Startseite Medizin LncRNA EBLN3P inhibits myocardial inflammation and apoptosis after acute myocardial infarction via targeting miR-675-3p
Artikel Open Access

LncRNA EBLN3P inhibits myocardial inflammation and apoptosis after acute myocardial infarction via targeting miR-675-3p

  • Wanrong Bi ORCID logo EMAIL logo , Qing Peng ORCID logo , Yushi Feng ORCID logo und Jingjing Liu ORCID logo
Veröffentlicht/Copyright: 15. September 2025

Abstract

Objectives

Acute myocardial infarction (AMI) is myocardial necrosis caused by acute and persistent ischemia and hypoxia of the coronary arteries. This study aimed to investigate the potential mechanisms of lncRNA EBLN3P in the development of AMI.

Methods

This study recruited 109 patients with AMI and 117 healthy controls. The hypoxia/reoxygenation (H/R) cell model was primarily utilized to simulate in vivo ischemia-reperfusion injury of tissues and organs. The expression levels of EBLN3P were assessed using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The concentrations of TNF-α, IL-6, and IL-1β were quantified via enzyme-linked immunosorbent assay (ELISA). Apoptosis was evaluated using flow cytometry. Additionally, the dual-luciferase reporter assay was utilized to confirm the targeting relationship between EBLN3P and miR-675-3p.

Results

EBLN3P was significantly downregulated in the serum of AMI patients. The receiver operating characteristic (ROC) curve analysis demonstrated that EBLN3P exhibited high diagnostic accuracy for AMI. Hypoxia/reoxygenation (H/R) treatment led to a marked decrease in EBLN3P expression levels, which could be significantly restored by transfecting EBLN3P post H/R treatment. H/R treatment significantly elevated the levels of TNF-α, IL-1β, and IL-6, but EBLN3P transfection partially reversed this effect. Additionally, H/R treatment resulted in a significant increase in the apoptosis rate, which EBLN3P transfection markedly reduced following H/R. EBLN3P targeted and regulated miR-675-3p, with their expression levels being negatively correlated. Furthermore, under H/R conditions, co-expression of EBLN3P and miR-mimic partially alleviated the effects of EBLN3P on cell behavior.

Conclusions

EBLN3P was involved in the regulatory mechanism of AMI by targeting miR-675-3p.

Introduction

Acute myocardial infarction (AMI) is primarily caused by the rupture or erosion of atherosclerotic plaques, leading to acute thrombotic occlusion of the epicardial coronary arteries [1], 2]. AMI affects approximately three million individuals globally each year [3], with a notably high incidence and mortality rate in China [4]. Following AMI onset, an inflammatory response is initiated, characterized by the infiltration of inflammatory cells into the infarcted area and the release of numerous inflammatory mediators, such as interleukin-1 (IL-1). While these mediators can promote tissue repair to some extent, an excessive inflammatory response exacerbates myocardial damage and increases myocardial cell apoptosis [5]. Research indicates that reducing myocardial cell apoptosis is crucial for improving cardiac function and preventing adverse cardiac remodeling [6]. Additionally, ischemia-reperfusion injury plays a significant role in AMI pathophysiology. Upon reperfusion following coronary artery occlusion, a series of complex pathophysiological processes are triggered [7]. Recent advancements in medical technology have significantly extended the survival period of AMI patients. However, AMI remains a leading cause of mortality [8]. A deeper understanding of AMI mechanisms is essential for developing effective prevention and treatment strategies, which could positively impact patient outcomes and quality of life.

Long non-coding RNA (lncRNA) refers to a class of non-coding RNA molecules that exceed 200 nucleotides in length [9]. While lncRNAs do not directly encode proteins, they play crucial roles in various biological processes, including the regulation of gene expression, chromatin modification, and cell differentiation through interactions with DNA, RNA, or proteins [10]. LncRNAs typically regulate gene expression by interacting with microRNAs (miRNAs), thereby influencing cell apoptosis, and modulating various cellular biological processes [11], 12]. Both lncRNAs and miRNAs are critical regulators in numerous physiological activities, including cardiovascular diseases such as arrhythmia, myocardial hypertrophy, heart failure, and atrial fibrillation [13], 14]. A growing body of research has demonstrated that non-coding RNAs, including both miRNAs and lncRNAs, play pivotal roles in AMI pathogenesis [15], 16]. For instance, studies have shown that the expression profiles of specific lncRNAs in peripheral blood cells from AMI patients exhibit significant alterations [16], 17]. Notably, circulating lncRNA urothelial carcinoma associated 1 (UCA1) is downregulated within 3 days post-AMI onset [18]. Additionally, inhibition of miRNA-16 can prevent AMI progression by reversing the downregulation of β-adrenergic receptors [19], while miRNA-127–3p suppresses cardiomyocyte inflammation and mediates apoptosis following AMI [20]. Furthermore, lncRNA EBLN3P is classified within the long non-coding RNA family. Previous studies have demonstrated its influence on cell proliferation and invasion across multiple cancer types – including osteosarcoma [21], colorectal cancer [22], and cervical cancer cells [23]. Database analysis revealed that miR-675-3p was a target gene of EBLN3P. miR-675-3p, a microRNA regulates gene expression by binding to the 3′ untranslated region (3′ UTR) of target gene mRNA [24]. Research has shown that overexpression of miR-675-3p inhibits chondrocyte apoptosis and cartilage matrix degradation, promoting cell proliferation [25]. Notably, in atherosclerosis patients, serum levels of miR-675-3p are significantly elevated compared to healthy individuals [26]. And atherosclerosis serves as a critical pathological foundation for AMI [27]. Based on these findings, we hypothesized that the EBLN3P/miR-675-3p axis may play a role in the progression of AMI.

The primary objective of this study was to investigate the potential role of EBLN3P in AMI. Experimental evidence confirmed that miR-675-3p was the target gene of EBLN3P. The findings elucidated the significant roles of EBLN3P and miR-675-3p in the pathogenesis and progression of AMI, as well as their interrelationship. Both EBLN3P and miR-675-3p were implicated in the expression regulation of AMI-related cells and the modulation of pathological processes, offering crucial insights into the underlying mechanisms of AMI.

Materials and methods

Study objects

This study received approval from the Tongji Hospital Branch Affiliated to Tongji University Medical Ethics Committee (No.: 20,220,012, Date of approval: 07-03-2022). Additional, and all participants provided written informed consent. Additionally, all procedures adhered strictly to the principles outlined in the Helsinki Declaration.

This study recruited 109 patients with AMI and 117 healthy controls from Tongji Hospital Branch Affiliated to Tongji University from January 2022 to June 2024. All AMI patients met the predefined inclusion and exclusion criteria. The inclusion criteria were as follows: Participants aged 27–80 years. For inclusion, the diagnosis of AMI must be confirmed either at admission or within 24 h post-admission. Clinical manifestations: Typical chest pain symptoms include a compressive, oppressive, or constrictive pain located behind the sternum, potentially radiating to the left upper limb, neck, jaw, or back, lasting more than 30 min, and not fully relieved by rest or sublingual nitroglycerin administration. Atypical symptoms may present as dyspnea, nausea, vomiting, fatigue, or palpitations, necessitating objective examinations to exclude other differential diagnoses. Auxiliary examinations: Electrocardiogram (ECG) demonstrated characteristic changes indicative of myocardial ischemia or necrosis. Myocardial injury markers reveal elevated levels of cardiac troponin I (cTnI) and creatine kinase isoenzyme (CK-MB), consistent with the dynamic evolution pattern associated with myocardial infarction. Imaging evidence: Echocardiography identifies segmental wall motion abnormalities, while coronary angiography confirms at least one major coronary artery stenosis of ≥70 % or complete occlusion. Conformity with universal definition: The case satisfies criterion outlined in the 2018 revised “Universal Definition of Myocardial Infarction”.

The exclusion criteria were as follows: patients with a history of coronary stent implantation or coronary artery bypass grafting; chest pain attributable to non-myocardial infarction causes, such as pleurisy, costochondritis, pneumothorax, and other conditions; individuals with known cardiac diseases other than AMI, including stable angina pectoris, hypertrophic cardiomyopathy, and other cardiomyopathies; cases where systemic diseases could confound the diagnosis, such as acute cerebrovascular events, severe infections, shock, and other critical illnesses; patients with a history of infectious diseases or stroke; and those with hematological, immune-related disorders, or malignant tumors.

Inclusion criteria for healthy controls were as follows: age and gender matched to the AMI group, no history of cardiovascular diseases (e.g., AMI, angina pectoris, heart failure), no severe infections, autoimmune diseases, or malignant tumors, and no recent use of anti-inflammatory or immunomodulatory drugs. Exclusion criteria included: those with a history of infectious diseases or stroke, those with abnormal liver or renal function, and those with habits such as smoking or heavy drinking that could potentially interfere with inflammatory markers.

Five milliliters of blood were drawn from the cubital vein of the upper limb into a containing separation gel (for serum) (Sanli Medical Technology, Hunan, China) tube immediately after diagnosis of AMI upon admission or within 24 h post-admission. The blood sample was centrifuged within 15 min (3000 rpm) of collection to separate the serum, and the following parameters were measured: blood lipid indices (total cholesterol (TC), low-density lipoprotein (LDL), high-density lipoprotein (HDL), triglyceride (TG)), estimated glomerular filtration rate (eGFR), glycated hemoglobin (HbA1c), cardiac troponin I (cTnI), high-sensitivity C-reactive protein (hs-CRP), interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α). Selected inflammatory factors (e.g., TNF-α, IL-6) were quantified by ELISA on the same day, while the remaining serum was aliquoted and stored at −80 °C for subsequent analyses. RNA extraction was performed within one week of sample collection. The quality and concentration of the RNA were assessed using a NanoDrop spectrophotometer, with acceptable purity indicated by an A260/A280 ratio ranging from 1.8 to 2.0. Serum samples were maintained at −80 °C in a refrigerator to prevent repeated freeze-thaw cycles, thereby preserving the integrity and stability of RNA and proteins.

Cell culture and transfection

The human cardiomyocyte cell line AC16 (BeNa Culture Collection, BNCC339980) was obtained from the American Type Culture Collection (ATCC). The cells were maintained in DMEM medium (Solarbio, China), supplemented with 10–20 % fetal bovine serum (FBS) (Tianhang Biotechnology, China) and 1 % penicillin-streptomycin solution (double-antibiotic) to prevent microbial contamination. Cultivation was performed in a humidified incubator at 37 °C with 5 % CO2. When the cells reached 80–90 % confluence in the culture flask, they were passaged at a specific ratio into new flasks for continued growth.

The hypoxia/reoxygenation (H/R) cell model was primarily utilized to simulate in vivo ischemia-reperfusion injury of tissues and organs. Based on previously reported methods [28], 29]. We replicated the conditions of AMI in vitro by subjecting AC16 cells to hypoxia/reoxygenation treatment. Specifically, well-growing AC16 cells were placed in a serum-free DMEM medium within a hypoxia chamber (Billups-Rothenberg, Inc.). The chamber was sealed tightly, and a gas mixture (95 % N2, 5 % CO2, with residual O2 reduced to 1 %) was introduced to displace the oxygen, thereby establishing a hypoxic environment. The oxygen concentration was monitored by an oxygen sensor and maintained at≤1 %. Following an 18-h hypoxia period, the cells were transferred back to a standard incubator (37 °C, 5 % CO2, 95 % air) for a 6-h reoxygenation phase. Cells maintained under normal culture conditions served as the control group.

AC16 cells were seeded in 96-well plates at a density of 2 × 104 cells per well and cultured in an incubator at 37 °C with 5 % CO2. The coding sequence of EBLN3P was cloned into the pcDNA 3.1 vector (GenePharma, China) to construct the overexpression plasmid pcDNA-EBLN3P. Cells were divided into several groups: control, H/R, H/R + pcDNA3.1, H/R + pcDNA3.1-EBLN3P, and co-expression groups including H/R + pcDNA3.1-EBLN3P + mimic-NC, H/R + pcDNA3.1-EBLN3P + miR-mimic. The miR-675-3p mimic and its corresponding negative control (mimic-NC) were synthesized by MedChemExpress (catalog numbers: HY-R02064, China). The miR-675-3p-mimic was used to regulate the expression level of miR-675-3p. The concentration of the pcDNA3.1-EBLN3P plasmid was 1 μg/μL. The concentration of miR-675-3p-mimic was 50 nM. The sequence of the miR-675-3p mimic and the sequence of the mimics-NC were as follows: miRNA-675-3p-mimics sense: CUG​UAU​GCC​CUC​ACC​GCU​CA; miRNA-675-3p-mimics antisense: AGC​GGU​GAG​GGC​AUA​CAG​UU; mimics-NC: UUG​UAC​UAC​ACA​AAA​GUA​CUG. Cell transfection was performed using Lipofectamine 2000 (Thermo Fisher Scientific, USA). After 6 h of co-cultivation, the culture medium was replaced with a fresh medium. Transfection efficiency was assessed using RT-qPCR to confirm that the transfection efficiency in AC16 cells exceeded 80 %.

Quantitative real-time PCR (RT-qPCR)

Total RNA was extracted from serum or cell samples using the Trizol (Invitrogen, China) reagent, followed by reverse transcription into cDNA using the First-Strand cDNA Synthesis kit (A5001, Promega, USA). PT-qPCR was conducted based on the standard SYBR-Green PCR kit (4,367,659, Applied Biosystems, China) protocol on ABI 7600 (Applied Biosystems, China). U6 and GAPDH were used as internal controls for miR-675-3p and EBLN3P, respectively. For PCR, the reaction conditions were as follows: initial denaturation at 95 °C for 3 min, followed by 40 cycles consisting of denaturation at 95 °C for 10 s, annealing at 60 °C for 15 s, and extension at 72 °C for 30 s. The primer sequence was used in the RT-qPCR were as follows: EBLN3P forward, 5′-CAG​ACT​AAA​GGA​TCA​AGC​GAG​A-3′ and reverse 5′-ATC​AAT​TGC​CAC​AGG​TTG​AAG​A-3’; miR-675-3p forward 5′-GCC​GAG​CAT​CTT​ACC​GGA​CGT-3′ and reverse 5′-CTC​AAC​TGG​TGT​CGT​GGA-3’; U6 forward 5′-AAC​GCT​TCA​CGA​ATT​TGC​GT-3′ and reverse 5′-CTC​AAC​TGG​TGT​CGT​GGA-3’. GADPH forward: 5′-GGT​CGG​AGT​CAA​CGG​ATT​TG-3′ and reverse: 5′-ATG​AGC​CCC​AGC​CTT​CTC​CAT-3’. The relative mRNA expression levels of miR-675-3p and EBLN3P were quantified using the 2−ΔΔCt method.

Enzyme-linked immunosorbent assay (ELISA) was used to measure the inflammatory factors

The concentrations of TNF-α, IL-1β, and IL-6 in serum were measured using ELISA. Serum samples and protein standards were diluted and added to the wells. Following the addition of the stop solution, the absorbance was read at 450 nm. All procedures were conducted strictly according to the kit (FineTest, China)) instructions. TNF-α: EH0302 (Range: 15.625–1000 pg/mL (1 pg=76mIU); Sensitivity: 9.375 pg/mL; Intra-assay CV (%): 5.82; Inter-assay CV (%):5.62). IL-1β: EH0185 (Range: 3.906–250 pg/mL (1 pg=140mIU); Sensitivity: 2.344 pg/mL; Intra-assay CV (%): 4.58; Inter-assay CV (%): 4.60). IL-6: EH0201 (Range: 4.688–300 pg/mL (1 pg=110mIU); Sensitivity: 2.813 pg/mL; Intra-assay CV (%): 4.91; Inter-assay CV (%): 4.63). The standard curve was prepared in accordance with the instructions of each kit. Each experiment was repeated three times.

Dual-luciferase reporter assay

Primers targeting the 3′-untranslated region (UTR) of the EBLN3P gene were designed using Primer Premier 5.0 and subsequently used for amplification via PCR. The resulting PCR products were inserted into the GLO plasmid (Promega, USA) to construct the wild-type EBLN3P vector, designated as EBLN3P-WT. Additionally, site-directed mutagenesis was conducted on the 3′-UTR sequence of EBLN3P to generate the mutant vector, EBLN3P-MUT. Perform sequencing to precisely identify the position of the mutation site within the sequence. These vectors, along with either the miR-675-3p mimic or the corresponding control, were co-transfected into AC16 cells using Lipofectamine 3000 (Invitrogen, USA). The plasmid DNA was transfected at a concentration of 0.5–2 μg per well. Following a 48-h incubation period, luciferase activity was assessed using a luciferase reporter gene assay kit (16,186, Thermo Fisher Scientific, USA). Luciferase activity was normalized to the co-transfected reference gene (e.g., Renilla luciferase) to account for variations in transfection efficiency and cell viability.

Apoptosis assay

Samples were collected and subjected to multiple washes with phosphate-buffered saline (PBS) to remove impurities. The cells were then resuspended, and stained with Annexin V-FITC and propidium iodide (Sigma-Aldrich, USA) to label cells with damaged membranes. FSC-A/FSC-H was used to define single-cell populations and exclude adherent cells. Quadrants were then set in the dual-parameter dot plot of Annexin V-FITC (FL1 channel) and PI (FL3 channel): the lower left quadrant represents live cells (Annexin V/PI), the lower right quadrant represents early apoptotic cells (Annexin V+/PI), and the upper right quadrant represents late apoptotic/necrotic cells (Annexin V+/PI+). H9c2 cells were collected, washed twice with pre-cooled PBS, and adjusted to a concentration of (5 × 105) cells/mL. A total of 100 μL of the cell suspension was mixed with 5 μL of Annexin V-FITC and 5 μL of PI, gently vortexed, and incubated in the dark at room temperature for 15 min. Subsequently, 400 μL of 1 × Binding Buffer was added, and the samples were immediately analyzed by flow cytometry. A total of 10,000 single-cell events were acquired per sample. The cells that received the staining process underwent analysis based on FACSCalibur flow cytometer (no. 342973, BD Biosciences, Pudong, Shanghai) by the use of CellQuest software. The final apoptotic cell count was determined as a total percentage of early apoptotic cells staining positive for Annexin V and negative for PI and late apoptotic cells positive for both Annexin V and PI. The cell apoptosis rate was calculated using the following formula: Cell apoptosis rate (%) = (apoptotic cell number/total cell number) × 100.

Statistical analysis

SPSS 16.0 and GraphPad Prism 7.0 were utilized for statistical analysis of data. Data were checked for normality via the Kolmogorov–Smirnov (K-S) normality test. Differences between two groups were calculated using the Mann–Whitney U test for non-normally distributed continuous variables, Student’s t-test for normally distributed continuous variables, and chi-square tests were employed to analyze inter-group differences. The diagnostic value of EBLN3P for AMI was assessed using receiver operating characteristic (ROC) curve analysis. Pearson correlation analysis was conducted to quantify the strength and direction of the linear relationship between two continuous variables. The cell experiment was independently repeated three times under identical conditions to ensure the reliability of the results. A p<0.05 was considered statistically significant.

Results

Clinical features of study subjects

This study enrolled 109 patients with AMI and 117 healthy controls and compared the general clinical information and various characteristics between the two groups (Table 1). No significant differences were observed in age, sex, body mass index (BMI), smoking status, drinking, diastolic blood pressure (DBP), and HbA1c between the two groups (p>0.05). However, significant differences were noted in blood lipids (TC, LDL-C, HDL-C, TG), cardiac function (left ventricular ejection fraction (LVEF)), systolic blood pressure (SBP), eGFR, and cTnI, hs-CRP, TNF-α, IL-1β, IL-6 (p<0.05).

Table 1:

Characteristics of participants.

Characteristics AMI group n=109 Control group n=117 p-Value
Age, year 58.79 ± 11.22 58.93 ± 10.07 0.920
Sex 0.946
Male 61 66
Female 48 51
BMI, kg/m2 23.10 ± 2.23 23.27 ± 2.93 0.629
Smoking 0.546
No 90 104
Yes 19 13
Drinking 0.173
No 90 100
Yes 19 17
TC, mmol/L 4.42 ± 1.20 4.08 ± 0.94 0.017
LDL-C, mmol/L 2.82 ± 0.87 2.51 ± 0.56 0.002
HDL-C, mmol/L 1.14 ± 0.32 1.73 ± 0.36 <0.001
TG, mmol/L 1.89 ± 1.02 1.49 ± 0.89 0.002
LVEF, % 47.45 ± 3.48 57.67 ± 3.54 <0.001
SBP, mmHg 136.79 ± 18.16 128.84 ± 19.60 0.002
DBP, mmHg 87.16 ± 11.55 86.50 ± 9.78 0.643
eGFR (ml/(min × 1.73m2)) 86.88 ± 24.65 99.04 ± 12.07 <0.001
HbA1c, % 7.58 ± 1.56 7.29 ± 1.70 0.182
cTnI, μg/L 3.59 ± 0.28 0.17 ± 0.04 <0.001
Hs-CRP, mg/L 7.28 ± 1.94 3.37 ± 0.13 <0.001
TNF-α, ng/ml 387.29 ± 87.85 108.21 ± 40.78 <0.001
IL-1β, ng/ml 19.09 ± 4.41 5.03 ± 1.07 <0.001
IL-6, ng/L 56.73 ± 3.86 13.95 ± 2.71 <0.001
SYNTAX score 29.65 ± 9.76
  1. TC, total cholesterol; LDL, low-density lipoprotein cholesterol; HDL, high-density lipoprotein cholesterol; TG, triglycerides; LVEF, left ventricular ejection fraction; SBP, systolic blood pressure; DBP, diastolic blood pressure; eGFR, estimated glomerular filtration rate; HbA1c, glycosylated hemoglobin; cTnI, cardiac troponin I; hs-CRP, high-sensitivity C-reactive protein; TNF-α, tumor necrosis factor-α; IL, interleukin. The independent samples t-test and chi-square analysis were employed as statistical methods.

The expression of EBLN3P in AMI patients

The expression of EBLN3P was significantly decreased in AMI patients (Figure 1A). Additionally, the ROC curve analysis of EBLN3P demonstrated an area under the curve (AUC) of 0.881 (95%CI=0.8361–0.9253), with a sensitivity of 87.16 % and specificity of 78.63 % (p<0.0001) (Figure 1B), indicating that EBLN3P exhibited high diagnostic accuracy for distinguishing AMI patients from non-patients.

Figure 1: 
LncRNA EBLN3P demonstrates potential predictive value for AMI. (A) LncRNA EBLN3P expression was downregulated in AMI (p<0.001). (B) ROC curve analysis of EBLN3P demonstrated an AUC of 0.881 (95%CI=0.8361–0.9253), with a sensitivity of 87.16 % and specificity of 78.63 % indicating that EBLN3P exhibited high diagnostic accuracy for distinguishing AMI patients from non-patients (p<0.0001). *** means p<0.001. Non-parametric tests and ROC curve analysis were used as statistical methods.
Figure 1:

LncRNA EBLN3P demonstrates potential predictive value for AMI. (A) LncRNA EBLN3P expression was downregulated in AMI (p<0.001). (B) ROC curve analysis of EBLN3P demonstrated an AUC of 0.881 (95%CI=0.8361–0.9253), with a sensitivity of 87.16 % and specificity of 78.63 % indicating that EBLN3P exhibited high diagnostic accuracy for distinguishing AMI patients from non-patients (p<0.0001). *** means p<0.001. Non-parametric tests and ROC curve analysis were used as statistical methods.

The correlation between EBLN3P and clinical features of AMI patients

We further investigated the correlation between EBLN3P levels and various clinical features (Table 2). Pearson correlation analysis revealed no significant association between EBLN3P levels and TC, LDL, DBP, or HbA1c (p>0.05). Conversely, EBLN3P levels exhibited a negative correlation with TG, SBP, cTnI, hs-CRP, TNF-α, IL-1β, IL-6, and SYNTAX scores.

Table 2:

Correlation between EBLN3P level and clinical features.

Correlation coefficient r p-Value
TC −0.072 0.281
LDL −0.126 0.058
HDL 0.461 <0.001
TG −0.155 0.019
LVEF 0.552 <0.001
SBP −0.138 0.039
DBP −0.002 0.982
eGFR 0.211 0.001
HbA1c 0.080 0.229
cTnI −0.638 <0.001
hs-CRP −0.519 <0.001
TNF-α −0.580 <0.001
IL-1β −0.556 <0.001
IL-6 −0.635 <0.001
SYNTAX score −0.388 <0.001
  1. TC, total cholesterol; LDL, low-density lipoprotein cholesterol; HDL, high-density lipoprotein cholesterol; TG, triglycerides; LVEF, left ventricular ejection fraction; SBP, systolic blood pressure; DBP, diastolic blood pressure; eGFR, estimated glomerular filtration rate; HbA1c, glycosylated hemoglobin; cTnI, cardiac troponin I; hs-CRP, high-sensitivity C-reactive protein; TNF-α, tumor necrosis factor-α; IL, interleukin. Pearson correlation coefficient was employed as a statistical method.

The impact of EBLN3P on cardiomyocyte injury

We examined the expression levels of EBLN3P under various treatment conditions and its effects on inflammatory factors and apoptosis in cardiomyocytes (Figure 2). Transfection with EBLN3P significantly restored its expression following H/R treatment (Figure 2A). H/R treatment markedly increased the levels of inflammatory cytokines TNF-α, IL-1β, and IL-6. Transfection with pcDNA3.1 had no significant effect on these cytokine levels. In contrast, transfection with EBLN3P partially attenuated the increase in TNF-α, IL-1β, and IL-6 levels but did not fully restore them to control levels (Figure 2B–D). Transfection with EBLN3P significantly decreased the apoptosis rate after H/R treatment (Figure 2E).

Figure 2: 
Effects of EBLN3P on inflammatory factors and apoptosis of cardiomyocytes. (A) H/R treatment can reduce the expression level of EBLN3P (p<0.001). (B-D) H/R treatment significantly increased levels of TNF-α, IL-1β, and IL-6 (p<0.001). (E) H/R treatment resulted in a significant increase in apoptosis rate (p<0.001). *** means p<0.001. Non-parametric tests were used as statistical methods.
Figure 2:

Effects of EBLN3P on inflammatory factors and apoptosis of cardiomyocytes. (A) H/R treatment can reduce the expression level of EBLN3P (p<0.001). (B-D) H/R treatment significantly increased levels of TNF-α, IL-1β, and IL-6 (p<0.001). (E) H/R treatment resulted in a significant increase in apoptosis rate (p<0.001). *** means p<0.001. Non-parametric tests were used as statistical methods.

EBLN3P targeted and regulated miR-675-3p

The potential target genes associated with EBLN3P were identified using the ENCORI database, revealing that miR-675-3p was a likely target gene of EBLN3P (Figure 3A). The luciferase reporter assay further validated the direct interaction between EBLN3P and miR-675-3p (Figure 3B). Specifically, transfection of the miR-675-3p mimic into the EBLN3P-WT group resulted in a significant reduction in luciferase activity. Conversely, no significant change in luciferase activity was observed in the EBLN3P-MUT group following miR-675-3p mimic transfection (Figure 3B). Additionally, miR-675-3p expression were significantly elevated in the serum of AMI patients (Figure 3C). Pearson correlation analysis revealed a negative correlation between miR-675-3p and EBLN3P levels in AMI (r = −0.5245, p<0.0001) (Figure 3D). In addition, H/R treatment resulted in a significant increase in the expression level of miR-675-3p. Transfecting EBLN3P partially attenuated this upregulation following H/R treatment (Figure 3E).

Figure 3: 
Targeting relationship of EBLN3P-miR-675-3p. (A) EBLN3P and miR-675-3p target binding sites. (B) The dual-luciferase reporter assay was employed to validate the direct targeting interaction between EBLN3P and miR-675-3p. (C) Expression level of miR-675-3p was upregulated in patients with AMI (p<0.001). (D) The expression levels of EBLN3P and miR-675-3p exhibit a negative correlation (p<0.0001). (E) H/R treatment resulted in increased expression levels of miR-675-3p (p<0.001). *** Means p<0.001. Non-parametric tests and pearson correlation coefficient were employed as statistical methods.
Figure 3:

Targeting relationship of EBLN3P-miR-675-3p. (A) EBLN3P and miR-675-3p target binding sites. (B) The dual-luciferase reporter assay was employed to validate the direct targeting interaction between EBLN3P and miR-675-3p. (C) Expression level of miR-675-3p was upregulated in patients with AMI (p<0.001). (D) The expression levels of EBLN3P and miR-675-3p exhibit a negative correlation (p<0.0001). (E) H/R treatment resulted in increased expression levels of miR-675-3p (p<0.001). *** Means p<0.001. Non-parametric tests and pearson correlation coefficient were employed as statistical methods.

A regulatory network where EBLN3P and miR-675-3p were interconnected

Under H/R conditions, transfecting cells with EBLN3P or co-expressing EBLN3P with mimic-NC significantly reduced the relative expression level of miR-675-3p. However, co-expression of EBLN3P with miR-mimic partially counteracted this effect on miR-675-3p expression (Figure 4A). Transfection with EBLN3P or co-expression of EBLN3P with mimic-NC also significantly decreased the levels of inflammatory factors TNF-α, IL-1β, and IL-6, while co-expression of EBLN3P with miR-mimic partially mitigated these reductions (Figure 4B–D). Additionally, transfection with EBLN3P or co-expression of EBLN3P with mimic-NC significantly reduced cell apoptosis, whereas co-expression of EBLN3P with miR-mimic partially alleviated this anti-apoptotic effect (Figure 4E).

Figure 4: 
Effects of EBLN3P-miR-675-3p co-transfection on inflammatory factors and apoptosis of cardiomyocytes. (A) Verification of co-expression of miR-675-3p. (B-D) co-expression of EBLN3P+ miR-mimic could partially increased the effects of EBLN3P on the levels of TNF-α, IL-1β, and IL-6 (p<0.001). (E) Co-expression of EBLN3P+ miR-mimic could partially increase the effect of EBLN3P on apoptosis (p<0.001). *** Means p<0.001. Non-parametric tests were used as statistical methods.
Figure 4:

Effects of EBLN3P-miR-675-3p co-transfection on inflammatory factors and apoptosis of cardiomyocytes. (A) Verification of co-expression of miR-675-3p. (B-D) co-expression of EBLN3P+ miR-mimic could partially increased the effects of EBLN3P on the levels of TNF-α, IL-1β, and IL-6 (p<0.001). (E) Co-expression of EBLN3P+ miR-mimic could partially increase the effect of EBLN3P on apoptosis (p<0.001). *** Means p<0.001. Non-parametric tests were used as statistical methods.

Discussion

AMI is characterized by myocardial necrosis resulting from acute and sustained ischemia and hypoxia of the coronary arteries [30], 31]. While emergency coronary reperfusion remains the most effective approach to minimize infarct size, no current therapies effectively delay adverse cardiac remodeling or prevent cardiomyocyte death [32]. Consequently, early prevention is crucial for inhibiting AMI development. This study aimed to investigate the expression levels of EBLN3P and miR-675-3p in AMI and their interactions by statistically analyzing the clinical characteristics of AMI patients and conducting relevant pathological experiments, thereby providing insights for developing novel therapeutic strategies for AMI.

AMI is often triggered by the rupture or erosion of atherosclerotic plaques, leading to acute thrombotic occlusion of epicardial coronary arteries [1], 2]. Regarding cardiac function, AMI patients exhibited significantly lower LVEF values, indicating impaired cardiac pumping capacity. Following AMI onset, an inflammatory response is initiated, characterized by the infiltration of inflammatory cells into the infarcted area and the release of numerous inflammatory mediators [5]. Our study enrolled 109 patients with AMI and 117 healthy controls. The results demonstrated that AMI patients exhibited significant dyslipidemia, impaired cardiac function, and enhanced inflammatory responses. These three factors may be interconnected and collectively drive disease progression.

EBLN3P expression is low in primary spiral ganglion neurons (SGNs) of deaf model mice [33], and its knockdown significantly reduces proliferation, migration, and invasion in HepG2 cells [34]. Additionally, down-regulation of EBLN3P decreases methotrexate (MTX) resistance in osteosarcoma cells [35]. Our research demonstrated that EBLN3P expression was significantly downregulated in AMI, suggesting its potential as a critical regulatory factor in the pathological process of AMI. EBLN3P not only held diagnostic value for AMI but also exerted multi-dimensional regulatory effects on the cardiovascular system. The downregulation of EBLN3P may contribute to disease progression by impairing myocardial protective functions, exacerbating inflammatory responses, and inducing metabolic disorders.

Studies have shown that following AMI, an inflammatory response is initiated, characterized by the infiltration of inflammatory cells into the infarcted area and the release of numerous inflammatory mediators. While these mediators can promote tissue repair to some extent, excessive inflammation can exacerbate myocardial damage and increase cardiomyocyte apoptosis [5]. Our experimental results demonstrated that H/R treatment resulted in a significant decrease in the expression level of EBLN3P. The downregulation of EBLN3P expression may serve as a critical driving factor for both pro-inflammatory and pro-apoptotic responses. Exogenous restoration of EBLN3P could protect the myocardium by suppressing inflammatory factor release and reducing cell apoptosis.

Previous studies have demonstrated that miR-675-3p is significantly upregulated in various cancers, including gastric cancer (GC) tissues [36], indicating its potential role in multiple disease dysregulations [37]. Overexpression of miR-675-3p inhibits apoptosis and cartilage matrix degradation in human chondrocytes while promoting cell proliferation [25]. In patients with atherosclerosis, the serum level of miR-675-3p was significantly higher compared to healthy individuals [26]. Our research has demonstrated that miR-675-3p is a target gene of EBLN3P. In AMI cells, the expression level of miR-675-3p was significantly upregulated and negatively correlated with the expression of EBLN3P. Under H/R conditions, EBLN3P established a negative regulatory feedback loop with miR-675-3p. High expression of EBLN3P suppressed the expression of miR-675-3p, thereby reducing inflammation and cell apoptosis and exerting a protective effect on the myocardium. EBLN3P inhibited inflammation and apoptosis in AMI by acting as a molecular sponge for miR-675-3p. This mechanism exhibited methodological similarities to the canonical functions of lncRNAs in oncology. For example, in breast cancer, HEIH functions as a miR-98-5p-targeted competing endogenous RNA (ceRNA) sponge for STAT3, thereby modulating the proliferation, migration, and invasion of hepatocellular carcinoma (HCC) cells [38]; in colorectal cancer, lncRNA UCA1 adsorbs miR-185–5p via the competing endogenous RNA (ceRNA) mechanism to regulate the β-catenin signaling pathway [39]. These examples highlight the capacity of lncRNAs to reshape gene expression networks through competitive binding to miRNAs. Nevertheless, there are notable distinctions between these processes. In cardiovascular injury, the protective role of EBLN3P is achieved by suppressing dual anti-inflammatory and anti-apoptotic pathways, whereas in tumors, lncRNAs often drive disease progression by regulating cell proliferation, metabolic reprogramming, or immune evasion. For instance, lncRNA GHET1 enhanced the proliferation of gastric cancer cells by stabilizing c-Myc mRNA [40].

LncRNA plays a crucial role in cardiovascular diseases. It regulates gene expression via multi-level epigenetic mechanisms and exerts physiological effects within cells, such as remodeling chromatin structure in the nucleus, participating in transcriptional regulatory networks, modulating mRNA stability in the cytoplasm, and activating signaling pathways [41], 42]. In the context of cardiovascular diseases, EBLN3P in AMI functions may as a ceRNA to target miR-675-3p, thereby inhibiting its pro-inflammatory and pro-apoptotic activities. This leads to reduced release of pro-inflammatory cytokines, such as TNF-α and IL-6, decreased cardiomyocyte apoptosis rates, and alleviation of myocardial injury. Furthermore, the serum level of EBLN3P in AMI patients is negatively correlated with myocardial injury markers (e.g., cTnI) and SYNTAX scores, suggesting its potential as a diagnostic or prognostic biomarker. The EBLN3P/miR-675-3p axis may serve as a novel therapeutic target for AMI treatment, while antisense oligonucleotides (ASO) or small molecule inhibitors targeting lncRNA also exhibit promising translational potential. Further research is warranted to address unresolved issues, such as identifying downstream target genes of this mechanism, validating the regulatory network in vivo, analyzing the complex ceRNA network, and developing targeted myocardial delivery systems for therapeutic intervention.

Previous research has shown the crucial value of HbA1c in assessing the prognosis of patients with AMI [43]. In this study, the HbA1c levels between the two groups were comparable and exhibited no correlation, suggesting that blood glucose fluctuations may not be the primary driver of EBLN3P expression changes. The pathological role of EBLN3P primarily involves regulating the core pathological processes of AMI (inflammation and apoptosis) via targeting miR-675-3p, rather than through diabetes-associated pathways. The homogeneity of our study cohort and the balanced HbA1c levels effectively excluded diabetes as a major confounding factor, thereby reinforcing the evidence that EBLN3P serves as a specific biomarker for AMI. Future research directions include further validating the role of EBLN3P in patients with both diabetes and AMI, potentially expanding the sample size and conducting stratified analyses to determine whether blood glucose levels influence the function of EBLN3P in AMI.

EBLN3P, as a novel biomarker and therapeutic target for AMI, demonstrated its potential through comparative analysis with established biomarkers and therapeutic strategies. Specifically, in terms of diagnostic performance, the AUC of EBLN3P for diagnosing AMI was 0.881, which is comparable to miR-499 (AUC=0.91) [44]. Moreover, EBLN3P offered the advantage of ultra-early diagnosis, as differences can be detected within 24 h of symptom onset, surpassing UCA1, which requires detection within 3 days post-AMI [45]. Additionally, the expression level of EBLN3P is negatively correlated with myocardial injury markers (e.g., cTnI) and inflammatory factors (e.g., TNF-α), thereby providing more comprehensive pathological insights into AMI. Clinically, measuring serum EBLN3P levels may serve as an auxiliary diagnostic marker for AMI. Furthermore, dynamically monitoring the EBLN3P/miR-675-3p ratio may help evaluate the inflammatory burden and disease progression risk in AMI patients, thereby guiding clinical decision-making (e.g., determining whether to intensify anti-inflammatory therapy).

This study also had some limitations. The sample size was relatively small, the population was fairly homogeneous, and direct evidence from in vivo animal models (e.g., AMI mice) was lacking. In the future, we plan to conduct a multi-center, large-sample prospective study, establish an AMI animal model, and comprehensively investigate the role of the EBLN3P/miR-675-3p axis in AMI.

In summary, the expression of EBLN3P and miR-675-3p exhibited a negative correlation. Furthermore, co-expression of EBLN3P with miR-675-3p could partially mitigate the effects of EBLN3P on inflammatory factors and cell apoptosis. These findings provided robust evidence that EBLN3P was involved in the regulatory mechanism of AMI by targeting miR-675-3p.


Corresponding author: Wanrong Bi, Department of General Practice, Tongji Hospital Branch Affiliated to Tongji University, No.50, Chifeng Road, Shanghai, 200092, China, E-mail:

  1. Research ethics: This study received approval from the Tongji Hospital Branch Affiliated to Tongji University Medical Ethics Committee (No.: 20220012, Date of approval: 07-03-2022). Additionally, all procedures adhered strictly to the principles outlined in the Helsinki Declaration.

  2. Informed consent: All participants provided written informed consent.

  3. Author contributions: study conception and design: Wanrong Bi, Qing Peng, Yushi Feng, Jingjing Liu; data collection: Wanrong Bi, Qing Peng, Yushi Feng, Jingjing Liu; analysis and interpretation of results: Wanrong Bi, Qing Peng, Yushi Feng, Jingjing Liu; draft manuscript preparation: Wanrong Bi. All authors reviewed the results and approved the final version of the manuscript.

  4. Use of Large Language Models, AI and Machine Learning Tools: Not applicable.

  5. Conflict of interest: The author states no conflict of interest.

  6. Research funding: None declared.

  7. Data availability: The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.

References

1. Abdullah, Rashid, M, Soto, CJ, Virk, GS, Mekowulu, FC, Chaudhari, SS, et al.. The safety and efficacy of the early use of sacubitril/valsartan After acute myocardial infarction: a meta-analysis of randomized controlled trials. Cureus 2024;16:e53784. https://doi.org/10.7759/cureus.53784.Suche in Google Scholar PubMed PubMed Central

2. Guo, Y, Luo, F, Liu, Q, Xu, DJJ, Medicine m. regulatory non‐coding RNA s in acute myocardial infarction. 2017;21(5):1013-23, https://doi.org/10.1111/jcmm.13032.Suche in Google Scholar PubMed PubMed Central

3. Guerchicoff, A, Brener, SJ, Maehara, A, Witzenbichler, B, Fahy, M, Xu, K, et al.. Impact of delay to reperfusion on reperfusion success, infarct size, and clinical outcomes in patients with ST-segment elevation myocardial infarction: the INFUSE-AMI trial (INFUSE-Anterior Myocardial Infarction). JACC Cardiovasc Interv 2014;7:733–40. https://doi.org/10.1016/j.jcin.2014.01.166.Suche in Google Scholar PubMed

4. Zhao, Y, Guo, Y, Chen, Y, Liu, S, Wu, N, Jia, DJI. Curculigoside attenuates myocardial ischemia-reperfusion injury by inhibiting the opening of the mitochondrial permeability transition pore. 2020;45(5):1514-24, https://doi.org/10.3892/ijmm.2020.4513.Suche in Google Scholar PubMed PubMed Central

5. Fang, L, Moore, XL, Dart, AM, Wang, LM. Systemic inflammatory response following acute myocardial infarction. Journal of geriatric cardiology: JGC 2015;12:305–12. https://doi.org/10.11909/j.issn.1671-5411.2015.03.020.Suche in Google Scholar PubMed PubMed Central

6. Huang, W, Zhang, Q, Qi, H, Shi, P, Song, C, Liu, Y, et al.. Deletion of neuropeptide Y attenuates cardiac dysfunction and apoptosis during acute myocardial infarction. 2019;10:1268. https://doi.org/10.3389/fphar.2019.01268.Suche in Google Scholar PubMed PubMed Central

7. Neri, M, Riezzo, I, Pascale, N, Pomara, C, Turillazzi, E. Ischemia/reperfusion injury following acute myocardial infarction: a critical issue for Clinicians and forensic pathologists. Mediat Inflamm 2017;2017:7018393. https://doi.org/10.1155/2017/7018393.Suche in Google Scholar PubMed PubMed Central

8. Kim, SJ. Global awareness of myocardial infarction symptoms in general population. Korean Circ J 2021;51:997–1000. https://doi.org/10.4070/kcj.2021.0320.Suche in Google Scholar PubMed PubMed Central

9. Zhang, Y, Zhang, Y, Wang, S, Cao, B, Hu, D, Jia, J, et al.. LINC00467 facilitates the proliferation, migration and invasion of glioma via promoting the expression of inositol hexakisphosphate kinase 2 by binding to miR-339-3p. Bioengineered 2022;13:3370–82. https://doi.org/10.1080/21655979.2021.2018098.Suche in Google Scholar PubMed PubMed Central

10. Tan, L, Mengshan, L, Yu, F, Yelin, L, Jihong, Z, Lixin, G. Predicting lncRNA-protein interactions using a hybrid deep learning model with dinucleotide-codon fusion feature encoding. BMC Genom 2024;25:1253. https://doi.org/10.1186/s12864-024-11168-3.Suche in Google Scholar PubMed PubMed Central

11. Chen, L, Li, J, Li, Q, Yan, H, Zhou, B, Gao, Y, et al.. Non-coding RNAs: the new insight on hypertrophic scar. J Cell Biochem 2017;118:1965–8. https://doi.org/10.1002/jcb.25873.Suche in Google Scholar PubMed

12. Eftekhari, H. Interaction between long non-coding RNA and micro RNA in hypertrophic scar formation. Burns : journal of the International Society for Burn Injuries 2024;50:107239. https://doi.org/10.1016/j.burns.2024.08.005.Suche in Google Scholar PubMed

13. Huang, YJJ, medicine, m. The novel regulatory role of lnc RNA‐mi RNA‐mRNA axis in cardiovascular diseases. 2018;22(12):5768-75.10.1111/jcmm.13866Suche in Google Scholar PubMed PubMed Central

14. Xu, X, Zhang, Q, Song, H, Ao, Z, Li, X, Cheng, C, et al.. Effects of artemisinin on ventricular arrhythmias in response to left ventricular afterload increase and microRNA expression profiles in wistar rats. 2018;6:e6110. https://doi.org/10.7717/peerj.6110.Suche in Google Scholar PubMed PubMed Central

15. Adachi, T, Nakanishi, M, Otsuka, Y, Nishimura, K, Hirokawa, G, Goto, Y, et al.. Plasma microRNA 499 as a biomarker of acute myocardial infarction. Clin Chem 2010;56:1183–5. https://doi.org/10.1373/clinchem.2010.144121.Suche in Google Scholar PubMed

16. Vausort, M, Wagner, DR, Devaux, Y. Long noncoding RNAs in patients with acute myocardial infarction. Circ Res 2014;115:668–77. https://doi.org/10.1161/circresaha.115.303836.Suche in Google Scholar

17. Yang, KC, Nerbonne, JM. Mechanisms contributing to myocardial potassium channel diversity, regulation and remodeling. Trends Cardiovasc Med 2016;26:209–18. https://doi.org/10.1016/j.tcm.2015.07.002.Suche in Google Scholar PubMed PubMed Central

18. Guo, Y, Luo, F, Liu, Q, Xu, D. Regulatory non-coding RNAs in acute myocardial infarction. J Cell Mol Med 2017;21:1013–23. https://doi.org/10.1111/jcmm.13032.Suche in Google Scholar PubMed PubMed Central

19. Liu, J, Sun, F, Wang, Y, Yang, W, Xiao, H, Zhang, Y, et al.. Suppression of microRNA-16 protects against acute myocardial infarction by reversing beta2-adrenergic receptor down-regulation in rats. Oncotarget 2017;8:20122–32. https://doi.org/10.18632/oncotarget.15391.Suche in Google Scholar PubMed PubMed Central

20. Zhu, S, Fang, Z. MicroRNA-127-3p inhibits cardiomyocyte inflammation and apoptosis after acute myocardial infarction via targeting CDKN3. Int Heart J 2023;64:1133–9. https://doi.org/10.1536/ihj.23-379.Suche in Google Scholar PubMed

21. Dai, S, Li, N, Zhou, M, Yuan, Y, Yue, D, Li, T, et al.. LncRNA EBLN3P promotes the progression of osteosarcoma through modifying the miR-224-5p/Rab10 signaling axis. Sci Rep 2021;11:1992. https://doi.org/10.1038/s41598-021-81641-6.Suche in Google Scholar PubMed PubMed Central

22. Wang, X, Yue, Y, Tan, J, Kou, F, Su, B, Xie, J, et al.. LncRNA EBLN3P accelerates cell proliferation and invasion of Colon cancer through modulating the miR-519d-3p/ZFP91 axis. Cancer Biother Rad 2024. https://doi.org/10.1089/cbr.2022.0089.Suche in Google Scholar PubMed

23. Zou, T, Gao, Y, Qie, M. Promotive actions of lncRNA EBLN3P involved in cervical cancer progression via interacting with miR-29c-3p and TAF15 to modify RCC2. Arch Biochem Biophys 2024;755:109980. https://doi.org/10.1016/j.abb.2024.109980.Suche in Google Scholar PubMed

24. van der Ree, MH, de Bruijne, J, Kootstra, NA, Jansen, PL, Reesink, HW. MicroRNAs: role and therapeutic targets in viral hepatitis. Antivir Ther 2014;19:533–41. https://doi.org/10.3851/imp2766.Suche in Google Scholar PubMed

25. Shen, XF, Cheng, Y, Dong, QR, Zheng, MQ. MicroRNA-675-3p regulates IL-1β-stimulated human chondrocyte apoptosis and cartilage degradation by targeting GNG5. Biochem Biophys Res Commun 2020;527:458–65. https://doi.org/10.1016/j.bbrc.2020.04.044.Suche in Google Scholar PubMed

26. Wang, S, Shao, W, Gao, Y, Zhao, H, Du, D. Diagnostic and prognostic significance of miR-675-3p in patients with atherosclerosis. Clin Appl Thrombosis/Hemostasis: Off J Int Acad Clin Appl Thrombosis/Hemostasis 2021;27:10760296211024754. https://doi.org/10.1177/10760296211024754.Suche in Google Scholar PubMed PubMed Central

27. Zhang, Y, Zhong, P, Xu, Y, Wang, B, Zhu, T, Zhang, W, et al.. Differential expression of TXNIP isoforms in the peripheral leukocytes of patients with acute myocardial infarction. Dis Markers 2018;2018:9051481. https://doi.org/10.1155/2018/9051481.Suche in Google Scholar PubMed PubMed Central

28. Liu, H, Mo, H, Yang, C, Mei, X, Song, X, Lu, W, et al.. A novel function of ATF3 in suppression of ferroptosis in mouse heart suffered ischemia/reperfusion. Free Radic Biol Med 2022;189:122–35. https://doi.org/10.1016/j.freeradbiomed.2022.07.006.Suche in Google Scholar PubMed

29. Li, W, Li, W, Leng, Y, Xiong, Y, Xia, Z. Ferroptosis is involved in diabetes myocardial ischemia/reperfusion injury through endoplasmic reticulum stress. DNA Cell Biol 2020;39:210–25. https://doi.org/10.1089/dna.2019.5097.Suche in Google Scholar PubMed

30. Rojas-Humpire, R, Olarte-Durand, M, Medina-Ramirez, S, Gutierrez-Ajalcriña, R, Canaza, JF, Huancahuire-Vega, SJJN, et al.. Insulin resistance indexes as biomarkers of lifetime cardiovascular risk among adults from Peru. 2021;2021(1):6633700, https://doi.org/10.1155/2021/6633700.Suche in Google Scholar PubMed PubMed Central

31. Niccoli, G, Kharbanda, RK, Crea, F, Banning, APJE. No-reflow: again prevention is better than treatment. 2010;31(20):2449-55, https://doi.org/10.1093/eurheartj/ehq299.Suche in Google Scholar PubMed

32. Xu, H, Yu, W, Sun, S, Li, C, Ren, J, Zhang, YJSB. TAX1BP1 protects against myocardial infarction-associated cardiac anomalies through inhibition of inflammasomes in a RNF34/MAVS/NLRP3-dependent manner. 2021;66(16):1669-83, https://doi.org/10.1016/j.scib.2021.01.030.Suche in Google Scholar PubMed

33. Jiang, W, Peng, A, Chen, Y, Pang, B, Zhang, Z. Long non-coding RNA EBLN3P promotes the recovery of the function of impaired spiral ganglion neurons by competitively binding to miR-204-5p and regulating TMPRSS3 expression. Int J Mol Med 2020;45:1851–63. https://doi.org/10.3892/ijmm.2020.4545.Suche in Google Scholar PubMed PubMed Central

34. Li, H, Wang, M, Zhou, H, Lu, S, Zhang, B. Long noncoding RNA EBLN3P promotes the progression of liver cancer via alteration of microRNA-144-3p/DOCK4 signal. Cancer Manag Res 2020;12:9339–49. https://doi.org/10.2147/cmar.s261976.Suche in Google Scholar PubMed PubMed Central

35. Sun, MX, An, HY, Sun, YB, Sun, YB, Bai, B. LncRNA EBLN3P attributes methotrexate resistance in osteosarcoma cells through miR-200a-3p/O-GlcNAc transferase pathway. J Orthop Surg Res 2022;17:557. https://doi.org/10.1186/s13018-022-03449-y.Suche in Google Scholar PubMed PubMed Central

36. Chen, R, Li, Y, Ouyang, W, Chen, S. Bioinformatics analysis reveals the biomarker value and potential mechanism of miR-675-3p in gastric cancer. Comput Intell Neurosci 2022;2022:5456554. https://doi.org/10.1155/2022/5456554.Suche in Google Scholar PubMed PubMed Central

37. Intelligence, Neuroscience C. Retracted: bioinformatics analysis reveals the biomarker value and potential mechanism of miR-675-3p in gastric cancer. Comput Intell Neurosci. 2023;2023:9804746, https://doi.org/10.1155/2023/9804746.Suche in Google Scholar PubMed PubMed Central

38. Ai, JH, Wen, YZ, Dai, SJ, Zhang, LD, Huang, ZJ, Shi, J. Exosomal lncRNA HEIH, an essential communicator for hepatocellular carcinoma cells and macrophage M2 polarization through the miR-98-5p/STAT3 axis. J Biochem Mol Toxicol 2024;38:e23686. https://doi.org/10.1002/jbt.23686.Suche in Google Scholar PubMed

39. Liu, C, Ji, L, Song, X. Long non coding RNA UCA1 contributes to the autophagy and survival of colorectal cancer cells via sponging miR-185-5p to up-regulate the WISP2/β-catenin pathway. RSC Adv 2019;9:14160–6. https://doi.org/10.1039/c8ra10468a.Suche in Google Scholar PubMed PubMed Central

40. Yang, F, Xue, X, Zheng, L, Bi, J, Zhou, Y, Zhi, K, et al.. Long non-coding RNA GHET1 promotes gastric carcinoma cell proliferation by increasing c-Myc mRNA stability. FEBS J 2014;281:802–13. https://doi.org/10.1111/febs.12625.Suche in Google Scholar PubMed

41. Poloni, JF, Oliveira, FHS, Feltes, BC. Localization is the key to action: regulatory peculiarities of lncRNAs. Front Genet 2024;15:1478352. https://doi.org/10.3389/fgene.2024.1478352.Suche in Google Scholar PubMed PubMed Central

42. Dong, Y, Lyu, L, Wen, H, Shi, B. Brain and pituitary transcriptome analyses reveal the differential regulation of reproduction-related LncRNAs and mRNAs in Cynoglossus semilaevis. Front Genet 2021;12:802953. https://doi.org/10.3389/fgene.2021.802953.Suche in Google Scholar PubMed PubMed Central

43. Choi, IJ, Choo, EH, Kim, HJ, Lim, S, Moon, D, Lee, KY, et al.. J-curve relationship between long term glycemic control and mortality in diabetic patients with acute myocardial infarction undergoing percutaneous coronary intervention. Cardiovasc Diabetol 2021;20:234. https://doi.org/10.1186/s12933-021-01428-x.Suche in Google Scholar PubMed PubMed Central

44. Liu, X, Fan, Z, Zhao, T, Cao, W, Zhang, L, Li, H, et al.. Plasma miR-1, miR-208, miR-499 as potential predictive biomarkers for acute myocardial infarction: an independent study of Han population. Exp Gerontol 2015;72:230–8. https://doi.org/10.1016/j.exger.2015.10.011.Suche in Google Scholar PubMed

45. Yan, Y, Zhang, B, Liu, N, Qi, C, Xiao, Y, Tian, X, et al.. Circulating long noncoding RNA UCA1 as a novel biomarker of acute myocardial infarction. BioMed Res Int 2016;2016:8079372. https://doi.org/10.1155/2016/8079372.Suche in Google Scholar PubMed PubMed Central

Received: 2025-02-12
Accepted: 2025-08-19
Published Online: 2025-09-15

© 2025 the author(s), published by De Gruyter, Berlin/Boston

This work is licensed under the Creative Commons Attribution 4.0 International License.

Heruntergeladen am 15.12.2025 von https://www.degruyterbrill.com/document/doi/10.1515/tjb-2025-0060/html
Button zum nach oben scrollen