1
|
Yu J, Waresi M, Zhong H, Wu H, Ge J. 20-HETE induced platelet activation via a GPR75-independent pathway. Thromb Res 2025; 247:109277. [PMID: 39914277 DOI: 10.1016/j.thromres.2025.109277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/26/2025] [Accepted: 01/29/2025] [Indexed: 02/26/2025]
Abstract
BACKGROUND Arachidonic acid (AA) metabolism is a critical regulator of platelet activation. The cytochrome P450 (CYP450) pathway represents a key metabolic route for AA, yet the precise roles of CYP450 enzymes and their primary product, 20-hydroxyeicosatetraenoic acid (20-HETE), in platelet activation and thrombosis remain incompletely elucidated. METHODS We assessed the impact of aspirin on AA-induced platelet aggregation in human platelets. We further explored the influence of 20-HETE on platelet aggregation, granule secretion, and integrin activation. To elucidate the underlying mechanisms of 20-HETE action, we employed the antagonist 20-hydroxyeicosa-6(Z),15(Z)-dienoic acid (20-HEDE) and G protein-coupled receptor 75 (GPR75) knockout mice. Additionally, we evaluated the antiplatelet potential of the CYP450 inhibitor 17-octadecynoic acid (17-ODYA). RESULTS Aspirin suppressed platelet aggregation induced by low dose of AA and has no effect on high dose AA-induced aggregation.20-HETE indirectly induced platelet aggregation, granule release, and integrin αIIbβ3 activation in a concentration-dependent manner, independent of GPR75. The effects of 20-HETE were mediated through Gαq-coupled GPCRs. The CYP450 inhibitor 17-ODYA potently suppressed platelet activation and thrombus formation. CONCLUSIONS Thromboxane A2 (TXA2) is not indispensable for AA induced platelet activation. Moreover, 20-HETE has been identified as a potent platelet activator that acts through Gαq-coupled GPCRs. Its effects are mediated by downstream metabolites rather than direct interaction with GPR75.
Collapse
Affiliation(s)
- Jianhan Yu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China
| | - Maieryemu Waresi
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haoxuan Zhong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China
| | - Hongyi Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China; NHC Key Laboratory of Ischemic Heart Diseases, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China; NHC Key Laboratory of Ischemic Heart Diseases, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China.
| |
Collapse
|
2
|
Li Y, Ge JP, Ma K, Yin YY, He J, Gu JP. The combination of EGCG with warfarin reduces deep vein thrombosis in rabbits through modulating HIF-1α and VEGF via the PI3K/AKT and ERK1/2 signaling pathways. Chin J Nat Med 2022; 20:679-690. [PMID: 36162953 DOI: 10.1016/s1875-5364(22)60172-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Indexed: 11/28/2022]
Abstract
Deep venous thrombosis (DVT) poses a major challenge to public health worldwide. Endothelial cell injury evokes inflammatory and oxidative responses that contribute to thrombus formation. Tea polyphenol (TP) in the form of epigallocatechin-3-gallate (EGCG) has anti-inflammatory and oxidative effect that may ameliorate DVT. However, the precise mechanism remains incompletely understood. The current study was designed to investigate the anti-DVT mechanism of EGCG in combination with warfarin (an oral anticoagulant). Rabbits were randomly divided into five groups. A DVT model of rats was established through ligation of the inferior vena cava (IVC) and left common iliac vein, and the animals were orally administered with EGCG, warfarin, or vehicle for seven days. In vitro studies included pretreatment of human umbilical vein endothelial cells (HUVECs) with different concentrations of EGCG for 2 h before exposure to hydrogen peroxide. Thrombus weight and length were examined. Histopathological changes were observed by hematoxylin-eosin staining. Blood samples were collected for detecting coagulation function, including thrombin and prothrombin times, activated partial thromboplastin time, and fibrinogen levels. Protein expression in thrombosed IVCs and HUVECs was evaluated by Western blot, immunohistochemical analysis, and/or immunofluorescence staining. RT-qPCR was used to determine the levels of AGTR-1 and VEGF mRNA in IVCs and HUVECs. The viability of HUVECs was examined by CCK-8 assay. Flow cytometry was performed to detect cell apoptosis and ROS generation was assessed by 2',7'-dichlorofluorescein diacetate reagent. In vitro and invivo studies showed that EGCG combined with warfarin significantly reduced thrombus weight and length, and apoptosis in HUVECs. Our findings indicated that the combination of EGCG and warfarin protects HUVECs from oxidative stress and prevents apoptosis. However, HIF-1α silencing weakened these effects, which indicated that HIF-1α may participate in DVT. Furthermore, HIF-1α silencing significantly up-regulated cell apoptosis and ROS generation, and enhanced VEGF expression and the activation of the PI3K/AKT and ERK1/2 signaling pathways. In conclusion, our results indicate that EGCG combined with warfarin modifies HIF-1α and VEGF to prevent DVT in rabbits through anti-inflammation via the PI3K/AKT and ERK1/2 signaling pathways.
Collapse
Affiliation(s)
- Yan Li
- Department of Vascular and Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Jing-Ping Ge
- Department of Vascular and Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Ke Ma
- Department of Acupuncture, Qinhuai District Hospital of Traditional Chinese Medicine, Nanjing 210000, China
| | - Yuan-Yuan Yin
- Department of Vascular and Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Juan He
- Department of Vascular and Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Jian-Ping Gu
- Department of Vascular and Interventional Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China.
| |
Collapse
|
3
|
Lin P, Chen S, Liao M, Wang W. Physicochemical Characterization of Fucoidans from Sargassum henslowianum C.Agardh and Their Antithrombotic Activity In Vitro. Mar Drugs 2022; 20:300. [PMID: 35621950 PMCID: PMC9144781 DOI: 10.3390/md20050300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 12/31/2022] Open
Abstract
Sargassum fucoidan is a kind of sulfated heteropolysaccharide with a variety of biological activities. The aim of this study was to investigate the extraction, purification, physicochemical characterization and in vitro antithrombotic activity of fucoidan from Sargassum henslowianum C.Agardh. Hot-water-assisted ultrasound was used to extract fucoidan (F). Fucoidan was purified by DEAE cellulose 52 (F1), Vc-H2O2 (FD1) and Superdex 75 gel (FDS1). The physical and chemical properties of fucoidans were analyzed by chemical composition, monosaccharide composition, average molecular weight (Mw) and FTIR. The sulfate contents of F, F1, FD1 and FDS1 were 11.45%, 16.35% and 17.52%, 9.66%, respectively; the Mw was 5.677 × 105, 4.393 × 105, 2.176 × 104 and 6.166 × 103, respectively. The results of monosaccharide composition showed that the four fucoidans contained l-fucose, d-galactose, l-mannose, d-xylose, l-rhamnose and d-glucose, but the mass fraction ratio was different. The results of FTIR showed that fucoidan contained characteristic peaks of sugar and sulfate. In vitro, F1, FD1 and FDS1 could alleviate HUVEC damage induced by adrenaline (Adr). F1, FD1 and FDS1 decreased vWF and TF and increased the ratio of t-PA/PAI-1 in Adr-induced HUVEC.
Collapse
Affiliation(s)
- Peichun Lin
- School of Chemistry and Environment, Guangdong Ocean University, Zhanjiang 524088, China;
| | - Suhua Chen
- School of Chemistry and Environment, Guangdong Ocean University, Zhanjiang 524088, China;
| | - Min Liao
- School of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (M.L.); (W.W.)
| | - Weimin Wang
- School of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (M.L.); (W.W.)
| |
Collapse
|
4
|
Hamers A, Primus CP, Whitear C, Kumar NA, Masucci M, Montalvo Moreira SA, Rathod K, Chen J, Bubb K, Colas R, Khambata RS, Dalli J, Ahluwalia A. 20-HETE is a pivotal endogenous ligand for TRPV1-mediated neurogenic inflammation in the skin. Br J Pharmacol 2021; 179:1450-1469. [PMID: 34755897 DOI: 10.1111/bph.15726] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 10/11/2021] [Accepted: 10/26/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Transient receptor potential cation channel subfamily V member 1 (TRPV1) is localised to sensory C-fibres and its opening leads to membrane depolarization, resulting in neuropeptide release and neurogenic inflammation. However, the identity of the endogenous activator of TRPV1 in this setting is unknown. The arachidonic acid (AA) metabolites 12-hydroperoxyeicosatetraenoyl acid (12-HpETE) and 20-hydroxyeicosatetraenoic acid (20-HETE) have emerged as potential endogenous activators of TRPV1 however, whether these lipids underlie TRPV1-mediated neurogenic inflammation remains unknown. EXPERIMENTAL APPROACH we analysed human cantharidin-induced blister samples and inflammatory responses in TRPV1 transgenic mice. KEY RESULTS In a human cantharidin-blister model the potent TRPV1 activators 20-HETE but not 12-HETE (stable metabolite of 12-HpETE) correlated with AA levels. Similarly, in mice levels of 20-HETE (but not 12-HETE) and AA were strongly positively correlated within the inflammatory milieu. Furthermore, LPS-induced oedema formation and neutrophil recruitment were substantially and significantly attenuated by pharmacological block or genetic deletion of TRPV1 channels, inhibition of 20-HETE formation or SP receptor neurokinin 1 (NK1 ) blockade. LPS treatment also increased cytochrome-P450 ώ-hydroxylase gene expression, the enzyme responsible for 20-HETE production. CONCLUSIONS AND IMPLICATIONS Taken together, our findings suggest that endogenously generated 20-HETE activates TRPV1 causing C-fibre activation and consequent oedema formation. These findings identify a novel pathway that may be useful in the therapeutics of diseases/conditions characterized by a prominent neurogenic inflammation, as in several skin diseases.
Collapse
Affiliation(s)
- Alexander Hamers
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Christopher P Primus
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Charlotte Whitear
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Nitin Ajit Kumar
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Michael Masucci
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Shanik A Montalvo Moreira
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Krishnaraj Rathod
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Jianmin Chen
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Kristen Bubb
- Monash Cardiovascular Research Centre, Monash University, Melbourne, Australia
| | - Romain Colas
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Rayomand S Khambata
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Jesmond Dalli
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Amrita Ahluwalia
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| |
Collapse
|
5
|
He L, Lin Y, Wang X, Liu XL, Wang Y, Qin J, Wang X, Day D, Xiang J, Mo J, Zhang Y, Zhang JJ. Associations of ozone exposure with urinary metabolites of arachidonic acid. ENVIRONMENT INTERNATIONAL 2020; 145:106154. [PMID: 33038623 DOI: 10.1016/j.envint.2020.106154] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/20/2020] [Accepted: 09/21/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Ozone (O3) exposure has been associated with biomarkers of platelet activation and oxidative stress. The metabolism of arachidonic acid (AA) plays an important role in platelet activation and oxidative stress. However, AA metabolic pathways have not been examined in relation to O3 and other air pollutants. METHODS Early morning urine and fasting blood were longitudinally collected up to four times from 89 healthy adults (22-52 years old, 25 women) in Changsha City, China. We measured three urinary AA metabolites, namely 11-dehydro-Thromboxane B2 (11-dhTXB2) produced from the arachidonic cyclooxygenase pathway, 20-hydroxyeicosatetraenoic acid (20-HETE) from the CYPs pathway, and 8-isoprostane from the non-enzymatic pathway. Urinary malondialdehyde (MDA) and 8-hydroxy-2'-deoxyguanosine (8-OHdG) were measured as indicators of oxidative damage to lipids and DNA, respectively. We measured soluble P-selectin (sCD62p) concentrations in plasma as an indicator of platelet activation. Indoor and outdoor air pollutants were measured and combined with participants' time-activity pattern to calculate personal exposure to O3, PM2.5, NO2, and SO2 averaged over 12-hour, 24-hour, 1-week, and 2-week periods prior to biospecimen collection, respectively. Linear mixed-effects models were used to examine the relationships of AA metabolites with air pollutant exposures, plasma sCD62p, and urinary MDA & 8-OHdG. RESULTS We found that a 10% increase in 12 h and 24 h O3 exposure were associated with increases in urinary11-dhTXB2 by 1.4% (95%, 0.1% to 2.6%) and 1.3% (0.05% to 2.5%), respectively. These associations remained robust after adjusting for co-pollutant exposures. No significant associations were observed between 11-dhTXB2 and other pollutants or between O3 exposures and other AA metabolites. All the three AA metabolites were significantly and positively associated with urinary MDA and 8-OHdG, whereas only urinary 11-dhTXB2 was significantly and positively associated with plasma sCD62p. CONCLUSIONS A metabolite of AA from the cyclooxygenase pathway was positively associated with short-term O3 exposure, and with a plasma marker of platelet activation and two urinary markers of oxidative stress. The results suggest that O3 exposure may contribute to increased platelet activation and oxidative damages via altering the metabolism of AA.
Collapse
Affiliation(s)
- Linchen He
- Nicholas School of the Environment, Duke University, Durham, NC 27705, USA; Global Health Institute, Duke University, Durham, NC 27708, USA
| | - Yan Lin
- Nicholas School of the Environment, Duke University, Durham, NC 27705, USA; Global Health Institute, Duke University, Durham, NC 27708, USA
| | - Xiangtian Wang
- Nicholas School of the Environment, Duke University, Durham, NC 27705, USA
| | - Xing Lucy Liu
- Global Health Institute, Duke University, Durham, NC 27708, USA
| | - Yang Wang
- Nicholas School of the Environment, Duke University, Durham, NC 27705, USA
| | - Jian Qin
- Guangxi Medical University, Nanning, Guangxi Province 530021, China
| | - Xiaoli Wang
- Tianjin University of Technology, Tianjin 300384, China
| | - Drew Day
- Seattle Children's Research Institute, Seattle, WA 98121, United States
| | - Jianbang Xiang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, United States
| | - Jinhan Mo
- Department of Building Science, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory of Indoor Air Quality Evaluation and Control, Beijing 100084, China
| | - Yinping Zhang
- Department of Building Science, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory of Indoor Air Quality Evaluation and Control, Beijing 100084, China.
| | - Junfeng Jim Zhang
- Nicholas School of the Environment, Duke University, Durham, NC 27705, USA; Global Health Institute, Duke University, Durham, NC 27708, USA; Duke Kunshan University, Kunshan City, Jiangsu Province 215316, China.
| |
Collapse
|
6
|
Effects of resveratrol on coagulative, fibrinolytic, and inflammatory marker expression and secretion by endothelial cells (human umbilical vein endothelial cells). Blood Coagul Fibrinolysis 2020; 31:207-212. [PMID: 32108678 DOI: 10.1097/mbc.0000000000000900] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
: Increasing the prevalence of cardiovascular disease (CVD) has led to an investigation into components that might influence CVD. Accordingly, many recent studies have reported the benefits of resveratrol (RSV). Therefore, this study aimed to scrutinize the direct effect of RSV on human umbilical vein endothelial cells (HUVECs) by detecting coagulative, fibrinolytic, and inflammatory markers. HUVECs were cultured and treated with different concentrations of RSV. The effects of RSV were identified by representative markers of coagulation, fibrinolysis pathway, and inflammation, including von Willebrand factor (VWF), factor VIII, tissue plasminogen activator-1 (t-PA-1), and interleukin-8 (IL-8). The detection process was carried out using real-time PCR (qPCR), flow cytometry, ELISA, and immunocytochemistry (ICC) methods. The present findings demonstrated a significant decrease in VWF, t-PA-1, and IL-8 secretion levels. Furthermore, RSV diminished the activity of factor VIII and mRNA expression levels of VWF and t-PA-1. The ICC results also showed a decrease in the level of intracellular t-PA. Our data revealed the anti-inflammatory, anticoagulation, and antifibrinolytic effect of RSV in cell culture.
Collapse
|
7
|
Du Y, Taylor CG, Aukema HM, Zahradka P. Role of oxylipins generated from dietary PUFAs in the modulation of endothelial cell function. Prostaglandins Leukot Essent Fatty Acids 2020; 160:102160. [PMID: 32717531 DOI: 10.1016/j.plefa.2020.102160] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/17/2020] [Accepted: 07/17/2020] [Indexed: 12/13/2022]
Abstract
Oxylipins, which are circulating bioactive lipids generated from polyunsaturated fatty acids (PUFAs) by cyclooxygenase, lipooxygenase and cytochrome P450 enzymes, have diverse effects on endothelial cells. Although studies of the effects of oxylipins on endothelial cell function are accumulating, a review that provides a comprehensive compilation of current knowledge and recent advances in the context of vascular homeostasis is lacking. This is the first compilation of the various in vitro, ex vivo and in vivo reports to examine the effects and potential mechanisms of action of oxylipins on endothelial cells. The aggregate data indicate docosahexaenoic acid-derived oxylipins consistently show beneficial effects related to key endothelial cell functions, whereas oxylipins derived from other PUFAs exhibit both positive and negative effects. Furthermore, information is lacking for certain oxylipin classes, such as those derived from α-linolenic acid, which suggests additional studies are required to achieve a full understanding of how oxylipins affect endothelial cells.
Collapse
Affiliation(s)
- Youjia Du
- Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; Department of Physiology and Pathophysiology, University of Manitoba, MB R3E 0J9, Canada
| | - Carla G Taylor
- Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; Department of Physiology and Pathophysiology, University of Manitoba, MB R3E 0J9, Canada; Department of Food and Human Nutritional Sciences, University of Manitoba, MB R3T 2N2, Canada
| | - Harold M Aukema
- Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; Department of Food and Human Nutritional Sciences, University of Manitoba, MB R3T 2N2, Canada
| | - Peter Zahradka
- Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; Department of Physiology and Pathophysiology, University of Manitoba, MB R3E 0J9, Canada; Department of Food and Human Nutritional Sciences, University of Manitoba, MB R3T 2N2, Canada.
| |
Collapse
|
8
|
Effects of Saccharides from Arctium lappa L. Root on FeCl 3-Induced Arterial Thrombosis via the ERK/NF- κB Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7691352. [PMID: 32308808 PMCID: PMC7132581 DOI: 10.1155/2020/7691352] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 02/11/2020] [Accepted: 03/02/2020] [Indexed: 12/20/2022]
Abstract
Saccharides from Arctium lappa. L. root (ALR-S) is a high-purity fructosaccharide separated from the medicinal plant Arctium lappa. L. root. These compounds showed many pharmacological effects in previous studies. In the present study, the antithrombotic effects of ALR-S in arterial thrombosis via inhibiting platelet adhesion and rebalancing thrombotic and antithrombotic factor expression and secretion were found in rats and human aortic endothelial cells (HAECs). This study also showed that inhibition of oxidative stress (OS), which is closely involved in the expression of coagulation- and thrombosis-related proteins, was involved in the antithrombotic effects of ALR-S. Furthermore, studies using FeCl3-treated HAECs showed that ALR-S induced the abovementioned effects at least partly by blocking the ERK/NF-κB pathway. Moreover, U0126, a specific inhibitor of ERK, exhibited the same effects with ALR-S on a thrombotic process in FeCl3-injured HAECs, suggesting the thrombotic role of the ERK/NF-κB pathway and the antithrombotic role of blocking the ERK/NF-κB pathway by ALR-S. In conclusion, our study revealed that the ERK/NF-κB pathway is a potential therapeutic target in arterial thrombosis and that ALR-S has good characteristics for the cure of arterial thrombosis via regulating the ERK/NF-κB signaling pathway.
Collapse
|
9
|
Afshinnia F, Zeng L, Byun J, Wernisch S, Deo R, Chen J, Hamm L, Miller ER, Rhee EP, Fischer MJ, Sharma K, Feldman HI, Michailidis G, Pennathur S. Elevated lipoxygenase and cytochrome P450 products predict progression of chronic kidney disease. Nephrol Dial Transplant 2020; 35:303-312. [PMID: 30137494 PMCID: PMC7391277 DOI: 10.1093/ndt/gfy232] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 06/14/2018] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND The clinical relevance of arachidonic acid (AA) metabolites in chronic kidney disease (CKD) progression is poorly understood. We aimed to compare the concentrations of 85 enzymatic pathway products of AA metabolism in patients with CKD who progressed to end-stage kidney disease (ESKD) versus patients who did not in a subcohort of Chronic Renal Insufficiency Cohort (CRIC) and to estimate the risk of CKD progression and major cardiovascular events by levels of AA metabolites and their link to enzymatic metabolic pathways. METHODS A total 123 patients in the CRIC study who progressed to ESKD were frequency matched with 177 nonprogressors and serum eicosanoids were quantified by mass spectrometry. We applied serum collected at patients' Year 1 visit and outcome of progression to ESKD was ascertained over the next 10 years. We used logistic regression models for risk estimation. RESULTS Baseline 15-hydroxyeicosatetraenoate (HETE) and 20-HETE levels were significantly elevated in progressors (false discovery rate Q ≤ 0.026). The median 20-HETE level was 7.6 pmol/mL [interquartile range (IQR) 4.2-14.5] in progressors and 5.4 pmol/mL (IQR 2.8-9.4) in nonprogressors (P < 0.001). In an adjusted model, only 20-HETE independently predicted CKD progression. Each 1 standard deviation increase in 20-HETE was independently associated with 1.45-fold higher odds of progression (95% confidence interval 1.07-1.95; P = 0.017). Principal components of lipoxygenase (LOX) and cytochrome P450 (CYP450) pathways were independently associated with CKD progression. CONCLUSIONS We found higher odds of CKD progression associated with higher 20-HETE, LOX and CYP450 metabolic pathways. These alterations precede CKD progression and may serve as targets for interventions aimed at halting progression.
Collapse
Affiliation(s)
- Farsad Afshinnia
- Department of Internal Medicine-Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Lixia Zeng
- Department of Internal Medicine-Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Jaeman Byun
- Department of Internal Medicine-Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Stefanie Wernisch
- Department of Internal Medicine-Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Rajat Deo
- Division of Cardiovascular Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jing Chen
- Division of Nephrology and Hypertension, Tulane University, New Orleans, LA, USA
| | - Lee Hamm
- Division of Nephrology and Hypertension, Tulane University, New Orleans, LA, USA
| | - Edgar R Miller
- Department of Internal Medicine, Jones Hopkins University, Baltimore, MD, USA
| | - Eugene P Rhee
- Department of Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Michael J Fischer
- Department of Medicine, University of Illinois, Center of Innovation for Complex Chronic Healthcare, Jesse Brown VAMC, Chicago, IL, USA
| | - Kumar Sharma
- Department of Internal Medicine-Nephrology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Harold I Feldman
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Subramaniam Pennathur
- Department of Internal Medicine-Nephrology, University of Michigan, Ann Arbor, MI, USA
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
10
|
Lou Z, Li X, Zhao X, Du K, Li X, Wang B. Resveratrol attenuates hydrogen peroxide‑induced apoptosis, reactive oxygen species generation, and PSGL‑1 and VWF activation in human umbilical vein endothelial cells, potentially via MAPK signalling pathways. Mol Med Rep 2017; 17:2479-2487. [PMID: 29207192 DOI: 10.3892/mmr.2017.8124] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 10/24/2017] [Indexed: 11/06/2022] Open
Abstract
Reactive oxygen species (ROS) are implicated in the pathogenesis of thrombosis. Studies have reported that resveratrol exhibits antioxidative activities, however, the effect and underlying mechanisms of resveratrol on venous thrombosis remain largely unknown. To investigate the effect of resveratrol on venous thrombosis and the underlying mechanisms, the present study investigated the effects of resveratrol on cell viability, apoptosis, ROS generation and the expression of thrombosis‑associated markers in human umbilical vein endothelial cells (HUVECs). HUVECs were pretreated with resveratrol for 2 h and incubated with hydrogen peroxide (H2O2) for 24 h prior to the evaluation of cell viability, ROS generation, apoptosis and thrombosis‑associated marker expression by performing MTT assays, 2',7'‑dichlorofluorescin diacetate reagent, flow cytometry, and reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and western blot analysis, respectively. Subsequently, to validate whether resve-ratrol functions via mitogen‑activated protein kinase (MAPK) pathways, the expression of thrombosis‑associated markers was detected by western blot analysis and RT‑qPCR following treatment of cells with resveratrol and the MAPK pathway activators anisomycin and curcumin. The results demonstrated that cell viability was markedly reduced by H2O2, and resveratrol treatment reversed the reductions in cell viability in a dose‑dependent manner. In addition, the levels of cell apoptosis and ROS generation were significantly increased by H2O2 alone, and resveratrol also reduced these effects in a dose‑dependent manner. Furthermore, the mRNA and protein expression of caspase‑3, P‑selectin glycoprotein ligand‑1 and von Willebrand factor was upregulated by H2O2 treatment in HUVECs. However, resveratrol decreased the protein expression these proteins in a dose‑dependent manner. Resveratrol also significantly inhibited the induction of phosphorylated (p)‑p38, P‑c‑Jun N‑terminal kinase and P‑extracellular signal‑regulated kinase by H2O2, and these effects were attenuated by the MAPK pathway activators anisomycin and curcumin. In conclusion, these results indicate that resveratrol protected HUVECs against oxidative stress and apoptosis. Furthermore, to the best of our knowledge, the present study is the first to demonstrate that resveratrol attenuates the expression of thrombosis‑associated markers induced by H2O2, which may occur through the suppression of the MAPK signalling pathways, indicating a potential novel therapeutic approach to prevent venous thrombosis.
Collapse
Affiliation(s)
- Zhenkai Lou
- Department of Orthopaedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Xingguo Li
- Department of Orthopaedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Xueling Zhao
- Department of Orthopaedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Kaili Du
- Department of Orthopaedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Xing Li
- Department of Ultrasound, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Bing Wang
- Department of Orthopaedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
11
|
Fan F, Ge Y, Lv W, Elliott MR, Muroya Y, Hirata T, Booz GW, Roman RJ. Molecular mechanisms and cell signaling of 20-hydroxyeicosatetraenoic acid in vascular pathophysiology. Front Biosci (Landmark Ed) 2016; 21:1427-63. [PMID: 27100515 DOI: 10.2741/4465] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cytochrome P450s enzymes catalyze the metabolism of arachidonic acid to epoxyeicosatrienoic acids (EETs), dihydroxyeicosatetraenoic acid and hydroxyeicosatetraeonic acid (HETEs). 20-HETE is a vasoconstrictor that depolarizes vascular smooth muscle cells by blocking K+ channels. EETs serve as endothelial derived hyperpolarizing factors. Inhibition of the formation of 20-HETE impairs the myogenic response and autoregulation of renal and cerebral blood flow. Changes in the formation of EETs and 20-HETE have been reported in hypertension and drugs that target these pathways alter blood pressure in animal models. Sequence variants in CYP4A11 and CYP4F2 that produce 20-HETE, UDP-glucuronosyl transferase involved in the biotransformation of 20-HETE and soluble epoxide hydrolase that inactivates EETs are associated with hypertension in human studies. 20-HETE contributes to the regulation of vascular hypertrophy, restenosis, angiogenesis and inflammation. It also promotes endothelial dysfunction and contributes to cerebral vasospasm and ischemia-reperfusion injury in the brain, kidney and heart. This review will focus on the role of 20-HETE in vascular dysfunction, inflammation, ischemic and hemorrhagic stroke and cardiac and renal ischemia reperfusion injury.
Collapse
Affiliation(s)
- Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Ying Ge
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Wenshan Lv
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216 and Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Matthew R Elliott
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Yoshikazu Muroya
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216 and Department of General Medicine and Rehabilitation, Tohoku Medical and Pharmaceutical University School of Medicine, Sendai, Japan
| | - Takashi Hirata
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216 and Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - George W Booz
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216,
| |
Collapse
|