1
|
Forbes LM, Bauer N, Bhadra A, Bogaard HJ, Choudhary G, Goss KN, Gräf S, Heresi GA, Hopper RK, Jose A, Kim Y, Klouda T, Lahm T, Lawrie A, Leary PJ, Leopold JA, Oliveira SD, Prisco SZ, Rafikov R, Rhodes CJ, Stewart DJ, Vanderpool RR, Yuan K, Zimmer A, Hemnes AR, de Jesus Perez VA, Wilkins MR. Precision Medicine for Pulmonary Vascular Disease: The Future Is Now (2023 Grover Conference Series). Pulm Circ 2025; 15:e70027. [PMID: 39749110 PMCID: PMC11693987 DOI: 10.1002/pul2.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025] Open
Abstract
Pulmonary vascular disease is not a single condition; rather it can accompany a variety of pathologies that impact the pulmonary vasculature. Applying precision medicine strategies to better phenotype, diagnose, monitor, and treat pulmonary vascular disease is increasingly possible with the growing accessibility of powerful clinical and research tools. Nevertheless, challenges exist in implementing these tools to optimal effect. The 2023 Grover Conference Series reviewed the research landscape to summarize the current state of the art and provide a better understanding of the application of precision medicine to managing pulmonary vascular disease. In particular, the following aspects were discussed: (1) Clinical phenotypes, (2) genetics, (3) epigenetics, (4) biomarker discovery, (5) application of precision biology to clinical trials, (6) the right ventricle (RV), and (7) integrating precision medicine to clinical care. The present review summarizes the content of these discussions and the prospects for the future.
Collapse
Affiliation(s)
- Lindsay M. Forbes
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of ColoradoAuroraColoradoUSA
| | - Natalie Bauer
- Department of PharmacologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileAlabamaUSA
| | - Aritra Bhadra
- Department of PharmacologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
- Center for Lung BiologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
| | - Harm J. Bogaard
- Department of Pulmonary MedicineAmsterdam UMCAmsterdamNetherlands
| | - Gaurav Choudhary
- Division of CardiologyWarren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
- Lifespan Cardiovascular InstituteRhode Island and Miriam HospitalsProvidenceRhode IslandUSA
- Department of CardiologyProvidence VA Medical CenterProvidenceRhode IslandUSA
| | - Kara N. Goss
- Department of Medicine and PediatricsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Stefan Gräf
- Division of Computational Genomics and Genomic Medicine, Department of MedicineUniversity of Cambridge, Victor Phillip Dahdaleh Heart & Lung Research InstituteCambridgeUK
| | | | - Rachel K. Hopper
- Department of PediatricsStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Arun Jose
- Division of Pulmonary, Critical Care, and Sleep MedicineUniversity of CincinnatiCincinnatiOhioUSA
| | - Yunhye Kim
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Timothy Klouda
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Tim Lahm
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of ColoradoAuroraColoradoUSA
- Division of Pulmonary, Critical Care, and Sleep MedicineNational Jewish HealthDenverColoradoUSA
- Pulmonary and Critical Care SectionRocky Mountain Regional VA Medical CenterDenverColoradoUSA
| | - Allan Lawrie
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Peter J. Leary
- Departments of Medicine and EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Jane A. Leopold
- Division of Cardiovascular MedicineBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Suellen D. Oliveira
- Department of Anesthesiology, Department of Physiology and BiophysicsUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Sasha Z. Prisco
- Division of CardiovascularLillehei Heart Institute, University of MinnesotaMinneapolisMinnesotaUSA
| | - Ruslan Rafikov
- Department of MedicineIndiana UniversityIndianapolisIndianaUSA
| | | | - Duncan J. Stewart
- Ottawa Hospital Research InstituteFaculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | | | - Ke Yuan
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Alexsandra Zimmer
- Department of MedicineBrown UniversityProvidenceRhode IslandUSA
- Lifespan Cardiovascular InstituteRhode Island HospitalProvidenceRhode IslandUSA
| | - Anna R. Hemnes
- Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Vinicio A. de Jesus Perez
- Division of Pulmonary and Critical Care MedicineStanford University Medical CenterStanfordCaliforniaUSA
| | | |
Collapse
|
2
|
Ejikeme C, Safdar Z. Exploring the pathogenesis of pulmonary vascular disease. Front Med (Lausanne) 2024; 11:1402639. [PMID: 39050536 PMCID: PMC11267418 DOI: 10.3389/fmed.2024.1402639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Pulmonary hypertension (PH) is a complex cardiopulmonary disorder impacting the lung vasculature, resulting in increased pulmonary vascular resistance that leads to right ventricular dysfunction. Pulmonary hypertension comprises of 5 groups (PH group 1 to 5) where group 1 pulmonary arterial hypertension (PAH), results from alterations that directly affect the pulmonary arteries. Although PAH has a complex pathophysiology that is not completely understood, it is known to be a multifactorial disease that results from a combination of genetic, epigenetic and environmental factors, leading to a varied range of symptoms in PAH patients. PAH does not have a cure, its incidence and prevalence continue to increase every year, resulting in higher morbidity and mortality rates. In this review, we discuss the different pathologic mechanisms with a focus on epigenetic modifications and their roles in the development and progression of PAH. These modifications include DNA methylation, histone modifications, and microRNA dysregulation. Understanding these epigenetic modifications will improve our understanding of PAH and unveil novel therapeutic targets, thus steering research toward innovative treatment strategies.
Collapse
Affiliation(s)
| | - Zeenat Safdar
- Department of Pulmonary-Critical Care Medicine, Houston Methodist Lung Center, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
3
|
Dignam JP, Sharma S, Stasinopoulos I, MacLean MR. Pulmonary arterial hypertension: Sex matters. Br J Pharmacol 2024; 181:938-966. [PMID: 37939796 DOI: 10.1111/bph.16277] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a complex disease of multifactorial origin. While registries have demonstrated that women are more susceptible to the disease, females with PAH have superior right ventricle (RV) function and a better prognosis than their male counterparts, a phenomenon referred to as the 'estrogen paradox'. Numerous pre-clinical studies have investigated the involvement of sex hormones in PAH pathobiology, often with conflicting results. However, recent advances suggest that abnormal estrogen synthesis, metabolism and signalling underpin the sexual dimorphism of this disease. Other sex hormones, such as progesterone, testosterone and dehydroepiandrosterone may also play a role. Several non-hormonal factor including sex chromosomes and epigenetics have also been implicated. Though the underlying pathophysiological mechanisms are complex, several compounds that modulate sex hormones levels and signalling are under investigation in PAH patients. Further elucidation of the estrogen paradox will set the stage for the identification of additional therapeutic targets for this disease.
Collapse
Affiliation(s)
- Joshua P Dignam
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Smriti Sharma
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Ioannis Stasinopoulos
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, UK
| | - Margaret R MacLean
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| |
Collapse
|
4
|
Novara ME, Di Martino E, Stephens B, Nayrouz M, Vitulo P, Carollo A, Provenzani A. Future Perspectives of Pulmonary Arterial Hypertension: A Review of Novel Pipeline Treatments and Indications. Drugs R D 2024; 24:13-28. [PMID: 38514585 PMCID: PMC11035521 DOI: 10.1007/s40268-024-00453-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2024] [Indexed: 03/23/2024] Open
Abstract
Pulmonary arterial hypertension is characterized by elevated blood pressure and pathological changes in the pulmonary arterioles, leading to the development of right-heart failure and potentially fatal outcomes if left untreated. This review aims to provide an overview of novel drugs or formulations and new drug indications for pulmonary arterial hypertension that are currently in phases II-III of randomized controlled trials, and describe the rationale for the use of these targeted therapies, as well as their efficacy, safety profile, and impact on quality of life and survival. The literature research was conducted using data from ClinicalTrials.gov for the period between 1 January 2016 up to 31 December 2022. The population of interest includes individuals aged ≥ 18 years who have been diagnosed with pulmonary arterial hypertension. The review selection criteria included trials with recruiting, enrolling by invitation, active, terminated or completed status in 2022 and 2023. A total of 24 studies were selected for evaluation based on the inclusion and exclusion criteria. This review summarizes the updated information from randomized clinical trials involving novel therapies for pulmonary arterial hypertension. However, larger clinical trials are required to validate their clinical safety and effects. In the future, clinicians should choose therapies based on the patient's individual situation and requirements when developing treatment strategies.
Collapse
Affiliation(s)
- Maria Eugenia Novara
- Clinical Pharmacy Service, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Palermo, Italy
| | - Enrica Di Martino
- Clinical Pharmacy Service, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Palermo, Italy
| | - Brandon Stephens
- School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Mary Nayrouz
- School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Patrizio Vitulo
- Pneumology Unit, Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic Transplantation, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Palermo, Italy
| | - Anna Carollo
- Clinical Pharmacy Service, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Palermo, Italy
| | - Alessio Provenzani
- Clinical Pharmacy Service, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Palermo, Italy.
| |
Collapse
|
5
|
Benza RL, Adamson PB, Bhatt DL, Frick F, Olsson G, Bergh N, Dahlöf B. CS1, a controlled-release formulation of valproic acid, for the treatment of patients with pulmonary arterial hypertension: Rationale and design of a Phase 2 clinical trial. Pulm Circ 2024; 14:e12323. [PMID: 38174159 PMCID: PMC10763516 DOI: 10.1002/pul2.12323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/13/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
Although rare, pulmonary arterial hypertension (PAH) is associated with substantial morbidity and a median survival of approximately 7 years, even with treatment. Current medical therapies have a primarily vasodilatory effect and do not modify the underlying pathology of the disease. CS1 is a novel oral, controlled-release formulation of valproic acid, which exhibits a multi-targeted mode of action (pulmonary pressure reduction, reversal of vascular remodeling, anti-inflammatory, anti-fibrotic, and anti-thrombotic) and therefore potential for disease modification and right ventricular modeling in patients with PAH. A Phase 1 study conducted in healthy volunteers indicated favorable safety and tolerability, with no increased risk of bleeding and significant reduction of plasminogen activator inhibitor 1. In an ongoing randomized Phase 2 clinical trial, three doses of open-label CS1 administered for 12 weeks is evaluating the use of multiple outcome measures. The primary endpoint is safety and tolerability, as measured by the occurrence of adverse events. Secondary outcome measures include the use of the CardioMEMS™ HF System, which provides a noninvasive method of monitoring pulmonary artery pressure, as well as cardiac magnetic resonance imaging and echocardiography. Other outcomes include changes in risk stratification (using the REVEAL 2.0 and REVEAL Lite 2 tools), patient reported outcomes, functional capacity, 6-min walk distance, actigraphy, and biomarkers. The pharmacokinetic profile of CS1 will also be evaluated. Overall, the novel design and unique, extensive clinical phenotyping of participants in this trial will provide ample evidence to inform the design of any future Phase 3 studies with CS1.
Collapse
Affiliation(s)
- Raymond L. Benza
- Ohio State Wexner Medical CenterThe Ohio State UniversityColumbusOhioUSA
| | | | - Deepak L. Bhatt
- Mount Sinai HeartIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | | | - Gunnar Olsson
- Institute of MedicineUniversity of GothenburgGothenburgSweden
| | - Niklas Bergh
- Institute of MedicineUniversity of GothenburgGothenburgSweden
- Early Clinical Development, Biopharmaceuticals Research and Development—CardiovascularRenal and Metabolism, AstraZenecaMölndalSweden
| | - Björn Dahlöf
- Cereno ScientificGothenburgSweden
- Institute of MedicineUniversity of GothenburgGothenburgSweden
| |
Collapse
|
6
|
Toro V, Jutras-Beaudoin N, Boucherat O, Bonnet S, Provencher S, Potus F. Right Ventricle and Epigenetics: A Systematic Review. Cells 2023; 12:2693. [PMID: 38067121 PMCID: PMC10705252 DOI: 10.3390/cells12232693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/08/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
There is an increasing recognition of the crucial role of the right ventricle (RV) in determining the functional status and prognosis in multiple conditions. In the past decade, the epigenetic regulation (DNA methylation, histone modification, and non-coding RNAs) of gene expression has been raised as a critical determinant of RV development, RV physiological function, and RV pathological dysfunction. We thus aimed to perform an up-to-date review of the literature, gathering knowledge on the epigenetic modifications associated with RV function/dysfunction. Therefore, we conducted a systematic review of studies assessing the contribution of epigenetic modifications to RV development and/or the progression of RV dysfunction regardless of the causal pathology. English literature published on PubMed, between the inception of the study and 1 January 2023, was evaluated. Two authors independently evaluated whether studies met eligibility criteria before study results were extracted. Amongst the 817 studies screened, 109 studies were included in this review, including 69 that used human samples (e.g., RV myocardium, blood). While 37 proposed an epigenetic-based therapeutic intervention to improve RV function, none involved a clinical trial and 70 are descriptive. Surprisingly, we observed a substantial discrepancy between studies investigating the expression (up or down) and/or the contribution of the same epigenetic modifications on RV function or development. This exhaustive review of the literature summarizes the relevant epigenetic studies focusing on RV in human or preclinical setting.
Collapse
Affiliation(s)
| | | | | | | | | | - François Potus
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, QC G1V 4G5, Canada; (V.T.); (N.J.-B.); (O.B.); (S.B.); (S.P.)
| |
Collapse
|
7
|
Balsa A, Adão R, Brás-Silva C. Therapeutic Approaches in Pulmonary Arterial Hypertension with Beneficial Effects on Right Ventricular Function-Preclinical Studies. Int J Mol Sci 2023; 24:15539. [PMID: 37958522 PMCID: PMC10647677 DOI: 10.3390/ijms242115539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 11/15/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive condition that affects the pulmonary vessels, but its main prognostic factor is the right ventricle (RV) function. Many mice/rat models are used for research in PAH, but results fail to translate to clinical trials. This study reviews studies that test interventions on pulmonary artery banding (PAB), a model of isolated RV disfunction, and PH models. Multiple tested drugs both improved pulmonary vascular hemodynamics in PH models and improved RV structure and function in PAB animals. PH models and PAB animals frequently exhibited similar results (73.1% concordance). Macitentan, sildenafil, and tadalafil improved most tested pathophysiological parameters in PH models, but almost none in PAB animals. Results are frequently not consistent with other studies, possibly due to the methodology, which greatly varied. Some research groups start treating the animals immediately, and others wait up to 4 weeks from model induction. Treatment duration and choice of anaesthetic are other important differences. This review shows that many drugs currently under research for PAH have a cardioprotective effect on animals that may translate to humans. However, a uniformization of methods may increase comparability between studies and, thus, improve translation to clinical trials.
Collapse
Affiliation(s)
- André Balsa
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (A.B.); (R.A.)
| | - Rui Adão
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (A.B.); (R.A.)
- Department of Pharmacology and Toxicology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- CIBER of Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| | - Carmen Brás-Silva
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (A.B.); (R.A.)
- Faculty of Nutrition and Food Sciences, University of Porto, 4150-180 Porto, Portugal
| |
Collapse
|
8
|
Dave J, Jagana V, Janostiak R, Bisserier M. Unraveling the epigenetic landscape of pulmonary arterial hypertension: implications for personalized medicine development. J Transl Med 2023; 21:477. [PMID: 37461108 DOI: 10.1186/s12967-023-04339-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/10/2023] [Indexed: 07/20/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a multifactorial disease associated with the remodeling of pulmonary blood vessels. If left unaddressed, PAH can lead to right heart failure and even death. Multiple biological processes, such as smooth muscle proliferation, endothelial dysfunction, inflammation, and resistance to apoptosis, are associated with PAH. Increasing evidence suggests that epigenetic factors play an important role in PAH by regulating the chromatin structure and altering the expression of critical genes. For example, aberrant DNA methylation and histone modifications such as histone acetylation and methylation have been observed in patients with PAH and are linked to vascular remodeling and pulmonary vascular dysfunction. In this review article, we provide a comprehensive overview of the role of key epigenetic targets in PAH pathogenesis, including DNA methyltransferase (DNMT), ten-eleven translocation enzymes (TET), switch-independent 3A (SIN3A), enhancer of zeste homolog 2 (EZH2), histone deacetylase (HDAC), and bromodomain-containing protein 4 (BRD4). Finally, we discuss the potential of multi-omics integration to better understand the molecular signature and profile of PAH patients and how this approach can help identify personalized treatment approaches.
Collapse
Affiliation(s)
- Jaydev Dave
- Department of Cell Biology and Anatomy, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA
- Department of Physiology, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA
| | - Vineeta Jagana
- Department of Cell Biology and Anatomy, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA
- Department of Physiology, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA
| | - Radoslav Janostiak
- First Faculty of Medicine, BIOCEV, Charles University, Vestec, 25250, Czech Republic
| | - Malik Bisserier
- Department of Cell Biology and Anatomy, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA.
- Department of Physiology, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA.
| |
Collapse
|
9
|
Shanmukha KD, Paluvai H, Lomada SK, Gokara M, Kalangi SK. Histone deacetylase (HDACs) inhibitors: Clinical applications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 198:119-152. [DOI: 10.1016/bs.pmbts.2023.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
10
|
Ho L, Hossen N, Nguyen T, Vo A, Ahsan F. Epigenetic Mechanisms as Emerging Therapeutic Targets and Microfluidic Chips Application in Pulmonary Arterial Hypertension. Biomedicines 2022; 10:170. [PMID: 35052850 PMCID: PMC8773438 DOI: 10.3390/biomedicines10010170] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 12/15/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a disease that progress over time and is defined as an increase in pulmonary arterial pressure and pulmonary vascular resistance that frequently leads to right-ventricular (RV) failure and death. Epigenetic modifications comprising DNA methylation, histone remodeling, and noncoding RNAs (ncRNAs) have been established to govern chromatin structure and transcriptional responses in various cell types during disease development. However, dysregulation of these epigenetic mechanisms has not yet been explored in detail in the pathology of pulmonary arterial hypertension and its progression with vascular remodeling and right-heart failure (RHF). Targeting epigenetic regulators including histone methylation, acetylation, or miRNAs offers many possible candidates for drug discovery and will no doubt be a tempting area to explore for PAH therapies. This review focuses on studies in epigenetic mechanisms including the writers, the readers, and the erasers of epigenetic marks and targeting epigenetic regulators or modifiers for treatment of PAH and its complications described as RHF. Data analyses from experimental cell models and animal induced PAH models have demonstrated that significant changes in the expression levels of multiple epigenetics modifiers such as HDMs, HDACs, sirtuins (Sirt1 and Sirt3), and BRD4 correlate strongly with proliferation, apoptosis, inflammation, and fibrosis linked to the pathological vascular remodeling during PAH development. The reversible characteristics of protein methylation and acetylation can be applied for exploring small-molecule modulators such as valproic acid (HDAC inhibitor) or resveratrol (Sirt1 activator) in different preclinical models for treatment of diseases including PAH and RHF. This review also presents to the readers the application of microfluidic devices to study sex differences in PAH pathophysiology, as well as for epigenetic analysis.
Collapse
Affiliation(s)
- Linh Ho
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA
| | - Nazir Hossen
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA
| | - Trieu Nguyen
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA
- East Bay Institute for Research & Education (EBIRE), Mather, CA 95655, USA
| | - Au Vo
- Department of Life Sciences, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Fakhrul Ahsan
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA
| |
Collapse
|
11
|
Applegate TJ, Krafsur GM, Boon JA, Zhang H, Li M, Holt TN, Ambler SK, Abrams BA, Gustafson DL, Bartels K, Garry FB, Stenmark KR, Brown RD. Brief Report: Case Comparison of Therapy With the Histone Deacetylase Inhibitor Vorinostat in a Neonatal Calf Model of Pulmonary Hypertension. Front Physiol 2021; 12:712583. [PMID: 34552503 PMCID: PMC8450341 DOI: 10.3389/fphys.2021.712583] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/16/2021] [Indexed: 11/13/2022] Open
Abstract
Pulmonary hypertension (PH) is an incurable condition in humans; driven by pulmonary vascular remodeling partially mediated by epigenetic mechanisms; and leading to right ventricular hypertrophy, failure, and death. We hypothesized that targeting chromatin-modifying histone deacetylases may provide benefit. In this Brief Report we describe case comparison studies using the histone deacetylase inhibitor vorinostat (suberanilohydroxamic acid, 5 mg/kg/day for the first 5 study days) in an established model of severe neonatal bovine PH induced by 14 days of environmental hypoxia. Echocardiographic, hemodynamic, and pharmacokinetic data were obtained in hypoxia-exposed (one each, vorinostat-treated vs. untreated) and normoxic vorinostat-treated control animals (n = 2). Echocardiography detected PH changes by day 4 and severe PH over 14 days of continued hypoxic exposure. RV dysfunction at day 4 was less severe in vorinostat-treated compared to untreated hypoxic calves. Cardioprotective effects were partially maintained following cessation of treatment through the duration of hypoxic exposure, accompanied by hemodynamic evidence suggestive of reduced pulmonary vascular stiffening, and modulated expression of HDAC1 protein and genes involved in RV and pulmonary vascular remodeling and pathological RV hypertrophy. Control calves did not develop PH, nor show adverse cardiac or clinical effects. These results provide novel translation of epigenetic-directed therapy to a large animal severe PH model that recapitulates important features of human disease.
Collapse
Affiliation(s)
- Tanya J. Applegate
- Department of Clinical Sciences, Colorado State University College of Veterinary Medicine and Biomedical Sciences, Fort Collins, CO, United States
| | - Greta M. Krafsur
- Division of Clinical Research, Medicine and Pathobiologic Services, RTI, L.L.C., Brookings, SD, United States
- Departments of Pediatrics and Medicine, University of Colorado Denver, Aurora, CO, United States
| | - June A. Boon
- Department of Clinical Sciences, Colorado State University College of Veterinary Medicine and Biomedical Sciences, Fort Collins, CO, United States
| | - Hui Zhang
- Departments of Pediatrics and Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Min Li
- Departments of Pediatrics and Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Timothy N. Holt
- Department of Clinical Sciences, Colorado State University College of Veterinary Medicine and Biomedical Sciences, Fort Collins, CO, United States
| | - S. Kelly Ambler
- Departments of Pediatrics and Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Benjamin A. Abrams
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO, United States
| | - Daniel L. Gustafson
- Department of Clinical Sciences, Colorado State University College of Veterinary Medicine and Biomedical Sciences, Fort Collins, CO, United States
| | - Karsten Bartels
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO, United States
| | - Franklyn B. Garry
- Department of Clinical Sciences, Colorado State University College of Veterinary Medicine and Biomedical Sciences, Fort Collins, CO, United States
| | - Kurt R. Stenmark
- Departments of Pediatrics and Medicine, University of Colorado Denver, Aurora, CO, United States
| | - R. Dale Brown
- Departments of Pediatrics and Medicine, University of Colorado Denver, Aurora, CO, United States
| |
Collapse
|
12
|
Novel Therapeutic Targets for the Treatment of Right Ventricular Remodeling: Insights from the Pulmonary Artery Banding Model. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18168297. [PMID: 34444046 PMCID: PMC8391744 DOI: 10.3390/ijerph18168297] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 12/15/2022]
Abstract
Right ventricular (RV) function is the main determinant of the outcome of patients with pulmonary hypertension (PH). RV dysfunction develops gradually and worsens progressively over the course of PH, resulting in RV failure and premature death. Currently, approved therapies for the treatment of left ventricular failure are not established for the RV. Furthermore, the direct effects of specific vasoactive drugs for treatment of pulmonary arterial hypertension (PAH, Group 1 of PH) on RV are not fully investigated. Pulmonary artery banding (PAB) allows to study the pathogenesis of RV failure solely, thereby testing potential therapies independently of pulmonary vascular changes. This review aims to discuss recent studies of the mechanisms of RV remodeling and RV-directed therapies based on the PAB model.
Collapse
|
13
|
Qin J, Guo N, Tong J, Wang Z. Function of histone methylation and acetylation modifiers in cardiac hypertrophy. J Mol Cell Cardiol 2021; 159:120-129. [PMID: 34175302 DOI: 10.1016/j.yjmcc.2021.06.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 06/14/2021] [Accepted: 06/19/2021] [Indexed: 12/15/2022]
Abstract
Cardiac hypertrophy is an adaptive response of the heart to increased workload induced by various physiological or pathological stimuli. It is a common pathological process in multiple cardiovascular diseases, and it ultimately leads to heart failure. The development of cardiac hypertrophy is accompanied by gene expression reprogramming, a process that is largely dependent on epigenetic regulation. Histone modifications such as methylation and acetylation are dynamically regulated under cardiac stress. These consequently contribute to the pathogenesis of cardiac hypertrophy via compensatory or maladaptive transcriptome reprogramming. Histone methylation and acetylation modifiers play crucial roles in epigenetic remodeling during the pathogenesis of cardiac hypertrophy. Regulation of histone methylation and acetylation modifiers serves as a bridge between signal transduction and downstream gene reprogramming. Exploring the role of histone modifiers in cardiac hypertrophy provides novel therapeutic strategies to treat cardiac hypertrophy and heart failure. In this review, we summarize the recent advancements in functional histone methylation and acetylation modifiers in cardiac hypertrophy, with an emphasis on the underlying mechanisms and the therapeutic potential.
Collapse
Affiliation(s)
- Jian Qin
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ningning Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingjing Tong
- School of Life Sciences, Central China Normal University, Wuhan, China
| | - Zhihua Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen, China; State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
14
|
Prisco SZ, Thenappan T, Prins KW. Treatment Targets for Right Ventricular Dysfunction in Pulmonary Arterial Hypertension. JACC Basic Transl Sci 2020; 5:1244-1260. [PMID: 33426379 PMCID: PMC7775863 DOI: 10.1016/j.jacbts.2020.07.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/27/2020] [Accepted: 07/27/2020] [Indexed: 01/10/2023]
Abstract
Right ventricle (RV) dysfunction is the strongest predictor of mortality in pulmonary arterial hypertension (PAH), but, at present, there are no therapies directly targeting the failing RV. Although there are shared molecular mechanisms in both RV and left ventricle (LV) dysfunction, there are important differences between the 2 ventricles that may allow for the development of RV-enhancing or RV-directed therapies. In this review, we discuss the current understandings of the dysregulated pathways that promote RV dysfunction, highlight RV-enriched or RV-specific pathways that may be of particular therapeutic value, and summarize recent and ongoing clinical trials that are investigating RV function in PAH. It is hoped that development of RV-targeted therapies will improve quality of life and enhance survival for this deadly disease.
Collapse
Key Words
- FAO, fatty acid oxidation
- IPAH, idiopathic pulmonary arterial hypertension
- LV, left ventricle/ventricular
- PAH, pulmonary arterial hypertension
- PH, pulmonary hypertension
- RAAS, renin-angiotensin-aldosterone system
- RV, right ventricle/ventricular
- RVH, right ventricular hypertrophy
- SSc-PAH, systemic sclerosis-associated pulmonary arterial hypertension
- clinical trials
- miRNA/miR, micro-ribonucleic acid
- pulmonary arterial hypertension
- right ventricle
Collapse
Affiliation(s)
- Sasha Z. Prisco
- Cardiovascular Division, Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Thenappan Thenappan
- Cardiovascular Division, Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kurt W. Prins
- Cardiovascular Division, Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
15
|
Chun P. Therapeutic effects of histone deacetylase inhibitors on heart disease. Arch Pharm Res 2020; 43:1276-1296. [PMID: 33245518 DOI: 10.1007/s12272-020-01297-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/22/2020] [Indexed: 01/04/2023]
Abstract
A wide range of histone deacetylase (HDAC) inhibitors have been studied for their therapeutic potential because the excessive activity and expression of HDACs have been implicated in the pathogenesis of cardiac diseases. An increasing number of preclinical studies have demonstrated the cardioprotective effects of numerous HDAC inhibitors, suggesting a wide variety of mechanisms by which the inhibitors protect against cardiac stress, such as the suppression of cardiac fibrosis and fetal gene expression, enhancement of angiogenesis and mitochondrial biogenesis, prevention of electrical remodeling, and regulation of apoptosis, autophagy, and cell cycle arrest. For the development of isoform-selective HDAC inhibitors with high efficacy and low toxicity, it is important to identify and understand the mechanisms responsible for the effects of the inhibitors. This review highlights the preclinical effects of HDAC inhibitors that act against Zn2+-dependent HDACs and the underlying mechanisms of their protective effects against cardiac hypertrophy, hypertension, myocardial infarction, heart failure, and atrial fibrillation.
Collapse
Affiliation(s)
- Pusoon Chun
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, 197 Inje-ro, Gimhae, Gyeongnam, 50834, Republic of Korea.
| |
Collapse
|
16
|
Bisserier M, Pradhan N, Hadri L. Current and emerging therapeutic approaches to pulmonary hypertension. Rev Cardiovasc Med 2020; 21:163-179. [PMID: 32706206 PMCID: PMC7389678 DOI: 10.31083/j.rcm.2020.02.597] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 05/25/2020] [Indexed: 12/15/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and fatal lung disease of multifactorial etiology. Most of the available drugs and FDA-approved therapies for treating pulmonary hypertension attempt to overcome the imbalance between vasoactive and vasodilator mediators, and restore the endothelial cell function. Traditional medications for treating PAH include the prostacyclin analogs and receptor agonists, phosphodiesterase 5 inhibitors, endothelin-receptor antagonists, and cGMP activators. While the current FDA-approved drugs showed improvements in quality of life and hemodynamic parameters, they have shown only very limited beneficial effects on survival and disease progression. None of them offers a cure against PAH, and the median survival rate remains less than three years from diagnosis. Extensive research efforts have led to the emergence of innovative therapeutic approaches in the area of PAH. In this review, we provide an overview of the current FDA-approved therapies in PAH and discuss the associated clinical trials and reported-side effects. As recent studies have led to the emergence of innovative therapeutic approaches in the area of PAH, we also focus on the latest promising therapies in preclinical studies such as stem cell-based therapies, gene transfer, and epigenetic therapies.
Collapse
Affiliation(s)
- Malik Bisserier
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Natasha Pradhan
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lahouaria Hadri
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
17
|
Roles of Histone Acetylation Modifiers and Other Epigenetic Regulators in Vascular Calcification. Int J Mol Sci 2020; 21:ijms21093246. [PMID: 32375326 PMCID: PMC7247359 DOI: 10.3390/ijms21093246] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/28/2020] [Accepted: 05/01/2020] [Indexed: 02/07/2023] Open
Abstract
Vascular calcification (VC) is characterized by calcium deposition inside arteries and is closely associated with the morbidity and mortality of atherosclerosis, chronic kidney disease, diabetes, and other cardiovascular diseases (CVDs). VC is now widely known to be an active process occurring in vascular smooth muscle cells (VSMCs) involving multiple mechanisms and factors. These mechanisms share features with the process of bone formation, since the phenotype switching from the contractile to the osteochondrogenic phenotype also occurs in VSMCs during VC. In addition, VC can be regulated by epigenetic factors, including DNA methylation, histone modification, and noncoding RNAs. Although VC is commonly observed in patients with chronic kidney disease and CVD, specific drugs for VC have not been developed. Thus, discovering novel therapeutic targets may be necessary. In this review, we summarize the current experimental evidence regarding the role of epigenetic regulators including histone deacetylases and propose the therapeutic implication of these regulators in the treatment of VC.
Collapse
|
18
|
Schiano C, Benincasa G, Franzese M, Della Mura N, Pane K, Salvatore M, Napoli C. Epigenetic-sensitive pathways in personalized therapy of major cardiovascular diseases. Pharmacol Ther 2020; 210:107514. [PMID: 32105674 DOI: 10.1016/j.pharmthera.2020.107514] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The complex pathobiology underlying cardiovascular diseases (CVDs) has yet to be explained. Aberrant epigenetic changes may result from alterations in enzymatic activities, which are responsible for putting in and/or out the covalent groups, altering the epigenome and then modulating gene expression. The identification of novel individual epigenetic-sensitive trajectories at single cell level might provide additional opportunities to establish predictive, diagnostic and prognostic biomarkers as well as drug targets in CVDs. To date, most of studies investigated DNA methylation mechanism and miRNA regulation as epigenetics marks. During atherogenesis, big epigenetic changes in DNA methylation and different ncRNAs, such as miR-93, miR-340, miR-433, miR-765, CHROME, were identified into endothelial cells, smooth muscle cells, and macrophages. During man development, lipid metabolism, inflammation and homocysteine homeostasis, alter vascular transcriptional mechanism of fundamental genes such as ABCA1, SREBP2, NOS, HIF1. At histone level, increased HDAC9 was associated with matrix metalloproteinase 1 (MMP1) and MMP2 expression in pro-inflammatory macrophages of human carotid plaque other than to have a positive effect on toll like receptor signaling and innate immunity. HDAC9 deficiency promoted inflammation resolution and reverse cholesterol transport, which might block atherosclerosis progression and promote lesion regression. Here, we describe main human epigenetic mechanisms involved in atherosclerosis, coronary heart disease, ischemic stroke, peripheral artery disease; cardiomyopathy and heart failure. Different epigenetics mechanisms are activated, such as regulation by circular RNAs, as MICRA, and epitranscriptomics at RNA level. Moreover, in order to open new frontiers for precision medicine and personalized therapy, we offer a panoramic view on the most innovative bioinformatic tools designed to identify putative genes and molecular networks underlying CVDs in man.
Collapse
Affiliation(s)
- Concetta Schiano
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Giuditta Benincasa
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | | | | | | | - Claudio Napoli
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; IRCCS SDN, Naples, Italy
| |
Collapse
|
19
|
Chelladurai P, Boucherat O, Stenmark K, Kracht M, Seeger W, Bauer UM, Bonnet S, Pullamsetti SS. Targeting histone acetylation in pulmonary hypertension and right ventricular hypertrophy. Br J Pharmacol 2020; 178:54-71. [PMID: 31749139 DOI: 10.1111/bph.14932] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/21/2019] [Accepted: 11/06/2019] [Indexed: 12/22/2022] Open
Abstract
Epigenetic mechanisms, including DNA methylation and histone post-translational modifications (PTMs), have been known to regulate chromatin structure and lineage-specific gene expression during cardiovascular development and disease. However, alterations in the landscape of histone PTMs and their contribution to the pathogenesis of incurable cardiovascular diseases such as pulmonary hypertension (PH) and associated right heart failure (RHF) remain largely unexplored. This review focusses on the studies in PH and RHF that investigated the gene families that write (histone acetyltransferases), read (bromodomain-containing proteins) or erase (histone deacetylases [HDACs] and sirtuins [SIRT]) acetyl moieties from the ε-amino group of lysine residues of histones and non-histone proteins. Analysis of cells and tissues isolated from the in vivo preclinical models of PH and human pulmonary arterial hypertension not only confirmed significant alterations in the expression levels of multiple HDACs, SIRT1, SIRT3 and BRD4 proteins but also demonstrated their strong association to proliferative, inflammatory and fibrotic phenotypes linked to the pathological vascular remodelling process. Due to the reversible nature of post-translational protein acetylation, the therapeutic efficacy of numerous small-molecule inhibitors (vorinostat, valproic acid, sodium butyrate, mocetinostat, entinostat, tubastatin A, apabetalone, JQ1 and resveratrol) have been evaluated in different preclinical models of cardiovascular disease, which revealed the promising therapeutic benefits of targeting histone acetylation pathways in the attenuation of cardiac hypertrophy, fibrosis, left heart dysfunction, PH and RHF. This review also emphasizes the need for deeper molecular insights into the contribution of epigenetic changes to PH pathogenesis and therapeutic evaluation of isoform-specific modulation in ex vivo and in vivo models of PH and RHF. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.1/issuetoc.
Collapse
Affiliation(s)
- Prakash Chelladurai
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Olivier Boucherat
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Kurt Stenmark
- Cardiovascular Pulmonary Research Laboratories, Division of Pulmonary Sciences and Critical Care Medicine, Division of Pediatrics-Critical Care, Depts of Medicine and Pediatrics, University of Colorado, Aurora, CO, USA
| | - Michael Kracht
- Rudolf-Buchheim-Institute of Pharmacology, Justus Liebig University Giessen, Giessen, Germany
| | - Werner Seeger
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the DZL, Member of CPI, Justus-Liebig University, Giessen, Germany
| | - Uta-Maria Bauer
- Institute for Molecular Biology and Tumor Research (IMT), Philipps-University Marburg, Marburg, Germany
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Soni Savai Pullamsetti
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the DZL, Member of CPI, Justus-Liebig University, Giessen, Germany
| |
Collapse
|
20
|
Bisserier M, Janostiak R, Lezoualc’h F, Hadri L. Targeting epigenetic mechanisms as an emerging therapeutic strategy in pulmonary hypertension disease. VASCULAR BIOLOGY (BRISTOL, ENGLAND) 2020; 2:R17-R34. [PMID: 32161845 PMCID: PMC7065685 DOI: 10.1530/vb-19-0030] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a multifactorial cardiopulmonary disease characterized by an elevation of pulmonary artery pressure (PAP) and pulmonary vascular resistance (PVR), which can lead to right ventricular (RV) failure, multi-organ dysfunction, and ultimately to premature death. Despite the advances in molecular biology, the mechanisms underlying pulmonary hypertension (PH) remain unclear. Nowadays, there is no curative treatment for treating PH. Therefore, it is crucial to identify novel, specific therapeutic targets and to offer more effective treatments against the progression of PH. Increasing amounts of evidence suggest that epigenetic modification may play a critical role in the pathogenesis of PAH. In the presented paper, we provide an overview of the epigenetic mechanisms specifically, DNA methylation, histone acetylation, histone methylation, and ncRNAs. As the recent identification of new pharmacological drugs targeting these epigenetic mechanisms has opened new therapeutic avenues, we also discuss the importance of epigenetic-based therapies in the context of PH.
Collapse
Affiliation(s)
- Malik Bisserier
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Radoslav Janostiak
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Frank Lezoualc’h
- Inserm, UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, University of Toulouse, Toulouse Cedex 4, France
| | - Lahouaria Hadri
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
21
|
Jung H, Lee E, Kim I, Song JH, Kim GJ. Histone deacetylase inhibition has cardiac and vascular protective effects in rats with pressure overload cardiac hypertrophy. Physiol Res 2019; 68:727-737. [PMID: 31424255 DOI: 10.33549/physiolres.934110] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Histone deacetylase (HDAC) inhibitors have shown beneficial effects in animal models of cardiovascular diseases. We hypothesized that HDAC inhibitor, sodium valproate (VPA), has cardiac and vascular protective effects in rats with pressure overload cardiac hypertrophy induced by transverse aortic constriction (TAC). Sections of the heart were visualized after hematoxylin and eosin staining, picrosirius red staining and immunohistochemistry. The expression of genes related to cardiac hypertrophy, fibrosis, and oxidative stress was determined by quantitative real-time polymerase chain reaction. The aortic ring tension analysis was conducted using both the ascending aorta and descending thoracic aorta. TAC increased the expression of hypertrophic, fibrotic, and oxidative stress genes, which was attenuated by VPA. In the ascending aorta with intact endothelium, there was a significant decrease in the relaxation response, which was recovered by VPA treatment. These results indicate that VPA has cardiac and vascular protective effects in rats with pressure overload cardiac hypertrophy.
Collapse
Affiliation(s)
- H Jung
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea.
| | | | | | | | | |
Collapse
|
22
|
Lyu X, Hu M, Peng J, Zhang X, Sanders YY. HDAC inhibitors as antifibrotic drugs in cardiac and pulmonary fibrosis. Ther Adv Chronic Dis 2019; 10:2040622319862697. [PMID: 31367296 PMCID: PMC6643173 DOI: 10.1177/2040622319862697] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 06/17/2019] [Indexed: 12/14/2022] Open
Abstract
Fibrosis usually results from dysregulated wound repair and is characterized by
excessive scar tissue. It is a complex process with unclear mechanisms.
Accumulating evidence indicates that epigenetic alterations, including histone
acetylation, play a pivotal role in this process. Histone acetylation is
governed by histone acetyltransferases (HATs) and histone deacetylases (HDACs).
HDACs are enzymes that remove the acetyl groups from both histone and nonhistone
proteins. Aberrant HDAC activities are observed in fibrotic diseases, including
cardiac and pulmonary fibrosis. HDAC inhibitors (HDACIs) are molecules that
block HDAC functions. HDACIs have been studied extensively in a variety of
tumors. Currently, there are four HDACIs approved by the US Food and Drug
Administration for cancer treatment yet none for fibrotic diseases. Emerging
evidence from in vitro and in vivo preclinical
studies has presented beneficial effects of HDACIs in preventing or reversing
fibrogenesis. In this review, we summarize the latest findings of the roles of
HDACs in the pathogenesis of cardiac and pulmonary fibrosis and highlight the
potential applications of HDACIs in these two fibrotic diseases.
Collapse
Affiliation(s)
- Xing Lyu
- Laboratory of Clinical Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Min Hu
- Laboratory of Clinical Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jieting Peng
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiangyu Zhang
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yan Y Sanders
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, 901 19 Street South, BMRII Room 408, Birmingham, AL 35294, USA
| |
Collapse
|
23
|
Yuan L, Liu C, Wan Y, Yan H, Li T. Effect of HDAC2/Inpp5f on neuropathic pain and cognitive function through regulating PI3K/Akt/GSK-3β signal pathway in rats with neuropathic pain. Exp Ther Med 2019; 18:678-684. [PMID: 31281447 PMCID: PMC6580097 DOI: 10.3892/etm.2019.7622] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022] Open
Abstract
The effect of histone deacetylase (HDAC)2/Inositol polyphosphate-5-phosphatase F (Inpp5f) on neuropathic pain and cognitive dysfunction through regulating PI3K/Akt/GSK-3β signal pathway in rats with neuropathic pain was investigated. A total of 80 SPF mature male SD rats were averagely randomized into the sham operation group, the model group, the HDAC2 intervention group (group A) and the Inpp5f intervention group (group B). The rat models of neuropathic pain were established in the model group, and groups A and B. At the 15th day after modeling, rats in group A were transfected with the interference vector of HDAC2, and rats in group B were transfected with the overexpression vector of Inpp5f. Rats in the four groups were observed before modeling, after modeling/before intervention and 3 days after intervention in terms of paw thermal withdrawal latency (PWL), paw withdrawal mechanical threshold (PWT) and changes in cognitive function (Morris water maze and passive avoidance task). Then the rats were sacrificed. RT-qPCR and western blot analysis were used to detect the levels of HDAC2 mRNA, Inpp5f mRNA, phosphorylated PI3K (p-PI3K), phosphorylated AKT (p-AKT), phosphorylated GSK-3β (p-GSK-3β) in rat brain tissue. Correlation of HDAC2 mRNA with Inpp5f mRNA expression levels was detected by Pearsons correlation analysis. Compared with the sham operation group, PWL was significantly lower while PWT was higher in the other 3 groups (P<0.05). Three days after intervention, PWL was significantly higher while PWT was significantly lower (P<0.05). Inhibiting the expression of HDAC2 or promoting the expression of Inpp5f can effectively improve cognitive function in rats (P<0.05). After intervention, compared with the sham operation group, rats in the other 3 groups had higher HDAC2 mRNA level and lower Inpp5f mRNA level (P<0.05). In conclusion, neuropathic pain can cause an increase in HDAC2 expression level and a decrease in Inpp5f expression level, and activate the PI3K/Akt/GSK-3β signal pathway. Inhibition of HDAC2 expression can inhibit the activation of PI3K/Akt/GSK-3β signal pathway through increasing Inpp5f expression, thus improving the condition and cognitive disorder of rats with neuropathic pain.
Collapse
Affiliation(s)
- Lili Yuan
- Department of Anesthesiology, Fifth Hospital in Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430050, P.R. China
| | - Caihua Liu
- Department of Anesthesiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430050, P.R. China
| | - Yingchun Wan
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Hong Yan
- Department of Anesthesiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430050, P.R. China
| | - Tao Li
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
24
|
Bagchi RA, Weeks KL. Histone deacetylases in cardiovascular and metabolic diseases. J Mol Cell Cardiol 2019; 130:151-159. [PMID: 30978343 DOI: 10.1016/j.yjmcc.2019.04.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/29/2019] [Accepted: 04/06/2019] [Indexed: 12/13/2022]
Abstract
Histone deacetylases (HDACs) regulate gene transcription by catalyzing the removal of acetyl groups from key lysine residues in nucleosomal histones and via the recruitment of other epigenetic regulators to DNA promoter/enhancer regions. Over the past two decades, HDACs have been implicated in multiple processes pertinent to cardiovascular and metabolic diseases, including cardiac hypertrophy and remodeling, fibrosis, calcium handling, inflammation and energy metabolism. The development of small molecule HDAC inhibitors and genetically modified loss- and gain-of-function mouse models has allowed interrogation of the roles of specific HDAC isoforms in these processes. Isoform-selective HDAC inhibitors may prove to be powerful therapeutic agents for the treatment of cardiovascular diseases, obesity and diabetes.
Collapse
Affiliation(s)
- Rushita A Bagchi
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States of America
| | - Kate L Weeks
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
25
|
Ren X, Johns RA, Gao WD. EXPRESS: Right Heart in Pulmonary Hypertension: From Adaptation to Failure. Pulm Circ 2019; 9:2045894019845611. [PMID: 30942134 PMCID: PMC6681271 DOI: 10.1177/2045894019845611] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/27/2019] [Indexed: 01/24/2023] Open
Abstract
Right ventricular (RV) failure (RVF) has garnered significant attention in recent years because of its negative impact on clinical outcomes in patients with pulmonary hypertension (PH). PH triggers a series of events, including activation of several signaling pathways that regulate cell growth, metabolism, extracellular matrix remodeling, and energy production. These processes render the RV adaptive to PH. However, RVF develops when PH persists, accompanied by RV ischemia, alterations in substrate and mitochondrial energy metabolism, increased free oxygen radicals, increased cell loss, downregulation of adrenergic receptors, increased inflammation and fibrosis, and pathologic microRNAs. Diastolic dysfunction is also an integral part of RVF. Emerging non-invasive technologies such as molecular or metallic imaging, cardiac MRI, and ultrafast Doppler coronary flow mapping will be valuable tools to monitor RVF, especially the transition to RVF. Most PH therapies cannot treat RVF once it has occurred. A variety of therapies are available to treat acute and chronic RVF, but they are mainly supportive, and no effective therapy directly targets the failing RV. Therapies that target cell growth, cellular metabolism, oxidative stress, and myocyte regeneration are being tested preclinically. Future research should include establishing novel RVF models based on existing models, increasing use of human samples, creating human stem cell-based in vitro models, and characterizing alterations in cardiac excitation–contraction coupling during transition from adaptive RV to RVF. More successful strategies to manage RVF will likely be developed as we learn more about the transition from adaptive remodeling to maladaptive RVF in the future.
Collapse
Affiliation(s)
- Xianfeng Ren
- Department of Anesthesiology,
China-Japan
Friendship Hospital, Beijing, China
| | - Roger A. Johns
- Department of Anesthesiology and
Critical Care Medicine,
Johns
Hopkins University School of Medicine,
Baltimore, MD, USA
| | - Wei Dong Gao
- Department of Anesthesiology and
Critical Care Medicine,
Johns
Hopkins University School of Medicine,
Baltimore, MD, USA
| |
Collapse
|
26
|
HDAC Inhibitors: Therapeutic Potential in Fibrosis-Associated Human Diseases. Int J Mol Sci 2019; 20:ijms20061329. [PMID: 30884785 PMCID: PMC6471162 DOI: 10.3390/ijms20061329] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/05/2019] [Accepted: 03/12/2019] [Indexed: 02/07/2023] Open
Abstract
Fibrosis is characterized by excessive deposition of the extracellular matrix and develops because of fibroblast differentiation during the process of inflammation. Various cytokines stimulate resident fibroblasts, which differentiate into myofibroblasts. Myofibroblasts actively synthesize an excessive amount of extracellular matrix, which indicates pathologic fibrosis. Although initial fibrosis is a physiologic response, the accumulated fibrous material causes failure of normal organ function. Cardiac fibrosis interferes with proper diastole, whereas pulmonary fibrosis results in chronic hypoxia; liver cirrhosis induces portal hypertension, and overgrowth of fibroblasts in the conjunctiva is a major cause of glaucoma surgical failure. Recently, several reports have clearly demonstrated the functional relevance of certain types of histone deacetylases (HDACs) in various kinds of fibrosis and the successful alleviation of the condition in animal models using HDAC inhibitors. In this review, we discuss the therapeutic potential of HDAC inhibitors in fibrosis-associated human diseases using results obtained from animal models.
Collapse
|
27
|
Zhang J, Xu Z, Gu J, Jiang S, Liu Q, Zheng Y, Freedman JH, Sun J, Cai L. HDAC3 inhibition in diabetic mice may activate Nrf2 preventing diabetes-induced liver damage and FGF21 synthesis and secretion leading to aortic protection. Am J Physiol Endocrinol Metab 2018; 315:E150-E162. [PMID: 29634312 DOI: 10.1152/ajpendo.00465.2017] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Vascular complications are common pathologies associated with type 1 diabetes. In recent years, histone deacetylation enzyme (HDAC) inhibitors have been shown to be successful in preventing atherosclerosis. To investigate the mechanism for HDAC3 inhibition in preventing diabetic aortic pathologies, male OVE26 type 1 diabetic mice and age-matched wild-type (FVB) mice were given the HDAC3-specific inhibitor RGFP-966 or vehicle for 3 mo. These mice were then euthanized immediately or maintained for an additional 3 mo without treatment. Levels of aortic inflammation and fibrosis and plasma and fibroblast growth factor 21 (FGF21) levels were determined. Because the liver is the major organ for FGF21 synthesis in diabetic animals, the effects of HDAC3 inhibition on hepatic FGF21 synthesis were examined. Additionally, hepatic miR-200a and kelch-like ECH-associated protein 1 (Keap1) expression and nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation were measured. HDAC3 inhibition significantly reduced aortic fibrosis and inflammation in OVE26 mice at both 3 and 6 mo. Plasma FGF21 levels were significantly higher in RGFP-966-treated OVE26 mice compared with vehicle-treated mice at both time points. It also significantly reduced hepatic pathologies associated with diabetes, accompanied by increased FGF21 mRNA and protein expression. HDAC3 inhibition also increased miR-200a expression, reduced Keap1 protein levels, and increased Nrf2 nuclear translocation with an upregulation of antioxidant gene and FGF21 transcription. Our results support a model where HDAC3 inhibition may promote Nrf2 activity by increasing miR-200a expression with a concomitant decrease in Keap1 to preserve hepatic FGF21 synthesis. The preservation of hepatic FGF21 synthesis ultimately leads to a reduction in diabetes-induced aorta pathologies.
Collapse
Affiliation(s)
- Jian Zhang
- Cardiovascular Center, the First Hospital of Jilin University , Changchun, Jilin , China
- Department of Pediatrics, Pediatric Research Institute, University of Louisville , Louisville, Kentucky
| | - Zheng Xu
- Cardiovascular Center, the First Hospital of Jilin University , Changchun, Jilin , China
| | - Junlian Gu
- Department of Pediatrics, Pediatric Research Institute, University of Louisville , Louisville, Kentucky
| | - Saizhi Jiang
- Department of Pediatrics, Pediatric Research Institute, University of Louisville , Louisville, Kentucky
- Department of Pediatrics, the First Affiliated Hospital of Wenzhou Medical University , Wenzhou, Zhejiang , China
| | - Quan Liu
- Cardiovascular Center, the First Hospital of Jilin University , Changchun, Jilin , China
| | - Yang Zheng
- Cardiovascular Center, the First Hospital of Jilin University , Changchun, Jilin , China
| | - Jonathan H Freedman
- Department of Pharmacology and Toxicology, University of Louisville , Louisville, Kentucky
| | - Jian Sun
- Cardiovascular Center, the First Hospital of Jilin University , Changchun, Jilin , China
| | - Lu Cai
- Department of Pediatrics, Pediatric Research Institute, University of Louisville , Louisville, Kentucky
- Department of Pharmacology and Toxicology, University of Louisville , Louisville, Kentucky
| |
Collapse
|
28
|
Chabert C, Khochbin S, Rousseaux S, Veyrenc S, Furze R, Smithers N, Prinjha RK, Schlattner U, Pison C, Dubouchaud H. Inhibition of BET Proteins Reduces Right Ventricle Hypertrophy and Pulmonary Hypertension Resulting from Combined Hypoxia and Pulmonary Inflammation. Int J Mol Sci 2018; 19:ijms19082224. [PMID: 30061518 PMCID: PMC6121304 DOI: 10.3390/ijms19082224] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 07/22/2018] [Indexed: 12/17/2022] Open
Abstract
Pulmonary hypertension is a co-morbidity, which strongly participates in morbi-mortality in patients with chronic obstructive pulmonary disease (COPD). Recent findings showed that bromodomain-containing proteins, in charge of reading histone acetylation, could be involved in pulmonary arterial hypertension. Our aim was to study the effect of I-BET151, an inhibitor of bromodomain and extra-terminal domain (BET), on the right ventricle hypertrophy and pulmonary hypertension, induced by a combination of chronic hypoxia and pulmonary inflammation, as the two main stimuli encountered in COPD. Adult Wistar male rats, exposed to chronic hypoxia plus pulmonary inflammation (CHPI), showed a significant right ventricle hypertrophy (+57%, p < 0.001), an increase in systolic pressure (+46%, p < 0.001) and in contraction speed (+36%, p < 0.001), when compared to control animals. I-BET151 treated animals (CHPI-iB) showed restored hemodynamic parameters to levels similar to control animals, despite chronic hypoxia plus exposure to pulmonary inflammation. They displayed lower right ventricle hypertrophy and hematocrit compared to the CHPI group (respectively -16%, p < 0.001; and -9%, p < 0.05). Our descriptive study shows a valuable effect of the inhibition of bromodomain and extra-terminal domain proteins on hemodynamic parameters, despite the presence of chronic hypoxia and pulmonary inflammation. This suggests that such inhibition could be of potential interest for COPD patients with pulmonary hypertension. Further studies are needed to unravel the underlying mechanisms involved and the net benefits of inhibiting adaptations to chronic hypoxia.
Collapse
MESH Headings
- Animals
- Blood Pressure/drug effects
- Heterocyclic Compounds, 4 or More Rings/therapeutic use
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/physiopathology
- Hypertrophy, Right Ventricular/drug therapy
- Hypertrophy, Right Ventricular/etiology
- Hypertrophy, Right Ventricular/pathology
- Hypertrophy, Right Ventricular/physiopathology
- Hypoxia/complications
- Hypoxia/pathology
- Hypoxia/physiopathology
- Male
- Pneumonia/complications
- Pneumonia/pathology
- Pneumonia/physiopathology
- Pulmonary Disease, Chronic Obstructive/complications
- Pulmonary Disease, Chronic Obstructive/pathology
- Pulmonary Disease, Chronic Obstructive/physiopathology
- Rats, Wistar
- Transcription Factors/antagonists & inhibitors
Collapse
Affiliation(s)
- Clovis Chabert
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée, 38058 Grenoble, France.
| | - Saadi Khochbin
- CNRS UMR 5309, Inserm U1209, Université Grenoble Alpes, Institute for Advanced Biosciences, 38700 Grenoble, France.
| | - Sophie Rousseaux
- CNRS UMR 5309, Inserm U1209, Université Grenoble Alpes, Institute for Advanced Biosciences, 38700 Grenoble, France.
| | - Sylvie Veyrenc
- Université Grenoble Alpes, CNRS UMR 5553, Laboratoire d'Ecologie Alpine, 38058 Grenoble, France.
| | - Rebecca Furze
- Epigenetics DPU, Immuno-Inflammation Therapy Area, GlaxoSmithKline R&D, Medicines Research Centre, Stevenage SG1 2NY, UK.
| | - Nicholas Smithers
- Epigenetics DPU, Immuno-Inflammation Therapy Area, GlaxoSmithKline R&D, Medicines Research Centre, Stevenage SG1 2NY, UK.
| | - Rab K Prinjha
- Epigenetics DPU, Immuno-Inflammation Therapy Area, GlaxoSmithKline R&D, Medicines Research Centre, Stevenage SG1 2NY, UK.
| | - Uwe Schlattner
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée, 38058 Grenoble, France.
| | - Christophe Pison
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée, 38058 Grenoble, France.
- Centre Hospitalier Universitaire Grenoble Alpes, Université Grenoble Alpes, 38700 Grenoble, France.
| | - Hervé Dubouchaud
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée, 38058 Grenoble, France.
| |
Collapse
|
29
|
Abstract
Pulmonary hypertension is defined as a resting mean pulmonary artery pressure of 25 mm Hg or above. This review deals with pulmonary arterial hypertension (PAH), a type of pulmonary hypertension that primarily affects the pulmonary vasculature. In PAH, the pulmonary vasculature is dynamically obstructed by vasoconstriction, structurally obstructed by adverse vascular remodeling, and pathologically non-compliant as a result of vascular fibrosis and stiffening. Many cell types are abnormal in PAH, including vascular cells (endothelial cells, smooth muscle cells, and fibroblasts) and inflammatory cells. Progress has been made in identifying the causes of PAH and approving new drug therapies. A cancer-like increase in cell proliferation and resistance to apoptosis reflects acquired abnormalities of mitochondrial metabolism and dynamics. Mutations in the type II bone morphogenetic protein receptor (BMPR2) gene dramatically increase the risk of developing heritable PAH. Epigenetic dysregulation of DNA methylation, histone acetylation, and microRNAs also contributes to disease pathogenesis. Aberrant bone morphogenetic protein signaling and epigenetic dysregulation in PAH promote cell proliferation in part through induction of a Warburg mitochondrial-metabolic state of uncoupled glycolysis. Complex changes in cytokines (interleukins and tumor necrosis factor), cellular immunity (T lymphocytes, natural killer cells, macrophages), and autoantibodies suggest that PAH is, in part, an autoimmune, inflammatory disease. Obstructive pulmonary vascular remodeling in PAH increases right ventricular afterload causing right ventricular hypertrophy. In some patients, maladaptive changes in the right ventricle, including ischemia and fibrosis, reduce right ventricular function and cause right ventricular failure. Patients with PAH have dyspnea, reduced exercise capacity, exertional syncope, and premature death from right ventricular failure. PAH targeted therapies (prostaglandins, phosphodiesterase-5 inhibitors, endothelin receptor antagonists, and soluble guanylate cyclase stimulators), used alone or in combination, improve functional capacity and hemodynamics and reduce hospital admissions. However, these vasodilators do not target key features of PAH pathogenesis and have not been shown to reduce mortality, which remains about 50% at five years. This review summarizes the epidemiology, pathogenesis, diagnosis, and treatment of PAH.
Collapse
Affiliation(s)
| | - Mark L Ormiston
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - John J Ryan
- Department of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, ON, Canada
| |
Collapse
|
30
|
Hensley MK, Levine A, Gladwin MT, Lai YC. Emerging therapeutics in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2018; 314:L769-L781. [PMID: 29388467 DOI: 10.1152/ajplung.00259.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive and often fatal illness presenting with nonspecific symptoms of dyspnea, lower extremity edema, and exercise intolerance. Pathologically, endothelial dysfunction leads to abnormal intimal and smooth muscle proliferation along with reduced apoptosis, resulting in increased pulmonary vascular resistance and elevated pulmonary pressures. PH is subdivided into five World Health Organization groups based on the disease pathology and specific cause. While there are Food and Drug Administration-approved medications for the treatment of pulmonary arterial hypertension (PAH; Group 1 PH), as well as for chronic thromboembolic PH (Group 4 PH), the morbidity and mortality remain high. Moreover, there are no approved therapies for other forms of PH (Groups 2, 3, and 5) at present. New research has identified molecular targets that mediate vasodilation, anti-inflammatory, and antifibrotic changes within the pulmonary vasculature. Given that PAH is the most commonly studied form of PH worldwide and because recent studies have led to better mechanistic understanding of this devastating disease, in this review we attempt to provide an updated overview of new therapeutic approaches under investigation for the treatment of PH, with a particular focus on PAH, as well as to offer guidelines for future investigations.
Collapse
Affiliation(s)
- Matthew K Hensley
- Division of Pulmonary and Critical Care Medicine, University of Michigan , Ann Arbor, Michigan
| | - Andrea Levine
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Yen-Chun Lai
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|
31
|
Xu Z, Tong Q, Zhang Z, Wang S, Zheng Y, Liu Q, Qian LB, Chen SY, Sun J, Cai L. Inhibition of HDAC3 prevents diabetic cardiomyopathy in OVE26 mice via epigenetic regulation of DUSP5-ERK1/2 pathway. Clin Sci (Lond) 2017; 131:1841-1857. [PMID: 28533215 PMCID: PMC5737625 DOI: 10.1042/cs20170064] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/17/2017] [Accepted: 05/19/2017] [Indexed: 02/07/2023]
Abstract
Inhibition of total histone deacetylases (HDACs) was phenomenally associated with the prevention of diabetic cardiomyopathy (DCM). However, which specific HDAC plays the key role in DCM remains unclear. The present study was designed to determine whether DCM can be prevented by specific inhibition of HDAC3 and to elucidate the mechanisms by which inhibition of HDAC3 prevents DCM. Type 1 diabetes OVE26 and age-matched wild-type (WT) mice were given the selective HDAC3 inhibitor RGFP966 or vehicle for 3 months. These mice were then killed immediately or 3 months later for cardiac function and pathological examination. HDAC3 activity was significantly increased in the heart of diabetic mice. Administration of RGFP966 significantly prevented DCM, as evidenced by improved diabetes-induced cardiac dysfunction, hypertrophy, and fibrosis, along with diminished cardiac oxidative stress, inflammation, and insulin resistance, not only in the mice killed immediately or 3 months later following the 3-month treatment. Furthermore, phosphorylated extracellular signal-regulated kinases (ERK) 1/2, a well-known initiator of cardiac hypertrophy, was significantly increased, while dual specificity phosphatase 5 (DUSP5), an ERK1/2 nuclear phosphatase, was substantially decreased in diabetic hearts. Both of these changes were prevented by RGFP966. Chromatin immunoprecipitation (ChIP) assay showed that HDAC3 inhibition elevated histone H3 acetylation on the DUSP5 gene promoter at both two time points. These findings suggest that diabetes-activated HDAC3 inhibits DUSP5 expression through deacetylating histone H3 on the primer region of DUSP5 gene, leading to the derepression of ERK1/2 and the initiation of DCM. The present study indicates the potential application of HDAC3 inhibitor for the prevention of DCM.
Collapse
MESH Headings
- Acrylamides/therapeutic use
- Animals
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/genetics
- Diabetic Cardiomyopathies/etiology
- Diabetic Cardiomyopathies/genetics
- Diabetic Cardiomyopathies/prevention & control
- Drug Evaluation, Preclinical/methods
- Dual-Specificity Phosphatases/metabolism
- Epigenesis, Genetic/drug effects
- Histone Deacetylase Inhibitors/pharmacology
- Histone Deacetylase Inhibitors/therapeutic use
- Histone Deacetylases/drug effects
- Histone Deacetylases/metabolism
- Histone Deacetylases/physiology
- MAP Kinase Signaling System/drug effects
- MAP Kinase Signaling System/genetics
- Male
- Mice, Transgenic
- Myocardium/enzymology
- Oxidative Stress/drug effects
- Phenylenediamines/therapeutic use
- Receptor, Insulin/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Zheng Xu
- Cardiovascular Center, the First Hospital of Jilin University, Changchun 130021, China
- Pediatric Research Institute at the Department of Pediatrics, the University of Louisville, Louisville, KY 40202, U.S.A
| | - Qian Tong
- Cardiovascular Center, the First Hospital of Jilin University, Changchun 130021, China
| | - Zhiguo Zhang
- Cardiovascular Center, the First Hospital of Jilin University, Changchun 130021, China
| | - Shudong Wang
- Cardiovascular Center, the First Hospital of Jilin University, Changchun 130021, China
| | - Yang Zheng
- Cardiovascular Center, the First Hospital of Jilin University, Changchun 130021, China
| | - Qiuju Liu
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China
| | - Ling-Bo Qian
- Pediatric Research Institute at the Department of Pediatrics, the University of Louisville, Louisville, KY 40202, U.S.A
- Department of Basic Medical Sciences, Hangzhou Medical College, Hangzhou 310053, China
| | - Shao-Yu Chen
- Department of Pharmacology and Toxicology, Alcohol Research Center, University of Louisville, Louisville, KY 40202, U.S.A
| | - Jian Sun
- Cardiovascular Center, the First Hospital of Jilin University, Changchun 130021, China
| | - Lu Cai
- Cardiovascular Center, the First Hospital of Jilin University, Changchun 130021, China
- Pediatric Research Institute at the Department of Pediatrics, the University of Louisville, Louisville, KY 40202, U.S.A
| |
Collapse
|
32
|
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: 13] [Impact Index Per Article: 1.6] [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
|
33
|
Chelladurai P, Seeger W, Pullamsetti SS. Epigenetic mechanisms in pulmonary arterial hypertension: the need for global perspectives. Eur Respir Rev 2017; 25:135-40. [PMID: 27246590 PMCID: PMC9487251 DOI: 10.1183/16000617.0036-2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 05/06/2016] [Indexed: 02/07/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe and progressive disease, characterised by high pulmonary artery pressure that usually culminates in right heart failure. Recent findings of alterations in the DNA methylation state of superoxide dismutase 2 and granulysin gene loci; histone H1 levels; aberrant expression levels of histone deacetylases and bromodomain-containing protein 4; and dysregulated microRNA networks together suggest the involvement of epigenetics in PAH pathogenesis. Thus, PAH pathogenesis evidently involves the interplay of a predisposed genetic background, epigenetic state and injurious events. Profiling the genome-wide alterations in the epigenetic mechanisms, such as DNA methylation or histone modification pattern in PAH vascular cells, may explain the great variability in susceptibility and disease severity that is frequently associated with pronounced remodelling and worse clinical outcome. Moreover, the influence of genetic predisposition and the acquisition of epigenetic alterations in response to environmental cues in PAH progression and establishment has largely been unexplored on a genome-wide scale. In order to gain insights into the molecular mechanisms leading to the development of PAH and to design novel therapeutic strategies, high-throughput approaches have to be adopted to facilitate systematic identification of the disease-specific networks using next-generation sequencing technologies, the application of these technologies in PAH has been relatively trivial to date. An epigenetic component is hypothesised in PAH: an overview of the current literature and future perspectiveshttp://ow.ly/7miS3002BYw
Collapse
Affiliation(s)
- Prakash Chelladurai
- Max-Planck-Institute for Heart and Lung Research, Dept of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Werner Seeger
- Max-Planck-Institute for Heart and Lung Research, Dept of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), Bad Nauheim, Germany University of Giessen Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Soni Savai Pullamsetti
- Max-Planck-Institute for Heart and Lung Research, Dept of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), Bad Nauheim, Germany University of Giessen Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| |
Collapse
|
34
|
Maarman GJ, Schulz R, Sliwa K, Schermuly RT, Lecour S. Novel putative pharmacological therapies to protect the right ventricle in pulmonary hypertension: a review of current literature. Br J Pharmacol 2017; 174:497-511. [PMID: 28099680 DOI: 10.1111/bph.13721] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 09/06/2016] [Accepted: 09/27/2016] [Indexed: 12/15/2022] Open
Abstract
Pulmonary hypertension (PH) is defined by elevated mean pulmonary artery pressure following the pathological remodelling of small pulmonary arteries. An increase in right ventricular (RV) afterload results in RV hypertrophy and RV failure. The pathophysiology of PH, and RV remodelling in particular, is not well understood, thus explaining, at least in part, why current PH therapies have a limited effect. Existing therapies mostly target the pulmonary circulation. Because the remodelled RV fails to support normal cardiac function, patients eventually succumb from RV failure. Developing novel therapies that directly target the function of the RV may therefore benefit patients with PH. In the past decade, several promising studies have investigated novel cardioprotective strategies in experimental models of PH. This review aims to comprehensively discuss and highlight these novel experimental approaches to confer, in the long-term, greater health benefit in patients with PH.
Collapse
Affiliation(s)
- Gerald J Maarman
- Hatter Institute for Cardiovascular Research in Africa (HICRA) and MRC Inter-University Cape Heart Group, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Karen Sliwa
- Hatter Institute for Cardiovascular Research in Africa (HICRA) and MRC Inter-University Cape Heart Group, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Ralph Theo Schermuly
- Universities of Giessen and Marburg Lung Centre, Member of the German Lung Centre (DZL), Justus Liebig University Giessen, Giessen, Germany
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa (HICRA) and MRC Inter-University Cape Heart Group, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
35
|
Chowdhury SK, Liu W, Zi M, Li Y, Wang S, Tsui H, Prehar S, Castro S, Zhang H, Ji Y, Zhang X, Xiao R, Zhang R, Lei M, Cyganek L, Guan K, Millar CB, Liao X, Jain MK, Boyett MR, Cartwright EJ, Shiels HA, Wang X. Stress-Activated Kinase Mitogen-Activated Kinase Kinase-7 Governs Epigenetics of Cardiac Repolarization for Arrhythmia Prevention. Circulation 2016; 135:683-699. [PMID: 27899394 DOI: 10.1161/circulationaha.116.022941] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 11/16/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND Ventricular arrhythmia is a leading cause of cardiac mortality. Most antiarrhythmics present paradoxical proarrhythmic side effects, culminating in a greater risk of sudden death. METHODS We describe a new regulatory mechanism linking mitogen-activated kinase kinase-7 deficiency with increased arrhythmia vulnerability in hypertrophied and failing hearts using mouse models harboring mitogen-activated kinase kinase-7 knockout or overexpression. The human relevance of this arrhythmogenic mechanism is evaluated in human-induced pluripotent stem cell-derived cardiomyocytes. Therapeutic potentials by targeting this mechanism are explored in the mouse models and human-induced pluripotent stem cell-derived cardiomyocytes. RESULTS Mechanistically, hypertrophic stress dampens expression and phosphorylation of mitogen-activated kinase kinase-7. Such mitogen-activated kinase kinase-7 deficiency leaves histone deacetylase-2 unphosphorylated and filamin-A accumulated in the nucleus to form a complex with Krüppel-like factor-4. This complex leads to Krüppel-like factor-4 disassociation from the promoter regions of multiple key potassium channel genes (Kv4.2, KChIP2, Kv1.5, ERG1, and Kir6.2) and reduction of their transcript levels. Consequent repolarization delays result in ventricular arrhythmias. Therapeutically, targeting the repressive function of the Krüppel-like factor-4/histone deacetylase-2/filamin-A complex with the histone deacetylase-2 inhibitor valproic acid restores K+ channel expression and alleviates ventricular arrhythmias in pathologically remodeled hearts. CONCLUSIONS Our findings unveil this new gene regulatory avenue as a new antiarrhythmic target where repurposing of the antiepileptic drug valproic acid as an antiarrhythmic is supported.
Collapse
Affiliation(s)
- Sanjoy K Chowdhury
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Wei Liu
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Min Zi
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Yatong Li
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Shunyao Wang
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Hoyee Tsui
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Sukhpal Prehar
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Simon Castro
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Henggui Zhang
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Yong Ji
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Xiuqin Zhang
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Ruiping Xiao
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Rongli Zhang
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Ming Lei
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Lukas Cyganek
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Kaomei Guan
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Catherine B Millar
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Xudong Liao
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Mukesh K Jain
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Mark R Boyett
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Elizabeth J Cartwright
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Holly A Shiels
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.)
| | - Xin Wang
- From Faculty of Biology, Medicine and Health (S.K.C., W.L., M.Z., Y.L., S.W., H.T., S.P., C.B.M., M.R.B., E.J.C., H.A.S., X.W.) and School of Physics and Astronomy (S.C., H.Z.), University of Manchester, United Kingdom; Atherosclerosis Research Centre, Nanjing Medical University, Jiangsu, China (Y.J.); Institute of Molecular Medicine, Peking University, Beijing, China (X.Z., R.X.); Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (R.Z., X.L., M.K.J.); Department of Pharmacology, University of Oxford, United Kingdom (M.L.); and Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (L.C., K.G.).
| |
Collapse
|
36
|
Mumby S, Gambaryan N, Meng C, Perros F, Humbert M, Wort SJ, Adcock IM. Bromodomain and extra-terminal protein mimic JQ1 decreases inflammation in human vascular endothelial cells: Implications for pulmonary arterial hypertension. Respirology 2016; 22:157-164. [PMID: 27539364 PMCID: PMC5215513 DOI: 10.1111/resp.12872] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 04/17/2016] [Accepted: 05/31/2016] [Indexed: 12/22/2022]
Abstract
Background and objective Nuclear factor kappa B (NF‐kB)‐mediated inflammatory gene expression and vascular endothelial cell proliferation/remodelling are implicated in the pathophysiology of the fatal disease, pulmonary arterial hypertension (PAH). Bromodomain and extra‐terminal (BET) proteins are essential for the expression of a subset of NF‐kB‐induced inflammatory genes. BET mimics including JQ1+ prevent binding of BETs to acetylated histones and down‐regulate the expression of selected genes. Methods The effects of JQ1+ on the proliferation of primary human pulmonary microvascular endothelial cells (HPMECs) from healthy subjects were measured by bromodeoxyuridine (BrdU) incorporation. Cell cycle progression was assessed by flow cytometry; mRNA and protein levels of cyclin‐dependent kinases (CDKs), inhibitors and cytokines were determined by reverse transcription‐quantitative PCR (RT‐qPCR), Western blotting or ELISA. Histone acetyltransferase (HAT) and deacetylase (HDAC) activities were determined in nuclear extracts from whole lung of PAH and control patients. Results JQ1+ significantly inhibited IL6 and IL8 (IL6 and CXCL8) mRNA and protein in HPMECs compared with its inactive enantiomer JQ1−. JQ1+ decreased NF‐kB p65 recruitment to native IL6 and IL8 promoters. JQ1+ showed a concentration‐dependent decrease in HPMEC proliferation compared with JQ1−‐treated cells. JQ1+ induced G1 cell cycle arrest by increasing the expression of the CDK inhibitors (CDKN) 1A (p21cip) and CDKN2D (p19INK4D) and decreasing that of CDK2, CDK4 and CDK6. JQ1+ also inhibited serum‐stimulated migration of HPMECs. Finally, HAT activity was significantly increased in the lung of PAH patients. Conclusion Inhibition of BETs in primary HPMECs decreases inflammation and remodelling. BET proteins could be a target for future therapies for PAH. Endothelial cell inflammation and proliferation are important in the pathogenesis of pulmonary arterial hypertension (PAH). Bromodomain and extra‐terminal (BET) mimics reduced inflammation and cell proliferation in primary human pulmonary vascular endothelial cells. The enhanced histone acetyltransferase (HAT) activity in PAH suggests that BET mimics may be effective in PAH although experiments in disease cells/tissues are required.
Collapse
Affiliation(s)
- Sharon Mumby
- Vascular Biology, Imperial College London, London, UK.,Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Chao Meng
- Vascular Biology, Imperial College London, London, UK
| | - Frederic Perros
- Faculty of Medicine, South Paris University, Clamart, France.,Pulmonary Hypertension: Pathophysiology and Therapeutic Innovation, INSERM Research Unit 999, Clamart, France
| | - Marc Humbert
- Faculty of Medicine, South Paris University, Clamart, France.,Pulmonary Hypertension: Pathophysiology and Therapeutic Innovation, INSERM Research Unit 999, Clamart, France.,Pulmonary Resuscitation Respiratory and Service, National Reference Centre for Pulmonary Hypertension Severe, Assistance Publique Hôpitaux de Paris, Hôpital Antoine Béclère, Paris, France
| | - S John Wort
- Vascular Biology, Imperial College London, London, UK
| | - Ian M Adcock
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
37
|
Khan S, Ahirwar K, Jena G. Anti-fibrotic effects of valproic acid: role of HDAC inhibition and associated mechanisms. Epigenomics 2016; 8:1087-101. [PMID: 27411759 DOI: 10.2217/epi-2016-0034] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Tissue injuries and pathological insults produce oxidative stress, genetic and epigenetic alterations, which lead to an imbalance between pro- and anti-fibrotic molecules, and subsequent accumulation of extracellular matrix, thereby fibrosis. Various molecular pathways play a critical role in fibroblasts activation, which promotes the extracellular matrix production and accumulation. Recent reports highlighted that histone deacetylases (HDACs) are upregulated in various fibrotic disorders and play a central role in fibrosis, while HDAC inhibitors exert antifibrotic effects. Valproic acid is a first-line anti-epileptic drug and a proven HDAC inhibitor. This review provides the current research and novel insights on antifibrotic effects of valproic acid in various fibrotic conditions with an emphasis on the possible strategies for treatment of fibrosis.
Collapse
Affiliation(s)
- Sabbir Khan
- Facility for Risk Assessment & Intervention Studies, Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research (NIPER), Sector-67, S.A.S. Nagar, Punjab 160062, India
| | - Kailash Ahirwar
- Facility for Risk Assessment & Intervention Studies, Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research (NIPER), Sector-67, S.A.S. Nagar, Punjab 160062, India
| | - Gopabandhu Jena
- Facility for Risk Assessment & Intervention Studies, Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research (NIPER), Sector-67, S.A.S. Nagar, Punjab 160062, India
| |
Collapse
|
38
|
Yoon S, Eom GH. HDAC and HDAC Inhibitor: From Cancer to Cardiovascular Diseases. Chonnam Med J 2016; 52:1-11. [PMID: 26865995 PMCID: PMC4742605 DOI: 10.4068/cmj.2016.52.1.1] [Citation(s) in RCA: 320] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 11/27/2015] [Accepted: 12/01/2015] [Indexed: 01/15/2023] Open
Abstract
Histone deacetylases (HDACs) are epigenetic regulators that regulate the histone tail, chromatin conformation, protein-DNA interaction, and even transcription. HDACs are also post-transcriptional modifiers that regulate the protein acetylation implicated in several pathophysiologic states. HDAC inhibitors have been highlighted as a novel category of anti-cancer drugs. To date, four HDAC inhibitors, Vorinostat, Romidepsin, Panobinostat, and Belinostat, have been approved by the United States Food and Drug Administration. Principally, these HDAC inhibitors are used for hematologic cancers in clinic with less severe side effects. Clinical trials are continuously expanding to address other types of cancer and also nonmalignant diseases. HDAC inhibition also results in beneficial outcomes in various types of neurodegenerative diseases, inflammation disorders, and cardiovascular diseases. In this review, we will briefly discuss 1) the roles of HDACs in the acquisition of a cancer's phenotype and the general outcome of the HDAC inhibitors in cancer, 2) the functional relevance of HDACs in cardiovascular diseases and the possible therapeutic implications of HDAC inhibitors in cardiovascular disease.
Collapse
Affiliation(s)
- Somy Yoon
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, Korea
| | - Gwang Hyeon Eom
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
39
|
Eom GH, Kook H. Role of histone deacetylase 2 and its posttranslational modifications in cardiac hypertrophy. BMB Rep 2015; 48:131-8. [PMID: 25388210 PMCID: PMC4453031 DOI: 10.5483/bmbrep.2015.48.3.242] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Indexed: 11/20/2022] Open
Abstract
Cardiac hypertrophy is a form of global remodeling, although the initial step seems to be an adaptation to increased hemodynamic demands. The characteristics of cardiac hypertrophy include the functional reactivation of the arrested fetal gene program, where histone deacetylases (HDACs) are closely linked in the development of the process. To date, mammalian HDACs are divided into four classes: I, II, III, and IV. By structural similarities, class II HDACs are then subdivided into IIa and IIb. Among class I and II HDACs, HDAC2, 4, 5, and 9 have been reported to be involved in hypertrophic responses; HDAC4, 5, and 9 are negative regulators, whereas HDAC2 is a pro-hypertrophic mediator. The molecular function and regulation of class IIa HDACs depend largely on the phosphorylation-mediated cytosolic redistribution, whereas those of HDAC2 take place primarily in the nucleus. In response to stresses, posttranslational modification (PTM) processes, dynamic modifications after the translation of proteins, are involved in the regulation of the activities of those hypertrophy-related HDACs. In this article, we briefly review 1) the activation of HDAC2 in the development of cardiac hypertrophy and 2) the PTM of HDAC2 and its implications in the regulation of HDAC2 activity.
Collapse
Affiliation(s)
- Gwang Hyeon Eom
- Department of Pharmacology and Medical Research Center for Gene Regulation, Chonnam National University Medical School, Gwangju 501-746, Korea
| | - Hyun Kook
- Department of Pharmacology and Medical Research Center for Gene Regulation, Chonnam National University Medical School, Gwangju 501-746, Korea
| |
Collapse
|
40
|
Wang J, Saren G, Jiang H. HDAC inhibition: A novel therapeutic target for attenuating pulmonary hypertension by regulating Tregs. Int J Cardiol 2015; 198:176-7. [DOI: 10.1016/j.ijcard.2015.06.172] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 06/30/2015] [Indexed: 11/28/2022]
|
41
|
Sardana M, Moll M, Farber HW. Novel investigational therapies for treating pulmonary arterial hypertension. Expert Opin Investig Drugs 2015; 24:1571-96. [DOI: 10.1517/13543784.2015.1098616] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
42
|
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]
|
43
|
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
|
44
|
Kang SH, Seok YM, Song MJ, Lee HA, Kurz T, Kim I. Histone deacetylase inhibition attenuates cardiac hypertrophy and fibrosis through acetylation of mineralocorticoid receptor in spontaneously hypertensive rats. Mol Pharmacol 2015; 87:782-91. [PMID: 25667225 DOI: 10.1124/mol.114.096974] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Inhibition of histone deacetylases (HDACs) by valproic acid (VPA) attenuates inflammatory, hypertrophic, and fibrotic responses in the hearts of spontaneously hypertensive rats (SHRs); however, the molecular mechanism is still unclear. We hypothesized that HDAC inhibition (HDACi) attenuates cardiac hypertrophy and fibrosis through acetylation of mineralocorticoid receptor (MR) in SHRs. Seven-week-old SHRs and Wistar-Kyoto rats were treated with an HDAC class I inhibitor (0.71% w/v in drinking water; VPA) for 11 weeks. Sections of heart were visualized after trichrome stain as well as H&E stain. Histone modifications, such as acetylation (H3Ac [acetylated histone 3]) and fourth lysine trimethylation (H3K4me3) of histone 3, and recruitment of MR and RNA polymerase II (Pol II) into promoters of target genes were measured by quantitative real-time polymerase chain reaction after chromatin immunoprecipitation assay. MR acetylation was determined by Western blot with anti-acetyl-lysine antibody after immunoprecipitation with anti-MR antibody. Treatment with VPA attenuated cardiac hypertrophy and fibrosis. Although treatment with VPA increased H3Ac and H3K4me3 on promoter regions of MR target genes, expression of MR target genes as well as recruitment of MR and Pol II on promoters of target genes were decreased. Although HDACi did not affect MR expression, it increased MR acetylation. These results indicate that HDACi attenuates cardiac hypertrophy and fibrosis through acetylation of MR in spontaneously hypertensive rats.
Collapse
Affiliation(s)
- Seol-Hee Kang
- Department of Pharmacology (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Cardiovascular Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Cell and Matrix Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Brain Korea 21 Plus Kyungpook National University Biomedical Convergence Program (S.-H.K., I.K.), Department of Biomedical Science, Kyungpook National University School of Medicine (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Daegu, Republic of Korea; and Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany (T.K.)
| | - Young Mi Seok
- Department of Pharmacology (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Cardiovascular Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Cell and Matrix Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Brain Korea 21 Plus Kyungpook National University Biomedical Convergence Program (S.-H.K., I.K.), Department of Biomedical Science, Kyungpook National University School of Medicine (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Daegu, Republic of Korea; and Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany (T.K.)
| | - Min-ji Song
- Department of Pharmacology (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Cardiovascular Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Cell and Matrix Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Brain Korea 21 Plus Kyungpook National University Biomedical Convergence Program (S.-H.K., I.K.), Department of Biomedical Science, Kyungpook National University School of Medicine (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Daegu, Republic of Korea; and Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany (T.K.)
| | - Hae-Ahm Lee
- Department of Pharmacology (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Cardiovascular Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Cell and Matrix Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Brain Korea 21 Plus Kyungpook National University Biomedical Convergence Program (S.-H.K., I.K.), Department of Biomedical Science, Kyungpook National University School of Medicine (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Daegu, Republic of Korea; and Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany (T.K.)
| | - Thomas Kurz
- Department of Pharmacology (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Cardiovascular Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Cell and Matrix Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Brain Korea 21 Plus Kyungpook National University Biomedical Convergence Program (S.-H.K., I.K.), Department of Biomedical Science, Kyungpook National University School of Medicine (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Daegu, Republic of Korea; and Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany (T.K.)
| | - InKyeom Kim
- Department of Pharmacology (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Cardiovascular Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Cell and Matrix Research Institute (S.-H.K., Y.M.S., H.-A.L., I.K.), Brain Korea 21 Plus Kyungpook National University Biomedical Convergence Program (S.-H.K., I.K.), Department of Biomedical Science, Kyungpook National University School of Medicine (S.-H.K., Y.M.S., M.S., H.-A.L., I.K.), Daegu, Republic of Korea; and Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany (T.K.)
| |
Collapse
|
45
|
Lan B, Hayama E, Kawaguchi N, Furutani Y, Nakanishi T. Therapeutic efficacy of valproic acid in a combined monocrotaline and chronic hypoxia rat model of severe pulmonary hypertension. PLoS One 2015; 10:e0117211. [PMID: 25629315 PMCID: PMC4309681 DOI: 10.1371/journal.pone.0117211] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 12/21/2014] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Pulmonary hypertension (PH) is a serious disease with poor prognosis. Reports show that cells in remodeled pulmonary arteries of PH patients have similar characteristics to cancer cells, such as exuberant inflammation, increased proliferation, and decreased apoptosis. An ideal strategy for developing PH therapies is to directly target pulmonary vascular remodeling. High levels of histone deacetylase (HDAC) expression and activity are found in certain cancers, and research has shown the potential of HDAC inhibitors in repressing tumor growth via anti-inflammatory and anti-proliferative effects. To date, little is known about the effectiveness of HDAC inhibitors against pulmonary vascular remodeling in severe PH. OBJECTIVE To investigate whether class I HDAC inhibitors suppress or reverse the development of severe PH in rats. METHODS Male Sprague-Dawley rats were injected with a single, subcutaneous dose of monocrotaline (60 mg/kg), and were exposed to chronic hypoxia to induce severe PH. Valproic acid, a class I HDAC inhibitor, was administered to rats daily via gastric gavage (300 mg/kg) in a PH prevention study (during the first 3 weeks) or a PH reversal study (from 3 to 5 weeks). At the end of experiment, hemodynamic indices were measured, ventricular hypertrophy indices were calculated and vascular remodeling phenotypes were analyzed. RESULTS After 3 weeks exposure to a combined stimulation of monocrotaline and chronic hypoxia, rats exhibited a reduced body weight, elevated right ventricular systolic pressure, an increased Fulton index, right ventricle weight ratio, medial wall thickness and muscularized peripheral pulmonary arteries. These parameters for PH evaluation were exacerbated from 3 to 5 weeks. Daily administration of valproic acid therapy prevented and partially reversed the development of severe PH in rats, and decreased inflammation and proliferation in remodeled pulmonary arteries. CONCLUSION These data show that class I HDAC inhibitors may be effective for treating severe PH.
Collapse
Affiliation(s)
- Beidi Lan
- Department of Pediatric Cardiology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Emiko Hayama
- Department of Pediatric Cardiology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Nanako Kawaguchi
- Department of Pediatric Cardiology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Yoshiyuki Furutani
- Department of Pediatric Cardiology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Toshio Nakanishi
- Department of Pediatric Cardiology, Tokyo Women’s Medical University, Tokyo, Japan
- * E-mail:
| |
Collapse
|
46
|
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
|
47
|
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
|
48
|
Paulin R, Sutendra G, Gurtu V, Dromparis P, Haromy A, Provencher S, Bonnet S, Michelakis ED. A miR-208-Mef2 axis drives the decompensation of right ventricular function in pulmonary hypertension. Circ Res 2014; 116:56-69. [PMID: 25287062 DOI: 10.1161/circresaha.115.303910] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
RATIONALE Right ventricular (RV) failure is a major cause of morbidity and mortality in pulmonary hypertension, but its mechanism remains unknown. Myocyte enhancer factor 2 (Mef2) has been implicated in RV development, regulating metabolic, contractile, and angiogenic genes. Moreover, Mef2 regulates microRNAs that have emerged as important determinants of cardiac development and disease, but for which the role in RV is still unclear. OBJECTIVE We hypothesized a critical role of a Mef2-microRNAs axis in RV failure. METHODS AND RESULTS In a rat pulmonary hypertension model (monocrotaline), we studied RV free wall tissues from rats with normal, compensated, and decompensated RV hypertrophy, carefully defined based on clinically relevant parameters, including RV systolic and end-diastolic pressures, cardiac output, RV size, and morbidity. Mef2c expression was sharply increased in compensating phase of RVH tissues but was lost in decompensation phase of RVH. An unbiased screening of microRNAs in our model resulted to a short microRNA signature of decompensated RV failure, which included the myocardium-specific miR-208, which was progressively downregulated as RV failure progressed, in contrast to what is described in left ventricular failure. With mechanistic in vitro experiments using neonatal and adult RV cardiomyocytes, we showed that miR-208 inhibition, as well as tumor necrosis factor-α, activates the complex mediator of transcription 13/nuclear receptor corepressor 1 axis, which in turn promotes Mef2 inhibition, closing a self-limiting feedback loop, driving the transition from compensating phase of RVH toward decompensation phase of RVH. In our model, serum tumor necrosis factor-α levels progressively increased with time while serum miR-208 levels decreased, mirroring its levels in RV myocardium. CONCLUSIONS We describe an RV-specific mechanism for heart failure, which could potentially lead to new biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Roxane Paulin
- From the Department of Medicine, University of Alberta, Edmonton, Canada (R.P., G.S., V.G., P.D., A.H., E.D.M.); and Pulmonary Hypertension Research Group, Department of Medicine, Laval University, CRIUCPQ, Québec City, QC, Canada (S.P., S.B.)
| | - Gopinath Sutendra
- From the Department of Medicine, University of Alberta, Edmonton, Canada (R.P., G.S., V.G., P.D., A.H., E.D.M.); and Pulmonary Hypertension Research Group, Department of Medicine, Laval University, CRIUCPQ, Québec City, QC, Canada (S.P., S.B.)
| | - Vikram Gurtu
- From the Department of Medicine, University of Alberta, Edmonton, Canada (R.P., G.S., V.G., P.D., A.H., E.D.M.); and Pulmonary Hypertension Research Group, Department of Medicine, Laval University, CRIUCPQ, Québec City, QC, Canada (S.P., S.B.)
| | - Peter Dromparis
- From the Department of Medicine, University of Alberta, Edmonton, Canada (R.P., G.S., V.G., P.D., A.H., E.D.M.); and Pulmonary Hypertension Research Group, Department of Medicine, Laval University, CRIUCPQ, Québec City, QC, Canada (S.P., S.B.)
| | - Alois Haromy
- From the Department of Medicine, University of Alberta, Edmonton, Canada (R.P., G.S., V.G., P.D., A.H., E.D.M.); and Pulmonary Hypertension Research Group, Department of Medicine, Laval University, CRIUCPQ, Québec City, QC, Canada (S.P., S.B.)
| | - Steeve Provencher
- From the Department of Medicine, University of Alberta, Edmonton, Canada (R.P., G.S., V.G., P.D., A.H., E.D.M.); and Pulmonary Hypertension Research Group, Department of Medicine, Laval University, CRIUCPQ, Québec City, QC, Canada (S.P., S.B.)
| | - Sebastien Bonnet
- From the Department of Medicine, University of Alberta, Edmonton, Canada (R.P., G.S., V.G., P.D., A.H., E.D.M.); and Pulmonary Hypertension Research Group, Department of Medicine, Laval University, CRIUCPQ, Québec City, QC, Canada (S.P., S.B.)
| | - Evangelos D Michelakis
- From the Department of Medicine, University of Alberta, Edmonton, Canada (R.P., G.S., V.G., P.D., A.H., E.D.M.); and Pulmonary Hypertension Research Group, Department of Medicine, Laval University, CRIUCPQ, Québec City, QC, Canada (S.P., S.B.).
| |
Collapse
|
49
|
Eom GH, Kook H. Posttranslational modifications of histone deacetylases: Implications for cardiovascular diseases. Pharmacol Ther 2014; 143:168-80. [DOI: 10.1016/j.pharmthera.2014.02.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 02/25/2014] [Indexed: 02/08/2023]
|
50
|
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
|