1
|
Zuo JY, Chen HX, Yang Q, Liu ZG, He GW. Tetralogy of Fallot: variants of MYH6 gene promoter and cellular functional analyses. Pediatr Res 2024; 96:338-346. [PMID: 38135727 DOI: 10.1038/s41390-023-02955-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 11/10/2023] [Accepted: 11/18/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND Tetralogy of Fallot (TOF) is a common form of congenital heart disease. The MYH6 gene has important effects on cardiovascular growth and development. METHODS In 608 subjects, including 315 TOF patients, we investigated the MYH6 gene promoter variants and verified the effect on gene expression by using cellular functional experiments with three cell lines (HEK-293, HL-1, and H9C2 cells) and bioinformatics analysis. RESULTS In the MYH6 gene promoter, 12 variants were identified from 608 subjects. Five variants were found only in patients with TOF and two of them (g.3384G>T and g.4518T>C) were novel. Electrophoretic mobility shift assay with three cell lines (HEK-293, HL-1, and H9C2) showed significant changes in the transcription factors bound by the promoter variants compared to the wild-type. Dual luciferase reporter showed that four of the five variants reduced the transcriptional activity of the MYH6 gene promoter (p < 0.05). CONCLUSIONS This study is the first to test the cellular function of variants in the promoter region of the MYH6 gene in patients with TOF, which provides new insights into the genetic basis of TOF and provides a basis for further study of the mechanism of TOF formation. IMPACT DNA from 608 human subjects was sequenced for MYH6 gene promoter region variants with five variants found only in TOF patients and two were novel. EMSA and dual luciferase reporter experiments in three cell lines found these variants pathological. Prediction by JASPAR database indicated that these variants alter the transcription factor binding sites. The study, for the first time, confirmed that there are variants at the MYH6 gene promoter region and these variants alter the cellular function. The variants found in this study suggest the possible pathological role in the formation of TOF.
Collapse
Affiliation(s)
- Ji-Yang Zuo
- The Institute of Cardiovascular Diseases & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, 300457, China
- Clinical School of Cardiovascular Disease, Tianjin Medical University, Tianjin, China
| | - Huan-Xin Chen
- The Institute of Cardiovascular Diseases & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, 300457, China
| | - Qin Yang
- The Institute of Cardiovascular Diseases & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, 300457, China
| | - Zhi-Gang Liu
- The Institute of Cardiovascular Diseases & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin, 300457, China.
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, 300457, China.
| | - Guo-Wei He
- The Institute of Cardiovascular Diseases & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin, 300457, China.
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, 300457, China.
| |
Collapse
|
2
|
Wang J, Wang Z, Xia F, Duan Q, Peng X. Atorvastatin reduces renal interstitial fibrosis caused by unilateral ureteral obstruction through inhibiting the transcriptional activity of YAP. Biochem Biophys Res Commun 2023; 678:109-114. [PMID: 37633180 DOI: 10.1016/j.bbrc.2023.08.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/01/2023] [Accepted: 08/15/2023] [Indexed: 08/28/2023]
Abstract
Renal interstitial fibrosis is the primary pathological basis for the progression and development of various chronic kidney diseases, ultimately leading to renal failure. Obstructive kidney disease caused by conditions such as kidney stones, is a common cause of renal fibrosis. The Hippo pathway is a crucial signaling pathway that senses mechanical forces and is involved in the pathophysiology of fibrosis. In this study, we established a mouse model of obstructive kidney disease induced by unilateral ureteral obstruction (UUO). The UUO procedure significantly upregulated YAP and fibrosis-related gene expression in a time-dependent manner. Morphologically, the renal fibrotic lesions associated with hydronephrosis progressively worsened over time in the UUO group. Atorvastatin, which is widely used to lower blood cholesterol levels, has recently been shown to inhibit Yes1 associated protein (YAP). We treated UUO mice with atorvastatin for 3 and 10 days and observed a decrease in the expression of YAP and fibrosis-related genes at the mRNA and protein levels, along with a reduction in the renal fibrosis analyzed by Masson's staining. These findings suggest that atorvastatin may serve as a preventive agent for fibrosis associated with obstructive kidney disease.
Collapse
Affiliation(s)
- Jun Wang
- First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Zhichao Wang
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China; Jiangxi Hypertension Research Institute, Nanchang, 330006, China
| | - Fan Xia
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China; Jiangxi Hypertension Research Institute, Nanchang, 330006, China
| | - Qiong Duan
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China; Jiangxi Hypertension Research Institute, Nanchang, 330006, China.
| | - Xiaoping Peng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China; Jiangxi Hypertension Research Institute, Nanchang, 330006, China.
| |
Collapse
|
3
|
Chaulin AM. The Negative Effects of Statin Drugs on Cardiomyocytes: Current Review of Laboratory and Experimental Data (Mini-Review). Cardiovasc Hematol Agents Med Chem 2023; 22:7-16. [PMID: 36918788 DOI: 10.2174/1871525721666230314101019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 01/03/2023] [Accepted: 01/30/2023] [Indexed: 03/16/2023]
Abstract
Statin drugs have long been used as a key component of lipid-lowering therapy, which is necessary for the prevention and treatment of atherosclerosis and cardiovascular diseases. Many studies focus on finding and refining new effects of statin drugs. In addition to the main lipidlowering effect (blocking cholesterol synthesis), statin drugs have a number of pleiotropic effects, including negative effects. The main beneficial effects of statin drugs on the components of the cardiovascular system are: anti-ischemic, antithrombotic, anti-apoptotic, antioxidant, endothelioprotective, anti-inflammatory properties, and a number of other beneficial effects. Due to these effects, statin drugs are considered one of the main therapeutic agents for the management of patients with cardiovascular pathologies. To date, many review manuscripts have been published on the myotoxicity, hepatotoxicity, nephrotoxicity, neurotoxicity and diabetogenic effects of statins. However, there are no review manuscripts considering the negative effect of statin drugs on myocardial contractile cells (cardiomyocytes). The purpose of this review is to discuss the negative effects of statin drugs on cardiomyocytes. Special attention is paid to the cardiotoxic action of statin drugs on cardiomyocytes and the mechanisms of increased serum levels of cardiac troponins. In the process of preparing this review, a detailed analysis of laboratory and experimental data devoted to the study of the negative effects of statin drugs on cardiomyocytes was carried out. The literature search was carried out with the keywords: statin drugs, negative effects, mechanisms, cardiac troponins, oxidative stress, apoptosis. Thus, statin drugs can have a number of negative effects on cardiomyocytes, in particular, increased oxidative stress, endoplasmic reticulum stress, damage to mitochondria and intercalated discs, and inhibition of glucose transport into cardiomyocytes. Additional studies are needed to confirm and clarify the mechanisms and clinical consequences of the negative effects of statin drugs on cardiomyocytes.
Collapse
Affiliation(s)
- Aleksey Michailovich Chaulin
- Department of Cardiology and Cardiovascular Surgery, Samara State Medical University, Samara, 443099, Russia
- Department of Histology and Embryology, Samara State Medical University, Samara, 443099, Russia
| |
Collapse
|
4
|
Chaulin AM. Review of Recent Laboratory and Experimental Data on Cardiotoxicity of Statins. J Cardiovasc Dev Dis 2022; 9:403. [PMID: 36421938 PMCID: PMC9696927 DOI: 10.3390/jcdd9110403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/30/2022] [Accepted: 10/08/2022] [Indexed: 11/22/2022] Open
Abstract
Due to the fact that statins are among the most high-demand therapeutic agents used for the treatment and prevention of the most common cardiovascular diseases, a significant amount of research is focused on these drugs. As a result, the study and discovery of new effects in statin drugs continues. Research methods are constantly being improved in terms of their sensitivity and specificity, which leads to a change in ideas. In addition to the main lipid-lowering effect, statins have a number of additional effects, which can be conditionally divided into positive (pleiotropic) and negative (side effects). Moreover, information about many of the pleiotropic effects of statins is controversial and may subsequently change as new data become available. To a large extent, this is due to the introduction of new and the improvement of old methods of study: clinical, laboratory and morphological ones. Recent studies report the possibility of statins having potential cardiotoxic properties, which is expressed by an increase in the concentration of highly sensitive cardiac troponins, as well as various adverse changes in cardiac myocytes at the ultrastructural and molecular levels. This paper discusses possible mechanisms of statin cardiotoxicity. This narrative review is based on an analysis of publications in the Medline, PubMed, PubMed Central and Embase databases. The terms "statins", "troponin", "troponin I", "troponin T" in combination with "cardiotoxicity", "false positive", "mechanisms of increase", "pathophysiological mechanisms", "oxidative stress" and "cardiomyocyte apoptosis" were used to search publications.
Collapse
Affiliation(s)
- Aleksey M. Chaulin
- Department of Histology and Embryology, Samara State Medical University, 443099 Samara, Russia; ; Tel.: +7-(927)-770-25-87
- Department of Cardiology and Cardiovascular Surgery, Samara State Medical University, 443099 Samara, Russia
- Research Institute of Cardiology, Samara State Medical University, 443099 Samara, Russia
| |
Collapse
|
5
|
Katanasaka Y, Hirano S, Sunagawa Y, Miyazaki Y, Sato H, Funamoto M, Shimizu K, Shimizu S, Sari N, Hasegawa K, Morimoto T. Clinically Administered Doses of Pitavastatin and Rosuvastatin. Int Heart J 2021; 62:1379-1386. [PMID: 34853228 DOI: 10.1536/ihj.21-231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Clinical studies have indicated that 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors, also known as statins, can potentially inhibit chronic heart failure. In the Stat-LVDF study, a difference was noted in terms of the effect of lipophilic pitavastatin (PTV) and hydrophilic rosuvastatin (RSV) on plasma BNP, suggesting that statin lipophilicity and pharmacokinetics change the pleiotropic effect on heart failure in humans. Therefore, we assessed the beneficial effects of PTV on hypertrophy in cardiac myocytes compared with RSV at clinically used doses. Cultured cardiomyocytes were stimulated with 30 μM phenylephrine (PE) in the presence of PTV (250 nM) or RSV (50 nM). These doses were calculated based on the maximum blood concentration of statins used in clinical situations in Japan. The results showed that PTV, but not RSV, significantly inhibits the PE-induced increase in cell size and leucine incorporation without causing cell toxicity. In addition, PTV significantly suppressed PE-induced mRNA expression of hypertrophic response genes. PE-induced ERK phosphorylation was inhibited by PTV, but not by RSV. Furthermore, PTV significantly suppressed the angiotensin-II-induced proline incorporation in primary cultured cardiac fibroblasts. In conclusion, a clinical dose of PTV was noted to directly inhibit cardiomyocyte hypertrophy and cardiac fibrosis, suggesting that lipophilic PTV can be a potential drug candidate against chronic heart failure.
Collapse
Affiliation(s)
- Yasufumi Katanasaka
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
- Division of Translational Research, Clinical Research Institute, Kyoto Medical Center, National Hospital Organization
- Shizuoka General hospital
| | - Sae Hirano
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
| | - Yoichi Sunagawa
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
- Division of Translational Research, Clinical Research Institute, Kyoto Medical Center, National Hospital Organization
- Shizuoka General hospital
| | - Yusuke Miyazaki
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
- Division of Translational Research, Clinical Research Institute, Kyoto Medical Center, National Hospital Organization
- Shizuoka General hospital
| | - Hikaru Sato
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
| | - Masafumi Funamoto
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
- Division of Translational Research, Clinical Research Institute, Kyoto Medical Center, National Hospital Organization
| | - Kana Shimizu
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
- Division of Translational Research, Clinical Research Institute, Kyoto Medical Center, National Hospital Organization
| | - Satoshi Shimizu
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
- Division of Translational Research, Clinical Research Institute, Kyoto Medical Center, National Hospital Organization
| | - Nurmila Sari
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
- Division of Translational Research, Clinical Research Institute, Kyoto Medical Center, National Hospital Organization
| | - Koji Hasegawa
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
- Division of Translational Research, Clinical Research Institute, Kyoto Medical Center, National Hospital Organization
| | - Tatsuya Morimoto
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka
- Division of Translational Research, Clinical Research Institute, Kyoto Medical Center, National Hospital Organization
- Shizuoka General hospital
| |
Collapse
|
6
|
Sun Y, Jin Z, Zhang X, Cui T, Zhang W, Shao S, Li H, Wang N. GATA Binding Protein 3 Is a Direct Target of Kruppel-Like Transcription Factor 7 and Inhibits Chicken Adipogenesis. Front Physiol 2020; 11:610. [PMID: 32587528 PMCID: PMC7298121 DOI: 10.3389/fphys.2020.00610] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/15/2020] [Indexed: 12/21/2022] Open
Abstract
Kruppel-like transcription factor 7 (KLF7) is a negative regulator of adipogenesis, however, its precise mechanism is poorly understood. Our previous KLF7 ChIP-seq analysis showed that one of the KLF7 binding peaks was present upstream of GATA binding protein 3 (GATA3) in chicken preadipocytes. In the present study, we identified GATA3 as a target of KLF7. Overexpression analysis showed KLF7 markedly enhanced the endogenous expression of GATA3 in the immortalized chicken preadipcyte cell line (ICP2), and the luciferase reporter assay showed that KLF7 overexpression increased the reporter gene activity of the cloned upstream region (-5285/-4336 relative to the translation initiation codon ATG) of GATA3 in ICP2 and DF1 cells, and mutation of the putative KLF7 binding site abolished the promotive effect of KLF7 overexpression on the reporter gene activity of the cloned GATA3 upstream region. ChIP-qPCR further demonstrated that KLF7 directly bound to the GATA3 upstream region. Gene expression analysis showed that GATA3 mRNA expression in abdominal adipose tissue was significantly higher in lean chicken line than in the fat line at 2, 3, and 6 weeks of age. In addition, GATA3 mRNA expression markedly decreased during the preadipocyte differentiation. Furthermore, a functional study showed that GATA3 overexpression inhibited the differentiation of the ICP2 cells. Taken together, our results demonstrated that KLF7 inhibits chicken adipogenesis, at least in part through direct upregulation of GATA3.
Collapse
Affiliation(s)
- Yingning Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.,College of Life Science and Agriculture Forestry, Qiqihar University, Qiqihar, China.,Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China
| | - Zhao Jin
- College of Life Science and Agriculture Forestry, Qiqihar University, Qiqihar, China
| | - Xinyang Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.,Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China
| | - Tingting Cui
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.,College of Life Science and Agriculture Forestry, Qiqihar University, Qiqihar, China.,Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China
| | - Wenjian Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.,Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China
| | - Shuli Shao
- College of Life Science and Agriculture Forestry, Qiqihar University, Qiqihar, China
| | - Hui Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.,Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China
| | - Ning Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.,Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China
| |
Collapse
|
7
|
Zhang J, Wang J, Yu H, Wang G, Zhang J, Zhu R, Liu X, Li J. Comparison between Atorvastatin and Rosuvastatin on Secondary Percutaneous Coronary Intervention Rate and the Risk Factors in Patients with Coronary Heart Disease. Curr Drug Metab 2020; 21:818-828. [PMID: 32156233 DOI: 10.2174/1389200221666200310110410] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/29/2019] [Accepted: 01/06/2020] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The aim is to compare atorvastatin versus rosuvastatin on secondary percutaneous coronary intervention (PCI) rate and explore risk factors in coronary heart disease (CHD) patients. METHODS A cohort study with 283 CHD subjects was launched from 2011 to 2015. Cox proportional hazards regression model, Receiver Operating Characteristic (ROC) and nomogram were used to compare the effect of atorvastatin and rosuvastatin on secondary PCI rate and disease risk factors. Even why the two statins had different effects based on gene expression profile analysis has been explored. RESULTS Gene FFA (Freely fatty acid), AST (Aspartate Transaminase) and ALT (Alanine transaminase) showed the statistical difference between the four statin groups (P<0.05). In the AA group (Continuous Atorvastatin usage), albumin was a risk factor (Hazard Ratio (HR):1.076, 95%CI (1.001, 1.162), p<0.05). In the AR group (Start with Atorvastatin usage, then change to Rosuvastatin usage), ApoA was a protective factor (HR:0.004, 95%CI (0.001, 0.665), p<0.05). GLB (Galactosidase Beta) was a risk factor (HR:1.262, 95%CI (1.010, 1.576), p<0.05). In RR group (Continuous Rosuvastatin usage), ApoE was a protective factor (HR:0.943, 95%CI (0.890, 1.000), <0.05). ALT was a risk factor (HR:1.030, 95%CI (1.000, 1.060), p<0.05). CONCLUSION Patients in the RA group had the lowest secondary PCI rate. ALT was a risk factor in the RR group. Gene Gpt (Glutamic Pyruvic Transaminase) encoded for one subtype of ALT had a significantly different expression in different statin groups.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Cardiology, Shanghai Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200065, China
| | - Jiaqi Wang
- Institute of Clinical Epidemiology and Evidence-based Medicine, Tongji University School of Medicine, Shanghai, 200092, China
| | - Han Yu
- Department of Cardiology, Shanghai Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200065, China
| | - Guanghua Wang
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 201204, China
| | - Junfang Zhang
- Institute of Clinical Epidemiology and Evidence-based Medicine, Tongji University School of Medicine, Shanghai, 200092, China
| | - Rui Zhu
- Department of Cardiology, Shanghai Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200065, China
| | - Xuebo Liu
- Department of Cardiology, Shanghai Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200065, China
| | - Jue Li
- Department of Cardiology, Shanghai Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200065, China
| |
Collapse
|
8
|
Godoy JC, Niesman IR, Busija AR, Kassan A, Schilling JM, Schwarz A, Alvarez EA, Dalton ND, Drummond JC, Roth DM, Kararigas G, Patel HH, Zemljic-Harpf AE. Atorvastatin, but not pravastatin, inhibits cardiac Akt/mTOR signaling and disturbs mitochondrial ultrastructure in cardiac myocytes. FASEB J 2018; 33:1209-1225. [PMID: 30169110 DOI: 10.1096/fj.201800876r] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Statins, which reduce LDL-cholesterol by inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A reductase, are among the most widely prescribed drugs. Skeletal myopathy is a known statin-induced adverse effect associated with mitochondrial changes. We hypothesized that similar effects would occur in cardiac myocytes in a lipophilicity-dependent manner between 2 common statins: atorvastatin (lipophilic) and pravastatin (hydrophilic). Neonatal cardiac ventricular myocytes were treated with atorvastatin and pravastatin for 48 h. Both statins induced endoplasmic reticular (ER) stress, but only atorvastatin inhibited ERK1/2T202/Y204, AktSer473, and mammalian target of rapamycin signaling; reduced protein abundance of caveolin-1, dystrophin, epidermal growth factor receptor, and insulin receptor-β; decreased Ras homolog gene family member A activation; and induced apoptosis. In cardiomyocyte-equivalent HL-1 cells, atorvastatin, but not pravastatin, reduced mitochondrial oxygen consumption. When male mice underwent atorvastatin and pravastatin administration per os for up to 7 mo, only long-term atorvastatin, but not pravastatin, induced elevated serum creatine kinase; swollen, misaligned, size-variable, and disconnected cardiac mitochondria; alteration of ER structure; repression of mitochondria- and endoplasmic reticulum-related genes; and a 21% increase in mortality in cardiac-specific vinculin-knockout mice during the first 2 months of administration. To our knowledge, we are the first to demonstrate in vivo that long-term atorvastatin administration alters cardiac ultrastructure, a finding with important clinical implications.-Godoy, J. C., Niesman, I. R., Busija, A. R., Kassan, A., Schilling, J. M., Schwarz, A., Alvarez, E. A., Dalton, N. D., Drummond, J. C., Roth, D. M., Kararigas, G., Patel, H. H., Zemljic-Harpf, A. E. Atorvastatin, but not pravastatin, inhibits cardiac Akt/mTOR signaling and disturbs mitochondrial ultrastructure in cardiac myocytes.
Collapse
Affiliation(s)
- Joseph C Godoy
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Ingrid R Niesman
- Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Anna R Busija
- Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Adam Kassan
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, West Coast University, North Hollywood, California, USA
| | - Jan M Schilling
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Anna Schwarz
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Erika A Alvarez
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Nancy D Dalton
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - John C Drummond
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - David M Roth
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Georgios Kararigas
- Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Hemal H Patel
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| | - Alice E Zemljic-Harpf
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Anesthesiology, University of California, San Diego, San Diego, California, USA
| |
Collapse
|
9
|
El-Shinnawy NA, Abd Elhalem SS, Haggag NZ, Badr G. Ameliorative role of camel whey protein and rosuvastatin on induced dyslipidemia in mice. Food Funct 2018; 9:1038-1047. [PMID: 29349446 DOI: 10.1039/c7fo01871a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The incidence of obesity is rapidly increasing throughout the world. Dyslipidemia is a major risk factor for a number of chronic diseases, including diabetes and cardiovascular diseases. This work presents a novel approach to study the activity of camel whey protein (WP) with antioxidant and anti-inflammatory properties as a cheap dietary protein substance extracted from camel milk to produce satiety and help in building muscles. Mice model suffering from dyslipidemia as a result of feeding on high fat-cholesterol diet for 8 weeks were administrated with either camel WP and/or rosuvastatin for 4 weeks. Dyslipidemia revealed significant increase in anthropometrical measurements, levels of glucose, insulin, cholesterol, triglycerides, low-density lipoprotein, total leucocyte count, inflammatory cytokines and reactive oxygen species, accompanied by a significant elevation in activating transcription factor-3 and inducible nitric oxide synthase expressions. These alterations were correlated with a profound reduction in high-density lipoprotein, peroxisome proliferator-activated receptor alpha and adiponectin along with a decrease in liver and muscle mitochondrial proteins. Rosuvastatin treatment to mice suffering from dyslipidemia in combination with camel WP for 4 weeks ameliorated these parameters. Notably, animals treated with both camel WP and rosuvastatin exhibited a remarkable decrease in the incidence of dyslipidemia. In addition, camel WP succeeded to overcome the therapeutic drawback posed from rosuvastatin therapy alone with minimal side effects.
Collapse
Affiliation(s)
- Nashwa Ahmed El-Shinnawy
- Zoology Department, Faculty of Women for Arts, Science and Education, Ain Shams University, 11757 Cairo, Egypt.
| | | | | | | |
Collapse
|
10
|
Perrino C, Barabási AL, Condorelli G, Davidson SM, De Windt L, Dimmeler S, Engel FB, Hausenloy DJ, Hill JA, Van Laake LW, Lecour S, Leor J, Madonna R, Mayr M, Prunier F, Sluijter JPG, Schulz R, Thum T, Ytrehus K, Ferdinandy P. Epigenomic and transcriptomic approaches in the post-genomic era: path to novel targets for diagnosis and therapy of the ischaemic heart? Position Paper of the European Society of Cardiology Working Group on Cellular Biology of the Heart. Cardiovasc Res 2018; 113:725-736. [PMID: 28460026 PMCID: PMC5437366 DOI: 10.1093/cvr/cvx070] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 04/27/2017] [Indexed: 01/19/2023] Open
Abstract
Despite advances in myocardial reperfusion therapies, acute myocardial ischaemia/reperfusion injury and consequent ischaemic heart failure represent the number one cause of morbidity and mortality in industrialized societies. Although different therapeutic interventions have been shown beneficial in preclinical settings, an effective cardioprotective or regenerative therapy has yet to be successfully introduced in the clinical arena. Given the complex pathophysiology of the ischaemic heart, large scale, unbiased, global approaches capable of identifying multiple branches of the signalling networks activated in the ischaemic/reperfused heart might be more successful in the search for novel diagnostic or therapeutic targets. High-throughput techniques allow high-resolution, genome-wide investigation of genetic variants, epigenetic modifications, and associated gene expression profiles. Platforms such as proteomics and metabolomics (not described here in detail) also offer simultaneous readouts of hundreds of proteins and metabolites. Isolated omics analyses usually provide Big Data requiring large data storage, advanced computational resources and complex bioinformatics tools. The possibility of integrating different omics approaches gives new hope to better understand the molecular circuitry activated by myocardial ischaemia, putting it in the context of the human ‘diseasome’. Since modifications of cardiac gene expression have been consistently linked to pathophysiology of the ischaemic heart, the integration of epigenomic and transcriptomic data seems a promising approach to identify crucial disease networks. Thus, the scope of this Position Paper will be to highlight potentials and limitations of these approaches, and to provide recommendations to optimize the search for novel diagnostic or therapeutic targets for acute ischaemia/reperfusion injury and ischaemic heart failure in the post-genomic era.
Collapse
Affiliation(s)
- Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Via Pansini 5, 80131 Naples, Italy
| | - Albert-Laszló Barabási
- Center for Complex Networks Research and Department of Physics, Northeastern University, Boston, MA, USA.,Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.,Center for Network Science, Central European University, Budapest, Hungary.,Department of Medicine, and Division of Network Medicine, Brigham and Womens Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Gianluigi Condorelli
- Department of Cardiovascular Medicine, Humanitas Research Hospital and Humanitas University, Rozzano, Italy.,Institute of Genetic and Biomedical Research, National Research Council of Italy, Rozzano, Milan, Italy
| | - Sean Michael Davidson
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK
| | - Leon De Windt
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, University Frankfurt, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), RheinMain, Germany
| | - Felix Benedikt Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Derek John Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, UK.,The National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, UK.,Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore.,Yong Loo Lin School of Medicine, National University Singapore, Singapore.,Barts Heart Centre, St Bartholomew's Hospital, London, UK
| | - Joseph Addison Hill
- Departments of Medicine (Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Linda Wilhelmina Van Laake
- Division of Heart and Lungs, Hubrecht Institute, University Medical Center Utrecht, Utrecht, The Netherlands.,UMC Utrecht Regenerative Medicine Center and Hubrecht Institute, Utrecht, The Netherlands
| | - Sandrine Lecour
- Hatter Cardiovascular Research Institute, University of Cape Town, Cape Town, South Africa
| | - Jonathan Leor
- Neufeld Cardiac Research Institute, Tel-Aviv University, Tel-Aviv, Israel.,Tamman Cardiovascular Research Institute, Sheba Medical Center; Sheba Center for Regenerative Medicine, Stem Cell, and Tissue Engineering, Tel Hashomer, Israel
| | - Rosalinda Madonna
- Center of Aging Sciences and Translational Medicine - CESI-MeT, "G. d'Annunzio" University, Chieti, Italy; Institute of Cardiology, Department of Neurosciences, Imaging, and Clinical Sciences, "G. d'Annunzio" University, Chieti, Italy.,The Texas Heart Institute and Center for Cardiovascular Biology and Atherosclerosis Research, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Manuel Mayr
- King's British Heart Foundation Centre, King's College London, London, UK
| | - Fabrice Prunier
- Department of Cardiology, Institut MITOVASC, University of Angers, University Hospital of Angers, Angers, France
| | - Joost Petrus Geradus Sluijter
- Cardiology and UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Kirsti Ytrehus
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged, Hungary.,Pharmahungary Group, Szeged, Hungary
| |
Collapse
|