Cargando…

Acid sphingomyelinase promotes diabetic cardiomyopathy via NADPH oxidase 4 mediated apoptosis

BACKGROUND: Increased acid sphingomyelinase (ASMase) activity is associated with insulin resistance and cardiac dysfunction. However, the effects of ASMase on diabetic cardiomyopathy (DCM) and the molecular mechanism(s) underlying remain to be elucidated. We here investigated whether ASMase caused D...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Ruijiao, Duan, Tengfei, Yu, Li, Tang, Yongzhong, Liu, Shikun, Wang, Chunjiang, Fang, Wei-Jin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9896821/
https://www.ncbi.nlm.nih.gov/pubmed/36732747
http://dx.doi.org/10.1186/s12933-023-01747-1
_version_ 1784882128547217408
author Liu, Ruijiao
Duan, Tengfei
Yu, Li
Tang, Yongzhong
Liu, Shikun
Wang, Chunjiang
Fang, Wei-Jin
author_facet Liu, Ruijiao
Duan, Tengfei
Yu, Li
Tang, Yongzhong
Liu, Shikun
Wang, Chunjiang
Fang, Wei-Jin
author_sort Liu, Ruijiao
collection PubMed
description BACKGROUND: Increased acid sphingomyelinase (ASMase) activity is associated with insulin resistance and cardiac dysfunction. However, the effects of ASMase on diabetic cardiomyopathy (DCM) and the molecular mechanism(s) underlying remain to be elucidated. We here investigated whether ASMase caused DCM through NADPH oxidase 4-mediated apoptosis. METHODS AND RESULTS: We used pharmacological and genetic approaches coupled with study of murine and cell line samples to reveal the mechanisms initiated by ASMase in diabetic hearts. The protein expression and activity of ASMase were upregulated, meanwhile ceramide accumulation was increased in the myocardium of HFD mice. Inhibition of ASMase with imipramine (20 mg Kg(−1) d(−1)) or siRNA reduced cardiomyocyte apoptosis, fibrosis, and mitigated cardiac hypertrophy and cardiac dysfunction in HFD mice. The similar effects were observed in cardiomyocytes treated with high glucose (HG, 30 mmol L(−1)) + palmitic acid (PA, 100 μmol L(−1)) or C16 ceramide (CER, 20 μmol L(−1)). Interestingly, the cardioprotective effect of ASMase inhibition was not accompanied by reduced ceramide accumulation, indicating a ceramide-independent manner. The mechanism may involve activated NADPH oxidase 4 (NOX4), increased ROS generation and triggered apoptosis. Suppression of NOX4 with apocynin prevented HG + PA and CER incubation induced Nppb and Myh7 pro-hypertrophic gene expression, ROS production and apoptosis in H9c2 cells. Furthermore, cardiomyocyte-specific ASMase knockout (ASMase(Myh6KO)) restored HFD-induced cardiac dysfunction, remodeling, and apoptosis, whereas NOX4 protein expression was downregulated. CONCLUSIONS: These results demonstrated that HFD-mediated activation of cardiomyocyte ASMase could increase NOX4 expression, which may stimulate oxidative stress, apoptosis, and then cause metabolic cardiomyopathy. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12933-023-01747-1.
format Online
Article
Text
id pubmed-9896821
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-98968212023-02-04 Acid sphingomyelinase promotes diabetic cardiomyopathy via NADPH oxidase 4 mediated apoptosis Liu, Ruijiao Duan, Tengfei Yu, Li Tang, Yongzhong Liu, Shikun Wang, Chunjiang Fang, Wei-Jin Cardiovasc Diabetol Research BACKGROUND: Increased acid sphingomyelinase (ASMase) activity is associated with insulin resistance and cardiac dysfunction. However, the effects of ASMase on diabetic cardiomyopathy (DCM) and the molecular mechanism(s) underlying remain to be elucidated. We here investigated whether ASMase caused DCM through NADPH oxidase 4-mediated apoptosis. METHODS AND RESULTS: We used pharmacological and genetic approaches coupled with study of murine and cell line samples to reveal the mechanisms initiated by ASMase in diabetic hearts. The protein expression and activity of ASMase were upregulated, meanwhile ceramide accumulation was increased in the myocardium of HFD mice. Inhibition of ASMase with imipramine (20 mg Kg(−1) d(−1)) or siRNA reduced cardiomyocyte apoptosis, fibrosis, and mitigated cardiac hypertrophy and cardiac dysfunction in HFD mice. The similar effects were observed in cardiomyocytes treated with high glucose (HG, 30 mmol L(−1)) + palmitic acid (PA, 100 μmol L(−1)) or C16 ceramide (CER, 20 μmol L(−1)). Interestingly, the cardioprotective effect of ASMase inhibition was not accompanied by reduced ceramide accumulation, indicating a ceramide-independent manner. The mechanism may involve activated NADPH oxidase 4 (NOX4), increased ROS generation and triggered apoptosis. Suppression of NOX4 with apocynin prevented HG + PA and CER incubation induced Nppb and Myh7 pro-hypertrophic gene expression, ROS production and apoptosis in H9c2 cells. Furthermore, cardiomyocyte-specific ASMase knockout (ASMase(Myh6KO)) restored HFD-induced cardiac dysfunction, remodeling, and apoptosis, whereas NOX4 protein expression was downregulated. CONCLUSIONS: These results demonstrated that HFD-mediated activation of cardiomyocyte ASMase could increase NOX4 expression, which may stimulate oxidative stress, apoptosis, and then cause metabolic cardiomyopathy. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12933-023-01747-1. BioMed Central 2023-02-02 /pmc/articles/PMC9896821/ /pubmed/36732747 http://dx.doi.org/10.1186/s12933-023-01747-1 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Liu, Ruijiao
Duan, Tengfei
Yu, Li
Tang, Yongzhong
Liu, Shikun
Wang, Chunjiang
Fang, Wei-Jin
Acid sphingomyelinase promotes diabetic cardiomyopathy via NADPH oxidase 4 mediated apoptosis
title Acid sphingomyelinase promotes diabetic cardiomyopathy via NADPH oxidase 4 mediated apoptosis
title_full Acid sphingomyelinase promotes diabetic cardiomyopathy via NADPH oxidase 4 mediated apoptosis
title_fullStr Acid sphingomyelinase promotes diabetic cardiomyopathy via NADPH oxidase 4 mediated apoptosis
title_full_unstemmed Acid sphingomyelinase promotes diabetic cardiomyopathy via NADPH oxidase 4 mediated apoptosis
title_short Acid sphingomyelinase promotes diabetic cardiomyopathy via NADPH oxidase 4 mediated apoptosis
title_sort acid sphingomyelinase promotes diabetic cardiomyopathy via nadph oxidase 4 mediated apoptosis
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9896821/
https://www.ncbi.nlm.nih.gov/pubmed/36732747
http://dx.doi.org/10.1186/s12933-023-01747-1
work_keys_str_mv AT liuruijiao acidsphingomyelinasepromotesdiabeticcardiomyopathyvianadphoxidase4mediatedapoptosis
AT duantengfei acidsphingomyelinasepromotesdiabeticcardiomyopathyvianadphoxidase4mediatedapoptosis
AT yuli acidsphingomyelinasepromotesdiabeticcardiomyopathyvianadphoxidase4mediatedapoptosis
AT tangyongzhong acidsphingomyelinasepromotesdiabeticcardiomyopathyvianadphoxidase4mediatedapoptosis
AT liushikun acidsphingomyelinasepromotesdiabeticcardiomyopathyvianadphoxidase4mediatedapoptosis
AT wangchunjiang acidsphingomyelinasepromotesdiabeticcardiomyopathyvianadphoxidase4mediatedapoptosis
AT fangweijin acidsphingomyelinasepromotesdiabeticcardiomyopathyvianadphoxidase4mediatedapoptosis