1
|
Chen Y, Lin Y, Hong J, Wang J, Li B, Liu Z, Ma Y, Sun X, Wang S, Li M, Wu M, Fu D, Li J, Wang M, Hu L. Oroxylin A reverses SHP-2 oxidative inactivation in GPVI signaling to suppress platelet activation and thrombus formation. Thromb J 2025; 23:26. [PMID: 40102927 PMCID: PMC11921655 DOI: 10.1186/s12959-025-00709-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Arterial thrombotic events are the leading causes of death worldwide, and the therapeutic effects of current antiplatelet drugs are not fully satisfactory. Oroxylin A (OroA), a flavone compound extracted from Scutellaria radix, possesses cardioprotective and many other pharmacological effects. While platelets play a crucial role in the development of myocardial infarction, the direct effects of OroA on platelet activation and thrombosis have yet to be investigated. METHODS FeCl₃-induced arteriole thrombosis and whole-blood perfusion were used to assess the inhibitory effect of OroA on thrombus formation. A myocardial ischemia model was employed to evaluate the protective effect of OroA on myocardial injury. Multiple platelet function studies including platelet aggregation, platelet spreading, clot retraction were performed. Network pharmacology, flow cytometry, enzyme-linked immunosorbent assay, co-immunoprecipitation and western blot were utilized to explore the mechanism of OroA on platelet activation. RESULTS OroA inhibited thrombus formation with less bleeding risk compared with aspirin. OroA protected against myocardial injury by suppressing microvascular thrombosis and platelet infiltration. OroA suppressed different agonist-induced platelet activation in a concentration-dependent manner, showing greater antiplatelet activity against collagen-induced platelet aggregation compared to ADP or thrombin-induced aggregation. OroA decreased granule release, integrin αIIbβ3 activation, platelet spreading and clot retraction. As a flavone, OroA boosted superoxide dismutase (SOD) and glutathione (GSH) activities and decreased malondialdehyde (MDA), oxidized glutathione (GSSG) and ROS levels in platelets during oxidative stress. OroA binds to SHP-2 and prevents its oxidative inactivation, leading to the tyrosine dephosphorylation of Src, Syk and PLCγ2, as well as the reduction of Ca2+ influx and PKC phosphorylation in GPVI signaling. CONCLUSIONS OroA inhibits platelet activation, thrombus formation and myocardial injury via reversing SHP-2 oxidative inactivation thereby attenuating collagen-induced GPVI signaling. With minor bleeding risk and no obvious pharmacological toxicity, OroA holds promising therapeutic potential as an antithrombotic drug.
Collapse
Affiliation(s)
- Yufei Chen
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuan Lin
- The Research Center for Cardiovascular diseases, The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jin Hong
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiaorui Wang
- The Research Center for Cardiovascular diseases, The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Biling Li
- The Research Center for Cardiovascular diseases, The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zixian Liu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yongbo Ma
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaolan Sun
- The Research Center for Cardiovascular diseases, The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shufang Wang
- The Research Center for Cardiovascular diseases, The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Mingjie Li
- The Research Center for Cardiovascular diseases, The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Meiling Wu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, 999077, China
| | - Deyu Fu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jian Li
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Mingzhu Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Liang Hu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- The Research Center for Cardiovascular diseases, The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
2
|
Cao C, Yang Q, Xia X, Chen Z, Liu P, Wu X, Hu H, Ding Z, Li X. WY-14643, a novel antiplatelet and antithrombotic agent targeting the GPIbα receptor. Thromb Res 2024; 238:41-51. [PMID: 38669962 DOI: 10.1016/j.thromres.2024.04.011] [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: 10/28/2023] [Revised: 03/26/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND AND PURPOSE Hypolipidemia and platelet activation play key roles in atherosclerotic diseases. Pirinixic acid (WY-14643) was originally developed as a lipid-lowering drug. Here we focused on its antiplatelet and antithrombotic abilities and the underlying mechanism. EXPERIMENTAL APPROACH The effects of WY-14643 on platelet aggregation was measured using a lumi-aggregometer. Clot retraction and spreading on fibrinogen were also assayed. PPARα-/- platelets were used to identify the target of WY-14643. The interaction between WY-14643 and glycoprotein Ibα (GPIbα) was detected using cellular thermal shift assay (CETSA), surface plasmon resonance (SPR) spectroscopy and molecular docking. GPIbα downstream signaling was examined by Western blot. The antithrombotic effect was investigated using mouse mesenteric arteriole thrombosis model. Mouse tail bleeding model was used to study its effect on bleeding side effects. KEY RESULTS WY-14643 concentration-dependently inhibits human washed platelet aggregation, clot retraction, and spreading. Significantly, WY-14643 inhibits thrombin-induced activation of human washed platelets with an IC50 of 7.026 μM. The antiplatelet effect of WY-14643 is mainly dependent of GPIbα. CESTA, SPR and molecular docking results indicate that WY-14643 directly interacts with GPIbα and acts as a GPIbα antagonist. WY-14643 also inhibits phosphorylation of PLCγ2, Akt, p38, and Erk1/2 induced by thrombin. Noteworthily, 20 mg/kg oral administration of WY-14643 inhibits FeCl3-induced thrombosis of mesenteric arteries in mice similarly to clopidogrel without increasing bleeding. CONCLUSION AND IMPLICATIONS WY-14643 is not only a PPARα agonist with lipid-lowering effect, but also an antiplatelet agent as a GPIbα antagonist. It may have more significant therapeutic advantages than current antiplatelet agents for the treatment of atherosclerotic thrombosis, which have lipid-lowering effects without bleeding side effects.
Collapse
Affiliation(s)
- Chen Cao
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Qingyuan Yang
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xiaoshuang Xia
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Zhuangzhuang Chen
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Peilin Liu
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xiaowen Wu
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Hu Hu
- Department of Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310012, China
| | - Zhongren Ding
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
| | - Xin Li
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| |
Collapse
|
3
|
Yu J, Wang T, Zhang X, Chen Q, Hu Y, Liang Q, Shi Y, Wang Y, Zhang Y, Yu M, Zhao B. Anti-thrombotic Effects Mediated by a Novel Dual-Target Peptide Inhibiting Both Platelet Aggregation and Thrombin Activity without Causing Bleeding. Thromb Haemost 2024; 124:108-121. [PMID: 37657484 DOI: 10.1055/a-2165-1000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/03/2023]
Abstract
BACKGROUND Classical anticoagulants and antiplatelets are associated with high frequencies of bleeding complications or treatment failure when used as single agents. Thrombin plays an important role in the blood coagulation system. GP IIb/IIIa is the central receptor of platelets, which can recognize the Arg-Gly-Asp (RGD) sequence and activate platelets. MATERIAL AND METHODS Molecular simulation and homology modeling were performed to design a novel dual-target anticoagulant short peptide (PTIP ). The activities of PTIP on coagulation and platelet in vitro were analyzed. The antithrombotic activity of PTIP was determined by pulmonary thromboembolism model, ferric chloride injury model and arteriovenous bypass thrombosis model. Bleeding effect and toxicity of PTIP were evaluated. RESULTS We have constructed a novel dual-target peptide (PTIP) based on the direct thrombin inhibitor peptide (DTIP). PTIP was expressed at high levels in Pichia pastoris. PTIP interfered with thrombin-mediated coagulation and ADP-induced platelet aggregation in vitro. When injected intravenously or subcutaneously, PTIP showed potent and dose-dependent extension of aPTT and PT which were similar to DTIP; but only PTIP was capable of inhibiting platelet aggregation. PTIP (1.0 mg/kg) decelerated thrombosis formation in venous and arterial vessels induced by FeCl3 injury. PTIP (1.0 mg/kg) also prevented deep venous thrombosis and increased the survival rate associated with pulmonary thromboembolism. And PTIP effectively reduced thrombus length in arteriovenous bypass thrombosis model. Moreover, the antithrombotic dose of PTIP could not induce bleeding. CONCLUSION These data establish that PTIP represents a novel antithrombotic agent whose effects involve both inhibition of platelet activation and reduction of fibrin generation. And PTIP not only can be used in venous thrombosis and arterial thrombosis, it can also replace the combined treatment of antiplatelet and anticoagulant drugs in thrombotic diseases.
Collapse
Affiliation(s)
- Jinchao Yu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
- The Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Tianfa Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
- The Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaohan Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
- The Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qing Chen
- The Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai, China
| | - Yajun Hu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
- The Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qiaoyan Liang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
- The Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yun Shi
- Department of Immunology and Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, California, United States
| | - Yuxiong Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
- The Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yanling Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
- The Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Min Yu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
- The Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Bing Zhao
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
- The Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Design and fabrication of r-hirudin loaded dissolving microneedle patch for minimally invasive and long-term treatment of thromboembolic disease. Asian J Pharm Sci 2022; 17:284-297. [PMID: 35582638 PMCID: PMC9091604 DOI: 10.1016/j.ajps.2022.02.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/12/2022] [Accepted: 02/26/2022] [Indexed: 11/30/2022] Open
Abstract
Cardiovascular disease is the leading cause of global mortality, with anticoagulant therapy being the main prevention and treatment strategy. Recombinant hirudin (r-hirudin) is a direct thrombin inhibitor that can potentially prevent thrombosis via subcutaneous (SC) and intravenous (IV) administration, but there is a risk of haemorrhage via SC and IV. Thus, microneedle (MN) provides painless and sanitary alternatives to syringes and oral administration. However, the current technological process for the micro mould is complicated and expensive. The micro mould obtained via three-dimensional (3D) printing is expected to save time and cost, as well as provide a diverse range of MNs. Therefore, we explored a method for MNs array model production based on 3D printing and translate it to micro mould that can be used for fabrication of dissolving MNs patch. The results show that r-hirudin-loaded and hyaluronic acid (HA)-based MNs can achieve transdermal drug delivery and exhibit significant potential in the prevention of thromboembolic disease without bleeding in animal models. These results indicate that based on 3D printing technology, MNs combined with r-hirudin are expected to achieve diverse customizable MNs and thus realize personalized transdermal anticoagulant delivery for minimally invasive and long-term treatment of thrombotic disease.
Collapse
|
5
|
Yang Y, Li B. Novel Peptide Motifs Containing Asp-Glu-Gly Target P 2Y 12 and Thromboxane A2 Receptors to Inhibit Platelet Aggregation and Thrombus Formation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:785-793. [PMID: 35016500 DOI: 10.1021/acs.jafc.1c06159] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Increasing evidence has shown that collagen peptides have multiple biological activities. Our previous study has separated and identified antiplatelet aggregation peptides Asp-Glu-Gly-Pro (DEGP) from Salmo salar skin. This study is to investigate the cellular target of DEGP on platelets and its underlying mechanism. DEGP inhibited platelet aggregation in a dose-dependent manner induced by 2MeS-ADP and U46619 and significantly attenuated tail thrombosis formation by 30% in mice at the dose of 50 mg/kg body weight. Mechanically, DEGP displayed apparent antagonism effects on TP and P2Y12 receptors by the drug affinity responsive target stability (DARTS) technique to regulate the phosphorylation of RhoAS188, PLCβ3S537, as well as VASPS157. The molecular docking results revealed a stronger binding energy with the target protein of modified peptides DEGI and DDEGL. Practically, DEGI exhibited the highest inhibition activity against 2MeS-ADP- and U46619-induced platelet aggregation in vitro with IC50 values of 0.88 ± 0.10 and 0.85 ± 0.10 mM, respectively, and comparable antithrombosis activity with aspirin at the dose of 25 mg/kg body weight in vivo. These results indicated the possibility that the peptide motifs containing Asp-Glu-Gly could potentially be developed as a novel therapeutic agent in the prevention and treatment of thrombotic diseases.
Collapse
Affiliation(s)
- Yijie Yang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Bo Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
- Key Laboratory of Functional Dairy, Ministry of Education, Beijing 100083, China
| |
Collapse
|
6
|
Huang M, Deng M, Nie W, Zou D, Wu H, Xu D. Naringenin Inhibits Platelet Activation and Arterial Thrombosis Through Inhibition of Phosphoinositide 3-Kinase and Cyclic Nucleotide Signaling. Front Pharmacol 2021; 12:722257. [PMID: 34475824 PMCID: PMC8406801 DOI: 10.3389/fphar.2021.722257] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/30/2021] [Indexed: 11/19/2022] Open
Abstract
Citrus flavanoids intake can reduce the risk of cardiovascular diseases. Naringenin, a natural predominant flavonoid abundant in citrus fruits, possesses protective effects against atherothrombotic diseases. As platelet activation plays central roles in atherothrombogenesis, we studied the effects of naringenin on platelet activation, signaling, thrombosis and hemostasis. Naringenin dose-dependently inhibited agonist-induced platelet aggregation in vitro, and exhibited more-potent efficacy on ADP-induced platelet aggregation. It also suppressed platelet aggregation stimulated by ADP ex vivo. Naringenin inhibited ADP-induced platelet α-granule secretion, fibrinogen binding, intracellular calcium mobilization and platelet adhesion on collagen-coated surface. Naringenin also inhibited platelet spreading on fibrinogen and clot retraction, processes mediated by outside-in integrin signaling. Mechanism studies indicated that naringenin suppressed PI3K-mediated signaling and phosphodiesterase activity in platelets, in addition to increasing cGMP levels and VASP phosphorylation at Ser239. Furthermore, naringenin-induced VASP phosphorylation and inhibition of platelet aggregation were reversed by a PKA inhibitor treatment. Interestingly, naringenin inhibited thrombus formation in the (FeCl3)-induced rat carotid arterial thrombus model, but not cause a prolonged bleeding time in mice. This study suggests that naringenin may represent a potential antiplatelet agent targeting PI3K and cyclic nucleotide signaling, with a low bleeding risk.
Collapse
Affiliation(s)
- Manting Huang
- Department of Vascular Intervention, Zhongshan Hospital of Traditional Chinese Medicine, Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, China
- Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Minzhen Deng
- Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenqiang Nie
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Dezhi Zou
- Emergency Department, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Huanlin Wu
- Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Huanlin Wu, ; Danping Xu,
| | - Danping Xu
- Department of Traditional Chinese Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- *Correspondence: Huanlin Wu, ; Danping Xu,
| |
Collapse
|
7
|
Jiang Q, Mao R, Wu J, Chang L, Zhu H, Zhang G, Ding Z, Zhang J. Platelet activation during chronic hepatitis B infection exacerbates liver inflammation and promotes fibrosis. J Med Virol 2020; 92:3319-3326. [PMID: 31769518 DOI: 10.1002/jmv.25641] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 11/19/2019] [Indexed: 01/16/2023]
Abstract
Recurrent hepatitis activity during chronic hepatitis B virus infection results in fibrosis and even hepatocellular carcinoma. It is still unclear what causes acute exacerbation. As platelets have recently been identified as a significant role in inflammation, we here investigated the role of platelets in mediating liver damage in patients with chronic hepatitis B virus infection. Platelet aggregation testing and flow cytometry were carried out to evaluate platelet activation status in 121 patients chronically infected with hepatitis B across different phases of the condition. The correlation between platelet aggregation rate and liver inflammation or liver fibrosis index was evaluated. To investigate the genesis of platelet activation, several serum cytokines were also assessed by MILLIPLEX microsphere-based multiplex cytokine assay. Active hepatitis patients showed a higher aggregation rate than others. Levels of CD62p, a marker of platelet activation, were also increased in this group of patients. Positive correlations between platelet aggregation rate and liver inflammation or liver fibrosis were also noted, indicating a significant role of platelet in the progression of liver disease. The level of tumor necrosis factor-alpha, which is known to trigger platelet activation, was markedly higher in the active hepatitis group (P < .005). Based on the findings in our study, platelet activation plays a vital role in the progression of chronic hepatitis B virus infection. Antiplatelet therapy may provide a new means of hepatitis B infection treatment.
Collapse
Affiliation(s)
- Qirong Jiang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Richeng Mao
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Jingwen Wu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Lin Chang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Haoxiang Zhu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Ge Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhongren Ding
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jiming Zhang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- Department of Infectious Diseases, Jing'An District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Pan G, Chang L, Zhang J, Liu Y, Hu L, Zhang S, Zhang J, Qiao J, Jakopin Ž, Hu H, Dong J, Ding Z. GSK669, a NOD2 receptor antagonist, inhibits thrombosis and oxidative stress via targeting platelet GPVI. Biochem Pharmacol 2020; 183:114315. [PMID: 33152345 DOI: 10.1016/j.bcp.2020.114315] [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: 08/17/2020] [Revised: 10/21/2020] [Accepted: 10/29/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND PURPOSE Previously, we discovered that the activation of nucleotide-binding oligomerization domain 2 (NOD2) enhances platelet activation. We here investigated the antiplatelet and antithrombotic potential of GSK669, a NOD2 antagonist. EXPERIMENTAL APPROACH Effects of GSK669 on platelet functions, reactive oxygen species (ROS) and proinflammatory cytokine generation were detected. NOD2-/- platelets were used to confirm GSK669 target. The interaction between GSK669 and glycoprotein VI (GPVI) was detected using surface plasmon resonance (SPR) spectroscopy. GPVI downstream signaling was examined by Western blot. The antithrombotic and antioxidative effects were investigated using mouse mesenteric arteriole thrombosis model and pulmonary embolism model. KEY RESULTS GSK669 significantly inhibits platelet proinflammatory cytokine release induced by muramyl dipeptide, platelet aggregation, ATP release, and ROS generation induced by collagen and collagen related peptide (CRP). Platelet spreading and clot retraction are also inhibited. GSK669 also decreases collagen-induced phosphorylation of Src, Syk, PLCγ2, and Akt. The antiplatelet effect of GSK669 is NOD2-independent and mediated by GPVI antagonism. Consistent with its antiplatelet activity as a GPVI antagonist, GSK669 inhibits platelet adhesion on collagen in flow condition. Notably, GSK669 inhibits mouse mesenteric arteriole thrombosis similarly to aspirin without bleeding. The antithrombotic effect of GSK669 is further confirmed in the pulmonary embolism model; decreased malonaldehyde (MDA) and increased superoxide dismutase (SOD) levels in mouse plasma reveal a significant antioxidant effect of GSK669. CONCLUSION AND IMPLICATIONS Beyond its anti-inflammatory effect as a NOD2 antagonist, GSK669 is also an efficient and safe antiplatelet agent combined with antioxidant effect by targeting GPVI. An antiplatelet agent bearing antioxidative and anti-inflammatory activities without bleeding risk may have therapeutic advantage over current antiplatelet drugs for atherothrombosis.
Collapse
Affiliation(s)
- Guanxing Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Lin Chang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jianjun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yangyang Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450018, China
| | - Liang Hu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450018, China
| | - Si Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jian Zhang
- Department of Pathophysiology, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, China
| | - Jianlin Qiao
- Department of Hematology, Blood Disease Institute, Xuzhou Medical University, Xuzhou 221004, China
| | - Žiga Jakopin
- Faculty of Pharmacy, University of Ljubljana, Askerceva 7, SI-1000 Ljubljana, Slovenia
| | - Hu Hu
- Department of Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310012, China
| | - Jianzeng Dong
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450018, China
| | - Zhongren Ding
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450018, China
| |
Collapse
|
9
|
Zhang S, Liu Y, Wang X, Yang L, Li H, Wang Y, Liu M, Zhao X, Xie Y, Yang Y, Zhang S, Fan Z, Dong J, Yuan Z, Ding Z, Zhang Y, Hu L. SARS-CoV-2 binds platelet ACE2 to enhance thrombosis in COVID-19. J Hematol Oncol 2020; 13:120. [PMID: 32887634 PMCID: PMC7471641 DOI: 10.1186/s13045-020-00954-7] [Citation(s) in RCA: 471] [Impact Index Per Article: 94.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 08/19/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Critically ill patients diagnosed with COVID-19 may develop a pro-thrombotic state that places them at a dramatically increased lethal risk. Although platelet activation is critical for thrombosis and is responsible for the thrombotic events and cardiovascular complications, the role of platelets in the pathogenesis of COVID-19 remains unclear. METHODS Using platelets from healthy volunteers, non-COVID-19 and COVID-19 patients, as well as wild-type and hACE2 transgenic mice, we evaluated the changes in platelet and coagulation parameters in COVID-19 patients. We investigated ACE2 expression and direct effect of SARS-CoV-2 virus on platelets by RT-PCR, flow cytometry, Western blot, immunofluorescence, and platelet functional studies in vitro, FeCl3-induced thrombus formation in vivo, and thrombus formation under flow conditions ex vivo. RESULTS We demonstrated that COVID-19 patients present with increased mean platelet volume (MPV) and platelet hyperactivity, which correlated with a decrease in overall platelet count. Detectable SARS-CoV-2 RNA in the blood stream was associated with platelet hyperactivity in critically ill patients. Platelets expressed ACE2, a host cell receptor for SARS-CoV-2, and TMPRSS2, a serine protease for Spike protein priming. SARS-CoV-2 and its Spike protein directly enhanced platelet activation such as platelet aggregation, PAC-1 binding, CD62P expression, α granule secretion, dense granule release, platelet spreading, and clot retraction in vitro, and thereby Spike protein enhanced thrombosis formation in wild-type mice transfused with hACE2 transgenic platelets, but this was not observed in animals transfused with wild-type platelets in vivo. Further, we provided evidence suggesting that the MAPK pathway, downstream of ACE2, mediates the potentiating role of SARS-CoV-2 on platelet activation, and that platelet ACE2 expression decreases following SARS-COV-2 stimulation. SARS-CoV-2 and its Spike protein directly stimulated platelets to facilitate the release of coagulation factors, the secretion of inflammatory factors, and the formation of leukocyte-platelet aggregates. Recombinant human ACE2 protein and anti-Spike monoclonal antibody could inhibit SARS-CoV-2 Spike protein-induced platelet activation. CONCLUSIONS Our findings uncovered a novel function of SARS-CoV-2 on platelet activation via binding of Spike to ACE2. SARS-CoV-2-induced platelet activation may participate in thrombus formation and inflammatory responses in COVID-19 patients.
Collapse
Affiliation(s)
- Si Zhang
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| | - Yangyang Liu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Cardiovascular Institute of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaofang Wang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Cardiovascular Institute of Zhengzhou University, Zhengzhou, 450052, China
| | - Li Yang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Haishan Li
- Department of Emergency, Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuyan Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), and Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mengduan Liu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Cardiovascular Institute of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaoyan Zhao
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Cardiovascular Institute of Zhengzhou University, Zhengzhou, 450052, China
| | - Youhua Xie
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), and Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Yang
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Shenghui Zhang
- Department of Hematology, Wenzhou Key Laboratory of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT, 06030, USA
| | - Jianzeng Dong
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Cardiovascular Institute of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), and Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhongren Ding
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Cardiovascular Institute of Zhengzhou University, Zhengzhou, 450052, China
| | - Yi Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Liang Hu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Cardiovascular Institute of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
10
|
Zhang J, Zhang Y, Zheng S, Liu Y, Chang L, Pan G, Hu L, Zhang S, Liu J, Kim S, Dong J, Ding Z. PAK Membrane Translocation and Phosphorylation Regulate Platelet Aggregation Downstream of Gi and G12/13 Pathways. Thromb Haemost 2020; 120:1536-1547. [PMID: 32854120 DOI: 10.1055/s-0040-1714745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Platelet activation plays a pivotal role in physiological hemostasis and pathological thrombosis causing heart attack and stroke. Previous studies conclude that simultaneous activation of Gi and G12/13 signaling pathways is sufficient to cause platelet aggregation. However, using Gq knockout mice and Gq-specific inhibitors, we here demonstrated that platelet aggregation downstream of coactivation of Gi and G12/13 depends on agonist concentrations; coactivation of Gi and G12/13 pathways only induces platelet aggregation under higher agonist concentrations. We confirmed Gi and G12/13 pathway activation by showing cAMP (cyclic adenosine monophosphate) decrease and RhoA activation in platelets stimulated at both low and high agonist concentrations. Interestingly, we found that though Akt and PAK (p21-activated kinase) translocate to the platelet membrane upon both low and high agonist stimulation, membrane-translocated Akt and PAK only phosphorylate at high agonist concentrations, correlating well with platelet aggregation downstream of concomitant Gi and G12/13 pathway activation. PAK inhibitor abolishes Akt phosphorylation, inhibits platelet aggregation in vitro and arterial thrombus formation in vivo. We propose that the PAK-PI3K/Akt pathway mediates platelet aggregation downstream of Gi and G12/13, and PAK may represent a potential antiplatelet and antithrombotic target.
Collapse
Affiliation(s)
- Jianjun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Shuang Zheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yangyang Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lin Chang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Guanxing Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Liang Hu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Si Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Junling Liu
- Department of Biochemistry and Molecular Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Soochong Kim
- College of Veterinary Medicine, Chungbuk National University, Cheongju, Korea
| | - Jianzeng Dong
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhongren Ding
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Sepúlveda C, Hernández B, Burgos CF, Fuentes E, Palomo I, Alarcón M. The cAMP/PKA Pathway Inhibits Beta-amyloid Peptide Release from Human Platelets. Neuroscience 2018; 397:159-171. [PMID: 30496824 DOI: 10.1016/j.neuroscience.2018.11.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/30/2018] [Accepted: 11/15/2018] [Indexed: 12/12/2022]
Abstract
The main component of Alzheimer's disease (AD) is the amyloid-beta peptide (Aβ), the brain of these patients is characterized by deposits in the parenchyma and cerebral blood vessels known as cerebral amyloid angiopathy (CAA). On the other hand, the platelets are the major source of the Aβ peptide in circulation and once secreted can activate the platelets and endothelial cells producing the secretion of several inflammatory mediators that finally end up unchaining the CAA and later AD. In the present study we demonstrate that cAMP/PKA pathway plays key roles in the regulation of calpain activation and secretion of Aβ in human platelets. We confirmed that inhibition of platelet functionality occurred when platelets were incubated with forskolin (molecule that rapidly increased cAMP levels). In this sense we found that platelets pre-incubated with forskolin (20 μM) present a complete inhibition of calpain activity and this effect is reversed using an inhibitor of protein kinase A. Consequentially, when platelets were inhibited by forskolin a reduction in the processing of the APP with the consequent decrease in the Aβ peptide secretion was observed. Therefore our study provides novel insight in relation to the mechanism of processing and release of the Aβ peptide from human platelets.
Collapse
Affiliation(s)
- C Sepúlveda
- Thrombosis Reasearch Center, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001 Talca, Chile
| | - B Hernández
- Thrombosis Reasearch Center, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001 Talca, Chile
| | - C F Burgos
- Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, Chile
| | - E Fuentes
- Thrombosis Reasearch Center, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Chile; Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca 3460000, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001 Talca, Chile
| | - I Palomo
- Thrombosis Reasearch Center, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Chile; Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca 3460000, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001 Talca, Chile
| | - M Alarcón
- Thrombosis Reasearch Center, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Chile; Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca 3460000, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001 Talca, Chile.
| |
Collapse
|
12
|
Baqi Y, Müller CE. Antithrombotic P2Y 12 receptor antagonists: recent developments in drug discovery. Drug Discov Today 2018; 24:325-333. [PMID: 30291899 DOI: 10.1016/j.drudis.2018.09.021] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 08/31/2018] [Accepted: 09/27/2018] [Indexed: 12/16/2022]
Abstract
The P2Y12 receptor is one of eight known P2Y receptor subtypes, and belongs to the G-protein-coupled receptor (GPCR) family. The P2Y12 receptor is highly expressed on blood platelets and in the brain. Potent, selective, peripherally acting antagonists for the P2Y12 receptor are used clinically as antithrombotic drugs. Several different scaffolds have been identified as P2Y12 receptor antagonists, including irreversibly acting thienotetrahydropyridines (prodrugs), and reversible competitive antagonists, including adenine nucleotide analogs, piperazinyl-glutamate-quinolines, -pyridines, and -pyrimidines, and anthraquinone derivatives. Here, we provide an overview of the different scaffolds that have been developed as P2Y12 receptor antagonists, some of which have become important therapeutics.
Collapse
Affiliation(s)
- Younis Baqi
- Department of Chemistry, Faculty of Science, Sultan Qaboos University, PO Box 36, Postal Code 123, Muscat, Oman.
| | - Christa E Müller
- Pharma-Center Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, An der Immenburg 4, D-53121 Bonn, Germany
| |
Collapse
|
13
|
Chen L, Han L, Hu L, Liang J, Wang J, Ding Z. Preparation and characterization of solid dispersion of novel dual antiplatelet agent BF061 for oral use. J Drug Deliv Sci Technol 2018. [DOI: 10.1016/j.jddst.2017.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
14
|
Role of P2Y 12 Receptor in Thrombosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 906:307-324. [PMID: 27628007 DOI: 10.1007/5584_2016_123] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
P2Y12 receptor is a 342 amino acid Gi-coupled receptor predominantly expressed on platelets. P2Y12 receptor is physiologically activated by ADP and inhibits adenyl cyclase (AC) to decrease cyclic AMP (cAMP) level, resulting in platelet aggregation. It also activates PI3 kinase (PI3K) pathway leading to fibrinogen receptor activation, and may protect platelets from apoptosis. Abnormalities of P2Y12 receptor include congenital deficiencies or high activity in diseases like diabetes mellitus (DM) and chronic kidney disease (CKD), exposing such patients to a prothrombotic condition. A series of clinical antiplatelet drugs, such as clopidogrel and ticagrelor, are designed as indirect or direct antagonists of P2Y12 receptor to reduce incidence of thrombosis mainly for patients of acute coronary syndrome (ACS) who are at high risk of thrombotic events. Studies on novel dual-/multi-target antiplatelet agents consider P2Y12 receptor as a promising part in combined targets. However, the clinical practical phenomena, such as "clopidogrel resistance" due to gene variations of cytochrome P450 or P2Y12 receptor constitutive activation, call for better antiplatelet agents. Researches also showed inverse agonist of P2Y12 receptor could play a better role over neutral antagonists. Personalized antiplatelet therapy is the most ideal destination for antiplatelet therapy in ACS patients with or without other underlying diseases like DM or CKD, however, there is still a long way to go.
Collapse
|
15
|
Sugidachi A, Ohno K, Jakubowski JA, Ito Y, Tomizawa A, Mizuno M. Induction of Diabetes Abolishes the Antithrombotic Effect of Clopidogrel in Apolipoprotein E-Deficient Mice. TH OPEN 2017; 1:e92-e100. [PMID: 31249914 PMCID: PMC6524843 DOI: 10.1055/s-0037-1605361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Patients with acute coronary syndrome with diabetes mellitus (DM) exhibit an impaired platelet inhibitory response to clopidogrel which is only partially understood. DM was induced by the administration of streptozotocin (STZ) to 9-week-old mice. The antithrombotic effects of clopidogrel (10 mg/kg/d, orally × 5 days) were determined using a FeCl
3
-induced thrombosis model employing wild-type (WT), apolipoprotein E (apoE)-deficient, and diabetic apoE-deficient mice at 21 weeks. Antiplatelet effects were determined using flow cytometry. The antithrombotic effects of clopidogrel were similar in WT and apoE-deficient mice but were attenuated in diabetic apoE-deficient mice with the percent inhibition of thrombus area (µm
2
) by clopidogrel being 85.5% (WT mice), 75.0% (apoE-deficient mice), and 1.9% (diabetic apoE-deficient mice). The time to first occlusion and lumen stenosis also reflected a significant loss of the antithrombotic effects of clopidogrel in diabetic apoE-deficient mice. Ex vivo platelet activation, which was assessed using ADP-induced expression of activated glycoprotein IIb/IIIa, was completely inhibited by clopidogrel in these three groups of mice. In contrast, the effect of clopidogrel on the ex vivo expression of platelet P-selectin induced by protease-activated receptor 4–activating peptide was diminished in diabetic apoE-deficient mice compared with that in WT and apoE-deficient mice. These data suggest that diabetic apoE-deficient mice may serve as a useful model to better understand the impaired responses to clopidogrel in patients with DM, which may partially reflect a reduction of the effect of clopidogrel on thrombin-induced platelet activation.
Collapse
Affiliation(s)
- A Sugidachi
- Rare Disease and LCM Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - K Ohno
- Rare Disease and LCM Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - J A Jakubowski
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States
| | - Y Ito
- Rare Disease and LCM Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - A Tomizawa
- Rare Disease and LCM Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - M Mizuno
- Rare Disease and LCM Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| |
Collapse
|
16
|
Hu L, Chang L, Zhang Y, Zhai L, Zhang S, Qi Z, Yan H, Yan Y, Luo X, Zhang S, Wang Y, Kunapuli SP, Ye H, Ding Z. Platelets Express Activated P2Y 12 Receptor in Patients With Diabetes Mellitus. Circulation 2017. [PMID: 28637879 DOI: 10.1161/circulationaha.116.026995] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Platelets from patients with diabetes mellitus are hyperactive. Hyperactivated platelets may contribute to cardiovascular complications and inadequate responses to antiplatelet agents in the setting of diabetes mellitus. However, the underlying mechanism of hyperactivated platelets is not completely understood. METHODS We measured P2Y12 expression on platelets from patients with type 2 diabetes mellitus and on platelets from rats with diabetes mellitus. We also assayed platelet P2Y12 activation by measuring cAMP and VASP phosphorylation. The antiplatelet and antithrombotic effects of AR-C78511 and cangrelor were compared in rats. Finally, we explored the role of the nuclear factor-κB pathway in regulating P2Y12 receptor expression in megakaryocytes. RESULTS Platelet P2Y12 levels are 4-fold higher in patients with type 2 diabetes mellitus compared with healthy subjects. P2Y12 expression correlates with ADP-induced platelet aggregation (r=0.89, P<0.01). P2Y12 in platelets from patients with diabetes mellitus is constitutively activated. Although both AR-C78511, a potent P2Y12 inverse agonist, and cangrelor have similar antiplatelet efficacy on platelets from healthy subjects, AR-C78511 exhibits more powerful antiplatelet effects on diabetic platelets than cangrelor (aggregation ratio 36±3% versus 49±5%, respectively, P<0.05). Using a FeCl3-injury mesenteric arteriole thrombosis model in rats and an arteriovenous shunt thrombosis model in rats, we found that the inverse agonist AR-C78511 has greater antithrombotic effects on GK rats with diabetes mellitus than cangrelor (thrombus weight 4.9±0.3 mg versus 8.3±0.4 mg, respectively, P<0.01). We also found that a pathway involving high glucose-reactive oxygen species-nuclear factor-κB increases platelet P2Y12 receptor expression in diabetes mellitus. CONCLUSIONS Platelet P2Y12 receptor expression is significantly increased and the receptor is constitutively activated in patients with type 2 diabetes mellitus, which contributes to platelet hyperactivity and limits antiplatelet drug efficacy in type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Liang Hu
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lin Chang
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan Zhang
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lili Zhai
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shenghui Zhang
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhiyong Qi
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hongmei Yan
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan Yan
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinping Luo
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Si Zhang
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiping Wang
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Satya P Kunapuli
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hongying Ye
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhongren Ding
- From Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (L.H., L.C., Y.Z., L.Z., Shenghui Z., Si Z., Z.D.); Division of Cardiovascular Disease (Z.Q.), Division of Endocrinology and Metabolism (H.Y.), Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology and Metabolism (H.Y.), Division of Cardiovascular Disease (Y.Y.), Zhongshan Hospital, Fudan University, Shanghai, China; Department of Pharmacology I, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (L.H., Y.W.); Department of Physiology and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA (S.P.K.). Dr Shenghui Zhang is presently at Department of Hematology, Wenzhou Key Laboratory of Hematology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
17
|
Abstract
Thrombosis is a complex process involving multiple pathways. Currently, therapy relies on the combination of two or more antithrombotic drugs, showing that inhibiting more than one target provides benefits in the prevention and treatment of thrombosis. This review focuses on structure-activity relationship studies of molecules possessing multiple actions against thrombosis, namely, dual inhibitors of coagulation, dual inhibitors of coagulation and platelet aggregation, and also dual inhibitors of platelet aggregation. EP217609 has just entered clinical trials, which raise the expectations on the multitarget strategy to prevent or treat thrombosis.
Collapse
|
18
|
Ye J, Zhai L, Zhang S, Zhang Y, Chen L, Hu L, Zhang S, Ding Z. DL-3-n-butylphthalide inhibits platelet activation via inhibition of cPLA2-mediated TXA2 synthesis and phosphodiesterase. Platelets 2015; 26:736-44. [PMID: 25734213 DOI: 10.3109/09537104.2014.989826] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Aberrant platelet activation plays a critical role in the pathogenesis of heart attack and stroke. DL-3-n-butylphthalide (NBP) has been approved in China to treat stroke with multiple mechanisms. The anti-stroke effects of NBP may be related to its antiplatelet effects reported in rats in addition to its antioxidative, antiapoptotic, and angiogenic effects. However, the effects and the underlying mechanisms of NBP on human platelets are not yet clear. In this study, we found that NBP concentration-dependently inhibited human platelet aggregation and ATP release induced by ADP, thrombin, U46619, arachidonic acid, or collagen. NBP also inhibited PAC-1 binding induced by ADP or thrombin and platelet spreading on immobilized fibrinogen. NBP reduced TXA2 synthesis induced by thrombin or collagen via inhibiting cPLA2 phosphorylation, concomitantly with a marked decrease in intracellular calcium mobilization. Moreover, NBP also inhibited human platelet phosphodiesterase (PDE) and elevated 3,5-cyclic adenosine monophosphate level in platelets. In conclusion, NBP significantly inhibits human platelet activation via inhibition of cPLA2-mediated TXA2 synthesis and PDE, and may be effective as an antiplatelet drug to treat other arterial thrombotic diseases.
Collapse
Affiliation(s)
- Jianqin Ye
- a Key Laboratory of Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology , Shanghai Medical College, Fudan University , Shanghai , China
| | - Lili Zhai
- a Key Laboratory of Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology , Shanghai Medical College, Fudan University , Shanghai , China
| | - Shenghui Zhang
- a Key Laboratory of Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology , Shanghai Medical College, Fudan University , Shanghai , China
| | - Yan Zhang
- a Key Laboratory of Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology , Shanghai Medical College, Fudan University , Shanghai , China
| | - Leilei Chen
- a Key Laboratory of Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology , Shanghai Medical College, Fudan University , Shanghai , China
| | - Liang Hu
- a Key Laboratory of Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology , Shanghai Medical College, Fudan University , Shanghai , China
| | - Si Zhang
- a Key Laboratory of Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology , Shanghai Medical College, Fudan University , Shanghai , China
| | - Zhongren Ding
- a Key Laboratory of Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology , Shanghai Medical College, Fudan University , Shanghai , China
| |
Collapse
|
19
|
Du H, He Q, Chen N, Xu J, Chen F, Liu G. Proton NMR investigations on 6-alkylamino-2-alkylthioadenosine derivatives. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2015; 53:218-222. [PMID: 25279994 DOI: 10.1002/mrc.4151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 08/26/2014] [Accepted: 08/29/2014] [Indexed: 06/03/2023]
Affiliation(s)
- Hongguang Du
- State Key Laboratory of Chemical Resource Engineering, Department of Organic Chemistry, College of Science, Beijing University of Chemical Technology, Beijing, 100029, China
| | | | | | | | | | | |
Collapse
|
20
|
Vistoli G, Brizzolari A, Faioni E, Razzari C, Santaniello E. Naturally occurring N(6)-substituted adenosines (cytokinin ribosides) are in vitro inhibitors of platelet aggregation: an in silico evaluation of their interaction with the P2Y(12) receptor. Bioorg Med Chem Lett 2014; 24:5652-5655. [PMID: 25467153 DOI: 10.1016/j.bmcl.2014.10.080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 10/22/2014] [Accepted: 10/23/2014] [Indexed: 12/21/2022]
Abstract
A few naturally occurring N(6)-substituted adenosine derivatives (cytokinin ribosides) were investigated as inhibitors of platelet aggregation induced in vitro by collagen and their activity range was demonstrated (IC50: 6.77-141 μM). A docking study suggests that anti-aggregation activity of these compounds could involve an interaction with the P2Y12 receptor binding site.
Collapse
Affiliation(s)
- Giulio Vistoli
- Department of Pharmaceutical Science, Università degli Studi, Via Celoria 2, 20100 Milano, Italy
| | - Andrea Brizzolari
- Department of Health Sciences, Università degli Studi, Via A. Di Rudinì 8, 20142 Milano, Italy
| | - Elena Faioni
- Department of Health Sciences, Università degli Studi, Via A. Di Rudinì 8, 20142 Milano, Italy; S. Paolo Hospital, Via A. Di Rudinì 8, 20142 Milano, Italy
| | | | - Enzo Santaniello
- Department of Health Sciences, Università degli Studi, Via A. Di Rudinì 8, 20142 Milano, Italy.
| |
Collapse
|
21
|
Liu L, Li J, Zhang Y, Zhang S, Ye J, Wen Z, Ding J, Kunapuli SP, Luo X, Ding Z. Salvianolic acid B inhibits platelets as a P2Y12 antagonist and PDE inhibitor: Evidence from clinic to laboratory. Thromb Res 2014; 134:866-76. [DOI: 10.1016/j.thromres.2014.07.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 06/05/2014] [Accepted: 07/13/2014] [Indexed: 11/30/2022]
|
22
|
Cross-modulatory effects of clopidogrel and heparin on platelet and fibrin incorporation in thrombosis. Blood Coagul Fibrinolysis 2014; 24:593-8. [PMID: 23492917 DOI: 10.1097/mbc.0b013e3283602a03] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Pharmacologic inhibition of platelet activation and aggregation is a mainstay for reducing the incidence of arterial thrombosis, whereas anticoagulation is the primary approach for preventing the development of venous thrombosis. The effect of standard pharmacologic agents on their reciprocal vessel - anticoagulants on arterial thrombosis and platelet inhibitor on venous thrombosis - is relatively understudied. This study was designed to evaluate murine large-vessel arterial or venous thrombosis under conditions of either fibrin or platelet inhibition. In this study, heparin and clopidogrel were used as standard anticoagulant and platelet inhibitor, respectively, evaluating both large artery and vein thrombosis in mice, using in-vivo fluorescence imaging to simultaneously measure fibrin and platelet levels at the thrombus induction site. Heparin reduced both fibrin and platelet development in both arteries and veins, with stronger influences on fibrin accrual. Clopidogrel had a stronger effect in arteries, reducing both platelet and fibrin accumulation. Clopidogrel also reduced platelet accumulation with venous thrombosis, but the reductions in fibrin formation did not reach statistical significance. These findings illustrate the interactive role of platelet activity and coagulation in the development of large-vessel thrombosis, with inhibition of one thrombotic component showing profound effects on the other component in both arterial and venous thrombosis.
Collapse
|
23
|
Fuentes E, Palomo I. Regulatory mechanisms of cAMP levels as a multiple target for antiplatelet activity and less bleeding risk. Thromb Res 2014; 134:221-6. [PMID: 24830902 DOI: 10.1016/j.thromres.2014.04.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 04/22/2014] [Accepted: 04/25/2014] [Indexed: 12/19/2022]
Abstract
Platelet activation is a critical component of atherothrombosis. The multiple pathways of platelet activation limit the effect of specific receptor/pathway inhibitors, resulting in limited clinical efficacy. Recent research has confirmed that combination therapy results in enhanced antithrombotic efficacy without increasing bleeding risk. In this way, the best-known inhibitor and turn off signaling in platelet activation is cAMP. In this article we discuss the mechanisms of regulation of intraplatelet cAMP levels, a) platelet-dependent pathway: Gi/Gs protein-coupled receptors, phosphodiesterase inhibition and activation of PPARs and b) platelet-independent pathway: inhibition of adenosine uptake by erythrocytes. With respect to the association between intraplatelet cAMP levels and bleeding risk it is possible to establish that compounds/drugs with pleitropic effect for increased intraplatelet cAMP level could have an antithrombotic activity with less risk of bleeding.
Collapse
Affiliation(s)
- Eduardo Fuentes
- Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001, Chile.
| | - Iván Palomo
- Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001, Chile.
| |
Collapse
|
24
|
Zhang SH, Zhang Y, Shen J, Zhang S, Chen L, Gu J, Mruk JS, Cheng G, Zhu L, Kunapuli SP, Ding Z. Tumor vascular disrupting agent 5,6-dimethylxanthenone-4-acetic acid inhibits platelet activation and thrombosis via inhibition of thromboxane A2 signaling and phosphodiesterase. J Thromb Haemost 2013; 11:1855-66. [PMID: 23902231 DOI: 10.1111/jth.12362] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Indexed: 12/01/2022]
Abstract
BACKGROUND 5,6-Dimethylxanthenone-4-acetic acid (DMXAA) is a tumor vascular disrupting agent under clinical trials as an adjacent antitumor agent. DMXAA is structurally similar to flavone-8-acetic acid (FAA), an old tumor vascular disrupting agent with antiplatelet and antithrombotic effects. In contrast to FAA, which causes bleeding in tumor patients, no bleeding has been reported in patients receiving DMXAA. Whether DMXAA also affects platelet function is not clear. OBJECTIVES To determine the effects of DMXAA on platelet function and explore the underlying mechanisms. METHODS AND RESULTS DMXAA concentration-dependently inhibited human platelet aggregation and ATP release induced by U46619, arachidonic acid, ADP, collagen, or ristocetin. Furthermore, DMXAA inhibited phosphorylation of Erk1/2 and Akt downstream of thromboxane A2 signaling inhibition. DMXAA also inhibited human platelet phosphodiesterase. The antiplatelet effects were further confirmed using mice administered DMXAA intravenously. DMXAA dramatically inhibited thrombus formation in FeCl3 -injured mouse mesenteric arterial thrombus model and laser-injured mouse cremaster arteriole thrombus model. Notably, at a dose exhibiting antithrombotic effects similar to those of clopidogrel in mice, DMXAA did not significantly increase bleeding. CONCLUSIONS For the first time, we found that tumor vascular disrupting agent DMXAA has potent antiplatelet and antithrombotic effects without any bleeding diathesis. As DMXAA inhibits platelet activity with safe profile, DMXAA could be used as an efficacious and safe antiplatelet drug.
Collapse
Affiliation(s)
- S H Zhang
- Key Laboratory of Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Zhang Y, Ye J, Hu L, Zhang S, Zhang SH, Li Y, Kunapuli SP, Ding Z. Increased platelet activation and thrombosis in transgenic mice expressing constitutively active P2Y12. J Thromb Haemost 2012; 10:2149-57. [PMID: 22906019 PMCID: PMC3495164 DOI: 10.1111/j.1538-7836.2012.04894.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND In our previous in vitro study, we reported a constitutively active chimeric P2Y(12) (cP2Y(12)) and found that AR-C78511 is a potent inverse agonist at this receptor. The role of cP2Y(12) in platelet activation and thrombosis is not clear. OBJECTIVES To investigate the physiologic implications of cP2Y(12) for platelet activation and thrombus formation, and to evaluate the antiplatelet activity of AR-C78511 as an inverse agonist. METHODS AND RESULTS We generated transgenic mice conditionally and platelet-specifically expressing cP2Y(12). High-level expression of cP2Y(12) in platelets increased platelet reactivity, as shown by increased platelet aggregation in response to multiple platelet agonists. Moreover, transgenic mice showed a shortened bleeding time, and more rapid and stable thrombus formation in mesenteric artery injured with FeCl(3). The constitutive activity of cP2Y(12) in platelets was confirmed by decreased platelet cAMP levels and constitutive Akt phosphorylation in the absence of agonists. AR-C78511 reversed the cAMP decrease in transgenic mouse platelets, and exhibited a superior antiplatelet effect to that of AR-C69931MX in transgenic mice. CONCLUSIONS These findings further emphasize the importance of P2Y(12) in platelet activation, hemostasis, and thrombosis, as well as the prothrombotic role of the constitutive activity of P2Y(12). Our data also validate the in vivo inverse agonist activity of AR-C78511, and confirm its superior antiplatelet activity over neutral antagonists.
Collapse
Affiliation(s)
- Y Zhang
- Key Laboratory of Molecular Medicine, Ministry of Education and Department of Biochemistry and Molecular Biology, Fudan University Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Pan C, Wei X, Ye J, Liu G, Zhang S, Zhang Y, Du H, Ding Z. BF066, a novel dual target antiplatelet agent without significant bleeding. PLoS One 2012; 7:e40451. [PMID: 22815749 PMCID: PMC3398006 DOI: 10.1371/journal.pone.0040451] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 06/07/2012] [Indexed: 01/16/2023] Open
Abstract
In this study, we report BF066, a novel adenine derivative, inhibits platelet activation and thrombosis via the adenosine receptor (A(2A)) activation and phosphodiesterase (PDE) inhibition. BF066 inhibits platelet aggregation and ATP releasing induced by multiple platelet agonists in a dose-dependent manner. The inhibition of BF066 on ADP-induced aggregation is potentiated by adenosine and can be dramatically antagonized by the A(2A) antagonist SCH58261. BF066 also inhibits the PDE activity and platelet spreading on fibrinogen. In FeCl(3)-injured mouse mesenteric arterial thrombosis model, BF066 prevents thrombus formation effectively, similar to clopidogrel. Intriguingly, at dose achieving similar antithrombotic effect compared to clopidogrel, BF066 does not increase bleeding significantly. Taken together, these results suggest that BF066 may be an effective and safe antiplatelet agent targeting both PDE and A(2A). Considering the successful use of combined antiplatelet therapy, BF066 may be further developed as a novel dual target antiplatelet agent.
Collapse
Affiliation(s)
- ChangE Pan
- School of Life Science, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xunbin Wei
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- * E-mail: (XW); (ZD)
| | - Jianqin Ye
- Key Laboratory of Molecular Medicine, Ministry of Education, and Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
| | - Guangda Liu
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Si Zhang
- Key Laboratory of Molecular Medicine, Ministry of Education, and Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
| | - Yan Zhang
- Key Laboratory of Molecular Medicine, Ministry of Education, and Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
| | - Hongguang Du
- College of Science, Beijing University of Chemical Technology, Chaoyang District, Beijing, China
| | - Zhongren Ding
- Key Laboratory of Molecular Medicine, Ministry of Education, and Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
- * E-mail: (XW); (ZD)
| |
Collapse
|