Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2023 Nov 17:2023:1108280.
doi: 10.1155/2023/1108280. eCollection 2023.

Depletion of lncRNA MEG3 Ameliorates Imatinib-Induced Injury of Cardiomyocytes via Regulating miR-129-5p/HMGB1 Axis

Affiliations

Depletion of lncRNA MEG3 Ameliorates Imatinib-Induced Injury of Cardiomyocytes via Regulating miR-129-5p/HMGB1 Axis

Peng Tang et al. Anal Cell Pathol (Amst). .

Abstract

Imatinib is a classical targeted drug to treat chronic myeloid leukemia (CML). However, it shows cardiotoxicity, which limits its clinical application. Long noncoding RNA (lncRNA) maternally expressed gene 3 (MEG3) shows proapoptotic properties in human cells. This study is performed to investigate whether targeting MEG3 can attenuate imatinib-mediated cardiotoxicity to cardiomyocytes. In this work, H9c2 cells were divided into four groups: control group, hypoxia group, hypoxia + imatinib, and hypoxia + imatinib + MEG3 knockdown group. MEG3 and microRNA-129-5p (miR-129-5p) expression levels were detected by the quantitative real-time PCR (qRT-PCR). The viability and apoptosis of H9c2 cells were then evaluated by cell counting kit-8 (CCK-8), flow cytometry, and TUNEL assays. The targeting relationships between MEG3 and miR-129-5p, between miR-129-5p and high-mobility group box 1 (HMBG1), were validated by dual-luciferase reporter assay and RNA Immunoprecipitation (RIP) assay. The protein expression level of HMGB1 was detected by western blot. It was revealed that, Imatinib-inhibited cell viability and aggravated the apoptosis of H9c2 cells cultured in hypoxic condition, and MEG3 knockdown significantly counteracted this effect. MiR-129-5p was a downstream target of MEG3 and it directly targeted HMGB1, and knockdown of MEG3 inhibited HMGB1 expression in H9c2 cells. In conclusion, targeting MEG3 ameliorates imatinib-induced injury of cardiomyocytes via regulating miR-129-5p/HMGB1 axis.

PubMed Disclaimer

Conflict of interest statement

The authors declare that they have no conflicts of interest.

Figures

Figure 1
Figure 1
MEG3 is highly expressed in imatinib-induced cardiomyocyte in hypoxic condition. (a) Chemical structure of imatinib. (b) H9c2 cells were pretreated with imatinib followed by hypoxia induction, and the viability of each group of cells was subsequently measured using a CCK-8 assay. (c and d) H9c2 cells were pretreated with imatinib followed by hypoxia induction, followed by TUNEL assay and flow cytometry to detect apoptosis in each group of cells. (e) Western blot was performed to detect the expression levels of caspase 3 and cleaved caspase 3 in H9c2 cells of each group.  P < 0.05,  P < 0.01, and  P < 0.001.
Figure 2
Figure 2
Effect of knockdown of MEG3 on the viability and apoptosis of H9c2 cells cultured with imatinib. (a) Dataset GSE161151 was downloaded to analyze the differences in lncRNA expression pattern in normal and ischemic myocardial tissues of mice. (b) H9c2 was transfected with si-NC or si-MEG3#1/2, followed by imatinib pretreatment, then hypoxia induction, and finally qRT-PCR was performed to detect MEG3 expression. (c) After H9c2 treatment, a CCK-8 assay was performed to detect the viability of H9c2 cells in each group. (d) A TUNEL assay was performed to detect apoptosis of H9c2 in each group. (e) Apoptosis of H9c2 cells was detected by flow cytometry for each group of H9c2.  P < 0.05,  P < 0.01, and  P < 0.001.
Figure 3
Figure 3
MEG3 directly targets miR-129-5p. (a) The downstream targets of MEG3 were predicted by StarBase and LncBase databases, and a Venn diagrams was plotted. (b) The binding site of MiR-129-5p to MEG3 was shown. (c) MiR-NC or miR-129-5p mimics were transfected with MEG3-WT or MEG3-MUT, respectively, into H9c2 cells, and the luciferase activity of each group was detected using a dual-luciferase reporter gene assay. (d) The enrichment of miR-129-5p with MEG3 in H9c2 cells in lgG group or Ago2 group was analyzed by RIP assay. (e) MEG3 overexpression plasmid and si-MEG3#2 were transfected into H9c2 cells, respectively, and then miR-129-5p expression was detected by qRT-PCR.  P < 0.001.
Figure 4
Figure 4
MEG3 is involved in imatinib-induced myocardial injury by targeting miR-129-5p. (a) si-MEG3#2 and miR-129-5p inhibitors were cotransfected into H9c2 cells, followed by imatinib treatment, then hypoxia induction, and finally qRT-PCR was performed to detect miR-129-5p expression. (b) A CCK-8 assay was performed to detect the cell viability of H9c2 cells in each group. (c) A TUNEL assay was executed to detect apoptosis of H9c2 cells in each group. (d) The apoptosis of H9c2 cells was detected by flow cytometry.  P < 0.001.
Figure 5
Figure 5
MEG3 upregulates HMGB1 expression by targeting miR-129-5p. (a) The downstream target genes of miR-129-5p were predicted by StarBase and TargetScan databases, and a Venn diagram was plotted. (b) The binding site between HMGB1 3′UTR and miR-129-5p is shown. (c) miR-NC or miR-129-5p mimics were transfected with HMGB1-WT or HMGB1-MUT, respectively, into H9c2 cells, and the luciferase activity of each group was detected using a dual-luciferase reporter gene assay. (d) miR-129-5p mimics or miR-129-5p inhibitors were transfected with H9c2, respectively, and HMGB1 expression was detected by the western blot. (e) si-MEG3#2 + miR-129-5p inhibitors were cotransfected with imatinib-cultured H9c2, and HMGB1 expression was detected by the western blot.  P < 0.001.

Similar articles

Cited by

References

    1. Castillo-Casas J. M., Caño-Carrillo S., Sánchez-Fernández C., Franco D., Lozano-Velasco E. Comparative analysis of heart regeneration: searching for the key to heal the heart—part II: molecular mechanisms of cardiac regeneration. Journal of Cardiovascular Development and Disease . 2023;10(9) doi: 10.3390/jcdd10090357.357 - DOI - PMC - PubMed
    1. Stagno F., Stella S., Spitaleri A., Pennisi M. S., Di Raimondo F., Vigneri P. Imatinib mesylate in chronic myeloid leukemia: frontline treatment and long-term outcomes. Expert Review of Anticancer Therapy . 2016;16(3):273–278. doi: 10.1586/14737140.2016.1151356. - DOI - PubMed
    1. Orphanos G. S., Ioannidis G. N., Ardavanis A. G. Cardiotoxicity induced by tyrosine kinase inhibitors. Acta Oncologica . 2009;48(7):964–970. doi: 10.1080/02841860903229124. - DOI - PubMed
    1. Garcia-Alvarez A., Garcia-Albeniz X., Esteve J., Rovira M., Bosch X. Cardiotoxicity of tyrosine-kinase-targeting drugs. Cardiovascular & Hematological Agents in Medicinal Chemistry . 2010;8(1):11–21. doi: 10.2174/187152510790796192. - DOI - PubMed
    1. Singh A. P., Umbarkar P., Tousif S., Lal H. Cardiotoxicity of the BCR-ABL1 tyrosine kinase inhibitors: emphasis on ponatinib. International Journal of Cardiology . 2020;316:214–221. doi: 10.1016/j.ijcard.2020.05.077. - DOI - PMC - PubMed

LinkOut - more resources