201
|
Tsujikawa LM, Fu L, Das S, Halliday C, Rakai BD, Stotz SC, Sarsons CD, Gilham D, Daze E, Wasiak S, Studer D, Rinker KD, Sweeney M, Johansson JO, Wong NCW, Kulikowski E. Apabetalone (RVX-208) reduces vascular inflammation in vitro and in CVD patients by a BET-dependent epigenetic mechanism. Clin Epigenetics 2019; 11:102. [PMID: 31300040 PMCID: PMC6626370 DOI: 10.1186/s13148-019-0696-z] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 06/11/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Apabetalone (RVX-208) is a bromodomain and extraterminal protein inhibitor (BETi) that in phase II trials reduced the relative risk (RR) of major adverse cardiac events (MACE) in patients with cardiovascular disease (CVD) by 44% and in diabetic CVD patients by 57% on top of statins. A phase III trial, BETonMACE, is currently assessing apabetalone's ability to reduce MACE in statin-treated post-acute coronary syndrome type 2 diabetic CVD patients with low high-density lipoprotein C. The leading cause of MACE is atherosclerosis, driven by dysfunctional lipid metabolism and chronic vascular inflammation (VI). In vitro studies have implicated the BET protein BRD4 as an epigenetic driver of inflammation and atherogenesis, suggesting that BETi may be clinically effective in combating VI. Here, we assessed apabetalone's ability to regulate inflammation-driven gene expression and cell adhesion in vitro and investigated the mechanism by which apabetalone suppresses expression. The clinical impact of apabetalone on mediators of VI was assessed with proteomic analysis of phase II CVD patient plasma. RESULTS In vitro, apabetalone prevented inflammatory (TNFα, LPS, or IL-1β) induction of key factors that drive endothelial activation, monocyte recruitment, adhesion, and plaque destabilization. BRD4 abundance on inflammatory and adhesion gene promoters and enhancers was reduced by apabetalone. BRD2-4 degradation by MZ-1 also prevented TNFα-induced transcription of monocyte and endothelial cell adhesion molecules and inflammatory mediators, confirming BET-dependent regulation. Transcriptional regulation by apabetalone translated into a reduction in monocyte adhesion to an endothelial monolayer. In a phase II trial, apabetalone treatment reduced the abundance of multiple VI mediators in the plasma of CVD patients (SOMAscan® 1.3 k). These proteins correlate with CVD risk and include adhesion molecules, cytokines, and metalloproteinases. Ingenuity® Pathway Analysis (IPA®) predicted that apabetalone inhibits pro-atherogenic regulators and pathways and prevents disease states arising from leukocyte recruitment. CONCLUSIONS Apabetalone suppressed gene expression of VI mediators in monocytes and endothelial cells by inhibiting BET-dependent transcription induced by multiple inflammatory stimuli. In CVD patients, apabetalone treatment reduced circulating levels of VI mediators, an outcome conducive with atherosclerotic plaque stabilization and MACE reduction. Inhibition of inflammatory and adhesion molecule gene expression by apabetalone is predicted to contribute to MACE reduction in the phase III BETonMACE trial.
Collapse
Affiliation(s)
- Laura M Tsujikawa
- Resverlogix Corp., 300, 4820 Richard Road SW, Calgary, AB, T3E 6 L1, Canada
| | - Li Fu
- Resverlogix Corp., 300, 4820 Richard Road SW, Calgary, AB, T3E 6 L1, Canada
| | - Shovon Das
- Resverlogix Corp., 300, 4820 Richard Road SW, Calgary, AB, T3E 6 L1, Canada
| | | | - Brooke D Rakai
- Resverlogix Corp., 300, 4820 Richard Road SW, Calgary, AB, T3E 6 L1, Canada
| | - Stephanie C Stotz
- Resverlogix Corp., 300, 4820 Richard Road SW, Calgary, AB, T3E 6 L1, Canada
| | | | - Dean Gilham
- Resverlogix Corp., 300, 4820 Richard Road SW, Calgary, AB, T3E 6 L1, Canada
| | - Emily Daze
- Resverlogix Corp., 300, 4820 Richard Road SW, Calgary, AB, T3E 6 L1, Canada
| | - Sylwia Wasiak
- Resverlogix Corp., 300, 4820 Richard Road SW, Calgary, AB, T3E 6 L1, Canada
| | - Deborah Studer
- Cellular and Molecular Bioengineering Research Lab, Libin Cardiovascular Institute of Alberta, University of Calgary, HMRB 358/361 3330 University Drive NW, Calgary, AB, T2N 4 N1, Canada
| | - Kristina D Rinker
- Cellular and Molecular Bioengineering Research Lab, Libin Cardiovascular Institute of Alberta, University of Calgary, HMRB 358/361 3330 University Drive NW, Calgary, AB, T2N 4 N1, Canada
| | - Michael Sweeney
- Resverlogix Inc., Suite 4010, 44 Montgomery Street, San Francisco, CA, 94104, USA
| | - Jan O Johansson
- Resverlogix Inc., Suite 4010, 44 Montgomery Street, San Francisco, CA, 94104, USA
| | - Norman C W Wong
- Resverlogix Corp., 300, 4820 Richard Road SW, Calgary, AB, T3E 6 L1, Canada
| | - Ewelina Kulikowski
- Resverlogix Corp., 300, 4820 Richard Road SW, Calgary, AB, T3E 6 L1, Canada.
| |
Collapse
|
202
|
Song HS, Kwon JE, Baek HJ, Kim CW, Jeon H, Ra JS, Lee HK, Kang SC. Sorghum Fermented by Aspergillus oryzae NK Enhances Inhibition of Vascular Inflammation in TNF-α-stimulated Human Aortic Smooth Muscle Cells. INT J VITAM NUTR RES 2019; 88:309-318. [PMID: 31237194 DOI: 10.1024/0300-9831/a000496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Sorghum bicolor L. Moench is widely grown all over the world for food and feed. The effects of sorghum extracts on general inflammation have been previously studied, but its anti-vascular inflammatory effects are unknown. Therefore, this study investigated the anti-vascular inflammation effects of sorghum extract (SBE) and fermented extract of sorghum (fSBE) on human aortic smooth muscle cells (HASMCs). After the cytotoxicity test of the sorghum extract, a series of experiments were conducted. The inhibition effects of SBE and fSBE on the inflammatory response and adhesion molecule expression were measured using treatment with tumor necrosis factor-α (TNF-α), a crucial promoter for the development of atherosclerotic lesions, on HASMCs. After TNF-α (10 ng/mL) treatment for 2 h, then SBE and fSBE (100 and 200 μg/mL) were applied for 12h. Western blotting analysis showed that the expression of vascular cell adhesion molecule-1 (VCAM-1) (2.4-fold) and cyclooxygenase-2 (COX-2) (6.7-fold) decreased, and heme oxygenase-1 (HO-1) (3.5-fold) increased compared to the TNF-α control when treated with 200 μg/mL fSBE (P<0.05). In addition, the fSBE significantly increased the expression of HO-1 and significantly decreased the expression of VCAM-1 and COX-2 compared to the TNF-α control in mRNA level (P<0.05). These reasons of results might be due to the increased concentrations of procyanidin B1 (about 6-fold) and C1 (about 30-fold) produced through fermentation with Aspergillus oryzae NK for 48 h, at 37 °C. Overall, the results demonstrated that fSBE enhanced the inhibition of the inflammatory response and adherent molecule expression in HASMCs.
Collapse
Affiliation(s)
- Hae Seong Song
- 1 Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Kyunggi-do, 17104, Korea.,a Authors contributed equally to the paper
| | - Jung-Eun Kwon
- 1 Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Kyunggi-do, 17104, Korea
| | - Hyun Jin Baek
- 1 Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Kyunggi-do, 17104, Korea
| | - Chang Won Kim
- 1 Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Kyunggi-do, 17104, Korea
| | - Hyelin Jeon
- 1 Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Kyunggi-do, 17104, Korea.,a Authors contributed equally to the paper
| | - Jong Sung Ra
- 1 Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Kyunggi-do, 17104, Korea.,a Authors contributed equally to the paper
| | - Hyo Kyu Lee
- 1 Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Kyunggi-do, 17104, Korea
| | - Se Chan Kang
- 1 Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Kyunggi-do, 17104, Korea.,b Corresponding author at: Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Korea, Tel) +82-31-201-2687 Fax) +82-31-204-8116, (Se Chan Kang)
| |
Collapse
|
203
|
Rajamani A, Borkowski K, Akre S, Fernandez A, Newman JW, Simon SI, Passerini AG. Oxylipins in triglyceride-rich lipoproteins of dyslipidemic subjects promote endothelial inflammation following a high fat meal. Sci Rep 2019; 9:8655. [PMID: 31209255 PMCID: PMC6572825 DOI: 10.1038/s41598-019-45005-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 05/28/2019] [Indexed: 11/26/2022] Open
Abstract
Elevated triglyceride-rich lipoproteins (TGRL) in circulation is a risk factor for atherosclerosis. TGRL from subjects consuming a high saturated fat test meal elicited a variable inflammatory response in TNFα-stimulated endothelial cells (EC) that correlated strongly with the polyunsaturated fatty acid (PUFA) content. This study investigates how the relative abundance of oxygenated metabolites of PUFA, oxylipins, is altered in TGRL postprandially, and how these changes promote endothelial inflammation. Human aortic EC were stimulated with TNFα and treated with TGRL, isolated from subjects’ plasma at fasting and 3.5 hrs postprandial to a test meal high in saturated fat. Endothelial VCAM-1 surface expression stimulated by TNFα provided a readout for atherogenic inflammation. Concentrations of esterified and non-esterified fatty acids and oxylipins in TGRL were quantified by mass spectrometry. Dyslipidemic subjects produced TGRL that increased endothelial VCAM-1 expression by ≥35%, and exhibited impaired fasting lipogenesis activity and a shift in soluble epoxide hydrolase and lipoxygenase activity. Pro-atherogenic TGRL were enriched in eicosapentaenoic acid metabolites and depleted in esterified C18-PUFA-derived diols. Abundance of these metabolites was strongly predictive of VCAM-1 expression. We conclude the altered metabolism in dyslipidemic subjects produces TGRL with a unique oxylipin signature that promotes a pro-atherogenic endothelial phenotype.
Collapse
Affiliation(s)
- Anita Rajamani
- Department of Biomedical Engineering, University of California, Davis, 451 Health Sciences Dr., Davis, CA, 95616, USA
| | - Kamil Borkowski
- West Coast Metabolomics Center, Genome Center, University of California, Davis, 451 Health Sciences Dr., Davis, CA, 95616, USA
| | - Samir Akre
- Department of Biomedical Engineering, University of California, Davis, 451 Health Sciences Dr., Davis, CA, 95616, USA
| | - Andrea Fernandez
- Department of Biomedical Engineering, University of California, Davis, 451 Health Sciences Dr., Davis, CA, 95616, USA
| | - John W Newman
- West Coast Metabolomics Center, Genome Center, University of California, Davis, 451 Health Sciences Dr., Davis, CA, 95616, USA.,Department of Nutrition, University of California, Davis, 3135 Meyer Hall, One Shields Avenue, Davis, CA, 95616, USA.,Western Human Nutrition Research Center, Obesity and Metabolism Research Unit, Agricultural Research Service, United States Department of Agriculture, 430 West Health Sciences Dr., Davis, CA, 95616, USA
| | - Scott I Simon
- Department of Biomedical Engineering, University of California, Davis, 451 Health Sciences Dr., Davis, CA, 95616, USA
| | - Anthony G Passerini
- Department of Biomedical Engineering, University of California, Davis, 451 Health Sciences Dr., Davis, CA, 95616, USA.
| |
Collapse
|
204
|
Woodside DG. Nanoparticle Imaging of Vascular Inflammation and Remodeling in Atherosclerotic Disease. CURRENT CARDIOVASCULAR IMAGING REPORTS 2019. [DOI: 10.1007/s12410-019-9501-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
205
|
Brauner S, Jiang X, Thorlacius GE, Lundberg AM, Östberg T, Yan ZQ, Kuchroo VK, Hansson GK, Wahren-Herlenius M. Augmented Th17 differentiation in Trim21 deficiency promotes a stable phenotype of atherosclerotic plaques with high collagen content. Cardiovasc Res 2019; 114:158-167. [PMID: 29016728 DOI: 10.1093/cvr/cvx181] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 09/01/2017] [Indexed: 12/22/2022] Open
Abstract
Aims Patients with hyperlipidemia are at risk of atherosclerosis, but not all develop cardiovascular disease, highlighting the importance of other risk factors such as inflammation. Both the innate and adaptive arms of the immune system have been suggested in the initiation and propagation of plaque formation. Tri-partite motif (TRIM) 21 is a regulator of tissue inflammation and pro-inflammatory cytokine production, and has been implicated in chronic inflammatory disease. Here, we investigate a potential role for TRIM21 in coronary artery disease. Methods and results Trim21-deficient or wild-type bone marrow was transplanted into Ldlr-/- mice fed a hypercholesterolemic diet. The Trim21-/-->Ldlr-/- mice developed larger atherosclerotic plaques, with significantly higher collagen content compared to mice transplanted with wild-type cells. High collagen content of the atheroma is stabilizing, and has recently been linked to IL-17. Interestingly, Trim21-/-->Ldlr-/- mice had elevated CD4 and IL-17 mRNA expression in plaques, and increased numbers of activated CD4+ T cells in the periphery. An increased differentiation of naïve T cells lacking Trim21 into Th17 cells was confirmed in vitro, with transcriptomic analysis revealing upregulation of genes of a non-pathogenic Th17 phenotype. Also, decreased expression of matrix metalloproteinases (MMPs) was noted in aortic plaques. Analysis of human carotid plaques confirmed that TRIM21 expression negatively correlates with the expression of key Th17 genes and collagen, but positively to MMPs also in patients, linking our findings to a clinical setting. Conclusion In this study, we demonstrate that TRIM21 influences atherosclerosis via regulation of Th17 responses, with TRIM21 deficiency promoting IL-17 expression and a more fibrous, stable, phenotype of the plaques.
Collapse
Affiliation(s)
- Susanna Brauner
- Unit of Experimental Rheumatology, Department of Medicine, Karolinska Insititutet, Center for Molecular Medicine L8:04, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Xintong Jiang
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gudny Ella Thorlacius
- Unit of Experimental Rheumatology, Department of Medicine, Karolinska Insititutet, Center for Molecular Medicine L8:04, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Anna M Lundberg
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Therese Östberg
- Unit of Experimental Rheumatology, Department of Medicine, Karolinska Insititutet, Center for Molecular Medicine L8:04, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Zhong-Qun Yan
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Vijay K Kuchroo
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Harvard University, Boston, USA
| | - Göran K Hansson
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marie Wahren-Herlenius
- Unit of Experimental Rheumatology, Department of Medicine, Karolinska Insititutet, Center for Molecular Medicine L8:04, Karolinska University Hospital, 171 76 Stockholm, Sweden
| |
Collapse
|
206
|
Chai Y, Yin Z, Fan Q, Zhang Z, Ye K, Xu Y, Xiao W, Chai X, Zhu T, Nie H. Protective Effects of Angong Niuhuang Pill on Early Atherosclerosis in ApoE -/- Mice by Reducing the Inflammatory Response. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:9747212. [PMID: 31236126 PMCID: PMC6545748 DOI: 10.1155/2019/9747212] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/25/2019] [Accepted: 05/02/2019] [Indexed: 12/28/2022]
Abstract
Atherosclerosis (AS) is the primary cause of cardiocerebrovascular disease, and inflammation is responsible for the initiation of its pathogenesis. Therefore, targeting inflammatory pathways to prevent AS progression is an ideal strategy. Angong Niuhuang pill (ANP) is a well-known traditional Chinese medicine and has been widely used for thousands of years to treat central nervous system and cardiovascular diseases. In this study, we investigated the role of ANP in reducing inflammation during early AS, using a high-fat diet-induced ApoE-/- mouse model of AS. Compared to those with simvastatin, ANP had no significant effect on serum triglyceride, low-density lipoprotein, and high-density lipoprotein levels. However, it effectively inhibited splenic and vascular inflammation. This agent also reduced the Th17/CD4+T ratio and mRNA expression of IL-6 and increased the Treg/CD4+T ratio and mRNA expression of TGF-β1. Thus, ANP restored Th17/Treg homeostasis in the spleen. It also regulated pro- and anti-inflammatory cytokine expression in the aorta in a similar manner. Further, it downregulated the expression of chemokine receptors (CCR2, CXCR3), their ligands (MCP-1, MCP-2, and MCP-3), and cell adhesion molecules (VCAM-1, ICAM-1) in arterial vessels. These results indicate that ANP can ameliorate the development of early AS, mainly by reducing inflammation instead of acting as an antihyperlipidemic drug.
Collapse
Affiliation(s)
- Yushuang Chai
- Guangzhou Baiyunshan Zhongyi Pharmaceutical Co., Ltd, Guangzhou 510530, China
| | - Zhen Yin
- Guangzhou Baiyunshan Zhongyi Pharmaceutical Co., Ltd, Guangzhou 510530, China
| | - Qinghong Fan
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, Guangdong, China
| | - Zhe Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, Guangdong, China
| | - Kaihe Ye
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, Guangdong, China
| | - Yimin Xu
- Guangzhou Baiyunshan Zhongyi Pharmaceutical Co., Ltd, Guangzhou 510530, China
| | - Wei Xiao
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, Guangdong, China
| | - Xiaomeng Chai
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, Guangdong, China
| | - Tao Zhu
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, Guangdong, China
| | - Hong Nie
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, Guangdong, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, China
| |
Collapse
|
207
|
The Effects of Known Cardioprotective Drugs on Proinflammatory Cytokine Secretion From Human Coronary Artery Endothelial Cells. Am J Ther 2019; 26:e321-e332. [DOI: 10.1097/mjt.0000000000000648] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
208
|
A Pilot, Randomized Study in Women of Nutrition-Related Clinical Chemistry at 6 Weeks after Roux en Y Gastric Bypass: Comparison of Two Nutrition Support Plans. Obes Surg 2019; 29:2781-2789. [DOI: 10.1007/s11695-019-03895-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
209
|
Lawal IO, Ankrah AO, Stoltz AC, Sathekge MM. Radionuclide imaging of inflammation in atherosclerotic vascular disease among people living with HIV infection: current practice and future perspective. Eur J Hybrid Imaging 2019; 3:5. [PMID: 34191183 PMCID: PMC8218042 DOI: 10.1186/s41824-019-0053-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/11/2019] [Indexed: 01/03/2023] Open
Abstract
People living with human immunodeficiency virus (HIV) infection have twice the risk of atherosclerotic vascular disease compared with non-infected individuals. Inflammation plays a critical role in the development and progression of atherosclerotic vascular disease. Therapies targeting inflammation irrespective of serum lipid levels have been shown to be effective in preventing the occurrence of CVD. Radionuclide imaging is a viable method for evaluating arterial inflammation. This evaluation is useful in quantifying CVD risk and for assessing the effectiveness of anti-inflammatory treatment. The most tested radionuclide method for quantifying arterial inflammation among people living with HIV infection has been with F-18 FDG PET/CT. The level of arterial uptake of F-18 FDG correlates with vascular inflammation and with the risk of development and progression of atherosclerotic disease. Several limitations exist to the use of F-18 FDG for PET quantification of arterial inflammation. Many targets expressed on macrophage, a significant player in arterial inflammation, have the potential for use in evaluating arterial inflammation among people living with HIV infection. The review describes the clinical utility of F-18 FDG PET/CT in assessing arterial inflammation as a risk for atherosclerotic disease among people living with HIV infection. It also outlines potential newer probes that may quantify arterial inflammation in the HIV-infected population by targeting different proteins expressed on macrophages.
Collapse
Affiliation(s)
- Ismaheel O. Lawal
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001 South Africa
| | - Alfred O. Ankrah
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001 South Africa
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen & University of Groningen, Groningen, The Netherlands
| | - Anton C. Stoltz
- Infectious Disease Unit, Department of Internal Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, South Africa
| | - Mike M. Sathekge
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001 South Africa
| |
Collapse
|
210
|
Yue W, Li Y, Ou D, Yang Q. The GLP-1 receptor agonist liraglutide protects against oxidized LDL-induced endothelial inflammation and dysfunction via KLF2. IUBMB Life 2019; 71:1347-1354. [PMID: 30969479 DOI: 10.1002/iub.2046] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/22/2019] [Accepted: 03/22/2019] [Indexed: 02/05/2023]
Abstract
Cardiovascular complications are the major causes of the mortality and morbidities in diabetic patients. The diabetic patients have an increased risk of developing atherosclerosis, which could lead to heart attack and stroke. Glucagon-like peptide 1 (GLP-1) receptor agonists are a class of potent anti-glycemic agents to treat diabetes. Recently, several GLP-1 receptor agonists have been found to have cardiovascular benefit independent of their glucose lowing ability. Liraglutide is one of clinically approved effective GLP-1 receptor agonists. In this study, we explored the molecular mechanism of Liraglutide against oxidized low-density lipoprotein (ox-LDL) in cultured endothelial cells. Our data show that Liraglutide treatment ameliorates ox-LDL caused reduction of the transcriptional factor KLF2. In the same experiment, Liraglutide also rescues ox-LDL induced reduction of mitogen-activated protein kinase (MAPK) kinase extracellular signal regulated kinase 5 (ERK5) phosphorylation, and blockage of ERK5 activity by its inhibitor XMD8-92 abolishes the protection of Liraglutide on KLF2 expression. These facts suggest that the action of Liraglutide on endothelial KLF2 is dependent on ERK5. Liraglutide also recovers ox-LDL caused reduction of endothelial tight junctions protein Occludin and ameliorates ox-LDL induced endothelial monolayer permeability increase. On the other hand, Liraglutide inhibits ox-LDL induced expression of vascular adhesion molecules (E-selectin and vascular cell adhesion molecule 1), and prevents ox-LDL induced attachment of monocytes adhesion to endothelial cells. Moreover, Liraglutide mitigates ox-LDL triggered reduction of endothelial nitric oxide synthase (eNOS) expression and NO release. Collectively, our study provides multiple facets of the mechanisms that Liraglutide is a protective agent in endothelial cells and has the potential implication in therapeutic usage of vascular complication in diabetes patients. © 2019 IUBMB Life, 71(9):1347-1354, 2019.
Collapse
Affiliation(s)
- Wen Yue
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yi Li
- Department of Cardiology, Jiajiang Rehabilitation Hospital, Leshan, Sichuan, People's Republic of China
| | - Dengke Ou
- Department of Cardiology, Chengdu Fifth People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Qing Yang
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
211
|
Schick D, Babendreyer A, Wozniak J, Awan T, Noels H, Liehn E, Bartsch JW, Vlacil AK, Grote K, Zayat R, Goetzenich A, Ludwig A, Dreymueller D. Elevated expression of the metalloproteinase ADAM8 associates with vascular diseases in mice and humans. Atherosclerosis 2019; 286:163-171. [PMID: 30910225 DOI: 10.1016/j.atherosclerosis.2019.03.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/17/2019] [Accepted: 03/12/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Members of the family of a disintegrin and metalloproteinases (ADAMs) and their substrates have been previously shown to modulate the inflammatory response in cardiac diseases, but studies investigating the relevance of ADAM8 are still rare. Our aim is to provide evidence for the inflammatory dysregulation of ADAM8 in vascular diseases and its association with disease severity. METHODS Western-type diet fed Apoe-/- and Ldlr-/- mice and artery ligation served as murine model for atherosclerosis and myocardial infarction, respectively. Human bypass grafts were used to study the association with coronary artery disease (CAD), with the simplified acute physiology score II (SAPS II) as a measure of postoperative organ dysfunction. Human primary vascular and blood cells were analyzed under basal and inflammatory conditions. mRNA levels were determined by RT-qPCR, ADAM8 protein levels by ELISA, immunohistochemistry or flow cytometry. RESULTS ADAM8/ADAM8 expression is associated with atherosclerosis and CAD such as myocardial infarction in both mice and humans, especially in endothelial cells and leukocytes. We observed a strong in vivo and in vitro correlation of ADAM8 with the vascular disease markers VCAM-1, ICAM-1, TNF, IL-6, and CCL-2. Serum analysis revealed a significant elevation of soluble ADAM8 serum levels correlating with soluble CXCL16 levels and SAPS II. CONCLUSIONS We demonstrate a general association of ADAM8 with cardiovascular diseases in mice and humans predominantly acting in endothelial cells and leukocytes. The correlation with postoperative organ dysfunctions in CAD patients highlights the value of further studies investigating the specific function of ADAM8 in cardiovascular diseases.
Collapse
Affiliation(s)
- Daniel Schick
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Aaron Babendreyer
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Justyna Wozniak
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Tanzeela Awan
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Heidi Noels
- Institute of Molecular Cardiovascular Research, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Elisa Liehn
- Institute of Molecular Cardiovascular Research, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany; National Heart Center Singapore, Singapore, Human Genetic Laboratory, University of Medicine Craiova, Romania
| | - Jörg-W Bartsch
- Department of Neurosurgery, Philipps University Marburg, University Hospital Marburg, Baldingerstrasse, 35033, Marburg, Germany
| | - Ann-Kathrin Vlacil
- Clinic for Internal Medicine, Cardiology, Philipps University Marburg, University Hospital Marburg, Marburg, Germany
| | - Karsten Grote
- Clinic for Internal Medicine, Cardiology, Philipps University Marburg, University Hospital Marburg, Marburg, Germany
| | - Rashad Zayat
- Department of Thoracic and Cardiovascular Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52066, Aachen, Germany
| | - Andreas Goetzenich
- Department of Thoracic and Cardiovascular Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52066, Aachen, Germany
| | - Andreas Ludwig
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Daniela Dreymueller
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, ZHMB, Saarland University, UKS Bldg. 46, 66421, Homburg, Germany.
| |
Collapse
|
212
|
Skeletal Muscle-specific PGC-1α Overexpression Suppresses Atherosclerosis in Apolipoprotein E-Knockout Mice. Sci Rep 2019; 9:4077. [PMID: 30858489 PMCID: PMC6411944 DOI: 10.1038/s41598-019-40643-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 02/21/2019] [Indexed: 12/12/2022] Open
Abstract
Endurance exercise training prevents atherosclerosis. Peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) increases myokine secretion from the skeletal muscle, and these myokines have been shown to affect the function of multiple organs. Since endurance exercise training increases PGC-1α expression in skeletal muscles, we investigated whether skeletal muscle-specific PGC-1α overexpression suppresses atherosclerosis. Apolipoprotein E-knockout (ApoE-KO)/PGC-1α mice, which overexpress PGC-1α in the skeletal muscle of ApoE-KO mice, were sacrificed, and the atherosclerotic plaque area, spontaneous activity, plasma lipid profile, and aortic gene expression were measured. Immunohistochemical analyses were also performed. The atherosclerotic lesions in ApoE-KO/PGC-1α mice were 40% smaller than those in ApoE-KO mice, concomitant with the reduction in vascular cell adhesion molecule-1 (VCAM-1) and monocyte chemoattractant protein-1 (MCP-1) mRNA and protein levels in the aorta. Spontaneous activity and plasma lipid profiles were not changed by the overexpression of PGC-1α in the skeletal muscle. In human umbilical vein endothelial cells, Irisin and β-aminoisobutyric acid (BAIBA), PGC-1α-dependent myokines, inhibited the tumor necrosis factor α-induced VCAM-1 gene and protein expression. BAIBA also inhibited TNFα-induced MCP-1 gene expression. These results showed that the skeletal muscle-specific overexpression of PGC-1α suppresses atherosclerosis and that PGC-1α-dependent myokines may be involved in the preventive effects observed.
Collapse
|
213
|
The effects of occupational lead exposure on selected inflammatory biomarkers. Arh Hig Rada Toksikol 2019; 70:36-41. [PMID: 30956219 DOI: 10.2478/aiht-2019-70-3214] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 03/01/2019] [Indexed: 11/20/2022] Open
Abstract
In exposure to toxic metals such as lead, determining lead and cytokine levels (IL-6, IL-10, and TNF-α) is essential for early detection of diseases. The aim of this study was to develop an model for early detection of inflammation and onset of atherosclerosis in the absence of clinical findings in young workers, which could help physicians take timely an action and start treatment. This study included 49 metal workers exposed to lead occupationally and 50 unexposed administrative workers (controls) who underwent immunological analysis for cytokines (IL-6, IL-10, and TNF-α) and atherosclerosis markers (h-FABP and VCAM-1), toxicological analysis for lead, and routine biochemical analysis (ALT, AST, creatinine) at the Ankara Occupational and Environmental Diseases Hospital in 2017. Lead levels correlated with IL-6, IL-10, and TNF-α (r=0.469, r=0.521 and r=0.279, respectively, p<0.01) but did not significantly affect h-FABP and VCAM-1 levels.
Collapse
|
214
|
Zheng J, Chen K, Zhu Y, Wang H, Chen Z, Yong X, Yin H, Chen J, Lai K, Liu Y. The neurokinin-1 receptor antagonist aprepitant ameliorates oxidized LDL-induced endothelial dysfunction via KLF2. Mol Immunol 2019; 106:29-35. [DOI: 10.1016/j.molimm.2018.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/15/2018] [Accepted: 12/09/2018] [Indexed: 12/15/2022]
|
215
|
Fan X, Chen X, Feng Q, Peng K, Wu Q, Passerini AG, Simon SI, Sun C. Downregulation of GATA6 in mTOR-inhibited human aortic endothelial cells: effects on TNF-α-induced VCAM-1 expression and monocytic cell adhesion. Am J Physiol Heart Circ Physiol 2019; 316:H408-H420. [PMID: 30462552 PMCID: PMC6397389 DOI: 10.1152/ajpheart.00411.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 10/19/2018] [Accepted: 11/07/2018] [Indexed: 01/13/2023]
Abstract
Increased expression of vascular cell adhesion molecule 1 (VCAM-1) on the aortic endothelium is an early marker of atherogenesis, promoted in part by elevated levels of inflammatory cytokines such as TNF-α. Mammalian target of rapamycin (mTOR) is a ubiquitous signaling molecule that has been considered to contribute to diverse cellular processes through mTOR complex 1 (mTORC1) or complex 2 (mTORC2). This study aimed to elucidate the role of mTOR signaling in TNF-α-induced VCAM-1 expression by the arterial endothelium. Primary human aortic endothelial cells (HAECs) were treated with low-dose (0.1 ng/ml) TNF-α, and VCAM-1 expression was measured by real-time quantitative PCR, Western blot analysis, and flow cytometry. Inhibition of mTOR through siRNA-mediated depletion or treatment with chemical inhibitors rapamycin or torin 1 suppressed VCAM1 transcription, which translated to inhibition of VCAM-1 surface expression by HAECs and concomitant decreased adhesion of monocytes. A promoter luciferase assay and chromatin immunoprecipitation indicated that mTOR regulated VCAM1 transcription through a mechanism involving transcription factor GATA6. Activation of PKC-α and an increase in miR-200a-3p expression, caused by mTOR inhibition but not disruption of mTORC1 or mTORC2 singly or together, decreased TNF-α-induced GATA6 expression and its enrichment at the VCAM1 promoter. In conclusion, mTOR inhibition activates PKC-α independently of disruption of mTORC1 and/or mTORC2, which challenges the conventional wisdom regarding mTOR signaling. Moreover, mTOR signals through transcriptional and posttranscriptional mechanisms to elicit maximal cytokine-induced endothelial inflammation that precedes atherosclerosis. NEW & NOTEWORTHY Both mammalian target of rapamycin (mTOR) complex 1 (mTORC1) and mTORC2 contribute to PKC-α activation in the human aortic endothelium. Inhibition of mTOR is not equivalent to disruption of mTORC1 and/or mTORC2 in affecting human aortic endothelial cell signaling. Specifically, inhibition of mTOR causes PKC-α activation and miR-200a-3p upregulation, which independently suppresses TNF-α-induced transcription factor GATA6 expression and subsequently inhibits VCAM-1 expression and monocytic cell adhesion onto the aortic endothelium.
Collapse
Affiliation(s)
- Xing Fan
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University , Nanjing , China
| | - Xiaolin Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University , Nanjing , China
| | - Qi Feng
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University , Nanjing , China
| | - Kai Peng
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University , Nanjing , China
| | - Qianqian Wu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University , Nanjing , China
| | - Anthony G Passerini
- Department of Biomedical Engineering, University of California , Davis, California
| | - Scott I Simon
- Department of Biomedical Engineering, University of California , Davis, California
| | - ChongXiu Sun
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University , Nanjing , China
| |
Collapse
|
216
|
Wadey RM, Connolly KD, Mathew D, Walters G, Rees DA, James PE. Inflammatory adipocyte-derived extracellular vesicles promote leukocyte attachment to vascular endothelial cells. Atherosclerosis 2019; 283:19-27. [PMID: 30771557 DOI: 10.1016/j.atherosclerosis.2019.01.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 11/21/2018] [Accepted: 01/10/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS Obesity is associated with an increased risk of cardiovascular disease, but the mechanisms involved are not completely understood. In obesity, the adipocyte microenvironment is characterised by both hypoxia and inflammation. Therefore, we sought to determine whether extracellular vesicles (EVs) derived from adipocytes in this setting might be involved in mediating cardiovascular disease, specifically by promoting leukocyte attachment to vascular endothelial cells. METHODS Mature 3T3-L1 adipocytes were incubated for 24 h under control, TNF-α (30 ng/mL), hypoxia (1% O2), or TNF-α+hypoxia (30 ng/mL, 1% O2) conditions. EVs were isolated by differential ultracentrifugation and analysed by nanoparticle tracking analysis. Primary human umbilical vein endothelial cells (HUVECs) were treated with EVs for 6 h before being lysed for Western blotting to investigate changes in adhesion molecule production, or for use in leukocyte attachment assays. RESULTS EVs from adipocytes treated with TNF-α and TNF-α+hypoxia increased vascular cell adhesion molecule (VCAM-1) production in HUVECs compared to basal level (4.2 ± 0.6 and 3.8 ± 0.3-fold increase, respectively (p < 0.05)), an effect that was inhibited by an anti-TNF-α neutralising antibody. Production of other adhesion molecules (E-selectin, P-selectin, platelet endothelial cell adhesion molecule and VE-Cadherin) was unchanged. Pre-incubating HUVECs with TNF-α+hypoxia EVs significantly increased leukocyte attachment compared to basal level (3.0 ± 0.4-fold increase (p < 0.05)). CONCLUSIONS Inflammatory adipocyte EVs induce VCAM-1 production in vascular endothelial cells, accompanied by enhanced leukocyte attachment. Preventing adipocyte derived EV-induced VCAM-1 upregulation may offer a novel therapeutic target in the prevention of obesity-driven cardiovascular disease.
Collapse
Affiliation(s)
- Rebecca M Wadey
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, 200 Western Avenue, Cardiff, CF5 2YB, UK
| | - Katherine D Connolly
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, 200 Western Avenue, Cardiff, CF5 2YB, UK
| | - Donna Mathew
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, 200 Western Avenue, Cardiff, CF5 2YB, UK
| | - Gareth Walters
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, 200 Western Avenue, Cardiff, CF5 2YB, UK
| | - D Aled Rees
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Philip E James
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, 200 Western Avenue, Cardiff, CF5 2YB, UK.
| |
Collapse
|
217
|
Ma S, Bai Z, Wu H, Wang W. The DPP-4 inhibitor saxagliptin ameliorates ox-LDL-induced endothelial dysfunction by regulating AP-1 and NF-κB. Eur J Pharmacol 2019; 851:186-193. [PMID: 30639312 DOI: 10.1016/j.ejphar.2019.01.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/16/2018] [Accepted: 01/08/2019] [Indexed: 12/12/2022]
Abstract
Diabetes-associated cardiovascular complications are the leading cause of death for diabetic patients. Dipeptidyl peptidase 4 (DPP-4) inhibitor agents, known as gliptins, are a class of potent anti-glycemic agents developed to treat diabetes. Recently, gliptins have been shown to have independent cardiovascular benefits. In this study, we revealed the protective role of saxagliptin in vascular endothelial cells. Our data show that saxagliptin suppresses oxidized low-density lipoprotein cholesterol (ox-LDL)-induced expression of its receptor lectin-like ox-LDL receptor-1 (LOX-1). Saxagliptin treatment reduces ox-LDL-induced production of cytokines and vascular adhesion molecules including tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), vascular cell adhesion molecule 1 (VCAM-1), and intercellular cell adhesion molecule-1 (ICAM-1). The presence of saxagliptin suppressed ox-LDL-induced adhesion of monocytes to endothelial cells in co-culture adhesion experiments. Moreover, saxagliptin mitigated ox-LDL-induced production of reactive oxygen species and suppressed elevated expression of endothelial nicotinamide adenine dinucleotide phosphate oxidase subunit (NOX-4) induced by ox-LDL. Mechanistically, saxagliptin exerted inhibitory effects against ox-LDL-induced phosphorylation of JNK kinase, expression of the activator protein 1 (AP-1) subunits c-Jun/c-fos, and AP-1 promoter activity. Saxagliptin also suppressed nuclear factor κB (NF-κB) p65 accumulation and inhibited its promoter activity. Our data elaborate the molecular mechanism of saxagliptin-mediated endothelial protection and indicate that saxagliptin could have vascular benefits independent on its anti-glycemic function.
Collapse
Affiliation(s)
- Suxia Ma
- Cardiology Department 2, Shangqiu First People's Hospital, Shangqiu, Henan 476100, China
| | - Zhifeng Bai
- Cardiology Department 2, Shangqiu First People's Hospital, Shangqiu, Henan 476100, China.
| | - Huiying Wu
- Cardiology Department, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan 450007, China
| | - Wei Wang
- Cardiology Department, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan 450007, China
| |
Collapse
|
218
|
Deciphering Endothelial Dysfunction in the HIV-Infected Population. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1134:193-215. [PMID: 30919339 DOI: 10.1007/978-3-030-12668-1_11] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cardiovascular disease (CVD), as a possible consequence of endothelial dysfunction, is prevalent among HIV-infected patients despite successful administration of antiretroviral drugs. This warrants the routine clinical assessment of endothelial function in HIV-positive patients to circumvent potential CVD events. Several different non-invasive strategies have been employed to assess endothelial function in clinical research studies yielding inconsistencies among these reports. This review summarises the different techniques used for assessing endothelial function, with a focus on proposed blood-based biomarkers, such as endothelial leukocyte adhesion molecule-1 (E-selectin), soluble intercellular adhesion molecule-1 (sICAM-1), soluble vascular cell adhesion molecule-1 (sVCAM-1), von Willebrand factor (vWF), TNF-α, interleukin 6 (IL6) and soluble thrombomodulin (sTM). The identification of suitable blood-based biomarkers, especially those that can be measured using a point-of-care device, would be more applicable in under-resourced countries where the prevalence of HIV is high.
Collapse
|
219
|
Bruxel MA, Tavares AMV, Zavarize Neto LD, de Souza Borges V, Schroeder HT, Bock PM, Rodrigues MIL, Belló-Klein A, Homem de Bittencourt PI. Chronic whole-body heat treatment relieves atherosclerotic lesions, cardiovascular and metabolic abnormalities, and enhances survival time restoring the anti-inflammatory and anti-senescent heat shock response in mice. Biochimie 2019; 156:33-46. [DOI: 10.1016/j.biochi.2018.09.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 09/26/2018] [Indexed: 12/15/2022]
|
220
|
Crane ED, Al-Hashimi AA, Chen J, Lynn EG, Won KD, Lhoták Š, Naeim M, Platko K, Lebeau P, Byun JH, Shayegan B, Krepinsky JC, Rayner KJ, Marchiò S, Pasqualini R, Arap W, Austin RC. Anti-GRP78 autoantibodies induce endothelial cell activation and accelerate the development of atherosclerotic lesions. JCI Insight 2018; 3:99363. [PMID: 30568038 DOI: 10.1172/jci.insight.99363] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 11/09/2018] [Indexed: 12/31/2022] Open
Abstract
The 78-kDa glucose-regulated protein (GRP78) is an ER molecular chaperone that aids in protein folding and secretion. However, pathological conditions that cause ER stress can promote the relocalization of GRP78 to the cell surface (csGRP78), where it acts as a signaling receptor to promote cancer progression. csGRP78 also possesses antigenic properties, leading to the production of anti-GRP78 autoantibodies, which contribute to tumor growth. In contrast, the presence and role of anti-GRP78 autoantibodies in atherosclerosis is unknown. Here, we show that atherosclerotic-prone ApoE-/- mice develop circulating anti-GRP78 autoantibodies that bind to csGRP78 on lesion-resident endothelial cells. Moreover, GRP78-immunized ApoE-/- mice exhibit a marked increase in circulating anti-GRP78 autoantibody titers that correlated with accelerated lesion growth. Mechanistically, engagement of anti-GRP78 autoantibodies with csGRP78 on human endothelial cells activated NF-κB, thereby inducing the expression of ICAM-1 and VCAM-1, a process blocked by NF-κB inhibitors. Disrupting the autoantibody/csGRP78 complex with enoxaparin, a low-molecular-weight heparin, reduced the expression of adhesion molecules and attenuated lesion growth. In conclusion, anti-GRP78 autoantibodies play a crucial role in atherosclerosis development, and disruption of the interaction between anti-GRP78 autoantibodies and csGRP78 represents a therapeutic strategy.
Collapse
Affiliation(s)
| | - Ali A Al-Hashimi
- Department of Medicine, Division of Nephrology, and.,Division of Urology, Department of Surgery, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, Ontario, Canada
| | - Jack Chen
- Department of Medicine, Division of Nephrology, and
| | | | | | - Šárka Lhoták
- Department of Medicine, Division of Nephrology, and
| | - Magda Naeim
- Department of Medicine, Division of Nephrology, and
| | | | - Paul Lebeau
- Department of Medicine, Division of Nephrology, and
| | | | - Bobby Shayegan
- Division of Urology, Department of Surgery, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, Ontario, Canada
| | | | - Katey J Rayner
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa and University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Serena Marchiò
- Department of Oncology, University of Turin, Candiolo, Italy.,Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia, Istituto di Ricerca e Cura a Carattere Scientifico, Candiolo, Italy
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, USA.,Division of Cancer Biology, Department of Radiation Oncology, and
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, USA.,Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Richard C Austin
- Department of Biochemistry and Biomedical Sciences.,Department of Medicine, Division of Nephrology, and
| |
Collapse
|
221
|
Bhatt SP, Nath HP, Kim YI, Ramachandran R, Watts JR, Terry NLJ, Sonavane S, Deshmane SP, Woodruff PG, Oelsner EC, Bodduluri S, Han MK, Labaki WW, Michael Wells J, Martinez FJ, Barr RG, Dransfield MT. Centrilobular emphysema and coronary artery calcification: mediation analysis in the SPIROMICS cohort. Respir Res 2018; 19:257. [PMID: 30563576 PMCID: PMC6299495 DOI: 10.1186/s12931-018-0946-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/20/2018] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is associated with a two-to-five fold increase in the risk of coronary artery disease independent of shared risk factors. This association is hypothesized to be mediated by systemic inflammation but this link has not been established. METHODS We included 300 participants enrolled in the SPIROMICS cohort, 75 each of lifetime non-smokers, smokers without airflow obstruction, mild-moderate COPD, and severe-very severe COPD. We quantified emphysema and airway disease on computed tomography, characterized visual emphysema subtypes (centrilobular and paraseptal) and airway disease, and used the Weston visual score to quantify coronary artery calcification (CAC). We used the Sobel test to determine whether markers of systemic inflammation mediated a link between spirometric and radiographic features of COPD and CAC. RESULTS FEV1/FVC but not quantitative emphysema or airway wall thickening was associated with CAC (p = 0.036), after adjustment for demographics, diabetes mellitus, hypertension, statin use, and CT scanner type. To explain this discordance, we examined visual subtypes of emphysema and airway disease, and found that centrilobular emphysema but not paraseptal emphysema or bronchial thickening was independently associated with CAC (p = 0.019). MMP3, VCAM1, CXCL5 and CXCL9 mediated 8, 8, 7 and 16% of the association between FEV1/FVC and CAC, respectively. Similar biomarkers partially mediated the association between centrilobular emphysema and CAC. CONCLUSIONS The association between airflow obstruction and coronary calcification is driven primarily by the centrilobular subtype of emphysema, and is linked through bioactive molecules implicated in the pathogenesis of atherosclerosis. TRIAL REGISTRATION ClinicalTrials.gov: Identifier: NCT01969344 .
Collapse
Affiliation(s)
- Surya P Bhatt
- Division of Pulmonary, Allergy and Critical Care Medicine and Lung Health Center, University of Alabama at Birmingham, THT 422, 1720, 2nd Avenue South, Birmingham, AL, 35294, USA.
- UAB Lung Imaging Core, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Hrudaya P Nath
- UAB Lung Imaging Core, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Young-Il Kim
- Division of Pulmonary, Allergy and Critical Care Medicine and Lung Health Center, University of Alabama at Birmingham, THT 422, 1720, 2nd Avenue South, Birmingham, AL, 35294, USA
- Department of Preventive Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Rekha Ramachandran
- Department of Preventive Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jubal R Watts
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Nina L J Terry
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Sushil Sonavane
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Swati P Deshmane
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Prescott G Woodruff
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University California San Francisco, San Francisco, CA, 94143, USA
| | - Elizabeth C Oelsner
- Division of Pulmonary, Allergy and Critical Care Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Sandeep Bodduluri
- Division of Pulmonary, Allergy and Critical Care Medicine and Lung Health Center, University of Alabama at Birmingham, THT 422, 1720, 2nd Avenue South, Birmingham, AL, 35294, USA
- UAB Lung Imaging Core, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - MeiLan K Han
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Wassim W Labaki
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - J Michael Wells
- Division of Pulmonary, Allergy and Critical Care Medicine and Lung Health Center, University of Alabama at Birmingham, THT 422, 1720, 2nd Avenue South, Birmingham, AL, 35294, USA
- UAB Lung Imaging Core, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Birmingham Veterans Affairs Hospital, Birmingham, AL, 35294, USA
| | - Fernando J Martinez
- Division of Pulmonary and Critical Care Medicine, Weill Cornell School of Medicine, New York, NY, 10065, USA
| | - R Graham Barr
- Division of Pulmonary, Allergy and Critical Care Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Mark T Dransfield
- Division of Pulmonary, Allergy and Critical Care Medicine and Lung Health Center, University of Alabama at Birmingham, THT 422, 1720, 2nd Avenue South, Birmingham, AL, 35294, USA
- UAB Lung Imaging Core, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Birmingham Veterans Affairs Hospital, Birmingham, AL, 35294, USA
| |
Collapse
|
222
|
Barron AMS, Mantero JC, Ho JD, Nazari B, Horback KL, Bhawan J, Lafyatis R, Lam C, Browning JL. Perivascular Adventitial Fibroblast Specialization Accompanies T Cell Retention in the Inflamed Human Dermis. THE JOURNAL OF IMMUNOLOGY 2018; 202:56-68. [PMID: 30510068 DOI: 10.4049/jimmunol.1801209] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022]
Abstract
Perivascular accumulation of lymphocytes can be a prominent histopathologic feature of various human inflammatory skin diseases. Select examples include systemic sclerosis, spongiotic dermatitis, and cutaneous lupus. Although a large body of work has described various aspects of the endothelial and vascular smooth muscle layers in these diseases, the outer adventitial compartment is poorly explored. The goal of the current study was to characterize perivascular adventitial fibroblast states in inflammatory human skin diseases and relate these states to perivascular lymphocyte accumulation. In normal skin, adventitial fibroblasts are distinguished by CD90 expression, and dense perivascular lymphocytic infiltrates are uncommon. In systemic sclerosis, this compartment expands, but lymphocyte infiltrates remain sparse. In contrast, perivascular adventitial fibroblast expression of VCAM1 is upregulated in spongiotic dermatitis and lupus and is associated with a dense perivascular T cell infiltrate. VCAM1 expression marks transitioned fibroblasts that show some resemblance to the reticular stromal cells in secondary lymphoid organs. Expanded adventitial compartments with perivascular infiltrates similar to the human settings were not seen in the inflamed murine dermis. This species difference may hinder the dissection of aspects of perivascular adventitial pathology. The altered perivascular adventitial compartment and its associated reticular network form a niche for lymphocytes and appear to be fundamental in the development of an inflammatory pattern.
Collapse
Affiliation(s)
- Alexander M S Barron
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
| | - Julio C Mantero
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
| | - Jonathan D Ho
- Department of Dermatology, Boston University School of Medicine, Boston, MA 02118
| | - Banafsheh Nazari
- Section of Rheumatology, Boston University School of Medicine, Boston, MA 02118
| | - Katharine L Horback
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and
| | - Jag Bhawan
- Department of Dermatology, Boston University School of Medicine, Boston, MA 02118
| | - Robert Lafyatis
- Section of Rheumatology, Boston University School of Medicine, Boston, MA 02118.,Division of Rheumatology and Clinical Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213
| | - Christina Lam
- Department of Dermatology, Boston University School of Medicine, Boston, MA 02118
| | - Jeffrey L Browning
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118; .,Section of Rheumatology, Boston University School of Medicine, Boston, MA 02118
| |
Collapse
|
223
|
Interactions between Roseburia intestinalis and diet modulate atherogenesis in a murine model. Nat Microbiol 2018; 3:1461-1471. [PMID: 30397344 PMCID: PMC6280189 DOI: 10.1038/s41564-018-0272-x] [Citation(s) in RCA: 354] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 09/24/2018] [Indexed: 12/14/2022]
Abstract
Humans with metabolic and inflammatory diseases frequently harbour lower levels of butyrate-producing bacteria in their gut. However, it is not known whether variation in the levels of these organisms is causally linked with disease development and whether diet modifies the impact of these bacteria on health. Here we show that a prominent gut-associated butyrate-producing bacterial genus (Roseburia) is inversely correlated with atherosclerotic lesion development in a genetically diverse mouse population. We use germ-free apolipoprotein E-deficient mice colonized with synthetic microbial communities that differ in their capacity to generate butyrate to demonstrate that Roseburia intestinalis interacts with dietary plant polysaccharides to: impact gene expression in the intestine, directing metabolism away from glycolysis and toward fatty acid utilization; lower systemic inflammation; and ameliorate atherosclerosis. Furthermore, intestinal administration of butyrate reduces endotoxaemia and atherosclerosis development. Together, our results illustrate how modifiable diet-by-microbiota interactions impact cardiovascular disease, and suggest that interventions aimed at increasing the representation of butyrate-producing bacteria may provide protection against atherosclerosis.
Collapse
|
224
|
Choi HJ, Kim NE, Kim BM, Seo M, Heo JH. TNF-α-Induced YAP/TAZ Activity Mediates Leukocyte-Endothelial Adhesion by Regulating VCAM1 Expression in Endothelial Cells. Int J Mol Sci 2018; 19:ijms19113428. [PMID: 30388809 PMCID: PMC6274800 DOI: 10.3390/ijms19113428] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 12/14/2022] Open
Abstract
YAP/TAZ, a transcriptional co-activator of Hippo pathway, has emerged as a central player in vessel homeostasis such as sprouting angiogenesis and vascular barrier stabilization, during development. However, the role of YAP/TAZ in pathological angiogenesis remains unclear. Here, we demonstrated that YAP/TAZ is a critical mediator in leukocyte-endothelial adhesion induced by the vascular inflammatory cytokine TNF-α. YAP/TAZ was dephosphorylated, translocated from the cytosol to the nucleus, and activated by TNF-α in endothelial cells. A specific inhibitor of Rho GTPases suppressed the TNF-α-induced dephosphorylation of YAP. Knockdown of YAP/TAZ using siRNA significantly reduced the expression of the leukocyte adhesion molecule VCAM1 induced by TNF-α. The adhesion of monocytes to endothelial cells was also markedly reduced by YAP/TAZ silencing. However, knockdown of YAP/TAZ did not affect TNF-α-induced NF-κB signaling. Overall, these results suggest that YAP/TAZ plays critical roles in regulating TNF-α-induced endothelial cell adhesive properties without affecting the NF-κB pathway, and implicate YAP/TAZ as a potential therapeutic target for treating inflammatory vascular diseases.
Collapse
Affiliation(s)
- Hyun-Jung Choi
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases (SIRIC), Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Na-Eun Kim
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases (SIRIC), Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Byeong Mo Kim
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases (SIRIC), Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Miran Seo
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases (SIRIC), Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Ji Hoe Heo
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases (SIRIC), Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
- Department of Neurology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| |
Collapse
|
225
|
Chang WC, Yu YM, Cheng AC. Curcumin suppresses pro-inflammatory adhesion response in Human Umbilical Vein Endothelial Cells. J Food Biochem 2018. [DOI: 10.1111/jfbc.12623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Weng-Cheng Chang
- Department of Otolaryngology; Taichung Tzu Chi Hospital; Buddhist Tzu Chi Medical Foundation; Taiwan
| | - Ya-Mei Yu
- Department of Senior Citizen Service Management; National Taichung University of Science and Technology; Taichung Taiwan
| | - An-Chin Cheng
- Department of Nutrition and Health Sciences; Chang Jung Christian University; Tainan Taiwan
| |
Collapse
|
226
|
Lin-Wang HT, Cipullo R, Dias França JI, Finger MA, Rossi Neto JM, Correia EDB, Dinkhuysen JJ, Hirata MH. Intragraft vasculitis and gene expression analysis: Association with acute rejection and prediction of mortality in long-term heart transplantation. Clin Transplant 2018; 32:e13373. [PMID: 30080295 DOI: 10.1111/ctr.13373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/31/2018] [Accepted: 08/01/2018] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Vasculitis entails heterogeneous origins; it starts with an inflammatory process that leads to small vessels' necrosis, hemorrhage, and ischemic lesion, and may further result in occlusion of the vascular lumen. Vasculitis' contribution to allograft rejection is still unclear. This study aims to investigate the incidence of vasculitis in the early stages of heart transplantation as well as to assess the intragraft genes' expression associated with vascular function and subsequently to verify the way in which it affects the outcome of the allograft. METHODS In this retrospective study, 300 archive paraffin-embedded endomyocardial biopsies from 63 heart allograft recipients were assessed. Cellular rejection and vasculitis were diagnosed through histological analysis, and antibody-mediated rejection was performed with immunohistochemical C4d staining. The transcripts of ICAM, VCAM, VEGF, CCL2, IFNG, TGFB, TNF, ADIPOR1, and ADIPOR2 genes were examined through quantitative polymerase chain reaction using B2M for normalization. RESULTS We observed a higher prevalence of severe vasculitis in the early period of post-transplant, and recovery was observed to take place around 1 year post-transplant. Additionally, vasculitis was found to be directly associated with acute cellular rejection and antibody-mediated rejection. The intense C4d capillary positivity predicts higher long-term cardiovascular disease mortality. In comparison with the vasculitis-free group, the group with severe vasculitis displayed reduced left ventricular ejection fraction and an upregulation of VCAM and IFNG associated with the downregulation of VEGF, ADIPOR1, and ADIPOR2. CONCLUSION The vasculitis associated with the presence of C4d and the change in intragraft gene expression profile may contribute to poor allograft outcomes.
Collapse
Affiliation(s)
- Hui Tzu Lin-Wang
- Laboratory of Molecular Investigation in Cardiology, Dante Pazzanese Institute of Cardiology, São Paulo, Brazil
| | - Reginaldo Cipullo
- Department of Heart Transplantation, Dante Pazzanese Institute of Cardiology, São Paulo, Brazil
| | - João Italo Dias França
- Statistic and Epidemiology Laboratory, Dante Pazzanese Institute of Cardiology, São Paulo, Brazil
| | - Marco Aurelio Finger
- Department of Heart Transplantation, Dante Pazzanese Institute of Cardiology, São Paulo, Brazil
| | - Joao Manoel Rossi Neto
- Department of Heart Transplantation, Dante Pazzanese Institute of Cardiology, São Paulo, Brazil
| | | | | | - Mário Hiroyuki Hirata
- Laboratory of Molecular Investigation in Cardiology, Dante Pazzanese Institute of Cardiology, São Paulo, Brazil.,School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
227
|
Maguire EM, Pearce SWA, Xiao Q. Foam cell formation: A new target for fighting atherosclerosis and cardiovascular disease. Vascul Pharmacol 2018; 112:54-71. [PMID: 30115528 DOI: 10.1016/j.vph.2018.08.002] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/17/2018] [Accepted: 08/03/2018] [Indexed: 12/23/2022]
Abstract
During atherosclerosis, the gradual accumulation of lipids into the subendothelial space of damaged arteries results in several lipid modification processes followed by macrophage uptake in the arterial wall. The way in which these modified lipoproteins are dealt with determines the likelihood of cholesterol accumulation within the monocyte-derived macrophage and thus its transformation into the foam cell that makes up the characteristic fatty streak observed in the early stages of atherosclerosis. The unique expression of chemokine receptors and cellular adhesion molecules expressed on the cell surface of monocytes points to a particular extravasation route that they can take to gain entry into atherosclerotic site, in order to undergo differentiation into the phagocytic macrophage. Indeed several GWAS and animal studies have identified key genes and proteins required for monocyte recruitment as well cholesterol handling involving lipid uptake, cholesterol esterification and cholesterol efflux. A re-examination of the previously accepted paradigm of macrophage foam cell origin has been called into question by recent studies demonstrating shared expression of scavenger receptors, cholesterol transporters and pro-inflammatory cytokine release by alternative cell types present in the neointima, namely; endothelial cells, vascular smooth muscle cells and stem/progenitor cells. Thus, therapeutic targets aimed at a more heterogeneous foam cell population with shared functions, such as enhanced protease activity, and signalling pathways, mediated by non-coding RNA molecules, may provide greater therapeutic outcome in patients. Finally, studies targeting each aspect of foam cell formation and death using both genetic knock down and pharmacological inhibition have provided researchers with a clearer understanding of the cellular processes at play, as well as helped researchers to identify key molecular targets, which may hold significant therapeutic potential in the future.
Collapse
Affiliation(s)
- Eithne M Maguire
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Stuart W A Pearce
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK.
| |
Collapse
|
228
|
Zarezadeh M, Saedisomeolia A, Khorshidi M, Kord Varkane H, Makhdoomi Arzati M, Abdollahi M, Yekaninejad MS, Hashemi R, Effatpanah M, Mohammadzadeh Honarvar N. Asymmetric dimethylarginine and soluble inter-cellular adhesion molecule-1 serum levels alteration following ginger supplementation in patients with type 2 diabetes: a randomized double-blind, placebo-controlled clinical trial. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2018; 16:/j/jcim.2019.16.issue-2/jcim-2018-0019/jcim-2018-0019.xml. [PMID: 30099412 DOI: 10.1515/jcim-2018-0019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 07/09/2018] [Indexed: 12/11/2022]
Abstract
Aims Patients with type 2 diabetes mellitus (T2DM) are prone to cardiovascular disease (CVD) due to inflammation process and oxidative stress. ADMA (Asymmetric dimethylarginine) and ICAM-1 (inter-cellular adhesion molecule-1) play an important role in CVD pathogenesis. Ginger as an anti-oxidant and anti-inflammation can effect on these biomarkers. The aim of present study was to characterize the effect of ginger supplementation on ADMA and ICAM-1 serum levels in patients with T2DM. Methods The present study is a randomized double-blind clinical trial which is conducted among 45 diabetic patients (nginger=23, nplacebo=22). The participants were randomly divided into two intervention and placebo groups which were received 2 g ginger powder and 2 g wheat flour for 10 weeks, respectively. ADMA and ICAM-1 concentration were measured by ELISA method. Results Ginger supplementation decreased ADMA serum levels significantly (P=0.002) and sICAM-1 serum levels marginally (P=0.097) in supplementation group after intervention. No significant difference was observed between placebo and supplementation groups. Conclusions Present study was conducted among patients with type 2 diabetes mellitus to investigate the effect of ginger supplementation on ADMA and sICAM-1 levels. There was a significant decrement in ADMA serum concentration and slight reduction in sICAM-1 levels in intervention group. The amount of reduction in both biomarkers was not statistically significant in between-groups comparison.
Collapse
Affiliation(s)
- Meysam Zarezadeh
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Saedisomeolia
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Khorshidi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Kord Varkane
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Motahareh Makhdoomi Arzati
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Abdollahi
- AmirAlam Hospital Complex, Marvasti Hospital, Tehran University of Medical Sciences, Tehran, Iran (Islamic Republic of)
| | - Mir Saeed Yekaninejad
- Department of Biostatistics and Epidemiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Rezvan Hashemi
- Department of Geriatric Medicine, Ziaeian Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Effatpanah
- School of Medicine, Ziaeian Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Niyaz Mohammadzadeh Honarvar
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
229
|
Beyond the Foam Cell: The Role of LXRs in Preventing Atherogenesis. Int J Mol Sci 2018; 19:ijms19082307. [PMID: 30087224 PMCID: PMC6121590 DOI: 10.3390/ijms19082307] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is a chronic condition associated with cardiovascular disease. While largely identified by the accumulation of lipid-laden foam cells within the aorta later on in life, atherosclerosis develops over several stages and decades. During atherogenesis, various cell types of the aorta acquire a pro-inflammatory phenotype that initiates the cascade of signaling events facilitating the formation of these foam cells. The liver X receptors (LXRs) are nuclear receptors that upon activation induce the expression of transporters responsible for promoting cholesterol efflux. In addition to promoting cholesterol removal from the arterial wall, LXRs have potent anti-inflammatory actions via the transcriptional repression of key pro-inflammatory cytokines. These beneficial functions sparked an interest in the potential to target LXRs and the development of agonists as anti-atherogenic agents. These early studies focused on mediating the contributions of macrophages to the underlying pathogenesis. However, further evidence has since demonstrated that LXRs reduce atherosclerosis through their actions in multiple cell types apart from those monocytes/macrophages that infiltrate the lesion. LXRs and their target genes have profound effects on multiple other cells types of the hematopoietic system. Furthermore, LXRs can also mediate dysfunction within vascular cell types of the aorta including endothelial and smooth muscle cells. Taken together, these studies demonstrate the whole-body benefits of LXR activation with respect to anti-atherogenesis, and that LXRs remain a viable target for the treatment of atherosclerosis, with a reach which extends beyond plaque macrophages.
Collapse
|
230
|
Barnard SA, Smith W, Mels CMC, Botha S, Schutte AE. Bioavailable IGF-1 is beneficially associated with biomarkers of endothelial function in young healthy adults: The African-PREDICT study. Growth Horm IGF Res 2018; 41:28-33. [PMID: 29936324 DOI: 10.1016/j.ghir.2018.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 06/01/2018] [Accepted: 06/10/2018] [Indexed: 10/14/2022]
Abstract
INTRODUCTION Low circulating levels of insulin-like growth factor-1 (IGF-1) are associated with endothelial dysfunction, subsequently leading to the development of cardiovascular disease. OBJECTIVE To better understand the early phases of vascular deterioration in a young, healthy population, we investigated, cross-sectionally, whether biomarkers of endothelial function (intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1) and von Willebrand factor antigen (vWFag)) are associated with IGF-1 in a healthy study population forming part of the larger African Prospective study on the Early Detection and Identification of Cardiovascular diseases and Hypertension (African-PREDICT). METHOD We included 825 black and white men and women (aged 20-30 years) and determined IGF-1, IGF binding protein-3 (IGFBP-3), ICAM-1, VCAM-1 and vWFag from blood samples. We also measured 24-h blood pressure and health behaviours namely waist circumference, accelerometery, cotinine and gamma glutamyl transferase. We used the IGF-1/IGFBP-3 M ratio as an estimate of bioavailable IGF-1. RESULTS In multivariable-adjusted regression analyses performed in the total group, VCAM-1 associated positively with IGFBP-3 (β = 0.21; p < .001) and negatively with IGF-1/IGFBP-3 (β = -0.18; p < .001). ICAM-1 showed a borderline negative association with IGF-1 (β = -0.09; p = .054) and IGF-1/IGFBP-3 (β = -0.08; p = .057). vWFag was not associated with IGF-1, IGFBP-3 or bioavailable IGF-1. CONCLUSION VCAM-1 is beneficially associated with IGF-1 in a young healthy cohort, independent of sex, ethnicity, blood pressure and health behaviours - thereby confirming the potential importance of bioavailable IGF-1 in early vascular endothelial protection.
Collapse
Affiliation(s)
- Sunelle A Barnard
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa.
| | - Wayne Smith
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa; MRC Research Unit for Hypertension and Cardiovascular disease, North-West University, Potchefstroom, South Africa.
| | - Catharina M C Mels
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa; MRC Research Unit for Hypertension and Cardiovascular disease, North-West University, Potchefstroom, South Africa.
| | - Shani Botha
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa; MRC Research Unit for Hypertension and Cardiovascular disease, North-West University, Potchefstroom, South Africa.
| | - Aletta E Schutte
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa; MRC Research Unit for Hypertension and Cardiovascular disease, North-West University, Potchefstroom, South Africa.
| |
Collapse
|
231
|
7-Ketocholesterol enhances leukocyte adhesion to endothelial cells via p38MAPK pathway. PLoS One 2018; 13:e0200499. [PMID: 30063760 PMCID: PMC6067699 DOI: 10.1371/journal.pone.0200499] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/27/2018] [Indexed: 12/25/2022] Open
Abstract
7-Ketocholesterol is a major dietary cholesterol oxidation product found in high concentrations in atherosclerotic plaques, which contribute to the development of atherosclerosis. This study aimed to investigate the effects of 7-ketocholesterol on endothelial inflammation, as well as the underlying mechanisms. Pretreatment of human umbilical vein endothelial cells (HUVEC) with 7-ketocholesterol significantly enhanced the total interactions between human monocytic cells (THP-1 cell line) and TNFα-activated HUVECs under physiological flow conditions, compared to pretreatment with cholesterol (TNFα+50 μM cholesterol: 13.1 ± 0.54 cells/CPF, TNFα+50 μM 7-ketocholesterol: 18.9 ± 0.35 cells/CPF, p < 0.01). 7-Ketocholesterol enhanced the expression of E-selectin, ICAM-1, and VCAM-1 proteins. It also activated p38 mitogen-activated protein kinase (MAPK), and treatment with a p38 MAPK inhibitor inhibited both E-selectin expression via ATF-2 activation and 7-ketocholesterol-induced THP-1 adhesion to HUVECs. These findings suggest that 7-ketocholesterol enhances leukocyte–endothelial interactions by upregulating the expression of adhesion molecules, presumably via the p38 MAPK-dependent pathway.
Collapse
|
232
|
Nuclear Transcription Factor Kappa B (NF-кB) and Molecular Damage Mechanisms in Acute Cardiovascular Diseases. A Review. JOURNAL OF CARDIOVASCULAR EMERGENCIES 2018. [DOI: 10.2478/jce-2018-0008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Abstract
Worldwide, cardiovascular diseases (CVDs) represent one of the main causes of morbidity and mortality, and acute coronary syndromes are responsible for a large number of sudden cardiac deaths. One of the main challenges that still exist in this area is represented by the early detection and targeted monitoring of the pathophysiology involved in CVDs. During the last couple of years, researchers have highlighted the importance of molecular and epigenetic mechanisms involved in the initiation and augmentation of CVDs, culminating in their most severe form represented by acute myocardial infarction. One of the most studied molecular factors involved in this type of pathology is represented by nuclear transcription factor kappa B (NF-κB), as well as the involvement of microRNAs (miRNAs). It has been suggested that miRNAs can also be involved in the complex process of atheromatous plaque vulnerabilization that leads to an acute cardiac event. In this review paper, we describe the most important molecular mechanisms involved in the pathogenesis of CVDs and atheromatous plaque progression and vulnerabilization, which include molecular mechanisms dependent on NF-κB. For this paper, we used international databases (PubMed and Scopus). The keywords used for the search were “miRNAs biomarkers”, “miRNAs in cardiovascular disease”, “NF-κB in cardiovascular disease”, “molecular mechanism in cardiovascular disease”, and “myocardial NF-κB mechanisms”. Numerous molecular reactions that have NF-κB as a trigger are involved in the pathogenesis of CVDs. Moreover, miRNAs play an important role in initiating and aggravating certain segments of CVDs. Therefore, miRNAs can be used as biomarkers for early evaluation of CVDs. Furthermore, in the future, miRNAs could be used as a targeted molecular therapy in order to block certain mechanisms responsible for inducing CVDs and leading to acute cardiovascular events.
Collapse
|
233
|
Li Y, Huang S, Huang X, Li X, Falcon A, Soutar A, Bornancin F, Jiang Z, Xin HB, Fu M. Pharmacological inhibition of MALT1 protease activity suppresses endothelial activation via enhancing MCPIP1 expression. Cell Signal 2018; 50:1-8. [PMID: 29913212 DOI: 10.1016/j.cellsig.2018.05.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 05/09/2018] [Accepted: 05/23/2018] [Indexed: 01/29/2023]
Abstract
Mucosa associated lymphoid tissue lymphoma translocation protein 1 (MALT1) is not only an intracellular signaling scaffold protein but also a paracaspase that plays a key role in the signal transduction and cellular activation of lymphocytes and macrophages. However, its role in endothelial cells remains unknown. Here we report that pharmacological inhibition of MALT1 protease activity strongly suppresses endothelial activation via enhancing MCPIP1 expression. Treatment with MALT1 protease inhibitors selectively inhibited TNFα-induced VCAM-1 expression in HUVECs and LPS-induced VCAM-1 expression in mice. In addition, Inhibition of MALT1 protease activity also significantly inhibited TNFα-induced adhesion of THP-1 monocytic cells to HUVECs. To explore the mechanisms, MALT1 inhibitors does not affect the activation of NF-κB signaling pathway in HUVEC. However, they can stabilize MCPIP1 protein and significantly enhance MCPIP1 protein level in endothelial cells. These results suggest that MALT1 paracaspase also targets MCPIP1 and degrade MCPIP1 protein in endothelial cells similar as it does in immune cells. Taken together, the study suggest inhibition of MALT1 protease activity may represent a new strategy for prevention/therapy of vascular inflammatory diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Yong Li
- Department of Biomedical Science and Shock/Trauma Research Center, School of Medicine, University of Missouri-Kansas City, 2411 Holmes Street, Kansas City, MO 64108, USA; Institute of Translational Medicine, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi 330031, PR China
| | - Shengping Huang
- Department of Biomedical Science and Shock/Trauma Research Center, School of Medicine, University of Missouri-Kansas City, 2411 Holmes Street, Kansas City, MO 64108, USA
| | - Xuan Huang
- Department of Biomedical Science and Shock/Trauma Research Center, School of Medicine, University of Missouri-Kansas City, 2411 Holmes Street, Kansas City, MO 64108, USA; Institute of Translational Medicine, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi 330031, PR China
| | - Xiuzhen Li
- Department of Biomedical Science and Shock/Trauma Research Center, School of Medicine, University of Missouri-Kansas City, 2411 Holmes Street, Kansas City, MO 64108, USA
| | - Adrian Falcon
- Department of Biomedical Science and Shock/Trauma Research Center, School of Medicine, University of Missouri-Kansas City, 2411 Holmes Street, Kansas City, MO 64108, USA
| | - Adele Soutar
- Department of Biomedical Science and Shock/Trauma Research Center, School of Medicine, University of Missouri-Kansas City, 2411 Holmes Street, Kansas City, MO 64108, USA
| | - Frederic Bornancin
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, Switzerland
| | - Zhisheng Jiang
- Institute of Cardiovascular Diseases, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, China
| | - Hong-Bo Xin
- Institute of Translational Medicine, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi 330031, PR China
| | - Mingui Fu
- Department of Biomedical Science and Shock/Trauma Research Center, School of Medicine, University of Missouri-Kansas City, 2411 Holmes Street, Kansas City, MO 64108, USA.
| |
Collapse
|
234
|
Huang A, Patel S, McAlpine CS, Werstuck GH. The Role of Endoplasmic Reticulum Stress-Glycogen Synthase Kinase-3 Signaling in Atherogenesis. Int J Mol Sci 2018; 19:ijms19061607. [PMID: 29848965 PMCID: PMC6032052 DOI: 10.3390/ijms19061607] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 05/25/2018] [Accepted: 05/28/2018] [Indexed: 01/31/2023] Open
Abstract
Cardiovascular disease (CVD) is the number one cause of global mortality and atherosclerosis is the underlying cause of most CVD. However, the molecular mechanisms by which cardiovascular risk factors promote the development of atherosclerosis are not well understood. The development of new efficient therapies to directly block or slow disease progression will require a better understanding of these mechanisms. Accumulating evidence supports a role for endoplasmic reticulum (ER) stress in all stages of the developing atherosclerotic lesion however, it was not clear how ER stress may contribute to disease progression. Recent findings have shown that ER stress signaling through glycogen synthase kinase (GSK)-3α may significantly contribute to macrophage lipid accumulation, inflammatory cytokine production and M1macrophage polarization. In this review we summarize our knowledge of the potential role of ER stress-GSK3 signaling in the development and progression of atherosclerosis as well as the possible therapeutic implications of this pathway.
Collapse
Affiliation(s)
- Aric Huang
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON L9L 2X2, Canada.
| | - Sarvatit Patel
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON L9L 2X2, Canada.
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4L8, Canada.
| | - Cameron S McAlpine
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON L9L 2X2, Canada.
| | - Geoff H Werstuck
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON L9L 2X2, Canada.
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4L8, Canada.
- Department of Medicine, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada.
| |
Collapse
|
235
|
Gordts PLSM, Esko JD. The heparan sulfate proteoglycan grip on hyperlipidemia and atherosclerosis. Matrix Biol 2018; 71-72:262-282. [PMID: 29803939 DOI: 10.1016/j.matbio.2018.05.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 12/20/2022]
Abstract
Heparan sulfate proteoglycans are found at the cell surface and in the extracellular matrix, where they interact with a plethora of proteins involved in lipid homeostasis and inflammation. Over the last decade, new insights have emerged regarding the mechanism and biological significance of these interactions in the context of cardiovascular disease. The majority of cardiovascular disease-related deaths are caused by complications of atherosclerosis, a disease that results in narrowing of the arterial lumen, thereby reducing blood flow to critical levels in vital organs, such as the heart and brain. Here, we discuss novel insights into how heparan sulfate proteoglycans modulate risk factors such as hyperlipidemia and inflammation that drive the initiation and progression of atherosclerotic plaques to their clinical critical endpoint.
Collapse
Affiliation(s)
- Philip L S M Gordts
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, USA; Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA.
| | - Jeffrey D Esko
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
236
|
Srivastava RAK, Cornicelli JA, Markham B, Bisgaier CL. Gemcabene, a First-in-Class Hypolipidemic Small Molecule in Clinical Development, Attenuates Osteoarthritis and Pain in Animal Models of Arthritis and Pain. Front Pharmacol 2018; 9:471. [PMID: 29867478 PMCID: PMC5958179 DOI: 10.3389/fphar.2018.00471] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 04/23/2018] [Indexed: 01/23/2023] Open
Abstract
Our clinical studies have demonstrated that gemcabene, a small molecule in late-stage clinical development, lowers pro-inflammatory acute-phase protein, C-reactive protein (CRP). This observation was further confirmed in a cell-based study showing inhibition of cytokine-induced CRP production. Based on these observations, in the present study, we tested the hypothesis that gemcabene may possess anti-inflammatory activities in animal models of inflammatory disease. Efficacy of gemcabene was investigated in rat models of carrageenan-induced thermal hyperalgesia (CITH), monosodium iodoacetate (MIA)-induced osteoarthritis (OA), and IL-6/IL-6sR-induced inflammation. We also evaluated efficacy of gemcabene in collagen antibody-induced joint swelling and arthritis in BALB/c mice. In CITH rat model, gemcabene administration attenuated paw withdrawal latency (60% at 30 mg/kg/d and 97% at 100 mg/kg/d) and showed improvement in joint swelling (-50% at 30 mg/kg/d) in MIA model of OA. These findings were further corroborated by IL-6/IL-6sR knee injection model in rat, showing 63 and 71% reduction in hind paw weight distribution at 10 and 30 mg/kg/d doses, respectively. In mouse model of monoclonal antibody-induced arthritis, a dose-dependent attenuation of joint swelling was observed. These results demonstrate that the anti-inflammatory activity of gemcabene previously observed in cell-based and in clinical studies also occurred in animal models of inflammation-induced arthritis and hyperalgesia. Thus, in addition to hypolipidemic efficacy, the anti-inflammatory activity of gemcabene may have additional benefits to patients with elevated vascular inflammation.
Collapse
|
237
|
Protective Role of Antioxidant Huskless Barley Extracts on TNF- α-Induced Endothelial Dysfunction in Human Vascular Endothelial Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3846029. [PMID: 29861828 PMCID: PMC5971280 DOI: 10.1155/2018/3846029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/17/2018] [Accepted: 03/27/2018] [Indexed: 12/01/2022]
Abstract
Oxidative stress and inflammation are considered as two key factors that contribute to the development of atherosclerosis. This study was to investigate the antioxidant capacity of huskless barley and to explore its protective functions through the regulation of the antioxidant defense and inflammatory response in human umbilical vein endothelial cells (HUVEC). The oxygen radical absorbance capacity (ORAC), ferric-reducing antioxidant power (FRAP), and 2,2-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) diammonium salt (ABTS) scavenging capacity of water and alkali extracts of the polysaccharides from nine huskless barley varieties were investigated in vitro. The antioxidant properties of the alkaline extracts were more pronounced than those of the water extracts. The results from the cell model showed that pretreatment of HUVEC with the water or alkaline extracts of the polysaccharides from the huskless barley cultivars QHH and NLGL decreased the levels of reactive oxygen species (ROS), monocyte chemotactic protein 1 (MCP-1), and vascular cell adhesion molecule 1 (VCAM-1) but increased the level of superoxide dismutase (SOD) and maintained cell viability. Huskless barley polysaccharide extracts exhibited the vasodilatory effect of inhibiting angiotensin-converting enzyme (ACE) production. These discoveries revealed the potent protective functions of barley in oxidative damage and a potential role for barley in preventing chronic inflammation in cardiovascular diseases.
Collapse
|
238
|
Abstract
Over the past decade, studies have repeatedly found single-nucleotide polymorphisms located in the collagen ( COL) 4A1 and COL4A2 genes to be associated with cardiovascular disease (CVD), and the 13q34 locus harboring these genes is one of ~160 genome-wide significant risk loci for coronary artery disease. COL4A1 and COL4A2 encode the α1- and α2-chains of collagen type IV, a major component of basement membranes in various tissues including arteries. Despite the growing body of evidence indicating a role for collagen type IV in CVD, remarkably few studies have aimed to directly investigate such a role. The purpose of this review is to summarize the clinical reports linking 13q34 to coronary artery disease, atherosclerosis, and artery stiffening and to assemble the scattered pieces of evidence from experimental studies based on vascular cells and tissue collectively supporting a role for collagen type IV in atherosclerosis and other macrovascular disease conditions.
Collapse
Affiliation(s)
- L B Steffensen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital , Odense , Denmark.,Centre for Individualized Medicine in Arterial Diseases, Odense University Hospital , Odense , Denmark.,Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark , Odense , Denmark
| | - L M Rasmussen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital , Odense , Denmark.,Centre for Individualized Medicine in Arterial Diseases, Odense University Hospital , Odense , Denmark
| |
Collapse
|
239
|
Kumar KS, Sabu V, Sindhu G, Rauf AA, Helen A. Isolation, identification and characterization of apigenin from Justicia gendarussa and its anti-inflammatory activity. Int Immunopharmacol 2018; 59:157-167. [PMID: 29655057 DOI: 10.1016/j.intimp.2018.04.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 03/23/2018] [Accepted: 04/04/2018] [Indexed: 12/26/2022]
Abstract
Inflammatory responses during chronic diseases such as atherosclerosis, cancer etc., are harmful to host organisms. Generally NSAIDs are used to treat against these severe conditions but due to its adverse effects studies are going on with medicinal plants, since they are rich in bioactive compounds. Justicia gendarussa is one such plant which has been used as a remedial measure for treating inflammatory diseases since ancient time. Thus the present study involved in the isolation, characterization and identification of apigenin (flavonoid) from this plant and to elucidate its molecular mechanism against inflammation via TLR-NF-κB signaling pathway using ox-LDL induced hPBMCs in in vitro model. Methanolic extract was used for the isolation process and results showed that the F6 fraction collected from ethyl acetate through column chromatography showed 89% paw edema inhibition at a dose of 10 mg/kg in carrageenan induced rats. Purification of F6 by TLC with toluene: chloroform: acetone (8:5:7) and further characterization by 1HNMR indicated the presence of bioactive compound, apigenin. In vitro studies revealed that pretreatment of ox-LDL induced hPBMCs with apigenin (25 μM) significantly (P < 0.05) reduced the levels of TLR4, MyD88, TRIF, TRAF6, NF-κB, COX-2, PGE2, IL-1β and TNF-α responsible for generating inflammation and elevated the level of anti-inflammatory cytokine, IL-10. These results indicate the therapeutic efficacy of bioflavonoid apigenin which was isolated from Justicia gendarussa against ox-LDL induced inflammation. Therefore apigenin can be treated as a suitable therapeutic agent against inflammatory diseases.
Collapse
Affiliation(s)
- K S Kumar
- Department of Biochemistry, University of Kerala, Kariavattom Campus, Thiruvananthapuram 695581, Kerala, India
| | - V Sabu
- Department of Biochemistry, University of Kerala, Kariavattom Campus, Thiruvananthapuram 695581, Kerala, India
| | - G Sindhu
- Department of Biochemistry, University of Kerala, Kariavattom Campus, Thiruvananthapuram 695581, Kerala, India
| | - A A Rauf
- Department of Biochemistry, University of Kerala, Kariavattom Campus, Thiruvananthapuram 695581, Kerala, India
| | - A Helen
- Department of Biochemistry, University of Kerala, Kariavattom Campus, Thiruvananthapuram 695581, Kerala, India.
| |
Collapse
|
240
|
Srivastava RAK, Cornicelli JA, Markham B, Bisgaier CL. Gemcabene, a first-in-class lipid-lowering agent in late-stage development, down-regulates acute-phase C-reactive protein via C/EBP-δ-mediated transcriptional mechanism. Mol Cell Biochem 2018; 449:167-183. [PMID: 29644527 PMCID: PMC6223808 DOI: 10.1007/s11010-018-3353-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 04/07/2018] [Indexed: 12/11/2022]
Abstract
Inflammation plays a key role in setting the stage leading to atherosclerosis progression, and high-sensitivity C-reactive protein (CRP) has been recognized as a predictor of cardiovascular risk. As a monotherapy and in combination with statins, gemcabene markedly reduced CRP in humans. Present investigation was undertaken to understand the mechanism of CRP reduction. In human hepatoma cells, gemcabene inhibited IL-6 plus IL-1β-induced CRP production in a concentration-dependent manner, reaching 70% inhibition at 2 mM. In TNF-α-stimulated primary human coronary artery endothelial cells, both CRP and IL-6 productions were reduced by 70% at 2 mM gemcabene concentration. To investigate the mechanism of gemcabene-mediated reduction of CRP, transfection studies were performed with human CRP regulatory sequences in luciferase/β-gal system that showed 25-fold increase in IL-6- and IL-6 plus IL-1β-stimulated CRP transcription. Luciferase activity was reduced by 50% by gemcabene, suggesting transcriptional down-regulation of CRP. Site-directed mutagenesis of human CRP promoter revealed that the overlapping downstream C/EBP and NF-κB binding sites are important for gemcabene-mediated CRP transcription. Gel shift assays identified the transcription factor that binds to the downstream CRP promoter as C/EBP-δ. In conclusion, gemcabene decreases CRP by C/EBP-δ and NF-κB-mediated transcriptional mechanism and suppresses IL-6 and IL-1β-induced CRP production.
Collapse
|
241
|
Guo L, Akahori H, Harari E, Smith SL, Polavarapu R, Karmali V, Otsuka F, Gannon RL, Braumann RE, Dickinson MH, Gupta A, Jenkins AL, Lipinski MJ, Kim J, Chhour P, de Vries PS, Jinnouchi H, Kutys R, Mori H, Kutyna MD, Torii S, Sakamoto A, Choi CU, Cheng Q, Grove ML, Sawan MA, Zhang Y, Cao Y, Kolodgie FD, Cormode DP, Arking DE, Boerwinkle E, Morrison AC, Erdmann J, Sotoodehnia N, Virmani R, Finn AV. CD163+ macrophages promote angiogenesis and vascular permeability accompanied by inflammation in atherosclerosis. J Clin Invest 2018; 128:1106-1124. [PMID: 29457790 PMCID: PMC5824873 DOI: 10.1172/jci93025] [Citation(s) in RCA: 243] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 01/02/2018] [Indexed: 12/26/2022] Open
Abstract
Intake of hemoglobin by the hemoglobin-haptoglobin receptor CD163 leads to a distinct alternative non-foam cell antiinflammatory macrophage phenotype that was previously considered atheroprotective. Here, we reveal an unexpected but important pathogenic role for these macrophages in atherosclerosis. Using human atherosclerotic samples, cultured cells, and a mouse model of advanced atherosclerosis, we investigated the role of intraplaque hemorrhage on macrophage function with respect to angiogenesis, vascular permeability, inflammation, and plaque progression. In human atherosclerotic lesions, CD163+ macrophages were associated with plaque progression, microvascularity, and a high level of HIF1α and VEGF-A expression. We observed irregular vascular endothelial cadherin in intraplaque microvessels surrounded by CD163+ macrophages. Within these cells, activation of HIF1α via inhibition of prolyl hydroxylases promoted VEGF-mediated increases in intraplaque angiogenesis, vascular permeability, and inflammatory cell recruitment. CD163+ macrophages increased intraplaque endothelial VCAM expression and plaque inflammation. Subjects with homozygous minor alleles of the SNP rs7136716 had elevated microvessel density, increased expression of CD163 in ruptured coronary plaques, and a higher risk of myocardial infarction and coronary heart disease in population cohorts. Thus, our findings highlight a nonlipid-driven mechanism by which alternative macrophages promote plaque angiogenesis, leakiness, inflammation, and progression via the CD163/HIF1α/VEGF-A pathway.
Collapse
MESH Headings
- Adult
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Atherosclerosis/metabolism
- Coronary Disease/metabolism
- Coronary Vessels/metabolism
- Disease Progression
- Female
- Hemoglobins/metabolism
- Humans
- Inflammation/metabolism
- Macrophages/cytology
- Macrophages/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Middle Aged
- Myocardial Infarction/metabolism
- Neovascularization, Pathologic
- Oxidative Stress
- Permeability
- Phenotype
- Polymorphism, Single Nucleotide
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Liang Guo
- CVPath Institute, Gaithersburg, Maryland, USA
| | - Hirokuni Akahori
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | | | - Rohini Polavarapu
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Vinit Karmali
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | | | | | | | - Anuj Gupta
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Audrey L. Jenkins
- MedStar Heart and Vascular Institute and MedStar Health Research Institute, MedStar Washington Hospital Center, Washington, DC, USA
| | - Michael J. Lipinski
- MedStar Heart and Vascular Institute and MedStar Health Research Institute, MedStar Washington Hospital Center, Washington, DC, USA
| | - Johoon Kim
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peter Chhour
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paul S. de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | | | | | | | | | - Sho Torii
- CVPath Institute, Gaithersburg, Maryland, USA
| | | | - Cheol Ung Choi
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Qi Cheng
- CVPath Institute, Gaithersburg, Maryland, USA
| | - Megan L. Grove
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Mariem A. Sawan
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yin Zhang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | | | - David P. Cormode
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dan E. Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | - Alanna C. Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, and German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Germany
| | - Nona Sotoodehnia
- Division of Cardiology, Department of Medicine and Epidemiology, Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, USA
| | | | - Aloke V. Finn
- CVPath Institute, Gaithersburg, Maryland, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
242
|
Woodside DG, Tanifum EA, Ghaghada KB, Biediger RJ, Caivano AR, Starosolski ZA, Khounlo S, Bhayana S, Abbasi S, Craft JW, Maxwell DS, Patel C, Stupin IV, Bakthavatsalam D, Market RV, Willerson JT, Dixon RAF, Vanderslice P, Annapragada AV. Magnetic Resonance Imaging of Atherosclerotic Plaque at Clinically Relevant Field Strengths (1T) by Targeting the Integrin α4β1. Sci Rep 2018; 8:3733. [PMID: 29487319 PMCID: PMC5829217 DOI: 10.1038/s41598-018-21893-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 02/12/2018] [Indexed: 02/07/2023] Open
Abstract
Inflammation drives the degradation of atherosclerotic plaque, yet there are no non-invasive techniques available for imaging overall inflammation in atherosclerotic plaques, especially in the coronary arteries. To address this, we have developed a clinically relevant system to image overall inflammatory cell burden in plaque. Here, we describe a targeted contrast agent (THI0567-targeted liposomal-Gd) that is suitable for magnetic resonance (MR) imaging and binds with high affinity and selectivity to the integrin α4β1(very late antigen-4, VLA-4), a key integrin involved in recruiting inflammatory cells to atherosclerotic plaques. This liposomal contrast agent has a high T1 relaxivity (~2 × 105 mM-1s-1 on a particle basis) resulting in the ability to image liposomes at a clinically relevant MR field strength. We were able to visualize atherosclerotic plaques in various regions of the aorta in atherosclerosis-prone ApoE-/- mice on a 1 Tesla small animal MRI scanner. These enhanced signals corresponded to the accumulation of monocyte/macrophages in the subendothelial layer of atherosclerotic plaques in vivo, whereas non-targeted liposomal nanoparticles did not demonstrate comparable signal enhancement. An inflammatory cell-targeted method that has the specificity and sensitivity to measure the inflammatory burden of a plaque could be used to noninvasively identify patients at risk of an acute ischemic event.
Collapse
Affiliation(s)
- Darren G Woodside
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA.
| | - Eric A Tanifum
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Ketan B Ghaghada
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Ronald J Biediger
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Amy R Caivano
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Zbigniew A Starosolski
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Sayadeth Khounlo
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Saakshi Bhayana
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Shahrzad Abbasi
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - John W Craft
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA.,Department of Biology and Chemistry, University of Houston, 4800 Calhoun Road, Houston, Texas, 77004, USA
| | - David S Maxwell
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas, 77030, USA.,Department of Institutional Analytics and Informatics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Chandreshkumar Patel
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Igor V Stupin
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | | | - Robert V Market
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - James T Willerson
- Division of Cardiology Research, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Richard A F Dixon
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Peter Vanderslice
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Ananth V Annapragada
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA.
| |
Collapse
|
243
|
Piao S, Lee JW, Nagar H, Jung SB, Choi S, Kim S, Lee I, Kim SM, Shin N, Lee YR, Lee SD, Park JB, Irani K, Won M, Hur GM, Jeon BH, Kim DW, Kim CS. CR6 interacting factor 1 deficiency promotes endothelial inflammation by SIRT1 downregulation. PLoS One 2018; 13:e0192693. [PMID: 29474366 PMCID: PMC5825004 DOI: 10.1371/journal.pone.0192693] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 01/29/2018] [Indexed: 11/29/2022] Open
Abstract
Aims CR6 interacting factor 1 (CRIF1) deficiency impairs mitochondrial oxidative phosphorylation complexes, contributing to increased mitochondrial and cellular reactive oxygen species (ROS) production. CRIF1 downregulation has also been revealed to decrease sirtuin 1 (SIRT1) expression and impair vascular function. Inhibition of SIRT1 disturbs oxidative energy metabolism and stimulates nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)-induced inflammation. Therefore, we hypothesized that both CRIF1 deficiency-induced mitochondrial ROS production and SIRT1 reduction play stimulatory roles in vascular inflammation. Methods and results Plasma levels and mRNA expression of proinflammatory cytokines (tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6) were markedly elevated in endothelium-specific CRIF1-knockout mice and CRIF1-silenced endothelial cells, respectively. Moreover, CRIF1 deficiency-induced vascular adhesion molecule-1 (VCAM-1) expression was consistently attenuated by the antioxidant N-acetyl-cysteine and NF-κB inhibitor (BAY11). We next showed that siRNA-mediated CRIF1 downregulation markedly activated NF-κB. SIRT1 overexpression not only rescued CRIF1 deficiency-induced NF-κB activation but also decreased inflammatory cytokines (TNF-α, IL-1β, and IL-6) and VCAM-1 expression levels in endothelial cells. Conclusions These results strongly suggest that CRIF1 deficiency promotes endothelial cell inflammation by increasing VCAM-1 expression, elevating inflammatory cytokines levels, and activating the transcription factor NF-κB, all of which were inhibited by SIRT1 overexpression.
Collapse
Affiliation(s)
- Shuyu Piao
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jun Wan Lee
- Emergency ICU, Regional Emergency Center, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Harsha Nagar
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Saet-byel Jung
- Department of Endocrinology, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Sujeong Choi
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Seonhee Kim
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Ikjun Lee
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Sung-min Kim
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Nara Shin
- Department of Anatomy & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Yu Ran Lee
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Sang Do Lee
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jin Bong Park
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Kaikobad Irani
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA United States of America
| | - Minho Won
- Department of Pharmacology, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Gang Min Hur
- Department of Pharmacology, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Byeong Hwa Jeon
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Dong Woon Kim
- Department of Anatomy & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Cuk-Seong Kim
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
- * E-mail:
| |
Collapse
|
244
|
Ismail SM, Sundar UM, Hui CK, Aminuddin A, Ugusman A. Piper sarmentosum attenuates TNF-α-induced VCAM-1 and ICAM-1 expression in human umbilical vein endothelial cells. J Taibah Univ Med Sci 2018; 13:225-231. [PMID: 31435328 PMCID: PMC6694970 DOI: 10.1016/j.jtumed.2018.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/22/2018] [Accepted: 01/22/2018] [Indexed: 01/24/2023] Open
Abstract
Objectives Inflammation plays a key role in the pathogenesis of atherosclerosis. Piper sarmentosum is an herb with antioxidant and anti-atherosclerotic activities. The aim of this study was to evaluate the anti-inflammatory properties of an aqueous extract of P. sarmentosum (AEPS) in human umbilical vein endothelial cells (HUVECs). Methods HUVECs were divided into six groups: control, treatment with 10 ng/ml TNF-α, and co-treatment of 10 ng/ml TNF-α with four different concentrations of AEPS (100, 150, 250, and 300 μg/ml) for 24 h. Subsequently, vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) protein expression, U937 monocyte cells adhesion, and nuclear factor-kappaB (NF-κB) p65 expression in HUVECs were measured. Results Treatment of TNF-α-stimulated HUVECs with AEPS at different concentrations resulted in decreased VCAM-1 and ICAM-1 protein expression in a dose-dependent manner. Furthermore, AEPS also inhibited TNF-α-stimulated U937 monocyte cells adhesion to HUVECs. In addition, AEPS reduced TNF-α-induced NF-κB p65 expression in a dose-dependent manner. Conclusions The results indicated that AEPS suppressed TNF-α-induced VCAM-1 and ICAM-1 expression NF-κB signaling. Piper sarmentosum attenuates VCAM-1 and ICAM-1 expression in TNF-α-induced HUVEC. P. sarmentosum inhibits TNF-α-stimulated monocytes adhesion to HUVEC. P. sarmentosum reduces TNF-α-induced NF-κB p65 expression in HUVEC.
Collapse
Affiliation(s)
| | | | | | | | - Azizah Ugusman
- Corresponding address: Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latiff, 56000 Cheras, Kuala Lumpur, Malaysia.
| |
Collapse
|
245
|
Farhangi MA, Najafi M. Dietary inflammatory index: a potent association with cardiovascular risk factors among patients candidate for coronary artery bypass grafting (CABG) surgery. Nutr J 2018; 17:20. [PMID: 29439738 PMCID: PMC5812038 DOI: 10.1186/s12937-018-0325-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 01/15/2018] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Recently, the clinical importance of dietary inflammatory index (DII) in predicting the inflammatory potential of diet and its role in pathogenesis of several chronic disease including some types of cancers, osteoporosis, cardiovascular disease and renal disease has been proposed. However, its association with the disease severity and progression and cardio-metabolic risk factors among patients candidate for coronary artery bypass graft surgery (CABG) has not been evaluated yet. In the current study, the association of DII with cardiovascular risk factors among patients candidate for CABG has been investigated. METHODS In the current cross-sectional study, 454 patients aged 35-80 years as candidates of CABG and hospitalized in Tehran Heart Center were enrolled. Anthropometric and demographic characteristics were obtained from all of the participants. Dietary intakes were evaluated with a semi-quantitative food-frequency questionnaire (FFQ) and DII was calculated. Biochemical parameters including hemoglobin (Hb) A1C, serum lipids, albumin, creatinine and high-sensitive C-reactive protein (hs-CRP), lipoprotein (a), creatinine, blood urea nitrogen (BUN), albumin and hematocrit (HCT) were also assessed by commercial laboratory methods. Left ventricular ejection- fraction, number of diseased vessels, New York Heart Association (NYHA) functional class and the European system for cardiac operative risk evaluation (EuroSCORE) were estimated for clinical assessment. One way analysis of variance and chi square tests were used for comparison of demographic parameters between groups. Beta estimates and 95% confidence intervals (CI) for the association between DII and clinical parameters were estimated using linear regression adjusted for the confounders. RESULTS According to our findings, high DII scores were associated with higher age, lower body mass index (BMI), higher prevalence of diabetes and myocardial infarction (MI) and lower educational attainment (P < 0.05). Male patients in 4th and 3rd quartile of DII had significantly higher total cholesterol (TC), triglyceride (TG), albumin, creatinine, BUN and hs-CRP concentrations and lower high density lipoprotein cholesterol (HDL) concentrations compared with male patients in lower quartiles (P < 0.05). While in female patients, only lipoprotein (a) concentrations and hematocrit (HCT) percentage in the 4th and 2nd quartile were significantly higher than lower quartiles. EuroSCORE was also significantly higher in top quartiles of DII (P = 0.006). CONCLUSION As shown in our results, DII was in a positive association with several cardiovascular risk factors. The higher inflammatory potential of diet denoted higher values of serum lipids, CRP and kidney function tests and higher EuroSCORE as a predictor of post-operative mortality. Therefore, DII can be demonstrated as a target of nutritional interventions for ameliorating the CVD risk factors among patients candidate for CABG especially in male patients.
Collapse
Affiliation(s)
| | - Mahdi Najafi
- Department of Research, Tehran Heart Center, Tehran University of Medical Sciences, North Karegar Street, Tehran, 1411713138 Iran
- Cardiac Outcome Research and Education (CORE), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
246
|
Kim MH, Kim CE, Kim SW. Rosuvastatin inhibits high glucose-stimulated upregulation of VCAM-1 via the MAPK-signalling pathway in endothelial cells. Acta Cardiol 2018; 73:13-18. [PMID: 28885098 DOI: 10.1080/00015385.2017.1321908] [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: 01/21/2023]
Abstract
OBJECTIVE The aim of this study is to investigate the molecular mechanisms and effect of rosuvastatin on adhesion molecule induction in human endothelial cells under high-glucose conditions (HG). METHODS AND RESULTS The effects of rosuvastatin on vascular cell adhesion molecule (VCAM)-1 production and pERK phosphorylation were measured in HG-induced human umbilical vein endothelial cells (HUVECs) with inhibitors targeting the mitogen-activated protein kinase (MAPK) signal pathway. HG increased levels of VCAM-1. Treatment with rosuvastatin inhibited VCAM-1 expression in a concentration- and time-dependent manner. In addition, we investigated the effects of rosuvastatin on the extracellular signal-regulated kinase (ERK) 1/2 signal pathway. Rosuvastatin completely inhibited HG-induced phosphorylation of ERK. ERK/MAPK inhibitors completely prevented the VCAM-1 inhibition effect of rosuvastatin under HG condition. CONCLUSIONS This study demonstrated that rosuvastatin suppresses HG-induced VCAM-1 production via the MAPK signalling pathway, playing a role in the suppression of atherosclerosis.
Collapse
Affiliation(s)
- Moo Hyun Kim
- Department of Cardiology, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Chae-Eun Kim
- Department of Cardiology, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Sung-Whan Kim
- Department of Medicine, Catholic Kwandong University College of Medicine, Gangneung, Republic of Korea
| |
Collapse
|
247
|
Russo L, Muturi HT, Ghadieh HE, Wisniewski AM, Morgan EE, Quadri SS, Landesberg GP, Siragy HM, Vazquez G, Scalia R, Gupta R, Najjar SM. Liver-specific rescuing of CEACAM1 reverses endothelial and cardiovascular abnormalities in male mice with null deletion of Ceacam1 gene. Mol Metab 2018; 9:98-113. [PMID: 29396368 PMCID: PMC5870095 DOI: 10.1016/j.molmet.2018.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/07/2018] [Accepted: 01/14/2018] [Indexed: 12/22/2022] Open
Abstract
Objective Mice with global null mutation of Ceacam1 (Cc1−/−), display impairment of insulin clearance that causes hyperinsulinemia followed by insulin resistance, elevated hepatic de novo lipogenesis, and visceral obesity. In addition, they manifest abnormal vascular permeability and elevated blood pressure. Liver-specific rescuing of Ceacam1 reversed all of the metabolic abnormalities in Cc1−/−liver+ mice. The current study examined whether Cc1−/− male mice develop endothelial and cardiac dysfunction and whether this relates to the metabolic abnormalities caused by defective insulin extraction. Methods and results Myography studies showed reduction of agonist-stimulated nitric oxide production in resistance arterioles in Cc1−/−, but not Cc1−/−liver+ mice. Liver-based rescuing of CEACAM1 also attenuated the abnormal endothelial adhesiveness to circulating leukocytes in parallel to reducing plasma endothelin-1 and recovering plasma nitric oxide levels. Echocardiography studies revealed increased septal wall thickness, cardiac hypertrophy and reduced cardiac performance in Cc1−/−, but not Cc1−/−xliver+ mice. Insulin signaling experiments indicated compromised IRS1/Akt/eNOS pathway leading to lower nitric oxide level, and activated Shc/MAPK pathway leading to more endothelin-1 production in the aortae and hearts of Cc1−/−, but not Cc1−/−xliver+ mice. The increase in the ratio of endothelin-1 receptor A/B indicated an imbalance in the vasomotor activity of Cc1−/− mice, which was normalized in Cc1−/−xliver+ mice. Conclusions The data underscore a critical role for impaired CEACAM1-dependent hepatic insulin clearance pathways and resulting hyperinsulinemia and lipid accumulation in aortae and heart in regulating the cardiovascular function. Mice with global deletion of Ceacam1 gene (Cc1−/−) manifest endothelial dysfunction which is reversed by liver-specific rescuing of CEACAM1. Restoring CEACAM1 expression in the liver reversed cardiac hypertrophy and rescued cardiac performance. Hyperinsulinemia emerging from impaired insulin clearance regulates endothelial and cardiovascular functions.
Collapse
Affiliation(s)
- Lucia Russo
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Harrison T Muturi
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Hilda E Ghadieh
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Alexander M Wisniewski
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Eric E Morgan
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Syed S Quadri
- Department of Endocrinology and Metabolism, College of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Gavin P Landesberg
- Department of Physiology and Cardiovascular Research Center, School of Medicine, Temple University, Philadelphia, PA, USA
| | - Helmy M Siragy
- Department of Endocrinology and Metabolism, College of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Guillermo Vazquez
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Rosario Scalia
- Department of Physiology and Cardiovascular Research Center, School of Medicine, Temple University, Philadelphia, PA, USA
| | - Rajesh Gupta
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Sonia M Najjar
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
248
|
Zhang S, Zou J, Li P, Zheng X, Feng D. Curcumin Protects against Atherosclerosis in Apolipoprotein E-Knockout Mice by Inhibiting Toll-like Receptor 4 Expression. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:449-456. [PMID: 29224353 DOI: 10.1021/acs.jafc.7b04260] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Toll-like receptor 4 (TLR4) has been reported to play a critical role in the pathogenesis of atherosclerosis, the current study aimed to investigate whether curcumin suppresses atherosclerosis development in ApoE-knockout (ApoE-/-) mice by inhibiting TLR4 expression. ApoE-/- mice were fed a high-fat diet supplemented with or without curcumin (0.1% w/w) for 16 weeks. Curcumin supplementation significantly reduced TLR4 expression and macrophage infiltration in atherosclerotic plaques. Curcumin also reduced aortic interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) expression, nuclear factor-κB (NF-κB) activity, and plasma IL-1β, TNF-α, soluble VCAM-1 and ICAM-1 levels. In addition, aortic sinus sections revealed that curcumin treatment reduced the extent of atherosclerotic lesions and inhibited atherosclerosis development. In vitro, curcumin inhibited NF-κB activation in macrophages and reduced TLR4 expression induced by lipopolysaccharide. Our results indicate that curcumin protects against atherosclerosis at least partially by inhibiting TLR4 expression and its related inflammatory reaction.
Collapse
Affiliation(s)
- Shanshan Zhang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Preventive Medicine, School of Public Health, Sun Yat-sen University , Guangzhou 510080, China
| | - Jun Zou
- Department of Cardiology, Affiliated NanHai Hospital of Southern Medical University , Guangzhou 528200, China
| | - Peiyang Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Preventive Medicine, School of Public Health, Sun Yat-sen University , Guangzhou 510080, China
| | - Xiumei Zheng
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Preventive Medicine, School of Public Health, Sun Yat-sen University , Guangzhou 510080, China
| | - Dan Feng
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Preventive Medicine, School of Public Health, Sun Yat-sen University , Guangzhou 510080, China
| |
Collapse
|
249
|
Guan L, Geng X, Shen J, Yip J, Li F, Du H, Ji Z, Ding Y. PM2.5 inhalation induces intracranial atherosclerosis which may be ameliorated by omega 3 fatty acids. Oncotarget 2018; 9:3765-3778. [PMID: 29423081 PMCID: PMC5790498 DOI: 10.18632/oncotarget.23347] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 11/26/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Intracranial atherosclerosis (ICA) a major health problem. This study investigated whether inhalation of fine airborne particulate matters (PM2.5) causes ICA and whether omega-3 fatty acids (O3FA) attenuated the development of ICA. RESULTS Twelve but not 6 week exposure significantly increased triglycerides (TG) in normal chow diet (NCD), while PM2.5 enhanced all lipid profiles (TG, low density lipoprotein (LDL) and cholesterol (CHO)) after both 6 and 12-week exposure with high-cholesterol diet (HCD). PM2.5 exposure for 12 but not 6 weeks significantly induced middle cerebral artery (MCA) narrowing and thickening, in association with the enhanced expression of inflammatory cytokines, (interleukin 6 (IL-6), tumor necrosis factor alpha (TNF-α), monocyte chemoattractant protein-1 (MCP-1), interferon gamma (IFN-γ)), vascular cell adhesion molecule 1 (VCAM-1) and inducible nitric oxide synthase (iNOS). O3FA significantly attenuated vascular alterations, even without favorable changes in lipid profiles, in association with reduced expression of IL-6, TNF-α, MCP-1, IFN-γ, VCAM-1 and iNOS in brain vessels. CONCLUSIONS PM2.5 exposure for 12 weeks aggravates ICA in a dietary model (HCD + short-term L-NAME), which may be mediated by vascular inflammation. O3FA dietary supplementation prevents ICA development and inflammatory reaction in cerebral vessels. METHODS Adult Sprague-Dawly rats were under filtered air (FA) or PM2.5 exposure with NCD or HCD for 6 or 12 weeks. Half of the HCD rats were treated with O3FA (5 mg/kg/day) by gavage. A total of 600 mg NG-nitro-L-arginine methyl ester (L-NAME, 3 mg/mL) per rat was administered over two weeks as supplementation in the HCD group. Blood lipids, including LDL, CHO, TG and high density lipoprotein (HDL), were measured at 6 and 12 weeks. ICA was determined by lumen diameter and thickness of the MCA. Inflammatory markers, IL-6, TNF-α, MCP-1, IFN-γ, VCAM-1 and iNOS were assessed by real-time PCR for mRNA and Western blot for protein expression.
Collapse
Affiliation(s)
- Longfei Guan
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Tongzhou Qu, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Xiaokun Geng
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Tongzhou Qu, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Tongzhou Qu, China
| | - Jiamei Shen
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Tongzhou Qu, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - James Yip
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Fengwu Li
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Tongzhou Qu, China
| | - Huishan Du
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Tongzhou Qu, China
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Tongzhou Qu, China
| | - Zhili Ji
- Department of General Surgery, Beijing Luhe Hospital, Capital Medical University, Tongzhou Qu, China
| | - Yuchuan Ding
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Tongzhou Qu, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
250
|
Yu L, Yin M, Yang X, Lu M, Tang F, Wang H. Calpain inhibitor I attenuates atherosclerosis and inflammation in atherosclerotic rats through eNOS/NO/NF-κB pathway. Can J Physiol Pharmacol 2018; 96:60-67. [DOI: 10.1139/cjpp-2016-0652] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously reported that calpain, the Ca2+-sensitive cysteine protease, gets involved in atherogenesis. This study aimed to investigate the effects of calpain inhibitor I (CAI, 5 mg/kg per day) with or without NG-nitro-l-arginine-methyl ester (l-NAME) (100 mg/kg per day), the inhibitor of nitric oxide synthase (NOS), on atherosclerosis and inflammation in a rat model induced by high-cholesterol diet (HCD). The results demonstrated HCD increased protein expression of calpain-1 but not calpain-2 in aortic tissue. In addition, CAI reduced the thickness of atherosclerotic intima compared with HCD group, which was weakened by the l-NAME combination. CAI with or without l-NAME decreased the activity of calpain in the aorta. Also, CAI decreased the expressions of vascular cell adhesion molecule-1 (VCAM-1), intracellular cell adhesion molecule-1 (ICAM-1), and monocyte chemoattractant protein-1 (MCP-1) in the aorta at the levels of both mRNA and protein. Furthermore, CAI increased the activity and the protein expression of endothelial NOS (eNOS) accompanied by increased content of NO and downregulated the protein expression of nuclear factor κB (NF-κB) of the nucleus in the aorta. However, the abovementioned effects were at least partly cancelled by l-NAME except for the protein expression of eNOS. The results suggested that CAI attenuated atherosclerosis and inflammation through eNOS/NO/NF-κB pathway.
Collapse
Affiliation(s)
- Lan Yu
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou 121001, China
- Central Hospital of Yingkou Development Areas, Yingkou 115007, China
| | - Meihui Yin
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou 121001, China
| | - Xueyan Yang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou 121001, China
- Internal Medicine-Cardiovascular Departments, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Meili Lu
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou 121001, China
| | - Futian Tang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou 121001, China
| | - Hongxin Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou 121001, China
| |
Collapse
|