AccScience Publishing / GTM / Volume 1 / Issue 2 / DOI: 10.36922/gtm.v1i2.137
ORIGINAL RESEARCH ARTICLE

Comparative proteomic analysis of hearts from mice with high-fat diet-induced metabolic cardiomyopathy

Zongzhe Jiang1 Mingyang Pang2 Wei Huang2,3,4*
Show Less
1 Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
2 Department of Cardiology, Chinese PLA Central War Command General Hospital, Wuhan, 430 010, China
3 Division of Cardiology, Department of Internal Medicine, and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430 010, China
4 Gene Therapy Center and the Institute of Hypertension, Huazhong University of Science and Technology, Wuhan, 430 010, China
Global Translational Medicine 2022, 1(2), 137 https://doi.org/10.36922/gtm.v1i2.137
Submitted: 17 June 2022 | Accepted: 12 August 2022 | Published: 15 September 2022
© 2022 by the Author(s). This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Abstract

In patients with obesity or type 2 diabetes, the accumulation of lipotoxic by-products in cardiomyocytes leads to apoptosis and contractile dysfunction, eventually resulting in metabolic cardiomyopathy (MC). However, the underlying mechanisms remain unclear. Inb this study, a comparative proteome analysis was conducted to evaluate the differentially expressed proteins (DEPs) in the hearts of normal mice on standard diet (control group) and of high-fat diet (HFD)-induced MC mice (HFD group). We identified 90 DEPs unique to the control group and 18 DEPs unique to the HFD group. In 90 DEPs unique to the control group, only 74 DEPs were annotated in the gene ontology (GO) database. These annotated DEPs are involved in 114 biological processes, 68 molecular functions, and 174 cellular components. In 18 DEPs unique to the HFD group, only 14 DEPs were annotated in the GO database. These annotated DEPs are involved in 24 biological processes, 22 molecular functions, and six cellular components. Protein levels of two fatty acid metabolism-related enzymes, carnitine palmitoyltransferase 1B (CPT1B) and acetyl-CoA acyltransferase 2 (ACAA2), in the hearts of the mice in control group and HFD group were analyzed by immunostaining and Western blot. The results showed that the protein levels of CPT1B and ACAA2 were elevated in hearts of the mice in HFD group, which were consistent with the proteomic analysis. Our results reveal the differentially expressed proteome related to the progression of MC, providing a series of potential therapeutic targets for MC.

Keywords
High-fat diet
Metabolic cardiomyopathy
Proteome analysis
Funding
Sichuan Science and Technology Department
Doctoral Research Initiation Fund of Affiliated Hospital of Southwest Medical University
References
[1]

Ogurtsova K, Da Rocha Fernandes JD, Huang Y, et al., 2017, IDF diabetes atlas: global estimates for the prevalence of diabetes for 2015 and 2040. Diabetes Res. Clin. Pract, 128: 40–50. https://doi.org/10.1016/j.diabres.2017.03.024

[2]

Nishida K, Otsu K, 2017, Inflammation and metabolic cardiomyopathy. Cardiovasc Res, 113: 389–398. https://doi.org/10.1093/cvr/cvx012

[3]

Maack C, Murphy E, 2017, Metabolic cardiomyopathies— fighting the next epidemic. Cardiovasc Res, 113: 367–369. https://doi.org/10.1093/cvr/cvx022

[4]

Obokata M, Reddy YNV, Pislaru SV, et al., 2017, Evidence supporting the existence of a distinct obese phenotype of heart failure with preserved ejection fraction., Circulation, 136: 6–19. https://doi.org/10.1161/CIRCULATIONAHA.116.026807

[5]

Ponikowski P, Voors AA, Anker SD, et al., 2016, 2016 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure: the task force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC). Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J, 37: 2129–2200. https://doi.org/10.1093/eurheartj/ehw128

[6]

Isfort M, Stevens SC, Schaffer S, et al., 2014, Metabolic dysfunction in dia betic cardiomyopathy. Heart Fail Rev, 19: 35–48. https://doi.org/10.1007/s10741-013-9377-8

[7]

Seferovic PM, Paulus WJ, 2015, Clinical diabetic cardiomyopathy: A two-faced disease with restrictive and dilated phenotypes. Eur Heart J, 36: 1718–1727, 1727a–1727c. https://doi.org/10.1093/eurheartj/ehv134

[8]

Authors/Task Force Members, Ryden L, Grant PJ, et al., 2013, ESC guidelines on diabetes, prediabetes, and cardiovascular diseases developed in collaboration with the EASD: the task force on diabetes, pre-diabetes, and cardiovascular diseases of the European Society of Cardiology (ESC) and developed in collaboration with the European association for the study of Diabetes (EASD). Eur Heart J, 34: 3035–3087. https://doi.org/10.1093/eurheartj/eht108

[9]

Schulze PC, Drosatos K, Goldberg IJ, 2016, Lipid use and misuse by the heart. Circ Res, 118: 1736–1751. https://doi.org/10.1161/CIRCRESAHA.116.306842

[10]

Gibbs EM, Stock JL, McCoid SC, et al., 1995, Glycemic improvement in diabetic db/db mice by overexpression of the human insulinregulatable glucose transporter (GLUT4). J Clin Invest, 95: 1512–1218. https://doi.org/10.1172/JCI117823

[11]

Niture SK, Khatri R, Jaiswal AK, 2014, Regulation of Nrf2-an update. Free Radic Biol Med, 66: 36–44. https://doi.org/10.1016/j.freeradbiomed.2013.02.008

[12]

Tan Y, Ichikawa T, Li J, et al., 2011, Diabetic downregulation of Nrf2 activity via ERK contributes to oxidative stress-induced insulin resistance in cardiac cells in vitro and in vivo. Diabetes, 60: 625–633. https://doi.org/10.2337/db10-1164

[13]

Jia G, DeMarco VG, Sowers JR, 2016, Insulin resistance and hyperinsulinaemia in diabetic cardiomyopathy. Nat Rev Endocrinol, 12: 144–53. https://doi.org/10.1038/nrendo.2015.216

[14]

Wang Y, Zhou S, Sun W, et al., 2014, Inhibition of JNK by novel curcumin analog C66 prevents diabetic cardiomyopathy with a preservation of cardiac metallothionein expression. Am J Physiol Endocrinol Metab, 306: E1239–E1247. https://doi.org/10.1152/ajpendo.00629.2013

[15]

Winzell MS, Ahren B, 2004, The high-fat diet-fed mouse: A model for studying mecha-nisms and treatment of impaired glucose tolerance and Type 2 diabetes. Diabetes, 53: S215–S219. https://doi.org/10.2337/diabetes.53.suppl_3.s215

[16]

Kanehisa M, Furumichi M, Tanabe M, et al., 2017, KEGG: New perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res, 45(D1): D353–D61. https://doi.org/10.1093/nar/gkw1092

[17]

Costantino S, Akhmedov A, Melina G, et al., 2019, Obesity-induced activation of JunD promotes myocardial lipid accumulation and metabolic cardiomyopathy. Eur Heart J, 40: 997–1008. https://doi.org/10.1093/eurheartj/ehy903

[18]

Bardella C, Pollard P, Tomlinson I, 2011, SDH mutations in cancer. Biochim Biophys Acta, 1807: 1432–1443. https://doi.org/10.1016/j.bbabio.2011.07.003

[19]

Burnichon N, Briere JJ, Libe R, et al., 2010, Tissier, Sdha is a tumor suppressor gene causing paraganglioma. Human Mol Genet, 19: 3011–3020. https://doi.org/10.1093/hmg/ddq206

[20]

Gill AJ, Pachter NS, Chou A, et al., 2011, Tucker, renal tumors associated with germline sdhb mutation show distinctive morphology. Am J Surg Pathol, 35: 1578–1585. https://doi.org/10.1097/PAS.0b013e318227e7f4

[21]

Ricketts CJ, Shuch B, Vocke CD, et al., 2012, Middelton, succinate dehydrogenase kidney cancer: an aggressive example of the warburg effffect in cancer. J Urol, 188: 2063–2071. https://doi.org/10.1016/j.juro.2012.08.030

[22]

Kropotov A, Usmanova N, Serikov V, et al., 2007, Mitochondrial targeting of human peroxiredoxin V protein and regulation of PRDX5 gene expression by nuclear transcription factors controlling biogenesis of mitochondria. FEBS J, 274: 5804–5814. https://doi.org/10.1111/j.1742-4658.2007.06103.x

[23]

Wu J, Tang Q, Shen J, et al., 2010, Comparative proteome profile during the early period of small-for-size liver transplantation in rats revealed the protective role of Prdx5. J Hepatol, 53: 73–83. https://doi.org/10.1016/j.jhep.2010.01.032

[24]

Sun Y, Fan W, Xue R, 2020, Transcribed ultraconserved regions, Uc.323, ameliorates cardiac hypertrophy by regulating the transcription of CPT1b (Carnitine Palmitoyl transferase 1b). Hypertension, 75: 79–90. https://doi.org/10.1161/HYPERTENSIONAHA.119.13173

[25]

He L, Kim T, Long Q, et al., 2012, Carnitine palmitoyltransferase-1b deficiency aggravates pressure overload-induced cardiac hypertrophy caused by lipotoxicity. Circulation, 126: 1705–1716. https://doi.org/10.1161/CIRCULATIONAHA.111.075978

[26]

Miltiadou D, Hager-Theodorides AL, Symeou S, et al., 2017, Variants in the 3’ untranslated region of the ovine acetyl-coenzyme a acyltransferase 2 gene are associated with dairy traits and exhibit differential allelic expression. J Dairy Sci, 100: 6285–6297. https://doi.org/10.3168/jds.2016-12326

Conflict of interest
The authors declare that there are no conflicts of interest regarding the publication of this paper.
Share
Back to top
Global Translational Medicine, Electronic ISSN: 2811-0021 Published by AccScience Publishing