A novel circPIK3C2A/miR-31-5p/TFRC axis drives ferroptosis and accelerates myocardial injury

Shuo Miao1, Lanting Yang1, Tao Xu2, Zhantao Liu1, Yixiao Zhang1, Lin Ding1, Wei Ding3(), Xiang Ao1(), Jianxun Wang1()

PDF
MedComm ›› 2024, Vol. 5 ›› Issue (6) : e571. DOI: 10.1002/mco2.571
ORIGINAL ARTICLE

A novel circPIK3C2A/miR-31-5p/TFRC axis drives ferroptosis and accelerates myocardial injury

  • Shuo Miao1, Lanting Yang1, Tao Xu2, Zhantao Liu1, Yixiao Zhang1, Lin Ding1, Wei Ding3(), Xiang Ao1(), Jianxun Wang1()
Author information +
History +

Abstract

Iron overload is common in cardiovascular disease, it is also the factor that drives ferroptosis. Noncoding RNAs play an important role in heart disease; however, their regulatory role in iron overload-mediated ferroptosis remains much unknown. In our study, the iron overload model in mice was constructed through a high-iron diet, and ammonium iron citrate treatment was used to mimic iron overload in vitro. We found iron overload induced ferroptosis in cardiomyocytes, which was dependent on the high expression of transferrin receptor (TFRC). MiR-31-5p was downregulated during iron overload; it inhibited cardiomyocyte ferroptosis by targeting TFRC. CircPIK3C2A, a highly expressed circRNA in the heart, was upregulated when iron was overloaded. CircPIK3C2A enhanced the expression of TFRC by sponging miR-31-5p and promoted ferroptosis during iron overload. Our results reveal a novel mechanistic insight into noncoding RNA-based ferroptosis and identify the circPIK3C2A/miR-31-5p/TFRC axis as a promising therapeutic target for myocardial damage.

Keywords

circPIK3C2A / ferroptosis / Iron overload / miR-31-5p / TFRC

Cite this article

Download citation ▾
Shuo Miao, Lanting Yang, Tao Xu, Zhantao Liu, Yixiao Zhang, Lin Ding, Wei Ding, Xiang Ao, Jianxun Wang. A novel circPIK3C2A/miR-31-5p/TFRC axis drives ferroptosis and accelerates myocardial injury. MedComm, 2024, 5(6): e571 https://doi.org/10.1002/mco2.571

References

1 X Jiang, BR Stockwell, M Conrad. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 2021;22(4):266-282.
2 A Sharma, SJS Flora. Positive and negative regulation of ferroptosis and its role in maintaining metabolic and redox homeostasis. Oxid Med Cell Longev. 2021;2021:9074206.
3 Y Lu, Q Yang, Y Su, et al. MYCN mediates TFRC-dependent ferroptosis and reveals vulnerabilities in neuroblastoma. Cell Death Dis. 2021;12(6):511.
4 S Zhuang, Y Ma, Y Zeng, et al. METTL14 promotes doxorubicin-induced cardiomyocyte ferroptosis by regulating the KCNQ1OT1-miR-7-5p-TFRC axis. Cell Biol Toxicol. 2021;39(3):1015-1035.
5 H Zhang, P Zhabyeyev, S Wang, GY Oudit. Role of iron metabolism in heart failure: from iron deficiency to iron overload. Biochim Biophys Acta Mol Basis Dis. 2019;1865(7):1925-1937.
6 KT Sawicki, A De Jesus, H Ardehali. Iron metabolism in cardiovascular disease: physiology, mechanisms, and therapeutic targets. Circ Res. 2023;132(3):379-396.
7 X Li, L Yang, LL Chen. The biogenesis, functions, and challenges of circular RNAs. Mol Cell. 2018;71(3):428-442.
8 S Misir, N Wu, BB Yang. Specific expression and functions of circular RNAs. Cell Death Differ. 2022;29(3):481-491.
9 A Balihodzic, F Prinz, MA Dengler, GA Calin, PJ Jost, M Pichler. Non-coding RNAs and ferroptosis: potential implications for cancer therapy. Cell Death Differ. 2022;29(6):1094-1106.
10 R Qi, Y Bai, Y Wei, N Liu, B Shi. The role of non-coding RNAs in ferroptosis regulation. J Trace Elem Med Biol. 2022;70:126911.
11 S Miao, Q Zhang, W Ding, et al. Platelet internalization mediates ferroptosis in myocardial infarction. Arterioscler Thromb Vasc Biol. 2023;43(2):218-230.
12 S Werfel, S Nothjunge, T Schwarzmayr, TM Strom, T Meitinger, S Engelhardt. Characterization of circular RNAs in human, mouse and rat hearts. J Mol Cell Cardiol. 2016;98:103-107.
13 K Kuo, L Palmer. Pathophysiology of hemorrhagic shock. J Vet Emerg Crit Care (San Antonio). 2022;32(S1):22-31.
14 K Usuki. Diagnosis and treatment for aplastic anemia. Rinsho Ketsueki. 2021;62(8):922-930.
15 T Pan, Y Ji, H Liu, et al. Impact of iron overload and iron chelation with deferasirox on outcomes of patients with severe aplastic anemia after allogeneic hematopoietic stem cell transplantation. Transplant Cell Ther. 2023;29(8):507.e1-507.e8.
16 X Fang, Z Cai, H Wang, et al. Loss of cardiac ferritin H facilitates cardiomyopathy via Slc7a11-mediated ferroptosis. Circ Res. 2020;127(4):486-501.
17 Y Baba, JK Higa, BK Shimada, et al. Protective effects of the mechanistic target of rapamycin against excess iron and ferroptosis in cardiomyocytes. Am J Physiol Heart Circ Physiol. 2018;314(3):H659-H668.
18 HD Miyamoto, M Ikeda, T Ide, et al. Iron overload via heme degradation in the endoplasmic reticulum triggers ferroptosis in myocardial ischemia-reperfusion injury. JACC Basic Transl Sci. 2022;7(8):800-819.
19 E Park, SW Chung. ROS-mediated autophagy increases intracellular iron levels and ferroptosis by ferritin and transferrin receptor regulation. Cell Death Dis. 2019;10(11):822.
20 T Tadokoro, M Ikeda, T Ide, et al. Mitochondria-dependent ferroptosis plays a pivotal role in doxorubicin cardiotoxicity. JCI Insight. 2023;8(6).
21 S Zhuang, Y Ma, Y Zeng, et al. METTL14 promotes doxorubicin-induced cardiomyocyte ferroptosis by regulating the KCNQ1OT1-miR-7-5p-TFRC axis. Cell Biol Toxicol. 2023;39(3):1015-1035.
22 W Yu, Y Hu, Z Liu, et al. Sorting nexin 3 exacerbates doxorubicin-induced cardiomyopathy via regulation of TFRC-dependent ferroptosis. Acta Pharm Sin B. 2023;13(12):4875-4892.
23 Y Pan, J Yang, J Dai, X Xu, X Zhou, W Mao. TFRC in cardiomyocytes promotes macrophage infiltration and activation during the process of heart failure through regulating Ccl2 expression mediated by hypoxia inducible factor-1alpha. Immun Inflamm Dis. 2023;11(8):e835.
24 K Kang, N Li, Y Gao, et al. circ-Katnal1 enhances inflammatory pyroptosis in sepsis-induced liver injury through the miR-31-5p/GSDMD axis. Mediat Inflamm. 2022;2022:8950130.
25 TJ Ren, C Liu, JF Hou, FX Shan. CircDDX17 reduces 5-fluorouracil resistance and hinders tumorigenesis in colorectal cancer by regulating miR-31-5p/KANK1 axis. Eur Rev Med Pharmacol Sci. 2020;24(4):1743-1754.
26 SN Reilly, X Liu, R Carnicer, et al. Up-regulation of miR-31 in human atrial fibrillation begets the arrhythmia by depleting dystrophin and neuronal nitric oxide synthase. Sci Transl Med. 2016;8(340):340ra74.
27 H Li, H Shi, F Zhang, et al. LncRNA Tincr regulates PKCvarepsilon expression in a miR-31-5p-dependent manner in cardiomyocyte hypertrophy. Naunyn Schmiedebergs Arch Pharmacol. 2020;393(12):2495-2506.
28 X Fang, H Wang, D Han, et al. Ferroptosis as a target for protection against cardiomyopathy. Proc Natl Acad Sci USA. 2019;116(7):2672-2680.
29 Z Zeng, L Xia, S Fan, et al. Circular RNA CircMAP3K5 acts as a microRNA-22-3p sponge to promote resolution of intimal hyperplasia via TET2-mediated smooth muscle cell differentiation. Circulation. 2021;143(4):354-371.
30 M Li, W Ding, MA Tariq, et al. A circular transcript of ncx1 gene mediates ischemic myocardial injury by targeting miR-133a-3p. Theranostics. 2018;8(21):5855-5869.
31 ZX Wang, J Ma, XY Li, et al. Quercetin induces p53-independent cancer cell death through lysosome activation by the transcription factor EB and Reactive Oxygen Species-dependent ferroptosis. Br J Pharmacol. 2021;178(5):1133-1148.
32 JX Ren, C Li, XL Yan, Y Qu, Y Yang, ZN Guo. Crosstalk between oxidative stress and ferroptosis/oxytosis in ischemic stroke: possible targets and molecular mechanisms. Oxid Med Cell Longev. 2021;2021:6643382.
33 MP Chen, ZI Cabantchik, S Chan, GC Chan, YF Cheung. Iron overload and apoptosis of HL-1 cardiomyocytes: effects of calcium channel blockade. PLoS One. 2014;9(11):e112915.
34 Y Wang, M Wu, R Al-Rousan, et al. Iron-induced cardiac damage: role of apoptosis and deferasirox intervention. J Pharmacol Exp Ther. 2011;336(1):56-63.
35 K Wang, B Long, F Liu, et al. A circular RNA protects the heart from pathological hypertrophy and heart failure by targeting miR-223. Eur Heart J. 2016;37(33):2602-2611.
PDF

Accesses

Citations

Detail

Sections
Recommended

/