1
|
Nasiri-Ansari N, Spilioti E, Kyrou I, Kalotychou V, Chatzigeorgiou A, Sanoudou D, Dahlman-Wright K, Randeva HS, Papavassiliou AG, Moutsatsou P, Kassi E. Estrogen Receptor Subtypes Elicit a Distinct Gene Expression Profile of Endothelial-Derived Factors Implicated in Atherosclerotic Plaque Vulnerability. Int J Mol Sci 2022; 23:ijms231810960. [PMID: 36142876 PMCID: PMC9506323 DOI: 10.3390/ijms231810960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 11/21/2022] Open
Abstract
In the presence of established atherosclerosis, estrogens are potentially harmful. MMP-2 and MMP-9, their inhibitors (TIMP-2 and TIMP-1), RANK, RANKL, OPG, MCP-1, lysyl oxidase (LOX), PDGF-β, and ADAMTS-4 play critical roles in plaque instability/rupture. We aimed to investigate (i) the effect of estradiol on the expression of the abovementioned molecules in endothelial cells, (ii) which type(s) of estrogen receptors mediate these effects, and (iii) the role of p21 in the estrogen-mediated regulation of the aforementioned factors. Human aortic endothelial cells (HAECs) were cultured with estradiol in the presence or absence of TNF-α. The expression of the aforementioned molecules was assessed by qRT-PCR and ELISA. Zymography was also performed. The experiments were repeated in either ERα- or ERβ-transfected HAECs and after silencing p21. HAECs expressed only the GPR-30 estrogen receptor. Estradiol, at low concentrations, decreased MMP-2 activity by 15-fold, increased LOX expression by 2-fold via GPR-30, and reduced MCP-1 expression by 3.5-fold via ERβ. The overexpression of ERα increased MCP-1 mRNA expression by 2.5-fold. In a low-grade inflammation state, lower concentrations of estradiol induced the mRNA expression of MCP-1 (3.4-fold) and MMP-9 (7.5-fold) and increased the activity of MMP-2 (1.7-fold) via GPR-30. Moreover, p21 silencing resulted in equivocal effects on the expression of the abovementioned molecules. Estradiol induced different effects regarding atherogenic plaque instability through different ERs. The balance of the expression of the various ER subtypes may play an important role in the paradoxical characterization of estrogens as both beneficial and harmful.
Collapse
Affiliation(s)
- Narjes Nasiri-Ansari
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Eliana Spilioti
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Laboratory of Toxicological Control of Pesticides, Scientific Directorate of Pesticides’ Control and Phytopharmacy, Benaki Phytopathological Institute, 14561 Athens, Greece
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Laboratory of Dietetics and Quality of Life, Department of Food Science and Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855 Athens, Greece
- Centre for Sport, Exercise and Life Sciences, Research Institute for Health & Wellbeing, Coventry University, Coventry CV1 5FB, UK
| | - Vassiliki Kalotychou
- Department of Internal Medicine, Laikon General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Despina Sanoudou
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, Attikon Hospital Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Karin Dahlman-Wright
- Department of Biosciences and Nutrition, Novum, Karolinska Institute, SE-14183 Huddinge, Sweden
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Paraskevi Moutsatsou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Endocrine Unit, 1st Department of Propaedeutic Internal Medicine, Laiko General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Correspondence: ; Tel.: +30-21-0746-2699; Fax: +30-21-0746-2703
| |
Collapse
|
2
|
Medina-Gil JM, Pérez-García A, Saavedra-Santana P, Díaz-Carrasco A, Martínez-Quintana E, Rodríguez-González F, Ramírez CM, Riaño M, Garay-Sánchez P, Tugores A. A Common Variant at the 3'untranslated Region of the CCL7 Gene (rs17735770) Is Associated With Decreased Susceptibility to Coronary Heart Disease. Front Cardiovasc Med 2022; 9:908070. [PMID: 35711383 PMCID: PMC9194478 DOI: 10.3389/fcvm.2022.908070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/12/2022] [Indexed: 11/13/2022] Open
Abstract
Monocytes participate in the development of atherosclerosis through the action of cytokines and other inflammatory mediators. Among them, CCR2 and its ligands, CCL2 and CCL7 play an important role, so the main objective of this work was to determine whether genetic variants affecting their activity were associated with cardiovascular disease. A cohort of 519 patients that have suffered coronary events was analyzed under a propensity score-matching protocol selecting a homogeneous set of cases and controls, according to age, sex, smoking status, dyslipidemia, arterial hypertension and type 2 diabetes as risk factors. While dyslipidemia and arterial hypertension were more prevalent among patients with angina pectoris, current smoking status and elevated inflammatory markers, including total leukocyte and monocyte counts, were more likely associated with acute coronary events. Propensity score matching analysis, performed to eliminate the influence of these risk factors and highlight genetic modifiers, revealed that a single nucleotide variant, rs17735770 at the 3'untranslated region of the CCL7 gene transcript, was associated with decreased cardiovascular risk in a group represented mostly by men, with an average age of 57, and without significant differences in traditional risk factors. Furthermore, the presence of this variant altered the local mRNA structure encompassing a binding site for miR-23ab, resulting in increased translation of a reporter gene in a miR23 independent fashion. The rs17735770 genetic variant led to increased expression of CCL7, a potential antagonist of CCR2 at inflammatory sites, where it could play a meaningful role during the evolution of atherosclerosis.
Collapse
Affiliation(s)
- José María Medina-Gil
- Servicio de Cardiología, Complejo Hospitalario Universitario Insular Materno-Infantil, Las Palmas de Gran Canaria, Spain
| | - Ana Pérez-García
- IMDEA Research Institute of Food and Health Sciences, Madrid, Spain
| | - Pedro Saavedra-Santana
- Departamento de Matemáticas, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | | | - Efrén Martínez-Quintana
- Servicio de Cardiología, Complejo Hospitalario Universitario Insular Materno-Infantil, Las Palmas de Gran Canaria, Spain
| | - Fayna Rodríguez-González
- Servicio de Oftalmología, Hospital Universitario Gran Canaria Doctor Negrín, Las Palmas de Gran Canaria, Spain
| | | | - Marta Riaño
- Servicio de Bioquímica Clínica y Análisis Clínicos, Complejo Hospitalario Universitario Insular Materno-Infantil, Las Palmas de Gran Canaria, Spain
| | - Paloma Garay-Sánchez
- Unidad de Investigación, Complejo Hospitalario Universitario Insular Materno-Infantil, Las Palmas de Gran Canaria, Spain
| | - Antonio Tugores
- Unidad de Investigación, Complejo Hospitalario Universitario Insular Materno-Infantil, Las Palmas de Gran Canaria, Spain
- *Correspondence: Antonio Tugores
| |
Collapse
|
3
|
Fan ZX, Yuan SJ, Li XQ, Yang TT, Niu TT, Ma L, Sun K, Wang L, Liu GZ. Preliminary study on the differentiation of vulnerable carotid plaques via analysis of calcium content and spectral curve slope by using gemstone spectral imaging. Exp Ther Med 2022; 23:325. [PMID: 35386621 PMCID: PMC8972840 DOI: 10.3892/etm.2022.11254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 02/23/2022] [Indexed: 11/05/2022] Open
Abstract
Growing evidence indicates that vulnerable carotid plaque rupture is an important cause of stroke. However, the role of novel gemstone spectral imaging (GSI) in the assessment of vulnerable carotid plaques has remained to be sufficiently explored. Therefore, the aim of the present study was to provide a comprehensive evaluation of carotid atherosclerotic plaques using both GSI imaging biomarkers and serological biomarkers, and further explore their possible roles in the atherogenic process. The present study analyzed GSI data, including calcium content of carotid atherosclerotic plaques and spectral curve slope, as well as serum high-sensitivity C-reactive protein (Hs-CRP) and monocyte chemotactic protein-1 (MCP-1) levels in patients with a carotid atherosclerotic plaque using GSI-computed tomographic angiography and immunoturbidimetry. Patients with unstable plaque exhibited a significantly lower calcium content and higher spectral curve slope than those of the stable plaque group. In addition, patients with unstable plaque exhibited an increase in Hs-CRP and MCP-1 levels compared with those of the stable plaque and normal control groups. The alteration in GSI calcium content and spectral curve slope reflects a close link between calcification and plaque instability, while aberrant Hs-CRP and MCP-1 expression are involved in the formation or development of vulnerable plaques. Taken together, the present results strongly support the feasibility of using these serological and newly identified imaging parameters as multiple potential biomarkers relevant to plaque vulnerability or stroke progression.
Collapse
Affiliation(s)
- Ze-Xin Fan
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Shao-Jie Yuan
- Department of Neurology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xiao-Qing Li
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Ting-Ting Yang
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Tian-Tong Niu
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Lin Ma
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Kai Sun
- Department of Imaging, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Li Wang
- Department of Neurology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Guang-Zhi Liu
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| |
Collapse
|
4
|
Retro-orbital injection of FITC-dextran combined with isolectin B4 in assessing the retinal neovascularization defect. BMC Ophthalmol 2021; 21:208. [PMID: 33975571 PMCID: PMC8112026 DOI: 10.1186/s12886-021-01969-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 04/28/2021] [Indexed: 11/15/2022] Open
Abstract
Background A reliable and effective method is required to deliver agent that can aid the in vivo imaging of retinal vessels. The aim of the present study was to evaluate retro-orbital (RO) injection of fluorescein-labeled isothiocyanate dextran (FITC-dextran) as a method of demonstrating retinal neovascularization (NV) and avascular areas in oxygen-induced retinopathy (OIR) mice. Methods Different concentrations of FITC-dextran were used to compare the efficacy of this agent in perfusing the retinal vessels. Hematoxylin–eosin (HE) staining was used to evaluate the safety of RO injection. The vitreous blood vessels and extent of NV were assessed in P17 OIR mice using FITC-dextran and compared with the corresponding measurements obtained following isolectin B4 staining or the combination of both methods. Results The fluorescence of small vessels and neovascular tufts could be observed clearly following RO injection of 0.05 ml of 25 mg/ml or 50 mg/ml FITC-dextran. No visible damage to tissues adjacent to the injection site was discovered. Vitreous blood flow was gradually reduced from P0 to P5 and eventually disappeared in P17 OIR mice, as demonstrated by FITC-dextran perfusion. The retinal NV areas assessed by isolectin B4 were larger than those assessed by FITC-dextran, but the retinal avascular areas were smaller. The combination of both methods could conduce to distinguish non-functional blood vessels. Conclusions RO injection of FITC-dextran combined with isolectin B4 is an effective, optimal method for assessing the NV area and avascular area.
Collapse
|
5
|
Zhao Y, Chen W, Liu Y, Li H, Chi J, Chang Q, Shen L, Yan R, Li J, Yin X, Fu Y. Promoting plaque stability by gene silencing of monocyte chemotactic protein-3 or overexpression of tissue factor pathway inhibitor in ApoE-/- mice. J Drug Target 2021; 29:669-675. [PMID: 33472448 DOI: 10.1080/1061186x.2021.1878363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Chemokines may promote the formation and instability of atherosclerotic plaque, which is the most common cause of acute coronary syndrome. The aim of this study was to clarify the function of monocyte chemotactic protein-3 (MCP-3) in the stability of atherosclerotic plaque, to determine the role of tissue factor pathway inhibitor (TFPI) on the development and stability of atherosclerotic plaques, and to further elucidate the anti-atherosclerotic mechanism of TFPI with the emphasis on chemokine MCP-3. We constructed an adenovirus-mediated shRNA against mouse MCP-3 (Ad-MCP-3-shRNA) and an adenovirus-containing TFPI (Ad-TFPI), and tranferred them in a model of vulnerable plaque in ApoE-/- mice respectively. Here, we reported that MCP-3-shRNA and TFPI could both reduce the plaque area and decrease the content of lipids and macrophages, on the contrary, the fibrous cap thickness and content of collagen and smooth muscle cells were increased. In addition, the expression of MCP-3 and CC chemokine receptor 2 (CCR2) was decreased by TFPI transfer. These data provide the first in vivo evidence that MCP-3 is a major contributor to the unstability of atherosclerotic plaque and TFPI may exert its anti-atherosclerotic effects and promote stabilisation of plaque at least partly through inhibiting MCP-3/CCR2 pathway, which may be a new therapeutic method for atherosclerosis.
Collapse
Affiliation(s)
- Yong Zhao
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenjia Chen
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yue Liu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hui Li
- Department of Cardiology, The Fifth Hospital in Harbin, Harbin, China
| | - Jinyu Chi
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qing Chang
- Department of Cardiology, Heilongjiang Provincial Hospital, Harbin, China
| | - Li Shen
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Runan Yan
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiashu Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinhua Yin
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu Fu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
6
|
Ding Q, Shao C, Rose P, Zhu YZ. Epigenetics and Vascular Senescence-Potential New Therapeutic Targets? Front Pharmacol 2020; 11:535395. [PMID: 33101015 PMCID: PMC7556287 DOI: 10.3389/fphar.2020.535395] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 08/31/2020] [Indexed: 12/14/2022] Open
Abstract
Epigenetics is defined as the heritable alterations of gene expression without changes to the coding sequence of DNA. These alterations are mediated by processes including DNA methylation, histone modifications, and non-coding RNAs mechanisms. Vascular aging consists of both structural and functional changes in the vasculature including pathological processes that drive progression such as vascular cell senescence, inflammation, oxidation stress, and calcification. As humans age, these pathological conditions gradually accumulate, driven by epigenetic alterations, and are linked to various aging-related diseases. The development of drugs targeting a spectrum of epigenetic processes therefore offers novel treatment strategies for the targeting of age-related diseases. In our previous studies, we identified HDAC4, JMJD3, Fra-1, and GATA4 as potential pharmacological targets for regulating vascular inflammation, injury, and senescence.
Collapse
Affiliation(s)
- Qian Ding
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China.,School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Chunhong Shao
- Department of Psychiatry, Huashan Hospital, Fudan University, Shanghai, China
| | - Peter Rose
- School of Biosciences, University of Nottingham, Loughborough, United Kingdom
| | - Yi Zhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China
| |
Collapse
|
7
|
Li M, Chen Y, Zhang Y, Li D, Liu J. Correlation between monocyte chemoattractant protein-1/chemokine (C-C motif) ligand 2 and coronary plaque characteristics. Exp Biol Med (Maywood) 2020; 245:1335-1343. [PMID: 32640896 DOI: 10.1177/1535370220941424] [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] [Indexed: 11/16/2022] Open
Abstract
IMPACT STATEMENT Vulnerable plaques are plaques which are susceptible to rupture or thrombosis and trigger a series of adverse events such as coronary disorders. CCL2 is a soluble basic protein belonging to the CC subfamily. Previous studies have been investigated on the correlation between inflammatory factors and clinical events, but there are few studies on the correlation between CCL2 and plaque characteristics. Our study found that the high expression of CCL2 is involved in multiple processes in the genesis and progression of coronary artery disease, and would be a potential clinical prognostic indicator. In addition, high expression of CCL2 may be related to gene pathways such as Nod-like receptor signaling pathway, suggesting that CCL2 is involved in the inflammatory response and immune process of coronary artery disease.
Collapse
Affiliation(s)
- Meng Li
- Department of Cardiology, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Zhengzhou University, Zhengzhou 450003, China
| | - Yan Chen
- Department of Cardiology, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Zhengzhou University, Zhengzhou 450003, China
| | - Yan Zhang
- Department of Cardiology, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Zhengzhou University, Zhengzhou 450003, China
| | - Danna Li
- Department of Ultrasound, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jun Liu
- Department of Cardiology, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Zhengzhou University, Zhengzhou 450003, China
| |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW Atherosclerosis is characterized by accumulation of lipids and chronic inflammation in medium size to large arteries. Recently, RNA-based antisense oligonucleotides (ASOs) and small interfering RNAs (siRNAs) are being developed, along with small molecule-based drugs and monoclonal antibodies, for the treatment of risk factors associated with atherosclerosis.. The purpose of this review is to describe nucleic acid-based therapeutics and introduce novel RNAs that might become future tools for treatment of atherosclerosis. RECENT FINDINGS RNA-based inhibitors for PCSK9, Lp(a), ApoCIII, and ANGPTL3 have been successfully tested in phase II-III clinical trials. Moreover, multiple microRNA and long non-coding RNAs have been found to reduce atherogenesis in preclinical animal models. Clinical trials especially with ASOs and siRNAs directed to liver, targeting cholesterol and lipoprotein metabolism, have shown promising results. Additional research in larger patient cohorts is needed to fully evaluate the therapeutic potential of these new drugs.
Collapse
Affiliation(s)
- Petri Mäkinen
- A.I. Virtanen Institute, University of Eastern Finland, Neulaniementie 2, P.O. Box 1627, 70211, Kuopio, Finland
| | - Anna-Kaisa Ruotsalainen
- A.I. Virtanen Institute, University of Eastern Finland, Neulaniementie 2, P.O. Box 1627, 70211, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute, University of Eastern Finland, Neulaniementie 2, P.O. Box 1627, 70211, Kuopio, Finland.
- Heart Center and Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland.
| |
Collapse
|
9
|
Tsuchida S, Matsuzaki T, Yamato M, Okuda K, Fu HY, Araki R, Sanada S, Asanuma H, Asano Y, Asakura M, Hao H, Takashima S, Kitakaze M, Sakata Y, Mekada E, Minamino T. Anti-HB-EGF Antibody-Mediated Delivery of siRNA to Atherosclerotic Lesions in Mice. Int Heart J 2018; 59:1425-1431. [DOI: 10.1536/ihj.17-644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Shota Tsuchida
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Takashi Matsuzaki
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University
| | - Masaki Yamato
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Keiji Okuda
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Hai Ying Fu
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Ryo Araki
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Shoji Sanada
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Hiroshi Asanuma
- Department of Internal Medicine, Meiji University of Integrative Medicine
| | - Yoshihiro Asano
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Masanori Asakura
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine
| | - Hiroyuki Hao
- Department of Pathology, Nihon University School of Medicine
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine
| | - Masafumi Kitakaze
- Clinical Research and Development, National Cerebral and Cardiovascular Center Research Institute
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Eisuke Mekada
- Department of Cell Biology, Research Institute for Microbial Diseases, Osaka University
| | - Tetsuo Minamino
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University
| |
Collapse
|
10
|
Quesada I, Cejas J, García R, Cannizzo B, Redondo A, Castro C. Vascular dysfunction elicited by a cross talk between periaortic adipose tissue and the vascular wall is reversed by pioglitazone. Cardiovasc Ther 2018; 36:e12322. [PMID: 29464937 DOI: 10.1111/1755-5922.12322] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/30/2018] [Accepted: 02/15/2018] [Indexed: 01/17/2023] Open
Abstract
AIM Perivascular adipose tissue (PVAT) is in intimate contact with the vessel wall and extravascular PVAT-derived inflammatory mediators may adversely influence atherosclerotic plaque formation and stability through outside-to-inside signaling. We sought to investigate the role of PVAT on the atheroma development in an experimental animal model of metabolic syndrome (MS) associated with oxidative stress and low-grade inflammatory state. We also studied the effect of pioglitazone an insulin sensitizer, on the aortic wall and its surrounding PVAT, considering a bi-directional communication between both layers. METHODS Apolipoprotein E-deficient mice (ApoE-/- ) were fed with standard diet (CD, control diet) or fructose overload (10% w/v) (FD, fructose diet) for 8 weeks and treated with or without pioglitazone the latest 4 weeks. RESULTS Biochemical variables show that glycemia and lipid peroxidation determined by thiobarbituric acid reactive species (TBARS) significantly increased in FD-fed ApoE-/- mice. FD significantly increased aortic PVAT expression of oxidative stress associated genes: p22phox , Nox1, Nox2, Nox4 and p47phox , and proinflammatory genes: Visfatin, MCP-1, and MMP-9. Pioglitazone diminished PVAT-oxidative damage elicited by fructose treatment and markedly down-regulated proinflammatory markers. Even pioglitazone did not prevent the development of the aortic atheroma plaques stimulated by FD, significantly diminished VCAM-1 expression, MMP-9 expression and activity in aortic media wall and significantly reduced the accumulation of lipids and macrophages in atheroma plaques. CONCLUSION Our results support the fact that PVAT contributes to the development and progression of cardiovascular disease by underlying mechanisms elicited by "outside-in" signaling. Treatment with pioglitazone may offer a new effect on the whole vessel wall, promoting the stability of advanced atherosclerotic plaques.
Collapse
Affiliation(s)
- Isabel Quesada
- Vascular Biology Lab, Institute of Experimental Medicine and Biology of Cuyo (IMBECU) CONICET, School of Medical Sciences, National University of Cuyo, Mendoza, Argentina
| | - Jimena Cejas
- Vascular Biology Lab, Institute of Experimental Medicine and Biology of Cuyo (IMBECU) CONICET, School of Medical Sciences, National University of Cuyo, Mendoza, Argentina
| | - Rodrigo García
- Laboratory of Cardiovascular Physiopathology, Institute of Experimental Medicine and Biology of Cuyo (IMBECU) - CONICET, Mendoza, Argentina
| | - Beatriz Cannizzo
- Vascular Biology Lab, Institute of Experimental Medicine and Biology of Cuyo (IMBECU) CONICET, School of Medical Sciences, National University of Cuyo, Mendoza, Argentina
| | - Analía Redondo
- Vascular Biology Lab, Institute of Experimental Medicine and Biology of Cuyo (IMBECU) CONICET, School of Medical Sciences, National University of Cuyo, Mendoza, Argentina
| | - Claudia Castro
- Vascular Biology Lab, Institute of Experimental Medicine and Biology of Cuyo (IMBECU) CONICET, School of Medical Sciences, National University of Cuyo, Mendoza, Argentina
| |
Collapse
|
11
|
Epigenetic Regulation of Vascular Aging and Age-Related Vascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1086:55-75. [PMID: 30232752 DOI: 10.1007/978-981-13-1117-8_4] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Vascular aging refers to the structural and functional defects that occur in the aorta during the aging process and is characterized by increased vascular cell senescence, vascular dyshomeostasis, and vascular remodeling. Vascular aging is a major risk factor for vascular diseases. However, the current understanding of the biological process of vascular aging and age-related diseases is insufficient. Epigenetic regulation can influence gene expression independently of the gene sequence and mainly includes DNA methylation, histone modifications, and RNA-based gene regulation. Epigenetic regulation plays important roles in many physiological and pathophysiological processes and may explain some gaps in our knowledge regarding the interaction between genes and diseases. In this review, we summarize recent advances in the understanding of the epigenetic regulation of vascular aging and age-related diseases in terms of vascular cell senescence, vascular dyshomeostasis, and vascular remodeling. Moreover, the possibility of targeting epigenetic regulation to delay vascular aging and treat age-related vascular diseases is also discussed.
Collapse
|
12
|
Ma L, Ni M, Hao P, Lu H, Yang X, Xu X, Zhang C, Huang S, Zhao Y, Liu X, Zhang Y. Tongxinluo mitigates atherogenesis by regulating angiogenic factors and inhibiting vasa vasorum neovascularization in apolipoprotein E-deficient mice. Oncotarget 2017; 7:16194-204. [PMID: 26908443 PMCID: PMC4941307 DOI: 10.18632/oncotarget.7477] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 02/08/2016] [Indexed: 02/05/2023] Open
Abstract
Vasa vasorum (VV) neovascularization contributes to atherogenesis and its expansion and distribution is correlated with intraplaque expression of angiogenic factors. The present study investigated the roles of Tongxinluo (TXL), a traditional Chinese medication, on VV proliferation and atherogenesis. In vitro, TXL pre-treatment reversed the tumor necrosis factor-a (TNF-a) induced expression of vascular endothelial growth factor A (VEGF-A) and angiopoietin-1 (ANGPT-1) but not ANGPT-2, leading to increased ratio of ANGPT-1 to ANGPT-2. Consistently, TXL treatment (at a dosage of 0.38, 0.75, 1.5 g/kg/d, respectively) decreased the expression of VEGF-A while increased that of ANGPT-1 in early atherosclerotic lesions of apolipoprotein E deficient (apoE−/−) mice. On aortic ring assay, microvessels sprouting from aortas were significantly inhibited in TXL-treated mice. Moreover, VV neovascularization in plaques was markedly reduced with TXL treatment. Histological and morphological analysis demonstrated that TXL treatment reduced plaque burden, plaque size and changed the plaque composition. These data suggest that TXL inhibits early atherogenesis through regulating angiogenic factor expression and inhibiting VV proliferation in atherosclerotic plaque. Our study shed new light on the anti-atherosclerotic effect of TXL.
Collapse
Affiliation(s)
- Lianyue Ma
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China.,The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Mei Ni
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Panpan Hao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Huixia Lu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Xiaoyan Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Xingli Xu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Shanying Huang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Yuxia Zhao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Xiaoling Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China.,The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Shandong 250012, P.R. China.,The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Shandong 250012, P.R. China
| |
Collapse
|
13
|
Cui S, Li W, Lv X, Wang P, Gao Y, Huang G. Folic Acid Supplementation Delays Atherosclerotic Lesion Development by Modulating MCP1 and VEGF DNA Methylation Levels In Vivo and In Vitro. Int J Mol Sci 2017; 18:ijms18050990. [PMID: 28475147 PMCID: PMC5454903 DOI: 10.3390/ijms18050990] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 04/25/2017] [Accepted: 05/02/2017] [Indexed: 12/11/2022] Open
Abstract
The pathogenesis of atherosclerosis has been partly acknowledged to result from aberrant epigenetic mechanisms. Accordingly, low folate levels are considered to be a contributing factor to promoting vascular disease because of deregulation of DNA methylation. We hypothesized that increasing the levels of folic acid may act via an epigenetic gene silencing mechanism to ameliorate atherosclerosis. Here, we investigated the atheroprotective effects of folic acid and the resultant methylation status in high-fat diet-fed ApoE knockout mice and in oxidized low-density lipoprotein-treated human umbilical vein endothelial cells. We analyzed atherosclerotic lesion histology, folate concentration, homocysteine concentration, S-adenosylmethionine (SAM) and S-adenosylhomocysteine (SAH), and DNA methyltransferase activity, as well as monocyte chemotactic protein-1 (MCP1) and vascular endothelial growth factor (VEGF) expression and promoter methylation. Folic acid reduced atherosclerotic lesion size in ApoE knockout mice. The underlying folic acid protective mechanism appears to operate through regulating the normal homocysteine state, upregulating the SAM: SAH ratio, elevating DNA methyltransferase activity and expression, altering MCP1 and VEGF promoter methylation, and inhibiting MCP1 and VEGF expression. We conclude that folic acid supplementation effectively prevented atherosclerosis by modifying DNA methylation through the methionine cycle, improving DNA methyltransferase activity and expression, and thus changing the expression of atherosclerosis-related genes.
Collapse
Affiliation(s)
- Shanshan Cui
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China.
| | - Wen Li
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China.
| | - Xin Lv
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China.
| | - Pengyan Wang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China.
| | - Yuxia Gao
- Department of Cardiology, General Hospital of Tianjin Medical University, Tianjin 300052, China.
| | - Guowei Huang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China.
| |
Collapse
|
14
|
Cytokines: roles in atherosclerosis disease progression and potential therapeutic targets. Future Med Chem 2016; 8:1317-30. [PMID: 27357616 DOI: 10.4155/fmc-2016-0072] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis, the primary cause of cardiovascular disease (CVD), is a chronic inflammatory disorder in the walls of medium and large arteries. CVD is currently responsible for about one in three global deaths and this is expected to rise in the future due to an increase in the prevalence of obesity and diabetes. Current therapies for atherosclerosis mainly modulate lipid homeostasis and while successful at reducing the risk of a CVD-related death, they are associated with considerable residual risk and various side effects. There is, therefore, a need for alternative therapies aimed at regulating inflammation in order to reduce atherogenesis. This review will highlight the key role cytokines play during disease progression as well as potential therapeutic strategies to target them.
Collapse
|
15
|
Updates on cardiovascular comorbidities associated with psoriatic diseases: epidemiology and mechanisms. Rheumatol Int 2016; 37:97-105. [PMID: 27221457 DOI: 10.1007/s00296-016-3487-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 05/03/2016] [Indexed: 02/06/2023]
Abstract
Psoriasis and psoriatic arthritis are associated with a significantly increased risk of cardiovascular risk factors and major adverse cardiovascular events (MACE). Active research is ongoing to elucidate this relationship between psoriatic diseases and cardiovascular comorbidities, as well as their shared pathogenic mechanisms. This review focuses on (1) the epidemiologic association between psoriasis and cardiovascular risk factors, (2) the epidemiologic association between psoriasis and MACE, (3) the epidemiologic association between psoriatic arthritis, cardiovascular risk factors, and MACE, and (4) proposed mechanisms for the contribution of psoriatic diseases to cardiovascular diseases. The proposed mechanisms for shared pathogenesis between psoriatic diseases and cardiovascular diseases are inflammation, insulin resistance, dyslipidemia, angiogenesis, oxidative stress, and endothelial dysfunction. There is complex interplay and overlap among these mechanisms and their contributions to shared pathogenesis. Future translational research is necessary to elucidate the link between psoriatic diseases and cardiovascular diseases. Such findings may be applied clinically to improve the lives of psoriasis patients.
Collapse
|
16
|
Grosse GM, Schulz-Schaeffer WJ, Teebken OE, Schuppner R, Dirks M, Worthmann H, Lichtinghagen R, Maye G, Limbourg FP, Weissenborn K. Monocyte Subsets and Related Chemokines in Carotid Artery Stenosis and Ischemic Stroke. Int J Mol Sci 2016; 17:433. [PMID: 27023515 PMCID: PMC4848889 DOI: 10.3390/ijms17040433] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 03/08/2016] [Accepted: 03/17/2016] [Indexed: 01/01/2023] Open
Abstract
Carotid stenosis (CS) is an important cause of ischemic stroke. However, reliable markers for the purpose of identification of high-risk, so-called vulnerable carotid plaques, are still lacking. Monocyte subsets are crucial players in atherosclerosis and might also contribute to plaque rupture. In this study we, therefore, aimed to investigate the potential role of monocyte subsets and associated chemokines as clinical biomarkers for vulnerability of CS. Patients with symptomatic and asymptomatic CS (n = 21), patients with cardioembolic ischemic strokes (n = 11), and controls without any cardiovascular disorder (n = 11) were examined. Cardiovascular risk was quantified using the Essen Stroke Risk Score (ESRS). Monocyte subsets in peripheral blood were measured by quantitative flow cytometry. Plaque specimens were histologically analyzed. Furthermore, plasma levels of monocyte chemotactic protein 1 (MCP-1) and fractalkine were measured. Intermediate monocytes (Mon2) were significantly elevated in symptomatic and asymptomatic CS-patients compared to controls. Mon2 counts positively correlated with the ESRS. Moreover, stroke patients showed an elevation of Mon2 compared to controls, independent of the ESRS. MCP-1 levels were significantly higher in patients with symptomatic than in those with asymptomatic CS. Several histological criteria significantly differed between symptomatic and asymptomatic plaques. However, there was no association of monocyte subsets or chemokines with histological features of plaque vulnerability. Due to the multifactorial influence on monocyte subsets, the usability as clinical markers for plaque vulnerability seems to be limited. However, monocyte subsets may be critically involved in the pathology of CS.
Collapse
Affiliation(s)
- Gerrit M Grosse
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.
| | | | - Omke E Teebken
- Department of Vascular Surgery, Klinikum Peine, 31226 Peine, Germany.
| | - Ramona Schuppner
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.
| | - Meike Dirks
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.
| | - Hans Worthmann
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.
| | - Ralf Lichtinghagen
- Department of Clinical Chemistry, Hannover Medical School, 30625 Hannover, Germany.
| | - Gerrit Maye
- Department of Nephrology and Hypertension, Hannover Medical School, 30625 Hannover, Germany.
| | - Florian P Limbourg
- Department of Nephrology and Hypertension, Hannover Medical School, 30625 Hannover, Germany.
| | - Karin Weissenborn
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.
- Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany.
| |
Collapse
|
17
|
Voelter-Mahlknecht S. Epigenetic associations in relation to cardiovascular prevention and therapeutics. Clin Epigenetics 2016; 8:4. [PMID: 26779291 PMCID: PMC4714496 DOI: 10.1186/s13148-016-0170-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 01/06/2016] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVD) increasingly burden societies with vast financial and health care problems. Therefore, the importance of improving preventive and therapeutic measures against cardiovascular diseases is continually growing. To accomplish such improvements, research must focus particularly on understanding the underlying mechanisms of such diseases, as in the field of epigenetics, and pay more attention to strengthening primary prevention. To date, preliminary research has found a connection between DNA methylation, histone modifications, RNA-based mechanisms and the development of CVD like atherosclerosis, cardiac hypertrophy, myocardial infarction, and heart failure. Several therapeutic agents based on the findings of such research projects are currently being tested for use in clinical practice. Although these tests have produced promising data so far, no epigenetically active agents or drugs targeting histone acetylation and/or methylation have actually entered clinical trials for CVDs, nor have they been approved by the FDA. To ensure the most effective prevention and treatment possible, further studies are required to understand the complex relationship between epigenetic regulation and the development of CVD. Similarly, several classes of RNA therapeutics are currently under development. The use of miRNAs and their targets as diagnostic or prognostic markers for CVDs is promising, but has not yet been realized. Further studies are necessary to improve our understanding of the involvement of lncRNA in regulating gene expression changes underlying heart failure. Through the data obtained from such studies, specific therapeutic strategies to avoid heart failure based on interference with incRNA pathways could be developed. Together, research and testing findings raise hope for enhancing the therapeutic armamentarium. This review presents the currently available data concerning epigenetic mechanisms and compounds involved in cardiovascular diseases, as well as preventive and therapeutic approaches against them.
Collapse
Affiliation(s)
- Susanne Voelter-Mahlknecht
- University Hospital of Tuebingen, Institute of Occupational and Social Medicine and Health Services Research, Wilhelmstr. 27, 72074 Tuebingen, Germany
| |
Collapse
|
18
|
Tissue factor pathway inhibitor gene transfer prevents vascular smooth muscle cell proliferation by interfering with the MCP-3/CCR2 pathway. J Transl Med 2015; 95:1246-57. [PMID: 26302185 DOI: 10.1038/labinvest.2015.106] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 06/12/2015] [Accepted: 06/16/2015] [Indexed: 01/13/2023] Open
Abstract
Increased vascular smooth muscle cell (VSMC) proliferation substantially contributes to the pathogenesis of atherosclerosis and intimal hyperplasia after vascular injury. The importance of inflammation in VSMC proliferation is now being recognized. Preventing the inflammatory response is one therapeutic strategy that can be used to inhibit atherosclerosis in the clinic. The present study, using RNA interference and gene transfer techniques, was conducted to investigate the effect of monocyte chemotactic protein-3 (MCP-3) on VSMC proliferation that is a result of TNF-α stimulation, and whether overexpression of the tissue factor pathway inhibitor (TFPI) gene could prevent VSMC proliferation by blocking the MCP-3/CC chemokine receptor 2 (CCR2) pathway. Mouse VSMCs were infected in vitro with recombinant adenoviruses containing either mouse MCP-3-shRNA (Ad-MCP-3-shRNA), the TFPI gene (Ad-TFPI), or the negative control, which was shRNA encoding the sequence for EGFP (Ad-EGFP) or DMEM only. The cells were then stimulated with TNF-α for different time periods on the third day after gene transfer. The data show that VSMC proliferation in the Ad-MCP-3-shRNA and Ad-TFPI groups was markedly decreased using BrdU ELISA and MTT assays; MCP-3-shRNA and TFPI inhibited the expression of MCP-3 and CCR2 after long-term stimulation and inhibited the phosphorylation of ERK1/2 and AKT after short-term stimulation, as shown by ELISA and western blot analysis. This study provides convincing evidence that clarifies the effect of the proinflammatory factor MCP-3 in promoting VSMC proliferation. Our data also show, for the first time, that TFPI has an anti-proliferative role in TNF-α stimulated-VSMCs at least partly by interfering with the MCP-3/CCR2 pathway and then via suppression of the ERK1/2 and PI3K/AKT signaling pathways. We conclude that TFPI gene transfer may be a safe and effective therapeutic tool for treating atherosclerosis and intimal hyperplasia.
Collapse
|
19
|
Kivelä AM, Huusko J, Ylä-Herttuala S. Prospect and progress of gene therapy in treating atherosclerosis. Expert Opin Biol Ther 2015; 15:1699-712. [PMID: 26328616 DOI: 10.1517/14712598.2015.1084282] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Despite considerable improvements in therapies, atherosclerotic cardiovascular diseases remain the leading cause of death worldwide. Therefore, in addition to current treatment options, new therapeutic approaches are still needed. AREAS COVERED In this review, novel gene and RNA interference-based therapy approaches and promising target genes for treating atherosclerosis are addressed. In addition, relevant animal models for the demonstration of the efficacy of different gene therapy applications, and current progress toward more efficient, targeted and safer gene transfer vectors are reviewed. EXPERT OPINION Atherosclerosis represents a complex multifactorial disease that is dependent on the interplay between lipoprotein metabolism, cellular reactions and inflammation. Recent advances and novel targets, especially in the field of RNA interference-based therapies, are very promising. However, it should be noted that the modulation of a particular gene is not as clearly associated with a complex polygenic disease as it is in the case of monogenic diseases. A deeper understanding of molecular mechanisms of atherosclerosis, further progress in vector development and the demonstration of treatment efficacy in relevant animal models will be required before gene therapy of atherosclerosis meets its clinical reality.
Collapse
Affiliation(s)
- Annukka M Kivelä
- a 1 University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Department of Biotechnology and Molecular Medicine , Kuopio, Finland +358 403 552 075 ;
| | - Jenni Huusko
- a 1 University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Department of Biotechnology and Molecular Medicine , Kuopio, Finland +358 403 552 075 ;
| | - Seppo Ylä-Herttuala
- a 1 University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Department of Biotechnology and Molecular Medicine , Kuopio, Finland +358 403 552 075 ; .,b 2 Science Service Center , Kuopio, Finland.,c 3 Kuopio University Hospital, Gene Therapy Unit , Kuopio, Finland
| |
Collapse
|
20
|
Cytokines in atherosclerosis: Key players in all stages of disease and promising therapeutic targets. Cytokine Growth Factor Rev 2015; 26:673-85. [PMID: 26005197 PMCID: PMC4671520 DOI: 10.1016/j.cytogfr.2015.04.003] [Citation(s) in RCA: 327] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/27/2015] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, a chronic inflammatory disorder of the arteries, is responsible for most deaths in westernized societies with numbers increasing at a marked rate in developing countries. The disease is initiated by the activation of the endothelium by various risk factors leading to chemokine-mediated recruitment of immune cells. The uptake of modified lipoproteins by macrophages along with defective cholesterol efflux gives rise to foam cells associated with the fatty streak in the early phase of the disease. As the disease progresses, complex fibrotic plaques are produced as a result of lysis of foam cells, migration and proliferation of vascular smooth muscle cells and continued inflammatory response. Such plaques are stabilized by the extracellular matrix produced by smooth muscle cells and destabilized by matrix metalloproteinase from macrophages. Rupture of unstable plaques and subsequent thrombosis leads to clinical complications such as myocardial infarction. Cytokines are involved in all stages of atherosclerosis and have a profound influence on the pathogenesis of this disease. This review will describe our current understanding of the roles of different cytokines in atherosclerosis together with therapeutic approaches aimed at manipulating their actions.
Collapse
|
21
|
Wang M, Shah AM. Age-associated pro-inflammatory remodeling and functional phenotype in the heart and large arteries. J Mol Cell Cardiol 2015; 83:101-11. [PMID: 25665458 PMCID: PMC4459900 DOI: 10.1016/j.yjmcc.2015.02.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 01/20/2015] [Accepted: 02/02/2015] [Indexed: 01/12/2023]
Abstract
The aging population is increasing dramatically. Aging–associated stress simultaneously drives proinflammatory remodeling, involving angiotensin II and other factors, in both the heart and large arteries. The structural remodeling and functional changes that occur with aging include cardiac and vascular wall stiffening, systolic hypertension and suboptimal ventricular-arterial coupling, features that are often clinically silent and thus termed a silent syndrome. These age-related effects are the result of responses initiated by cardiovascular proinflammatory cells. Local proinflammatory signals are coupled between the heart and arteries due to common mechanical and humoral messengers within a closed circulating system. Thus, targeting proinflammatory signaling molecules would be a promising approach to improve age-associated suboptimal ventricular-arterial coupling, a major predisposing factor for the pathogenesis of clinical cardiovascular events such as heart failure.
Collapse
Affiliation(s)
- Mingyi Wang
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Blvd, Baltimore, MD 21224, USA.
| | - Ajay M Shah
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, UK.
| |
Collapse
|
22
|
Abstract
Atherosclerosis is an inflammatory disease of the vessel wall characterized by activation of the innate immune system, with macrophages as the main players, as well as the adaptive immune system, characterized by a Th1-dominant immune response. Cytokines play a major role in the initiation and regulation of inflammation. In recent years, many studies have investigated the role of these molecules in experimental models of atherosclerosis. While some cytokines such as TNF or IFNγ clearly had atherogenic effects, others such as IL-10 were found to be atheroprotective. However, studies investigating the different cytokines in experimental atherosclerosis revealed that the cytokine system is complex with both disease stage-dependent and site-specific effects. In this review, we strive to provide an overview of the main cytokines involved in atherosclerosis and to shed light on their individual role during atherogenesis.
Collapse
Affiliation(s)
- Pascal J H Kusters
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Academic Medical Center, L01-146.1, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU), Munich, Germany.
| |
Collapse
|
23
|
Grimaldi V, Vietri MT, Schiano C, Picascia A, De Pascale MR, Fiorito C, Casamassimi A, Napoli C. Epigenetic reprogramming in atherosclerosis. Curr Atheroscler Rep 2015; 17:476. [PMID: 25433555 DOI: 10.1007/s11883-014-0476-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent data support the involvement of epigenetic alterations in the pathogenesis of atherosclerosis. The most widely investigated epigenetic mechanism is DNA methylation although also histone code changes occur during the diverse steps of atherosclerosis, such as endothelial cell proliferation, vascular smooth muscle cell (SMC) differentiation, and inflammatory pathway activation. In this review, we focus on the main genes that are epigenetically modified during the atherogenic process, particularly nitric oxide synthase (NOS), estrogen receptors (ERs), collagen type XV alpha 1 (COL15A1), vascular endothelial growth factor receptor (VEGFR), and ten-eleven translocation (TET), which are involved in endothelial dysfunction; gamma interferon (IFN-γ), forkhead box p3 (FOXP3), and tumor necrosis factor-α (TNF-α), associated with atherosclerotic inflammatory process; and p66shc, lectin-like oxLDL receptor (LOX1), and apolipoprotein E (APOE) genes, which are regulated by high cholesterol and homocysteine (Hcy) levels. Furthermore, we also discuss the role of non-coding RNAs (ncRNA) in atherosclerosis. NcRNAs are involved in epigenetic regulation of endothelial function, SMC proliferation, cholesterol synthesis, lipid metabolism, and inflammatory response.
Collapse
Affiliation(s)
- Vincenzo Grimaldi
- U.O.C. Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], Regional Reference Laboratory of Transplant Immunology [LIT], Azienda Universitaria Policlinico (AOU), Second University of Naples (SUN), Piazza L. Miraglia 2, 80138, Naples, Italy,
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Huang GB, Quan L, Zeng YL, Yang J, Lu KH, Lu SS. Role of linker histone H1c during the reprogramming of Chinese swamp buffalo (Bubalus Bubalis) embryos produced by somatic cell nuclear transfer. Reprod Fertil Dev 2014; 28:302-9. [PMID: 25145348 DOI: 10.1071/rd14051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 05/02/2014] [Indexed: 11/23/2022] Open
Abstract
During reprogramming, there is exchange of histone H1c and the oocyte-specific linker histone, and H1c may play a critically important role in the reprogramming process of somatic cell nuclear transfer (SCNT). The aim of the present study was to investigate the role of the H1c gene in SCNT reprogramming in Chinese swamp buffalo (Bubalus bubalis) using RNA interference (RNAi). Chinese swamp buffalo H1c gene sequences were obtained and H1c-RNAi vectors were designed, synthesised and then transfected into a buffalo fetal skin fibroblast cell line. Expression of H1c was determined by real-time polymerase chain reaction to examine the efficiency of vector interference. These cells were then used as a nuclear donor for SCNT so as to observe the further development of SCNT embryos. Inhibition of H1c gene expression in donor cells significantly improved the developmental speed of embryos from the 1-cell to 8-cell stage. Furthermore, compared with the control group, inhibition of H1c gene expression significantly reduced the blastocyst formation rate. It is concluded that linker histone H1c is very important in SCNT reprogramming in Chinese swamp buffalo. Correct expression of the H1c gene plays a significant role in preimplantation embryonic development in B. bubalis.
Collapse
Affiliation(s)
- Gao-Bo Huang
- State Key Laboratory for Conservation and Utilisation of Subtropical Agro-Bioresources, Guangxi University, Nanning, Guangxi 53004, China
| | - Li Quan
- State Key Laboratory for Conservation and Utilisation of Subtropical Agro-Bioresources, Guangxi University, Nanning, Guangxi 53004, China
| | - Yong-Lian Zeng
- State Key Laboratory for Conservation and Utilisation of Subtropical Agro-Bioresources, Guangxi University, Nanning, Guangxi 53004, China
| | - Jian Yang
- Tiandiyang Biotechnology Co. Ltd, Nanning, Guangxi 530004, China
| | - Ke-Huan Lu
- State Key Laboratory for Conservation and Utilisation of Subtropical Agro-Bioresources, Guangxi University, Nanning, Guangxi 53004, China
| | - Sheng-Sheng Lu
- State Key Laboratory for Conservation and Utilisation of Subtropical Agro-Bioresources, Guangxi University, Nanning, Guangxi 53004, China
| |
Collapse
|
25
|
Wang T, Green LA, Gupta SK, Kim C, Wang L, Almodovar S, Flores SC, Prudovsky IA, Jolicoeur P, Liu Z, Clauss M. Transfer of intracellular HIV Nef to endothelium causes endothelial dysfunction. PLoS One 2014; 9:e91063. [PMID: 24608713 PMCID: PMC3946685 DOI: 10.1371/journal.pone.0091063] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 02/07/2014] [Indexed: 12/17/2022] Open
Abstract
With effective antiretroviral therapy (ART), cardiovascular diseases (CVD) are emerging as a major cause of morbidity and death in the aging HIV-infected population. To address whether HIV-Nef, a viral protein produced in infected cells even when virus production is halted by ART, can lead to endothelial activation and dysfunction, we tested Nef protein transfer to and activity in endothelial cells. We demonstrated that Nef is essential for major endothelial cell activating effects of HIV-infected Jurkat cells when in direct contact with the endothelium. In addition, we found that Nef protein in endothelial cells is sufficient to cause apoptosis, ROS generation and release of monocyte attractant protein-1 (MCP-1). The Nef protein-dependent endothelial activating effects can be best explained by our observation that Nef protein rapidly transfers from either HIV-infected or Nef-transfected Jurkat cells to endothelial cells between these two cell types. These results are of in vivo relevance as we demonstrated that Nef protein induces GFP transfer from T cells to endothelium in CD4.Nef.GFP transgenic mice and Nef is present in chimeric SIV-infected macaques. Analyzing the signal transduction effects of Nef in endothelial cells, we found that Nef-induced apoptosis is mediated through ROS-dependent mechanisms, while MCP-1 production is NF-kB dependent. Together, these data indicate that inhibition of Nef-associated pathways may be promising new therapeutic targets for reducing the risk for cardiovascular disease in the HIV-infected population.
Collapse
Affiliation(s)
- Ting Wang
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Cellular & Integrative Physiology and Indiana Center for Vascular Biology & Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- R. L. Roudebush VA Medical Center, Indianapolis, Indiana, United States
| | - Linden A. Green
- Department of Cellular & Integrative Physiology and Indiana Center for Vascular Biology & Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- R. L. Roudebush VA Medical Center, Indianapolis, Indiana, United States
| | - Samir K. Gupta
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Chul Kim
- Department of Cellular & Integrative Physiology and Indiana Center for Vascular Biology & Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Liang Wang
- Department of Cellular & Integrative Physiology and Indiana Center for Vascular Biology & Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- R. L. Roudebush VA Medical Center, Indianapolis, Indiana, United States
| | - Sharilyn Almodovar
- Department of Medicine, Pulmonary Sciences & Critical Care Medicine, University of Colorado, Denver, Colorado, United States of America
| | - Sonia C. Flores
- Department of Medicine, Pulmonary Sciences & Critical Care Medicine, University of Colorado, Denver, Colorado, United States of America
| | - Igor A. Prudovsky
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, United States of America
| | - Paul Jolicoeur
- Institut de Recherches Cliniques de Montréal University of Montréal, Montréal, Quebec, Canada
| | - Ziyue Liu
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Matthias Clauss
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Cellular & Integrative Physiology and Indiana Center for Vascular Biology & Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- R. L. Roudebush VA Medical Center, Indianapolis, Indiana, United States
- * E-mail:
| |
Collapse
|
26
|
|
27
|
Wang Q, Dong Z, Liu X, Song X, Song Q, Shang Q, Jiang Y, Guo C, Zhang L. Programmed cell death-4 deficiency prevents diet-induced obesity, adipose tissue inflammation, and insulin resistance. Diabetes 2013; 62:4132-4143. [PMID: 23990362 PMCID: PMC3837052 DOI: 10.2337/db13-0097] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 08/26/2013] [Indexed: 12/18/2022]
Abstract
Programmed cell death-4 (PDCD4), a selective protein translation inhibitor, has shown proinflammatory effect in some inflammatory diseases, but its roles in obesity remain unestablished. This study aims to investigate the effects of PDCD4 on obesity, inflammation, and insulin resistance. Surprisingly, high-fat diet (HFD)-fed PDCD4-deficient (PDCD4(-/-)) mice exhibited an absolutely lean phenotype together with improved insulin sensitivity. Compared with wild-type obese mice, HFD-fed PDCD4(-/-) mice showed higher energy expenditure, lower epididymal fat weight, and reduced macrophage infiltration inflammatory cytokine secretion in white adipose tissue (WAT). Alleviated hepatic steatosis along with decreased plasma levels of triglyceride and cholesterol was also observed in these mice. Importantly, PDCD4 appeared to disturb lipid metabolism via inhibiting the expression of liver X receptor (LXR)-α, a master modulator of lipid homeostasis, which was elevated in HFD-fed PDCD4(-/-) mice accompanied by upregulation of its target genes and relieved endoplasmic reticulum stress in WAT. These data demonstrate that PDCD4 deficiency protects mice against diet-induced obesity, WAT inflammation, and insulin resistance through restoring the expression of LXR-α, thereby proposing PDCD4 as a potential target for treating obesity-associated diseases.
Collapse
Affiliation(s)
- Qun Wang
- Department of Immunology, Shandong University School of Medicine, Jinan, Shangdong, China
| | - Zhaojing Dong
- Department of Immunology, Shandong University School of Medicine, Jinan, Shangdong, China
| | - Xianglan Liu
- Department of Immunology, Shandong University School of Medicine, Jinan, Shangdong, China
| | - Xingguo Song
- Department of Immunology, Shandong University School of Medicine, Jinan, Shangdong, China
| | - Qiang Song
- Department of Radiology, Shandong Chest Hospital, Jinan, Shangdong, China
| | - Qianwen Shang
- Department of Immunology, Shandong University School of Medicine, Jinan, Shangdong, China
| | - Yang Jiang
- Department of Immunology, Shandong University School of Medicine, Jinan, Shangdong, China
| | - Chun Guo
- Department of Immunology, Shandong University School of Medicine, Jinan, Shangdong, China
| | - Lining Zhang
- Department of Immunology, Shandong University School of Medicine, Jinan, Shangdong, China
| |
Collapse
|
28
|
Wang X, Li L, Li M, Dang X, Wan L, Wang N, Bi X, Gu C, Qiu S, Niu X, Zhu X, Wang L. Knockdown of mTOR by lentivirus‑mediated RNA interference suppresses atherosclerosis and stabilizes plaques via a decrease of macrophages by autophagy in apolipoprotein E‑deficient mice. Int J Mol Med 2013; 32:1215-21. [PMID: 24043133 DOI: 10.3892/ijmm.2013.1494] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 09/06/2013] [Indexed: 11/06/2022] Open
Abstract
Atherosclerotic plaque destabilization and rupture leads to acute coronary syndromes which cause serious damage to human health worldwide. However, there is currently a lack of efficient therapeutic methods. Mammalian target of rapamycin (mTOR) has been suggested to be involved in the development of atherosclerotic plaques and serves as a therapeutic target. The present study was performed to determine whether RNA interference (RNAi) of mTOR in vivo by LV‑mediated small hairpin RNA (shRNA) was capable of inhibiting the progression of atherosclerotic plaques. LV‑mediated shRNA against mTOR (LV‑shmTOR) was designed and obtained. Male apolipoprotein E‑deficient mice were fed a high‑fat diet and a constrictive collar was placed around the right carotid arteries of these mice to induce plaque formation. Eight weeks after surgery, mice were randomly divided into the mTOR RNA interference (LV‑shmTOR) group, receiving treatment with LV‑mTOR‑shRNA; the LV‑shCON group, receiving treatment with LV‑non‑specific‑shRNA; and the control group, receiving treatment with phosphate‑buffered saline. Following transfection, the mice were sacrificed to evaluate the effects of mTOR expression silencing on atherosclerosis. Transfection of LV‑mTOR‑shRNA markedly inhibited the mRNA and protein expression levels. Knockdown of mTOR ameliorated dysregulated blood lipid metabolism and stabilized aortic atherosclerotic plaques by decreasing the plaque area and increasing the fibrous cap and cap‑to‑core ratio. Furthermore, macrophages were decreased by silencing mTOR in atherosclerotic plaques. In addition, western blot analysis revealed that the knockdown of mTOR increased autophagy‑related protein 13 (Atg13) dephosphorylation and light chain 3‑I/light chain 3‑II (LC3‑I/LC3‑II) ratios, both of which were associated with a high activity of autophagy, suggesting an increase of autophagy in atherosclerotic plaques. Moreover, genes including matrix metalloproteinase 2, monocyte chemoattractant protein 1 and tissue factor, which promote plaque instability, were downregulated by silencing mTOR. These results demonstrate that LV‑mediated mTOR silencing by RNAi treatment induces macrophage autophagy and is a potential strategy for the treatment of atherosclerotic plaques.
Collapse
Affiliation(s)
- Xiaochuang Wang
- Department of Emergency Medicine, the Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Wang J, Jiang Y, Yang A, Sun W, Ma C, Ma S, Gong H, Shi Y, Wei J. Hyperhomocysteinemia-Induced Monocyte Chemoattractant Protein-1 Promoter DNA Methylation by Nuclear Factor-κB/DNA Methyltransferase 1 in Apolipoprotein E-Deficient Mice. Biores Open Access 2013; 2:118-27. [PMID: 23593564 PMCID: PMC3620495 DOI: 10.1089/biores.2012.0300] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Hyperhomocysteinemia is considered to be a significant risk factor in atherosclerosis and plays an important role in it. The purpose of this study was to determine the molecular mechanism of blood monocyte chemoattractant protein-1 (MCP-1) promoter DNA hypomethylation in the formation of atherosclerosis induced by hyperhomocysteinemia, and to explore the effect of nuclear factor-κB (NF-κB)/DNA methyltransferase 1 (DNMT1) in this mechanism. The atherosclerotic effect of MCP-1 in apolipoprotein E–deficient (ApoE−/−) and wild-type C57BL/6J mice was evaluated using atherosclerotic lesion area; serum NF-κB, MCP-1, and DNMT1 levels; and MCP-1 promoter DNA methylation expression. In vitro, the mechanism responsible for the effect of NF-κB/DNMT1 on foam cells was investigated by measuring NF-κB and DNMT1 levels to determine whether NF-κB/DNMT1 had an effect on gene expression. Compared with the control group, atherosclerotic lesions in ApoE−/− mice fed a high methionine diet significantly increased, as did the expression of MCP-1. In vitro study showed that pyrrolidine dithiocarbamate treatment down-regulated levels of NF-κB and raised DNMT1 concentrations, confirming the effect of NF-κB/DNMT1 in the MCP-1 promoter DNA methylation process. In conclusion, our results suggest that through NF-κB/DNMT1, MCP-1 promoter DNA hypomethylation may play a key role in formation of atherosclerosis under hyperhomocysteinemia.
Collapse
Affiliation(s)
- Ju Wang
- Department of Inspection, Ningxia Medical University , Yinchuan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Mäkinen PI, Ylä-Herttuala S. Therapeutic gene targeting approaches for the treatment of dyslipidemias and atherosclerosis. Curr Opin Lipidol 2013; 24:116-22. [PMID: 23314926 DOI: 10.1097/mol.0b013e32835da13c] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW Despite improved therapies, cardiovascular diseases are the leading cause of morbidity and mortality worldwide. Therefore, new therapeutic approaches are still needed. In the gene therapy field, RNA interference (RNAi) and regulation of microRNAs (miRNAs) have gained a lot of attention in addition to traditional overexpression based strategies. Here, recent findings in therapeutic gene silencing and modulation of small RNA expression related to atherogenesis and dyslipidemia are summarized. RECENT FINDINGS Novel gene therapy approaches for the treatment of hyperlipidemia have been addressed. Antisense oligonucleotide and RNAi-based therapies against apolipoprotein B100 and proprotein convertase subtilisin/kexin type 9 have shown already efficacy in preclinical and clinical trials. In addition, several miRNAs dysregulated in atherosclerotic lesions and regulating cholesterol homeostasis have been found, which may represent novel targets for future therapies. SUMMARY New therapies for lowering lipid levels are now being tested in clinical trials, and both antisense oligonucleotide and RNAi-based therapies have shown promising results in lowering cholesterol levels. However, the modulation of inflammatory component in atherosclerosis by gene therapy and targeting of the effects to plaques are still difficult challenges.
Collapse
MESH Headings
- Atherosclerosis/genetics
- Atherosclerosis/therapy
- Cholesterol, HDL/genetics
- Cholesterol, HDL/metabolism
- Cholesterol, LDL/genetics
- Cholesterol, LDL/metabolism
- Clinical Trials as Topic
- Dyslipidemias/genetics
- Dyslipidemias/therapy
- Epigenesis, Genetic
- Genetic Therapy
- Humans
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/metabolism
- RNA Interference
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
Collapse
Affiliation(s)
- Petri I Mäkinen
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland
| | | |
Collapse
|
31
|
Mukhopadhyay R. Mouse models of atherosclerosis: explaining critical roles of lipid metabolism and inflammation. J Appl Genet 2013; 54:185-92. [PMID: 23361320 DOI: 10.1007/s13353-013-0134-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 01/14/2013] [Accepted: 01/15/2013] [Indexed: 10/27/2022]
Abstract
Atherosclerosis is the most common cause of death globally. It is a complex disease involving morphological and cellular changes in vascular walls. Studying molecular mechanism of the disease is hindered by disease complexity and lack of robust noninvasive diagnostics in human. Mouse models are the most popular animal models that allow researchers to study the mechanism of disease progression. In this review we discuss the advantage and development of mouse as a model for atherosclerotic research. Along with commonly used models, this review discusses strains that are used to study the role of two critical processes associated with the disease-lipid metabolism and inflammation.
Collapse
Affiliation(s)
- Rupak Mukhopadhyay
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Assam, 784 028, India.
| |
Collapse
|