101
|
Blakeslee WW, Lin YH, Stratton MS, Tatman PD, Hu T, Ferguson BS, McKinsey TA. Class I HDACs control a JIP1-dependent pathway for kinesin-microtubule binding in cardiomyocytes. J Mol Cell Cardiol 2017; 112:74-82. [PMID: 28886967 DOI: 10.1016/j.yjmcc.2017.09.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/25/2017] [Accepted: 09/04/2017] [Indexed: 01/01/2023]
Abstract
Class I histone deacetylase (HDAC) inhibitors block hypertrophy and fibrosis of the heart by suppressing pathological signaling and gene expression programs in cardiac myocytes and fibroblasts. The impact of HDAC inhibition in unstressed cardiac cells remains poorly understood. Here, we demonstrate that treatment of cultured cardiomyocytes with small molecule HDAC inhibitors leads to dramatic induction of c-Jun amino-terminal kinase (JNK)-interacting protein-1 (JIP1) mRNA and protein expression. In contrast to prior findings, elevated levels of endogenous JIP1 in cardiomyocytes failed to significantly alter JNK signaling or cardiomyocyte hypertrophy. Instead, HDAC inhibitor-mediated induction of JIP1 was required to stimulate expression of the kinesin heavy chain family member, KIF5A. We provide evidence for an HDAC-dependent regulatory circuit that promotes formation of JIP1:KIF5A:microtubule complexes that regulate intracellular transport of cargo such as autophagosomes. These findings define a novel role for class I HDACs in the control of the JIP1/kinesin axis in cardiomyocytes, and suggest that HDAC inhibitors could be used to alter microtubule transport in the heart.
Collapse
Affiliation(s)
- Weston W Blakeslee
- Department of Medicine, Division of Cardiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Ying-Hsi Lin
- Department of Medicine, Division of Cardiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; Consortium for Fibrosis Research & Translation, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew S Stratton
- Department of Medicine, Division of Cardiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; Consortium for Fibrosis Research & Translation, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Philip D Tatman
- Department of Medicine, Division of Cardiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; Medical Scientist Training Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Tianjing Hu
- Department of Medicine, Division of Cardiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; Consortium for Fibrosis Research & Translation, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Bradley S Ferguson
- Department of Medicine, Division of Cardiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Timothy A McKinsey
- Department of Medicine, Division of Cardiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; Consortium for Fibrosis Research & Translation, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
102
|
Kumar N, Tomar R, Pandey A, Tomar V, Singh VK, Chandra R. Preclinical evaluation and molecular docking of 1,3-benzodioxole propargyl ether derivatives as novel inhibitor for combating the histone deacetylase enzyme in cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:1288-1299. [PMID: 28847179 DOI: 10.1080/21691401.2017.1369423] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Even after huge strides in medicine, cancer continues to be a formidable disease, which is slated to become the leading cause of death worldwide. The present study investigates the 1,3-benzodioxole and its propargyl ether derivatives as a novel histone deacetylase enzyme inhibitor in order to cure cancer, as aberrant expression of histone deacetylases (HDACs) is associated with carcinogenesis. Bioinformatics approaches were employed to carry out preclinical and pharmacological evaluations of designed benzodioxole derivatives. Furthermore, their interaction with HDAC-1 enzyme was studied through computational methods for their specific inhibitory effects and evaluated for their LD50 (oral rat acute toxicity) value. In addition to this work, three-dimensional (3D) structure of HDAC-1 enzyme was extracted and evaluated using various parameters including Ramachandran plot and molecular docking stimulation. In our study, we found that compound 7 and compound 9 have higher binding score than approved drugs (SAHA, TSA and VPA). Importantly, these compounds were found to possess good pharmacological and pharmacokinetic properties and can be considered as potent novel compound to combat the HDAC-1 enzyme to cure cancer. Compounds were also analyzed and validated with parameters like absorption, metabolism, excretion, toxicity and synthetic accessibility during the preclinical evaluation. This study paves way to search for novel and potent small chemical compounds for inhibiting HDAC-1 enzyme and in particular to combat the cancer progression by interrupting the cell cycle.
Collapse
Affiliation(s)
- Neeraj Kumar
- a Department of Chemistry, Drug Discovery and Development Laboratory , University of Delhi , Delhi , India.,b Department of Biotechnology, Stem Cell Research Laboratory , Delhi Technological University , Delhi , India
| | - Ravi Tomar
- a Department of Chemistry, Drug Discovery and Development Laboratory , University of Delhi , Delhi , India
| | - Apurva Pandey
- a Department of Chemistry, Drug Discovery and Development Laboratory , University of Delhi , Delhi , India
| | - Vartika Tomar
- a Department of Chemistry, Drug Discovery and Development Laboratory , University of Delhi , Delhi , India
| | - Vimal Kishor Singh
- c Dr. B. R. Ambedkar Centre for Biomedical Research , University of Delhi , Delhi , India
| | - Ramesh Chandra
- a Department of Chemistry, Drug Discovery and Development Laboratory , University of Delhi , Delhi , India.,c Dr. B. R. Ambedkar Centre for Biomedical Research , University of Delhi , Delhi , India
| |
Collapse
|
103
|
Manivasagam S, Velusamy T, Sowndharajan B, Chandrasekar N, Dhanusu S, Vellaichamy E. Valporic acid enhances the Atrial Natriuretic Peptide (ANP) mediated anti-hypertrophic activity by modulating the Npr1 gene transcription in H9c2 cells in vitro. Eur J Pharmacol 2017; 813:94-104. [PMID: 28743391 DOI: 10.1016/j.ejphar.2017.07.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 07/21/2017] [Accepted: 07/21/2017] [Indexed: 12/21/2022]
Abstract
The present study was aimed to determine whether stimulating Npr1 gene activity using Valporic acid (VA), a small short chain fatty acid molecule can enhance ANP mediated anti-hypertrophic activity in isoproterenol (ISO) - treated H9c2 cells in vitro. H9c2 cells were treated with ISO (10-5 M) and co-treated with VA (10-5 M) in the presence and absence of ANP (10-8M), for 48h. ATRA (10-5 M) was used as a positive inducer of Npr1 gene transcription. The mRNA expression of Npr1 and PKG-I genes, proto-oncogenes (c-fos, c-jun and c-myc) and hypertrophic markers (ANP, BNP, α-sk and β-MyHC), genes were determined by quantitative PCR (qPCR). The protein profiling of NPR-A, PKG-I and cGMP were evaluated by Western blot, immunofluorescence and ELISA respectively. A marked reduction in the level of expression of Npr1 (3- fold) and PKG-I (2.5-fold) genes and increased expression of proto-oncogenes (p< 0.001, respectively) and hypertrophic marker genes (p<0.001, respectively) were noticed in the ISO-treated H9c2 cells as compared with control cells. In contrast, the VA treated cells showed maximal Npr1 gene expression (3.5-fold) as compared with ATRA treated cells (2 fold), which is well correlated with the intracellular cGMP levels (80% vs 60%) and reduced (2.5-fold) HDAC -1&-2 mRNA expression. Furthermore, VA or ATRA treatment effectively reversed the ISO-induced altered expression of Npr1 and PKG-I genes, proto-oncogenes, and hypertrophic markers genes. Interestingly, the results of the present study suggest that ANP mediated anti-hypertrophic activity was enhanced with either VA (p<0.001) or ATRA (p<0.01) co-treatment. Together, we conclude that VA in combination with ANP can be a novel therapeutical approach for the treatment and management of left ventricular cardiac hypertrophy.
Collapse
Affiliation(s)
| | - Tamilselvi Velusamy
- Department of Biochemistry, University of Madras, Guindy Campus,Chennai 600025, India
| | - Boopathi Sowndharajan
- Department of Biochemistry, University of Madras, Guindy Campus,Chennai 600025, India
| | - Navvi Chandrasekar
- Department of Biochemistry, University of Madras, Guindy Campus,Chennai 600025, India
| | - Suresh Dhanusu
- Department of Biochemistry, University of Madras, Guindy Campus,Chennai 600025, India
| | - Elangovan Vellaichamy
- Department of Biochemistry, University of Madras, Guindy Campus,Chennai 600025, India.
| |
Collapse
|
104
|
Samson N, Paulin R. Epigenetics, inflammation and metabolism in right heart failure associated with pulmonary hypertension. Pulm Circ 2017; 7:572-587. [PMID: 28628000 PMCID: PMC5841893 DOI: 10.1177/2045893217714463] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/19/2017] [Indexed: 12/19/2022] Open
Abstract
Right ventricular failure (RVF) is the most important prognostic factor for both morbidity and mortality in pulmonary arterial hypertension (PAH), but also occurs in numerous other common diseases and conditions, including left ventricle dysfunction. RVF remains understudied compared with left ventricular failure (LVF). However, right and left ventricles have many differences at the morphological level or the embryologic origin, and respond differently to pressure overload. Therefore, knowledge from the left ventricle cannot be extrapolated to the right ventricle. Few studies have focused on the right ventricle and have permitted to increase our knowledge on the right ventricular-specific mechanisms driving decompensation. Here we review basic principles such as mechanisms accounting for right ventricle hypertrophy, dysfunction, and transition toward failure, with a focus on epigenetics, inflammatory, and metabolic processes.
Collapse
Affiliation(s)
- Nolwenn Samson
- Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Roxane Paulin
- Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
105
|
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
|
106
|
The Impact of Environmental Factors in Influencing Epigenetics Related to Oxidative States in the Cardiovascular System. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:2712751. [PMID: 28607629 PMCID: PMC5457758 DOI: 10.1155/2017/2712751] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 04/03/2017] [Accepted: 04/12/2017] [Indexed: 12/14/2022]
Abstract
Oxidative states exert a significant influence on a wide range of biological and molecular processes and functions. When their balance is shifted towards enhanced amounts of free radicals, pathological phenomena can occur, as the generation of reactive oxygen species (ROS) in tissue microenvironment or in the systemic circulation can be detrimental. Epidemic chronic diseases of western societies, such as cardiovascular disease, obesity, and diabetes correlate with the imbalance of redox homeostasis. Current advances in our understanding of epigenetics have revealed a parallel scenario showing the influence of oxidative stress as a major regulator of epigenetic gene regulation via modification of DNA methylation, histones, and microRNAs. This has provided both the biological link and a potential molecular explanation between oxidative stress and cardiovascular/metabolic phenomena. Accordingly, in this review, we will provide current insights on the physiological and pathological impact of changes in oxidative states on cardiovascular disorders, by specifically focusing on the influence of epigenetic regulation. A special emphasis will highlight the effect on epigenetic regulation of human's current life habits, external and environmental factors, including food intake, tobacco, air pollution, and antioxidant-based approaches. Additionally, the strategy to quantify oxidative states in humans in order to determine which biological marker could best match a subject's profile will be discussed.
Collapse
|
107
|
Keating ST, Plutzky J, El-Osta A. Epigenetic Changes in Diabetes and Cardiovascular Risk. Circ Res 2017; 118:1706-22. [PMID: 27230637 DOI: 10.1161/circresaha.116.306819] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 04/30/2016] [Indexed: 01/03/2023]
Abstract
Cardiovascular complications remain the leading causes of morbidity and premature mortality in patients with diabetes mellitus. Studies in humans and preclinical models demonstrate lasting gene expression changes in the vasculopathies initiated by previous exposure to high glucose concentrations and the associated overproduction of reactive oxygen species. The molecular signatures of chromatin architectures that sensitize the genome to these and other cardiometabolic risk factors of the diabetic milieu are increasingly implicated in the biological memory underlying cardiovascular complications and now widely considered as promising therapeutic targets. Atherosclerosis is a complex heterocellular disease where the contributing cell types possess distinct epigenomes shaping diverse gene expression. Although the extent that pathological chromatin changes can be manipulated in human cardiovascular disease remains to be established, the clinical applicability of epigenetic interventions will be greatly advanced by a deeper understanding of the cell type-specific roles played by writers, erasers, and readers of chromatin modifications in the diabetic vasculature. This review details a current perspective of epigenetic mechanisms of macrovascular disease in diabetes mellitus and highlights recent key descriptions of chromatinized changes associated with persistent gene expression in endothelial, smooth muscle, and circulating immune cells relevant to atherosclerosis. Furthermore, we discuss the challenges associated with pharmacological targeting of epigenetic networks to correct abnormal or deregulated gene expression as a strategy to alleviate the clinical burden of diabetic cardiovascular disease.
Collapse
Affiliation(s)
- Samuel T Keating
- From the Epigenetics in Human Health and Disease Laboratory (S.T.K., A.E.-O.) and Epigenomics Profiling Facility (A.E.-O.), Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia; Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.P.); Department of Pathology, The University of Melbourne, Victoria, Australia (A.E.-O.); and Central Clinical School, Department of Medicine, Monash University, Victoria, Australia (A.E.-O.)
| | - Jorge Plutzky
- From the Epigenetics in Human Health and Disease Laboratory (S.T.K., A.E.-O.) and Epigenomics Profiling Facility (A.E.-O.), Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia; Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.P.); Department of Pathology, The University of Melbourne, Victoria, Australia (A.E.-O.); and Central Clinical School, Department of Medicine, Monash University, Victoria, Australia (A.E.-O.)
| | - Assam El-Osta
- From the Epigenetics in Human Health and Disease Laboratory (S.T.K., A.E.-O.) and Epigenomics Profiling Facility (A.E.-O.), Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia; Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.P.); Department of Pathology, The University of Melbourne, Victoria, Australia (A.E.-O.); and Central Clinical School, Department of Medicine, Monash University, Victoria, Australia (A.E.-O.).
| |
Collapse
|
108
|
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: 36] [Impact Index Per Article: 4.5] [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
|
109
|
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
|
110
|
The pro-inflammatory signalling regulator Stat4 promotes vasculogenesis of great vessels derived from endothelial precursors. Nat Commun 2017; 8:14640. [PMID: 28256502 PMCID: PMC5338034 DOI: 10.1038/ncomms14640] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 01/17/2017] [Indexed: 01/05/2023] Open
Abstract
Vasculogenic defects of great vessels (GVs) are a major cause of congenital cardiovascular diseases. However, genetic regulators of endothelial precursors in GV vasculogenesis remain largely unknown. Here we show that Stat4, a transcription factor known for its regulatory role of pro-inflammatory signalling, promotes GV vasculogenesis in zebrafish. We find stat4 transcripts highly enriched in nkx2.5+ endothelial precursors in the pharynx and demonstrate that genetic ablation of stat4 causes stenosis of pharyngeal arch arteries (PAAs) by suppressing PAAs 3–6 angioblast development. We further show that stat4 is a downstream target of nkx2.5 and that it autonomously promotes proliferation of endothelial precursors of the mesoderm. Mechanistically, stat4 regulates the emerging PAA angioblasts by inhibiting the expression of hdac3 and counteracting the effect of stat1a. Altogether, our study establishes a role for Stat4 in zebrafish great vessel development, and suggests that Stat4 may serve as a therapeutic target for GV defects. Stat4 is a transcription factor known to regulate pro-inflammatory signalling. Here, Meng et al. show that Stat4 is not only regulating inflammation but it is also crucial for great vessels development and endothelial precursor proliferation in zebrafish, by inhibiting the expression of hdac3 and counteracting the effect of Stat1a.
Collapse
|
111
|
Perspectivas moleculares en cardiopatía hipertrófica: abordaje epigenético desde la modificación de la cromatina. REVISTA COLOMBIANA DE CARDIOLOGÍA 2017. [DOI: 10.1016/j.rccar.2016.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
112
|
Zagni C, Floresta G, Monciino G, Rescifina A. The Search for Potent, Small-Molecule HDACIs in Cancer Treatment: A Decade After Vorinostat. Med Res Rev 2017; 37:1373-1428. [PMID: 28181261 DOI: 10.1002/med.21437] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 12/05/2016] [Accepted: 12/12/2016] [Indexed: 12/12/2022]
Abstract
Histone deacetylases (HDACs) play a crucial role in the remodeling of chromatin, and are involved in the epigenetic regulation of gene expression. In the last decade, inhibition of HDACs came out as a target for specific epigenetic changes associated with cancer and other diseases. Until now, more than 20 HDAC inhibitors (HDACIs) have entered clinical studies, and some of them (e.g., vorinostat, romidepsin) have been approved for the treatment of cutaneous T-cell lymphoma. This review provides an overview of current knowledge, progress, and molecular mechanisms of HDACIs, covering a period from 2011 until 2015.
Collapse
Affiliation(s)
- Chiara Zagni
- Dipartimento di Scienze del Farmaco, Università degli Studi di Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| | - Giuseppe Floresta
- Dipartimento di Scienze del Farmaco, Università degli Studi di Catania, Viale Andrea Doria 6, 95125, Catania, Italy.,Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| | - Giulia Monciino
- Dipartimento di Scienze del Farmaco, Università degli Studi di Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| | - Antonio Rescifina
- Dipartimento di Scienze del Farmaco, Università degli Studi di Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| |
Collapse
|
113
|
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
|
114
|
Ono T, Kamimura N, Matsuhashi T, Nagai T, Nishiyama T, Endo J, Hishiki T, Nakanishi T, Shimizu N, Tanaka H, Ohta S, Suematsu M, Ieda M, Sano M, Fukuda K, Kaneda R. The histone 3 lysine 9 methyltransferase inhibitor chaetocin improves prognosis in a rat model of high salt diet-induced heart failure. Sci Rep 2017; 7:39752. [PMID: 28051130 PMCID: PMC5209701 DOI: 10.1038/srep39752] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 11/18/2016] [Indexed: 12/20/2022] Open
Abstract
Histone acetylation has been linked to cardiac hypertrophy and heart failure. However, the pathological implications of changes in histone methylation and the effects of interventions with histone methyltransferase inhibitors for heart failure have not been fully clarified. Here, we focused on H3K9me3 status in the heart and investigated the effects of the histone H3K9 methyltransferase inhibitor chaetocin on prognoses in Dahl salt-sensitive rats, an animal model of chronic heart failure. Chaetocin prolonged survival and restored mitochondrial dysfunction. ChIP-seq analysis demonstrated that chronic stress to the heart induced H3K9me3 elevation in thousands of repetitive elements, including intronic regions of mitochondria-related genes, such as the gene encoding peroxisome proliferator-activated receptor-gamma coactivator 1 alpha. Furthermore, chaetocin reversed this effect on these repetitive loci. These data suggested that excessive heterochromatinization of repetitive elements of mitochondrial genes in the failing heart may lead to the silencing of genes and impair heart function. Thus, chaetocin may be a potential therapeutic agent for chronic heart failure.
Collapse
Affiliation(s)
- Tomohiko Ono
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku Tokyo, Japan
| | - Naomi Kamimura
- Department of Biochemistry and Cell Biology, Institute of Development and Aging Sciences, Graduate School of Medicine, Nippon Medical School, Kawasaki, Kanagawa, Japan
| | - Tomohiro Matsuhashi
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku Tokyo, Japan
| | - Toshihiro Nagai
- Electron Microscope Laboratory, Keio University Hospital, Shinjuku-ku, Tokyo, Japan
| | - Takahiko Nishiyama
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku Tokyo, Japan
| | - Jin Endo
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku Tokyo, Japan
| | - Takako Hishiki
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Clinical and Translational Research Center, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Tsuyoshi Nakanishi
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- MS Business Unit, Shimadzu Corporation, Kyoto, Japan
| | - Noriaki Shimizu
- Division of Rheumatology, Center for Antibody and Vaccine Therapy, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Hirotoshi Tanaka
- Division of Rheumatology, Center for Antibody and Vaccine Therapy, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Shigeo Ohta
- Department of Biochemistry and Cell Biology, Institute of Development and Aging Sciences, Graduate School of Medicine, Nippon Medical School, Kawasaki, Kanagawa, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Masaki Ieda
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku Tokyo, Japan
| | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku Tokyo, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku Tokyo, Japan
| | - Ruri Kaneda
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku Tokyo, Japan
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsukeshi, Tochigi, Japan
- JST, PRESTO, Kawaguchi, Saitama, Japan
| |
Collapse
|
115
|
Wu Y, Leng Y, Meng Q, Xue R, Zhao B, Zhan L, Xia Z. Suppression of Excessive Histone Deacetylases Activity in Diabetic Hearts Attenuates Myocardial Ischemia/Reperfusion Injury via Mitochondria Apoptosis Pathway. J Diabetes Res 2017; 2017:8208065. [PMID: 28191472 PMCID: PMC5278197 DOI: 10.1155/2017/8208065] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 11/11/2016] [Accepted: 11/22/2016] [Indexed: 02/06/2023] Open
Abstract
Background. Histone deacetylases (HDACs) play a pivotal role in signaling modification and gene transcriptional regulation that are essential for cardiovascular pathophysiology. Diabetic hearts with higher HDACs activity were more vulnerable to myocardial ischemia/reperfusion (MI/R) injury compared with nondiabetic hearts. We are curious about whether suppression of excessive HDACs activity in diabetic heart protects against MI/R injury. Methods. Diabetic rats were subjected to 45 min of ischemia, followed by 3 h of reperfusion. H9C2 cardiomyocytes were exposed to high glucose for 24 h, followed by 4 h of hypoxia and 2 h of reoxygenation (H/R). Results. Both MI/R injury and diabetes mellitus elevated myocardium HDACs activity. MI/R induced apoptotic cell death was significantly decreased in diabetic rats treated with HDACs inhibitor trichostatin A (TSA). TSA administration markedly moderated dissipation of mitochondrial membrane potential, protected the integrity of mitochondrial permeability transition pore (mPTP), and decreased cell apoptosis. Notably, cotreatment with Akt inhibitor partly or absolutely inhibited the protective effect of TSA in vivo and in vitro. Furthermore, TSA administration activated Akt/Foxo3a pathway, leading to Foxo3a cytoplasm translocation and attenuation proapoptosis protein Bim expression. Conclusions. Both diabetes mellitus and MI/R injury increased cardiac HDACs activity. Suppression of HDACs activity triggered protective effects against MI/R and H/R injury under hyperglycemia conditions through Akt-modulated mitochondrial apoptotic pathways via Foxo3a/Bim.
Collapse
Affiliation(s)
- Yang Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Yan Leng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Qingtao Meng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Rui Xue
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Bo Zhao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Liying Zhan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
- *Zhongyuan Xia:
| |
Collapse
|
116
|
Ding YY, Li JM, Guo FJ, Liu Y, Tong YF, Pan XC, Lu XL, Ye W, Chen XH, Zhang HG. Triptolide Upregulates Myocardial Forkhead Helix Transcription Factor p3 Expression and Attenuates Cardiac Hypertrophy. Front Pharmacol 2016; 7:471. [PMID: 27965581 PMCID: PMC5127789 DOI: 10.3389/fphar.2016.00471] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 11/18/2016] [Indexed: 12/21/2022] Open
Abstract
The forkhead/winged helix transcription factor (Fox) p3 can regulate the expression of various genes, and it has been reported that the transfer of Foxp3-positive T cells could ameliorate cardiac hypertrophy and fibrosis. Triptolide (TP) can elevate the expression of Foxp3, but its effects on cardiac hypertrophy remain unclear. In the present study, neonatal rat ventricular myocytes (NRVM) were isolated and stimulated with angiotensin II (1 μmol/L) to induce hypertrophic response. The expression of Foxp3 in NRVM was observed by using immunofluorescence assay. Fifty mice were randomly divided into five groups and received vehicle (control), isoproterenol (Iso, 5 mg/kg, s.c.), one of three doses of TP (10, 30, or 90 μg/kg, i.p.) for 14 days, respectively. The pathological morphology changes were observed after Hematoxylin and eosin, lectin and Masson's trichrome staining. The levels of serum brain natriuretic peptide (BNP) and troponin I were determined by enzyme-linked immunosorbent assay and chemiluminescence, respectively. The mRNA and protein expressions of α- myosin heavy chain (MHC), β-MHC and Foxp3 were determined using real-time PCR and immunohistochemistry, respectively. It was shown that TP (1, 3, 10 μg/L) treatment significantly decreased cell size, mRNA and protein expression of β-MHC, and upregulated Foxp3 expression in NRVM. TP also decreased heart weight index, left ventricular weight index and, improved myocardial injury and fibrosis; and decreased the cross-scetional area of the myocardium, serum cardiac troponin and BNP. Additionally, TP markedly reduced the mRNA and protein expression of myocardial β-MHC and elevated the mRNA and protein expression of α-MHC and Foxp3 in a dose-dependent manner. In conclusion, TP can effectively ameliorate myocardial damage and inhibit cardiac hypertrophy, which is at least partly related to the elevation of Foxp3 expression in cardiomyocytes.
Collapse
Affiliation(s)
- Yuan-Yuan Ding
- Department of Pharmacology, College of Pharmacy, Third Military Medical University Chongqing, China
| | - Jing-Mei Li
- Department of Pharmacology, College of Pharmacy, Third Military Medical University Chongqing, China
| | - Feng-Jie Guo
- The People's Liberation Army No. 309 Hospital Beijing, China
| | - Ya Liu
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Third Military Medical University Chongqing, China
| | - Yang-Fei Tong
- Department of Pharmacology, College of Pharmacy, Third Military Medical UniversityChongqing, China; Department of Pharmacy, Chongqing Traditional Medicine HospitalChongqing, China
| | - Xi-Chun Pan
- Department of Pharmacology, College of Pharmacy, Third Military Medical University Chongqing, China
| | - Xiao-Lan Lu
- Department of Pharmacology, College of Pharmacy, Third Military Medical UniversityChongqing, China; Department of Clinical Laboratory, First Affiliated Hospital of North Sichuan Medical CollegeNanchong, China
| | - Wen Ye
- Department of Pharmacology, College of Pharmacy, Third Military Medical University Chongqing, China
| | - Xiao-Hong Chen
- Department of Pharmacology, College of Pharmacy, Third Military Medical University Chongqing, China
| | - Hai-Gang Zhang
- Department of Pharmacology, College of Pharmacy, Third Military Medical University Chongqing, China
| |
Collapse
|
117
|
Abstract
PURPOSE OF REVIEW Here, we provide a summary of the current knowledge on the impact of early life nutrition on cardiovascular diseases that have emerged from studies in humans and experimental animal models. The involvement of epigenetic mechanisms in the Developmental Origins of Health and Disease will be discussed in relation to the implications for the heart and the cardiovascular system. RECENT FINDINGS Environmental cues, such as parental diet and a suboptimal in utero environment can shape growth and development, causing long-lasting cardiometabolic perturbations. Increasing evidence suggest that these effects are mediated at the epigenomic level, and can be passed onto future generations. In the last decade, epigenetic mechanisms (DNA methylation, histone modifications) and RNA-based mechanisms (microRNAs, piRNAs, and tRNAs) have therefore emerged as potential candidates for mediating inheritance of cardiometabolic diseases. SUMMARY The burden of obesity and associated cardiometabolic diseases is believed to arise through interaction between an individual's genetics and the environment. Moreover, the risk of developing poor cardiometabolic health in adulthood is defined by early life exposure to pathological cues and can be inherited by future generations, initiating a vicious cycle of transmission of disease. Elucidating the molecular triggers of such a process will help tackle and prevent the uncontrolled rise in obesity and cardiometabolic disease.
Collapse
Affiliation(s)
- Elena Loche
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
118
|
Oncometabolite d-2-hydroxyglutarate impairs α-ketoglutarate dehydrogenase and contractile function in rodent heart. Proc Natl Acad Sci U S A 2016; 113:10436-41. [PMID: 27582470 DOI: 10.1073/pnas.1601650113] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hematologic malignancies are frequently associated with cardiac pathologies. Mutations of isocitrate dehydrogenase 1 and 2 (IDH1/2) occur in a subset of acute myeloid leukemia patients, causing metabolic and epigenetic derangements. We have now discovered that altered metabolism in leukemic cells has a profound effect on cardiac metabolism. Combining mathematical modeling and in vivo as well as ex vivo studies, we found that increased amounts of the oncometabolite d-2-hydroxyglutarate (D2-HG), produced by IDH2 mutant leukemic cells, cause contractile dysfunction in the heart. This contractile dysfunction is associated with impaired oxidative decarboxylation of α-ketoglutarate, a redirection of Krebs cycle intermediates, and increased ATP citrate lyase (ACL) activity. Increased availability of D2-HG also leads to altered histone methylation and acetylation in the heart. We propose that D2-HG promotes cardiac dysfunction by impairing α-ketoglutarate dehydrogenase and induces histone modifications in an ACL-dependent manner. Collectively, our results highlight the impact of cancer cell metabolism on function and metabolism of the heart.
Collapse
|
119
|
Stratton MS, Lin CY, Anand P, Tatman PD, Ferguson BS, Wickers ST, Ambardekar AV, Sucharov CC, Bradner JE, Haldar SM, McKinsey TA. Signal-Dependent Recruitment of BRD4 to Cardiomyocyte Super-Enhancers Is Suppressed by a MicroRNA. Cell Rep 2016; 16:1366-1378. [PMID: 27425608 PMCID: PMC4972677 DOI: 10.1016/j.celrep.2016.06.074] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 05/04/2016] [Accepted: 06/16/2016] [Indexed: 12/20/2022] Open
Abstract
BRD4 governs pathological cardiac gene expression by binding acetylated chromatin, resulting in enhanced RNA polymerase II (Pol II) phosphorylation and transcription elongation. Here, we describe a signal-dependent mechanism for the regulation of BRD4 in cardiomyocytes. BRD4 expression is suppressed by microRNA-9 (miR-9), which targets the 3' UTR of the Brd4 transcript. In response to stress stimuli, miR-9 is downregulated, leading to derepression of BRD4 and enrichment of BRD4 at long-range super-enhancers (SEs) associated with pathological cardiac genes. A miR-9 mimic represses stimulus-dependent targeting of BRD4 to SEs and blunts Pol II phosphorylation at proximal transcription start sites, without affecting BRD4 binding to SEs that control constitutively expressed cardiac genes. These findings suggest that dynamic enrichment of BRD4 at SEs genome-wide serves a crucial role in the control of stress-induced cardiac gene expression and define a miR-dependent signaling mechanism for the regulation of chromatin state and Pol II phosphorylation.
Collapse
Affiliation(s)
- Matthew S Stratton
- Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - Charles Y Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Priti Anand
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Philip D Tatman
- Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA; Medical Scientist Training Program, University of Colorado Denver, Aurora, CO 80045, USA
| | - Bradley S Ferguson
- Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - Sean T Wickers
- Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - Amrut V Ambardekar
- Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - Carmen C Sucharov
- Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - James E Bradner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Saptarsi M Haldar
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA; Division of Cardiology, Department of Medicine and Cardiovascular Research Institute, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Timothy A McKinsey
- Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA; Medical Scientist Training Program, University of Colorado Denver, Aurora, CO 80045, USA.
| |
Collapse
|
120
|
Lother A, Hein L. Pharmacology of heart failure: From basic science to novel therapies. Pharmacol Ther 2016; 166:136-49. [PMID: 27456554 DOI: 10.1016/j.pharmthera.2016.07.004] [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: 03/18/2016] [Accepted: 07/08/2016] [Indexed: 01/10/2023]
Abstract
Chronic heart failure is one of the leading causes for hospitalization in the United States and Europe, and is accompanied by high mortality. Current pharmacological therapy of chronic heart failure with reduced ejection fraction is largely based on compounds that inhibit the detrimental action of the adrenergic and the renin-angiotensin-aldosterone systems on the heart. More than one decade after spironolactone, two novel therapeutic principles have been added to the very recently released guidelines on heart failure therapy: the HCN-channel inhibitor ivabradine and the combined angiotensin and neprilysin inhibitor valsartan/sacubitril. New compounds that are in phase II or III clinical evaluation include novel non-steroidal mineralocorticoid receptor antagonists, guanylate cyclase activators or myosine activators. A variety of novel candidate targets have been identified and the availability of gene transfer has just begun to accelerate translation from basic science to clinical application. This review provides an overview of current pharmacology and pharmacotherapy in chronic heart failure at three stages: the updated clinical guidelines of the American Heart Association and the European Society of Cardiology, new drugs which are in clinical development, and finally innovative drug targets and their mechanisms in heart failure which are emerging from preclinical studies will be discussed.
Collapse
Affiliation(s)
- Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Heart Center, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
121
|
Abstract
Epigenetic regulatory mechanisms play key roles in cardiac development, differentiation, homeostasis, response to stress and injury, and disease. Human heart failure (HF) epigenetic regulatory mechanisms have not been deciphered to date. This 2-part review distills the rapidly evolving research focused on human HF epigenetic regulatory mechanisms. Part I, which was published in the September/October issue, focused on epigenetic regulatory mechanisms involving RNA, specifically the role of short, intermediate, and long noncoding RNAs (lncRNAs) and endogenous competing RNA regulatory networks. Part II, now in the November/December issue, focuses on the epigenetic regulatory mechanisms involving DNA, including DNA methylation, histone modifications, and chromatin conformational changes. Part II concludes with 2 examples of well-studied integrated epigenetic regulatory mechanisms: the structural and functional roles of the Mediator complex in regulating transcription and the epigenetic networked "cross-talk" regulating atrial natriuretic peptide and brain natriuretic peptide promoter activation.
Collapse
|
122
|
Wright LH, Menick DR. A class of their own: exploring the nondeacetylase roles of class IIa HDACs in cardiovascular disease. Am J Physiol Heart Circ Physiol 2016; 311:H199-206. [PMID: 27208161 PMCID: PMC5005290 DOI: 10.1152/ajpheart.00271.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/13/2016] [Indexed: 11/22/2022]
Abstract
Histone deacetylases (HDACs) play integral roles in many cardiovascular biological processes ranging from transcriptional and translational regulation to protein stabilization and localization. There are 18 known HDACs categorized into 4 classes that can differ on the basis of substrate targets, subcellular localization, and regulatory binding partners. HDACs are classically known for their ability to remove acetyl groups from histone and nonhistone proteins that have lysine residues. However, despite their nomenclature and classical functions, discoveries from many research groups over the past decade have suggested that nondeacetylase roles exist for class IIa HDACs. This is not surprising given that class IIa HDACs have, for example, relatively poor deacetylase capabilities and are often shuttled in and out of nuclei upon specific pathological and nonpathological cardiac events. This review aims to consolidate and elucidate putative nondeacetylase roles for class IIa HDACs and, where possible, highlight studies that provide evidence for their noncanonical roles, especially in the context of cardiovascular maladies. There has been great interest recently in exploring the pharmacological regulators of HDACs for use in therapeutic interventions for treating cardiovascular diseases and inflammation. Thus it is of interest to earnestly consider nonenzymatic and or nondeacetylase roles of HDACs that might be key in potentiating or abrogating pathologies. These noncanonical HDAC functions may possibly yield new mechanisms and targets for drug discovery.
Collapse
Affiliation(s)
- Lillianne H Wright
- Department of Medicine, Division of Cardiology, Medical University of South Carolina; and
| | - Donald R Menick
- Department of Medicine, Division of Cardiology, Medical University of South Carolina; and Ralph Johnson Veteran's Hospital, Charleston, South Carolina
| |
Collapse
|
123
|
Abstract
SUMOylation is a ubiquitin-related transient posttranslational modification pathway catalyzing the conjugation of small ubiquitin-like modifier (SUMO) proteins (SUMO1, SUMO2, and SUMO3) to lysine residues of proteins. SUMOylation targets a wide variety of cellular regulators and thereby affects a multitude of different cellular processes. SUMO/sentrin-specific proteases are able to remove SUMOs from targets, contributing to a tight control of SUMOylated proteins. Genetic and cell biological experiments indicate a critical role of balanced SUMOylation/deSUMOylation for proper cardiac development, metabolism, and stress adaptation. Here, we review the current knowledge about SUMOylation/deSUMOylation in the heart and provide an integrated picture of cardiac functions of the SUMO system under physiologic or pathologic conditions. We also describe potential therapeutic approaches targeting the SUMO machinery to combat heart disease.
Collapse
Affiliation(s)
- Luca Mendler
- From the Institute of Biochemistry II, Goethe University, Medical School, Frankfurt, Germany (L.M., S.M.); Institute of Biochemistry, Faculty of General Medicine, University of Szeged, Szeged, Hungary (L.M.); and Department I - Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.)
| | - Thomas Braun
- From the Institute of Biochemistry II, Goethe University, Medical School, Frankfurt, Germany (L.M., S.M.); Institute of Biochemistry, Faculty of General Medicine, University of Szeged, Szeged, Hungary (L.M.); and Department I - Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.).
| | - Stefan Müller
- From the Institute of Biochemistry II, Goethe University, Medical School, Frankfurt, Germany (L.M., S.M.); Institute of Biochemistry, Faculty of General Medicine, University of Szeged, Szeged, Hungary (L.M.); and Department I - Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.).
| |
Collapse
|
124
|
Abstract
PURPOSE OF REVIEW This article provides an overview, highlighting recent findings, of a major mechanism of gene regulation and its relevance to the pathophysiology of heart failure. RECENT FINDINGS The syndrome of heart failure is a complex and highly prevalent condition, one in which the heart undergoes substantial structural remodeling. Triggered by a wide range of disease-related cues, heart failure pathophysiology is governed by both genetic and epigenetic events. Epigenetic mechanisms, such as chromatin/DNA modifications and noncoding RNAs, have emerged as molecular transducers of environmental stimuli to control gene expression. Here, we emphasize metabolic milieu, aging, and hemodynamic stress as they impact the epigenetic landscape of the myocardium. SUMMARY Recent studies in multiple fields, including cancer, stem cells, development, and cardiovascular biology, have uncovered biochemical ties linking epigenetic machinery and cellular energetics and mitochondrial function. Elucidation of these connections will afford molecular insights into long-established epidemiological observations. With time, exploitation of the epigenetic machinery therapeutically may emerge with clinical relevance.
Collapse
Affiliation(s)
- Soo Young Kim
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Cyndi Morales
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Thomas G. Gillette
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph A. Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
125
|
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
|
126
|
Morales CR, Li DL, Pedrozo Z, May HI, Jiang N, Kyrychenko V, Cho GW, Kim SY, Wang ZV, Rotter D, Rothermel BA, Schneider JW, Lavandero S, Gillette TG, Hill JA. Inhibition of class I histone deacetylases blunts cardiac hypertrophy through TSC2-dependent mTOR repression. Sci Signal 2016; 9:ra34. [PMID: 27048565 DOI: 10.1126/scisignal.aad5736] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Altering chromatin structure through histone posttranslational modifications has emerged as a key driver of transcriptional responses in cells. Modulation of these transcriptional responses by pharmacological inhibition of class I histone deacetylases (HDACs), a group of chromatin remodeling enzymes, has been successful in blocking the growth of some cancer cell types. These inhibitors also attenuate the pathogenesis of pathological cardiac remodeling by blunting and even reversing pathological hypertrophy. The mechanistic target of rapamycin (mTOR) is a critical sensor and regulator of cell growth that, as part of mTOR complex 1 (mTORC1), drives changes in protein synthesis and metabolism in both pathological and physiological hypertrophy. We demonstrated through pharmacological and genetic methods that inhibition of class I HDACs suppressed pathological cardiac hypertrophy through inhibition of mTOR activity. Mice genetically silenced for HDAC1 and HDAC2 had a reduced hypertrophic response to thoracic aortic constriction (TAC) and showed reduced mTOR activity. We determined that the abundance of tuberous sclerosis complex 2 (TSC2), an mTOR inhibitor, was increased through a transcriptional mechanism in cardiomyocytes when class I HDACs were inhibited. In neonatal rat cardiomyocytes, loss of TSC2 abolished HDAC-dependent inhibition of mTOR activity, and increased expression of TSC2 was sufficient to reduce hypertrophy in response to phenylephrine. These findings point to mTOR and TSC2-dependent control of mTOR as critical components of the mechanism by which HDAC inhibitors blunt pathological cardiac growth. These results also suggest a strategy to modulate mTOR activity and facilitate the translational exploitation of HDAC inhibitors in heart disease.
Collapse
Affiliation(s)
- Cyndi R Morales
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Dan L Li
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Zully Pedrozo
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA. Advanced Center for Chronic Diseases, Facultad Ciencias Químicas y Farmacéuticas & Facultad Medicina, Universidad de Chile, Santiago 8380492, Chile
| | - Herman I May
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Nan Jiang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Viktoriia Kyrychenko
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Geoffrey W Cho
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Soo Young Kim
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Zhao V Wang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - David Rotter
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Beverly A Rothermel
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA. Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Jay W Schneider
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Sergio Lavandero
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA. Advanced Center for Chronic Diseases, Facultad Ciencias Químicas y Farmacéuticas & Facultad Medicina, Universidad de Chile, Santiago 8380492, Chile
| | - Thomas G Gillette
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | - Joseph A Hill
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA. Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA.
| |
Collapse
|
127
|
Kopljar I, Gallacher DJ, De Bondt A, Cougnaud L, Vlaminckx E, Van den Wyngaert I, Lu HR. Functional and Transcriptional Characterization of Histone Deacetylase Inhibitor-Mediated Cardiac Adverse Effects in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Stem Cells Transl Med 2016; 5:602-12. [PMID: 27034410 DOI: 10.5966/sctm.2015-0279] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/14/2016] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED Histone deacetylase (HDAC) inhibitors possess therapeutic potential to reverse aberrant epigenetic changes associated with cancers, neurological diseases, and immune disorders. Unfortunately, clinical studies with some HDAC inhibitors displayed delayed cardiac adverse effects, such as atrial fibrillation and ventricular tachycardia. However, the underlying molecular mechanism(s) of HDAC inhibitor-mediated cardiotoxicity remains poorly understood and is difficult to detect in the early stages of preclinical drug development because of a delayed onset of effects. In the present study, we show for the first time in human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) that HDAC inhibitors (dacinostat, panobinostat, vorinostat, entinostat, and tubastatin-a) induce delayed dose-related cardiac dysfunction at therapeutic concentrations associated with cardiac adverse effects in humans. HDAC inhibitor-mediated delayed effects on the beating properties of hiPS-CMs developed after 12 hours by decreasing the beat rate, shortening the field potential duration, and inducing arrhythmic behavior under form of sustained contractions and fibrillation-like patterns. Transcriptional changes that are common between the cardiotoxic HDAC inhibitors but different from noncardiotoxic treatments identified cardiac-specific genes and pathways related to structural and functional changes in cardiomyocytes. Combining the functional data with epigenetic changes in hiPS-CMs allowed us to identify molecular targets that might explain HDAC inhibitor-mediated cardiac adverse effects in humans. Therefore, hiPS-CMs represent a valuable translational model to assess HDAC inhibitor-mediated cardiotoxicity and support identification of better HDAC inhibitors with an improved benefit-risk profile. SIGNIFICANCE Histone deacetylase (HDAC) inhibitors are a promising class of drugs to treat certain cancers, autoimmune, and neurodegenerative diseases. However, treated patients can experience various cardiac adverse events such as hearth rhythm disorders. This study found that human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) can predict cardiac adverse events in patients caused by HDAC inhibitors. Furthermore, transcriptional changes at the level of gene expression supported the effects on the beating properties of hiPS-CMs and highlight targets that might cause these cardiac adverse effects. hiPS-CMs represent a valuable translational model to assess HDAC inhibitor-mediated cardiotoxicity and to support development of safer HDAC inhibitors.
Collapse
MESH Headings
- Action Potentials
- Arrhythmias, Cardiac/chemically induced
- Arrhythmias, Cardiac/enzymology
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/physiopathology
- Cells, Cultured
- Dose-Response Relationship, Drug
- Epigenesis, Genetic/drug effects
- Gene Expression Profiling/methods
- Gene Expression Regulation
- Genotype
- Heart Diseases/chemically induced
- Heart Diseases/enzymology
- Heart Diseases/genetics
- Heart Diseases/physiopathology
- Heart Rate/drug effects
- Histone Deacetylase Inhibitors/toxicity
- Humans
- Induced Pluripotent Stem Cells/drug effects
- Induced Pluripotent Stem Cells/enzymology
- Myocardial Contraction/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Oligonucleotide Array Sequence Analysis
- Phenotype
- Risk Assessment
- Time Factors
- Transcription, Genetic/drug effects
Collapse
Affiliation(s)
- Ivan Kopljar
- Discovery Sciences, Janssen Research and Development, Janssen Pharmaceutica, Beerse, Belgium
| | - David J Gallacher
- Discovery Sciences, Janssen Research and Development, Janssen Pharmaceutica, Beerse, Belgium
| | - An De Bondt
- Discovery Sciences, Janssen Research and Development, Janssen Pharmaceutica, Beerse, Belgium
| | | | - Eddy Vlaminckx
- Discovery Sciences, Janssen Research and Development, Janssen Pharmaceutica, Beerse, Belgium
| | - Ilse Van den Wyngaert
- Discovery Sciences, Janssen Research and Development, Janssen Pharmaceutica, Beerse, Belgium
| | - Hua Rong Lu
- Discovery Sciences, Janssen Research and Development, Janssen Pharmaceutica, Beerse, Belgium
| |
Collapse
|
128
|
Wang J, Hu X, Jiang H. HDAC inhibition: A novel therapeutic approach for attenuating heart failure by suppressing cardiac remodeling. Int J Cardiol 2016; 214:41-2. [PMID: 27057972 DOI: 10.1016/j.ijcard.2016.03.188] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 03/23/2016] [Indexed: 11/18/2022]
Affiliation(s)
- Jichun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Hubei Key Laboratory of Cardiology, Jiefang Road 238, Wuchang, 430060 Wuhan, PR China
| | - Xiaorong Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Hubei Key Laboratory of Cardiology, Jiefang Road 238, Wuchang, 430060 Wuhan, PR China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Hubei Key Laboratory of Cardiology, Jiefang Road 238, Wuchang, 430060 Wuhan, PR China.
| |
Collapse
|
129
|
Zhang D, Hu X, Henning RH, Brundel BJJM. Keeping up the balance: role of HDACs in cardiac proteostasis and therapeutic implications for atrial fibrillation. Cardiovasc Res 2015; 109:519-26. [PMID: 26645980 DOI: 10.1093/cvr/cvv265] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 10/29/2015] [Indexed: 12/16/2022] Open
Abstract
Cardiomyocytes are long-lived post-mitotic cells with limited regenerative capacity. Proper cardiomyocyte function depends critically on the maintenance of a healthy homeostasis of protein expression, folding, assembly, trafficking, function, and degradation, together commonly referred to as proteostasis. Impairment of proteostasis has a prominent role in the pathophysiology of ageing-related neurodegenerative diseases including Huntington's, Parkinson's, and Alzheimer's disease. Emerging evidence reveals also a role for impaired proteostasis in the pathophysiology of common human cardiac diseases such as cardiac hypertrophy, dilated and ischaemic cardiomyopathies, and atrial fibrillation (AF). Histone deacetylases (HDACs) have recently been recognized as key modulators which control cardiac proteostasis by deacetylating various proteins. By deacetylating chromatin proteins, including histones, HDACs modulate epigenetic regulation of pathological gene expression. Also, HDACs exert a broad range of functions outside the nucleus by deacetylating structural and contractile proteins. The cytosolic actions of HDACs result in changed protein function through post-translational modifications and/or modulation of their degradation. This review describes the mechanisms underlying the derailment of proteostasis in AF and subsequently focuses on the role of HDACs herein. In addition, the therapeutic potential of HDAC inhibition to maintain a healthy proteostasis resulting in a delay in AF onset and progression is discussed.
Collapse
Affiliation(s)
- Deli Zhang
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, PO Box 30 001, 9700RB Groningen, The Netherlands
| | - Xu Hu
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, PO Box 30 001, 9700RB Groningen, The Netherlands
| | - Robert H Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, PO Box 30 001, 9700RB Groningen, The Netherlands
| | - Bianca J J M Brundel
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, PO Box 30 001, 9700RB Groningen, The Netherlands Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
130
|
Komaravelli N, Tian B, Ivanciuc T, Mautemps N, Brasier AR, Garofalo RP, Casola A. Respiratory syncytial virus infection down-regulates antioxidant enzyme expression by triggering deacetylation-proteasomal degradation of Nrf2. Free Radic Biol Med 2015; 88:391-403. [PMID: 26073125 PMCID: PMC4628892 DOI: 10.1016/j.freeradbiomed.2015.05.043] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 05/21/2015] [Accepted: 05/27/2015] [Indexed: 11/23/2022]
Abstract
Respiratory syncytial virus (RSV) is the most important cause of viral acute respiratory tract infections and hospitalizations in children, for which no vaccine or treatment is available. RSV infection in cells, mice, and children leads to rapid generation of reactive oxygen species, which are associated with oxidative stress and lung damage, due to a significant decrease in the expression of airway antioxidant enzymes (AOEs). Oxidative stress plays an important role in the pathogenesis of RSV-induced lung disease, as antioxidants ameliorate clinical disease and inflammation in vivo. The aim of this study is to investigate the unknown mechanism(s) of virus-induced inhibition of AOE expression. RSV infection is shown to induce a progressive reduction in nuclear and total cellular levels of the transcription factor NF-E2-related factor 2 (Nrf2), resulting in decreased binding to endogenous AOE gene promoters and decreased AOE expression. RSV induces Nrf2 deacetylation and degradation via the proteasome pathway in vitro and in vivo. Histone deacetylase and proteasome inhibitors block Nrf2 degradation and increase Nrf2 binding to AOE endogenous promoters, resulting in increased AOE expression. Known inducers of Nrf2 are able to increase Nrf2 activation and subsequent AOE expression during RSV infection in vitro and in vivo, with significant amelioration of oxidative stress. This is the first study to investigate the mechanism(s) of virus-induced inhibition of AOE expression. RSV-induced inhibition of Nrf2 activation, due to deacetylation and proteasomal degradation, could be targeted for therapeutic intervention aimed to increase airway antioxidant capacity during infection.
Collapse
Affiliation(s)
- Narayana Komaravelli
- Department of Pediatrics, University of Texas Medical Branch at Galveston, TX 77555, USA
| | - Bing Tian
- Department of Internal Medicine, University of Texas Medical Branch at Galveston, TX 77555, USA
| | - Teodora Ivanciuc
- Department of Pediatrics, University of Texas Medical Branch at Galveston, TX 77555, USA
| | - Nicholas Mautemps
- Department of Pediatrics, University of Texas Medical Branch at Galveston, TX 77555, USA
| | - Allan R Brasier
- Department of Internal Medicine, University of Texas Medical Branch at Galveston, TX 77555, USA; Department of Sealy Center for Molecular Medicine, University of Texas Medical Branch at Galveston, TX 77555, USA
| | - Roberto P Garofalo
- Department of Pediatrics, University of Texas Medical Branch at Galveston, TX 77555, USA
| | - Antonella Casola
- Department of Pediatrics, University of Texas Medical Branch at Galveston, TX 77555, USA; Department of Sealy Center for Molecular Medicine, University of Texas Medical Branch at Galveston, TX 77555, USA.
| |
Collapse
|
131
|
Noor Z, Afzal N, Rashid S. Exploration of Novel Inhibitors for Class I Histone Deacetylase Isoforms by QSAR Modeling and Molecular Dynamics Simulation Assays. PLoS One 2015; 10:e0139588. [PMID: 26431201 PMCID: PMC4592208 DOI: 10.1371/journal.pone.0139588] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 09/15/2015] [Indexed: 12/20/2022] Open
Abstract
Histone deacetylases (HDAC) are metal-dependent enzymes and considered as important targets for cell functioning. Particularly, higher expression of class I HDACs is common in the onset of multiple malignancies which results in deregulation of many target genes involved in cell growth, differentiation and survival. Although substantial attempts have been made to control the irregular functioning of HDACs by employing various inhibitors with high sensitivity towards transformed cells, limited success has been achieved in epigenetic cancer therapy. Here in this study, we used ligand-based pharmacophore and 2-dimensional quantitative structure activity relationship (QSAR) modeling approaches for targeting class I HDAC isoforms. Pharmacophore models were generated by taking into account the known IC50 values and experimental energy scores with extensive validations. The QSAR model having an external R2 value of 0.93 was employed for virtual screening of compound libraries. 10 potential lead compounds (C1-C10) were short-listed having strong binding affinities for HDACs, out of which 2 compounds (C8 and C9) were able to interact with all members of class I HDACs. The potential binding modes of HDAC2 and HDAC8 to C8 were explored through molecular dynamics simulations. Overall, bioactivity and ligand efficiency (binding energy/non-hydrogen atoms) profiles suggested that proposed hits may be more effective inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Zainab Noor
- National Center for Bioinformatics, Quaid I Azam University, Islamabad, Pakistan
| | - Noreen Afzal
- National Center for Bioinformatics, Quaid I Azam University, Islamabad, Pakistan
| | - Sajid Rashid
- National Center for Bioinformatics, Quaid I Azam University, Islamabad, Pakistan
| |
Collapse
|
132
|
Huang YY, Fang ZF, Hu XQ, Zhou SH. HDAC inhibition: A novel therapeutic target for pulmonary hypertension by reducing right ventricular hypertrophy through diverse pathological mechanisms. Int J Cardiol 2015; 196:125-6. [DOI: 10.1016/j.ijcard.2015.05.170] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 05/29/2015] [Indexed: 10/23/2022]
|
133
|
Renaud L, Harris LG, Mani SK, Kasiganesan H, Chou JC, Baicu CF, Van Laer A, Akerman AW, Stroud RE, Jones JA, Zile MR, Menick DR. HDACs Regulate miR-133a Expression in Pressure Overload-Induced Cardiac Fibrosis. Circ Heart Fail 2015; 8:1094-104. [PMID: 26371176 DOI: 10.1161/circheartfailure.114.001781] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 09/02/2015] [Indexed: 01/04/2023]
Abstract
BACKGROUND MicroRNAs (miRNAs) and histone deacetylases (HDACs) serve a significant role in the pathogenesis of a variety of cardiovascular diseases. The transcriptional regulation of miRNAs is poorly understood in cardiac hypertrophy. We investigated whether the expression of miR-133a is epigenetically regulated by class I and IIb HDACs during hypertrophic remodeling. METHODS AND RESULTS Transverse aortic constriction (TAC) was performed in CD1 mice to induce pressure overload hypertrophy. Mice were treated with class I and IIb HDAC inhibitor (HDACi) via drinking water for 2 and 4 weeks post TAC. miRNA expression was determined by real-time polymerase chain reaction. Echocardiography was performed at baseline and post TAC end points for structural and functional assessment. Chromatin immunoprecipitation was used to identify HDACs and transcription factors associated with miR-133a promoter. miR-133a expression was downregulated by 0.7- and 0.5-fold at 2 and 4 weeks post TAC, respectively, when compared with vehicle control (P<0.05). HDAC inhibition prevented this significant decrease 2 weeks post TAC and maintained miR-133a expression near vehicle control levels, which coincided with (1) a decrease in connective tissue growth factor expression, (2) a reduction in cardiac fibrosis and left atrium diameter (marker of end-diastolic pressure), suggesting an improvement in diastolic function. Chromatin immunoprecipitation analysis revealed that HDAC1 and HDAC2 are present on the miR-133a enhancer regions. CONCLUSIONS The results reveal that HDACs play a role in the regulation of pressure overload-induced miR-133a downregulation. This work is the first to provide insight into an epigenetic-miRNA regulatory pathway in pressure overload-induced cardiac fibrosis.
Collapse
Affiliation(s)
- Ludivine Renaud
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - Lillianne G Harris
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - Santhosh K Mani
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - Harinath Kasiganesan
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - James C Chou
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - Catalin F Baicu
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - An Van Laer
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - Adam W Akerman
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - Robert E Stroud
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - Jeffrey A Jones
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - Michael R Zile
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.)
| | - Donald R Menick
- From the Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute (L.R., L.G.H., S.K.M., H.K., C.F.B., A.V.L., M.R.Z., D.R.M.), Division of Cardiothoracic Surgery, Department of Cardiothoracic Surgical Research (A.W.A., R.E.S., J.A.J.), and Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (J.C.C.), The Medical University of South Carolina, Charleston; and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (J.A.J., M.R.Z., D.R.M.).
| |
Collapse
|
134
|
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
|
135
|
Mayer SC, Gilsbach R, Preissl S, Monroy Ordonez EB, Schnick T, Beetz N, Lother A, Rommel C, Ihle H, Bugger H, Rühle F, Schrepper A, Schwarzer M, Heilmann C, Bönisch U, Gupta SK, Wilpert J, Kretz O, von Elverfeldt D, Orth J, Aktories K, Beyersdorf F, Bode C, Stiller B, Krüger M, Thum T, Doenst T, Stoll M, Hein L. Adrenergic Repression of the Epigenetic Reader MeCP2 Facilitates Cardiac Adaptation in Chronic Heart Failure. Circ Res 2015. [PMID: 26195221 PMCID: PMC4568894 DOI: 10.1161/circresaha.115.306721] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Supplemental Digital Content is available in the text. In chronic heart failure, increased adrenergic activation contributes to structural remodeling and altered gene expression. Although adrenergic signaling alters histone modifications, it is unknown, whether it also affects other epigenetic processes, including DNA methylation and its recognition.
Collapse
Affiliation(s)
- Sandra C Mayer
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Ralf Gilsbach
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Sebastian Preissl
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Elsa Beatriz Monroy Ordonez
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Tilman Schnick
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Nadine Beetz
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Achim Lother
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Carolin Rommel
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Hannah Ihle
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Heiko Bugger
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Frank Rühle
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Andrea Schrepper
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Michael Schwarzer
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Claudia Heilmann
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Ulrike Bönisch
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Shashi Kumar Gupta
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Jochen Wilpert
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Oliver Kretz
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Dominik von Elverfeldt
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Joachim Orth
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Klaus Aktories
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Friedhelm Beyersdorf
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Christoph Bode
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Brigitte Stiller
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Markus Krüger
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Thomas Thum
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Torsten Doenst
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Monika Stoll
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.)
| | - Lutz Hein
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (S.C.M., R.G., S.P., E.B.M.O., T.S., N.B., A.L., C.R., H.I., J.O., K.A., L.H.), Hermann-Staudinger-Graduiertenschule (S.P.), University Heart Center Freiburg-Bad Krozingen (T.S., A.L., H.B., C.H., F.B., C.B., B.S.), Department of Medicine IV, Nephrology and Primary Care, Medical Center (J.W.), Institute of Anatomy and Cell Biology (O.K.), Renal Division, University Clinic Freiburg (O.K.), Medical Physics (D.E.), and BIOSS Centre for Biological Signalling Studies (L.H.), University of Freiburg, Freiburg, Germany; Department of Molecular Biology, UT Southwestern Medical Center at Dallas, TX (N.B.); Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany (F.R., M.S.); Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany (A.S., M.S., T.D.); Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany (U.B.); Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx (S.K.G., T.T.) and REBIRTH Excellence Cluster (T.T.), Hannover Medical School, Hannover, Germany; Max-Planck-Institut für Herz- und Lungenforschung, Bad Nauheim, Germany (M.K.); and National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.).
| |
Collapse
|
136
|
Aune SE, Herr DJ, Kutz CJ, Menick DR. Histone Deacetylases Exert Class-Specific Roles in Conditioning the Brain and Heart Against Acute Ischemic Injury. Front Neurol 2015; 6:145. [PMID: 26175715 PMCID: PMC4485035 DOI: 10.3389/fneur.2015.00145] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 06/15/2015] [Indexed: 12/12/2022] Open
Abstract
Ischemia-reperfusion (IR) injury comprises a significant portion of morbidity and mortality from heart and brain diseases worldwide. This enduring clinical problem has inspired myriad reports in the scientific literature of experimental interventions seeking to elucidate the pathology of IR injury. Elective cardiac surgery presents perhaps the most viable scenario for protecting the heart and brain from IR injury due to the opportunity to condition the organs prior to insult. The physiological parameters for the preconditioning of vital organs prior to insult through mechanical and pharmacological maneuvers have been heavily examined. These investigations have revealed new insights into how preconditioning alters cellular responses to IR injury. However, the promise of preconditioning remains unfulfilled at the clinical level, and research seeking to implicate cell signals essential to this protection continues. Recent discoveries in molecular biology have revealed that gene expression can be controlled through posttranslational modifications, without altering the chemical structure of the genetic code. In this scenario, gene expression is repressed by enzymes that cause chromatin compaction through catalytic removal of acetyl moieties from lysine residues on histones. These enzymes, called histone deacetylases (HDACs), can be inhibited pharmacologically, leading to the de-repression of protective genes. The discovery that HDACs can also alter the function of non-histone proteins through posttranslational deacetylation has expanded the potential impact of HDAC inhibitors for the treatment of human disease. HDAC inhibitors have been applied in a very small number of experimental models of IR. However, the scientific literature contains an increasing number of reports demonstrating that HDACs converge on preconditioning signals in the cell. This review will describe the influence of HDACs on major preconditioning signaling pathways in the heart and brain.
Collapse
Affiliation(s)
- Sverre E Aune
- Gazes Cardiac Research Institute, Medical University of South Carolina , Charleston, SC , USA
| | - Daniel J Herr
- Gazes Cardiac Research Institute, Medical University of South Carolina , Charleston, SC , USA
| | - Craig J Kutz
- Gazes Cardiac Research Institute, Medical University of South Carolina , Charleston, SC , USA
| | - Donald R Menick
- Gazes Cardiac Research Institute, Medical University of South Carolina , Charleston, SC , USA
| |
Collapse
|
137
|
Passacquale G, Phinikaridou A, Warboys C, Cooper M, Lavin B, Alfieri A, Andia ME, Botnar RM, Ferro A. Aspirin-induced histone acetylation in endothelial cells enhances synthesis of the secreted isoform of netrin-1 thus inhibiting monocyte vascular infiltration. Br J Pharmacol 2015; 172:3548-64. [PMID: 25824964 PMCID: PMC4507159 DOI: 10.1111/bph.13144] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 02/19/2015] [Accepted: 03/23/2015] [Indexed: 12/18/2022] Open
Abstract
Background and Purpose There are conflicting data regarding whether netrin-1 retards or accelerates atherosclerosis progression, as it can lead either to monocyte repulsion from or retention within plaques depending on its cellular source. We investigated the effect of aspirin, which is widely used in cardiovascular prophylaxis, on the synthesis of different isoforms of netrin-1 by endothelial cells under pro-inflammatory conditions, and defined the net effect of aspirin-dependent systemic modulation of netrin-1 on atherosclerosis progression. Experimental Approach Netrin-1 synthesis was studied in vitro using human endothelial cells stimulated with TNF-α, with or without aspirin treatment. In vivo experiments were conducted in ApoE−/− mice fed with a high-fat diet (HFD), receiving either aspirin or clopidogrel. Key Results TNF-α-induced NF-κB activation up-regulated the nuclear isoform of netrin-1, while simultaneously reducing secreted netrin-1. Down-regulation of the secreted isoform compromised the chemorepellent action of the endothelium against monocyte chemotaxis. Aspirin counteracted TNF-α-mediated effects on netrin-1 synthesis by endothelial cells through COX-dependent inhibition of NF-κB and concomitant histone hyperacetylation. Administration of aspirin to ApoE−/− mice on HFD increased blood and arterial wall levels of netrin-1 independently of its effects on platelets, accompanied by reduced plaque size and content of monocytes/macrophages, compared with untreated or clopidogrel-treated mice. In vivo blockade of netrin-1 enhanced monocyte plaque infiltration in aspirin-treated ApoE−/− mice. Conclusions and Implications Aspirin counteracts down-regulation of secreted netrin-1 induced by pro-inflammatory stimuli in endothelial cells. The aspirin-dependent increase of netrin-1 in ApoE−/− mice exerts anti-atherogenic effects by preventing arterial accumulation of monocytes.
Collapse
Affiliation(s)
- Gabriella Passacquale
- Department of Clinical Pharmacology, BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| | - Alkystis Phinikaridou
- Division of Imaging Sciences and Biomedical Engineering, BHF Centre of Research Excellence and the Wellcome Trust/EPSRC Medical Engineering Centre, King's College London, London, UK
| | - Christina Warboys
- Department of Clinical Pharmacology, BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| | - Margaret Cooper
- Division of Imaging Sciences and Biomedical Engineering, BHF Centre of Research Excellence and the Wellcome Trust/EPSRC Medical Engineering Centre, King's College London, London, UK
| | - Begona Lavin
- Division of Imaging Sciences and Biomedical Engineering, BHF Centre of Research Excellence and the Wellcome Trust/EPSRC Medical Engineering Centre, King's College London, London, UK
| | - Alessio Alfieri
- Department of Vascular Biology, BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| | - Marcelo E Andia
- Division of Imaging Sciences and Biomedical Engineering, BHF Centre of Research Excellence and the Wellcome Trust/EPSRC Medical Engineering Centre, King's College London, London, UK
| | - Rene M Botnar
- Division of Imaging Sciences and Biomedical Engineering, BHF Centre of Research Excellence and the Wellcome Trust/EPSRC Medical Engineering Centre, King's College London, London, UK
| | - Albert Ferro
- Department of Clinical Pharmacology, BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| |
Collapse
|
138
|
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
|
139
|
Epigenetic pathways in macrophages emerge as novel targets in atherosclerosis. Eur J Pharmacol 2015; 763:79-89. [PMID: 26004034 DOI: 10.1016/j.ejphar.2015.03.101] [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: 11/23/2014] [Revised: 01/25/2015] [Accepted: 03/05/2015] [Indexed: 12/13/2022]
Abstract
Atherosclerosis is a lipid-driven chronic inflammatory disorder. Monocytes and macrophages are key immune cells in the development of disease and clinical outcome. It is becoming increasingly clear that epigenetic pathways govern many aspects of monocyte and macrophage differentiation and activation. The dynamic regulation of epigenetic patterns provides opportunities to alter disease-associated epigenetic states. Therefore, pharmaceutical companies have embraced the targeting of epigenetic processes as new approaches for interventions. Particularly histone deacetylase (Hdac) inhibitors and DNA-methyltransferase inhibitors have long received attention and several of them have been approved for clinical use in relation to hematological malignancies. The key focus is still on oncology, but Alzheimer's disease, Huntington's disease and inflammatory disorders are coming in focus as well. These developments raise opportunities for the epigenetic targeting in cardiovascular disease (CVD). In this review we discuss the epigenetic regulation of the inflammatory pathways in relation to atherosclerosis with a specific attention to monocyte- and macrophage-related processes. What are the opportunities for future therapy of atherosclerosis by epigenetic interventions?
Collapse
|
140
|
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: 24] [Impact Index Per Article: 2.4] [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
|
141
|
Wang J, Hu X, Jiang H. HDAC inhibition: A novel therapeutic target for attenuating myocardial ischemia and reperfusion injury by reversing cardiac remodeling. Int J Cardiol 2015; 190:126-7. [PMID: 25918063 DOI: 10.1016/j.ijcard.2015.04.172] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Accepted: 04/20/2015] [Indexed: 12/19/2022]
Affiliation(s)
- Jichun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Jiefang Road 238, Wuchang, 430060 Wuhan, PR China
| | - Xiaorong Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Jiefang Road 238, Wuchang, 430060 Wuhan, PR China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Jiefang Road 238, Wuchang, 430060 Wuhan, PR China.
| |
Collapse
|
142
|
Mani SK, Kern CB, Kimbrough D, Addy B, Kasiganesan H, Rivers WT, Patel RK, Chou JC, Spinale FG, Mukherjee R, Menick DR. Inhibition of class I histone deacetylase activity represses matrix metalloproteinase-2 and -9 expression and preserves LV function postmyocardial infarction. Am J Physiol Heart Circ Physiol 2015; 308:H1391-401. [PMID: 25795711 DOI: 10.1152/ajpheart.00390.2014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 03/10/2015] [Indexed: 11/22/2022]
Abstract
Left ventricular (LV) remodeling, after myocardial infarction (MI), can result in LV dilation and LV pump dysfunction. Post-MI induction of matrix metalloproteinases (MMPs), particularly MMP-2 and MMP-9, have been implicated as causing deleterious effects on LV and extracellular matrix remodeling in the MI region and within the initially unaffected remote zone. Histone deacetylases (HDACs) are a class of enzymes that affect the transcriptional regulation of genes during pathological conditions. We assessed the efficacy of both class I/IIb- and class I-selective HDAC inhibitors on MMP-2 and MMP-9 abundance and determined if treatment resulted in the attenuation of adverse LV and extracellular matrix remodeling and improved LV pump function post-MI. MI was surgically induced in MMP-9 promoter reporter mice and randomized for treatment with a class I/IIb HDAC inhibitor for 7 days post-MI. After MI, LV dilation, LV pump dysfunction, and activation of the MMP-9 gene promoter were significantly attenuated in mice treated with either the class I/IIb HDAC inhibitor tichostatin A or suberanilohydroxamic acid (voronistat) compared with MI-only mice. Immunohistological staining and zymographic levels of MMP-2 and MMP-9 were reduced with either tichostatin A or suberanilohydroxamic acid treatment. Class I HDAC activity was dramatically increased post-MI. Treatment with the selective class I HDAC inhibitor PD-106 reduced post-MI levels of both MMP-2 and MMP-9 and attenuated LV dilation and LV pump dysfunction post-MI, similar to class I/IIb HDAC inhibition. Taken together, these unique findings demonstrate that selective inhibition of class I HDACs may provide a novel therapeutic means to attenuate adverse LV remodeling post-MI.
Collapse
Affiliation(s)
- Santhosh K Mani
- Gazes Cardiac Research Institute, Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Christine B Kern
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Denise Kimbrough
- Gazes Cardiac Research Institute, Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Benjamin Addy
- Gazes Cardiac Research Institute, Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Harinath Kasiganesan
- Gazes Cardiac Research Institute, Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - William T Rivers
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Risha K Patel
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - James C Chou
- Department of Pharmaceutical Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Francis G Spinale
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Rupak Mukherjee
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Donald R Menick
- Gazes Cardiac Research Institute, Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina; Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, South Carolina; and
| |
Collapse
|
143
|
Histone deacetylases: structural determinants of inhibitor selectivity. Drug Discov Today 2015; 20:718-35. [PMID: 25687212 DOI: 10.1016/j.drudis.2015.01.007] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/17/2014] [Accepted: 01/14/2015] [Indexed: 12/22/2022]
Abstract
Histone deacetylases (HDACs) are epigenetic targets with an important role in cancer, neurodegeneration, inflammation, and metabolic disorders. Although clinically effective HDAC inhibitors have been developed, the design of inhibitors with the desired isoform(s) selectivity remains a challenge. Selective inhibitors could help clarify the function of each isoform, and provide therapeutic agents having potentially fewer adverse effects. Crystal structures of several HDACs have been reported, enabling structure-based drug design and providing important information to understand enzyme function. Here, we provide a comprehensive review of the structural information available on HDACs, discussing both conserved and isoform-specific structural and mechanistic features. We focus on distinctive aspects that help rationalize inhibitor selectivity, and provide structure-based recommendations for achieving the desired selectivity.
Collapse
|
144
|
Abstract
The molecular signatures of epigenetic regulation and chromatin architectures are fundamental to genetically determined biological processes. Covalent and post-translational chemical modification of the chromatin template can sensitize the genome to changing environmental conditions to establish diverse functional states. Recent interest and research focus surrounds the direct connections between metabolism and chromatin dynamics, which now represents an important conceptual challenge to explain many aspects of metabolic dysfunction. Several components of the epigenetic machinery require intermediates of cellular metabolism for enzymatic function. Furthermore, changes to intracellular metabolism can alter the expression of specific histone methyltransferases and acetyltransferases conferring widespread variations in epigenetic modification patterns. Specific epigenetic influences of dietary glucose and lipid consumption, as well as undernutrition, are observed across numerous organs and pathways associated with metabolism. Studies have started to define the chromatin-dependent mechanisms underlying persistent and pathophysiological changes induced by altered metabolism. Importantly, numerous recent studies demonstrate that gene regulation underlying phenotypic determinants of adult metabolic health is influenced by maternal and early postnatal diet. These emerging concepts open new perspectives to combat the rising global epidemic of metabolic disorders.
Collapse
Affiliation(s)
- Samuel T. Keating
- From the Epigenetics in Human Health and Disease Laboratory (S.T.K., A.E.-O.) and Epigenomics Profiling Facility (S.T.K., A.E.-O.), Baker IDI Heart & Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia; Department of Pathology, The University of Melbourne, Victoria, Australia (A.E.-O.); and Central Clinical School, Department of Medicine, Monash University, Melbourne, Victoria, Australia (A.E.-O.)
| | - Assam El-Osta
- From the Epigenetics in Human Health and Disease Laboratory (S.T.K., A.E.-O.) and Epigenomics Profiling Facility (S.T.K., A.E.-O.), Baker IDI Heart & Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia; Department of Pathology, The University of Melbourne, Victoria, Australia (A.E.-O.); and Central Clinical School, Department of Medicine, Monash University, Melbourne, Victoria, Australia (A.E.-O.)
| |
Collapse
|
145
|
Clinical applications of epigenetics in cardiovascular disease: the long road ahead. Transl Res 2015; 165:143-53. [PMID: 24768945 PMCID: PMC4190107 DOI: 10.1016/j.trsl.2014.04.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/01/2014] [Accepted: 04/01/2014] [Indexed: 12/18/2022]
Abstract
Epigenetic processes, defined as heritable changes in gene expression that occur without changes to the DNA sequence, have emerged as a promising area of cardiovascular disease research. Epigenetic information transcends that of the genotype alone and provides for an integrated etiologic picture of cardiovascular disease pathogenesis because of the interaction of the epigenome with the environment. Epigenetic biomarkers, which include DNA methylation, histone modifications, and RNA-based mechanisms, are both modifiable and cell-type specific, which makes them not only responsive to the environment, but also an attractive target for drug development. However, the enthusiasm surrounding possible applications of cardiovascular epigenetics currently outpaces available evidence. In this review, the authors synthesize the evidence linking epigenetic changes with cardiovascular disease, emphasizing the gap between the translational potential and the clinical reality of cardiovascular epigenetics.
Collapse
|
146
|
Stratton MS, McKinsey TA. Acetyl-lysine erasers and readers in the control of pulmonary hypertension and right ventricular hypertrophy. Biochem Cell Biol 2014; 93:149-57. [PMID: 25707943 DOI: 10.1139/bcb-2014-0119] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Acetylation of lysine residues within nucleosomal histone tails provides a crucial mechanism for epigenetic control of gene expression. Acetyl groups are coupled to lysine residues by histone acetyltransferases (HATs) and removed by histone deacetylases (HDACs), which are also commonly referred to as "writers" and "erasers", respectively. In addition to altering the electrostatic properties of histones, lysine acetylation often creates docking sites for bromodomain-containing "reader" proteins. This review focuses on epigenetic control of pulmonary hypertension (PH) and associated right ventricular (RV) cardiac hypertrophy and failure. Effects of small molecule HDAC inhibitors in pre-clinical models of PH are highlighted. Furthermore, we describe the recently discovered role of bromodomain and extraterminal (BET) reader proteins in the control of cardiac hypertrophy, and provide evidence suggesting that one member of this family, BRD4, contributes to the pathogenesis of RV failure. Together, the data suggest intriguing potential for pharmacological epigenetic therapies for the treatment of PH and right-sided heart failure.
Collapse
Affiliation(s)
- Matthew S Stratton
- Department of Medicine, Division of Cardiology, University of Colorado Denver, 12700 E. 19th Ave, Aurora, CO 80045-0508, USA
| | | |
Collapse
|
147
|
Weeks KL, Avkiran M. Roles and post-translational regulation of cardiac class IIa histone deacetylase isoforms. J Physiol 2014; 593:1785-97. [PMID: 25362149 PMCID: PMC4405742 DOI: 10.1113/jphysiol.2014.282442] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 10/17/2014] [Indexed: 12/25/2022] Open
Abstract
Cardiomyocyte hypertrophy is an integral component of pathological cardiac remodelling in response to mechanical and chemical stresses in settings such as chronic hypertension or myocardial infarction. For hypertrophy to ensue, the pertinent mechanical and chemical signals need to be transmitted from membrane sensors (such as receptors for neurohormonal mediators) to the cardiomyocyte nucleus, leading to altered transcription of the genes that regulate cell growth. In recent years, nuclear histone deacetylases (HDACs) have attracted considerable attention as signal-responsive, distal regulators of the transcriptional reprogramming that in turn precipitates cardiomyocyte hypertrophy, with particular focus on the role of members of the class IIa family, such as HDAC4 and HDAC5. These histone deacetylase isoforms appear to repress cardiomyocyte hypertrophy through mechanisms that involve protein interactions in the cardiomyocyte nucleus, particularly with pro-hypertrophic transcription factors, rather than via histone deacetylation. In contrast, evidence indicates that class I HDACs promote cardiomyocyte hypertrophy through mechanisms that are dependent on their enzymatic activity and thus sensitive to pharmacological HDAC inhibitors. Although considerable progress has been made in understanding the roles of post-translational modifications (PTMs) such as phosphorylation, oxidation and proteolytic cleavage in regulating class IIa HDAC localisation and function, more work is required to explore the contributions of other PTMs, such as ubiquitination and sumoylation, as well as potential cross-regulatory interactions between distinct PTMs and between class IIa and class I HDAC isoforms.
Collapse
Affiliation(s)
| | - Metin Avkiran
- Corresponding author M. Avkiran: Cardiovascular Division, King's College London British Heart Foundation Centre, The Rayne Institute, St Thomas’ Hospital, Westminster Bridge Road, London SE1 7EH, UK.
| |
Collapse
|
148
|
Lkhagva B, Chang SL, Chen YC, Kao YH, Lin YK, Chiu CTH, Chen SA, Chen YJ. Histone deacetylase inhibition reduces pulmonary vein arrhythmogenesis through calcium regulation. Int J Cardiol 2014; 177:982-9. [PMID: 25449511 DOI: 10.1016/j.ijcard.2014.09.175] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 09/21/2014] [Accepted: 09/28/2014] [Indexed: 11/27/2022]
Abstract
Pulmonary veins (PVs) play a critical role in the pathophysiology of atrial fibrillation (AF). Histone deacetylases (HDACs) are vital to calcium homeostasis and AF genesis. However, the electrophysiological effects of HDAC inhibition were unclear. This study evaluated whether HDAC inhibition can regulate PV electrical activity through calcium modulation. Whole-cell patch-clamp, confocal microscopic with fluorescence, and Western blot were used to evaluate electrophysiological characteristics and Ca(2+) dynamics in isolated rabbit PV cardiomyocytes with and without MPT0E014 (a pan HDAC inhibitor), MS-275 (HDAC1 and 3 inhibitor), and MC-1568 (HDAC4 and 6 inhibitor) for 5~8h. Atrial electrical activity and induced-AF (rapid atrial pacing and acetylcholine infusion) were measured in rabbits with and without MPT0E014 (10mg/kg treated for 5 hours) in vivo. MPT0E014 (1 μM)-treated PV cardiomyocytes (n=12) had slower beating rates (2.1 ± 0.2 vs. 2.8 ± 0.1 Hz, p < 0.05) than control PV cardiomyocytes. However, control (n=11) and MPT0E014 (1 μM)-treated (n = 12) SAN cardiomyocytes had similar beating rates (3.2 ± 0.2 vs. 2.9 ± 0.3 Hz). MS-275-treated PV cardiomyocytes (n = 12, 2.3 ± 0.2 Hz), but not MC-1568-treated PV cardiomyocytes (n=14, 3.1 ± 0.3 Hz) had slower beating rates than control PV cardiomocytes. MPT0E014-treated PV cardiomyocytes (n=14) had a lower frequency (2.4 ± 0.6 vs. 0.3 ± 0.1 spark/mm/s, p < 0.05) of Ca(2+) sparks than control PV (n=17) cardiomyocytes. As compared to control, MPT0E014-treated PV cardiomyocytes had reduced Ca(2+) transient amplitudes, sodium-calcium exchanger currents, and ryanodine receptor expressions. Moreover, MPT0E014-treated rabbits had less AF and shorter AF duration than control rabbits. In conclusions, HDAC inhibition reduced PV arrhythmogenesis and AF inducibility with modulation on calcium homeostasis.
Collapse
Affiliation(s)
- Baigalmaa Lkhagva
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Lin Chang
- Division of Cardiology and Cardiovascular Research Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, 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
| | - Yung-Kuo Lin
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | | | - Shih-Ann Chen
- Division of Cardiology and Cardiovascular Research Center, Taipei Veterans General Hospital, 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
|
149
|
The Huntington's disease-related cardiomyopathy prevents a hypertrophic response in the R6/2 mouse model. PLoS One 2014; 9:e108961. [PMID: 25268775 PMCID: PMC4182603 DOI: 10.1371/journal.pone.0108961] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 09/05/2014] [Indexed: 11/29/2022] Open
Abstract
Huntington's disease (HD) is neurodegenerative disorder for which the mutation results in an extra-long tract of glutamines that causes the huntingtin protein to aggregate. It is characterized by neurological symptoms and brain pathology that is associated with nuclear and cytoplasmic aggregates and with transcriptional deregulation. Despite the fact that HD has been recognized principally as a neurological disease, there are multiple epidemiological studies showing that HD patients exhibit a high rate of cardiovascular events leading to heart failure. To unravel the mechanistic basis of cardiac dysfunction in HD, we employed a wide range of molecular techniques using the well-established genetic R6/2 mouse model that develop a considerable degree of the cardiac atrophy at end stage disease. We found that chronic treatment with isoproterenol, a potent beta-adrenoreceptor agonist, did not change the overall gross morphology of the HD murine hearts. However, there was a partial response to the beta-adrenergenic stimulation by the further re-expression of foetal genes. In addition we have profiled the expression level of Hdacs in the R6/2 murine hearts and found that the isoproterenol stimulation of Hdac expression was partially blocked. For the first time we established the Hdac transcriptional profile under hypertrophic conditions and found 10 out of 18 Hdacs to be markedly deregulated. Therefore, we conclude that R6/2 murine hearts are not able to respond to the chronic isoproterenol treatment to the same degree as wild type hearts and some of the hypertrophic signals are likely attenuated in the symptomatic HD animals.
Collapse
|
150
|
Blakeslee WW, Wysoczynski CL, Fritz KS, Nyborg JK, Churchill MEA, McKinsey TA. Class I HDAC inhibition stimulates cardiac protein SUMOylation through a post-translational mechanism. Cell Signal 2014; 26:2912-20. [PMID: 25220405 DOI: 10.1016/j.cellsig.2014.09.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 08/28/2014] [Accepted: 09/05/2014] [Indexed: 12/27/2022]
Abstract
Lysine residues are subject to a multitude of reversible post-translational modifications, including acetylation and SUMOylation. In the heart, enhancement of lysine acetylation or SUMOylation using histone deacetylase (HDAC) inhibitors or SUMO-1 gene transfer, respectively, has been shown to be cardioprotective. Here, we addressed whether there is crosstalk between lysine acetylation and SUMOylation in the heart. Treatment of cardiomyocytes and cardiac fibroblasts with pharmacological inhibitors of HDAC catalytic activity robustly increased conjugation of SUMO-1, but not SUMO-2/3, to several high molecular weight proteins in both cell types. The use of a battery of selective HDAC inhibitors and short hairpin RNAs demonstrated that HDAC2, which is a class I HDAC, is the primary HDAC isoform that controls cardiac protein SUMOylation. HDAC inhibitors stimulated protein SUMOylation in the absence of de novo gene transcription or protein synthesis, revealing a post-translational mechanism of HDAC inhibitor action. HDAC inhibition did not suppress the activity of de-SUMOylating enzymes, suggesting that increased protein SUMOylation in HDAC inhibitor-treated cells is due to stimulation of SUMO-1 conjugation rather than blockade of SUMO-1 cleavage. Consistent with this, multiple components of the SUMO conjugation machinery were capable of being acetylated in vitro. These findings reveal a novel role for reversible lysine acetylation in the control of SUMOylation in the heart, and suggest that cardioprotective actions of HDAC inhibitors are in part due to stimulation of protein SUMO-1-ylation in myocytes and fibroblasts.
Collapse
Affiliation(s)
- Weston W Blakeslee
- Department of Medicine, Division of Cardiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Christina L Wysoczynski
- Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Kristofer S Fritz
- Department of Pharmaceutical Sciences, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Jennifer K Nyborg
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Mair E A Churchill
- Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Timothy A McKinsey
- Department of Medicine, Division of Cardiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|