151
|
Han L, Li Y, Tchao J, Kaplan AD, Lin B, Li Y, Mich-Basso J, Lis A, Hassan N, London B, Bett GCL, Tobita K, Rasmusson RL, Yang L. Study familial hypertrophic cardiomyopathy using patient-specific induced pluripotent stem cells. Cardiovasc Res 2014; 104:258-69. [PMID: 25209314 PMCID: PMC4217687 DOI: 10.1093/cvr/cvu205] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Aims Familial hypertrophic cardiomyopathy (HCM) is one the most common heart disorders, with gene mutations in the cardiac sarcomere. Studying HCM with patient-specific induced pluripotent stem-cell (iPSC)-derived cardiomyocytes (CMs) would benefit the understanding of HCM mechanism, as well as the development of personalized therapeutic strategies. Methods and results To investigate the molecular mechanism underlying the abnormal CM functions in HCM, we derived iPSCs from an HCM patient with a single missense mutation (Arginine442Glycine) in the MYH7 gene. CMs were next enriched from HCM and healthy iPSCs, followed with whole transcriptome sequencing and pathway enrichment analysis. A widespread increase of genes responsible for ‘Cell Proliferation’ was observed in HCM iPSC-CMs when compared with control iPSC-CMs. Additionally, HCM iPSC-CMs exhibited disorganized sarcomeres and electrophysiological irregularities. Furthermore, disease phenotypes of HCM iPSC-CMs were attenuated with pharmaceutical treatments. Conclusion Overall, this study explored the possible patient-specific and mutation-specific disease mechanism of HCM, and demonstrates the potential of using HCM iPSC-CMs for future development of therapeutic strategies. Additionally, the whole methodology established in this study could be utilized to study mechanisms of other human-inherited heart diseases.
Collapse
Affiliation(s)
- Lu Han
- Department of Developmental Biology, University of Pittsburgh School of Medicine, 8117 Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA
| | - Yang Li
- Department of Developmental Biology, University of Pittsburgh School of Medicine, 8117 Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA
| | - Jason Tchao
- Department of Developmental Biology, University of Pittsburgh School of Medicine, 8117 Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA
| | - Aaron D Kaplan
- Center for Cellular and Systems Electrophysiology, Department of Physiology and Biophysics, SUNY, Buffalo, NY 14214, USA
| | - Bo Lin
- Department of Developmental Biology, University of Pittsburgh School of Medicine, 8117 Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA
| | - You Li
- Department of Developmental Biology, University of Pittsburgh School of Medicine, 8117 Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA
| | - Jocelyn Mich-Basso
- Department of Developmental Biology, University of Pittsburgh School of Medicine, 8117 Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA
| | - Agnieszka Lis
- Center for Cellular and Systems Electrophysiology, Department of Physiology and Biophysics, SUNY, Buffalo, NY 14214, USA
| | - Narmeen Hassan
- Department of Developmental Biology, University of Pittsburgh School of Medicine, 8117 Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA
| | - Barry London
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Glenna C L Bett
- Department of Obstetrics and Gynecology, SUNY, Buffalo, NY 14214, USA
| | - Kimimasa Tobita
- Department of Developmental Biology, University of Pittsburgh School of Medicine, 8117 Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA
| | - Randall L Rasmusson
- Center for Cellular and Systems Electrophysiology, Department of Physiology and Biophysics, SUNY, Buffalo, NY 14214, USA
| | - Lei Yang
- Department of Developmental Biology, University of Pittsburgh School of Medicine, 8117 Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA
| |
Collapse
|
152
|
Affiliation(s)
- Thomas G. Di Salvo
- Division of Cardiovascular Medicine, Vanderbilt Heart and Vascular Institute, Nashville TN
| | - Saptarsi M. Haldar
- Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland OH
- Harrington Heart & Vascular Institute, University Hospitals Case Medical Center, Cleveland, OH
| |
Collapse
|
153
|
Hyndman KA, Ho DH, Sega MF, Pollock JS. Histone deacetylase 1 reduces NO production in endothelial cells via lysine deacetylation of NO synthase 3. Am J Physiol Heart Circ Physiol 2014; 307:H803-9. [PMID: 25015965 DOI: 10.1152/ajpheart.00243.2014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The lysine acetylation state of nonhistone proteins may be regulated through histone deacetylases (HDACs). Evidence suggests that nitric oxide (NO) synthase 3 (NOS3; endothelial NOS) is posttranslationally lysine acetylated, leading to increased NO production in the endothelium. We tested the hypothesis that NOS3 is lysine acetylated and that upregulated HDAC1-mediated deacetylation leads to reduced NO production in endothelial cells. We determined that NOS3 is basally lysine acetylated in cultured bovine aortic endothelial cells (BAECs). In BAECs, HDAC1 is expressed in the nucleus and cytosol and forms a novel protein-protein interaction with NOS3. Overexpression of HDAC1 in BAECs resulted in a significant reduction in NOS3 lysine acetylation (control = 1.0 ± 0.1 and HDAC1 = 0.59 ± 0.08 arbitrary units, P < 0.01) and significantly blunted basal nitrite production (control 287.7 ± 29.1 and HDAC1 172.4 ± 31.7 pmol·mg(-1)·h(-1), P < 0.05) as well as attenuating endothelin-1-stimulated nitrite production (control = 481.8 ± 50.3 and HDAC1 243.1 ± 48.2 pmol·mg(-1)·h(-1), P < 0.05). While HDAC1 knockdown with small-interfering RNA resulted in no change in NOS3 acetylation level, yet increased basal nitrite production (730.6 ± 99.1 pmol·mg(-1)·h(-1)) and further exaggerated increases in endothelin-1 stimulated nitrite production (1276.9 ± 288.2 pmol·mg(-1)·h(-1)) was observed. Moreover, overexpression or knockdown of HDAC1 resulted in no significant effect on NOS3 protein expression or NOS3 phosphorylation sites T497, S635, or S1179. Thus these data indicate that upregulated HDAC1 decreases NOS3 activity, most likely through direct lysine deacetylation of NOS3. We propose that HDAC1-mediated deacetylation of NOS3 may represent a novel target for endothelial dysfunction.
Collapse
Affiliation(s)
- Kelly A Hyndman
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Dao H Ho
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Martiana F Sega
- Department of Medicine, Georgia Regents University, Augusta, Georgia
| | - Jennifer S Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and Department of Medicine, Georgia Regents University, Augusta, Georgia
| |
Collapse
|
154
|
De Raaf MA, Hussaini AA, Gomez-Arroyo J, Kraskaukas D, Farkas D, Happé C, Voelkel NF, Bogaard HJ. Histone deacetylase inhibition with trichostatin A does not reverse severe angioproliferative pulmonary hypertension in rats (2013 Grover Conference series). Pulm Circ 2014; 4:237-43. [PMID: 25006442 DOI: 10.1086/675986] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 01/09/2014] [Indexed: 01/08/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rapidly progressive and devastating disease characterized by remodeling of lung vessels, increased pulmonary vascular resistance, and eventually right ventricular hypertrophy and failure. Because histone deacetylase (HDAC) inhibitors are agents hampering tumor growth and cardiac hypertrophy, they have been attributed a therapeutic potential for patients with PAH. Outcomes of studies evaluating the use of HDAC inhibitors in models of PAH and right ventricular pressure overload have been equivocal, however. Here we describe the levels of HDAC activity in the lungs and hearts of rats with pulmonary hypertension and right heart hypertrophy or failure, experimentally induced by monocrotaline (MCT), the combined exposure to the VEGF-R inhibitor SU5416 and hypoxia (SuHx), and pulmonary artery banding (PAB). We show that HDAC activity levels are reduced in the lungs of rat with experimentally induced hypertension, whereas activity levels are increased in the hypertrophic hearts. In contrast to what was previously found in the MCT model, the HDAC inhibitor trichostatin A had no effect on pulmonary vascular remodeling in the SuHx model. When our results and those in the published literature are taken together, it is suggested that the effects of HDAC inhibitors in humans with PAH and associated RV failure are, at best, unpredictable. Significant progress can perhaps be made by using more specific HDAC inhibitors, but before clinical tests in human PAH can be undertaken, careful preclinical studies are required to determine potential cardiotoxicity.
Collapse
Affiliation(s)
- Michiel Alexander De Raaf
- Department of Pulmonology, Pulmonary Arterial Hypertension Knowledge Centre, VU University Medical Center, Amsterdam, The Netherlands
| | - Aysar Al Hussaini
- Pulmonary and Critical Care Medicine Division, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jose Gomez-Arroyo
- Pulmonary and Critical Care Medicine Division, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Donatas Kraskaukas
- Pulmonary and Critical Care Medicine Division, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Daniela Farkas
- Pulmonary and Critical Care Medicine Division, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Chris Happé
- Department of Pulmonology, Pulmonary Arterial Hypertension Knowledge Centre, VU University Medical Center, Amsterdam, The Netherlands
| | - Norbert F Voelkel
- Pulmonary and Critical Care Medicine Division, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Harm Jan Bogaard
- Department of Pulmonology, Pulmonary Arterial Hypertension Knowledge Centre, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
155
|
Aune SE, Herr DJ, Mani SK, Menick DR. Selective inhibition of class I but not class IIb histone deacetylases exerts cardiac protection from ischemia reperfusion. J Mol Cell Cardiol 2014; 72:138-45. [PMID: 24632412 PMCID: PMC4046340 DOI: 10.1016/j.yjmcc.2014.03.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 02/18/2014] [Accepted: 03/05/2014] [Indexed: 12/20/2022]
Abstract
While inhibition of class I/IIb histone deacetylases (HDACs) protects the mammalian heart from ischemia reperfusion (IR) injury, class selective effects remain unexamined. We hypothesized that selective inhibition of class I HDACs would preserve left ventricular contractile function following IR in isolated hearts. Male Sprague Dawley rats (n=6 per group) were injected with vehicle (dimethylsulfoxide, 0.63mg/kg), the class I/IIb HDAC inhibitor trichostatin A (1mg/kg), the class I HDAC inhibitor entinostat (MS-275, 10mg/kg), or the HDAC6 (class IIb) inhibitor tubastatin A (10mg/kg). After 24h, hearts were isolated and perfused in Langendorff mode for 30min (Sham) or subjected to 30min global ischemia and 120min global reperfusion (IR). A saline filled balloon attached to a pressure transducer was placed in the LV to monitor contractile function. After perfusion, LV tissue was collected for measurements of antioxidant protein levels and infarct area. At the conclusion of IR, MS-275 pretreatment was associated with significant preservation of developed pressure, rate of pressure generation, rate of pressure relaxation and rate pressure product, as compared to vehicle treated hearts. There was significant reduction of infarct area with MS-275 pretreatment. Contractile function was not significantly restored in hearts treated with trichostatin A or tubastatin A. Mitochondrial superoxide dismutase (SOD2) and catalase protein and mRNA in hearts from animals pretreated with MS-275 were increased following IR, as compared to Sham. This was associated with a dramatic enrichment of nuclear FOXO3a transcription factor, which mediates the expression of SOD2 and catalase. Tubastatin A treatment was associated with significantly decreased catalase levels after IR. Class I HDAC inhibition elicits protection of contractile function following IR, which is associated with increased expression of endogenous antioxidant enzymes. Class I/IIb HDAC inhibition with trichostatin A or selective inhibition of HDAC6 with tubastatin A was not protective. This study highlights the need for the development of new strategies that target specific HDAC isoforms in cardiac ischemia reperfusion.
Collapse
Affiliation(s)
- Sverre E Aune
- Gazes Cardiac Research Institute, Medical University of South Carolina, 114 Doughty St., Charleston, SC 29425, USA
| | - Daniel J Herr
- Gazes Cardiac Research Institute, Medical University of South Carolina, 114 Doughty St., Charleston, SC 29425, USA
| | - Santhosh K Mani
- Gazes Cardiac Research Institute, Medical University of South Carolina, 114 Doughty St., Charleston, SC 29425, USA
| | - Donald R Menick
- Gazes Cardiac Research Institute, Medical University of South Carolina, 114 Doughty St., Charleston, SC 29425, USA.
| |
Collapse
|
156
|
Sharifi-Sanjani M, Shoushtari AH, Quiroz M, Baust J, Sestito SF, Mosher M, Ross M, McTiernan CF, St Croix CM, Bilonick RA, Champion HC, Isenberg JS. Cardiac CD47 drives left ventricular heart failure through Ca2+-CaMKII-regulated induction of HDAC3. J Am Heart Assoc 2014; 3:e000670. [PMID: 24922625 PMCID: PMC4309049 DOI: 10.1161/jaha.113.000670] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Left ventricular heart failure (LVHF) remains progressive and fatal and is a formidable health problem because ever‐larger numbers of people are diagnosed with this disease. Therapeutics, while relieving symptoms and extending life in some cases, cannot resolve this process and transplant remains the option of last resort for many. Our team has described a widely expressed cell surface receptor (CD47) that is activated by its high‐affinity secreted ligand, thrombospondin 1 (TSP1), in acute injury and chronic disease; however, a role for activated CD47 in LVHF has not previously been proposed. Methods and Results In experimental LVHF TSP1‐CD47 signaling is increased concurrent with up‐regulation of cardiac histone deacetylase 3 (HDAC3). Mice mutated to lack CD47 displayed protection from transverse aortic constriction (TAC)‐driven LVHF with enhanced cardiac function, decreased cellular hypertrophy and fibrosis, decreased maladaptive autophagy, and decreased expression of HDAC3. In cell culture, treatment of cardiac myocyte CD47 with a TSP1‐derived peptide, which binds and activates CD47, increased HDAC3 expression and myocyte hypertrophy in a Ca2+/calmodulin protein kinase II (CaMKII)‐dependent manner. Conversely, antibody blocking of CD47 activation, or pharmacologic inhibition of CaMKII, suppressed HDAC3 expression, decreased myocyte hypertrophy, and mitigated established LVHF. Downstream gene suppression of HDAC3 mimicked the protective effects of CD47 blockade and decreased hypertrophy in myocytes and mitigated LVHF in animals. Conclusions These data identify a proximate role for the TSP1‐CD47 axis in promoting LVHF by CaKMII‐mediated up‐regulation of HDAC3 and suggest novel therapeutic opportunities.
Collapse
Affiliation(s)
- Maryam Sharifi-Sanjani
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA (M.S.S., M.Q., J.B., S.F.S., H.C.C., J.S.I.) Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA (M.S.S., H.C.C., J.S.I.)
| | - Ali Hakim Shoushtari
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA (A.H.S., H.C.C.)
| | - Marisol Quiroz
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA (M.S.S., M.Q., J.B., S.F.S., H.C.C., J.S.I.)
| | - Jeffrey Baust
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA (M.S.S., M.Q., J.B., S.F.S., H.C.C., J.S.I.)
| | - Samuel F Sestito
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA (M.S.S., M.Q., J.B., S.F.S., H.C.C., J.S.I.)
| | - Mackenzie Mosher
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA (M.M., M.R., C.M.S.C.)
| | - Mark Ross
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA (M.M., M.R., C.M.S.C.)
| | - Charles F McTiernan
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA (C.F.M.T.)
| | - Claudette M St Croix
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA (M.M., M.R., C.M.S.C.)
| | - Richard A Bilonick
- Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA (R.A.B.)
| | - Hunter C Champion
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA (M.S.S., M.Q., J.B., S.F.S., H.C.C., J.S.I.) Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA (M.S.S., H.C.C., J.S.I.) Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA (A.H.S., H.C.C.)
| | - Jeffrey S Isenberg
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA (M.S.S., M.Q., J.B., S.F.S., H.C.C., J.S.I.) Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA (M.S.S., H.C.C., J.S.I.)
| |
Collapse
|
157
|
Abstract
Studies of transcriptional mechanisms in heart failure have focused heavily on roles of sequence-specific DNA-binding factors such as NFAT, MEF2 and GATA4. Recent findings have illuminated crucial functions for epigenetic regulators in the control of cardiac structural remodeling and mechanical dysfunction in response to pathological stress. Here, we review the current understanding of chromatin-dependent signal transduction in cardiac gene control, and highlight the potential for pharmacologic regulation of BET acetyl-lysine binding proteins as a means of treating heart failure.
Collapse
|
158
|
Mathiyalagan P, Keating ST, Du XJ, El-Osta A. Chromatin modifications remodel cardiac gene expression. Cardiovasc Res 2014; 103:7-16. [PMID: 24812277 DOI: 10.1093/cvr/cvu122] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Signalling and transcriptional control involve precise programmes of gene activation and suppression necessary for cardiovascular physiology. Deep sequencing of DNA-bound transcription factors reveals a remarkable complexity of co-activators or co-repressors that serve to alter chromatin modification and regulate gene expression. The regulated complexes characterized by genome-wide mapping implicate the recruitment and exchange of proteins with specific enzymatic activities that include roles for histone acetylation and methylation in key developmental programmes of the heart. As for transcriptional changes in response to pathological stress, co-regulatory complexes are also differentially utilized to regulate genes in cardiac disease. Members of the histone deacetylase (HDAC) family catalyse the removal of acetyl groups from proteins whose pharmacological inhibition has profound effects preventing heart failure. HDACs interact with a complex co-regulatory network of transcription factors, chromatin-remodelling complexes, and specific histone modifiers to regulate gene expression in the heart. For example, the histone methyltransferase (HMT), enhancer of zeste homolog 2 (Ezh2), is regulated by HDAC inhibition and associated with pathological cardiac hypertrophy. The challenge now is to target the activity of enzymes involved in protein modification to prevent or reverse the expression of genes implicated with cardiac hypertrophy. In this review, we discuss the role of HDACs and HMTs with a focus on chromatin modification and gene function as well as the clinical treatment of heart failure.
Collapse
Affiliation(s)
- Prabhu Mathiyalagan
- Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia
| | - Samuel T Keating
- Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia
| | - Xiao-Jun Du
- Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia Central Clinical School, Faculty of Medicine, Monash University, Victoria, Australia
| | - Assam El-Osta
- Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia Central Clinical School, Faculty of Medicine, Monash University, Victoria, Australia Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
159
|
GATA-dependent regulatory switches establish atrioventricular canal specificity during heart development. Nat Commun 2014; 5:3680. [PMID: 24770533 PMCID: PMC4015328 DOI: 10.1038/ncomms4680] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 03/17/2014] [Indexed: 12/17/2022] Open
Abstract
The embryonic vertebrate heart tube develops an atrioventricular canal that divides the atrial and ventricular chambers, forms atrioventricular conduction tissue and organizes valve development. Here we assess the transcriptional mechanism underlying this localized differentiation process. We show that atrioventricular canal-specific enhancers are GATA-binding site-dependent and act as switches that repress gene activity in the chambers. We find that atrioventricular canal-specific gene loci are enriched in H3K27ac, a marker of active enhancers, in atrioventricular canal tissue and depleted in H3K27ac in chamber tissue. In the atrioventricular canal, Gata4 activates the enhancers in synergy with Bmp2/Smad signalling, leading to H3K27 acetylation. In contrast, in chambers, Gata4 cooperates with pan-cardiac Hdac1 and Hdac2 and chamber-specific Hey1 and Hey2, leading to H3K27 deacetylation and repression. We conclude that atrioventricular canal-specific enhancers are platforms integrating cardiac transcription factors, broadly active histone modification enzymes and localized co-factors to drive atrioventricular canal-specific gene activity. The atrioventricular canal partitions the developing vertebrate heart. Here, the authors show that the cardiac transcription factor Gata4 together with histone modification enzymes and localized co-factors binds atrioventricular canal-specific enhancers, thereby repressing gene activity in the cardiac chambers.
Collapse
|
160
|
Turgeon PJ, Sukumar AN, Marsden PA. Epigenetics of Cardiovascular Disease - A New "Beat" in Coronary Artery Disease. ACTA ACUST UNITED AC 2014; 2:37-52. [PMID: 25408699 DOI: 10.1159/000360766] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Genome-wide association studies (GWAS) have become a powerful tool in the identification of disease-associated variants. Unfortunately, many of these studies have found that the estimated variability in cardiovascular disease risk cannot be fully explained by traditional paradigms of genetic variation in protein coding genes. Moreover, traditional views do not sufficiently explain the well-known link between cardiovascular disease and environmental influence. We posit that epigenetics, defined as chromatin-based mechanisms important in the regulation of gene expression that do not involve changes in the DNA sequence per se, represents the missing link. The nuclear-based mechanisms that contribute to epigenetic gene regulation can be broadly separated into three unique but highly interrelated processes: DNA methylation and hydroxymethylation; histone density and post-translational modifications; and RNA-based mechanisms. Together they complement the cis/trans perspective on transcriptional control paradigms in blood vessels. Moreover, it provides a molecular basis for understanding how the environment impacts the genome to modify cardiovascular disease risk over the lifetime of a cell and its offspring. This review provides an introduction to epigenetic function and cardiovascular disease, with a focus on endothelial cell biology. Additionally, we highlight emerging concepts on epigenetic gene regulation that are highly relevant to atherosclerosis and coronary artery disease.
Collapse
Affiliation(s)
- Paul J Turgeon
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada ; Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Aravin N Sukumar
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada ; Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Philip A Marsden
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada ; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada ; Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
161
|
Soubrier F, Chung WK, Machado R, Grünig E, Aldred M, Geraci M, Loyd JE, Elliott CG, Trembath RC, Newman JH, Humbert M. Genetics and genomics of pulmonary arterial hypertension. J Am Coll Cardiol 2014; 62:D13-21. [PMID: 24355637 DOI: 10.1016/j.jacc.2013.10.035] [Citation(s) in RCA: 229] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 10/22/2013] [Indexed: 12/24/2022]
Abstract
Major discoveries have been obtained within the last decade in the field of hereditary predisposition to pulmonary arterial hypertension (PAH). Among them, the identification of bone morphogenetic protein receptor type 2 (BMPR2) as the major predisposing gene and activin A receptor type II-like kinase-1 (ACVRL1, also known as ALK1) as the major gene when PAH is associated with hereditary hemorrhagic telangiectasia. The mutation detection rate for the known genes is approximately 75% in familial PAH, but the mutation shortfall remains unexplained even after careful molecular investigation of these genes. To identify additional genetic variants predisposing to PAH, investigators harnessed the power of next-generation sequencing to successfully identify additional genes that will be described in this report. Furthermore, common genetic predisposing factors for PAH can be identified by genome-wide association studies and are detailed in this paper. The careful study of families and routine genetic diagnosis facilitated natural history studies based on large registries of PAH patients to be set up in different countries. These longitudinal or cross-sectional studies permitted the clinical characterization of PAH in mutation carriers to be accurately described. The availability of molecular genetic diagnosis has opened up a new field for patient care, including genetic counseling for a severe disease, taking into account that the major predisposing gene has a highly variable penetrance between families. Molecular information can be drawn from the genomic study of affected tissues in PAH, in particular, pulmonary vascular tissues and cells, to gain insight into the mechanisms leading to the development of the disease. High-throughput genomic techniques, on the basis of next-generation sequencing, now allow the accurate quantification and analysis of ribonucleic acid, species, including micro-ribonucleic acids, and allow for a genome-wide investigation of epigenetic or regulatory mechanisms, which include deoxyribonucleic acid methylation, histone methylation, and acetylation, or transcription factor binding.
Collapse
Affiliation(s)
- Florent Soubrier
- Genetics Department, Hospital Pitié-Salpêtrière, Assistance Publique Hôpitaux de Paris (APHP), Unité Mixte de Recherche en Sante (UMRS) 956 Institut National de la Sante et de la Recherche Medicale INSERM, Université Pierre et Marie Curie Paris 06 (UPMC), and Institute of Cardiometabolism and Nutrition (ICAN), Paris, France.
| | - Wendy K Chung
- Departments of Pediatrics and Medicine, Columbia University Medical Center, New York, New York
| | - Rajiv Machado
- University of Lincoln, School of Life Sciences, Lincoln, United Kingdom
| | - Ekkehard Grünig
- Centre for Pulmonary Hypertension at University Hospital Heidelberg, Heidelberg, Germany
| | - Micheala Aldred
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Mark Geraci
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Aurora, Colorado
| | - James E Loyd
- Pulmonary Hypertension Center, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center North, Nashville, Tennessee
| | - C Gregory Elliott
- Departments of Medicine at Intermountain Medical Center and the University of Utah, Salt Lake City, Utah
| | - Richard C Trembath
- Division of Genetics and Molecular Medicine, Kings College, London, United Kingdom
| | - John H Newman
- Pulmonary Hypertension Center, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center North, Nashville, Tennessee
| | - Marc Humbert
- Centre de Référence de l'Hypertension Pulmonaire Sévère, Service de Pneumologie, Hôpital de Bicêtre, APHP, Le Kremlin Bicêtre, Université Paris-Sud, Faculté de Médecine, Le Kremlin Bicêtre; Département Hospitalo-Universitaire (DHU) thorax Innovation, AP-HP, Le Kremlin Bicêtre; UMR_S 999, INSERM and Université Paris-Sud, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| |
Collapse
|
162
|
Zhang Y, Ren J. Targeting autophagy for the therapeutic application of histone deacetylase inhibitors in ischemia/reperfusion heart injury. Circulation 2014; 129:1088-91. [PMID: 24396040 DOI: 10.1161/circulationaha.113.008115] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Yingmei Zhang
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie (Y.Z., J.R.); and Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (Y.Z.)
| | | |
Collapse
|
163
|
Xie M, Kong Y, Tan W, May H, Battiprolu PK, Pedrozo Z, Wang ZV, Morales C, Luo X, Cho G, Jiang N, Jessen ME, Warner JJ, Lavandero S, Gillette TG, Turer AT, Hill JA. Histone deacetylase inhibition blunts ischemia/reperfusion injury by inducing cardiomyocyte autophagy. Circulation 2014; 129:1139-51. [PMID: 24396039 DOI: 10.1161/circulationaha.113.002416] [Citation(s) in RCA: 261] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Reperfusion accounts for a substantial fraction of the myocardial injury occurring with ischemic heart disease. Yet, no standard therapies are available targeting reperfusion injury. Here, we tested the hypothesis that suberoylanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor approved for cancer treatment by the US Food and Drug Administration, will blunt reperfusion injury. METHODS AND RESULTS Twenty-one rabbits were randomly assigned to 3 groups: (1) vehicle control, (2) SAHA pretreatment (1 day before and at surgery), and (3) SAHA treatment at the time of reperfusion only. Each arm was subjected to ischemia/reperfusion surgery (30 minutes coronary ligation, 24 hours reperfusion). In addition, cultured neonatal and adult rat ventricular cardiomyocytes were subjected to simulated ischemia/reperfusion to probe mechanism. SAHA reduced infarct size and partially rescued systolic function when administered either before surgery (pretreatment) or solely at the time of reperfusion. SAHA plasma concentrations were similar to those achieved in patients with cancer. In the infarct border zone, SAHA increased autophagic flux, assayed in both rabbit myocardium and in mice harboring an RFP-GFP-LC3 transgene. In cultured myocytes subjected to simulated ischemia/reperfusion, SAHA pretreatment reduced cell death by 40%. This reduction in cell death correlated with increased autophagic activity in SAHA-treated cells. RNAi-mediated knockdown of ATG7 and ATG5, essential autophagy proteins, abolished SAHA's cardioprotective effects. CONCLUSIONS The US Food and Drug Administration-approved anticancer histone deacetylase inhibitor, SAHA, reduces myocardial infarct size in a large animal model, even when delivered in the clinically relevant context of reperfusion. The cardioprotective effects of SAHA during ischemia/reperfusion occur, at least in part, through the induction of autophagic flux.
Collapse
Affiliation(s)
- Min Xie
- Departments of Internal Medicine (Cardiology) (M.X., Y.K., W.Y., H.M., P.K.B., Z.P., Z.V.W., C.M., X.L., G.C., N.J., J.J.W., S.L., T.G.G., A.T.T., J.A.H.), Cardiovascular and Thoracic Surgery (M.E.J.), Advanced Center for Chronic Diseases (ACCDiS) & Centro Estudios Moleculares de la Celula, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile (S.L.); and the Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas (J.A.H.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
164
|
Tao H, Shi KH, Yang JJ, Huang C, Zhan HY, Li J. Histone deacetylases in cardiac fibrosis: current perspectives for therapy. Cell Signal 2013; 26:521-7. [PMID: 24321371 DOI: 10.1016/j.cellsig.2013.11.037] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 11/30/2013] [Accepted: 11/30/2013] [Indexed: 12/17/2022]
Abstract
Cardiac fibrosis is an important pathological feature of cardiac remodeling in heart diseases. The molecular mechanisms of cardiac fibrosis are unknown. Histone deacetylases (HDACs) are enzymes that balance the acetylation activities of histone acetyltransferases on chromatin remodeling and play essential roles in regulating gene transcription. In recent years, the role of HDACs in cardiac fibrosis initiation and progression, as well as the therapeutic effects of HDAC inhibitors, has been well studied. Moreover, numerous studies indicated that HDAC activity is associated with the development and progression of cardiac fibrosis. In this review, the innovative aspects of HDACs are discussed, with respect to biogenesis, their role in cardiac fibrosis. Furthermore, the potential applications of HDAC inhibitors in the treatment of cardiac fibrosis associated with fibroblast activation and proliferation.
Collapse
Affiliation(s)
- Hui Tao
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei 230601, China; Cardiovascular Research Center, Anhui Medical University, Hefei 230601, China
| | - Kai-Hu Shi
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei 230601, China; Cardiovascular Research Center, Anhui Medical University, Hefei 230601, China.
| | - Jing-Jing Yang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Department of Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Cheng Huang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Hong-Ying Zhan
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei 230601, China; Cardiovascular Research Center, Anhui Medical University, Hefei 230601, China
| | - Jun Li
- School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
165
|
Lehmann LH, Worst BC, Stanmore DA, Backs J. Histone deacetylase signaling in cardioprotection. Cell Mol Life Sci 2013; 71:1673-90. [PMID: 24310814 PMCID: PMC3983897 DOI: 10.1007/s00018-013-1516-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 10/23/2013] [Accepted: 11/07/2013] [Indexed: 12/17/2022]
Abstract
Cardiovascular disease (CVD) represents a major challenge for health care systems, both in terms of the high mortality associated with it and the huge economic burden of its treatment. Although CVD represents a diverse range of disorders, they share common compensatory changes in the heart at the structural, cellular, and molecular level that, in the long term, can become maladaptive and lead to heart failure. Treatment of adverse cardiac remodeling is therefore an important step in preventing this fatal progression. Although previous efforts have been primarily focused on inhibition of deleterious signaling cascades, the stimulation of endogenous cardioprotective mechanisms offers a potent therapeutic tool. In this review, we discuss class I and class II histone deacetylases, a subset of chromatin-modifying enzymes known to have critical roles in the regulation of cardiac remodeling. In particular, we discuss their molecular modes of action and go on to consider how their inhibition or the stimulation of their intrinsic cardioprotective properties may provide a potential therapeutic route for the clinical treatment of CVD.
Collapse
Affiliation(s)
- Lorenz H. Lehmann
- Research Unit Cardiac Epigenetics, Internal Medicine III, Heidelberg University and DZHK (German Center for Cardiovascular Research), partner site Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Barbara C. Worst
- Research Unit Cardiac Epigenetics, Internal Medicine III, Heidelberg University and DZHK (German Center for Cardiovascular Research), partner site Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - David A. Stanmore
- Research Unit Cardiac Epigenetics, Internal Medicine III, Heidelberg University and DZHK (German Center for Cardiovascular Research), partner site Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Johannes Backs
- Research Unit Cardiac Epigenetics, Internal Medicine III, Heidelberg University and DZHK (German Center for Cardiovascular Research), partner site Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| |
Collapse
|
166
|
Singh RK, Lall N, Leedahl TS, McGillivray A, Mandal T, Haldar M, Mallik S, Cook G, Srivastava DK. Kinetic and thermodynamic rationale for suberoylanilide hydroxamic acid being a preferential human histone deacetylase 8 inhibitor as compared to the structurally similar ligand, trichostatin a. Biochemistry 2013; 52:8139-49. [PMID: 24079912 DOI: 10.1021/bi400740x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Of the different hydroxamate-based histone deacetylase (HDAC) inhibitors, suberoylanilide hydroxamic acid (SAHA) has been approved by the Food and Drug Administration for the treatment of T-cell lymphoma. Interestingly, a structurally similar inhibitor, trichostatin A (TSA), which has a higher in vitro inhibitory potency against HDAC8, reportedly shows poor efficacy in clinical settings. To gain molecular insight into this discriminatory feature, we performed transient kinetic and isothermal titration calorimetric studies for the interaction of SAHA and TSA with the recombinant form of human HDAC8. The transient kinetic data revealed that the binding of both inhibitors to the enzyme showed biphasic profiles, which represented an initial encounter of the enzyme with the inhibitor followed by the isomerization of the transient enzyme-inhibitor complexes. The temperature-dependent transient kinetic studies with these inhibitors revealed that the bimolecular process is primarily dominated by favorable enthalpic changes, as opposed to the isomerization step, which is solely contributed by entropic changes. The standard binding enthalpy (ΔH°) of SAHA, deduced from the transient kinetic as well as the isothermal titration calorimetric experiments, was 2-3 kcal/mol higher than that of TSA. The experimental data presented herein suggest that SAHA serves as a preferential (target-specific and -selective) HDAC8 inhibitor as compared to TSA. Arguments that the detailed kinetic and thermodynamic studies may guide the rational design of HDAC inhibitors as therapeutic agents are presented.
Collapse
Affiliation(s)
- Raushan K Singh
- Department of Chemistry and Biochemistry, North Dakota State University , Fargo, North Dakota 58102, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Walker LA, Buttrick PM. The right ventricle: biologic insights and response to disease: updated. Curr Cardiol Rev 2013; 9:73-81. [PMID: 23092273 PMCID: PMC3584309 DOI: 10.2174/157340313805076296] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2012] [Accepted: 10/27/2012] [Indexed: 02/07/2023] Open
Abstract
Despite ample evidence that right ventricular function is a critical determinant of the clinical response to a spectrum of cardiovascular diseases, there has been only a limited analysis of the unique and distinguishing physiologic properties of the RV under normal circumstances and in response to pathologic insults. This knowledge deficit is increasingly acknowledged. This review highlights some of these features and underscores the fact that rational therapy in RV failure needs to acknowledge its unique physiology and ought to be chamber specific. That is proven therapies for LV dysfunction do not necessarily apply to the RV. The updated version of this review now acknowledges recent advances in the understanding of metabolic, inflammatory and gender-specific influences on the right ventricle.
Collapse
|
168
|
Kao YH, Liou JP, Chung CC, Lien GS, Kuo CC, Chen SA, Chen YJ. Histone deacetylase inhibition improved cardiac functions with direct antifibrotic activity in heart failure. Int J Cardiol 2013; 168:4178-83. [PMID: 23931972 DOI: 10.1016/j.ijcard.2013.07.111] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 07/03/2013] [Accepted: 07/13/2013] [Indexed: 01/01/2023]
Abstract
BACKGROUND Histone deacetylases (HDACs), important epigenetic regulatory enzymes, can reduce cardiac hypertrophy and cardiac fibrosis. However, the mechanisms underlying the antifibrotic activity of HDAC inhibitors remain unclear. The purposes of this study were to evaluate the effects of an HDAC inhibitor on systolic heart failure (HF) and investigate the potential mechanisms. METHODS Echocardiographic, histologic, atrial natriuretic peptide (ANP), and Western blot measurements were performed in HF rats (isoproterenol 100 mg/kg, subcutaneous injection) with and without orally administered (100 mg/kg for 7 consecutive days) MPT0E014 (a novel HDAC inhibitor). Western blot, migration and proliferation assays were carried out on primary isolated cardiac fibroblasts with and without MPT0E014 (0.1 and 1 μM) for 24 h. RESULTS MPT0E014-treated HF rats (n = 6) had better fraction shortening (48 ± 2 vs. 33 ± 4%, p = 0.006) and smaller left ventricular end diastolic diameter (4.6 ± 0.2 vs. 5.6 ± 0.3 mm, p = 0.031) and systolic diameter (2.4 ± 0.2 vs. 3.9 ± 0.3 mm, p = 0.006) than HF (n = 7) rats. MPT0E014-treated HF rats had lower ANP, cardiac fibrosis, and angiotensin II type I receptor (AT1R), transforming growth factor (TGF)-β, and CaMKIIδ protein levels compared to HF rats. MPT0E014 (at 1 μM, but not 0.1 μM) decreased the migration and proliferation of cardiac fibroblasts. MPT0E014 (0.1 and 1 μM) decreased expression of the AT1R and TGF-β. CONCLUSIONS MPT0E014 improved cardiac contractility and attenuated structural remodeling in isoproterenol-induced dilated cardiomyopathy. The direct antifibrotic activity may have contributed to these beneficial effects.
Collapse
Affiliation(s)
- Yu-Hsun Kao
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
169
|
Romano SL, Lionetti V. From cell phenotype to epigenetic mechanisms: new insights into regenerating myocardium. Can J Physiol Pharmacol 2013; 91:579-85. [DOI: 10.1139/cjpp-2012-0392] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The self-regenerating property of the adult myocardium is not a new discovery. Even though we could not confirm that the adult myocardium is a post-mitotic tissue, we should consider that its plasticity is extremely low. Studies are still in progress to decipher the mechanisms underlying the abovementioned potential fetal features of the adult heart. The modest results of several clinical trials based on the transplantation of millions of autologous stem cells into the dysfunctional heart have confirmed that the cross-talk of different signals, such as the microenvironment, promotes the regeneration of adult myocardium. Recent scientific evidence has revealed that cellular cross-talk does not depend on the action of a single cell phenotype. It is conceivable that the limited turnover of cardiomyocytes is ensured by the interplay of adult cardiac cells in response to environmental changes. The epigenetic state of a cell serves as a dynamic interface between the environment and phenotype. The epigenetic modulation of the adult cardiac cells by natural active compounds encourages further studies to improve myocardial plasticity. In this review, we will highlight the most relevant studies demonstrating the epigenetic modulation of myocardial regeneration without the use of stem cell transplantation.
Collapse
Affiliation(s)
- Simone Lorenzo Romano
- Laboratory of Medical Science, Institute of Life Sciences, Via G. Moruzzi, 1, Scuola Superiore Sant’Anna, 56124 Pisa, Italy
- Cardiac and Thoracic Department, Azienda Ospedaliero – Universitaria Pisana, Pisa, Italy
| | - Vincenzo Lionetti
- Laboratory of Medical Science, Institute of Life Sciences, Via G. Moruzzi, 1, Scuola Superiore Sant’Anna, 56124 Pisa, Italy
- Fondazione CNR – Regione Toscana “G. Monasterio”, Pisa, Italy
- Institute of Clinical Physiology, CNR, Pisa, Italy
| |
Collapse
|
170
|
Lauffer BEL, Mintzer R, Fong R, Mukund S, Tam C, Zilberleyb I, Flicke B, Ritscher A, Fedorowicz G, Vallero R, Ortwine DF, Gunzner J, Modrusan Z, Neumann L, Koth CM, Lupardus PJ, Kaminker JS, Heise CE, Steiner P. Histone deacetylase (HDAC) inhibitor kinetic rate constants correlate with cellular histone acetylation but not transcription and cell viability. J Biol Chem 2013; 288:26926-43. [PMID: 23897821 DOI: 10.1074/jbc.m113.490706] [Citation(s) in RCA: 294] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Histone deacetylases (HDACs) are critical in the control of gene expression, and dysregulation of their activity has been implicated in a broad range of diseases, including cancer, cardiovascular, and neurological diseases. HDAC inhibitors (HDACi) employing different zinc chelating functionalities such as hydroxamic acids and benzamides have shown promising results in cancer therapy. Although it has also been suggested that HDACi with increased isozyme selectivity and potency may broaden their clinical utility and minimize side effects, the translation of this idea to the clinic remains to be investigated. Moreover, a detailed understanding of how HDACi with different pharmacological properties affect biological functions in vitro and in vivo is still missing. Here, we show that a panel of benzamide-containing HDACi are slow tight-binding inhibitors with long residence times unlike the hydroxamate-containing HDACi vorinostat and trichostatin-A. Characterization of changes in H2BK5 and H4K14 acetylation following HDACi treatment in the neuroblastoma cell line SH-SY5Y revealed that the timing and magnitude of histone acetylation mirrored both the association and dissociation kinetic rates of the inhibitors. In contrast, cell viability and microarray gene expression analysis indicated that cell death induction and changes in transcriptional regulation do not correlate with the dissociation kinetic rates of the HDACi. Therefore, our study suggests that determining how the selective and kinetic inhibition properties of HDACi affect cell function will help to evaluate their therapeutic utility.
Collapse
|
171
|
Spallotta F, Tardivo S, Nanni S, Rosati JD, Straino S, Mai A, Vecellio M, Valente S, Capogrossi MC, Farsetti A, Martone J, Bozzoni I, Pontecorvi A, Gaetano C, Colussi C. Detrimental effect of class-selective histone deacetylase inhibitors during tissue regeneration following hindlimb ischemia. J Biol Chem 2013; 288:22915-29. [PMID: 23836913 DOI: 10.1074/jbc.m113.484337] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Histone deacetylase inhibitors (DIs) are promising drugs for the treatment of several pathologies including ischemic and failing heart where they demonstrated efficacy. However, adverse side effects and cardiotoxicity have also been reported. Remarkably, no information is available about the effect of DIs during tissue regeneration following acute peripheral ischemia. In this study, mice made ischemic by femoral artery excision were injected with the DIs MS275 and MC1568, selective for class I and IIa histone deacetylases (HDACs), respectively. In untreated mice, soon after damage, class IIa HDAC phosphorylation and nuclear export occurred, paralleled by dystrophin and neuronal nitric-oxide synthase (nNOS) down-regulation and decreased protein phosphatase 2A activity. Between 14 and 21 days after ischemia, dystrophin and nNOS levels recovered, and class IIa HDACs relocalized to the nucleus. In this condition, the MC1568 compound increased the number of newly formed muscle fibers but delayed their terminal differentiation, whereas MS275 abolished the early onset of the regeneration process determining atrophy and fibrosis. The selective DIs had differential effects on the vascular compartment: MC1568 increased arteriogenesis whereas MS275 inhibited it. Capillarogenesis did not change. Chromatin immunoprecipitations revealed that class IIa HDAC complexes bind promoters of proliferation-associated genes and of class I HDAC1 and 2, highlighting a hierarchical control between class II and I HDACs during tissue regeneration. Our findings indicate that class-selective DIs interfere with normal mouse ischemic hindlimb regeneration and suggest that their use could be limited by alteration of the regeneration process in peripheral ischemic tissues.
Collapse
Affiliation(s)
- Francesco Spallotta
- Laboratorio di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino, 20138 Milano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
172
|
Zhao L, Chen CN, Hajji N, Oliver E, Cotroneo E, Wharton J, Wilkins MR, Wang D, Li M, Stenmark KR, McKinsey TA, Buttrick P. Response to letter regarding article, “histone deacetylation inhibition in pulmonary hypertension: therapeutic potential of valproic acid and suberoylanilide hydroxamic acid”. Circulation 2013; 127:e540. [PMID: 23691553 DOI: 10.1161/circulationaha.112.154757] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
173
|
Morrell NW, Archer SL, Defelice A, Evans S, Fiszman M, Martin T, Saulnier M, Rabinovitch M, Schermuly R, Stewart D, Truebel H, Walker G, Stenmark KR. Anticipated classes of new medications and molecular targets for pulmonary arterial hypertension. Pulm Circ 2013; 3:226-44. [PMID: 23662201 PMCID: PMC3641734 DOI: 10.4103/2045-8932.109940] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) remains a life-limiting condition with a major impact on the ability to lead a normal life. Although existing therapies may improve the outlook in some patients there remains a major unmet need to develop more effective therapies in this condition. There have been significant advances in our understanding of the genetic, cell and molecular basis of PAH over the last few years. This research has identified important new targets that could be explored as potential therapies for PAH. In this review we discuss whether further exploitation of vasoactive agents could bring additional benefits over existing approaches. Approaches to enhance smooth muscle cell apotosis and the potential of receptor tyrosine kinase inhibition are summarised. We evaluate the role of inflammation, epigenetic changes and altered glycolytic metabolism as potential targets for therapy, and whether inherited genetic mutations in PAH have revealed druggable targets. The potential of cell based therapies and gene therapy are also discussed. Potential candidate pathways that could be explored in the context of experimental medicine are identified.
Collapse
|
174
|
Xu Q, Lin X, Andrews L, Patel D, Lampe PD, Veenstra RD. Histone deacetylase inhibition reduces cardiac connexin43 expression and gap junction communication. Front Pharmacol 2013; 4:44. [PMID: 23596417 PMCID: PMC3625725 DOI: 10.3389/fphar.2013.00044] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 03/27/2013] [Indexed: 01/02/2023] Open
Abstract
Histone deacetylase inhibitors (HDACIs) are being investigated as novel therapies for cancer, inflammation, neurodegeneration, and heart failure. The effects of HDACIs on the functional expression of cardiac gap junctions (GJs) are essentially unknown. The purpose of this study was to determine the effects of trichostatin A (TSA) and vorinostat (VOR) on functional GJ expression in ventricular cardiomyocytes. The effects of HDAC inhibition on connexin43 (Cx43) expression and functional GJ assembly were examined in primary cultured neonatal mouse ventricular myocytes. TSA and VOR reduced Cx43 mRNA, protein expression, and immunolocalized Cx43 GJ plaque area within ventricular myocyte monolayer cultures in a dose-dependent manner. Chromatin immunoprecipitation experiments revealed altered protein interactions with the Cx43 promoter. VOR also altered the phosphorylation state of several key regulatory Cx43 phospho-serine sites. Patch clamp analysis revealed reduced electrical coupling between isolated ventricular myocyte pairs, altered transjunctional voltage-dependent inactivation kinetics, and steady state junctional conductance inactivation and recovery relationships. Single GJ channel conductance was reduced to 54 pS only by maximum inhibitory doses of TSA (≥ 100 nM). These two hydroxamate pan-HDACIs exert multiple levels of regulation on ventricular GJ communication by altering Cx43 expression, GJ area, post-translational modifications (e.g., phosphorylation, acetylation), gating, and channel conductance. Although a 50% downregulation of Cx43 GJ communication alone may not be sufficient to slow ventricular conduction or induce arrhythmias, the development of class-selective HDACIs may help avoid the potential negative cardiovascular effects of pan-HDACI.
Collapse
Affiliation(s)
- Qin Xu
- Department of Pharmacology, State University of New York Upstate Medical University Syracuse, NY, USA
| | | | | | | | | | | |
Collapse
|
175
|
Xie M, Hill JA. HDAC-dependent ventricular remodeling. Trends Cardiovasc Med 2013; 23:229-35. [PMID: 23499301 DOI: 10.1016/j.tcm.2012.12.006] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 12/17/2012] [Accepted: 12/18/2012] [Indexed: 01/07/2023]
Abstract
Heart failure, a syndrome culminating the pathogenesis of many forms of heart disease, is highly prevalent and projected to be increasingly so for years to come. Major efforts are directed at identifying the means of preventing, slowing, or possibly reversing the unremitting progression of pathological stress leading to myocardial injury and ultimately heart failure. Indeed, despite widespread use of evidence-based therapies, heart failure morbidity and mortality remain high. Recent work has uncovered a fundamental role of reversible protein acetylation in the regulation of many biological processes, including pathological remodeling of the heart. This reversible acetylation is governed by enzymes that attach (histone acetyltransferases, HATs) or remove (histone deacetylases, HDACs) acetyl groups. In the latter case, small molecule inhibitors of HDACs are currently being tested for a variety of oncological indications. Now, evidence has revealed that HDAC inhibitors blunt pathological cardiac remodeling in the settings of pressure overload and ischemia/reperfusion, thereby diminishing the emergence of heart failure. Mechanistically, HDAC inhibitors reduce stress-induced cardiomyocyte death, hypertrophy, and ventricular fibrosis. Looking to the future, HDAC inhibitor therapy may emerge as a novel means of arresting the untoward consequences of pathological cardiac stress, conferring clinical benefit to millions of patients with heart failure.
Collapse
Affiliation(s)
- Min Xie
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | | |
Collapse
|
176
|
Gopalan B, Ponpandian T, Kachhadia V, Bharathimohan K, Vignesh R, Sivasudar V, Narayanan S, Mandar B, Praveen R, Saranya N, Rajagopal S, Rajagopal S. Discovery of adamantane based highly potent HDAC inhibitors. Bioorg Med Chem Lett 2013; 23:2532-7. [PMID: 23538115 DOI: 10.1016/j.bmcl.2013.03.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 02/25/2013] [Accepted: 03/01/2013] [Indexed: 01/04/2023]
Abstract
Herein, we report the development of highly potent HDAC inhibitors for the treatment of cancer. A series of adamantane and nor-adamantane based HDAC inhibitors were designed, synthesized and screened for the inhibitory activity of HDAC. A number of compounds exhibited GI50 of 10-100 nM in human HCT116, NCI-H460 and U251 cancer cells, in vitro. Compound 32 displays efficacy in human tumour animal xenograft model.
Collapse
Affiliation(s)
- Balasubramanian Gopalan
- Drug Discovery Research Centre, Orchid Chemicals & Pharmaceuticals Ltd, Chennai 600119, India
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
177
|
Azakie A, Fineman J, He Y. Differential responses of the right ventricle to abnormal loading conditions in vivo: possible pathophysiologic mechanisms. J Thorac Cardiovasc Surg 2013; 145:1335-44. [PMID: 23434296 DOI: 10.1016/j.jtcvs.2013.01.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 12/13/2012] [Accepted: 01/11/2013] [Indexed: 11/17/2022]
Abstract
OBJECTIVE The right ventricle (RV) demonstrates differential adaptations in response to pressure versus volume loading, a phenomenon that may be important in the management of children and adults with congenital heart disease (CHD). The purpose of this study is to elucidate possible transcriptional mechanisms of the RV response to pressure versus volume loading in vivo. METHODS Fetal lambs had aortopulmonary shunting or pulmonary artery (PA) banding. Four weeks after spontaneous delivery, ovine hearts were evaluated for hemodynamic changes and changes in expression of sarcomeric gene proteins and transcriptional factors. Western blot densitometry and chromatin immunoprecipitation were applied using standard techniques. Transactivation assays were performed using transient transfections in Schneider's Drosophila line 2 cells in culture. RESULTS After PA banding, the RV pressure increased from 36 ± 4 mm Hg (n = 4) to 96 ± 8 mm Hg (n = 4, P < .05). The RVs of shunted and banded animals showed significant increases in the expression levels and promoter binding of activators myocyte enhancer factor 2, GATA-4, Nkx2.5, transcriptional enhancer factor 1, and specificity protein (Sp) 1. The transcriptional repressor Sp3 was downregulated in shunted animals, but its expression was increased paradoxically in the RV of the PA band group. Immunoprecipitation of Sp3 showed posttranslational modification to the acetylated isoform. In transient transfections of Schneider's Drosophila line 2 cells, acetylation of Sp3 converted it from a transcriptional repressor to an activator. CONCLUSIONS Posttranslational modifications of the transcriptional repressor Sp3, by acetylation, may be an important mechanism in the differential response of the RV to abnormal loading conditions. Sp3 may serve as a biomarker for RV failure for various heart defects in children and adults with CHD. These findings may have therapeutic implications in the management of right heart failure.
Collapse
MESH Headings
- Acetylation
- Animals
- Binding Sites
- Blotting, Western
- Cell Line
- Chromatin Immunoprecipitation
- Disease Models, Animal
- Female
- GATA4 Transcription Factor/genetics
- GATA4 Transcription Factor/metabolism
- Gene Expression Regulation
- Gestational Age
- Heart Ventricles/metabolism
- Heart Ventricles/physiopathology
- Hemodynamics
- Hypertrophy, Right Ventricular/genetics
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/physiopathology
- MEF2 Transcription Factors
- Myocardium/metabolism
- Myogenic Regulatory Factors/genetics
- Myogenic Regulatory Factors/metabolism
- Pregnancy
- Promoter Regions, Genetic
- Protein Processing, Post-Translational
- Pulmonary Artery/physiopathology
- Pulmonary Artery/surgery
- Sheep
- Sp1 Transcription Factor/genetics
- Sp1 Transcription Factor/metabolism
- Sp3 Transcription Factor/genetics
- Sp3 Transcription Factor/metabolism
- Time Factors
- Transcriptional Activation
- Transfection
- Ventricular Dysfunction, Right/genetics
- Ventricular Dysfunction, Right/metabolism
- Ventricular Dysfunction, Right/physiopathology
- Ventricular Function, Right/genetics
- Ventricular Pressure
Collapse
Affiliation(s)
- Anthony Azakie
- Department of Surgery, University of California at San Francisco, San Francisco, CA 94143, USA.
| | | | | |
Collapse
|
178
|
Hewitson R, Dargan J, Collis D, Green A, Moorjani N, Ohri S, Townsend PA. Heart failure: The pivotal role of histone deacetylases. Int J Biochem Cell Biol 2013. [DOI: 10.1016/j.biocel.2012.11.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
179
|
Bonney S, Hughes K, Harter PN, Mittelbronn M, Walker L, Eckle T. Cardiac period 2 in myocardial ischemia: clinical implications of a light dependent protein. Int J Biochem Cell Biol 2013; 45:667-71. [PMID: 23291353 DOI: 10.1016/j.biocel.2012.12.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 12/17/2012] [Accepted: 12/23/2012] [Indexed: 01/04/2023]
Abstract
Since the onset of myocardial infarction and stroke has distinct circadian patterns, the disruption of circadian rhythms may contribute to cardiovascular disease. A recent clinical study, reporting that the severity of myocardial ischemia depends on the time-of-day when ischemia occurs, highlights the impact of circadian rhythms on cardiovascular disease. In support of these observations, we found a cardioprotective role of the circadian rhythm protein Period 2 (Per2) during myocardial ischemia in mice. In these studies, exposing mice to daylight induced cardiac Per2, which was associated with protection from myocardial ischemia. Recent epidemiological studies found sunlight to be the dominant regulator of the human circadian rhythm, suggesting sunlight cycles are critical for maintaining a healthy cardiovascular system. However, the impact of circadian rhythm proteins on human disease remains unclear. This current review aims to make a link to current and future clinical practice by targeting cardiac Per2.
Collapse
Affiliation(s)
- Stephanie Bonney
- Department of Anesthesiology and Mucosal Inflammation Program, University of Colorado Denver, CO, USA
| | | | | | | | | | | |
Collapse
|
180
|
Menick DR, Li MS, Chernysh O, Renaud L, Kimbrough D, Kasiganesan H, Mani SK. Transcriptional pathways and potential therapeutic targets in the regulation of Ncx1 expression in cardiac hypertrophy and failure. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 961:125-35. [PMID: 23224875 PMCID: PMC3624972 DOI: 10.1007/978-1-4614-4756-6_11] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Changes in cardiac gene expression contribute to the progression of heart failure by affecting cardiomyocyte growth, function, and survival. The Na(+)-Ca(2+) exchanger gene (Ncx1) is upregulated in hypertrophy and is often found elevated in end-stage heart failure. Studies have shown that the change in its expression contributes to contractile dysfunction. Several transcriptional pathways mediate Ncx1 expression in pathological cardiac remodeling. Both α-adrenergic receptor (α-AR) and β-adrenergic receptor (β-AR) signaling can play a role in the regulation of calcium homeostasis in the cardiomyocyte, but chronic activation in periods of cardiac stress contributes to heart failure by mechanisms which include Ncx1 upregulation. Our studies have even demonstrated that NCX1 can directly act as a regulator of "activity-dependent signal transduction" mediating changes in its own expression. Finally, we present evidence that histone deacetylases (HDACs) and histone acetyltransferases (HATs) act as master regulators of Ncx1 expression. We show that many of the transcription factors regulating Ncx1 expression are important in cardiac development and also in the regulation of many other genes in the so-called fetal gene program, which are activated by pathological stimuli. Importantly, studies have revealed that the transcriptional network regulating Ncx1 expression is also mediating many of the other changes in genetic remodeling contributing to the development of cardiac dysfunction and revealed potential therapeutic targets for the treatment of hypertrophy and failure.
Collapse
|
181
|
Majumdar G, Adris P, Bhargava N, Chen H, Raghow R. Pan-histone deacetylase inhibitors regulate signaling pathways involved in proliferative and pro-inflammatory mechanisms in H9c2 cells. BMC Genomics 2012; 13:709. [PMID: 23249388 PMCID: PMC3561284 DOI: 10.1186/1471-2164-13-709] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 12/12/2012] [Indexed: 01/19/2023] Open
Abstract
Background We have shown previously that pan-HDAC inhibitors (HDACIs) m-carboxycinnamic acid bis-hydroxamide (CBHA) and trichostatin A (TSA) attenuated cardiac hypertrophy in BALB/c mice by inducing hyper-acetylation of cardiac chromatin that was accompanied by suppression of pro-inflammatory gene networks. However, it was not feasible to determine the precise contribution of the myocytes- and non-myocytes to HDACI-induced gene expression in the intact heart. Therefore, the current study was undertaken with a primary goal of elucidating temporal changes in the transcriptomes of cardiac myocytes exposed to CBHA and TSA. Results We incubated H9c2 cardiac myocytes in growth medium containing either of the two HDACIs for 6h and 24h and analyzed changes in gene expression using Illumina microarrays. H9c2 cells exposed to TSA for 6h and 24h led to differential expression of 468 and 231 genes, respectively. In contrast, cardiac myocytes incubated with CBHA for 6h and 24h elicited differential expression of 768 and 999 genes, respectively. We analyzed CBHA- and TSA-induced differentially expressed genes by Ingenuity Pathway (IPA), Kyoto Encyclopedia of Genes and Genomes (KEGG) and Core_TF programs and discovered that CBHA and TSA impinged on several common gene networks. Thus, both HDACIs induced a repertoire of signaling kinases (PTEN-PI3K-AKT and MAPK) and transcription factors (Myc, p53, NFkB and HNF4A) representing canonical TGFβ, TNF-α, IFNγ and IL-6 specific networks. An overrepresentation of E2F, AP2, EGR1 and SP1 specific motifs was also found in the promoters of the differentially expressed genes. Apparently, TSA elicited predominantly TGFβ- and TNF-α-intensive gene networks regardless of the duration of treatment. In contrast, CBHA elicited TNF-α and IFNγ specific networks at 6 h, followed by elicitation of IL-6 and IFNγ-centered gene networks at 24h. Conclusions Our data show that both CBHA and TSA induced similar, but not identical, time-dependent, gene networks in H9c2 cardiac myocytes. Initially, both HDACIs impinged on numerous genes associated with adipokine signaling, intracellular metabolism and energetics, and cell cycle. A continued exposure to either CBHA or TSA led to the emergence of a number of apoptosis- and inflammation-specific gene networks that were apparently suppressed by both HDACIs. Based on these data we posit that the anti-inflammatory and anti-proliferative actions of HDACIs are myocyte-intrinsic. These findings advance our understanding of the mechanisms of actions of HDACIs on cardiac myocytes and reveal potential signaling pathways that may be targeted therapeutically.
Collapse
Affiliation(s)
- Gipsy Majumdar
- Department of Veterans Affairs Medical Center, 1030 Jefferson Avenue, Memphis, TN 38104, USA
| | | | | | | | | |
Collapse
|
182
|
Shi H, Chen L, Wang H, Zhu S, Dong C, Webster KA, Wei J. Synergistic induction of miR-126 by hypoxia and HDAC inhibitors in cardiac myocytes. Biochem Biophys Res Commun 2012. [PMID: 23201405 DOI: 10.1016/j.bbrc.2012.11.061] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
HDAC inhibitors are under clinical development for the treatment of hypertrophic cardiomyopathy and heart failure although the mechanisms of protection are incompletely understood. Micro-RNA 126, an endothelium-specific miR has been assigned essential developmental roles in the heart by activating survival kinases ERK1/2 and Akt and increasing pro-angiogenic signaling. Here we provide the first evidence that hypoxia and HDAC inhibitors selectively and synergistically stimulate expression of miR-126 in cardiac myocytes. MiR-126 expression was increased 1.7-fold (p<0.05) after 1h of hypoxic exposure and this was further enhanced to 3.0-fold (p<0.01) by simultaneously blocking HDAC with the pan-HDAC inhibitor Tricostatin A (TSA). TSA alone did not increase miR-126. In parallel, hypoxia and TSA synergistically increased p-ERK and p-Akt without effecting VEGF-A level. Knockdown of miR-126 with si-RNA eliminated inductions of p-ERK and p-Akt by hypoxia, whereas miR-126 overexpression mimicked hypoxia and amplified p-ERK and p-Akt in parallel with miR-126. The results suggest that miR-126 is a hypoxia-inducible target of HAT/HDAC and its activation in cardiac myocytes may contribute to cardioprotection by activating cell survival and pro-angiogenic pathways selectively during ischemia.
Collapse
Affiliation(s)
- Huaping Shi
- Hangzhou Red Cross Hospital, Zhejiang, China
| | | | | | | | | | | | | |
Collapse
|
183
|
Chang CWJ, Lee L, Yu D, Dao K, Bossuyt J, Bers DM. Acute β-adrenergic activation triggers nuclear import of histone deacetylase 5 and delays G(q)-induced transcriptional activation. J Biol Chem 2012; 288:192-204. [PMID: 23161540 DOI: 10.1074/jbc.m112.382358] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
During hemodynamic stress, catecholamines and neurohumoral stimuli may induce co-activation of G(q)-coupled receptors and β-adrenergic receptors (β-AR), leading to cardiac remodeling. Dynamic regulation of histone deacetylase 5 (HDAC5), a transcriptional repressor, is crucial during stress signaling due to its role in epigenetic control of fetal gene markers. Little is known about its regulation during acute and chronic β-AR stimulation and its cross-interaction with G(q) signaling in adult cardiac myocytes. Here, we evaluate the potential cross-talk between G(q)-driven and β-AR mediated signaling at the level of nucleocytoplasmic shuttling of HDAC5. We show the translocation of GFP-tagged wild type HDAC5 or mutants (S279A and S279D) in response to β-AR or G(q) agonists. Isoproterenol (ISO) or PKA activation results in strong nuclear accumulation of HDAC5 in contrast to nuclear export driven by Ca(2+)-calmodulin protein kinase II and protein kinase D. Moreover, nuclear accumulation of HDAC5 under acute ISO/PKA signaling is dependent on phosphorylation of Ser-279 and can block subsequent G(q)-mediated nuclear HDAC5 export. Intriguingly, the attenuation of G(q)-induced export is abolished after chronic PKA activation, yet nuclear HDAC5 remains elevated. Last, the effect of chronic β-AR signaling on HDAC5 translocation was examined in adult myocytes from a rabbit model of heart failure, where ISO-induced nuclear import is ablated, but G(q)-agonist mediated export is preserved. Acute β-AR/PKA activation protects against hypertrophic signaling by delaying G(q)-mediated transcriptional activation. This serves as a key physiological control switch before allowing genetic reprogramming via HDAC5 nuclear export during more severe stress, such as heart failure.
Collapse
Affiliation(s)
- Chia-Wei Jenny Chang
- Department of Pharmacology, University of California, Davis, California 95616-8636, USA
| | | | | | | | | | | |
Collapse
|
184
|
Ferrari A, Fiorino E, Giudici M, Gilardi F, Galmozzi A, Mitro N, Cermenati G, Godio C, Caruso D, De Fabiani E, Crestani M. Linking epigenetics to lipid metabolism: focus on histone deacetylases. Mol Membr Biol 2012; 29:257-66. [PMID: 23095054 DOI: 10.3109/09687688.2012.729094] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A number of recent studies revealed that epigenetic modifications play a central role in the regulation of lipid and of other metabolic pathways such as cholesterol homeostasis, bile acid synthesis, glucose and energy metabolism. Epigenetics refers to aspects of genome functions regulated in a DNA sequence-independent fashion. Chromatin structure is controlled by epigenetic mechanisms through DNA methylation and histone modifications. The main modifications are histone acetylation and deacetylation on specific lysine residues operated by two different classes of enzymes: Histone acetyltransferases (HATs) and histone deacetylases (HDACs), respectively. The interaction between these enzymes and histones can activate or repress gene transcription: Histone acetylation opens and activates chromatin, while deacetylation of histones and DNA methylation compact chromatin making it transcriptionally silent. The new evidences on the importance of HDACs in the regulation of lipid and other metabolic pathways will open new perspectives in the comprehension of the pathophysiology of metabolic disorders.
Collapse
Affiliation(s)
- Alessandra Ferrari
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Abstract
Fibrosis is a pathological feature of most chronic inflammatory diseases. Fibrosis, or scarring, is defined by the accumulation of excess extracellular matrix components. If highly progressive, the fibrotic process eventually leads to organ malfunction and death. Fibrosis affects nearly every tissue in the body. Here we discuss how key components of the innate and adaptive immune response contribute to the pathogenesis of fibrosis. We also describe how cell-intrinsic changes in important structural cells can perpetuate the fibrotic response by regulating the differentiation, recruitment, proliferation and activation of extracellular matrix-producing myofibroblasts. Finally, we highlight some of the key mechanisms and pathways of fibrosis that are being targeted as potential therapies for a variety of important human diseases.
Collapse
|
186
|
Abstract
Fibrosis is a pathological feature of most chronic inflammatory diseases. Fibrosis, or scarring, is defined by the accumulation of excess extracellular matrix components. If highly progressive, the fibrotic process eventually leads to organ malfunction and death. Fibrosis affects nearly every tissue in the body. Here we discuss how key components of the innate and adaptive immune response contribute to the pathogenesis of fibrosis. We also describe how cell-intrinsic changes in important structural cells can perpetuate the fibrotic response by regulating the differentiation, recruitment, proliferation and activation of extracellular matrix-producing myofibroblasts. Finally, we highlight some of the key mechanisms and pathways of fibrosis that are being targeted as potential therapies for a variety of important human diseases.
Collapse
Affiliation(s)
- Thomas A Wynn
- Immunopathogenesis Section, Program in Barrier Immunity and Repair, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| | | |
Collapse
|
187
|
Cavasin MA, Demos-Davies K, Horn TR, Walker LA, Lemon DD, Birdsey N, Weiser-Evans MCM, Harral J, Irwin DC, Anwar A, Yeager ME, Li M, Watson PA, Nemenoff RA, Buttrick PM, Stenmark KR, McKinsey TA. Selective class I histone deacetylase inhibition suppresses hypoxia-induced cardiopulmonary remodeling through an antiproliferative mechanism. Circ Res 2012; 110:739-48. [PMID: 22282194 DOI: 10.1161/circresaha.111.258426] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
RATIONALE Histone deacetylase (HDAC) inhibitors are efficacious in models of hypertension-induced left ventricular heart failure. The consequences of HDAC inhibition in the context of pulmonary hypertension with associated right ventricular cardiac remodeling are poorly understood. OBJECTIVE This study was performed to assess the utility of selective small-molecule inhibitors of class I HDACs in a preclinical model of pulmonary hypertension. METHODS AND RESULTS Rats were exposed to hypobaric hypoxia for 3 weeks in the absence or presence of a benzamide HDAC inhibitor, MGCD0103, which selectively inhibits class I HDACs 1, 2, and 3. The compound reduced pulmonary arterial pressure more dramatically than tadalafil, a standard-of-care therapy for human pulmonary hypertension that functions as a vasodilator. MGCD0103 improved pulmonary artery acceleration time and reduced systolic notching of the pulmonary artery flow envelope, which suggests a positive impact of the HDAC inhibitor on pulmonary vascular remodeling and stiffening. Similar results were obtained with an independent class I HDAC-selective inhibitor, MS-275. Reduced pulmonary arterial pressure in MGCD0103-treated animals was associated with blunted pulmonary arterial wall thickening because of suppression of smooth muscle cell proliferation. Right ventricular function was maintained in MGCD0103-treated animals. Although the class I HDAC inhibitor only modestly reduced right ventricular hypertrophy, it had multiple beneficial effects on the right ventricle, which included suppression of pathological gene expression, inhibition of proapoptotic caspase activity, and repression of proinflammatory protein expression. CONCLUSIONS By targeting distinct pathogenic mechanisms, isoform-selective HDAC inhibitors have potential as novel therapeutics for pulmonary hypertension that will complement vasodilator standards of care.
Collapse
Affiliation(s)
- Maria A Cavasin
- Department of Medicine, Division of Cardiology, University of Colorado Denver, Aurora, CO, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
188
|
Bernardo BC, Ooi JY, McMullen JR. The yin and yang of adaptive and maladaptive processes in heart failure. ACTA ACUST UNITED AC 2012. [DOI: 10.1016/j.ddstr.2013.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|