101
|
Zhang X, Xu X, Lu L, Wan X, Qin Y, Ruan W, Lv C, He L, Guo X. A new Mfn-2 related synthetic peptide promotes vascular smooth muscle cell apoptosis via regulating the mitochondrial apoptotic pathway by inhibiting Akt signaling. J Transl Med 2021; 19:395. [PMID: 34538249 PMCID: PMC8451139 DOI: 10.1186/s12967-021-03064-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 09/02/2021] [Indexed: 01/11/2023] Open
Abstract
Background Restenosis after angioplasty is a major challenge for the treatment of coronary artery diseases. Facilitation of vascular smooth muscle cell (VSMC) apoptosis may be an attractive approach to decrease the incidence of restenosis. We synthesized a 16-amino acid mitofusin-2 (Mfn-2) gene related peptide (MRSP) based on the sequence of the p21ras signature motif, the smallest functional sequence of the Mfn-2 gene with proapoptotic properties in VSMC. We investigated whether MRSP enhanced apoptotic activities to inhibit VSMC accumulation and neointimal hyperplasia in rats with carotid balloon injury. Methods VSMCs were treated with different concentrations of MRSP, the PI3K agonist 740 Y-P and the inhibitor LY294002. Cell apoptosis and related pathway molecules were assessed. MRSP was also given to rats with carotid artery balloon injury. Neointimal hyperplasia and cell apoptotic pathways were detected. Results In vitro experiments revealed that MRSP treatment significantly increased VSMC apoptosis and induced increases in procaspase-9 cleavage, caspase-3 activation, cytochrome c release from mitochondria to the cytoplasm and the Bax/Bcl-2 ratio but not caspase-8 expression, indicating that the mitochondrial apoptotic cascade was activated by MRSP, which might be attributed to suppression of the PI3K/Akt signaling pathway. We further found that the PI3K agonist 740 Y-P prevented and that the inhibitor LY294002 strengthened the proapoptotic effects of MRSP. MRSP strongly inhibited neointimal hyperplasia and VSMC accumulation, but increased VSMC apoptosis in the vascular wall after balloon injury. Moreover, MRSP substantially enhanced Bax and cleaved caspase-3 expression and decreased Bcl-2 levels in intima, accompanied by decreased levels of phosphorylated Akt and PI3K in vivo. Conclusions Taken together, the present study showed that MRSP treatment results in a strong proapoptotic effect by activating the mitochondrial apoptotic cascade through suppression of the PI3K/Akt pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03064-1.
Collapse
Affiliation(s)
- Xinxin Zhang
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiangyu Xu
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Li Lu
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoning Wan
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yating Qin
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weibin Ruan
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chao Lv
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lin He
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiaotong University, Shanghai, China
| | - Xiaomei Guo
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
102
|
Guo J, Yang Z, Wang X, Xu Y, Lu Y, Qin Z, Zhang L, Xu J, Wang W, Zhang J, Tang J. Advances in Nanomaterials for Injured Heart Repair. Front Bioeng Biotechnol 2021; 9:686684. [PMID: 34513807 PMCID: PMC8424111 DOI: 10.3389/fbioe.2021.686684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 08/09/2021] [Indexed: 11/30/2022] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is one of the leading causes of mortality worldwide. Because of the limited regenerative capacity of adult myocardium to compensate for the loss of heart tissue after ischemic infarction, scientists have been exploring the possible mechanisms involved in the pathological process of ASCVD and searching for alternative means to regenerate infarcted cardiac tissue. Although numerous studies have pursued innovative solutions for reversing the pathological process of ASCVD and improving the effectiveness of delivering therapeutics, the translation of those advances into downstream clinical applications remains unsatisfactory because of poor safety and low efficacy. Recently, nanomaterials (NMs) have emerged as a promising new strategy to strengthen both the efficacy and safety of ASCVD therapy. Thus, a comprehensive review of NMs used in ASCVD treatment will be useful. This paper presents an overview of the pathophysiological mechanisms of ASCVD and the multifunctional mechanisms of NM-based therapy, including antioxidative, anti-inflammation and antiapoptosis mechanisms. The technological improvements of NM delivery are summarized and the clinical transformations concerning the use of NMs to treat ASCVD are examined. Finally, this paper discusses the challenges and future perspectives of NMs in cardiac regeneration to provide insightful information for health professionals on the latest advancements in nanotechnologies for ASCVD treatment.
Collapse
Affiliation(s)
- Jiacheng Guo
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Zhenzhen Yang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xu Wang
- Department of Medical Record Management, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanyan Xu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Yongzheng Lu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Zhen Qin
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Li Zhang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Jing Xu
- Department of Cardiac Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Wang
- Henan Medical Association, Zhengzhou, China
| | - Jinying Zhang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Junnan Tang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| |
Collapse
|
103
|
Handke J, Kummer L, Weigand MA, Larmann J. Modulation of Peripheral CD4 +CD25 +Foxp3 + Regulatory T Cells Ameliorates Surgical Stress-Induced Atherosclerotic Plaque Progression in ApoE-Deficient Mice. Front Cardiovasc Med 2021; 8:682458. [PMID: 34485396 PMCID: PMC8416168 DOI: 10.3389/fcvm.2021.682458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/12/2021] [Indexed: 11/18/2022] Open
Abstract
Systemic inflammation associated with major surgery rapidly accelerates atherosclerotic plaque progression in mice. Regulatory T cells (Tregs) have emerged as important modulators of atherogenesis. In coronary artery disease patients, low frequency of Tregs constitutes an independent risk factor for cardiovascular complications after non-cardiac surgery. In this exploratory analysis, we investigate whether preoperative Treg levels affect surgery-induced atherosclerotic lesion destabilization in a murine model of perioperative stress. After 9 weeks of high-cholesterol diet, atherosclerotic apolipoprotein E-deficient mice with modulated Treg levels were subjected to a 30-minute surgical procedure consisting of general isoflurane anesthesia, laparotomy and moderate blood loss. Controls underwent general anesthesia only. Brachiocephalic arteries were harvested 3 days after the intervention for histomorphological analyses of atherosclerotic plaques. Tregs were depleted by a single dose of anti-CD25 monoclonal antibody (mAb) administered 6 days prior to the intervention. Expansion of Tregs was induced by daily injections of IL-2/anti-IL-2 complex (IL-2C) on three consecutive days starting 3 days before surgery. Isotype-matched antibodies and PBS served as controls. Antibody-mediated modulation was Treg-specific. IL-2C treatment resulted in an eight-fold elevation of peripheral CD4+CD25+Foxp3+ Tregs compared to mice administered with anti-CD25 mAb. In mice treated with PBS and anti-CD25 mAb, surgical stress response caused a significant increase of atherosclerotic plaque necrosis (PBS: p < 0.001; anti-CD25 mAb: p = 0.037). Preoperative Treg expansion abrogated perioperative necrotic core formation (p = 0.556) and significantly enhanced postoperative atherosclerotic plaque stability compared to PBS-treated mice (p = 0.036). Postoperative plaque volume (p = 0.960), stenosis (p = 0.693), lesional collagen (p = 0.258), as well as the relative macrophage (p = 0.625) and smooth muscle cell content (p = 0.178) remained largely unaffected by preoperative Treg levels. In atherosclerotic mice, therapeutic expansion of Tregs prior to major surgery mitigates rapid effects on perioperative stress-driven atherosclerotic plaque destabilization. Future studies will show, whether short-term interventions modulating perioperative inflammation qualify for prevention of cardiovascular events associated with major non-cardiac surgery.
Collapse
Affiliation(s)
- Jessica Handke
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Laura Kummer
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Markus A Weigand
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jan Larmann
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
104
|
Yu H, Kang D, Whang M, Kim T, Kim J. A Microfluidic Model Artery for Studying the Mechanobiology of Endothelial Cells. Adv Healthc Mater 2021; 10:e2100508. [PMID: 34297476 DOI: 10.1002/adhm.202100508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/25/2021] [Indexed: 11/07/2022]
Abstract
Recent vascular mechanobiology studies find that endothelial cells (ECs) convert multiple mechanical forces into functional responses in a nonadditive way, suggesting that signaling pathways such as those regulating cytoskeleton may be shared among the processes of converting individual forces. However, previous in vitro EC-culture platforms are inherent with extraneous mechanical components, which may saturate or insufficiently activate the shared signaling pathways and accordingly, may misguide EC mechanobiological responses being investigated. Here, a more physiologically relevant model artery is reported that accurately reproduces most of the mechanical forces found in vivo, which can be individually varied in any combination to pathological levels to achieve diseased states. Arterial geometries of normal and diseased states are also realized. By mimicking mechanical microenvironments of early-stage atherosclerosis, it is demonstrated that the elevated levels of the different types of stress experienced by ECs strongly correlate with the disruption of barrier integrity, suggesting that boundaries of an initial lesion could be sites for efficient disease progression.
Collapse
Affiliation(s)
- Hyeonji Yu
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107, Republic of Korea
| | - Dongwon Kang
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107, Republic of Korea
| | - Minji Whang
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107, Republic of Korea
| | - Taeyoung Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107, Republic of Korea
| | - Jungwook Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107, Republic of Korea
| |
Collapse
|
105
|
Hawkins CL, Davies MJ. Role of myeloperoxidase and oxidant formation in the extracellular environment in inflammation-induced tissue damage. Free Radic Biol Med 2021; 172:633-651. [PMID: 34246778 DOI: 10.1016/j.freeradbiomed.2021.07.007] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/04/2021] [Accepted: 07/05/2021] [Indexed: 12/30/2022]
Abstract
The heme peroxidase family generates a battery of oxidants both for synthetic purposes, and in the innate immune defence against pathogens. Myeloperoxidase (MPO) is the most promiscuous family member, generating powerful oxidizing species including hypochlorous acid (HOCl). Whilst HOCl formation is important in pathogen removal, this species is also implicated in host tissue damage and multiple inflammatory diseases. Significant oxidant formation and damage occurs extracellularly as a result of MPO release via phagolysosomal leakage, cell lysis, extracellular trap formation, and inappropriate trafficking. MPO binds strongly to extracellular biomolecules including polyanionic glycosaminoglycans, proteoglycans, proteins, and DNA. This localizes MPO and subsequent damage, at least partly, to specific sites and species, including extracellular matrix (ECM) components and plasma proteins/lipoproteins. Biopolymer-bound MPO retains, or has enhanced, catalytic activity, though evidence is also available for non-catalytic effects. These interactions, particularly at cell surfaces and with the ECM/glycocalyx induce cellular dysfunction and altered gene expression. MPO binds with higher affinity to some damaged ECM components, rationalizing its accumulation at sites of inflammation. MPO-damaged biomolecules and fragments act as chemo-attractants and cell activators, and can modulate gene and protein expression in naïve cells, consistent with an increasing cycle of MPO adhesion, activity, damage, and altered cell function at sites of leukocyte infiltration and activation, with subsequent tissue damage and dysfunction. MPO levels are used clinically both diagnostically and prognostically, and there is increasing interest in strategies to prevent MPO-mediated damage; therapeutic aspects are not discussed as these have been reviewed elsewhere.
Collapse
Affiliation(s)
- Clare L Hawkins
- Department of Biomedical Sciences, University of Copenhagen, Panum Institute, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark
| | - Michael J Davies
- Department of Biomedical Sciences, University of Copenhagen, Panum Institute, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark.
| |
Collapse
|
106
|
Wang J, Cai Y, Lu H, Zhang F, Zheng J. LncRNA APOA1-AS facilitates proliferation and migration and represses apoptosis of VSMCs through TAF15-mediated SMAD3 mRNA stabilization. Cell Cycle 2021; 20:1642-1652. [PMID: 34382908 DOI: 10.1080/15384101.2021.1951940] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Coronary atherosclerosis (CAS) is a major cause of cardiovascular disease. Long non-coding RNAs (lncRNAs) have been implicated as novel biomarkers in coronary artery disease (CAD). APOA1 antisense RNA (APOA1-AS) was proven to show high expression during atherosclerotic development, but no report has uncovered the detailed mechanism of APOA1-AS in CAS. Thus, this paper aims to explore the role of APOA1-AS in CAS. Vascular smooth muscle cells (VSMCs) were treated with oxidized low-density lipoprotein (ox-LDL) to mimic atherosclerosis-like injury. Quantitative real-time PCR (RT-qPCR) and western blot analysis analyzed gene expression. Cell counting kit-8 (CCK-8), wound healing assay, and flow cytometry were implemented to assess the function of APOA1-AS in modulating pathological phenotype of VSMCs. Results demonstrated that APOA1-AS was notably up-regulated in ox-LDL treated VSMCs (ox-LDL-VSMCs). The deficiency of APOA1-AS hindered proliferation and migration and stimulated apoptosis in ox-LDL-VSMCs. Mechanistically, APOA1-AS recruited TATA-box binding protein associated factor 15 (TAF15) protein to stabilized SMAD family member 3 (SMAD3) mRNA and activate the TGF-β/SMAD3 signaling pathway. In conclusion, APOA1-AS contributed to proliferation and migration and repressed apoptosis of VSMCs through TAF15-mediated SMAD3 mRNA stabilization, indicating that APOA1-AS could be a promising target for CAS.
Collapse
Affiliation(s)
- Jixiang Wang
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Ying Cai
- Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin, China
| | - Hui Lu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Fugeng Zhang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Junyi Zheng
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| |
Collapse
|
107
|
Contraction Measurements Using Three-Dimensional Fibrillar Collagen Gel Lattices. Methods Mol Biol 2021. [PMID: 34028737 DOI: 10.1007/978-1-0716-1382-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The ability of cells to contract is an important feature of many cell types. Methods to quantitate changes in the degree of contraction are important to study how cells respond to stimuli or change due to various pathologies. Here we describe a method of embedding cells in three-dimensional collagen lattices to measure contractile properties of cells in vitro.
Collapse
|
108
|
Gaul S, Schaeffer KM, Opitz L, Maeder C, Kogel A, Uhlmann L, Kalwa H, Wagner U, Haas J, Behzadi A, Pelegrin P, Boeckel JN, Laufs U. Extracellular NLRP3 inflammasome particles are internalized by human coronary artery smooth muscle cells and induce pro-atherogenic effects. Sci Rep 2021; 11:15156. [PMID: 34312415 PMCID: PMC8313534 DOI: 10.1038/s41598-021-94314-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 06/11/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammation driven by intracellular activation of the NLRP3 inflammasome is involved in the pathogenesis of a variety of diseases including vascular pathologies. Inflammasome specks are released into the extracellular compartment from disrupting pyroptotic cells. The potential uptake and function of extracellular NLRP3 inflammasomes in human coronary artery smooth muscle cells (HCASMC) are unknown. Fluorescently labeled NLRP3 inflammasome particles were isolated from a mutant NLRP3-YFP cell line and used to treat primary HCASMC for 4 and 24 h. Fluorescent and expressional analyses showed that extracellular NLRP3-YFP particles are internalized into HCASMC, where they remain active and stimulate intracellular caspase-1 (1.9-fold) and IL-1β (1.5-fold) activation without inducing pyroptotic cell death. Transcriptomic analysis revealed increased expression level of pro-inflammatory adhesion molecules (ICAM1, CADM1), NLRP3 and genes involved in cytoskleleton organization. The NLRP3-YFP particle-induced gene expression was not dependent on NLRP3 and caspase-1 activation. Instead, the effects were partly abrogated by blocking NFκB activation. Genes, upregulated by extracellular NLRP3 were validated in human carotid artery atheromatous plaques. Extracellular NLRP3-YFP inflammasome particles promoted the secretion of pro-atherogenic and inflammatory cytokines such as CCL2/MCP1, CXCL1 and IL-17E, and increased HCASMC migration (1.8-fold) and extracellular matrix production, such as fibronectin (5.8-fold) which was dependent on NFκB and NLRP3 activation. Extracellular NLRP3 inflammasome particles are internalized into human coronary artery smooth muscle cells where they induce pro-inflammatory and pro-atherogenic effects representing a novel mechanism of cell-cell communication and perpetuation of inflammation in atherosclerosis. Therefore, extracellular NLRP3 inflammasomes may be useful to improve the diagnosis of inflammatory diseases and the development of novel anti-inflammatory therapeutic strategies.
Collapse
Affiliation(s)
- Susanne Gaul
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany.
| | - Karen Marie Schaeffer
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany
| | - Lena Opitz
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany
| | - Christina Maeder
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany
| | - Alexander Kogel
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany
| | - Luisa Uhlmann
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany
| | - Hermann Kalwa
- Medical Faculty, Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Leipzig University, Leipzig, Germany
| | - Ulf Wagner
- Klinik für Gastroenterologie, Hepatologie, Infektionsmedizin, Rheumatologie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Jan Haas
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Amirhossein Behzadi
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany
| | - Pablo Pelegrin
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), Clinical University Hospital Virgen de La Arrixaca, Murcia, Spain
| | - Jes-Niels Boeckel
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany
| |
Collapse
|
109
|
Kota D, Kang L, Rickel A, Liu J, Smith S, Hong Z, Wang C. Low doses of zeolitic imidazolate framework-8 nanoparticles alter the actin organization and contractility of vascular smooth muscle cells. JOURNAL OF HAZARDOUS MATERIALS 2021; 414:125514. [PMID: 33647611 PMCID: PMC8144069 DOI: 10.1016/j.jhazmat.2021.125514] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/16/2021] [Accepted: 02/21/2021] [Indexed: 05/27/2023]
Abstract
Zeolitic imidazolate framework-8 (ZIF-8) nanoparticles have emerged as a promising platform for drug delivery and controlled release. Considering most ZIF-8 nanoparticle drug carriers are designed to be administered intravenously, and thus would directly contact vascular smooth muscle cells (VSMCs) in many circumstances, the potential interactions of ZIF-8 nanoparticles with VSMCs require investigation. Here, the effects of low doses of ZIF-8 nanoparticles on VSMC morphology, actin organization, and contractility are investigated. Two nanoscale imaging tools, atomic force microscopy, and direct stochastic optical reconstruction microscopy, show that even at the concentrations (12.5 and 25 µg/ml) that were deemed "safe" by conventional biochemical cell assays (MTT and LDH assays), ZIF-8 nanoparticles can still cause changes in cell morphology and actin cytoskeleton organization at the cell apical and basal surfaces. These cytoskeletal structural changes impair the contractility function of VSMCs in response to Angiotensin II, a classic vasoconstrictor. Based on intracellular zinc and actin polymerization assays, we conclude that the increased intracellular Zn2+ concentration due to the uptake and dissociation of ZIF-8 nanoparticles could cause the actin cytoskeleton dis-organization, as the elevated Zn2+ directly disrupts the actin assembly process, leading to altered actin organization such as branches and networks. Since the VSMC phenotype change and loss of contractility are fundamental to the development of atherosclerosis and related cardiovascular diseases, it is worth noting that these low doses of ZIF-8 nanoparticles administered intravenously could still be a safety concern in terms of cardiovascular risks. Moving forward, it is imperative to re-consider the "safe" nanoparticle dosages determined by biochemical cell assays alone, and take into account the impact of these nanoparticles on the biophysical characteristics of VSMCs, including changes in the actin cytoskeleton and cell morphology.
Collapse
Affiliation(s)
- Divya Kota
- Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, 501 East Saint Joseph Street, Rapid City, SD, USA 57701; BioSystems Networks & Translational Research (BioSNTR), 501 East Saint Joseph Street, Rapid City, SD, USA 57701
| | - Lin Kang
- Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, 501 East Saint Joseph Street, Rapid City, SD, USA 57701; BioSystems Networks & Translational Research (BioSNTR), 501 East Saint Joseph Street, Rapid City, SD, USA 57701
| | - Alex Rickel
- Biomedical Engineering, University of South Dakota, 4800 N Career Avenue, Sioux Falls, SD, USA 57107; BioSystems Networks & Translational Research (BioSNTR), 501 East Saint Joseph Street, Rapid City, SD, USA 57701
| | - Jinyuan Liu
- Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, 501 East Saint Joseph Street, Rapid City, SD, USA 57701; BioSystems Networks & Translational Research (BioSNTR), 501 East Saint Joseph Street, Rapid City, SD, USA 57701
| | - Steve Smith
- Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, 501 East Saint Joseph Street, Rapid City, SD, USA 57701; BioSystems Networks & Translational Research (BioSNTR), 501 East Saint Joseph Street, Rapid City, SD, USA 57701
| | - Zhongkui Hong
- Biomedical Engineering, University of South Dakota, 4800 N Career Avenue, Sioux Falls, SD, USA 57107; BioSystems Networks & Translational Research (BioSNTR), 501 East Saint Joseph Street, Rapid City, SD, USA 57701.
| | - Congzhou Wang
- Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, 501 East Saint Joseph Street, Rapid City, SD, USA 57701; BioSystems Networks & Translational Research (BioSNTR), 501 East Saint Joseph Street, Rapid City, SD, USA 57701.
| |
Collapse
|
110
|
Zhang Y, Ma A, Xi H, Chen N, Wang R, Yang C, Chen J, Lv P, Zheng F, Kang W. Antrodia cinnamomea ameliorates neointimal formation by inhibiting inflammatory cell infiltration through downregulation of adhesion molecule expression in vitro and in vivo. FOOD SCIENCE AND HUMAN WELLNESS 2021. [DOI: 10.1016/j.fshw.2021.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
111
|
Promyelocytic leukemia protein: an atherosclerosis suppressor protein? Clin Sci (Lond) 2021; 135:1557-1561. [PMID: 34192313 DOI: 10.1042/cs20210314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 11/17/2022]
Abstract
As many as 70% of cells in atherosclerotic plaque are vascular smooth muscle cell (VSMC) in origin, and pathways and proteins which regulate VSMC migration, proliferation, and phenotype modulation represent novel targets for rational drug design to reduce atherosclerotic vascular disease. In this volume of Clinical Science, Karle et al. demonstrate that tumor suppressor, promyelocytic leukemia protein (PML) plays an important role in regulation of VSMC phenotype and response to inflammatory stimuli (Clin Sci (2021) 135(7), 887-905; DOI: 10.1042/CS20201399). This important work demonstrates that PML, previously unrecognized as a participant in development of atherosclerosis, may represent a novel target for anti-atherosclerotic therapeutic modalities.
Collapse
|
112
|
Su C, Menon NV, Xu X, Teo YR, Cao H, Dalan R, Tay CY, Hou HW. A novel human arterial wall-on-a-chip to study endothelial inflammation and vascular smooth muscle cell migration in early atherosclerosis. LAB ON A CHIP 2021; 21:2359-2371. [PMID: 33978037 DOI: 10.1039/d1lc00131k] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Mechanistic understanding of atherosclerosis is largely hampered by the lack of a suitable in vitro human arterial model that recapitulates the arterial wall structure, and the interplay between different cell types and the surrounding extracellular matrix (ECM). This work introduces a novel microfluidic endothelial cell (EC)-smooth muscle cell (SMC) 3D co-culture platform that replicates the structural and biological aspects of the human arterial wall for modeling early atherosclerosis. Using a modified surface tension-based ECM patterning method, we established a well-defined intima-media-like structure, and identified an ECM composition (collagen I and Matrigel mixture) that retains the SMCs in a quiescent and aligned state, characteristic of a healthy artery. Endothelial stimulation with cytokines (IL-1β and TNFα) and oxidized low-density lipoprotein (oxLDL) was performed on-chip to study various early atherogenic events including endothelial inflammation (ICAM-1 expression), EC/SMC oxLDL uptake, SMC migration, and monocyte-EC adhesion. As a proof-of-concept for drug screening applications, we demonstrated the atheroprotective effects of vitamin D (1,25(OH)2D3) and metformin in mitigating cytokine-induced monocyte-EC adhesion and SMC migration. Overall, the developed arterial wall model facilitates quantitative and multi-factorial studies of EC and SMC phenotype in an atherogenic environment, and can be readily used as a platform technology to reconstitute multi-layered ECM tissue biointerfaces.
Collapse
Affiliation(s)
- Chengxun Su
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore. and Interdisciplinary Graduate School, Nanyang Technological University, Singapore, 639798, Singapore
| | - Nishanth Venugopal Menon
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore.
| | - Xiaohan Xu
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore.
| | - Yu Rong Teo
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore.
| | - Huan Cao
- School of Materials Science & Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Rinkoo Dalan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore and Endocrinology Department, Tan Tock Seng Hospital, Singapore, 308433, Singapore
| | - Chor Yong Tay
- School of Materials Science & Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Han Wei Hou
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore. and Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| |
Collapse
|
113
|
Circulating Biomarkers Reflecting Destabilization Mechanisms of Coronary Artery Plaques: Are We Looking for the Impossible? Biomolecules 2021; 11:biom11060881. [PMID: 34198543 PMCID: PMC8231770 DOI: 10.3390/biom11060881] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/11/2021] [Accepted: 06/12/2021] [Indexed: 12/12/2022] Open
Abstract
Despite significant strides to mitigate the complications of acute coronary syndrome (ACS), this clinical entity still represents a major global health burden. It has so far been well-established that most of the plaques leading to ACS are not a result of gradual narrowing of the vessel lumen, but rather a result of sudden disruption of vulnerable atherosclerotic plaques. As most of the developed imaging modalities for vulnerable plaque detection are invasive, multiple biomarkers were proposed to identify their presence. Owing to the pivotal role of lipids and inflammation in the pathophysiology of atherosclerosis, most of the biomarkers originated from one of those processes, whereas recent advancements in molecular sciences shed light on the use of microRNAs. Yet, at present there are no clinically implemented biomarkers or any other method for that matter that could non-invasively, yet reliably, diagnose the vulnerable plaque. Hence, in this review we summarized the available knowledge regarding the pathophysiology of plaque instability, the current evidence on potential biomarkers associated with plaque destabilization and finally, we discussed if search for biomarkers could one day bring us to non-invasive, cost-effective, yet valid way of diagnosing the vulnerable, rupture-prone coronary artery plaques.
Collapse
|
114
|
Chin DD, Poon C, Wang J, Joo J, Ong V, Jiang Z, Cheng K, Plotkin A, Magee GA, Chung EJ. miR-145 micelles mitigate atherosclerosis by modulating vascular smooth muscle cell phenotype. Biomaterials 2021; 273:120810. [PMID: 33892346 PMCID: PMC8152375 DOI: 10.1016/j.biomaterials.2021.120810] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/31/2021] [Accepted: 04/04/2021] [Indexed: 12/14/2022]
Abstract
In atherosclerosis, resident vascular smooth muscle cells (VSMCs) in the blood vessels become highly plastic and undergo phenotypic switching from the quiescent, contractile phenotype to the migratory and proliferative, synthetic phenotype. Additionally, recent VSMC lineage-tracing mouse models of atherosclerosis have found that VSMCs transdifferentiate into macrophage-like and osteochondrogenic cells and make up to 70% of cells found in atherosclerotic plaques. Given VSMC phenotypic switching is regulated by microRNA-145 (miR-145), we hypothesized that nanoparticle-mediated delivery of miR-145 to VSMCs has the potential to mitigate atherosclerosis development by inhibiting plaque-propagating cell types derived from VSMCs. To test our hypothesis, we synthesized miR-145 micelles targeting the C-C chemokine receptor-2 (CCR2), which is highly expressed on synthetic VSMCs. When miR-145 micelles were incubated with human aortic VSMCs in vitro, >90% miR-145 micelles escaped the lysosomal pathway in 4 hours and released the miR cargo under cytosolic levels of glutathione, an endogenous reducing agent. As such, miR-145 micelles rescued atheroprotective contractile markers, myocardin, α-SMA, and calponin, in synthetic VSMCs in vitro. In early-stage atherosclerotic ApoE-/- mice, one dose of miR-145 micelles prevented lesion growth by 49% and sustained an increased level of miR-145 expression after 2 weeks post-treatment. Additionally, miR-145 micelles inhibited 35% and 43% plaque growth compared to free miR-145 and PBS, respectively, in mid-stage atherosclerotic ApoE-/- mice. Collectively, we present a novel therapeutic strategy and cell target for atherosclerosis, and present miR-145 micelles as a viable nanotherapeutic that can intervene atherosclerosis progression at both early and later stages of disease.
Collapse
Affiliation(s)
- Deborah D Chin
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Christopher Poon
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Jonathan Wang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Johan Joo
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Victor Ong
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Zhangjingyi Jiang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Kayley Cheng
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Anastasia Plotkin
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States
| | - Gregory A Magee
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States; Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States; Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, 90089, United States; Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States; Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States; Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, United States.
| |
Collapse
|
115
|
Turpin C, Catan A, Meilhac O, Bourdon E, Canonne-Hergaux F, Rondeau P. Erythrocytes: Central Actors in Multiple Scenes of Atherosclerosis. Int J Mol Sci 2021; 22:ijms22115843. [PMID: 34072544 PMCID: PMC8198892 DOI: 10.3390/ijms22115843] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 12/16/2022] Open
Abstract
The development and progression of atherosclerosis (ATH) involves lipid accumulation, oxidative stress and both vascular and blood cell dysfunction. Erythrocytes, the main circulating cells in the body, exert determinant roles in the gas transport between tissues. Erythrocytes have long been considered as simple bystanders in cardiovascular diseases, including ATH. This review highlights recent knowledge concerning the role of erythrocytes being more than just passive gas carriers, as potent contributors to atherosclerotic plaque progression. Erythrocyte physiology and ATH pathology is first described. Then, a specific chapter delineates the numerous links between erythrocytes and atherogenesis. In particular, we discuss the impact of extravasated erythrocytes in plaque iron homeostasis with potential pathological consequences. Hyperglycaemia is recognised as a significant aggravating contributor to the development of ATH. Then, a special focus is made on glycoxidative modifications of erythrocytes and their role in ATH. This chapter includes recent data proposing glycoxidised erythrocytes as putative contributors to enhanced atherothrombosis in diabetic patients.
Collapse
Affiliation(s)
- Chloé Turpin
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
| | - Aurélie Catan
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
| | - Olivier Meilhac
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
- Centre Hospitalier Universitaire de La Réunion, 97400 Saint Denis, France
| | - Emmanuel Bourdon
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
| | | | - Philippe Rondeau
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
- Correspondence: ; Tel.: +262(0)-2-62-93-88-43; Fax: +262-(0)-2-62-93-88-01
| |
Collapse
|
116
|
Wang X, Chen S, Gao Y, Yu C, Nie Z, Lu R, Sun Y, Guan Z. MicroRNA-125b inhibits the proliferation of vascular smooth muscle cells induced by platelet-derived growth factor BB. Exp Ther Med 2021; 22:791. [PMID: 34093747 DOI: 10.3892/etm.2021.10223] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/29/2021] [Indexed: 12/14/2022] Open
Abstract
Excessive proliferation and migration of vascular smooth muscle cells (VSMCs) is the main cause of arteriosclerosis obliterans (ASO). The present study aimed to investigate the role of microRNA (miR)-125b on the proliferation and migration of VSMCs. Platelet-derived growth factor-BB (PDGF-BB; 20 ng/ml) was used to treat VSMCs to establish an in vitro model of ASO. VSMCs were transfected with miR-125b mimic to overexpress miR-125. Cell Counting kit-8 (CCK-8) and BrdU assays were performed to assess the proliferative ability of VSMCs, while Transwell and wound healing assays were performed to assess the migratory ability of VSMCs. Western blot and immunofluorescence analyses were performed to detect the expression levels of angio-associated migratory cell protein (AAMP) and serum response factor (SRF) in VSMCs following transfection with miR-125b mimic or inhibitor. The results demonstrated that miR-125b expression decreased following treatment with PDGF-BB, the effects of which were reversed following transfection with miR-125b mimic. According to the CCK-8 assay, the cell proliferative ability decreased by ~50% compared with the negative control (NC) group, and ~40% at day 4 based on the BrdU assay. The results of the Transwell and wound healing assays indicated that the migratory ability of VSMCs significantly decreased in the miR-125b mimic group compared with the NC group. Furthermore, western blot and immunofluorescence analyses demonstrated that AAMP and SRF expression levels decreased following transfection with miR-125b mimic compared with the NC group, the effects of which were reversed following transfection with miR-125 inhibitor. Taken together, the results of the present study suggested that miR-125b inhibits the proliferative and migratory abilities of VSMCs by regulating the expression levels of AAMP and SRF.
Collapse
Affiliation(s)
- Xiaogao Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Shiyuan Chen
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Yong Gao
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Chaowen Yu
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Zhonglin Nie
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Ran Lu
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Yong Sun
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Zeyu Guan
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| |
Collapse
|
117
|
Choi HY, Ruel I, Genest J. Identification of Docetaxel as a Potential Drug to Promote HDL Biogenesis. Front Pharmacol 2021; 12:679456. [PMID: 34093205 PMCID: PMC8176524 DOI: 10.3389/fphar.2021.679456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/07/2021] [Indexed: 12/13/2022] Open
Abstract
Objective: Our recent studies showed that desmocollin 1 (DSC1) binds to apoA-I in order to inhibit apoA-I-mediated high density lipoprotein (HDL) biogenesis in atherosclerotic plaques. To promote HDL biogenesis in the plaque, here we search for small molecules that block apoA-I-DSC1 interactions. Approach and Results: We combined mutational and computational mapping methods to show that amino acid residues 442-539 in the mature DSC1 protein form an apoA-I binding site (AIBS). Using a crystal structure of the AIBS, we carried out virtual screening of 10 million small molecules to estimate their binding affinities to the AIBS, followed by the selection of 51 high-affinity binding molecules as potential inhibitors of apoA-I-DSC1 interactions. Among the 51, the chemotherapy drug docetaxel showed the highest potency in promoting apoA-I-mediated HDL biogenesis in primary human skin fibroblasts with the half-maximal effective concentration of 0.72 nM. In silico docking studies suggest that the taxane ring in docetaxel binds to the AIBS and that the carbon-13 sidechain of the taxane tightens/stabilizes the binding. The HDL biogenic effect of docetaxel was also observed in two predominant cell types in atherosclerosis, macrophages and smooth muscle cells. Importantly, docetaxel promoted HDL biogenesis at concentrations much lower than those required for inducing cytotoxicity. Conclusion: Determination of the AIBS in DSC1 and AIBS structure-based virtual screening allowed us to identify docetaxel as a strong HDL biogenic agent. With the remarkable potency in promoting HDL biogenesis, a chemotherapy drug docetaxel may be repurposed to enhance atheroprotective HDL functions.
Collapse
Affiliation(s)
- Hong Y Choi
- Cardiovascular Research Laboratories, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - Isabelle Ruel
- Cardiovascular Research Laboratories, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - Jacques Genest
- Cardiovascular Research Laboratories, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
118
|
Ma G, Bi S, Zhang P. Long non-coding RNA MIAT regulates ox-LDL-induced cell proliferation, migration and invasion by miR-641/STIM1 axis in human vascular smooth muscle cells. BMC Cardiovasc Disord 2021; 21:248. [PMID: 34016053 PMCID: PMC8139145 DOI: 10.1186/s12872-021-02048-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/05/2021] [Indexed: 11/16/2022] Open
Abstract
Background Atherosclerosis (AS) is a primary cause of coronary heart and vascular diseases. Long non-coding RNAs (lncRNAs) are indicated to regulate AS progression. This study aimed to reveal the biological roles of lncRNA myocardial infarction associated transcript (MIAT) in oxidized low-density lipoprotein (ox-LDL)-induced human vascular smooth muscle cells (VSMCs). Methods The RNA levels of MIAT, microRNA-641 (miR-641) and stromal interaction molecule 1 (STIM1) were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The protein levels were determined by western blot analysis. Cell proliferation was assessed by cell colony formation and DNA content quantitation assays. Cell migration and invasion were demonstrated by wound-healing and transwell assays. The putative binding relationships between miR-641 and MIAT or STIM1 were predicted by starbase online database, and identified by dual-luciferase reporter and RNA immunoprecipitation assays. Results MIAT and STIM1 expression were substantially upregulated, whereas miR-641 expression was downregulated in ox-LDL-induced VSMCs compared with control groups. Functionally, MIAT silencing attenuated ox-LDL-induced cell proliferation, migration and invasion in VSMCs; however, these effects were impaired by miR-641 inhibitor. STIM1 overexpression also restrained miR-641-mediated impacts on cell proliferation and metastasis under ox-LDL. Mechanistically, MIAT acted as a sponge for miR-641, and miR-641 was associated with STIM1. Conclusions MIAT silencing hindered ox-LDL-induced cell proliferation, migration and invasion by downregulating STIM1 expression through binding to miR-641 in VSMCs. The mechanism provided us with a new target for AS therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-021-02048-9.
Collapse
Affiliation(s)
- Gang Ma
- Deptment of Cardiac Surgury, Zibo Central Hospital, Zibo, 255036, Shandong, People's Republic of China
| | - Shuting Bi
- Deptment of Cardiac Surgury, Zibo Central Hospital, Zibo, 255036, Shandong, People's Republic of China
| | - Pengfei Zhang
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, No.105, Jiefang Road, Jinan, 250013, Shandong, People's Republic of China. .,Department of Cardiac Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China.
| |
Collapse
|
119
|
Farra YM, Matz J, Ramkhelawon B, Oakes JM, Bellini C. Structural and functional remodeling of the female Apoe-/- mouse aorta due to chronic cigarette smoke exposure. Am J Physiol Heart Circ Physiol 2021; 320:H2270-H2282. [PMID: 33834870 DOI: 10.1152/ajpheart.00893.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Despite a decline in popularity over the past several decades, cigarette smoking remains a leading cause of cardiovascular morbidity and mortality. Yet, the effects of cigarette smoking on vascular structure and function are largely unknown. To evaluate changes in the mechanical properties of the aorta that occur with chronic smoking, we exposed female apolipoprotein E-deficient mice to mainstream cigarette smoke daily for 24 wk, with room air as control. By the time of euthanasia, cigarette-exposed mice had lower body mass but experienced larger systolic/diastolic blood pressure when compared with controls. Smoking was associated with significant wall thickening, reduced axial stretch, and circumferential material softening of the aorta. Although this contributed to maintaining intrinsic tissue stiffness at control levels despite larger pressure loads, the structural stiffness became significantly larger. Furthermore, the aorta from cigarette-exposed mice exhibited decreased ability to store elastic energy and augment diastolic blood flow. Histological analysis revealed a region-dependent increase in the cross-sectional area due to smoking. Increased smooth muscle and extracellular matrix content led to medial thickening in the ascending aorta, whereas collagen deposition increased the thickness of the descending thoracic and abdominal aorta. Atherosclerotic lesions were larger in exposed vessels and featured a necrotic core overlaid by a thinned fibrous cap and macrophage infiltration, consistent with a vulnerable phenotype. Collectively, our data indicate that cigarette smoking decreases the mechanical functionality of the aorta, inflicts morphometric alterations to distinct segments of the aorta, and accelerates the progression of atherosclerosis.NEW & NOTEWORTHY We studied the effects of chronic cigarette smoking on the structure and function of the aorta in a mouse model of nose-only aerosol inhalation. Our data indicated that exposure to cigarette smoke impairs vascular function by reducing the ability of the aorta to store elastic energy and by decreasing aortic distensibility. Combined with a more vulnerable atherosclerotic phenotype, these findings reveal the biomechanical mechanisms that support the development of cardiovascular disease due to cigarette smoking.
Collapse
Affiliation(s)
- Yasmeen M Farra
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Jacqueline Matz
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Bhama Ramkhelawon
- Division of Vascular Surgery, Department of Surgery, New York University Langone Health, New York City, New York.,Department of Cell Biology, New York University Langone Health, New York City, New York
| | - Jessica M Oakes
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Chiara Bellini
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| |
Collapse
|
120
|
Targeting Nanostrategies for Imaging of Atherosclerosis. CONTRAST MEDIA & MOLECULAR IMAGING 2021; 2021:6664471. [PMID: 33880112 PMCID: PMC8032543 DOI: 10.1155/2021/6664471] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/02/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023]
Abstract
Despite the progress in cardiovascular research, atherosclerosis still represents the main cause of death worldwide. Clinically, the diagnosis of Atherosclerotic Cardiovascular Disease (ASCVD) relies on imaging methodologies including X-ray angiography and computed tomography (CT), which however still fails in the identification of patients at high risk of plaque rupture, the main cause of severe clinical events as stroke and heart attack. Magnetic resonance imaging, which is characterized by very high spatial resolution, could provide a better characterization of atherosclerotic plaque (AP) anatomy and composition, aiding in the identification of “vulnerable” plaques. In this context, hydrogel matrices, which have been demonstrated able to boost relaxometric properties of Gd-based contrast agents (CAs) by the effect of Hydrodenticity, represent a valuable tool towards the precision imaging of ASCVD improving the performance of this class of CAs while reducing systemic toxicity. In particular, hydrogel nanoparticles encapsulating Gd-DTPA can further contribute to providing CA-specific accumulation in the AP by nanoparticle surface decoration triggering an active targeting of the AP with the overall effect of allowing an earlier and more accurate diagnosis. In this work, we tested crosslinked Hyaluronic Acid Nanoparticles (cHANPs) in the complex environment of human atherosclerotic plaque. In addition, the surface of cHANPs was decorated with the antibody anti-CD36 (Ab36-cHANPs) for the active targeting of AP-associated macrophages. Results demonstrate that the Hydrodenticity of cHANPs and Ab36-cHANPs is preserved in this complex system and, preliminarily, that interaction of these probes with the AP is present.
Collapse
|
121
|
Pan J, Cai Y, Wang L, Maehara A, Mintz GS, Tang D, Li Z. A prediction tool for plaque progression based on patient-specific multi-physical modeling. PLoS Comput Biol 2021; 17:e1008344. [PMID: 33780445 PMCID: PMC8057612 DOI: 10.1371/journal.pcbi.1008344] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 04/20/2021] [Accepted: 03/10/2021] [Indexed: 11/19/2022] Open
Abstract
Atherosclerotic plaque rupture is responsible for a majority of acute vascular syndromes and this study aims to develop a prediction tool for plaque progression and rupture. Based on the follow-up coronary intravascular ultrasound imaging data, we performed patient-specific multi-physical modeling study on four patients to obtain the evolutional processes of the microenvironment during plaque progression. Four main pathophysiological processes, i.e., lipid deposition, inflammatory response, migration and proliferation of smooth muscle cells (SMCs), and neovascularization were coupled based on the interactions demonstrated by experimental and clinical observations. A scoring table integrating the dynamic microenvironmental indicators with the classical risk index was proposed to differentiate their progression to stable and unstable plaques. The heterogeneity of plaque microenvironment for each patient was demonstrated by the growth curves of the main microenvironmental factors. The possible plaque developments were predicted by incorporating the systematic index with microenvironmental indicators. Five microenvironmental factors (LDL, ox-LDL, MCP-1, SMC, and foam cell) showed significant differences between stable and unstable group (p < 0.01). The inflammatory microenvironments (monocyte and macrophage) had negative correlations with the necrotic core (NC) expansion in the stable group, while very strong positive correlations in unstable group. The inflammatory microenvironment is strongly correlated to the NC expansion in unstable plaques, suggesting that the inflammatory factors may play an important role in the formation of a vulnerable plaque. This prediction tool will improve our understanding of the mechanism of plaque progression and provide a new strategy for early detection and prediction of high-risk plaques. Besides the traditional systematic factors, the influences of the local microenvironmental factors on atherosclerotic plaque progression have been demonstrated. Mathematical and computational modeling is an important tool to investigate the complex interplay between plaque progression and the microenvironment, and provides a potential way toward the prediction of plaque vulnerability according to the comprehensive evaluation of both morphological and/or biochemical factors in tissue level with microenvironmental factors in cellular level. We performed patient-specific multi-physical modeling study on four patients to obtain the evolutional processes of the microenvironment during plaque progression and predicted the possible plaque developments. A scoring table integrating the dynamic microenvironmental indicators with the classical risk index was proposed to differentiate their progression to stable and unstable plaques. Based on patient-specific imaging data, the mathematical model will provide a novel method to predict the changes of plaque microenvironment and improve ability to access the personal therapeutic strategy for atherosclerotic plaque.
Collapse
Affiliation(s)
- Jichao Pan
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing Jiangsu, China
| | - Yan Cai
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing Jiangsu, China
| | - Liang Wang
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing Jiangsu, China
| | - Akiko Maehara
- The Cardiovascular Research Foundation, New York, New York, United States of America
| | - Gary S Mintz
- The Cardiovascular Research Foundation, New York, New York, United States of America
| | - Dalin Tang
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing Jiangsu, China
- Mathematical Sciences Department, Worcester Polytechnic Institute, Massachusetts, United States of America
| | - Zhiyong Li
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing Jiangsu, China
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
122
|
Liberale L, Ministrini S, Carbone F, Camici GG, Montecucco F. Cytokines as therapeutic targets for cardio- and cerebrovascular diseases. Basic Res Cardiol 2021; 116:23. [PMID: 33770265 PMCID: PMC7997823 DOI: 10.1007/s00395-021-00863-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023]
Abstract
Despite major advances in prevention and treatment, cardiac and cerebral atherothrombotic complications still account for substantial morbidity and mortality worldwide. In this context, inflammation is involved in the chronic process leading atherosclerotic plaque formation and its complications, as well as in the maladaptive response to acute ischemic events. For this reason, modulation of inflammation is nowadays seen as a promising therapeutic strategy to counteract the burden of cardio- and cerebrovascular disease. Being produced and recognized by both inflammatory and vascular cells, the complex network of cytokines holds key functions in the crosstalk of these two systems and orchestrates the progression of atherothrombosis. By binding to membrane receptors, these soluble mediators trigger specific intracellular signaling pathways eventually leading to the activation of transcription factors and a deep modulation of cell function. Both stimulatory and inhibitory cytokines have been described and progressively reported as markers of disease or interesting therapeutic targets in the cardiovascular field. Nevertheless, cytokine inhibition is burdened by harmful side effects that will most likely prevent its chronic use in favor of acute administrations in well-selected subjects at high risk. Here, we summarize the current state of knowledge regarding the modulatory role of cytokines on atherosclerosis, myocardial infarction, and stroke. Then, we discuss evidence from clinical trials specifically targeting cytokines and the potential implication of these advances into daily clinical practice.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952, Schlieren, Switzerland.
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.
| | - Stefano Ministrini
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- Internal Medicine, Angiology and Atherosclerosis, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Genoa, Italy
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952, Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| |
Collapse
|
123
|
Dhaouadi N, Nehme A, Faour WH, Feugier P, Cerutti C, Kacem K, Eid AH, Li JY, Zibara K. Transforming growth factor-β1 inhibits interleukin-1β-induced expression of inflammatory genes and Cathepsin S activity in human vascular smooth muscle cells. Fundam Clin Pharmacol 2021; 35:979-988. [PMID: 33683760 DOI: 10.1111/fcp.12666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/28/2021] [Accepted: 03/04/2021] [Indexed: 12/26/2022]
Abstract
OBJECTIVE AND DESIGN This study investigated the opposite mechanisms by which IL-1β and TGF-β1 modulated the inflammatory and migratory phenotypes in cultured human intimal vascular smooth muscle cells vSMCs. MATERIALS AND TREATMENT Primary human vSMCs, obtained from twelve hypertensive patients who underwent carotid endarterectomy, were incubated for 24 hours with either 40 pM TGF-β1, or 1 nmol/L IL-1β, or their combination in presence or absence of anti-TGF-β neutralizing antibody. METHODS The expression levels of matrix metalloproteases and their inhibitors, and the elastolytic enzyme cathepsin S (CTSS) and its inhibitor cystatin C were evaluated with RT-PCR. CTSS activity was measured by fluorometry. RESULTS TGF-β1 reversed IL-1β-induced expression of iNOS, CXCL6, IL1R1, MMP12, and CTSS, while upregulated TIMP2 expression. Furthermore, anti-TGF-β neutralizing antibody abrogated TGF-β effects. Combination with IL-1β and TGF-β1 induced the expression of IL1α, IL1β, IL1R1, and CTSS, but suppressed CST3 expression. CTSS expression in the combination treatment was higher than that of cells treated with anti-TGF-β antibodies alone. Moreover, IL-1β-induced CTSS enzymatic activity was reduced when human vSMCs were co-treated with TGF-β, whereas this reduction was abrogated by anti-TGF-β neutralizing antibody. CONCLUSION TGF-β1 abrogated IL-1β-induced expression of inflammatory genes and elastolytic activity in cultured human vSMCs. Thus, TGF-β1 can play a crucial role in impairing IL-1β-induced vascular inflammation and damage involved in the etiology of cardiovascular diseases.
Collapse
Affiliation(s)
- Nedra Dhaouadi
- EA 4173 Génomique Fonctionnelle de l'Hypertension Artérielle, Université Lyon 1, Lyon, France.,Unité de Physiologie Intégrée, Laboratoire de Pathologies Vasculaires, Faculté des Sciences de Bizerte, Université de Carthage, Bizerte, Tunisia
| | - Ali Nehme
- EA 4173 Génomique Fonctionnelle de l'Hypertension Artérielle, Université Lyon 1, Lyon, France.,PRASE, Lebanese University, Beirut, Lebanon
| | - Wissam H Faour
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Patrick Feugier
- EA 4173 Génomique Fonctionnelle de l'Hypertension Artérielle, Université Lyon 1, Lyon, France
| | - Catherine Cerutti
- EA 4173 Génomique Fonctionnelle de l'Hypertension Artérielle, Université Lyon 1, Lyon, France
| | - Kamel Kacem
- Unité de Physiologie Intégrée, Laboratoire de Pathologies Vasculaires, Faculté des Sciences de Bizerte, Université de Carthage, Bizerte, Tunisia
| | - Ali H Eid
- Biomedical and Pharmaceutical Research Unit and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Jacques-Yuan Li
- EA 4173 Génomique Fonctionnelle de l'Hypertension Artérielle, Université Lyon 1, Lyon, France
| | - Kazem Zibara
- PRASE, Lebanese University, Beirut, Lebanon.,Department of Biology, Faculty of sciences - I, Lebanese University, Beirut, Lebanon
| |
Collapse
|
124
|
Sharma A, Choi JSY, Stefanovic N, Al-Sharea A, Simpson DS, Mukhamedova N, Jandeleit-Dahm K, Murphy AJ, Sviridov D, Vince JE, Ritchie RH, de Haan JB. Specific NLRP3 Inhibition Protects Against Diabetes-Associated Atherosclerosis. Diabetes 2021; 70:772-787. [PMID: 33323396 DOI: 10.2337/db20-0357] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 12/01/2020] [Indexed: 11/13/2022]
Abstract
Low-grade persistent inflammation is a feature of diabetes-driven vascular complications, in particular activation of the Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome to trigger the maturation and release of the inflammatory cytokine interleukin-1β (IL-1β). We investigated whether inhibiting the NLRP3 inflammasome, through the use of the specific small-molecule NLRP3 inhibitor MCC950, could reduce inflammation, improve vascular function, and protect against diabetes-associated atherosclerosis in the streptozotocin-induced diabetic apolipoprotein E-knockout mouse. Diabetes led to an approximately fourfold increase in atherosclerotic lesions throughout the aorta, which were significantly attenuated with MCC950 (P < 0.001). This reduction in lesions was associated with decreased monocyte-macrophage content, reduced necrotic core, attenuated inflammatory gene expression (IL-1β, tumor necrosis factor-α, intracellular adhesion molecule 1, and MCP-1; P < 0.05), and reduced oxidative stress, while maintaining fibrous cap thickness. Additionally, vascular function was improved in diabetic vessels of mice treated with MCC950 (P < 0.05). In a range of cell lines (murine bone marrow-derived macrophages, human monocytic THP-1 cells, phorbol 12-myristate 13-acetate-differentiated human macrophages, and aortic smooth muscle cells from humans with diabetes), MCC950 significantly reduced IL-1β and/or caspase-1 secretion and attenuated leukocyte-smooth muscle cell interactions under high glucose or lipopolysaccharide conditions. In summary, MCC950 reduces plaque development, promotes plaque stability, and improves vascular function, suggesting that targeting NLRP3-mediated inflammation is a novel therapeutic strategy to improve diabetes-associated vascular disease.
Collapse
Affiliation(s)
- Arpeeta Sharma
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Judy S Y Choi
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Nada Stefanovic
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Annas Al-Sharea
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Daniel S Simpson
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | | | - Karin Jandeleit-Dahm
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Dmitri Sviridov
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - James E Vince
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Rebecca H Ritchie
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Judy B de Haan
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia
- Faculty of Science, Engineering and Technology, Swinburne University, Melbourne, Victoria, Australia
| |
Collapse
|
125
|
Vacante F, Rodor J, Lalwani MK, Mahmoud AD, Bennett M, De Pace AL, Miller E, Van Kuijk K, de Bruijn J, Gijbels M, Williams TC, Clark MB, Scanlon JP, Doran AC, Montgomery R, Newby DE, Giacca M, O'Carroll D, Hadoke PWF, Denby L, Sluimer JC, Baker AH. CARMN Loss Regulates Smooth Muscle Cells and Accelerates Atherosclerosis in Mice. Circ Res 2021; 128:1258-1275. [PMID: 33622045 DOI: 10.1161/circresaha.120.318688] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Francesca Vacante
- Queens Medical Research Institute, BHF Centre for Cardiovascular Sciences (F.V., J.R., M.K.L., A.D.M., M.B., E.M., J.P.S., D.E.N., P.W.F.H., L.D., J.C.S., A.H.B.), University of Edinburgh, Scotland
| | - Julie Rodor
- Queens Medical Research Institute, BHF Centre for Cardiovascular Sciences (F.V., J.R., M.K.L., A.D.M., M.B., E.M., J.P.S., D.E.N., P.W.F.H., L.D., J.C.S., A.H.B.), University of Edinburgh, Scotland
| | - Mukesh K Lalwani
- Queens Medical Research Institute, BHF Centre for Cardiovascular Sciences (F.V., J.R., M.K.L., A.D.M., M.B., E.M., J.P.S., D.E.N., P.W.F.H., L.D., J.C.S., A.H.B.), University of Edinburgh, Scotland
| | - Amira D Mahmoud
- Queens Medical Research Institute, BHF Centre for Cardiovascular Sciences (F.V., J.R., M.K.L., A.D.M., M.B., E.M., J.P.S., D.E.N., P.W.F.H., L.D., J.C.S., A.H.B.), University of Edinburgh, Scotland
| | - Matthew Bennett
- Queens Medical Research Institute, BHF Centre for Cardiovascular Sciences (F.V., J.R., M.K.L., A.D.M., M.B., E.M., J.P.S., D.E.N., P.W.F.H., L.D., J.C.S., A.H.B.), University of Edinburgh, Scotland
| | - Azzurra L De Pace
- Institute for Regeneration and Repair, Centre for Regenerative Medicine (A.D.P., D.O.), University of Edinburgh, Scotland
| | - Eileen Miller
- Queens Medical Research Institute, BHF Centre for Cardiovascular Sciences (F.V., J.R., M.K.L., A.D.M., M.B., E.M., J.P.S., D.E.N., P.W.F.H., L.D., J.C.S., A.H.B.), University of Edinburgh, Scotland
| | - Kim Van Kuijk
- Pathology, Maastricht Medical Center, the Netherlands (K.V.K., J.d., J.C.S., A.H.B.)
| | - Jenny de Bruijn
- Pathology, Maastricht Medical Center, the Netherlands (K.V.K., J.d., J.C.S., A.H.B.)
| | - Marion Gijbels
- Pathology CARIM, Cardiovascular Research Institute Maastricht, GROW-School for Oncology and Developmental Biology, Maastricht University, the Netherlands (M. Gijbels)
| | - Thomas C Williams
- Insitute of Genetics and Molecular Medicine (T.C.W.), University of Edinburgh, Scotland
| | - Michael B Clark
- Centre for Stem Cell Systems, Department of Anatomy and Neuroscience, The University of Melbourne, Australia (M.B.C.)
| | - Jessica P Scanlon
- Queens Medical Research Institute, BHF Centre for Cardiovascular Sciences (F.V., J.R., M.K.L., A.D.M., M.B., E.M., J.P.S., D.E.N., P.W.F.H., L.D., J.C.S., A.H.B.), University of Edinburgh, Scotland
| | - Amanda C Doran
- Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (A.C.D)
| | | | - David E Newby
- Queens Medical Research Institute, BHF Centre for Cardiovascular Sciences (F.V., J.R., M.K.L., A.D.M., M.B., E.M., J.P.S., D.E.N., P.W.F.H., L.D., J.C.S., A.H.B.), University of Edinburgh, Scotland
| | - Mauro Giacca
- Medical Biochemistry, Experimental Vascular Biology, Amsterdam UMC, University of Amsterdam, the Netherlands (M. Gijbels).,King's College London, England (M. Giacca)
| | - Dónal O'Carroll
- Institute for Regeneration and Repair, Centre for Regenerative Medicine (A.D.P., D.O.), University of Edinburgh, Scotland
| | - Patrick W F Hadoke
- Queens Medical Research Institute, BHF Centre for Cardiovascular Sciences (F.V., J.R., M.K.L., A.D.M., M.B., E.M., J.P.S., D.E.N., P.W.F.H., L.D., J.C.S., A.H.B.), University of Edinburgh, Scotland
| | - Laura Denby
- Queens Medical Research Institute, BHF Centre for Cardiovascular Sciences (F.V., J.R., M.K.L., A.D.M., M.B., E.M., J.P.S., D.E.N., P.W.F.H., L.D., J.C.S., A.H.B.), University of Edinburgh, Scotland
| | - Judith C Sluimer
- Queens Medical Research Institute, BHF Centre for Cardiovascular Sciences (F.V., J.R., M.K.L., A.D.M., M.B., E.M., J.P.S., D.E.N., P.W.F.H., L.D., J.C.S., A.H.B.), University of Edinburgh, Scotland.,Pathology, Maastricht Medical Center, the Netherlands (K.V.K., J.d., J.C.S., A.H.B.)
| | - Andrew H Baker
- Queens Medical Research Institute, BHF Centre for Cardiovascular Sciences (F.V., J.R., M.K.L., A.D.M., M.B., E.M., J.P.S., D.E.N., P.W.F.H., L.D., J.C.S., A.H.B.), University of Edinburgh, Scotland.,Pathology, Maastricht Medical Center, the Netherlands (K.V.K., J.d., J.C.S., A.H.B.)
| |
Collapse
|
126
|
Quiles-Jiménez A, Gregersen I, Segers FM, Skarpengland T, Kroustallaki P, Yang K, Kong XY, Lauritzen KH, Olsen MB, Karlsen TR, Nyman TA, Sagen EL, Bjerkeli V, Suganthan R, Nygård S, Scheffler K, Prins J, Van der Veer E, Øgaard JD, Fløisand Y, Jørgensen HF, Holven KB, Biessen EA, Nilsen H, Dahl TB, Holm S, Bennett MR, Aukrust P, Bjørås M, Halvorsen B. DNA glycosylase Neil3 regulates vascular smooth muscle cell biology during atherosclerosis development. Atherosclerosis 2021; 324:123-132. [PMID: 33714552 DOI: 10.1016/j.atherosclerosis.2021.02.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 02/12/2021] [Accepted: 02/19/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Atherogenesis involves a complex interaction between immune cells and lipids, processes greatly influenced by the vascular smooth muscle cell (VSMC) phenotype. The DNA glycosylase NEIL3 has previously been shown to have a role in atherogenesis, though whether this is due to its ability to repair DNA damage or to other non-canonical functions is not yet clear. Hereby, we investigate the role of NEIL3 in atherogenesis, specifically in VSMC phenotypic modulation, which is critical in plaque formation and stability. METHODS Chow diet-fed atherosclerosis-prone Apoe-/- mice deficient in Neil3, and NEIL3-abrogated human primary aortic VSMCs were characterized by qPCR, and immunohistochemical and enzymatic-based assays; moreover, single-cell RNA sequencing, mRNA sequencing, and proteomics were used to map the molecular effects of Neil3/NEIL3 deficiency in the aortic VSMC phenotype. Furthermore, BrdU-based proliferation assays and Western blot were performed to elucidate the involvement of the Akt signaling pathway in the transdifferentiation of aortic VSMCs lacking Neil3/NEIL3. RESULTS We show that Neil3 deficiency increases atherosclerotic plaque development without affecting systemic lipids. This observation was associated with a shift in VSMC phenotype towards a proliferating, lipid-accumulating and secretory macrophage-like cell phenotype, without changes in DNA damage. VSMC transdifferentiation in Neil3-deficient mice encompassed increased activity of the Akt signaling pathway, supported by cell experiments showing Akt-dependent proliferation in NEIL3-abrogated human primary aortic VSMCs. CONCLUSIONS Our findings show that Neil3 deficiency promotes atherosclerosis development through non-canonical mechanisms affecting VSMC phenotype involving activation of the Akt signaling pathway.
Collapse
Affiliation(s)
- Ana Quiles-Jiménez
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ida Gregersen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Filip M Segers
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Department of Pharmacology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Tonje Skarpengland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Penelope Kroustallaki
- Department of Clinical Molecular Biology, University of Oslo, Akershus University Hospital, Lørenskog, Norway
| | - Kuan Yang
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Xiang Yi Kong
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Knut H Lauritzen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Maria B Olsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Tom Rune Karlsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Tuula A Nyman
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Ellen L Sagen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Vigdis Bjerkeli
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Rajikala Suganthan
- Department of Microbiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Ståle Nygård
- Bioinformatics Core Facility, Institute for Medical Informatics, Oslo University Hospital, Oslo, Norway
| | - Katja Scheffler
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jurriën Prins
- Einthoven Laboratory of Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands; Department of Internal Medicine, Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Eric Van der Veer
- Einthoven Laboratory of Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands; Department of Internal Medicine, Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jonas Ds Øgaard
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Yngvar Fløisand
- Department of Hematology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Helle F Jørgensen
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Kirsten B Holven
- Norwegian National Advisory Unit on Familial Hypercholesterolemia, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway; Faculty of Medicine, Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Erik A Biessen
- Department of Pathology, University of Maastricht, Maastricht, the Netherlands
| | - Hilde Nilsen
- Department of Clinical Molecular Biology, University of Oslo, Akershus University Hospital, Lørenskog, Norway
| | - Tuva B Dahl
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Sverre Holm
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Martin R Bennett
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Magnar Bjørås
- Department of Microbiology, Oslo University Hospital Rikshospitalet, Oslo, Norway; Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
127
|
Zheng L, Wang Z, Li Z, Wang M, Wang W, Chang G. MicroRNA-130a inhibits proliferation of vascular smooth muscle cells by suppressing autophagy via ATG2B. J Cell Mol Med 2021; 25:3829-3839. [PMID: 33611856 PMCID: PMC8051697 DOI: 10.1111/jcmm.16305] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 11/24/2020] [Accepted: 12/14/2020] [Indexed: 01/01/2023] Open
Abstract
Numerous microRNAs participate in regulating the pathological process of atherosclerosis. We have found miR-130a is one of the most significantly down-regulated microRNAs in arteriosclerosis obliterans. Our research explored the function of miR-130a in regulating proliferation by controlling autophagy in arteriosclerosis obliterans development. A Gene Ontology (GO) enrichment analysis of miR-130a target genes indicated a correlation between miR-130a and cell proliferation. Thus, cell cycle, CCK-8 assays and Western blot analysis were performed, and the results indicated that miR-130a overexpression in vascular smooth muscle cells (VSMCs) significantly attenuated cell proliferation, which was validated by an in vivo assay in a rat model. Moreover, autophagy is thought to be involved in the regulation of proliferation. As our results indicated, miR-130a could inhibit autophagy, and ATG2B was predicted to be a target of miR-130a. The autophagy inhibition effect of miR-130a overexpression was consistent with the effect of ATG2B knockdown. The results that ATG2B plasmids and miR-130a mimics were cotransfected in VSMCs further confirmed our conclusion. In addition, by using immunohistochemistry, the positive results of LC3 II/I and ATG2B in the rat model and artery vascular tissues from the patient were in accordance with in vitro data. In conclusion, our data demonstrate that miR-130a inhibits VSMCs proliferation via ATG2B, which indicates that miR-130a could be a potential therapeutic target that regulates autophagy in atherosclerosis obliterans.
Collapse
Affiliation(s)
- Liang Zheng
- Laboratory of General Surgery, Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhecun Wang
- Laboratory of General Surgery, Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zilun Li
- Laboratory of General Surgery, Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mian Wang
- Laboratory of General Surgery, Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenjian Wang
- Laboratory of General Surgery, Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guangqi Chang
- Laboratory of General Surgery, Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
128
|
Qian LL, Ji JJ, Guo JQ, Wu YP, Ma GS, Yao YY. Protective role of serpina3c as a novel thrombin inhibitor against atherosclerosis in mice. Clin Sci (Lond) 2021; 135:447-463. [PMID: 33458764 DOI: 10.1042/cs20201235] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/13/2021] [Accepted: 01/18/2021] [Indexed: 12/26/2022]
Abstract
Abnormal vascular smooth muscle cell (VSMC) proliferation is a critical step in the development of atherosclerosis. Serpina3c is a serine protease inhibitor (serpin) that plays a key role in metabolic diseases. The present study aimed to investigate the role of serpina3c in atherosclerosis and regulation of VSMC proliferation and possible mechanisms. Serpina3c is down-regulated during high-fat diet (HFD)-induced atherosclerosis. An Apoe-/-/serpina3c-/--double-knockout mouse model was used to determine the role of serpina3c in atherosclerosis after HFD for 12 weeks. Compared with Apoe-/- mice, the Apoe-/-/serpina3c-/- mice developed more severe atherosclerosis, and the number of VSMCs and macrophages in aortic plaques was significantly increased. The present study revealed serpina3c as a novel thrombin inhibitor that suppressed thrombin activity. In circulating plasma, thrombin activity was high in the Apoe-/-/serpina3c-/- mice, compared with Apoe-/- mice. Immunofluorescence staining showed thrombin and serpina3c colocalization in the liver and aortic cusp. In addition, inhibition of thrombin by dabigatran in serpina3c-/- mice reduced neointima lesion formation due to partial carotid artery ligation. Moreover, an in vitro study confirmed that thrombin activity was also decreased by serpina3c protein, supernatant and cell lysate that overexpressed serpina3c. The results of experiments showed that serpina3c negatively regulated VSMC proliferation in culture. The possible mechanism may involve serpina3c inhibition of ERK1/2 and JNK signaling in thrombin/PAR-1 system-mediated VSMC proliferation. Our results highlight a protective role for serpina3c as a novel thrombin inhibitor in the development of atherosclerosis, with serpina3c conferring protection through the thrombin/PAR-1 system to negatively regulate VSMC proliferation through ERK1/2 and JNK signaling.
Collapse
Affiliation(s)
- Ling-Lin Qian
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Jing-Jing Ji
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Jia-Qi Guo
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Yan-Ping Wu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Gen-Shan Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Yu-Yu Yao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| |
Collapse
|
129
|
Altered Vascular Extracellular Matrix in the Pathogenesis of Atherosclerosis. J Cardiovasc Transl Res 2021; 14:647-660. [PMID: 33420681 DOI: 10.1007/s12265-020-10091-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/01/2020] [Indexed: 12/18/2022]
Abstract
Cardiovascular disease continues to grow as a massive global health burden, with coronary artery disease being one of its most lethal varieties. The pathogenesis of atherosclerosis induces changes in the blood vessel and its extracellular matrix (ECM) in each vascular layer. The alteration of the ECM homeostasis has significant modulatory effects on the inflammatory response, the proliferation and migration of vascular smooth muscle cells, neointimal formation, and vascular fibrosis seen in atherosclerosis. In this literature review, the role of the ECM, the multitude of components, and alterations to these components in the pathogenesis of atherosclerosis are discussed with a focus on versatile cellular phenotypes in the structure of blood vessel. An understanding of the various effects of ECM alterations opens up a plethora of therapeutic options that would mitigate the substantial health toll of atherosclerosis on the global population.
Collapse
|
130
|
Implications for MicroRNA involvement in the prognosis and treatment of atherosclerosis. Mol Cell Biochem 2021; 476:1327-1336. [PMID: 33389489 DOI: 10.1007/s11010-020-03992-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/16/2020] [Indexed: 12/30/2022]
Abstract
MicroRNAs (miRNAs) are important molecules which implicated in various processes, such as differentiation, development, cell survival, cell apoptosis and also cell metabolism. Investigations over decades have revealed that various genes and signaling pathways are implicated in beginning and development of atherosclerosis, several miRNAs being involved in these dysregulated genes and pathways. miRNAs have provided new molecular vision in the context of atherosclerosis. miRNAs are considered as important regulators of cellular migration, differentiation, proliferation, lipid uptake and efflux, as well as cytokine production. Application of miRNAs as a biomarker in diagnosis, prognosis and even therapy is quiet exciting. Although animal researches showed promising results, still some practical difficulties and technical challenges need to be addressed before translation from researches into clinical practices. In this review, we present important data about three critical cells endothelial cell (EC), vascular smooth muscle cell (VSMC), and monocyte/macrophage and regulation of these cells through miRNAs. Furthermore, we discuss about the potential of miRNAs as a prognostic and diagnostic biomarkers, therapeutic opportunities and challenges, and also future perspective.
Collapse
|
131
|
Su F, Shi M, Zhang J, Zheng Q, Wang H, Li X, Chen J. MiR-223/NFAT5 signaling suppresses arterial smooth muscle cell proliferation and motility in vitro. Aging (Albany NY) 2020; 12:26188-26198. [PMID: 33373321 PMCID: PMC7803580 DOI: 10.18632/aging.202395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 10/31/2020] [Indexed: 01/11/2023]
Abstract
Aberrant proliferation and migration of vascular smooth muscle cells contributes to cardiovascular diseases (CVDs), including atherosclerosis. MicroRNA-223 (miR-223) protects against atherosclerotic CVDs. We investigated the contribution of miR-223 to platelet-derived growth factor-BB (PDGF-BB)-induced proliferation and migration of human aortic smooth muscle cells (HASMCs). We found that miR-223 was downregulated in PDGF-BB-treated HASMCs in a dose- and time-dependent manner, while nuclear factor of activated T cells 5 (NFAT5) was upregulated. Gain- and loss-of-function studies demonstrated that miR-223 treatment reduced PDGF-BB-induced HASMC proliferation and motility, whereas miR-223 inhibitor enhanced these processes. Moreover, NFAT5 was identified as a direct target of miR-223 in HASMC. The inhibitory effects of miR-223 on HASMC proliferation and migration were partly rescued by NFAT5 restoration. Overall, these findings suggest that miR-223 inhibits the PDGF-BB-induced proliferation and motility of HASMCs by targeting NFAT5 and that miR-223 and NFAT5 may be potential therapeutic targets for atherosclerosis.
Collapse
Affiliation(s)
- Feifei Su
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, Shaanxi, China.,Department of Cardiology, Air Force General Hospital, PLA, Beijing 100142, China
| | - Miaoqian Shi
- Department of Cardiology, PLA Army General Hospital, Beijing 100700, China
| | - Jian Zhang
- Department of Cardiology, Beijing Chest Hospital Heart Center, Capital Medical University, Beijing 101149, China
| | - Qiangsun Zheng
- Division of Cardiology, Second Affiliated Hospital of Xi’an Jiao Tong University, Xi'an 710004, Shaanxi, China
| | - Haichang Wang
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, Shaanxi, China
| | - Xue Li
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, Shaanxi, China
| | - Jianghong Chen
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, Shaanxi, China
| |
Collapse
|
132
|
Kim JH. Glucagon-like peptide-1 receptor agonist reduces di(2-ethylhexyl) phthalate-induced atherosclerotic processes in vascular smooth muscle cells. Physiol Res 2020; 69:1095-1102. [PMID: 33129247 DOI: 10.33549/physiolres.934480] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Glucagon-like peptide-1 receptor (GLP1R) agonist is an incretin hormone and regulates glucose metabolism. However, phthalates, known as endocrine disruptors, can interfere with hormone homeostasis. In the present study, we aimed to estimate the impact of GLP1R agonist on di(2 ethylhexyl) phthalate (DEHP)-induced atherosclerosis. For this purpose, the effects of GLP1R agonist on various atherogenesis-related cellular processes and pathways were assessed in vascular smooth muscle cells (VSMCs). DEHP-induced cell proliferation and migration were significantly decreased by GLP1R agonist in VSMCs. Protein levels of matrix metalloproteinase (MMP)-2 and MMP-9 were significantly decreased in cells exposed to GLP1R agonist, compared with DEHP-treated cells. Expression levels of intercellular adhesion molecule 1 and vascular cell adhesion molecule 1 were also reduced in GLP1R agonist-treated cells. Similarly, DEHP-associated phosphorylation of protein kinase B and extracellular signal-regulated kinase 1/2 was decreased in GLP1R agonist-treated cells, compared with DEHP-treated cells. Our findings suggest that treatment with GLP1R agonist counteracts the activation of pathways related to atherosclerosis.
Collapse
Affiliation(s)
- Jin Hee Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea.
| |
Collapse
|
133
|
Tan XW, Kobayashi K, Shen L, Inagaki J, Ide M, Hwang SS, Matsuura E. Antioxidative attributes of rice bran extracts in ameliorative effects of atherosclerosis-associated risk factors. Heliyon 2020; 6:e05743. [PMID: 33376820 PMCID: PMC7758525 DOI: 10.1016/j.heliyon.2020.e05743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/28/2020] [Accepted: 12/11/2020] [Indexed: 01/22/2023] Open
Abstract
Oxidative stress, chronic inflammation, dyslipidemia, hyperglycemia, and shear stress (physical effect) are risk factors associated with the pathogenesis of atherosclerosis. Rice bran, a by-product of rice milling process, is known to house polyphenols and vitamins which exhibit potent antioxidant and anti-inflammatory properties. Through recent emerging knowledge of rice bran in health and wellness, the present study was aimed to assess the ameliorative effects of rice bran extracts (RBE) derived from Japanese colored rice varieties in modulating risk factors of atherosclerosis via in vitro and in vivo study models. Pre-treatment of lipopolysaccharide (LPS)-stimulated murine J774A.1 macrophage-like cells with RBE alleviated nitric oxide (NO) overproduction and downregulated gene expressions of pro-inflammatory modulators: tumor necrosis factor-α (TNF-α), interleukin (IL)-α (IL-1α), IL-1β, IL-6, and inducible nitric oxide synthase (iNOS). In addition, RBE also significantly attenuated LPS-stimulated protein expressions of iNOS, TNF-α, IL-1α, and IL-6 in J774A.1 macrophage-like cells as compared to non-treated LPS control group. In in vivo, 12 weeks of RBE dietary supplementations significantly reduced (p < 0.05) total cholesterol, triglycerides, and pro-atherogenic oxidized LDL/β2-glycoprotein I (oxLDL/β2GPI) complexes at plasma levels, in high fat diet (HFD) induced low density lipoprotein receptor knockout (Ldlr−/-) mice. En face pathological assessments of murine aortas also revealed significant reductions by 38% (p < 0.05) in plaque sizes of RBE-supplemented HFD mice groups as compared to non RBE-supplemented HFD control mice group. Moreover, gene expressions of aortic (iNOS, TNF-α, IL-1β) and hepatic (TNF-α, IL-1α, IL-1β) pro-inflammatory modulators were also downregulated in RBE-supplemented mice groups. Present study has revealed the potent health attributes and application of RBE as a dietary supplement to attenuate risks of inadvertent oxidative damage and chronic inflammation underlying the pathogenesis of atherosclerosis. Intrinsically, present preliminary findings may provide global health prospects for future dietary implementation of RBE in management of atherosclerosis.
Collapse
Affiliation(s)
- Xian Wen Tan
- Department of Cell Chemistry, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuko Kobayashi
- Collaborative Research Center for OMIC, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Lianhua Shen
- Department of Pathophysiology, Zunyi Medical University, Guizhou, China
| | - Junko Inagaki
- Department of Cell Chemistry, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Masahiro Ide
- Department of Cell Chemistry, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan.,Food Function Research Team, Saito Laboratories, Japan Food Research Laboratories, Osaka, Japan
| | - Siaw San Hwang
- School of Chemical Engineering and Science, Faculty of Engineering, Computing and Science, Swinburne University of Technology Sarawak Campus, Sarawak, Malaysia
| | - Eiji Matsuura
- Department of Cell Chemistry, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan.,Collaborative Research Center for OMIC, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan.,Neutron Therapy Research Center, Okayama University, Okayama, Japan
| |
Collapse
|
134
|
Infection of Porphyromonas gingivalis Increases Phosphate-Induced Calcification of Vascular Smooth Muscle Cells. Cells 2020; 9:cells9122694. [PMID: 33334022 PMCID: PMC7765351 DOI: 10.3390/cells9122694] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/11/2020] [Accepted: 12/12/2020] [Indexed: 12/15/2022] Open
Abstract
Accumulating evidence suggests a link between periodontal disease and cardiovascular diseases. Vascular calcification is the pathological precipitation of phosphate and calcium in the vasculature and is closely associated with increased cardiovascular risk and mortality. In this study, we have demonstrated that the infection with Porphyromonas gingivalis (P. gingivalis), one of the major periodontal pathogens, increases inorganic phosphate-induced vascular calcification through the phenotype transition, apoptosis, and matrix vesicle release of vascular smooth muscle cells. Moreover, P. gingivalis infection accelerated the phosphate-induced calcium deposition in cultured rat aorta ex vivo. Taken together, our findings indicate that P. gingivalis contributes to the periodontal infection-related vascular diseases associated with vascular calcification.
Collapse
|
135
|
Mishra V, Banerjee A, Gandhi AB, Kaleem I, Alexander J, Hisbulla M, Kannichamy V, Valaiyaduppu Subas S, Hamid P. Stroke and Fabry Disease: A Review of Literature. Cureus 2020; 12:e12083. [PMID: 33489501 PMCID: PMC7805529 DOI: 10.7759/cureus.12083] [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: 12/15/2022] Open
Abstract
Fabry disease is an X-linked lysosomal storage disorder caused by a mutation in the alpha-galactosidase A (GLA) gene, leading to the deficiency of alpha-galactosidase A enzyme. The natural history of the affected patients (both males and females) includes neurovascular complications, such as cerebrovascular disease at a relatively young age. The pathophysiology behind the vascular involvement is primarily attributed to the accumulation of globotriaosylceramide and its derivatives in the vascular endothelium and vascular smooth muscle cells. MRI is the gold standard radiological investigation to detect the white matter lesions characteristic of Fabry disease's neurological involvement. More studies should focus on the utility of universally screening patients with young stroke for Fabry disease and the effectiveness of enzyme replacement therapy to prevent stroke. This review offers a synopsis of the current knowledge of the pathophysiology, neuroradiology, treatment, and prognosis of cerebrovascular disease in Fabry patients.
Collapse
Affiliation(s)
- Vinayak Mishra
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Amit Banerjee
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Arohi B Gandhi
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Ifrah Kaleem
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Josh Alexander
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Mohamed Hisbulla
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Vishmita Kannichamy
- General Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | | | - Pousette Hamid
- Neurology, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| |
Collapse
|
136
|
Tian M, Zhao J, Mi X, Wang K, Kong D, Mao H, Wang T. Progress in research on effect of PM
2.5
on occurrence and development of atherosclerosis. J Appl Toxicol 2020; 41:668-682. [DOI: 10.1002/jat.4110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/22/2020] [Accepted: 10/24/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Mengya Tian
- Tianjin Key Laboratory of Urban Transport Emission Research, State Environmental Protection Key Laboratory of Urban Ambient Air Particulate Matter Pollution Prevention and Control, College of Environmental Science and Engineering Nankai University Tianjin China
| | - Jingbo Zhao
- Tianjin Key Laboratory of Urban Transport Emission Research, State Environmental Protection Key Laboratory of Urban Ambient Air Particulate Matter Pollution Prevention and Control, College of Environmental Science and Engineering Nankai University Tianjin China
| | - Xingyan Mi
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences Nankai University Tianjin China
| | - Kai Wang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences Nankai University Tianjin China
| | - Deling Kong
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences Nankai University Tianjin China
| | - Hongjun Mao
- Tianjin Key Laboratory of Urban Transport Emission Research, State Environmental Protection Key Laboratory of Urban Ambient Air Particulate Matter Pollution Prevention and Control, College of Environmental Science and Engineering Nankai University Tianjin China
| | - Ting Wang
- Tianjin Key Laboratory of Urban Transport Emission Research, State Environmental Protection Key Laboratory of Urban Ambient Air Particulate Matter Pollution Prevention and Control, College of Environmental Science and Engineering Nankai University Tianjin China
| |
Collapse
|
137
|
Bruijn LE, van den Akker BEWM, van Rhijn CM, Hamming JF, Lindeman JHN. Extreme Diversity of the Human Vascular Mesenchymal Cell Landscape. J Am Heart Assoc 2020; 9:e017094. [PMID: 33190596 PMCID: PMC7763765 DOI: 10.1161/jaha.120.017094] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 10/05/2020] [Indexed: 12/17/2022]
Abstract
Background Human mesenchymal cells are culprit factors in vascular (patho)physiology and are hallmarked by phenotypic and functional heterogeneity. At present, they are subdivided by classic umbrella terms, such as "fibroblasts," "myofibroblasts," "smooth muscle cells," "fibrocytes," "mesangial cells," and "pericytes." However, a discriminative marker-based subclassification has to date not been established. Methods and Results As a first effort toward a classification scheme, a systematic literature search was performed to identify the most commonly used phenotypical and functional protein markers for characterizing and classifying vascular mesenchymal cell subpopulation(s). We next applied immunohistochemistry and immunofluorescence to inventory the expression pattern of identified markers on human aorta specimens representing early, intermediate, and end stages of human atherosclerotic disease. Included markers comprise markers for mesenchymal lineage (vimentin, FSP-1 [fibroblast-specific protein-1]/S100A4, cluster of differentiation (CD) 90/thymocyte differentiation antigen 1, and FAP [fibroblast activation protein]), contractile/non-contractile phenotype (α-smooth muscle actin, smooth muscle myosin heavy chain, and nonmuscle myosin heavy chain), and auxiliary contractile markers (h1-Calponin, h-Caldesmon, Desmin, SM22α [smooth muscle protein 22α], non-muscle myosin heavy chain, smooth muscle myosin heavy chain, Smoothelin-B, α-Tropomyosin, and Telokin) or adhesion proteins (Paxillin and Vinculin). Vimentin classified as the most inclusive lineage marker. Subset markers did not separate along classic lines of smooth muscle cell, myofibroblast, or fibroblast, but showed clear temporal and spatial diversity. Strong indications were found for presence of stem cells/Endothelial-to-Mesenchymal cell Transition and fibrocytes in specific aspects of the human atherosclerotic process. Conclusions This systematic evaluation shows a highly diverse and dynamic landscape for the human vascular mesenchymal cell population that is not captured by the classic nomenclature. Our observations stress the need for a consensus multiparameter subclass designation along the lines of the cluster of differentiation classification for leucocytes.
Collapse
Affiliation(s)
- Laura E. Bruijn
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| | | | - Connie M. van Rhijn
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| | - Jaap F. Hamming
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| | - Jan H. N. Lindeman
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
138
|
Feng Z, Zhu Y, Zhang J, Yang W, Chen Z, Li B. Hsa-circ_0010283 Regulates Oxidized Low-Density Lipoprotein-Induced Proliferation and Migration of Vascular Smooth Muscle Cells by Targeting the miR-133a-3p/Pregnancy-Associated Plasma Protein A Axis. Circ J 2020; 84:2259-2269. [PMID: 33162460 DOI: 10.1253/circj.cj-20-0345] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND The dysfunction of vascular smooth muscle cells (VSMCs) contributes to the development of atherosclerosis. This study aimed to investigate the role of circular RNA-0010283 (circ_0010283) in oxidized low-density lipoprotein (ox-LDL)-treated VSMCs and the associated action mechanism. METHODS AND RESULTS The expression of circ_0010283 was investigated using quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation was monitored by using a 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay. Cell apoptosis was detected by using flow cytometry assay. A transwell assay was performed to observe migration and invasion, and a scratch assay was implemented to test migration. The expression of proliferation, apoptosis and migration/invasion-related proteins was measured by using a western blot. The targeted relationship was predicted by using a bioinformatics tool (Starbase) and verified by using a dual-luciferase reporter assay, a RNA immunoprecipitation (RIP) assay and a RNA pull-down assay. circ_0010283 was highly expressed in serum samples from atherosclerosis patients and ox-LDL-treated human VSMCs (HVSMCs). circ_0010283 knockdown suppressed ox-LDL-induced proliferation, migration and invasion in HVSMCs. MicroRNA-133a-3p (miR-133a-3p) was confirmed as a target of circ_0010283, and miR-133a-3p deficiency reversed the effects of circ_0010283 knockdown. Moreover, pregnancy-associated plasma protein A (PAPPA) was targeted by miR-133a-3p, and PAPPA overexpression reversed the effects of miR-133a-3p restoration. Interestingly, circ_0010283 could regulate PAPPA expression by mediating miR-133a-3p. CONCLUSIONS circ_0010283 participated in ox-LDL-induced dysfunctions of HVSMCs by modulating the miR-133a-3p/PAPPA pathway, suggesting that circ_0010283 might be associated with atherosclerosis pathogenesis.
Collapse
Affiliation(s)
- Zibo Feng
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Youpeng Zhu
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Jing Zhang
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Wenbo Yang
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Zhimin Chen
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Binghui Li
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology
| |
Collapse
|
139
|
Chen X, He Y, Fu W, Sahebkar A, Tan Y, Xu S, Li H. Histone Deacetylases (HDACs) and Atherosclerosis: A Mechanistic and Pharmacological Review. Front Cell Dev Biol 2020; 8:581015. [PMID: 33282862 PMCID: PMC7688915 DOI: 10.3389/fcell.2020.581015] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis (AS), the most common underlying pathology for coronary artery disease, is a chronic inflammatory, proliferative disease in large- and medium-sized arteries. The vascular endothelium is important for maintaining vascular health. Endothelial dysfunction is a critical early event leading to AS, which is a major risk factor for stroke and myocardial infarction. Accumulating evidence has suggested the critical roles of histone deacetylases (HDACs) in regulating vascular cell homeostasis and AS. The purpose of this review is to present an updated view on the roles of HDACs (Class I, Class II, Class IV) and HDAC inhibitors in vascular dysfunction and AS. We also elaborate on the novel therapeutic targets and agents in atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaona Chen
- Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanhong He
- The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenjun Fu
- The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Polish Mother's Memorial Hospital Research Institute, Łódź, Poland
| | - Yuhui Tan
- Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Suowen Xu
- Department of Endocrinology, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hong Li
- Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
140
|
Kumrić M, Tičinović Kurir T, Borovac JA, Božić J. The Role of Natural Killer (NK) Cells in Acute Coronary Syndrome: A Comprehensive Review. Biomolecules 2020; 10:E1514. [PMID: 33167533 PMCID: PMC7694449 DOI: 10.3390/biom10111514] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/31/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
With poor outcomes and an immense financial burden, acute coronary syndrome (ACS) and its ischemic repercussions still present a major global health problem. Unfavorable outcomes seem to be mainly due to adverse cardiac remodeling. Since the inflammatory response takes an important role in remodeling secondary to myocardial infarction (MI), and as inflammation in this manner has not been completely elucidated, we attempted to give rise to a further understanding of ACS pathophysiology. Hence, in this review, we integrated current knowledge of complex communication networks between natural killer (NK) cells and immune and resident heart cells in the context of ACS. Based on available data, the role of NK cells seems to be important in the infarcted myocardium, where it affects heart remodeling. On the other hand, in atherosclerotic plaque, NK cells seem to be mere passers-by, except in the case of chronic infections by atherogenic pathogens. In that case, NK cells seem to support proinflammatory milieu. NK cell research is challenging due to ethical reasons, convergent evolution, and phenotypic diversity among individuals. Therefore, we argue that further research of NK cells in ACS is valuable, given their therapeutic potential in improving postischemic heart remodeling.
Collapse
Affiliation(s)
- Marko Kumrić
- Department of Pathophysiology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia; (M.K.); (T.T.K.); (J.A.B.)
| | - Tina Tičinović Kurir
- Department of Pathophysiology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia; (M.K.); (T.T.K.); (J.A.B.)
- Endocrinology Clinic, University Hospital of Split, Spinčićeva 1, 21000 Split, Croatia
| | - Josip A. Borovac
- Department of Pathophysiology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia; (M.K.); (T.T.K.); (J.A.B.)
- Institute of Emergency Medicine of Split-Dalmatia County (ZHM SDZ), Spinčićeva 1, 21000 Split, Croatia
| | - Joško Božić
- Department of Pathophysiology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia; (M.K.); (T.T.K.); (J.A.B.)
| |
Collapse
|
141
|
Simion V, Zhou H, Pierce JB, Yang D, Haemmig S, Tesmenitsky Y, Sukhova G, Stone PH, Libby P, Feinberg MW. LncRNA VINAS regulates atherosclerosis by modulating NF-κB and MAPK signaling. JCI Insight 2020; 5:140627. [PMID: 33021969 PMCID: PMC7710319 DOI: 10.1172/jci.insight.140627] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/24/2020] [Indexed: 12/19/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) play important roles in regulating diverse cellular processes in the vessel wall, including atherosclerosis. RNA-Seq profiling of intimal lesions revealed a lncRNA, VINAS (Vascular INflammation and Atherosclerosis lncRNA Sequence), that is enriched in the aortic intima and regulates vascular inflammation. Aortic intimal expression of VINAS fell with atherosclerotic progression and rose with regression. VINAS knockdown reduced atherosclerotic lesion formation by 55% in LDL receptor-deficient (LDLR-/-) mice, independent of effects on circulating lipids, by decreasing inflammation in the vessel wall. Loss- and gain-of-function studies in vitro demonstrated that VINAS serves as a critical regulator of inflammation by modulating NF-κB and MAPK signaling pathways. VINAS knockdown decreased the expression of key inflammatory markers, such as MCP-1, TNF-α, IL-1β, and COX-2, in endothelial cells (ECs), vascular smooth muscle cells, and bone marrow-derived macrophages. Moreover, VINAS silencing decreased expression of leukocyte adhesion molecules VCAM-1, E-selectin, and ICAM-1 and reduced monocyte adhesion to ECs. DEP domain containing 4 (DEPDC4), an evolutionary conserved human ortholog of VINAS with approximately 74% homology, showed similar regulation in human and pig atherosclerotic specimens. DEPDC4 knockdown replicated antiinflammatory effects of VINAS in human ECs. These findings reveal a potentially novel lncRNA that regulates vascular inflammation, with broad implications for vascular diseases.
Collapse
Affiliation(s)
- Viorel Simion
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Haoyang Zhou
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jacob B. Pierce
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Dafeng Yang
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Stefan Haemmig
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yevgenia Tesmenitsky
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Galina Sukhova
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Peter H. Stone
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Peter Libby
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark W. Feinberg
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
142
|
Zhang H, Bai Z, Zhu L, Liang Y, Fan X, Li J, Wen H, Shi T, Zhao Q, Wang Z. Hydrogen sulfide donors: Therapeutic potential in anti-atherosclerosis. Eur J Med Chem 2020; 205:112665. [DOI: 10.1016/j.ejmech.2020.112665] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/09/2020] [Accepted: 07/12/2020] [Indexed: 12/15/2022]
|
143
|
Tułowiecka N, Kotlęga D, Prowans P, Szczuko M. The Role of Resolvins: EPA and DHA Derivatives Can Be Useful in the Prevention and Treatment of Ischemic Stroke. Int J Mol Sci 2020; 21:E7628. [PMID: 33076354 PMCID: PMC7589657 DOI: 10.3390/ijms21207628] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/27/2020] [Accepted: 10/13/2020] [Indexed: 01/09/2023] Open
Abstract
INTRODUCTION Most ischemic strokes develop as a result of atherosclerosis, in which inflammation plays a key role. The synthesis cascade of proinflammatory mediators participates in the process induced in the vascular endothelium and platelets. Resolvins are anti-inflammatory mediators originating from eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), which may improve the prognosis related to atherosclerosis by inhibiting the production of proinflammatory cytokines, limiting neutrophil migration, or positively influencing phagocytosis. Although clinical trials with resolvin in humans after stroke have not been realized, they may soon find application. AIM The aim of the study was to review the available literature on the scope of the possibilities of the prevention and treatment of stroke with the use of resolvins, EPA and DHA derivatives. MATERIALS AND METHODS The review features articles published until 31 January 2020. The search for adequate literature was conducted using the keywords: stroke and resolvins. Over 150 articles were found. Studies not written in English, letters to the editor, conference abstracts, and duplicate information were excluded. RESULTS In several studies using the animal model, the supplementation of resolvin D2 decreased brain damage caused by myocardial infarction, and it reversed the neurological dysfunction of the brain. A decrease in the concentration of proinflammatory cytokines, such as TNF-α, Il-6, and Il-1β, was also observed, as well as a decrease in the scope of brain damage. In the context of stroke in animals, the treatment with resolvin D2 (RvD2) (injection) has a better effect than supplementation with DHA. CONCLUSIONS Resolvins are characterised by strong anti-inflammatory properties. Resolvins improve prognosis and decrease the risk of developing cardiovascular disease, consequently lowering the risk of stroke, and may find application in the treatment of stroke.
Collapse
Affiliation(s)
- Nikola Tułowiecka
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland;
| | - Dariusz Kotlęga
- Department of Neurology, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland;
- Department of Applied and Clinical Physiology, Collegium Medicum University of Zielona Gora, 65-417 Zielona Gora, Poland
| | - Piotr Prowans
- Clinic of Plastic, Endocrine and General Surgery, Pomeranian Medical University in Szczecin, 72-009 Police, Poland;
| | - Małgorzata Szczuko
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland;
| |
Collapse
|
144
|
Navas-Madroñal M, Castelblanco E, Camacho M, Consegal M, Ramirez-Morros A, Sarrias MR, Perez P, Alonso N, Galán M, Mauricio D. Role of the Scavenger Receptor CD36 in Accelerated Diabetic Atherosclerosis. Int J Mol Sci 2020; 21:ijms21197360. [PMID: 33028031 PMCID: PMC7583063 DOI: 10.3390/ijms21197360] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 01/22/2023] Open
Abstract
Diabetes mellitus entails increased atherosclerotic burden and medial arterial calcification, but the precise mechanisms are not fully elucidated. We aimed to investigate the implication of CD36 in inflammation and calcification processes orchestrated by vascular smooth muscle cells (VSMCs) under hyperglycemic and atherogenic conditions. We examined the expression of CD36, pro-inflammatory cytokines, endoplasmic reticulum (ER) stress markers, and mineralization-regulating enzymes by RT-PCR in human VSMCs, cultured in a medium containing normal (5 mM) or high glucose (22 mM) for 72 h with or without oxidized low-density lipoprotein (oxLDL) (24 h). The uptake of 1,1′-dioctadecyl-3,3,3′,3-tetramethylindocarbocyanine perchlorate-fluorescently (DiI) labeled oxLDL was quantified by flow cytometry and fluorimetry and calcification assays were performed in VSMC cultured in osteogenic medium and stained by alizarin red. We observed induction in the expression of CD36, cytokines, calcification markers, and ER stress markers under high glucose that was exacerbated by oxLDL. These results were confirmed in carotid plaques from subjects with diabetes versus non-diabetic subjects. Accordingly, the uptake of DiI-labeled oxLDL was increased after exposure to high glucose. The silencing of CD36 reduced the induction of CD36 and the expression of calcification enzymes and mineralization of VSMC. Our results indicate that CD36 signaling is partially involved in hyperglycemia and oxLDL-induced vascular calcification in diabetes.
Collapse
MESH Headings
- Aged
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- CD36 Antigens/genetics
- CD36 Antigens/metabolism
- Calcinosis/genetics
- Calcinosis/metabolism
- Calcinosis/pathology
- Diabetes Complications/genetics
- Diabetes Complications/metabolism
- Diabetes Complications/pathology
- Diabetes Mellitus/genetics
- Diabetes Mellitus/metabolism
- Diabetes Mellitus/pathology
- Female
- Flow Cytometry
- Glucose/adverse effects
- Humans
- Hyperglycemia/genetics
- Hyperglycemia/metabolism
- Hyperglycemia/pathology
- Inflammation/genetics
- Inflammation/metabolism
- Inflammation/pathology
- Lipoproteins, LDL/genetics
- Lipoproteins, LDL/metabolism
- Male
- Middle Aged
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Receptors, Scavenger/genetics
- Receptors, Scavenger/metabolism
Collapse
Affiliation(s)
- Miquel Navas-Madroñal
- Sant Pau Biomedical Research Institute (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (M.N.-M.); (M.C.); (M.C.)
| | - Esmeralda Castelblanco
- Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau & Sant Pau Biomedical Research Institute (IIB Sant Pau), 08041 Barcelona, Spain;
- Center for Biomedical Research on Diabetes and Associated Metabolic Diseases (CIBERDEM), 08025 Barcelona, Spain;
| | - Mercedes Camacho
- Sant Pau Biomedical Research Institute (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (M.N.-M.); (M.C.); (M.C.)
- Center for Biomedical Research on Cardiovascular Disease (CIBERCV), 28029 Madrid, Spain
| | - Marta Consegal
- Sant Pau Biomedical Research Institute (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (M.N.-M.); (M.C.); (M.C.)
| | - Anna Ramirez-Morros
- Department of Endocrinology & Nutrition, University Hospital and Health Sciences Research Institute Germans Trias i Pujol, 08916 Badalona, Spain;
| | - Maria Rosa Sarrias
- Innate Immunity Group, Health Sciences Research Institute Germans Trias i Pujol, Center for Biomedical Research on Liver and Digestive Diseases (CIBEREHD), 28029 Madrid, Spain;
| | - Paulina Perez
- Department of Angiology & Vascular Surgery, University Hospital and Health Sciences Germans Trias i Pujol, Autonomous University of Barcelona, 08916 Badalona, Spain;
| | - Nuria Alonso
- Center for Biomedical Research on Diabetes and Associated Metabolic Diseases (CIBERDEM), 08025 Barcelona, Spain;
- Department of Endocrinology & Nutrition, University Hospital and Health Sciences Research Institute Germans Trias i Pujol, 08916 Badalona, Spain;
| | - María Galán
- Sant Pau Biomedical Research Institute (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (M.N.-M.); (M.C.); (M.C.)
- Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau & Sant Pau Biomedical Research Institute (IIB Sant Pau), 08041 Barcelona, Spain;
- Correspondence: (M.G.); (D.M.); Tel.: +34-93-556-56-22 (M.G.); +34-93-556-56-61 (D.M.); Fax: +34-93-556-55-59 (M.G.); +34-93-556-56-02 (D.M.)
| | - Dídac Mauricio
- Center for Biomedical Research on Diabetes and Associated Metabolic Diseases (CIBERDEM), 08025 Barcelona, Spain;
- Center for Biomedical Research on Cardiovascular Disease (CIBERCV), 28029 Madrid, Spain
- Correspondence: (M.G.); (D.M.); Tel.: +34-93-556-56-22 (M.G.); +34-93-556-56-61 (D.M.); Fax: +34-93-556-55-59 (M.G.); +34-93-556-56-02 (D.M.)
| |
Collapse
|
145
|
Wang DP, Yu ZX, He ZC, Liao JF, Shen XB, Zhu PL, Chen WN, Lin X, Xu SH. Apolipoprotein L1 is transcriptionally regulated by SP1, IRF1 and IRF2 in hepatoma cells. FEBS Lett 2020; 594:3108-3121. [PMID: 32671843 DOI: 10.1002/1873-3468.13887] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/21/2020] [Accepted: 07/05/2020] [Indexed: 11/07/2022]
Abstract
Apolipoprotein L1 (APOL1) participates in lipid metabolism. Here, we investigate the mechanisms regulating APOL1 gene expression in hepatoma cells. We demonstrate that the -80-nt to +31-nt region of the APOL1 promoter, which contains one SP transcription factor binding GT box and an interferon regulatory factor (IRF) binding ISRE element, maintains the maximum activity. Mutation of the GT box and ISRE element dramatically reduces APOL1 promoter activity. EMSA and chromatin immunoprecipitation assay reveal that the transcription factors Sp1, IRF1 and IRF2 could interact with their cognate binding sites on the APOL1 promoter. Overexpression of Sp1, IRF1 and IRF2 increases promoter activity, leading to increased APOL1 mRNA and protein levels, while knockdown of Sp1, IRF1 and IRF2 has the opposite effects. These results demonstrate that the APOL1 gene could be regulated by Sp1, IRF1 and IRF2 in hepatoma cells.
Collapse
Affiliation(s)
- De-Ping Wang
- Department of Medical Intensive Care Unit, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Department of Endocrinology and Metabolism, Hongqi Hospital of MuDanJiang Medical College, Mudanjiang, China
| | - Zhao-Xi Yu
- Department of Medical Intensive Care Unit, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Zong-Cun He
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Jin-Fu Liao
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xue-Bin Shen
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Department of Cardiology, Affiliated Nanping First Hospital, Fujian Medical University, Nanping, China
| | - Peng-Li Zhu
- Department of Medical Intensive Care Unit, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Wan-Nan Chen
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xu Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Shang-Hua Xu
- Department of Cardiology, Affiliated Nanping First Hospital, Fujian Medical University, Nanping, China
| |
Collapse
|
146
|
Vitamin A as a Transcriptional Regulator of Cardiovascular Disease. HEARTS 2020. [DOI: 10.3390/hearts1020013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Vitamin A is a micronutrient and signaling molecule that regulates transcription, cellular differentiation, and organ homeostasis. Additionally, metabolites of Vitamin A are utilized as differentiation agents in the treatment of hematological cancers and skin disorders, necessitating further study into the effects of both nutrient deficiency and the exogenous delivery of Vitamin A and its metabolites on cardiovascular phenotypes. Though vitamin A/retinoids are well-known regulators of cardiac formation, recent evidence has emerged that supports their role as regulators of cardiac regeneration, postnatal cardiac function, and cardiovascular disease progression. We here review findings from genetic and pharmacological studies describing the regulation of both myocyte- and vascular-driven cardiac phenotypes by vitamin A signaling. We identify the relationship between retinoids and maladaptive processes during the pathological hypertrophy of the heart, with a focus on the activation of neurohormonal signaling and fetal transcription factors (Gata4, Tbx5). Finally, we assess how this information might be leveraged to develop novel therapeutic avenues.
Collapse
|
147
|
Angiotensin-converting enzyme 2 augments the effects of endothelial progenitor cells-exosomes on vascular smooth muscle cell phenotype transition. Cell Tissue Res 2020; 382:509-518. [PMID: 32852610 DOI: 10.1007/s00441-020-03259-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 07/08/2020] [Indexed: 12/19/2022]
Abstract
Phenotype transition of vascular smooth muscle cells (VSMCs) is implicated in vascular diseases. Angiotensin-converting enzyme 2 (ACE2) is a perspective cardiovascular target due to its ability of converting angiotensin (Ang II) to Ang (1-7). Our group recently showed that ACE2 can regulate the function of endothelial progenitor cell-derived exosomes (EPC-EXs). Here, we investigate whether ACE2 could affect the role of EPC-EXs on phenotype transition of VSMCs. After co-incubation with EXs released from EPC overexpressed ACE2 (EPC-EXsACE2), the ACE2 level and Ang II/Ang (1-7), proliferation/migration, phenotype gene, cytokine and NF-κB level on VSMCs were assessed. To determine the EX uptake route, VSMCs were pretreated with inhibitors. We found that (1) EPC-EXs and EPC-EXsACE2 were uptaken by VSMCs dominantly through caveolin-dependent endocytosis. (2) EPC-EXsACE2 remarkably increased the ACE2 level and decreased Ang II/Ang (1-7) in VSMCs activated by Ang II, whereas EPC-EXsACE2 pretreated by proteinase A blocked this effect. (3) EPC-EXsACE2 had better effects than EPC-EXs on reducing proliferation/migration activities and cytokine (MCP-1, TNF-α) secretion of Ang II-activated VSMCs. (4) EPC-EXs attenuated Ang II-induced VSMC synthetic phenotype change as evidenced by upregulated expressions of calponin and a-SMA and downregulated expressions of CRBP-1 and MYH10, associated with a decreased NF-κB level. EPC-EXsACE2 augmented these effects, which were attenuated by ACE2 inhibitor (DX600). In conclusion, EPC-EXsACE2 reduced Ang II-induced VSMC phenotype change by conveying functional ACE2 to downregulate the activated NF-κB pathway.
Collapse
|
148
|
Liu D, Song J, Ji X, Liu Z, Li T, Hu B. PRDM16 Upregulation Induced by MicroRNA-448 Inhibition Alleviates Atherosclerosis via the TGF-β Signaling Pathway Inactivation. Front Physiol 2020; 11:846. [PMID: 32848826 PMCID: PMC7431868 DOI: 10.3389/fphys.2020.00846] [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: 02/28/2020] [Accepted: 06/24/2020] [Indexed: 12/14/2022] Open
Abstract
The dysregulated expression of microRNAs (miRs) has been associated with pathological and physiological processes of atherosclerosis (AS). In addition, PR domain-containing 16 (PRDM16), a transcriptional mediator of brown fat cell identity and smooth muscle cell activities, may be involved in the hypercholesterolemia during development of AS. The bioinformatic analysis identified a regulatory miR-448 of PRDM16. Hence, the current study aimed to explore whether miR-448 influenced the activities of aortic smooth muscle cell (ASMCs) in AS. We validated that miR-448 was highly expressed in peripheral blood of patients with AS and aortic smooth muscle of AS model mice. Whereas, PRDM16 was downregulated in the aortic smooth muscle of AS model mice. PRDM16 overexpression was observed to inhibit oxidative stress injury and cell proliferation, and promote apoptosis of ASMCs. Mechanistic studies revealed that miR-448 targeted PRDM16 and negatively regulated the PRDM16 expression, while PRDM16 blocked the TGF-β signaling pathway. Furthermore, Downregulated miR-448 alleviated oxidative stress injury, and attenuated ASMC cell proliferation, migration and enhanced cell apoptosis through upregulation of PRDM16. Taken together, silencing of miR-448 upregulates PRDM16 and inactivates the TGF-β signaling pathway, thereby impeding development of AS by repressing the proliferation, migration and invasion of ASMCs.
Collapse
Affiliation(s)
| | | | | | | | | | - Bo Hu
- Department of Emergency, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
149
|
Insights into pharmacological mechanisms of polydatin in targeting risk factors-mediated atherosclerosis. Life Sci 2020; 254:117756. [DOI: 10.1016/j.lfs.2020.117756] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/20/2022]
|
150
|
Wang W, Ma F, Zhang H. MicroRNA-374 is a potential diagnostic biomarker for atherosclerosis and regulates the proliferation and migration of vascular smooth muscle cells. Cardiovasc Diagn Ther 2020; 10:687-694. [PMID: 32968625 DOI: 10.21037/cdt-20-444] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Background The occurrence and development of atherosclerosis (AS) are closely related to the abnormality of vascular smooth muscle cells (VSMCs), and multiple microRNAs (miRNAs) have been reported to participate in the pathogenesis of AS. This study explored the expression and clinical value of miR-374 in the serum of AS patients, and analyzed its effect on the proliferation and migration of VSMCs. Methods The expression levels of miR-374 in the serum of 102 asymptomatic patients with AS and 89 healthy patients were detected by fluorescence quantitative PCR. The diagnostic value of miR-374 was evaluated through the receiver operating characteristic (ROC) curve. What's more, CCK-8 and Transwell assays were used to analyze the effects of miR-374 on the proliferation and migration of VSMCs. Results The expression level of miR-374 in the serum of AS patients was significantly higher than that of the control group. At the same time, the expression of miR-374 in AS patients was positively correlated with carotid intima-media thickness (CIMT). The area under the ROC curve is 0.824. Furthermore, overexpression of miR-374 significantly promoted the proliferation and migration of VSMCs, whereas reducing miR-374 inhibited the proliferation and migration of VSMCs. Conclusions The high expression of miR-374 may be a potential diagnostic marker for AS, and overexpression of miR-374 may play a role in AS by promoting the proliferation and migration of VSMCs.
Collapse
Affiliation(s)
- Weihong Wang
- Department of Healthcare, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fenghua Ma
- Department of Healthcare, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongyan Zhang
- Department of Thoracic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|