51
|
Osteopontin in Cardiovascular Diseases. Biomolecules 2021; 11:biom11071047. [PMID: 34356671 PMCID: PMC8301767 DOI: 10.3390/biom11071047] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/14/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Unprecedented advances in secondary prevention have greatly improved the prognosis of cardiovascular diseases (CVDs); however, CVDs remain a leading cause of death globally. These findings suggest the need to reconsider cardiovascular risk and optimal medical therapy. Numerous studies have shown that inflammation, pro-thrombotic factors, and gene mutations are focused not only on cardiovascular residual risk but also as the next therapeutic target for CVDs. Furthermore, recent clinical trials, such as the Canakinumab Anti-inflammatory Thrombosis Outcomes Study trial, showed the possibility of anti-inflammatory therapy for patients with CVDs. Osteopontin (OPN) is a matricellular protein that mediates diverse biological functions and is involved in a number of pathological states in CVDs. OPN has a two-faced phenotype that is dependent on the pathological state. Acute increases in OPN have protective roles, including wound healing, neovascularization, and amelioration of vascular calcification. By contrast, chronic increases in OPN predict poor prognosis of a major adverse cardiovascular event independent of conventional cardiovascular risk factors. Thus, OPN can be a therapeutic target for CVDs but is not clinically available. In this review, we discuss the role of OPN in the development of CVDs and its potential as a therapeutic target.
Collapse
|
52
|
Zhang X, Sun J, Canfrán-Duque A, Aryal B, Tellides G, Chang YJ, Suárez Y, Osborne TF, Fernández-Hernando C. Deficiency of histone lysine methyltransferase SETDB2 in hematopoietic cells promotes vascular inflammation and accelerates atherosclerosis. JCI Insight 2021; 6:147984. [PMID: 34003795 PMCID: PMC8262461 DOI: 10.1172/jci.insight.147984] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/12/2021] [Indexed: 02/05/2023] Open
Abstract
Epigenetic modifications of the genome, including DNA methylation, histone methylation/acetylation, and noncoding RNAs, have been reported to play a fundamental role in regulating immune response during the progression of atherosclerosis. SETDB2 is a member of the KMT1 family of lysine methyltransferases, and members of this family typically methylate histone H3 Lys9 (H3K9), an epigenetic mark associated with gene silencing. Previous studies have shown that SETDB2 is involved in innate and adaptive immunity, the proinflammatory response, and hepatic lipid metabolism. Here, we report that expression of SETDB2 is markedly upregulated in human and murine atherosclerotic lesions. Upregulation of SETDB2 was observed in proinflammatory M1 but not antiinflammatory M2 macrophages. Notably, we found that genetic deletion of SETDB2 in hematopoietic cells promoted vascular inflammation and enhanced the progression of atherosclerosis in BM transfer studies in Ldlr-knockout mice. Single-cell RNA-Seq analysis in isolated CD45+ cells from atherosclerotic plaques from mice transplanted with SETDB2-deficient BM revealed a significant increase in monocyte population and enhanced expression of genes involved in inflammation and myeloid cell recruitment. Additionally, we found that loss of SETDB2 in hematopoietic cells was associated with macrophage accumulation in atherosclerotic lesions and attenuated efferocytosis. Overall, these studies identify SETDB2 as an important inflammatory cell regulator that controls macrophage activation in atherosclerotic plaques.
Collapse
Affiliation(s)
- Xinbo Zhang
- Vascular Biology and Therapeutics Program.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, and
| | - Jonathan Sun
- Vascular Biology and Therapeutics Program.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, and
| | - Alberto Canfrán-Duque
- Vascular Biology and Therapeutics Program.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, and
| | - Binod Aryal
- Vascular Biology and Therapeutics Program.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, and
| | - George Tellides
- Vascular Biology and Therapeutics Program.,Department of Surgery, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ying Ju Chang
- Department of Medicine and.,Institute for Fundamental Biomedical Research, Johns Hopkins University School of Medicine, St. Petersburg, Florida, USA
| | - Yajaira Suárez
- Vascular Biology and Therapeutics Program.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, and
| | - Timothy F Osborne
- Department of Medicine and.,Institute for Fundamental Biomedical Research, Johns Hopkins University School of Medicine, St. Petersburg, Florida, USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, and
| |
Collapse
|
53
|
Wei X, Zhang Y, Xie L, Wang K, Wang X. Pharmacological inhibition of EZH2 by GSK126 decreases atherosclerosis by modulating foam cell formation and monocyte adhesion in apolipoprotein E-deficient mice. Exp Ther Med 2021; 22:841. [PMID: 34149887 PMCID: PMC8210282 DOI: 10.3892/etm.2021.10273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 12/01/2020] [Indexed: 12/31/2022] Open
Abstract
Histone modifications play an important role in the occurrence and development of atherosclerosis in human and atherosclerosis-prone mice. Histone methylation in macrophages, monocytes and endothelial cells markedly influence the progression of atherosclerosis. However, it remains unclear whether treatment with a histone methyltransferase enhancer of zeste homolog 2 (EZH2) inhibitor may suppress atherosclerosis. The present study aimed to determine the effects of the EZH2 inhibitor, GSK126, on the suppression and regression of atherosclerosis in apolipoprotein E-deficient mouse models. In vitro, it was found that pharmacological inhibition of EZH2 by GSK126 markedly reduced lipid transportation and monocyte adhesion during atherogenesis, predominantly through increasing the expression levels of ATP-binding cassette transporter A1 and suppressing vascular cell adhesion molecule 1 in human THP-1 cells. In vivo, it was found that atherosclerotic plaques in GSK126-treated mice were significantly decreased when comparing with the vehicle-treated animals. These results indicated that the GSK126 has the ability to attenuate the progression of atherosclerosis by reducing macrophage foam cell formation and monocyte adhesion in cell and mouse models. In conclusion, the present study provided new insights into the molecular mechanism behind the action of GSK126 and suggested its therapeutic potential for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Xianjing Wei
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116023, P.R. China
| | - Ying Zhang
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116023, P.R. China
| | - Lianna Xie
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116023, P.R. China
| | - Kaijun Wang
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116023, P.R. China
| | - Xiaoqing Wang
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116023, P.R. China
| |
Collapse
|
54
|
Khan A, Paneni F, Jandeleit-Dahm K. Cell-specific epigenetic changes in atherosclerosis. Clin Sci (Lond) 2021; 135:1165-1187. [PMID: 33988232 PMCID: PMC8314213 DOI: 10.1042/cs20201066] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/08/2021] [Accepted: 04/27/2021] [Indexed: 12/28/2022]
Abstract
Atherosclerosis is a disease of large and medium arteries that can lead to life-threatening cerebrovascular and cardiovascular consequences such as heart failure and stroke and is a major contributor to cardiovascular-related mortality worldwide. Atherosclerosis development is a complex process that involves specific structural, functional and transcriptional changes in different vascular cell populations at different stages of the disease. The application of single-cell RNA sequencing (scRNA-seq) analysis has discovered not only disease-related cell-specific transcriptomic profiles but also novel subpopulations of cells once thought as homogenous cell populations. Vascular cells undergo specific transcriptional changes during the entire course of the disease. Epigenetics is the instruction-set-architecture in living cells that defines and maintains the cellular identity by regulating the cellular transcriptome. Although different cells contain the same genetic material, they have different epigenomic signatures. The epigenome is plastic, dynamic and highly responsive to environmental stimuli. Modifications to the epigenome are driven by an array of epigenetic enzymes generally referred to as writers, erasers and readers that define cellular fate and destiny. The reversibility of these modifications raises hope for finding novel therapeutic targets for modifiable pathological conditions including atherosclerosis where the involvement of epigenetics is increasingly appreciated. This article provides a critical review of the up-to-date research in the field of epigenetics mainly focusing on in vivo settings in the context of the cellular role of individual vascular cell types in the development of atherosclerosis.
Collapse
Affiliation(s)
- Abdul Waheed Khan
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
| | - Francesco Paneni
- Cardiovascular Epigenetics and Regenerative Medicine, Centre for Molecular Cardiology, University of Zurich, Switzerland
| | - Karin A.M. Jandeleit-Dahm
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
- German Diabetes Centre, Leibniz Centre for Diabetes Research at the Heinrich Heine University, Dusseldorf, Germany
| |
Collapse
|
55
|
Cortese R. Epigenetics of Sleep Disorders: An Emerging Field in Diagnosis and Therapeutics. Diagnostics (Basel) 2021; 11:diagnostics11050851. [PMID: 34068472 PMCID: PMC8150507 DOI: 10.3390/diagnostics11050851] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 12/16/2022] Open
Affiliation(s)
- Rene Cortese
- Department of Child Health, Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
56
|
Isik B, Thaler R, Goksu BB, Conley SM, Al-Khafaji H, Mohan A, Afarideh M, Abumoawad AM, Zhu XY, Krier JD, Saadiq IM, Tang H, Eirin A, Hickson LJ, van Wijnen AJ, Textor SC, Lerman LO, Herrmann SM. Hypoxic preconditioning induces epigenetic changes and modifies swine mesenchymal stem cell angiogenesis and senescence in experimental atherosclerotic renal artery stenosis. Stem Cell Res Ther 2021; 12:240. [PMID: 33853680 PMCID: PMC8048283 DOI: 10.1186/s13287-021-02310-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/25/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Atherosclerotic renal artery stenosis (ARAS) is a risk factor for ischemic and hypertensive kidney disease (HKD) for which autologous mesenchymal stem cell (MSC) appears to be a promising therapy. However, MSCs from ARAS patients exhibit impaired function, senescence, and DNA damage, possibly due to epigenetic mechanisms. Hypoxia preconditioning (HPC) exerts beneficial effects on cellular proliferation, differentiation, and gene and protein expression. We hypothesized that HPC could influence MSC function and senescence in ARAS by epigenetic mechanisms and modulating gene expression of chromatin-modifying enzymes. METHODS Adipose-derived MSC harvested from healthy control (N = 8) and ARAS (N = 8) pigs were cultured under normoxia (20%O2) or hypoxia (1%O2) conditions. MSC function was assessed by migration, proliferation, and cytokine release in conditioned media. MSC senescence was evaluated by SA-β-gal activity. Specific pro-angiogenic and senescence genes were assessed by reverse transcription polymerase chain reaction (RT-PCR). Dot blotting was used to measure global genome 5-hydroxymethylcytosine (5hmC) levels on DNA and Western blotting of modified histone 3 (H3) proteins to quantify tri-methylated lysine-4 (H3K4me3), lysine-9 (H3K9me3), and lysine-27 (H3K27me3) residues. RESULTS Specific pro-angiogenic genes in ARAS assessed by RT-PCR were lower at baseline but increased under HPC, while pro-senescence genes were higher in ARAS at baseline as compared healthy MSCs. ARAS MSCs under basal conditions, displayed higher H3K4me3, H3K27me3, and 5hmC levels compared to healthy MSCs. During HPC, global 5hmC levels were decreased while no appreciable changes occurred in histone H3 tri-methylation. ARAS MSCs cultured under HPC had higher migratory and proliferative capacity as well as increased vascular endothelial growth factor and epidermal growth factor expression compared to normoxia, and SA-β-gal activity decreased in both animal groups. CONCLUSIONS These data demonstrate that swine ARAS MSCs have decreased angiogenesis and increased senescence compared to healthy MSCs and that HPC mitigates MSC dysfunction, senescence, and DNA hydroxymethylation in ARAS MSC. Thus, HPC for MSCs may be considered for their optimization to improve autologous cell therapy in patients with nephropathies.
Collapse
Affiliation(s)
- Busra Isik
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, MN, 55902, USA
| | - Roman Thaler
- Department of Biochemistry and Molecular Biology, Rochester, USA
- Department of Orthopedics, Rochester, USA
| | - Busra B Goksu
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, MN, 55902, USA
| | - Sabena M Conley
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, FL, USA
| | - Hayder Al-Khafaji
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, MN, 55902, USA
| | - Arjunmohan Mohan
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, MN, 55902, USA
| | - Mohsen Afarideh
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, MN, 55902, USA
| | - Abdelrhman M Abumoawad
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, MN, 55902, USA
| | - Xiang Y Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, MN, 55902, USA
| | - James D Krier
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, MN, 55902, USA
| | - Ishran M Saadiq
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, MN, 55902, USA
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, MN, 55902, USA
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, MN, 55902, USA
| | - LaTonya J Hickson
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, FL, USA
| | - Andre J van Wijnen
- Department of Biochemistry and Molecular Biology, Rochester, USA
- Department of Orthopedics, Rochester, USA
| | - Stephen C Textor
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, MN, 55902, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, MN, 55902, USA
| | - Sandra M Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, MN, 55902, USA.
| |
Collapse
|
57
|
Ciccone V, Genah S, Morbidelli L. Endothelium as a Source and Target of H 2S to Improve Its Trophism and Function. Antioxidants (Basel) 2021; 10:antiox10030486. [PMID: 33808872 PMCID: PMC8003673 DOI: 10.3390/antiox10030486] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/13/2021] [Accepted: 03/16/2021] [Indexed: 12/18/2022] Open
Abstract
The vascular endothelium consists of a single layer of squamous endothelial cells (ECs) lining the inner surface of blood vessels. Nowadays, it is no longer considered as a simple barrier between the blood and vessel wall, but a central hub to control blood flow homeostasis and fulfill tissue metabolic demands by furnishing oxygen and nutrients. The endothelium regulates the proper functioning of vessels and microcirculation, in terms of tone control, blood fluidity, and fine tuning of inflammatory and redox reactions within the vessel wall and in surrounding tissues. This multiplicity of effects is due to the ability of ECs to produce, process, and release key modulators. Among these, gasotransmitters such as nitric oxide (NO) and hydrogen sulfide (H2S) are very active molecules constitutively produced by endotheliocytes for the maintenance and control of vascular physiological functions, while their impairment is responsible for endothelial dysfunction and cardiovascular disorders such as hypertension, atherosclerosis, and impaired wound healing and vascularization due to diabetes, infections, and ischemia. Upregulation of H2S producing enzymes and administration of H2S donors can be considered as innovative therapeutic approaches to improve EC biology and function, to revert endothelial dysfunction or to prevent cardiovascular disease progression. This review will focus on the beneficial autocrine/paracrine properties of H2S on ECs and the state of the art on H2S potentiating drugs and tools.
Collapse
|
58
|
The Therapeutic Potential of Epigenome-Modifying Drugs in Cardiometabolic Disease. CURRENT GENETIC MEDICINE REPORTS 2021. [DOI: 10.1007/s40142-021-00198-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
59
|
Li Y, Zhang YX, Ning DS, Chen J, Li SX, Mo ZW, Peng YM, He SH, Chen YT, Zheng CJ, Gao JJ, Yuan HX, Ou JS, Ou ZJ. Simvastatin inhibits POVPC-mediated induction of endothelial-to-mesenchymal cell transition. J Lipid Res 2021; 62:100066. [PMID: 33711324 PMCID: PMC8063863 DOI: 10.1016/j.jlr.2021.100066] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 02/22/2021] [Accepted: 03/05/2021] [Indexed: 11/16/2022] Open
Abstract
Endothelial-to-mesenchymal transition (EndMT), the process by which an endothelial cell (EC) undergoes a series of molecular events that result in a mesenchymal cell phenotype, plays an important role in atherosclerosis. 1-Palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphocholine (POVPC), derived from the oxidation of 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphatidylcholine, is a proinflammatory lipid found in atherosclerotic lesions. Whether POVPC promotes EndMT and how simvastatin influences POVPC-mediated EndMT remains unclear. Here, we treated human umbilical vein ECs with POVPC, simvastatin, or both, and determined their effect on EC viability, morphology, tube formation, proliferation, and generation of NO and superoxide anion (O2•-). Expression of specific endothelial and mesenchymal markers was detected by immunofluorescence and immunoblotting. POVPC did not affect EC viability but altered cellular morphology from cobblestone-like ECs to a spindle-like mesenchymal cell morphology. POVPC increased O2- generation and expression of alpha-smooth muscle actin, vimentin, Snail-1, Twist-1, transforming growth factor-beta (TGF-β), TGF-β receptor II, p-Smad2/3, and Smad2/3. POVPC also decreased NO production and expression of CD31 and endothelial NO synthase. Simvastatin inhibited POVPC-mediated effects on cellular morphology, production of O2•- and NO, and expression of specific endothelial and mesenchymal markers. These data demonstrate that POVPC induces EndMT by increasing oxidative stress, which stimulates TGF-β/Smad signaling, leading to Snail-1 and Twist-1 activation. Simvastatin inhibited POVPC-induced EndMT by decreasing oxidative stress, suppressing TGF-β/Smad signaling, and inactivating Snail-1 and Twist-1. Our findings reveal a novel mechanism of atherosclerosis that can be inhibited by simvastatin.
Collapse
Affiliation(s)
- Yan Li
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Yi-Xin Zhang
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Da-Sheng Ning
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Jing Chen
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; Division of Hypertension and Vascular Diseases, Department of Cardiology, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Shang-Xuan Li
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Zhi-Wei Mo
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Yue-Ming Peng
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Shi-Hui He
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Ya-Ting Chen
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Chun-Juan Zheng
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Jian-Jun Gao
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Hao-Xiang Yuan
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Jing-Song Ou
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China.
| | - Zhi-Jun Ou
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; Division of Hypertension and Vascular Diseases, Department of Cardiology, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
60
|
Abstract
Atherosclerotic cardiovascular disease (ASCVD) proceeds through a series of stages: initiation, progression (or regression), and complications. By integrating known biology regarding molecular signatures of each stage with recent advances in high-dimensional molecular data acquisition platforms (to assay the genome, epigenome, transcriptome, proteome, metabolome, and gut microbiome), snapshots of each phase of atherosclerotic cardiovascular disease development can be captured. In this review, we will summarize emerging approaches for assessment of atherosclerotic cardiovascular disease risk in humans using peripheral blood molecular signatures and molecular imaging approaches. We will then discuss the potential (and challenges) for these snapshots to be integrated into a personalized movie providing dynamic readouts of an individual's atherosclerotic cardiovascular disease risk status throughout the life course.
Collapse
Affiliation(s)
- Matthew Nayor
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Kemar J. Brown
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Ramachandran S. Vasan
- Sections of Preventive Medicine & Epidemiology, and Cardiology, Department of Medicine, Boston University School of Medicine, Boston, MA; Department of Epidemiology, Boston University School of Public Health; Boston University Center for Computing and Data Sciences
| |
Collapse
|
61
|
Abstract
Emerging evidence suggests the growing importance of "nongenetic factors" in the pathogenesis of atherosclerotic vascular disease. Indeed, the inherited genome determines only part of the risk profile as genomic approaches do not take into account additional layers of biological regulation by "epi"-genetic changes. Epigenetic modifications are defined as plastic chemical changes of DNA/histone complexes which critically affect gene activity without altering the DNA sequence. These modifications include DNA methylation, histone posttranslational modifications, and non-coding RNAs and have the ability to modulate gene expression at both transcriptional and posttranscriptional level. Notably, epigenetic signals are mainly induced by environmental factors (i.e., pollution, smoking, noise) and, once acquired, may be transmitted to the offspring. The inheritance of adverse epigenetic changes may lead to premature deregulation of pathways involved in vascular damage and endothelial dysfunction. Here, we describe the emerging role of epigenetic modifications as fine-tuners of gene transcription in atherosclerosis. Specifically, the following aspects are described in detail: (1) discovery and impact of the epigenome in cardiovascular disease, (2) the epigenetic landscape in atherosclerosis; (3) inheritance of epigenetic signals and premature vascular disease; (4) epigenetic control of lipid metabolism, vascular oxidative stress, inflammation, autophagy, and apoptosis; (5) epigenetic biomarkers in patients with atherosclerosis; (6) novel therapeutic strategies to modulate epigenetic marks. Understanding the individual epigenetic profile may pave the way for new approaches to determine cardiovascular risk and to develop personalized therapies to treat atherosclerosis and its complications.
Collapse
|
62
|
Piko N, Bevc S, Hojs R, Naji FH, Ekart R. The association between pulse wave analysis, carotid-femoral pulse wave velocity and peripheral arterial disease in patients with ischemic heart disease. BMC Cardiovasc Disord 2021; 21:33. [PMID: 33441117 PMCID: PMC7807526 DOI: 10.1186/s12872-021-01859-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 12/15/2020] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Functional changes in peripheral arterial disease (PAD) could play a role in higher cardiovascular risk in these patients. METHODS 123 patients who underwent elective coronary angiography were included. Ankle-brachial index (ABI) was measured and arterial stiffness parameters were derived with applanation tonometry. RESULTS 6 patients (4.9%) had a previously known PAD (Rutherford grade I). Mean ABI was 1.04 ± 0.12, mean subendocardial viability ratio (SEVR) 166.6 ± 32.7% and mean carotid-femoral pulse wave velocity (cfPWV) 10.3 ± 2.4 m/s. Most of the patients (n = 81, 65.9%) had coronary artery disease (CAD). There was no difference in ABI among different degrees of CAD. Patients with zero- and three-vessel CAD had significantly lower values of SEVR, compared to patients with one- and two-vessel CAD (159.5 ± 32.9%/158.1 ± 31.5% vs 181.0 ± 35.2%/166.8 ± 27.8%; p = 0.048). No significant difference was observed in cfPWV values. Spearman's correlation test showed an important correlation between ABI and SEVR (r = 0.196; p = 0.037) and between ABI and cfPWV (r = - 0.320; p ≤ 0.001). Multiple regression analysis confirmed an association between cfPWV and ABI (β = - 0.210; p = 0.003), cfPWV and mean arterial pressure (β = 0.064; p < 0.001), cfPWV and age (β = 0.113; p < 0.001) and between cfPWV and body mass index (BMI (β = - 0.195; p = 0.028), but not with arterial hypertension, dyslipidemia, diabetes mellitus or smoking status. SEVR was not statistically significantly associated with ABI using the same multiple regression model. CONCLUSION Reduced ABI was associated with increased cfPWV, but not with advanced CAD or decreased SEVR.
Collapse
Affiliation(s)
- Nejc Piko
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000, Maribor, Slovenia.
| | - Sebastjan Bevc
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000, Maribor, Slovenia.,Medical Faculty Maribor, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia
| | - Radovan Hojs
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000, Maribor, Slovenia.,Medical Faculty Maribor, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia
| | - Franjo Husam Naji
- Department of Cardiology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000, Maribor, Slovenia.,Medical Faculty Maribor, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia
| | - Robert Ekart
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000, Maribor, Slovenia.,Medical Faculty Maribor, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia
| |
Collapse
|
63
|
Tao Z, Cao Z, Wang X, Pan D, Jia Q. Long noncoding RNA SNHG14 regulates ox-LDL-induced atherosclerosis cell proliferation and apoptosis by targeting miR-186-5p/WIPF2 axis. Hum Exp Toxicol 2021; 40:47-59. [PMID: 32735135 DOI: 10.1177/0960327120940363] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
To investigate the role of small nucleolus RNA host gene 14 (SNHG14) in the progression of atherosclerosis (AS), bioinformatics analysis, and other relevant experiments (cell counting kit-8, flow cytometry, quantitative real-time polymerase chain reaction, luciferase reporter, RNA immunoprecipitation, RNA pull-down, and western blot assays) were done. The current study revealed that SNHG14 level was high in the serum of AS patients and oxidized low-density lipoprotein (ox-LDL)-induced AS cell lines. Besides, we found that SNHG14 accelerated cell proliferation while inhibited cell apoptosis in ox-LDL-induced AS cell lines. Next, SNHG14 was confirmed to be a sponge for miR-186-5p in AS cells, and it was validated that SNHG14 regulated AS cell proliferation and apoptosis by sponging miR-186-5p. Moreover, we uncovered that WAS-interacting protein family member 2 (WIPF2) was a downstream target of miR-186-5p in AS cells. Finally, it was demonstrated that miR-186-5p modulated AS cell proliferation and apoptosis via targeting WIPF2. To conclude, our research disclosed that SNHG14 affected ox-LDL-induced AS cell proliferation and apoptosis through miR-186-5p/WIPF2 axis, which may provide a theoretical basis for the treatment and diagnosis of AS.
Collapse
Affiliation(s)
- Z Tao
- Department of Cardiology, 74734The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Z Cao
- Department of Cardiology, 74734The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - X Wang
- Department of Geriatrics, 74734The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - D Pan
- Department of Cardiology, 74734The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Q Jia
- Department of Cardiology, 74734The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
64
|
Chen SY, Chen YZ, Lee YJ, Jiang CL, Lu SC, Lin FJ. Maternal hypercholesterolemia exacerbates atherosclerosis lesions in female offspring through potentiating macrophage polarization toward an inflammatory M1 phenotype. J Nutr Biochem 2020; 90:108575. [PMID: 33387610 DOI: 10.1016/j.jnutbio.2020.108575] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 11/24/2020] [Accepted: 12/03/2020] [Indexed: 10/22/2022]
Abstract
Maternal hypercholesterolemia induces early onset of cardiovascular diseases in offspring; however, its underlying mechanism remains poorly understood. We hypothesized that maternal hypercholesterolemia increases offspring susceptibility to atherosclerosis in adulthood through developmental modifications of macrophages. Female apolipoprotein E (ApoE)-deficient mice were fed a Western-type diet (WD) or a control diet (CD) prior to and throughout gestation and lactation. The offspring were all fed a WD after weaning. Sixteen-week-old female offspring of WD-fed dams showed a significant increase in atherosclerotic lesions of the aorta and aortic root compared with those of CD-fed dams. This effect was associated with increased macrophage accumulation within lesions, macrophage inflammation and an increase in circulating Ly6Chigh monocyte and F4/80 macrophage counts. We further evidenced that in utero WD exposure promoted macrophage polarization toward the M1 phenotype by elevating M1 markers (Cd86, Inos/Nos2) without affecting M2 markers (Cd206, Arg1). Proinflammatory cytokine synthesis was also enhanced in response to LPS. Finally, maternal WD intake strongly inhibited the macrophage expression of Pparg and Lxra, which was associated with aberrant DNA methylation of Lxra promoter. Our findings demonstrate that maternal hypercholesterolemia exacerbates atherosclerosis, in part by altering the epigenetic state of the macrophage genome of the offspring, imprinting gene expression, and changing macrophage polarization, which ultimately contributes to plaque macrophage burden.
Collapse
Affiliation(s)
- Sin-Yu Chen
- Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Yi-Zhen Chen
- Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Yi-Jing Lee
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, Taiwan
| | - Chung-Lin Jiang
- Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Shao-Chun Lu
- Department of Biochemistry and Molecular Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Fu-Jung Lin
- Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan; Research Center for Development Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
65
|
Chu C, Xu G, Li X, Duan Z, Tao L, Cai H, Yang M, Zhang X, Chen B, Zheng Y, Shi H, Li X. Sustained expression of MCP-1 induced low wall shear stress loading in conjunction with turbulent flow on endothelial cells of intracranial aneurysm. J Cell Mol Med 2020; 25:110-119. [PMID: 33332775 PMCID: PMC7810920 DOI: 10.1111/jcmm.15868] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 07/30/2020] [Accepted: 08/24/2020] [Indexed: 12/28/2022] Open
Abstract
Shear stress was reported to regulate the expression of AC007362, but its underlying mechanisms remain to be explored. In this study, to isolate endothelial cells of blood vessels, unruptured and ruptured intracranial aneurysm (IA) tissues were collected from IA patients. Subsequently, quantitative real‐time PCR (qRT‐PCR), Western blot and luciferase assay were performed to investigate the relationships between AC007362, miRNAs‐493 and monocyte chemoattractant protein‐1 (MCP‐1) in human umbilical vein endothelial cells (HUVECs) exposed to shear stress. Reduced representation bisulphite sequencing (RRBS) was performed to assess the level of DNA methylation in AC007362 promoter. Accordingly, AC007362 and MCP‐1 were significantly up‐regulated while miR‐493 was significantly down‐regulated in HUVECs exposed to shear stress. AC007362 could suppress the miR‐493 expression and elevate the MCP‐1 expression, and miR‐493 was shown to respectively target AC007362 and MCP‐1. Moreover, shear stress in HUVECs led to the down‐regulated DNA methyltransferase 1 (DNMT1), as well as the decreased DNA methylation level of AC007362 promoter. Similar results were also observed in ruptured IA tissues when compared with unruptured IA tissues. In conclusion, this study presented a deep insight into the operation of the regulatory network of AC007362, miR‐493 and MCP‐1 upon shear stress. Under shear stress, the expression of AC007362 was enhanced by the inhibited promoter DNA methylation, while the expression of MCP‐1 was enhanced by sponging the expression of miR‐493.
Collapse
Affiliation(s)
- Cheng Chu
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Gang Xu
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Xiaocong Li
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Zuowei Duan
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Lihong Tao
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Hongxia Cai
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Ming Yang
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Xinjiang Zhang
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Bin Chen
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Yanyu Zheng
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Hongcan Shi
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Xiaoyu Li
- Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, China
| |
Collapse
|
66
|
Xue Y, Guo Y, Luo S, Zhou W, Xiang J, Zhu Y, Xiang Z, Shen J. Aberrantly Methylated-Differentially Expressed Genes Identify Novel Atherosclerosis Risk Subtypes. Front Genet 2020; 11:569572. [PMID: 33381146 PMCID: PMC7767999 DOI: 10.3389/fgene.2020.569572] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 11/16/2020] [Indexed: 12/15/2022] Open
Abstract
Increasing evidence has indicated that modulation of epigenetic mechanisms, especially methylation and long-non-coding RNA (lncRNA) regulation, plays a pivotal role in the process of atherosclerosis; however, few studies focused on revealing the epigenetic-related subgroups during atherosclerotic progression using unsupervised clustering analysis. Hence, we aimed to identify the epigenetics-related differentially expressed genes associated with atherosclerosis subtypes and characterize their clinical utility in atherosclerosis. Eighty samples with expression data (GSE40231) and 49 samples with methylation data (GSE46394) from a large artery plaque were downloaded from the GEO database, and aberrantly methylated-differentially expressed (AMDE) genes were identified based on the relationship between methylation and expression. Furthermore, we conducted weighted correlation network analysis (WGCNA) and co-expression analysis to identify the core AMDE genes strongly involved in atherosclerosis. K-means clustering was used to characterize two subtypes of atherosclerosis in GSE40231, and then 29 samples were recognized as validation dataset (GSE28829). In a blood sample cohort (GSE90074), chi-square test and logistic analysis were performed to explore the clinical implication of the K-means clusters. Furthermore, significance analysis of microarrays and prediction analysis of microarrays (PAM) were applied to identify the signature AMDE genes. Moreover, the classification performance of signature AMDE gene-based classifier from PAM was validated in another blood sample cohort (GSE34822). A total of 1,569 AMDE mRNAs and eight AMDE long non-coding RNAs (lncRNAs) were identified by differential analysis. Through the WGCNA and co-expression analysis, 32 AMDE mRNAs and seven AMDE lncRNAs were identified as the core genes involved in atherosclerosis development. Functional analysis revealed that AMDE genes were strongly related to inflammation and axon guidance. In the clinical analysis, the atherosclerotic subtypes were associated with the severity of coronary artery disease and risk of adverse events. Eight genes, including PARP15, SERGEF, PDGFD, MRPL45, UBR1, STAU1, WIZ, and LSM4, were selected as the signature AMDE genes that most significantly differentiated between atherosclerotic subtypes. Ultimately, the area under the curve of signature AMDE gene-based classifier for atherosclerotic subtypes was 0.858 and 0.812 in GSE90074 and GSE34822, respectively. This study identified the AMDE genes (lncRNAs and mRNAs) that could be implemented in clinical clustering to recognize high-risk atherosclerotic patients.
Collapse
Affiliation(s)
- Yuzhou Xue
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yongzheng Guo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Suxin Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Zhou
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Xiang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuansong Zhu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhenxian Xiang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian Shen
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
67
|
Zhong W, Deng Q, Deng X, Zhong Z, Hou J. Long non-coding RNA expression profiles in peripheral blood mononuclear cells of patients with coronary artery disease. J Thorac Dis 2020; 12:6813-6825. [PMID: 33282383 PMCID: PMC7711381 DOI: 10.21037/jtd-20-3105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Background Coronary artery disease (CAD) is a leading cause of death and often presents as a complex systemic disease. The aim of the presents study was to determine the expression profiles of long non-coding RNAs (lncRNAs) in peripheral blood mononuclear cells (PBMC) of CAD patients and controls. Methods The lncRNA expression profiling of PBMC obtained from 40 CAD patients and 10 non-obstructive coronary atherosclerosis (NOCA) subjects were analyzed using the Illumina Hiseq 4000 sequencer. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to validate the differentially expressed lncRNAs. Gene ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of mRNA were conducted to predict biologic functions. Results Our results indicated that lncRNAs were differentially expressed; 83 were upregulated lncRNAs and 114 were downregulated lncRNAs (P<0.05). A horizontal comparison of lncRNA expression indicated that the change in the expression profile of 48 lncRNAs was consistent with the degree of CAD. Six lncRNAs were validated using qRT-PCR, confirming the accuracy of the RNA sequencing analysis. GO analysis indicated that these dysregulated lncRNA transcripts were related to chromatin organization, cell and cell part, and protein heterodimerization activity. Pathway analysis indicated that these differentially expressed genes mainly included viral carcinogenesis, systemic lupus erythematosus and alcoholism. Conclusions Our preliminary findings indicate that lncRNAs could be used as potential biomarkers of subclinical cardiac abnormalities in the PBMC of CAD patients. However, further studies are needed to verify our findings and hypothesis.
Collapse
Affiliation(s)
- Wei Zhong
- Center for Cardiovascular Diseases, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, China.,Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China
| | - Qiaoting Deng
- Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China.,Research and Experimental Center, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Yat-sen University, Meizhou, China
| | - Xunwei Deng
- Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China.,Research and Experimental Center, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Yat-sen University, Meizhou, China
| | - Zhixiong Zhong
- Center for Cardiovascular Diseases, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, China.,Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China
| | - Jingyuan Hou
- Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China.,Research and Experimental Center, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Yat-sen University, Meizhou, China
| |
Collapse
|
68
|
Noori NM, shahraki Z, Karimi F, Miri-Moghaddam E. Rs4841587 in GATA4 and rs6999593 in DNMT1 gene associated with congenital heart diseases in the southeast of Iran. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
69
|
Burgos V, Paz C, Saavedra K, Saavedra N, Foglio MA, Salazar LA. Drimenol, isodrimeninol and polygodial isolated from Drimys winteri reduce monocyte adhesion to stimulated human endothelial cells. Food Chem Toxicol 2020; 146:111775. [DOI: 10.1016/j.fct.2020.111775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/17/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022]
|
70
|
Li W, Yu J, Xiao X, Li W, Zang L, Han T, Zhang D, Niu X. The inhibitory effect of (-)-Epicatechin gallate on the proliferation and migration of vascular smooth muscle cells weakens and stabilizes atherosclerosis. Eur J Pharmacol 2020; 891:173761. [PMID: 33249078 DOI: 10.1016/j.ejphar.2020.173761] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/22/2020] [Accepted: 11/23/2020] [Indexed: 02/08/2023]
Abstract
Vascular smooth muscle cells (VSMCs) lesions play an important role in atherosclerosis. The latest findings indicate that green tea extract has potential benefits for patients with atherosclerosis, but the components and mechanisms of action are unknown. (-)-Epicatechin gallate (ECG) is the main active ingredient extracted from green tea and has significant biological functions. However, the mechanism of ECG in atherosclerosis remains unclear. Therefore, we investigated the intervention of ECG on VSMCs induced by oxidized low-density lipoprotein (ox-LDL). The results show that ECG reduces the inflammatory response by preventing the overproduction of inflammatory mediators in VSMCs. ECG regulates the cell cycle and down-regulates the expression of proliferating cell nuclear antigen (PCNA) and cyclinD1, and then exerts an anti-proliferative effect. Furthermore, inhibition of the expression of matrix metalloproteinase 2 (MMP-2) and intercellular adhesion molecule 1 (ICAM-1) may be the mechanism by which ECG inhibits the migration of ox-LDL-induced VSMCs. Oil red O staining results show that ECG can improve cell foaming and reduce the content of total cholesterol (TC). In addition, ECG significantly reduces reactive oxygen species activity and also reduces the expression of p-p38, p-JNK, p-ERK1/2, p-IκBα, p-NF-κBp65, and TLR4. These results indicate that ECG has potential clinical applications for preventing atherosclerosis.
Collapse
Affiliation(s)
- Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Jinjin Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Xin Xiao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Wenqi Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Lulu Zang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Tengfei Han
- Shaanxi Panlong Pharmaceutical Group Limited By Share LTD, Xi'an, PR China
| | - Dezhu Zhang
- Shaanxi Panlong Pharmaceutical Group Limited By Share LTD, Xi'an, PR China
| | - Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China.
| |
Collapse
|
71
|
Gwon MH, Im YS, Seo AR, Kim KY, Moon HR, Yun JM. Phenethyl Isothiocyanate Protects against High Fat/Cholesterol Diet-Induced Obesity and Atherosclerosis in C57BL/6 Mice. Nutrients 2020; 12:nu12123657. [PMID: 33261070 PMCID: PMC7761196 DOI: 10.3390/nu12123657] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/25/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022] Open
Abstract
This study concerns obesity-related atherosclerosis, hyperlipidemia, and chronic inflammation. We studied the anti-obesity and anti-atherosclerosis effects of phenethyl isothiocyanate (PEITC) and explored their underlying mechanisms. We established an animal model of high fat/cholesterol-induced obesity in C57BL/6 mice fed for 13 weeks. We divided the mice into five groups: control (CON), high fat/cholesterol (HFCD), HFCD with 3 mg/kg/day gallic acid (HFCD + G), and HFCD with PEITC (30 and 75 mg/kg/day; HFCD + P30 and P75). The body weight, total cholesterol, and triglyceride were significantly lower in the HFCD + P75 group than in the HFCD group. Hepatic lipid accumulation and atherosclerotic plaque formation in the aorta were significantly lower in both HFCD + PEITC groups than in the HFCD group, as revealed by hematoxylin and eosin (H&E) staining. To elucidate the mechanism, we identified the expression of genes related to inflammation, reverse cholesterol transport, and lipid accumulation pathway in the liver. The expression levels of peroxisome proliferator activated receptor gamma (PPARγ), liver-X-receptor α (LXR-α), and ATP binding cassette subfamily A member 1 (ABCA1) were increased, while those of scavenger receptor A (SR-A1), cluster of differentiation 36 (CD36), and nuclear factor-kappa B (NF-κB) were decreased in the HFCD + P75 group compared with those in the HFCD group. Moreover, PEITC modulated H3K9 and H3K27 acetylation, H3K4 dimethylation, and H3K27 di-/trimethylation in the HFCD + P75 group. We, therefore, suggest that supplementation with PEITC may be a potential candidate for the treatment and prevention of atherosclerosis and obesity.
Collapse
Affiliation(s)
- Min-Hee Gwon
- Nutrition Education Major, Graduate School of Education, Chonnam National University, Gwangju 61186, Korea;
| | - Young-Sun Im
- Department of Food and Nutrition, Chonnam National University, Gwangju 61186, Korea; (Y.-S.I.); (A.-R.S.); (K.Y.K.); (H.-R.M.)
| | - A-Reum Seo
- Department of Food and Nutrition, Chonnam National University, Gwangju 61186, Korea; (Y.-S.I.); (A.-R.S.); (K.Y.K.); (H.-R.M.)
| | - Kyoung Yun Kim
- Department of Food and Nutrition, Chonnam National University, Gwangju 61186, Korea; (Y.-S.I.); (A.-R.S.); (K.Y.K.); (H.-R.M.)
| | - Ha-Rin Moon
- Department of Food and Nutrition, Chonnam National University, Gwangju 61186, Korea; (Y.-S.I.); (A.-R.S.); (K.Y.K.); (H.-R.M.)
| | - Jung-Mi Yun
- Department of Food and Nutrition, Chonnam National University, Gwangju 61186, Korea; (Y.-S.I.); (A.-R.S.); (K.Y.K.); (H.-R.M.)
- Correspondence: ; Tel.: +82-62-530-1332
| |
Collapse
|
72
|
Hou H, Zhao H. Epigenetic factors in atherosclerosis: DNA methylation, folic acid metabolism, and intestinal microbiota. Clin Chim Acta 2020; 512:7-11. [PMID: 33232735 DOI: 10.1016/j.cca.2020.11.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/07/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022]
Abstract
Atherosclerosis is a complex disease, influenced by both genetic and non-genetic factors. The most important epigenetic mechanism in the pathogenesis of atherosclerosis is DNA methylation, which involves modification of the gene without changes in the gene sequence. Nutrients involved in one-carbon metabolism interact to regulate DNA methylation, especially folic acid and B vitamins. Deficiencies in folic acid and other nutrients, such as vitamins B6 and B12, can increase homocysteine levels, induce endothelial dysfunction, and accelerate atherosclerotic pathological processes. Supplemented nutrients can improve DNA methylation status, reduce levels of inflammatory factors, and delay the process of atherosclerosis. In this review, the influence of intestinal flora on folate metabolism and epigenetics is also considered.
Collapse
Affiliation(s)
- Huimin Hou
- Department of Geriatrics, The First Hospital of Jilin University, Changchun 130021, China
| | - Huiying Zhao
- Department of Geriatrics, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
73
|
Li P, Xing J, Zhang J, Jiang J, Liu X, Zhao D, Zhang Y. Inhibition of long noncoding RNA HIF1A-AS2 confers protection against atherosclerosis via ATF2 downregulation. J Adv Res 2020; 26:123-135. [PMID: 33133688 PMCID: PMC7584671 DOI: 10.1016/j.jare.2020.07.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 01/17/2023] Open
Abstract
Introduction In atherosclerotic lesions, extensive inflammation of the vessel wall contributes to plaque instability. Long noncoding RNAs (lncRNAs) play important roles in diverse biological processes in atherosclerosis. Objectives Here, we aim to identify the functional role and regulatory mechanisms of lncRNA hypoxia-inducible factor 1 alpha-antisense RNA 2 (HIF1A-AS2) in atherosclerotic inflammation. Methods An atherosclerotic mouse model was induced in ApoE-/- mice by high fat diet (HFD). Endothelial cells (ECs), human aortic smooth muscle cells (SMCs) or human coronary artery endothelial cells (HCAECs) were exposed to ox-LDL to develop the in vitro model. The effects of lncRNA HIF1A-AS2 on inflammation were evaluated by determining levels of inflammatory factors tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6) and levels of adhesion molecules vascular cell adhesion molecule 1 (VCAM-1), intercellular adhesion molecule 1 (ICAM-1), and macrophage cationic peptide 1 (MCP-1). Results It was established that lncRNA HIF1A-AS2 and ATF2 were highly expressed in atherosclerotic ApoE-/- mice. Downregulating lncRNA HIF1A-AS2 in ox-LDL-exposed ECs, SMCs and HCAECs inhibited inflammation by reducing levels of pro-inflammatory factors and adhesion molecules. LncRNA HIF1A-AS2 bound to the transcription factor USF1 to elevate ATF2 expression. USF1 overexpression counteracted the suppressive effect of lncRNA HIF1A-AS2 silencing on ox-LDL-induced inflammation. Knockdown of lncRNA HIF1A-AS2 or ATF2 could also attenuate inflammation in atherosclerotic mice. Collectively, the present study demonstrates that downregulation of lncRNA HIF1A-AS2 represses the binding of USF1 to the ATF2 promoter region and then inhibits ATF2 expression, thereby suppressing atherosclerotic inflammation. Conclusion This study suggests lncRNA HIF1A-AS2 as an promising therapeutic target for atherosclerosis.
Collapse
Key Words
- ATCC, American Type Culture Collection
- ATF2, activating transcription factor 2
- Activating transcription factor
- Atherosclerosis
- CAD, coronary artery disease
- CCK-8, cell counting kit-8
- ChIP, Chromatin immunoprecipitation
- DMEM, Dulbecco’s modified Eagle’s medium
- ECs, endothelial cells
- ELISA, enzyme linked immunosorbent assay
- GAPDH, Glyceraldehyde-3-phosphate dehydrogenase
- HCAECs, human coronary artery endothelial cells
- HE, Hematoxylin-eosin
- HFD, high fat diet
- HIF1A-AS2, hypoxia-inducible factor 1 alpha-antisense RNA 2
- Hypoxia-inducible factor 1 alpha-antisense RNA 2
- ICAM-1, intercellular adhesion molecule-1
- IL-1β, interleukin-1β
- IL-6, interleukin-6
- IgG, immunoglobulin G
- Inflammation
- LDL, low-density lipoprotein
- Long noncoding RNA
- MCP-1, monocyte chemoattractant protein-1
- ND, normal diet
- PBS, phosphate buffered saline
- RIP, RNA binding protein immunoprecipitation
- RT-qPCR, reverse transcription quantitative polymerase chain reaction
- SMCs, smooth muscle cells
- TNF-α, tumor necrosis factor-α
- Transcription factor
- USF1, upstream stimulatory factor 1
- Upstream transcription factor 1
- VCAM-1, vascular cell adhesion molecule 1
- lncRNAs, long noncoding RNAs
- ox-LDL, oxidized-low-density lipoprotein
- sh, short hairpin RNA
- si-NC, small interfering RNA-negative control
Collapse
Affiliation(s)
- Pengcheng Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Junhui Xing
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Jielei Zhang
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Jianwu Jiang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Xuemeng Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Di Zhao
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
- Corresponding authors at: Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou 450052, Henan Province, PR China (D. Zhao). Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou 450052, Henan Province, PR China (Y. Zhang).
| | - Yanzhou Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
- Corresponding authors at: Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou 450052, Henan Province, PR China (D. Zhao). Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou 450052, Henan Province, PR China (Y. Zhang).
| |
Collapse
|
74
|
Quiles-Jiménez A, Gregersen I, Mittelstedt Leal de Sousa M, Abbas A, Kong XY, Alseth I, Holm S, Dahl TB, Skagen K, Skjelland M, Aukrust P, Bjørås M, Halvorsen B. N6-methyladenosine in RNA of atherosclerotic plaques: An epitranscriptomic signature of human carotid atherosclerosis. Biochem Biophys Res Commun 2020; 533:631-637. [PMID: 33004177 DOI: 10.1016/j.bbrc.2020.09.057] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 09/16/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND More than 170 post-transcriptional RNA modifications regulate the localization, processing and function of cellular RNAs, and aberrant RNA modifications have been linked to a range of human diseases. The RNA modification landscape in atherosclerosis, the main underlying cause of cardiovascular diseases, is still largely unknown. METHODS We used mass spectrometry to analyse a selection of RNA-modifying enzymes and the N6-methyladenosine (m6A) in carotid atherosclerotic lesion samples representing early and advanced stages of atherosclerosis as compared to non-atherosclerotic arteries from healthy controls. FINDINGS (i) the detection of different levels of several enzymes involved in methylations occurring in rRNA and mRNA; (ii) these findings included changes in the levels of methyltransferases ('writers'), binding proteins ('readers') and demethylases ('erasers') during atherosclerosis as compared to non-atherosclerotic control arteries, with generally the most prominent differences in samples from early atherosclerotic lesions; and (iii) these changes were accompanied by a marked downregulation of m6A in rRNA, the most abundant and well-studied modification in mRNA with a wide range of effects on cell biology. INTERPRETATION We show for the first time that RNA-modifying enzymes and the well-studied RNA modification m6A are differentially regulated in atherosclerotic lesions, which potentially could help creating new prognostic and treatment strategies.
Collapse
Affiliation(s)
- Ana Quiles-Jiménez
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Faculty of Medicine, University of Oslo, Norway
| | - Ida Gregersen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Faculty of Medicine, University of Oslo, Norway
| | - Mirta Mittelstedt Leal de Sousa
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway; PROMEC Core Facility for Proteomics and Metabolomics, NTNU and the Central Norway Regional Health Authority, Trondheim, Norway
| | - Azhar Abbas
- Department of Neurology, Østfold Hospital Trust Kalnes, Grålum, Norway
| | - Xiang Yi Kong
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway
| | - Ingrun Alseth
- Department of Microbiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Sverre Holm
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway
| | - Tuva B Dahl
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway
| | - Karolina Skagen
- Department of Neurology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Mona Skjelland
- Department of Neurology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Norway
| | - Magnar Bjørås
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway; PROMEC Core Facility for Proteomics and Metabolomics, NTNU and the Central Norway Regional Health Authority, Trondheim, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Faculty of Medicine, University of Oslo, Norway.
| |
Collapse
|
75
|
Zhang C, Chen X, Wang JK, Li Y, Cui SJ, Wang Z, Luo T. Phenotypic Switching of Atherosclerotic Smooth Muscle Cells is Regulated by Activated PARP1-Dependent TET1 Expression. J Atheroscler Thromb 2020; 28:716-729. [PMID: 32981917 PMCID: PMC8265424 DOI: 10.5551/jat.55343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aim:
During the development of atherosclerosis, the vascular smooth muscle cells (SMCs) undergo phenotypic switching from contractile phenotype to synthetic phenotype. This study aimed at examining the role of DNA modification mediated by the oxidative stress dependent ten eleven translocation enzymes (TETs) expression at early stage of phenotypic switching.
Methods:
Based on the
in vitro
SMCs calcification model, DNA damage, phenotypic switching and 5-hydroxymethylcytosine (5hmC) were examined by comet assay, alkaline DNA unwinding assay, immunofluorescence staining, Dot blotting and Western blotting. Then Western blotting and qRT-PCR were performed to analyze the TETs expression and the relationship between the activity of poly(ADP-ribose) polymerase 1 (PARP1) and TETs expression. We further alter 5hmC modification by inhibition of TET1 or PARP1 to rescue the phenotypic switching of SMCs using immunofluorescence staining, Dot blotting and qRT-PCR. We performed immunochemistry staining to examine the activated PARP1-TET1 pathway
in vivo
.
Results:
The phenotypic switching was observed in the SMCs cultured with calcification medium as the expression of the cell markers of contractile SMCs decreased and cell proliferation increased. In contrast, PAR and 5hmC were markedly increased in SMCs with calcification due to DNA damage. Our study further demonstrated that oxidative stress-activated PARP1, promotes TET1 expression and 5hmC increase during the phenotypic switching. Inhibition of TET1 or PARP1 can rescue the phenotypic switching of SMCs with calcification.
Conclusion:
Our study demonstrated the important role of PARylation dependent 5hmC, in SMCs phenotypic switching. It raises the possibility to target TET1 and PARP1 for atherosclerosis treatment.
Collapse
Affiliation(s)
- Chao Zhang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University
| | - Xin Chen
- Department of General Surgery, Xuanwu Hospital, Capital Medical University
| | - Ju-Kun Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University
| | - Yu Li
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University
| | - Shi-Jun Cui
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University
| | - Zhonggao Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University.,Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University
| | - Tao Luo
- Department of General Surgery, Xuanwu Hospital, Capital Medical University
| |
Collapse
|
76
|
Lee HT, Oh S, Ro DH, Yoo H, Kwon YW. The Key Role of DNA Methylation and Histone Acetylation in Epigenetics of Atherosclerosis. J Lipid Atheroscler 2020; 9:419-434. [PMID: 33024734 PMCID: PMC7521974 DOI: 10.12997/jla.2020.9.3.419] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 12/17/2022] Open
Abstract
Atherosclerosis, which is the most common chronic disease of the coronary artery, constitutes a vascular pathology induced by inflammation and plaque accumulation within arterial vessel walls. Both DNA methylation and histone modifications are epigenetic changes relevant for atherosclerosis. Recent studies have shown that the DNA methylation and histone modification systems are closely interrelated and mechanically dependent on each other. Herein, we explore the functional linkage between these systems, with a particular emphasis on several recent findings suggesting that histone acetylation can help in targeting DNA methylation and that DNA methylation may control gene expression during atherosclerosis.
Collapse
Affiliation(s)
- Han-Teo Lee
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science, Seoul National University, Seoul, Korea.,Interdisciplinary Program in Stem Cell Biology, Graduate School of Medicine, Seoul National University, Seoul, Korea
| | - Sanghyeon Oh
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science, Seoul National University, Seoul, Korea.,Interdisciplinary Program in Stem Cell Biology, Graduate School of Medicine, Seoul National University, Seoul, Korea
| | - Du Hyun Ro
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science, Seoul National University, Seoul, Korea.,Interdisciplinary Program in Stem Cell Biology, Graduate School of Medicine, Seoul National University, Seoul, Korea.,Department of Orthopedic Surgery, Seoul National University Hospital, Seoul, Korea
| | - Hyerin Yoo
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science, Seoul National University, Seoul, Korea.,Interdisciplinary Program in Stem Cell Biology, Graduate School of Medicine, Seoul National University, Seoul, Korea
| | - Yoo-Wook Kwon
- Strategic Center of Cell and Bio Therapy for Heart, Diabetes & Cancer, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,Department of Medicine, College of Medicine, Seoul National University, Seoul, Korea
| |
Collapse
|
77
|
Li N, Liu S, Zhang Y, Yu L, Hu Y, Wu T, Fang M, Xu Y. Transcriptional Activation of Matricellular Protein Spondin2 (SPON2) by BRG1 in Vascular Endothelial Cells Promotes Macrophage Chemotaxis. Front Cell Dev Biol 2020; 8:794. [PMID: 32974343 PMCID: PMC7461951 DOI: 10.3389/fcell.2020.00794] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 07/28/2020] [Indexed: 12/18/2022] Open
Abstract
The matricellular protein SPON2 plays diverse roles in the development of cardiovascular diseases. SPON2 is expressed in endothelial cells, but its transcription regulation in the context of atherogenesis remains incompletely appreciated. Here we report that SPON2 expression was up-regulated by pro-atherogenic stimuli (oxLDL and TNF-α) in vascular endothelia cells. In addition, endothelial SPON2 was elevated in Apoe–/– mice fed on a Western diet compared to the control mice. Induction of SPON2 in endothelial cells by pro-atherogenic stimuli was mediated by BRG1, a chromatin remodeling protein, both in vitro and in vivo. Further analysis revealed that BRG1 interacted with the sequence-specific transcription factor Egr-1 to activate SPON2 transcription. BRG1 contributed to SPON2 trans-activation by modulating chromatin structure surrounding the SPON2 promoter. Functionally, activation of SPON2 transcription by the Egr-1/BRG1 complex provided chemoattractive cues for macrophage trafficking. SPON2 depletion abrogated the ability of BRG1 or Egr-1 to stimulate endothelial derived chemoattractive cue for macrophage migration. On the contrary, recombinant SPON2 rescued endothelial chemo-attractability in the absence of BRG1 or Egr-1. In conclusion, our data have identified a novel transcriptional cascade in endothelial cells that may potentially promote macrophage recruitment and vascular inflammation leading to atherogenesis.
Collapse
Affiliation(s)
- Nan Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Shuai Liu
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research and Key Laboratory of Emergency and Trauma of Ministry of Education, Institute of Cardiovascular Research of the First Affiliated Hospital, Hainan Medical University, Haikou, China.,Department of Cardiology, Kaifeng People's Hospital, Kaifeng, China
| | - Yuanyuan Zhang
- Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Liming Yu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Yanjiang Hu
- Department of Cardiothoracic Surgery, Liyang People's Hospital, Liyang, China
| | - Teng Wu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Mingming Fang
- Department of Clinical Medicine and Laboratory Center for Experimental Medicine, Jiangsu Health Vocational Institute, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| |
Collapse
|
78
|
Li SY, Yoshida Y, Kobayashi E, Adachi A, Hirono S, Matsutani T, Mine S, Machida T, Ohno M, Nishi E, Maezawa Y, Takemoto M, Yokote K, Kitamura K, Sumazaki M, Ito M, Shimada H, Takizawa H, Kashiwado K, Tomiyoshi G, Shinmen N, Nakamura R, Kuroda H, Zhang XM, Wang H, Goto K, Iwadate Y, Hiwasa T. Association between serum anti‑ASXL2 antibody levels and acute ischemic stroke, acute myocardial infarction, diabetes mellitus, chronic kidney disease and digestive organ cancer, and their possible association with atherosclerosis and hypertension. Int J Mol Med 2020; 46:1274-1288. [PMID: 32945427 PMCID: PMC7447314 DOI: 10.3892/ijmm.2020.4690] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/29/2020] [Indexed: 11/09/2022] Open
Abstract
The aim of the present study was to identify novel antibody markers for the early diagnosis of atherosclerosis in order to improve the prognosis of patients at risk for acute ischemic stroke (AIS) and acute myocardial infarction (AMI). A first screening involved the serological identification of antigens by recombinant cDNA expression cloning and identified additional sex combs-like 2 (ASXL2) as a target antigen recognized by serum IgG antibodies in the sera of patients with atherosclerosis. Antigens, including the recombinant glutathione S-transferasefused ASXL2 protein and its synthetic peptide were then prepared to examine serum antibody levels. Amplified luminescence proximity homoge-neous assay-linked immunosorbent assay, which incorporates glutathione-donor beads and anti-human-IgG-acceptor beads, revealed significantly higher serum antibody levels against the ASXL2 protein and its peptide in the patients with AIS, diabetes mellitus, AMI, chronic kidney disease, esophageal squamous cell carcinoma, or colorectal carcinoma compared with those in healthy donors. The ASXL2 antibody levels were well associated with hypertension complication, but not with sex, body mass index, habitual smoking, or alcohol intake. These results suggest that the serum ASXL2 antibody marker can discriminate between hypertension-induced atherosclerotic AIS and AMI, as well as a number of digestive organ cancers.
Collapse
Affiliation(s)
- Shu-Yang Li
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260‑8670, Japan
| | - Yoichi Yoshida
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260‑8670, Japan
| | - Eiichi Kobayashi
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260‑8670, Japan
| | - Akihiko Adachi
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260‑8670, Japan
| | - Seiichiro Hirono
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260‑8670, Japan
| | - Tomoo Matsutani
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260‑8670, Japan
| | - Seiichiro Mine
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260‑8670, Japan
| | - Toshio Machida
- Department of Neurological Surgery, Chiba Cerebral and Cardiovascular Center, Chiba 290‑0512, Japan
| | - Mikiko Ohno
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Eiichiro Nishi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Yoshiro Maezawa
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba 260‑8670, Japan
| | - Minoru Takemoto
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba 260‑8670, Japan
| | - Koutaro Yokote
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba 260‑8670, Japan
| | - Kenichiro Kitamura
- Department of Internal Medicine 3, University of Yamanashi School of Medicine, Yamanashi 409‑3898, Japan
| | - Makoto Sumazaki
- Department of Gastroenterological Surgery and Clinical Oncology, Toho University Graduate School of Medicine, Tokyo 143‑8541, Japan
| | - Masaaki Ito
- Department of Gastroenterological Surgery and Clinical Oncology, Toho University Graduate School of Medicine, Tokyo 143‑8541, Japan
| | - Hideaki Shimada
- Department of Gastroenterological Surgery and Clinical Oncology, Toho University Graduate School of Medicine, Tokyo 143‑8541, Japan
| | - Hirotaka Takizawa
- Port Square Kashiwado Clinic, Kashiwado Memorial Foundation, Chiba 260‑0025, Japan
| | - Koichi Kashiwado
- Department of Neurology, Kashiwado Hospital, Chiba 260‑8656, Japan
| | - Go Tomiyoshi
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260‑8670, Japan
| | - Natsuko Shinmen
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260‑8670, Japan
| | - Rika Nakamura
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260‑8670, Japan
| | - Hideyuki Kuroda
- Medical Project Division, Research Development Center, Fujikura Kasei Co., Saitama 340‑0203, Japan
| | - Xiao-Meng Zhang
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260‑8670, Japan
| | - Hao Wang
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260‑8670, Japan
| | - Kenichiro Goto
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba 260‑8670, Japan
| | - Yasuo Iwadate
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260‑8670, Japan
| | - Takaki Hiwasa
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Chiba 260‑8670, Japan
| |
Collapse
|
79
|
Pierce JB, Feinberg MW. Long Noncoding RNAs in Atherosclerosis and Vascular Injury: Pathobiology, Biomarkers, and Targets for Therapy. Arterioscler Thromb Vasc Biol 2020; 40:2002-2017. [PMID: 32698685 DOI: 10.1161/atvbaha.120.314222] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite major advances in the primary and secondary prevention of atherosclerosis and its risk factors, atherosclerotic cardiovascular disease remains a major clinical and financial burden on individuals and health systems worldwide. In addition, neointima formation and proliferation due to mechanical trauma to the vessel wall during percutaneous coronary interventions can lead to vascular restenosis and limit the longevity and effectiveness of coronary revascularization. Long noncoding RNAs (lncRNAs) have emerged as a novel class of epigenetic regulators with critical roles in the pathogenesis of atherosclerosis and restenosis following vascular injury. Here, we provide an in-depth review of lncRNAs that regulate the development of atherosclerosis or contribute to the pathogenesis of restenosis following mechanical vascular injury. We describe the diverse array of intracellular mechanisms by which lncRNAs exert their regulatory effects. We highlight the utility and challenges of lncRNAs as biomarkers. Finally, we discuss the immense translational potential of lncRNAs and strategies for targeting them therapeutically using oligonucleotide-based therapeutics and novel gene therapy platforms.
Collapse
Affiliation(s)
- Jacob B Pierce
- From the Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.B.P., M.W.F.).,Feinberg School of Medicine, Northwestern University, Chicago, IL (J.B.P.)
| | - Mark W Feinberg
- From the Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.B.P., M.W.F.)
| |
Collapse
|
80
|
Chen WD, Song T, Cao QH, Li R, Wang H, Chen XB, Chen ZT. Atherosclerosis prediction by microarray-based DNA methylation analysis. Exp Ther Med 2020; 20:2863-2869. [PMID: 32765783 DOI: 10.3892/etm.2020.9025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 04/29/2020] [Indexed: 12/23/2022] Open
Abstract
Using a series of DNA methylation analysis, pathogenesis was investigated to identify the specific DNA methylation markers for diagnosing atherosclerosis. Firstly, with the chip platform of Illumina Human Methylation 450 BeadChip, a total of 1,458 CpGs, covering 971 differential methylated genes were extracted with stringent filtering criteria. Secondly, hierarchical clustering as a heat map was used to check on the dependability of differential methylated genes. Thirdly, the related GO terms and pathways were enriched by up- and down-methylated genes, respectively, after verifying the capacity of these differential methylated genes to distinguish between atherosclerosis and healthy controls. In total, 971 differential DNA methylated genes were identified (1,458 CpGs). Several important function regions were also identified, including cell adhesion, PI3K-Akt signaling pathway and transcription from RNA polymerase II promoter. This study indicates that patients with atherosclerosis have high levels of DNA methylation, which is promising for early diagnosis and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Wei-da Chen
- Health Care Department, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China.,Health Care Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| | - Ting Song
- Health Care Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| | - Qiu-Hong Cao
- Department of Anesthesiology, Jinan Center Hospital, Jinan, Shandong 250013, P.R. China
| | - Rui Li
- Health Care Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| | - Hua Wang
- Health Care Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| | - Xiu-Bao Chen
- Health Care Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| | - Ze-Tao Chen
- Health Care Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| |
Collapse
|
81
|
Almeida SO, Ram RJ, Kinninger A, Budoff MJ. Effect of 5-lipoxygenase inhibitor, VIA-2291 (Atreleuton), on epicardial fat volume in patients with recent acute coronary syndrome. J Cardiovasc Comput Tomogr 2020; 14:343-348. [DOI: 10.1016/j.jcct.2019.12.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 12/21/2019] [Accepted: 12/24/2019] [Indexed: 12/01/2022]
|
82
|
Jurcikova-Novotna L, Mrazova L, Mičová K, Friedecký D, Hubacek JA, Poledne R. Global DNA methylation in rats´ liver is not affected by hypercholesterolemic diet. Physiol Res 2020; 69:347-252. [PMID: 32199015 DOI: 10.33549/physiolres.934313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Increased plasma cholesterol levels are listed between the major atherosclerosis risk factors. The final plasma cholesterol levels result from the interplay between the genetic and environmental (diet, physical activity) factors. Little is known, how dietary factor influence epigenetics. We have analyzed, if an over-generation feeding of rat with cholesterol influences total liver-DNA methylation, and if total liver-DNA methylation differ between the different rat strains (Prague hereditary hypercholesterolemic rats, Prague hereditary hypertriglyceridemic rats and Wistar Kyoto rats). The animals were feed with high fat (additional 5 % over normal capacity) high cholesterol (2 %) diet for 14 days. DNA methylation in the liver tissue in different generations was analyzed using the liquid chromatography coupled with tandem mass spectrometry. We have not observed any significant changes in total liver-DNA methylation over the 9 generations of animals feed by fat/cholesterol enriched diet. Additionally, there were no differences in DNA methylation between different rat strains. In animal model, the dietary changes (hypercholesterolemic diet) not significantly influence the total DNA methylation status within the liver.
Collapse
Affiliation(s)
- L Jurcikova-Novotna
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | | | | | | | | | | |
Collapse
|
83
|
Mur J, McCartney DL, Walker RM, Campbell A, Bermingham ML, Morris SW, Porteous DJ, McIntosh AM, Deary IJ, Evans KL, Marioni RE. DNA methylation in APOE: The relationship with Alzheimer's and with cardiovascular health. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2020; 6:e12026. [PMID: 32346601 PMCID: PMC7185210 DOI: 10.1002/trc2.12026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 12/16/2019] [Accepted: 01/01/2020] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Genetic variation in the apolipoprotein E (APOE) gene is associated with Alzheimer's disease (AD) and risk factors for cardiovascular disease (CVD). DNA methylationat APOE has been associated with altered cognition and AD. It is unclear if epigenetic marks could be used for predicting future disease. METHODS We assessed blood-based DNA methylation at 13 CpGs in the APOE gene in 5828 participants from the Generation Scotland (GS) cohort. Using linear mixed models regression, we examined the relationships among APOE methylation, cognition, cholesterol, the family history of AD and the risk for CVD. RESULTS DNA methylation at two CpGs was associated with the ratio of total cholesterol and HDL cholesterol, but not with cognition, family history of AD, or the risk of CVD. DISCUSSION APOE methylation is associated with the levels of blood cholesterol, but there is no evidence for the utility of APOE methylation as a biomarker for predicting AD or CVD.
Collapse
Affiliation(s)
- Jure Mur
- Department of PsychologyUniversity of EdinburghEdinburghUK
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - Daniel L. McCartney
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - Rosie M. Walker
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - Archie Campbell
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - Mairead L. Bermingham
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - Stewart W. Morris
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - David J. Porteous
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - Andrew M. McIntosh
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
- Division of PsychiatryCentre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | - Ian J. Deary
- Department of PsychologyUniversity of EdinburghEdinburghUK
| | - Kathryn L. Evans
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - Riccardo E. Marioni
- Centre for Genomic and Experimental MedicineInstitute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| |
Collapse
|
84
|
Abstract
Cardiac hypertrophy is a significant risk factor for cardiovascular disease, including heart failure, arrhythmia, and sudden death. Cardiac hypertrophy involves both embryonic gene expression and transcriptional reprogramming, which are tightly regulated by epigenetic mechanisms. An increasing number of studies have demonstrated that epigenetics plays an influential role in the occurrence and development of cardiac hypertrophy. Here, we summarize the latest research progress on epigenetics in cardiac hypertrophy involving DNA methylation, histone modification, and non-coding RNA, to help understand the mechanism of epigenetics in cardiac hypertrophy. The expression of both embryonic and functional genes can be precisely regulated by epigenetic mechanisms during cardiac hypertrophy, providing a substantial number of therapeutic targets. Thus, epigenetic treatment is expected to become a novel therapeutic strategy for cardiac hypertrophy. According to the research performed to date, epigenetic mechanisms associated with cardiac hypertrophy remain far from completely understood. Therefore, epigenetic mechanisms require further exploration to improve the prevention, diagnosis, and treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Hao Lei
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Jiahui Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Kaijun Sun
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
85
|
Grbić E, Gorkič N, Pleskovič A, Zorc M, Ljuca F, Gasparini M, Mrđa B, Cilenšek I, Mankoč S, Banach M, Petrovič D, Fras Z. Association between rs2107595 HDAC9 gene polymorphism and advanced carotid atherosclerosis in the Slovenian cohort. Lipids Health Dis 2020; 19:71. [PMID: 32284067 PMCID: PMC7155263 DOI: 10.1186/s12944-020-01255-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 03/31/2020] [Indexed: 12/14/2022] Open
Abstract
Background Histone deacetylase 9 (HDAC9) plays an important role in transcriptional regulation, cell cycle progression and developmental events; moreover, it has been investigated as a candidate gene in a number of conditions, including the onset and progression of atherosclerosis. We hypothesized that the rs2107595 HDAC9 gene polymorphism may be associated with advanced carotid artery disease in a Slovenian cohort. We also investigated the effect of this polymorphism on HDAC9 receptor expression in the internal carotid artery (ICA) specimens obtained by endarterectomy. Methods This case-control study enrolled 619 unrelated Slovenian patients: 311 patients with ICA stenosis > 75% as the study group and 308 patients with ICA stenosis < 50% as the control group. Patient laboratory and clinical data were obtained from the medical records. The rs2107595 polymorphisms were genotyped using TaqMan SNP Genotyping assay. HDAC9 expression was assessed by immunohistochemistry in 30 ICA specimens from patients with ICA atherosclerosis > 75%, and the numerical areal density of HDAC9 positive cells was calculated. Results The occurrence of advanced ICA atherosclerosis in the Slovenian cohort was 3.81 times higher in the codominant genetic model (OR = 3.81, 95%CI = 1.06–13.77, p = 0.04), and 3.10 times higher in the recessive genetic model (OR = 3.10, 95%CI = 1.16–8.27, p = 0.02). In addition, the A allele of rs2107595 was associated with increased HDAC9 expression in the ICA specimens obtained by endarterectomy. Conclusions We observed a significant association between the AA genotype of rs2107595 with the advanced carotid artery disease in our Slovenian cohort, indicating that this polymorphism may be a genetic risk factor for ICA atherosclerosis.
Collapse
Affiliation(s)
- Emin Grbić
- Department of Physiology, Faculty of Medicine, University of Tuzla, Tuzla, Bosnia and Herzegovina
| | - Nataša Gorkič
- International Center for Cardiovascular Diseases MC Medicor d.d, Izola, Slovenia
| | - Aleš Pleskovič
- Department of Cardiology, Division of Medicine, University Medical Centre of Ljubljana, Ljubljana, Slovenia
| | - Marjeta Zorc
- International Center for Cardiovascular Diseases MC Medicor d.d, Izola, Slovenia
| | - Farid Ljuca
- Department of Physiology, Faculty of Medicine, University of Tuzla, Tuzla, Bosnia and Herzegovina
| | - Mladen Gasparini
- Department of Vascular Surgery, General Hospital Izola, Izola, Slovenia
| | - Božidar Mrđa
- Department of Vascular Surgery, University Medical Centre Maribor, Maribor, Slovenia
| | - Ines Cilenšek
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000, Ljubljana, Slovenia
| | - Sara Mankoč
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000, Ljubljana, Slovenia
| | - Maciej Banach
- Cardiovascular Research Centre, University of Zielona-Gora, Zielona Gora, Poland.,Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Daniel Petrovič
- International Center for Cardiovascular Diseases MC Medicor d.d, Izola, Slovenia. .,Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000, Ljubljana, Slovenia.
| | - Zlatko Fras
- Department of Cardiology, Division of Medicine, University Medical Centre of Ljubljana, Ljubljana, Slovenia. .,Division of Medicine, Centre for Preventive Cardiology, Division of Medicine, University Medical Centre Ljubljana, Zaloška cesta 7, SI-1525, Ljubljana, Slovenia. .,Chair of Internal Medicine, Medical Faculty, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
86
|
Basu A, Dvorina N, Baldwin WM, Mazumder B. High-fat diet-induced GAIT element-mediated translational silencing of mRNAs encoding inflammatory proteins in macrophage protects against atherosclerosis. FASEB J 2020; 34:6888-6906. [PMID: 32232901 DOI: 10.1096/fj.201903119r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/28/2020] [Accepted: 03/16/2020] [Indexed: 11/11/2022]
Abstract
Previously, we identified a mechanism of inflammation control directed by ribosomal protein L13a and "GAIT" (Gamma Activated Inhibitor of Translation) elements in target mRNAs and showed that its elimination in myeloid cell-specific L13a knockout mice (L13a KO) increased atherosclerosis susceptibility and severity. Here, we investigated the mechanistic basis of this endogenous defense against atherosclerosis. We compared molecular and cellular aspects of atherosclerosis in high-fat diet (HFD)-fed L13a KO and intact (control) mice. HFD treatment of control mice induced release of L13a from 60S ribosome, formation of RNA-binding complex, and subsequent GAIT element-mediated translational silencing. Atherosclerotic plaques from HFD-treated KO mice showed increased infiltration of M1 type inflammatory macrophages. Macrophages from KO mice showed increased phagocytic activity and elevated expression of LDL receptor and pro-inflammatory mediators. NanoString analysis of the plaques from KO mice showed upregulation of a number of mRNAs encoding inflammatory proteins. Bioinformatics analysis suggests the presence of the potential GAIT elements in the 3'UTRs of several of these mRNAs. Macrophage induces L13a/GAIT-dependent translational silencing of inflammatory genes in response to HFD as an endogenous defense against atherosclerosis in ApoE-/- model.
Collapse
Affiliation(s)
- Abhijit Basu
- Department of Biology, Geology and Environmental Sciences, Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH, USA
| | - Nina Dvorina
- Department of Inflammation and Immunity, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - William M Baldwin
- Department of Inflammation and Immunity, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Barsanjit Mazumder
- Department of Biology, Geology and Environmental Sciences, Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH, USA
| |
Collapse
|
87
|
In Search for Genes Related to Atherosclerosis and Dyslipidemia Using Animal Models. Int J Mol Sci 2020; 21:ijms21062097. [PMID: 32197550 PMCID: PMC7139774 DOI: 10.3390/ijms21062097] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is a multifactorial chronic disease that affects large arteries and may lead to fatal consequences. According to current understanding, inflammation and lipid accumulation are the two key mechanisms of atherosclerosis development. Animal models based on genetically modified mice have been developed to investigate these aspects. One such model is low-density lipoprotein (LDL) receptor knockout (KO) mice (ldlr-/-), which are characterized by a moderate increase of plasma LDL cholesterol levels. Another widely used genetically modified mouse strain is apolipoprotein-E KO mice (apoE-/-) that lacks the primary lipoprotein required for the uptake of lipoproteins through the hepatic receptors, leading to even greater plasma cholesterol increase than in ldlr-/- mice. These and other animal models allowed for conducting genetic studies, such as genome-wide association studies, microarrays, and genotyping methods, which helped identifying more than 100 mutations that contribute to atherosclerosis development. However, translation of the results obtained in animal models for human situations was slow and challenging. At the same time, genetic studies conducted in humans were limited by low sample sizes and high heterogeneity in predictive subclinical phenotypes. In this review, we summarize the current knowledge on the use of KO mice for identification of genes implicated in atherosclerosis and provide a list of genes involved in atherosclerosis-associated inflammatory pathways and their brief characteristics. Moreover, we discuss the approaches for candidate gene search in animals and humans and discuss the progress made in the field of epigenetic studies that appear to be promising for identification of novel biomarkers and therapeutic targets.
Collapse
|
88
|
Abstract
The vasculature not only transports oxygenated blood, metabolites, and waste products but also serves as a conduit for hormonal communication between distant tissues. Therefore, it is important to maintain homeostasis within the vasculature. Recent studies have greatly expanded our understanding of the regulation of vasculature development and vascular-related diseases at the epigenetic level, including by protein posttranslational modifications, DNA methylation, and noncoding RNAs. Integrating epigenetic mechanisms into the pathophysiologic conceptualization of complex and multifactorial vascular-related diseases may provide promising therapeutic approaches. Several reviews have presented detailed discussions of epigenetic mechanisms not including histone methylation in vascular biology. In this review, we primarily discuss histone methylation in vascular development and maturity, and in vascular diseases.
Collapse
|
89
|
LNCRNA OIP5-AS1 regulates oxidative low-density lipoprotein-mediated endothelial cell injury via miR-320a/LOX1 axis. Mol Cell Biochem 2020; 467:15-25. [DOI: 10.1007/s11010-020-03688-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 01/18/2020] [Indexed: 01/26/2023]
|
90
|
Huminiecki L, Atanasov AG, Horbańczuk J. Etiology of atherosclerosis informs choice of animal models and tissues for initial functional genomic studies of resveratrol. Pharmacol Res 2020; 156:104598. [PMID: 32067842 DOI: 10.1016/j.phrs.2019.104598] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 12/09/2019] [Accepted: 12/11/2019] [Indexed: 01/03/2023]
Abstract
Resveratrol, a phytoalexin, is a natural polyphenol synthesized exclusively by plants in response to environmental stresses. However, the molecule has also many exogenous bioactivities in animal cells. These bioactivities may lead to anti-cancer and cardio-protective health benefits. Because cellular responses to the treatment with resveratrol include the changes of expression patterns, functional genomics is an attractive tool to study them. In recent and today's experimental practice, this mostly means microarray profiling of gene expression (using RNAs isolated from bulk tissues). Herein, we review such published studies undertaken in the context of cardiovascular diseases (CVDs). CVDs are a number one public health problem in developed countries, outweighing in magnitude even cancer. In particular, we review the studies of resveratrol in several animal models relevant to CVDs. These models included: normal and pre-mature aging in mice, as well as atherogenic diet in mice / pigs / non-human primates. Additionally, there were few clinical studies published in the context of the comorbidities of atherosclerosis in humans (e.g. obesity, diabetes, hypertension). For the purposes of these studies, three types of samples were most commonly profiled with microarrays: the liver, the skeletal muscle, and peripheral blood mononuclear cells. Resveratrol-induced changes of gene expression typically mimicked those associated with calorie restriction and lifespan extension. They also opposed changes induced by the atherogenic diet. We conclude by discussing few experimental factors that were relatively neglected thus far, but which could be interesting to investigate in the future. These factors include sex and the exact formulation of resveratrol (plant extract, or synthetic chemical).
Collapse
Affiliation(s)
- Lukasz Huminiecki
- The Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Postępu 36A, Jastrzębiec, 05-552 Magdalenka, Poland.
| | - Atanas G Atanasov
- The Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Postępu 36A, Jastrzębiec, 05-552 Magdalenka, Poland; Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Jarosław Horbańczuk
- The Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Postępu 36A, Jastrzębiec, 05-552 Magdalenka, Poland
| |
Collapse
|
91
|
Navarro E, Mallén A, Cruzado JM, Torras J, Hueso M. Unveiling ncRNA regulatory axes in atherosclerosis progression. Clin Transl Med 2020; 9:5. [PMID: 32009226 PMCID: PMC6995802 DOI: 10.1186/s40169-020-0256-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 01/05/2020] [Indexed: 02/06/2023] Open
Abstract
Completion of the human genome sequencing project highlighted the richness of the cellular RNA world, and opened the door to the discovery of a plethora of short and long non-coding RNAs (the dark transcriptome) with regulatory or structural potential, which shifted the balance of pathological gene alterations from coding to non-coding RNAs. Thus, disease risk assessment currently has to also evaluate the expression of new RNAs such as small micro RNAs (miRNAs), long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), competing endogenous RNAs (ceRNAs), retrogressed elements, 3'UTRs of mRNAs, etc. We are interested in the pathogenic mechanisms of atherosclerosis (ATH) progression in patients suffering Chronic Kidney Disease, and in this review, we will focus in the role of the dark transcriptome (non-coding RNAs) in ATH progression. We will focus in miRNAs and in the formation of regulatory axes or networks with their mRNA targets and with the lncRNAs that function as miRNA sponges or competitive inhibitors of miRNA activity. In this sense, we will pay special attention to retrogressed genomic elements, such as processed pseudogenes and Alu repeated elements, that have been recently seen to also function as miRNA sponges, as well as to the use or miRNA derivatives in gene silencing, anti-ATH therapies. Along the review, we will discuss technical developments associated to research in lncRNAs, from sequencing technologies to databases, repositories and algorithms to predict miRNA targets, as well as new approaches to miRNA function, such as integrative or enrichment analysis and their potential to unveil RNA regulatory networks.
Collapse
Affiliation(s)
- Estanislao Navarro
- Independent Researcher, Barcelona, Spain. .,Department of Nephrology, Hospital Universitari Bellvitge and Bellvitge Research Institute (IDIBELL), C/Feixa Llarga, s/n; L'Hospitalet de Llobregat, 08907, Barcelona, Spain.
| | - Adrian Mallén
- Department of Nephrology, Hospital Universitari Bellvitge and Bellvitge Research Institute (IDIBELL), C/Feixa Llarga, s/n; L'Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Josep M Cruzado
- Department of Nephrology, Hospital Universitari Bellvitge and Bellvitge Research Institute (IDIBELL), C/Feixa Llarga, s/n; L'Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Joan Torras
- Department of Nephrology, Hospital Universitari Bellvitge and Bellvitge Research Institute (IDIBELL), C/Feixa Llarga, s/n; L'Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Miguel Hueso
- Department of Nephrology, Hospital Universitari Bellvitge and Bellvitge Research Institute (IDIBELL), C/Feixa Llarga, s/n; L'Hospitalet de Llobregat, 08907, Barcelona, Spain.
| |
Collapse
|
92
|
Fan Z, Zhang Y, Xiao D, Ma J, Liu H, Shen L, Zhang M, He B. Long noncoding RNA UC.98 stabilizes atherosclerotic plaques by promoting the proliferation and adhesive capacity in murine aortic endothelial cells. Acta Biochim Biophys Sin (Shanghai) 2020; 52:141-149. [PMID: 31942916 DOI: 10.1093/abbs/gmz155] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/01/2019] [Accepted: 11/06/2019] [Indexed: 12/25/2022] Open
Abstract
Pathological studies have shown that the vulnerability of plaques affects outcomes in patients with atherosclerosis (AS), a chronic inflammatory disease and common cause of morbidity and mortality worldwide. Although emerging technologies have enabled early diagnosis of AS with high-risk vulnerable plaques, more accurate and noninvasive diagnostic methods are urgently required. To this end, molecules involved in genetic or epigenetic regulation of the vulnerability of atherosclerotic plaques have been extensively studied. Here, we evaluated long noncoding RNA (lncRNA) variability by microarray assay in murine aortic endothelial cells (MAECs) bearing vulnerable plaques and identified the novel functional lncRNA UC.98, whose expression pattern was associated with the vulnerability of atherosclerotic plaques. Consistent with this, clinical statistics comparing the peripheral blood specimens from sets of patients with AS with or without vulnerable plaques confirmed the linear relationship between the expression pattern of UC.98 and plaque instability. Moreover, MTT assays and western blot analysis showed that silencing of intrinsic UC.98 in MAECs not only suppressed cell proliferation but also decreased the expressions of vascular cell adhesion molecule-1 and intercellular adhesion molecule-1, thereby inactivating the nuclear factor-κB pathway. In conclusion, our results highlighted the pivotal role of UC.98 in regulating the vulnerability of plaques during AS progression and suggested that UC.98 may be a biomarker of the early diagnosis and prognosis of AS with vulnerable plaques and a potential therapeutic target for slowing AS progression.
Collapse
Affiliation(s)
- Zixu Fan
- Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Ying Zhang
- Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Danrui Xiao
- Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Jianwei Ma
- Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Hua Liu
- Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Linghong Shen
- Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Min Zhang
- Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Ben He
- Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| |
Collapse
|
93
|
SULT2B1b inhibits reverse cholesterol transport and promotes cholesterol accumulation and inflammation in lymphocytes from AMI patients with low LDL-C levels. Clin Sci (Lond) 2020; 134:273-287. [PMID: 31957803 DOI: 10.1042/cs20190459] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 01/19/2023]
Abstract
The current main treatment for coronary artery disease (CAD) is to reduce low-density lipoprotein cholesterol (LDL-C) by statins, which could decrease the incidence of major adverse cardiovascular events (MACEs) by 30%. However, many residual risks still remain. To clarify the mechanism involved, we studied patients with acute myocardial infarction (AMI) with low LDL-C levels. Lymphocytes were isolated, and it was found that despite no difference in plasma LDL-C level, the lymphocyte cholesterol content was higher in AMI patient than those in non-CAD patients; thus, the decrease in intracellular cholesterol content was inconsistent with that in the plasma. Additionally, [3H]-cholesterol efflux rates were lower and mRNA levels of the inflammatory factors tumour necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) higher in AMI lymphocytes. It was found that sulphotransferase 2B1b (SULT2B1b) expression was higher in AMI lymphocytes. Further research using Jurkat T lymphocytes confirmed that SULT2B1b knockdown increased cholesterol efflux capacity and decreased mRNA levels of TNF-α and IFN-γ by increasing liver X receptor (LXR)-β levels. Furthermore, the degree of CpG island methylation in the SULT2B1b promoter was reduced in cells from AMI patients. In conclusion, SULT2B1b up-regulation due to hypomethylation of its promoter promotes cholesterol accumulation and inflammation by inhibiting LXR-β in lymphocytes of AMI patients with low LDL-C levels. Therefore, reducing intracellular cholesterol is also important as plasma cholesterol levels. Therapeutic approaches to decrease SULT2B1b expression might be potentially beneficial for CAD prevention by decreasing intracellular cholesterol.
Collapse
|
94
|
Abstract
Epigenetic regulatory mechanisms, encompassing diverse molecular processes including DNA methylation, histone post-translational modifications, and noncoding RNAs, are essential to numerous processes such as cell differentiation, growth and development, environmental adaptation, aging, and disease states. In many cases, epigenetic changes occur in response to environmental cues and lifestyle factors, resulting in persistent changes in gene expression that affect vascular disease risk during the lifetime of the individual. Biological aging-a powerful cardiovascular risk factor-is partly genetically determined yet strongly influenced by traditional risk factors, reflecting epigenetic modulation. Quantification of specific DNA methylation patterns may serve as an accurate predictor of biological age-a concept known as the epigenetic clock, which could help to refine cardiovascular risk assessment. Epigenetic reprogramming of monocytes rewires cellular immune signaling and induces a metabolic shift toward aerobic glycolysis, thereby increasing innate immune responses. This form of trained epigenetic memory can be maladaptive, thus augmenting vascular inflammation. Somatic mutations in epigenetic regulatory enzymes lead to clonal hematopoiesis of indeterminate potential, a precursor of hematologic malignancies and a recently recognized cardiovascular risk factor; moreover, epigenetic regulators are increasingly being targeted in cancer therapeutics. Thus, understanding epigenetic regulatory mechanisms lies at the intersection between cancer and cardiovascular disease and is of paramount importance to the burgeoning field of cardio-oncology (Graphic Abstract).
Collapse
Affiliation(s)
- Abdalrahman Zarzour
- From the Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University
| | - Ha Won Kim
- From the Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University
| | - Neal L Weintraub
- From the Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University
| |
Collapse
|
95
|
Shan H, Guo D, Zhang S, Qi H, Liu S, Du Y, He Y, Wang B, Xu M, Yu X. SNHG6 modulates oxidized low-density lipoprotein-induced endothelial cells injury through miR-135a-5p/ROCK in atherosclerosis. Cell Biosci 2020; 10:4. [PMID: 31921409 PMCID: PMC6947907 DOI: 10.1186/s13578-019-0371-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 12/26/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Plenty of long non-coding RNAs (lncRNAs) play vital roles in the progression of atherosclerosis. Small nucleolar RNA host gene 6 (SNHG6) is a well known lncRNA that is aberrantly high expressed in atherosclerosis patients. However, its function and basic mechanism in atherosclerosis events have not been well clarified. METHODS The expression patterns of SNHG6, miR-135a-5p, ROCK1 and ROCK2 in clinical samples and cells were detected by RT-qPCR assays. Cell Counting Kit-8 (CCK-8), flow cytometry assays, ELISA and reactive oxygen species (ROS) and malondialdehyde (MDA) detection, were performed to assess cell viability, apoptosis, inflammation and oxidative stress, respectively. Western blot analysis was carried out to examine the protein levels of Bax, Bcl-2, and SNHG6. Luciferase reporter and RIP assays were used to confirm the true interaction between SNHG6 and miR-135a-5p, or miR-135a-5p and ROCK. RESULTS The levels of SNHG6, ROCK1 and ROCK2 were notably increased and miR-135a-5p was decreased in atherosclerosis patients and oxidized low-density lipoprotein (ox-LDL)-treated HUVECs. Knockdown of SNHG6 alleviated ox-LDL-induced injury of HUVECs, while this effect was partly reversed by miR-135a-5p inhibitor. Moreover, overexpression of ROCKs aggravated miR-135a-5p-alleviated atherosclerosis cell injury. SNHG6 contributed to ROCK expression through sequestering miR-135a-5p as a molecular sponge. CONCLUSION SNHG6 functions as a promoter in atherosclerosis events by targeting miR-135a-5p/ROCK axis in ox-LDL-stimulated HUVECs. This finding will help to develop a novel therapeutic strategy for atherosclerosis.
Collapse
Affiliation(s)
- Haiyan Shan
- Department of General Practice, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, China
| | - Dawei Guo
- Department of the Fourth General Surgery, The Fourth Affiliated Hospital of China Medical University, No. 4 Chongshan East Road, Huanggu District, 110032, Shenyang, China
| | - Siyang Zhang
- Department of the Science and Experiment Center, The China Medical University, No. 77 Puhe Road, Shenbei New Area, 110122, Shenyang, China
| | - Huimeng Qi
- Department of General Practice, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, China
| | - Shen Liu
- Department of General Practice, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, China
| | - Yanmei Du
- Department of General Practice, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, China
| | - Yini He
- Department of General Practice, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, China
| | - Bofu Wang
- Department of General Practice, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, China
| | - Ming Xu
- Department of General Practice, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, China
| | - Xiaosong Yu
- Department of General Practice, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, China.
| |
Collapse
|
96
|
Association of N 6-methyladenine DNA with plaque progression in atherosclerosis via myocardial infarction-associated transcripts. Cell Death Dis 2019; 10:909. [PMID: 31797919 PMCID: PMC6892866 DOI: 10.1038/s41419-019-2152-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/01/2019] [Accepted: 11/05/2019] [Indexed: 01/14/2023]
Abstract
Modification of the novel N6-methyladenine (m6A) DNA implicates this epigenetic mark in human malignant disease, but its role in atherosclerosis (AS) is largely unknown. Here, we found that the leukocyte level of m6A but not 5mC DNA modification was decreased with increasing of carotid plaque size and thickness in 207 AS patients as compared with 142 sex- and age-matched controls. Serum low-density lipoprotein (LDL) and leukocyte m6A levels were associated with the progression of carotid plaque size and thickness. Both LDL level and plaque thickness were also independently and negatively related to m6A level. Reduced m6A level was further confirmed in leukocytes and endothelium in western diet-induced AS mice and in oxidized-LDL (ox-LDL)-treated human endothelium and monocyte cells. Decreased m6A level was closely related to the upregulation of AlkB homolog 1 (ALKBH1), the demethylase of m6A. Silencing of ALKBH1 or hypoxia-inducible factor 1α (HIF1α) could rescue the ox-LDL–increased level of MIAT, a hypoxia-response gene. Mechanically, ox-LDL induced HIF1α for transfer into the nucleus. Nuclear HIF1α bound to the ALKBH1-demethylated MIAT promoter and transcriptionally upregulated its expression. Therefore, elevated ALKBH1 level in endothelium and leukocytes reduced m6A level, which is a novel and sensitive biomarker for AS progression.
Collapse
|
97
|
Lu Q, Meng Q, Qi M, Li F, Liu B. Shear-Sensitive lncRNA AF131217.1 Inhibits Inflammation in HUVECs via Regulation of KLF4. Hypertension 2019; 73:e25-e34. [PMID: 30905197 DOI: 10.1161/hypertensionaha.118.12476] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Atherosclerosis is one of the most common vascular diseases, and inflammation participates in all stages of its progression. Laminar shear stress protects arteries from atherosclerosis and reduces endothelial inflammation. Long noncoding RNAs have emerged as critical regulators in many diseases, including atherosclerosis. However, the expression and functions of long noncoding RNAs subjected to laminar shear stress in endothelial cells remain unclear. This study aimed to reveal the mechanism by which shear stress-regulated long noncoding RNAs contribute to anti-inflammation. In this study, we identified a novel long noncoding RNA AF131217.1, which was upregulated after laminar shear stress treatment in human umbilical vein endothelial cells. Knockdown of AF131217.1 inhibited flow-mediated reduction of monocyte adhesion VCAM-1 (vascular cell adhesion molecule-1) and ICAM-1 (intercellular adhesion molecule-1) expression and inhibited flow-mediated enhancement of flow-responsive expression of KLF (Kruppel-like factor) 2 and eNOS (endothelial NO synthase). Furthermore, TNF-α (tumor necrosis factor-α) was used to induce an inflammatory response in human umbilical vein endothelial cells. Knockdown of AF131217.1 promoted ICAM-1 and VCAM-1 expression, as well as changes in monocyte adhesion and KLF2 and eNOS expression induced by TNF-α. Mechanistic investigations indicated that AF131217.1 acted as a competing endogenous RNA for miR-128-3p, leading to regulation of its target gene KLF4. In conclusion, our study demonstrates for the first time that laminar shear stress regulates the expression of AF131217.1 in human umbilical vein endothelial cells, and the AF131217.1/miR-128-3p/KLF4 axis plays a vital role in atherosclerosis development.
Collapse
Affiliation(s)
- Qing Lu
- From the Department of Pathogenobiology, Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, Jilin, China (Q.L., Q.M., M.Q., F.L.)
| | - Qingyu Meng
- From the Department of Pathogenobiology, Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, Jilin, China (Q.L., Q.M., M.Q., F.L.)
| | - Mingran Qi
- From the Department of Pathogenobiology, Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, Jilin, China (Q.L., Q.M., M.Q., F.L.)
| | - Fan Li
- From the Department of Pathogenobiology, Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, Jilin, China (Q.L., Q.M., M.Q., F.L.)
| | - Bin Liu
- Cardiovascular Disease Center, The First Hospital of Jilin University, Changchun, Jilin, China (B.L.)
| |
Collapse
|
98
|
Abuzhalihan J, Wang YT, Ma YT, Fu ZY, Yang YN, Ma X, Li XM, Liu F, Chen BD. SOAT1 methylation is associated with coronary heart disease. Lipids Health Dis 2019; 18:192. [PMID: 31684966 PMCID: PMC6829990 DOI: 10.1186/s12944-019-1138-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/21/2019] [Indexed: 12/13/2022] Open
Abstract
Background This study was designed to investigate whether differential DNA methylationin of cholesterol absorption candidate genes can function as a biomarker for patients with coronary heart disease (CHD). Methods DNA methylation levels of the candidate genes FLOT1, FLOT2 and SOAT1 were measured in peripheral blood leukocytes (PBLs) from 99 patients diagnosed with CHD and 89 control subjects without CHD. A total of 110 CPG sites around promoter regions of them were examined. Results Compared with groups without CHD, patients with CHD had lower methylation levels of SOAT1 (P<0.001). When each candidate genes were divided into different target segments, patients with CHD also had lower methylation levels of SOAT1 than patients without (P = 0.005). After adjustment of other confounders, methylation levels of SOAT1 were still associated with CHD (P = 0.001, OR = 0.290, 95% CI: 0.150–0.561). Conclusions SOAT1 methylation may be associated with development of CHD. Patients with lower methylation levels in SOAT1 may have increased risks for CHD. Further studies on the specific mechanisms of this relationship are necessary.
Collapse
Affiliation(s)
- Jialin Abuzhalihan
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People's Republic of China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China
| | - Yong-Tao Wang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People's Republic of China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China
| | - Yi-Tong Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People's Republic of China. .,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China.
| | - Zhen-Yan Fu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People's Republic of China. .,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China.
| | - Yi-Ning Yang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People's Republic of China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China
| | - Xiang Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People's Republic of China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China
| | - Xiao-Mei Li
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People's Republic of China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China
| | - Fen Liu
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China
| | - Bang-Dang Chen
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China
| |
Collapse
|
99
|
Jukema RA, Ahmed TAN, Tardif JC. Does low-density lipoprotein cholesterol induce inflammation? If so, does it matter? Current insights and future perspectives for novel therapies. BMC Med 2019; 17:197. [PMID: 31672136 PMCID: PMC6824020 DOI: 10.1186/s12916-019-1433-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/26/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Dyslipidemia and inflammation are closely interrelated contributors in the pathogenesis of atherosclerosis. Disorders of lipid metabolism initiate an inflammatory and immune-mediated response in atherosclerosis, while low-density lipoprotein cholesterol (LDL-C) lowering has possible pleiotropic anti-inflammatory effects that extend beyond lipid lowering. MAIN TEXT Activation of the immune system/inflammasome destabilizes the plaque, which makes it vulnerable to rupture, resulting in major adverse cardiac events (MACE). The activated immune system potentially accelerates atherosclerosis, and atherosclerosis activates the immune system, creating a vicious circle. LDL-C enhances inflammation, which can be measured through multiple parameters like high-sensitivity C-reactive protein (hsCRP). However, multiple studies have shown that CRP is a marker of residual risk and not, itself, a causal factor. Recently, anti-inflammatory therapy has been shown to decelerate atherosclerosis, resulting in fewer MACE. Nevertheless, an important side effect of anti-inflammatory therapy is the potential for increased infection risk, stressing the importance of only targeting patients with high residual inflammatory risk. Multiple (auto-)inflammatory diseases are potentially related to/influenced by LDL-C through inflammasome activation. CONCLUSIONS Research suggests that LDL-C induces inflammation; inflammation is of proven importance in atherosclerotic disease progression; anti-inflammatory therapies yield promise in lowering (cardiovascular) disease risk, especially in selected patients with high (remaining) inflammatory risk; and intriguing new anti-inflammatory developments, for example, in nucleotide-binding leucine-rich repeat-containing pyrine receptor inflammasome targeting, are currently underway, including novel pathway interventions such as immune cell targeting and epigenetic interference. Long-term safety should be carefully monitored for these new strategies and cost-effectiveness carefully evaluated.
Collapse
Affiliation(s)
- Ruurt A Jukema
- Department of Medicine, VU University Medical Centre Amsterdam, Amsterdam, the Netherlands. .,Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Canada.
| | | | - Jean-Claude Tardif
- Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Canada
| |
Collapse
|
100
|
Manea SA, Vlad ML, Fenyo IM, Lazar AG, Raicu M, Muresian H, Simionescu M, Manea A. Pharmacological inhibition of histone deacetylase reduces NADPH oxidase expression, oxidative stress and the progression of atherosclerotic lesions in hypercholesterolemic apolipoprotein E-deficient mice; potential implications for human atherosclerosis. Redox Biol 2019; 28:101338. [PMID: 31634818 PMCID: PMC6807290 DOI: 10.1016/j.redox.2019.101338] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/27/2019] [Accepted: 10/03/2019] [Indexed: 01/01/2023] Open
Abstract
NADPH oxidase (Nox)-derived reactive oxygen species (ROS) are instrumental in all inflammatory phases of atherosclerosis. Dysregulated histone deacetylase (HDAC)-related epigenetic pathways have been mechanistically linked to alterations in gene expression in experimental models of cardiovascular disorders. Hitherto, the relation between HDAC and Nox in atherosclerosis is not known. We aimed at uncovering whether HDAC plays a role in mediating Nox up-regulation, oxidative stress, inflammation, and atherosclerotic lesion progression. Human non-atherosclerotic and atherosclerotic arterial samples, ApoE-/- mice, and in vitro polarized monocyte-derived M1/M2-macrophages (Mac) were examined. Male ApoE-/- mice, maintained on normal or high-fat, cholesterol-rich diet, were randomized to receive 10 mg/kg suberoylanilide hydroxamic acid (SAHA), a pan-HDAC inhibitor, or its vehicle, for 4 weeks. In the human/animal studies, real-time PCR, Western blot, lipid staining, lucigenin-enhanced chemiluminescence assay, and enzyme-linked immunosorbent assay were employed. The protein levels of class I, class IIa, class IIb, and class IV HDAC isoenzymes were significantly elevated both in human atherosclerotic tissue samples and in atherosclerotic aorta of ApoE-/- mice. Treatment of ApoE-/- mice with SAHA reduced significantly the extent of atherosclerotic lesions, and the aortic expression of Nox subtypes, NADPH-stimulated ROS production, oxidative stress and pro-inflammatory markers. Significantly up-regulated HDAC and Nox subtypes were detected in inflammatory M1-Mac. In these cells, SAHA reduced the Nox1/2/4 transcript levels. Collectively, HDAC inhibition reduced atherosclerotic lesion progression in ApoE-/- mice, possibly by intertwined mechanisms involving negative regulation of Nox expression and inflammation. The data propose that HDAC-oriented pharmacological interventions could represent an effective therapeutic strategy in atherosclerosis.
Collapse
Affiliation(s)
- Simona-Adriana Manea
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Mihaela-Loredana Vlad
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Ioana Madalina Fenyo
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Alexandra-Gela Lazar
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Monica Raicu
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Horia Muresian
- University Hospital Bucharest, Cardiovascular Surgery Department, Bucharest, Romania
| | - Maya Simionescu
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Adrian Manea
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania.
| |
Collapse
|