2Central Laboratory, Jinhua Municipal General Hospital, Jinhua, Zhejiang, China
3Department of Cardiology, Jinhua Municipal General Hospital, Jinhua, Zhejiang, China
4Department of Cardiology, Heart Center, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China
Abstract
Background: Growth arrest specific 5 (GAS5) is a long noncoding RNA (lncRNA) that regulates the function of cardiovascular cells in various cardiovascular diseases. The current study delved into the regulation of GAS5 on the function of endothelial progenitor cells (EPCs) and its potential regulatory mechanism in coronary heart disease (CHD).
Methods: Reverse transcription-quantitative polymerase chain reaction was used to detect GAS5 expression in the blood samples and EPCs from CHD patients and healthy controls. Cell Counting Kit-8, colony formation, flow cytometry, and transwell assays were performed to evaluate cell phenotype of EPCs. Ferroptosis was detected by the measurement of Fe2+, malondialdehyde, GSH, and reactive oxygen species (ROS) levels. Glycolysis was determined by extracellular acidification rate (ECAR), oxygen consumption rate (OCR), glucose uptake and lactate production.
Results: Growth arrest specific 5 was downregulated in the blood samples and EPCs from CHD patients. Growth arrest specific 5 deficiency suppressed EPC proliferative capacity, migration, invasion and facilitated EPC apoptosis while GAS5 overexpression showed contrary effects. Moreover, GAS5 silencing inhibited the glucose metabolic reprogramming, as evidenced by the reduced ECAR, glycolysis capacity, ATP, glucose uptake and lactate production, and elevated OCR. Additionally, GAS5 overexpression attenuated the erastin-induced ferroptosis of EPCs. Growth arrest specific 5 could bind to IGF2BP1 to enhance the mRNA stability of glycolysis transcriptional regulator SIX1. Growth arrest specific 5 interacted with miR-23a-3p to regulate SLC7A11 expression. GAS5 promoted glucose metabolic reprogramming of EPCs by upregulating SIX1 and inhibited EPC ferroptosis by elevating SLC7A11.
Conclusion: Growth arrest specific 5 promotes glucose metabolic reprogramming and represses ferroptosis of EPCs via the IGF2BP1/SIX1 and miR-23a-3p/SLC7A11 dual-regulatory pathways in CHD.
Highlights
- Growth arrest specific 5 contributes to the growth, migration and invasion of endothelial progenitor cells.
- Growth arrest specific 5 promoted the glucose metabolic reprogramming of endothelial progenitor cells by upregulating SIX1.
- Growth arrest specific 5 inhibits ferroptosis of endothelial progenitor cells by upregulating SLC7A11.
- Growth arrest specific 5 interacts with IGF2BP1 to stabilize SIX1.
- Growth arrest specific 5 binds to miR-23a-3p to upregulate SLC7A11 in EPCs.
Introduction
Coronary heart disease (CHD) caused by coronary atherosclerosis is a common cause of heart attack. Clinically, patients with CHD exhibit symptoms such as angina pectoris induced by temporary myocardial ischemia. According to the China Cardiovascular Health and Disease Report 2022, there are an estimated 330 million people with cardiovascular diseases in China, among which 11.39 million cases are CHD.1 The high incidence and mortality rate of CHD pose a high burden to society. Currently, the treatment of CHD mainly includes percutaneous coronary intervention, bypass surgery, and pharmacological therapy. However, many patients are not allowed to receive interventional procedures, and for those receiving treatment, the clinical outcomes are limited by myocardial damage and subsequent heart failure.2 The exploration of novel effective therapeutic strategies is still in urgent need.
Endothelial dysfunction is closely associated with the onset of CHD.3 Endothelial progenitor cells (EPCs) can differentiate into mature endothelial cells and are considered as critical contributors to neovascularization and vascular repair, showing the potential for CHD therapy.4 Studies have demonstrated that EPCs facilitate the vascular remodeling of ischemic and damaged tissues in animal models and human cases of ischemia.5,
Long noncoding RNAs (lncRNAs) are noncoding transcripts over 200 nucleotides long and possess no protein-coding ability.8,
This study intended to investigate the role of the novel molecular mechanism of GAS5 in CHD. It was hypothesized that GAS5 affected CHD progression by regulating the function of EPCs via upregulating the expression of the ferroptosis inhibitor SLC7A11 and the glycolysis transcriptional regulator SIX1, which might provide novel insight into CHD therapy.
Methods
Patient Sample Collection
The atherosclerotic peripheral blood (2 mL) was collected from 30 CHD patients and 30 healthy individuals in ethylenediaminetetraacetic acid (EDTA) tubes in our hospital. The plasma was collected after centrifugation for 15 minutes at 3000 rpm at 4°C and then stored at −80°C for further analysis. All participants have signed the informed consent. The Ethics Committee of our hospital has approved this study.
Isolation and Incubation of Endothelial Progenitor Cells
Peripheral blood from CHD patients and healthy individuals were collected and diluted with phosphate buffer saline (PBS). Next, the isolation of peripheral blood mononuclear cells (PBMCs) was conducted with ficoll density gradient centrifugation, followed by washing and centrifugation to remove the platelet. The supernatant was removed and PBMCs were resuspended in culture medium to adjust a concentration at 1 × 106. Next, cells were cultured on 6-well plates coated with fibronectin, and each well was added with 2 mL EGM-2 (LONZA, USA) and maintained at 37 °C with 95% air and 5% CO2.
After culturing for 4 days, to select EPCs, the plates were washed with PBS for removing the non-attached cells. The attached cells were collected and subject to flow cytometry analysis with for surface markers including CD34, CD31, and CD45.17
Cell Transfection
Two specific siRNAs targeting GAS5 (si-GAS5#1/#2) and negative control (si-NC), SIX1 and IGF2BP1 overexpression vector and pcDNA3.1 empty vector (oe-NC), miR-23a-3p mimic and NC mimic were designed and synthesized by GenePharma (Shanghai, China). The transfection vector or plasmid was conducted with Lipofectamine 3000 (Invitrogen, Carlsbad, CA, USA) for 48 hours.
Reverse Transcription-Quantitative Polymerase Chain Reaction
Total RNA was extracted using TRIzol reagent. A High-Capacity cDNA Reverse Transcription Kit (Applied Biosystems, USA) was applied for synthesizing cDNAs. Quantitative polymerase chain reaction was carried out with a LightCycler FastStart DNA MasterPLUS SYBR Green I mix (Roche, Germany) on the ABI-7500 platform. Gene expression was calculated with the 2−ΔΔCt method, and β-actin and U6 served as internal references.
Western Blot
Radioimmunoprecipitation assay lysis buffer was used for total protein collection. A Bicinchoninic Acid Kit was adopted for the detection of protein concentration. Next, the protein sample was electrophoresed by SDS–polyacrylamide gel electrophoresis (Bio-Rad, USA) and electro-transferred to nitrocellulose membranes. Subsequently, 5% skim milk was applied for blocking these membranes, which were then incubated overnight with the primary antibodies at 4°C, with β-actin as a loading control. After rinsing with Tris-Borate-Sodium Tween-20 (TBST), the membranes were incubated with a secondary antibody for 60 minutes. The enhanced chemiluminescence (Pierce, USA) was used for the visualization of protein signals.
Fluorescent In Situ Hybridization
Fluorescent In Situ Hybridization Kit (RiboBio, Guangzhou, China) was used to determine the subcellular localization of GAS5 following the producer’s protocol. Endothelial progenitor cells were seeded in slides, processed with 4% paraformaldehyde, rinsed with PBS, and processed using 0.5% Triton X-100. Subsequently, EPCs were hybridized with probes for GAS5 and IGF2BP1 (RiboBio, China) overnight at 37 °C. 4’,6-diamidino-2-phenylindole (DAPI) was applied to stain the nucleus. The images were captured by a confocal fluorescence microscope (Leica, Wetzlar, Germany).
Co-Immunoprecipitation
Endothelial progenitor cells were treated using lysis buffer, centrifuged, and the supernatant was incubated with anti-IgG or anti-SIX1 overnight at 4°C. Next, the mixture was captured by incubation with protein A/G agarose beads for 4 hours. After washing with ice-cold IP buffer, the samples were centrifuged at 1000 ×
RNA Immunoprecipitation
Endothelial progenitor cells were lysed and the lysate was incubated with anti-IgG (negative control) or anti-Ago2 antibody conjugated with magnetic beads at 4°C overnight. The beads were collected and washed, followed by proteinase K treatment. RNA extraction from the complex was conducted using phenol: chloroform: isoamyl alcohol solution. Immunoprecipitated RNAs from anti-IgG or anti-Ago2 were subject to reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis.
RNA Pull-Down
Biotinylated GAS5, miR-23a-3p, and control NC probes were provided by Ribobio. Endothelial progenitor cells were lysed and incubated overnight with biotinylated probes-conjugated with magnetic beads at 4°C. After collecting and washing beads, the RNA or protein in the complex was purified. The enrichment of IGF2BP1, miRNAs, or SLC7A11 was subject to western blot or RT-qPCR analysis.
Dual-Luciferase Reporter Assays
Growth arrest specific 5 and SLC7A11 3′UTR containing including wild-type (Wt) and mutated (Mut) binding sequence with miR-23a-3p were subcloned into pmirGLO vector (Promega, USA) for the construction of GAS5-wt/mut or SLC7A11-wt/mut vector. The vector was co-transfected in EPCs with miR-23a-3p/NC mimic for 48 hours. The luciferase activities were determined with a Promega dual-luciferase reporter kit.
Cell Viability
The viability of EPCs was detected using CCK-8 assays. Briefly, EPCs were seeded in 96-well plates (5 × 103 cells in each well) and cultured for indicated time (1, 2, 3, 4 days). EPCs in each well were supplemented with 10 μL CCK-8 reagent and cultured for 120 minutes. The absorbance was analyzed at 450 nm.
Colony Formation Assay
Endothelial progenitor cells were seeded in 6-well plates at 500 cells per well. After a 2-week incubation, 4% polyformaldehyde was applied to fasten colonies, which were then dyed by 0.5% crystal violet for 1 hour. The colony number was finally calculated with a microscope.
Flow Cytometry Analysis
The apoptosis of EPCs was measured using an Annexin V-FITC/PI Apoptosis Kit (Mutisiences, China) following the producer’s guide. Endothelial progenitor cells after transfection were harvested, and stained with Annexin V-FITC and Propidium Iodide for 20 minutes at 4°C in darkness. Cell apoptotic rate was evaluated by FACScan Flow Cytometer, BD Biosciences, USA) and analyzed with FlowJo software.
Transwell Assays
The transwell assays were performed for evaluating the migration and invasion abilities of EPCs using Transwell chambers (Costar, USA) coated with or without matrigel (BD Biosciences, USA). Endothelial progenitor cells (5 × 104) were suspended in culture medium (0.5 mL) containing 0.1% bovine serum albumin and added in the top Transwell chambers, and culture media with 20% FBS in the lower chambers. The incubation last for 24 hours. After PBS washing, and processing in 4% paraformaldehyde, EPCs were stained for 5 minutes with crystal violet solution. The migrated or invaded cells were counted and imaged with an inverted microscope.
Glucose Uptake Measurement
Endothelial progenitor cells were grown into 96-well plates at 1 × 104 cells/well, and incubated at 37°C overnight. Next, EPCs were deprived of sugar for 2 hours. Then each well was supplemented with 10 μL 2-deoxy-d-glucose (2-DG) and cultured for 20 minutes. The glucose intake was determined at 412 nm wavelength.
Lactic Acid Production Assay
The production of lactic acid in EPCs was assessed using an L-lactic acid test kit (colorimetry). Endothelial progenitor cells were first grown in 96-well plates and cultured overnight at 37°C. After starvation for 120 minutes, the supernatant was harvested and lactic acid production was evaluated at 450 nm.
Extracellular Acidification Rate and Oxygen Consumption Rate Measurement
Endothelial progenitor cells were grown in 96-well plates added with 10% FBS and cultured at 37°C overnight. After measuring the baseline concentration, glucose, oligomycin, and 2-DG were supplemented into each well successively for extracellular acidification rate(ECAR) measurement. For oxygen consumption rate(OCR) measurement, oligomycin, FCCP, antimycin A, and rotenone were added successively. The results were analyzed using XF-96Wave software.
Malondialdehyde Detection
The malondialdehyde (MDA) content in the lysate of EPCs was detected using a Lipid Peroxidation Assay Kit (Abcam, UK) based on the manufacturer’s protocol. Briefly, EPCs after erastin treatment and indicated transfection were harvested and lysed, and subsequently centrifuged for 10 minutes at 13 000 ×
Glutathione (GSH)/oxidized glutathione (GSSG) Detection
The level of GSH/GSSG in EPCs was measured using a GSH and GSSG Assay Kit (Beyotime, China) based on the producer’s guide. The resulting mixture was measured using a microplate reader at 412 nm. The GSH level is calculated as GSH = Total Glutathione−GSSG × 2.
Iron Assay
Intracellular ferrous iron (Fe2+) level in EPCs was detected using an iron assay kit (Abcam, UK) following the manufacturer’s protocol. Endothelial progenitor cells were grown in a culture plate and exposed to erastin for 12 hours. Then cells were harvested, washed, and homogenized in iron assay buffer, followed by for 10-minute centrifugation at 4°C. Next, the supernatant was harvested and supplemented iron reducer into each well, mixed, and incubated for 30 minutes at ambient temperature. Subsequently, the iron probe was supplemented and mixed, followed by a 1-hour incubation in darkness. Finally, a colorimetric microplate reader was applied for absorbance detection at 593 nm.
Statistical Analysis
GraphPad Prism software (GraphPad Software Inc., San Diego, CA) was used for statistical analysis. The experiments were independently conducted at least 3 times. The normality of data distribution was evaluated with the Shapiro–Wilk test. Data were presented as the mean ± SD. Two-tailed Student’s
Results
Growth Arrest Specific 5 is Downregulated in Coronary Heart Disease and Accelerates Endothelial Progenitor Cell Proliferation, Migration, and Invasion
The GAS5 level was determined in the blood samples and EPCs collected from CHD patients (n = 30) and healthy individuals (n = 30). The results of RT-qPCR analysis identified that GAS5 was downregulated in both blood samples and EPCs of CHD patients compared with healthy controls (
Growth Arrest Specific 5 is Located in the Cell Cytoplasm of Endothelial Progenitor Cells
To determine the subcellular localization of GAS5 in EPCs, Fluorescence
Growth Arrest Specific 5 Promotes Glucose Metabolism Reprogramming and Inhibits Ferroptosis of Endothelial Progenitor Cells Via the IGF2BP1/SIX1 Axis
Growth arrest specific 5 can regulate cellular metabolism, which is closely associated with the fate of EPCs. Thus, the effects of GAS5 on glucose metabolic reprogramming of EPCs were further investigated. As revealed by immunofluorescence staining of 2-NBDG, the glucose uptake of EPCs was significantly suppressed by GAS5 knockdown (
Growth Arrest Specific 5 Interacts with IGF2BP1 to Stabilize SIX1
The underlying mechanism of GAS5 to regulate glucose metabolism reprogramming and ferroptosis of EPCs was further investigated. As shown in
Growth Arrest Specific 5 Promotes Glucose Metabolism Reprogramming by Upregulating SIX1
Whether SIX1 is involved in GAS5-mediated metabolic reprogramming of EPCs was further explored. SIX1 was overexpressed in EPCs, and the overexpression efficiency was verified by RT-qPCR analysis as well as western blot analysis (
Growth Arrest Specific 5 Binds to miR-23a-3p to Modulate SLC7A11 Expression
The mechanism of GAS5 to regulate ferroptosis of EPCs was further explored. Substantial literature has revealed that lncRNAs can also function as ceRNAs to regulate gene expression. Based on bioinformatics analysis, the candidate miRNA targets of GAS5 were explored on the ENCORI and lncbase databases. A total of 22 candidate miRNAs were screened by the 2 databases (
Growth Arrest Specific 5 Inhibits Ferroptosis of Endothelial Progenitor Cells Via SLC7A11
Further investigation was conducted on whether SLC7A11 was involved in GAS5-mediated ferroptosis of EPCs. The viability of EPCs reduced in response to erastin treatment was reversed after GAS5 overexpression, while SLC7A11 silencing attenuated the protective effects of GAS5 on EPCs, as evidenced by the reduced viability of EPCs (
Discussion
Cardiovascular diseases remain a major cause of death globally, taking nearly 18 million lives annually, among which CHD is responsible for an estimated 7.4 million death cases.20 However, the therapeutic interventions for CHD are still limited, and patients suffer from complications with unfavorable outcomes. Thus, it is essential to explore effective therapeutic targets to improve the treatment of CHD.
LncRNAs are closely associated with the progression of cardiovascular diseases by regulating diverse biological processes including inflammation, cell proliferation, apoptosis, as well as senescence.21 Functionally, lncRNAs are known as regulators for mRNA decay or sponging miRNAs to prevent the inhibitory effects of miRNAs on the stability or the translation of their targets at post-transcriptional levels.22,
In recent decades, the potential of endothelial progenitor cells has been noticed in the treatment of cardiovascular diseases. As a group of cells released from bone marrow into peripheral blood, EPCs effectively promote endothelial repair and neovasculogenesis, and are thus considered useful for the cell therapy of ischemic heart disease.31 Clinical studies have revealed that the proliferation, and migration abilities of EPCs are impaired and the apoptosis rate is elevated in acute coronary syndrome patients.32,
Glucose metabolic reprogramming is induced by myocardial infarction. However, the changes cannot support cardiomyocyte proliferation to replace lost cells caused by the ischemic injury.37 Metabolic reprogramming is indicated to promote cardiac regeneration following injury, while the underlying targets need further investigation. Wang et al38 have found that PPARγ agonist promotes cardiac glucose metabolic reprogramming by enhancing glycolytic capacity and reducing mitochondrial reactive ROS production via HIF-1α to prevent hypoxia-induced cardiac dysfunction. A study reveals that VEGF-B reprograms myocardial metabolism to enhance cardiac function in ischemic heart disease.39 In our study, GAS5 silencing was demonstrated to inhibit glucose metabolic reprogramming by reducing glycolysis capacity, glucose uptake, and lactate production. SIX1 is a critical transcription regulator of glycolysis. In our previous study, GAS5 was found to regulate glucose metabolism reprogramming of EPCs via the miR-495-3p/SIX1 and IGF2BP2/NRF2 dual-regulatory pathways.16 In this study, GAS5 was further found to directly bind to IGF2BP1 to enhance the mRNA stability of SIX1 in EPCs. Moreover, rescue assays proved that GAS5 promoted the glucose metabolic reprogramming of EPCs by upregulating SIX1.
Ferroptosis is a novel form of iron-dependent programmed cell death. Studies have shown that anti-ferroptosis strategies can alleviate cell injury in coronary heart disease. For example, SGK1 deficiency attenuates the erastin-caused ferroptosis of mouse aortic endothelial cells by reversing the erastin-caused decrease in SLC7A11, GPX4, and GSH/GSSG and elevation in lipid peroxidation as well as Fe accumulation.40 SLC7A11 as an amino acid transporter plays a crucial role in regulating GSH synthesis and cellular ferroptosis. In our previous study, GAS5 was found to participate in the progression of CHD by regulating ferroptosis of EPCs through the miR-495-3p/SIX1 and IGF2BP2/NRF2 dual-regulatory pathways.16 The current study has confirmed for the first time that GAS5 can interact with miR-23a-3p to upregulate SLC7A11, thereby protecting EPCs from ferroptosis in CHD.
Conclusion
Growth arrest specific 5 is downregulated in EPCs collected from CHD patients, and overexpression of GAS5 promotes glucose metabolism reprogramming via the IGF2BP1/SIX1 axis and inhibits ferroptosis of EPCs via the miR-23a-3p/SLC7A11 axis. The findings of our study might provide novel therapeutic targets for EPC protection in CHD treatment.
Supplementary Materials
Footnotes
References
- Wang Z, Ma L, Liu M, Fan J, Hu S. Summary of the 2022 report on cardiovascular health and diseases in china. Chin Med J (Engl). 2023;136(24):2899-2908.
- Chepeleva EV. Cell therapy in the treatment of coronary heart disease. Int J Mol Sci. 2023;24(23):16844-.
- Matsuzawa Y, Guddeti RR, Kwon TG, Lerman LO, Lerman A. Treating coronary disease and the impact of endothelial dysfunction. Prog Cardiovasc Dis. 2015;57(5):431-442.
- Morrone D, Picoi MEL, Felice F. Endothelial progenitor cells: an appraisal of relevant data from bench to bedside. Int J Mol Sci. 2021;22(23):12874-.
- Hu Z, Wang H, Fan G. Danhong injection mobilizes endothelial progenitor cells to repair vascular endothelium injury via upregulating the expression of Akt, eNOS and MMP-9. Phytomedicine. 2019;61():152850-.
- Vasa M, Fichtlscherer S, Adler K. Increase in circulating endothelial progenitor cells by statin therapy in patients with stable coronary artery disease. Circulation. 2001;103(24):2885-2890.
- Yan F, Li J, Zhang W. Transplantation of Endothelial Progenitor Cells: summary and prospect. Acta Histochem. 2023;125(1):151990-.
- Kopp F, Mendell JT. Functional classification and experimental dissection of long noncoding RNAs. Cell. 2018;172(3):393-407.
- Rinn JL, Chang HY. Long noncoding RNAs: molecular modalities to organismal functions. Annu Rev Biochem. 2020;89():283-308.
- Yao RW, Wang Y, Chen LL. Cellular functions of long noncoding RNAs. Nat Cell Biol. 2019;21(5):542-551.
- Li P, Li Y, Chen L. Long noncoding RNA uc003pxg.1 regulates endothelial cell proliferation and migration via miR255p in coronary artery disease. Int J Mol Med. 2021;48(2):-.
- Zhang H, Ji N, Gong X, Ni S, Wang Y. NEAT1/miR-140-3p/MAPK1 mediates the viability and survival of coronary endothelial cells and affects coronary atherosclerotic heart disease. Acta Biochim Biophys Sin (Shanghai). 2020;52(9):967-974.
- Hao S, Liu X, Sui X, Pei Y, Liang Z, Zhou N. Long non-coding RNA GAS5 reduces cardiomyocyte apoptosis induced by MI through sema3a. Int J Biol Macromol. 2018;120(Pt A):371-377.
- Yao J, Shi Z, Ma X, Xu D, Ming G. lncRNA GAS5/miR-223/NAMPT axis modulates the cell proliferation and senescence of endothelial progenitor cells through PI3K/AKT signaling. J Cell Biochem. 2019;120(9):14518-14530.
- Chen T, Liang Q, Xu J. MiR-665 regulates vascular smooth muscle cell senescence by interacting with LncRNA GAS5/SDC1. Front Cell Dev Biol. 2021;9():700006-.
- Zhong M, Xu W, Tang B, Zhao Q, Jiang Z, Liu Y. GAS5 promotes glucose metabolism reprogramming and resistance to ferroptosis of endothelial progenitor cells through the miR-495-3p/SIX1 and IGF2BP2/NRF2 dual-regulatory pathways in coronary heart disease. Cell Mol Biol (Noisy-Le-Grand). 2024;70(9):121-128.
- Ikutomi M, Sahara M, Nakajima T. Diverse contribution of bone marrow-derived late-outgrowth endothelial progenitor cells to vascular repair under pulmonary arterial hypertension and arterial neointimal formation. J Mol Cell Cardiol. 2015;86():121-135.
- Ren Y, Zhao X. Bone marrow mesenchymal stem cells-derived exosomal lncRNA GAS5 mitigates heart failure by inhibiting UL3/Hippo pathway-mediated ferroptosis. Eur J Med Res. 2024;29(1):303-.
- Liu D, Yang M, Yao Y. Cardiac fibroblasts promote ferroptosis in atrial fibrillation by secreting Exo-miR-23a-3p targeting SLC7A11. Oxid Med Cell Longev. 2022;2022():3961495-.
- . . Fact. ;():-.
- Yeh CF, Chang YE, Lu CY, Hsuan CF, Chang WT, Yang KC. Expedition to the missing link: long noncoding RNAs in cardiovascular diseases. J Biomed Sci. 2020;27(1):48-.
- Zhang L, Yang Z, Trottier J, Barbier O, Wang L. Long noncoding RNA MEG3 induces cholestatic liver injury by interaction with PTBP1 to facilitate shp mRNA decay. Hepatology. 2017;65(2):604-615.
- Tay Y, Rinn J, Pandolfi PP. The multilayered complexity of ceRNA crosstalk and competition. Nature. 2014;505(7483):344-352.
- Ni W, Yao S, Zhou Y. Long noncoding RNA GAS5 inhibits progression of colorectal cancer by interacting with and triggering YAP phosphorylation and degradation and is negatively regulated by the m(6)A reader YTHDF3. Mol Cancer. 2019;18(1):143-.
- Zhang Z, Liu T, Cheng C. LncRNA GAS5 regulates the Wnt/β-catenin pathway through the miR-18a-5p/AXIN2/GSK3β axis to inhibit the proliferation and migration of bladder cancer cells. Carcinogenesis. 2022;43(12):1176-1189.
- Zhu L, Zhou D, Guo T. LncRNA GAS5 inhibits Invasion and Migration of Lung Cancer through influencing EMT process. J Cancer. 2021;12(11):3291-3298.
- Zhou XH, Chai HX, Bai M, Zhang Z. LncRNA-GAS5 regulates PDCD4 expression and mediates myocardial infarction-induced cardiomyocytes apoptosis via targeting MiR-21. Cell Cycle. 2020;19(11):1363-1377.
- Wu N, Zhang X, Bao Y, Yu H, Jia D, Ma C. Down-regulation of GAS5 ameliorates myocardial ischaemia/reperfusion injury via the miR-335/ROCK1/AKT/GSK-3β axis. J Cell Mol Med. 2019;23(12):8420-8431.
- Zhuo X, Bai K, Wang Y. Long-chain noncoding RNA-GAS5/hsa-miR-138-5p attenuates high glucose-induced cardiomyocyte damage by targeting CYP11B2. Biosci Rep. 2021;41(9):BSR20202232-.
- Diao L, Bai L, Jiang X, Li J, Zhang Q. Long-chain noncoding RNA GAS5 mediates oxidative stress in cardiac microvascular endothelial cells injury. J Cell Physiol. 2019;234(10):17649-17662.
- Bianconi V, Sahebkar A, Kovanen P. Endothelial and cardiac progenitor cells for cardiovascular repair: a controversial paradigm in cell therapy. Pharmacol Ther. 2018;181():156-168.
- Zhang LJ, Liu WX, Chen YD, Song XT, Jin ZN, Lü SZ. Proliferation, migration and apoptosis activities of endothelial progenitor cells in acute coronary syndrome. Chin Med J (Engl). 2010;123(19):2655-2661.
- Solomon A, Blum A, Peleg A, Lev EI, Leshem-Lev D, Hasin Y. Endothelial progenitor cells are suppressed in anemic patients with acute coronary syndrome. Am J Med. 2012;125(6):604-611.
- Jin Y, Yang CJ, Xu X, Cao JN, Feng QT, Yang J. MiR-214 regulates the pathogenesis of patients with coronary artery disease by targeting VEGF. Mol Cell Biochem. 2015;402(1-2):111-122.
- Ouyang S, Li Y, Wu X. GPR4 signaling is essential for the promotion of acid-mediated angiogenic capacity of endothelial progenitor cells by activating STAT3/VEGFA pathway in patients with coronary artery disease. Stem Cell Res Ther. 2021;12(1):149-.
- Li Y, Cui W, Song B, Ye X, Li Z, Lu C. Autophagy-sirtuin1(SIRT1) alleviated the coronary atherosclerosis (AS)in mice through regulating the proliferation and migration of endothelial progenitor cells (EPCs) via wnt/β-catenin/GSK3β signaling pathway. J Nutr Health Aging. 2022;26(3):297-306.
- Chen X, Wu H, Liu Y, Liu L, Houser SR, Wang WE. Metabolic reprogramming: a byproduct or a driver of cardiomyocyte proliferation?. Circulation. 2024;149(20):1598-1610.
- Wang Y, Zhang R, Chen Q. PPARγ agonist pioglitazone prevents hypoxia-induced cardiac dysfunction by reprogramming glucose metabolism. Int J Biol Sci. 2024;20(11):4297-4313.
- Kivelä R, Bry M, Robciuc MR. VEGF-B-induced vascular growth leads to metabolic reprogramming and ischemia resistance in the heart. EMBO Mol Med. 2014;6(3):307-321.
- Peng Y, Jiang Y, Zhou Q, Jia Z, Tang H. SGK1 contributes to ferroptosis in coronary heart disease through the NEDD4L/NF-κB pathway. J Mol Cell Cardiol. 2024;196():71-83.