151
|
Kharaba ZJ, Buabeid MA, Alfoteih YA. Effectiveness of testosterone therapy in hypogonadal patients and its controversial adverse impact on the cardiovascular system. Crit Rev Toxicol 2020; 50:491-512. [PMID: 32689855 DOI: 10.1080/10408444.2020.1789944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Testosterone is the major male hormone produced by testicles which are directly associated with man's appearance and secondary sexual developments. Androgen deficiency starts when the male hormonal level falls from its normal range though, in youngsters, the deficiency occurs due to disruption of the normal functioning of pituitary, hypothalamus glands, and testes. Thus, testosterone replacement therapy was already known for the treatment of androgen deficiency with lesser risks of producing cardiovascular problems. Since from previous years, the treatment threshold in the form of testosterone replacement therapy has effectively increased to that extent that it was prescribed for those conditions which it was considered as inappropriate. However, there are some research studies and clinical trials available that proposed the higher risk of inducing cardiovascular disease with the use of testosterone replacement therapy. Thus under the light of these results, the FDA has published the report of the increased risk of cardiovascular disease with the increased use of testosterone replacement therapy. Nevertheless, there is not a single trial available or designed that could evaluate the risk of cardiovascular events with the use of testosterone replacement therapy. As a result, the use of testosterone still questioned the cardiovascular safety of this replacement therapy. Thus, this literature outlines the distribution pattern of disease by investigating the data and link between serum testosterone level and the cardiovascular disease, also the prescription data of testosterone replacement therapy patients and their tendency of inducing cardiovascular disease, meta-analysis and the trials regarding testosterone replacement therapy and its connection with the risks of causing cardiovascular disease and lastly, the possible effects of testosterone replacement therapy on the cardiovascular system. This study aims to evaluate the available evidence regarding the use of testosterone replacement therapy when choosing it as a treatment plan for their patients.
Collapse
Affiliation(s)
- Zelal Jaber Kharaba
- Department of Clinical Sciences, College of Pharmacy, Al-Ain University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Manal Ali Buabeid
- Department of Clinical Sciences, Ajman University, Ajman, United Arab Emirates
| | | |
Collapse
|
152
|
Chen LW, Tsai MC, Chern CY, Tsao TP, Lin FY, Chen SJ, Tsui PF, Liu YW, Lu HJ, Wu WL, Lin WS, Tsai CS, Lin CS. A chalcone derivative, 1m-6, exhibits atheroprotective effects by increasing cholesterol efflux and reducing inflammation-induced endothelial dysfunction. Br J Pharmacol 2020; 177:5375-5392. [PMID: 32579243 DOI: 10.1111/bph.15175] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 05/27/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Atherosclerosis, resulting from lipid dysregulation and vascular inflammation, causes atherosclerotic cardiovascular disease (ASCVD), which contributes to morbidity and mortality worldwide. Chalcone and its derivatives possess beneficial properties, including anti-inflammatory, antioxidant and antitumour activity with unknown cardioprotective effects. We aimed to develop an effective chalcone derivative with antiatherogenic potential. EXPERIMENTAL APPROACH Human THP-1 cells and HUVECs were used as in vitro models. Western blots and real-time PCRs were performed to quantify protein, mRNA and miRNA expressions. The cholesterol efflux capacity was assayed by 3 H labelling of cholesterol. LDL receptor knockout (Ldlr-/- ) mice fed a high-fat diet were used as an in vivo atherogenesis model. Haematoxylin and eosin and oil red O staining were used to analyse plaque formation. KEY RESULTS Using ATP-binding cassette transporter A1 (ABCA1) expression we identified the chalcone derivative, 1m-6, which enhances ABCA1 expression and promotes cholesterol efflux in THP-1 macrophages. Moreover, 1m-6 stabilizes ABCA1 mRNA and suppresses the expression of potential ABCA1-regulating miRNAs through nuclear factor erythroid 2-related factor 2 (Nrf2)/haem oxygenase-1 (HO-1) signalling. Additionally, 1m-6 significantly inhibits TNF-α-induced expression of adhesion molecules, vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1), plus production of proinflammatory cytokines via inhibition of JAK/STAT3 activation and the modulation of Nrf2/HO-1 signalling in HUVECs. In atherosclerosis-prone mice, 1m-6 significantly reduces lipid accumulation and atherosclerotic plaque formation. CONCLUSION AND IMPLICATIONS Our study demonstrates that 1m-6 produces promising atheroprotective effects by enhancing cholesterol efflux and suppressing inflammation-induced endothelial dysfunction, which opens a new avenue for treating ASCVD. LINKED ARTICLES This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc.
Collapse
Affiliation(s)
- Liv Weichien Chen
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Min-Chien Tsai
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Ching-Yuh Chern
- Department of Applied Chemistry, National Chiayi University, Chiayi City, Taiwan
| | - Tien-Ping Tsao
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Division of Cardiology, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Feng-Yen Lin
- Taipei Heart Research Institute and Department of Internal Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Cardiology and Cardiovascular Research Center, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Sy-Jou Chen
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Pi-Fen Tsui
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Yao-Wen Liu
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsien-Jui Lu
- Department of Applied Chemistry, National Chiayi University, Chiayi City, Taiwan
| | - Wan-Lin Wu
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Wei-Shiang Lin
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Sung Tsai
- Division of Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei, Taiwan.,Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Chin-Sheng Lin
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
153
|
Potor L, Sikura KÉ, Hegedűs H, Pethő D, Szabó Z, Szigeti ZM, Pócsi I, Trencsényi G, Szikra D, Garai I, Gáll T, Combi Z, Kappelmayer J, Balla G, Balla J. The Fungal Iron Chelator Desferricoprogen Inhibits Atherosclerotic Plaque Formation. Int J Mol Sci 2020; 21:ijms21134746. [PMID: 32635347 PMCID: PMC7369830 DOI: 10.3390/ijms21134746] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/26/2020] [Accepted: 07/01/2020] [Indexed: 12/14/2022] Open
Abstract
Hemoglobin, heme and iron are implicated in the progression of atherosclerosis. Therefore, we investigated whether the hydrophobic fungal iron chelator siderophore, desferricoprogen (DFC) inhibits atherosclerosis. DFC reduced atherosclerotic plaque formation in ApoE-/- mice on an atherogenic diet. It lowered the plasma level of oxidized LDL (oxLDL) and inhibited lipid peroxidation in aortic roots. The elevated collagen/elastin content and enhanced expression of adhesion molecule VCAM-1 were decreased. DFC diminished oxidation of Low-density Lipoprotein (LDL) and plaque lipids catalyzed by heme or hemoglobin. Formation of foam cells, uptake of oxLDL by macrophages, upregulation of CD36 and increased expression of TNF-α were reduced by DFC in macrophages. TNF-triggered endothelial cell activation (vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecules (ICAMs), E-selectin) and increased adhesion of monocytes to endothelium were attenuated. The increased endothelial permeability and intracellular gap formation provoked by TNF-α was also prevented by DFC. DFC acted as a cytoprotectant in endothelial cells and macrophages challenged with a lethal dose of oxLDL and lowered the expression of stress-responsive heme oxygenase-1 as sublethal dose was employed. Saturation of desferrisiderophore with iron led to the loss of the beneficial effects. We demonstrated that DFC accumulated within the atheromas of the aorta in ApoE-/- mice. DFC represents a novel therapeutic approach to control the progression of atherosclerosis.
Collapse
Affiliation(s)
- László Potor
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, 4012 Debrecen, Hungary; (L.P.); (K.É.S.); (T.G.)
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4012 Debrecen, Hungary
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, 4012 Debrecen, Hungary; (H.H.); (D.P.); (Z.C.)
| | - Katalin Éva Sikura
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, 4012 Debrecen, Hungary; (L.P.); (K.É.S.); (T.G.)
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4012 Debrecen, Hungary
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, 4012 Debrecen, Hungary; (H.H.); (D.P.); (Z.C.)
| | - Hajnalka Hegedűs
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, 4012 Debrecen, Hungary; (H.H.); (D.P.); (Z.C.)
| | - Dávid Pethő
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, 4012 Debrecen, Hungary; (H.H.); (D.P.); (Z.C.)
| | - Zsuzsa Szabó
- Department of Molecular Biotechnology and Microbiology, Institute of Technology, Faculty of Science and Technology, University of Debrecen, 4012 Debrecen, Hungary; (Z.S.); (Z.M.S.); (I.P.)
| | - Zsuzsa M Szigeti
- Department of Molecular Biotechnology and Microbiology, Institute of Technology, Faculty of Science and Technology, University of Debrecen, 4012 Debrecen, Hungary; (Z.S.); (Z.M.S.); (I.P.)
| | - István Pócsi
- Department of Molecular Biotechnology and Microbiology, Institute of Technology, Faculty of Science and Technology, University of Debrecen, 4012 Debrecen, Hungary; (Z.S.); (Z.M.S.); (I.P.)
| | - György Trencsényi
- Scanomed Ltd., University of Debrecen, 4012 Debrecen, Hungary; (G.T.); (D.S.); (I.G.)
| | - Dezső Szikra
- Scanomed Ltd., University of Debrecen, 4012 Debrecen, Hungary; (G.T.); (D.S.); (I.G.)
| | - Ildikó Garai
- Scanomed Ltd., University of Debrecen, 4012 Debrecen, Hungary; (G.T.); (D.S.); (I.G.)
| | - Tamás Gáll
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, 4012 Debrecen, Hungary; (L.P.); (K.É.S.); (T.G.)
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, 4012 Debrecen, Hungary; (H.H.); (D.P.); (Z.C.)
| | - Zsolt Combi
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, 4012 Debrecen, Hungary; (H.H.); (D.P.); (Z.C.)
| | - János Kappelmayer
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, 4012 Debrecen, Hungary;
| | - György Balla
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, 4012 Debrecen, Hungary; (L.P.); (K.É.S.); (T.G.)
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4012 Debrecen, Hungary
- Correspondence: (G.B.); (J.B.)
| | - József Balla
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, 4012 Debrecen, Hungary; (L.P.); (K.É.S.); (T.G.)
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, 4012 Debrecen, Hungary; (H.H.); (D.P.); (Z.C.)
- Correspondence: (G.B.); (J.B.)
| |
Collapse
|
154
|
Zabriskie MS, Wang C, Wang S, Alexander MD. Apolipoprotein E knockout rabbit model of intracranial atherosclerotic disease. Animal Model Exp Med 2020; 3:208-213. [PMID: 32613180 PMCID: PMC7323697 DOI: 10.1002/ame2.12125] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/15/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022] Open
Abstract
Intracranial atherosclerotic disease (ICAD) is the most common cause of ischemic stroke. Poor understanding of the disease due to limited human data leads to imprecise treatment. Apolipoprotein E knockout (ApoE-KO) rabbits were compared to an existing model, the Watanabe heritable hyperlipidemic (WHHL) rabbit, and wild-type New Zealand white (NZW) rabbit controls. Intracranial artery samples were assessed on histopathology for the presence of ICAD. Logistic and ordinal regression analyses were performed to assess for disease presence and severity, respectively. Eighteen rabbits and 54 artery segments were analyzed. Univariate logistic analysis confirmed the presence of ICAD in model rabbits (P < .001), while no difference was found between WHHL and ApoE-KO rabbits (P = .178). In multivariate analysis, only classification as a model vs wild-type animal (P < .001) was associated with the presence of ICAD. Univariate ordinal regression analysis demonstrated an association between ICAD severity and model animals (P = .001), with no difference was noted between WHHL and ApoE-KO rabbits (P = .528). In multivariate ordinal regression analysis, only classification as a model retained significance (P < .001). ICAD can be reliably produced in ApoE-KO rabbits, developing the disease comparably to the older WHHL model. Further analysis is warranted to optimize accelerated development of ICAD in ApoE-KO rabbits to more efficiently study this disease.
Collapse
Affiliation(s)
- Matthew S. Zabriskie
- Department of Radiology and Imaging SciencesUniversity of UtahSalt Lake CityUTUSA
| | - Chuanzhuo Wang
- Department of RadiologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Shuping Wang
- Department of Radiology and Imaging SciencesUniversity of UtahSalt Lake CityUTUSA
| | - Matthew D. Alexander
- Department of Radiology and Imaging SciencesUniversity of UtahSalt Lake CityUTUSA
- Department of NeurosurgeryUniversity of UtahSalt Lake CityUTUSA
| |
Collapse
|
155
|
Kefir peptides alleviate high-fat diet-induced atherosclerosis by attenuating macrophage accumulation and oxidative stress in ApoE knockout mice. Sci Rep 2020; 10:8802. [PMID: 32472055 PMCID: PMC7260220 DOI: 10.1038/s41598-020-65782-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 05/11/2020] [Indexed: 02/01/2023] Open
Abstract
In the past decade, the high morbidity and mortality of atherosclerotic disease have been prevalent worldwide. High-fat food consumption has been suggested to be an overarching factor for atherosclerosis incidence. This study aims to investigate the effects of kefir peptides on high-fat diet (HFD)-induced atherosclerosis in apolipoprotein E knockout (ApoE−/−) mice. 7-week old male ApoE−/− and normal C57BL/6 mice were randomly divided into five groups (n = 8). Atherosclerotic lesion development in ApoE−/− mice was established after fed the HFD for 12 weeks compared to standard chow diet (SCD)-fed C57BL/6 and ApoE−/− control groups. Kefir peptides oral administration significantly improved atherosclerotic lesion development by protecting against endothelial dysfunction, decreasing oxidative stress, reducing aortic lipid deposition, attenuating macrophage accumulation, and suppressing the inflammatory immune response compared with the HFD/ApoE−/− mock group. Moreover, the high dose of kefir peptides substantially inhibited aortic fibrosis and restored the fibrosis in the aorta root close to that observed in the C57BL/6 normal control group. Our findings show, for the first time, anti-atherosclerotic progression via kefir peptides consumption in HFD-fed ApoE−/− mice. The profitable effects of kefir peptides provide new perspectives for its use as an anti-atherosclerotic agent in the preventive medicine.
Collapse
|
156
|
Kheradmandi M, Ackers I, Burdick MM, Malgor R, Farnoud AM. Targeting Dysfunctional Vascular Endothelial Cells Using Immunoliposomes Under Flow Conditions. Cell Mol Bioeng 2020; 13:189-199. [PMID: 32426057 DOI: 10.1007/s12195-020-00616-1] [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] [Received: 10/24/2019] [Accepted: 04/24/2020] [Indexed: 02/08/2023] Open
Abstract
Introduction Atherosclerosis (ATH), the build up of fat in the arteries, is a principal cause of heart attack and stroke. Drug instability and lack of target specificity are major drawbacks of current clinical therapeutics. These undesirable effects can be eliminated by site-specific drug delivery. The endothelial surface over ATH lesions has been shown to overexpress vascular cell adhesion molecule1 (VCAM1), which can be used for targeted therapy. Methods Here, we report the synthesis, characterization, and development of anti VCAM1-functionalized liposomes to target cells overexpressing VCAM1 under static and flow conditions. Liposomes were composed of dioleoyl-phosphatidylcholine, sphingomyelin, cholesterol, and distearoyl-phosphatidylethanolamine-polyethylene glycol-cyanur (31.67:31.67:31.67:5 mol%). VCAM1 expression in endothelial cells was induced by lipopolysaccharide (LPS) treatment. Results Characterization study revealed that liposomes were negatively charged (- 7.7 ± 2.6 mV) with an average diameter of 201.3 ± 3.3 nm. Liposomes showed no toxicity toward THP-1 derived macrophages and endothelial cells. Liposomes were able to target both fixed and non-fixed endothelial cells, in vitro, with significantly higher localization observed in non-fixed conditions. To mimic biological and physiologically-relevant conditions, liposome targeting was also examined under flow (4 dyn/cm2) with or without erythrocytes (40% v/v hematocrit). Liposomes were able to target LPS-treated endothelial cells under dynamic culture, in the presence or absence of erythrocytes, although targeting efficiency was five-fold lower in flow compared to static conditions. Conclusions This liposomal delivery system showed a significant improvement in localization on dysfunctional endothelium after surface functionalization. We conclude that VCAM1-functionalized liposomes can target and potentially deliver therapeutic compounds to ATH regions.
Collapse
Affiliation(s)
- Mahsa Kheradmandi
- Department of Chemical and Biomolecular Engineering, Ohio University, 161 Stocker Center, Athens, OH 45701 USA
| | - Ian Ackers
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701 USA.,Translational Biomedical Science Program, Ohio University, Athens, OH 45701 USA
| | - Monica M Burdick
- Department of Chemical and Biomolecular Engineering, Ohio University, 161 Stocker Center, Athens, OH 45701 USA.,Translational Biomedical Science Program, Ohio University, Athens, OH 45701 USA
| | - Ramiro Malgor
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701 USA.,Translational Biomedical Science Program, Ohio University, Athens, OH 45701 USA
| | - Amir M Farnoud
- Department of Chemical and Biomolecular Engineering, Ohio University, 161 Stocker Center, Athens, OH 45701 USA.,Translational Biomedical Science Program, Ohio University, Athens, OH 45701 USA
| |
Collapse
|
157
|
Lawal IO, Stoltz AC, Sathekge MM. Molecular imaging of cardiovascular inflammation and infection in people living with HIV infection. Clin Transl Imaging 2020. [DOI: 10.1007/s40336-020-00370-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
158
|
Lee J, Ha SJ, Park J, Kim YH, Lee NH, Kim YE, Hong YS, Song KM. Arctium lappa root extract containing L-arginine prevents TNF-α-induced early atherosclerosis in vitro and in vivo. Nutr Res 2020; 77:85-96. [PMID: 32388084 DOI: 10.1016/j.nutres.2020.03.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 03/12/2020] [Accepted: 03/17/2020] [Indexed: 11/28/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease affecting the aorta and is a major cause of cardiovascular disease. Arctium lappa root is a plant widely used in traditional Chinese medicine (TCM), and Arctium lappa root extract (ALE) has been reported to exhibit anti-inflammatory capacity and to ameliorate endothelial dysfunction. Thus, we hypothesized that ALE would inhibit the early atherosclerotic stage. In this study, we evaluated the inhibitory effect of ALE on early arteriosclerosis and its mechanisms of action. ALE suppressed TNF-α-induced monocyte adhesion to the vascular endothelium by suppressing NF-κB signaling in HUVECs. In an acute mouse model of atherosclerosis, ALE suppressed TNF-α-induced monocyte infiltration of the vascular endothelium and the expression of genes encoding inflammatory cytokines including IL-1β, IL-6, TNF-α, and MCP-1 in the mouse aorta. Moreover, inulin-type fructan and amino acids, especially L-aspartate and L-arginine (60.27 and 42.17 mg/g, respectively) were detected by NMR, MALDI-TOF MS, and HPLC analysis as the main components of ALE. Notably, L-arginine suppressed TNF-α-induced monocyte adhesion to HUVECs. Therefore, these results suggest that ALE may be a functional food for the suppression or prevention of early stages of atherosclerosis.
Collapse
Affiliation(s)
- Jangho Lee
- Division of Food Functionality Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea.
| | - Su Jeong Ha
- Division of Strategic Food Technology Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea; Department of Agricultural Biotechnology, Seoul National University, Seoul 151-921, Republic of Korea.
| | - Joon Park
- Division of Food Functionality Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea; Department of Food Biotechnology, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea.
| | - Young Ho Kim
- Division of Strategic Food Technology Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea.
| | - Nam Hyouck Lee
- Division of Strategic Food Technology Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea.
| | - Young Eon Kim
- Division of Strategic Food Technology Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea.
| | - Young-Shick Hong
- Division of Food and Nutrition, Chonnam National University, Yongbong-ro, Buk-gu, Gwangju 500-757, Republic of Korea.
| | - Kyung-Mo Song
- Division of Strategic Food Technology Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea.
| |
Collapse
|
159
|
Wang S, Wang F, Yang L, Li Q, Huang Y, Cheng Z, Chu H, Song Y, Shang L, Hao W, Wei X. Effects of coal-fired PM 2.5 on the expression levels of atherosclerosis-related proteins and the phosphorylation level of MAPK in ApoE -/- mice. BMC Pharmacol Toxicol 2020; 21:34. [PMID: 32384920 PMCID: PMC7206822 DOI: 10.1186/s40360-020-00411-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/30/2020] [Indexed: 12/19/2022] Open
Abstract
Background Air pollution increases the morbidity and mortality of cardiovascular disease (CVD). Atherosclerosis (AS) is the pathological basis of most CVD, and the progression of atherosclerosis and the increase of fragile plaque rupture are the mechanism basis of the relationship between atmospheric particulate pollution and CVD. The aim of the present study was to investigate the effects of coal-fired fine particulate matter (PM2.5) on the expression levels of atherosclerosis-related proteins (von Willebrand factor (vWF), Endothelin-1 (ET-1), intercellular adhesion molecule-1 (ICAM-1), and E-selectin, and to explore the role and mechanism of the progression of atherosclerosis induced by coal-fired PM2.5 via the mitogen-activated protein kinase (MAPK) signaling pathways. Methods Different concentrations of PM2.5 were given to apolipoprotein-E knockout (ApoE−/−) mice via intratracheal instillation for 8 weeks. Enzyme-linked immunosorbent assay (ELISA) was used to detect the levels of vWF, ET-1 in serum of mice. Immunohistochemistry was used to observe the expression and distribution of ICAM-1 and E-selectin in the aorta of mice. Western blot was used to investigate the phosphoylation of proteins relevant to MAPK signaling pathways. Results Coal-fired PM2.5 exacerbated atherosclerosis induced by a high-fat diet. Fibrous cap formation, foam cells accumulation, and atherosclerotic lesions were observed in the aortas of PM2.5-treated mice. Coal-fired PM2.5 increased the protein levels of ET-1, ICAM-1, and E-selectin, but there was no significant difference in the vWF levels between the PM2.5-treatment mice and the HFD control mice. Coal-fired PM2.5 promoted the phosphorylation of p38, c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK) in aortic tissues of mice. Conclusion Coal-derived PM2.5 exacerbated the formation of atherosclerosis in mice, increased the expression levels of atherosclerosis-related proteins in mice serum, and promoted the phosphorylation of proteins relevant to MAPK signaling pathway. Thus, MAPK signaling pathway may play a role in the atherosclerosis pathogenesis induced by Coal-derived PM2.5.
Collapse
Affiliation(s)
- Siqi Wang
- Department of Toxicology, School of Public Health, Peking University Health Science Center, No.38 XueYuan Road, HaiDian District, Beijing, 100191, People's Republic of China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, People's Republic of China
| | - Feifei Wang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, People's Republic of China
| | - Lixin Yang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, People's Republic of China
| | - Qin Li
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, People's Republic of China
| | - Yao Huang
- Department of Toxicology, School of Public Health, Peking University Health Science Center, No.38 XueYuan Road, HaiDian District, Beijing, 100191, People's Republic of China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, People's Republic of China
| | - Zhiyuan Cheng
- Department of Toxicology, School of Public Health, Peking University Health Science Center, No.38 XueYuan Road, HaiDian District, Beijing, 100191, People's Republic of China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, People's Republic of China
| | - Hongqian Chu
- Department of Toxicology, School of Public Health, Peking University Health Science Center, No.38 XueYuan Road, HaiDian District, Beijing, 100191, People's Republic of China.,Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, People's Republic of China.,Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, People's Republic of China
| | - Yiming Song
- Department of Toxicology, School of Public Health, Peking University Health Science Center, No.38 XueYuan Road, HaiDian District, Beijing, 100191, People's Republic of China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, People's Republic of China
| | - Lanqin Shang
- Department of Toxicology, School of Public Health, Peking University Health Science Center, No.38 XueYuan Road, HaiDian District, Beijing, 100191, People's Republic of China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, People's Republic of China
| | - Weidong Hao
- Department of Toxicology, School of Public Health, Peking University Health Science Center, No.38 XueYuan Road, HaiDian District, Beijing, 100191, People's Republic of China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, People's Republic of China
| | - Xuetao Wei
- Department of Toxicology, School of Public Health, Peking University Health Science Center, No.38 XueYuan Road, HaiDian District, Beijing, 100191, People's Republic of China. .,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, People's Republic of China.
| |
Collapse
|
160
|
Stanley S, Vanarsa K, Soliman S, Habazi D, Pedroza C, Gidley G, Zhang T, Mohan S, Der E, Suryawanshi H, Tuschl T, Buyon J, Putterman C, Mok CC, Petri M, Saxena R, Mohan C. Comprehensive aptamer-based screening identifies a spectrum of urinary biomarkers of lupus nephritis across ethnicities. Nat Commun 2020; 11:2197. [PMID: 32366845 PMCID: PMC7198599 DOI: 10.1038/s41467-020-15986-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 04/02/2020] [Indexed: 02/08/2023] Open
Abstract
Emerging urinary biomarkers continue to show promise in evaluating lupus nephritis (LN). Here, we screen urine from active LN patients for 1129 proteins using an aptamer-based platform, followed by ELISA validation in two independent cohorts comprised of 127 inactive lupus, 107 active LN, 67 active non-renal lupus patients and 74 healthy controls, of three different ethnicities. Urine proteins that best distinguish active LN from inactive disease are ALCAM, PF-4, properdin, and VCAM-1 among African-Americans, sE-selectin, VCAM-1, BFL-1 and Hemopexin among Caucasians, and ALCAM, VCAM-1, TFPI and PF-4 among Asians. Most of these correlate significantly with disease activity indices in the respective ethnic groups, and surpass conventional metrics in identifying active LN, with better sensitivity, and negative/positive predictive values. Several elevated urinary molecules are also expressed within the kidneys in LN, based on single-cell RNAseq analysis. Longitudinal studies are warranted to assess the utility of these biomarkers in tracking lupus nephritis.
Collapse
Affiliation(s)
- Samantha Stanley
- Department Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Kamala Vanarsa
- Department Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Samar Soliman
- Department Biomedical Engineering, University of Houston, Houston, TX, USA
- Rheumatology and Rehabilitation Department, Faculty of Medicine, Minia University, Minya, Egypt
| | - Deena Habazi
- Department Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Claudia Pedroza
- Center for Clinical Research and Evidence-Based Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Gabriel Gidley
- Department Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Ting Zhang
- Department Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Shree Mohan
- Department Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Evan Der
- Department of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hemant Suryawanshi
- Department of Molecular Biology, Rockefeller University, New York, NY, USA
| | - Thomas Tuschl
- Department of Molecular Biology, Rockefeller University, New York, NY, USA
| | - Jill Buyon
- Department of Rheumatology, New York University, New York, NY, USA
| | - Chaim Putterman
- Department of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, USA
- Azrieli Faculty of Medicine, Bar-Ilan University, Zefat, Israel
- Research Institute, Galilee Medical Center, Nahariya, Israel
| | - Chi Chiu Mok
- Department of Medicine, Tuen Mun Hospital, New Territories, Hong Kong, China
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ramesh Saxena
- University Hospital Kidney & Liver Clinic, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chandra Mohan
- Department Biomedical Engineering, University of Houston, Houston, TX, USA.
| |
Collapse
|
161
|
Gabrielyan A, Quade M, Gelinsky M, Rösen-Wolff A. IL-11 and soluble VCAM-1 are important components of Hypoxia Conditioned Media and crucial for Mesenchymal Stromal Cells attraction. Stem Cell Res 2020; 45:101814. [PMID: 32334367 DOI: 10.1016/j.scr.2020.101814] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 04/03/2020] [Accepted: 04/09/2020] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Bone marrow stromal cells (BMSC) are highly attractive for tissue engineering due to their ability to differentiate into different cell types, to expand extensively in vitro and to release paracrine soluble factors with a high regenerative potential. They were observed to migrate towards the sites of injury in response to chemotactic signals in vivo. During the last years hypoxia has become a proven method to control proliferation, differentiation and multipotency of BMSC. Conditioned medium from hypoxia-treated BMSC (Hypoxia-conditioned Medium; HCM) has been shown to have various favorable properties on tissue regeneration - such as on cell recruitment, wound healing, angiogenesis and revascularization. Due to this regenerative potential many studies attempt to further characterize HCM and its main functional components. In this study we used HCM generated from umbilical cord mesenchymal stem cells (UC-MSC) instead of BMSC, because GMP-verified methods were used to isolate and cultivate the cells and ensure their constant quality. UC-MSC have a high regenerative potential and are still immunologically naive and therefore highly unlikely to cause an immune reaction. In our article we took the first steps to closer investigate the role of umbilical cord MSC-derived HCM components, namely stromal cell-derived factor 1 (SDF-1α), interleukin 11 (IL-11) and soluble vascular cell adhesion molecule 1 (sVCAM-1). RESULTS Our results show previously unknown roles of IL-11 and sVCAM-1 in the attraction of BMSC. The synergistic effect of the investigated protein mixture consisting of IL-11, sVCAM-1 and SDF-1α as well as those recombinant proteins alone revealed a significantly higher chemoattractive capacity towards human BMSC compared to normoxic control medium. Both, the protein mixtures and proteins alone as well as UC-HCM showed an angiogenic effect by promoting the formation of significantly longer tubule structures and higher amounts of junctions and tubules compared to normoxic control medium. CONCLUSIONS By showing the prominent upregulation of IL-11, sVCAM-1 and SDF-1α under hypoxic conditions compared to normoxic control and revealing their crucial role in migration of human BMSC we took a further step forward in characterization of the chemoattractive components of HCM.
Collapse
Affiliation(s)
- Anastasia Gabrielyan
- Department of Pediatrics, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307 Dresden, Germany.
| | - Mandy Quade
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Angela Rösen-Wolff
- Department of Pediatrics, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| |
Collapse
|
162
|
Suh JS, Lee SH, Fouladian Z, Lee JY, Kim T, Kang MK, Lusis AJ, Boström KI, Kim RH, Park NH. Rosuvastatin Prevents the Exacerbation of Atherosclerosis in Ligature-Induced Periodontal Disease Mouse Model. Sci Rep 2020; 10:6383. [PMID: 32286430 PMCID: PMC7156392 DOI: 10.1038/s41598-020-63350-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/04/2020] [Indexed: 01/06/2023] Open
Abstract
Periodontitis is a local and systemic inflammatory condition and a risk factor of atherosclerosis, but no studies investigated the effect of a statin on atherogenesis affected by severe periodontitis. In this study, we investigated the effect of rosuvastatin (RSV) on atherogenesis in Apolipoprotein E-deficient mice receiving silk ligature placement around the maxillary second molars. Mice with the ligature placement developed severe periodontitis and vascular inflammation. RSV significantly inhibited the development of periodontitis and vascular inflammation and remarkably blocked the increased lipid deposition and the atherogenic gene expression in the arterial wall and aortic sinus induced by severe periodontitis. To understand the mechanistic effect of RSV on periodontitis-associated atherogenesis, we investigated the in vitro effect of RSV on various effect of TNF-α, a major proinflammatory cytokine for periodontitis and atherogenesis. We found that RSV notably inhibited the TNF-α-induced osteoclast formation, endothelial cell phenotypic changes, foam cell formation, and the expression of CD47 and other oncogenes in arterial smooth muscle cells. Taken together, our study indicates that RSV prevents the exacerbation of atherosclerosis induced periodontitis by inhibiting local, systemic and vascular inflammation, as well as the expression of CD47 from arterial smooth muscle cells in mice.
Collapse
Affiliation(s)
- Jin Sook Suh
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, 10833 Le Conte Ave, Los Angeles, CA, USA
| | - Sung Hee Lee
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, 10833 Le Conte Ave, Los Angeles, CA, USA
| | - Zachary Fouladian
- Department of Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, USA
| | - Jae Young Lee
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, 10833 Le Conte Ave, Los Angeles, CA, USA
| | - Terresa Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, 10833 Le Conte Ave, Los Angeles, CA, USA
| | - Mo K Kang
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, 10833 Le Conte Ave, Los Angeles, CA, USA
| | - Aldons J Lusis
- Department of Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, USA
| | - Kristina I Boström
- Department of Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, USA
| | - Reuben H Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, 10833 Le Conte Ave, Los Angeles, CA, USA.
- UCLA Jonsson Comprehensive Cancer Center, 10833 Le Conte Ave, Los Angeles, CA, USA.
| | - No-Hee Park
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, 10833 Le Conte Ave, Los Angeles, CA, USA.
- Department of Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA, USA.
- UCLA Jonsson Comprehensive Cancer Center, 10833 Le Conte Ave, Los Angeles, CA, USA.
| |
Collapse
|
163
|
Badmus OO, Olatunji LA. Dexamethasone causes defective glucose-6-phosphate dehydrogenase dependent antioxidant barrier through endoglin in pregnant and nonpregnant rats. Can J Physiol Pharmacol 2020; 98:667-677. [PMID: 32259461 DOI: 10.1139/cjpp-2018-0351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glucocorticoid therapy has been associated with adverse cardiometabolic effects during pregnancy. Inflammation-mediated cardiac dysfunction, an independent risk factor for morbidity and mortality, has been linked to defective glucose-6-phosphate dehydrogenase (G6PD) dependent antioxidant defenses and increased endoglin expression. We therefore sought to investigate the effects of dexamethasone (DEX) on cardiac endoglin and G6PD-dependent antioxidant defense. Twenty-four rats were randomly assigned to nonpregnant (PRE(-)), DEX-exposed nonpregnant (PRE(-) + DEX), pregnant (PRE(+)), and DEX-exposed pregnant (PRE(+) + DEX) rats, respectively (n = 6 per group). PRE(-) and PRE(+) rats received vehicle (per oral (po)), while PRE(-) + DEX and PRE(+) + DEX groups were administered DEX (0.2 mg/kg po) between gestational days 14 and 19, respectively. Results showed that DEX caused increased cardiac pro-inflammatory markers (adenosine deaminase (ADA) activity, endoglin, vascular cell adhesion molecule-1 (VCAM-1), tissue injury markers (LDH, GGT, AST, ALT, and ALP), metabolic disturbances (elevated fasting plasma glucose, free fatty acid (FFA), lactate, cardiac FFA, and lactate) and depressed G6PD-dependent antioxidant defenses (G6PD activity, reduced glutathione/oxidized glutathione ratio, and nitric oxide) in pregnant and nonpregnant rats. The present study demonstrates that DEX led to increased cardiac endoglin and VCAM-1 that is accompanied by defective G6PD-dependent antioxidant defenses but not cardiac lipid accumulation in both pregnant and nonpregnant rats.
Collapse
Affiliation(s)
- Olufunto O Badmus
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria.,Department of Public Health, Kwara State University, Malete, Nigeria
| | - Lawrence A Olatunji
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| |
Collapse
|
164
|
Yoon JK, Kim DH, Kang ML, Jang HK, Park HJ, Lee JB, Yi SW, Kim HS, Baek S, Park DB, You J, Lee SD, Sei Y, Ahn SI, Shin YM, Kim CS, Bae S, Kim Y, Sung HJ. Anti-Atherogenic Effect of Stem Cell Nanovesicles Targeting Disturbed Flow Sites. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2000012. [PMID: 32239653 DOI: 10.1002/smll.202000012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 02/28/2020] [Accepted: 02/28/2020] [Indexed: 06/11/2023]
Abstract
Atherosclerosis development leads to irreversible cascades, highlighting the unmet need for improved methods of early diagnosis and prevention. Disturbed flow formation is one of the earliest atherogenic events, resulting in increased endothelial permeability and subsequent monocyte recruitment. Here, a mesenchymal stem cell (MSC)-derived nanovesicle (NV) that can target disturbed flow sites with the peptide GSPREYTSYMPH (PREY) (PMSC-NVs) is presented which is selected through phage display screening of a hundred million peptides. The PMSC-NVs are effectively produced from human MSCs (hMSCs) using plasmid DNA designed to functionalize the cell membrane with PREY. The potent anti-inflammatory and pro-endothelial recovery effects are confirmed, similar to those of hMSCs, employing mouse and porcine partial carotid artery ligation models as well as a microfluidic disturbed flow model with human carotid artery-derived endothelial cells. This nanoscale platform is expected to contribute to the development of new theragnostic strategies for preventing the progression of atherosclerosis.
Collapse
Affiliation(s)
- Jeong-Kee Yoon
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Dae-Hyun Kim
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Mi-Lan Kang
- TMD LAB Co., Ltd, Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hyeon-Ki Jang
- Department of Chemistry, Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, 04763, Republic of Korea
| | - Hyun-Ji Park
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, 30313, USA
| | - Jung Bok Lee
- Department of Biological Science, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Se Won Yi
- TMD LAB Co., Ltd, Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hye-Seon Kim
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Sewoom Baek
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Dan Bi Park
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jin You
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | | | - Yoshitaka Sei
- George W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, 30313, USA
| | - Song Ih Ahn
- George W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, 30313, USA
| | - Young Min Shin
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | | | - Sangsu Bae
- Department of Chemistry, Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, 04763, Republic of Korea
| | - YongTae Kim
- George W. Woodruff School of Mechanical Engineering, Wallace H. Coulter Department of Biomedical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience (IBB), Institute for Electronics and Nanotechnology (IEN), Georgia Institute of Technology, Atlanta, Georgia, 30313, USA
| | - Hak-Joon Sung
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| |
Collapse
|
165
|
Regal-McDonald K, Somarathna M, Lee T, Litovsky SH, Barnes J, Peretik JM, Traylor JG, Orr AW, Patel RP. Assessment of ICAM-1 N-glycoforms in mouse and human models of endothelial dysfunction. PLoS One 2020; 15:e0230358. [PMID: 32208424 PMCID: PMC7092995 DOI: 10.1371/journal.pone.0230358] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 02/27/2020] [Indexed: 12/31/2022] Open
Abstract
Endothelial dysfunction is a critical event in vascular inflammation characterized, in part, by elevated surface expression of adhesion molecules such as intercellular adhesion molecule-1 (ICAM-1). ICAM-1 is heavily N-glycosylated, and like other surface proteins, it is largely presumed that fully processed, complex N-glycoforms are dominant. However, our recent studies suggest that hypoglycosylated or high mannose (HM)-ICAM-1 N-glycoforms are also expressed on the cell surface during endothelial dysfunction, and have higher affinity for monocyte adhesion and regulate outside-in endothelial signaling by different mechanisms. Whether different ICAM-1 N-glycoforms are expressed in vivo during disease is unknown. In this study, using the proximity ligation assay, we assessed the relative formation of high mannose, hybrid and complex α-2,6-sialyated N-glycoforms of ICAM-1 in human and mouse models of atherosclerosis, as well as in arteriovenous fistulas (AVF) of patients on hemodialysis. Our data demonstrates that ICAM-1 harboring HM or hybrid epitopes as well as ICAM-1 bearing α-2,6-sialylated epitopes are present in human and mouse atherosclerotic lesions. Further, HM-ICAM-1 positively associated with increased macrophage burden in lesions as assessed by CD68 staining, whereas α-2,6-sialylated ICAM-1 did not. Finally, both HM and α-2,6-sialylated ICAM-1 N-glycoforms were present in hemodialysis patients who had AVF maturation failure compared to successful AVF maturation. Collectively, these data provide evidence that HM- ICAM-1 N-glycoforms are present in vivo, and at levels similar to complex α-2,6-sialylated ICAM-1 underscoring the need to better understand their roles in modulating vascular inflammation.
Collapse
Affiliation(s)
- Kellie Regal-McDonald
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Maheshika Somarathna
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Timmy Lee
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Silvio H. Litovsky
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jarrod Barnes
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - J. M. Peretik
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - J. G. Traylor
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - A. Wayne Orr
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - Rakesh P. Patel
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
166
|
Kim MJ, Jung SK. Nutraceuticals for prevention of atherosclerosis: Targeting monocyte infiltration to the vascular endothelium. J Food Biochem 2020; 44:e13200. [PMID: 32189369 DOI: 10.1111/jfbc.13200] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 01/14/2020] [Accepted: 01/20/2020] [Indexed: 12/28/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death, globally, and is a serious problem in developing countries. Preventing atherosclerosis is key to reducing the risk of developing CVD. Similar to carcinogenesis, atherogenesis can be divided into four stages: initiation, promotion, progression, and acute events. The current study focuses on the promotion stage, which is characterized by circular monocyte penetration into vascular endothelial cells, monocyte differentiation into macrophages, and the formation of foam cells. This early stage of atherogenesis is a major target for nutraceuticals. We discuss nutraceuticals that can potentially inhibit monocyte adhesion to the vascular endothelium, thereby preventing the promotional stage of atherosclerosis. The mechanisms through which these nutraceuticals prevent monocyte adhesion are classified according to the following targets: NF-κB, ROS, MAPKs, and AP-1. Additionally, we discuss promising targets for nutraceuticals that can regulate monocyte adhesion to the endothelium. PRACTICAL APPLICATIONS: Introduction of atherogenesis with initiation, promotion, progression, and acute events provide specific information and factors for each step in the development of atherosclerosis. Functional food or pharmaceutical researchers can set target stages and use them to develop materials that control atherosclerosis. In particular, because it focuses on vascular inflammation via interaction between monocytes and vascular endothelial cells, it provides specific information to researchers developing functional foods that regulate this process. Therefore, this manuscript, unlike previous papers, will provide material information and potential mechanisms of action to researchers who want to develop functional foods that control vascular inflammation rather than vascular lipids.
Collapse
Affiliation(s)
- Min Jeong Kim
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, Republic of Korea
| | - Sung Keun Jung
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, Republic of Korea.,Institute of Agricultural Science & Technology, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
167
|
Bindesbøll C, Aas A, Ogmundsdottir MH, Pankiv S, Reine T, Zoncu R, Simonsen A. NBEAL1 controls SREBP2 processing and cholesterol metabolism and is a susceptibility locus for coronary artery disease. Sci Rep 2020; 10:4528. [PMID: 32161285 PMCID: PMC7066131 DOI: 10.1038/s41598-020-61352-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/21/2020] [Indexed: 01/24/2023] Open
Abstract
Dysregulated cholesterol homeostasis promotes the pathology of atherosclerosis, myocardial infarction and strokes. Cellular cholesterol is mainly regulated at the transcriptional level by SREBP2, but also through uptake of extracellular cholesterol from low density lipoproteins (LDL) via expression of LDL receptors (LDLR) at the cell surface. Identification of the mechanisms involved in regulation of these processes are thus key to understand the pathology of coronary artery disease. Here, we identify the large and poorly characterized BEACH domain protein Neurobeachin-like (NBEAL) 1 as a Golgi- associated protein required for regulation of cholesterol metabolism. NBEAL1 is most abundantly expressed in arteries. Genetic variants in NBEAL1 are associated with decreased expression of NBEAL1 in arteries and increased risk of coronary artery disease in humans. We show that NBEAL1 regulates cholesterol metabolism by modulating LDLR expression in a mechanism involving interaction with SCAP and PAQR3 and subsequent SREBP2-processing. Thus, low expression of NBEAL1 may lead to increased risk of coronary artery disease by downregulation of LDLR levels.
Collapse
Affiliation(s)
- Christian Bindesbøll
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 1112 Blindern, 0317, Oslo, Norway.
| | - Aleksander Aas
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 1112 Blindern, 0317, Oslo, Norway
| | - Margret Helga Ogmundsdottir
- Department of Biochemistry and Molecular Biology, Biomedical Center, Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, 101, Reykjavik, Iceland
| | - Serhiy Pankiv
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 1112 Blindern, 0317, Oslo, Norway
| | - Trine Reine
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, 1112 Blindern, 0317, Oslo, Norway.,Section for Interphase genetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0424, Oslo, Norway
| | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 1112 Blindern, 0317, Oslo, Norway.
| |
Collapse
|
168
|
Huang HS, Liao PC, Liu CJ. Calcium Kidney Stones are Associated with Increased Risk of Carotid Atherosclerosis: The Link between Urinary Stone Risks, Carotid Intima-Media Thickness, and Oxidative Stress Markers. J Clin Med 2020; 9:jcm9030729. [PMID: 32182704 PMCID: PMC7141231 DOI: 10.3390/jcm9030729] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 02/27/2020] [Accepted: 03/06/2020] [Indexed: 12/15/2022] Open
Abstract
Previous studies have suggested that kidney stone formers are associated with a higher risk of cardiovascular events. To our knowledge, there have been no previous examinations of the relationship between carotid intima-media thickness (IMT) and urinary stone risk factors. This study was aimed toward an investigation of the association between dyslipidemia, IMT, and 24-hour urinalysis in patients with calcium oxalate (CaOx) or calcium phosphate (CaP) stones. We prospectively enrolled 114 patients with kidney stones and 33 controls between January 2016 and August 2016. All patients were divided into four groups, according to the stone compositions—CaOx ≥ 50% group, CaP group, struvite group, and uric acid stones group. Carotid IMT and the carotid score (CS) were evaluated using extracranial carotid artery doppler ultrasonography. The results of a multivariate analysis indicated that a higher serum total cholesterol (TC) and low-density lipoprotein (LDL) were all associated with lower urinary citrate and higher CS in both the CaOx ≥ 50% and CaP groups. Higher serum TC and LDL were also associated with increased serum 8-OHdG levels in both groups. The levels of carotid IMT and CS in the CaOx ≥ 50% and CaP groups were all significantly higher than in the controls. These findings suggest a strong link between dyslipidemia, carotid atherosclerosis, and calcium kidney stone disease.
Collapse
Affiliation(s)
- Ho Shiang Huang
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan;
- Department of Urology, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| | - Pao Chi Liao
- Department of Environmental and Occupational Health, Medical College, National Cheng Kung University, Tainan 70403, Taiwan;
| | - Chan Jung Liu
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan;
- Correspondence: ; Tel.: +886-6-235-3535 (ext. 5251); Fax: +886-6-276-6179
| |
Collapse
|
169
|
Cai S, Yang Q, Cao Y, Li Y, Liu J, Wang J, Zhang X, Liu L, Li X, Zhang Y. PF4 antagonizes retinal neovascularization via inhibiting PRAS40 phosphorylation in a mouse model of oxygen-induced retinopathy. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165604. [PMID: 31740404 DOI: 10.1016/j.bbadis.2019.165604] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 10/19/2019] [Accepted: 11/01/2019] [Indexed: 12/22/2022]
|
170
|
Amirfakhryan H. Vaccination against atherosclerosis: An overview. Hellenic J Cardiol 2020; 61:78-91. [DOI: 10.1016/j.hjc.2019.07.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/16/2019] [Accepted: 07/18/2019] [Indexed: 02/07/2023] Open
|
171
|
Szeto CC, Chow KM, Lam CWK, Cheung R, Kwan BCH, Chung KY, Leung CB, Li PKT. Peritoneal Albumin Excretion is a Strong Predictor of Cardiovascular Events in Peritoneal Dialysis Patients: A Prospective Cohort Study. Perit Dial Int 2020. [DOI: 10.1177/089686080502500508] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background Microalbuminuria is a marker of systemic endothelial dysfunction. We hypothesize that peritoneal albumin excretion in peritoneal dialysis (PD) patients, which is conceptually analogous to microalbuminuria in nonuremic patients, can predict cardiovascular disease in new PD patients. Method We studied peritoneal albumin excretion in 43 new PD patients. They were then followed prospectively for the development of cardiovascular events. All-cause mortality and duration of hospitalization for cardiovascular diseases were also recorded. Result The average duration of follow-up was 26.5 ± 17.6 months. During the follow-up period, 15 patients developed cardiovascular events. Event-free survival at 36 months was 81.4% and 53.6% for low (<300 mg/L) and high (≥300 mg/L) peritoneal albumin excretion groups respectively (log rank test, p = 0.042). By Cox regression analysis, the only independent factors for event-free survival were diabetic status and peritoneal albumin excretion rate. For every 100 mg/L increase in peritoneal albumin excretion, the adjusted hazard ratio of developing a cardiovascular event was 1.83 [95% confidence interval (CI) 1.11 – 3.02, p = 0.018]. Actuarial patient survival at 36 months was 85.7% and 59.1% for low and high peritoneal albumin excretion groups respectively (log rank test, p = 0.10). After adjusting for the duration of follow-up for individual patients, the average duration of hospitalization was 9.1 ± 16.2 and 21.7 ± 25.7 days per year of follow-up for low and high peritoneal albumin excretion groups respectively (Mann–Whitney U test, p = 0.012). Conclusion Although the sample size of our present study is small and does not have adequate statistical power, we conclude that peritoneal albumin excretion may be an important predictor of cardiovascular disease. Further studies are needed to examine the role of dialysate albumin excretion as a means of cardiovascular risk stratification in PD patients.
Collapse
Affiliation(s)
- Cheuk-Chun Szeto
- Department of Medicine & Therapeutics and Department of Chemical Pathology, Prince of Wales Hospital
| | - Kai-Ming Chow
- Department of Medicine & Therapeutics and Department of Chemical Pathology, Prince of Wales Hospital
| | | | - Robert Cheung
- The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Bonnie Ching-Ha Kwan
- Department of Medicine & Therapeutics and Department of Chemical Pathology, Prince of Wales Hospital
| | - Kwok-Yi Chung
- Department of Medicine & Therapeutics and Department of Chemical Pathology, Prince of Wales Hospital
| | - Chi-Bon Leung
- Department of Medicine & Therapeutics and Department of Chemical Pathology, Prince of Wales Hospital
| | - Philip Kam-Tao Li
- Department of Medicine & Therapeutics and Department of Chemical Pathology, Prince of Wales Hospital
| |
Collapse
|
172
|
Monslow J, Todd L, Chojnowski JE, Govindaraju PK, Assoian RK, Puré E. Fibroblast Activation Protein Regulates Lesion Burden and the Fibroinflammatory Response in Apoe-Deficient Mice in a Sexually Dimorphic Manner. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1118-1136. [PMID: 32084369 DOI: 10.1016/j.ajpath.2020.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 12/19/2019] [Accepted: 01/02/2020] [Indexed: 01/10/2023]
Abstract
Fibroblast activation protein (FAP) has been established as an inducible and mesenchymal cell-specific mediator of disease progression in cancer and fibrosis. Atherosclerosis is a fibroinflammatory disease, and FAP was previously reported to be up-regulated in human atherosclerotic plaques compared with normal vessel. We investigated the spatial and temporal distribution of Fap-expressing cells in a murine model of atherosclerosis and used a genetic approach to determine if and how Fap affected disease progression. Fap was found to be expressed predominantly on vascular smooth muscle cells in lesions of athero-prone Apoe-/- mice. Global deletion of Fap (Fap-/-) in Apoe-/- mice accelerated atherosclerotic disease progression in both males and females, with the effect observed earlier in males. Sex-specific effects on lesion morphology were observed. Relative levels of extracellular matrix, fibrotic, and inflammatory cell content were comparable in lesions in male mice regardless of Fap status. In contrast, lesions in Fap-/- female mice were characterized by a more fibrotic composition due to a reduction in inflammation, specifically a reduction in Mox macrophages. Combined, these data suggest that Fap restrains the progression of atherosclerosis and may contribute to the sexually dimorphic susceptibility to atherosclerosis by regulating the balance between inflammation (an indicator of vulnerability to plaque rupture) and fibrosis (an indicator of plaque stability).
Collapse
Affiliation(s)
- James Monslow
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia Pennsylvania.
| | - Leslie Todd
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia Pennsylvania
| | - John E Chojnowski
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia Pennsylvania
| | - Priya K Govindaraju
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia Pennsylvania
| | - Richard K Assoian
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia Pennsylvania
| | - Ellen Puré
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia Pennsylvania.
| |
Collapse
|
173
|
High-Density Lipoprotein (HDL) Inhibits Serum Amyloid A (SAA)-Induced Vascular and Renal Dysfunctions in Apolipoprotein E-Deficient Mice. Int J Mol Sci 2020; 21:ijms21041316. [PMID: 32075280 PMCID: PMC7072968 DOI: 10.3390/ijms21041316] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/09/2020] [Accepted: 02/11/2020] [Indexed: 01/09/2023] Open
Abstract
Serum amyloid A (SAA) promotes endothelial inflammation and dysfunction that is associated with cardiovascular disease and renal pathologies. SAA is an apoprotein for high-density lipoprotein (HDL) and its sequestration to HDL diminishes SAA bioactivity. Herein we investigated the effect of co-supplementing HDL on SAA-mediated changes to vascular and renal function in apolipoprotein E-deficient (ApoE-/-) mice in the absence of a high-fat diet. Male ApoE-/- mice received recombinant human SAA or vehicle (control) by intraperitoneal (i.p.) injection every three days for two weeks with or without freshly isolated human HDL supplemented by intravenous (i.v.) injection in the two weeks preceding SAA stimulation. Aorta and kidney were harvested 4 or 18 weeks after commencement of treatment. At 4 weeks after commencement of treatment, SAA increased aortic vascular cell adhesion molecule (VCAM)-1 expression and F2-isoprostane level and decreased cyclic guanosine monophosphate (cGMP), consistent with SAA stimulating endothelial dysfunction and promoting atherosclerosis. SAA also stimulated renal injury and inflammation that manifested as increased urinary protein, kidney injury molecule (KIM)-1, and renal tissue cytokine/chemokine levels as well as increased protein tyrosine chlorination and P38 MAPkinase activation and decreased in Bowman's space, confirming that SAA elicited a pro-inflammatory phenotype in the kidney. At 18 weeks, vascular lesions increased significantly in the cohort of ApoE-/- mice treated with SAA alone. By contrast, pretreatment of mice with HDL decreased SAA pro-inflammatory activity, inhibited SAA enhancement of aortic lesion size and renal function, and prevented changes to glomerular Bowman's space. Taken together, these data indicate that supplemented HDL reduces SAA-mediated endothelial and renal dysfunction in an atherosclerosis-prone mouse model.
Collapse
|
174
|
Xu J, Sun Y, Lu J. Knockdown of Long Noncoding RNA (lncRNA) AK094457 Relieved Angiotensin II Induced Vascular Endothelial Cell Injury. Med Sci Monit 2020; 26:e919854. [PMID: 32027625 PMCID: PMC7020760 DOI: 10.12659/msm.919854] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Hypertension could induce many serious diseases, including damage to vascular endothelial cells. As a non-coding RNA, long noncoding RNA (lncRNA) has received much attention in scientific research and has a regulating efficacy on many critical life activities in human body. The level of lncRNA AK094457 is thought to be elevated in hypertensive rats. However, there is no research indicating the relationship between the level of lncRNA AK094457 and vascular endothelial injury. Material/Methods In our study, we used lentiviral to knockdown lncRNA AK094457, and the human umbilical vein endothelial cells (HUVECs) were stimulated by the Ang II to imitate the vascular endothelial cell damage caused by hypertension. The Cell Counting Kit-8 assays were used to detect the cells viability. Western blotting was performed to detect the endothelial nitric oxide synthase (eNOS), p-eNOS and endothelin-1 (ET-1). After that the production of the NO was monitored. At last, the reactive oxygen species (ROS) levels and apoptosis rates were detected in this study. Results According to the results, we found that knockdown lncRNA AK094457 could alleviate the decrease of vascular endothelial cell viability induced by angiotensin II (Ang II). The knockdown of lncRNA AK094457 also relieved the downregulation of eNOS and p-eNOS, and the decreasing of NO release. At the same time, the knockdown of lncRNA inhibited the levels of Ang II-induced proinflammatory cytokines (tumor necrosis factor [TNF]-α, interleukin [IL]-1, and IL-6) and cell adhesion molecules (vascular cell adhesion molecule 1 [VCAM-1], intercellular adhesion molecule 1 [ICAM-1], and monocyte chemoattractant protein-1 [MCP-1]). The levels of ROS and apoptosis rates also decreased after the knockdown of lncRNA AK094457. Conclusions All these results indicated that lncRNA AK094457 could promote Ang II-induced vascular endothelial cell injury. On the contrary, knockdown of lncRNA AK094457 could alleviate this damage.
Collapse
Affiliation(s)
- JiaYi Xu
- Department of Gerontology, Minhang Hospital, Fudan University, Shanghai, China (mainland)
| | - Yingjie Sun
- Department of Critical Care Medicine, Minhang Hospital, Fudan University, Shanghai, China (mainland)
| | - Jie Lu
- Department of Gerontology, Minhang Hospital, Fudan University, Shanghai, China (mainland)
| |
Collapse
|
175
|
Peptide-based nanosystems for vascular cell adhesion molecule-1 targeting: a real opportunity for therapeutic and diagnostic agents in inflammation associated disorders. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2019.101461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
176
|
Turgeon PJ, Chan GC, Chen L, Jamal AN, Yan MS, Ho JJD, Yuan L, Ibeh N, Ku KH, Cybulsky MI, Aird WC, Marsden PA. Epigenetic Heterogeneity and Mitotic Heritability Prime Endothelial Cell Gene Induction. THE JOURNAL OF IMMUNOLOGY 2020; 204:1173-1187. [DOI: 10.4049/jimmunol.1900744] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/23/2019] [Indexed: 01/08/2023]
|
177
|
Cai L, Zhang X, Hou M, Gao F. Natural flavone tricetin suppresses oxidized LDL-induced endothelial inflammation mediated by Egr-1. Int Immunopharmacol 2020; 80:106224. [PMID: 31991371 DOI: 10.1016/j.intimp.2020.106224] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 01/02/2020] [Accepted: 01/16/2020] [Indexed: 02/08/2023]
Abstract
Atherosclerosis is the primary cause of many cardiovascular diseases. Endothelial dysfunction is recognized as a crucial early event in atherosclerotic lesion formation. Tricetin is a natural flavonoid derivative that has demonstrated a wide range of therapeutic properties. This study investigates the protective effect of tricetin in cultured endothelial cells. The results of our study show that tricetin suppressed oxidized low-density lipoprotein (ox-LDL)-induced expression of pro-inflammatory monocyte chemotactic protein-1 (MCP-1) and interleukin-1β (IL-1β), as well as the generation of reactive oxygen species (ROS). Furthermore, our findings indicate that tricetin suppressed ox-LDL-induced expression of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1). At the cellular level, the presence of tricetin inhibited ox-LDL-induced monocyte adhesion to endothelial cells. Mechanistically, we showed that tricetin suppressed the induction of the endothelial receptor for ox-LDL, lectin-like ox-LDL receptor-1 (LOX-1), and the transcriptional factor early growth response 1 (Egr-1) as well as extracellular signal-regulated protein kinase 1 and 2 (ERK1/2) activation. These data demonstrate that tricetin is a natural protective agent in vascular endothelial cells, indicating that tricetin could have a potentially beneficial effect in the modulation of atherosclerosis.
Collapse
Affiliation(s)
- Luming Cai
- Department of Cardiovascular Medicine, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, PR China
| | - Xuesong Zhang
- Department of Central Laboratory, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, PR China
| | - Meiling Hou
- Department of Cardiovascular Medicine, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, PR China
| | - Feng Gao
- Hospital Office, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, PR China.
| |
Collapse
|
178
|
Abstract
Testosterone is the main male sex hormone and is essential for the maintenance of male secondary sexual characteristics and fertility. Androgen deficiency in young men owing to organic disease of the hypothalamus, pituitary gland or testes has been treated with testosterone replacement for decades without reports of increased cardiovascular events. In the past decade, the number of testosterone prescriptions issued for middle-aged or older men with either age-related or obesity-related decline in serum testosterone levels has increased exponentially even though these conditions are not approved indications for testosterone therapy. Some retrospective studies and randomized trials have suggested that testosterone replacement therapy increases the risk of cardiovascular disease, which has led the FDA to release a warning statement about the potential cardiovascular risks of testosterone replacement therapy. However, no trials of testosterone replacement therapy published to date were designed or adequately powered to assess cardiovascular events; therefore, the cardiovascular safety of this therapy remains unclear. In this Review, we provide an overview of epidemiological data on the association between serum levels of endogenous testosterone and cardiovascular disease, prescription database studies on the risk of cardiovascular disease in men receiving testosterone therapy, randomized trials and meta-analyses evaluating testosterone replacement therapy and its association with cardiovascular events and mechanistic studies on the effects of testosterone on the cardiovascular system. Our aim is to help clinicians to make informed decisions when considering testosterone replacement therapy in their patients.
Collapse
|
179
|
Haas MJ, Jurado-Flores M, Hammoud R, Feng V, Gonzales K, Onstead-Haas L, D Mooradian A. Inhibition of Pro-Inflammatory Cytokine Secretion by Select Antioxidants in Human Coronary Artery Endothelial Cells. INT J VITAM NUTR RES 2020; 90:103-112. [DOI: 10.1024/0300-9831/a000520] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Abstract. Inflammatory and oxidative stress in endothelial cells are implicated in the pathogenesis of premature atherosclerosis in diabetes. To determine whether high-dextrose concentrations induce the expression of pro-inflammatory cytokines, human coronary artery endothelial cells (HCAEC) were exposed to either 5.5 or 27.5 mM dextrose for 24-hours and interleukin-1β (IL-1β), interleukin-2 (IL-2), interleukin-6 (IL-6), interleukin-8 (IL-8), and tumor necrosis factor α (TNF α) levels were measured by enzyme immunoassays. To determine the effect of antioxidants on inflammatory cytokine secretion, cells were also treated with α-tocopherol, ascorbic acid, and the glutathione peroxidase mimetic ebselen. Only the concentration of IL-1β in culture media from cells exposed to 27.5 mM dextrose increased relative to cells maintained in 5.5 mM dextrose. Treatment with α-tocopherol (10, 100, and 1,000 μM) and ascorbic acid (15, 150, and 1,500 μM) at the same time that the dextrose was added reduced IL-1β, IL-6, and IL-8 levels in culture media from cells maintained at 5.5 mM dextrose but had no effect on IL-1β, IL-6, and IL-8 levels in cells exposed to 27.5 mM dextrose. However, ebselen treatment reduced IL-1β, IL-6, and IL-8 levels in cells maintained in either 5.5 or 27.5 mM dextrose. IL-2 and TNF α concentrations in culture media were below the limit of detection under all experimental conditions studied suggesting that these cells may not synthesize detectable quantities of these cytokines. These results suggest that dextrose at certain concentrations may increase IL-1β levels and that antioxidants have differential effects on suppressing the secretion of pro-inflammatory cytokines in HCAEC.
Collapse
Affiliation(s)
- Michael J. Haas
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Florida Jacksonville College of Medicine, Jacksonville, FL 32209
| | - Marilu Jurado-Flores
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Florida Jacksonville College of Medicine, Jacksonville, FL 32209
| | - Ramadan Hammoud
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Florida Jacksonville College of Medicine, Jacksonville, FL 32209
| | - Victoria Feng
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Florida Jacksonville College of Medicine, Jacksonville, FL 32209
| | - Krista Gonzales
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Florida Jacksonville College of Medicine, Jacksonville, FL 32209
| | - Luisa Onstead-Haas
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Florida Jacksonville College of Medicine, Jacksonville, FL 32209
| | - Arshag D Mooradian
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Florida Jacksonville College of Medicine, Jacksonville, FL 32209
| |
Collapse
|
180
|
Yang X, Wan M, Cheng Z, Wang Z, Wu Q. Tofacitinib inhibits ox-LDL-induced adhesion of THP-1 monocytes to endothelial cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2775-2782. [PMID: 31284768 DOI: 10.1080/21691401.2019.1573740] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of the blood vasculature. Endothelial dysfunction is an early event in the development of atherosclerosis and the endothelium plays an important role in the innate immune defense in the pathology of cardiovascular diseases. New therapies are being developed based on the involvement of the immune system in atherosclerosis. In this study, we demonstrate that a commonly used anti-rheumatic drug, tofacitinib, possesses vascular protective properties in cultured primary human aortic endothelial cells (HAECs). Tofacitinib ameliorates oxidized low-density lipoprotein (ox-LDL)-induced adhesion of THP-1 cells to HAECs, suppresses the expression of vascular adhesion molecules and production of cytokines, including vascular cell adhesion molecule 1 (VCAM-1), intercellular cell adhesion molecule-1 (ICAM-1), tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β). Moreover, tofacitinib inhibits elevation of endothelial lectin-like ox-LDL receptor-1 (LOX-1) and production of reactive oxygen species (ROS) triggered by ox-LDL. As a result, the presence of tofacitinib reduces ox-LDL-induced cytotoxicity and improves endothelial viability. Mechanistically, we demonstrate that tofacitinib suppresses ox-LDL-mediated activation of NF-κB inhibitor α (IκB-α), accumulation of nuclear p65 and activation of nuclear factor κB (NF-κB) promoter, indicating that tofacitinib inhibits NF-κB activation. Collectively, our data support that tofacitinib possesses a novel protective function in endothelial cells, implying that tofacitinib could have the therapeutic potential to modulate inflammation in cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaoyang Yang
- a Department of Hematology, Affiliated Haikou Hospital Xiangya School Central South University and Haikou Municipal Municipal People's Hospital , Haikou , Hainan , China
| | - Mengjie Wan
- a Department of Hematology, Affiliated Haikou Hospital Xiangya School Central South University and Haikou Municipal Municipal People's Hospital , Haikou , Hainan , China
| | - Zhiyong Cheng
- a Department of Hematology, Affiliated Haikou Hospital Xiangya School Central South University and Haikou Municipal Municipal People's Hospital , Haikou , Hainan , China
| | - Zhiming Wang
- a Department of Hematology, Affiliated Haikou Hospital Xiangya School Central South University and Haikou Municipal Municipal People's Hospital , Haikou , Hainan , China
| | - Qingxia Wu
- b Department of Nursing, Affiliated Haikou Hospital Xiangya School Central South University and Haikou Municipal Municipal People's Hospital , Haikou , Hainan , China
| |
Collapse
|
181
|
Xu H, Jiang J, Chen W, Li W, Chen Z. Vascular Macrophages in Atherosclerosis. J Immunol Res 2019; 2019:4354786. [PMID: 31886303 PMCID: PMC6914912 DOI: 10.1155/2019/4354786] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/19/2019] [Accepted: 10/23/2019] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is the main pathological basis for the occurrence of most cardiovascular diseases, the leading global health threat, and a great burden for society. It has been well established that atherosclerosis is not only a metabolic disorder but also a chronic, sterile, and maladaptive inflammatory process encompassing both innate and adaptive immunity. Macrophages, the major immune cell population in atherosclerotic lesions, have been shown to play critical roles in all stages of atherosclerosis, including the initiation and progression of advanced atherosclerosis. Macrophages have emerged as a novel potential target for antiatherosclerosis therapy. In addition, the macrophage phenotype is greatly influenced by microenvironmental stimuli in the plaques and presents complex heterogeneity. This article reviews the functions of macrophages in different stages of atherosclerosis, as well as the phenotypes and functions of macrophage subsets. New treatment strategies based on macrophage-related inflammation are also discussed.
Collapse
Affiliation(s)
- Hailin Xu
- Department of General Surgery, The First People's Hospital of Jiande, Hangzhou, China
| | - Jingxin Jiang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wuzhen Chen
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| | - Wenlu Li
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Zhigang Chen
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou, China
| |
Collapse
|
182
|
Badmus OO, Olatunji LA. Glucocorticoid exposure causes disrupted glucoregulation, cardiac inflammation and elevated dipeptidyl peptidase-4 activity independent of glycogen synthase kinase-3 in female rats. Arch Physiol Biochem 2019; 125:414-422. [PMID: 29912577 DOI: 10.1080/13813455.2018.1479426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Objective: We tested the hypothesis that glucocorticoid (GC) exposure in female rats would lead to glucose dysregulation and elevated cardiac inflammatory biomarkers, which are dipeptidyl peptidase-4 (DPP-4)- and glycogen synthase kinase-3 (GSK-3)-dependent. Methods: Female Wistar rats received vehicle (control; n = 6) or GC (dexamethasone; n = 6; 0.2 mg/kg; p.o.) for six days. Insulin resistance was determined by HOMA-IR. DPP-4 activity was determined by fluorescence method, whereas vascular cell adhesion molecule-1 (VCAM-1), uric acid, malondialdehyde (MDA), lactate dehydrogenase (LDH) and nitric oxide (NO) from plasma and cardiac homogenate were estimated as cardiac pro-inflammatory biomarkers. Results: Results showed that GC exposure resulted in glucose dysregulation and increased plasma and cardiac pro-inflammatory markers which are associated with elevated DPP-4 activity but reduced GSK-3. Conclusions: The present results demonstrate that GC exposure would cause glucose dysregulation, increased DPP-4 activity and cardiac inflammation that is independent of GSK-3.
Collapse
Affiliation(s)
- Olufunto O Badmus
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin , Ilorin , Nigeria
- Department of Public Health, Kwara State University , Malete , Nigeria
| | - Lawrence A Olatunji
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin , Ilorin , Nigeria
| |
Collapse
|
183
|
Sun Z, Pang S, Cui Y, Yan B. Genetic and Functional Variants Analysis of the GATA6 Gene Promoter in Acute Myocardial Infarction. Front Genet 2019; 10:1100. [PMID: 31781165 PMCID: PMC6851265 DOI: 10.3389/fgene.2019.01100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 10/11/2019] [Indexed: 01/13/2023] Open
Abstract
Background: Acute myocardial infarction (AMI) which is a specific type of coronary artery disease (CAD), is caused by the combination of genetic factors and acquired environment. Although some common genetic variations have been recorded to contribute to the development of CAD and AMI, more genetic factors and potential molecular mechanisms remain largely unknown. The GATA6 gene is expressed in the heart during embryogenesis and is also detected in vascular smooth muscle cells (VSMCs), different human primary endothelial cells (ECs), and vascular ECs in mice. To date, no studies have directly linked GATA6 gene with regulation of the CAD. Methods: In this study, we used a case-control study to investigate and analyze the genetic variations and functional variations of the GATA6 gene promoter region in AMI patients and controls. A variety of statistical analysis methods were utilized to analyze the association of single nucleotide polymorphisms (SNPs) with AMI. Functional analysis of DNA sequence variants (DSVs) was performed using a dual luciferase reporter assay. In vitro, electrophoretic mobility shift assay (EMSA) was selected to examine DNA-protein interactions. Results: A total of 705 subjects were enrolled in the study. Ten DSVs were found in AMI patients (n = 352) and controls (n = 353), including seven SNPs. One novel heterozygous DSV, (g.22168409 A > G), and two SNPs, [g.22168362 C > A(rs1416421760) and g.22168521 G > T(rs1445501474)], were reported in three AMI patients, which were not found in controls. The relevant statistical analysis, including allele and genotype frequencies between AMI patients and controls, five genetic models, linkage disequilibrium (LD) and haplotype analysis, and SNP–SNP interactions, suggested no statistical significance (P > 0.05). The transcriptional activity of GATA6 gene promoter was significantly increased by the DSV (g.22168409 A > G) and SNP [g.22168362 C > A(rs1416421760)]. The EMSA revealed that the DSV (g.22168409 A > G) and SNP [g.22168362 C > A(rs1416421760)] evidently influenced the binding of transcription factors. Conclusion: In conclusion, the DSV (g.22168409 A > G) and SNP [g.22168362 C > A(rs1416421760)] may increase GATA6 levels in both HEK-293 and H9c2 cell lines by affecting the binding of transcription factors. Whether the two variants identified in the GATA6 gene promoter can promote the development and progression of human AMI by altering GATA6 levels still requires further studies to verify.
Collapse
Affiliation(s)
- Zhaoqing Sun
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shuchao Pang
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Yinghua Cui
- Division of Cardiology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China.,The Center for Molecular Genetics of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Bo Yan
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China.,The Center for Molecular Genetics of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China.,Shandong Provincial Sino-US Cooperation Research Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| |
Collapse
|
184
|
Zhang L, Wang X, Zhang L, Virgous C, Si H. Combination of curcumin and luteolin synergistically inhibits TNF-α-induced vascular inflammation in human vascular cells and mice. J Nutr Biochem 2019; 73:108222. [DOI: 10.1016/j.jnutbio.2019.108222] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/01/2019] [Accepted: 07/30/2019] [Indexed: 01/24/2023]
|
185
|
Melchinger H, Jain K, Tyagi T, Hwa J. Role of Platelet Mitochondria: Life in a Nucleus-Free Zone. Front Cardiovasc Med 2019; 6:153. [PMID: 31737646 PMCID: PMC6828734 DOI: 10.3389/fcvm.2019.00153] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 10/08/2019] [Indexed: 12/19/2022] Open
Abstract
Platelets are abundant, small, anucleate circulating cells, serving many emerging pathophysiological roles beyond hemostasis; including active critical roles in thrombosis, injury response, and immunoregulation. In the absence of genomic DNA transcriptional regulation (no nucleus), platelets require strategic prepackaging of all the needed RNA and organelles from megakaryocytes, to sense stress (e.g., hyperglycemia), to protect themselves from stress (e.g., mitophagy), and to communicate a stress response to other cells (e.g., granule and microparticle release). Distinct from avian thrombocytes that have a nucleus, the absence of a nucleus allows the mammalian platelet to maintain its small size, permits morphological flexibility, and may improve speed and efficiency of protein expression in response to stress. In the absence of a nucleus, platelet lifespan of 7–10 days, is largely determined by the mitochondria. The packaging of 5–8 mitochondria is critical in aerobic respiration and yielding metabolic substrates needed for function and survival. Mitochondria damage or dysfunction, as observed with several disease processes, results in greatly attenuated platelet survival and increased risk for thrombovascular events. Here we provide insights into the emerging roles of platelets despite the lack of a nucleus, and the key role played by mitochondria in platelet function and survival both in health and disease.
Collapse
Affiliation(s)
- Hannah Melchinger
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| | - Kanika Jain
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| | - Tarun Tyagi
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| | - John Hwa
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
186
|
Herrero-Fernandez B, Gomez-Bris R, Somovilla-Crespo B, Gonzalez-Granado JM. Immunobiology of Atherosclerosis: A Complex Net of Interactions. Int J Mol Sci 2019; 20:E5293. [PMID: 31653058 PMCID: PMC6862594 DOI: 10.3390/ijms20215293] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is the leading cause of mortality worldwide, and atherosclerosis the principal factor underlying cardiovascular events. Atherosclerosis is a chronic inflammatory disease characterized by endothelial dysfunction, intimal lipid deposition, smooth muscle cell proliferation, cell apoptosis and necrosis, and local and systemic inflammation, involving key contributions to from innate and adaptive immunity. The balance between proatherogenic inflammatory and atheroprotective anti-inflammatory responses is modulated by a complex network of interactions among vascular components and immune cells, including monocytes, macrophages, dendritic cells, and T, B, and foam cells; these interactions modulate the further progression and stability of the atherosclerotic lesion. In this review, we take a global perspective on existing knowledge about the pathogenesis of immune responses in the atherosclerotic microenvironment and the interplay between the major innate and adaptive immune factors in atherosclerosis. Studies such as this are the basis for the development of new therapies against atherosclerosis.
Collapse
Affiliation(s)
- Beatriz Herrero-Fernandez
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
| | - Raquel Gomez-Bris
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
| | | | - Jose Maria Gonzalez-Granado
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares, 28029 Madrid, Spain.
| |
Collapse
|
187
|
Taghizadeh E, Taheri F, Renani PG, Reiner Ž, Navashenaq JG, Sahebkar A. Macrophage: A Key Therapeutic Target in Atherosclerosis? Curr Pharm Des 2019; 25:3165-3174. [DOI: 10.2174/1381612825666190830153056] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/22/2019] [Indexed: 12/19/2022]
Abstract
Background:
Atherosclerosis is a chronic inflammatory disease and a leading cause of coronary artery
disease, peripheral vascular disease and stroke. Lipid-laden macrophages are derived from circulating monocytes
and form fatty streaks as the first step of atherogenesis.
Methods:
An electronic search in major databases was performed to review new therapeutic opportunities for
influencing the inflammatory component of atherosclerosis based on monocytes/macrophages targeting.
Results:
In the past two decades, macrophages have been recognized as the main players in atherogenesis but also
in its thrombotic complications. There is a growing interest in immunometabolism and recent studies on metabolism
of macrophages have created new therapeutic options to treat atherosclerosis. Targeting recruitment, polarization,
cytokine profile extracellular matrix remodeling, cholesterol metabolism, oxidative stress, inflammatory
activity and non-coding RNAs of monocyte/macrophage have been proposed as potential therapeutic approaches
against atherosclerosis.
Conclusion:
Monocytes/macrophages have a crucial role in progression and pathogenesis of atherosclerosis.
Therefore, targeting monocyte/macrophage therapy in order to achieve anti-inflammatory effects might be a good
option for prevention of atherosclerosis.
Collapse
Affiliation(s)
- Eskandar Taghizadeh
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Forough Taheri
- Sharekord Branch, Islamic Azad University, Sharekord, Iran
| | | | - Željko Reiner
- University Hospital Centre Zagreb, School of Medicine, University of Zagreb, Department of Internal Medicine, Zagreb, Croatia
| | - Jamshid G. Navashenaq
- Immunogenetic and Cell Culture Department, Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
188
|
Glucagon-like peptide-1 receptor agonist dulaglutide prevents ox-LDL-induced adhesion of monocytes to human endothelial cells: An implication in the treatment of atherosclerosis. Mol Immunol 2019; 116:73-79. [PMID: 31630078 DOI: 10.1016/j.molimm.2019.09.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 09/09/2019] [Accepted: 09/29/2019] [Indexed: 02/06/2023]
Abstract
Atherosclerosis is a common comorbidity of type II diabetes and a leading cause of death worldwide. The presence of oxidized low-density lipoprotein (ox-LDL) drives atherogenesis by inducing oxidative stress, mitochondrial dysfunction, expression of proinflammatory cytokines and chemokines including interleukin (IL)-1β, IL-6, and monocyte chemoattractant protein 1 (MCP-1), adhesion molecules including vascular cellular adhesion molecule 1 (VCAM-1) and E-selectin, and downregulating expression of the Krüppel-like factor 2 (KLF2) transcription factor. Importantly, ox-LDL induced the attachment of THP-1 monocytes to endothelial cells. In the present study, we demonstrate for the first time that the specific glucagon-like peptide 1 receptor (GLP-1R) agonist dulaglutide may prevent these atherosclerotic effects of ox-LDL by preventing suppression of KLF2 by p53 protein in human aortic endothelial cells. KLF2 has been shown to play a major role in protecting vascular endothelial cells from damage induced by ox-LDL and oscillatory shear, and therefore, therapies capable of mediating KLF2 signaling may be an attractive treatment option for preventing the development and progression of atherosclerosis.
Collapse
|
189
|
Zeng X, Li X, Chen Z, Yao Q. DPP-4 inhibitor saxagliptin ameliorates oxygen deprivation/reoxygenation-induced brain endothelial injury. Am J Transl Res 2019; 11:6316-6325. [PMID: 31737185 PMCID: PMC6834500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 01/29/2019] [Indexed: 06/10/2023]
Abstract
Cardiovascular diseases are the main cause of death and disability among diabetes patients. Atherosclerosis-associated stroke is one of the most severe complications in diabetes patients. DPP-4 inhibitors are a class of potent anti-glycemic agents used to treat diabetes. Recently, some DPP-4 inhibitors have been shown to have cardiovascular benefits. In this study, we reveal that saxagliptin, one of the most widely used DPP-4 inhibitors, exhibits vascular protective effects against oxygen and glucose depletion/reoxygenation (OGD/R) in human brain vascular endothelial cells. Our data show that DPP-4 is fairly expressed in brain endothelial cells and its expression is induced by OGD/R. The results of MTT assay show that inhibition of DPP-4 by saxagliptin ameliorates OGD/R-induced reduced cell viability, and LDH assay demonstrated that saxagliptin reduces cellular toxicity. Furthermore, we show that saxagliptin mitigates OGD/R-induced collapse of mitochondrial membrane potential (MMP). Saxagliptin also reduces oxidative stress-induced release of 4-HNE and the NAPDH oxidase catalytic subunit NOX-4. At the molecular level, saxagliptin suppresses OGD/R-induced expression of pro-inflammatory cytokines and production of vascular adhesion molecules including tumor necrosis factor-α (TNF-α), interleukin (IL)-6, monocyte chemoattractant protein 1 (MCP-1), vascular cellular adhesion molecule 1 (VCAM-1), and E-selectin. Mechanistically, saxagliptin inhibits activation of the NF-κB pathway by OGD/R via its inhibitory effect on nuclear p65 and NF-κB promoter activity. Collectively, our study explicitly demonstrates the cellular protective effect of saxagliptin against OGD/R-induced brain endothelial injury. Our findings extend our recognition of the protective roles of DPP-4 inhibitors in brain vascular cells.
Collapse
Affiliation(s)
- Xudong Zeng
- Department of Neurosurgery, Luoyang Central Hospital Affiliated to Zhengzhou University Luoyang 471039, Henan Province, China
| | - Xiaohui Li
- Department of Neurosurgery, Luoyang Central Hospital Affiliated to Zhengzhou University Luoyang 471039, Henan Province, China
| | - Zhenbo Chen
- Department of Neurosurgery, Luoyang Central Hospital Affiliated to Zhengzhou University Luoyang 471039, Henan Province, China
| | - Qinghe Yao
- Department of Neurosurgery, Luoyang Central Hospital Affiliated to Zhengzhou University Luoyang 471039, Henan Province, China
| |
Collapse
|
190
|
Farzaei MH, Singh AK, Kumar R, Croley CR, Pandey AK, Coy-Barrera E, Kumar Patra J, Das G, Kerry RG, Annunziata G, Tenore GC, Khan H, Micucci M, Budriesi R, Momtaz S, Nabavi SM, Bishayee A. Targeting Inflammation by Flavonoids: Novel Therapeutic Strategy for Metabolic Disorders. Int J Mol Sci 2019; 20:4957. [PMID: 31597283 PMCID: PMC6801776 DOI: 10.3390/ijms20194957] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 09/28/2019] [Accepted: 09/30/2019] [Indexed: 12/16/2022] Open
Abstract
A balanced metabolic profile is essential for normal human physiological activities. Disproportions in nutrition give rise to imbalances in metabolism that are associated with aberrant immune function and an elevated risk for inflammatory-associated disorders. Inflammation is a complex process, and numerous mediators affect inflammation-mediated disorders. The available clinical modalities do not effectively address the underlying diseases but rather relieve the symptoms. Therefore, novel targeted agents have the potential to normalize the metabolic system and, thus, provide meaningful therapy to the underlying disorder. In this connection, polyphenols, the well-known and extensively studied phytochemical moieties, were evaluated for their effective role in the restoration of metabolism via various mechanistic signaling pathways. The various flavonoids that we observed in this comprehensive review interfere with the metabolic events that induce inflammation. The mechanisms via which the polyphenols, in particular flavonoids, act provide a promising treatment option for inflammatory disorders. However, detailed clinical studies of such molecules are required to decide their clinical fate.
Collapse
Affiliation(s)
- Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran.
| | - Amit Kumar Singh
- Department of Biochemistry, University of Allahabad, Allahabad 211 002, India.
| | - Ramesh Kumar
- Department of Biochemistry, University of Allahabad, Allahabad 211 002, India.
| | - Courtney R Croley
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA.
| | - Abhay K Pandey
- Department of Biochemistry, University of Allahabad, Allahabad 211 002, India.
| | - Ericsson Coy-Barrera
- Bioorganic Chemistry Laboratory, Facultad de Ciencias Básicas y Aplicadas, Campus Nueva Granada, Universidad Militar Nueva Granada, Cajicá 250247, Colombia.
| | - Jayanta Kumar Patra
- Research Institute of Biotechnology and Medical Converged Science, Dongguk University-Seoul, Ilsandong-gu, Gyeonggi-do 10326, Korea.
| | - Gitishree Das
- Research Institute of Biotechnology and Medical Converged Science, Dongguk University-Seoul, Ilsandong-gu, Gyeonggi-do 10326, Korea.
| | - Rout George Kerry
- Post Graduate Department of Biotechnology, Utkal University, Vani Vihar, Bhubaneswar 751 004, Odisha, India.
| | - Giuseppe Annunziata
- Department of Pharmacy, University of Naples "Federico II", Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Gian Carlo Tenore
- Department of Pharmacy, University of Naples "Federico II", Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan.
| | - Matteo Micucci
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, 40126 Bologna, Italy.
| | - Roberta Budriesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, 40126 Bologna, Italy.
| | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, Karaj 141554364, Iran.
- Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences, Tehran University of Medical Sciences, Tehran 141556451, Iran.
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran.
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA.
| |
Collapse
|
191
|
Mury P, Chirico EN, Mura M, Millon A, Canet-Soulas E, Pialoux V. Oxidative Stress and Inflammation, Key Targets of Atherosclerotic Plaque Progression and Vulnerability: Potential Impact of Physical Activity. Sports Med 2019; 48:2725-2741. [PMID: 30302720 DOI: 10.1007/s40279-018-0996-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, a complex cardiovascular disease, is a leading cause of mortality and morbidity worldwide. Oxidative stress and inflammation are both involved in the development of atherosclerotic plaque as they increase the biological processes associated with this pathology, such as endothelial dysfunction and macrophage recruitment and adhesion. Atherosclerotic plaque rupture leading to major ischemic events is the result of vulnerable plaque progression, which is a result of the detrimental effect of oxidative stress and inflammation on risk factors for atherosclerotic plaque rupture, such as intraplaque hemorrhage, neovascularization, and fibrous cap thickness. Thus, both are key targets for primary and secondary interventions. It is well recognized that chronic physical activity attenuates oxidative stress in healthy subjects via the improvement of antioxidant enzyme capacities and inflammation via the enhancement of anti-inflammatory molecules. Moreover, it was recently shown that chronic physical activity could decrease oxidative stress and inflammation in atherosclerotic patients. The aim of this review is to summarize the role of oxidative stress and inflammation in atherosclerosis and the results of therapeutic interventions targeting them in both preclinical and clinical studies. The effects of chronic physical activity on these two key processes are then reviewed in vulnerable atherosclerotic plaques in both coronary and carotid arteries.
Collapse
Affiliation(s)
- Pauline Mury
- Team Vascular Biology and Red Blood Cell, Interuniversity Laboratory of Human Movement Biology, University Claude Bernard Lyon 1, University of Lyon, Faculté de Médecine Lyon Est, 8 Avenue Rockefeller, 69008, Lyon, France.,Laboratory of Excellence GR-Ex, Paris, France
| | - Erica N Chirico
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Mathilde Mura
- Team Vascular Biology and Red Blood Cell, Interuniversity Laboratory of Human Movement Biology, University Claude Bernard Lyon 1, University of Lyon, Faculté de Médecine Lyon Est, 8 Avenue Rockefeller, 69008, Lyon, France.,Laboratory of Excellence GR-Ex, Paris, France
| | - Antoine Millon
- University of Lyon, University Claude Bernard Lyon 1, CarMeN Laboratory, INSERM U1060, Bron, France.,Department of Vascular Surgery, Edouard Herriot Hospital, Lyon, France
| | - Emmanuelle Canet-Soulas
- University of Lyon, University Claude Bernard Lyon 1, CarMeN Laboratory, INSERM U1060, Bron, France
| | - Vincent Pialoux
- Team Vascular Biology and Red Blood Cell, Interuniversity Laboratory of Human Movement Biology, University Claude Bernard Lyon 1, University of Lyon, Faculté de Médecine Lyon Est, 8 Avenue Rockefeller, 69008, Lyon, France. .,Laboratory of Excellence GR-Ex, Paris, France. .,Institut Universitaire de France, Paris, France.
| |
Collapse
|
192
|
Nagarajan UM, Sikes JD, Burris RL, Jha R, Popovic B, Fraungruber P, Hennings L, Haggerty CL, Nagarajan S. Genital Chlamydia infection in hyperlipidemic mouse models exacerbates atherosclerosis. Atherosclerosis 2019; 290:103-110. [PMID: 31604170 DOI: 10.1016/j.atherosclerosis.2019.09.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 09/11/2019] [Accepted: 09/26/2019] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND AIMS Atherosclerosis is a chronic inflammatory disease, and recent studies have shown that infection at remote sites can contribute to the progression of atherosclerosis in hyperlipidemic mouse models. In this report, we tested the hypothesis that genital Chlamydia infection could accelerate the onset and progression of atherosclerosis. METHODS Apolipoprotein E (Apoe-/-) and LDL receptor knockout (Ldlr-/-) mice on a high-fat diet were infected intra-vaginally with Chlamydia muridarum. Atherosclerotic lesions on the aortic sinuses and in the descending aorta were assessed at 8-weeks post-infection. Systemic, macrophage, and vascular site inflammatory responses were assessed and quantified. RESULTS Compared to the uninfected groups, infected Apoe-/- and Ldlr-/- mice developed significantly more atherosclerotic lesions in the aortic sinus and in the descending aorta. Increased lesions were associated with higher circulating levels of serum amyloid A-1, IL-1β, TNF-α, and increased VCAM-1 expression in the aortic sinus, suggesting an association with inflammatory responses observed during C. muridarum infection. Genital infection courses were similar in Apoe-/-, Ldlr-/-, and wild type mice. Further, Apoe-/- mice developed severe uterine pathology with increased dilatations. Apoe-deficiency also augmented cytokine/chemokine response in C. muridarum infected macrophages, suggesting that the difference in macrophage response could have contributed to the genital pathology in Apoe-/- mice. CONCLUSIONS Overall, these studies demonstrate that genital Chlamydia infection exacerbates atherosclerotic lesions in hyperlipidemic mouse and suggest a novel role for Apoe in full recovery of uterine anatomy after chlamydial infection.
Collapse
Affiliation(s)
- Uma M Nagarajan
- Department of Pediatrics and Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA
| | - James D Sikes
- Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA
| | - Ramona L Burris
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA
| | - Rajneesh Jha
- Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA
| | - Branimir Popovic
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Pamelia Fraungruber
- Department of Pathology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Leah Hennings
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Catherine L Haggerty
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Shanmugam Nagarajan
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, AR, USA; Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pathology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
193
|
Li R, Dong Z, Zhuang X, Liu R, Yan F, Chen Y, Gao X, Shi H. Salidroside prevents tumor necrosis factor-α-induced vascular inflammation by blocking mitogen-activated protein kinase and NF-κB signaling activation. Exp Ther Med 2019; 18:4137-4143. [PMID: 31656544 DOI: 10.3892/etm.2019.8064] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 09/10/2019] [Indexed: 02/07/2023] Open
Abstract
Vascular inflammation is a key factor in the pathogenesis of atherosclerosis. Salidroside is an important active ingredient extracted from the root of the Rhodiola rosea plant, which has been reported to have antioxidative, anti-cancer, neuroprotective and cardioprotective effects. However, the effects of salidroside on vascular inflammation have not been clarified. The purpose of the present study was to investigate the protective effects of salidroside against tumor necrosis factor (TNF)-α-induced vascular inflammation in cardiac microvascular endothelial cells (CMECs), a specific cell type derived from coronary micro-vessels. Over a 24-h period, salidroside did not exert any significant cytotoxicity up to a dose of 100 µM. Additionally, salidroside decreased the expression levels of the cell adhesion molecule vascular cell adhesion molecule-1 (VCAM-1) in TNF-α-stimulated CMECs, thus suppressing monocyte-to-CMEC adhesion. Salidroside also decreased the production of inflammatory cytokines such as interleukin (IL)-1β, IL-6 and monocyte chemotactic protein 1 (MCP-1) in TNF-α-induced CMECs, as well as suppressing TNF-α-activated mitogen-activated protein kinase (MAPK) and NF-κB activation. Since MAPKs and NF-κB both serve notable roles in regulating the expression of VCAM-1, IL-1β, IL-6 and MCP-1, the present study provided a preliminary understanding of the mechanism underlying the protective effects of salidroside. Overall, salidroside alleviated vascular inflammation by mediating MAPK and NF-κB activation in TNF-α-induced CMECs. These results indicated that salidroside may have potential applications as a therapeutic agent against vascular inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Ruoshui Li
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai 200036, P.R. China
| | - Zhen Dong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Xinyu Zhuang
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai 200036, P.R. China
| | - Rongchen Liu
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai 200036, P.R. China
| | - Fangying Yan
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai 200036, P.R. China
| | - Yufei Chen
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai 200036, P.R. China
| | - Xiufang Gao
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai 200036, P.R. China
| | - Haiming Shi
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai 200036, P.R. China
| |
Collapse
|
194
|
Wettschureck N, Strilic B, Offermanns S. Passing the Vascular Barrier: Endothelial Signaling Processes Controlling Extravasation. Physiol Rev 2019; 99:1467-1525. [PMID: 31140373 DOI: 10.1152/physrev.00037.2018] [Citation(s) in RCA: 195] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A central function of the vascular endothelium is to serve as a barrier between the blood and the surrounding tissue of the body. At the same time, solutes and cells have to pass the endothelium to leave or to enter the bloodstream to maintain homeostasis. Under pathological conditions, for example, inflammation, permeability for fluid and cells is largely increased in the affected area, thereby facilitating host defense. To appropriately function as a regulated permeability filter, the endothelium uses various mechanisms to allow solutes and cells to pass the endothelial layer. These include transcellular and paracellular pathways of which the latter requires remodeling of intercellular junctions for its regulation. This review provides an overview on endothelial barrier regulation and focuses on the endothelial signaling mechanisms controlling the opening and closing of paracellular pathways for solutes and cells such as leukocytes and metastasizing tumor cells.
Collapse
Affiliation(s)
- Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Boris Strilic
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| |
Collapse
|
195
|
Kuang Y, Liu H, Guo S, Wang Y, Zhang H, Qiao Y. The antagonist of P2Y11 receptor NF157 ameliorates oxidized LDL-induced vascular endothelial inflammation. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:1839-1845. [PMID: 31066305 DOI: 10.1080/21691401.2019.1610412] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Atherosclerosis is the chronic inflammatory disease, and inflammation-elicited endothelial activation is an early event in the development of atherosclerosis. The P2Y11 receptor is a purinergic receptor and a member of the P2 family of G coupled protein which has been shown to modulate vascular function. Progress in the study of purine receptors has been tremendous and these receptors have become pharmacological targets for various diseases. In this study, we show that the P2Y11R antagonist NF157 can mitigate oxidized LDL (ox-LDL)-induced endothelial inflammation. Our study demonstrates that P2Y11R is expressed to a fair degree in human aortic endothelial cells and is induced by treatment with ox-LDL. Blockage of P2Y11R by its selective antagonist NF157 ameliorates ox-LDL-induced adhesion of THP-1 monocytes to endothelial cells. NF157 inhibits ox-LDL-induced expression of adhesion molecules including E-selectin and VCAM-1. NF157 also suppresses ox-LDL-associated ROS production and induction of the NADPH oxidase subunit NOX-4. Moreover, NF157 has an inhibitory effect on the production of major cytokines including IL-6 and TNF-α. Mechanistically, we show that NF157 mitigates ox-LDL-induced phosphorylation of MAPK kinase p38 and NF-κB activation. Our findings indicate that blockage of P2Y11R signalling by its antagonist NF157 may protect endothelial cells from ox-LDL-induced endothelial inflammation. Therefore, NF157 may have therapeutic implications in the modulation of atherosclerosis-associated inflammation.
Collapse
Affiliation(s)
- Yun Kuang
- a Department of Health Care , Shenzhen Hospital Affiliated to Guangzhou University of Traditional Chinese Medicine , Shenzhen , China
| | - Hui Liu
- b Institute of Metabolic Diseases , the 5th Affiliated Hospital of Shenzhen University Health Science Center , Shenzhen , China
| | - Siyu Guo
- b Institute of Metabolic Diseases , the 5th Affiliated Hospital of Shenzhen University Health Science Center , Shenzhen , China
| | - Yufei Wang
- b Institute of Metabolic Diseases , the 5th Affiliated Hospital of Shenzhen University Health Science Center , Shenzhen , China
| | - Huijuan Zhang
- b Institute of Metabolic Diseases , the 5th Affiliated Hospital of Shenzhen University Health Science Center , Shenzhen , China
| | - Yanxiang Qiao
- b Institute of Metabolic Diseases , the 5th Affiliated Hospital of Shenzhen University Health Science Center , Shenzhen , China.,c Department of Internal Medicine , Baoan Central Hospital of Shenzhen , Shenzhen , China.,d Department of Emergency and Critical Care Medicine , the 5th Affiliated Hospital of Shenzhen University Health Science Center , Shenzhen , China
| |
Collapse
|
196
|
Wen J, Lin T, Wu W, Yang Y, Luo C, Zhou C, Wan J, Liu S, Wang D, Wang P, Li J. Tiaopi huxin recipe improved endothelial dysfunction and attenuated atherosclerosis by decreasing the expression of caveolin-1 in ApoE-deficient mice. J Cell Physiol 2019; 234:15369-15379. [PMID: 30729525 DOI: 10.1002/jcp.28184] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
The Tiaopi Huxin recipe (TPHXR) is widely used in traditional Chinese medicine for the clinical treatment of coronary heart disease. However, the mechanism of TPHXR treatment of atherosclerosis (AS) has not been fully elucidated. In this study, we have aimed to explore the potential antiatherosclerotic effect of TPHXR and its underlying mechanisms. Male ApoE knockout (ApoE-/- ) mice were fed a high-fat diet for 12 weeks and were randomly divided into four groups: the control group, and the low-dose, medium-dose, and high-dose TPHXR groups. The nitric oxide (NO) levels in arterial tissue and human umbilical vein endothelial cells (HUVECs) were measured by diaminofluorescein-2 diacetate staining. Vasorelaxation of mice aorta was performed by wire myograph. Inflammatory cytokines, including tumor necrosis factor-α (TNF-α), hs-CRP, IL-6, and IL-1β, in mice plasma were analyzed by enzyme-linked immunosorbent assay. Western blot analysis was applied to observe protein expression. Oil Red O staining was utilized for the quantification of atherosclerotic plaques. Results showed that 4 weeks of high- and medium-dose TPHXR treatment by oral gavage reduced atheromatous lesions in ApoE -/- mice. The high- and medium-dose TPHXR treatment, but not the low-dose treatment, promoted eNOS phosphorylation, increased NO levels and improved endothelium-dependent vasorelaxation in ApoE -/- mice. High- and medium-dose TPHXR, but not low-dose TPHXR, decreased the expression of cav-1, NF-κB p50, NF-κB p65, ICAM1, VCAM-1, TNF-α, IL-6, and IL-1β in the vasculature of ApoE -/- mice. Enzyme-linked immunosorbent assay analysis indicated that high- and medium-dose TPHXR decreased the levels of TNF-α, IL-6, hs-CRP, and IL-1β. In conclusion, our findings show that TPHXR improved the endothelial function and reduced atheromatous lesions in ApoE -/- mice. This result may be due to the decreased expression of caveolin-1 and NF-κB and, hence, the attenuated inflammatory response in AS mice vasculature. TPHXR may represent a promising intervention in patients with AS.
Collapse
Affiliation(s)
- Junmao Wen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tong Lin
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Wu
- Department of Cardiovascular Disease, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi Yang
- Department of Cardiology, The First Affiliated Hospital, Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Chuanjin Luo
- Department of Cardiovascular Disease, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chi Zhou
- Department of Cardiovascular Disease, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jindong Wan
- Department of Cardiology, The First Affiliated Hospital, Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Sen Liu
- Department of Cardiology, The First Affiliated Hospital, Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Dan Wang
- Department of Cardiology, The First Affiliated Hospital, Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Peijian Wang
- Department of Cardiology, The First Affiliated Hospital, Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Junzhe Li
- Department of Cardiovascular Disease, Guangdong Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
197
|
Arumugasaamy N, Gudelsky A, Hurley-Novatny A, Kim PC, Fisher JP. Model Placental Barrier Phenotypic Response to Fluoxetine and Sertraline: A Comparative Study. Adv Healthc Mater 2019; 8:e1900476. [PMID: 31407872 PMCID: PMC6752965 DOI: 10.1002/adhm.201900476] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/10/2019] [Indexed: 12/20/2022]
Abstract
Medications taken during pregnancy may significantly impact fetal development, yet there are few studies that rigorously assess medication safety due to ethical concerns. Selective serotonin reuptake inhibitors (SSRIs) are a class of drug increasingly being prescribed for depression, yet multiple studies have shown that taking SSRIs during pregnancy can lead to preterm birth and potential health concerns for the baby. Therefore, a biomimetic placental barrier model is utilized herein to assess transport profiles and phenotypic effects resulting from SSRI exposure, comparing fluoxetine and sertraline. Results show that the placental barrier quickly uptakes drug from the maternal side, but slowly releases on the fetal side. Phenotypically, there is a dose-dependent change in cell adhesion molecule (CAM) and transforming growth factor beta (TGFβ) secretions, markers of cell adhesion and angiogenesis. Both drugs impact CAM secretions, whereas sertraline alone impacts TGFβ secretions. When evaluating cell type, it becomes clear that endothelial cells, not trophoblast, are the main cell type involved in these phenotypic changes. Overall, these findings further the understanding of SSRI transplacental transport and drug-induced effects on the placental barrier.
Collapse
Affiliation(s)
- Navein Arumugasaamy
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, D.C. 20010
| | - Alana Gudelsky
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, D.C. 20010
| | - Amelia Hurley-Novatny
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742
| | - Peter C.W. Kim
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, D.C. 20010
- Department of Surgery, The George Washington University, Washington, D.C. 20037
| | - John P. Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742
| |
Collapse
|
198
|
Iwashima T, Kudome Y, Kishimoto Y, Saita E, Tanaka M, Taguchi C, Hirakawa S, Mitani N, Kondo K, Iida K. Aronia berry extract inhibits TNF-α-induced vascular endothelial inflammation through the regulation of STAT3. Food Nutr Res 2019; 63:3361. [PMID: 31452653 PMCID: PMC6698673 DOI: 10.29219/fnr.v63.3361] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 07/04/2019] [Accepted: 07/15/2019] [Indexed: 12/19/2022] Open
Abstract
Background Inflammation in endothelial cells induces production of inflammatory cytokines and monocytes adhesion, which are crucial events in the initiation of atherosclerosis. Aronia berry (Aronia meranocalpa), also called black chokeberry, contains abundant anthocyanins that have received considerable interest for their possible relations to vascular health. Objective The aim of this study was to investigate whether an anthocyanin-rich extract obtained from aronia berry can attenuate inflammatory responses in vascular endothelial cells. Methods As a model of vascular endothelial inflammation, human umbilical vein endothelial cells (HUVECs) pretreated with aronia berry extract were stimulated with tumor necrosis factor-alpha (TNF-α). The expression levels of cytokines and adhesion molecules were analyzed. To investigate the effects of aronia berry extract on the adhesion of THP-1 monocytic cell, the static adhesion assay was carried out. The possible molecular mechanisms by which aronia berry extract regulated vascular inflammatory responses were explored. Results The mRNA expressions of interleukins (IL-1β, IL-6, and IL-8) and monocyte chemoattractant protein-1 (MCP-1) upregulated by TNF-α were significantly suppressed by pretreatment with aronia berry extract. Aronia berry extract decreased TNF-α-induced monocyte/endothelial adhesion and suppressed vascular cell adhesion molecule-1 (VCAM-1) expression, but did not affect intercellular adhesion molecule-1 (ICAM-1) expression. Moreover, aronia berry extract decreased the phosphorylation of signal transducer and activator of transcription 3 (STAT3) and the nuclear levels of STAT3 and interferon regulatory transcription factor-1 (IRF1). The nuclear translocation of nuclear factor-kappa B (NF-κB) was not inhibited by aronia berry extract. Conclusion Aronia berry extract could exert anti-atherosclerotic effects on TNF-α-induced inflammation through inhibition of STAT3/IRF1 pathway in vascular endothelial cells.
Collapse
Affiliation(s)
- Tomomi Iwashima
- Department of Food and Nutritional Sciences, Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo, Japan
| | - Yuki Kudome
- Department of Food and Nutritional Sciences, Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo, Japan
| | - Yoshimi Kishimoto
- Endowed Research Department "Food for Health," Ochanomizu University, Tokyo, Japan
| | - Emi Saita
- Endowed Research Department "Food for Health," Ochanomizu University, Tokyo, Japan
| | - Miori Tanaka
- Department of Food and Nutritional Sciences, Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo, Japan
| | - Chie Taguchi
- Endowed Research Department "Food for Health," Ochanomizu University, Tokyo, Japan
| | | | - Nobu Mitani
- Pola Chemical Industries Inc., Kanagawa, Japan
| | - Kazuo Kondo
- Endowed Research Department "Food for Health," Ochanomizu University, Tokyo, Japan.,Institute of Life Innovation Studies, Toyo University, Gunma, Japan
| | - Kaoruko Iida
- Department of Food and Nutritional Sciences, Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo, Japan.,Institute for Human Life Innovation, Ochanomizu University, Tokyo, Japan
| |
Collapse
|
199
|
Feng YM, Thijs L, Zhang ZY, Yang WY, Huang QF, Wei FF, Kuznetsova T, Jennings AM, Delles C, Lennox R, Verhamme P, Dominiczak A, Staessen JA. Glomerular function in relation to circulating adhesion molecules and inflammation markers in a general population. Nephrol Dial Transplant 2019; 33:426-435. [PMID: 28992257 PMCID: PMC6018976 DOI: 10.1093/ndt/gfx256] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/29/2017] [Indexed: 01/14/2023] Open
Abstract
Background Inflammation is a hallmark of chronic kidney disease (CKD) and stimulates glomerular expression of vascular adhesion molecules (VCAMs). We investigated in a general population whether estimated glomerular filtration rate (eGFR) is associated with circulating adhesion molecules, inflammation markers or both. Methods We measured serum levels of five adhesion molecules [VCAM-1, intracellular adhesion molecule-1 (ICAM-1), P-selectin, E-selectin and monocyte chemoattractant protein-1 (MCP-1)] and seven inflammation markers [C-reactive protein (CRP), neutrophil gelatinase-associated lipocalin (NGAL), tumour necrosis factor receptor 1 (TNF-R1), TNF-α, interleukin 6 (IL-6), IL-8 and vascular endothelial growth factor] in 1338 randomly recruited people (50.8% women, mean age 51.7 years, eGFR 79.9 mL/min/1.73 m2). Results In multivariable-adjusted analyses, eGFR decreased (P ≤ 0.004) with higher VCAM-1 (association size expressed in mL/min/1.73 m2 for a doubling of the marker, −2.99), MCP-1 (−1.19), NGAL (−1.19), TNF receptor 1 (−2.78), TNF-α (−2.28) and IL-6 (−0.94). The odds ratios of having eGFR <60 versus ≥60 mL/min/1.73 m2 (n = 138 versus 1200) were significant (P ≤ 0.001) for VCAM-1 (1.77), MCP-1 (1.32), NGAL (1.26), TNF-R1 (1.49), TNF-α (1.45) and IL-6 (1.20). Compared with 24-h albuminuria, VCAM-1 increased (P <0.0001) the area under the curve from 0.57 to 0.65, MCP-1 to 0.67 and TNF-R1 to 0.79, but TNF-R1 outperformed both adhesion molecules (P < 0.0001). Conclusions In a general population, eGFR is inversely associated with circulating adhesion molecules VCAM-1 and MCP-1 and several inflammation markers, but inflammation markers, in particular TNF-R1 and TNF-α, identify patients with eGFR <60 mL/min/1.73 m2 more accurately.
Collapse
Affiliation(s)
- Ying-Mei Feng
- Beijing Key Laboratory of Diabetes Prevention and Research, Department of Endocrinology, Luhe Hospital, Capital Medical University, Beijing, China.,Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Lutgarde Thijs
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Zhen-Yu Zhang
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Wen-Yi Yang
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Qi-Fang Huang
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Fang-Fei Wei
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Tatiana Kuznetsova
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | | | - Christian Delles
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, UK
| | | | - Peter Verhamme
- Research Unit Molecular and Vascular Biology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Anna Dominiczak
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, UK
| | - Jan A Staessen
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium.,R&D Group VitaK, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
200
|
Inflammation as a Therapeutic Target in Atherosclerosis. J Clin Med 2019; 8:jcm8081109. [PMID: 31357404 PMCID: PMC6722844 DOI: 10.3390/jcm8081109] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/18/2019] [Accepted: 07/20/2019] [Indexed: 12/18/2022] Open
Abstract
Atherosclerotic coronary artery disease (CAD) results from build-up of cholesterol-rich plaques in the walls of the coronary arteries and is a leading cause of death. Inflammation is central to atherosclerosis. Uncontrolled inflammation makes coronary plaques “unstable” and vulnerable to rupture or erosion, leading to thrombosis and myocardial infarction (MI). As multiple inflamed plaques often co-exist in the coronary system, patients are at risk of repeated atherothrombotic cardiovascular events after MI, with rates of 10–12% at one year and 18–20% at three years. This is largely because current therapies for CAD, such as lipid-lowering statins, do not adequately control plaque inflammation. New anti-atherosclerotic agents are therefore needed, especially those that better target inflammation. The recent positive results for the anti-interleukin-1-beta (IL-1β) monoclonal antibody, Canakinumab, in the Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS) clinical trial has provided a major stimulant to the field. It highlights that not only is inflammation important from a pathogenic and risk prediction perspective in CAD, but that reducing inflammation can be beneficial. The challenge is now to find the best strategies to achieve this in real-world practice. This review outlines the role that inflammation plays in atherosclerosis and provides an update on anti-inflammatory therapies currently being investigated to target atherosclerosis.
Collapse
|