1
|
Crespo-Avilan GE, Hernandez-Resendiz S, Ramachandra CJ, Ungureanu V, Lin YH, Lu S, Bernhagen J, El Bounkari O, Preissner KT, Liehn EA, Hausenloy DJ. Metabolic reprogramming of immune cells by mitochondrial division inhibitor-1 to prevent post-vascular injury neointimal hyperplasia. Atherosclerosis 2024; 390:117450. [PMID: 38266625 DOI: 10.1016/j.atherosclerosis.2024.117450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/23/2023] [Accepted: 01/09/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND AND AIMS New treatments are needed to prevent neointimal hyperplasia that contributes to post-angioplasty and stent restenosis in patients with coronary artery disease (CAD) and peripheral arterial disease (PAD). We investigated whether modulating mitochondrial function using mitochondrial division inhibitor-1 (Mdivi-1) could reduce post-vascular injury neointimal hyperplasia by metabolic reprogramming of macrophages from a pro-inflammatory to anti-inflammatory phenotype. METHODS AND RESULTS In vivo Mdivi-1 treatment of Apoe-/- mice fed a high-fat diet and subjected to carotid-wire injury decreased neointimal hyperplasia by 68%, reduced numbers of plaque vascular smooth muscle cells and pro-inflammatory M1-like macrophages, and decreased plaque inflammation, endothelial activation, and apoptosis, when compared to control. Mdivi-1 treatment of human THP-1 macrophages shifted polarization from a pro-inflammatory M1-like to an anti-inflammatory M2-like phenotype, reduced monocyte chemotaxis and migration to CCL2 and macrophage colony stimulating factor (M-CSF) and decreased secretion of pro-inflammatory mediators. Finally, treatment of pro-inflammatory M1-type-macrophages with Mdivi-1 metabolically reprogrammed them to an anti-inflammatory M2-like phenotype by inhibiting oxidative phosphorylation and attenuating the increase in succinate levels and correcting the decreased levels of arginine and citrulline. CONCLUSIONS We report that treatment with Mdivi-1 inhibits post-vascular injury neointimal hyperplasia by metabolic reprogramming macrophages towards an anti-inflammatory phenotype thereby highlighting the therapeutic potential of Mdivi-1 for preventing neointimal hyperplasia and restenosis following angioplasty and stenting in CAD and PAD patients.
Collapse
Affiliation(s)
- Gustavo E Crespo-Avilan
- Department of Biochemistry, Medical Faculty, Justus Liebig-University, Giessen, Germany; Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Sauri Hernandez-Resendiz
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Chrishan J Ramachandra
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Victor Ungureanu
- National Institute of Pathology, "Victor Babes", Bucharest, Romania
| | - Ying-Hsi Lin
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Shengjie Lu
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Jürgen Bernhagen
- Division of Vascular Biology, Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-University, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Munich Heart Alliance, Munich, Germany
| | - Omar El Bounkari
- Division of Vascular Biology, Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Klaus T Preissner
- Department of Biochemistry, Medical Faculty, Justus Liebig-University, Giessen, Germany; Kerckhoff-Heart-Research-Institute, Department of Cardiology, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Elisa A Liehn
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore; National Institute of Pathology, "Victor Babes", Bucharest, Romania; Institute for Molecular Medicine, University of South Denmark, Odense, Denmark.
| | - Derek J Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore; The Hatter Cardiovascular Institute, University College London, London, WC1E 6BT, UK; Yong Loo Lin School of Medicine, National University Singapore, Singapore.
| |
Collapse
|
2
|
Wei JH, Lee WJ, Luo JL, Huang HL, Wang SC, Chou RH, Huang PH, Lin SJ. Vertical Sleeve Gastrectomy Offers Protection against Disturbed Flow-Induced Atherosclerosis in High-Fat Diet-Fed Mice. Int J Mol Sci 2023; 24:ijms24065669. [PMID: 36982743 PMCID: PMC10051344 DOI: 10.3390/ijms24065669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
Bariatric surgery reduces body weight, enhances metabolic and diabetic control, and improves outcomes on obesity-related comorbidities. However, the mechanisms mediating this protection against cardiovascular diseases remain unclear. We investigated the effect of sleeve gastrectomy (SG) on vascular protection in response to shear stress-induced atherosclerosis using an overweighted and carotid artery ligation mouse model. Eight-week-old male wild-type mice (C57BL/6J) were fed a high-fat diet (HFD) for two weeks to induce weight gain and dysmetabolism. SG was performed in HFD-fed mice. Two weeks after the SG procedure, partial carotid-artery ligation was performed to promote disturbed flow-induced atherosclerosis. Compared with the control mice, HFD-fed wild-type mice exhibited increased body weight, total cholesterol level, hemoglobin A1c, and enhanced insulin resistance; SG significantly reversed these adverse effects. As expected, HFD-fed mice exhibited greater neointimal hyperplasia and atherosclerotic plaques than the control group, and the SG procedure attenuated HFD-promoted ligation-induced neointimal hyperplasia and arterial elastin fragmentation. Besides, HFD promoted ligation-induced macrophage infiltration, matrix metalloproteinase-9 expression, upregulation of inflammatory cytokines, and increased vascular endothelial growth factor secretion. SG significantly reduced the above-mentioned effects. Moreover, HFD restriction partially reversed the intimal hyperplasia caused by carotid artery ligation; however, this protective effect was significantly lower than that observed in SG-operated mice. Our study demonstrated that HFD deteriorates shear stress-induced atherosclerosis and SG mitigates vascular remodeling, and this protective effect was not comparable in HFD restriction group. These findings provide a rationale for using bariatric surgery to counter atherosclerosis in morbid obesity.
Collapse
Affiliation(s)
- Jih-Hua Wei
- Division of Cardiology, Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan 330, Taiwan; (J.-H.W.)
- School of Medicine, National Defense Medical Center, Taipei 114, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Wei-Jei Lee
- Department of Surgery, Min-Sheng General Hospital, Taoyuan 330, Taiwan
| | - Jing-Lin Luo
- Division of Cardiology, Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan 330, Taiwan; (J.-H.W.)
| | - Hsin-Lei Huang
- School of Nursing, National Taipei University of Nursing and Health Sciences, Taipei 112, Taiwan
| | - Shen-Chih Wang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Ruey-Hsing Chou
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Department of Anesthesiology, Taipei Veteran General Hospital, Taipei 112, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, No.201, Sec. 2, Shipai Rd., Beitou District, Taipei 112, Taiwan
| | - Po-Hsun Huang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Department of Anesthesiology, Taipei Veteran General Hospital, Taipei 112, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, No.201, Sec. 2, Shipai Rd., Beitou District, Taipei 112, Taiwan
- Correspondence: ; Tel.: +886-2-2875-7374; Fax: +886-2-2875-7375
| | - Shing-Jong Lin
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Anesthesiology, Taipei Veteran General Hospital, Taipei 112, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, No.201, Sec. 2, Shipai Rd., Beitou District, Taipei 112, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
3
|
Zhang Y, Fu Y, Zhang C, Jia L, Yao N, Lin Y, Dong Y, Fatima N, Alam N, Wang R, Wang W, Bai L, Zhao S, Liu E. MED1 Deficiency in Macrophages Accelerates Intimal Hyperplasia via ROS Generation and Inflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3010577. [PMID: 34853629 PMCID: PMC8629658 DOI: 10.1155/2021/3010577] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/17/2021] [Accepted: 11/07/2021] [Indexed: 11/17/2022]
Abstract
Mediator complex subunit 1 (MED1) is a component of the mediator complex and functions as a coactivator involved in the regulated transcription of nearly all RNA polymerase II-dependent genes. Previously, we showed that MED1 in macrophages has a protective effect on atherosclerosis; however, the effect of MED1 on intimal hyperplasia and mechanisms regulating proinflammatory cytokine production after macrophage MED1 deletion are still unknown. In this study, we report that MED1 macrophage-specific knockout (MED1 ΔMac) mice showed aggravated neointimal hyperplasia, vascular smooth muscle cells (VSMCs), and macrophage accumulation in injured arteries. Moreover, MED1 ΔMac mice showed increased proinflammatory cytokine production after an injury to the artery. After lipopolysaccharide (LPS) treatment, MED1 ΔMac macrophages showed increased generation of reactive oxygen species (ROS) and reduced expression of peroxisome proliferative activated receptor gamma coactivator-1α (PGC1α) and antioxidant enzymes, including catalase and glutathione reductase. The overexpression of PGC1α attenuated the effects of MED1 deficiency in macrophages. In vitro, conditioned media from MED1 ΔMac macrophages induced more proliferation and migration of VSMCs. To explore the potential mechanisms by which MED1 affects inflammation, macrophages were treated with BAY11-7082 before LPS treatment, and the results showed that MED1 ΔMac macrophages exhibited increased expression of phosphorylated-p65 and phosphorylated signal transducer and activator of transcription 1 (p-STAT1) compared with the control macrophages, suggesting the enhanced activation of NF-κB and STAT1. In summary, these data showed that MED1 deficiency enhanced inflammation and the proliferation and migration of VSMCs in injured vascular tissue, which may result from the activation of NF-κB and STAT1 due to the accumulation of ROS.
Collapse
Affiliation(s)
- Yali Zhang
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Yu Fu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Chenyang Zhang
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Linying Jia
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Nuo Yao
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Yuhao Lin
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Yue Dong
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Nazira Fatima
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Naqash Alam
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Rong Wang
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Weirong Wang
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Liang Bai
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Sihai Zhao
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| | - Enqi Liu
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, China
| |
Collapse
|
4
|
Zhang Y, Fatima M, Hou S, Bai L, Zhao S, Liu E. Research methods for animal models of atherosclerosis (Review). Mol Med Rep 2021; 24:871. [PMID: 34713295 PMCID: PMC8569513 DOI: 10.3892/mmr.2021.12511] [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: 02/25/2021] [Accepted: 05/20/2021] [Indexed: 11/17/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that threatens human health and lives by causing vascular stenosis and plaque rupture. Various animal models have been employed for elucidating the pathogenesis, drug development and treatment validation studies for atherosclerosis. To the best of our knowledge, the species used for atherosclerosis research include mice, rats, hamsters, rabbits, pigs, dogs, non-human primates and birds, among which the most commonly used ones are mice and rabbits. Notably, apolipoprotein E knockout (KO) or low-density lipoprotein receptor KO mice have been the most widely used animal models for atherosclerosis research since the late 20th century. Although the aforementioned animal models can form atherosclerotic lesions, they cannot completely simulate those in humans with respect to lesion location, lesion composition, lipoprotein composition and physiological structure. Hence, an appropriate animal model needs to be selected according to the research purpose. Additionally, it is necessary for atherosclerosis research to include quantitative analysis results of atherosclerotic lesion size and plaque composition. Laboratory animals can provide not only experimental tissues for in vivo studies but also cells needed for in vitro experiments. The present review first summarizes the common animal models and their practical applications, followed by focus on mouse and rabbit models and elucidating the methods to quantify atherosclerotic lesions. Finally, the methods of culturing endothelial cells, macrophages and smooth muscle cells were elucidated in detail and the experiments involved in atherosclerosis research were discussed.
Collapse
Affiliation(s)
- Yali Zhang
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Mahreen Fatima
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Siyuan Hou
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Liang Bai
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Sihai Zhao
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, P.R. China
| | - Enqi Liu
- Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Centre, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
5
|
Wang Q, Ozer HG, Wang B, Zhang M, Urabe G, Huang Y, Kent KC, Guo LW. A hierarchical and collaborative BRD4/CEBPD partnership governs vascular smooth muscle cell inflammation. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:54-66. [PMID: 33768129 PMCID: PMC7966960 DOI: 10.1016/j.omtm.2021.02.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/23/2021] [Indexed: 12/17/2022]
Abstract
Bromodomain protein BRD4 reads histone acetylation (H3K27ac), an epigenomic mark of transcription enhancers. CCAAT enhancer binding protein delta (CEBPD) is a transcription factor typically studied in metabolism. While both are potent effectors and potential therapeutic targets, their relationship was previously unknown. Here we investigated their interplay in vascular smooth muscle cell (SMC) inflammation. Chromatin immunoprecipitation followed by high-throughput sequencing (ChIP-seq) revealed H3K27ac/BRD4 enrichment at Cebpd in injured rat carotid arteries. While genomic deletion of BRD4-associated enhancer in SMCs in vitro decreased Cebpd transcripts, BRD4 gene silencing also diminished Cebpd mRNA and protein, indicative of a BRD4 control over CEBPD expression. Bromodomain-1, but not bromodomain-2, accounted for this BRD4 function. Moreover, endogenous BRD4 protein co-immunoprecipitated with CEBPD, and both proteins co-immunoprecipitated the Cebpd promoter and enhancer DNA fragments. These co-immunoprecipitations (coIPs) were all abolished by the BRD4-bromodomain blocker JQ1, suggesting a BRD4/CEBPD /promoter/enhancer complex. While BRD4 and CEBPD were both upregulated upon tumor necrosis factor alpha (TNF-α) stimulation of SMC inflammation (increased interleukin [IL]-1b, IL-6, and MCP-1), they mediated this stimulation via preferentially elevated expression of platelet-derived growth factor receptor alpha (PDGFRα, versus PDGFRβ), as indicated by loss- and gain-of-function experiments. Taken together, our study unravels a hierarchical yet collaborative BRD4/CEBPD relationship, a previously unrecognized mechanism that prompts SMC inflammation and may underlie other pathophysiological processes as well.
Collapse
Affiliation(s)
- Qingwei Wang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Hatice Gulcin Ozer
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Bowen Wang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Mengxue Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Go Urabe
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Yitao Huang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - K Craig Kent
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Lian-Wang Guo
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
6
|
Satish M, Gunasekar P, Agrawal DK. Pro-inflammatory and pro-resolving mechanisms in the immunopathology of arteriovenous fistula maturation. Expert Rev Cardiovasc Ther 2019; 17:369-376. [PMID: 31056981 DOI: 10.1080/14779072.2019.1612745] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction: With high rates of arteriovenous fistula (AVF) failure, there is a continued need to predict other factors and mechanisms associated with maturation deficits. Given the central association of inflammation with AVF failure, with neointimal hyperplasia (NIH) as one such mechanism, inflammation must be considered in two endogenous ways, either pro-inflammatory or pro-resolving, resulting in inward or outward vascular remodeling. Areas covered: This review summarizes and critically evaluates the preclinical and interventional data underlying AVF failure in attempts to elucidate the necessary balance between inflammation and its resolution. Expert opinion: Understanding the pro-inflammatory and pro-resolving mechanisms underlying inward and outward vascular remodeling and NIH prevention with AVF maturation is a necessary effort to develop key diagnostic and therapeutic interventions towards the ongoing issue of long-term AVF patency. The ability for clinical application has progressed but is limited to the identification of key targets and pathways with little understanding of how they are related synergistically or antagonistically. Likewise, the balance between acute inflammation and pro-resolution requires pertinent temporal considerations necessary for timely therapeutic application and predictive measurement.
Collapse
Affiliation(s)
- Mohan Satish
- a Department of Clinical and Translational Science , Creighton University School of Medicine , Omaha , NE , USA
| | - Palanikumar Gunasekar
- a Department of Clinical and Translational Science , Creighton University School of Medicine , Omaha , NE , USA
| | - Devendra K Agrawal
- a Department of Clinical and Translational Science , Creighton University School of Medicine , Omaha , NE , USA
| |
Collapse
|
7
|
Chen CF, Lin CC. The association of genotype polymorphisms with vascular access patency in hemodialysis patients. J Vasc Access 2018; 20:24-30. [PMID: 29512407 DOI: 10.1177/1129729818758556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
Some hemodialysis patients suffer from repeat dysfunction of dialysis vascular access and need procedures of angioplasty, thrombectomy, and even temporary catheter use. Why these patients are vulnerable to vascular access dysfunction and how to improve its patency are imperative to be discovered. Traditional risk factors for vascular access function had been widely investigated but could not fully explain this question. Several genotype polymorphisms were demonstrated to increase the incidence of cardiovascular disease and might also be linked to higher risk of vascular access dysfunction. As the major causes of arteriovenous access thrombosis are hypercoagulable status and arteriovenous access stenosis, the investigated genes mainly focus on the mediators of the coagulation cascade, inflammatory process, and endothelial dysfunction. The reported polymorphisms of genes significantly associated with arteriovenous access dysfunction included genes encoding methylene tetrahydrofolate reductase, coagulation factors, heme oxygenase-1, matrix metalloproteinase, transforming growth factor-β1, tumor necrosis factor-α, vascular endothelial growth factor-A, renin-angiotensin-aldosterone system, and protein methyl transferase. However, further prospective study is indispensable to elucidate the association between the genotype polymorphisms and the outcome of vascular access. More and more therapeutic options that focus on genotype polymorphisms may generate a great benefit to the patency of vascular access of uremic patients.
Collapse
Affiliation(s)
- Chun-Fan Chen
- 1 School of Medicine, National Yang-Ming University, Taipei, Taiwan.,2 National Yang-Ming University Hospital, Yilan, Taiwan
| | - Chih-Ching Lin
- 1 School of Medicine, National Yang-Ming University, Taipei, Taiwan.,3 Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
8
|
Getzin T, Krishnasamy K, Gamrekelashvili J, Kapanadze T, Limbourg A, Häger C, Napp LC, Bauersachs J, Haller H, Limbourg FP. The chemokine receptor CX 3CR1 coordinates monocyte recruitment and endothelial regeneration after arterial injury. EMBO Mol Med 2018; 10:151-159. [PMID: 29229785 PMCID: PMC5801509 DOI: 10.15252/emmm.201707502] [Citation(s) in RCA: 31] [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: 01/04/2017] [Revised: 11/15/2017] [Accepted: 11/17/2017] [Indexed: 12/12/2022] Open
Abstract
Regeneration of arterial endothelium after injury is critical for the maintenance of normal blood flow, cell trafficking, and vascular function. Using mouse models of carotid injury, we show that the transition from a static to a dynamic phase of endothelial regeneration is marked by a strong increase in endothelial proliferation, which is accompanied by induction of the chemokine CX3CL1 in endothelial cells near the wound edge, leading to progressive recruitment of Ly6Clo monocytes expressing high levels of the cognate CX3CR1 chemokine receptor. In Cx3cr1-deficient mice recruitment of Ly6Clo monocytes, endothelial proliferation and regeneration of the endothelial monolayer after carotid injury are impaired, which is rescued by acute transfer of normal Ly6Clo monocytes. Furthermore, human non-classical monocytes induce proliferation of endothelial cells in co-culture experiments in a VEGFA-dependent manner, and monocyte transfer following carotid injury promotes endothelial wound closure in a hybrid mouse model in vivo Thus, CX3CR1 coordinates recruitment of specific monocyte subsets to sites of endothelial regeneration, which promote endothelial proliferation and arterial regeneration.
Collapse
Affiliation(s)
- Tobias Getzin
- Vascular Medicine Research, Hannover Medical School, Hannover, Germany
| | - Kashyap Krishnasamy
- Vascular Medicine Research, Hannover Medical School, Hannover, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Jaba Gamrekelashvili
- Vascular Medicine Research, Hannover Medical School, Hannover, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Tamar Kapanadze
- Vascular Medicine Research, Hannover Medical School, Hannover, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Anne Limbourg
- Vascular Medicine Research, Hannover Medical School, Hannover, Germany
| | - Christine Häger
- Vascular Medicine Research, Hannover Medical School, Hannover, Germany
| | - L Christian Napp
- Vascular Medicine Research, Hannover Medical School, Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Hermann Haller
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Florian P Limbourg
- Vascular Medicine Research, Hannover Medical School, Hannover, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| |
Collapse
|
9
|
The epigenetic factor PCAF regulates vascular inflammation and is essential for intimal hyperplasia development. PLoS One 2017; 12:e0185820. [PMID: 29016683 PMCID: PMC5634597 DOI: 10.1371/journal.pone.0185820] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 09/20/2017] [Indexed: 12/20/2022] Open
Abstract
Objective Genetic P300/CBP-associated factor (PCAF) variation affects restenosis-risk in patients. PCAF has lysine acetyltransferase activity and promotes nuclear factor kappa-beta (NFκB)-mediated inflammation, which drives post-interventional intimal hyperplasia development. We studied the contributing role of PCAF in post-interventional intimal hyperplasia. Methods and results PCAF contribution to inflammation and intimal hyperplasia was assessed in leukocytes, macrophages and vascular smooth muscle cells (vSMCs) in vitro and in a mouse model for intimal hyperplasia, in which a cuff is placed around the femoral artery. PCAF deficiency downregulate CCL2, IL-6 and TNF-alpha expression, as demonstrated on cultured vSMCs, leukocytes and macrophages. PCAF KO mice showed a 71.8% reduction of vSMC-rich intimal hyperplasia, a 73.4% reduction of intima/media ratio and a 63.7% reduction of luminal stenosis after femoral artery cuff placement compared to wild type (WT) mice. The association of PCAF and vascular inflammation was further investigated using the potent natural PCAF inhibitor garcinol. Garcinol treatment reduced CCL2 and TNF-alpha expression, as demonstrated on cultured vSMCs and leukocytes. To assess the effect of garcinol treatment on vascular inflammation we used hypercholesterolemic ApoE*3-Leiden mice. After cuff placement, garcinol treatment resulted in reduced arterial leukocyte and macrophage adherence and infiltration after three days compared to untreated animals. Conclusions These results identify a vital role for the lysine acetyltransferase PCAF in the regulation of local inflammation after arterial injury and likely the subsequent vSMC proliferation, responsible for intimal hyperplasia.
Collapse
|
10
|
The Renin-Angiotensin-aldosterone system in vascular inflammation and remodeling. Int J Inflam 2014; 2014:689360. [PMID: 24804145 PMCID: PMC3997861 DOI: 10.1155/2014/689360] [Citation(s) in RCA: 228] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 02/28/2014] [Accepted: 03/03/2014] [Indexed: 12/31/2022] Open
Abstract
The RAAS through its physiological effectors plays a key role in promoting and maintaining inflammation. Inflammation is an important mechanism in the development and progression of CVD such as hypertension and atherosclerosis. In addition to its main role in regulating blood pressure and its role in hypertension, RAAS has proinflammatory and profibrotic effects at cellular and molecular levels. Blocking RAAS provides beneficial effects for the treatment of cardiovascular and renal diseases. Evidence shows that inhibition of RAAS positively influences vascular remodeling thus improving CVD outcomes. The beneficial vascular effects of RAAS inhibition are likely due to decreasing vascular inflammation, oxidative stress, endothelial dysfunction, and positive effects on regeneration of endothelial progenitor cells. Inflammatory factors such as ICAM-1, VCAM-1, TNFα, IL-6, and CRP have key roles in mediating vascular inflammation and blocking RAAS negatively modulates the levels of these inflammatory molecules. Some of these inflammatory markers are clinically associated with CVD events. More studies are required to establish long-term effects of RAAS inhibition on vascular inflammation, vascular cells regeneration, and CVD clinical outcomes. This review presents important information on RAAS's role on vascular inflammation, vascular cells responses to RAAS, and inhibition of RAAS signaling in the context of vascular inflammation, vascular remodeling, and vascular inflammation-associated CVD. Nevertheless, the review also equates the need to rethink and rediscover new RAAS inhibitors.
Collapse
|
11
|
Lei J, Vodovotz Y, Tzeng E, Billiar TR. Nitric oxide, a protective molecule in the cardiovascular system. Nitric Oxide 2013; 35:175-85. [DOI: 10.1016/j.niox.2013.09.004] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Revised: 09/02/2013] [Accepted: 09/24/2013] [Indexed: 12/19/2022]
|
12
|
Zhang JM, Wang Y, Miao YJ, Zhang Y, Wu YN, Jia LX, Qi YF, Du J. Knockout of CD8 delays reendothelialization and accelerates neointima formation in injured arteries of mouse via TNF-α inhibiting the endothelial cells migration. PLoS One 2013; 8:e62001. [PMID: 23658704 PMCID: PMC3642119 DOI: 10.1371/journal.pone.0062001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 03/17/2013] [Indexed: 01/26/2023] Open
Abstract
Objective Delayed or impaired reendothelialization is a major cause of stent thrombosis in the interventional treatment of coronary heart disease. T cells are involved in neointima formation of injured arteries. However, the regulated mechanism of reendothelialization and the role of CD8 T cell in reendothelialization are unclear. Methods and Results Immunofluorescence staining showed that CD8 positive cells were increased in wire injured femoral artery of mice. On day 21 after injury, elastin staining showed that knockout of CD8 (CD8−/−) significantly increased intimal thickness and a ratio of intima to media by 1.8 folds and 1.9 folds respectively in injured arteries. Evans blue staining showed that knockout of CD8 delayed the reendothelialization area on day 7 after injury (18.8±0.5% versus 42.1±5.6%, p<0.05). In vitro, a migration assay revealed that CD8−/− T cells co-cultured with WT macrophages significantly inhibited the migration of the endothelial cells (ECs); compared to CD4+ T cells, and CD8+ T cells could promote the ECs migration. Furthermore, real-time PCR analysis showed that knockout of CD8 increased the level of tumor necrosis factor α (TNF-α) in injured arteries and cytometric bead cytokine array showed that TNF-α was elevated in cultured CD8−/− T cells. Finally, a wound-healing assay showed that recombinant TNF-α significantly inhibited the migration of ECs. Conclusion Our study suggested that CD8+ T cells could promote the reendothelialization and inhibit the neointima formation after the artery wire injury, and this effect is at least partly dependent on decreasing TNF-α production promoting ECs migration.
Collapse
Affiliation(s)
- Jun-Meng Zhang
- Beijing An Zhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Ying Wang
- Beijing An Zhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Yan-Ju Miao
- Beijing An Zhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Yi Zhang
- Beijing An Zhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Yi-Na Wu
- Beijing An Zhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Li-Xin Jia
- Beijing An Zhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Yong-Fen Qi
- Beijing An Zhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Jie Du
- Beijing An Zhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
- * E-mail:
| |
Collapse
|
13
|
Divergent effects of 17-β-estradiol on human vascular smooth muscle and endothelial cell function diminishes TNF-α-induced neointima formation. Biochem Biophys Res Commun 2012; 420:828-33. [PMID: 22465119 DOI: 10.1016/j.bbrc.2012.03.082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 03/15/2012] [Indexed: 01/01/2023]
Abstract
Coronary heart disease (CHD) is a condition characterized by increased levels of proinflammatory cytokines, including tumor necrosis factor-α (TNF-α). TNF-α can induce vascular endothelial cell (EC) and smooth muscle cell (SMC) dysfunction, central events in development of neointimal lesions. The reduced incidence of CHD in young women is believed to be due to the protective effects of estradiol (E2). We therefore investigated the effects of TNF-α on human neointima formation and SMC/EC functions and any modulatory effects of E2. Saphenous vein (SV) segments were cultured in the presence of TNF-α (10 ng/ml), E2 (2.5 nM) or both in combination. Neointimal thickening was augmented by incubation with TNF-α, an effect that was abolished by co-culture with E2. TNF-α increased SV-SMC proliferation in a concentration-dependent manner that was optimal at 10 ng/ml (1.5-fold increase), and abolished by E2 at all concentrations studied (1-50 nM). Surprisingly, E2 itself at low concentrations (1 and 5 nM) stimulated SV-SMC proliferation to a level comparable to that of TNF-α alone. SV-EC migration was significantly impaired by TNF-α (42% of control), and co-culture with E2 partially restored the ability of SV-EC to migrate and repair the wound. In contrast, TNF-α increased SV-SMC migration by 1.7-fold, an effect that was completely reversed by co-incubation with E2. Finally, TNF-α potently induced ICAM-1 and VCAM-1 expression in both SV-EC and SV-SMC. However there was no modulation by E2 in either cell-type. In conclusion, TNF-α induced SV neointima formation, increased SMC proliferation and migration, impaired SV-EC migration and increased expression of adhesion molecules. E2 exerted distinct cell-type and function-specific modulation, the mechanisms underlying which are worthy of further detailed study.
Collapse
|
14
|
Chu X, Filali M, Stanic B, Takapoo M, Sheehan A, Bhalla R, Lamb FS, Miller FJ. A critical role for chloride channel-3 (CIC-3) in smooth muscle cell activation and neointima formation. Arterioscler Thromb Vasc Biol 2011; 31:345-51. [PMID: 21071705 PMCID: PMC3025755 DOI: 10.1161/atvbaha.110.217604] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 10/14/2010] [Indexed: 02/03/2023]
Abstract
OBJECTIVE We have shown that the chloride-proton antiporter chloride channel-3 (ClC-3) is required for endosome-dependent signaling by the Nox1 NADPH oxidase in SMCs. In this study, we tested the hypothesis that ClC-3 is necessary for proliferation of smooth muscle cells (SMCs) and contributes to neointimal hyperplasia following vascular injury. METHODS AND RESULTS Studies were performed in SMCs isolated from the aorta of ClC-3-null and littermate control (wild-type [WT]) mice. Thrombin and tumor necrosis factor-α (TNF-α) each caused activation of both mitogen activated protein kinase extracellular signal-regulated kinases 1 and 2 and the matrix-degrading enzyme matrix metalloproteinase-9 and cell proliferation of WT SMCs. Whereas responses to thrombin were preserved in ClC-3-null SMCs, the responses to TNF-α were markedly impaired. These defects normalized following gene transfer of ClC-3. Carotid injury increased vascular ClC-3 expression, and compared with WT mice, ClC-3-null mice exhibited a reduction in neointimal area of the carotid artery 28 days after injury. CONCLUSIONS ClC-3 is necessary for the activation of SMCs by TNF-α but not thrombin. Deficiency of ClC-3 markedly reduces neointimal hyperplasia following vascular injury. In view of our previous findings, this observation is consistent with a role for ClC-3 in endosomal Nox1-dependent signaling. These findings identify ClC-3 as a novel target for the prevention of inflammatory and proliferative vascular diseases.
Collapse
Affiliation(s)
- Xi Chu
- Department of Internal Medicine, The University of Iowa, Iowa City, IA 52242
- Department of Health Care, Shandong University and Shandong Provincial Hospital, Shandong, 250021, P.R. China
| | - Mohammed Filali
- Department of Pediatrics, The University of Iowa, Iowa City, IA 52242
| | - Bojana Stanic
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, IA 52242
| | | | - Andrea Sheehan
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, IA 52242
| | - Ramesh Bhalla
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, IA 52242
| | - Fred S. Lamb
- Department of Pediatrics, The University of Iowa, Iowa City, IA 52242
| | - Francis J. Miller
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, IA 52242
- Department of Internal Medicine, The University of Iowa, Iowa City, IA 52242
- Free Radical and Radiation Biology Program, The University of Iowa, Iowa City, IA 52242
- Veterans Affair Medical Center, Iowa City, IA 52242
| |
Collapse
|
15
|
Chen JS, Huang PH, Wang CH, Lin FY, Tsai HY, Wu TC, Lin SJ, Chen JW. Nrf-2 mediated heme oxygenase-1 expression, an antioxidant-independent mechanism, contributes to anti-atherogenesis and vascular protective effects of Ginkgo biloba extract. Atherosclerosis 2010; 214:301-9. [PMID: 21144518 DOI: 10.1016/j.atherosclerosis.2010.11.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2010] [Revised: 11/01/2010] [Accepted: 11/08/2010] [Indexed: 10/18/2022]
Abstract
AIMS Vascular protective effects of Ginkgo biloba extract (GBE) may involve both antioxidant-related and anti-inflammatory mechanisms. GBE was recently suggested as a heme oxygenase (HO)-1 inducer. The role of HO-1 in anti-atherogenesis and related vascular protective effects of GBE awaited further clarification. METHODS AND RESULTS Tumor necrosis factor (TNF)-α was used to stimulate adhesiveness of human aortic endothelial cells (HAECs) to monocytes, an in vitro sign simulating atherogenesis. Pretreatment with GBE reduced TNF-α-stimulated endothelial adhesiveness, which could be attenuated by HO-1 inhibitors ZnPP IX or SnPP IX. GBE increased HO-1 expression and enzyme activity in HAECs. Pretreatment with MAP kinase inhibitor SB203580 significantly reduced GBE-induced HO-1 expression. Furthermore, GBE activated the translocation of the transcription factor nuclear factor-erythroid 2-related factor 2 (Nrf2), and increased its binding to the antioxidant response element (ARE) of the HO-1 gene. Pretreatment with PEG-SOD or other antioxidant reagents did not alter GBE-induced endothelial HO-1 expression. In vivo study also showed that GBE treatment could reduce leukocyte adherence to injury arteries, and enhance HO-1 expression in circulating monocytes and in arteries after wire injury, suggesting the in vivo induction of HO-1 by GBE. CONCLUSION GBE could inhibit cytokine-induced endothelial adhesiveness by inducing HO-1 expression via the activation of p38 and Nrf-2 pathways, a mechanism in which oxidative stress is not directly involved. GBE might exert its anti-atherogenesis and vascular protective effects by inducing vascular HO-1 expression.
Collapse
Affiliation(s)
- Jia-Shiong Chen
- Institute of Pharmacology, National Yang-Ming University School of Medicine, Taipei, Taiwan, ROC
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Lambert CM, Roy M, Meloche J, Robitaille GA, Agharazii M, Richard DE, Bonnet S. Tumor necrosis factor inhibitors as novel therapeutic tools for vascular remodeling diseases. Am J Physiol Heart Circ Physiol 2010; 299:H995-1001. [PMID: 20709868 DOI: 10.1152/ajpheart.00562.2010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Vascular remodeling diseases (VRDs) are characterized by enhanced inflammation and proliferative and apoptosis-resistant vascular smooth muscle cells (VSMCs). The sustainability of this phenotype has been attributed in part to the activation of the transcription factor hypoxia-inducible factor-1 (HIF-1). There is evidence that circulating cytokines can act as HIF-1 activators in a variety of tissues, including VSMCs. Increased circulating tumor necrosis factor (TNF) levels have been associated with vascular diseases, but the mechanisms involved remain unknown. We hypothesized that increased circulating levels of TNF promotes VRDs by the activation of HIF-1, resulting in VSMC proliferation and resistance to apoptosis. Circulating TNF levels were significantly increased in patients with vascular diseases (n = 19) compared with healthy donors (n = 15). Using human carotid artery smooth muscle cells (CASMCs), we demonstrated that TNF (100 ng/ml) activates HIF-1 (HIF-1α expression), leading to increased CASMC proliferation (Ki-67 and PCNA staining) and resistance to mitochondrial-dependent apoptosis [tetramethylrhodamine methyl ester perchlorate (TMRM), terminal deoxynucleotide transferase-mediated dUTP nick end labeling (TUNEL), annexin-V staining]. In vivo, TNF inhibition using polyethylene glycol coupled with TNF membrane receptor 1 (PEGsTNFR1), a soluble TNF receptor inhibiting circulating TNF, prevented carotid artery postinjury media remodeling and neointima development in rats. This effect was associated with lowered HIF-1 activation and decreased CASMC proliferation. In conclusion, we demonstrate for the first time that the inhibition of the TNF/Akt/HIF-1 axis prevents vascular remodeling. TNF inhibitors may therefore represent new and interesting therapeutic tools against VRDs.
Collapse
Affiliation(s)
- Caroline M Lambert
- Centre de recherche du Centre Hospitalier Universitaire de Québec, L'Hôtel-Dieu de Québec, Faculté de Médecine, Université Laval, Québec, Canada
| | | | | | | | | | | | | |
Collapse
|
17
|
Takaoka M, Suzuki H, Shioda S, Sekikawa K, Saito Y, Nagai R, Sata M. Endovascular Injury Induces Rapid Phenotypic Changes in Perivascular Adipose Tissue. Arterioscler Thromb Vasc Biol 2010; 30:1576-82. [DOI: 10.1161/atvbaha.110.207175] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Accumulating evidence suggests that adipose tissue not only stores energy but also secretes various bioactive substances called adipocytokines. Periadventitial fat is distributed ubiquitously around arteries throughout the body. It was reported that inflammatory changes in the periadventitial fat may have a direct role in the pathogenesis of vascular diseases accelerated by obesity. We investigated the effect of endovascular injury on the phenotype of perivascular fat.
Methods and Results—
Endovascular injury significantly upregulated proinflammatory adipocytokines and downregulated adiponectin within periadventitial fat tissue in models of mouse femoral artery wire injury and rat iliac artery balloon injury. Genetic disruption of tumor necrosis factor (TNF)-α attenuated upregulation of proinflammatory adipocytokine expression, with reduced neointimal hyperplasia after vascular injury. Local delivery of TNF-α to the periadventitial area enhanced inflammatory adipocytokine expression, which was associated with augmented neointimal hyperplasia in TNF-α-deficient mice. Conditioned medium from a coculture of 3T3-L1 and RAW264 cells stimulated vascular smooth muscle cell proliferation. An anti-TNF-α neutralizing antibody in the coculture abrogated the stimulating effect of the conditioned medium.
Conclusion—
Our findings indicate that endovascular injury induces rapid and marked changes in perivascular adipose tissue, mainly mediated by TNF-α. It is suggested that the phenotypic changes in perivascular adipose tissue may have a role in the pathogenesis of neointimal hyperplasia after angioplasty.
Collapse
Affiliation(s)
- Minoru Takaoka
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (M.T., R.N.); Division of Cardiology, Department of Internal Medicine, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (H.S.); First Department of Anatomy, Showa University School of Medicine, Tokyo, Japan (S.S.); PrevenTec, Tsukuba, Japan (K.S.); First Department of Internal Medicine, Nara Medical University, Nara, Japan (Y.S.); Department of Cardiovascular Medicine,
| | - Hiroshi Suzuki
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (M.T., R.N.); Division of Cardiology, Department of Internal Medicine, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (H.S.); First Department of Anatomy, Showa University School of Medicine, Tokyo, Japan (S.S.); PrevenTec, Tsukuba, Japan (K.S.); First Department of Internal Medicine, Nara Medical University, Nara, Japan (Y.S.); Department of Cardiovascular Medicine,
| | - Seiji Shioda
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (M.T., R.N.); Division of Cardiology, Department of Internal Medicine, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (H.S.); First Department of Anatomy, Showa University School of Medicine, Tokyo, Japan (S.S.); PrevenTec, Tsukuba, Japan (K.S.); First Department of Internal Medicine, Nara Medical University, Nara, Japan (Y.S.); Department of Cardiovascular Medicine,
| | - Kenji Sekikawa
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (M.T., R.N.); Division of Cardiology, Department of Internal Medicine, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (H.S.); First Department of Anatomy, Showa University School of Medicine, Tokyo, Japan (S.S.); PrevenTec, Tsukuba, Japan (K.S.); First Department of Internal Medicine, Nara Medical University, Nara, Japan (Y.S.); Department of Cardiovascular Medicine,
| | - Yoshihiko Saito
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (M.T., R.N.); Division of Cardiology, Department of Internal Medicine, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (H.S.); First Department of Anatomy, Showa University School of Medicine, Tokyo, Japan (S.S.); PrevenTec, Tsukuba, Japan (K.S.); First Department of Internal Medicine, Nara Medical University, Nara, Japan (Y.S.); Department of Cardiovascular Medicine,
| | - Ryozo Nagai
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (M.T., R.N.); Division of Cardiology, Department of Internal Medicine, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (H.S.); First Department of Anatomy, Showa University School of Medicine, Tokyo, Japan (S.S.); PrevenTec, Tsukuba, Japan (K.S.); First Department of Internal Medicine, Nara Medical University, Nara, Japan (Y.S.); Department of Cardiovascular Medicine,
| | - Masataka Sata
- From the Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan (M.T., R.N.); Division of Cardiology, Department of Internal Medicine, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (H.S.); First Department of Anatomy, Showa University School of Medicine, Tokyo, Japan (S.S.); PrevenTec, Tsukuba, Japan (K.S.); First Department of Internal Medicine, Nara Medical University, Nara, Japan (Y.S.); Department of Cardiovascular Medicine,
| |
Collapse
|
18
|
Naringenin Inhibits Neointimal Hyperplasia Following Arterial Reconstruction With Interpositional Vein Graft. Ann Plast Surg 2010; 64:105-13. [DOI: 10.1097/sap.0b013e31819b03cd] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
19
|
Abarbanell AM, Herrmann JL, Weil BR, Wang Y, Tan J, Moberly SP, Fiege JW, Meldrum DR. Animal models of myocardial and vascular injury. J Surg Res 2009; 162:239-49. [PMID: 20053409 DOI: 10.1016/j.jss.2009.06.021] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2009] [Revised: 06/06/2009] [Accepted: 06/16/2009] [Indexed: 01/09/2023]
Abstract
Over the past century, numerous animal models have been developed in an attempt to understand myocardial and vascular injury. However, the successful translation of results observed in animals to human therapy remains low. To understand this problem, we present several animal models of cardiac and vascular injury that are of particular relevance to the cardiac or vascular surgeon. We also explore the potential clinical implications and limitations of each model with respect to the human disease state. Our results underscore the concept that animal research requires an in-depth understanding of the model, animal physiology, and the potential confounding factors. Future outcome analyses with standardized animal models may improve translation of animal research from the bench to the bedside.
Collapse
Affiliation(s)
- Aaron M Abarbanell
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Murayama H, Takahashi M, Takamoto M, Shiba Y, Ise H, Koyama J, Tagawa YI, Iwakura Y, Ikeda U. Deficiency of tumour necrosis factor-alpha and interferon-gamma in bone marrow cells synergistically inhibits neointimal formation following vascular injury. Cardiovasc Res 2008; 80:175-80. [PMID: 18791204 DOI: 10.1093/cvr/cvn250] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS Neointimal formation after percutaneous coronary intervention (PCI), termed restenosis, limits therapeutic revascularization. Since it is now known that vascular injury involves an inflammatory response, we examined the role of tumour necrosis factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma) in the neointimal formation after injury. METHODS AND RESULTS Control (BALB/c), TNF-alpha-deficient (Tnf(-/-)), IFN-gamma-deficient (Ifng(-/-)), or double-deficient (Tnf(-/-)Ifng(-/-)) mice were subjected to wire-mediated vascular injury of the right femoral artery. Neointimal formation after injury was significantly reduced after the injury in the Tnf(-/-)Ifng(-/-) mice, compared to that in the control, Tnf(-/-), and Ifng(-/-) mice. Immunohistochemical analysis showed that TNF-alpha and IFN-gamma were expressed in neointimal lesions in the control mice, but not in mice with deficiency of the corresponding cytokine. No significant difference in re-endothelialization was observed among these groups. The number of proliferating cell nuclear antigen in the neointimal lesions was significantly decreased in the Tnf(-/-)Ifng(-/-) mice. Bone marrow transplantation experiments revealed that deficiency of TNF-alpha and IFN-gamma specifically in bone marrow cells significantly inhibited neointimal formation after vascular injury. CONCLUSION The absence of TNF-alpha and IFN-gamma in bone marrow cells synergistically inhibits neointimal formation following vascular injury, and thus, may provide new insights into the mechanisms underlying restenosis after PCI.
Collapse
Affiliation(s)
- Hideki Murayama
- Department of Cardiovascular Medicine, Shinshu University Graduate School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Myocardin inhibits cellular proliferation by inhibiting NF-kappaB(p65)-dependent cell cycle progression. Proc Natl Acad Sci U S A 2008; 105:3362-7. [PMID: 18296632 DOI: 10.1073/pnas.0705842105] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We previously reported the importance of the serum response factor (SRF) cofactor myocardin in controlling muscle gene expression as well as the fundamental role for the inflammatory transcription factor NF-kappaB in governing cellular fate. Inactivation of myocardin has been implicated in malignant tumor growth. However, the underlying mechanism of myocardin regulation of cellular growth remains unclear. Here we show that NF-kappaB(p65) represses myocardin activation of cardiac and smooth muscle genes in a CArG-box-dependent manner. Consistent with their functional interaction, p65 directly interacts with myocardin and inhibits the formation of the myocardin/SRF/CArG ternary complex in vitro and in vivo. Conversely, myocardin decreases p65-mediated target gene activation by interfering with p65 DNA binding and abrogates LPS-induced TNF-alpha expression. Importantly, myocardin inhibits cellular proliferation by interfering with NF-kappaB-dependent cell-cycle regulation. Cumulatively, these findings identify a function for myocardin as an SRF-independent transcriptional repressor and cell-cycle regulator and provide a molecular mechanism by which interaction between NF-kappaB and myocardin plays a central role in modulating cellular proliferation and differentiation.
Collapse
|
22
|
Abstract
The A2b adenosine receptor (A2bAR) is highly abundant in bone marrow macrophages and vascular smooth muscle cells (VSMC). To examine the functional significance of this receptor expression, we applied a femoral artery injury model to A2bAR knockout (KO) mice and showed that the A2bAR prevents vascular lesion formation in an injury model that resembles human restenosis after angioplasty. While considering related mechanisms, we noted higher levels of TNF-alpha, an up-regulator of CXCR4, and of VSMC proliferation in the injured KO mice. In accordance, CXCR4, which is known to attract progenitor cells during tissue regeneration, is up-regulated in lesions of the KO mice. In addition, aortic smooth muscle cells derived from A2bAR KO mice display greater proliferation in comparison with controls. Bone marrow transplantation experiments indicated that the majority of the signal for lesion formation in the null mice originates from bone marrow cells. Thus, this study highlights the significance of the A2bAR in regulating CXCR4 expression in vivo and in protecting against vascular lesion formation.
Collapse
|
23
|
Ozaki CK, Jiang Z, Berceli SA. TNF-alpha and shear stress-induced large artery adaptations. J Surg Res 2007; 141:299-305. [PMID: 17574273 PMCID: PMC2032015 DOI: 10.1016/j.jss.2006.12.563] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2006] [Revised: 12/18/2006] [Accepted: 12/29/2006] [Indexed: 10/23/2022]
Abstract
BACKGROUND Tumor necrosis factor-alpha (TNF-alpha) up-regulation has been associated with both low and high shear-induced arterial remodeling. To address this apparent paradox and to define the biology of TNF-alpha signaling in large arteries, we tested the hypotheses that differential temporal expression of TNF-alpha drives shear-regulated arterial remodeling. MATERIALS AND METHODS Both low- and high-shear environments in the same rabbit were surgically created for common carotid arteries. Common carotid arteries (n = 60 total) were harvested after d0, d1, d3, d7, and d14 and analyses included morphology, TNF-alpha, and IL-10 mRNA quantitation. In separate experiments, animals received pegylated soluble TNF-alpha Type 1 receptor (PEG sTNF-RI) or vehicle via either short- or long-term dosing to define the effect of TNF-alpha blockade. RESULTS The model yielded a 14-fold shear differential (P < 0.001) with medial thickening under low shear (P = 0.025), and evidence of outward remodeling with high shear (P = 0.007). Low shear immediately up-regulated TNF-alpha expression approximately 50 fold (P < 0.001) at d1. Conversely, high shear-induced delayed and sustained TNF-alpha expression (22-fold at d7, P = 0.012; 23-fold at d14, P = 0.007). Both low and high shear gradually induced IL-10 expression (P = 0.002 and P = 0.004, respectively). Neither short-term (5-day) nor long-term (14-day) blockage of TNF-alpha signaling resulted in treatment-induced changes in the remodeling of low- or high-shear arteries. CONCLUSIONS Shear stress differentially and temporally regulates TNF-alpha expression in remodeling large arteries. However, TNF-alpha blockage did not substantially impact the final shear-induced morphology, suggesting that large arteries can remodel in response to flow perturbations independent of TNF-alpha signaling.
Collapse
Affiliation(s)
- C Keith Ozaki
- University of Florida College of Medicine and the Malcom Randall VAMC, Gainesville, Florida, USA.
| | | | | |
Collapse
|
24
|
Ozaki CK. Cytokines and the early vein graft: strategies to enhance durability. J Vasc Surg 2007; 45 Suppl A:A92-8. [PMID: 17544029 PMCID: PMC2031915 DOI: 10.1016/j.jvs.2007.02.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2007] [Accepted: 02/13/2007] [Indexed: 11/21/2022]
Abstract
This brief review focuses on experimental studies linking the proinflammatory cytokine tumor necrosis factor-alpha to accelerated vein graft failure in the broader historical context of vein graft research. From some perspectives, the field appears ripe for transfer of cytokine knowledge and therapeutic approaches that have evolved in other systems to vascular surgery problems. However, the complexity of vein graft disease suggests that more robust research approaches, such as broadening of the scope beyond focus on single mediators and neointimal hyperplasia, will be necessary to reach translatable strategies to prolong human vein graft durability.
Collapse
Affiliation(s)
- C Keith Ozaki
- University of Florida College of Medicine and the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL, USA.
| |
Collapse
|
25
|
Jiang Z, Shukla A, Miller BL, Espino DR, Tao M, Berceli SA, Ozaki CK. Tumor necrosis factor-alpha and the early vein graft. J Vasc Surg 2007; 45:169-76. [PMID: 17210403 DOI: 10.1016/j.jvs.2006.08.049] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2006] [Accepted: 08/23/2006] [Indexed: 10/23/2022]
Abstract
BACKGROUND Tumor necrosis factor-alpha (TNF-alpha) has been implicated in the blood vessel wall response to hemodynamic forces. We hypothesized that TNF-alpha activity drives neointimal hyperplasia (NIH) during vein graft arterialization and that anti-TNF-alpha therapy would inhibit NIH. METHODS Rabbits underwent bilateral vein grafting using jugular vein. All distal branches except the occipital artery were unilaterally ligated to create distinct flow environments between the bilateral grafts. Vein grafts were harvested sequentially up to 28 days for TNF-alpha messenger RNA (mRNA) quantitation. In separate experiments, animals received short-term or long-term dosing with pegylated soluble TNF-alpha type I receptor (PEG sTNF-RI) or vehicle. After 14 to 28 days, grafts were analyzed for morphometry, proliferation, apoptosis, and PEG sTNF-RI distribution. RESULTS Quantitative mRNA assay (TaqMan) revealed shear-dependent (P < .001) and time-dependent (P < .001) TNF-alpha expression. TNF-alpha induction was maximal at day 1 and gradually decreased over time, but was persistently elevated even 4 weeks later (P < .001). Low shear (associated with increased NIH) resulted in significantly higher TNF-alpha mRNA expression (P = .03). PEG sTNF-RI was found in high concentrations in the serum and localized to NIH. The high-flow and low-flow vein grafts from treated animals demonstrated similar volumes of NIH compared with controls. PEG-sTNF-RI had only modest impact on vascular wall cell turnover, as reflected by terminal deoxynucleotide transferase-mediated deoxy uridine triphosphate nick-end labeling (P = .064) and anti-Ki-67 (P = .12) assays. CONCLUSIONS Placement of a vein into the arterial circulation acutely upregulates TNF-alpha; this expression level correlates with the degree of subsequent NIH. Pharmacologic interruption of this signaling pathway has no significant impact on NIH or wall cellular proliferation/apoptosis, suggesting that early vein graft adaptations can proceed via TNF-alpha-independent mechanisms.
Collapse
Affiliation(s)
- Zhihua Jiang
- University of Florida College of Medicine, Gainesville, FL. USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Schepers A, Pires NMM, Eefting D, de Vries MR, van Bockel JH, Quax PHA. Short-term dexamethasone treatment inhibits vein graft thickening in hypercholesterolemic ApoE3Leiden transgenic mice. J Vasc Surg 2006; 43:809-15. [PMID: 16616241 DOI: 10.1016/j.jvs.2005.11.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2005] [Accepted: 11/12/2005] [Indexed: 11/21/2022]
Abstract
OBJECTIVE The aim of this study was to assess whether the anti-inflammatory agent dexamethasone can inhibit vein graft thickening without the occurrence of serious side effects. METHODS Venous interposition grafting was performed in the common carotid artery of hypercholesterolemic ApoE3Leiden transgenic mice. Mice were treated with dexamethasone (0.15 mg.kg(-1).d(-1) orally), and after 28 days, vein graft thickening was quantified. RESULTS Treatment with dexamethasone resulted in a significant 43% reduction in lesion area without changes in lesion composition when compared with nontreated controls. However, dexamethasone, when administered for a prolonged period of time, is known for its potentially serious side effects. To overcome these potential side effects of prolonged dexamethasone treatment, the effect of a short-term 7-day dexamethasone treatment was studied. This short dexamethasone treatment resulted in a 49% decrease of vein graft thickening at 28 days. Furthermore, it was demonstrated that dexamethasone treatment led to reduced local expression of several proinflammatory cytokines and factors in the vein grafts 24 hours after surgery. Finally, observations in mice were verified in human saphenous organ cultures. Exposure to dexamethasone for either 7 or 28 days significantly reduced intimal hyperplasia formation on cultured saphenous vein segments. CONCLUSIONS Short-term anti-inflammatory treatment with dexamethasone leads to a significant reduction in vein graft thickening over an extended period, possibly by the reduction of early expression of proinflammatory cytokines. This 7-day treatment minimizes the risk of unwanted side effects of long-term dexamethasone treatment and may be a new approach to prevent graft failure.
Collapse
Affiliation(s)
- Abbey Schepers
- Gaubius Laboratory, TNO Quality of Life, Leiden University Medical Center, The Netherlands
| | | | | | | | | | | |
Collapse
|
27
|
Rao JS, Langenbach R, Bosetti F. Down-regulation of brain nuclear factor-kappa B pathway in the cyclooxygenase-2 knockout mouse. ACTA ACUST UNITED AC 2005; 139:217-24. [PMID: 16055227 DOI: 10.1016/j.molbrainres.2005.05.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2005] [Revised: 04/18/2005] [Accepted: 05/12/2005] [Indexed: 01/04/2023]
Abstract
Cyclooxygenase (COX) is the rate-limiting enzyme in the synthesis of prostaglandins (PGs) from arachidonic acid. Evidence suggests that neuronal COX-2 gene expression is mainly regulated by the transcription factor nuclear factor kappa-B (NF-kappaB). The present study was undertaken to determine whether there is a shared regulation of NF-kappaB or of nuclear factor of activated T-cells cytoplasmic (NFATc) with COX-2 and whether these transcription factors known to regulate COX-2 expression are altered in brain from COX-2 knockout (COX-2-/-) mice compared to wild type. We found a decrease in NF-kappaB DNA-protein binding activity, which was accompanied by a reduction of the phosphorylation state of both I-kappaBalpha and p65 proteins in the COX-2-/- mice. The mRNA and protein levels of p65 were also reduced in COX-2-/- mice, whereas total cytoplasmic I-kappaB protein level was not significantly changed. Taken together, these changes may be responsible for the observed decrease in NF-kappaB DNA binding activity. NF-kappaB DNA binding activity was selectively affected in the COX-2-/- mice compared to the wild type as there was no significant change in NFATc DNA binding activity. Overall, our data indicate that constitutive brain NF-kappaB activity is decreased in COX-2 deficient mice and suggest a reciprocal coupling between NF-kappaB and COX-2.
Collapse
Affiliation(s)
- Jagadeesh S Rao
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, 9000 Rockville Pike, Building 9, Room 1S 128, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
28
|
Zuckerbraun BS, Barbato JE, Hamilton A, Sebti S, Tzeng E. Inhibition of geranylgeranyltransferase I decreases generation of vascular reactive oxygen species and increases vascular nitric oxide production. J Surg Res 2005; 124:256-63. [PMID: 15820256 DOI: 10.1016/j.jss.2004.10.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2004] [Indexed: 01/07/2023]
Abstract
BACKGROUND Vascular injury with endothelial dysfunction results in an imbalance between the production of vasoprotective molecules such as nitric oxide (NO) and deleterious reactive oxygen species (ROS). The purpose of this work was to test the hypothesis that inhibition of geranylgeranyltransferase I (GG Tase I) reduces vascular injury by increasing vascular NO production while decreasing ROS generation. METHODS AND RESULTS GGTI-298 decreased the formation of intimal hyperplasia at 14 days following balloon injury. GGTI-298 (10 microm) inhibited activation of RhoA and Rac1 as well as inhibited SMC proliferation. GGTI increased SMC-inducible NO synthase (iNOS) levels and NO production in vitro. Additionally, the activation of NAD(P)H oxidase subunits was decreased by GGTI in vitro. This correlated with a decrease in TNF-alpha- or angiotensin-II-induced ROS production assayed by DCF fluorescence. In vivo, GGTI treatment increased endothelial NOS (eNOS) expression in uninjured arteries and iNOS expression in balloon-injured arteries. Furthermore, GGTI treatment attenuated balloon-injury-induced superoxide generation assayed by MCLA luminescence. CONCLUSIONS GGTI decreases the production of ROS and increases the production of NO both in vitro and in vivo. These effects may be mediated via the inhibition of activation of the small GTPases Rac1 and RhoA. Pharmacological inhibition of GGTase I may prove to be a useful clinical adjunct in the treatment of cardiovascular diseases.
Collapse
MESH Headings
- Alkyl and Aryl Transferases/antagonists & inhibitors
- Alkyl and Aryl Transferases/metabolism
- Angioplasty, Balloon/adverse effects
- Animals
- Aorta, Thoracic/cytology
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/injuries
- Benzamides/pharmacology
- Cell Cycle Proteins/metabolism
- Cell Division
- Cells, Cultured
- Cyclin-Dependent Kinase Inhibitor p21
- Cyclin-Dependent Kinase Inhibitor p27
- Enzyme Inhibitors/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- NADPH Oxidases/metabolism
- Nitric Oxide/metabolism
- Nitric Oxide Synthase/metabolism
- Nitric Oxide Synthase Type II
- Phosphoproteins/metabolism
- Rats
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Tumor Suppressor Proteins/metabolism
- Tunica Intima/enzymology
- Tunica Intima/injuries
- rac1 GTP-Binding Protein/metabolism
- rhoA GTP-Binding Protein/metabolism
Collapse
Affiliation(s)
- Brian S Zuckerbraun
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | | | | | | | | |
Collapse
|
29
|
Zimmerman MA, Reznikov LL, Raeburn CD, Selzman CH. Interleukin-10 attenuates the response to vascular injury. J Surg Res 2004; 121:206-13. [PMID: 15501460 DOI: 10.1016/j.jss.2004.03.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2004] [Indexed: 11/23/2022]
Abstract
BACKGROUND The inflammatory response to vascular injury is characterized by expression of cytokines, growth factors, and chemokines that conspire to promote vessel remodeling and intimal hyperplasia (IH). Interleukin-10 (IL-10) is a multifunctional cytokine that has several anti-inflammatory properties in vitro. Few studies have evaluated the effects of IL-10 in experimental atherosclerosis. The purpose of the present study was to determine the influence of IL-10 on vascular inflammation and IH following mechanical injury. METHODS Wire carotid injury was performed in wild-type (WT) mice with and without IL-10 treatment. Immunohistochemistry, PCR, and ELISA assays were used to examine vessel production of basic fibroblast growth factor (bFGF), monocyte chemotactic protein-1 (MCP-1), and nuclear factor kappa B (NFkappaB). Vessels were morphometrically analyzed for IH. RESULTS Carotid injury induced early expression of MCP-1 and bFGF that was abrogated in mice treated with IL-10. Similarly, injury-induced expression of NFkappaB message and protein was attenuated in mice receiving exogenous IL-10. Compared to untreated mice, IL-10 markedly decreased levels of IH. Interestingly, carotid injury in IL-10-deficient mice resulted in an augmented IH response compared to injured WT mice. CONCLUSIONS In an in vivo model of direct vascular injury, IL-10 decreased expression of the pro-inflammatory transcription factor, NFkappaB, and the mitogenic chemokine and growth factor, MCP-1 and bFGF, respectively. These observations were associated with IL-10-induced attenuation of IH. Furthermore, endogenous IL-10 appeared to suppress the injury response. In conclusion, exogenously delivered IL-10 may represent a clinically relevant anti-inflammatory strategy for post-injury intimal hyperplasia.
Collapse
Affiliation(s)
- Michael A Zimmerman
- Department of Surgery, University of Colorado Health, Sciences Center, Denver, Colorado, USA
| | | | | | | |
Collapse
|
30
|
Zhang L, Peppel K, Brian L, Chien L, Freedman NJ. Vein graft neointimal hyperplasia is exacerbated by tumor necrosis factor receptor-1 signaling in graft-intrinsic cells. Arterioscler Thromb Vasc Biol 2004; 24:2277-83. [PMID: 15486311 DOI: 10.1161/01.atv.0000147766.68987.0d] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Vein graft remodeling and neointimal hyperplasia involve inflammation, graft-intrinsic cells, and recruitment of vascular progenitor cells. We sought to examine if the inflammatory cytokine tumor necrosis factor (TNF) affects vein graft remodeling via its p55 TNF receptor-1 (p55). METHODS AND RESULTS Inferior vena cava-to-carotid artery interposition grafting was performed between p55-/- and congenic (C57Bl/6) wild-type (WT) mice. Immunofluorescence revealed TNF in early (2-week) vein grafts. Six weeks postoperatively, luminal and medial areas were indistinguishable among all vein graft groups. However, neointimal area was reduced in p55-/- grafts: by 40% in p55-/- grafts placed in p55-/- recipients, and by 21% in p55-/- grafts placed in WT recipients, compared with WT grafts in WT recipients (P<0.05). In 2-week-old vein grafts, p55 deficiency reduced intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and monocyte chemoattractant protein-1 expression by 50% to 60%, and increased the extent of graft endothelialization. In vitro, TNF promoted chemokine expression and [3H]thymidine incorporation in vascular smooth muscle cells (SMCs) from WT, but not from p55-/- mice. However, responses of WT and p55-/- SMCs to other growth factors were equivalent. CONCLUSIONS Signaling via p55, in vein graft-intrinsic cells, contributes to the pathogenesis of vein graft neointimal hyperplasia.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Carotid Artery, Common/surgery
- Cell Adhesion Molecules/biosynthesis
- Cell Line
- Chemokine CCL2/biosynthesis
- Chemokines/biosynthesis
- Enzyme Activation/physiology
- Hyperplasia/enzymology
- Hyperplasia/genetics
- Hyperplasia/pathology
- Male
- Mice
- Mice, Congenic
- Mice, Inbred C57BL
- Neovascularization, Pathologic/enzymology
- Neovascularization, Pathologic/metabolism
- Receptors, Tumor Necrosis Factor, Type I/deficiency
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Receptors, Tumor Necrosis Factor, Type I/physiology
- Signal Transduction/physiology
- Tumor Necrosis Factor-alpha/biosynthesis
- Tunica Intima/chemistry
- Tunica Intima/enzymology
- Tunica Intima/metabolism
- Tunica Intima/pathology
- Veins/enzymology
- Veins/metabolism
- Veins/transplantation
- Vena Cava, Inferior/cytology
- Vena Cava, Inferior/transplantation
Collapse
Affiliation(s)
- Lisheng Zhang
- Duke University Department of Medicine (Cardiology), Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
31
|
Blessing E, Bea F, Kuo CC, Campbell LA, Chesebro B, Rosenfeld ME. Lesion progression and plaque composition are not altered in older apoE−/− mice lacking tumor necrosis factor-α receptor p55. Atherosclerosis 2004; 176:227-32. [PMID: 15380444 DOI: 10.1016/j.atherosclerosis.2004.05.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2003] [Revised: 05/19/2004] [Accepted: 05/25/2004] [Indexed: 11/25/2022]
Abstract
BACKGROUND Inflammatory processes are an integral component of the initiation, progression, and destabilization of atherosclerotic lesions. Tumor necrosis factor-alpha (TNF-alpha) is considered a primary mediator of inflammatory processes. METHODS AND RESULTS The role of TNF-alpha in plaque progression and plaque destabilization was investigated in the innominate arteries of older TNF-alpha receptor p55 deficient mice that were generated on a hyperlipidemic apolipoprotein E deficient background (p55-/- apoE-/-). There were no significant differences in levels of circulating cytokines, plaque progression, plaque composition or features of plaque destabilization in p55-/- apoE-/- compared to wild type (p55+/+ apoE-/-) mice. CONCLUSIONS Progression and destabilization of advanced atherosclerotic lesions does not seem to be mediated via the TNF-alpha receptor p55.
Collapse
Affiliation(s)
- Erwin Blessing
- Department of Pathobiology, University of Washington, P.O. Box 353410, Seattle, WA 98195, USA
| | | | | | | | | | | |
Collapse
|
32
|
Xu Q. Mouse models of arteriosclerosis: from arterial injuries to vascular grafts. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 165:1-10. [PMID: 15215157 PMCID: PMC2216680 DOI: 10.1016/s0002-9440(10)63270-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Animal models are designed to be preliminary tools for better understanding of the pathogenesis, improvement in diagnosis, prevention, and therapy of arteriosclerosis in humans. Attracted by the well-defined genetic systems, a number of investigators have begun to use the mouse as an experimental system for arteriosclerosis research. Hundreds of inbred lines have been established, and the genetic map is relatively well defined, and both congenic strains and recombinant strains are available to facilitate genetic experimentation. Because arteriosclerosis is a complicated disease, which includes spontaneous (native) atherosclerosis, transplant arteriosclerosis, vein graft atherosclerosis, and angioplasty-induced restenosis, several mouse models for studying all types of arteriosclerosis have recently been established. Using these mouse models, much knowledge concerning the pathogenesis of the disease and therapeutic intervention has been gained, eg, origins of endothelial and smooth muscle cells in lesions of transplant and vein graft atherosclerosis. This review will not attempt to cover all aspects of mouse models, rather focus on models of arterial injuries, vein grafts, and transplant arteriosclerosis, by which the major progress in understanding the mechanisms of the disease has been made. This article will also point out (dis)advantages of a variety of models, and how the models can be appropriately chosen for different purposes of study.
Collapse
Affiliation(s)
- Qingbo Xu
- Department of Cardiac and Vascular Sciences, St. George's Hospital Medical School, Cranmer Terrace, London SW17 0RE, United Kingdom.
| |
Collapse
|
33
|
Zimmerman MA, Reznikov LL, Sorensen AC, Selzman CH. Relative contribution of the TNF-alpha receptors to murine intimal hyperplasia. Am J Physiol Regul Integr Comp Physiol 2003; 284:R1213-8. [PMID: 12531783 DOI: 10.1152/ajpregu.00434.2002] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tumor necrosis factor-alpha (TNF-alpha) is an important mediator in the inflammatory response to vascular injury. The present study sought to determine the relative contribution of each TNF-alpha receptor subtype (p55 and p75) to intimal hyperplasia (IH) and characterize the mechanisms of transcriptional regulation after vascular injury. A murine model of wire carotid arterial injury was employed to induce IH in wild-type (WT), p55-deficient (p55-/-), and p75-deficient (p75-/-) mice. Compared with injured WT and p75-/- animals, p55-/- mice demonstrated a twofold reduction in IH. Additionally, p55-/- mice demonstrated a decrease in expression of nuclear factor-kappaB mRNA and protein. These observations suggest an important role for the p55 receptor in IH after mechanical endoluminal injury. Suppression of the transcriptional activator nuclear factor-kappaB may provide a mechanism by which p55-mediated IH is attenuated.
Collapse
Affiliation(s)
- Michael A Zimmerman
- Divisions of Cardiothoracic Surgery and Infectious Disease, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA
| | | | | | | |
Collapse
|
34
|
Affiliation(s)
- Heimo Ehmke
- Institut für Vegetative Physiologie und Pathophysiologie, Universität Hamburg, D-20246 Hamburg, Germany.
| |
Collapse
|