Korean Circ J. 2024;54:e30. Forthcoming. English.
Published online Mar 25, 2024.
Copyright © 2024. The Korean Society of Cardiology
Original Article

LncRNA PART1 Attenuates Myocardial Ischemia-Reperfusion Injury by Regulating TFAP2C/DUSP5 Axis via miR-302a-3p

Min Zeng, MD,1,* Xin Wei, MD,2,* Jinchao Zhou, MM,3 and Siqi Luo, MM3
    • 1Medical Care Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China.
    • 2Department of Otorhinolaryngology Head and Neck Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China.
    • 3Hainan Medical University, Haikou, China.
Received May 16, 2023; Revised January 10, 2024; Accepted February 13, 2024.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted noncommercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Author's summary

Preventing myocardial ischemia-reperfusion injury (MIRI) is essential for treatment of ischemic heart diseases. However, the molecular mechanisms of MIRI are far from understood. Here, we explored long non-coding RNA prostate androgen regulated transcript 1 (PART1)’ role on MIRI. We found PART1 was downregulated in hypoxia/reoxygenation (H/R)-treated AC16 cells. PART1 overexpression protected cardiomyocytes against H/R-triggered autophagy and apoptosis. Mechanistically, PART1 induced transcription factor activating enhancer-binding protein 2C (TFAP2C) expression by targeting miR-302a-3p, TFAP2C bond to dual-specificity phosphatase 5 (DUSP5) promoter to upregulate DUSP5. Functional analyses demonstrated PART1 exhibited cardioprotective effects through the miR-302a-3p/TFAP2C/DUSP5 axis, representing a potential target for MIRI treatment.

Graphical Abstract

Abstract

Background and Objectives

Myocardial ischemia-reperfusion injury (MIRI) refers to the damage of cardiac function caused by restoration of blood flow perfusion in ischemic myocardium. However, long non-coding RNA prostate androgen regulated transcript 1 (PART1)’s role in MIRI remain unclear.

Methods

Immunofluorescence detected LC3 expression. Intermolecular relationships were verified by dual luciferase reporter assay. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, flow cytometry and transferase-mediated dUTP nick-end labeling (TUNEL) assays analyzed cell viability and apoptosis. The release of lactate dehydrogenase was tested via enzyme-linked immunosorbent assay (ELISA). Left anterior descending coronary artery surgery induced a MIRI mouse model. Infarct area was detected by 2,3,5-triphenyltetrazolium chloride staining. Hematoxylin and eosin staining examined myocardial injury. ELISA evaluated myocardial marker (creatine kinase MB) level.

Results

PART1 was decreased in H/R induced AC16 cells and MIRI mice. PART1 upregulation attenuated the increased levels of Bax, beclin-1 and the ratio of LC3II/I, and enhanced the decrease of Bcl-2 and p62 expression in H/R-treated cells. PART1 upregulation alleviated H/R-triggered autophagy and apoptosis via miR-302a-3p. Mechanically, PART1 targeted miR-302a-3p to upregulate transcription factor activating enhancer-binding protein 2C (TFAP2C). TFAP2C silencing reversed the protected effects of miR-302a-3p inhibitor on H/R treated AC16 cells. We further established TFAP2C combined to dual-specificity phosphatase 5 (DUSP5) promoter and activated DUSP5. TFAP2C upregulation suppressed H/R-stimulated autophagy and apoptosis through upregulating DUSP5. Overexpressed PART1 reduced myocardial infarction area and attenuated MIRI in mice.

Conclusion

PART1 improved the autophagy and apoptosis in H/R-exposed AC16 cells through miR-302a-3p/TFAP2C/DUSP5 axis, which might provide novel targets for MIRI treatment.

Keywords
LncRNA; Ischemia; Reperfusion

INTRODUCTION

Ischemic heart disease (IHD) is the most common cardiovascular-related disease and an important cause of death in patients with cardiovascular disease.1) In recent years, the morbidity and mortality of IHD are still rising and it is a serious threat to human health. Early reperfusion treatment can achieve the highest efficacy to rescue the injured myocardium. However, reperfusion therapy can also lead to cardiometabolic and structural damage, called myocardial ischemia-reperfusion injury (MIRI).2) MIRI can cause lethal arrhythmias, sudden cardiac death or heart failure. Autophagy is related to the pathophysiological process of MIRI. Aberrant autophagy represents a major factor causing ischemia/reperfusion (I/R) myocardial cell death.3) Therefore, exploring the molecular mechanism of cardiomyocyte autophagy after MIRI is important for shedding more lights on MIRI intervention.

Long non-coding RNAs (lncRNAs) are a class of non-coding RNAs that do not encode proteins and whose transcripts exceed 200 nucleotides. Various lncRNAs have been found to be abnormally expressed in I/R-induced myocardial injury, such as lncRNA OIP5-AS1.4) LncRNA prostate androgen regulated transcript 1 (PART1), as a novel lncRNA, was decreased in I/R heart and hypoxia/reoxygenation (H/R) cardiomyocytes, and depleted PART1 could aggravate cardiomyocyte apoptosis and mitochondrial damage,5) suggesting that PART1 may play a protective role in MIRI. However, whether PART1 affects MIRI by regulating autophagy remains to be further explored.

MicroRNAs (miRNAs) are a kind of small endogenous non coding RNAs with the length of 18–22 nucleotides, which play an important role in regulating different biological processes of many diseases. I/R-induced myocardial injury is associated with a variety of miRNAs. For example, miR-451-3p attenuated autophagy and apoptosis to improved MIRI.6) Recently, miR-302a-3p was reported to be increased in I/R induced mice, and miR-302a-3p knockdown could inhibit I/R-mediated apoptosis and mitochondrial damage,7) indicating that miR-302a-3p inhibition may be a therapeutic strategy for MIRI. StarBase analysis showed that there were the binding sites between PART1 and miR-302a-3p, but the regulatory mechanism between them has not been reported.

Transcription factor activating enhancer-binding protein 2C (TFAP2C), a member of AP2 transcription factor family, controls specific expression of target genes and is involved in various cancers.8) Evidence showed that TFAP2C expression was decreased in tissues with myocardial septal injury.9) Meanwhile, we found that miR-302a-3p and TFAP2C had the bind sites. Dual-specificity phosphatase 5 (DUSP5) is the endogenous phosphatase of extracellular signal regulated kinase 1/2 (ERK1/2).10) DUSP5 could repress the occurrence of autophagy in RAW264.7 cells by inhibiting the phosphorylation of signaling molecules in ERK1/2 signaling cascade.10) Additionally, we previously reported that the expression of DUSP5 was significantly reduced in MIRI.11) Besides, we found that there were the binding sites between TFAP2C and the promoter of DUSP5 through JASPAR. Nevertheless, the relationships between these molecular have not been investigated.

Based on the aforementioned evidences, we hypothesized that PART1 might sponge miR-302a-3p to regulate TFAP2C expression, thereby transcriptionally upregulating DUSP5 level, thus inhibiting H/R-induced cardiomyocyte autophagy and apoptosis. Our study may provide novel molecular targets for MIRI treatment.

METHODS

Ethical statement

All the animal procedures were performed in accordance with the Guiding Principles in the Use and Care of Animals and was authorized by the Animal Ethics Committee of Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University) (Med-Eth-Re[2022] 750).

Cell culture and treatment

Human AC16 cells were purchased from Cell Resource Center of Shanghai Institutes for Biological Sciences (SIBS, Shanghai, China) and cultured in Dulbecco’s Modified Eagle Medium (Corning, Bedford, MA, USA) with 10% fetal bovine serum (Gibco, Grand Island, NY, USA) and 1% Penicillin-Streptomycin (Sigma, St. Louis, MO, USA) at 37°C and 5% CO2. For H/R treatment, cells (1×106/well) were plated on 6-well plates and incubated in a hypoxic incubator (5% CO2, 94% N2, 1% O2) for 1, 2, 4, 6 hours. Subsequently, cells were put into an atmosphere of 21% O2 and 5% CO2 for 6 hours. Control cells were cultured normally (in vitro experimental cell processing timeline can be seen in Supplementary Figure 1).

Cell transfection

Short hairpin RNAs against TFAP2C (sh-TFAP2C), DUSP5 (sh-DUSP5), miR-302a-3p (mimics and inhibitor) and negative controls (NCs) were purchased from GeneChem (Shanghai, China). The empty vector and plasmids containing TFAP2C (overexpression plasmids of TFAP2C; oe-TFAP2C) were synthesized by Sangon Biotech (Shanghai, China). Cells were transfected with the above vectors utilizing Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA) and harvested after 24 hours. The lentivirus for overexpressing PART1 and the control lentivirus were obtained from GenePharma Co., Ltd. (Shanghai, China). AC16 cells were infected with oe-PART1 or oe-NC in enhanced infection solution supplemented with polybrene according to the manufacturer’s instructions. A stable cell line of AC16 cells with PART1 overexpression was constructed after screening with puromycin for 2 weeks.

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay

Cells (1×103/well) were inoculated into 96-well plates, followed by designated treatment for 24 hours. Later, every well was introduced with 5 mg/mL MTT solution (10 μL; Sigma) for 4 hours incubation under 37°C, followed by adding dimethyl sulfoxide (150 μL/well, 10 minutes). Optical density (OD) values were identified with the microplate reader (Perkin Elmer, Waltham, MA, USA) at 570 nm.

Flow cytometry

Cells were resuspended within binding buffer at 1×106 cells/mL, followed by 15 minutes staining using Annexin V-fluorescein isothiocyanate (FITC) (5 μL) according to the cell apoptosis assay kit (BD Biosciences, San Jose, CA, USA). Later, cell suspension was introduced with propidium iodide (10 μL) in dark for 15 minutes. The FACScan flow cytometry (BD Biosciences) was conducted to examine stained cells. FlowJo 7.6 software (TreeStar, Ashland, OR, USA) was adopted for data analysis.

Immunofluorescence

Cells on coverslips were fixed with 4% paraformaldehyde for 30 min, permeabilized with 0.1% Triton X-100, and blocked with 5% bovine serum albumin for 1 hour, followed by incubation with LC3 antibody (#4108, 1:500; Cell Signaling Technology, Danvers, MA, USA) at 4°C overnight. After washing with phosphate-buffered saline, cells were stained with FITC-labeled goat anti-rabbit IgG (ab6717, 1:1,000; Abcam, Cambridge, MA, USA) at 37°C for 1 hour. Cells were observed utilizing a confocal microscopy (Olympus, Tokyo, Japan).

Quantitative real-time polymerase chain reaction (RT-qPCR)

TRIzol reagent (Invitrogen) was utilized to separate total RNAs from cells. For mRNA, RNA was reverse transcribed to cDNA with PrimeScript RT Reagent Kit (Invitrogen). For miRNA, cDNA was synthesized with PrimeScript miRNA cDNA Synthesis Kit (Takara, Dalian, China). qPCR was conducted on ABI 7500 system (Applied Biosystems, Foster City, CA, USA) with SYBR Premix ExTaq II (Takara). The following specific primers were used: PART1 F: 5′-GGACTCGT GCTTCTCGTACGCTGG-3′, R: 5′-GCCTGCCCTTTGGTTTCTGGGAC-3′; miR-302a-3p F: 5′-ACACUCCAGCUGGGAGUGGUUUUGUACCUUC-3′, R: 5′-CUCAACUGGUGUCGUGGAGUCGGCAAUUCAGUUGAGUCGUGAAU-3′; TFAP2C F: 5'-ACAGGATCCATGTTGTGGAAAATAACCGAT-3', R: 5'-ATACTCGAGTTTCCTGTGTTTCTCCATTTT-3'; DUSP5 F: 5′-TCAGCCAGTGTGGAAAACCAG-3′, R: 5′-AGGCACTTCCAAGGTAGAGGA-3′; glyceraldehyde 3-phosphate dehydrogenase (GAPDH) F: 5′-CCAGGTGGTCTCCTCTGA-3′, R: 5′-GCTGTAGCCAAATCGTTGT-3′; U6 F: 5′-CTCGCTTCGGCAGCACA-3′, R: 5′-AACGCTTCACGAATTTGCGT-3′. GAPDH or U6 was used as endogenous controls. Data were calculated utilizing 2−∆∆Ct method.

Western blot

We prepared protein lysates using RIPA lysis buffer. Protein concentration was tested with BCA Protein Quantitation Kit (Beyotime, Shanghai, China). Protein aliquots were separated through 10–12% sodium dodecyl-sulfate polyacrylamide gel electrophoresis and transferred onto polyvinylidene fluoride membranes (Millipore, Boston, MA, USA), which were blocked using 5% defatted milk (Bio-Rad, Hercules, CA, USA) under ambient temperature for 1 hour and incubated using primary antibodies overnight under 4°C: TFAP2C (AV38284, 1:1,000; Sigma), DUSP5 (ab200708; 1:1,000; Abcam), p62 (ab56416, 1:1,000; Abcam), LC3 (#4108, 1:1,000; Cell Signaling Technology), Beclin1 (ab210498, 1:1,000; Abcam), Bax (ab182733, 1:2,000; Abcam), Bcl-2 (MA5-11757, 1:1,000; Invitrogen), GAPDH (ab9485, 1:1,000; Abcam), followed by incubation with horseradish peroxidase (HRP)-labeled secondary antibody (#7074, 1:1,000; Cell Signaling Technology) for 1 hour at room temperature. Signals were visualized using SuperSignal Pico PLUS chemiluminescent substrate (Pierce, Rockford, IL, USA). GAPDH served as a loading control. Data analysis was conducted using ImageJ software (NIH, Bethesda, MD, USA).

Dual luciferase reporter assay

The wild-type PART1 (PART1-WT) or mutant PART1 (PART1-MUT), which contained miR-302a-3p binding site, were inserted in a dual-luciferase reporter vector (pRL-TK) by Promega (Madison, WI, USA). Besides, the sequences of wild (TFAP2C-WT) and mutant type (TFAP2C-MUT) of TFAP2C 3’untranslated region (UTR) containing miR-302a-3p binding sites were synthesized and cloned into pRL-TK vector (Promega). The above constructs were co-transfected into cells with miR-302a-3p mimics or mimics NC by Lipofectamine 2000 (Invitrogen).

To verify the interaction between DUSP5 and TFAP2C, sequences of DUSP5, including wild-type or mutant binding sites of TFAP2C, were cloned into pGL3 vector (Promega). Cells were co-transfected with vector or OE-TFAP2C and DUSP5-WT or DUSP5-MUT using Lipofectamine 2000 (Invitrogen). After 48 hours, cells were subjected to luciferase assays (Promega).

Lactate dehydrogenase (LDH) assay

A enzyme-linked immunosorbent assay (ELISA) kit (Abcam) was utilized to evaluate LDH levels. Briefly, cell media was collected and cleared to remove cell debris. Samples were enzymically reacted with substrates in 96-well plates. Reacted plates were determined with a microplate reader at 450 nm (Bio-Rad).

A mouse model of myocardial ischemia-reperfusion injury

The left anterior descending coronary artery (LAD) was used to establish a MIRI mouse model.12) Male C57BL/6 mice (Shanghai Animal Center, Shanghai, China; 20–25 g, n=40, 8–10 weeks) were subjected to 12-hour light and dark cycles and the temperature was maintained at 20–25°C. Mice were randomly divided into 4 groups (n=5/group, we conducted two batches of model construction, one for pathological detection and the other for RNA and protein isolation and identification): control group, I/R group, I/R+vector group, and I/R+oe-PART1 group. Briefly, mice were anesthetized by intraperitoneal injection of 3.5% chloral hydrate (10 mL/kg). After a left thoracotomy between the fourth and fifth ribs, the LAD was seen and ligated with a 6-0 prolene suture. Ischemia is confirmed by visual examination of the discoloration of the occlusive distal myocardium. After 30 minutes of ischemia, the ligation was removed. The sham group used the same method of thoracotomy without ligation. In the process of I/R modeling, an injection of lentivirus particles (1×108 PFU/mL, 100 μL) carrying oe-PART1 or vector was administered into the left ventricle, followed by clamping of the aorta for a duration of 10 seconds. The viral transfection into the myocardium was achieved by delivering the virus through the coronary artery, taking advantage of cardiac contraction.13)

2,3,5-Triphenyltetrazolium chloride (TTC) staining

Measurement of infarct volume was conducted as previously described.14) Briefly, myocardial tissues from mice were sliced into sections (1-mm-thick coronal sections), and stained with a 2% solution of TTC (Sigma) for 20 minutes at 37°C, followed by fixation with 4% paraformaldehyde. Afterwards, TTC stained sections were imaged using a digital camera and analyzed by Image J (NIH).

Histopathological examination (hematoxylin and eosin [H&E] and transferase-mediated dUTP nick-end labeling [TUNEL] staining)

Paraformaldehyde (4%) was adopted to fix the hearts from mice. Myocardial tissues were dehydrated and embedded in paraffin. The 4-um sections were stained with H&E staining and then analyzed utilizing light microscopy. Terminal deoxynucleotidyl TUNEL method with Onestep TUNEL Apoptosis Assay Kit (Beyotime) was used to identify apoptotic cells in myocardial sections according to the manufacturer’s instructions. TUNEL-positive nuclei were identified using a microscopy, and apoptosis rate of cardiomyocytes was calculated.

Immunohistochemistry (IHC)

Mouse myocardial tissue sections were initially subjected to a 2-hour baking process at 55°C in an oven. Subsequently, deparaffinization and rehydration steps were performed, followed by antigen retrieval using sodium citrate. Immunostaining was then conducted on the samples using anti-cleaved caspase3 (#9661, 1:1,000; Cell Signaling Technology) antibody at 4°C overnight. Afterward, sections were incubated with a secondary antibody labeled with HRP for 1 hour. Diaminobenzidine (DAB; MXB Biotechnologies, Fuzhou, China) was utilized for section development. Finally, sections were counterstained with hematoxylin and examined utilizing a microscope (Olympus).

ELISA assay

The serum of mice was collected after centrifugation, and serum concentration of creatine kinase MB (CK-MB) was detected utilizing an ELISA kit (Biosciences) according to manufacturer’s instructions.

Statistical analysis

Data were analyzed using GraphPad Prism 8 and expressed as mean ± standard deviation (SD) of 3 independent biological replicates and technical replicates in vitro and 5 mice/per group in vivo. The Kolmogorov-Smirnov test was used to determine the normality of the distribution of the data in each group, and the in vitro data conforms to a normal distribution. Therefore, all in vitro data are analyzed using parametric analysis, differences between two groups were calculated utilizing Student’s t-test. One-way analysis of variance was conducted for multiple comparisons. While in vivo data were analyzed using non-parametric analysis, differences between groups using Mann-Whitney test. The p value <0.05 was taken as statistically significant.

RESULTS

Overexpression of PART1 inhibited H/R-induced cardiomyocyte autophagy and apoptosis

Firstly, we used MTT assay to detect the cell viability at different hypoxia time points. As shown in Figure 1A, cell viability was about 50% at the time point of 4 hours. At this time, cells had both certain damage and certain activity. Therefore, this hypoxia time was used for subsequent experiments. Afterwards, AC16 cells were transfected with oe-PART1 and overexpressed PART1 significantly promoted PART1 expression compared with NC group (Figure 1B). Next, the transfected AC16 cells were cultured under H/R condition to establish a MIRI model in vitro. Compared to control cells, H/R treatment inhibited PART1 expression, but PART1 upregulation reversed the effect of H/R on PART1 expression (Figure 1C). Additionally, H/R treatment greatly repressed cell viability compared with control group, whereas overexpressed PART1 induced cell viability (Figure 1D). Moreover, the secretion of LDH was increased by H/R treatment, while overexpression of PART1 reversed the induction of H/R on LDH (Figure 1E). Flow cytometry and TUNEL staining showed that H/R stimulation enhanced cell apoptosis, but oe-PART1 inhibited cell apoptosis (Figure 1F and G). Meanwhile, H/R treatment increased Bax expression, while decreased Bcl-2 level. However, PART1 upregulation overturned these effects (Figure 1H). Immunofluorescence analysis revealed that fluorescence of LC3 in cells with H/R treatment was elevated, but fluorescence intensity of LC3 was reduced after overexpression of PART1 (Figure 1I). Furthermore, H/R treatment enhanced beclin-1 expression and the ratio of LC3II/I, but decreased p62 expression. Nevertheless, overexpressed PART1 showed the opposite effects (Figure 1J). Therefore, PART1 upregulation alleviated H/R-stimulated cardiomyocyte autophagy and apoptosis.

Figure 1
Overexpression of PART1 inhibited autophagy and apoptosis in H/R-challenged cardiomyocytes. (A) MTT assay detected cell viability at different hypoxia time points. (B) PART1 transfection efficiency was verified by RT-qPCR. (C) RT-qPCR detection of PART1 level after overexpression of PART1 in H/R-treated AC16 cells. (D) MTT tested cell viability. (E) ELISA tested LDH concentration. (F) Cells apoptosis was detected utilizing Flow cytometry. (G) TUNEL staining detected apoptosis. (H) Bcl-2 and Bax expressions were measured using Western blot. (I) Immunofluorescence detection of LC3 expression in AC16 cells. (J) Western blot detected p62, LC3II/I, Beclin-1 levels. Data were shown as mean ± SD based on three independent experiments (n=3).
ELISA = enzyme-linked immunosorbent assay; H/R = hypoxia/reoxygenation; LDH = lactate dehydrogenase; MTT = 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; oe- = overexpression plasmids; PART1 = prostate androgen regulated transcript 1; RT-qPCR = quantitative real-time polymerase chain reaction; SD = standard deviation; TUNEL = transferase-mediated dUTP nick-end labeling.

*p<0.05, **p<0.01, ***p<0.001.

PART1 targeted miR-302a-3p to upregulate TFAP2C expression

Next, bioinformatics tool StarBase predicted binding site of miR-302a-3p with PART1 or TFAP2C (Figure 2A). Dual luciferase reporter assay demonstrated that luciferase activities were reduced by co-transfection with PART1-WT or TFAP2C-WT with miR-302a-3p mimics in AC16 cells, whereas luciferase activities were not changed in PART1-MUT and TFAP2C-MUT groups (Figure 2B). Furthermore, overexpression of PART1 inhibited miR-302a-3p expression, but enhanced TFAP2C expression (Figure 2C-E). Additionally, compared to NC group, miR-302a-3p upregulation promoted its expression but inhibited TFAP2C expression, while miR-302a-3p downregulation had the opposite effects (Figure 2F-H). Therefore, PART1 interacted with miR-302a-3p to increase TFAP2C expression in AC16 cells.

Figure 2
PART1 targeted miR-302a-3p and upregulated TFAP2C level. (A) StarBase predicted binding site of miR-302a-3p with PART1 or TFAP2C. (B) Luciferase activity of PART1-WT/MUT (left panel) or TFAP2C-WT/MUT (right panel) in AC16 cells treated with miR-302a-3p mimics or mimics NC. (C, D) RT-qPCR assessed miR-302a-3p (C) and TFAP2C (D) levels after overexpression of PART1 in AC16 cells. (E) Western blot analyzed TFAP2C protein level. (F, G) RT-qPCR detected miR-302a-3p (F) and TFAP2C (G) after miR-302a-3p overexpression or inhibition in AC16 cells. (H) Western blot tested the protein level of TFAP2C. Data were shown as mean ± SD based on three independent experiments (n=3).
MUT = mutant type; NC = negative control; PART1 = prostate androgen regulated transcript 1; RT-qPCR = quantitative real-time polymerase chain reaction; SD = standard deviation; TFAP2C = transcription factor activating enhancer-binding protein 2C; WT = wild type.

*p<0.05, **p<0.01, ***p<0.001.

PART1 participated in H/R-induced cardiomyocyte autophagy and apoptosis by regulating miR-302a-3p

To explore the mechanism of PART1 in H/R-triggered cardiomyocyte autophagy and apoptosis, AC16 cells were transfected with oe-PART1, miR-302a-3p mimics and NCs, and then cultured under H/R condition. Compared to control group, miR-302a-3p was elevated by H/R treatment. Although PART1 overexpression suppressed miR-302a-3p expression, this effect was abolished by miR-302a-3p upregulation (Figure 3A). H/R treatment inhibited cell viability, while overexpression of PART1 increased cell viability, but co-transfection of miR-302a-3p mimics inhibited cell viability (Figure 3B). Moreover, LDH release and cell apoptosis were suppressed by PART1 upregulation in H/R-challenged cells, and they were recovered by co-transfection with miR-302a-3p mimics (Figure 3C-E). Additionally, after H/R treatment, miR-302a-3p upregulation ameliorated the inhibition of oe-PART1 on Bax expression, decreased the promotion of oe-PART1 on Bcl-2 expression (Figure 3F). H/R treatment enhanced LC3 fluorescence intensity, Beclin-1 expression and LC3II/I ratio, and decreased p62 expression, while overexpression of PART1 decreased LC3 fluorescence intensity, Beclin-1 expression and LC3 II/I ratio, increased p62 level. However, miR-302a-3p upregulation reversed the regulation of PART1 on autophagy-related proteins (Figure 3G and H). In short, PART1 regulated H/R-triggered cell autophagy and apoptosis through inhibiting miR-302a-3p.

Figure 3
PART1 participated in H/R-mediated cardiomyocyte function injury via regulating miR-302a-3p. (A) RT-qPCR determined miR-302a-3p level. (B) MTT tested cell viability. (C) ELISA tested LDH concentration. (D) Flow cytometry analyzed cell apoptosis. (E) TUNEL staining detected apoptosis. (F) Western blot tested Bcl-2 and Bax levels. (G) Immunofluorescence measured LC3 expression in AC16 cells. (H) Western blot detected the protein levels of p62, LC3II/I, Beclin-1. Data were shown as mean ± SD based on three independent experiments (n=3).
ELISA = enzyme-linked immunosorbent assay; LDH = lactate dehydrogenase; MTT = 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; PART1 = prostate androgen regulated transcript 1; RT-qPCR = quantitative real-time polymerase chain reaction; SD = standard deviation; TUNEL = transferase-mediated dUTP nick-end labeling.

*p<0.05, **p<0.01, ***p<0.001.

Knockdown of miR-302a-3p alleviated H/R-triggered cardiomyocyte autophagy and apoptosis through TFAP2C

We next depleted TFAP2C in AC16 cells and found that sh-TFAP2C significantly reduced TFAP2C expression compared with NC group (Figure 4A). Next, AC16 cells were transfected with miR-302a-3p inhibitor, sh-TFAP2C and NCs, and then cultured under H/R condition. MiR-302a-3p inhibition eliminated the inhibitory effect of H/R treatment on cell viability, whereas co-treatment of sh-TFAP2C and miR-302a-3p inhibitor could reverse the effect of miR-302a-3p inhibition on cell viability (Figure 4B). Additionally, miR-302a-3p inhibition reduced LDH release and apoptosis in H/R-exposed cells, whereas co-transfection of sh-TFAP2C and miR-302a-3p inhibitor had the opposite effect (Figure 4C-E). Furthermore, in H/R-challenged cells, the level of Bax was reduced and Bcl-2 was increased by miR-302a-3p depletion, while co-treatment of sh-TFAP2C and miR-302a-3p inhibitor reversed the effect of miR-302a-3p knockdown (Figure 4F). Moreover, miR-302a-3p knockdown mediated-decrease of LC3 fluorescence, Beclin-1 expression and LC3II/I ratio, and increased p62 level were abrogated by sh-TFAP2C (Figure 4G and H). All in all, miR-302a-3p knockdown attenuated H/R-mediated cell autophagy and apoptosis via regulating TFAP2C in AC16 cells.

Figure 4
MiR-302a-3p inhibitor attenuated H/R-triggered cardiomyocyte autophagy and apoptosis through TFAP2C. (A) TFAP2C transfection efficiency was verified by RT-qPCR and Western blot. (B) MTT tested cell viability. (C) ELISA tested LDH concentration. (D) Flow cytometry determined apoptosis. (E) TUNEL staining detected apoptosis. (F) Western blot analyzed Bcl-2 and Bax expression levels. (G) Immunofluorescence detected LC3 expression in AC16 cells. (H) Western blot measured p62, LC3II/I, Beclin-1 levels. Data were shown as mean ± SD based on three independent experiments (n=3).
ELISA = enzyme-linked immunosorbent assay; H/R = hypoxia/reoxygenation; LDH = lactate dehydrogenase; MTT = 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; RT-qPCR = quantitative real-time polymerase chain reaction; SD = standard deviation; TFAP2C = transcription factor activating enhancer-binding protein 2C; TUNEL = transferase-mediated dUTP nick-end labeling.

*p<0.05, **p<0.01, ***p<0.001.

TFAP2C bound to DUSP5 promoter and upregulated DUSP5 expression

Furthermore, we found the binding site between TFAP2C and DUSP5 by bioinformatics tool JASPAR (Figure 5A). Dual luciferase reporter assay revealed that the fluorescence value of DUSP5 wild-type group increased in AC16 cells with oe-TFAP2C, while that of DUSP5 mutant group did not change significantly (Figure 5B). Additionally, compared to control group, overexpression of TFAP2C significantly promoted the expression of TFAP2C and DUSP5 (Figure 5C and D). Therefore, TFAP2C induced DUSP5 expression by binding with DUSP5 promoter.

Figure 5
TFAP2C bound to DUSP5 promoter and upregulated DUSP5. (A) Binding site between TFAP2C and DUSP5 by JASPAR. (B) Dual luciferase reporter assay verified the binding of TFAP2C to DUSP5 promoter. (C, D) RT-qPCR and Western blot detected the expression of TFAP2C and DUSP5 after overexpression of TFAP2C. Data were shown as mean ± SD based on three independent experiments (n=3).
DUSP5 = dual-specificity phosphatase 5; RT-qPCR = quantitative real-time polymerase chain reaction; SD = standard deviation; TFAP2C = transcription factor activating enhancer-binding protein 2C.

*p<0.05, **p<0.01, ***p<0.001.

TFAP2C upregulation suppressed H/R-induced cardiomyocyte autophagy and apoptosis through upregulating DUSP5

To explore the molecular mechanism of the protective role of TFAP2C in MIRI. We overexpressed TFAP2C and downregulated DUSP5 in AC16 cells, and then cultured under H/R condition. H/R treatment inhibited DUSP5 expression compared with control group. When TFAP2C was overexpressed, DUSP5 level was significantly increased, which could be rescued by co-transfecting sh-DUSP5 (Figure 6A). Meanwhile, cell viability was elevated in response to TFAP2C overexpression in H/R-treated cells, while co-transfection of oe-TFAP2C and sh-DUSP5 ameliorated this effect (Figure 6B). Moreover, TFAP2C upregulation induced a reduction in LDH release and apoptosis in H/R-induced cells that was completely abolished by sh-DUSP5 (Figure 6C-E). Additionally, in H/R-exposed cells, overexpression of TFAP2C downregulated Bax and upregulated Bcl-2, these effects were reversed after co-transfection with sh-DUSP5 (Figure 6F). In addition, oe-TFAP2C transfection reduced LC3 fluorescence, Beclin-1 expression and LC3 II/I ratio, and induced p62 level in H/R-treated cells. However, these effects were overturned by sh-DUSP5 (Figure 6G and H). Taken together, TFAP2C inhibited H/R-treated cell autophagy and apoptosis via DUSP5.

Figure 6
TFAP2C upregulation suppressed H/R-challenged cardiomyocyte autophagy and apoptosis through regulating DUSP5. (A) RT-qPCR and Western blot analysis of DUSP5 expression. (B) MTT tested cell viability. (C) ELISA tested LDH concentration. (D) Flow cytometry detected apoptosis. (E) TUNEL staining detected apoptosis. (F) Western blot analyzed Bcl-2 and Bax expression levels. (G) Immunofluorescence detected LC3 in AC16 cells. (H) Western blot detected p62, LC3II/I, Beclin-1 levels. Data were shown as mean ± SD based on three independent experiments (n=3).
DUSP5 = dual-specificity phosphatase 5; ELISA = enzyme-linked immunosorbent assay; H/R = hypoxia/reoxygenation; LDH = lactate dehydrogenase; MTT = 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; RT-qPCR = quantitative real-time polymerase chain reaction; SD = standard deviation; TFAP2C = transcription factor activating enhancer-binding protein 2C; TUNEL = transferase-mediated dUTP nick-end labeling.

*p<0.05, **p<0.01, ***p<0.001.

Enforced expression of PART1 relieved I/R-induced MIRI

To explore the effect of PART1 on MIRI in vivo, a mouse model of MIRI was established by LAD surgery. oe-PART1 vector was used to treat these mice. As shown in Figure 7A, it presents the in vitro model timeline of MIRI. TTC staining showed that myocardial infarct volume was increased in response to I/R treatment, while this effect was attenuated by PART1 upregulation (Figure 7B). H&E staining showed that I/R led to obvious damage to myocardial function, manifested as inflammatory infiltration, as well as swelling and necrosis of cardiomyocytes. However, oe-PART1 reduced severe myocardial damage (Figure 7C). ELISA assay showed that level of CK-MB in mouse serum was increased after I/R. PART1 upregulation reduced the secretion of CK-MB (Figure 7D). Furthermore, mRNA expression of PART1 in mouse myocardial tissues was reduced in response to I/R treatment; oe-PART1 ameliorated this effect (Figure 7E). TUNEL staining discovered that cardiomyocyte apoptosis increased after I/R in mouse myocardial tissues, but oe-PART1 inhibited the apoptosis (Figure 7F). IHC analysis further demonstrated that cleaved caspase3 was elevated in myocardial tissue of I/R mice, and which was inhibited by PART1 upregulation (Figure 7G). Moreover, I/R dramatically increased LC3 II/I ratio and Beclin-1 expression while decreased p62 level, however, forced PART1 expression abolished the dysregulation of these molecules (Figure 7H). Taken together, these findings revealed that oe-PART1 attenuated MIRI in mice.

Figure 7
Enforced expression of PART1 relieved I/R-induced MIRI. A mice model of MIRI was prepared and treated with a PART1 overexpression vector. (A) Mouse MIRI model construction timeline. (B) TTC staining detected myocardial infarct volume. (C) H&E staining detected myocardial histopathological changes in mice. (D) Blood level of myocardial marker CK-MB was measured by ELISA. (E) RT-qPCR detection of PART1 expression in mouse myocardial tissue. (F) TUNEL staining detected cardiomyocyte apoptosis. (G) Cleaved caspase3 expression in mouse myocardial tissue was assessed with IHC. (H) Western blot detected p62, LC3II/I, Beclin-1 levels. Data are the means ± SD for three independent experiments (n=5 mice/group).
CK-MB = creatine kinase MB; ELISA = enzyme-linked immunosorbent assay; H&E = hematoxylin and eosin; IHC = immunohistochemistry; I/R = ischemia/reperfusion; MIRI = myocardial ischemia-reperfusion injury; PART1 = prostate androgen regulated transcript 1; RT-qPCR = quantitative real-time polymerase chain reaction; SD = standard deviation; TTC = 2,3,5-Triphenyltetrazolium chloride; TUNEL = transferase-mediated dUTP nick-end labeling.

*p<0.05, **p<0.01, ***p<0.001.

DISCUSSION

MIRI is a common clinical complication in ischemic heart disease after treatment with recanalization. MIRI can induce myocardial remodeling including cardiomyocyte death (autophagy and apoptosis), cardiac hypertrophy, myocardial fibrosis and cardiac dysfunction.15) At present, the pathogenesis of MIRI is not fully defined, and there is still a lack of precise means of prevention and treatment. This study investigated the role of PART1 in MIRI. It was revealed that PART1 acted as a ceRNA of miR-302a-3p and inhibited H/R-triggered cardiomyocyte autophagy and apoptosis through mediating TFAP2C/DUSP5 pathway, providing a novel therapeutic target for MIRI treatment.

Evidence showed that lncRNAs were associated with autophagy and apoptosis of cardiomyocytes as well as regulation of vascular endothelial function during MIRI. For example, lncRNA cardiac autophagy inhibitory factor suppressed cardiac autophagy and attenuated myocardial infarction.16) LncRNA TUG1 aggravated cardiomyocyte apoptosis and MIRI in a mouse model.17) Therefore, lncRNAs are promising as novel targets for clinical treatment of MIRI. LncRNA PART1 acts as a carcinogenic factor in many cancers. A previous study indicated that PART1 suppressed mitochondrial apoptosis and alleviated MIRI via miR-503-5p/BIRC5 pathway.5) Meanwhile, PART1 overexpression inhibited the inhibition of H/R treatment on neonatal mice ventricle cells viability.5) Our results implied that PART1 was reduced in H/R-exposed AC16 cells. Moreover, PART1 overexpression significantly suppressed autophagy and apoptosis, and promoted cell viability in H/R-triggered cardiomyocytes. Our data highlighted therapeutic potential role of PART1 in MIRI.

MiR-302a-3p is differentially expressed in various diseases. Recently, miR-302a-3p modulated apoptosis related genes and participated in cardiomyocyte apoptosis and mitochondrial autophagy in mice after MIRI.7) Additionally, miR-302a-3p was involved in the regulation of cell apoptosis in left ventricular murine HL-1 cardiomyocytes under H/R treatment.18) Besides, miR-302a-3p was increased in H/R-treated rat cardiomyocytes and its inhibition attenuated cardiomyocyte apoptosis.19) LncRNAs act as sponges of miRNAs, which are key targets in regulating myocardial stress genes under physiological and pathological conditions and involved in MIRI. For example, knockdown of lncRNA AK139328 alleviated MIRI through directly regulating miR-204-3p and suppressing autophagy in diabetic mice.20) LncRNA highly up-regulated in liver cancer attenuated MIRI in rat models and apoptosis of H/R-induced cardiomyocytes by targeting miR-377-5p.21) In our work, PART1 was ascertained to directly bind to miR-302a-3p and remarkably reduced miR-302a-3p expression. Interestingly, the protective effects of PART1 were abolished when H/R-treated AC16 cells overexpressed miR-302a-3p, suggesting that miR-302a-3p may function as a downstream effector in PART1-mediated protective effects in MIRI.

TFAP2C has been proved to participate in the development of multiple cancers.22) Interestingly, TFAP2C has been shown to be decreased in tissues with myocardial septal injury.9) A previous study reported that miR-302a-3p inhibitor protected H/R-induced mouse cardiomyocytes apoptosis, suggesting inhibition of miR-302a-3p could promoted cell viability in H/R treatment cardiomyocytes.7) Additionally, after H/R treatment, miR-302a-3p was elevated in HK-2 cells, and down-regulation of miR-302a-3p limited the H/ R-induced NLRP3 inflammasome-mediated apoptosis.23) Accumulated evidence indicates that miRNAs govern their biological functions through target gene mRNAs.24) We further proved that miR-302a-3p could bind to TFAP2C and inhibited TFAP2C expression in AC16 cells. Functionally, we found that H/R treatment inhibited cell viability, while knockdown of miR-302a-3p increased cell viability. Studies have shown that the miRNA-mRNA axis has an important effect on MIRI.25) We further demonstrated that knockdown of TFAP2C could rescue the decreased of autophagy and apoptosis in H/R-stimulated AC16 cells caused by miR-302a-3p inhibition. DUSP5 is a nuclear phosphatase and is responsible for diverse biological processes.26) Recent data have proven that DUSP5 silencing increased autophagy-related proteins including LC3II and Beclin1 in Bacillus Calmette-Guérin-infected murine macrophages.10) We previously found that DUSP5 level was decreased during MIRI.11) In this study, we revealed that TFAP2C could increase DUSP5 expression, and TFAP2C overexpression suppressed H/R-triggered cardiomyocyte autophagy and apoptosis through regulating DUSP5 expression to against MIRI. Taken together, our results revealed an important function for TFAP2C in regulation of MIRI through DUSP5.

The principle of MTT assay is that succinate dehydrogenase in living cell mitochondria can reduce exogenous MTT to water-insoluble blue-purple crystal formazan and deposit in cells, while dead cells do not have this function, so cell viability can be quantified. At the same time, MTT assay is the result of a complex process, depending on many variables, including aging, necrosis, apoptosis and superoxide.27) Secondly, the principle of flow cytometry to detect apoptosis is to use fluorescently labeled phospholipid molecules and DNA dyes to detect the signal of phospholipid eversion and DNA fragments on the cell membrane by flow cytometry. Rapid and accurate detection of apoptosis.28) In addition, cell viability assays have proved insufficient in the past to determine whether apoptosis occurs in cancer cells.29) Additionally, when cells are damaged, LDH will be quickly released into the cell culture medium, which may be reflected in cell viability and apoptosis.30) Therefore, we use a variety of experimental methods to confirm our hypothesis and play a complementary role.

In summary, PART1 reduced cardiomyocyte autophagy and apoptosis, and exerted protective effects in H/R-stimulated cardiomyocytes via miR-302a-3p/TFAP2C/DUSP5 pathway. These results suggested that PART1 might be a novel therapeutic target for MIRI. However, there are some limitations in this study. In the H/R induced in vitro model, in addition to the commonly used cell lines of AC16, other cardiomyocyte models will also be an exploration direction of our subsequent research. Additionally, the lack of clinical data is also a main limitation of current study, which will be addressed in the future.

SUPPLEMENTARY MATERIALS

Supplementary Figure 1

The processing time of each experiment.

Click here to view.(495K, ppt)

Notes

Funding:This work was supported by Hainan Clinical Medical Research Center Project (LCYX202207, LCYX202305), Hainan Key R&D Plan Project (ZDYF2020118), Hainan general Hospital Clinical Innovation and Transformation Cultivation 550 Project (2022CXZH01) and Hainan Provincial Health Industry Research Project (22A200216).

Conflict of Interest:The authors have no financial conflicts of interest.

Data Sharing Statement:All data generated or analyzed during this study are included in this published article (and its supplementary information files).

Author Contributions:

  • Conceptualization: Zeng M.

  • Data curation: Zeng M.

  • Funding acquisition: Wei X.

  • Investigation: Wei X, Zhou J.

  • Methodology: Wei X, Zhou J, Luo S.

  • Project administration: Zhou J, Luo S.

  • Resources: Luo S.

  • Validation: Zeng M, Luo S.

  • Writing - original draft: Zeng M.

  • Writing - review & editing: Zeng M.

References

    1. Pagliaro BR, Cannata F, Stefanini GG, Bolognese L. Myocardial ischemia and coronary disease in heart failure. Heart Fail Rev 2020;25:53–65.
    1. Davidson SM, Ferdinandy P, Andreadou I, et al. Multitarget strategies to reduce myocardial ischemia/reperfusion injury: JACC review topic of the week. J Am Coll Cardiol 2019;73:89–99.
    1. Du J, Li Y, Zhao W. Autophagy and myocardial ischemia. Adv Exp Med Biol 2020;1207:217–222.
    1. Niu X, Pu S, Ling C, et al. lncRNA Oip5-as1 attenuates myocardial ischaemia/reperfusion injury by sponging miR-29a to activate the SIRT1/AMPK/PGC1α pathway. Cell Prolif 2020;53:e12818
    1. Guo Z, Zhao M, Jia G, Ma R, Li M. LncRNA PART1 alleviated myocardial ischemia/reperfusion injury via suppressing miR-503-5p/BIRC5 mediated mitochondrial apoptosis. Int J Cardiol 2021;338:176–184.
    1. Lv XW, He ZF, Zhu PP, Qin QY, Han YX, Xu TT. miR-451-3p alleviates myocardial ischemia/reperfusion injury by inhibiting MAP1LC3B-mediated autophagy. Inflamm Res 2021;70:1089–1100.
    1. Lv W, Jiang J, Li Y, Fu L, Meng F, Li J. MiR-302a-3p aggravates myocardial ischemia-reperfusion injury by suppressing mitophagy via targeting FOXO3. Exp Mol Pathol 2020;117:104522
    1. Gu Z, Zhou Y, Cao C, Wang X, Wu L, Ye Z. TFAP2C-mediated LINC00922 signaling underpins doxorubicin-resistant osteosarcoma. Biomed Pharmacother 2020;129:110363
    1. Hammer S, Toenjes M, Lange M, et al. Characterization of TBX20 in human hearts and its regulation by TFAP2. J Cell Biochem 2008;104:1022–1033.
    1. Luo J, Xue D, Song F, Liu X, Li W, Wang Y. DUSP5 (dual-specificity protein phosphatase 5) suppresses BCG-induced autophagy via ERK 1/2 signaling pathway. Mol Immunol 2020;126:101–109.
    1. Zeng M, Wei X, He YL, Chen JX, Lin WT, Xu WX. EGCG protects against myocardial I/RI by regulating lncRNA Gm4419-mediated epigenetic silencing of the DUSP5/ERK1/2 axis. Toxicol Appl Pharmacol 2021;433:115782
    1. Wang J, Hu X, Fu W, Xie J, Zhou X, Jiang H. Isoproterenol-mediated heme oxygenase-1 induction inhibits high mobility group box 1 protein release and protects against rat myocardial ischemia/reperfusion injury in vivo. Mol Med Rep 2014;9:1863–1868.
    1. Xu Z, Mo Y, Li X, et al. The novel lncRNA AK035396 drives cardiomyocyte apoptosis through Mterf1 in myocardial ischemia/reperfusion injury. Front Cell Dev Biol 2021;9:773381
    1. Xu S, Wu B, Zhong B, et al. Naringenin alleviates myocardial ischemia/reperfusion injury by regulating the nuclear factor-erythroid factor 2-related factor 2 (Nrf2) /System xc-/ glutathione peroxidase 4 (GPX4) axis to inhibit ferroptosis. Bioengineered 2021;12:10924–10934.
    1. Valikeserlis I, Athanasiou AA, Stakos D. Cellular mechanisms and pathways in myocardial reperfusion injury. Coron Artery Dis 2021;32:567–577.
    1. Liu CY, Zhang YH, Li RB, et al. LncRNA CAIF inhibits autophagy and attenuates myocardial infarction by blocking p53-mediated myocardin transcription. Nat Commun 2018;9:29.
    1. Fu D, Gao T, Liu M, et al. LncRNA TUG1 aggravates cardiomyocyte apoptosis and myocardial ischemia/reperfusion injury. Histol Histopathol 2021;36:1261–1272.
    1. Sun Y, Zhang Y, Ye Z, et al. circRNA-miRNA complex participates in the apoptosis of myocardial cells in myocardial ischemia/reperfusion injury. Discov Med 2022;33:13–26.
    1. Fang YC, Yeh CH. Inhibition of miR-302 suppresses hypoxia-reoxygenation-induced H9c2 cardiomyocyte death by regulating Mcl-1 expression. Oxid Med Cell Longev 2017;2017:7968905
    1. Yu SY, Dong B, Fang ZF, Hu XQ, Tang L, Zhou SH. Knockdown of lncRNA AK139328 alleviates myocardial ischaemia/reperfusion injury in diabetic mice via modulating miR-204-3p and inhibiting autophagy. J Cell Mol Med 2018;22:4886–4898.
    1. Liang H, Li F, Li H, Wang R, Du M. Overexpression of lncRNA HULC attenuates myocardial ischemia/reperfusion Injury in rat models and apoptosis of hypoxia/reoxygenation cardiomyocytes via targeting miR-377-5p through NLRP3/Caspase-1/IL-1β signaling pathway inhibition. Immunol Invest 2021;50:925–938.
    1. Jiang X, Guo S, Xu M, et al. TFAP2C-mediated lncRNA PCAT1 inhibits ferroptosis in docetaxel-resistant prostate cancer through c-Myc/miR-25-3p/SLC7A11 signaling. Front Oncol 2022;12:862015
    1. Bai T, Cui Y, Yang X, et al. miR-302a-3p targets FMR1 to regulate pyroptosis of renal tubular epithelial cells induced by hypoxia-reoxygenation injury. Exp Physiol 2021;106:2531–2541.
    1. Fabian MR, Sonenberg N, Filipowicz W. Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem 2010;79:351–379.
    1. Makkos A, Ágg B, Petrovich B, Varga ZV, Görbe A, Ferdinandy P. Systematic review and network analysis of microRNAs involved in cardioprotection against myocardial ischemia/reperfusion injury and infarction: involvement of redox signalling. Free Radic Biol Med 2021;172:237–251.
    1. Kutty RG, Talipov MR, Bongard RD, et al. Dual specificity phosphatase 5-substrate interaction: a mechanistic perspective. Compr Physiol 2017;7:1449–1461.
    1. Ghasemi M, Turnbull T, Sebastian S, Kempson I. The MTT assay: utility, limitations, pitfalls, and interpretation in bulk and single-cell analysis. Int J Mol Sci 2021;22:12827.
    1. Wallberg F, Tenev T, Meier P. Analysis of apoptosis and necroptosis by fluorescence-activated cell sorting. Cold Spring Harb Protoc 2016;2016:pdb.prot087387
    1. Brunelle JK, Zhang B. Apoptosis assays for quantifying the bioactivity of anticancer drug products. Drug Resist Updat 2010;13:172–179.
    1. Klein R, Nagy O, Tóthová C, Chovanová F. Clinical and diagnostic significance of lactate dehydrogenase and its isoenzymes in animals. Vet Med Int 2020;2020:5346483

Metrics
Share
Figures

1 / 7

PERMALINK