 cells, rmHMGB1 induction significantly upregulated KBTBD7 mRNA expression levels compared with in the control, whereas crocin inhibited KBTBD7 mRNA expression levels in rmHMGB1-treated cells (Fig. 4B). Similar results were also determined in the rmHSP60-induced RAW264.7 cells (Fig. 4C). These results therefore suggested that KBTBD7 is a target of crocin.

KBTBD7 knockdown reduces the production of DAMP-induced inflammatory cytokines

The effect of KBTBD7 on DAMP-induced inflammatory cytokines in RAW264.7 cells was further observed. The transfection efficiency of si-KBTBD7 was first analyzed. The results demonstrated that si-KBTBD7 could significantly reduce mRNA expression levels of KBTBD7 compared with si-NC (Fig. 5A). Furthermore, the results demonstrated that KBTBD7 knockdown significantly decreased the levels of the rmHMGB1- and rmHSP60-induced inflammatory cytokines compared with si-NC (Fig. 5B and C).

KBTBD7 knockdown reduces DAMP-induced p38 and NF-κB signaling activation

The molecular mechanism by which KBTBD7 affects the inflammatory response was investigated. Western blotting was used to detect the expression levels of proteins associated with the MAPK and NF-κB pathways following rmHMGB1, rmHSP60 and si-KBTBD7 treatments. The results demonstrated that the protein expression levels of p-p38, p-JNK and p-p65 were significantly increased following rmHMGB1 and rmHSP60 treatment compared with those in the control group. However, KBTBD7 knockdown significantly reduced the protein expression levels of p-p38, p-JNK and p-p65 compared with the rmHMGB1 and rmHSP60 only treatments (Fig. 6A and B). These results indicated that rmHMGB1 and rmHSP60 stimulation activated the p38 and NF-κB signaling pathways, whereas KBTBD7 knockdown potentially inhibited the production of inflammatory cytokines.

Discussion

Ischemic cardiomyopathy is a complex heart disease worldwide (36). The primary and most common cause of ischemic cardiomyopathy is coronary atherosclerosis, which causes lumen stenosis or occlusion (37). Early reperfusion therapy is an important strategy to save dying myocardial tissues; however, reperfusion itself can also cause injury. Reperfusion can induce the production of high levels of reactive oxygen species, resulting in the peroxidation damage of proteins, lipids and chromosomes, calcium overload, leukocyte accumulation, abnormal pH and ATP energy metabolism disorder (38). The inflammatory response plays an important role in this phenomenon by triggering the release of numerous pro-inflammatory factors, such as TNFα, IL-6 and IL-1β, which can lead to tissue damage and in severe cases to apoptosis and necrosis (39). Therefore, inhibiting the inflammatory response can reduce MIRI to a certain extent (40).

Crocin is a traditional Chinese medicine extract with strong bioactivity (17). Crocin has been reported to exhibit a variety of pharmacological activities, such as antioxidative, anticancer, anti-inflammatory and anti-atherosclerosis effects, and cardiovascular protection (41). Crocin has also been shown to act on mitochondria and inhibit mitochondrial dysfunction in cardiovascular-related tissues and cells (4244). Crocin has previously been reported to reduce the hydrogen peroxide-induced mitochondrial membrane potential of platelets (44) and increase the release rate of cytochrome c into rat heart tissues (42). Crocin may also act on cardiovascular diseases via reducing platelet aggregation and apoptosis, oxidative stress and cardiovascular apoptosis (45,46). In the present study, the literature and preliminary experiments were used to determine the concentrations of crocin to use in animals and cells (4750). The preliminary results demonstrated that the current dose of crocin (200 mg/kg in mice) would reflect the therapeutic effect of crocin on myocardial ischemia.

Following MIRI, DAMPs are released, which consequently activate toll-like receptors expressed in macrophages, triggering a series of inflammatory responses. Notably, HMGB1 and HSP60 are major DAMP triggers (51,52). HMGB1 is a highly conserved and widely present protein in the nuclei of eukaryotic cells (53,54). Under normal physiological conditions, HMBG1 has the function of a nuclear-binding protein, whereas in the pathological state, it can enter the intercellular space from the nucleus through passive release and active secretion, thus triggering an inflammatory response (55). As a pro-inflammatory protein, HMGB1 plays an important role in MIRI and is associated with inflammatory factors, such as TNFα, IL-6 and inducible nitric oxide synthase (56). Inhibition of HMGB1 activity has demonstrated the protective effects of HMGB1 on cardiopulmonary function injury (57). The present study further investigated the inhibitory effects of crocin on HMGB1 and HSP60-induced inflammatory responses. The results demonstrated that treatment with HMGB1 and HSP60 markedly upregulated the levels of IL-1β, IL-6, and TNFα in RAW 264.7 cells. Conversely, the levels of these pro-inflammatory cytokines were significantly inhibited by crocin. The cardiac protective effect of crocin may be related to the inhibition of inflammatory responses. The release of inflammatory factors is closely related to the occurrence and development of MIRI (58). During and after MIRI, a high level of inflammatory cytokines, such as TNFα and IL-6, are released into the blood, which can cause neutrophils to adhere to endothelial cells (59). This phenomenon results in no reflow after reperfusion and a large amount of oxygen free radicals are generated in response (60). In addition, macrophages are encouraged to secrete a large amount of inflammatory factors, which eventually cause or aggravate the degree of MIRI (61). In the present study, crocin treatment reduced the levels of TNFα and IL-6 in the serum of rats with MIRI. This result suggested that crocin may protect against MIRI in rats by reducing inflammatory responses.

The MAPK and NF-κB signaling pathways serve key roles in inflammatory responses in MIRI; therefore, inhibition of the MAPK and NF-κB signaling pathways serves an important role in reducing the inflammatory response (62,63). The present study revealed that crocin suppressed the production of inflammatory cytokines, which may be mediated through inactivation of the MAPK and NF-κB signaling pathways.

The BTB protein is a transcription factor in eukaryotes, which is closely related to the development of various multicellular organisms, including fruit flies and vertebrates. Yang et al (6) demonstrated that microRNA-21 inhibited the inflammatory response in the early stages of MI by targeting KBTBD7, and inhibiting p38 and NF-κB signaling activation, thereby preventing excessive scarring and improving cardiac function. In the present study, crocin reduced the inflammation caused by MIRI and the apoptosis of myocardial cells. The protective effect of crocin may be realized by inhibiting the expression of KBTBD7.

In conclusion, the present study demonstrated that crocin protects against MIRI by inhibiting myocardial inflammatory response and regulating the expression of inflammatory factors. The anti-inflammatory effects of crocin were possibly related to the inhibition of KBTBD7. The present study has provided a new therapeutic approach for MIRI and lays the foundations for further research on the myocardial-protective mechanism of crocin and the development of novel treatments. Moreover, KBTBD7 may be a potential target of MI therapy.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

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

CY designed the study and wrote the manuscript. CY, ZC and QZ performed all experiments. ZC is responsible for data analysis. CY and QZ confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All animal experiments were operated according to the Guide for the Care and Use of Laboratory Animals (64) and were approved by the Ethics Committee of Zhongda Hospital Southeast University (approval no. 190501017; Nanjing, China).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Mui D and Zhang Y: Mitochondrial scenario: Roles of mitochondrial dynamics in acute myocardial ischemia/reperfusion injury. J Recept Signal Transduct Res. 41:1–5. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Rout A, Tantry US, Novakovic M, Sukhi A and Gurbel PA: Targeted pharmacotherapy for ischemia reperfusion injury in acute myocardial infarction. Expert Opin Pharmacother. 21:1851–1865. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Hausenloy DJ and Yellon DM: Myocardial ischemia-reperfusion injury: A neglected therapeutic target. J Clin Invest. 123:92–100. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Turer AT and Hill JA: Pathogenesis of myocardial ischemia-reperfusion injury and rationale for therapy. Am J Cardiol. 106:360–368. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Davidson SM, Ferdinandy P, Andreadou I, Bøtker HE, Heusch G, Ibáñez B, Ovize M, Schulz R, Yellon DM, Hausenloy DJ, et al: Multitarget strategies to reduce myocardial ischemia/reperfusion injury: JACC review topic of the week. J Am Coll Cardiol. 73:89–99. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Yang L, Wang B, Zhou Q, Wang Y, Liu X, Liu Z and Zhan Z: MicroRNA-21 prevents excessive inflammation and cardiac dysfunction after myocardial infarction through targeting KBTBD7. Cell Death Dis. 9:7692018. View Article : Google Scholar : PubMed/NCBI

7 

Frantz S and Nahrendorf M: Cardiac macrophages and their role in ischaemic heart disease. Cardiovasc Res. 102:240–248. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Lambert JM, Lopez EF and Lindsey ML: Macrophage roles following myocardial infarction. Int J Cardiol. 130:147–158. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Zhang Y, Wang Y, Xu J, Tian F, Hu S, Chen Y and Fu Z: Melatonin attenuates myocardial ischemia-reperfusion injury via improving mitochondrial fusion/mitophagy and activating the AMPK-OPA1 signaling pathways. J Pineal Res. 66:e125422019. View Article : Google Scholar : PubMed/NCBI

10 

Ye B, Chen X, Dai S, Han J, Liang X, Lin S, Cai X, Huang Z and Huang W: Emodin alleviates myocardial ischemia/reperfusion injury by inhibiting gasdermin D-mediated pyroptosis in cardiomyocytes. Drug Des Devel Ther. 13:975–990. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Ryu SM, Kim HJ, Cho KR and Jo WM: Myocardial protective effect of tezosentan, an endothelin receptor antagonist, for ischemia-reperfusion injury in experimental heart failure models. J Korean Med Sci. 24:782–788. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Xi X, Liu N, Wang Q, Chu Y, Yin Z, Ding Y and Lu Y: ACT001, a novel PAI-1 inhibitor, exerts synergistic effects in combination with cisplatin by inhibiting PI3K/AKT pathway in glioma. Cell Death Dis. 10:7572019. View Article : Google Scholar : PubMed/NCBI

13 

Wang H, Zhong W, Zhao J, Zhang H, Zhang Q, Liang Y, Chen S, Liu H, Zong S, Tian Y, et al: Oleanolic acid inhibits epithelial-mesenchymal transition of hepatocellular carcinoma by promoting iNOS dimerization. Mol Cancer Ther. 18:62–74. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Zhong W, Sun B, Gao W, Qin Y, Zhang H, Huai L, Tang Y, Liang Y, He L, Zhang X, et al: Salvianolic acid A targeting the transgelin-actin complex to enhance vasoconstriction. EBioMedicine. 37:246–258. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Zhong W, Hou H, Liu T, Su S, Xi X, Liao Y, Xie R, Jin G, Liu X, Zhu L, et al: Cartilage oligomeric matrix protein promotes epithelial-mesenchymal transition by interacting with transgelin in colorectal cancer. Theranostics. 10:8790–8806. 2020. View Article : Google Scholar : PubMed/NCBI

16 

Zhong W, Yang W, Qin Y, Gu W, Xue Y, Tang Y, Xu H, Wang H, Zhang C, Wang C, et al: 6-Gingerol stabilized the p-VEGFR2/VE-cadherin/β-catenin/actin complex promotes microvessel normalization and suppresses tumor progression. J Exp Clin Cancer Res. 38:2852019. View Article : Google Scholar : PubMed/NCBI

17 

Chen L, Qi Y and Yang X: Neuroprotective effects of crocin against oxidative stress induced by ischemia/reperfusion injury in rat retina. Ophthalmic Res. 54:157–168. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Lee IA, Lee JH, Baek NI and Kim DH: Antihyperlipidemic effect of crocin isolated from the fructus of Gardenia jasminoides and its metabolite crocetin. Biol Pharm Bull. 28:2106–2110. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Hashemzaei M, Mamoulakis C, Tsarouhas K, Georgiadis G, Lazopoulos G, Tsatsakis A, Shojaei Asrami E and Rezaee R: Crocin: A fighter against inflammation and pain. Food Chem Toxicol. 143:1115212020. View Article : Google Scholar : PubMed/NCBI

20 

Hashemzaei M, Rezaee R, Nabatzehi M, Tsarouhas K, Konstantinos Nikolouzakis T, Lazopoulos G, A Spandidos D, Tsatsakis A and Shahraki J: Anti-hypertensive effect of crocin and hesperidin combination in high-fat diet treated rats. Exp Ther Med. 19:3840–3844. 2020.PubMed/NCBI

21 

Rezaee R, Mahmoudi M, Abnous K, Zamani Taghizadeh Rabe S, Tabasi N, Hashemzaei M and Karimi G: Cytotoxic effects of crocin on MOLT-4 human leukemia cells. J Complement Integr Med. 10:1–8. 2013.PubMed/NCBI

22 

El-Baz FK, Aly HF and Abd-Alla HI: The ameliorating effect of carotenoid rich fraction extracted from Dunaliella salina microalga against inflammation-associated cardiac dysfunction in obese rats. Toxicol Rep. 7:118–124. 2020. View Article : Google Scholar : PubMed/NCBI

23 

Tian Y, Pu X, Yu H, Ji A, Gao R, Hu Y, Xu Z and Wang H: Genome-wide characterization and analysis of bHLH transcription factors related to crocin biosynthesis in Gardenia jasminoides ellis (rubiaceae). Biomed Res Int. 2020:29038612020. View Article : Google Scholar : PubMed/NCBI

24 

Xu G, Gong Z, Yu W, Gao L, He S and Qian Z: Increased expression ratio of Bcl-2/Bax is associated with crocin-mediated apoptosis in bovine aortic endothelial cells. Basic Clin Pharmacol Toxicol. 100:31–35. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Ochiai T, Ohno S, Soeda S, Tanaka H, Shoyama Y and Shimeno H: Crocin prevents the death of rat pheochromyctoma (PC-12) cells by its antioxidant effects stronger than those of alpha-tocopherol. Neurosci Lett. 362:61–64. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Sebastin Santhosh M, Hemshekhar M, Thushara RM, Devaraja S, Kemparaju K and Girish KS: Vipera russelli venom-induced oxidative stress and hematological alterations: Amelioration by crocin a dietary colorant. Cell Biochem Funct. 31:41–50. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Ordoudi SA, Befani CD, Nenadis N, Koliakos GG and Tsimidou MZ: Further examination of antiradical properties of Crocus sativus stigmas extract rich in crocins. J Agric Food Chem. 57:3080–3086. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Oruc S, Gönül Y, Tunay K, Oruc OA, Bozkurt MF, Karavelioğlu E, Bağcıoğlu E, Coşkun KS and Celik S: The antioxidant and antiapoptotic effects of crocin pretreatment on global cerebral ischemia reperfusion injury induced by four vessels occlusion in rats. Life Sci. 154:79–86. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Genau HM, Huber J, Baschieri F, Akutsu M, Dötsch V, Farhan H, Rogov V and Behrends C: CUL3-KBTBD6/KBTBD7 ubiquitin ligase cooperates with GABARAP proteins to spatially restrict TIAM1-RAC1 signaling. Mol Cell. 57:995–1010. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Hu J, Yuan W, Tang M, Wang Y, Fan X, Mo X, Li Y, Ying Z, Wan Y, Ocorr K, et al: KBTBD7, a novel human BTB-kelch protein, activates transcriptional activities of SRE and AP-1. BMB Rep. 43:17–22. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Liu YT, Liu F, Cao L, Xue L, Gu WT, Zheng YZ, Tang H, Wang Y, Yao H, Zhang Y, et al: The KBTBD6/7-DRD2 axis regulates pituitary adenoma sensitivity to dopamine agonist treatment. Acta Neuropathol. 140:377–396. 2020. View Article : Google Scholar : PubMed/NCBI

32 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

33 

Gao Y, Wang C, Wang Z, Li W, Liu Y, Shou S and Chai Y: Semaphorin 3A contributes to sepsis-induced immunosuppression by impairing CD4+ T cell anergy. Mol Med Rep. 23:3022021. View Article : Google Scholar : PubMed/NCBI

34 

Li J, Wang X, Ma C, Xu S, Xu M, Yang J, Wang R and Xue L: Dual PI3K/mTOR inhibitor NVP-BEZ235 decreases the proliferation of doxorubicin-resistant K562 cells. Mol Med Rep. 23:3012021. View Article : Google Scholar : PubMed/NCBI

35 

Wu H, Tao Y, Zhang W, Wang G and Zhang Q: circ-0000212 promotes cell proliferation of colorectal cancer by sponging miR-491 and modulating FOXP4 expression. Mol Med Rep. 23:3002021. View Article : Google Scholar : PubMed/NCBI

36 

Liu NB, Wu M, Chen C, Fujino M, Huang JS, Zhu P and Li XK: Novel molecular targets participating in myocardial ischemia-reperfusion injury and cardioprotection. Cardiol Res Pract. 2019:69351472019. View Article : Google Scholar : PubMed/NCBI

37 

Mozaffari MS, Liu JY, Abebe W and Baban B: Mechanisms of load dependency of myocardial ischemia reperfusion injury. Am J Cardiovasc Dis. 3:180–196. 2013.PubMed/NCBI

38 

Fischesser DM, Bo B, Benton RP, Su H, Jahanpanah N and Haworth KJ: Controlling reperfusion injury with controlled reperfusion: Historical perspectives and new paradigms. J Cardiovasc Pharmacol Ther. 26:504–523. 2021. View Article : Google Scholar : PubMed/NCBI

39 

Liu H, Wu X, Luo J, Wang X, Guo H, Feng D, Zhao L, Bai H, Song M, Liu X, et al: Pterostilbene attenuates astrocytic inflammation and neuronal oxidative injury after ischemia-reperfusion by inhibiting NF-κB phosphorylation. Front Immunol. 10:24082019. View Article : Google Scholar : PubMed/NCBI

40 

Wang C, Sun H, Song Y, Ma Z, Zhang G, Gu X and Zhao L: Pterostilbene attenuates inflammation in rat heart subjected to ischemia-reperfusion: Role of TLR4/NF-κB signaling pathway. Int J Clin Exp Med. 8:1737–1746. 2015.PubMed/NCBI

41 

Farkhondeh T and Samarghandian S: The effect of saffron (Crocus sativus L.) and its ingredients on the management of diabetes mellitus and dislipidemia. Afr J Pharm Pharmacol. 8:541–549. 2014. View Article : Google Scholar

42 

Razavi BM, Hosseinzadeh H, Movassaghi AR, Imenshahidi M and Abnous K: Protective effect of crocin on diazinon induced cardiotoxicity in rats in subchronic exposure. Chem Biol Interact. 203:547–555. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Sarshoori JR, Asadi MH and Mohammadi MT: Neuroprotective effects of crocin on the histopathological alterations following brain ischemia-reperfusion injury in rat. Iran J Basic Med Sci. 17:895–902. 2014.PubMed/NCBI

44 

Thushara RM, Hemshekhar M, Santhosh MS, Jnaneshwari S, Nayaka SC, Naveen S, Kemparaju K and Girish KS: Crocin, a dietary additive protects platelets from oxidative stress-induced apoptosis and inhibits platelet aggregation. Mol Cell Biochem. 373:73–83. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Vakili A, Einali MR and Bandegi AR: Protective effect of crocin against cerebral ischemia in a dose-dependent manner in a rat model of ischemic stroke. J Stroke Cerebrovasc Dis. 23:106–113. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Zhang X, Fan Z and Jin T: Crocin protects against cerebral-ischemia-induced damage in aged rats through maintaining the integrity of blood-brain barrier. Restor Neurol Neurosci. 35:65–75. 2017.PubMed/NCBI

47 

Naghizadeh B, Boroushaki MT, Vahdati Mashhadian N and Mansouri MT: Protective effects of crocin against cisplatin-induced acute renal failure and oxidative stress in rats. Iran Biomed J. 12:93–100. 2008.PubMed/NCBI

48 

He SY, Qian ZY, Tang FT, Wen N, Xu GL and Sheng L: Effect of crocin on experimental atherosclerosis in quails and its mechanisms. Life Sci. 77:907–921. 2005. View Article : Google Scholar : PubMed/NCBI

49 

Roshankhah S, Salahshoor MR, Jalili F, Karimi F, Sohrabi M and Jalili C: Crocin effects on the nicotine-induce ovary injuries in female rat. Int J Life Sci Pharm Res. 7:1–8. 2017.

50 

Hussain MA, Abogresha NM, AbdelKader G, Hassan R, Abdelaziz EZ and Greish SM: Antioxidant and anti-inflammatory effects of crocin ameliorate doxorubicin-induced nephrotoxicity in rats. Oxid Med Cell Longev. 2021:88417262021. View Article : Google Scholar : PubMed/NCBI

51 

Khwaja B, Thankam FG and Agrawal DK: Mitochondrial DAMPs and altered mitochondrial dynamics in OxLDL burden in atherosclerosis. Mol Cell Biochem. 476:1915–1928. 2021. View Article : Google Scholar : PubMed/NCBI

52 

Yang H, Wang H and Andersson U: Targeting Inflammation Driven by HMGB1. Front Immunol. 11:4842020. View Article : Google Scholar : PubMed/NCBI

53 

Andrassy M, Volz HC, Riedle N, Gitsioudis G, Seidel C, Laohachewin D, Zankl AR, Kaya Z, Bierhaus A, Giannitsis E, et al: HMGB1 as a predictor of infarct transmurality and functional recovery in patients with myocardial infarction. J Intern Med. 270:245–253. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Ding HS, Yang J, Chen P, Yang J, Bo SQ, Ding JW and Yu QQ: The HMGB1-TLR4 axis contributes to myocardial ischemia/reperfusion injury via regulation of cardiomyocyte apoptosis. Gene. 527:389–393. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Hu X, Fu W and Jiang H: HMGB1: A potential therapeutic target for myocardial ischemia and reperfusion injury. Int J Cardiol. 155:4892012. View Article : Google Scholar : PubMed/NCBI

56 

Li Q, Xu M, Li Z, Li T, Wang Y, Chen Q, Wang Y, Feng J, Yin X and Lu C: Maslinic acid attenuates ischemia/reperfusion injury-induced myocardial inflammation and apoptosis by regulating HMGB1-TLR4 axis. Front Cardiovasc Med. 8:7689472021. View Article : Google Scholar : PubMed/NCBI

57 

Giallauria F, Cirillo P, D'agostino M, Petrillo G, Vitelli A, Pacileo M, Angri V, Chiariello M and Vigorito C: Effects of exercise training on high-mobility group box-1 levels after acute myocardial infarction. J Card Fail. 17:108–114. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Vinten-Johansen J, Jiang R, Reeves JG, Mykytenko J, Deneve J and Jobe LJ: Inflammation, proinflammatory mediators and myocardial ischemia-reperfusion Injury. Hematol Oncol Clin North Am. 21:123–145. 2007. View Article : Google Scholar : PubMed/NCBI

59 

Hou M, Wu X, Zhao Z, Deng Q, Chen Y and Yin L: Endothelial cell-targeting, ROS-ultrasensitive drug/siRNA co-delivery nanocomplexes mitigate early-stage neutrophil recruitment for the anti-inflammatory treatment of myocardial ischemia reperfusion injury. Acta Biomater. 143:344–355. 2022. View Article : Google Scholar : PubMed/NCBI

60 

Gao C, Liu Y, Yu Q, Yang Q, Li B, Sun L, Yan W, Cai X, Gao E, Xiong L, et al: TNF-α antagonism ameliorates myocardial ischemia-reperfusion injury in mice by upregulating adiponectin. Am J Physiol Heart Circ Physiol. 308:H1583–H1591. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Cao J, Xie H, Sun Y, Zhu J, Ying M, Qiao S, Shao Q, Wu H and Wang C: Sevoflurane post-conditioning reduces rat myocardial ischemia reperfusion injury through an increase in NOS and a decrease in phopshorylated NHE1 levels. Int J Mol Med. 36:1529–1537. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Xie C, Li X, Zhu J, Wu J, Geng S and Zhong C: Magnesium isoglycyrrhizinate suppresses LPS-induced inflammation and oxidative stress through inhibiting NF-κB and MAPK pathways in RAW264.7 cells. Bioorg Med Chem. 27:516–524. 2019. View Article : Google Scholar : PubMed/NCBI

63 

Wang HW, Liu HJ, Cao H, Qiao ZY and Xu YW: Diosgenin protects rats from myocardial inflammatory injury induced by ischemia-reperfusion. Med Sci Monit. 24:246–253. 2018. View Article : Google Scholar : PubMed/NCBI

64 

National Research Council (US) Committee for the Update of the Guide for the Care and Use of Laboratory Animals, . Guide for the care and use of laboratory animals. 8th edition. Washington (DC): National Academies Press (US); 2011

Related Articles

Journal Cover

January-2023
Volume 27 Issue 1

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
Yuan C, Chen Z and Zhou Q: Crocin inhibits KBTBD7 to prevent excessive inflammation and cardiac dysfunction following myocardial infarction. Mol Med Rep 27: 20, 2023
APA
Yuan, C., Chen, Z., & Zhou, Q. (2023). Crocin inhibits KBTBD7 to prevent excessive inflammation and cardiac dysfunction following myocardial infarction. Molecular Medicine Reports, 27, 20. https://doi.org/10.3892/mmr.2022.12907
MLA
Yuan, C., Chen, Z., Zhou, Q."Crocin inhibits KBTBD7 to prevent excessive inflammation and cardiac dysfunction following myocardial infarction". Molecular Medicine Reports 27.1 (2023): 20.
Chicago
Yuan, C., Chen, Z., Zhou, Q."Crocin inhibits KBTBD7 to prevent excessive inflammation and cardiac dysfunction following myocardial infarction". Molecular Medicine Reports 27, no. 1 (2023): 20. https://doi.org/10.3892/mmr.2022.12907