Open Access

CTRP12 alleviates cardiomyocyte ischemia‑reperfusion injury via regulation of KLF15

  • Authors:
    • Bo Liao
    • Xiaoyuan Tian
  • View Affiliations

  • Published online on: June 2, 2022     https://doi.org/10.3892/mmr.2022.12763
  • Article Number: 247
  • Copyright: © Liao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Myocardial ischemia‑reperfusion (I/R) serves a crucial role in myocardial infarction. C1q/TNF‑related protein 12 (CTRP12) is a secretory protein involved in metabolism. It has been reported that CTRP12 participates in the regulation of numerous cardiovascular diseases. However, its role in myocardial I/R injury remains unclear. In the present study, the left anterior descending coronary artery in mice was ligated to establish a mouse I/R model. A myocardial hypoxia‑reoxygenation (H/R) cell model was also established. Cardiomyocyte injury was evaluated using hematoxylin and eosin staining, Cell Counting Kit‑8 and a lactate dehydrogenase (LDH) kit. The expression levels of CTRP12 and Krueppel‑like factor 15 (KLF15) in murine myocardial tissues and H9c2 cells were determined using reverse transcription‑quantitative PCR and western blotting, as KLF15 was previously reported to protect against I/R‑induced cardiomyocyte damage. Furthermore, inflammatory factors TNF‑α, IL‑1β and IL‑6 were analyzed using ELISA while apoptosis was assessed using TUNEL assays and western blotting. Moreover, the activity of the CTRP12 promoter was determined using a dual‑luciferase reporter assay. The results demonstrated that I/R surgery markedly exacerbated myocardial tissue damage, whereas H/R treatment significantly reduced cell viability and significantly increased LDH activity as well as the release of inflammatory factors and apoptosis. I/R and H/R induction significantly reduced the expression levels of CTRP12 and KLF15. CTRP12 overexpression significantly alleviated H/R‑induced cell injury and significantly inhibited inflammation and apoptosis. Further analysis demonstrated that KLF15 could significantly promote the activity of the CTRP12 promoter. However, following CTRP12 knockdown, KLF15 overexpression exacerbated cell injury, inflammation and apoptosis. In conclusion, the present study demonstrated that CTRP12 may mitigate inflammation and apoptosis in H/R‑induced cardiomyocytes, possibly via the regulation of KLF15, which provided a theoretical basis for the potential treatment of I/R‑induced myocardial infarction.

Introduction

Myocardial infarction (MI) is an ischemic heart condition that poses a significant burden to society and may lead to death or disability (13). Myocardial ischemia-reperfusion (I/R) injury is considered as the main pathophysiology of ischemic heart diseases (4). It has been reported that acute and chronic cell death following MI affects cardiac function and patient prognosis (5,6). A previous study has reported that the apoptotic cascade in myocardial I/R is initiated either by mitochondrial damage and activation of caspase-9 or by death receptor ligation and activation of caspase-8 (7). Myocardial ischemia may induce inflammatory responses, while reperfusion may promote cell apoptosis, thus resulting in impaired myocardial structure and function (810). Therefore, inhibiting inflammatory responses and apoptosis following I/R could be considered as an effective strategy for preventing myocardial I/R injury.

C1q/TNF-related proteins (CTRPs) are a family of adipokines, similar to adiponectin, which exert numerous biological activities, including anti-atherosclerotic, insulin sensitivity, anti-inflammatory and vascular functions (11). CTRP12, also known as adipolin, is a member of the CTRP family, which is abundantly expressed in the fat tissue (12). It has previously been reported that CTRP12 is involved in the protection against metabolic disorders such as type 2 diabetes, atherosclerosis and obesity (1315). Furthermore, it has been suggested that CTRP12 may serve a critical role in cardiovascular injury (16). Another study has demonstrated that CTRP12 could alleviate lipopolysaccharide (LPS)-induced cardiomyocyte injury by preventing inflammation and apoptosis (17), which suggests that CTRP12 may be tightly linked with the cardiovascular system. However, its role in myocardial I/R injury has not been previously reported.

Krueppel-like factor 15 (KLF15), a member of the zinc-finger family of transcription factors, is tightly associated with several disorders, including inflammation, obesity and metabolic dysfunction (18). Emerging evidence has indicated that KLF15 enhances the activity of the CTRP12 promoter and regulates CTRP12 expression in adipocytes (19). Furthermore, another study reported that KLF15 attenuates hypoxia-induced apoptosis and oxidative stress in myocardial cells (20). Therefore, the aim of the present study was to evaluate the functional role of CTRP12 in myocardial I/R injury and explore the possible underlying molecular mechanisms of CTRP12 and KLF15.

Materials and methods

Animals

The present study was approved by the Animal Care and Use Committee of the Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center (Shenzhen, China) and conducted in accordance with Chinese legislation regarding animal experiments (21). A total of 30 c57BL6/J male mice (age, 8 weeks; weight, 18–22 g) were purchased from the Comparative Medicine Centre of Yangzhou University and housed under standard conditions of 25°C, with a relative humidity of 60%, in a 12-h light/dark cycle with free access to standard laboratory food and water.

Establishment of the myocardial I/R model

To establish the I/R mouse model, the left anterior descending coronary artery (LAD) was ligated (22). Mice were anesthetized using isoflurane (4% for induction and 2% for maintenance). Subsequently, a 6–0 silk suture slipknot was placed around the LAD and was released after 30 min. During the operation, mice were subcutaneously injected with 0.05 mg/kg buprenorphine hydrochloride as an intraoperative analgesic. Mice in the sham group underwent the same surgical procedure but without LAD ligation. Following the operation, 0.1 ml 5% glucose was subcutaneously injected into the mice to prevent dehydration. Following recovery from anesthesia, mice received buprenorphine hydrochloride (0.05 mg/kg subcutaneously) for post-operative analgesia (2326). Animals were sacrificed by cervical dislocation following anesthesia at 24 h following reperfusion and myocardial tissue samples were subsequently collected and stored at −80°C for further analysis.

Histopathological examination

Myocardial tissue specimens derived from mice who underwent myocardial I/R and sham surgery were collected and fixed using 4% paraformaldehyde for 24 h at room temperature. Subsequently, the tissues were dehydrated in different concentrations of ethanol, embedded in paraffin and sliced into 4-µm-thick sections. The tissue sections were then stained with hematoxylin (0.4%) and eosin (0.1%) (H&E) solution at 37°C for 5 min and the pathological changes after myocardial I/R injury were observed using a light microscope.

Cell culture and hypoxia-reoxygenation (H/R) treatment

The rat myoblast H9c2 cell line was obtained from the American Type Culture Collection. Cells were maintained in DMEM (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% FBS (Gibco; Thermo Fisher Scientific, Inc.) and 1% penicillin/streptomycin solution (Gibco; Thermo Fisher Scientific, Inc.) in a humidified atmosphere with 5% CO2 at 37°C. To establish an in vitro cardiomyocyte I/R model, H9c2 cells were cultured in glucose-free/serum-free DMEM and maintained in an incubator containing a gaseous mixture of 94% N2, 5% CO2 and 1% O2 at 37°C for 4 h. Following incubation, cells were transferred to 30 mM high-glucose DMEM, containing 10% FBS, was cultured under normoxic conditions for 4 h at 37°C for reoxygenation. H9c2 cells were not previously cultured under hypoxic conditions and these cells were cultured in high glucose DMEM containing 10% FBS under normoxic conditions and served as the control group.

Cell transfection

CTRP12- or KLF15-specific pcDNA overexpression plasmids [overexpressed (Ov)-CTRP12 or Ov-KLF15] and the corresponding negative control (NC; Ov-NC), CTRP12-specific small interfering (si) RNA-CTRP12 (siRNA-CTRP12) and the corresponding NC (siRNA-NC), were synthesized by Shanghai GenePharma Co., Ltd. The following siRNA sequences were used: CTRP12 forward (F), 5′-CUGUGACGGUAGACAAGAAGA-3′ and reverse (R), 5′-UUCUUGUCUACCGUCACAGGG-3′; and siRNA-NC F, 5′-GAUCAUACGUGCGAUCAGA-3′ and R, 5′-UCUGAUCGCACGUAUGAUC-3′. H9c2 cells (1×105 cells/well) were transfected with the recombinants (1 µg) using Lipofectamine® 2000 reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions for 6 h at room temperature. After 48 h cells underwent H/R injury.

Cell viability assay

The Cell Counting Kit-8 (CCK-8) assay was utilized to detect cell viability (27). Briefly, at 48 h following transfection, H9c2 cells were seeded into 96-well plates at a density of 5×103 cells/well and treated with H/R for 8 h (4 h hypoxia followed by 4 h reperfusion) at 37°C. Subsequently, each well was supplemented with 10 µl CCK-8 solution (Beyotime Institute of Biotechnology) and cells were incubated at 37°C for a further 2 h. The absorbance of each well was measured at a wavelength of 450 nm using a microplate reader (Bio-Rad Laboratories, Inc.).

Detection of lactate dehydrogenase (LDH) activity

H9c2 cells were seeded into 96-well plates at a density of 5×103 cells/well and cultured for 4 h of hypoxia and 4 h of reperfusion at 37°C. The cell supernatants were harvested using centrifugation at 1,000 × g for 15 min at 4°C to quantify the secretory levels of LDH using the LDH Assay Kit (cat. no. A020-2-2; Nanjing Jiancheng Bioengineering Institute) according to the manufacturer's protocol.

ELISA

The secretory levels of TNF-α and IL-6 were measured using the TNF-α Assay Kit (cat. no. H052-1; Nanjing Jiancheng Bioengineering Institute) and IL-6 Kit (cat. no. H007-1-1; Nanjing Jiancheng Bioengineering Institute) according to the manufacturer's protocol. IL-1β levels were detected using the Human IL-1β ELISA Kit (cat. no. ab214025; Abcam) according to the manufacturer's protocol. Culture supernatants from H9c2 cells (2×104 cells/well) were cultured at 37°C under 4 h of hypoxia and 4 h of reperfusion and were collected. Subsequently, 50 µl supernatant was added to each well. The optical density was detected at 450 nm using a microplate reader (BioTek Instruments, Inc.). A total of eight parallel wells were used for each ELISA.

Cell apoptosis

The TUNEL assay was performed to evaluate cell apoptosis (28). Briefly, following treatment, H9c2 cells (2×106 cells/well) were washed twice with PBS. Following fixation with 4% paraformaldehyde for 15 min at room temperature, cells were incubated with 15 µg/ml proteinase K for 15 min at 37°C and endogenous peroxidase activity was blocked using 3% H2O2 for 15 min at room temperature. After washing, cells were treated with TUNEL working solution for 60 min at 37°C, followed by co-labeling with DAPI (0.5 µg/ml) for 5 min at room temperature. The cells were mounted in Antifade Mounting Medium (Beyotime Institute of Biotechnology) and cells from five randomly selected fields of view were captured using a fluorescence microscope (Leica Microsystems GmbH). Apoptosis was quantified using ImageJ v1.8.0 software (National Institutes of Health).

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from murine myocardial tissues and H9c2 cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). Subsequently, RNA was reverse transcribed into first-strand cDNA using the PrimeScript RT Master Mix (Takara Bio, Inc.) according to the manufacturer's instructions. qPCR was performed using the SYBR Premix ExTaq Kit (Takara Bio, Inc.) using the ABI PRISM 7900 Real-Time System (Applied Biosystems; Thermo Fisher Scientific, Inc.). The thermocycling conditions were as follows: initial denaturation at 95°C for 30 sec; 40 cycles of denaturation at 95°C for 5 sec, annealing at 60°C for 30 sec and extension at 72°C for 60 sec. The program was terminated via final extension at 72°C for 1 min and cooling at 40°C. The primer sequences used for qPCR were as follows: CTRP12 F, 5′-CCTGTCCTTGGGCCGATTTA-3′ and R, 5′-CAGGGACGTATGACGGTGAC-3′; KLF15 F, 5′-GGGATCGTGGAGGAGAGCCT-3′ and R, 5′-CCAGCTGAGAGCTGGCTACA-3′; and GAPDH F, 5′-GTCAAGGCTGAGAACGGGAA-3′ and R 5′-AAATGAGCCCCAGCCTTCTC-3′. The mRNA expression levels were quantified using the 2−ΔΔCq method and were normalized using the internal reference gene GAPDH (29).

Western blotting

Total protein was extracted from myocardial tissues and H9c2 cells using RIPA buffer (Auragene; Hunan Aijia Biotechnology Co., Ltd.). Total protein (30 µg protein/lane) was separated via SDS-PAGE (Bio-Rad Laboratories, Inc.) on a 10% gel. Separated protein was transferred onto a PVDF membrane (MilliporeSigma). Following blocking with 5% skimmed milk for 1 h at room temperature, the membranes were incubated overnight at 4°C with primary antibodies targeting CTRP12 (1:1,000; cat. no. ab177991), KLF15 (1:1,000; cat. no. ab2647), Bax (1:2,000; cat. no. ab182733), cleaved caspase-3 (1:500; cat. no. ab32042), cleaved poly (ADP-ribose) polymerase (PARP; 1:1,000; cat. no. ab32064), Bcl-2 (1:1,000; cat. no. ab194583) and GAPDH (1:3,000; cat. no. ab125247) (all purchased from Abcam). Subsequently, the membranes were washed four times with PBS and incubated with horseradish peroxidase-labeled goat anti-rabbit IgG secondary antibodies (1:3,000; cat. no. 7074S; Cell Signaling Technology, Inc.) for 1 h at room temperature. The protein bands were visualized using an Amersham ECL Prime Western Blotting Detection Reagent (Amersham; Cytiva) and analyzed using ImageJ software v1.8.0 (National Institutes of Health).

Bioinformatic analysis

The binding site between KLF15 and the CTRP12 promoter was analyzed using the JASPAR database 2018 (https://jaspar.genereg.net/). The JASPAR database is an open-access database containing manually curated, non-redundant transcription factor (TF) binding profiles for TFs across six taxonomic groups.

Dual-luciferase reporter assay

The activity of the CTRP12 promoter was evaluated using a dual-luciferase reporter assay (30). Briefly, the CTRP12 promoter covering predicted binding sites was cloned into the pGL3 basic plasmid (Shanghai Genomeditech Co., Ltd.) with the luciferase/Renilla reporter gene. The Ov-KLF15 vector or the empty vector and luciferase/Renilla plasmids were co-transfected into H9c2 cells (1×105 cells/well) using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) for 24 h at room temperature. Each well was then supplemented with 100 µl Renilla Luciferase Reporter Gene Assay Cell Lysis Buffer (Beyotime Institute of Biotechnology) and the lysed cells were centrifuged at 5,000 × g for 1 min at 4°C to collect the supernatants. Subsequently, the cells were supplemented with 20 µl lysis buffer and then with 100 µl 1X firefly and 1X Renilla luciferase reaction solutions (Wuhan Scithera Microbiological Technologies, Inc.) to detect firefly and Renilla luciferase activity, respectively. After 48 h, luciferase activity was measured using the Dual Luciferase Reporter System, purchased from Promega Corporation, according to the manufacturer's instructions. The results are presented as the ratio of luciferase to Renilla activity.

Statistical analysis

All data are presented as the mean ± SD. All statistical analyses were carried out using GraphPad Prism 6 software (GraphPad Software, Inc.) using unpaired two-tailed Student's t-tests or a one-way ANOVA followed by a Bonferroni's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

CTRP12 is downregulated in I/R-induced myocardial tissues and H/R-induced cardiomyocytes

To explore the role of CTRP12 in myocardial I/R injury, a myocardial I/R mouse model was established. H&E staining demonstrated that mice who underwent the sham operation exhibited normal myocardial tissue structure, whereas those in the I/R group displayed damaged myocardial fiber structure, broken vascular walls and hemocyte and inflammatory molecule infiltration in the myocardial tissue (Fig. 1A), indicating that the model was successfully established. Subsequently, the mRNA expression levels of CTRP12 were determined in myocardial tissue samples and H9c2 cardiomyocytes using RT-qPCR and western blotting. The mRNA and protein expression levels of CTRP12 were significantly decreased in both I/R-induced myocardial tissues and H/R-induced H9c2 cells compared with the corresponding control groups (Fig. 1B-E). These results suggested that the decreased expression of CTRP12 was associated with myocardial I/R injury.

CTRP12 overexpression alleviates H/R-induced cell injury in H9c2 cells

To uncover the biological role of CTRP12 in hypoxia-induced cardiomyocytes, H9c2 cells were transfected with Ov-CTRP12 to enhance CTRP12 expression. The transfection efficiency of Ov-CTRP12 was evaluated using RT-qPCR and it was demonstrated that the mRNA expression levels of CTRP12 were significantly elevated compared with the Ov-NC group (Fig. 2A). The results demonstrated that CTRP12 mRNA and protein expression levels were significantly downregulated under H/R conditions compared with the control group, which was significantly reversed following transfection with Ov-CTRP12 in H/R cells compared with the H/R only group (Fig. 2B and C). The CCK-8 assay was performed to measure the effects of CTRP12 overexpression on cell viability and the results demonstrated that H/R injury significantly reduced cell viability compared with the control. However, CTRP12 overexpression significantly restored the H/R-mediated reduced cell viability compared with the H/R group (Fig. 2D). Furthermore, the levels of LDH were measured to determine the severity of cardiomyocyte injury. The results demonstrated that LDH activity was significantly enhanced in hypoxic cardiomyocytes compared with the control, whereas CTRP12 overexpression significantly reversed this effect compared with the H/R group (Fig. 2E). The aforementioned findings indicated that CTRP12 overexpression exerted a protective role against H/R-induced myocardial cell injury.

CTRP12 overexpression relieves the secretion of inflammatory factors and apoptosis in H/R-induced cells

Subsequently, the present study aimed to investigate whether CTRP12 overexpression could potentially protect H9c2 cells from H/R-induced inflammation and apoptosis. The secretory levels of TNF-α, IL-1β and IL-6 were significantly elevated by H/R injury compared with the control, whereas their levels were significantly reduced following CTRP12 overexpression compared with the H/R group (Fig. 3A-C). Furthermore, the TUNEL assay results demonstrated that H/R treatment significantly promoted cardiomyocyte apoptosis compared with the control, whereas CTRP12 overexpression significantly prevented the effects of H/R induction on cardiomyocyte apoptosis compared with the H/R group (Fig. 3D and E). Moreover, western blotting demonstrated that compared to the control the protein expression levels of Bax, cleaved caspase-3 and cleaved PARP were significantly increased, whereas those of Bcl-2 were significantly decreased in H/R-treated cells. However, CTRP12 overexpression significantly reversed the effects of H/R exposure on the expression profile of the aforementioned proteins, compared with the H/R group (Fig. 3F). These results suggested that CTRP12 overexpression may ameliorate hypoxia-induced H9c2 cell injury via attenuating inflammation and apoptosis.

KLF15 enhances the activity of the CTRP12 promoter and regulates CTRP12 expression levels

To elucidate whether KLF15 was involved in the regulation of CTRP12 expression in myocardial I/R injury, the expression levels of KLF15 were determined in I/R-induced myocardial tissues and H/R-induced cardiomyocytes. The results demonstrated that the mRNA and protein expression levels of KLF15 were significantly reduced in both myocardial tissues and H9c2 cells compared with those in the untreated control groups (Fig. 4A-D). The transfection efficiency of Ov-KLF15 was assessed using RT-qPCR and the results demonstrated that that the KLF5 mRNA expression levels were significantly elevated in the Ov-KLF15 group (Fig. 4E). Subsequently, H/R-treated H9c2 cells were transfected with Ov-KLF15 to overexpress KLF15 and the results demonstrated that KLF15 mRNA expression in the Ov-KLF15 + H/R group was significantly increased compared with the Ov-NC + H/R group (Fig. 4F and G). The mRNA and protein expression levels of CTRP12 were significantly increased in KLF15-overexpressing H9c2 cells compared with the H/R group (Fig. 4H and I). The binding site of the KLF15 and CTRP12 promoter was predicted using the JASPAR database (Fig. 4J). The results of the dual-luciferase reporter assay demonstrated that the luciferase activity was significantly enhanced in cells transfected with Ov-KLF15 after exposure to H/R compared with the H/R group (Fig. 4K). These results therefore suggested that KLF15 overexpression may increase the promoter activity of CTRP12.

KLF15 overexpression attenuates H/R-induced H9c2 cell injury via regulation of CTRP12

Based on the aforementioned findings, the present study further investigated the effect of KLF15 on regulating CTRP12 expression levels in cardiomyocytes. The transfection efficiency of siRNA-CTRP12 was assessed using RT-qPCR and the results demonstrated that CTRP12 mRNA expression were significantly reduced in the siRNA-CTRP12 group compared with the siRNA-NC (Fig. 5A). Compared with the H/R group, KLF15 overexpression significantly rescued H/R-mediated reduced cell viability in H/R-treated H9c2 cells (Fig. 5B). Furthermore, CTRP12 silencing significantly reduced the effects of H/R on cell viability compared with the Ov-KLF15 + siRNA-NC + H/R group. LDH activity was also assessed to determine the degree of H9c2 cell injury and the results demonstrated that CTRP12 silencing significantly elevated LDH activity in H/R-treated H9c2 cells transfected with Ov-KLF15 compared with the Ov-KLF15 + siRNA-NC + H/R group (Fig. 5C). These results therefore supported the involvement of KLF15 in CTRP12-mediated H/R-induced cardiomyocyte injury.

KLF15 overexpression inhibits inflammation and apoptosis in H/R-induced H9c2 cells via regulation of CTRP12

The effects of KLF15 overexpression on H/R-induced inflammation and apoptosis were evaluated in CTRP12-silenced H9c2 cells. The secretory levels of TNF-α, IL-1β and IL-6 were significantly reduced by KLF15 overexpression compared with the H/R group, whereas this effect was significantly reversed following CTRP12 knockdown compared with the Ov-KLF15 + siRNA-NC + H/R group (Fig. 6A-C). Furthermore, the apoptotic rate of H/R-induced cells was significantly decreased following KLF15 overexpression compared with the H/R group, whereas CTRP12 silencing resulted in a significant increase in the apoptotic rate in H/R-treated H9c2 cells transfected with Ov-KLF15 compared with the Ov-KLF15 + siRNA-NC + H/R group (Fig. 6D and E). Furthermore, western blotting demonstrated that the protein expression levels of Bax, cleaved caspase-3 and cleaved PARP were significantly decreased, whereas Bcl-2 protein expression levels were significantly increased in the Ov-KLF15 + H/R group compared with the H/R group. However, transfection of H9c2 cells with si-CTRP12 significantly upregulated Bax, cleaved caspase-3 and cleaved PARP, and significantly downregulated Bcl-2 compared with the Ov-KLF15 + siRNA-NC + H/R group (Fig. 6F). These data suggested that KLF15 overexpression may attenuate H/R-induced inflammation and apoptosis in H9c2 cells via the regulation of CTRP12 expression.

Discussion

The present study demonstrated that the expression levels of CTRP12 could be modulated by cardiac I/R, while its overexpression exerted a protective effect against I/R injury via alleviating reperfusion-induced inflammation and apoptosis. Furthermore, mechanistic investigations demonstrated that CTRP12 exerted its therapeutic benefits via its KLF15-regulated enhanced promotor activity.

Myocardial I/R injury is involved in complicated pathophysiological mechanisms and is considered to be a significant cause of heart diseases, which makes it a severe global burden (31). Myocardial ischemia may lead to hypoxia, thus resulting in cell injury and increased cytotoxicity, which in turn can promote cell apoptosis and oxidative stress (32,33). In the present study, a myocardial I/R mouse model was established after LAD ligation and reperfusion (34). H&E staining demonstrated typical myocardial injury, characterized by damaged myocardial fiber structure, broken vascular walls and hemocyte infiltration. Furthermore, H9c2 cells were treated with H/R to construct an in vitro myocardial I/R model. Subsequently, the mRNA and protein expression levels of CTRP12 in I/R-exposed myocardial tissues and H/R-induced cardiomyocytes was determined. The results demonstrated that CTRP12 was significantly downregulated both in vivo and in vitro models. CCK-8 assays and the determination of LDH activity are sensitive tools for evaluating myocardial cell viability and injury, respectively (35). In the present study, following CTRP12 overexpression, cell viability was significantly improved and LDH activity was significantly reduced, supporting the hypothesis that CTRP12 has a protective effect against I/R-induced cardiomyocyte injury, which is consistent with previous studies (17,36).

It has also been reported that CTRP12 regulates inflammation, glucose metabolism, vascular remodeling and cardiac fibrosis (15,37,38). Zhou et al (17) demonstrated that CTRP12 ameliorated LPS-induced inflammatory responses and cell apoptosis in cardiomyocytes in a nuclear factor erythroid 2-related factor 2 (Nrf2)-dependent manner. Nrf2, a critical antioxidant gene in scavenging reactive oxygen species (ROS) and maintaining redox homeostasis, could mediate redox balance (3941). Moreover, Fadaei et al (16) reported that the serum levels of CTRP12 were decreased and negatively associated with TNF-α and IL-6 in patients with coronary artery disease. Consistent with previously published findings (36), the results of the present study demonstrated that the secretory levels of TNF-α, IL-1β and IL-6 were significantly increased in cell culture supernatants from H/R-treated cells, whereas CTRP12 overexpression significantly abrogated this effect. These findings indicated that CTRP12 overexpression may protect cardiomyocytes against H/R-induced inflammation.

Myocardial cell apoptosis contributes to cardiomyocyte loss in ischemic heart diseases (42). A previous study suggested that cell apoptosis is a crucial ‘blasting fuse’ for hypoxia-induced cardiac ischemic injury (43). Therefore, in the present study a TUNEL staining assay and western blotting were carried out to assess the effects of CTRP12 overexpression on H/R-induced cardiomyocyte apoptosis. The results demonstrated that CTRP12 significantly attenuated cardiomyocyte apoptosis induced by H/R, accompanied by the significant upregulation of the anti-apoptotic protein Bcl-2 and significantly decreased expression of the pro-apoptotic proteins, Bax, cleaved caspase-3 and cleaved PARP. The aforementioned findings suggested that CTRP12 exhibited an inhibitory effect on the apoptosis of cardiomyocytes with H/R injury. However, it was also demonstrated that cell apoptosis is not completely reversed by CTRP12 silencing. These results therefore suggested that CTRP12 silencing may reverse cell apoptosis in part and the degree of reversal is associated with numerous factors such as concentration of transfection reagent, transfection efficiency, cell type and culture time.

It has previously been reported that the zinc finger transcription factor KLF15 serves a significant role in heart diseases, including pathological forms of left ventricular hypertrophy and heart failure (44,45). Furthermore, it has been proposed that KLF15 protects cardiomyocytes via attenuating hypertrophic remodeling, regulating cardiomyocyte gene expression and lipid oxidation and inhibiting cardiomyocyte apoptosis and oxidative stress (20,45,46). In the present study the mRNA and protein expression levels of KLF15 were significantly downregulated in both I/R-exposed myocardial tissues ad H/R-induced cardiomyocytes. Moreover, the results determined that KLF15 overexpression significantly promoted the mRNA and protein expression of CTRP12, which therefore indicated that KLF15 may have exerted a regulatory role on CTRP12 expression. Furthermore, the dual-luciferase reporter assay was performed and demonstrated KLF15 overexpression increased CTRP12 promoter activity. It was therefore hypothesized that KLF15 regulated CTRP12 expression at the transcriptional level by transcriptionally activating the CTRP12 promoter. Rescue experiments demonstrated that KLF15 overexpression significantly relieved cardiomyocyte cell viability and ameliorated H/R-induced cardiomyocyte inflammation and apoptosis. However, CTRP12 silencing significantly reversed the effects of KLF15 overexpression on H/R-mediated cardiomyocyte injury. These data provided direct evidence to suggest that KLF15 may be involved in the CTRP12-mediated protection of cardiomyocytes against H/R injury.

There are several limitations of the present study. First, due to the aim of the present study investigating the biological role of CTRP12 in myocardial I/R injury, the expression levels of other CTRPs were not explored in the in vivo and in vitro models. Furthermore, although the relationship between CTRP12 and KLF15 was verified in the present study, whether other factors are related to the pathological mechanism in myocardial I/R injury is unclear. Moreover, whether the KLF15/CTRP12 signaling pathway affects other apoptotic proteins by endoplasmic reticulum stress-related or mitochondria-related apoptotic signaling is unknown. To the best of our knowledge, there are no studies that have reported the association between other CTRPs and KLFs, especially in myocardial I/R injury. Therefore, it is necessary to further study the associations of other CTRPs and KLFs in myocardial I/R injury and identify the downstream genes of the KLF15/CTRP12 signaling pathway in myocardial I/R injury. Second, even though the expression changes of CTRP12 and KLF15 in I/R tissues were explored, localization studies of CTRP and KLF15 proteins were not performed and the expression level of KLF15 in CTRP12-overexpressed H/R cells was not investigated. In the present study KLF15 overexpression and its effects were investigated; however, the role of downregulated KLF15 was not and therefore the expression of the KLF15 inhibitor microRNA-223-3p will be investigated to assess the effects of KLF15 silencing both in vivo and in vitro. Future work will also explore more signaling pathways that are potentially controlled by KLF15 in myocardial I/R injury. Third, in the present study, it was demonstrated that KLF15 and KLF15-mediated CRTP12 significantly regulated the production of inflammatory cytokines, but the mechanism which decreases inflammatory cytokine levels by CTRP12 was not explored. Fourth the interaction between the CTRP12 promotor and KLF15 was preliminarily verified using the dual-luciferase reporter assay but needs to be further confirmed. Finally, both KLF15 and CTRP12 expression should be considered following hypoxia or reperfusion alone, in order to evaluate the effect of reperfusion-induced ROS. Furthermore, the effects of CTRP12 and KLF15 on different parts of the heart, such as the infarct, the border zone and on the volume of infarcted heart, were not explored, as well as the effects of the time and degree of I/R and H/R treatment on the expression and role of CTRP12 and KLF15. These issues are important and will be investigated in future studies.

In conclusion, the present study demonstrated that CTRP12 may potentially exert a protective effect against H/R-induced H9c2 cell injury via attenuating inflammation and apoptosis. These effects were regulated by KLF15. Overall, the aforementioned findings suggested that CTRP12 may serve as a novel target for treating ischemic heart diseases.

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

BL and XT designed the study, performed the experiments, wrote and revised the manuscript. BL analyzed the datasets. XT searched the literature. Both authors read and approved the final manuscript. BL and XT confirm the authenticity of all the raw data.

Ethics approval and consent to participate

This study was approved by the Animal Care and Use Committee of Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center (Shenzhen, China; approval no. 2020-630).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Reed GW, Rossi JE and Cannon CP: Acute myocardial infarction. Lancet. 389:197–210. 2017. View Article : Google Scholar : PubMed/NCBI

2 

White HD and Chew DP: Acute myocardial infarction. Lancet. 372:570–584. 2008. 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 

Wang Z, Yu J, Wu J, Qi F, Wang H, Wang Z and Xu Z: Scutellarin protects cardiomyocyte ischemia-reperfusion injury by reducing apoptosis and oxidative stress. Life Sci. 157:200–207. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Ibanez B, Fuster V, Jiménez-Borreguero J and Badimon JJ: Lethal myocardial reperfusion injury: A necessary evil? Int J Cardiol. 151:3–11. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Prech M, Marszałek A, Schröder J, Filas V, Lesiak M, Jemielity M, Araszkiewicz A and Grajek S: Apoptosis as a mechanism for the elimination of cardiomyocytes after acute myocardial infarction. Am J Cardiol. 105:1240–1245. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Scarabelli TM, Stephanou A, Pasini E, Comini L, Raddino R, Knight RA and Latchman DS: Different signaling pathways induce apoptosis in endothelial cells and cardiac myocytes during ischemia/reperfusion injury. Circ Res. 90:745–748. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Anselmi A, Abbate A, Girola F, Nasso G, Biondi-Zoccai GG, Possati G and Gaudino M: Myocardial ischemia, stunning, inflammation, and apoptosis during cardiac surgery: A review of evidence. Eur J Cardiothorac Surg. 25:304–311. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Hoffman JW Jr, Gilbert TB, Poston RS and Silldorff EP: Myocardial reperfusion injury: Etiology, mechanisms, and therapies. J Extra Corpor Technol. 36:391–411. 2004.PubMed/NCBI

10 

Al-Salam S and Hashmi S: Myocardial ischemia reperfusion injury: Apoptotic, inflammatory and oxidative stress role of galectin-3. Cell Physiol Biochem. 50:1123–1139. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Kawano J and Arora R: The role of adiponectin in obesity, diabetes, and cardiovascular disease. J Cardiometab Syndr. 4:44–49. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Omidifar A, Toolabi K, Rahimipour A, Emamgholipour S and Shanaki M: The gene expression of CTRP12 but not CTRP13 is upregulated in both visceral and subcutaneous adipose tissue of obese subjects. Diabetes Metab Syndr. 13:2593–2599. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Du J, Xu J, Wang X, Liu Y, Zhao X and Zhang H: Reduced serum CTRP12 levels in type 2 diabetes are associated with renal dysfunction. Int Urol Nephrol. 52:2321–2327. 2020. View Article : Google Scholar : PubMed/NCBI

14 

Wang G, Chen JJ, Deng WY, Ren K, Yin SH and Yu XH: CTRP12 ameliorates atherosclerosis by promoting cholesterol efflux and inhibiting inflammatory response via the miR-155-5p/LXRα pathway. Cell Death Dis. 12:2542021. View Article : Google Scholar : PubMed/NCBI

15 

Enomoto T, Ohashi K, Shibata R, Higuchi A, Maruyama S, Izumiya Y, Walsh K, Murohara T and Ouchi N: Adipolin/C1qdc2/CTRP12 protein functions as an adipokine that improves glucose metabolism. J Biol Chem. 286:34552–34558. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Fadaei R, Moradi N, Kazemi T, Chamani E, Azdaki N, Moezibady SA, Shahmohamadnejad S and Fallah S: Decreased serum levels of CTRP12/adipolin in patients with coronary artery disease in relation to inflammatory cytokines and insulin resistance. Cytokine. 113:326–331. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Zhou MQ, Jin E, Wu J, Ren F, Yang YZ and Duan DD: CTRP12 ameliorated lipopolysaccharide-induced cardiomyocyte injury. Chem Pharm Bull (Tokyo). 68:133–139. 2020. View Article : Google Scholar : PubMed/NCBI

18 

McConnell BB and Yang VW: Mammalian Krüppel-like factors in health and diseases. Physiol Rev. 90:1337–1381. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Enomoto T, Ohashi K, Shibata R, Kambara T, Uemura Y, Yuasa D, Kataoka Y, Miyabe M, Matsuo K, Joki Y, et al: Transcriptional regulation of an insulin-sensitizing adipokine adipolin/CTRP12 in adipocytes by Krüppel-like factor 15. PLoS One. 8:e831832013. View Article : Google Scholar : PubMed/NCBI

20 

Tang Q, Li MY, Su YF, Fu J, Zou ZY, Wang Y and Li SN: Absence of miR-223-3p ameliorates hypoxia-induced injury through repressing cardiomyocyte apoptosis and oxidative stress by targeting KLF15. Eur J Pharmacol. 841:67–74. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Bayne K, Ramachandra GS, Rivera EA and Wang JF: The evolution of animal welfare and the 3Rs in Brazil, China, and India. J Am Assoc Lab Anim Sci. 54:181–191. 2015.PubMed/NCBI

22 

Cao X, Li B, Han X, Zhang X, Dang M, Wang H, Du F, Zeng X and Guo C: Soluble receptor for advanced glycation end-products promotes angiogenesis through activation of STAT3 in myocardial ischemia/reperfusion injury. Apoptosis. 25:341–353. 2020. View Article : Google Scholar : PubMed/NCBI

23 

Roughan JV and Flecknell PA: Buprenorphine: A reappraisal of its antinociceptive effects and therapeutic use in alleviating post-operative pain in animals. Lab Anim. 36:322–343. 2002. View Article : Google Scholar : PubMed/NCBI

24 

Haubner BJ, Adamowicz-Brice M, Khadayate S, Tiefenthaler V, Metzler B, Aitman T and Penninger JM: Complete cardiac regeneration in a mouse model of myocardial infarction. Aging (Albany NY). 4:966–977. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Haubner BJ, Schuetz T and Penninger JM: A reproducible protocol for neonatal ischemic injury and cardiac regeneration in neonatal mice. Basic Res Cardiol. 111:642016. View Article : Google Scholar : PubMed/NCBI

26 

Ding M, Chen Y, Luan H, Zhang X, Zhao Z and Wu Y: Dexmedetomidine reduces inflammation in traumatic brain injury by regulating the inflammatory responses of macrophages and splenocytes. Exp Ther Med. 18:2323–2331. 2019.PubMed/NCBI

27 

Zhang C, Liang R, Gan X, Yang X, Chen L and Jian J: MicroRNA-384-5p/beclin-1 as potential indicators for epigallocatechin gallate against cardiomyocytes ischemia reperfusion injury by inhibiting autophagy via PI3K/Akt pathway. Drug Des Devel Ther. 13:3607–3623. 2019. View Article : Google Scholar : PubMed/NCBI

28 

Shen S, He F, Cheng C, Xu B and Sheng J: Uric acid aggravates myocardial ischemia-reperfusion injury via ROS/NLRP3 pyroptosis pathway. Biomed Pharmacother. 133:1109902021. View Article : Google Scholar : PubMed/NCBI

29 

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

30 

Shi Y, Liu M, Huang Y, Zhang J and Yin L: Promotion of cell autophagy and apoptosis in cervical cancer by inhibition of long noncoding RNA LINC00511 via transcription factor RXRA-regulated PLD1. J Cell Physiol. 235:6592–6604. 2020. View Article : Google Scholar : PubMed/NCBI

31 

Cadenas S: ROS and redox signaling in myocardial ischemia-reperfusion injury and cardioprotection. Free Radic Biol Med. 117:76–89. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Gao X, Zhang H, Zhuang W, Yuan G, Sun T, Jiang X, Zhou Z, Yuan H, Zhang Z and Dong H: PEDF and PEDF-derived peptide 44mer protect cardiomyocytes against hypoxia-induced apoptosis and necroptosis via anti-oxidative effect. Sci Rep. 4:56372014. View Article : Google Scholar : PubMed/NCBI

33 

Jing L, Li Q, He L, Sun W, Jia Z and Ma H: Protective effect of tempol against hypoxia-induced oxidative stress and apoptosis in H9c2 cells. Med Sci Monit Basic Res. 23:159–165. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Huang ZQ, Xu W, Wu JL, Lu X and Chen XM: MicroRNA-374a protects against myocardial ischemia-reperfusion injury in mice by targeting the MAPK6 pathway. Life Sci. 232:1166192019. View Article : Google Scholar : PubMed/NCBI

35 

Maisch B, Seferović PM, Ristić AD, Erbel R, Rienmüller R, Adler Y, Tomkowski WZ, Thiene G and Yacoub MH; Task Force on the Diagnosis and Management of Pricardial Diseases of the European Society of Cardiology, : Guidelines on the diagnosis and management of pericardial diseases executive summary; The Task force on the diagnosis and management of pericardial diseases of the European society of cardiology. Eur Heart J. 25:587–610. 2004. View Article : Google Scholar : PubMed/NCBI

36 

Jin AP, Zhang QR, Yang CL, Ye S, Cheng HJ and Zheng YY: Up-regulation of CTRP12 ameliorates hypoxia/re-oxygenation-induced cardiomyocyte injury by inhibiting apoptosis, oxidative stress, and inflammation via the enhancement of Nrf2 signaling. Hum Exp Toxicol. 40:2087–2098. 2021. View Article : Google Scholar : PubMed/NCBI

37 

Ogawa H, Ohashi K, Ito M, Shibata R, Kanemura N, Yuasa D, Kambara T, Matsuo K, Hayakawa S, Hiramatsu-Ito M, et al: Adipolin/CTRP12 protects against pathological vascular remodelling through suppression of smooth muscle cell growth and macrophage inflammatory response. Cardiovasc Res. 116:237–249. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Wang X, Huang T and Xie H: CTRP12 alleviates isoproterenol induced cardiac fibrosis via inhibiting the activation of P38 pathway. Chem Pharm Bull (Tokyo). 69:178–184. 2021. View Article : Google Scholar : PubMed/NCBI

39 

Kovac S, Angelova PR, Holmström KM, Zhang Y, Dinkova-Kostova AT and Abramov AY: Nrf2 regulates ROS production by mitochondria and NADPH oxidase. Biochim Biophys Acta. 1850:794–801. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Schmidlin CJ, Dodson MB, Madhavan L and Zhang DD: Redox regulation by NRF2 in aging and disease. Free Radic Biol Med. 134:702–707. 2019. View Article : Google Scholar : PubMed/NCBI

41 

Abrescia P, Treppiccione L, Rossi M and Bergamo P: Modulatory role of dietary polyunsaturated fatty acids in Nrf2-mediated redox homeostasis. Prog Lipid Res. 80:1010662020. View Article : Google Scholar : PubMed/NCBI

42 

Krijnen PA, Nijmeijer R, Meijer CJ, Visser CA, Hack CE and Niessen HW: Apoptosis in myocardial ischaemia and infarction. J Clin Pathol. 55:801–811. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Zhou Q, Peng X, Liu X, Chen L, Xiong Q, Shen Y, Xie J, Xu Z, Huang L, Hu J, et al: FAT10 attenuates hypoxia-induced cardiomyocyte apoptosis by stabilizing caveolin-3. J Mol Cell Cardiol. 116:115–124. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Leenders JJ, Wijnen WJ, Hiller M, van der Made I, Lentink V, van Leeuwen REW, Herias V, Pokharel S, Heymans S, de Windt LJ, et al: Regulation of cardiac gene expression by KLF15, a repressor of myocardin activity. J Biol Chem. 285:27449–27456. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Haldar SM, Lu Y, Jeyaraj D, Kawanami D, Cui Y, Eapen SJ, Hao C, Li Y, Doughman YQ, Watanabe M, et al: Klf15 deficiency is a molecular link between heart failure and aortic aneurysm formation. Sci Transl Med. 2:26ra262010. View Article : Google Scholar : PubMed/NCBI

46 

Noack C, Haupt LP, Zimmermann WH, Streckfuss-Bömeke K and Zelarayán LC: Generation of a KLF15 homozygous knockout human embryonic stem cell line using paired CRISPR/Cas9n, and human cardiomyocytes derivation. Stem Cell Res. 23:127–131. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2022
Volume 26 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
Liao B and Liao B: CTRP12 alleviates cardiomyocyte ischemia‑reperfusion injury via regulation of KLF15. Mol Med Rep 26: 247, 2022
APA
Liao, B., & Liao, B. (2022). CTRP12 alleviates cardiomyocyte ischemia‑reperfusion injury via regulation of KLF15. Molecular Medicine Reports, 26, 247. https://doi.org/10.3892/mmr.2022.12763
MLA
Liao, B., Tian, X."CTRP12 alleviates cardiomyocyte ischemia‑reperfusion injury via regulation of KLF15". Molecular Medicine Reports 26.1 (2022): 247.
Chicago
Liao, B., Tian, X."CTRP12 alleviates cardiomyocyte ischemia‑reperfusion injury via regulation of KLF15". Molecular Medicine Reports 26, no. 1 (2022): 247. https://doi.org/10.3892/mmr.2022.12763