101
|
Kwa FA, Jackson DE. Manipulating the epigenome for the treatment of disorders with thrombotic complications. Drug Discov Today 2018; 23:719-726. [DOI: 10.1016/j.drudis.2018.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/11/2017] [Accepted: 01/04/2018] [Indexed: 11/25/2022]
|
102
|
Egemnazarov B, Crnkovic S, Nagy BM, Olschewski H, Kwapiszewska G. Right ventricular fibrosis and dysfunction: Actual concepts and common misconceptions. Matrix Biol 2018; 68-69:507-521. [PMID: 29343458 DOI: 10.1016/j.matbio.2018.01.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 01/10/2018] [Indexed: 12/25/2022]
Abstract
Fibrosis and remodeling of the right ventricle (RV) are associated with RV dysfunction and mortality of patients with pulmonary hypertension (PH) but it is unknown how much RV fibrosis contributes to RV dysfunction and mortality. RV fibrosis manifests as fibroblast accumulation and collagen deposition which may be excessive. Although extracellular matrix deposition leads to elevated ventricular stiffness, it is not known to which extent it affects RV function. Various animal models of pulmonary hypertension have been established to investigate the role of fibrosis in RV dysfunction and failure. However, they do not perfectly resemble the human disease. In the current review we describe the major characteristics of RV fibrosis, molecular mechanisms regulating the fibrotic process, and discuss how therapeutic targeting of fibrosis might affect RV function.
Collapse
Affiliation(s)
| | - Slaven Crnkovic
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Bence M Nagy
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Horst Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria; Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria; Institute of Physiology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
103
|
Li Y, Zhang Z, Zhou X, Li R, Cheng Y, Shang B, Han Y, Liu B, Xie X. Histone Deacetylase 1 Inhibition Protects Against Hypoxia-Induced Swelling in H9c2 Cardiomyocytes Through Regulating Cell Stiffness. Circ J 2017; 82:192-202. [PMID: 28747611 DOI: 10.1253/circj.cj-17-0022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND The process of cardiomyocyte swelling involves changes of biomechanical properties and profiles of cellular genes. Although many genes have been proved to regulate cell edema of cardiomyocyte, the mechanisms involved in this event, as well as the biomechanical properties of swelling cell, remain unknown. METHODS AND RESULTS Whether histone deacetylase 1 (HDAC1) inhibition protects against hypoxia-induced H9c2 cardiomyocyte swelling is examined in this study. Hypoxia-induced changes in the biomechanical properties and cytoskeletal structure that are relevant to cell swelling were also determined. H9c2 cells were treated under a chemical hypoxia situation (cobalt chloride) with HDAC1 inhibition (chemical inhibitor or siRNA) for 5 h, followed by in vitro biological and mechanical characterization. The results showed that expression of HDAC1 instead of HDAC4 was upregulated by chemical hypoxia. HDAC1 inhibition protects H9c2 cells against chemical hypoxia-induced hypoxic injury and cell swelling. HDAC1 inhibition improved cell viability, decreased lactate dehydrogenase leakage, cell apoptosis, malondialdehyde concentration, cell volume, and particles on the cell surface, and increased superoxide dismutase activity. Moreover, chemical hypoxia induced a decrease of Young's modulus, accompanied by alterations in the integrity of acetylated histone and organization of the cytoskeletal network. HDAC1 inhibition significantly reversed these processes. CONCLUSIONS Based on the ideal physical model, HDAC1 inhibition protects against hypoxia-induced swelling in H9c2 cardiomyocytes through enhancing cell stiffness. Overall, HDAC1 is a potential therapeutic target for myocardial edema.
Collapse
Affiliation(s)
- Yi Li
- The Institute of Medical Genetics, School of Basic Medical Sciences, Lanzhou University
- Gansu Cardiovascular Institute
| | - Zhengyi Zhang
- Cardiac Hospital, Lanzhou University Second Hospital
| | - Xiangnan Zhou
- School of Physics and Information Engineering, Shanxi Normal University
| | - Rui Li
- School of Stomatology, Lanzhou University
| | - Yan Cheng
- Experimental Center, Northwest University for Nationalities
- Department of Biochemistry and Medical Genetics, University of Manitoba
| | - Bo Shang
- Cardiac Hospital, Lanzhou University Second Hospital
| | - Yu Han
- College of Life Science & Technology, Huazhong University of Science & Technology
| | - Bin Liu
- School of Stomatology, Lanzhou University
| | - Xiaodong Xie
- The Institute of Medical Genetics, School of Basic Medical Sciences, Lanzhou University
- Gansu Cardiovascular Institute
| |
Collapse
|
104
|
Yan H, Yi S, Zhuang H, Wu L, Wang DW, Jiang J. Sphingosine-1-phosphate ameliorates the cardiac hypertrophic response through inhibiting the activity of histone deacetylase-2. Int J Mol Med 2017; 41:1704-1714. [PMID: 29286094 DOI: 10.3892/ijmm.2017.3325] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 11/29/2017] [Indexed: 11/06/2022] Open
Abstract
Inhibition of histone deacetylase-2 (HDAC2), which is a prohypertrophic factor in the heart, can functionally attenuate cardiac hypertrophy. The present study aimed to investigate whether sphingosine‑1‑phosphate (S1P), which has recently been reported to suppress HDAC2 activity, could ameliorate the cardiac hypertrophic response and improve cardiac function in mice with transverse aortic constriction (TAC), as well as to determine the underlying mechanisms. Briefly, 8‑week‑old male C57BL/6 mice were randomly divided into sham, TAC and TAC + S1P groups; the results indicated that S1P treatment attenuated TAC‑induced cardiac dysfunction. In addition, heart size and the expression levels of fetal cardiac genes were reduced in the TAC + S1P group compared with in the TAC group. Furthermore, in cultured H9c2 cells exposed to phenylephrine, S1P was revealed to decrease cardiomyocyte size and the exaggerated expression of fetal cardiac genes. The present study also demonstrated that S1P had no effect on HDAC2 expression, but it did suppress its activity and increase acetylation of histone H3 in vivo and in vitro. Krüppel‑like factor 4 (KLF4) is an antihypertrophic transcriptional regulator, which mediates HDAC inhibitor‑induced prevention of cardiac hypertrophy; in the present study, KLF4 was upregulated by S1P. Finally, the results indicated that S1P receptor 2 (S1PR2) may be involved in the antihypertrophic effects, whereas the suppressive effects of S1P on HDAC2 activity were independent of S1PR2. In conclusion, the present study demonstrated that S1P treatment may ameliorate the cardiac hypertrophic response, which may be partly mediated by the suppression of HDAC2 activity and the upregulation of KLF4; it was suggested that S1PR2 may also be involved. Therefore, S1P may be considered a potential therapy for the treatment of heart diseases caused by cardiac hypertrophy.
Collapse
Affiliation(s)
- Hui Yan
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Shaowei Yi
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Hang Zhuang
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Lujin Wu
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Dao Wen Wang
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jiangang Jiang
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
105
|
Liu Y, Li S, Zhang Z, Lv Z, Jiang H, Tan X, Liu F. Effects of valproic acid on sympathetic activity and left ventricularmyocardial remodelling in rats during pressure overload. Turk J Med Sci 2017; 47:1651-1660. [PMID: 29152949 DOI: 10.3906/sag-1704-142] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Background/aim: Pressure overload induces cardiac remodelling and results in heart failure. Enhanced sympathetic outflow participates in the development of cardiac remodelling for the duration of pressure overload as an independent factor. Valproic acid has recently been shown to reduce neuronal injury and have antiinflammatory and antiapoptotic effects as a histone deacetylase inhibitor. We speculate that the drug plays a specific role in alleviating cardiac remodelling by inhibiting sympathetic activity. Materials and methods: Surgery of partial abdominal aortic constriction was performed on male Sprague-Dawley rats. After 4 weeks, animal models of pressure overload were validated and then valproic acid (300 mg/kg) was administrated to rats once a day for the next 4 weeks. Experimental parameters were detected 4 weeks after validation. Results: The administration of valproic acid alleviated cardiomyocyte hypertrophy, myocardial interstitial fibrosis and left ventricular diastolic dysfunction caused by partial abdominal aortic constriction. Valproic acid reduced the levels of plasma and local norepinephrine, augmented concentrations of hypothalamic gamma-aminobutyric acid, and had no side effects on the hepatic and renal function of the animals. Conclusion: These results suggest that valproic acid may be a safe and effective therapeutic strategy for the inhibition of sympathetic outflow and cardiac remodelling.
Collapse
|
106
|
Kwon DH, Kim YK, Kook H. New Aspects of Vascular Calcification: Histone Deacetylases and Beyond. J Korean Med Sci 2017; 32:1738-1748. [PMID: 28960024 PMCID: PMC5639052 DOI: 10.3346/jkms.2017.32.11.1738] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 08/19/2017] [Indexed: 11/20/2022] Open
Abstract
Vascular calcification is a pathologic phenomenon in which calcium phosphate is ectopically deposited in the arteries. Previously, calcification was considered to be a passive process in response to metabolic diseases, vascular or valvular diseases, or even aging. However, now calcification is recognized as a highly-regulated consequence, like bone formation, and many clinical trials have been carried out to elucidate the correlation between vascular calcification and cardiovascular events and mortality. As a result, vascular calcification has been implicated as an independent risk factor in cardiovascular diseases. Many molecules are now known to be actively associated with this process. Recently, our laboratory found that posttranslational modification of histone deacetylase (HDAC) 1 is actively involved in the development of vascular calcification. In addition, we found that modulation of the activity of HDAC as well as its protein stability by MDM2, an HDAC1-E3 ligase, may be a therapeutic target in vascular calcification. In the present review, we overview the pathomechanism of vascular calcification and the involvement of posttranslational modification of epigenetic regulators.
Collapse
Affiliation(s)
- Duk Hwa Kwon
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, Korea
- Basic Research Laboratory for Cardiac Remodeling, Chonnam National University Medical School, Gwangju, Korea
| | - Young Kook Kim
- Basic Research Laboratory for Cardiac Remodeling, Chonnam National University Medical School, Gwangju, Korea
- Center for Creative Biomedical Scientists at Chonnam National University, Gwangju, Korea
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, Korea
| | - Hyun Kook
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, Korea
- Basic Research Laboratory for Cardiac Remodeling, Chonnam National University Medical School, Gwangju, Korea.
| |
Collapse
|
107
|
Pereyra AS, Hasek LY, Harris KL, Berman AG, Damen FW, Goergen CJ, Ellis JM. Loss of cardiac carnitine palmitoyltransferase 2 results in rapamycin-resistant, acetylation-independent hypertrophy. J Biol Chem 2017; 292:18443-18456. [PMID: 28916721 DOI: 10.1074/jbc.m117.800839] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/05/2017] [Indexed: 12/20/2022] Open
Abstract
Cardiac hypertrophy is closely linked to impaired fatty acid oxidation, but the molecular basis of this link is unclear. Here, we investigated the loss of an obligate enzyme in mitochondrial long-chain fatty acid oxidation, carnitine palmitoyltransferase 2 (CPT2), on muscle and heart structure, function, and molecular signatures in a muscle- and heart-specific CPT2-deficient mouse (Cpt2M-/-) model. CPT2 loss in heart and muscle reduced complete oxidation of long-chain fatty acids by 87 and 69%, respectively, without altering body weight, energy expenditure, respiratory quotient, or adiposity. Cpt2M-/- mice developed cardiac hypertrophy and systolic dysfunction, evidenced by a 5-fold greater heart mass, 60-90% reduction in blood ejection fraction relative to control mice, and eventual lethality in the absence of cardiac fibrosis. The hypertrophy-inducing mammalian target of rapamycin complex 1 (mTORC1) pathway was activated in Cpt2M-/- hearts; however, daily rapamycin exposure failed to attenuate hypertrophy in Cpt2M-/- mice. Lysine acetylation was reduced by ∼50% in Cpt2M-/- hearts, but trichostatin A, a histone deacetylase inhibitor that improves cardiac remodeling, failed to attenuate Cpt2M-/- hypertrophy. Strikingly, a ketogenic diet increased lysine acetylation in Cpt2M-/- hearts 2.3-fold compared with littermate control mice fed a ketogenic diet, yet it did not improve cardiac hypertrophy. Together, these results suggest that a shift away from mitochondrial fatty acid oxidation initiates deleterious hypertrophic cardiac remodeling independent of fibrosis. The data also indicate that CPT2-deficient hearts are impervious to hypertrophy attenuators, that mitochondrial metabolism regulates cardiac acetylation, and that signals derived from alterations in mitochondrial metabolism are the key mediators of cardiac hypertrophic growth.
Collapse
Affiliation(s)
| | | | | | - Alycia G Berman
- the Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907
| | - Frederick W Damen
- the Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907
| | - Craig J Goergen
- the Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907
| | | |
Collapse
|
108
|
Anti-fibrotic effects of valproic acid in experimental peritoneal fibrosis. PLoS One 2017; 12:e0184302. [PMID: 28873458 PMCID: PMC5584960 DOI: 10.1371/journal.pone.0184302] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 08/21/2017] [Indexed: 01/06/2023] Open
Abstract
Background Progressive fibrous thickening of the peritoneal membrane is a complication of long-term peritoneal dialysis (PD). TGF-β/Smad pathway activation, inflammation, and neoangiogenesis play important roles in peritoneal membrane (PM) changes induced by PD. Recently, histone deacetilase inhibitors (HDACi) have shown anti-fibrotic and anti-inflammatory effects in different experimental models. These drugs prevent deacetylation of histones causing a loosen chromatin, which in turn induce the expression of some anti-fibrotic genes. In addition, acetylation may increase the activity of proteins involved in tissue fibrosis, such as Smad7. Here, we explored the effect of valproic acid (VPA), an HDACi, on the development of peritoneal fibrosis (PF) in rats. Methods PF was induced by daily intraperitoneal injections of 0.1% chlorhexidine gluconate (CG) for 15 consecutive days. Male Wistar rats (250–300 g) were divided into 3 groups: CONTROL, control rats receiving only vehicle; PF, peritoneal fibrosis induced in rats; PF+VPA, rats with PF treated with VPA (300 mg/kg/day by gavage). PF was assessed by Masson’s trichrome staining. Inflammation and fibrosis-associated factors were assessed by immunohistochemistry, immunofluorescence, multiplex analysis, and qPCR. Results Treatment with VPA significantly reduced PM thickness and the expression of myofibroblasts, besides preventing loss of ultrafiltration capacity of the PM. The upregulation of profibrotic factors (TGF-β, fibronectin, and Smad3) in the PF group was significantly ameliorated by VPA. VPA modulated the TGF/Smad pathway, inhibiting phosphorylated Smad3 expression and inducing an increased Smad7 expression in the FP+VPA group. The neoangiogenesis and the expression of proinflammatory cytokines (TNF-α, IL-1β, MCP-1) observed in the PF group was significantly reduced by VPA. Conclusions Our results indicate that VPA suppressed experimental PF through modulation of the TGF-β/Smad pathway. Interestingly, VPA treatment induced a higher expression of antifibrotic factors, such as Smad7. These results suggest that VPA may represent a potential strategy for treating long term PD complications.
Collapse
|
109
|
Wang Z, Zhu Q, Wang W, Yi F, Li PL, Boini KM, Li N. Infusion of Valproic Acid Into the Renal Medulla Activates Stem Cell Population and Attenuates Salt-Sensitive Hypertension in Dahl S Rats. Cell Physiol Biochem 2017; 42:1264-1273. [PMID: 28693025 DOI: 10.1159/000478955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 04/25/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Our previous study has detected a stem cell deficiency in the renal medulla in Dahl salt-sensitive (S) rats. This study determined whether infusion of valproic acid (VA), an agent known to stimulate the stem cell function, attenuated salt-sensitive hypertension in Dahl S rats. METHODS Uninephrectomized Dahl S rats were infused with vehicle or VA (50mg/kg/d) into the renal medulla and fed with a low (LS) or high salt diet (HS). Stem cell marker and number were analyzed by immunohistochemistry, Real-time RT-PCR and Western blot. Sodium excretion and blood pressure were measured. RESULTS VA significantly increased the mRNA and protein levels of FGF2, a stem cell niche factor, and CD133, a stem cell marker. The number of CD133+ cells was significantly increased in the renal medulla in VA-treated rats. Meanwhile, high salt-induced increases in the mRNA level of proinflammatory factors interleukin-1β and interleukin-6 were blocked in VA-treated rats. Functionally, sodium excretion in response to the blood pressure increase and acute sodium loading was significantly enhanced, sodium retention attenuated, high salt-induced increase of blood pressure reduced in VA-treated rats. CONCLUSION Activation of stem cell function by VA inhibits the activation of proinflammatory factors and attenuates salt-sensitive hypertension in Dahl S rats.
Collapse
Affiliation(s)
- Zhengchao Wang
- Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, China.,Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Qing Zhu
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA.,Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Weili Wang
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Fan Yi
- Department of Pharmacology, Shandong University School of Medicine, Jinan, China
| | - Pin-Lan Li
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Krishna M Boini
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Ningjun Li
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
110
|
Gallic acid attenuates hypertension, cardiac remodeling, and fibrosis in mice with N G-nitro-L-arginine methyl ester-induced hypertension via regulation of histone deacetylase 1 or histone deacetylase 2. J Hypertens 2017; 35:1502-1512. [DOI: 10.1097/hjh.0000000000001327] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
111
|
Early transcriptional alteration of histone deacetylases in a murine model of doxorubicin-induced cardiomyopathy. PLoS One 2017; 12:e0180571. [PMID: 28662206 PMCID: PMC5491252 DOI: 10.1371/journal.pone.0180571] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 06/16/2017] [Indexed: 12/22/2022] Open
Abstract
Doxorubicin is a potent chemotherapeutic agent that is widely-used to treat a variety of cancers but causes acute and chronic cardiac injury, severely limiting its use. Clinically, the acute side effects of doxorubicin are mostly manageable, whereas the delayed consequences can lead to life-threatening heart failure, even decades after cancer treatment. The cardiotoxicity of doxorubicin is subject to a critical cumulative dose and so dosage limitation is considered to be the best way to reduce these effects. Hence, a number of studies have defined a "safe dose" of the drug, both in animal models and clinical settings, with the aim of avoiding long-term cardiac effects. Here we show that a dose generally considered as safe in a mouse model can induce harmful changes in the myocardium, as early as 2 weeks after infusion. The adverse changes include the development of fibrotic lesions, disarray of cardiomyocytes and a major transcription dysregulation. Importantly, low-dose doxorubicin caused specific changes in the transcriptional profile of several histone deacetylases (HDACs) which are epigenetic regulators of cardiac remodelling. This suggests that cardioprotective therapies, aimed at modulating HDACs during doxorubicin treatment, deserve further exploration.
Collapse
|
112
|
Zhang L, Du J, Yano N, Wang H, Zhao YT, Dubielecka PM, Zhuang S, Chin YE, Qin G, Zhao TC. Sodium Butyrate Protects -Against High Fat Diet-Induced Cardiac Dysfunction and Metabolic Disorders in Type II Diabetic Mice. J Cell Biochem 2017; 118:2395-2408. [PMID: 28109123 DOI: 10.1002/jcb.25902] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 01/18/2017] [Indexed: 12/31/2022]
Abstract
Histone deacetylases are recently identified to act as key regulators for cardiac pathophysiology and metabolic disorders. However, the function of histone deacetylase (HDAC) in controlling cardiac performance in Type II diabetes and obesity remains unknown. Here, we determine whether HDAC inhibition attenuates high fat diet (HFD)-induced cardiac dysfunction and improves metabolic features. Adult mice were fed with either HFD or standard chow food for 24 weeks. Starting at 12 weeks, mice were divided into four groups randomly, in which sodium butyrate (1%), a potent HDAC inhibitor, was provided to chow and HFD-fed mice in drinking water, respectively. Glucose intolerance, metabolic parameters, cardiac function, and remodeling were assessed. Histological analysis and cellular signaling were examined at 24 weeks following euthanization of mice. HFD-fed mice demonstrated myocardial dysfunction and profound interstitial fibrosis, which were attenuated by HDAC inhibition. HFD-induced metabolic syndrome features insulin resistance, obesity, hyperinsulinemia, hyperglycemia, lipid accumulations, and cardiac hypertrophy, these effects were prevented by HDAC inhibition. Furthermore, HDAC inhibition attenuated myocyte apoptosis, reduced production of reactive oxygen species, and increased angiogenesis in the HFD-fed myocardium. Notably, HFD induced decreases in MKK3, p38, p38 regulated/activated protein kinase (PRAK), and Akt-1, but not p44/42 phosphorylation, which were prevented by HDAC inhibition. These results suggest that HDAC inhibition plays a critical role to preserve cardiac performance and mitigate metabolic disorders in obesity and diabetes, which is associated with MKK3/p38/PRAK pathway. The study holds promise in developing a new therapeutic strategy in the treatment of Type II diabetic-induced heart failure and metabolic disorders. J. Cell. Biochem. 118: 2395-2408, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Surgery, Roger Williams Medical Center, Boston University School of Medicine, Providence, Rhode Island.,Department of Emergency Medicine, Rhode Island Hospital, Providence, Rhode Island
| | - Jianfeng Du
- Department of Surgery, Roger Williams Medical Center, Boston University School of Medicine, Providence, Rhode Island
| | - Naohiro Yano
- Women and Infants Hospital, Brown University, Providence, Rhode Island
| | - Hao Wang
- Department of Surgery, Roger Williams Medical Center, Boston University School of Medicine, Providence, Rhode Island
| | - Yu Tina Zhao
- Department of Surgery, Roger Williams Medical Center, Boston University School of Medicine, Providence, Rhode Island
| | | | - Shougang Zhuang
- Department of Medicine, Rhode Island Hospital, Providence, Rhode Island
| | - Y Eugene Chin
- Key Laboratory of Stem Cell Biology, Institutes of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Gangjian Qin
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ting C Zhao
- Department of Surgery, Roger Williams Medical Center, Boston University School of Medicine, Providence, Rhode Island
| |
Collapse
|
113
|
Bauer AJ, Martin KA. Coordinating Regulation of Gene Expression in Cardiovascular Disease: Interactions between Chromatin Modifiers and Transcription Factors. Front Cardiovasc Med 2017; 4:19. [PMID: 28428957 PMCID: PMC5382160 DOI: 10.3389/fcvm.2017.00019] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 03/20/2017] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease is a leading cause of death with increasing economic burden. The pathogenesis of cardiovascular diseases is complex, but can arise from genetic and/or environmental risk factors. This can lead to dysregulated gene expression in numerous cell types including cardiomyocytes, endothelial cells, vascular smooth muscle cells, and inflammatory cells. While initial studies addressed transcriptional control of gene expression, epigenetics has been increasingly appreciated to also play an important role in this process through alterations in chromatin structure and gene accessibility. Chromatin-modifying proteins including enzymes that modulate DNA methylation, histone methylation, and histone acetylation can influence gene expression in numerous ways. These chromatin modifiers and their marks can promote or prevent transcription factor recruitment to regulatory regions of genes through modifications to DNA, histones, or the transcription factors themselves. This review will focus on the emerging question of how epigenetic modifiers and transcription factors interact to coordinately regulate gene expression in cardiovascular disease. While most studies have addressed the roles of either epigenetic or transcriptional control, our understanding of the integration of these processes is only just beginning. Interrogating these interactions is challenging, and improved technical approaches will be needed to fully dissect the temporal and spatial relationships between transcription factors, chromatin modifiers, and gene expression in cardiovascular disease. We summarize the current state of the field and provide perspectives on limitations and future directions. Through studies of epigenetic and transcriptional interactions, we can advance our understanding of the basic mechanisms of cardiovascular disease pathogenesis to develop novel therapeutics.
Collapse
Affiliation(s)
- Ashley J Bauer
- Department of Medicine (Cardiovascular Medicine), Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA.,Department of Pharmacology, Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| | - Kathleen A Martin
- Department of Medicine (Cardiovascular Medicine), Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA.,Department of Pharmacology, Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
114
|
Schuetze KB, Stratton MS, Blakeslee WW, Wempe MF, Wagner FF, Holson EB, Kuo YM, Andrews AJ, Gilbert TM, Hooker JM, McKinsey TA. Overlapping and Divergent Actions of Structurally Distinct Histone Deacetylase Inhibitors in Cardiac Fibroblasts. J Pharmacol Exp Ther 2017; 361:140-150. [PMID: 28174211 PMCID: PMC5363768 DOI: 10.1124/jpet.116.237701] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 01/23/2017] [Indexed: 01/05/2023] Open
Abstract
Inhibitors of zinc-dependent histone deacetylases (HDACs) profoundly affect cellular function by altering gene expression via changes in nucleosomal histone tail acetylation. Historically, investigators have employed pan-HDAC inhibitors, such as the hydroxamate trichostatin A (TSA), which simultaneously targets members of each of the three zinc-dependent HDAC classes (classes I, II, and IV). More recently, class- and isoform-selective HDAC inhibitors have been developed, providing invaluable chemical biology probes for dissecting the roles of distinct HDACs in the control of various physiologic and pathophysiological processes. For example, the benzamide class I HDAC-selective inhibitor, MGCD0103 [N-(2-aminophenyl)-4-[[(4-pyridin-3-ylpyrimidin-2-yl)amino]methyl] benzamide], was shown to block cardiac fibrosis, a process involving excess extracellular matrix deposition, which often results in heart dysfunction. Here, we compare the mechanisms of action of structurally distinct HDAC inhibitors in isolated primary cardiac fibroblasts, which are the major extracellular matrix-producing cells of the heart. TSA, MGCD0103, and the cyclic peptide class I HDAC inhibitor, apicidin, exhibited a common ability to enhance histone acetylation, and all potently blocked cardiac fibroblast cell cycle progression. In contrast, MGCD0103, but not TSA or apicidin, paradoxically increased expression of a subset of fibrosis-associated genes. Using the cellular thermal shift assay, we provide evidence that the divergent effects of HDAC inhibitors on cardiac fibroblast gene expression relate to differential engagement of HDAC1- and HDAC2-containing complexes. These findings illustrate the importance of employing multiple compounds when pharmacologically assessing HDAC function in a cellular context and during HDAC inhibitor drug development.
Collapse
Affiliation(s)
- Katherine B Schuetze
- Division of Cardiology and Consortium for Fibrosis Research and Translation, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado (K.B.S., M.S.S., W.W.B., T.A.M.); Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical, Sciences, University of Colorado Denver, Aurora, Colorado (M.F.W.); Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts (F.F.W., E.B.H.); Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania (Y.-M.K., A.J.A.); and Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts (T.M.G., J.M.H.)
| | - Matthew S Stratton
- Division of Cardiology and Consortium for Fibrosis Research and Translation, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado (K.B.S., M.S.S., W.W.B., T.A.M.); Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical, Sciences, University of Colorado Denver, Aurora, Colorado (M.F.W.); Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts (F.F.W., E.B.H.); Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania (Y.-M.K., A.J.A.); and Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts (T.M.G., J.M.H.)
| | - Weston W Blakeslee
- Division of Cardiology and Consortium for Fibrosis Research and Translation, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado (K.B.S., M.S.S., W.W.B., T.A.M.); Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical, Sciences, University of Colorado Denver, Aurora, Colorado (M.F.W.); Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts (F.F.W., E.B.H.); Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania (Y.-M.K., A.J.A.); and Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts (T.M.G., J.M.H.)
| | - Michael F Wempe
- Division of Cardiology and Consortium for Fibrosis Research and Translation, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado (K.B.S., M.S.S., W.W.B., T.A.M.); Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical, Sciences, University of Colorado Denver, Aurora, Colorado (M.F.W.); Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts (F.F.W., E.B.H.); Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania (Y.-M.K., A.J.A.); and Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts (T.M.G., J.M.H.)
| | - Florence F Wagner
- Division of Cardiology and Consortium for Fibrosis Research and Translation, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado (K.B.S., M.S.S., W.W.B., T.A.M.); Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical, Sciences, University of Colorado Denver, Aurora, Colorado (M.F.W.); Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts (F.F.W., E.B.H.); Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania (Y.-M.K., A.J.A.); and Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts (T.M.G., J.M.H.)
| | - Edward B Holson
- Division of Cardiology and Consortium for Fibrosis Research and Translation, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado (K.B.S., M.S.S., W.W.B., T.A.M.); Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical, Sciences, University of Colorado Denver, Aurora, Colorado (M.F.W.); Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts (F.F.W., E.B.H.); Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania (Y.-M.K., A.J.A.); and Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts (T.M.G., J.M.H.)
| | - Yin-Ming Kuo
- Division of Cardiology and Consortium for Fibrosis Research and Translation, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado (K.B.S., M.S.S., W.W.B., T.A.M.); Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical, Sciences, University of Colorado Denver, Aurora, Colorado (M.F.W.); Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts (F.F.W., E.B.H.); Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania (Y.-M.K., A.J.A.); and Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts (T.M.G., J.M.H.)
| | - Andrew J Andrews
- Division of Cardiology and Consortium for Fibrosis Research and Translation, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado (K.B.S., M.S.S., W.W.B., T.A.M.); Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical, Sciences, University of Colorado Denver, Aurora, Colorado (M.F.W.); Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts (F.F.W., E.B.H.); Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania (Y.-M.K., A.J.A.); and Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts (T.M.G., J.M.H.)
| | - Tonya M Gilbert
- Division of Cardiology and Consortium for Fibrosis Research and Translation, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado (K.B.S., M.S.S., W.W.B., T.A.M.); Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical, Sciences, University of Colorado Denver, Aurora, Colorado (M.F.W.); Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts (F.F.W., E.B.H.); Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania (Y.-M.K., A.J.A.); and Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts (T.M.G., J.M.H.)
| | - Jacob M Hooker
- Division of Cardiology and Consortium for Fibrosis Research and Translation, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado (K.B.S., M.S.S., W.W.B., T.A.M.); Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical, Sciences, University of Colorado Denver, Aurora, Colorado (M.F.W.); Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts (F.F.W., E.B.H.); Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania (Y.-M.K., A.J.A.); and Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts (T.M.G., J.M.H.)
| | - Timothy A McKinsey
- Division of Cardiology and Consortium for Fibrosis Research and Translation, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado (K.B.S., M.S.S., W.W.B., T.A.M.); Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical, Sciences, University of Colorado Denver, Aurora, Colorado (M.F.W.); Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts (F.F.W., E.B.H.); Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania (Y.-M.K., A.J.A.); and Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts (T.M.G., J.M.H.)
| |
Collapse
|
115
|
Chen Z, Li S, Subramaniam S, Shyy JYJ, Chien S. Epigenetic Regulation: A New Frontier for Biomedical Engineers. Annu Rev Biomed Eng 2017; 19:195-219. [PMID: 28301736 DOI: 10.1146/annurev-bioeng-071516-044720] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Gene expression in mammalian cells depends on the epigenetic status of the chromatin, including DNA methylation, histone modifications, promoter-enhancer interactions, and noncoding RNA-mediated regulation. The coordinated actions of these multifaceted regulations determine cell development, cell cycle regulation, cell state and fate, and the ultimate responses in health and disease. Therefore, studies of epigenetic modulations are critical for our understanding of gene regulation mechanisms at the molecular, cellular, tissue, and organ levels. The aim of this review is to provide biomedical engineers with an overview of the principles of epigenetics, methods of study, recent findings in epigenetic regulation in health and disease, and computational and sequencing tools for epigenetics analysis, with an emphasis on the cardiovascular system. This review concludes with the perspectives of the application of bioengineering to advance epigenetics and the utilization of epigenetics to translate bioengineering research into clinical medicine.
Collapse
Affiliation(s)
- Zhen Chen
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, Duarte, California 91016; .,Department of Medicine, University of California at San Diego, La Jolla, California 92093; ,
| | - Shuai Li
- Department of Medicine, University of California at San Diego, La Jolla, California 92093; ,
| | - Shankar Subramaniam
- Department of Bioengineering and Institute of Engineering in Medicine, University of California at San Diego, La Jolla, California 92093; ,
| | - John Y-J Shyy
- Department of Medicine, University of California at San Diego, La Jolla, California 92093; ,
| | - Shu Chien
- Department of Medicine, University of California at San Diego, La Jolla, California 92093; , .,Department of Bioengineering and Institute of Engineering in Medicine, University of California at San Diego, La Jolla, California 92093; ,
| |
Collapse
|
116
|
Hulsurkar M, Li Z, Zhang Y, Li X, Zheng D, Li W. Beta-adrenergic signaling promotes tumor angiogenesis and prostate cancer progression through HDAC2-mediated suppression of thrombospondin-1. Oncogene 2017; 36:1525-1536. [PMID: 27641328 DOI: 10.1038/onc.2016.319] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 06/22/2016] [Accepted: 07/22/2016] [Indexed: 12/19/2022]
Abstract
Chronic behavioral stress and beta-adrenergic signaling have been shown to promote cancer progression, whose underlying mechanisms are largely unclear, especially the involvement of epigenetic regulation. Histone deacetylase-2 (HDAC2), an epigenetic regulator, is critical for stress-induced cardiac hypertrophy. It is unknown whether it is necessary for beta-adrenergic signaling-promoted cancer progression. Using xenograft models, we showed that chronic behavioral stress and beta-adrenergic signaling promote angiogenesis and prostate cancer progression. HDAC2 was induced by beta-adrenergic signaling in vitro and in mouse xenografts. We next uncovered that HDAC2 is a direct target of cAMP response element-binding protein (CREB) that is activated by beta-adrenergic signaling. Notably, HDAC2 is necessary for beta-adrenergic signaling to induce angiogenesis. We further demonstrated that, upon CREB activation, HDAC2 represses thrombospondin-1 (TSP1), a potent angiogenesis inhibitor, through epigenetic regulation. Together, these data establish a novel pathway that HDAC2 and TSP1 act downstream of CREB activation in beta-adrenergic signaling to promote cancer progression.
Collapse
Affiliation(s)
- M Hulsurkar
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| | - Z Li
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
- Breast and Thyroid Surgery Center, The Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Y Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
- The Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - X Li
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
- The Liaocheng People's Hospital, Liaocheng Clinical School of Taishan Medical University, Liaocheng, China
| | - D Zheng
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Medical Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - W Li
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| |
Collapse
|
117
|
Ghosh AK, Rai R, Flevaris P, Vaughan DE. Epigenetics in Reactive and Reparative Cardiac Fibrogenesis: The Promise of Epigenetic Therapy. J Cell Physiol 2017; 232:1941-1956. [PMID: 27883184 DOI: 10.1002/jcp.25699] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/21/2016] [Indexed: 12/20/2022]
Abstract
Epigenetic changes play a pivotal role in the development of a wide spectrum of human diseases including cardiovascular diseases, cancer, diabetes, and intellectual disabilities. Cardiac fibrogenesis is a common pathophysiological process seen during chronic and stress-induced accelerated cardiac aging. While adequate production of extracellular matrix (ECM) proteins is necessary for post-injury wound healing, excessive synthesis and accumulation of extracellular matrix protein in the stressed or injured hearts causes decreased or loss of lusitropy that leads to cardiac failure. This self-perpetuating deposition of collagen and other matrix proteins eventually alter cellular homeostasis; impair tissue elasticity and leads to multi-organ failure, as seen during pathogenesis of cardiovascular diseases, chronic kidney diseases, cirrhosis, idiopathic pulmonary fibrosis, and scleroderma. In the last 25 years, multiple studies have investigated the molecular basis of organ fibrosis and highlighted its multi-factorial genetic, epigenetic, and environmental regulation. In this minireview, we focus on five major epigenetic regulators and discuss their central role in cardiac fibrogenesis. Additionally, we compare and contrast the epigenetic regulation of hypertension-induced reactive fibrogenesis and myocardial infarction-induced reparative or replacement cardiac fibrogenesis. As microRNAs-one of the major epigenetic regulators-circulate in plasma, we also advocate their potential diagnostic role in cardiac fibrosis. Lastly, we discuss the evolution of novel epigenetic-regulating drugs and predict their clinical role in the suppression of pathological cardiac remodeling, cardiac aging, and heart failure. J. Cell. Physiol. 232: 1941-1956, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Asish K Ghosh
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Rahul Rai
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Panagiotis Flevaris
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Douglas E Vaughan
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
118
|
Godoy LD, Lucas JE, Bender AJ, Romanick SS, Ferguson BS. Targeting the epigenome: Screening bioactive compounds that regulate histone deacetylase activity. Mol Nutr Food Res 2017; 61. [PMID: 27981795 DOI: 10.1002/mnfr.201600744] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 11/07/2016] [Accepted: 12/05/2016] [Indexed: 12/18/2022]
Abstract
SCOPE Nutrigenomics is a rapidly expanding field that elucidates the link between diet-genome interactions. Recent evidence demonstrates that regulation of the epigenome, and in particular inhibition of histone deacetylases (HDACs), impact pathogenetic mechanisms involved in chronic disease. Few studies, to date, have screened libraries of bioactive compounds that act as epigenetic modifiers. This study screened a library of 131 natural compounds to determine bioactive compounds that inhibit Zn-dependent HDAC activity. METHODS AND RESULTS Using class-specific HDAC substrates, we screened 131 natural compounds for HDAC activity in bovine cardiac tissue. From this screen, we identified 18 bioactive compound HDAC inhibitors. Using our class-specific HDAC substrates, we next screened these 18 bioactive compounds against recombinant HDAC proteins. Consistent with inhibition of HDAC activity, these compounds were capable of inhibiting activity of individual HDAC isoforms. Lastly, we report that treatment of H9c2 cardiac myoblasts with bioactive HDAC inhibitors was sufficient to increase lysine acetylation as assessed via immunoblot. CONCLUSION This study provided the first step in identifying multiple bioactive compound HDAC inhibitors. Taken together, this report sets the stage for future exploration of these bioactive compounds as epigenetic regulators to potentially ameliorate chronic disease.
Collapse
Affiliation(s)
- Luis D Godoy
- Department Agriculture, Nutrition, & Veterinary Sciences, University of Nevada, Reno, NV, USA
| | - Julianna E Lucas
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, NV, USA
| | - Abigail J Bender
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, NV, USA
| | | | - Bradley S Ferguson
- Department Agriculture, Nutrition, & Veterinary Sciences, University of Nevada, Reno, NV, USA
| |
Collapse
|
119
|
Raghunathan S, Goyal RK, Patel BM. Selective inhibition of HDAC2 by magnesium valproate attenuates cardiac hypertrophy. Can J Physiol Pharmacol 2016; 95:260-267. [PMID: 28177689 DOI: 10.1139/cjpp-2016-0542] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The regulatory paradigm in cardiac hypertrophy involves alterations in gene expression that is mediated by chromatin remodeling. Various data suggest that class I and class II histone deacetylases (HDACs) play opposing roles in the regulation of hypertrophic pathways. To address this, we tested the effect of magnesium valproate (MgV), an HDAC inhibitor with 5 times more potency on class I HDACs. Cardiac hypertrophy was induced by partial abdominal aortic constriction in Wistar rats, and at the end of 6 weeks, we evaluated hypertrophic, hemodynamic, and oxidative stress parameters, and mitochondrial DNA concentration. Treatment with MgV prevented cardiac hypertrophy, improved hemodynamic functions, prevented oxidative stress, and increased mitochondrial DNA concentration. MgV treatment also increased the survival rate of the animals as depicted by the Kaplan-Meier curve. Improvement in hypertrophy due to HDAC inhibition was further confirmed by HDAC mRNA expression studies, which revealed that MgV decreases expression of pro-hypertrophic HDAC (i.e., HDAC2) without altering the expression of anti-hypertrophic HDAC5. Selective class I HDAC inhibition is required for controlling cardiac hypertrophy. Newer HDAC inhibitors that are class I inhibitors and class II promoters can be designed to obtain "pan" or "dual" natural HDAC "regulators".
Collapse
Affiliation(s)
| | - Ramesh K Goyal
- b Delhi Pharmaceutical Sciences Research University, Delhi, India
| | - Bhoomika M Patel
- a Institute of Pharmacy, Nirma University, Ahmedabad 382 481, India
| |
Collapse
|
120
|
Thiagarajan D, Vedantham S, Ananthakrishnan R, Schmidt AM, Ramasamy R. Mechanisms of transcription factor acetylation and consequences in hearts. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1862:2221-2231. [PMID: 27543804 PMCID: PMC5159280 DOI: 10.1016/j.bbadis.2016.08.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 08/12/2016] [Accepted: 08/14/2016] [Indexed: 01/06/2023]
Abstract
Acetylation of proteins as a post-translational modification is gaining rapid acceptance as a cellular control mechanism on par with other protein modification mechanisms such as phosphorylation and ubiquitination. Through genetic manipulations and evolving proteomic technologies, identification and consequences of transcription factor acetylation is beginning to emerge. In this review, we summarize the field and discuss newly unfolding mechanisms and consequences of transcription factor acetylation in normal and stressed hearts. This article is part of a Special Issue entitled: The role of post-translational protein modifications on heart and vascular metabolism edited by Jason R.B. Dyck & Jan F.C. Glatz.
Collapse
Affiliation(s)
- Devi Thiagarajan
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, NYU Langone Medical Center, NY, New York 10016, United States
| | | | - Radha Ananthakrishnan
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, NYU Langone Medical Center, NY, New York 10016, United States
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, NYU Langone Medical Center, NY, New York 10016, United States
| | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, NYU Langone Medical Center, NY, New York 10016, United States.
| |
Collapse
|
121
|
Choi SY, Piao ZH, Jin L, Kim JH, Kim GR, Ryu Y, Lin MQ, Kim HS, Kee HJ, Jeong MH. Piceatannol Attenuates Renal Fibrosis Induced by Unilateral Ureteral Obstruction via Downregulation of Histone Deacetylase 4/5 or p38-MAPK Signaling. PLoS One 2016; 11:e0167340. [PMID: 27902771 PMCID: PMC5130266 DOI: 10.1371/journal.pone.0167340] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/11/2016] [Indexed: 02/05/2023] Open
Abstract
Piceatannol, a resveratrol metabolite, is a phenolic compound found in red wine and grapes. We investigated the effect of piceatannol on renal fibrosis and histone deacetylase (HDAC) expression in a mouse model of unilateral ureteral obstruction (UUO). Fibrosis was established by UUO and piceatannol was intraperitoneally injected for 2 weeks. Piceatannol suppressed extracellular matrix (ECM) protein deposition including collagen type I and fibronectin as well as connective tissue growth factor (CTGF) and α-smooth muscle actin (α-SMA) in UUO kidneys. However, the expressions of epithelial-mesenchymal transition (EMT) marker genes, such as N-cadherin and E-cadherin, were not changed in the kidneys after UUO. Masson’s trichrome staining and fluorescence immunostaining showed that piceatannol administration attenuated collagen deposition in UUO kidneys. HDAC1, HDAC4, HDAC5, HDAC6, and HDAC10 protein expression was upregulated in UUO kidneys, whereas that of HDAC8 was downregulated. Piceatannol treatment significantly reduced HDAC4 and HDAC5 protein expression. Further, piceatannol attenuated phosphorylation of p38 mitogen-activated protein kinase (p38-MAPK) in UUO kidneys, but not that of transforming growth factor beta1-Smad2/3. These results suggest that class I HDACs and class IIa/b HDACs are involved in renal fibrosis development. Piceatannol may be a beneficial therapeutic agent for treating renal fibrosis via reduction of HDAC4 and HDAC5 protein expression or suppression of the p38-MAPK signaling pathway.
Collapse
Affiliation(s)
- Sin Young Choi
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Zhe Hao Piao
- The Second Hospital of Jilin University, Changchun, China
| | - Li Jin
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- Jilin Hospital Affiliated with Jilin University, Jilin, China
| | - Jung Ha Kim
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Gwi Ran Kim
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Yuhee Ryu
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Ming Quan Lin
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- Yanbian University Hospital, Jilin Yanbian, China
| | - Hyung-Seok Kim
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Hae Jin Kee
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- * E-mail: (HJK); (MHJ)
| | - Myung Ho Jeong
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- * E-mail: (HJK); (MHJ)
| |
Collapse
|
122
|
Irisin Ameliorates Hypoxia/Reoxygenation-Induced Injury through Modulation of Histone Deacetylase 4. PLoS One 2016; 11:e0166182. [PMID: 27875543 PMCID: PMC5119735 DOI: 10.1371/journal.pone.0166182] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/24/2016] [Indexed: 11/30/2022] Open
Abstract
Irisin is a recently identified myokine which brings increases in energy expenditure and contributes to the beneficial effects of exercise through the browning of white adipose tissues. However, its effects in the heart remains unknown. This study sought to determine the effects of irisin on hypoxia/reoxygenation injury and its relationship with HDAC4. Wild type and stable HDAC4-overexpression cells were generated from H9c2 cardiomyoblasts. HDAC4 overexpression cells and wild type H9c2 cells were exposed to 24 hours of hypoxia followed by one hour of reoxygenation in vitro in the presence or absence of irisin (5 ng/ml). Cell cytotoxicity, apoptosis, mitochondrial respiration, and mitochondrial permeability transition pore (mPTP) were determined. Western blotting was employed to determine active-caspase 3, annexin V, and HDAC4 expression. As compared to wild type H9c2 group, HDAC4 overexpression remarkably led to a great increase in cell death as evident by the increased lactate dehydrogenase (LDH) leakage, ratio of caspase-3-positive cells as well as the upregulated levels of active-caspase 3 and annexin V shown by western blot analysis. In addition, HDAC4 overexpression also induced much severe mitochondrial dysfunction, as indicated by apoptotic mitochondria and increased mPTP. However, irisin treatment significantly attenuated all of these effects. Though irisin treatment did not influence the expression of HDAC4 at the transcriptional level, western blot analysis showed that HDAC4 protein levels decreased in a time-dependent way after administration of irisin, which is associated with the degradation of HDAC4 mediated by small ubiquitin-like modification (SUMO). Our results are the first to demonstrate that the protective effects of irisin in cardiomyoblasts exposed to hypoxia/reoxygenation might be associated with HDAC4 degradation.
Collapse
|
123
|
Chistiakov DA, Orekhov AN, Bobryshev YV. Treatment of cardiovascular pathology with epigenetically active agents: Focus on natural and synthetic inhibitors of DNA methylation and histone deacetylation. Int J Cardiol 2016; 227:66-82. [PMID: 27852009 DOI: 10.1016/j.ijcard.2016.11.204] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 11/06/2016] [Indexed: 12/20/2022]
Abstract
Cardiovascular disease (CVD) retains a leadership as a major cause of human death worldwide. Although a substantial progress was attained in the development of cardioprotective and vasculoprotective drugs, a search for new efficient therapeutic strategies and promising targets is under way. Modulation of epigenetic CVD mechanisms through administration epigenetically active agents is one of such new approaches. Epigenetic mechanisms involve heritable changes in gene expression that are not linked to the alteration of DNA sequence. Pathogenesis of CVDs is associated with global genome-wide changes in DNA methylation and histone modifications. Epigenetically active compounds that influence activity of epigenetic modulators such as DNA methyltransferases (DNMTs), histone acetyltransferases, histone deacetylases (HDACs), etc. may correct these pathogenic changes in the epigenome and therefore be used for CVD therapy. To date, many epigenetically active natural substances (such as polyphenols and flavonoids) and synthetic compounds such as DNMT inhibitors or HDAC inhibitors are known. Both native and chemical DNMT and HDAC inhibitors possess a wide range of cytoprotective activities such as anti-inflammatory, antioxidant, anti-apoptotic, anti-anfibrotic, and anti-hypertrophic properties, which are beneficial of treatment of a variety of CVDs. However, so far, only synthetic DNMT inhibitors enter clinical trials while synthetic HDAC inhibitors are still under evaluation in preclinical studies. In this review, we consider epigenetic mechanisms such as DNA methylation and histone modifications in cardiovascular pathology and the epigenetics-based therapeutic approaches focused on the implementation of DNMT and HDAC inhibitors.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Molecular Genetic Diagnostics and Cell Biology, Division of Laboratory Medicine, Institute of Pediatrics, Research Center for Children's Health, 119991, Moscow, Russia
| | - Alexander N Orekhov
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, 125315, Russia; Department of Biophysics, Biological Faculty, Moscow State University, Moscow, 119991, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, 121609, Russia; National Research Center for Preventive Medicine, Moscow, 101000, Russia
| | - Yuri V Bobryshev
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, 125315, Russia; Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia; School of Medicine, University of Western Sydney, Campbelltown, NSW 2560, Australia.
| |
Collapse
|
124
|
Cencioni C, Atlante S, Savoia M, Martelli F, Farsetti A, Capogrossi MC, Zeiher AM, Gaetano C, Spallotta F. The double life of cardiac mesenchymal cells: Epimetabolic sensors and therapeutic assets for heart regeneration. Pharmacol Ther 2016; 171:43-55. [PMID: 27742569 DOI: 10.1016/j.pharmthera.2016.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Organ-specific mesenchymal cells naturally reside in the stroma, where they are exposed to some environmental variables affecting their biology and functions. Risk factors such as diabetes or aging influence their adaptive response. In these cases, permanent epigenetic modifications may be introduced in the cells with important consequences on their local homeostatic activity and therapeutic potential. Numerous results suggest that mesenchymal cells, virtually present in every organ, may contribute to tissue regeneration mostly by paracrine mechanisms. Intriguingly, the heart is emerging as a source of different cells, including pericytes, cardiac progenitors, and cardiac fibroblasts. According to phenotypic, functional, and molecular criteria, these should be classified as mesenchymal cells. Not surprisingly, in recent years, the attention on these cells as therapeutic tools has grown exponentially, although only very preliminary data have been obtained in clinical trials to date. In this review, we summarized the state of the art about the phenotypic features, functions, regenerative properties, and clinical applicability of mesenchymal cells, with a particular focus on those of cardiac origin.
Collapse
Affiliation(s)
- Chiara Cencioni
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Sandra Atlante
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Matteo Savoia
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Universitá Cattolica, Institute of Medical Pathology, 00138 Rome, Italy; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, San Donato Milanese, Milan 20097, Italy.
| | - Antonella Farsetti
- Consiglio Nazionale delle Ricerche, Istituto di Biologia Cellulare e Neurobiologia, Roma, Italy; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Maurizio C Capogrossi
- Laboratorio di Patologia Vascolare, Istituto Dermopatico dell'Immacolata, Roma, Italy.
| | - Andreas M Zeiher
- Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Carlo Gaetano
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Francesco Spallotta
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| |
Collapse
|
125
|
Rivers ZT, Oostra DR, Westholder JS, Vercellotti GM. Romidepsin-associated cardiac toxicity and ECG changes: A case report and review of the literature. J Oncol Pharm Pract 2016; 24:56-62. [DOI: 10.1177/1078155216673229] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Romidepsin is a novel histone deacetylase inhibitor that is approved for the treatment of cutaneous and peripheral T-cell lymphoma in patients who have had at least one prior therapy. Romidepsin is generally well tolerated, though it comes with a risk of cardiac toxicities. Objective We report a case of electrocardiogram changes in a 64-year-old male with enteropathy-associated T-cell lymphoma, type 2, treated with salvage romidepsin therapy who relapsed after non-myeloablative allogeneic sibling peripheral blood stem cell transplant. Discussion Although histone deacetylase inhibitors have been investigated for many years, they have only recently been translated to clinical use as a therapy for malignancies. Furthermore, given their approval for a rare disease, clinicians often have limited experience with the dosing and side effects of histone deacetylase inhibitors. Conclusion This case report and literature review investigates the cardiac side effects of histone deacetylase inhibitors and illustrates the importance of cardiac monitoring prior to and during treatment.
Collapse
Affiliation(s)
- Zachary T Rivers
- Department of Pharmacy, University of Minnesota Medical Center, Minneapolis, MN, USA
- Department of Pharmaceutical Care and Health Systems, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Drew R Oostra
- Division of Hematology, Oncology, and Transplant, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - James S Westholder
- Department of Pharmacy, University of Minnesota Medical Center, Minneapolis, MN, USA
| | - Gregory M Vercellotti
- Division of Hematology, Oncology, and Transplant, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
126
|
Histone Deacetylase Inhibitor Phenylbutyrate Exaggerates Heart Failure in Pressure Overloaded Mice independently of HDAC inhibition. Sci Rep 2016; 6:34036. [PMID: 27667442 PMCID: PMC5036044 DOI: 10.1038/srep34036] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 09/05/2016] [Indexed: 12/18/2022] Open
Abstract
4-Sodium phenylbutyrate (PBA) has been reported to inhibit endoplasmic reticulum stress and histone deacetylation (HDAC), both of which are novel therapeutic targets for cardiac hypertrophy and heart failure. However, it is unclear whether PBA can improve heart function. Here, we tested the effects of PBA and some other HDAC inhibitors on cardiac dysfunction induced by pressure overload. Transverse aortic constriction (TAC) was performed on male C57BL/6 mice. PBA treatment (100 mg/kg, 6 weeks) unexpectedly led to a higher mortality, exacerbated cardiac remodelling and dysfunction. Similar results were noted in TAC mice treated with butyrate sodium (BS), a PBA analogue. In contrast, other HDAC inhibitors, valproic acid (VAL) and trichostatin A (TSA), improved the survival. All four HDAC inhibitors induced histone H3 acetylation and inhibited HDAC total activity. An individual HDAC activity assay showed that rather than class IIa members (HDAC4 and 7), PBA and BS predominantly inhibited class I members (HDAC2 and 8), whereas VAL and TSA inhibited all of them. These findings indicate that PBA and BS accelerate cardiac hypertrophy and dysfunction, whereas VAL and TSA have opposing effects.
Collapse
|
127
|
Fukushima A, Lopaschuk GD. Acetylation control of cardiac fatty acid β-oxidation and energy metabolism in obesity, diabetes, and heart failure. Biochim Biophys Acta Mol Basis Dis 2016; 1862:2211-2220. [PMID: 27479696 DOI: 10.1016/j.bbadis.2016.07.020] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/22/2016] [Accepted: 07/25/2016] [Indexed: 12/31/2022]
Abstract
Alterations in cardiac energy metabolism are an important contributor to the cardiac pathology associated with obesity, diabetes, and heart failure. High rates of fatty acid β-oxidation with cardiac insulin resistance represent a cardiac metabolic hallmark of diabetes and obesity, while a marginal decrease in fatty acid oxidation and a prominent decrease in insulin-stimulated glucose oxidation are commonly seen in the early stages of heart failure. Alterations in post-translational control of energy metabolic processes have recently been identified as an important contributor to these metabolic changes. In particular, lysine acetylation of non-histone proteins, which controls a diverse family of mitochondrial metabolic pathways, contributes to the cardiac energy derangements seen in obesity, diabetes, and heart failure. Lysine acetylation is controlled both via acetyltransferases and deacetylases (sirtuins), as well as by non-enzymatic lysine acetylation due to increased acetyl CoA pool size or dysregulated nicotinamide adenine dinucleotide (NAD+) metabolism (which stimulates sirtuin activity). One of the important mitochondrial acetylation targets are the fatty acid β-oxidation enzymes, which contributes to alterations in cardiac substrate preference during the course of obesity, diabetes, and heart failure, and can ultimately lead to cardiac dysfunction in these disease states. This review will summarize the role of lysine acetylation and its regulatory control in the context of mitochondrial fatty acid β-oxidation. The functional contribution of cardiac protein lysine acetylation to the shift in cardiac energy substrate preference that occurs in obesity, diabetes, and especially in the early stages of heart failure will also be reviewed. This article is part of a Special Issue entitled: The role of post-translational protein modifications on heart and vascular metabolism edited by Jason R.B. Dyck & Jan F.C. Glatz.
Collapse
Affiliation(s)
- Arata Fukushima
- Cardiovascular Translational Science Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Gary D Lopaschuk
- Cardiovascular Translational Science Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
128
|
Subramanian U, Kumar P, Mani I, Chen D, Kessler I, Periyasamy R, Raghavaraju G, Pandey KN. Retinoic acid and sodium butyrate suppress the cardiac expression of hypertrophic markers and proinflammatory mediators in Npr1 gene-disrupted haplotype mice. Physiol Genomics 2016; 48:477-90. [PMID: 27199456 PMCID: PMC4967220 DOI: 10.1152/physiolgenomics.00073.2015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 04/12/2016] [Indexed: 01/15/2023] Open
Abstract
The objective of the present study was to examine the genetically determined differences in the natriuretic peptide receptor-A (NPRA) gene (Npr1) copies affecting the expression of cardiac hypertrophic markers, proinflammatory mediators, and matrix metalloproteinases (MMPs) in a gene-dose-dependent manner. We determined whether stimulation of Npr1 by all-trans retinoic acid (RA) and histone deacetylase (HDAC) inhibitor sodium butyric acid (SB) suppress the expression of cardiac disease markers. In the present study, we utilized Npr1 gene-disrupted heterozygous (Npr1(+/-), 1-copy), wild-type (Npr1(+/+), 2-copy), gene-duplicated (Npr1(++/+), 3-copy) mice, which were treated intraperitoneally with RA, SB, and a combination of RA/SB, a hybrid drug (HB) for 2 wk. Untreated 1-copy mice showed significantly increased heart weight-body weight (HW/BW) ratio, blood pressure, hypertrophic markers, including beta-myosin heavy chain (β-MHC) and proto-oncogenes (c-fos and c-jun), proinflammatory mediator nuclear factor kappa B (NF-κB), and MMPs (MMP-2, MMP-9) compared with 2-copy and 3-copy mice. The heterozygous (haplotype) 1-copy mice treated with RA, SB, or HB, exhibited significant reduction in the expression of β-MHC, c-fos, c-jun, NF-κB, MMP-2, and MMP-9. In drug-treated animals, the activity and expression levels of HDAC were significantly reduced and histone acetyltransferase activity and expression levels were increased. The drug treatments significantly increased the fractional shortening and reduced the systolic and diastolic parameters of the Npr1(+/-) mice hearts. Together, the present results demonstrate that a decreased Npr1 copy number enhanced the expression of hypertrophic markers, proinflammatory mediators, and MMPs, whereas an increased Npr1 repressed the cardiac disease markers in a gene-dose-dependent manner.
Collapse
Affiliation(s)
- Umadevi Subramanian
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine, New Orleans, Louisiana
| | - Prerna Kumar
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine, New Orleans, Louisiana
| | - Indra Mani
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine, New Orleans, Louisiana
| | - David Chen
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine, New Orleans, Louisiana
| | - Isaac Kessler
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine, New Orleans, Louisiana
| | - Ramu Periyasamy
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine, New Orleans, Louisiana
| | - Giri Raghavaraju
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine, New Orleans, Louisiana
| | - Kailash N Pandey
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine, New Orleans, Louisiana
| |
Collapse
|
129
|
Tsigkas G, Katsanos K, Apostolakis E, Papadimitriou E, Koutsioumpa M, Kagadis GC, Koumoundourou D, Hahalis G, Alexopoulos D. A minimally invasive endovascular rabbit model for experimental induction of progressive myocardial hypertrophy. Hypertens Res 2016; 39:840-847. [DOI: 10.1038/hr.2016.66] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 05/10/2016] [Accepted: 05/19/2016] [Indexed: 01/30/2023]
|
130
|
Schiattarella GG, Sannino A, Toscano E, Cattaneo F, Trimarco B, Esposito G, Perrino C. Cardiovascular effects of histone deacetylase inhibitors epigenetic therapies: Systematic review of 62 studies and new hypotheses for future research. Int J Cardiol 2016; 219:396-403. [PMID: 27362830 DOI: 10.1016/j.ijcard.2016.06.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 06/12/2016] [Indexed: 10/21/2022]
Affiliation(s)
- Gabriele Giacomo Schiattarella
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy; Departments of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Anna Sannino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy; Baylor Heart and Vascular Hospital, Baylor Research Institute, Dallas, TX, USA
| | - Evelina Toscano
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Fabio Cattaneo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Giovanni Esposito
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy.
| |
Collapse
|
131
|
Lkhagva B, Kao YH, Chen YC, Chao TF, Chen SA, Chen YJ. Targeting histone deacetylases: A novel therapeutic strategy for atrial fibrillation. Eur J Pharmacol 2016; 781:250-7. [PMID: 27089819 DOI: 10.1016/j.ejphar.2016.04.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/18/2016] [Accepted: 04/15/2016] [Indexed: 12/28/2022]
Abstract
Atrial fibrillation (AF) is a common cardiac arrhythmia associated with high mortality and morbidity. Current treatments of AF have limited efficacy and considerable side effects. Histone deacetylases (HDACs) play critical roles in the pathophysiology of cardiovascular diseases and contribute to the genesis of AF. Therefore, HDAC inhibition may prove a novel therapeutic strategy for AF through upstream therapy and modifications of AF electrical and structural remodeling. In this review, we provide an update of the knowledge of the effects of HDACs and HDAC inhibitors on AF, and dissect potential underlying mechanisms.
Collapse
Affiliation(s)
- Baigalmaa Lkhagva
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Tze-Fan Chao
- Division of Cardiology and Cardiovascular Research Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shih-Ann Chen
- Division of Cardiology and Cardiovascular Research Center, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
132
|
Galán M, Varona S, Orriols M, Rodríguez JA, Aguiló S, Dilmé J, Camacho M, Martínez-González J, Rodriguez C. Induction of histone deacetylases (HDACs) in human abdominal aortic aneurysm: therapeutic potential of HDAC inhibitors. Dis Model Mech 2016; 9:541-52. [PMID: 26989193 PMCID: PMC4892665 DOI: 10.1242/dmm.024513] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/14/2016] [Indexed: 02/01/2023] Open
Abstract
Clinical management of abdominal aortic aneurysm (AAA) is currently limited to elective surgical repair because an effective pharmacotherapy is still awaited. Inhibition of histone deacetylase (HDAC) activity could be a promising therapeutic option in cardiovascular diseases. We aimed to characterise HDAC expression in human AAA and to evaluate the therapeutic potential of class I and IIa HDAC inhibitors in the AAA model of angiotensin II (Ang II)-infused apolipoprotein-E-deficient (ApoE−/−) mice. Real-time PCR, western blot and immunohistochemistry evidenced an increased expression of HDACs 1, 2 (both class I), 4 and 7 (both class IIa) in abdominal aorta samples from patients undergoing AAA open repair (n=22) compared with those from donors (n=14). Aortic aneurysms from Ang-II-infused ApoE−/− mice exhibited a similar HDAC expression profile. In these animals, treatment with a class I HDAC inhibitor (MS-275) or a class IIa inhibitor (MC-1568) improved survival, reduced the incidence and severity of AAA and limited aneurysmal expansion evaluated by Doppler ultrasonography. These beneficial effects were more potent in MC-1568-treated mice. The disorganisation of elastin and collagen fibres and lymphocyte and macrophage infiltration were effectively reduced by both inhibitors. Additionally, HDAC inhibition attenuated the exacerbated expression of pro-inflammatory markers and the increase in metalloproteinase-2 and -9 activity induced by Ang II in this model. Therefore, our data evidence that HDAC expression is deregulated in human AAA and that class-selective HDAC inhibitors limit aneurysm expansion in an AAA mouse model. New-generation HDAC inhibitors represent a promising therapeutic approach to overcome human aneurysm progression. Summary: This study reports the upregulation of HDACs in human AAA, evidences that HDAC inhibitors limit aneurysm progression in a preclinical model and suggests the therapeutic interest of HDAC inhibition in AAA.
Collapse
Affiliation(s)
- María Galán
- Centro de Investigación Cardiovascular (CSIC-ICCC), Instituto de Investigación Biomédica (IIB-Sant Pau), 08025 Barcelona, Spain
| | - Saray Varona
- Centro de Investigación Cardiovascular (CSIC-ICCC), Instituto de Investigación Biomédica (IIB-Sant Pau), 08025 Barcelona, Spain
| | - Mar Orriols
- Centro de Investigación Cardiovascular (CSIC-ICCC), Instituto de Investigación Biomédica (IIB-Sant Pau), 08025 Barcelona, Spain
| | - José Antonio Rodríguez
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Center for Applied Medical Research, University of Navarra, 31008 Pamplona, Spain
| | - Silvia Aguiló
- Centro de Investigación Cardiovascular (CSIC-ICCC), Instituto de Investigación Biomédica (IIB-Sant Pau), 08025 Barcelona, Spain
| | - Jaume Dilmé
- Laboratorio de Angiología, Biología Vascular e Inflamación y Servicio de Cirugía Vascular, Instituto de Investigación Biomédica (IIB-Sant Pau), 08025 Barcelona, Spain
| | - Mercedes Camacho
- Laboratorio de Angiología, Biología Vascular e Inflamación y Servicio de Cirugía Vascular, Instituto de Investigación Biomédica (IIB-Sant Pau), 08025 Barcelona, Spain
| | - José Martínez-González
- Centro de Investigación Cardiovascular (CSIC-ICCC), Instituto de Investigación Biomédica (IIB-Sant Pau), 08025 Barcelona, Spain
| | - Cristina Rodriguez
- Centro de Investigación Cardiovascular (CSIC-ICCC), Instituto de Investigación Biomédica (IIB-Sant Pau), 08025 Barcelona, Spain
| |
Collapse
|
133
|
MDM2 E3 ligase-mediated ubiquitination and degradation of HDAC1 in vascular calcification. Nat Commun 2016; 7:10492. [PMID: 26832969 PMCID: PMC4740400 DOI: 10.1038/ncomms10492] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 12/04/2015] [Indexed: 12/15/2022] Open
Abstract
Vascular calcification (VC) is often associated with cardiovascular and metabolic diseases. However, the molecular mechanisms linking VC to these diseases have yet to be elucidated. Here we report that MDM2-induced ubiquitination of histone deacetylase 1 (HDAC1) mediates VC. Loss of HDAC1 activity via either chemical inhibitor or genetic ablation enhances VC. HDAC1 protein, but not mRNA, is reduced in cell and animal calcification models and in human calcified coronary artery. Under calcification-inducing conditions, proteasomal degradation of HDAC1 precedes VC and it is mediated by MDM2 E3 ubiquitin ligase that initiates HDAC1 K74 ubiquitination. Overexpression of MDM2 enhances VC, whereas loss of MDM2 blunts it. Decoy peptide spanning HDAC1 K74 and RG 7112, an MDM2 inhibitor, prevent VC in vivo and in vitro. These results uncover a previously unappreciated ubiquitination pathway and suggest MDM2-mediated HDAC1 ubiquitination as a new therapeutic target in VC. Vascular calcification (VC) increases morbidity and mortality in cardiovascular and metabolic diseases. Here, Kwon et al. show that calcification stimuli induce MDM2- mediated ubiquitination and proteasomal degradation of HDAC1, suggesting a possible therapeutic strategy for treatment of VC patients.
Collapse
|
134
|
Yoon S, Eom GH. HDAC and HDAC Inhibitor: From Cancer to Cardiovascular Diseases. Chonnam Med J 2016; 52:1-11. [PMID: 26865995 PMCID: PMC4742605 DOI: 10.4068/cmj.2016.52.1.1] [Citation(s) in RCA: 336] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 11/27/2015] [Accepted: 12/01/2015] [Indexed: 01/15/2023] Open
Abstract
Histone deacetylases (HDACs) are epigenetic regulators that regulate the histone tail, chromatin conformation, protein-DNA interaction, and even transcription. HDACs are also post-transcriptional modifiers that regulate the protein acetylation implicated in several pathophysiologic states. HDAC inhibitors have been highlighted as a novel category of anti-cancer drugs. To date, four HDAC inhibitors, Vorinostat, Romidepsin, Panobinostat, and Belinostat, have been approved by the United States Food and Drug Administration. Principally, these HDAC inhibitors are used for hematologic cancers in clinic with less severe side effects. Clinical trials are continuously expanding to address other types of cancer and also nonmalignant diseases. HDAC inhibition also results in beneficial outcomes in various types of neurodegenerative diseases, inflammation disorders, and cardiovascular diseases. In this review, we will briefly discuss 1) the roles of HDACs in the acquisition of a cancer's phenotype and the general outcome of the HDAC inhibitors in cancer, 2) the functional relevance of HDACs in cardiovascular diseases and the possible therapeutic implications of HDAC inhibitors in cardiovascular disease.
Collapse
Affiliation(s)
- Somy Yoon
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, Korea
| | - Gwang Hyeon Eom
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
135
|
Miao R, Lu Y, Xing X, Li Y, Huang Z, Zhong H, Huang Y, Chen AF, Tang X, Li H, Cai J, Yuan H. Regulator of G-Protein Signaling 10 Negatively Regulates Cardiac Remodeling by Blocking Mitogen-Activated Protein Kinase–Extracellular Signal-Regulated Protein Kinase 1/2 Signaling. Hypertension 2016; 67:86-98. [PMID: 26573707 DOI: 10.1161/hypertensionaha.115.05957] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 10/28/2015] [Indexed: 11/16/2022]
Abstract
Regulator of G-protein signaling 10 (RGS10) is an important member of the RGS family and produces biological effects in multiple organs. We used a genetic approach to study the role of RGS10 in the regulation of pathological cardiac hypertrophy and found that RGS10 can negatively influence pressure overload–induced cardiac remodeling. RGS10 expression was markedly decreased in failing human hearts and hypertrophic murine hearts. The extent of aortic banding–induced cardiac hypertrophy, dysfunction, and fibrosis in RGS10-knockout mice was exacerbated, whereas the heart of transgenic mice with cardiac-specific RGS10 overexpression exhibited an alleviated response to pressure overload. Consistently, RGS10 also inhibited an angiotensin II–induced hypertrophic response in isolated cardiomyocytes. Mechanistically, cardiac remodeling improvement elicited by RGS10 was associated with the abrogation of mitogen-activated protein kinase kinase 1/2–extracellular signal-regulated protein kinase 1/2 signaling. Furthermore, the inhibition of mitogen-activated protein kinase kinase–extracellular signal-regulated protein kinase 1/2 transduction abolished RGS10 deletion-induced hypertrophic aggravation. These findings place RGS10 and its downstream signaling mitogen-activated protein kinase kinase–extracellular signal-regulated protein kinase 1/2 as crucial regulators of pathological cardiac hypertrophy after pressure overload and identify this pathway as a potential therapeutic target to attenuate the pressure overload–driven cardiac remodeling.
Collapse
Affiliation(s)
- Rujia Miao
- From the Department of Cardiology (R.M., H.Z., A.F.C., X.T., J.C., H.Y.) and Center of Clinical Pharmacology (Y.L., X.X., Y.L., Z.H., Y.H., J.C., H.Y.), the Third Xiangya Hospital, Central South University, Changsha, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, China (H.L.)
| | - Yao Lu
- From the Department of Cardiology (R.M., H.Z., A.F.C., X.T., J.C., H.Y.) and Center of Clinical Pharmacology (Y.L., X.X., Y.L., Z.H., Y.H., J.C., H.Y.), the Third Xiangya Hospital, Central South University, Changsha, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, China (H.L.)
| | - Xiaowei Xing
- From the Department of Cardiology (R.M., H.Z., A.F.C., X.T., J.C., H.Y.) and Center of Clinical Pharmacology (Y.L., X.X., Y.L., Z.H., Y.H., J.C., H.Y.), the Third Xiangya Hospital, Central South University, Changsha, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, China (H.L.)
| | - Ying Li
- From the Department of Cardiology (R.M., H.Z., A.F.C., X.T., J.C., H.Y.) and Center of Clinical Pharmacology (Y.L., X.X., Y.L., Z.H., Y.H., J.C., H.Y.), the Third Xiangya Hospital, Central South University, Changsha, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, China (H.L.)
| | - Zhijun Huang
- From the Department of Cardiology (R.M., H.Z., A.F.C., X.T., J.C., H.Y.) and Center of Clinical Pharmacology (Y.L., X.X., Y.L., Z.H., Y.H., J.C., H.Y.), the Third Xiangya Hospital, Central South University, Changsha, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, China (H.L.)
| | - Hua Zhong
- From the Department of Cardiology (R.M., H.Z., A.F.C., X.T., J.C., H.Y.) and Center of Clinical Pharmacology (Y.L., X.X., Y.L., Z.H., Y.H., J.C., H.Y.), the Third Xiangya Hospital, Central South University, Changsha, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, China (H.L.)
| | - Yun Huang
- From the Department of Cardiology (R.M., H.Z., A.F.C., X.T., J.C., H.Y.) and Center of Clinical Pharmacology (Y.L., X.X., Y.L., Z.H., Y.H., J.C., H.Y.), the Third Xiangya Hospital, Central South University, Changsha, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, China (H.L.)
| | - Alex F. Chen
- From the Department of Cardiology (R.M., H.Z., A.F.C., X.T., J.C., H.Y.) and Center of Clinical Pharmacology (Y.L., X.X., Y.L., Z.H., Y.H., J.C., H.Y.), the Third Xiangya Hospital, Central South University, Changsha, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, China (H.L.)
| | - Xiaohong Tang
- From the Department of Cardiology (R.M., H.Z., A.F.C., X.T., J.C., H.Y.) and Center of Clinical Pharmacology (Y.L., X.X., Y.L., Z.H., Y.H., J.C., H.Y.), the Third Xiangya Hospital, Central South University, Changsha, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, China (H.L.)
| | - Hongliang Li
- From the Department of Cardiology (R.M., H.Z., A.F.C., X.T., J.C., H.Y.) and Center of Clinical Pharmacology (Y.L., X.X., Y.L., Z.H., Y.H., J.C., H.Y.), the Third Xiangya Hospital, Central South University, Changsha, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, China (H.L.)
| | - Jingjing Cai
- From the Department of Cardiology (R.M., H.Z., A.F.C., X.T., J.C., H.Y.) and Center of Clinical Pharmacology (Y.L., X.X., Y.L., Z.H., Y.H., J.C., H.Y.), the Third Xiangya Hospital, Central South University, Changsha, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, China (H.L.)
| | - Hong Yuan
- From the Department of Cardiology (R.M., H.Z., A.F.C., X.T., J.C., H.Y.) and Center of Clinical Pharmacology (Y.L., X.X., Y.L., Z.H., Y.H., J.C., H.Y.), the Third Xiangya Hospital, Central South University, Changsha, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (H.L.); and Cardiovascular Research Institute of Wuhan University, Wuhan, China (H.L.)
| |
Collapse
|
136
|
Zelko IN, Folz RJ. Regulation of Oxidative Stress in Pulmonary Artery Endothelium. Modulation of Extracellular Superoxide Dismutase and NOX4 Expression Using Histone Deacetylase Class I Inhibitors. Am J Respir Cell Mol Biol 2015; 53:513-24. [PMID: 25749103 DOI: 10.1165/rcmb.2014-0260oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
An imbalance between oxidants and antioxidants is considered a major factor in the development of pulmonary vascular diseases. Oxidative stress seen in pulmonary vascular cells is regulated by increased expression of prooxidant enzymes (e.g., nicotinamide adenine dinucleotide phosphate reduced oxidases) and/or decreased production of antioxidants and antioxidant enzymes (e.g., superoxide dismutases). We and others have shown that expression of antioxidant genes in pulmonary artery cells is regulated by epigenetic mechanisms. In this study, we investigate the regulation of oxidative stress in pulmonary artery cells using inhibitors of histone deacetylases (HDACs). Human pulmonary artery endothelial cells (HPAECs) and human pulmonary artery smooth muscle cells were exposed to an array of HDAC inhibitors followed by analysis of anti- and prooxidant gene expression using quantitative RT-PCR and quantitative RT-PCR array. We found that exposure of HPAECs to scriptaid, N-[4-[(hydroxyamino)carbonyl]phenyl]-α-(1-methylethyl)-benzeneacetamide, and trichostatin A for 24 hours induced expression of extracellular superoxide dismutase (EC-SOD) up to 10-fold, whereas expression of the prooxidant gene NADPH oxidase 4 was decreased by more than 95%. We also found that this differential regulation of anti- and prooxidant gene expression resulted in significant attenuation in the cellular levels of reactive oxygen species. Induction of EC-SOD expression was attenuated by the Janus kinase 2 protein kinase inhibitor AG490 and by silencing Janus kinase 2 expression. Augmentation of EC-SOD expression using scriptaid was associated with increased histone H3 (Lys27) acetylation and H3 (Lys4) trimethylation at the gene promoter. We have determined that oxidative stress in pulmonary endothelial cells is regulated by epigenetic mechanisms and can be modulated using HDAC inhibitors.
Collapse
Affiliation(s)
- Igor N Zelko
- Departments of Medicine and Biochemistry and Molecular Biology, University of Louisville, Louisville, Kentucky
| | - Rodney J Folz
- Departments of Medicine and Biochemistry and Molecular Biology, University of Louisville, Louisville, Kentucky
| |
Collapse
|
137
|
Eom GH, Kook H. Role of histone deacetylase 2 and its posttranslational modifications in cardiac hypertrophy. BMB Rep 2015; 48:131-8. [PMID: 25388210 PMCID: PMC4453031 DOI: 10.5483/bmbrep.2015.48.3.242] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Indexed: 11/20/2022] Open
Abstract
Cardiac hypertrophy is a form of global remodeling, although the initial step seems to be an adaptation to increased hemodynamic demands. The characteristics of cardiac hypertrophy include the functional reactivation of the arrested fetal gene program, where histone deacetylases (HDACs) are closely linked in the development of the process. To date, mammalian HDACs are divided into four classes: I, II, III, and IV. By structural similarities, class II HDACs are then subdivided into IIa and IIb. Among class I and II HDACs, HDAC2, 4, 5, and 9 have been reported to be involved in hypertrophic responses; HDAC4, 5, and 9 are negative regulators, whereas HDAC2 is a pro-hypertrophic mediator. The molecular function and regulation of class IIa HDACs depend largely on the phosphorylation-mediated cytosolic redistribution, whereas those of HDAC2 take place primarily in the nucleus. In response to stresses, posttranslational modification (PTM) processes, dynamic modifications after the translation of proteins, are involved in the regulation of the activities of those hypertrophy-related HDACs. In this article, we briefly review 1) the activation of HDAC2 in the development of cardiac hypertrophy and 2) the PTM of HDAC2 and its implications in the regulation of HDAC2 activity.
Collapse
Affiliation(s)
- Gwang Hyeon Eom
- Department of Pharmacology and Medical Research Center for Gene Regulation, Chonnam National University Medical School, Gwangju 501-746, Korea
| | - Hyun Kook
- Department of Pharmacology and Medical Research Center for Gene Regulation, Chonnam National University Medical School, Gwangju 501-746, Korea
| |
Collapse
|
138
|
Tao H, Shi KH, Yang JJ, Li J. Epigenetic mechanisms in atrial fibrillation: New insights and future directions. Trends Cardiovasc Med 2015; 26:306-18. [PMID: 26475117 DOI: 10.1016/j.tcm.2015.08.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 08/23/2015] [Accepted: 08/28/2015] [Indexed: 11/28/2022]
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia. AF is a complex disease that results from genetic and environmental factors and their interactions. In recent years, numerous studies have shown that epigenetic mechanisms significantly participate in AF pathogenesis. Even though a poor understanding of the molecular and electrophysiologic mechanisms of AF, accumulated evidence has suggested that the relevance of epigenetic changes in the development of AF. The aim of this review is to describe the present knowledge about the epigenetic regulatory features significantly participates in AF, and look ahead on new perspectives of epigenetic mechanisms research. Epigenetic regulatory features such as DNA methylation, histone modification, and microRNA influence gene expression by epigenetic mechanisms and by directly binding to various factor response elements in the target gene promoters. Given the role of epigenetic alterations in regulating genes, there is potential for the integration of factors-induced epigenetic alterations as informative factors in the risk assessment process. In this review, new insight into the epigenetic mechanisms in AF pathogenesis is discussed, with special emphasis on DNA methylation, histone modification, and microRNA. Further studies are needed to reveal the potential targets of epigenetic mechanisms, and it can be developed as a therapeutic target for AF.
Collapse
Affiliation(s)
- Hui Tao
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, China; Cardiovascular Research Center, Anhui Medical University, Hefei, China
| | - Kai-Hu Shi
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, China; Cardiovascular Research Center, Anhui Medical University, Hefei, China.
| | - Jing-Jing Yang
- Department of Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China.
| | - Jun Li
- School of Pharmacy, Anhui Medical University, Hefei, China
| |
Collapse
|
139
|
Fatima N, Cohen DC, Sukumar G, Sissung TM, Schooley JF, Haigney MC, Claycomb WC, Cox RT, Dalgard CL, Bates SE, Flagg TP. Histone deacetylase inhibitors modulate KATP subunit transcription in HL-1 cardiomyocytes through effects on cholesterol homeostasis. Front Pharmacol 2015; 6:168. [PMID: 26321954 PMCID: PMC4534802 DOI: 10.3389/fphar.2015.00168] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/27/2015] [Indexed: 11/29/2022] Open
Abstract
Histone deacetylase inhibitors (HDIs) are under investigation for the treatment of a number of human health problems. HDIs have proven therapeutic value in refractory cases of cutaneous T-cell lymphoma. Electrocardiographic ST segment morphological changes associated with HDIs were observed during development. Because ST segment morphology is typically linked to changes in ATP sensitive potassium (KATP) channel activity, we tested the hypothesis that HDIs affect cardiac KATP channel subunit expression. Two different HDIs, romidepsin and trichostatin A, caused ~20-fold increase in SUR2 (Abcc9) subunit mRNA expression in HL-1 cardiomyocytes. The effect was specific for the SUR2 subunit as neither compound causes a marked change in SUR1 (Abcc8) expression. Moreover, the effect was cell specific as neither HDI markedly altered KATP subunit expression in MIN6 pancreatic β-cells. We observe significant enrichment of the H3K9Ac histone mark specifically at the SUR2 promoter consistent with the conclusion that chromatin remodeling at this locus plays a role in increasing SUR2 gene expression. Unexpectedly, however, we also discovered that HDI-dependent depletion of cellular cholesterol is required for the observed effects on SUR2 expression. Taken together, the data in the present study demonstrate that KATP subunit expression can be epigenetically regulated in cardiomyocytes, defines a role for cholesterol homeostasis in mediating epigenetic regulation and suggests a potential molecular basis for the cardiac effects of the HDIs.
Collapse
Affiliation(s)
- Naheed Fatima
- Department of Anatomy, Physiology and Genetics, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences Bethesda, MD, USA
| | - Devin C Cohen
- Department of Anatomy, Physiology and Genetics, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences Bethesda, MD, USA
| | - Gauthaman Sukumar
- Department of Anatomy, Physiology and Genetics, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences Bethesda, MD, USA
| | - Tristan M Sissung
- Developmental Therapeutic Branch, National Cancer Institute, National Institutes of Health Bethesda, MD, USA
| | - James F Schooley
- Department of Anatomy, Physiology and Genetics, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences Bethesda, MD, USA
| | - Mark C Haigney
- Department of Medicine, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences Bethesda, MD, USA
| | - William C Claycomb
- Department of Biochemistry and Molecular Biology, LSU Health Sciences Center New Orleans, LA, USA
| | - Rachel T Cox
- Department of Biochemistry, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences Bethesda, MD, USA
| | - Clifton L Dalgard
- Department of Anatomy, Physiology and Genetics, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences Bethesda, MD, USA
| | - Susan E Bates
- Developmental Therapeutic Branch, National Cancer Institute, National Institutes of Health Bethesda, MD, USA
| | - Thomas P Flagg
- Department of Anatomy, Physiology and Genetics, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences Bethesda, MD, USA
| |
Collapse
|
140
|
Chen Y, Du J, Zhao YT, Zhang L, Lv G, Zhuang S, Qin G, Zhao TC. Histone deacetylase (HDAC) inhibition improves myocardial function and prevents cardiac remodeling in diabetic mice. Cardiovasc Diabetol 2015; 14:99. [PMID: 26245924 PMCID: PMC4527099 DOI: 10.1186/s12933-015-0262-8] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 07/18/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Recent evidence indicates that inhibition of histone deacetylase (HDAC) protects the heart against myocardial injury and stimulates endogenous angiomyogenesis. However, it remains unknown whether HDAC inhibition produces the protective effect in the diabetic heart. We sought to determine whether HDAC inhibition preserves cardiac performance and suppresses cardiac remodeling in diabetic cardiomyopathy. METHODS Adult ICR mice received an intraperitoneal injection of either streptozotocin (STZ, 200 mg/kg) to establish the diabetic model or vehicle to serve as control. Once hyperglycemia was confirmed, diabetic mice received sodium butyrate (1%), a specific HDAC inhibitor, in drinking water on a daily basis to inhibit HDAC activity. Mice were randomly divided into following groups, which includes Control, Control + Sodium butyrate (NaBu), STZ and STZ + Sodium butyrate (NaBu), respectively. Myocardial function was serially assessed at 7, 14, 21 weeks following treatments. RESULTS Echocardiography demonstrated that cardiac function was depressed in diabetic mice, but HDAC inhibition resulted in a significant functional improvement in STZ-injected mice. Likewise, HDAC inhibition attenuates cardiac hypertrophy, as evidenced by a reduced heart/tibia ratio and areas of cardiomyocytes, which is associated with reduced interstitial fibrosis and decreases in active caspase-3 and apoptotic stainings, but also increased angiogenesis in diabetic myocardium. Notably, glucose transporters (GLUT) 1 and 4 were up-regulated following HDAC inhibition, which was accompanied with increases of GLUT1 acetylation and p38 phosphorylation. Furthermore, myocardial superoxide dismutase, an important antioxidant, was elevated following HDAC inhibition in the diabetic mice. CONCLUSION HDAC inhibition plays a critical role in improving cardiac function and suppressing myocardial remodeling in diabetic heart.
Collapse
Affiliation(s)
- Youfang Chen
- Department of Surgery, Boston University Medical School, Roger Williams Medical Center, Boston University, 50 Maude Street, Providence, RI, 02908, USA.
| | - Jianfeng Du
- Department of Surgery, Boston University Medical School, Roger Williams Medical Center, Boston University, 50 Maude Street, Providence, RI, 02908, USA.
| | - Yu Tina Zhao
- Department of Surgery, Boston University Medical School, Roger Williams Medical Center, Boston University, 50 Maude Street, Providence, RI, 02908, USA.
| | - Ling Zhang
- Department of Medicine, Rhode Island Hospital, Brown University, Providence, RI, USA.
| | - Guorong Lv
- Department of Ultrasound, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China.
| | - Shougang Zhuang
- Department of Medicine, Rhode Island Hospital, Brown University, Providence, RI, USA.
| | - Gangjian Qin
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, USA.
| | - Ting C Zhao
- Department of Surgery, Boston University Medical School, Roger Williams Medical Center, Boston University, 50 Maude Street, Providence, RI, 02908, USA.
| |
Collapse
|
141
|
Ferguson BS, McKinsey TA. Non-sirtuin histone deacetylases in the control of cardiac aging. J Mol Cell Cardiol 2015; 83:14-20. [PMID: 25791169 PMCID: PMC4459895 DOI: 10.1016/j.yjmcc.2015.03.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 02/19/2015] [Accepted: 03/10/2015] [Indexed: 02/08/2023]
Abstract
Histone deacetylases (HDACs) catalyze the removal of acetyl-groups from lysine residues within nucelosomal histone tails and thousands of non-histone proteins. The 18 mammalian HDACs are grouped into four classes. Classes I, II and IV HDACs employ zinc as a co-factor for catalytic activity, while class III HDACs (also known as sirtuins) require NAD+ for enzymatic function. Small molecule inhibitors of zinc-dependent HDACs are efficacious in multiple pre-clinical models of pressure overload and ischemic cardiomyopathy, reducing pathological hypertrophy and fibrosis, and improving contractile function. Emerging data have revealed numerous mechanisms by which HDAC inhibitors benefit the heart, including suppression of oxidative stress and inflammation, inhibition of MAP kinase signaling, and enhancement of cardiac protein aggregate clearance and autophagic flux. Here, we summarize recent findings with zinc-dependent HDACs and HDAC inhibitors in the heart, focusing on newly described functions for distinct HDAC isoforms (e.g. HDAC2, HDAC3 and HDAC6). Potential for pharmacological HDAC inhibition as a means of treating age-related cardiac dysfunction is also discussed. This article is part of a Special Issue entitled: CV Aging.
Collapse
Affiliation(s)
- Bradley S Ferguson
- Department of Medicine, Division of Cardiology, University of Colorado, Denver, 12700 E. 19th Ave Aurora, CO 80045-0508, USA
| | - Timothy A McKinsey
- Department of Medicine, Division of Cardiology, University of Colorado, Denver, 12700 E. 19th Ave Aurora, CO 80045-0508, USA.
| |
Collapse
|
142
|
Cavasin MA, Stenmark KR, McKinsey TA. Emerging roles for histone deacetylases in pulmonary hypertension and right ventricular remodeling (2013 Grover Conference series). Pulm Circ 2015; 5:63-72. [PMID: 25992271 DOI: 10.1086/679700] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 07/30/2014] [Indexed: 01/14/2023] Open
Abstract
Reversible lysine acetylation has emerged as a critical mechanism for controlling the function of nucleosomal histones as well as diverse nonhistone proteins. Acetyl groups are conjugated to lysine residues in proteins by histone acetyltransferases and removed by histone deacetylases (HDACs), which are also commonly referred to as lysine deacetylases. Over the past decade, many studies have shown that HDACs play crucial roles in the control of left ventricular (LV) cardiac remodeling in response to stress. Small molecule HDAC inhibitors block pathological hypertrophy and fibrosis and improve cardiac function in various preclinical models of LV failure. Only recently have HDACs been studied in the context of right ventricular (RV) failure, which commonly occurs in patients who experience pulmonary hypertension (PH). Here, we review recent findings with HDAC inhibitors in models of PH and RV remodeling, propose next steps for this newly uncovered area of research, and highlight potential for isoform-selective HDAC inhibitors for the treatment of PH and RV failure.
Collapse
Affiliation(s)
- Maria A Cavasin
- Department of Medicine, Division of Cardiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Kurt R Stenmark
- Department of Pediatrics, Division of Pulmonary and Critical Care Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Timothy A McKinsey
- Department of Medicine, Division of Cardiology, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
143
|
Nural-Guvener H, Zakharova L, Feehery L, Sljukic S, Gaballa M. Anti-Fibrotic Effects of Class I HDAC Inhibitor, Mocetinostat Is Associated with IL-6/Stat3 Signaling in Ischemic Heart Failure. Int J Mol Sci 2015; 16:11482-99. [PMID: 25997003 PMCID: PMC4463712 DOI: 10.3390/ijms160511482] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 04/26/2015] [Accepted: 05/05/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Recent studies have linked histone deacetylases (HDAC) to remodeling of the heart and cardiac fibrosis in heart failure. However, the molecular mechanisms linking chromatin remodeling events with observed anti-fibrotic effects are unknown. Here, we investigated the molecular players involved in anti-fibrotic effects of HDAC inhibition in congestive heart failure (CHF) myocardium and cardiac fibroblasts in vivo. METHODS AND RESULTS MI was created by coronary artery occlusion. Class I HDACs were inhibited in three-week post MI rats by intraperitoneal injection of Mocetinostat (20 mg/kg/day) for duration of three weeks. Cardiac function and heart tissue were analyzed at six week post-MI. CD90+ cardiac fibroblasts were isolated from ventricles through enzymatic digestion of heart. In vivo treatment of CHF animals with Mocetinostat reduced CHF-dependent up-regulation of HDAC1 and HDAC2 in CHF myocardium, improved cardiac function and decreased scar size and total collagen amount. Moreover, expression of pro-fibrotic markers, collagen-1, fibronectin and Connective Tissue Growth Factor (CTGF) were reduced in the left ventricle (LV) of Mocetinostat-treated CHF hearts. Cardiac fibroblasts isolated from Mocetinostat-treated CHF ventricles showed a decrease in expression of collagen I and III, fibronectin and Timp1. In addition, Mocetinostat attenuated CHF-induced elevation of IL-6 levels in CHF myocardium and cardiac fibroblasts. In parallel, levels of pSTAT3 were reduced via Mocetinostat in CHF myocardium. CONCLUSIONS Anti-fibrotic effects of Mocetinostat in CHF are associated with the IL-6/STAT3 signaling pathway. In addition, our study demonstrates in vivo regulation of cardiac fibroblasts via HDAC inhibition.
Collapse
Affiliation(s)
- Hikmet Nural-Guvener
- Cardiovascular Research Laboratory, Banner Sun Health Research Institute, Sun City, AZ 85351, USA.
| | - Liudmila Zakharova
- Cardiovascular Research Laboratory, Banner Sun Health Research Institute, Sun City, AZ 85351, USA.
| | - Lorraine Feehery
- Cardiovascular Research Laboratory, Banner Sun Health Research Institute, Sun City, AZ 85351, USA.
| | - Snjezana Sljukic
- Cardiovascular Research Laboratory, Banner Sun Health Research Institute, Sun City, AZ 85351, USA.
| | - Mohamed Gaballa
- Cardiovascular Research Laboratory, Banner Sun Health Research Institute, Sun City, AZ 85351, USA.
| |
Collapse
|
144
|
Du J, Zhang L, Zhuang S, Qin GJ, Zhao TC. HDAC4 degradation mediates HDAC inhibition-induced protective effects against hypoxia/reoxygenation injury. J Cell Physiol 2015; 230:1321-31. [PMID: 25475100 DOI: 10.1002/jcp.24871] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 11/12/2014] [Indexed: 12/12/2022]
Abstract
Histone deacetylases (HDACs) play a crucial role in the regulation of gene expression through remodeling of chromatin structures. However, the molecular mechanisms involved in this event remain unknown. In this study, we sought to examine whether HDAC inhibition-mediated protective effects involved HDAC4 sumoylation, degradation, and the proteasome pathway. Isolated neonatal mouse ventricular myocytes (NMVM) and H9c2 cardiomyoblasts were subjected to 48 h of hypoxia (H) (1% O2 ) and 2 h of reoxygenation (R). Treatment of cardiomyocytes with trichostatin A (TSA) attenuated H/R-elicited injury, as indicated by a reduction of lactate dehydrogenase (LDH) leakage, an increase in cell viability, and decrease in apoptotic positive cardiomyocytes. MG132, a potent proteasome pathway inhibitor, abrogated TSA-induced protective effects, which was associated with the accumulation of ubiquitinated HDAC4. NMVM transduced with adenoviral HDAC4 led to an exaggeration of H/R-induced injury. TSA treatment resulted in a decrease in HDAC4 in cardiomyocytes infected with adenoviral HDAC4, and HDAC4-induced injury was attenuated by TSA. HDAC inhibition resulted in a significant reduction in reactive oxygen species (ROS) in cardiomyoblasts exposed to H/R, which was attenuated by blockade of the proteasome pathway. Cardiomyoblasts carrying wild type and sumoylation mutation (K559R) were established to examine effects of HDAC4 sumoylation and ubiquitination on H/R injury. Disruption of HDAC4 sumoylation brought about HDAC4 accumulation and impairment of HDAC4 ubiquitination in association with enhanced susceptibility of cardiomyoblasts to H/R. Taken together, these results demonstrated that HDAC inhibition stimulates proteasome dependent degradation of HDAC4, which is associated with HDAC4 sumoylation to induce these protective effects.
Collapse
Affiliation(s)
- Jianfeng Du
- Department of Surgery, Boston University Medical School, Boston University, Roger Williams Medical Center, Providence, Rhode Island
| | | | | | | | | |
Collapse
|
145
|
Greco CM, Condorelli G. Epigenetic modifications and noncoding RNAs in cardiac hypertrophy and failure. Nat Rev Cardiol 2015; 12:488-97. [DOI: 10.1038/nrcardio.2015.71] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
146
|
Ooi JYY, Tuano NK, Rafehi H, Gao XM, Ziemann M, Du XJ, El-Osta A. HDAC inhibition attenuates cardiac hypertrophy by acetylation and deacetylation of target genes. Epigenetics 2015; 10:418-30. [PMID: 25941940 DOI: 10.1080/15592294.2015.1024406] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Pharmacological histone deacetylase (HDAC) inhibitors attenuate pathological cardiac remodeling and hypertrophic gene expression; yet, the direct histone targets remain poorly characterized. Since the inhibition of HDAC activity is associated with suppressing hypertrophy, we hypothesized histone acetylation would target genes implicated in cardiac remodeling. Trichostatin A (TSA) regulates cardiac gene expression and attenuates transverse aortic constriction (TAC) induced hypertrophy. We used chromatin immunoprecipitation (ChIP) coupled with massive parallel sequencing (ChIP-seq) to map, for the first time, genome-wide histone acetylation changes in a preclinical model of pathological cardiac hypertrophy and attenuation of pathogenesis with TSA. Pressure overload-induced cardiac hypertrophy was associated with histone acetylation of genes implicated in cardiac contraction, collagen deposition, inflammation, and extracellular matrix identified by ChIP-seq. Gene set enrichment analysis identified NF-kappa B (NF-κB) transcription factor activation with load induced hypertrophy. Increased histone acetylation was observed on the promoters of NFκB target genes (Icam1, Vcam1, Il21r, Il6ra, Ticam2, Cxcl10) consistent with gene activation in the hypertrophied heart. Surprisingly, TSA attenuated pressure overload-induced cardiac hypertrophy and the suppression of NFκB target genes by broad histone deacetylation. Our results suggest a mechanism for cardioprotection subject to histone deacetylation as a previously unknown target, implicating the importance of inflammation by pharmacological HDAC inhibition. The results of this study provides a framework for HDAC inhibitor function in the heart and argues the long held views of acetylation is subject to more flexibility than previously thought.
Collapse
Key Words
- ANP, Atrial natriuretic peptide
- BNP, Brain natriuretic peptide
- BW, Body Weight
- ChIP, Chromatin Immunoprecipitation
- Ct, threshold cycle number
- Cxcl10, Chemokine (C-X-C Motif) ligand 10
- ENCODE, Encyclopedia of DNA Elements Consortium
- FDR, False Discovery Rate
- FS, Fractional Shortening
- GAIIx, Genome Analyzer IIx
- HDAC inhibitor
- HDAC, Histone deacetylase
- Icam1, Intercellular adhesion molecule 1
- Il21r, Interleukin-21 receptor
- Il6ra, Interleukin-6 receptor
- LV, Left Ventricle
- LVDd, Left Ventricular Diastolic Dimension
- LVH, Left Ventricle Hypertrophy
- MACs, Model-based Analysis of ChIP-seq
- NES, normalized enrichment score
- NFκB, Nuclear factor of kappa light polypeptide gene enhancer in B-cells
- NGS, Next Generation Sequencing
- SEM, Standard Error of the Mean
- Serca2a, Sarcoplasmic reticulum Ca2+ ATPase
- TAC veh, TAC vehicle
- TAC, Transverse Aortic Constriction
- TF, transcription factor
- TL, Tibia Length
- TSA, Trichostatin A
- TSS, Transcription Start Site
- Ticam2, Toll-like receptor adaptor molecule 2
- Traf3, TNF receptor-associated factor 3
- UTR, Untranslated region
- Vcam1, Vascular cell adhesion molecule 1
- cDNA, complementary DNA
- cardiac hypertrophy
- chromatin
- epigenetics
- histone acetylation
- next generation sequencing
- α/βMHC, Alpha/Beta myosin heavy chain
Collapse
Affiliation(s)
- Jenny Y Y Ooi
- a Epigenetics in Human Health and Disease Laboratory; Baker IDI Heart and Diabetes Institute ; Melbourne , Victoria , Australia
| | | | | | | | | | | | | |
Collapse
|
147
|
Kang SH, Seok YM, Song MJ, Lee HA, Kurz T, Kim I. Histone deacetylase inhibition attenuates cardiac hypertrophy and fibrosis through acetylation of mineralocorticoid receptor in spontaneously hypertensive rats. Mol Pharmacol 2015; 87:782-91. [PMID: 25667225 DOI: 10.1124/mol.114.096974] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Inhibition of histone deacetylases (HDACs) by valproic acid (VPA) attenuates inflammatory, hypertrophic, and fibrotic responses in the hearts of spontaneously hypertensive rats (SHRs); however, the molecular mechanism is still unclear. We hypothesized that HDAC inhibition (HDACi) attenuates cardiac hypertrophy and fibrosis through acetylation of mineralocorticoid receptor (MR) in SHRs. Seven-week-old SHRs and Wistar-Kyoto rats were treated with an HDAC class I inhibitor (0.71% w/v in drinking water; VPA) for 11 weeks. Sections of heart were visualized after trichrome stain as well as H&E stain. Histone modifications, such as acetylation (H3Ac [acetylated histone 3]) and fourth lysine trimethylation (H3K4me3) of histone 3, and recruitment of MR and RNA polymerase II (Pol II) into promoters of target genes were measured by quantitative real-time polymerase chain reaction after chromatin immunoprecipitation assay. MR acetylation was determined by Western blot with anti-acetyl-lysine antibody after immunoprecipitation with anti-MR antibody. Treatment with VPA attenuated cardiac hypertrophy and fibrosis. Although treatment with VPA increased H3Ac and H3K4me3 on promoter regions of MR target genes, expression of MR target genes as well as recruitment of MR and Pol II on promoters of target genes were decreased. Although HDACi did not affect MR expression, it increased MR acetylation. These results indicate that HDACi attenuates cardiac hypertrophy and fibrosis through acetylation of MR in spontaneously hypertensive rats.
Collapse
Affiliation(s)
- Seol-Hee Kang
- Department of Pharmacology (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Cardiovascular Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Cell and Matrix Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Brain Korea 21 Plus Kyungpook National University Biomedical Convergence Program (S.-H.K., I.K.), Department of Biomedical Science, Kyungpook National University School of Medicine (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Daegu, Republic of Korea; and Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany (T.K.)
| | - Young Mi Seok
- Department of Pharmacology (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Cardiovascular Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Cell and Matrix Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Brain Korea 21 Plus Kyungpook National University Biomedical Convergence Program (S.-H.K., I.K.), Department of Biomedical Science, Kyungpook National University School of Medicine (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Daegu, Republic of Korea; and Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany (T.K.)
| | - Min-ji Song
- Department of Pharmacology (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Cardiovascular Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Cell and Matrix Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Brain Korea 21 Plus Kyungpook National University Biomedical Convergence Program (S.-H.K., I.K.), Department of Biomedical Science, Kyungpook National University School of Medicine (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Daegu, Republic of Korea; and Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany (T.K.)
| | - Hae-Ahm Lee
- Department of Pharmacology (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Cardiovascular Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Cell and Matrix Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Brain Korea 21 Plus Kyungpook National University Biomedical Convergence Program (S.-H.K., I.K.), Department of Biomedical Science, Kyungpook National University School of Medicine (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Daegu, Republic of Korea; and Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany (T.K.)
| | - Thomas Kurz
- Department of Pharmacology (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Cardiovascular Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Cell and Matrix Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Brain Korea 21 Plus Kyungpook National University Biomedical Convergence Program (S.-H.K., I.K.), Department of Biomedical Science, Kyungpook National University School of Medicine (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Daegu, Republic of Korea; and Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany (T.K.)
| | - InKyeom Kim
- Department of Pharmacology (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Cardiovascular Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Cell and Matrix Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Brain Korea 21 Plus Kyungpook National University Biomedical Convergence Program (S.-H.K., I.K.), Department of Biomedical Science, Kyungpook National University School of Medicine (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Daegu, Republic of Korea; and Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany (T.K.)
| |
Collapse
|
148
|
Choi SY, Ryu Y, Kee HJ, Cho SN, Kim GR, Cho JY, Kim HS, Kim IK, Jeong MH. Tubastatin A suppresses renal fibrosis via regulation of epigenetic histone modification and Smad3-dependent fibrotic genes. Vascul Pharmacol 2015; 72:130-40. [PMID: 25921924 DOI: 10.1016/j.vph.2015.04.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 03/02/2015] [Accepted: 04/20/2015] [Indexed: 11/29/2022]
Abstract
Inflammation and fibrosis are implicated in the pathogenesis of hypertensive kidney damage. We previously demonstrated that a nonspecific histone deacetylase (HDAC) inhibitor attenuates cardiac fibrosis in deoxycorticosterone acetate-salt hypertensive rats, which induces HDAC6 protein and enzymatic activity. However, the HDAC inhibitor's effect and mechanism have not yet been demonstrated. We sought to determine whether an HDAC6-selective inhibitor could treat hypertension and kidney damage in angiotensin II-infused mice. Hypertension was induced by infusion of ANG in mice. Tubastatin A, an HDAC6 selective inhibitor, did not regulate blood pressure. Hypertensive stimuli enhanced the expression of HDAC6 in vivo and in vitro. We showed that the inhibition of HDAC6 prevents fibrosis and inflammation as determined by quantitative real-time PCR, western blot, and immunohistochemistry. Small interfering RNA (siRNA) against HDAC6 or Smad3 attenuated hypertensive stimuli-induced fibrosis and inflammation, whereas Smad2 siRNA failed to inhibit fibrosis. Interestingly, the combination of the HDAC6 inhibitor and Smad3 knockdown synergistically blocked transforming growth factor β (TGF-β) or ANG-induced fibrosis. We also demonstrated for the first time, to our knowledge, that acetylation of collagen type I can be regulated by HDAC6/p300 acetyltransferase. The chromatin immunoprecipitation assay revealed that the HDAC6 inhibitor suppressed TGF-β-induced acetylated histone H4 or phospho-Smad2/3 to Smad3 binding elements in the fibrosis-associated gene promoters including collagen type I. These results suggest that HDAC6 may be a valuable therapeutic target for the treatment of hypertension-induced kidney fibrosis and inflammation.
Collapse
Affiliation(s)
- Sin Young Choi
- Heart Research Center of Chonnam National University Hospital, Gwangju 501-757, Republic of Korea
| | - Yuhee Ryu
- Heart Research Center of Chonnam National University Hospital, Gwangju 501-757, Republic of Korea
| | - Hae Jin Kee
- Heart Research Center of Chonnam National University Hospital, Gwangju 501-757, Republic of Korea.
| | - Soo-Na Cho
- Heart Research Center of Chonnam National University Hospital, Gwangju 501-757, Republic of Korea
| | - Gwi Ran Kim
- Heart Research Center of Chonnam National University Hospital, Gwangju 501-757, Republic of Korea
| | - Jae Yeong Cho
- Heart Research Center of Chonnam National University Hospital, Gwangju 501-757, Republic of Korea
| | - Hyung-Seok Kim
- Department of Anatomy, Chonnam National University Medical School, Gwangju 501-757, Republic of Korea
| | - In-Kyeom Kim
- Department of Pharmacology, Cardiovascular Research Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Myung Ho Jeong
- Heart Research Center of Chonnam National University Hospital, Gwangju 501-757, Republic of Korea.
| |
Collapse
|
149
|
Targeting histone deacetylases: perspectives for epigenetic-based therapy in cardio-cerebrovascular disease. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2015; 12:153-64. [PMID: 25870619 PMCID: PMC4394331 DOI: 10.11909/j.issn.1671-5411.2015.02.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Revised: 12/29/2014] [Accepted: 12/30/2014] [Indexed: 01/09/2023]
Abstract
Although the pathogenesis of cardio-cerebrovascular disease (CCVD) is multifactorial, an increasing number of experimental and clinical studies have highlighted the importance of histone deacetylase (HDAC)-mediated epigenetic processes in the development of cardio-cerebrovascular injury. HDACs are a family of enzymes to balance the acetylation activities of histone acetyltransferases on chromatin remodeling and play essential roles in regulating gene transcription. To date, 18 mammalian HDACs are identified and grouped into four classes based on similarity to yeast orthologs. The zinc-dependent HDAC family currently consists of 11 members divided into three classes (class I, II, and IV) on the basis of structure, sequence homology, and domain organization. In comparison, class III HDACs (also known as the sirtuins) are composed of a family of NAD+-dependent protein-modifying enzymes related to the Sir2 gene. HDAC inhibitors are a group of compounds that block HDAC activities typically by binding to the zinc-containing catalytic domain of HDACs and have displayed anti-inflammatory and antifibrotic effects in the cardio-cerebrovascular system. In this review, we summarize the current knowledge about classifications, functions of HDACs and their roles and regulatory mechanisms in the cardio-cerebrovascular system. Pharmacological targeting of HDAC-mediated epigenetic processes may open new therapeutic avenues for the treatment of CCVD.
Collapse
|
150
|
Abstract
Dynamic packaging of DNA into strings of nucleosomes is a major mechanism whereby eukaryotic cells regulate gene expression. Intricate control of nucleosomal structure and assembly governs access of RNA polymerase II to DNA and consequent RNA synthesis. As part of this, post-translational modifications of histone proteins are central to the regulation of chromatin structure, playing vital roles in regulating the activation and repression of gene transcription. In the heart, dynamic homeostasis of histone modification-driven by the actions of modifiers and recruitment of downstream effectors-is a fundamental regulator of the transcriptional reprogramming that occurs in the setting of disease-related stress. Here, we examine the growing evidence for histone modification as a key mechanism governing pathological growth and remodeling of the myocardium.
Collapse
Affiliation(s)
- Thomas G Gillette
- From the Departments of Internal Medicine (Cardiology) (T.G.G., J.A.H.) and Molecular Biology (J.A.H.), University of Texas Southwestern Medical Center, Dallas.
| | - Joseph A Hill
- From the Departments of Internal Medicine (Cardiology) (T.G.G., J.A.H.) and Molecular Biology (J.A.H.), University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|