1
|
Qi Z, Zhou L, Dai S, Zhang P, Zhong H, Zhou W, Zhao X, Xu H, Zhao G, Wu H, Ge J. Intermittent fasting inhibits platelet activation and thrombosis through the intestinal metabolite indole-3-propionate. LIFE METABOLISM 2025; 4:loaf002. [PMID: 40078933 PMCID: PMC11897983 DOI: 10.1093/lifemeta/loaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/09/2025] [Accepted: 01/21/2025] [Indexed: 03/14/2025]
Abstract
Platelet hyperreactivity contributes significantly to thrombosis in acute myocardial infarction and stroke. While antiplatelet drugs are used, residual ischemic risk remains. Intermittent fasting (IF), a dietary pattern characterized by alternating periods of eating and fasting, has shown cardiovascular benefits, but its effect on platelet activation is unclear. This study demonstrates that IF inhibits platelet activation and thrombosis in both patients with coronary artery disease and apolipoprotein E (ApoE) knockout (ApoE -/- ) mice, by enhancing intestinal flora production of indole-3-propionic acid (IPA). Mechanistically, elevated IPA in plasma directly attenuates platelet activation by binding to the platelet pregnane X receptor (PXR) and suppressing downstream signaling pathways, including Src/Lyn/Syk and LAT/PLCγ/PKC/Ca2+. Importantly, IF alleviates myocardial and cerebral ischemia/reperfusion injury in ApoE -/- mice. These findings suggest that IF mitigates platelet activation and thrombosis risk in coronary atherosclerosis by enhancing intestinal flora production of IPA, which subsequently activates the platelet PXR-related signaling pathways.
Collapse
Affiliation(s)
- Zhiyong Qi
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Luning Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Shimo Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Peng Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Haoxuan Zhong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Wenxuan Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Xin Zhao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Huajie Xu
- Department of Infectious Disease, Zhongshan Hospital, Fudan University, 180 Fenglin Road , Shanghai 200032, China
| | - Gang Zhao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Hongyi Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
- Institutes of Biomedical Sciences, Fudan University, 131 Dong’an Road, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, 180 Fenglin Road, Shanghai 200032, China
| |
Collapse
|
2
|
Nissen PH, Pedersen OB. Unlocking the Potential of MicroRNA Expression: Biomarkers for Platelet Reactivity and Coronary Artery Disease. Semin Thromb Hemost 2025. [PMID: 40074010 DOI: 10.1055/s-0045-1805041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Coronary artery disease (CAD) is a leading cause of morbidity and mortality worldwide, with platelet reactivity playing a central role in its pathogenesis. Recent research has identified microRNAs (miRNAs; miRs) as potential biomarkers for CAD, due to their ability to regulate platelet function and reactivity. This review focuses on four key miRNAs-miR-223, miR-126, miR-21, and miR-150-known to influence platelet reactivity and their implications in CAD. miR-223, which is highly expressed in platelets, has shown associations with CAD and myocardial infarction, while miR-126 has been linked to thrombus formation and vascular health. Additionally, miR-21 and miR-150 have also emerged as important players, with roles in platelet reactivity and cardiovascular outcomes. However, despite their potential, the use of miRNAs as clinical biomarkers faces several challenges, including variability in reported results across studies. These inconsistencies often arise from differences in sample material, preanalytical conditions, and normalization strategies. Furthermore, the influence of antiplatelet therapy on miRNA expression adds another layer of complexity, making it difficult to determine whether observed changes in miRNA levels are due to disease states or therapeutic interventions. This review therefore highlights the need for standardization in miRNA research to enhance the reliability of findings. By addressing these methodological challenges, miRNAs could become powerful tools in personalized medicine, aiding in the development of tailored therapeutic strategies for CAD patients and ultimately improving clinical outcomes.
Collapse
Affiliation(s)
- Peter H Nissen
- Department of Clinical Biochemistry, Thrombosis and Haemostasis Research Group, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | |
Collapse
|
3
|
Liu Y, Zhu H, Dai Y, Zhang J, Li Y, Jiang H, Sun Y, Qiao J, Xu X. Isorhynchophylline Inhibits Platelet Activation and Thrombus Formation. J Cardiovasc Pharmacol 2025; 85:137-144. [PMID: 39620592 DOI: 10.1097/fjc.0000000000001655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/15/2024] [Indexed: 02/06/2025]
Abstract
ABSTRACT Isorhynchophylline is a Chinese herbal medicine and has multiple effects such as anti-inflammatory and neuroprotective effects. Whether isorhynchophylline has antithrombotic property is unknown. This study aims to evaluate its role in platelet function. Human platelets were incubated with isorhynchophylline (0, 10, 20, and 40 μM) at 37°C for 1 hour to detect platelet aggregation and activation, receptors level, spreading, and calcium mobilization. In addition, isorhynchophylline (5 mg/kg) was injected into mice to measure in vivo hemostasis and thrombosis. Isorhynchophylline dose-dependently reduced platelet aggregation, adenosine triphosphate secretion, P-selectin expression, and α IIb β 3 activation induced by collagen-related peptide or thrombin without affecting surface level of receptors α IIb β 3 , GPIbα, and glycoprotein VI. Meanwhile, isorhynchophylline-treated platelets showed reduced spreading. Moreover, isorhynchophylline reduced platelet calcium mobilization, phosphatidylserine exposure, and the phosphorylation of PLCγ2 and PKCα. Furthermore, administration of isorhynchophylline into mice impaired platelet hemostatic function and arterial/venous thrombosis without affecting coagulation. In conclusion, isorhynchophylline impairs platelet function and arterial/venous thrombosis, implying its potential to be a novel agent for treating thrombotic or cardiovascular diseases.
Collapse
Affiliation(s)
- Yun Liu
- Department of Clinical Laboratory, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China ; and
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Hui Zhu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Yue Dai
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Jie Zhang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Yingying Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Huimin Jiang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Yueyue Sun
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Xiaoqi Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
4
|
Tan YY, Liu J, Su QP. Advancing Platelet Research Through Live-Cell Imaging: Challenges, Techniques, and Insights. SENSORS (BASEL, SWITZERLAND) 2025; 25:491. [PMID: 39860861 PMCID: PMC11768609 DOI: 10.3390/s25020491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Platelet cells are essential to maintain haemostasis and play a critical role in thrombosis. They swiftly respond to vascular injury by adhering to damaged vessel surfaces, activating signalling pathways, and aggregating with each other to control bleeding. This dynamic process of platelet activation is intricately coordinated, spanning from membrane receptor maturation to intracellular interactions to whole-cell responses. Live-cell imaging has become an invaluable tool for dissecting these complexes. Despite its benefits, live imaging of platelets presents significant technical challenges. This review addresses these challenges, identifying key areas in need of further development and proposing possible solutions. We also focus on the dynamic processes of platelet adhesion, activation, and aggregation in haemostasis and thrombosis, applying imaging capacities from the microscale to the nanoscale. By exploring various live imaging techniques, we demonstrate how these approaches offer crucial insights into platelet biology and deepen our understanding of these three core events. In conclusion, this review provides an overview of the imaging methods currently available for studying platelet dynamics, guiding researchers in selecting suitable techniques for specific studies. By advancing our knowledge of platelet behaviour, these imaging methods contribute to research on haemostasis, thrombosis, and platelet-related diseases, ultimately aiming to improve clinical outcomes.
Collapse
Affiliation(s)
- Yuping Yolanda Tan
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia; (Y.Y.T.); (J.L.)
- Heart Research Institute, Newtown, NSW 2042, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Jinghan Liu
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia; (Y.Y.T.); (J.L.)
- Heart Research Institute, Newtown, NSW 2042, Australia
| | - Qian Peter Su
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia; (Y.Y.T.); (J.L.)
- Heart Research Institute, Newtown, NSW 2042, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
5
|
Tantry US, Becker RC, Singh S, Raghavakurup L, Navarese E, Bliden KP, Gurbel PA. Reassessing the role of aspirin in patients with coronary artery disease. Expert Opin Pharmacother 2024; 25:2307-2317. [PMID: 39505841 DOI: 10.1080/14656566.2024.2427338] [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: 09/02/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/08/2024]
Abstract
INTRODUCTION Recent data question the use of aspirin as a bedrock of antiplatelet therapy in patients with arterial diseases. There are controversies regarding the efficacy of aspirin therapy with respect to specific demographic characteristics, dose and formulations, benefit in primary prevention, and duration in secondary prevention. Importantly, to balance the ischemic benefits and the risk of excessive bleeding following a coronary event, recent studies have investigated strategies to discontinue aspirin therapy and continue with P2Y12 receptor inhibitor monotherapy. However, the precise time when to discontinue aspirin is still unresolved. AREAS COVERED Evidence from recent studies evaluating the role of aspirin in primary and secondary prevention studies was collected from a selective literature search. In this review, the authors discuss current recommendations, large-scale studies of aspirin therapy, controversies, and potential future opportunities for aspirin therapy. EXPERT OPINION With the new evidence showing lower bleeding risk with aspirin-free strategies in both primary and secondary prevention studies, the role of aspirin is being revaluated with P2Y12 receptor inhibitor monotherapy. The potential benefits of novel aspirin formulations and alternative delivery methods, such as inhaled aspirin, are undergoing much-needed investigation with the goal of optimizing care for a wide range of patients.
Collapse
Affiliation(s)
- Udaya S Tantry
- Sinai Center for Thrombosis Research and Drug Development, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Richard C Becker
- Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Sahib Singh
- Sinai Center for Thrombosis Research and Drug Development, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | | | - Eliano Navarese
- Clinical Experimental Cardiology, Clinical and Interventional Cardiology, University of Sassari, Sassari, Sardinia Island, Italy
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
- SIRIO MEDICINE Research Network, Sassari, Italy
| | - Kevin P Bliden
- Sinai Center for Thrombosis Research and Drug Development, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Paul A Gurbel
- Sinai Center for Thrombosis Research and Drug Development, Sinai Hospital of Baltimore, Baltimore, MD, USA
| |
Collapse
|
6
|
Abduljabbar MH. PCSK9 Inhibitors: Focus on Evolocumab and Its Impact on Atherosclerosis Progression. Pharmaceuticals (Basel) 2024; 17:1581. [PMID: 39770423 PMCID: PMC11676890 DOI: 10.3390/ph17121581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/22/2024] [Accepted: 11/15/2024] [Indexed: 01/11/2025] Open
Abstract
This paper investigates the therapeutic use of PCSK9 inhibitors, particularly Evolocumab, as monoclonal antibodies for the treatment of atherosclerosis based on recent literature reviews. PCSK9 is an outstanding example of a breakthrough in medical science, with advancements in understanding its biological function driving substantial progress in atherosclerosis treatment. Atherosclerotic cardiovascular disease (ASCVD) is a leading global cause of mortality, imposing substantial financial burdens on healthcare systems. Elevated low-density lipoprotein cholesterol (LDL-C), a modifiable risk factor, plays a pivotal role in the development of ASCVD. Emerging treatments such as PCSK9 inhibitors are now being introduced to combat this issue, with the goal of reducing ASCVD risk by directly targeting LDL-C levels. This discovery highlighted the potential of monoclonal antibodies to inhibit PCSK9, thereby enhancing LDL-C receptor activity. This breakthrough led to the development of Alirocumab and Evolocumab inhibitors, which typically reduce LDL-C levels by approximately 50%. This research underscores the importance of PCSK9 inhibitors in treating ASCVD, drawing on evidence from various randomized controlled trials such as FOURIER, ODYSSEY OUTCOMES, and VESALIUS-CV. These trials have also shown that PCSK9 inhibitors are effective and safe for the treatment of several cardiovascular disorders. PCSK9 inhibitors are therefore useful in patients who do not reach their target LDL-C levels when on the highest doses of statins or patients with very high cardiovascular risk who cannot tolerate statins at all.
Collapse
Affiliation(s)
- Maram H Abduljabbar
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
7
|
Helin TA, Lemponen M, Immonen K, Lakkisto P, Joutsi-Korhonen L. Circulating microRNAs targeting coagulation and fibrinolysis in patients with severe COVID-19. Thromb J 2024; 22:80. [PMID: 39237986 PMCID: PMC11375984 DOI: 10.1186/s12959-024-00649-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 08/26/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Coronavirus-19 disease (COVID-19) frequently causes coagulation disturbances. Data remains limited on the effects of microRNAs (miRNAs) on coagulation during COVID-19 infection. We aimed to analyze the comprehensive miRNA profile as well as coagulation markers and blood count in hospitalized COVID-19 patients. METHODS Citrated plasma samples from 40 patients (24 men and 16 women) hospitalized for COVID-19 were analyzed. Basic coagulation tests, von Willebrand factor (VWF), ADAMTS13, blood count, C-reactive protein, and 27 miRNAs known to associate with thrombosis or platelet activation were analyzed. MiRNAs were analyzed using quantitative reverse transcription polymerase chain reaction (RT qPCR), with 10 healthy controls serving as a comparator. RESULTS Among the patients, 15/36 (41%) had platelet count of over 360 × 109/L and 10/36 (28%) had low hemoglobin of < 100 g/L, while 26/37 (72%) had high VWF of over 200 IU/dL. Patients had higher levels of the miRNAs miR-27b-3p, miR-320a-3p, miR-320b-3p, and miR-424-5p, whereas levels of miR-103a-3p and miR-145-5p were lower than those in healthy controls. In total, 11 miRNAs were associated with platelet count. Let-7b-3p was associated with low hemoglobin levels of < 100 g/L. miR-24-3p, miR-27b-3p, miR-126-3p, miR-145-5p and miR-338-5p associated with high VWF. CONCLUSION COVID-19 patients differentially express miRNAs with target genes involved in fibrinolysis inhibition, coagulation activity, and increased inflammatory response. These findings support the notion that COVID-19 widely affects hemostasis, including platelets, coagulation and fibrinolysis.
Collapse
Affiliation(s)
- Tuukka A Helin
- HUS Diagnostic Center, Department of Clinical Chemistry, University of Helsinki and Helsinki University Hospital, POB 720, Helsinki, 00029, Finland.
| | - Marja Lemponen
- HUS Diagnostic Center, Department of Clinical Chemistry, University of Helsinki and Helsinki University Hospital, POB 720, Helsinki, 00029, Finland
| | - Katariina Immonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Päivi Lakkisto
- HUS Diagnostic Center, Department of Clinical Chemistry, University of Helsinki and Helsinki University Hospital, POB 720, Helsinki, 00029, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Lotta Joutsi-Korhonen
- HUS Diagnostic Center, Department of Clinical Chemistry, University of Helsinki and Helsinki University Hospital, POB 720, Helsinki, 00029, Finland
| |
Collapse
|
8
|
Chen J, Liu S, Ruan Z, Wang K, Xi X, Mao J. Thrombotic events associated with immune checkpoint inhibitors and novel antithrombotic strategies to mitigate bleeding risk. Blood Rev 2024; 67:101220. [PMID: 38876840 DOI: 10.1016/j.blre.2024.101220] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/23/2024] [Accepted: 06/05/2024] [Indexed: 06/16/2024]
Abstract
Although immunotherapy is expanding treatment options for cancer patients, the prognosis of advanced cancer remains poor, and these patients must contend with both cancers and cancer-related thrombotic events. In particular, immune checkpoint inhibitors are associated with an increased risk of atherosclerotic thrombotic events. Given the fundamental role of platelets in atherothrombosis, co-administration of antiplatelet agents is always indicated. Platelets are also involved in all steps of cancer progression. Classical antithrombotic drugs can cause inevitable hemorrhagic side effects due to blocking integrin β3 bidirectional signaling, which regulates simultaneously thrombosis and hemostasis. Meanwhile, many promising new targets are emerging with minimal bleeding risk and desirable anti-tumor effects. This review will focus on the issue of thrombosis during immune checkpoint inhibitor treatment and the role of platelet activation in cancer progression as well as explore the mechanisms by which novel antiplatelet therapies may exert both antithrombotic and antitumor effects without excessive bleeding risk.
Collapse
Affiliation(s)
- Jiayi Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shuang Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zheng Ruan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Kankan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Xiaodong Xi
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Jianhua Mao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
9
|
Wu C, Li Q, Ma J, Xu X, Sun S, Yang L, Li Y, Li P, Li W, Yu Y. Thrombus aspiration is associated with improved platelet inhibition rate following dual antiplatelet therapy in acute myocardial infarction patients. Eur J Med Res 2024; 29:420. [PMID: 39143607 PMCID: PMC11323623 DOI: 10.1186/s40001-024-02018-9] [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: 01/23/2024] [Accepted: 08/08/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND It is well-established that thrombus aspiration during primary percutaneous coronary intervention (PCI) in patients with acute myocardial infarction (AMI) indicates a higher thrombus burden and necessitates more intensive antithrombotic therapy. The bidirectional association between adverse events in AMI patients and platelet reactivity is typically observed during dual antiplatelet therapy (DAPT). OBJECTIVE To investigate platelet reactivity after DAPT in AMI patients with thrombus aspiration performed during PCI. METHODS In this retrospective study, we examined 269 consecutive AMI patients who underwent PCI and recorded their demographic, clinical and laboratory data. The platelet reactivity was measured with thromboelastogram (TEM). RESULTS Ultimately, 208 patients were included in this study and divided into a Thrombus Aspiration group (N = 97) and a PCI Alone group (N = 111) based on whether thrombus aspiration was performed or not. The adenosine diphosphate (ADP)-induced platelet inhibition rate in the Thrombus Aspiration group was higher than that in the PCI Alone group (P < 0.001). Furthermore, multivariate linear regression analysis revealed that the ADP-induced platelet inhibition rate was independently associated with leukocyte count, thrombus aspiration and the combination of aspirin and ticagrelor as DAPT after adjusting for potential covariates in all AMI patients. CONCLUSION In conclusion, clinicians should exercise heightened attention towards the bleeding risk among patients undergoing PCI concomitant with Thrombus Aspiration postoperatively.
Collapse
Affiliation(s)
- Chunxuan Wu
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qianyi Li
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Juan Ma
- Department of Laboratory Medicine, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoxing Xu
- Clinical Epidemiology Unit, CEU of INCLEN, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shiqun Sun
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lingchao Yang
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanyan Li
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Li
- Department of Pharmacy, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Li
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Ying Yu
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
10
|
Wang X, Yang G, Li J, Meng C, Xue Z. Dynamic molecular signatures of acute myocardial infarction based on transcriptomics and metabolomics. Sci Rep 2024; 14:10175. [PMID: 38702356 PMCID: PMC11068872 DOI: 10.1038/s41598-024-60945-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 04/29/2024] [Indexed: 05/06/2024] Open
Abstract
Acute myocardial infarction (AMI) commonly precedes ventricular remodeling, heart failure. Few dynamic molecular signatures have gained widespread acceptance in mainstream clinical testing despite the discovery of many potential candidates. These unmet needs with respect to biomarker and drug discovery of AMI necessitate a prioritization. We enrolled patients with AMI aged between 30 and 70. RNA-seq analysis was performed on the peripheral blood mononuclear cells collected from the patients at three time points: 1 day, 7 days, and 3 months after AMI. PLC/LC-MS analysis was conducted on the peripheral blood plasma collected from these patients at the same three time points. Differential genes and metabolites between groups were screened by bio-informatics methods to understand the dynamic changes of AMI in different periods. We obtained 15 transcriptional and 95 metabolite expression profiles at three time points after AMI through high-throughput sequencing. AMI-1d: enrichment analysis revealed the biological features of 1 day after AMI primarily included acute inflammatory response, elevated glycerophospholipid metabolism, and decreased protein synthesis capacity. Phosphatidylcholine (PC) and phosphatidylethanolamine (PE) might stand promising biomarkers to differentiate post-AMI stage. Anti-inflammatory therapy during the acute phase is an important direction for preventing related pathology. AMI-7d: the biological features of this stage primarily involved the initiation of cardiac fibrosis response and activation of platelet adhesion pathways. Accompanied by upregulated TGF-beta signaling pathway and ECM receptor interaction, GP5 help assess platelet activation, a potential therapeutic target to improve haemostasis. AMI-3m: the biological features of 3 months after AMI primarily showed a vascular regeneration response with VEGF signaling pathway, NOS3 and SHC2 widely activated, which holds promise for providing new therapeutic approaches for AMI. Our analysis highlights transcriptional and metabolomics signatures at different time points after MI, which deepens our understanding of the dynamic biological responses and associated molecular mechanisms that occur during cardiac repair.
Collapse
Affiliation(s)
- Xuejiao Wang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5 Beixiange, Xicheng District, Beijing, 100053, China
| | - Guang Yang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5 Beixiange, Xicheng District, Beijing, 100053, China
| | - Jun Li
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5 Beixiange, Xicheng District, Beijing, 100053, China.
| | - Chao Meng
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5 Beixiange, Xicheng District, Beijing, 100053, China
| | - Zengming Xue
- Department of Cardiology, Langfang People's Hospital, Hebei Medical University, No. 37, Xinhua Road, Langfang, 065000, China.
| |
Collapse
|
11
|
Bhatia HS, Becker RC, Leibundgut G, Patel M, Lacaze P, Tonkin A, Narula J, Tsimikas S. Lipoprotein(a), platelet function and cardiovascular disease. Nat Rev Cardiol 2024; 21:299-311. [PMID: 37938756 PMCID: PMC11216952 DOI: 10.1038/s41569-023-00947-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/10/2023] [Indexed: 11/09/2023]
Abstract
Lipoprotein(a) (Lp(a)) is associated with atherothrombosis through several mechanisms, including putative antifibrinolytic properties. However, genetic association studies have not demonstrated an association between high plasma levels of Lp(a) and the risk of venous thromboembolism, and studies in patients with highly elevated Lp(a) levels have shown that Lp(a) lowering does not modify the clotting properties of plasma ex vivo. Lp(a) can interact with several platelet receptors, providing biological plausibility for a pro-aggregatory effect. Observational clinical studies suggest that elevated plasma Lp(a) concentrations are associated with worse long-term outcomes in patients undergoing revascularization. Furthermore, in these patients, those with elevated plasma Lp(a) levels derive more benefit from prolonged dual antiplatelet therapy than those with normal Lp(a) levels. The ASPREE trial in healthy older individuals treated with aspirin showed a reduction in ischaemic events in those who had a single-nucleotide polymorphism in LPA that is associated with elevated Lp(a) levels in plasma, without an increase in bleeding events. In this Review, we re-examine the role of Lp(a) in the regulation of platelet function and suggest areas of research to define further the clinical relevance to cardiovascular disease of the observed associations between Lp(a) and platelet function.
Collapse
Affiliation(s)
- Harpreet S Bhatia
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, CA, USA
| | - Richard C Becker
- Heart, Lung and Vascular Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Gregor Leibundgut
- Division of Cardiology, University Hospital of Basel, Basel, Switzerland
| | - Mitul Patel
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, CA, USA
| | - Paul Lacaze
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Andrew Tonkin
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Jagat Narula
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
12
|
Abstract
Viscoelastic testing methods examine the real-time formation of a clot in a whole blood sample, and include thromboelastography (TEG), rotational thromboelastometry (ROTEM), and several other testing platforms. They allow for concurrent assessment of multiple aspects of clotting, including plasmatic coagulation factors, platelets, fibrinogen, and the fibrinolytic pathway. This testing is rapid and may be performed at the point-of-care, allowing for prompt identification of coagulopathies to guide focused and rational administration of blood products as well as the identification of anticoagulant effect. With recent industry progression towards user-friendly, cartridge-based, portable instruments, viscoelastic testing has emerged in the 21st century as a powerful tool to guide blood transfusions in the bleeding patient, and to identify and treat both bleeding and thrombotic conditions in many operative settings, including trauma surgery, liver transplant surgery, cardiac surgery, and obstetrics. In these settings, the use of transfusion algorithms guided by viscoelastic testing data has resulted in widespread improvements in patient blood management as well as modest improvements in select patient outcomes. To address the increasingly wide adoption of viscoelastic methods and the growing number of medical and laboratory personnel tasked with implementing, performing, and interpreting these methods, this chapter provides an overview of the history, physiology, and technology behind viscoelastic testing, as well as a practical review of its clinical utility and current evidence supporting its use. Also included is a review of testing limitations and the contextual role played by viscoelastic methods among all coagulation laboratory testing.
Collapse
Affiliation(s)
- Timothy Carll
- Department of Pathology, University of Chicago, Chicago, IL, United States.
| |
Collapse
|
13
|
Qin H, Wang W, Hu L, Yu Z, Chen Y, Zhao Y, Liao Y, Yang R. New Insights into the Role of HMGB2 in ST-Segment Elevation Myocardial Infarction. Int J Gen Med 2023; 16:4181-4191. [PMID: 37727529 PMCID: PMC10506601 DOI: 10.2147/ijgm.s429761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 08/31/2023] [Indexed: 09/21/2023] Open
Abstract
Background Ischemic heart disease is one of the leading causes of death in the world, of which ST-segment elevation myocardial infarction (STEMI) is an important type. Inappropriate activation and accumulation of platelets typically induced thrombosis, which may result in acute vessel occlusion and STEMI. Multiple cytokines have been shown to regulate platelet activation, but the relationship between HMGB2 and platelet activation has not been elucidated. Methods We collected peripheral blood of STEMI patients and healthy adults, and mass spectrometry analysis of platelet proteins was conducted. The "edgeR" package was used to identify the differentially expressed proteins (DEPs). The Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene ontology (GO) and Gene Set Enrichment Analysis (GSEA) were used to identify the significantly changed pathways. Western blot and ELISA were used to detect the expression of a high mobility group box 2 (HMGB2). Flow cytometric analysis and platelet aggregation rate were performed to evaluate the activation of platelets. Results We identified ALOX5, HIST1H1B, S100A11, HMGB2, and RPS15A were the top five up-regulated proteins by differential expression analysis. Western blot verified that the relative protein expression of HMGB2 in platelet was significantly higher in STEMI patients compared with control adults, and the results of ELISA indicated that the serum HMGB2 level increased and significantly correlated with neutrophil count in STEMI patients. Further investigation showed that the platelet aggregation induced by ADP, the activation of integrin αIIbβ3 and CD62P expression on platelet surface were all enhanced by the recombinant HMGB2 (rHMGB2). Conclusion In conclusion, HMGB2 may be the key molecule to regulate platelet activation in patients with STEMI, which may serve as a potential therapeutic target for STEMI.
Collapse
Affiliation(s)
- Hao Qin
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Wenjun Wang
- Department of Respiratory Diseases, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Longlong Hu
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Zuozhong Yu
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Yang Chen
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Yuanbin Zhao
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Yanhui Liao
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Renqiang Yang
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, People’s Republic of China
| |
Collapse
|
14
|
Di Costanzo A, Indolfi C, Sorrentino S, Esposito G, Spaccarotella CAM. The Effects of Statins, Ezetimibe, PCSK9-Inhibitors, Inclisiran, and Icosapent Ethyl on Platelet Function. Int J Mol Sci 2023; 24:11739. [PMID: 37511498 PMCID: PMC10380733 DOI: 10.3390/ijms241411739] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
This review aims to examine the complex interaction between dyslipidemia, platelet function, and related drug treatments. In particular, the manuscript provides an overview of the effects of major hypolipidemic drugs on platelet function. Indeed, growing evidence supports the view that statins, ezetimibe, PCSK9 inhibitors, inclisiran, and icosapent ethyl also act as antithrombotics. It is known that platelets play a key role not only in the acute phase of coronary syndromes but also in the early phase of atherosclerotic plaque formation. The goal of cholesterol-lowering therapy is to reduce cardiovascular events. The direct effects of cholesterol-lowering drugs are widely described in the literature. Lowering LDL-c (low-density lipoprotein cholesterol) by 1 mmol/L results in a 22-23% reduction in cardiovascular risk. Numerous studies have examined the direct antithrombotic effects of these drugs on platelets, endothelium, monocytes, and smooth muscle cells, and thus, potentially independent of blood LDL-cholesterol reduction. We reviewed in vitro and in vivo studies evaluating the complex interaction between hypercholesterolemia, hypertriglyceridemia, platelet function, and related drug treatments. First, we discussed the role of statins in modulating platelet activation. Discontinuation of statin therapy was associated with increased cardiovascular events with increased ox-LDL, P-selectin, and platelet aggregation. The effect of PCSK9-I (inhibitors of proprotein convertase subtilisin/kexin type 9, PCSK9 involved in the degradation of LDL receptors in the liver) was associated with a statistically significant reduction in platelet reactivity, calculated in P2Y12 reaction units (PRU), in the first 14 days and no difference at 30 days compared to placebo. Finally, in patients with hypertriglyceridemia, the REDUCE-IT study showed that icosapent ethyl (an ethyl ester of eicosapentaenoic acid that reduces triglyceride synthesis and improves triglyceride clearance) resulted in a 25% reduction in ischemic events and cardiovascular death. However, to date, there is not yet clear clinical evidence that the direct antithrombotic effects of the drugs may have a beneficial impact on outcomes independently from the reduction in LDL-C or triglycerides.
Collapse
Affiliation(s)
- Assunta Di Costanzo
- Division of Cardiology, Cardiovascular Research Center, University Magna Graecia Catanzaro, 88100 Catanzaro, Italy
| | - Ciro Indolfi
- Division of Cardiology, Cardiovascular Research Center, University Magna Graecia Catanzaro, 88100 Catanzaro, Italy
| | - Sabato Sorrentino
- Division of Cardiology, Cardiovascular Research Center, University Magna Graecia Catanzaro, 88100 Catanzaro, Italy
| | - Giovanni Esposito
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples Federico II, 80134 Naples, Italy
| | | |
Collapse
|
15
|
Tantry US, Ramotowski B, Kundan P, Gurbel PA. An evaluation of the AggreGuide A-100 ADP Assay for measuring the effect of antiplatelet agents targeting the P2Y 12 receptor. Expert Rev Mol Diagn 2023; 23:555-560. [PMID: 37293873 DOI: 10.1080/14737159.2023.2224166] [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: 02/06/2023] [Accepted: 06/06/2023] [Indexed: 06/10/2023]
Abstract
INTRODUCTION Currently available platelet function assays largely ignore the important characteristics of in vivo thrombus generation, such as flow conditions and shear. The AggreGuide A-100 ADP Assay detects platelet aggregation in whole blood using light scattering under flow conditions. AREAS COVERED In this review article, we discuss the limitations of currently available platelet function assays and the technology underlying the AggreGuide A-100 ADP assay. We also discuss the results of the validation assay study. EXPERT OPINION By incorporating arterial flow conditions and shear, the AggreGuide assay may be more indicative of in vivo thrombus generation as compared to currently available platelet function assays. As per the United States, Food and Drug administration, the AggreGuide A-100 ADP test has been cleared to assess antiplatelet effects of prasugrel and ticagrelor. The assay results are comparable to widely used VerifyNow PRU assay. The utility of AggreGuide A100-ADP Assay in guiding P2Y12 receptor inhibitor therapy in patients with cardiovascular disease needs to be explored in clinical studies.
Collapse
Affiliation(s)
- Udaya S Tantry
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Bogumił Ramotowski
- Department of Cardiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Parshotam Kundan
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Paul A Gurbel
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, MD, USA
| |
Collapse
|
16
|
Tantry US, Duhan S, Navarese E, Ramotowski B, Kundan P, Bliden KP, Gurbel P. An update on novel therapies for treating patients with arterial thrombosis. Expert Rev Hematol 2023; 16:593-605. [PMID: 37335893 DOI: 10.1080/17474086.2023.2227788] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
INTRODUCTION Antithrombotic therapy field is undergoing rapid and significant changes during the past decade. In addition to new therapeutic strategies with existing targets, investigators are exploring the potential use of new targets to address unmet needs to treat patients with arterial diseases. AREAS COVERED We aim to provide an update on and a comprehensive review of the antithrombic agents that are being explored in patients with arterial diseases. We discuss latest developments with respect to upstream antiplatelet agents, and collagen and thrombin pathway inhibitors. We searched PubMed databases for English language articles using keywords: antiplatelet agents, thrombin pathway inhibitors, collagen receptors, arterial disease. EXPERT OPINION Despite implementation of potent P2Y12 inhibitors, there are numerous unmet needs in the treatment of arterial diseases including ceiling effect of currently available antiplatelet agents along with and an elevated risk of bleeding. The latter observations encouraged investigators to explore new targets that can attenuate the generation of platelet-fibrin clot formation and subsequent ischemic event occurrences with minimal effect on bleeding. These targets include collagen receptors on platelets and thrombin generation including FXa, FXIa, and FXIIa. In addition, investigators are studying novel antiplatelet agents/strategies to facilitate upstream therapy in high-risk patients.
Collapse
Affiliation(s)
- Udaya S Tantry
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Sanchit Duhan
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Eliano Navarese
- Interventional Cardiology and Cardiovascular Medicine Research, Department of Cardiology and Internal Medicine, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Bogumil Ramotowski
- Department of Cardiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Parshotam Kundan
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Kevin P Bliden
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Paul Gurbel
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, MD, USA
| |
Collapse
|
17
|
Pan Y, Wang Y, Wang Y, Xu S, Jiang F, Han Y, Hu M, Liu Z. Platelet-derived microvesicles (PMVs) in cancer progression and clinical applications. Clin Transl Oncol 2023; 25:873-881. [PMID: 36417084 DOI: 10.1007/s12094-022-03014-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/12/2022] [Indexed: 11/24/2022]
Abstract
Platelet-derived microvesicles (PMVs), the microvesicles with the highest concentration in the bloodstream, play a key role in the regulation of hemostasis, inflammation, and angiogenesis. PMVs have recently been identified as key factors in the link between platelets and cancer. PMVs bind to both cancer cells and nontransformed cells in the microenvironment of the tumor, and then transfer platelet-derived contents to the target cell. These contents have the potential to either stimulate or modulate the target cell's response. PMVs are encased in a lipid bilayer that contains surface proteins and lipids as well as components found inside the PMV. Each of these components participates in known and potential PMV roles in cancer. The complicated roles played by PMVs in the onset, development, and progression of cancer and cancer-related comorbidities are summarized in this study.
Collapse
Affiliation(s)
- Yan Pan
- Department of Blood Transfusion, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, 100 Minjiang Road, Quzhou, 324000, Zhejiang, China
| | - Yingjian Wang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Yanzhong Wang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Shoufang Xu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Feiyu Jiang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Yetao Han
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Mengsi Hu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Zhiwei Liu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
| |
Collapse
|
18
|
Yamashita A, Asada Y. Underlying mechanisms of thrombus formation/growth in atherothrombosis and deep vein thrombosis. Pathol Int 2023; 73:65-80. [PMID: 36598039 PMCID: PMC11551813 DOI: 10.1111/pin.13305] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 12/03/2022] [Accepted: 12/17/2022] [Indexed: 01/05/2023]
Abstract
Thrombosis remains a leading cause of death worldwide despite technological advances in prevention, diagnosis, and treatment. The traditional view of arterial thrombus formation is that it is a platelet-dependent process, whereas that of venous thrombus formation is a coagulation-dependent process. Current pathological and basic studies on atherothrombosis and venous thrombosis have revealed the diverse participation of platelet and coagulation activation mechanisms in both thrombus initiation and growth processes during clinical thrombotic events. Atherosclerotic plaque cell-derived tissue factor contributes to fibrin formation and platelet aggregation. The degree of plaque disruption and a blood flow alteration promote atherothrombotic occlusion. While blood stasis/turbulent flow due to luminal stenosis itself initiates venous thrombus formation. The coagulation factor XI-driven propagation phase of blood coagulation plays a major role in venous thrombus growth, but a minor role in hemostasis. These lines of evidence indicate that atherothrombosis onset is affected by the thrombogenic potential of atherosclerotic plaques, the plaque disruption size, and an alteration in blood flow. Upon onset of venous thrombosis, enhancement of the propagation phase of blood coagulation under blood stasis and a hypercoagulable state contribute to large thrombus formation.
Collapse
Affiliation(s)
- Atsushi Yamashita
- Pathophysiology Section, Department of Pathology, Faculty of MedicineUniversity of MiyazakiMiyazakiJapan
| | - Yujiro Asada
- Pathophysiology Section, Department of Pathology, Faculty of MedicineUniversity of MiyazakiMiyazakiJapan
- Department of PathologyMiyazaki Medical Association HospitalMiyazakiJapan
| |
Collapse
|
19
|
Small Extracellular Vesicles as a New Class of Medicines. Pharmaceutics 2023; 15:pharmaceutics15020325. [PMID: 36839647 PMCID: PMC9961868 DOI: 10.3390/pharmaceutics15020325] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Extracellular vesicles (EVs) are nanovesicles that are naturally released from cells in a lipid bilayer-bound form. A subset population with a size of 200 nm, small EVs (sEVs), is enticing in many ways. Initially perceived as mere waste receptacles, sEVs have revealed other biological functions, such as cell-to-cell signal transduction and communication. Besides their notable biological functions, sEVs have profound advantages as future drug modalities: (i) excellent biocompatibility, (ii) high stability, and (iii) the potential to carry undruggable macromolecules as cargo. Indeed, many biopharmaceutical companies are utilizing sEVs, not only as diagnostic biomarkers but as therapeutic drugs. However, as all inchoate fields are challenging, there are limitations and hindrances in the clinical translation of sEV therapeutics. In this review, we summarize different types of sEV therapeutics, future improvements, and current strategies in large-scale production.
Collapse
|
20
|
Chen Y, Fu W, Zheng Y, Yang J, Liu Y, Qi Z, Wu M, Fan Z, Yin K, Chen Y, Gao W, Ding Z, Dong J, Li Q, Zhang S, Hu L. Galectin 3 enhances platelet aggregation and thrombosis via Dectin-1 activation: a translational study. Eur Heart J 2022; 43:3556-3574. [PMID: 35165707 PMCID: PMC9989600 DOI: 10.1093/eurheartj/ehac034] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/25/2021] [Accepted: 01/18/2022] [Indexed: 01/25/2023] Open
Abstract
AIMS Galectin-3, a β-galactoside-binding lectin, is abnormally increased in cardiovascular disease. Plasma Galectin-3 receives a Class II recommendation for heart failure management and has been extensively studied for multiple cellular functions. The direct effects of Galectin-3 on platelet activation remain unclear. This study explores the direct effects of Galectin-3 on platelet activation and thrombosis. METHODS AND RESULTS A strong positive correlation between plasma Galectin-3 concentration and platelet aggregation or whole blood thrombus formation was observed in patients with coronary artery disease (CAD). Multiple platelet function studies demonstrated that Galectin-3 directly potentiated platelet activation and in vivo thrombosis. Mechanistic studies using the Dectin-1 inhibitor, laminarin, and Dectin-1-/- mice revealed that Galectin-3 bound to and activated Dectin-1, a receptor not previously reported in platelets, to phosphorylate spleen tyrosine kinase and thus increased Ca2+ influx, protein kinase C activation, and reactive oxygen species production to regulate platelet hyperreactivity. TD139, a Galectin-3 inhibitor in a Phase II clinical trial, concentration dependently suppressed Galectin-3-potentiated platelet activation and inhibited occlusive thrombosis without exacerbating haemorrhage in ApoE-/- mice, which spontaneously developed increased plasma Galectin-3 levels. TD139 also suppressed microvascular thrombosis to protect the heart from myocardial ischaemia-reperfusion injury in ApoE-/- mice. CONCLUSION Galectin-3 is a novel positive regulator of platelet hyperreactivity and thrombus formation in CAD. As TD139 has potent antithrombotic effects without bleeding risk, Galectin-3 inhibitors may have therapeutic advantages as potential antiplatelet drugs for patients with high plasma Galectin-3 levels.
Collapse
Affiliation(s)
- Yufei Chen
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Wanrong Fu
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yunbo Zheng
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Yang
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yangyang Liu
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhiyong Qi
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Meiling Wu
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT, USA
| | - Kanhua Yin
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yunfeng Chen
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Wen Gao
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhongren Ding
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianzeng Dong
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qi Li
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Si Zhang
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Liang Hu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
21
|
Enhanced platelet NLRP3 inflammasome expression in patients with acute coronary syndrome and stable coronary artery disease: A prospective observational study. CARDIOLOGY PLUS 2022. [DOI: 10.1097/cp9.0000000000000018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
22
|
Liu C, Chen J, Chen H, Zhang T, He D, Luo Q, Chi J, Hong Z, Liao Y, Zhang S, Wu Q, Cen H, Chen G, Li J, Wang L. PCSK9 Inhibition: From Current Advances to Evolving Future. Cells 2022; 11:cells11192972. [PMID: 36230934 PMCID: PMC9562883 DOI: 10.3390/cells11192972] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/04/2022] [Accepted: 09/19/2022] [Indexed: 11/18/2022] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a secretory serine protease synthesized primarily by the liver. It mainly promotes the degradation of low-density lipoprotein receptor (LDL-R) by binding LDL-R, reducing low-density lipoprotein cholesterol (LDL-C) clearance. In addition to regulating LDL-R, PCSK9 inhibitors can also bind Toll-like receptors (TLRs), scavenger receptor B (SR-B/CD36), low-density lipoprotein receptor-related protein 1 (LRP1), apolipoprotein E receptor-2 (ApoER2) and very-low-density lipoprotein receptor (VLDL-R) reducing the lipoprotein concentration and slowing thrombosis. In addition to cardiovascular diseases, PCSK9 is also used in pancreatic cancer, sepsis, and Parkinson’s disease. Currently marketed PCSK9 inhibitors include alirocumab, evolocumab, and inclisiran, as well as small molecules, nucleic acid drugs, and vaccines under development. This review systematically summarized the application, preclinical studies, safety, mechanism of action, and latest research progress of PCSK9 inhibitors, aiming to provide ideas for the drug research and development and the clinical application of PCSK9 in cardiovascular diseases and expand its application in other diseases.
Collapse
Affiliation(s)
- Chunping Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510080, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou 510080, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
- Correspondence: (C.L.); (L.W.)
| | - Jing Chen
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Huiqi Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510080, China
| | - Tong Zhang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510080, China
| | - Dongyue He
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510080, China
| | - Qiyuan Luo
- Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Jiaxin Chi
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510080, China
| | - Zebin Hong
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510080, China
| | - Yizhong Liao
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510080, China
| | - Shihui Zhang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510080, China
| | - Qizhe Wu
- Department of Neurosurgery, Institute of Neuroscience, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Huan Cen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510080, China
| | - Guangzhong Chen
- Department of Neurosurgery, Institute of Neuroscience, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Jinxin Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510080, China
| | - Lei Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510080, China
- Correspondence: (C.L.); (L.W.)
| |
Collapse
|
23
|
Dual Antiplatelet Therapy: Guidance for Nurse Practitioners. J Nurse Pract 2022. [DOI: 10.1016/j.nurpra.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
24
|
Xue Y, Zhang L, Zhang L, Sun W, Fang Z, Leng Y, Li M, Ren X, Zhang R, Zhang Y, Chen L, Wang H. Danshensu prevents thrombosis by inhibiting platelet activation via SIRT1/ROS/mtDNA pathways without increasing bleeding risk. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154271. [PMID: 35777120 DOI: 10.1016/j.phymed.2022.154271] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/01/2022] [Accepted: 06/11/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Coronary thrombosis and its correlated disorders are main healthcare problems globally. The therapeutic effects of current treatments involving antiplatelet drugs are not fully satisfactory. Danshensu (DSS) is an important monomer obtained from Salvia miltiorrhiza roots that have been widely employed for vascular diseases in medicinal practices. Nonetheless, the underlying mechanisms of DSS are not fully unraveled. PURPOSE The objective of this study was to penetrate the antithrombotic and antiplatelet mechanisms of DSS. METHODS Network pharmacology assay was used to forecast the cellular mechanisms of DSS for treating thrombosis. The work focused the impacts of DSS on platelet activation by analyzing aggregation and adhesion in vitro. Flow cytometry, western blotting, CM-H2DCFDA staining and mitochondrial function assays were performed to reveal the molecular mechanisms. The model of common carotid artery thrombus induced by ferric chloride was established. The wet weight of thrombus was measured, and the thrombosis was observed by hematoxylin and eosin (H&E) staining, in order to support the inhibitory effect of DSS on thrombosis. RESULTS Data mining found the antithrombotic effect of DSS is related to platelet activation and the core target is silent information regulator 1 (SIRT1). We confirmed that DSS dose-dependently inhibited platelet activation in vitro. DSS was further demonstrated to induce the expression of SIRT1 and decreased reactive oxygen species (ROS) burden and thereby prevented mitochondrial dysfunction. Mitochondrial function tests further indicated that DSS prevented mitochondrial DNA (mtDNA) release, which induced activation of platelet in a dendritic cell specific intercellular-adhesion-molecule-3 grabbing non-integrin (DC-SIGN)-dependent manner. In carotid artery injury model induced by ferric chloride, DSS inhibited the development of carotid arterial thrombosis. More encouragingly, in tail bleeding time assay, DSS did not augment bleeding risk. CONCLUSION These findings indicated that DSS effectively inhibited platelet activation by depressing the collection of ROS and the release of platelet mtDNA without arousing hemorrhage risk. DSS might represent a promising candidate drug for thrombosis and cardiovascular disease therapeutics.
Collapse
Affiliation(s)
- Yuejin Xue
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China; School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China
| | - Liyuan Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China
| | - Lusha Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China
| | - Wei Sun
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China
| | - Zhirui Fang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China
| | - Yuze Leng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China
| | - Mengyao Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China
| | - Xiuyun Ren
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China
| | - Rui Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China
| | - Yingxue Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China
| | - Lu Chen
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, 301617 Tianjin, China; Tianjin State Key Laboratory of Modern Chinese Medicine, 301617 Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China.
| | - Hong Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, 301617 Tianjin, China; Tianjin State Key Laboratory of Modern Chinese Medicine, 301617 Tianjin, China; School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China.
| |
Collapse
|
25
|
Zhou H, Zhu J, Wan H, Shao C, Chen T, Yang J, He Y, Wan H. The combination of danhong injection plus tissue plasminogen activator ameliorates mouse tail thrombosis-induced by κ-carrageenan. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154320. [PMID: 35830758 DOI: 10.1016/j.phymed.2022.154320] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 06/22/2022] [Accepted: 07/05/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND After thrombosis, t-PA thrombolysis is the first choice, but the use of t-PA can easily lead to hemorrhagic injury and neurotoxicity. The combination of Danhong injection (DHI) and tissue plasminogen activator (t-PA) therapy may be a new strategy to find high-efficiency anti-thrombosis and low bleeding risk. However, nothing is about the effect of DHI plus t-PA on platelet activation. PURPOSE The present research was to explore the optimal dose of DHI and t-PA in vivo and mechanisms involved with the treatment of combining DHI and t-PA for thrombotic disease and determined whether DHI plus t-PA affects thrombotic processes related to platelet activation. METHODS Mice were induced by administering κ-carrageenan intraperitoneally, the ratio of different doses of DHI and t-PA in vivo, and the optimal dose effects on platelet aggregation, platelet adhesion, thrombosis formation, and platelet activation were determined. The effects of the αIIbβ3 signaling pathway were analyzed in mice. RESULTS In vitro, DHI (62% v/v), t-PA (1 mg/ml), and DHI + t-PA (62% v/v + 1 mg/ml) decreased rat platelet aggregation and adhesion, with a stronger effect from the combination as compared to t-PA monotherapy. In vivo, injections of κ-carrageenan were used to induce BALB/c mice. The optimal dose of DHI, t-PA, and DHI + t-PA is 12 ml/kg, 10 mg/kg, and 12 ml/kg + 7.5 mg/kg. The administration of DHI (12 ml/kg), t-PA (10 mg/kg), and DHI + t-PA (12 ml/kg + 7.5 mg/kg) decreased thrombi in mouse tissue vessels. Furthermore, the reduction of thrombosis formation by DHI, t-PA, and DHI + t-PA was related to lower collagen deposition, and lowered expressions of collagen I, matrix metalloproteinase 2 (MMP-2), and metalloproteinase 9 (MMP-9) in mouse tails, with increased efficacy in combination as compared to t-PA alone. The anti-thrombosis actions of DHI, t-PA, and their combination regulated the expression of CD41, purinergic receptor (P2Y12), guanine nucleotide-binding protein G (q) subunit alpha (GNAQ), phosphatidylinositol phospholipase c beta (PLCβ), Ras-related protein 1 (Rap1), RIAM, talin1, fibrinogen alpha chain (FG), kindlin-3, and RAS guany1-releasing protein 1 (RasGRP1). CONCLUSIONS Based on expression, the mechanism responsible for thrombosis may be attributed to platelet activation via the αIIbβ3 signaling pathway. Combination therapy with DHI and t-PA exerted potent thrombolytic effects. Thus, our data can be used as a foundation for further clinical studies examining the efficacy of traditional Chinese medicines for the treatment of thrombosis.
Collapse
Affiliation(s)
- Huifen Zhou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Jiaqi Zhu
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Haofang Wan
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Chongyu Shao
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Tianhang Chen
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Jiehong Yang
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, PR China.
| | - Yu He
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, PR China.
| | - Haitong Wan
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, PR China; Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China.
| |
Collapse
|
26
|
Lv H, Yang Z, Wu H, Liu M, Mao X, Liu X, Ding H, Shi Z, Zhou Y, Liu Q, Zhang Y, Zhou Y, Chen K, Li Z, Dong Q, Ma J, Han Y. High On-Treatment Platelet Reactivity as Predictor of Long-term Clinical Outcomes in Stroke Patients with Antiplatelet Agents. Transl Stroke Res 2022; 13:391-398. [PMID: 34596891 DOI: 10.1007/s12975-021-00949-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 08/20/2021] [Accepted: 09/17/2021] [Indexed: 01/01/2023]
Abstract
The purpose was to explore the value of high on-treatment platelet reactivity (HTPR) in predicting long-term clinical outcomes for stroke patients. The platelet reactivity was assayed after being treated with either 75 mg clopidogrel or 100 mg aspirin daily with VerifyNow System in stroke patients. HTPR for clopidogrel was defined as PRU ≥ 208, and that for aspirin was defined as ARU ≥ 550. CYP2C19 genotyping was performed using the Sequenom MassARRAY iPLEX platform. The primary endpoint was a composite of recurrent ischemic stroke, transient ischemic attack, myocardial infarction, or ischemic vascular death. The safety endpoint was bleeding. In the clopidogrel group, among 345 patients recruited, 174 of them were categorized as HTPR. A total of 270 patients were followed up for 54 months. There was a significant association between HTPR and the primary endpoint (HRadj 2.13 [95% CI, 1.43-3.15], p < 0.001). Among the 314 participants genotyped for CYP2C19, 187 (59.6%) were classified as CYP2C19 loss-of-function allele carriers. Patients with at least 1 loss-of-function allele were more likely to present with HTPR (ORadj 2.61 [95%CI, 1.43-4.77], p = 0.008), and had a higher risk of the primary endpoint (HRadj 2.05 [95% CI, 1.30, 3.25], p = 0.002). In the aspirin group, among 140 patients recruited, 28 of them were categorized as HTPR. A total of 121 patients were followed up for 30 months. Similarly, there was a significant association between HTPR and the primary endpoint (HRadj 3.28 [95% CI, 1.52-7.71], p = 0.002). HTPR is an independent risk factor for ischemic events during long-term follow-up in stroke patients. Platelet function testing is helpful to evaluate the effect of antiplatelet therapy for stroke patients.
Collapse
Affiliation(s)
- Huihui Lv
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, China
| | - Zidong Yang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Haibo Wu
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, China
| | - Mingyuan Liu
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, China
| | - Xiaowei Mao
- Department of Neurology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xu Liu
- School of Life Sciences, Fudan University, Shanghai, China
| | - Hongyan Ding
- Department of Neurology, Huashan Hospital, Fudan University, 12 Urumqi Zhong Lu, Shanghai, China
| | - Zhuqing Shi
- School of Life Sciences, Fudan University, Shanghai, China
| | - Yang Zhou
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, China
| | - Qianyun Liu
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, China
| | - Yongkang Zhang
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, China
| | - Yinting Zhou
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, China
| | - Kai Chen
- Maternal Fetal Medicine, Aurora Medical Group, Sinai Medical Center, Milwaukee, WI, USA
| | - Zezhi Li
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, Fudan University, 12 Urumqi Zhong Lu, Shanghai, China.
| | - Jianpeng Ma
- Multiscale Research Institute of Complex Systems, Fudan University, 220 Handan Road, Shanghai, China.
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA.
- Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, China.
| | - Yan Han
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, China.
| |
Collapse
|
27
|
Application and Prospect of Platelet Multi-Omics Technology in Study of Blood Stasis Syndrome. Chin J Integr Med 2021; 28:99-105. [PMID: 34935097 DOI: 10.1007/s11655-021-3349-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 10/19/2022]
Abstract
The abnormality of platelet function plays an important role in the pathogenesis and evolution of blood stasis syndrome (BSS). The explanation of its mechanism is a key scientific issue in the study of cardiovascular and cerebrovascular diseases and treatment. System biology technology provides a good technical platform for further development of platelet multi-omics, which is conducive to the scientific interpretation of the biological mechanism of BSS. The article summarized the pathogenesis of platelets in BSS, the mechanism of action of blood activating and stasis resolving drugs, and the application of genomics, proteomics, and metabonomics in platelet research, and put forward the concept of "plateletomics in BSS". Through the combination and cross-validation of multi-omics technology, it mainly focuses on the clinical and basic research of cardiovascular and cerebrovascular diseases; through the interactive verification of multi-omics technology and system biology, it mainly focuses on the platelet function and secretion system. The article systematically explains the molecular biological mechanism of platelet activation, aggregation, release, and other stages in the formation and development of BSS, and provides a new research idea and method for clarifying the pathogenesis of BSS and the mechanism of action of blood activating and stasis resolving drugs.
Collapse
|
28
|
Annarapu GK, Nolfi-Donegan D, Reynolds M, Wang Y, Kohut L, Zuckerbraun B, Shiva S. Heme stimulates platelet mitochondrial oxidant production to induce targeted granule secretion. Redox Biol 2021; 48:102205. [PMID: 34891098 PMCID: PMC8661700 DOI: 10.1016/j.redox.2021.102205] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/24/2021] [Accepted: 12/04/2021] [Indexed: 10/19/2022] Open
Abstract
Hemolysis, a pathological component of many diseases, is associated with thrombosis and vascular dysfunction. Hemolytic products, including cell-free hemoglobin and free heme directly activate platelets. However, the effect of hemolysis on platelet degranulation, a central process in not only thrombosis, but also inflammatory and mitogenic signaling, remains less clear. Our group showed that hemoglobin-induced platelet activation involved the production of mitochondrial reactive oxygen species (mtROS). However, the molecular mechanism by which extracellular hemolysis induces platelet mtROS production, and whether these mtROS regulate platelet degranulation remains unknown. Here, we demonstrate using isolated human platelets that cell free heme is a more potent agonist for platelet activation than hemoglobin, and stimulates the release of a specific set of molecules, including the glycoprotein thrombospondin-1 (TSP-1), from the α-granule of platelets. We uncover the mechanism of heme-mediated platelet mtROS production which is dependent on the activation of platelet toll-like receptor 4 (TLR4) signaling and leads to the downstream phosphorylation and inhibition of complex-V by the serine kinase Akt. Notably, inhibition of platelet TLR4 or Akt, or scavenging of mtROS prevents heme-induced granule release in vitro. Further, heme-dependent granule release is significantly attenuated in vivo in mice lacking TLR4 or those treated with the mtROS scavenger MitoTEMPO. These data elucidate a novel mechanism of TLR4-mediated mitochondrial regulation, establish the mechanistic link between hemolysis and platelet degranulation, and begin to define the heme and mtROS-dependent platelet secretome. These data have implications for hemolysis-induced thrombo-inflammatory signaling and for the consideration of platelet mitochondria as a therapeutic target in hemolytic disorders.
Collapse
Affiliation(s)
- Gowtham K Annarapu
- Pittsburgh Heart, Lung, Blood, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Deirdre Nolfi-Donegan
- Pittsburgh Heart, Lung, Blood, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA; Department of Pediatrics, Division of Hematology/Oncology, Children's Hospital of Pittsburgh, Pittsburgh, PA, 15224, USA
| | - Michael Reynolds
- Pittsburgh Heart, Lung, Blood, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Yinna Wang
- Pittsburgh Heart, Lung, Blood, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Lauryn Kohut
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Brian Zuckerbraun
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Sruti Shiva
- Pittsburgh Heart, Lung, Blood, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
29
|
Gurbel P, Tantry U. Is clopidogrel as the P2Y 12 inhibitor a wise choice for long-term monotherapy in patients undergoing stenting? EUROINTERVENTION 2021; 17:e865-e866. [PMID: 34870596 PMCID: PMC9707458 DOI: 10.4244/eijv17i11a144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Paul Gurbel
- Sinai Center for Thrombosis Research and Drug Development, Sinai Hospital of Baltimore, 2401 W Belvedere Ave, Baltimore, MD 21215, USA
| | - Udaya Tantry
- Sinai Center for Thrombosis Research and Drug Development, Sinai Hospital of Baltimore, Baltimore, MD, USA
| |
Collapse
|
30
|
Racine-Brzostek SE, Asmis LM. Assessment of platelet function utilizing viscoelastic testing. Transfusion 2021; 60 Suppl 6:S10-S20. [PMID: 33089932 DOI: 10.1111/trf.16081] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/29/2020] [Accepted: 08/29/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Sabrina E Racine-Brzostek
- Department of Pathology and Laboratory Medicine, New York-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, USA
| | - Lars M Asmis
- Centre for Perioperative Thrombosis and Haemostasis, Zurich, Switzerland
| |
Collapse
|
31
|
Gartshteyn Y, Mor A, Shimbo D, Khalili L, Kapoor T, Geraldino-Pardilla L, Alexander RV, Conklin J, Dervieux T, Askanase AD. Platelet bound complement split product (PC4d) is a marker of platelet activation and arterial vascular events in Systemic Lupus Erythematosus. Clin Immunol 2021; 228:108755. [PMID: 33984497 DOI: 10.1016/j.clim.2021.108755] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 05/05/2021] [Accepted: 05/08/2021] [Indexed: 11/25/2022]
Abstract
Platelet-bound complement activation products (PC4d) are associated with thrombosis in Systemic Lupus Erythematosus (SLE). This study investigated the effect of PC4d on platelet function, as a mechanistic link to arterial thrombosis. In a cohort of 150 SLE patients, 13 events had occurred within five years of enrollment. Patients with arterial events had higher PC4d levels (13.6 [4.4-24.0] vs. 4.0 [2.5-8.3] net MFI), with PC4d 10 being the optimal cutoff for event detection. The association of arterial events with PC4d remained significant after adjusting for antiphospholipid status, smoking, and prednisone use (p = 0.045). PC4d levels correlated with lower platelet counts (r = -0.26, p = 0.002), larger platelet volumes (r = 0.22, p = 0.009) and increased platelet aggregation: the adenosine diphosphate (ADP) concentration to achieve 50% maximal aggregation (EC50) was lower in patients with PC4d 10 compared with PC4d < 10 (1.6 vs. 3.7, p = 0.038, respectively). These results suggest that PC4d may be a mechanistic marker for vascular disease in SLE.
Collapse
Affiliation(s)
- Yevgeniya Gartshteyn
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States of America.
| | - Adam Mor
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States of America
| | - Daichi Shimbo
- Center for Behavioral Cardiovascular Health, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, United States of America
| | - Leila Khalili
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States of America
| | - Teja Kapoor
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States of America
| | - Laura Geraldino-Pardilla
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States of America
| | | | - John Conklin
- Exagen Diagnostics Inc, Vista, CA, United States of America
| | | | - Anca D Askanase
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States of America
| |
Collapse
|
32
|
Net platelet clot strength of thromboelastography platelet mapping assay for the identification of high on-treatment platelet reactivity in post-PCI patients. Biosci Rep 2021; 40:225736. [PMID: 32639536 PMCID: PMC7364512 DOI: 10.1042/bsr20201346] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/30/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023] Open
Abstract
High-on treatment platelet reactivity (HTPR) leads to more prevalence of thrombotic event in patients undergoing percutaneous coronary interventions (PCI). Dual antiplatelet therapy with aspirin in addition to one P2Y12 inhibitor is commonly administrated to reduce HTPR. However, ‘one size fits all’ antiplatelet strategy is widely implemented due to lacking benefits with tailored strategy. One reason for the failure of tailored treatment might be less specificity of the current indicators for HTPR. Therefore, searching for specific indicators for HTPR is critical. Thromboelastograph with platelet mapping (TEGpm) assay has been explored for identifying HTRP. Variables of TEGpm assay, including maximum amplitude (MA) induced by thrombin (MAthrombin), R time, platelet aggregation rate induced by ADP (TEGaradp) and MA induced by ADP (MAadp) have been demonstrated to be able to identify HTPR in post-PCI patients. However, these variables for HTPR might be less specific. Thus, in the present study, a novel variable nMAadp was derived by removing fibrin contribution from MAadp and analyzed for its usefulness in determining HTPR. In addition, MAthrombin, R time, MAadp and TEGaradp were also examined for determining HTPR. In conclusion, nMAadp and TEGaradp were demonstrated to be independent indicators for HTPR; nMAadp had the strongest power to identify HTPR with cutoff value of 26.3 mm; MAthrombin and R time were not significantly different between patients with and without HTPR; combination of TEGaradp and nMAadp further improved the ability to identify HTPR with an AUC of 0.893.
Collapse
|
33
|
Czajka P, Fitas A, Jakubik D, Eyileten C, Gasecka A, Wicik Z, Siller-Matula JM, Filipiak KJ, Postula M. MicroRNA as Potential Biomarkers of Platelet Function on Antiplatelet Therapy: A Review. Front Physiol 2021; 12:652579. [PMID: 33935804 PMCID: PMC8081881 DOI: 10.3389/fphys.2021.652579] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/22/2021] [Indexed: 11/25/2022] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding RNAs, able to regulate cellular functions by specific gene modifications. Platelets are the major source for circulating miRNAs, with significant regulatory potential on cardiovascular pathophysiology. MiRNAs have been shown to modify the expression of platelet proteins influencing platelet reactivity. Circulating miRNAs can be determined from plasma, serum, or whole blood, and they can be used as diagnostic and prognostic biomarkers of platelet reactivity during antiplatelet therapy as well as novel therapeutic targets in cardiovascular diseases (CVDs). Herein, we review diagnostic and prognostic value of miRNAs levels related to platelet reactivity based on human studies, presenting its interindividual variability as well as the substantial role of genetics. Furthermore, we discuss antiplatelet treatment in the context of miRNAs alterations related to pathways associated with drug response.
Collapse
Affiliation(s)
- Pamela Czajka
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology, Warsaw, Poland
| | - Alex Fitas
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology, Warsaw, Poland
| | - Daniel Jakubik
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology, Warsaw, Poland
| | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology, Warsaw, Poland
| | - Aleksandra Gasecka
- First Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Zofia Wicik
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology, Warsaw, Poland.,Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Paulo, Brazil
| | - Jolanta M Siller-Matula
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology, Warsaw, Poland.,Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Krzysztof J Filipiak
- First Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Marek Postula
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology, Warsaw, Poland
| |
Collapse
|
34
|
Kim HK, Tantry US, Smith SC, Jeong MH, Park SJ, Kim MH, Lim DS, Shin ES, Park DW, Huo Y, Chen SL, Bo Z, Goto S, Kimura T, Yasuda S, Chen WJ, Chan M, Aradi D, Geisler T, Gorog DA, Sibbing D, Lip GYH, Angiolillo DJ, Gurbel PA, Jeong YH. The East Asian Paradox: An Updated Position Statement on the Challenges to the Current Antithrombotic Strategy in Patients with Cardiovascular Disease. Thromb Haemost 2021; 121:422-432. [PMID: 33171520 DOI: 10.1055/s-0040-1718729] [Citation(s) in RCA: 183] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
East Asian patients have reduced anti-ischemic benefits and increased bleeding risk during antithrombotic therapies compared with Caucasian patients. As potent P2Y12 receptor inhibitors (e.g., ticagrelor and prasugrel) and direct oral anticoagulants are commonly used in current daily practice, the unique risk-benefit trade-off in East Asians has been a topic of emerging interest. In this article, we propose updated evidence and future directions of antithrombotic treatment in East Asian patients.
Collapse
Affiliation(s)
- Hyun Kuk Kim
- Department of Cardiology, Chosun University Hospital, Gwangju, South Korea
| | - Udaya S Tantry
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, Maryland, United States
| | - Sidney C Smith
- Division of Cardiology, University of North Carolina, Chapel Hill, North Carolina, United States
| | - Myung Ho Jeong
- Department of Cardiology, Chonnam National University Hospital, Gwangju, South Korea
| | - Seung-Jung Park
- The Heart Institute, Asan Medical Center, University of Ulsan, Seoul, South Korea
| | - Moo Hyun Kim
- Department of Cardiology, Dong-A University Hospital, Busan, South Korea
| | - Do-Sun Lim
- Department of Cardiology, Cardiovascular Center, Korea University Anam Hospital, Seoul, South Korea
| | - Eun-Seok Shin
- Division of Cardiology, Ulsan Hospital, Ulsan, South Korea
| | - Duk-Woo Park
- The Heart Institute, Asan Medical Center, University of Ulsan, Seoul, South Korea
| | - Yong Huo
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Shao-Liang Chen
- Cardiovascular Department, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Zheng Bo
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Shinya Goto
- Department of Medicine (Cardiology), Tokai University School of Medicine, Kanagawa, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Satoshi Yasuda
- National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Wen-Jone Chen
- Department of Internal Medicine, Cardiology Division, National Taiwan University Hospital, Taipei, Taiwan
| | - Mark Chan
- Department of Cardiology, National University Heart Centre Singapore, Singapore, Singapore
| | - Daniel Aradi
- Heart Centre Balatonfüred and Heart and Vascular Centre, Semmelweis University, Budapest, Hungary
| | - Tobias Geisler
- Department of Cardiology and Cardiovascular Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Diana A Gorog
- National Heart and Lung Institute, Imperial College, London, United Kingdom
- Postgraduate Medical School, University of Hertfordshire, Hertfordshire, United Kingdom
| | - Dirk Sibbing
- Department of Cardiology, LMU München, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom
| | - Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, Florida, United States
| | - Paul A Gurbel
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, Maryland, United States
| | - Young-Hoon Jeong
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Cardiovascular Center, Gyeongsang National University Changwon Hospital, Changwon, South Korea
- Institute of the Health Sciences, Gyeongsang National University, Jinju, South Korea
| |
Collapse
|
35
|
Zuo X, Li Q, Ya F, Ma LJ, Tian Z, Zhao M, Fan D, Zhao Y, Mao YH, Wan JB, Yang Y. Ginsenosides Rb2 and Rd2 isolated from Panax notoginseng flowers attenuate platelet function through P2Y 12-mediated cAMP/PKA and PI3K/Akt/Erk1/2 signaling. Food Funct 2021; 12:5793-5805. [PMID: 34041517 DOI: 10.1039/d1fo00531f] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Saponins derived from Panax notoginseng root are widely used as herbal medicines and dietary supplements due to their wide range of health benefits. However, the effects of those from Panax notoginseng flowers (PNF) on platelet function and thrombus formation remain largely unknown. Using a series of platelet function assays, we found that G-Rb2 and G-Rd2, among the ten PNF saponin monomers, significantly inhibited human platelet aggregation and activation induced by adenosine diphosphate (ADP) in vitro. The 50% inhibitory concentration (IC50) of G-Rb2 and G-Rd2 against ADP-induced platelet aggregation was 85.5 ± 4.5 μg mL-1 and 51.4 ± 4.6 μg mL-1, respectively. Mechanistically, G-Rb2 and G-Rd2 could effectively modulate platelet P2Y12-mediated signaling by up-regulating cAMP/PKA signaling and down-regulating PI3K/Akt/Erk1/2 signaling pathways. Co-incubation of the P2Y12 antagonist cangrelor with either G-Rb2 or G-Rd2 did not show significant additive inhibitory effects. G-Rb2 and G-Rd2 also substantially suppressed thrombus growth in a FeCl3-induced murine arteriole thrombosis model in vivo. Interestingly, G-Rd2 generally exhibited more potent inhibitory effects on platelet function and thrombus formation than G-Rb2. Thus, our data suggest that PNF-derived G-Rb2 and G-Rd2 effectively attenuate platelet hyperactivity through modulating signaling pathways downstream of P2Y12, which indicates G-Rb2 and G-Rd2 may play important preventive roles in thrombotic diseases.
Collapse
Affiliation(s)
- Xiao Zuo
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China. and Guangdong Provincial Key Laboratory for Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, China and Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, Guangdong Province 510080, China
| | - Qing Li
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China. and Guangdong Provincial Key Laboratory for Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, China and Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, Guangdong Province 510080, China
| | - Fuli Ya
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China. and Guangdong Provincial Key Laboratory for Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, China and Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, Guangdong Province 510080, China
| | - Li-Juan Ma
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau 999078, China.
| | - Zezhong Tian
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China. and Guangdong Provincial Key Laboratory for Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, China and Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, Guangdong Province 510080, China
| | - Mingzhu Zhao
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China. and Guangdong Provincial Key Laboratory for Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, China and Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, Guangdong Province 510080, China
| | - Die Fan
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China. and Guangdong Provincial Key Laboratory for Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, China and Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, Guangdong Province 510080, China
| | - Yimin Zhao
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China. and Guangdong Provincial Key Laboratory for Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, China and Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, Guangdong Province 510080, China
| | - Yu-Heng Mao
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China. and Guangdong Provincial Key Laboratory for Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, China and Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, Guangdong Province 510080, China
| | - Jian-Bo Wan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau 999078, China.
| | - Yan Yang
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China. and Guangdong Provincial Key Laboratory for Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, China and Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, Guangdong Province 510080, China
| |
Collapse
|
36
|
Qi Z, Hu L, Zhang J, Yang W, Liu X, Jia D, Yao Z, Chang L, Pan G, Zhong H, Luo X, Yao K, Sun A, Qian J, Ding Z, Ge J. PCSK9 (Proprotein Convertase Subtilisin/Kexin 9) Enhances Platelet Activation, Thrombosis, and Myocardial Infarct Expansion by Binding to Platelet CD36. Circulation 2020; 143:45-61. [PMID: 32988222 DOI: 10.1161/circulationaha.120.046290] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND PCSK9 (proprotein convertase subtilisin/kexin 9), mainly secreted by the liver and released into the blood, elevates plasma low-density lipoprotein cholesterol by degrading low-density lipoprotein receptor. Pleiotropic effects of PCSK9 beyond lipid metabolism have been shown. However, the direct effects of PCSK9 on platelet activation and thrombosis, and the underlying mechanisms, as well, still remain unclear. METHODS We detected the direct effects of PCSK9 on agonist-induced platelet aggregation, dense granule ATP release, integrin αIIbβ3 activation, α-granule release, spreading, and clot retraction. These studies were complemented by in vivo analysis of FeCl3-injured mouse mesenteric arteriole thrombosis. We also investigated the underlying mechanisms. Using the myocardial infarction (MI) model, we explored the effects of PCSK9 on microvascular obstruction and infarct expansion post-MI. RESULTS PCSK9 directly enhances agonist-induced platelet aggregation, dense granule ATP release, integrin αIIbβ3 activation, P-selectin release from α-granules, spreading, and clot retraction. In line, PCSK9 enhances in vivo thrombosis in a FeCl3-injured mesenteric arteriole thrombosis mouse model, whereas PCSK9 inhibitor evolocumab ameliorates its enhancing effects. Mechanism studies revealed that PCSK9 binds to platelet CD36 and thus activates Src kinase and MAPK (mitogen-activated protein kinase)-extracellular signal-regulated kinase 5 and c-Jun N-terminal kinase, increases the generation of reactive oxygen species, and activates the p38MAPK/cytosolic phospholipase A2/cyclooxygenase-1/thromboxane A2 signaling pathways downstream of CD36 to enhance platelet activation, as well. Using CD36 knockout mice, we showed that the enhancing effects of PCSK9 on platelet activation are CD36 dependent. It is important to note that aspirin consistently abolishes the enhancing effects of PCSK9 on platelet activation and in vivo thrombosis. Last, we showed that PCSK9 activating platelet CD36 aggravates microvascular obstruction and promotes MI expansion post-MI. CONCLUSIONS PCSK9 in plasma directly enhances platelet activation and in vivo thrombosis, and MI expansion post-MI, as well, by binding to platelet CD36 and thus activating the downstream signaling pathways. PCSK9 inhibitors or aspirin abolish the enhancing effects of PCSK9, supporting the use of aspirin in patients with high plasma PCSK9 levels in addition to PCSK9 inhibitors to prevent thrombotic complications.
Collapse
Affiliation(s)
- Zhiyong Qi
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (Z.Q., W.Y., D.J., Z.Y., K.Y., A.S., J.Q., J.G.)
| | - Liang Hu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, China (L.H., Z.D.)
| | - Jianjun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (J.Z., L.C., G.P., Z.D.)
| | - Wenlong Yang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (Z.Q., W.Y., D.J., Z.Y., K.Y., A.S., J.Q., J.G.)
| | - Xin Liu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (Z.Q., W.Y., D.J., Z.Y., K.Y., A.S., J.Q., J.G.)
| | - Daile Jia
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (Z.Q., W.Y., D.J., Z.Y., K.Y., A.S., J.Q., J.G.)
| | - Zhifeng Yao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (Z.Q., W.Y., D.J., Z.Y., K.Y., A.S., J.Q., J.G.)
| | - Lin Chang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (J.Z., L.C., G.P., Z.D.)
| | - Guanxing Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (J.Z., L.C., G.P., Z.D.)
| | - Haoxuan Zhong
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China (H.Z., X. Luo)
| | - Xinping Luo
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China (H.Z., X. Luo)
| | - Kang Yao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (Z.Q., W.Y., D.J., Z.Y., K.Y., A.S., J.Q., J.G.)
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (Z.Q., W.Y., D.J., Z.Y., K.Y., A.S., J.Q., J.G.)
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (Z.Q., W.Y., D.J., Z.Y., K.Y., A.S., J.Q., J.G.)
| | - Zhongren Ding
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, China (L.H., Z.D.).,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China (J.Z., L.C., G.P., Z.D.)
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (Z.Q., W.Y., D.J., Z.Y., K.Y., A.S., J.Q., J.G.)
| |
Collapse
|
37
|
Caiazzo G, Musci RL, Frediani L, Umińska J, Wanha W, Filipiak KJ, Kubica J, Navarese EP. State of the Art: No-Reflow Phenomenon. Cardiol Clin 2020; 38:563-573. [PMID: 33036718 DOI: 10.1016/j.ccl.2020.07.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Primary percutaneous coronary intervention is the preferred reperfusion strategy for the management of acute ST-segment elevation myocardial infarction. No reflow is characterized by the inadequate myocardial perfusion of a given segment without angiographic evidence of persistent mechanical obstruction of epicardial vessels. Both pharmacologic and device-based strategies have been tested to resolve coronary no reflow. This article provides an updated overview of the no-reflow phenomenon, discussing clinical evidence and ongoing investigations of existing and novel therapeutic strategies to counteract it.
Collapse
Affiliation(s)
- Gianluca Caiazzo
- ICCU, San Giuseppe Moscati Hospital, ASL CE, Via Gramsci 1, Aversa 81031, Italy
| | - Rita Leonarda Musci
- Department of Cardiology, Azienda Ospedaliera Bonomo, Viale Istria, Andria BT 76123, Italy
| | - Lara Frediani
- Department of Cardiology, Livorno Hospital, Azienda Usl Toscana Nord-Ovest, Ospedali Riuniti di Livorno, Viale Vittorio Alfieri, 36, Livorno LI 57124, Italy
| | - Julia Umińska
- Department of Cardiology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, SIRIO MEDICINE Network, ul. Jagiellońska 13-15, Bydgoszcz 85-067, Poland
| | - Wojciech Wanha
- Division of Cardiology and Structural Heart Diseases, Medical University of Silesia, ul. Józefa Poniatowskiego 15, Kato 40-055, Katowice, Poland
| | - Krzysztof J Filipiak
- Department of Cardiology, Medical University of Warsaw, Żwirki i Wigury 61, Warszawa 02-091, Poland
| | - Jacek Kubica
- Department of Cardiology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, SIRIO MEDICINE Network, ul. Jagiellońska 13-15, Bydgoszcz 85-067, Poland
| | - Eliano Pio Navarese
- Department of Cardiology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, SIRIO MEDICINE Network, ul. Jagiellońska 13-15, Bydgoszcz 85-067, Poland; University of Alberta, 116 Street & 85 Avenue, Edmonton, AB T6G 2R3, Canada.
| |
Collapse
|
38
|
Terrier J, Daali Y, Fontana P, Csajka C, Reny JL. Towards Personalized Antithrombotic Treatments: Focus on P2Y 12 Inhibitors and Direct Oral Anticoagulants. Clin Pharmacokinet 2020; 58:1517-1532. [PMID: 31250210 DOI: 10.1007/s40262-019-00792-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Oral anticoagulants and antiplatelet drugs are commonly prescribed to lower the risk of cardiovascular diseases, such as venous and arterial thrombosis, which represent the leading causes of mortality worldwide. A significant percentage of patients taking antithrombotics will nevertheless experience bleeding or recurrent ischemic events, and this represents a major public health issue. Cardiovascular medicine is now questioning the one-size-fits-all policy, and more personalized approaches are increasingly being considered. However, the available tools are currently limited and they are only moderately able to predict clinical events or have a significant impact on clinical outcomes. Predicting concentrations of antithrombotics in blood could be an effective means of personalization as they have been associated with bleeding and recurrent ischemia. Target concentration interventions could take advantage of physiologically based pharmacokinetic (PBPK) and population-based pharmacokinetic (POPPK) models, which are increasingly used in clinical settings and have attracted the interest of governmental regulatory agencies, to propose dosages adapted to specific population characteristics. These models have the benefit of combining parameters from different sources, such as experimental in vitro data and patients' demographic, genetic, and physiological in vivo data, to characterize the dose-concentration relationships of compounds of interest. As such, they can be used to predict individual drug exposure. In the near future, these models could therefore be a valuable means of predicting personalized antithrombotic blood concentrations and, hopefully, of preventing clinical non-response or bleeding in a given patient. Existing approaches for personalization of antithrombotic prescriptions will be reviewed using practical examples for P2Y12 inhibitors and direct oral anticoagulants. The review will additionally focus on the existing PBPK and POPPK models for these two categories of drugs. Lastly, we address potential scenarios for their implementation in clinics, along with the main limitations and challenges.
Collapse
Affiliation(s)
- Jean Terrier
- Division of General Internal Medicine, Geneva University Hospitals, Geneva, Switzerland.,Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Youssef Daali
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland.,Clinical Pharmacology and Toxicology Service, Anesthesiology, Pharmacology and Intensive Care Department, Geneva University Hospitals, Geneva, Switzerland
| | - Pierre Fontana
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Division of Angiology and Haemostasis, Geneva University Hospitals, Geneva, Switzerland
| | - Chantal Csajka
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Jean-Luc Reny
- Division of General Internal Medicine, Geneva University Hospitals, Geneva, Switzerland. .,Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland. .,Division of Internal Medicine and Rehabilitation, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 4, 1205, Geneva, Switzerland.
| |
Collapse
|
39
|
Cho H, Kang J, Kim HS, Park KW. Ethnic Differences in Oral Antithrombotic Therapy. Korean Circ J 2020; 50:645-657. [PMID: 32725974 PMCID: PMC7390713 DOI: 10.4070/kcj.2020.0098] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 03/22/2020] [Indexed: 02/04/2023] Open
Abstract
Oral antithrombotic therapy (antiplatelet therapy and anticoagulation therapy) is a key element of pharmacotherapy in patients with cardiovascular (CV) disease. Several reports of ethnic differences have suggested that there may be difference therapeutic requirements and response to therapy for antithrombotic therapy. In particular for East Asians, there seems to be a lower incidence of ischemic outcomes and a higher incidence of bleeding outcomes compared to Westerners. The purpose of this review is to describe the ethnicity-related differences in antithrombotic therapy for CV disease and to discuss the need to establish a more effective and targeted antithrombotic treatment strategy in East Asians.
Collapse
Affiliation(s)
- Haechan Cho
- Department of Internal Medicine, Cardiovascular Center, Seoul National University Hospital, Seoul, Korea
| | - Jeehoon Kang
- Department of Internal Medicine, Cardiovascular Center, Seoul National University Hospital, Seoul, Korea
| | - Hyo Soo Kim
- Department of Internal Medicine, Cardiovascular Center, Seoul National University Hospital, Seoul, Korea
| | - Kyung Woo Park
- Department of Internal Medicine, Cardiovascular Center, Seoul National University Hospital, Seoul, Korea.
| |
Collapse
|
40
|
microRNAs as promising biomarkers of platelet activity in antiplatelet therapy monitoring. Int J Mol Sci 2020; 21:ijms21103477. [PMID: 32423125 PMCID: PMC7278969 DOI: 10.3390/ijms21103477] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 12/13/2022] Open
Abstract
Given the high morbidity and mortality of cardiovascular diseases (CVDs), novel biomarkers for platelet reactivity are urgently needed. Ischemic events in CVDs are causally linked to platelets, small anucleate cells important for hemostasis. The major side-effect of antiplatelet therapy are life-threatening bleeding events. Current platelet function tests are not sufficient in guiding treatment decisions. Platelets host a broad spectrum of microRNAs (miRNAs) and are a major source of cell-free miRNAs in the blood stream. Platelet-related miRNAs have been suggested as biomarkers of platelet activation and assessment of antiplatelet therapy responsiveness. Platelets release miRNAs upon activation, possibly leading to alterations of plasma miRNA levels in conjunction with CVD or inadequate platelet inhibition. Unlike current platelet function tests, which measure platelet activation ex vivo, signatures of platelet-related miRNAs potentially enable the assessment of in vivo platelet reactivity. Evidence suggests that some miRNAs are responsive to platelet inhibition, making them promising biomarker candidates. In this review, we explain the secretion of miRNAs upon platelet activation and discuss the potential use of platelet-related miRNAs as biomarkers for CVD and antiplatelet therapy monitoring, but also highlight remaining gaps in our knowledge and uncertainties regarding clinical utility. We also elaborate on technical issues and limitations concerning plasma miRNA quantification.
Collapse
|
41
|
Wu HY, Zhang C, Zhao X, Qian JY, Wang QB, Ge JB. Residual platelet reactivity is preferred over platelet inhibition rate in monitoring antiplatelet efficacy: insights using thrombelastography. Acta Pharmacol Sin 2020; 41:192-197. [PMID: 31515526 PMCID: PMC7468573 DOI: 10.1038/s41401-019-0278-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/23/2019] [Indexed: 12/27/2022]
Abstract
Although thrombelastography (TEG) has been widely implemented in the clinical setting of endovascular intervention, consensus on the optimal parameter for defining high ischemic risk patients is lacking due to the limited data about the relationship between various TEG parameters and clinical outcomes. In this article, we report a post hoc analysis of a prospective, single-center cohort study, including 447 patients with acute coronary syndrome (ACS). Arachidonic acid (AA)- or adenosine diphosphate (ADP)-induced platelet-fibrin clot strength (MAAA or MAADP) was indicative of the net residual platelet reactivity after the treatment with aspirin or clopidogrel, respectively. AA% or ADP% was indices of the relative platelet inhibition rate on AA or ADP pathway. We found that each parameter alone was predictive of the risk of 6-month ischemic event, even after adjusting for confounding factors. However, the association between AA% and clinical outcome disappeared when further adjusted for MAAA. Likewise, inclusion of MAADP changed the significant relation between ADP% and clinical outcome. MAADP > 47.0 mm and MAAA > 15.1 mm were identified as the optimal cutoffs by receiver operating characteristic analysis. High MAAA (HR = 3.963; 95% CI: 1.152-13.632; P = 0.029) and high MAADP (HR = 5.185; 95% CI: 2.228-12.062; P < 0.001) were independent predictors when both were included in multivariable Cox regression hazards model. Interestingly, an even higher risk was found for the coexisting high MAAA and high MAADP (HR = 7.870; 95% CI: 3.462-17.899; P < 0.001). We conclude that when performing TEG to predict clinical efficacy, residual platelet reactivity has superiority over platelet inhibition rate as a measure of thrombotic risk in patients treated with aspirin and clopidogrel after ACS.
Collapse
Affiliation(s)
- Hong-Yi Wu
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chi Zhang
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xin Zhao
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ju-Ying Qian
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Qi-Bing Wang
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jun-Bo Ge
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
42
|
Abstract
Abstract
Collapse
Affiliation(s)
- Jean-Sébastien Hulot
- Centre d’Investigations Cliniques CIC1418, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
- Université de Paris, Paris Cardiovascular Research Center PARCC, INSERM, Paris, France
| | - Gilles Montalescot
- Sorbonne Université, ACTION Study Group, INSERM UMRS_1166, Institut de cardiologie (AP-HP), Paris, France
| |
Collapse
|
43
|
Wiley CD, Liu S, Limbad C, Zawadzka AM, Beck J, Demaria M, Artwood R, Alimirah F, Lopez-Dominguez JA, Kuehnemann C, Danielson SR, Basisty N, Kasler HG, Oron TR, Desprez PY, Mooney SD, Gibson BW, Schilling B, Campisi J, Kapahi P. SILAC Analysis Reveals Increased Secretion of Hemostasis-Related Factors by Senescent Cells. Cell Rep 2019; 28:3329-3337.e5. [PMID: 31553904 PMCID: PMC6907691 DOI: 10.1016/j.celrep.2019.08.049] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/26/2019] [Accepted: 08/14/2019] [Indexed: 01/07/2023] Open
Abstract
Cellular senescence irreversibly arrests cell proliferation, accompanied by a multi-component senescence-associated secretory phenotype (SASP) that participates in several age-related diseases. Using stable isotope labeling with amino acids (SILACs) and cultured cells, we identify 343 SASP proteins that senescent human fibroblasts secrete at 2-fold or higher levels compared with quiescent cell counterparts. Bioinformatic analysis reveals that 44 of these proteins participate in hemostasis, a process not previously linked with cellular senescence. We validated the expression of some of these SASP factors in cultured cells and in vivo. Mice treated with the chemotherapeutic agent doxorubicin, which induces widespread cellular senescence in vivo, show increased blood clotting. Conversely, selective removal of senescent cells using transgenic p16-3MR mice showed that clearing senescent cells attenuates the increased clotting caused by doxorubicin. Our study provides an in-depth, unbiased analysis of the SASP and unveils a function for cellular senescence in hemostasis.
Collapse
Affiliation(s)
| | - Su Liu
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | | | - Jennifer Beck
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Marco Demaria
- European Institute for the Biology of Aging, University of Groningen, Groningen, the Netherlands
| | - Robert Artwood
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | | | | | | | - Natan Basisty
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | | | - Pierre-Yves Desprez
- Buck Institute for Research on Aging, Novato, CA 94945, USA; California Pacific Medical Center, Research Institute, San Francisco, CA 94107, USA
| | - Sean D Mooney
- Department of Biomedical Informatics & Medical Education, University of Washington, Seattle, WA 98195, USA
| | - Bradford W Gibson
- Discovery Attribute Sciences, Amgen Inc., South San Francisco, CA 94080, USA
| | | | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA 94945, USA; Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| | - Pankaj Kapahi
- Buck Institute for Research on Aging, Novato, CA 94945, USA.
| |
Collapse
|
44
|
Pretorius E. Platelets as Potent Signaling Entities in Type 2 Diabetes Mellitus. Trends Endocrinol Metab 2019; 30:532-545. [PMID: 31196615 DOI: 10.1016/j.tem.2019.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 12/19/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a multifactorial disease with a dysregulated circulating inflammatory molecule tendency. T2DM is closely associated with systemic inflammation, endothelial dysfunction, cardiovascular risk, and increased clotting susceptibility. Platelets have fundamental roles in the development and propagation of inflammation and cardiovascular risk. They signal through membrane receptors, resulting in (hyper)activation and release of inflammatory molecules from platelet compartments. This review highlights how circulating inflammatory molecules, acting as platelet receptor ligands, interact with platelets, causing platelets to be potent drivers of systemic inflammation. We conclude by suggesting that focused platelet research in T2DM is an important avenue to pursue to identify novel therapeutic targets, and that platelets could be used as cellular activity sensors themselves.
Collapse
Affiliation(s)
- Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, 7602, South Africa.
| |
Collapse
|
45
|
Navarese EP, Wernly B, Lichtenauer M, Petrescu AM, Kołodziejczak M, Lauten A, Frediani L, Veulemanns V, Wanha W, Wojakowski W, Lesiak M, Ferrante G, Zeus T, Tantry U, Bliden K, Buffon A, Contegiacomo G, Jung C, Kubica J, Pestrichella V, Gurbel PA. Dual vs single antiplatelet therapy in patients with lower extremity peripheral artery disease – A meta-analysis. Int J Cardiol 2018; 269:292-297. [DOI: 10.1016/j.ijcard.2018.07.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/24/2018] [Accepted: 07/02/2018] [Indexed: 01/22/2023]
|
46
|
Dipyrone (metamizole) markedly interferes with platelet inhibition by aspirin in patients with acute and chronic pain: A case-control study. Eur J Anaesthesiol 2018; 34:288-296. [PMID: 28030443 DOI: 10.1097/eja.0000000000000581] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Nonopioid analgesic drugs may interfere with platelet inhibition by aspirin. Recent in vitro and clinical studies in patients with cardiovascular disease have suggested that this pharmacodynamic interaction may also occur with dipyrone, a nonopioid analgesic popular in Europe, Asia and South America. OBJECTIVE Dipyrone is used extensively in acute and chronic pain. This study was undertaken to provide clinical data, so far missing, on its interactions in this group of patients. DESIGN A case-control study. SETTING Primary care in one European university hospital centre. PATIENTS In total, 27 patients with stable cardiovascular, cerebrovascular or peripheral arterial disease and acute or chronic pain were identified and given dipyrone for at least 5 days in combination with low-dose aspirin. In total, 10 comparable patients on low-dose aspirin alone served as controls. MAIN OUTCOME MEASURES Platelet-rich plasma was prepared to determine arachidonic acid-induced aggregation (aggregometry) and thromboxane formation (immunoassay). Platelet sensitivity to aspirin was examined in vitro. The presence of dipyrone (metabolites) in plasma was confirmed by HPLC. Additional in vitro measurements examined the aspirin/dipyrone interaction in healthy donors. RESULTS Inhibition of aggregation was observed in only six of 27 patients receiving aspirin with dipyrone, with absence of complete inhibition by antiplatelet therapy showing in 78% of patients. In contrast, aggregation was completely inhibited in nine of 10 control patients (P < 0.001). Platelet thromboxane synthesis was higher in patients receiving dipyrone + aspirin compared with controls (387 ± 89 vs. 7 ± 1 ng ml, P < 0.001). Aspirin added in vitro failed to inhibit aggregation and thromboxane synthesis in platelet-rich plasma from dipyrone-treated patients. In vitro measurements with blood from healthy individuals confirmed that dipyrone dramatically reduces inhibition of platelet thromboxane synthesis by aspirin. CONCLUSIONS Dipyrone given for 5 days or longer blunts platelet inhibition by low-dose aspirin in the majority of recipients. TRIAL REGISTRATION German Clinical Trials Register: DRKS ID DRKS00000204. Universal Trial Number (UTN): U1111-1113-3946.
Collapse
|
47
|
Zhu W, Buffa JA, Wang Z, Warrier M, Schugar R, Shih DM, Gupta N, Gregory JC, Org E, Fu X, Li L, DiDonato JA, Lusis AJ, Brown JM, Hazen SL. Flavin monooxygenase 3, the host hepatic enzyme in the metaorganismal trimethylamine N-oxide-generating pathway, modulates platelet responsiveness and thrombosis risk. J Thromb Haemost 2018; 16:1857-1872. [PMID: 29981269 PMCID: PMC6156942 DOI: 10.1111/jth.14234] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 06/26/2018] [Indexed: 12/21/2022]
Abstract
Essentials Microbe-dependent production of trimethylamine N-oxide (TMAO) contributes to thrombosis risk. The impact of host flavin monooxygenase 3 (FMO3) modulation on platelet function is unknown. Genetic manipulation of FMO3 in mice alters systemic TMAO levels and thrombosis potential. Genetic manipulation of FMO3 is associated with alteration of gut microbial community structure. SUMMARY Background Gut microbes play a critical role in the production of trimethylamine N-oxide (TMAO), an atherogenic metabolite that impacts platelet responsiveness and thrombosis potential. Involving both microbe and host enzymatic machinery, TMAO generation utilizes a metaorganismal pathway, beginning with ingestion of trimethylamine (TMA)-containing dietary nutrients such as choline, phosphatidylcholine and carnitine, which are abundant in a Western diet. Gut microbial TMA lyases use these nutrients as substrates to produce TMA, which upon delivery to the liver via the portal circulation, is converted into TMAO by host hepatic flavin monooxygenases (FMOs). Gut microbial production of TMA is rate limiting in the metaorganismal TMAO pathway because hepatic FMO activity is typically in excess. Objectives FMO3 is the major FMO responsible for host generation of TMAO; however, a role for FMO3 in altering platelet responsiveness and thrombosis potential in vivo has not yet been explored. Methods The impact of FMO3 suppression (antisense oligonucleotide-targeting) and overexpression (as transgene) on plasma TMAO levels, platelet responsiveness and thrombosis potential was examined using a murine FeCl3 -induced carotid artery injury model. Cecal microbial composition was examined using 16S analyses. Results Modulation of FMO3 directly impacts systemic TMAO levels, platelet responsiveness and rate of thrombus formation in vivo. Microbial composition analyses reveal taxa whose proportions are associated with both plasma TMAO levels and in vivo thrombosis potential. Conclusions The present studies demonstrate that host hepatic FMO3, the terminal step in the metaorganismal TMAO pathway, participates in diet-dependent and gut microbiota-dependent changes in both platelet responsiveness and thrombosis potential in vivo.
Collapse
Affiliation(s)
- W. Zhu
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - J. A. Buffa
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Z. Wang
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - M. Warrier
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - R. Schugar
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - D. M. Shih
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - N. Gupta
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - J. C. Gregory
- Departments of Human Genetics and Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - E. Org
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - X. Fu
- Departments of Human Genetics and Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - L. Li
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - J. A. DiDonato
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - A. J. Lusis
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - J. M. Brown
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Departments of Human Genetics and Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - S. L. Hazen
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Heart and Vascular Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| |
Collapse
|
48
|
Polo E, Nitka TA, Neubert E, Erpenbeck L, Vuković L, Kruss S. Control of Integrin Affinity by Confining RGD Peptides on Fluorescent Carbon Nanotubes. ACS APPLIED MATERIALS & INTERFACES 2018; 10:17693-17703. [PMID: 29708725 DOI: 10.1021/acsami.8b04373] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Integrins are transmembrane receptors that mediate cell-adhesion, signaling cascades and platelet-mediated blood clotting. Most integrins bind to the common short peptide Arg-Gly-Asp (RGD). The conformational freedom of the RGD motif determines how strong and to which integrins it binds. Here, we present a novel approach to tune binding constants by confining RGD peptide motifs via noncovalent adsorption of single-stranded DNA (ssDNA) anchors onto single-walled carbon nanotubes (SWCNTs). Semiconducting SWCNTs display fluorescence in the near-infrared (nIR) region and are versatile fluorescent building blocks for imaging and biosensing. The basic idea of this approach is that the DNA adsorbed on the SWCNT surface determines the conformational freedom of the RGD motif and affects binding affinities. The RGD motif was conjugated to different ssDNA sequences in both linear ssDNA-RGD and bridged ssDNA-RGD-ssDNA geometries. Molecular dynamics (MD) simulations show that the RGD motif in all the synthesized systems is mostly exposed to solvent and thus available for binding, but its flexibility depends on the exact geometry. The affinity for the human platelet integrin αIIbβ3 could be modulated up to 15-fold by changing the ssDNA sequence. IC50 values varied from 309 nM for (C)20-RGD/SWCNT hybrids to 29 nM for (GT)15-RGD/SWCNT hybrids. When immobilized onto surface adhesion of epithelial cells increased 6-fold for (GT)15-RGD/SWCNTs. (GT)15-RGD/SWCNTs also increased the number of adhering human platelets by a factor of 4.8. Additionally, αIIbβ3 integrins on human platelets were labeled in the nIR by incubating them with these ssDNA-peptide/SWCNT hybrids. In summary, we show that ssDNA-peptide hybrid structures noncovalently adsorb onto SWCNTs and serve as recognition units for cell surface receptors such as integrins. The DNA sequence affects the overall RGD affinity, which is a versatile and straightforward approach to tune binding affinities. In combination with the nIR fluorescence properties of SWCNTs, these new hybrid materials promise many applications in integrin targeting and bioimaging.
Collapse
Affiliation(s)
- Elena Polo
- Institute of Physical Chemistry , Göttingen University , Tammanstrasse 6 , 37077 Göttingen , Germany
| | - Tara A. Nitka
- Department of Chemistry and Biochemistry , The University of Texas at El Paso , El Paso , Texas 79968 , United States
| | - Elsa Neubert
- Institute of Physical Chemistry , Göttingen University , Tammanstrasse 6 , 37077 Göttingen , Germany
- University Medical Center, Department of Dermatology , Göttingen University , 37077 Göttingen , Germany
| | - Luise Erpenbeck
- University Medical Center, Department of Dermatology , Göttingen University , 37077 Göttingen , Germany
| | - Lela Vuković
- Department of Chemistry and Biochemistry , The University of Texas at El Paso , El Paso , Texas 79968 , United States
| | - Sebastian Kruss
- Institute of Physical Chemistry , Göttingen University , Tammanstrasse 6 , 37077 Göttingen , Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB) , 37073 Göttingen , Germany
| |
Collapse
|
49
|
Zhang HH, Yu WY, Li L, Wu F, Chen Q, Yang Y, Yu CH. Protective effects of diketopiperazines from Moslae Herba against influenza A virus-induced pulmonary inflammation via inhibition of viral replication and platelets aggregation. JOURNAL OF ETHNOPHARMACOLOGY 2018; 215:156-166. [PMID: 29309861 DOI: 10.1016/j.jep.2018.01.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 12/27/2017] [Accepted: 01/04/2018] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Moslae Herba (MH) is broadly used as an antiviral, antipyretic and anticoagulant drug which effectively treats respiratory diseases including cough, asthma, throat, cold and flu. AIM OF THIS STUDY The excessive inflammation of the lungs is the hallmark of severe influenza A virus (IAV) infection, while platelet aggregation and its subsequent microvascular thrombosis can exacerbate IAV-induced lung injury. Thus, inhibition of platelet aggregation can be a potential target for IAV treatment. Previous studies focus on the flavonoids from MH and their anti-inflammatory activities, but the anticoagulant compounds and potential molecular mechanism of MH remains unclear. This study was to isolate and characterize diketopiperazines (DKPs) from MH and to explore the underlying anticoagulant mechanism on IAV infection models. MATERIALS AND METHODS EtOAc sub-extract separated from MH ethanolic extract was subjected to fractionation through column chromatography. The chemical structures of pure compounds were characterized by the spectral analysis. Antiviral activities of DKPs were assayed in IAV-infected Madin-Darby canine kidney (MDCK) cells and mice. Anticoagulant effects of DKPs were investigated on adenosine 5'-diphosphate (ADP)-induced acute pulmonary embolism and IAV-induced lung injury in vivo, as well as the inhibition on platelet activating factor (PAF), arachidonic acid (AA) and ADP-induced platelet aggregation in vitro. The serum levels of thromboxane B2 (TXB2) and 6-keto-PGF1α were detected by ELISA. The expressions of key proteins in CD41-mediated PI3K/AKT pathways were determined by western blotting analysis. RESULTS Six DKPs were, for the first time, isolated from MH and identified as cyclo(Tyr-Leu) (1), cyclo(Phe-Phe) (2), cyclo(Phe-Tyr) (3), cyclo(Ala-Ile) (4), cyclo(Ala-Leu) (5) and Bz-Phe-Phe-OMe (6). Among these DKPs, cyclo(Ala-Ile) and Bz-Phe-Phe-OMe possessed low cytotoxicities and significant inhibition against cytopathic effects induced by IAV (H1N1 and H3N2) replication in MDCK cells. Furthermore, cyclo(Ala-Ile) and Bz-Phe-Phe-OMe significantly alleviated IAV-induced platelet activation and lung inflammation in mice. They could reduce the expression of CD41 and the phosphorylation of PI3K and AKT in PLTs of IAV-infected mice. CONCLUSION These results suggested that cyclo(Ala-Ile) and Bz-Phe-Phe-OMe isolated from MH have antiviral and anticoagulant effects against IAV-induced PLT aggregation and lung inflammation via regulating CD41/PI3K/AKT pathway, and could be used as the potential agents for IAV treatment.
Collapse
Affiliation(s)
- Huan-Huan Zhang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Wen-Ying Yu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Lan Li
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Fang Wu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China; First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qin Chen
- Department of Clinical Laboratory Medicine, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou 310009, China
| | - Yang Yang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Chen-Huan Yu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China.
| |
Collapse
|
50
|
Bergmeijer TO, Reny JL, Pakyz RE, Gong L, Lewis JP, Kim EY, Aradi D, Fernandez-Cadenas I, Horenstein RB, Lee MTM, Whaley RM, Montaner J, Gensini GF, Cleator JH, Chang K, Holmvang L, Hochholzer W, Roden DM, Winter S, Altman RB, Alexopoulos D, Kim HS, Déry JP, Gawaz M, Bliden K, Valgimigli M, Marcucci R, Campo G, Schaeffeler E, Dridi NP, Wen MS, Shin JG, Simon T, Fontana P, Giusti B, Geisler T, Kubo M, Trenk D, Siller-Matula JM, Ten Berg JM, Gurbel PA, Hulot JS, Mitchell BD, Schwab M, Ritchie MD, Klein TE, Shuldiner AR. Genome-wide and candidate gene approaches of clopidogrel efficacy using pharmacodynamic and clinical end points-Rationale and design of the International Clopidogrel Pharmacogenomics Consortium (ICPC). Am Heart J 2018; 198:152-159. [PMID: 29653637 PMCID: PMC5903579 DOI: 10.1016/j.ahj.2017.12.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 12/10/2017] [Indexed: 02/07/2023]
Abstract
RATIONALE The P2Y12 receptor inhibitor clopidogrel is widely used in patients with acute coronary syndrome, percutaneous coronary intervention, or ischemic stroke. Platelet inhibition by clopidogrel shows wide interpatient variability, and high on-treatment platelet reactivity is a risk factor for atherothrombotic events, particularly in high-risk populations. CYP2C19 polymorphism plays an important role in this variability, but heritability estimates suggest that additional genetic variants remain unidentified. The aim of the International Clopidogrel Pharmacogenomics Consortium (ICPC) is to identify genetic determinants of clopidogrel pharmacodynamics and clinical response. STUDY DESIGN Based on the data published on www.clinicaltrials.gov, clopidogrel intervention studies containing genetic and platelet function data were identified for participation. Lead investigators were invited to share DNA samples, platelet function test results, patient characteristics, and cardiovascular outcomes to perform candidate gene and genome-wide studies. RESULTS In total, 17 study sites from 13 countries participate in the ICPC, contributing individual patient data from 8,829 patients. Available adenosine diphosphate-stimulated platelet function tests included vasodilator-stimulated phosphoprotein assay, light transmittance aggregometry, and the VerifyNow P2Y12 assay. A proof-of-principle analysis based on genotype data provided by each group showed a strong and consistent association between CYP2C19*2 and platelet reactivity (P value=5.1 × 10-40). CONCLUSION The ICPC aims to identify new loci influencing clopidogrel efficacy by using state-of-the-art genetic approaches in a large cohort of clopidogrel-treated patients to better understand the genetic basis of on-treatment response variability.
Collapse
Affiliation(s)
- Thomas O Bergmeijer
- St Antonius Center for Platelet Function Research, Department of Cardiology, St Antonius Hospital Nieuwegein, the Netherlands
| | - Jean-Luc Reny
- Internal Medicine, Béziers Hospital, France, Geneva Platelet Group, University of Geneva School of Medicine, Department of Internal Medicine, Rehabilitation and Geriatrics, University Hospitals of Geneva, Geneva, Switzerland
| | - Ruth E Pakyz
- Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Li Gong
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Joshua P Lewis
- Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eun-Young Kim
- Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Busan, South Korea
| | - Daniel Aradi
- Heart Center Balatonfüred and Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Israel Fernandez-Cadenas
- Stroke Pharmacogenomics and Genetics, Fundació Docència i Recerca Mútua Terrassa, Neurovascular Research Laboratory, Valle d'Hebron Hebron Institute of Research, Barcelona, Spain
| | - Richard B Horenstein
- Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Ryan M Whaley
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Barcelona, Spain
| | - Gian Franco Gensini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - John H Cleator
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Kiyuk Chang
- Cardiovascular Center and Cardiology Division, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Lene Holmvang
- Department of Cardiology and Cardiac Catheterization Laboratory, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Willibald Hochholzer
- University Heart Center Freiburg, Bad Krozingen, Department of Cardiology and Angiology II, Bad Krozingen, Germany
| | - Dan M Roden
- Departments of Medicine, Pharmacology, and Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Stefan Winter
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart and University of Tübingen, Tübingen, Germany
| | - Russ B Altman
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA; Departments of Bioengineering and Genetics, Stanford University, Stanford, CA, USA; Department of Medicine, Stanford University, Stanford, CA, USA
| | | | - Ho-Sook Kim
- Department of Pharmacology and Pharmacogenomics Research Center, College of Medicine, Inje University, Busan, South Korea
| | | | - Meinrad Gawaz
- Department of Cardiology and Cardiovascular Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Kevin Bliden
- Inova Center for Thrombosis Research and Drug Development. Inova Heart and Vascular Institute, Falls Church, VA, USA
| | - Marco Valgimigli
- Department of Cardiology, Swiss Cardiovascular Center Bern, Bern University Hospital, Bern, Switzerland
| | - Rossella Marcucci
- Department of Experimental and Clinical Medicine, University of Florence, Atherothrombotic Diseases Center, Careggi Hospital, Florence, Italy
| | - Gianluca Campo
- Cardiology Unit, Azienda Ospedaliera Universitria di Ferrara, Cona (FE) and Maria Cecilia Hospital, GVM Care and Research, Cotignola, (RA), Italy
| | - Elke Schaeffeler
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart and University of Tübingen, Tübingen, Germany
| | - Nadia P Dridi
- Department of Cardiology and Cardiac Catheterization Laboratory, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ming-Shien Wen
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou and School of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Jae Gook Shin
- Department of Pharmacology and Pharmacogenomics Research Center, College of Medicine, Inje University, Busan, South Korea
| | | | - Pierre Fontana
- Geneva Platelet Group, University of Geneva School of Medicine, Division of Angiology and Haemostasis, University Hospitals of Geneva, Geneva, Switzerland
| | - Betti Giusti
- Department of Experimental and Clinical Medicine, University of Florence, Atherothrombotic Diseases Center, Careggi Hospital, Florence, Italy
| | - Tobias Geisler
- Department of Cardiology and Cardiovascular Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Michiaki Kubo
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Dietmar Trenk
- Department of Cardiology and Cardiovascular Medicine, University Hospital Tübingen, Tübingen, Germany
| | | | - Jurriën M Ten Berg
- St Antonius Center for Platelet Function Research, Department of Cardiology, St Antonius Hospital Nieuwegein, the Netherlands
| | - Paul A Gurbel
- Inova Center for Thrombosis Research and Drug Development. Inova Heart and Vascular Institute, Falls Church, VA, USA
| | - Jean-Sebastien Hulot
- Sorbonne Universités, UPMC Univ Paris 06, Institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, F-75013 Paris, France
| | - Braxton D Mitchell
- Department of Medicine, University of Maryland, Baltimore, MD, USA; Geriatric Research, Education and Clinical Center, Veterans Affairs Medical Center, Baltimore, MD
| | - Matthias Schwab
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart and University of Tübingen, Tübingen, Germany; Department of Clinical Pharmacology, University Hospital, Tübingen, Germany
| | - Marylyn DeRiggi Ritchie
- Department of Biomedical and Translational Informatics, Geisinger Health System, Danville, PA, USA
| | - Teri E Klein
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA; Department of Medicine, Stanford University, Stanford, CA, USA
| | - Alan R Shuldiner
- Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|