HMOX1 silencing prevents doxorubicin-induced cardiomyocyte injury, mitochondrial dysfunction, and ferroptosis by downregulating CTGF
- PMID: 36008747
- DOI: 10.1007/s11748-022-01867-7
HMOX1 silencing prevents doxorubicin-induced cardiomyocyte injury, mitochondrial dysfunction, and ferroptosis by downregulating CTGF
Abstract
Objectives: Doxorubicin is a type of effective antitumor drug but can contribute to cardiomyocyte injuries. We aimed to dissect the mechanism of the HMOX1/CTGF axis in DOX-induced cardiomyocyte injury, mitochondrial dysfunction, and ferroptosis.
Methods: Bioinformatics analysis was conducted to retrieve differentially expressed genes in a DOX-induced mouse model. Mouse cardiomyocytes, HL-1 cells, were induced with l µM DOX, after which gain- or loss-of-function assays were applied. CCK-8, fluorescent probe assay, flow cytometry, and corresponding kits were employed to detect cell viability, ROS levels, mitochondrial membrane potential and cell apoptosis, and GSH and Fe2+ contents, respectively. qRT-PCR or Western blot assay was adopted to test HMOX1, CTGF, BCL-2, Caspase3, Cleaved-Caspase3, and GPX4 expression.
Results: Bioinformatics analysis showed that HMOX1 and CTGF were highly expressed in DOX-induced mice and correlated with each other. Also, HMOX1 and CTGF expression was high in HL-1 cells after DOX treatment, along with an obvious decrease in cell viability and GSH and GPX4 expression, an increase in ROS levels, apoptosis, and Fe2+ contents, and mitochondrial membrane potential dysfunction or loss. HMOX1 or CTGF silencing diminished cell apoptosis, Cleaved-Caspase3 expression, Fe2+ contents, and ROS levels, enhanced cell viability and the expression of GSH, GPX4, and BCL-2, and recovered mitochondrial membrane potential in DOX-induced HL-1 cells. Nevertheless, the effects of HMOX1 silencing on the viability, apoptosis, ferroptosis, and mitochondrial dysfunction of DOX-induced HL-1 cells were counteracted by CTGF overexpression.
Conclusions: In conclusion, HMOX1 silencing decreased CTGF expression to alleviate DOX-induced injury, mitochondrial dysfunction, and ferroptosis of mouse cardiomyocytes.
Keywords: CTGF; Cardiotoxicity; Cell apoptosis; Doxorubicin; Ferroptosis; HMOX1; Mitochondrial dysfunction.
© 2022. The Author(s), under exclusive licence to The Japanese Association for Thoracic Surgery.
Similar articles
-
Lapatinib induces mitochondrial dysfunction to enhance oxidative stress and ferroptosis in doxorubicin-induced cardiomyocytes via inhibition of PI3K/AKT signaling pathway.Bioengineered. 2022 Jan;13(1):48-60. doi: 10.1080/21655979.2021.2004980. Bioengineered. 2022. PMID: 34898356 Free PMC article.
-
Sheng-Mai-Yin inhibits doxorubicin-induced ferroptosis and cardiotoxicity through regulation of Hmox1.Aging (Albany NY). 2023 Sep 28;15(19):10133-10145. doi: 10.18632/aging.205062. Epub 2023 Sep 28. Aging (Albany NY). 2023. PMID: 37770231 Free PMC article.
-
WGX50 mitigates doxorubicin-induced cardiotoxicity through inhibition of mitochondrial ROS and ferroptosis.J Transl Med. 2023 Nov 17;21(1):823. doi: 10.1186/s12967-023-04715-1. J Transl Med. 2023. PMID: 37978379 Free PMC article.
-
Ferroptosis as a target for protection against cardiomyopathy.Proc Natl Acad Sci U S A. 2019 Feb 12;116(7):2672-2680. doi: 10.1073/pnas.1821022116. Epub 2019 Jan 28. Proc Natl Acad Sci U S A. 2019. PMID: 30692261 Free PMC article.
-
Ferroptosis: A novel therapeutic target of natural products against doxorubicin-induced cardiotoxicity.Biomed Pharmacother. 2024 Sep;178:117217. doi: 10.1016/j.biopha.2024.117217. Epub 2024 Jul 29. Biomed Pharmacother. 2024. PMID: 39079260 Review.
Cited by
-
HMOX1 Participates in Pre-Eclampsia by Regulating the Proliferation, Apoptosis, and Angiogenesis Modulation Potential of Mesenchymal Stem Cells via VEGF.Biochem Genet. 2024 Apr;62(2):1248-1262. doi: 10.1007/s10528-023-10474-x. Epub 2023 Aug 12. Biochem Genet. 2024. PMID: 37573262
-
Current Understanding of Pulmonary Fibrosis: Pathogenesis, Diagnosis, and Therapeutic Approaches.Can Respir J. 2025 Jul 15;2025:3183241. doi: 10.1155/carj/3183241. eCollection 2025. Can Respir J. 2025. PMID: 40697404 Free PMC article. Review.
-
The dual role of ferroptosis in anthracycline-based chemotherapy includes reducing resistance and increasing toxicity.Cell Death Discov. 2023 Jun 21;9(1):184. doi: 10.1038/s41420-023-01483-1. Cell Death Discov. 2023. PMID: 37344500 Free PMC article. Review.
-
Paeonol alleviates ox-LDL-induced endothelial cell injury by targeting the heme oxygenase-1/phosphoinositide 3-kinase/protein kinase B pathway.Naunyn Schmiedebergs Arch Pharmacol. 2025 Jan;398(1):591-600. doi: 10.1007/s00210-024-03307-0. Epub 2024 Jul 22. Naunyn Schmiedebergs Arch Pharmacol. 2025. PMID: 39037459
-
Ferroptosis, a Regulated Form of Cell Death, as a Target for the Development of Novel Drugs Preventing Ischemia/Reperfusion of Cardiac Injury, Cardiomyopathy and Stress-Induced Cardiac Injury.Int J Mol Sci. 2024 Jan 11;25(2):897. doi: 10.3390/ijms25020897. Int J Mol Sci. 2024. PMID: 38255971 Free PMC article. Review.
References
MeSH terms
Substances
LinkOut - more resources
Full Text Sources
Miscellaneous