1
|
Ren L, Pushpakumar S, Almarshood H, Das SK, Sen U. Epigenetic DNA Methylation and Protein Homocysteinylation: Key Players in Hypertensive Renovascular Damage. Int J Mol Sci 2024; 25:11599. [PMID: 39519150 PMCID: PMC11546175 DOI: 10.3390/ijms252111599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/24/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Hypertension has been a threat to the health of people, the mechanism of which, however, remains poorly understood. It is clinically related to loss of nephron function, glomerular sclerosis, or necrosis, resulting in renal functional declines. The mechanisms underlying hypertension's development and progression to organ damage, including hypertensive renal damage, remain to be fully elucidated. As a developing approach, epigenetics has been postulated to elucidate the phenomena that otherwise cannot be explained by genetic studies. The main epigenetic hallmarks, such as DNA methylation, histone acetylation, deacetylation, noncoding RNAs, and protein N-homocysteinylation have been linked with hypertension. In addition to contributing to endothelial dysfunction and oxidative stress, biologically active gases, including NO, CO, and H2S, are crucial regulators contributing to vascular remodeling since their complex interplay conducts homeostatic functions in the renovascular system. Importantly, epigenetic modifications also directly contribute to the pathogenesis of kidney damage via protein N-homocysteinylation. Hence, epigenetic modulation to intervene in renovascular damage is a potential therapeutic approach to treat renal disease and dysfunction. This review illustrates some of the epigenetic hallmarks and their mediators, which have the ability to diminish the injury triggered by hypertension and renal disease. In the end, we provide potential therapeutic possibilities to treat renovascular diseases in hypertension.
Collapse
Affiliation(s)
- Lu Ren
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; (L.R.)
| | - Sathnur Pushpakumar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; (L.R.)
| | - Hebah Almarshood
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; (L.R.)
| | - Swapan K. Das
- Department of Internal Medicine, Section on Endocrinology and Metabolism, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA
| | - Utpal Sen
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; (L.R.)
| |
Collapse
|
2
|
Zhang S, Xia Y, Chen W, Dong H, Cui B, Liu C, Liu Z, Wang F, Du J. Regulation and Therapeutic Application of Long non-Coding RNA in Tumor Angiogenesis. Technol Cancer Res Treat 2024; 23:15330338241273239. [PMID: 39110070 PMCID: PMC11307360 DOI: 10.1177/15330338241273239] [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: 04/20/2024] [Revised: 06/20/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Tumor growth and metastasis rely on angiogenesis. In recent years, long non-coding RNAs have been shown to play an important role in regulating tumor angiogenesis. Here, we review the multidimensional modes and relevant molecular mechanisms of long non-coding RNAs in regulating tumor angiogenesis. In addition, we summarize new strategies for tumor anti-angiogenesis therapies by targeting long non-coding RNAs. The aim of this study is to provide new diagnostic targets and treatment strategies for anti-angiogenic tumor therapy.
Collapse
Affiliation(s)
- Shuo Zhang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, P.R. China
- Department of Gynecology, Binzhou Medical University Hospital, Binzhou, P.R. China
- The First School of Clinical Medicine of Binzhou Medical University, Binzhou Medical University Hospital, Binzhou, P.R. China
| | - Yunxiu Xia
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, P.R. China
- Department of Gynecology, Binzhou Medical University Hospital, Binzhou, P.R. China
- The First School of Clinical Medicine of Binzhou Medical University, Binzhou Medical University Hospital, Binzhou, P.R. China
| | - Weiwei Chen
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, P.R. China
| | - Hongliang Dong
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, P.R. China
| | - Bingjie Cui
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, P.R. China
| | - Cuilan Liu
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, P.R. China
| | - Zhiqiang Liu
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, P.R. China
- Department of Gynecology, Binzhou Medical University Hospital, Binzhou, P.R. China
| | - Fei Wang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, P.R. China
- Medical Integration and Practice Center, Shandong University, Jinan, P.R. China
- Qilu Hospital of Shandong University, Jinan, P.R. China
| | - Jing Du
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, P.R. China
- Department of Gynecology, Binzhou Medical University Hospital, Binzhou, P.R. China
| |
Collapse
|
3
|
Meli A, McCormack A, Conte I, Chen Q, Streetley J, Rose ML, Bierings R, Hannah MJ, Molloy JE, Rosenthal PB, Carter T. Altered Storage and Function of von Willebrand Factor in Human Cardiac Microvascular Endothelial Cells Isolated from Recipient Transplant Hearts. Int J Mol Sci 2023; 24:ijms24054553. [PMID: 36901985 PMCID: PMC10003102 DOI: 10.3390/ijms24054553] [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: 01/27/2023] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
The assembly of von Willebrand factor (VWF) into ordered helical tubules within endothelial Weibel-Palade bodies (WPBs) is required for the efficient deployment of the protein at sites of vascular injury. VWF trafficking and storage are sensitive to cellular and environmental stresses that are associated with heart disease and heart failure. Altered storage of VWF manifests as a change in WPB morphology from a rod shape to a rounded shape and is associated with impaired VWF deployment during secretion. In this study, we examined the morphology, ultrastructure, molecular composition and kinetics of exocytosis of WPBs in cardiac microvascular endothelial cells isolated from explanted hearts of patients with a common form of heart failure, dilated cardiomyopathy (DCM; HCMECD), or from nominally healthy donors (controls; HCMECC). Using fluorescence microscopy, WPBs in HCMECC (n = 3 donors) showed the typical rod-shaped morphology containing VWF, P-selectin and tPA. In contrast, WPBs in primary cultures of HCMECD (n = 6 donors) were predominantly rounded in shape and lacked tissue plasminogen activator (t-PA). Ultrastructural analysis of HCMECD revealed a disordered arrangement of VWF tubules in nascent WPBs emerging from the trans-Golgi network. HCMECD WPBs still recruited Rab27A, Rab3B, Myosin-Rab Interacting Protein (MyRIP) and Synaptotagmin-like protein 4a (Slp4-a) and underwent regulated exocytosis with kinetics similar to that seen in HCMECc. However, secreted extracellular VWF strings from HCMECD were significantly shorter than for endothelial cells with rod-shaped WPBs, although VWF platelet binding was similar. Our observations suggest that VWF trafficking, storage and haemostatic potential are perturbed in HCMEC from DCM hearts.
Collapse
Affiliation(s)
- Athinoula Meli
- Transplant Immunology, Heart Science Centre, Harefield Hospital, Hill End Road, Harefield UB9 6JH, UK
| | - Ann McCormack
- Transplant Immunology, Heart Science Centre, Harefield Hospital, Hill End Road, Harefield UB9 6JH, UK
| | - Ianina Conte
- Molecular and Clinical Sciences Research Institute, St Georges University of London, London SW17 0RE, UK
| | - Qu Chen
- Structural Biology Science Technology Platform, The Francis Crick Institute, London NW1 1AT, UK
| | - James Streetley
- Structural Biology of Cells and Viruses Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Marlene L. Rose
- Transplant Immunology, Heart Science Centre, Harefield Hospital, Hill End Road, Harefield UB9 6JH, UK
| | - Ruben Bierings
- Hematology, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Matthew J. Hannah
- High Containment Microbiology, UK Health Security Agency, London NW9 5EQ, UK
| | - Justin E. Molloy
- Single Molecule Enzymology Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Peter B. Rosenthal
- Structural Biology of Cells and Viruses Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Tom Carter
- Molecular and Clinical Sciences Research Institute, St Georges University of London, London SW17 0RE, UK
- Correspondence: ; Tel.: +44-(208)-7255961
| |
Collapse
|
4
|
Raval M, Mishra S, Tiwari AK. Epigenetic regulons in Alzheimer's disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 198:185-247. [DOI: 10.1016/bs.pmbts.2023.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
|
5
|
Zemskov EA, Gross CM, Aggarwal S, Zemskova MA, Wu X, Gu C, Wang T, Tang H, Black SM. NF-κB-dependent repression of Sox18 transcription factor requires the epigenetic regulators histone deacetylases 1 and 2 in acute lung injury. Front Physiol 2022; 13:947537. [PMID: 35991176 PMCID: PMC9386230 DOI: 10.3389/fphys.2022.947537] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/11/2022] [Indexed: 11/30/2022] Open
Abstract
In acute lung injury (ALI), the NF-κB-mediated downregulation of Sox18 gene expression leads to the disruption of the pulmonary endothelial barrier. Previous studies have suggested that the action of NF-κB as a transcriptional repressor also requires the action of class I histone deacetylases (HDACs). Thus, the purpose of this study was to investigate and further delineate the mechanism of Sox18 repression during lipopolysaccharide (LPS) induced ALI. Using selective inhibitors and specific siRNA-driven depletion of HDACs 1-3 in human lung microvascular endothelial cells (HLMVEC) we were able to demonstrate a critical role for HDACs 1 and 2 in the LPS-mediated repression of Sox18 gene expression and the loss of endothelial monolayer integrity. Moreover, our data demonstrate that HDAC1 associates with a transcription-repressive complex within the NF-κB-binding site of Sox18 promoter. Further, we were able to show that the selective inhibitor of HDAC1, tacedinaline, significantly reduced the endothelial permeability and injury associated with LPS challenge in the mouse lung. Taken together, our data demonstrate, for the first time, that transcription repressors HDACs 1 and 2 are involved in pathological mechanism of ALI and can be considered as therapeutic targets.
Collapse
Affiliation(s)
- Evgeny A. Zemskov
- Center for Translational Science, Florida International University, Port St. Lucie, FL, United States
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Christine M. Gross
- Department of Medicine at Broward Health Medical Center, Fort Lauderdale, FL, United States
| | - Saurabh Aggarwal
- Department of Anesthesiology, The University of Alabama, Birmingham, AL, United States
| | - Marina A. Zemskova
- Center for Translational Science, Florida International University, Port St. Lucie, FL, United States
| | - Xiaomin Wu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, AZ, United States
| | - Chenxin Gu
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ting Wang
- Center for Translational Science, Florida International University, Port St. Lucie, FL, United States
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, United States
| | - Haiyang Tang
- Center for Translational Science, Florida International University, Port St. Lucie, FL, United States
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, United States
| | - Stephen M. Black
- Center for Translational Science, Florida International University, Port St. Lucie, FL, United States
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, United States
| |
Collapse
|
6
|
Bhamidipati T, Kumar M, Verma SS, Mohanty SK, Kacar S, Reese D, Martinez MM, Kamocka MM, Dunn KW, Sen CK, Singh K. Epigenetic basis of diabetic vasculopathy. Front Endocrinol (Lausanne) 2022; 13:989844. [PMID: 36568089 PMCID: PMC9780391 DOI: 10.3389/fendo.2022.989844] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) causes peripheral vascular disease because of which several blood-borne factors, including vital nutrients fail to reach the affected tissue. Tissue epigenome is sensitive to chronic hyperglycemia and is known to cause pathogenesis of micro- and macrovascular complications. These vascular complications of T2DM may perpetuate the onset of organ dysfunction. The burden of diabetes is primarily because of a wide range of complications of which nonhealing diabetic ulcers represent a major component. Thus, it is imperative that current research help recognize more effective methods for the diagnosis and management of early vascular injuries. This review addresses the significance of epigenetic processes such as DNA methylation and histone modifications in the evolution of macrovascular and microvascular complications of T2DM.
Collapse
Affiliation(s)
- Theja Bhamidipati
- Department of Vascular Surgery, Jefferson-Einstein Medical Center, Philadelphia, PA, United States
| | - Manishekhar Kumar
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sumit S. Verma
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sujit K. Mohanty
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sedat Kacar
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Diamond Reese
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Michelle M. Martinez
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Malgorzata M. Kamocka
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kenneth W. Dunn
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Chandan K. Sen
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN, United States
- *Correspondence: Kanhaiya Singh, ; Chandan K. Sen,
| | - Kanhaiya Singh
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN, United States
- *Correspondence: Kanhaiya Singh, ; Chandan K. Sen,
| |
Collapse
|
7
|
Subramaniam N, Nair R, Marsden PA. Epigenetic Regulation of the Vascular Endothelium by Angiogenic LncRNAs. Front Genet 2021; 12:668313. [PMID: 34512715 PMCID: PMC8427604 DOI: 10.3389/fgene.2021.668313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/17/2021] [Indexed: 12/15/2022] Open
Abstract
The functional properties of the vascular endothelium are diverse and heterogeneous between vascular beds. This is especially evident when new blood vessels develop from a pre-existing closed cardiovascular system, a process termed angiogenesis. Endothelial cells are key drivers of angiogenesis as they undergo a highly choreographed cascade of events that has both exogenous (e.g., hypoxia and VEGF) and endogenous regulatory inputs. Not surprisingly, angiogenesis is critical in health and disease. Diverse therapeutics target proteins involved in coordinating angiogenesis with varying degrees of efficacy. It is of great interest that recent work on non-coding RNAs, especially long non-coding RNAs (lncRNAs), indicates that they are also important regulators of the gene expression paradigms that underpin this cellular cascade. The protean effects of lncRNAs are dependent, in part, on their subcellular localization. For instance, lncRNAs enriched in the nucleus can act as epigenetic modifiers of gene expression in the vascular endothelium. Of great interest to genetic disease, they are undergoing rapid evolution and show extensive inter- and intra-species heterogeneity. In this review, we describe endothelial-enriched lncRNAs that have robust effects in angiogenesis.
Collapse
Affiliation(s)
- Noeline Subramaniam
- Marsden Lab, Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Marsden Lab, Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON, Canada
| | - Ranju Nair
- Marsden Lab, Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON, Canada
- Marsden Lab, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Philip A. Marsden
- Marsden Lab, Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Marsden Lab, Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON, Canada
- Marsden Lab, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
8
|
Wang TY, Chang MM, Li YSJ, Huang TC, Chien S, Wu CC. Maintenance of HDACs and H3K9me3 Prevents Arterial Flow-Induced Venous Endothelial Damage. Front Cell Dev Biol 2021; 9:642150. [PMID: 33898431 PMCID: PMC8063156 DOI: 10.3389/fcell.2021.642150] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/09/2021] [Indexed: 01/11/2023] Open
Abstract
The transition of flow microenvironments from veins to arteries in vein graft surgery induces “peel-off” of venous endothelial cells (vECs) and results in restenosis. Recently, arterial laminar shear stress (ALS) and oscillatory shear stress (OS) have been shown to affect the cell cycle and inflammation through epigenetic controls such as histone deacetylation by histone deacetylases (HDACs) and trimethylation on lysine 9 of histone 3 (H3K9me3) in arterial ECs. However, the roles of H3K9me3 and HDAC in vEC damage under ALS are not known. We hypothesized that the different responses of HDACs and H3K9me3 might cause vEC damage under the transition of venous flow to arterial flow. We found that arterial ECs showed high expression of H3K9me3 protein and were retained in the G0 phase of the cell cycle after being subjected to ALS. vECs became round under ALS with a decrease in the expression of H3K9me3, HDAC3, and HDAC5, and an increase in the expression of vascular cell adhesion molecule 1 (VCAM-1). Inhibition of HDACs activity by a specific inhibitor, phenylbutyrate, in arterial ECs caused similar ALS-induced inflammation and cell loss as observed in vECs. Activation of HDACs and H3K9me3 by ITSA-1, an HDAC activator, could prevent ALS-induced peel-off and reduced VCAM-1 expression in vECs. Moreover, shear stress modulates EC morphology by the regulation of focal adhesion kinase (FAK) expression. ITSA-1 or EGF could increase phosphorylated (p)-FAK expression in vECs under ALS. We found that perturbation of the activity of p-FAK and increase in p-FAK expression restored ALS-induced H3K9me3 expression in vECs. Hence, the abnormal mechanoresponses of H3K9me3 and HDAC in vECs after being subjected to ALS could be reversed by ITSA-1 or EGF treatment: this offers a strategy to prevent vein graft failure.
Collapse
Affiliation(s)
- Ting-Yun Wang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ming-Min Chang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Shuan Julie Li
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
| | - Tzu-Chieh Huang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States.,Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Chia-Ching Wu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan.,Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
9
|
Wurm CJ, Lindermayr C. Nitric oxide signaling in the plant nucleus: the function of nitric oxide in chromatin modulation and transcription. JOURNAL OF EXPERIMENTAL BOTANY 2021; 72:808-818. [PMID: 33128375 DOI: 10.1093/jxb/eraa404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/09/2020] [Indexed: 06/11/2023]
Abstract
Nitric oxide (NO) is involved in a vast number of physiologically important processes in plants, such as organ development, stress resistance, and immunity. Transduction of NO bioactivity is generally achieved by post-translational modification of proteins, with S-nitrosation of cysteine residues as the predominant form. While traditionally the subcellular location of the factors involved was of lesser importance, recent studies identified the connection between NO and transcriptional activity and thereby raised the question about the route of NO into the nuclear sphere. Identification of NO-affected transcription factors and chromatin-modifying histone deacetylases implicated the important role of NO signaling in the plant nucleus as a regulator of epigenetic mechanisms and gene transcription. Here, we discuss the relationship between NO and its directly regulated protein targets in the nuclear environment, focusing on S-nitrosated chromatin modulators and transcription factors.
Collapse
Affiliation(s)
- Christoph J Wurm
- Institute of Biochemical Plant Pathology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Christian Lindermayr
- Institute of Biochemical Plant Pathology, Helmholtz Zentrum München, Neuherberg, Germany
| |
Collapse
|
10
|
Ramahi A, Altorok N, Kahaleh B. Epigenetics and systemic sclerosis: An answer to disease onset and evolution? Eur J Rheumatol 2020; 7:S147-S156. [PMID: 32697935 PMCID: PMC7647676 DOI: 10.5152/eurjrheum.2020.19112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/06/2020] [Indexed: 12/22/2022] Open
Abstract
There is growing evidence that implicates epigenetic modification in the pathogenesis of systemic sclerosis (SSc). The complexity of epigenetic regulation and its dynamic nature complicate the investigation of its role in the disease. We will review the current literature for factors that link epigenetics to SSc by discussing DNA methylation, histone acetylation and methylation, and non-coding RNAs (ncRNAs), particularly microRNA changes in endothelial cells, fibroblasts (FBs), and lymphocytes. These three cell types are significantly involved in the early stages and throughout the course of the disease and are particularly vulnerable to epigenetic regulation. The pathogenesis of SSc is likely related to modifications of the epigenome by environmental signals in individuals with a specific genetic makeup. The epigenome is an attractive therapeutic target; however, successful epigenetics-based treatments require a better understanding of the molecular mechanisms controlling the epigenome and its alteration in the disease.
Collapse
Affiliation(s)
- Ahmad Ramahi
- Division of Rheumatology and Immunology, Department of Internal Medicine, University of Toledo Medical Center, Toledo, OH, USA
| | - Nezam Altorok
- Division of Rheumatology and Immunology, Department of Internal Medicine, University of Toledo Medical Center, Toledo, OH, USA
| | - Bashar Kahaleh
- Division of Rheumatology and Immunology, Department of Internal Medicine, University of Toledo Medical Center, Toledo, OH, USA
| |
Collapse
|
11
|
Ribatti D, Tamma R. Epigenetic control of tumor angiogenesis. Microcirculation 2020; 27:e12602. [PMID: 31863494 DOI: 10.1111/micc.12602] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/22/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022]
Abstract
The term "epigenetic" is used to refer to heritable alterations in chromatin that are not due to changes in DNA sequence. Different growth factors and vascular genes mediate the angiogenic process, which is regulated by epigenetic states of genes. The aim of this article is to analyze the role of epigenetic mechanisms in the control and regulation of tumor angiogenetic processes. The reversibility of epigenetic events in contrast to genetic aberrations makes them potentially suitable for therapeutic intervention. In this context, DNA methyltransferase (DNMT) and HDAC inhibitors indirectly-via the tumor cells-exhibit angiostatic effects in vivo, and inhibition of miRNAs can contribute to the development of novel anti-angiogenesis therapies.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| |
Collapse
|
12
|
Gomes AM, Pinto TS, Costa Fernandes CJ, Silva RA, Zambuzzi WF. Wortmannin targeting phosphatidylinositol 3‐kinase suppresses angiogenic factors in shear‐stressed endothelial cells. J Cell Physiol 2019; 235:5256-5269. [DOI: 10.1002/jcp.29412] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 12/05/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Anderson M. Gomes
- Bioassays and Cell Dynamics Laboratory, Department of Chemistry and BiochemistryBioscience Institute UNESP Botucatu Sao Paulo Brazil
| | - Thais S. Pinto
- Bioassays and Cell Dynamics Laboratory, Department of Chemistry and BiochemistryBioscience Institute UNESP Botucatu Sao Paulo Brazil
| | - Célio J. Costa Fernandes
- Bioassays and Cell Dynamics Laboratory, Department of Chemistry and BiochemistryBioscience Institute UNESP Botucatu Sao Paulo Brazil
| | - Rodrigo A. Silva
- Bioassays and Cell Dynamics Laboratory, Department of Chemistry and BiochemistryBioscience Institute UNESP Botucatu Sao Paulo Brazil
- Department of Biology, Dental SchoolUniversity of Taubaté Taubaté São Paulo Brazil
| | - Willian F. Zambuzzi
- Bioassays and Cell Dynamics Laboratory, Department of Chemistry and BiochemistryBioscience Institute UNESP Botucatu Sao Paulo Brazil
| |
Collapse
|
13
|
Alese OO, Mabandla MV. Transgenerational deep sequencing revealed hypermethylation of hippocampal mGluR1 gene with altered mRNA expression of mGluR5 and mGluR3 associated with behavioral changes in Sprague Dawley rats with history of prolonged febrile seizure. PLoS One 2019; 14:e0225034. [PMID: 31710636 PMCID: PMC6844483 DOI: 10.1371/journal.pone.0225034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/27/2019] [Indexed: 02/04/2023] Open
Abstract
The impact of febrile seizure has been shown to transcend immediate generation with the alteration of glutamatergic pathway being implicated. However, transgenerational effects of this neurological disorder particularly prolonged febrile seizure (PFS) on neurobehavioral study and methylation profile is unknown. We therefore hypothesized that transgenerational impact of prolonged febrile seizure is dependent on methylation of hippocampal mGluR1 gene. Prolonged febrile seizure was induced on post-natal day (PND) 14, by injecting lipopolysaccharide (LPS; 217μg/kg ip) and kainic acid (KA; 1.83 mg/kg ip). Sucrose preference test (SPT) and Forced swim test (FST) were carried out in the first generation (F0) of animals at PND37 and PND60. The F0 rats were decapitated at PND 14, 37 and 60 which corresponded to childhood, adolescent and adulthood respectively and their hippocampal tissue collected. The second generation (F1) rats were obtained by mating F0 generation at PND 60 across different groups, F1 rats were subjected to SPT and FST test on PND 37 only. Decapitation of F1rats and collection of hippocampal tissues were done on PND 14 and 37. Assessment of mGluR5 and mGluR3 mRNA was done with PCR while mGluR1 methylation profile was assessed with the Quantitative MassARRAY analysis. Results showed that PFS significantly leads to decreased sucrose consumption in the SPT and increased immobility time in the FST in both generations of rats. It also leads to significant decrease in mGluR5 mRNA expression with a resultant increased expression of mGluR3 mRNA expression and hypermethylation of mGluR1 gene across both generations of rats. This study suggested that PFS led to behavioral changes which could be transmitted on to the next generation in rats.
Collapse
MESH Headings
- Animals
- Base Sequence
- Behavior, Animal
- DNA Methylation/genetics
- High-Throughput Nucleotide Sequencing
- Hippocampus/metabolism
- Immobilization
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Receptor, Metabotropic Glutamate 5/genetics
- Receptor, Metabotropic Glutamate 5/metabolism
- Receptors, Metabotropic Glutamate/genetics
- Receptors, Metabotropic Glutamate/metabolism
- Seizures, Febrile/genetics
- Sucrose
- Swimming
Collapse
Affiliation(s)
- Oluwole Ojo Alese
- Department of Human Physiology, College of Health Sciences, University of Kwazulu-Natal, Durban, South Africa
| | - Musa V. Mabandla
- Department of Human Physiology, College of Health Sciences, University of Kwazulu-Natal, Durban, South Africa
| |
Collapse
|
14
|
Ku KH, Subramaniam N, Marsden PA. Epigenetic Determinants of Flow-Mediated Vascular Endothelial Gene Expression. Hypertension 2019; 74:467-476. [PMID: 31352815 DOI: 10.1161/hypertensionaha.119.13342] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Kyung Ha Ku
- From the Department of Laboratory Medicine and Pathobiology (K.H.K., P.A.M.), University of Toronto, Ontario, Canada.,Keenan Research Center for Biomedical Science, Li Ka Shing Knowledge Institute, St Michael's Hospital (K.H.K., N.S., P.A.M.) Toronto, Ontario, Canada
| | - Noeline Subramaniam
- Institute of Medical Science (N.S., P.A.M.), University of Toronto, Ontario, Canada.,Keenan Research Center for Biomedical Science, Li Ka Shing Knowledge Institute, St Michael's Hospital (K.H.K., N.S., P.A.M.) Toronto, Ontario, Canada
| | - Philip A Marsden
- From the Department of Laboratory Medicine and Pathobiology (K.H.K., P.A.M.), University of Toronto, Ontario, Canada.,Institute of Medical Science (N.S., P.A.M.), University of Toronto, Ontario, Canada.,Department of Medicine (P.A.M.), University of Toronto, Ontario, Canada.,Keenan Research Center for Biomedical Science, Li Ka Shing Knowledge Institute, St Michael's Hospital (K.H.K., N.S., P.A.M.) Toronto, Ontario, Canada
| |
Collapse
|
15
|
Xuan C, Li H, Li LL, Tian QW, Wang Q, Zhang BB, Guo JJ, He GW, Lun LM. Screening and Identification of Pregnancy Zone Protein and Leucine-Rich Alpha-2-Glycoprotein as Potential Serum Biomarkers for Early-Onset Myocardial Infarction using Protein Profile Analysis. Proteomics Clin Appl 2019; 13:e1800079. [PMID: 30411527 DOI: 10.1002/prca.201800079] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 10/19/2018] [Indexed: 12/29/2022]
Abstract
PURPOSE The present study aims to discover novel serum biomarkers of early-onset myocardial infarction (MI) using proteomic analysis. EXPERIMENTAL DESIGN In the first stage, the iTRAQ-coupled LC-MS/MS technique is utilized to investigate protein profiles of patients with early-onset MI. In the second stage, these candidate proteins are validated using ELISA. RESULTS A total of 538 proteins are quantified, with pregnancy zone protein (PZP), leucine-rich α-2-glycoprotein (LRG) and Apolipoprotein C-I (Apo C-I) being upregulated and Apolipoprotein A-I (Apo A-I) and Apolipoprotein A-IV (Apo A-IV) downregulated in early-onset MI patients. Results from the validation stage demonstrate that the serum concentrations of PZP and LRG are significantly increased in the early-onset MI group. The correlation between the concentrations of C-reactive protein (CRP) and the two candidate biomarkers is positive. Area under the curve values used to diagnose early-onset MI for LRG and PZP are 0.939 and 0.874, respectively. CONCLUSIONS AND CLINICAL RELEVANCE Five differential serum proteins are identified in early-onset MI using proteomic analysis. Lipoprotein-related biomarkers further demonstrate the close relationship between lipid metabolism and the disease. Inflammation-associated LRG and PZP may be novel biomarkers of the disease. In addition, changes in these proteins may partly reveal the possible mechanisms in the pathogenesis and pathophysiology of early-onset MI.
Collapse
Affiliation(s)
- Chao Xuan
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hui Li
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Le-Le Li
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qing-Wu Tian
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qing Wang
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bei-Bei Zhang
- Department of Molecular Microbiology, Oslo University Hospital, Oslo, Norway
| | - Jun-Jie Guo
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guo-Wei He
- Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital,, Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Surgery, Oregon Health and Science University, Portland, Oregon
| | - Li-Min Lun
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
16
|
Hepp P, Hutter S, Knabl J, Hofmann S, Kuhn C, Mahner S, Jeschke U. Histone H3 lysine 9 acetylation is downregulated in GDM Placentas and Calcitriol supplementation enhanced this effect. Int J Mol Sci 2018; 19:ijms19124061. [PMID: 30558244 PMCID: PMC6321349 DOI: 10.3390/ijms19124061] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/07/2018] [Accepted: 12/11/2018] [Indexed: 12/12/2022] Open
Abstract
Despite the ever-rising incidence of Gestational Diabetes Mellitus (GDM) and its implications for long-term health of mothers and offspring, the underlying molecular mechanisms remain to be elucidated. To contribute to this, the present study's objectives are to conduct a sex-specific analysis of active histone modifications in placentas affected by GDM and to investigate the effect of calcitriol on trophoblast cell's transcriptional status. The expression of Histone H3 lysine 9 acetylation (H3K9ac) and Histone H3 lysine 4 trimethylation (H3K4me3) was evaluated in 40 control and 40 GDM (20 male and 20 female each) placentas using immunohistochemistry and immunofluorescence. The choriocarcinoma cell line BeWo and primary human villous trophoblast cells were treated with calcitriol (48 h). Thereafter, western blots were used to quantify concentrations of H3K9ac and the transcription factor FOXO1. H3K9ac expression was downregulated in GDM placentas, while H3K4me3 expression was not significantly different. Cell culture experiments showed a slight downregulation of H3K9ac after calcitriol stimulation at the highest concentration. FOXO1 expression showed a dose-dependent increase. Our data supports previous research suggesting that epigenetic dysregulations play a key role in gestational diabetes mellitus. Insufficient transcriptional activity may be part of its pathophysiology and this cannot be rescued by calcitriol.
Collapse
Affiliation(s)
- Paula Hepp
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Maistraße 11, 80337 Munich, Germany.
| | - Stefan Hutter
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Maistraße 11, 80337 Munich, Germany.
| | - Julia Knabl
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Maistraße 11, 80337 Munich, Germany.
- Department of Obstetrics, Klinik Hallerwiese, 90419 Nürnberg, Germany.
| | - Simone Hofmann
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Maistraße 11, 80337 Munich, Germany.
| | - Christina Kuhn
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Maistraße 11, 80337 Munich, Germany.
| | - Sven Mahner
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Maistraße 11, 80337 Munich, Germany.
| | - Udo Jeschke
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Maistraße 11, 80337 Munich, Germany.
| |
Collapse
|
17
|
Tanaka T, Izawa K, Maniwa Y, Okamura M, Okada A, Yamaguchi T, Shirakura K, Maekawa N, Matsui H, Ishimoto K, Hino N, Nakagawa O, Aird WC, Mizuguchi H, Kawabata K, Doi T, Okada Y. ETV2-TET1/TET2 Complexes Induce Endothelial Cell-Specific Robo4 Expression via Promoter Demethylation. Sci Rep 2018; 8:5653. [PMID: 29618782 PMCID: PMC5884809 DOI: 10.1038/s41598-018-23937-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 03/23/2018] [Indexed: 01/18/2023] Open
Abstract
Although transcription factors regulating endothelial cell (EC)-specific gene expression have been identified, it is not known how those factors induce EC-specificity. We previously reported that DNA hypomethylation of the proximal promoter elicits EC-specific expression of Roundabout4 (Robo4). However, the mechanisms establishing EC-specific hypomethylation of the Robo4 promoter remain unknown. In this study, we demonstrated that the hypermethylated Robo4 proximal promoter is demethylated as human iPS cells differentiate into endothelial cells. Reporter assays demonstrated that ETV2, an ETS family transcription factor, bound to ETS motifs in the proximal promoter and activated Robo4 expression. Immunoprecipitation demonstrated direct interaction between ETV2 and methylcytosine-converting enzymes TET1 and TET2. Adenoviral expression of ETV2-TET1/TET2 complexes demethylated the Robo4 promoter and induced Robo4 expression in non-ECs. In summary, we propose a novel regulatory model of EC-specific gene expression via promoter demethylation induced by ETV2-TET1/TET2 complexes during endothelial differentiation.
Collapse
Affiliation(s)
- Toru Tanaka
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita City, Osaka, 565-0871, Japan
| | - Kohei Izawa
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita City, Osaka, 565-0871, Japan
| | - Yusuke Maniwa
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita City, Osaka, 565-0871, Japan
| | - Maki Okamura
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita City, Osaka, 565-0871, Japan
| | - Atsumasa Okada
- Laboratory of Stem Cell Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki City, Osaka, 567-0085, Japan
| | - Tomoko Yamaguchi
- Laboratory of Stem Cell Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki City, Osaka, 567-0085, Japan
| | - Keisuke Shirakura
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita City, Osaka, 565-0871, Japan
| | - Naoki Maekawa
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita City, Osaka, 565-0871, Japan
| | - Hayato Matsui
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita City, Osaka, 565-0871, Japan
| | - Kenji Ishimoto
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita City, Osaka, 565-0871, Japan
| | - Nobumasa Hino
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita City, Osaka, 565-0871, Japan
| | - Osamu Nakagawa
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita City, Osaka, 565-8565, Japan
| | - William C Aird
- Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Hiroyuki Mizuguchi
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita City, Osaka, 565-0871, Japan
| | - Kenji Kawabata
- Laboratory of Stem Cell Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki City, Osaka, 567-0085, Japan
| | - Takefumi Doi
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita City, Osaka, 565-0871, Japan
| | - Yoshiaki Okada
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita City, Osaka, 565-0871, Japan.
| |
Collapse
|
18
|
Wang Y, Yan L, Zhang Z, Prado E, Fu L, Xu X, Du L. Epigenetic Regulation and Its Therapeutic Potential in Pulmonary Hypertension. Front Pharmacol 2018; 9:241. [PMID: 29615911 PMCID: PMC5870037 DOI: 10.3389/fphar.2018.00241] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 03/05/2018] [Indexed: 12/21/2022] Open
Abstract
Recent advances in epigenetics have made a tremendous impact on our knowledge of biological phenomena and the environmental stressors on complex diseases. Understanding the mechanism of epigenetic reprogramming during the occurrence of pulmonary hypertension (PH) is important for advanced studies and clinical therapy. In this article, we review the discovery of novel epigenetic mechanisms associated with PH including DNA methylation, histone modification, and noncoding RNA interference. In addition, we highlight the role of epigenetic mechanisms in adult PAH resulting from undesirable perinatal environments-Extrauterine growth restriction (EUGR) and Intrauterine growth retardation (IUGR). Lastly, we give a comprehensive summary for the remaining challenges and discuss future methods of epigenetic targeted therapy for pulmonary hypertension.
Collapse
Affiliation(s)
- Yu Wang
- Department of Pediatrics, Children's Hospital of Zhejiang University, Hangzhou, China
| | - Lingling Yan
- Department of Pediatrics, Children's Hospital of Zhejiang University, Hangzhou, China
| | - Ziming Zhang
- Department of Pediatrics, Children's Hospital of Zhejiang University, Hangzhou, China
| | - Eric Prado
- Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Linchen Fu
- Department of Pediatrics, Children's Hospital of Zhejiang University, Hangzhou, China
| | - Xuefeng Xu
- Department of Pediatrics, Children's Hospital of Zhejiang University, Hangzhou, China
| | - Lizhong Du
- Department of Pediatrics, Children's Hospital of Zhejiang University, Hangzhou, China
| |
Collapse
|
19
|
Wong WT, Matrone G, Tian X, Tomoiaga SA, Au KF, Meng S, Yamazoe S, Sieveking D, Chen K, Burns DM, Chen JK, Blau HM, Cooke JP. Discovery of novel determinants of endothelial lineage using chimeric heterokaryons. eLife 2017; 6. [PMID: 28323620 PMCID: PMC5391207 DOI: 10.7554/elife.23588] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 03/17/2017] [Indexed: 12/29/2022] Open
Abstract
We wish to identify determinants of endothelial lineage. Murine embryonic stem cells (mESC) were fused with human endothelial cells in stable, non-dividing, heterokaryons. Using RNA-seq, it is possible to discriminate between human and mouse transcripts in these chimeric heterokaryons. We observed a temporal pattern of gene expression in the ESCs of the heterokaryons that recapitulated ontogeny, with early mesodermal factors being expressed before mature endothelial genes. A set of transcriptional factors not known to be involved in endothelial development was upregulated, one of which was POU class 3 homeobox 2 (Pou3f2). We confirmed its importance in differentiation to endothelial lineage via loss- and gain-of-function (LOF and GOF). Its role in vascular development was validated in zebrafish embryos using morpholino oligonucleotides. These studies provide a systematic and mechanistic approach for identifying key regulators in directed differentiation of pluripotent stem cells to somatic cell lineages. DOI:http://dx.doi.org/10.7554/eLife.23588.001 Endothelial cells form the inner surface of blood vessels, acting like a non-stick coating. In addition to making substances that keep blood from sticking to the vessel wall, endothelial cells generate compounds that relax the vessel, and prevent it from thickening. Endothelial cells also form capillaries, the smallest vessels that provide oxygen and nutrients for all tissues. A regenerating organ, or a bioengineered tissue, requires a system of capillaries and other microvessels. Thus, regenerative medicine could benefit from a knowledge of how to generate endothelial cells from pluripotent stem cells – cells that can “differentiate” to form almost any type of cell in the body. Wong, Matrone et al. have now used a cell fusion model (named heterokaryon) to track the changes in gene expression that occur as a pluripotent stem cell differentiates to ultimately become an endothelial cell. In this model, mouse embryonic stem cells (ESCs) are fused to human endothelial cells. Over time the human endothelial cells drive gene expression in the ESCs toward that of endothelial cells. Wong, Matrone et al. discovered changes in gene expression in many genes that have not previously been described as involved in the differentiation of endothelial cells. When one of these genes – named Pou3f2 – was inactivated in ESCs, they could not be differentiated into endothelial cells. The absence of Pou3f2 also drastically impaired how blood vessels developed in zebrafish embryos. Thus the heterokaryon model can generate important information regarding the dynamic changes in gene expression that occur as a pluripotent cell differentiates to become an endothelial cell. This model may also be useful for discovering other genes that control the differentiation of other cell types. DOI:http://dx.doi.org/10.7554/eLife.23588.002
Collapse
Affiliation(s)
- Wing Tak Wong
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, United States
| | - Gianfranco Matrone
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, United States
| | - XiaoYu Tian
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, United States
| | - Simion Alin Tomoiaga
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, United States
| | - Kin Fai Au
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, United States.,Department of Internal Medicine, University of Iowa, Iowa City, United States
| | - Shu Meng
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, United States
| | - Sayumi Yamazoe
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, United States
| | - Daniel Sieveking
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, United States
| | - Kaifu Chen
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, United States
| | - David M Burns
- Baxter Laboratory for Stem Cell Biology, Stanford University School of Medicine, Stanford, United States
| | - James K Chen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, United States
| | - Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Stanford University School of Medicine, Stanford, United States
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, United States
| |
Collapse
|
20
|
Min J, Weitian Z, Peng C, Yan P, Bo Z, Yan W, Yun B, Xukai W. Correlation between insulin-induced estrogen receptor methylation and atherosclerosis. Cardiovasc Diabetol 2016; 15:156. [PMID: 27832775 PMCID: PMC5105242 DOI: 10.1186/s12933-016-0471-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/26/2016] [Indexed: 01/30/2023] Open
Abstract
Background Hyperinsulinemia and insulin resistance have been recently recognized as an important cause of atherosclerosis. Clinical studies have also found that expression of the estrogen receptor is closely related to the incidence of atherosclerosis. This study investigate the effects of insulin and estrogen receptor α (ER-α) in atherosclerosis. Methods Double knockout ApoE/Lepr mice were given intraperitoneal injections of insulin, and their aortae were harvested for hematoxylin-eosin staining and immunohistochemical analysis. In addition, vascular smooth muscle cells (VSMCs) were treated with insulin or infected with a lentivirus encoding exogenous ER-α, and changes in gene expression were detected by real-time polymerase chain reaction and western blotting. The methylation levels of the ER-α gene were tested using bisulfite sequencing PCR, and flow cytometry and EdU assay were used to measure VSMCs proliferation. Results Our results showed that insulin can induce the formation of atherosclerosis. Gene expression analysis revealed that insulin promotes the expression of DNA methyltransferases and inhibits ER-α expression, while 5-aza-2′-deoxycytidine can inhibit this effect of insulin. Bisulfite sequencing PCR analysis showed that methylation of the ER-α second exon region increased in VSMCs treated with insulin. The results also showed that ER-α can inhibit VSMCs proliferation. Conclusions Our data suggest that insulin promotes the expression of DNA methyltransferases, induces methylation of ER-α second exon region and decreases the expression of ER-α, thereby interfering with estrogen regulation of VSMCs proliferation, resulting in atherosclerosis. Electronic supplementary material The online version of this article (doi:10.1186/s12933-016-0471-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jia Min
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Zhong Weitian
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Cai Peng
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Peng Yan
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Zhang Bo
- Department of Medical Genetics, College of Basic Medicine, Third Military, Medical University, Chongqing, 400038, China
| | - Wang Yan
- Department of Medical Genetics, College of Basic Medicine, Third Military, Medical University, Chongqing, 400038, China
| | - Bai Yun
- Department of Medical Genetics, College of Basic Medicine, Third Military, Medical University, Chongqing, 400038, China.
| | - Wang Xukai
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| |
Collapse
|
21
|
Recchioni R, Marcheselli F, Antonicelli R, Lazzarini R, Mensà E, Testa R, Procopio AD, Olivieri F. Physical activity and progenitor cell-mediated endothelial repair in chronic heart failure: Is there a role for epigenetics? Mech Ageing Dev 2016; 159:71-80. [DOI: 10.1016/j.mad.2016.03.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/17/2016] [Accepted: 03/21/2016] [Indexed: 02/09/2023]
|
22
|
Forte A, Galderisi U, Cipollaro M, De Feo M, Della Corte A. Epigenetic regulation of TGF-β1 signalling in dilative aortopathy of the thoracic ascending aorta. Clin Sci (Lond) 2016; 130:1389-1405. [PMID: 27389586 DOI: 10.1042/cs20160222] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 04/11/2016] [Indexed: 01/21/2023]
Abstract
The term 'epigenetics' refers to heritable, reversible DNA or histone modifications that affect gene expression without modifying the DNA sequence. Epigenetic modulation of gene expression also includes the RNA interference mechanism. Epigenetic regulation of gene expression is fundamental during development and throughout life, also playing a central role in disease progression. The transforming growth factor β1 (TGF-β1) and its downstream effectors are key players in tissue repair and fibrosis, extracellular matrix remodelling, inflammation, cell proliferation and migration. TGF-β1 can also induce cell switch in epithelial-to-mesenchymal transition, leading to myofibroblast transdifferentiation. Cellular pathways triggered by TGF-β1 in thoracic ascending aorta dilatation have relevant roles to play in remodelling of the vascular wall by virtue of their association with monogenic syndromes that implicate an aortic aneurysm, including Loeys-Dietz and Marfan's syndromes. Several studies and reviews have focused on the progression of aneurysms in the abdominal aorta, but research efforts are now increasingly being focused on pathogenic mechanisms of thoracic ascending aorta dilatation. The present review summarizes the most recent findings concerning the epigenetic regulation of effectors of TGF-β1 pathways, triggered by sporadic dilative aortopathy of the thoracic ascending aorta in the presence of a tricuspid or bicuspid aortic valve, a congenital malformation occurring in 0.5-2% of the general population. A more in-depth comprehension of the epigenetic alterations associated with TGF-β1 canonical and non-canonical pathways in dilatation of the ascending aorta could be helpful to clarify its pathogenesis, identify early potential biomarkers of disease, and, possibly, develop preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Amalia Forte
- Experimental Medicine, Second University of Naples, Naples, Italy
| | | | | | - Marisa De Feo
- Cardiothoracic Sciences, Second University of Naples, Naples, Italy
| | | |
Collapse
|
23
|
Man HSJ, Yan MS, Lee JJ, Marsden PA. Epigenetic determinants of cardiovascular gene expression: vascular endothelium. Epigenomics 2016; 8:959-79. [PMID: 27381277 DOI: 10.2217/epi-2016-0012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The modern landscape of gene regulation involves interacting factors that ultimately lead to gene activation or repression. Epigenetic mechanisms provide a perspective of cellular phenotype as dynamically regulated and responsive to input. This perspective is supported by the generation of induced pluripotent stem cells from fully differentiated cell types. In vascular endothelial cells, evidence suggests that epigenetic mechanisms play a major role in the expression of endothelial cell-specific genes such as the endothelial nitric oxide synthase (NOS3/eNOS). These mechanisms are also important for eNOS expression in response to environmental stimuli such as hypoxia and shear stress. A newer paradigm in epigenetics, long noncoding RNAs offer a link between genetic variation, epigenetic regulation and disease. While the understanding of epigenetic mechanisms is early in its course, it is becoming clear that approaches to understanding the interaction of these factors and their inputs will be necessary to improve outcomes in cardiovascular disease.
Collapse
Affiliation(s)
- Hon-Sum Jeffrey Man
- Department of Medicine, Keenan Research Centre, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Departments of Respirology & Critical Care, University Health Network & Mt Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Matthew S Yan
- Department of Medicine, Keenan Research Centre, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - John Jy Lee
- Department of Medicine, Keenan Research Centre, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Philip A Marsden
- Department of Medicine, Keenan Research Centre, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Nephrology, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
24
|
Han L, Liu Y, Duan S, Perry B, Li W, He Y. DNA methylation and hypertension: emerging evidence and challenges. Brief Funct Genomics 2016; 15:460-469. [PMID: 27142121 DOI: 10.1093/bfgp/elw014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Hypertension is a multifactorial disease influenced by an interaction of environmental and genetic factors. The exact molecular mechanism of hypertension remains unknown. Aberrant DNA methylation is the most well-defined epigenetic modification that regulates gene transcription. However, studies on the association between DNA methylation and hypertension are still in their infancy. This review summarizes the latest evidence and challenges regarding the role of DNA methylation on hypertension.
Collapse
|
25
|
Xiao FH, Kong QP, Perry B, He YH. Progress on the role of DNA methylation in aging and longevity. Brief Funct Genomics 2016; 15:454-459. [PMID: 27032421 DOI: 10.1093/bfgp/elw009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aging is a major risk factor for individuals' health problems. Moreover, environmental signals have a widespread influence on the aging process. Epigenetic modification, e.g. DNA methylation, represents a link between genetic and environmental signals via the regulation of gene transcription. An abundance of literature indicates that aberrant epigenetic change occurs throughout the aging process at both the cellular and the organismal level. In particular, DNA methylation presents globally decreasing and site-specific increasing in aging. In this review, we focus on the crucial roles of DNA methylation in aging and age-related disease and highlight the great potential of DNA methylation as a therapeutic target in preventing age-related diseases and promoting healthy longevity.
Collapse
|
26
|
Age-Dependent Demethylation of Sod2 Promoter in the Mouse Femoral Artery. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:8627384. [PMID: 26989455 PMCID: PMC4771915 DOI: 10.1155/2016/8627384] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 01/13/2016] [Indexed: 11/17/2022]
Abstract
We studied the age-dependent regulation of the expression of the antioxidant enzyme manganese superoxide dismutase (MnSOD encoded by Sod2) through promoter methylation. C57Bl/6 mice were either (i) sedentary (SED), (ii) treated with the antioxidant catechin (CAT), or (iii) voluntarily exercised (EX) from weaning (1-month old; mo) to 9 mo. Then, all mice aged sedentarily and were untreated until 12 mo. Sod2 promoter methylation was similar in all groups in 9 mo but decreased (p < 0.05) in 12 mo SED mice only, which was associated with an increased (p < 0.05) transcriptional activity in vitro. At all ages, femoral artery endothelial function was maintained; this was due to an increased (p < 0.05) contribution of eNOS-derived NO in 12 mo SED mice only. CAT and EX prevented these changes in age-related endothelial function. Thus, a ROS-dependent epigenetic positive regulation of Sod2 gene expression likely represents a defense mechanism prolonging eNOS function in aging mouse femoral arteries.
Collapse
|
27
|
Greißel A, Culmes M, Burgkart R, Zimmermann A, Eckstein HH, Zernecke A, Pelisek J. Histone acetylation and methylation significantly change with severity of atherosclerosis in human carotid plaques. Cardiovasc Pathol 2015; 25:79-86. [PMID: 26764138 DOI: 10.1016/j.carpath.2015.11.001] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 10/30/2015] [Accepted: 11/02/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The aim of the study was to analyze histone acetylation, methylation, and the expression of their corresponding transferases in atherosclerotic plaques of patients with carotid artery stenosis. METHODS Atherosclerotic tissue from our biobank (n=80) was divided into various segments covering all plaque stages and classified according to the American Heart Association. The plaques were assigned to early (types I-III) or advanced (types V-VII) stage group of atherosclerosis. Ten healthy carotid arteries from transplant donors served as controls. The expression of histone acetyltransferases (GNAT group: GCN5L, P300/CBP group: P300, MYST group: MYST1 and MYST2) and histone methyltransferases (H3K4: MLL2/4, SET7/9, and hSET1A; H3K9: SUV39H1, SUV39H2, ESET/SETDB1, and EHMT1; H3K27: EZH2 and G9a) was analyzed by SYBR-green-based real-time polymerase chain reaction. Histone acetylation/methylation in the cells within atherosclerotic plaques was determined by immunohistochemistry. RESULTS Increased histone acetylation was observed on H3K9 and H3K27 in smooth muscle cells (SMCs) in advanced atherosclerotic lesions compared to healthy vessels (P=.002 and .034). H3K9 acetylation in SMCs and macrophages was associated with plaque severity of atherosclerosis (P=.048 and <.001). Expression of GCN5L and MYST1 also correlated with the severity of atherosclerosis (P<.001). Methylation of H3K9 and H3K27 was significantly reduced in atherosclerotic plaques in SMCs and inflammatory cells (P<.001 and .026). Methylation on H3K4 was significantly associated with the severity of atherosclerosis. Expression of methyltransferase MLL2/4 was increased in advanced stages of atherosclerosis (P<.001). CONCLUSIONS Histone acetylation and methylation seem to play a decisive role in atherosclerosis, showing significant differences between healthy vessels and vessels at different stages of atherosclerosis.
Collapse
Affiliation(s)
- Anna Greißel
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar der Technischen Universitaet Muenchen, Germany
| | - Mihaela Culmes
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar der Technischen Universitaet Muenchen, Germany
| | - Rainer Burgkart
- Clinic of Orthopedics, Klinikum rechts der Isar der Technischen Universitaet Muenchen, Germany
| | - Alexander Zimmermann
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar der Technischen Universitaet Muenchen, Germany
| | - Hans-Henning Eckstein
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar der Technischen Universitaet Muenchen, Germany
| | - Alma Zernecke
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar der Technischen Universitaet Muenchen, Germany; Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital, Wuerzburg, Germany
| | - Jaroslav Pelisek
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar der Technischen Universitaet Muenchen, Germany.
| |
Collapse
|
28
|
Yan MS, Marsden PA. Epigenetics in the Vascular Endothelium: Looking From a Different Perspective in the Epigenomics Era. Arterioscler Thromb Vasc Biol 2015; 35:2297-306. [PMID: 26404488 DOI: 10.1161/atvbaha.115.305043] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 09/14/2015] [Indexed: 01/11/2023]
Abstract
Cardiovascular diseases are commonly thought to be complex, non-Mendelian diseases that are influenced by genetic and environmental factors. A growing body of evidence suggests that epigenetic pathways play a key role in vascular biology and might be involved in defining and transducing cardiovascular disease inheritability. In this review, we argue the importance of epigenetics in vascular biology, especially from the perspective of endothelial cell phenotype. We highlight and discuss the role of epigenetic modifications across the transcriptional unit of protein-coding genes, especially the role of intragenic chromatin modifications, which are underappreciated and not well characterized in the current era of genome-wide studies. Importantly, we describe the practical application of epigenetics in cardiovascular disease therapeutics.
Collapse
Affiliation(s)
- Matthew S Yan
- From the Department of Medical Biophysics (M.S.Y., P.A.M.) and Department of Medicine, Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital (M.S.Y., P.A.M.), University of Toronto, Toronto, Ontario, Canada
| | - Philip A Marsden
- From the Department of Medical Biophysics (M.S.Y., P.A.M.) and Department of Medicine, Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital (M.S.Y., P.A.M.), University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
29
|
Martinez SR, Gay MS, Zhang L. Epigenetic mechanisms in heart development and disease. Drug Discov Today 2015; 20:799-811. [PMID: 25572405 PMCID: PMC4492921 DOI: 10.1016/j.drudis.2014.12.018] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/05/2014] [Accepted: 12/29/2014] [Indexed: 12/11/2022]
Abstract
Suboptimal intrauterine development has been linked to predisposition to cardiovascular disease in adulthood, a concept termed 'developmental origins of health and disease'. Although the exact mechanisms underlying this developmental programming are unknown, a growing body of evidence supports the involvement of epigenetic regulation. Epigenetic mechanisms such as DNA methylation, histone modifications and micro-RNA confer added levels of gene regulation without altering DNA sequences. These modifications are relatively stable signals, offering possible insight into the mechanisms underlying developmental origins of health and disease. This review will discuss the role of epigenetic mechanisms in heart development as well as aberrant epigenetic regulation contributing to cardiovascular disease. Additionally, we will address recent advances targeting epigenetic mechanisms as potential therapeutic approaches to cardiovascular disease.
Collapse
Affiliation(s)
- Shannalee R Martinez
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Maresha S Gay
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Lubo Zhang
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| |
Collapse
|
30
|
Fraineau S, Palii CG, Allan DS, Brand M. Epigenetic regulation of endothelial-cell-mediated vascular repair. FEBS J 2015; 282:1605-29. [PMID: 25546332 DOI: 10.1111/febs.13183] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/17/2014] [Accepted: 12/19/2014] [Indexed: 01/16/2023]
Abstract
Maintenance of vascular integrity is essential for the prevention of vascular disease and for recovery following cardiovascular, cerebrovascular and peripheral vascular events including limb ischemia, heart attack and stroke. Endothelial stem/progenitor cells have recently gained considerable interest due to their potential use in stem cell therapies to mediate revascularization after ischemic injury. Therefore, there is an urgent need to understand fundamental mechanisms regulating vascular repair in specific cell types to develop new beneficial therapeutic interventions. In this review, we highlight recent studies demonstrating that epigenetic mechanisms (including post-translational modifications of DNA and histones as well as non-coding RNA-mediated processes) play essential roles in the regulation of endothelial stem/progenitor cell functions through modifying chromatin structure. Furthermore, we discuss the potential of using small molecules that modulate the activities of epigenetic enzymes to enhance the vascular repair function of endothelial cells and offer insight on potential strategies that may accelerate clinical applications.
Collapse
Affiliation(s)
- Sylvain Fraineau
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Canada; Ottawa Institute of Systems Biology, Canada
| | | | | | | |
Collapse
|
31
|
Aberrant hypermethylation of aldehyde dehydrogenase 2 promoter upstream sequence in rats with experimental myocardial infarction. BIOMED RESEARCH INTERNATIONAL 2015; 2015:503692. [PMID: 25629048 PMCID: PMC4299765 DOI: 10.1155/2015/503692] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/13/2014] [Accepted: 11/20/2014] [Indexed: 12/31/2022]
Abstract
Background. Aldehyde dehydrogenase 2 (ALDH2) plays a crucial role in myocardial protection against ischemia. Downregulation of ALDH2 was evidenced after myocardial infarction and the underlying mechanism is not fully understood. DNA methylation can regulate gene transcription in epigenetic level. We thus hypothesized that DNA methylation may affect ALDH2 expression in myocardial infarction (MI). Methods. MI was induced in Sprague-Dawley rats. MI border zone tissues were harvested at 1st week, 2nd week, and 3rd week after MI. Bisulfite sequencing PCR (BSP) was performed to detect the methylation levels of ALDH2 core promoter. Sequenom MassARRAY platform (MassARRAY) was used to examine the methylation levels of ALDH2 promoter upstream sequence. ALDH2 protein and mRNA expression were assayed by Western blot and real-time PCR, respectively. Results. Compared with Sham group, ALDH2 protein and mRNA expression of MI groups was significantly downregulated. Compared with Sham group, DNA methylation level of CpG sites in ALDH2 promoter upstream sequence was significantly higher in MI groups in a time-dependent manner (CpG1, CpG2, and CpG7, P < 0.01). DNA methylation did not affect ALDH2 core promoter sequence during the progress of MI. No significant difference was detected in DNA methylation level of ALDH2 promoter upstream sequence among MI groups. Conclusion. Aberrant hypermethylation of CpG sites in ALDH2 promoter upstream sequence is associated with myocardial ischemia injury and may partly result in ALDH2 downregulation after MI.
Collapse
|
32
|
Grimaldi V, Vietri MT, Schiano C, Picascia A, De Pascale MR, Fiorito C, Casamassimi A, Napoli C. Epigenetic reprogramming in atherosclerosis. Curr Atheroscler Rep 2015; 17:476. [PMID: 25433555 DOI: 10.1007/s11883-014-0476-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent data support the involvement of epigenetic alterations in the pathogenesis of atherosclerosis. The most widely investigated epigenetic mechanism is DNA methylation although also histone code changes occur during the diverse steps of atherosclerosis, such as endothelial cell proliferation, vascular smooth muscle cell (SMC) differentiation, and inflammatory pathway activation. In this review, we focus on the main genes that are epigenetically modified during the atherogenic process, particularly nitric oxide synthase (NOS), estrogen receptors (ERs), collagen type XV alpha 1 (COL15A1), vascular endothelial growth factor receptor (VEGFR), and ten-eleven translocation (TET), which are involved in endothelial dysfunction; gamma interferon (IFN-γ), forkhead box p3 (FOXP3), and tumor necrosis factor-α (TNF-α), associated with atherosclerotic inflammatory process; and p66shc, lectin-like oxLDL receptor (LOX1), and apolipoprotein E (APOE) genes, which are regulated by high cholesterol and homocysteine (Hcy) levels. Furthermore, we also discuss the role of non-coding RNAs (ncRNA) in atherosclerosis. NcRNAs are involved in epigenetic regulation of endothelial function, SMC proliferation, cholesterol synthesis, lipid metabolism, and inflammatory response.
Collapse
Affiliation(s)
- Vincenzo Grimaldi
- U.O.C. Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], Regional Reference Laboratory of Transplant Immunology [LIT], Azienda Universitaria Policlinico (AOU), Second University of Naples (SUN), Piazza L. Miraglia 2, 80138, Naples, Italy,
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Roy-Chaudhury P, Kruska L. Future Directions for Vascular Access for Hemodialysis. Semin Dial 2014; 28:107-13. [DOI: 10.1111/sdi.12329] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Prabir Roy-Chaudhury
- Dialysis Vascular Access Research Group; Division of Nephrology; University of Cincinnati and Cincinnati VA Medical Center; Cincinnati Ohio
| | - Lindsay Kruska
- Division of Nephrology; University of North Carolina; Chapel Hill North Carolina
| |
Collapse
|
34
|
Tousoulis D, Simopoulou C, Papageorgiou N, Oikonomou E, Hatzis G, Siasos G, Tsiamis E, Stefanadis C. Endothelial dysfunction in conduit arteries and in microcirculation. Novel therapeutic approaches. Pharmacol Ther 2014; 144:253-267. [PMID: 24928320 DOI: 10.1016/j.pharmthera.2014.06.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 05/28/2014] [Indexed: 11/22/2022]
Abstract
The vascular endothelium not only is a single monolayer of cells between the vessel lumen and the intimal wall, but also plays an important role by controlling vascular function and structure mainly via the production of nitric oxide (NO). The so called "cardiovascular risk factors" are associated with endothelial dysfunction, that reduces NO bioavailability, increases oxidative stress, and promotes inflammation contributing therefore to the development of atherosclerosis. The significant role of endothelial dysfunction in the development of atherosclerosis emphasizes the need for efficient therapeutic interventions. During the last years statins, angiotensin-converting enzyme inhibitors, angiotensin-receptor antagonists, antioxidants, beta-blockers and insulin sensitizers have been evaluated for their ability to restore endothelial function (Briasoulis et al., 2012). As there is not a straightforward relationship between therapeutic interventions and improvement of endothelial function but rather a complicated interrelationship between multiple cellular and sub-cellular targets, research has been focused on the understanding of the underlying mechanisms. Moreover, the development of novel diagnostic invasive and non-invasive methods has allowed the early detection of endothelial dysfunction expanding the role of therapeutic interventions and our knowledge. In the current review we present the available data concerning the contribution of endothelial dysfunction to atherogenesis and review the methods that assess endothelial function with a view to understand the multiple targets of therapeutic interventions. Finally we focus on the classic and novel therapeutic approaches aiming to improve endothelial dysfunction and the underlying mechanisms.
Collapse
Affiliation(s)
- Dimitris Tousoulis
- 1st Cardiology Department, Athens University Medical School, Hippokration Hospital, Greece.
| | - Chryssa Simopoulou
- 1st Cardiology Department, Athens University Medical School, Hippokration Hospital, Greece
| | - Nikos Papageorgiou
- 1st Cardiology Department, Athens University Medical School, Hippokration Hospital, Greece
| | - Evangelos Oikonomou
- 1st Cardiology Department, Athens University Medical School, Hippokration Hospital, Greece
| | - George Hatzis
- 1st Cardiology Department, Athens University Medical School, Hippokration Hospital, Greece
| | - Gerasimos Siasos
- 1st Cardiology Department, Athens University Medical School, Hippokration Hospital, Greece
| | - Eleftherios Tsiamis
- 1st Cardiology Department, Athens University Medical School, Hippokration Hospital, Greece
| | | |
Collapse
|
35
|
Byrne MM, Murphy RT, Ryan AW. Epigenetic modulation in the treatment of atherosclerotic disease. Front Genet 2014; 5:364. [PMID: 25389432 PMCID: PMC4211541 DOI: 10.3389/fgene.2014.00364] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 09/29/2014] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular disease is the single largest cause of death in the western world and its incidence is on the rise globally. Atherosclerosis, characterized by the development of atheromatus plaque, can trigger luminal narrowing and upon rupture result in myocardial infarction or ischemic stroke. Epigenetic phenomena are a focus of considerable research interest due to the role they play in gene regulation. Epigenetic mechanisms such as DNA methylation and histone acetylation have been identified as potential drug targets in the treatment of cardiovascular disease. miRNAs are known to play a role in gene silencing, which has been widely investigated in cancer. In comparison, the role they play in cardiovascular disease and plaque rupture is not well understood. Nutritional epigenetic modifiers from dietary components, for instance sulforaphane found in broccoli, have been shown to suppress the pro-inflammatory response through transcription factor activation. This review will discuss current and potential epigenetic therapeutics for the treatment of cardiovascular disease, focusing on the use of miRNAs and dietary supplements such as sulforaphane and protocatechuic aldehyde.
Collapse
Affiliation(s)
- Mikaela M. Byrne
- Department of Clinical Medicine and Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s HospitalDublin, Ireland
| | - Ross T. Murphy
- Department of Cardiology, St. James’s HospitalDublin, Ireland
| | - Anthony W. Ryan
- Department of Clinical Medicine and Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s HospitalDublin, Ireland
| |
Collapse
|
36
|
Abstract
Epigenetics refers to long-term modifications of gene activity that can be inherited, either somatically or transgenerationally, but that are independent of alterations in the primary base sequence of the organism's DNA. These changes can include chemical modifications of both the DNA bases and the proteins that associate with the DNA helices to form chromatin, the nucleic acid:protein complex of which the chromosomes are comprised. Epigenetic modifications can affect the accessibility of the DNA for transcription factors (the DNA-binding proteins that specify which genes are to be active or silent by modulating the recruitment of the transcriptional machinery that reads the information encoded in the sequence) and thereby regulate the expression of genes and alter the phenotype of the organism. Epigenetic marks can also be re-established following mitosis, allowing patterns of differential gene expression to be transmitted from one cell generation to the next, and can even be maintained through meiosis, allowing transgenerational transfer of regulatory cues. Unlike the information encoded in the DNA sequence, which is invariant between most cell types and over time, epigenetic information is tissue specific and can change in response to exogenous and endogenous perturbations. This responsive capacity enables a sensitive and reactive system that can optimize gene expression in relevant tissue in response to environmental change. The realization that organisms are capable of genetically 'reprograming' themselves as well as 'preprograming' future cells, and even future offspring to optimize gene expression for a given environment may have tremendous ramifications on our understanding of both acclimatization and adaptation to hypoxia.
Collapse
Affiliation(s)
- Carolyn J Brown
- 1 Department of Medical Genetics, Molecular Epigenetics Group, University of British Columbia , Vancouver, British Columbia, Canada
| | | |
Collapse
|
37
|
Cybulsky MI, Marsden PA. Effect of Disturbed Blood Flow on Endothelial Cell Gene Expression. Arterioscler Thromb Vasc Biol 2014; 34:1806-8. [DOI: 10.1161/atvbaha.114.304099] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Myron I. Cybulsky
- From the Toronto General Research Institute, University Health Network, and the Department of Laboratory Medicine and Pathobiology, University of Toronto (M.I.C.), and St Michael’s Hospital, Division of Nephrology, Department of Medicine, University of Toronto (P.A.M.), Toronto, Canada
| | - Philip A. Marsden
- From the Toronto General Research Institute, University Health Network, and the Department of Laboratory Medicine and Pathobiology, University of Toronto (M.I.C.), and St Michael’s Hospital, Division of Nephrology, Department of Medicine, University of Toronto (P.A.M.), Toronto, Canada
| |
Collapse
|
38
|
Miragoli M, Yacoub MH, El-Hamamsy I, Sanchez-Alonso JL, Moshkov A, Mongkoldhumrongkul N, Padala M, Paramagurunathan S, Sarathchandra P, Korchev YE, Gorelik J, Chester AH. Side-specific mechanical properties of valve endothelial cells. Am J Physiol Heart Circ Physiol 2014; 307:H15-24. [DOI: 10.1152/ajpheart.00228.2013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Aortic valve endothelial cells (ECs) function in vastly different levels of shear stress. The biomechanical characteristics of cells on each side of valve have not been investigated. We assessed the morphology and mechanical properties of cultured or native valve ECs on intact porcine aortic valve cusps using a scanning ion conductance microscope (SICM). The autocrine influence of several endothelial-derived mediators on cell compliance and the expression of actin were also examined. Cells on the aortic side of the valve are characterized by a more elongated shape and were aligned along a single axis. Measurement of EC membrane compliance using the SICM showed that the cells on the aortic side of intact valves were significantly softer than those on the ventricular side. A similar pattern was seen in cultured cells. Addition of 10−6 M of the nitric oxide donor sodium nitroprusside caused a significant reduction in the compliance of ventricular ECs but had no effect on cells on the aortic side of the valve. Conversely, endothelin-1 (10−10-10−8 M) caused an increase in the compliance of aortic cells but had no effect on cells on the ventricular side of the valve. Aortic side EC compliance was also increased by 10−4 M of the nitric oxide synthase inhibitor NG-nitro-l-arginine methyl ester. Immunofluorescent staining of actin filaments revealed a great density of staining in ECs on the ventricular surface. The expression of actin and the relative membrane compliance of ECs on both side of the valve were not affected by ventricular and aortic patterns of flow. This study has shown side-specific differences in the biomechanics of aortic valve ECs. These differences can have important implications for valve function.
Collapse
Affiliation(s)
- Michele Miragoli
- Imperial College, National Heart and Lung Institute, Department of Cardiac Medicine, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus, London, United Kingdom
- Humanitas Clinical and Research Center, Rozzano (Milan), Italy
| | - Magdi H. Yacoub
- Imperial College, National Heart and Lung Institute, Heart Science Centre, Harefield, Middlesex, United Kingdom
- Qatar Cardiovascular Research Centre, Qatar Foundation, Doha, Qatar
| | - Ismail El-Hamamsy
- Imperial College, National Heart and Lung Institute, Heart Science Centre, Harefield, Middlesex, United Kingdom
| | - Jose L. Sanchez-Alonso
- Imperial College, National Heart and Lung Institute, Department of Cardiac Medicine, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus, London, United Kingdom
| | - Alexey Moshkov
- Imperial College, National Heart and Lung Institute, Department of Cardiac Medicine, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus, London, United Kingdom
| | | | - Muralindar Padala
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia; and
| | - Saravanan Paramagurunathan
- Imperial College, National Heart and Lung Institute, Heart Science Centre, Harefield, Middlesex, United Kingdom
| | - Padmini Sarathchandra
- Imperial College, National Heart and Lung Institute, Heart Science Centre, Harefield, Middlesex, United Kingdom
| | - Yuri E. Korchev
- Division of Medicine, Imperial College, Hammersmith Campus, London, United Kingdom
| | - Julia Gorelik
- Imperial College, National Heart and Lung Institute, Department of Cardiac Medicine, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus, London, United Kingdom
| | - Adrian H. Chester
- Imperial College, National Heart and Lung Institute, Heart Science Centre, Harefield, Middlesex, United Kingdom
- Qatar Cardiovascular Research Centre, Qatar Foundation, Doha, Qatar
| |
Collapse
|
39
|
Campos B, Lee T, Roy-Chaudhury P. Arteriovenous fistula failure: is there a role for epigenetic regulation? Semin Nephrol 2014; 33:400-6. [PMID: 24011582 DOI: 10.1016/j.semnephrol.2013.05.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Epigenetics is the study of heritable changes in gene expression or cellular phenotype that occur without alterations in the DNA sequence. In the past decade, epigenetics has been identified as a key regulator of gene expression and therefore is likely to play a major role in multiple disease processes. More importantly, we now recognize epigenetics to be a sensitive, dynamic, and reversible process that has opened the door to multiple novel diagnostic, prognostic, and therapeutic strategies for human diseases. The focus of this review, however, is to explore the potential role of epigenetics in arteriovenous fistula (AVF) maturation. AVF maturation failure is currently the single most important cause of dialysis vascular access dysfunction and most important is the result of a peri-anastomotic stenosis thought to be caused by a combination of neointimal hyperplasia and inadequate outward remodeling. At a pathogenetic level, however, AVF maturation failure is likely the end result of the interaction between hemodynamic stressors (injury) and the vascular response to these stressors; the latter being influenced by uremia, oxidative stress, and inflammation. Interestingly, these same factors (hemodynamic shear stress, oxidative stress, inflammation, and uremia) are also important mediators of epigenetic modifications. We therefore believe that epigenetic factors potentially could play an important role in the pathogenesis of AVF maturation failure. The current review therefore tries to unravel some of these critical biological connections, with an emphasis on the future development of epigenetic-based diagnostic and therapeutic strategies for AVF maturation failure (a clinical problem for which there are currently no effective therapeutic interventions).
Collapse
Affiliation(s)
- Begoña Campos
- Dialysis Vascular Access Research Group, Division of Nephrology and Hypertension, Department of Medicine, University of Cincinnati, Cincinnati, OH
| | | | | |
Collapse
|
40
|
Barroso M, Florindo C, Kalwa H, Silva Z, Turanov AA, Carlson BA, de Almeida IT, Blom HJ, Gladyshev VN, Hatfield DL, Michel T, Castro R, Loscalzo J, Handy DE. Inhibition of cellular methyltransferases promotes endothelial cell activation by suppressing glutathione peroxidase 1 protein expression. J Biol Chem 2014; 289:15350-62. [PMID: 24719327 PMCID: PMC4140892 DOI: 10.1074/jbc.m114.549782] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
S-adenosylhomocysteine (SAH) is a negative regulator of most methyltransferases and the precursor for the cardiovascular risk factor homocysteine. We have previously identified a link between the homocysteine-induced suppression of the selenoprotein glutathione peroxidase 1 (GPx-1) and endothelial dysfunction. Here we demonstrate a specific mechanism by which hypomethylation, promoted by the accumulation of the homocysteine precursor SAH, suppresses GPx-1 expression and leads to inflammatory activation of endothelial cells. The expression of GPx-1 and a subset of other selenoproteins is dependent on the methylation of the tRNA(Sec) to the Um34 form. The formation of methylated tRNA(Sec) facilitates translational incorporation of selenocysteine at a UGA codon. Our findings demonstrate that SAH accumulation in endothelial cells suppresses the expression of GPx-1 to promote oxidative stress. Hypomethylation stress, caused by SAH accumulation, inhibits the formation of the methylated isoform of the tRNA(Sec) and reduces GPx-1 expression. In contrast, under these conditions, the expression and activity of thioredoxin reductase 1, another selenoprotein, is increased. Furthermore, SAH-induced oxidative stress creates a proinflammatory activation of endothelial cells characterized by up-regulation of adhesion molecules and an augmented capacity to bind leukocytes. Taken together, these data suggest that SAH accumulation in endothelial cells can induce tRNA(Sec) hypomethylation, which alters the expression of selenoproteins such as GPx-1 to contribute to a proatherogenic endothelial phenotype.
Collapse
Affiliation(s)
- Madalena Barroso
- From the Cardiovascular and ,the Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL) and
| | - Cristina Florindo
- the Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL) and
| | | | - Zélia Silva
- the Chronic Diseases Research Center, Departamento de Imunologia, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1099-085 Lisbon, Portugal
| | - Anton A. Turanov
- Genetics Divisions, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Bradley A. Carlson
- the Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Isabel Tavares de Almeida
- the Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL) and ,Department of Biochemistry and Human Biology, Faculty of Pharmacy, University of Lisbon, 1649-004 Lisbon, Portugal
| | - Henk J. Blom
- the Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, University Hospital, 79106 Freiburg, Germany
| | - Vadim N. Gladyshev
- Genetics Divisions, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Dolph L. Hatfield
- the Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, and
| | | | - Rita Castro
- the Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL) and ,Department of Biochemistry and Human Biology, Faculty of Pharmacy, University of Lisbon, 1649-004 Lisbon, Portugal
| | | | - Diane E. Handy
- From the Cardiovascular and , To whom correspondence should be addressed: Cardiovascular Div., Dept. of Medicine, Brigham and Women's Hospital and Harvard Medical School, 77 Ave. Louis Pasteur, Boston, MA, 02115. Tel.: 617-525-4845; Fax: 617-525-4830; E-mail:
| |
Collapse
|
41
|
Allen RP, Schelegle ES, Bennett SH. Diverse forms of pulmonary hypertension remodel the arterial tree to a high shear phenotype. Am J Physiol Heart Circ Physiol 2014; 307:H405-17. [PMID: 24858853 DOI: 10.1152/ajpheart.00144.2014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Pulmonary hypertension (PH) is associated with progressive changes in arterial network complexity. An allometric model is derived that integrates diameter branching complexity between pulmonary arterioles of generation n and the main pulmonary artery (MPA) via a power-law exponent (X) in dn = dMPA2(-n/X) and the arterial area ratio β = 2(1-2/X). Our hypothesis is that diverse forms of PH demonstrate early decrements in X independent of etiology and pathogenesis, which alters the arteriolar shear stress load from a low-shear stress (X > 2, β > 1) to a high-shear stress phenotype (X < 2, β < 1). Model assessment was accomplished by comparing theoretical predictions to retrospective morphometric and hemodynamic measurements made available from a total of 221 PH-free and PH subjects diagnosed with diverse forms (World Health Organization; WHO groups I-IV) of PH: mitral stenosis, congenital heart disease, chronic obstructive pulmonary lung disease, chronic thromboembolism, idiopathic pulmonary arterial hypertension (IPAH), familial (FPAH), collagen vascular disease, and methamphetamine exposure. X was calculated from pulmonary artery pressure (PPA), cardiac output (Q) and body weight (M), utilizing an allometric power-law prediction of X relative to a PH-free state. Comparisons of X between PAH-free and PAH subjects indicates a characteristic reduction in area that elevates arteriolar shear stress, which may contribute to mechanisms of endothelial dysfunction and injury before clinically defined thresholds of pulmonary vascular resistance and PH. We conclude that the evaluation of X may be of use in identifying reversible and irreversible phases of PH in the early course of the disease process.
Collapse
Affiliation(s)
- Roblee P Allen
- Department of Pulmonary and Critical Care Medicine, University of California Davis Health System, Sacramento, California
| | - Edward S Schelegle
- Department of Anatomy, Physiology and Cell Biology, Veterinary Medicine, University of California, Davis, California; Respiratory Disease Unit, California National Primate Center, University of California, Davis, California
| | - Stephen H Bennett
- Respiratory Disease Unit, California National Primate Center, University of California, Davis, California
| |
Collapse
|
42
|
Okada Y, Funahashi N, Tanaka T, Nishiyama Y, Yuan L, Shirakura K, Turjman AS, Kano Y, Naruse H, Suzuki A, Sakai M, Zhixia J, Kitajima K, Ishimoto K, Hino N, Kondoh M, Mukai Y, Nakagawa S, García-Cardeña G, Aird WC, Doi T. Endothelial cell-specific expression of roundabout 4 is regulated by differential DNA methylation of the proximal promoter. Arterioscler Thromb Vasc Biol 2014; 34:1531-8. [PMID: 24855053 DOI: 10.1161/atvbaha.114.303818] [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: 02/01/2023]
Abstract
OBJECTIVE The molecular basis of endothelial cell (EC)-specific gene expression is poorly understood. Roundabout 4 (Robo4) is expressed exclusively in ECs. We previously reported that the 3-kb 5'-flanking region of the human Robo4 gene contains information for lineage-specific expression in the ECs. Our studies implicated a critical role for GA-binding protein and specificity protein 1 (SP1) in mediating overall expression levels. However, these transcription factors are also expressed in non-ECs. In this study, we tested the hypothesis that epigenetic mechanisms contribute to EC-specific Robo4 gene expression. METHODS AND RESULTS Bisulfite sequencing analysis indicated that the proximal promoter of Robo4 is methylated in non-ECs but not in ECs. Treatment with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine increased Robo4 gene expression in non-ECs but not in ECs. Proximal promoter methylation significantly decreased the promoter activity in ECs. Electrophoretic mobility shift assays showed that DNA methylation of the proximal promoter inhibited SP1 binding to the -42 SP1 site. In DNase hypersensitivity assays, chromatin condensation of the Robo4 promoter was observed in some but not all nonexpressing cell types. In Hprt (hypoxanthine phosphoribosyltransferase)-targeted mice, a 0.3-kb proximal promoter directed cell-type-specific expression in the endothelium. Bisulfite sequencing analysis using embryonic stem cell-derived mesodermal cells and ECs indicated that the EC-specific methylation pattern of the promoter is determined by demethylation during differentiation and that binding of GA-binding protein and SP1 to the proximal promoter is not essential for demethylation. CONCLUSIONS The EC-specific DNA methylation pattern of the Robo4 proximal promoter is determined during cell differentiation and contributes to regulation of EC-specific Robo4 gene expression.
Collapse
Affiliation(s)
- Yoshiaki Okada
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.).
| | - Nobuaki Funahashi
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| | - Toru Tanaka
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| | - Yuji Nishiyama
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| | - Lei Yuan
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| | - Keisuke Shirakura
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| | - Alexis S Turjman
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| | - Yoshihiro Kano
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| | - Hiroki Naruse
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| | - Ayano Suzuki
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| | - Miki Sakai
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| | - Jiang Zhixia
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| | - Kenji Kitajima
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| | - Kenji Ishimoto
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| | - Nobumasa Hino
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| | - Masuo Kondoh
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| | - Yohei Mukai
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| | - Shinsaku Nakagawa
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| | - Guillermo García-Cardeña
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| | - William C Aird
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.).
| | - Takefumi Doi
- From the Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan (Y.O., N.F., T.T., Y.N., K.S., Y.K., H.N., A.S., M.S., J.Z., K.I., N.H., M.K., Y.M., S.N., T.D.); Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (L.Y., W.C.A.); Department of Pathology, Center for Excellence in Vascular Biology, Harvard Medical School, Boston, MA (A.S.T., G.G.-C.); Department of Material Sciences, Massachusetts Institute of Technology, Boston (A.S.T.); and Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan (K.K.)
| |
Collapse
|
43
|
Golub AS, Pittman RN. Bang-bang model for regulation of local blood flow. Microcirculation 2014; 20:455-83. [PMID: 23441827 DOI: 10.1111/micc.12051] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 02/19/2013] [Indexed: 11/27/2022]
Abstract
The classical model of metabolic regulation of blood flow in muscle tissue implies the maintenance of basal tone in arterioles of resting muscle and their dilation in response to exercise and/or tissue hypoxia via the evoked production of vasodilator metabolites by myocytes. A century-long effort to identify specific metabolites responsible for explaining active and reactive hyperemia has not been successful. Furthermore, the metabolic theory is not compatible with new knowledge on the role of physiological radicals (e.g., nitric oxide, NO, and superoxide anion, O2 (-) ) in the regulation of microvascular tone. We propose a model of regulation in which muscle contraction and active hyperemia are considered the physiologically normal state. We employ the "bang-bang" or "on/off" regulatory model which makes use of a threshold and hysteresis; a float valve to control the water level in a tank is a common example of this type of regulation. Active bang-bang regulation comes into effect when the supply of oxygen and glucose exceeds the demand, leading to activation of membrane NADPH oxidase, release of O2 (-) into the interstitial space and subsequent neutralization of the interstitial NO. Switching arterioles on/off when local blood flow crosses the threshold is realized by a local cell circuit with the properties of a bang-bang controller, determined by its threshold, hysteresis, and dead-band. This model provides a clear and unambiguous interpretation of the mechanism to balance tissue demand with a sufficient supply of nutrients and oxygen.
Collapse
Affiliation(s)
- Aleksander S Golub
- Department of Physiology and Biophysics, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA.
| | | |
Collapse
|
44
|
Shiva Shankar TV, Willems L. Epigenetic modulators mitigate angiogenesis through a complex transcriptomic network. Vascul Pharmacol 2014; 60:57-66. [PMID: 24445350 DOI: 10.1016/j.vph.2014.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 12/18/2013] [Accepted: 01/08/2014] [Indexed: 12/19/2022]
Abstract
In this review, we summarize the knowledge pertaining to the role of epigenetics in the regulation of angiogenesis. In particular, we show that lysine acetylation and cytosine methylation are important transcriptional regulators of angiogenic genes in endothelial cells. Lysine acetylation and cytosine methylation inhibitors idiosyncratically tune the transcriptome and affect expression of key modulators of angiogenesis such as VEGF and eNOS. Transcriptomic profiling also reveals a series of novel genes that are concomitantly affected by epigenetic modulators. The reversibility and overall tolerability of currently available epigenetic inhibitors open up the prospect of therapeutic intervention in pathologies where angiogenesis is exacerbated. This type of multitargeted strategy has the major advantage of overcoming the compensatory feedback mechanisms that characterize single anti-angiogenic factors.
Collapse
Affiliation(s)
- T V Shiva Shankar
- Molecular and Cellular Epigenetics (GIGA-Cancer) and Molecular Biology (GxABT), University of Liège (ULg), Liège, Belgium
| | - L Willems
- Molecular and Cellular Epigenetics (GIGA-Cancer) and Molecular Biology (GxABT), University of Liège (ULg), Liège, Belgium.
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Monocytes/macrophages play a decisive role in the development and progression of atherosclerosis. It is currently unknown what stimuli initiate and orchestrate the activation of these cells in atherogenesis. In this review, we postulate that the novel concept of 'trained immunity' modulates the development and progression of atherosclerosis. RECENT FINDINGS Recently, results from our laboratory challenged the current paradigm that innate immunity is static and does not have an immunological memory. Stimulation by various microbial products, including Candida albicans and bacille Calmette-Guérin, appeared to bring monocytes into a long-term enhanced functional state, showing a stronger proinflammatory response to a second stimulus. This 'trained immunity' was mediated by increased and stable histone methylation. SUMMARY We describe the hypothesis that this functional reprogramming of monocytes, either by microbial products or by metabolic products, contributes to atherogenesis and propose epigenetic reprogramming of monocytes as a novel pharmacological target for preventing or treating atherosclerosis in the future.
Collapse
Affiliation(s)
- Siroon Bekkering
- aDepartment of Internal Medicine, Division of Experimental Medicine, Radboud University Nijmegen Medical Centre bNijmegen Institute for Infection, Inflammation and Immunity (N4i) cDepartment of Internal Medicine, Division of Vascular Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | |
Collapse
|
46
|
Duygu B, Poels EM, da Costa Martins PA. Genetics and epigenetics of arrhythmia and heart failure. Front Genet 2013; 4:219. [PMID: 24198825 PMCID: PMC3812794 DOI: 10.3389/fgene.2013.00219] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 10/08/2013] [Indexed: 12/21/2022] Open
Abstract
Heart failure (HF) is the end stage of several pathological cardiac conditions including myocardial infarction, cardiac hypertrophy and hypertension. Various molecular and cellular mechanisms are involved in the development of HF. At the molecular level, the onset of HF is associated with reprogramming of gene expression, including downregulation of the alpha-myosin heavy chain (α-MHC) gene and sarcoplasmic reticulum Ca 2+ ATPase genes and reactivation of specific fetal cardiac genes such as atrial natriuretic factor and brain natriuretic peptide. These deviations in gene expression result in structural and electrophysiological changes, which eventually progress to HF. Cardiac arrhythmia is caused by altered conduction properties of the heart, which may arise in response to ischemia, inflammation, fibrosis, aging or from genetic factors. Because changes in the gene transcription program may have crucial consequences as deteriorated cardiac function, understanding the molecular mechanisms involved in the process has become a priority in the field. In this context, various studies besides having identified different DNA methylation patterns in HF patients, have also focused on specific disease processes and their underlying mechanisms, also introducing new concepts such as epigenomics. This review highlights specific genetic mutations associated with the onset and progression of HF, also providing an introduction to epigenetic mechanisms such as histone modifications, DNA methylation and RNA-based modification, and highlights the relation between epigenetics, arrhythmogenesis and HF.
Collapse
Affiliation(s)
- Burcu Duygu
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Maastricht, Netherlands
| | | | | |
Collapse
|
47
|
Joo JE, Hiden U, Lassance L, Gordon L, Martino DJ, Desoye G, Saffery R. Variable promoter methylation contributes to differential expression of key genes in human placenta-derived venous and arterial endothelial cells. BMC Genomics 2013; 14:475. [PMID: 23855827 PMCID: PMC3729658 DOI: 10.1186/1471-2164-14-475] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Accepted: 07/10/2013] [Indexed: 11/26/2022] Open
Abstract
Background The endothelial compartment, comprising arterial, venous and lymphatic cell types, is established prenatally in association with rapid phenotypic and functional changes. The molecular mechanisms underpinning this process in utero have yet to be fully elucidated. The aim of this study was to investigate the potential for DNA methylation to act as a driver of the specific gene expression profiles of arterial and venous endothelial cells. Results Placenta-derived venous and arterial endothelial cells were collected at birth prior to culturing. DNA methylation was measured at >450,000 CpG sites in parallel with expression measurements taken from 25,000 annotated genes. A consistent set of genomic loci was found to show coordinate differential methylation between the arterial and venous cell types. This included many loci previously not investigated in relation to endothelial function. An inverse relationship was observed between gene expression and promoter methylation levels for a limited subset of genes implicated in endothelial function, including NOS3, encoding endothelial Nitric Oxide Synthase. Conclusion Endothelial cells derived from the placental vasculature at birth contain widespread methylation of key regulatory genes. These are candidates involved in the specification of different endothelial cell types and represent potential target genes for environmentally mediated epigenetic disruption in utero in association with cardiovascular disease risk later in life.
Collapse
Affiliation(s)
- Jihoon E Joo
- Cancer and Disease Epigenetics, Murdoch Childrens Research Institute, Royal Children's Hospital, Flemington Road, Parkville, Melbourne, Australia
| | | | | | | | | | | | | |
Collapse
|
48
|
Kheirandish-Gozal L, Khalyfa A, Gozal D, Bhattacharjee R, Wang Y. Endothelial dysfunction in children with obstructive sleep apnea is associated with epigenetic changes in the eNOS gene. Chest 2013; 143:971-977. [PMID: 23328840 DOI: 10.1378/chest.12-2026] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is a highly prevalent disorder that has been associated with an increased risk for cardiovascular morbidity, even in children. However, not all children with OSA manifest alterations in endothelial postocclusive hyperemia, an endothelial nitric oxide synthase (eNOS)-dependent response. Since expression of the eNOS gene is regulated by epigenetic mechanisms and OSA may cause epigenetic modifications such as DNA hypermethylation, we hypothesized that epigenetic modifications in the eNOS gene may underlie the differential vascular phenotypes in pediatric OSA. METHODS Age-, sex-, ethnicity-, and BMI-matched prepubertal children with polysomnographically confirmed OSA and either normal (OSAn) or abnormal (OSAab) postocclusive hyperemic responses, assessed as the time to attain peak reperfusion flow (Tmax) by laser Doppler flowmetry, were recruited. Blood genomic DNA was assessed for epigenetic modifications in the eNOS gene using pyrosequencing. Children with no evidence of OSA or endothelial dysfunction served as a control group. RESULTS The study comprised 36 children with OSA (11 with OSAab and 25 with OSAn) and 35 children in the control group. Overall, the mean age was 7.5 ± 2.4 years, 65% were boys, and 30% were obese; mean apnea-hypopnea index was 18 ± 8.6/h of sleep for the children with OSA. Tmax was 66.7 ± 8.8 s in the OSAab group and 30.1 ± 8.3 s in the OSAn group (P < .001). Pyrosequencing of the proximal promoter region of the eNOS gene revealed no significant differences in six of the seven CpG sites. However, a CpG site located at position -171 (relative to transcription start site), approximating important transcriptional elements, displayed significantly higher methylation levels in the OSAab group as compared with the OSAn or control groups (81.5% ± 3.5%, 74.8% ± 1.4%, and 74.5% ± 1.7%, respectively; P < .001). eNOS mRNA expression levels were assessed in a separate group of children and were significantly reduced in the OSAab group in comparison with the OSAn group. CONCLUSIONS The presence of abnormal eNOS-dependent vascular responses in children with OSA is associated with epigenetic modifications in the eNOS gene.
Collapse
Affiliation(s)
- Leila Kheirandish-Gozal
- Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Abdelnaby Khalyfa
- Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - David Gozal
- Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Rakesh Bhattacharjee
- Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Yang Wang
- Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL.
| |
Collapse
|
49
|
Kim GH, Ryan JJ, Archer SL. The role of redox signaling in epigenetics and cardiovascular disease. Antioxid Redox Signal 2013; 18:1920-36. [PMID: 23480168 PMCID: PMC3624767 DOI: 10.1089/ars.2012.4926] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 12/24/2012] [Accepted: 01/15/2013] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE The term epigenetics refers to the changes in the phenotype and gene expression that occur without alterations in the DNA sequence. There is a rapidly growing body of evidence that epigenetic modifications are involved in the pathological mechanisms of many cardiovascular diseases (CVDs), which intersect with many of the pathways involved in oxidative stress. RECENT ADVANCES Most studies relating epigenetics and human pathologies have focused on cancer. There has been a limited study of epigenetic mechanisms in CVDs. Although CVDs have multiple established genetic and environmental risk factors, these explain only a portion of the total CVD risk. The epigenetic perspective is beginning to shed new light on how the environment influences gene expression and disease susceptibility in CVDs. Known epigenetic changes contributing to CVD include hypomethylation in proliferating vascular smooth muscle cells in atherosclerosis, changes in estrogen receptor-α (ER-α) and ER-β methylation in vascular disease, decreased superoxide dismutase 2 expression in pulmonary hypertension (PH), as well as trimethylation of histones H3K4 and H3K9 in congestive heart failure. CRITICAL ISSUES In this review, we discuss the epigenetic modifications in CVDs, including atherosclerosis, congestive heart failure, hypertension, and PH, with a focus on altered redox signaling. FUTURE DIRECTIONS As advances in both the methodology and technology accelerate the study of epigenetic modifications, the critical role they play in CVD is beginning to emerge. A fundamental question in the field of epigenetics is to understand the biochemical mechanisms underlying reactive oxygen species-dependent regulation of epigenetic modification.
Collapse
Affiliation(s)
- Gene H Kim
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, Illinois 60637, USA.
| | | | | |
Collapse
|
50
|
Webster ALH, Yan MSC, Marsden PA. Epigenetics and cardiovascular disease. Can J Cardiol 2013; 29:46-57. [PMID: 23261320 DOI: 10.1016/j.cjca.2012.10.023] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 10/26/2012] [Accepted: 10/28/2012] [Indexed: 12/18/2022] Open
Abstract
A commonly-assumed paradigm holds that the primary genetic determinant of cardiovascular disease resides within the DNA sequence of our genes. This paradigm can be challenged. For example, how do sequence changes in the non-coding region of the genome influence phenotype? Why are all diseases not shared between identical twins? Part of the answer lies in the fact that the environment or exogenous stimuli clearly influence disease susceptibility, but it was unclear in the past how these effects were signalled to the static DNA code. Epigenetics is providing a newer perspective on these issues. Epigenetics refers to chromatin-based mechanisms important in the regulation of gene expression that do not involve changes to the DNA sequence per se. The field can be broadly categorized into three areas: DNA base modifications (including cytosine methylation and cytosine hydroxymethylation), post-translational modifications of histone proteins, and RNA-based mechanisms that operate in the nucleus. Cardiovascular disease pathways are now being approached from the epigenetic perspective, including those associated with atherosclerosis, angiogenesis, ischemia-reperfusion damage, and the cardiovascular response to hypoxia and shear stress, among many others. With increasing interest and expanding partnerships in the field, we can expect new insights to emerge from epigenetic perspectives of cardiovascular health. This paper reviews the principles governing epigenetic regulation, discusses their presently-understood importance in cardiovascular disease, and considers the growing significance we are likely to attribute to epigenetic contributions in the future, as they provide new mechanistic insights and a host of novel clinical applications.
Collapse
Affiliation(s)
- Andrew L H Webster
- Keenan Research Centre and Li Ka Shing Knowledge Institute, St Michael's Hospital, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|