1
|
Arauna D, Navarrete S, Albala C, Wehinger S, Pizarro-Mena R, Palomo I, Fuentes E. Understanding the Role of Oxidative Stress in Platelet Alterations and Thrombosis Risk among Frail Older Adults. Biomedicines 2024; 12:2004. [PMID: 39335518 PMCID: PMC11429027 DOI: 10.3390/biomedicines12092004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
Frailty and cardiovascular diseases are increasingly prevalent in aging populations, sharing common pathological mechanisms, such as oxidative stress. The evidence shows that these factors predispose frail individuals to cardiovascular diseases but also increase the risk of thrombosis. Considering this background, this review aims to explore advances regarding the relationship between oxidative stress, platelet alterations, and cardiovascular diseases in frailty, examining the role of reactive oxygen species overproduction in platelet activation and thrombosis. The current evidence shows a bidirectional relationship between frailty and cardiovascular diseases, emphasizing how frailty not only predisposes individuals to cardiovascular diseases but also accelerates disease progression through oxidative damage and increased platelet function. Thus, oxidative stress is the central axis in the increase in platelet activation and secretion and the inadequate response to acetylsalicylic acid observed in frail people by mitochondrial mechanisms. Also, key biomarkers of oxidative stress, such as isoprostanes and derivate reactive oxygen metabolites, can be optimal predictors of cardiovascular risk and potential targets for therapeutic intervention. The potential of antioxidant therapies in mitigating oxidative stress and improving cardiovascular clinical outcomes such as platelet function is promising in frailty, although further research is necessary to establish the efficacy of these therapies. Understanding these mechanisms could prove essential in improving the health and quality of life of an aging population faced with the dual burden of frailty and cardiovascular diseases.
Collapse
Affiliation(s)
- Diego Arauna
- Thrombosis Research and Healthy Aging Center, Department of Clinical Biochemistry and Immunohematology, Interuniversity Center for Healthy Aging (CIES), Interuniversity Network of Healthy Aging in Latin America and Caribbean (RIES-LAC), Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile
| | - Simón Navarrete
- Thrombosis Research and Healthy Aging Center, Department of Clinical Biochemistry and Immunohematology, Interuniversity Center for Healthy Aging (CIES), Interuniversity Network of Healthy Aging in Latin America and Caribbean (RIES-LAC), Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile
| | - Cecilia Albala
- Unidad de Nutrición Pública, Instituto de Nutrición y Tecnología de los Alimentos, Interuniversity Center for Healthy Aging, Universidad de Chile, Santiago 7810000, Chile
| | - Sergio Wehinger
- Thrombosis Research and Healthy Aging Center, Department of Clinical Biochemistry and Immunohematology, Interuniversity Center for Healthy Aging (CIES), Interuniversity Network of Healthy Aging in Latin America and Caribbean (RIES-LAC), Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile
| | - Rafael Pizarro-Mena
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Sede Los Leones, Santiago 7500000, Chile
- Interuniversity Network of Healthy Aging in Latin America and Caribbean (RIES-LAC), Santiago 7810000, Chile
| | - Iván Palomo
- Thrombosis Research and Healthy Aging Center, Department of Clinical Biochemistry and Immunohematology, Interuniversity Center for Healthy Aging (CIES), Interuniversity Network of Healthy Aging in Latin America and Caribbean (RIES-LAC), Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile
| | - Eduardo Fuentes
- Thrombosis Research and Healthy Aging Center, Department of Clinical Biochemistry and Immunohematology, Interuniversity Center for Healthy Aging (CIES), Interuniversity Network of Healthy Aging in Latin America and Caribbean (RIES-LAC), Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile
| |
Collapse
|
2
|
Palomo I, Wehinger S, Andrés V, García‐García FJ, Fuentes E. RhoA/rho kinase pathway activation in age-associated endothelial cell dysfunction and thrombosis. J Cell Mol Med 2024; 28:e18153. [PMID: 38568071 PMCID: PMC10989549 DOI: 10.1111/jcmm.18153] [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: 09/27/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 04/05/2024] Open
Abstract
The small GTPase RhoA and the downstream Rho kinase (ROCK) regulate several cell functions and pathological processes in the vascular system that contribute to the age-dependent risk of cardiovascular disease, including endothelial dysfunction, excessive permeability, inflammation, impaired angiogenesis, abnormal vasoconstriction, decreased nitric oxide production and apoptosis. Frailty is a loss of physiological reserve and adaptive capacity with advanced age and is accompanied by a pro-inflammatory and pro-oxidative state that promotes vascular dysfunction and thrombosis. This review summarises the role of the RhoA/Rho kinase signalling pathway in endothelial dysfunction, the acquisition of the pro-thrombotic state and vascular ageing. We also discuss the possible role of RhoA/Rho kinase signalling as a promising therapeutic target for the prevention and treatment of age-related cardiovascular disease.
Collapse
Affiliation(s)
- Iván Palomo
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Medical Technology School, Thrombosis and Healthy Aging Research CenterUniversidad de TalcaTalcaChile
| | - Sergio Wehinger
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Medical Technology School, Thrombosis and Healthy Aging Research CenterUniversidad de TalcaTalcaChile
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV)MadridSpain
| | - Francisco J. García‐García
- Department of Geriatric MedicineHospital Universitario de Toledo, Instituto de Investigación de Castilla La Mancha (IDISCAM), CIBERFES (ISCIII)ToledoSpain
| | - Eduardo Fuentes
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Medical Technology School, Thrombosis and Healthy Aging Research CenterUniversidad de TalcaTalcaChile
| |
Collapse
|
3
|
Kullaya VI, Temba GS, Vadaq N, Njau J, Boahen CK, Nkambule BB, Thibord F, Chen MH, Pecht T, Lyamuya F, Kumar V, Netea MG, Mmbaga BT, van der Ven A, Johnson AD, de Mast Q. Genetic and nongenetic drivers of platelet reactivity in healthy Tanzanian individuals. J Thromb Haemost 2024; 22:805-817. [PMID: 38029856 DOI: 10.1016/j.jtha.2023.11.014] [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: 06/27/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Platelets play a key role in hemostasis, inflammation, and cardiovascular diseases. Platelet reactivity is highly variable between individuals. The drivers of this variability in populations from Sub-Saharan Africa remain largely unknown. OBJECTIVES We aimed to investigate the nongenetic and genetic determinants of platelet reactivity in healthy adults living in a rapidly urbanizing area in Northern Tanzania. METHODS Platelet activation and reactivity were measured by platelet P-selectin expression and the binding of fibrinogen in unstimulated blood and after ex vivo stimulation with adenosine diphosphate and PAR-1 and PAR-4 ligands. We then analyzed the associations of platelet parameters with host genetic and nongenetic factors, environmental factors, plasma inflammatory markers, and plasma metabolites. RESULTS Only a few associations were found between platelet reactivity parameters and plasma inflammatory markers and nongenetic host and environmental factors. In contrast, untargeted plasma metabolomics revealed a large number of associations with food-derived metabolites, including phytochemicals that were previously reported to inhibit platelet reactivity. Genome-wide single-nucleotide polymorphism genotyping identified 2 novel single-nucleotide polymorphisms (rs903650 and rs4789332) that were associated with platelet reactivity at the genome-wide level (P < 5 × 10-8) as well as a number of variants in the PAR4 gene (F2RL3) that were associated with PAR4-induced reactivity. CONCLUSION Our study uncovered factors that determine variation in platelet reactivity in a population in East Africa that is rapidly transitioning to an urban lifestyle, including the importance of genetic ancestry and the gradual abandoning of the traditional East African diet.
Collapse
Affiliation(s)
- Vesla I Kullaya
- Kilimanjaro Clinical Research Institute, Kilimanjaro Christian Medical Center, Moshi, Tanzania; Department of Medical Biochemistry and Molecular Biology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Godfrey S Temba
- Department of Medical Biochemistry and Molecular Biology, Kilimanjaro Christian Medical University College, Moshi, Tanzania; Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nadira Vadaq
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Judith Njau
- Kilimanjaro Clinical Research Institute, Kilimanjaro Christian Medical Center, Moshi, Tanzania
| | - Collins K Boahen
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Bongani B Nkambule
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Florian Thibord
- National Heart, Lung, and Blood Institute, Population Sciences Branch, Framingham, Massachusetts, USA
| | - Ming-Huei Chen
- National Heart, Lung, and Blood Institute, Population Sciences Branch, Framingham, Massachusetts, USA
| | - Tal Pecht
- Department for Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Furaha Lyamuya
- Kilimanjaro Clinical Research Institute, Kilimanjaro Christian Medical Center, Moshi, Tanzania
| | - Vinod Kumar
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department for Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Blandina T Mmbaga
- Kilimanjaro Clinical Research Institute, Kilimanjaro Christian Medical Center, Moshi, Tanzania; Department of Pediatrics, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Andre van der Ven
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Andrew D Johnson
- National Heart, Lung, and Blood Institute, Population Sciences Branch, Framingham, Massachusetts, USA
| | - Quirijn de Mast
- Department of Internal Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
4
|
Song Y, Lin Z, He J, Cui K, Song C, Zhang R, Liu Z, An T, Gao G, Gao Y, Dou K. Association of platelet-to-lymphocyte ratio levels with the risk of cardiac adverse events in people with type 2 diabetes undergoing percutaneous coronary intervention: A large-scale prospective cohort study. Diabetes Metab Syndr 2024; 18:102987. [PMID: 38518450 DOI: 10.1016/j.dsx.2024.102987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 03/24/2024]
Abstract
BACKGROUND The platelet-to-lymphocyte ratio (PLR), a promising inflammatory biomarker, contributes to the development of atherosclerosis and type 2 diabetes (T2D). Therefore, this study aimed to elucidate the importance of PLR in predicting adverse events in people undergoing percutaneous coronary intervention (PCI) with T2D. METHODS We consecutively enrolled 8831 people who underwent PCI and divided them into four groups according to PLR and glycemic metabolic status (PLR-Low/High without T2D, PLR-Low/High with T2D). The endpoints were major adverse cardiovascular and cerebrovascular events (MACCE) and stent thrombosis. A multivariate Cox regression analysis was performed to determine this association. RESULTS During the 2.4-year follow-up, 663 (7.5%) MACCE and 75 (0.85%) stent thromboses were recorded. The risk of MACCE (hazard ratio [HR]: 1.30, 95% confidence interval [CI]: 1.10-1.53, P = 0.002) and stent thrombosis (HR: 2.32, 95% CI: 1.38-3.90, P = 0.002) was significantly higher in people with high PLR levels than in those with low PLR. Among people with T2D, the PLR-High group showed a significantly higher risk of MACCE (HR: 1.59, 95% CI: 1.21-2.09, P = 0.001) and stent thrombosis (HR: 3.15, 95% CI: 1.32-7.52, P = 0.010). However, these associations were not significant in people without T2D. CONCLUSIONS PLR has been originally documented as a significant predictor of poor prognosis and a high incidence of stent thrombosis in people undergoing PCI, especially in those with T2D.
Collapse
Affiliation(s)
- Yanjun Song
- State Key Laboratory of Cardiovascular Disease, Beijing, China; Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Zhangyu Lin
- State Key Laboratory of Cardiovascular Disease, Beijing, China; Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Jining He
- State Key Laboratory of Cardiovascular Disease, Beijing, China; Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Kongyong Cui
- State Key Laboratory of Cardiovascular Disease, Beijing, China; Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Chenxi Song
- State Key Laboratory of Cardiovascular Disease, Beijing, China; Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Rui Zhang
- State Key Laboratory of Cardiovascular Disease, Beijing, China; Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Zechen Liu
- State Key Laboratory of Cardiovascular Disease, Beijing, China; Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Tao An
- State Key Laboratory of Cardiovascular Disease, Beijing, China; Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Guofeng Gao
- State Key Laboratory of Cardiovascular Disease, Beijing, China; Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Ying Gao
- State Key Laboratory of Cardiovascular Disease, Beijing, China; Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, China.
| | - Kefei Dou
- State Key Laboratory of Cardiovascular Disease, Beijing, China; Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing, 100037, China.
| |
Collapse
|
5
|
Luo T, Zhang Z, Xu J, Liu H, Cai L, Huang G, Wang C, Chen Y, Xia L, Ding X, Wang J, Li X. Atherosclerosis treatment with nanoagent: potential targets, stimulus signals and drug delivery mechanisms. Front Bioeng Biotechnol 2023; 11:1205751. [PMID: 37404681 PMCID: PMC10315585 DOI: 10.3389/fbioe.2023.1205751] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/31/2023] [Indexed: 07/06/2023] Open
Abstract
Cardiovascular disease (CVDs) is the first killer of human health, and it caused up at least 31% of global deaths. Atherosclerosis is one of the main reasons caused CVDs. Oral drug therapy with statins and other lipid-regulating drugs is the conventional treatment strategies for atherosclerosis. However, conventional therapeutic strategies are constrained by low drug utilization and non-target organ injury problems. Micro-nano materials, including particles, liposomes, micelles and bubbles, have been developed as the revolutionized tools for CVDs detection and drug delivery, specifically atherosclerotic targeting treatment. Furthermore, the micro-nano materials also could be designed to intelligently and responsive targeting drug delivering, and then become a promising tool to achieve atherosclerosis precision treatment. This work reviewed the advances in atherosclerosis nanotherapy, including the materials carriers, target sites, responsive model and treatment results. These nanoagents precisely delivery the therapeutic agents to the target atherosclerosis sites, and intelligent and precise release of drugs, which could minimize the potential adverse effects and be more effective in atherosclerosis lesion.
Collapse
Affiliation(s)
- Ting Luo
- Department of Cardiology, The Third People’s Hospital of Chengdu Affiliated to Southwest Jiaotong University, Key Laboratory of Advanced Technologies of Materials Ministry of Education, Southwest Jiaotong University, Chengdu, Sichuan, China
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Zhen Zhang
- Department of Cardiology, The Third People’s Hospital of Chengdu Affiliated to Southwest Jiaotong University, Key Laboratory of Advanced Technologies of Materials Ministry of Education, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Junbo Xu
- Department of Cardiology, The Third People’s Hospital of Chengdu Affiliated to Southwest Jiaotong University, Key Laboratory of Advanced Technologies of Materials Ministry of Education, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Hanxiong Liu
- Department of Cardiology, The Third People’s Hospital of Chengdu Affiliated to Southwest Jiaotong University, Key Laboratory of Advanced Technologies of Materials Ministry of Education, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Lin Cai
- Department of Cardiology, The Third People’s Hospital of Chengdu Affiliated to Southwest Jiaotong University, Key Laboratory of Advanced Technologies of Materials Ministry of Education, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Gang Huang
- Department of Cardiology, The Third People’s Hospital of Chengdu Affiliated to Southwest Jiaotong University, Key Laboratory of Advanced Technologies of Materials Ministry of Education, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Chunbin Wang
- Department of Cardiology, The Third People’s Hospital of Chengdu Affiliated to Southwest Jiaotong University, Key Laboratory of Advanced Technologies of Materials Ministry of Education, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Yingzhong Chen
- Department of Cardiology, The Third People’s Hospital of Chengdu Affiliated to Southwest Jiaotong University, Key Laboratory of Advanced Technologies of Materials Ministry of Education, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Long Xia
- Department of Cardiology, The Third People’s Hospital of Chengdu Affiliated to Southwest Jiaotong University, Key Laboratory of Advanced Technologies of Materials Ministry of Education, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Xunshi Ding
- Department of Cardiology, The Third People’s Hospital of Chengdu Affiliated to Southwest Jiaotong University, Key Laboratory of Advanced Technologies of Materials Ministry of Education, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Jin Wang
- Department of Cardiology, The Third People’s Hospital of Chengdu Affiliated to Southwest Jiaotong University, Key Laboratory of Advanced Technologies of Materials Ministry of Education, Southwest Jiaotong University, Chengdu, Sichuan, China
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Xin Li
- Department of Cardiology, The Third People’s Hospital of Chengdu Affiliated to Southwest Jiaotong University, Key Laboratory of Advanced Technologies of Materials Ministry of Education, Southwest Jiaotong University, Chengdu, Sichuan, China
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Plaza A, Rodríguez L, Concha-Meyer AA, Cabezas R, Zurob E, Merlet G, Palomo I, Fuentes E. Effects of Extraction Methods on Phenolic Content, Antioxidant and Antiplatelet Activities of Tomato Pomace Extracts. PLANTS (BASEL, SWITZERLAND) 2023; 12:1188. [PMID: 36904048 PMCID: PMC10005732 DOI: 10.3390/plants12051188] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/23/2023] [Accepted: 02/28/2023] [Indexed: 06/18/2023]
Abstract
Aqueous and ethanolic extracts of tomato pomace were examined with the aim of optimizing the extraction process of compounds with cardioprotective activity. Once the results of the ORAC response variables, total polyphenols, °Brix, and antiplatelet activity of the extracts were obtained, a multivariate statistical analysis was performed using the Statgraphics Centurion XIX software. This analysis showed that the most relevant positive effects in the inhibition of platelet aggregation were 83 ± 2% when using the agonist TRAP-6, when the working conditions were the type of tomato pomace conditioning (drum-drying process at 115 °C), phase ratio (1/8), type of solvent (ethanol 20%), and type of extraction (ultrasound-assisted solid-liquid extraction). The extracts with the best results were microencapsulated and characterized by HPLC. The presence of chlorogenic acid (0.729 mg/mg of dry sample) was found, a compound that has a potential cardioprotective effect documented in various studies, in addition to rutin (2.747 mg/mg of dry sample) and quercetin (0.255 mg/mg of dry sample). These results show that the extraction efficiency of compounds with cardioprotective activity depends largely on the polarity of the solvent, thus playing an important role in the antioxidant capacity of the extracts of tomato pomace.
Collapse
Affiliation(s)
- Andrea Plaza
- Centro de Estudios en Alimentos Procesados-CEAP, Conicyt, Programa Regional R19A10001, Gore Maule, Talca 3480094, Chile
| | - Lyanne Rodríguez
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Talca 3480094, Chile
| | - Anibal A. Concha-Meyer
- Centro de Estudios en Alimentos Procesados-CEAP, Conicyt, Programa Regional R19A10001, Gore Maule, Talca 3480094, Chile
- Instituto de Ciencia y Tecnología de los Alimentos, Facultad de Ciencias Agrarias y Alimentarias, Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia 5090000, Chile
| | - René Cabezas
- Departamento de Química Ambiental, Facultad de Ciencias, Universidad Católica de la Santísima Concepción, Concepción 4070129, Chile
| | - Elsie Zurob
- Laboratory of Membrane Separation Processes (LabProSeM), Department of Chemical Engineering, University of Santiago de Chile, Santiago 9170022, Chile
| | - Gastón Merlet
- Departamento de Agroindustrias, Facultad de Ingeniería Agrícola, Universidad de Concepción, Chillán 4070386, Chile
| | - Iván Palomo
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Talca 3480094, Chile
| | - Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Talca 3480094, Chile
| |
Collapse
|
7
|
A Comprehensive Literature Review on Cardioprotective Effects of Bioactive Compounds Present in Fruits of Aristotelia chilensis Stuntz (Maqui). Molecules 2022; 27:molecules27196147. [PMID: 36234679 PMCID: PMC9571323 DOI: 10.3390/molecules27196147] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
Some fruits and vegetables, rich in bioactive compounds such as polyphenols, flavonoids, and anthocyanins, may inhibit platelet activation pathways and therefore reduce the risk of suffering from CVD when consumed regularly. Aristotelia chilensis Stuntz (Maqui) is a shrub or tree native to Chile with outstanding antioxidant activity, associated with its high content in anthocyanins, polyphenols, and flavonoids. Previous studies reveal different pharmacological properties for this berry, but its cardioprotective potential has been little studied. Despite having an abundant composition, and being rich in bioactive products with an antiplatelet role, there are few studies linking this berry with antiplatelet activity. This review summarizes and discusses relevant information on the cardioprotective potential of Maqui, based on its composition of bioactive compounds, mainly as a nutraceutical antiplatelet agent. Articles published between 2000 and 2022 in the following bibliographic databases were selected: PubMed, ScienceDirect, and Google Scholar. Our search revealed that Maqui is a promising cardiovascular target since extracts from this berry have direct effects on the reduction in cardiovascular risk factors (glucose index, obesity, diabetes, among others). Although studies on antiplatelet activity in this fruit are recent, its rich chemical composition clearly shows that the presence of chemical compounds (anthocyanins, flavonoids, phenolic acids, among others) with high antiplatelet potential can provide this berry with antiplatelet properties. These bioactive compounds have antiplatelet effects with multiple targets in the platelet, particularly, they have been related to the inhibition of thromboxane, thrombin, ADP, and GPVI receptors, or through the pathways by which these receptors stimulate platelet aggregation. Detailed studies are needed to clarify this gap in the literature, as well as to specifically evaluate the mechanism of action of Maqui extracts, due to the presence of phenolic compounds.
Collapse
|
8
|
Tolksdorf C, Moritz E, Wolf R, Meyer U, Marx S, Bien-Möller S, Garscha U, Jedlitschky G, Rauch BH. Platelet-Derived S1P and Its Relevance for the Communication with Immune Cells in Multiple Human Diseases. Int J Mol Sci 2022; 23:ijms231810278. [PMID: 36142188 PMCID: PMC9499465 DOI: 10.3390/ijms231810278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 11/16/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a versatile signaling lipid involved in the regulation of numerous cellular processes. S1P regulates cellular proliferation, migration, and apoptosis as well as the function of immune cells. S1P is generated from sphingosine (Sph), which derives from the ceramide metabolism. In particular, high concentrations of S1P are present in the blood. This originates mainly from erythrocytes, endothelial cells (ECs), and platelets. While erythrocytes function as a storage pool for circulating S1P, platelets can rapidly generate S1P de novo, store it in large quantities, and release it when the platelet is activated. Platelets can thus provide S1P in a short time when needed or in the case of an injury with subsequent platelet activation and thereby regulate local cellular responses. In addition, platelet-dependently generated and released S1P may also influence long-term immune cell functions in various disease processes, such as inflammation-driven vascular diseases. In this review, the metabolism and release of platelet S1P are presented, and the autocrine versus paracrine functions of platelet-derived S1P and its relevance in various disease processes are discussed. New pharmacological approaches that target the auto- or paracrine effects of S1P may be therapeutically helpful in the future for pathological processes involving S1P.
Collapse
Affiliation(s)
- Céline Tolksdorf
- Division of Pharmacology and Toxicology, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
- Department of General Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Eileen Moritz
- Department of General Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Robert Wolf
- Department of General Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Ulrike Meyer
- Division of Pharmacology and Toxicology, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Sascha Marx
- Department of Neurosurgery, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Sandra Bien-Möller
- Department of General Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany
- Department of Neurosurgery, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Ulrike Garscha
- Institute of Pharmacy, University of Greifswald, 17489 Greifswald, Germany
| | - Gabriele Jedlitschky
- Department of General Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Bernhard H. Rauch
- Division of Pharmacology and Toxicology, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
- Correspondence:
| |
Collapse
|
9
|
Xu J, Wang J, Chen Y, Hou Y, Hu J, Wang G. Recent advances of natural and bioengineered extracellular vesicles and their application in vascular regeneration. Regen Biomater 2022; 9:rbac064. [PMID: 36176713 PMCID: PMC9514852 DOI: 10.1093/rb/rbac064] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/13/2022] [Accepted: 08/24/2022] [Indexed: 11/22/2022] Open
Abstract
The progression of cardiovascular diseases such as atherosclerosis and myocardial infarction leads to serious vascular injury, highlighting the urgent need for targeted regenerative therapy. Extracellular vesicles (EVs) composed of a lipid bilayer containing nuclear and cytosolic materials are relevant to the progression of cardiovascular diseases. Moreover, EVs can deliver bioactive cargo in pathological cardiovascular and regulate the biological function of recipient cells, such as inflammation, proliferation, angiogenesis and polarization. However, because the targeting and bioactivity of natural EVs are subject to several limitations, bioengineered EVs have achieved wide advancements in biomedicine. Bioengineered EVs involve three main ways to acquire including (i) modification of the EVs after isolation; (ii) modification of producer cells before EVs’ isolation; (iii) synthesize EVs using natural or modified cell membranes, and encapsulating drugs or bioactive molecules into EVs. In this review, we first summarize the cardiovascular injury-related disease and describe the role of different cells and EVs in vascular regeneration. We also discuss the application of bioengineered EVs from different producer cells to cardiovascular diseases. Finally, we summarize the surface modification on EVs which can specifically target abnormal cells in injured vascular.
Collapse
Affiliation(s)
| | | | - Yidan Chen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Yuanfang Hou
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Jianjun Hu
- Correspondence address. E-mail: (G.W.); (J.H.)
| | - Guixue Wang
- Correspondence address. E-mail: (G.W.); (J.H.)
| |
Collapse
|
10
|
An elevated platelet-to-lymphocyte ratio is associated with a higher risk of intracranial atherosclerotic stenosis. Chin Med J (Engl) 2022; 135:1425-1431. [PMID: 35868006 PMCID: PMC9481432 DOI: 10.1097/cm9.0000000000002228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Previous studies have shown that inflammation plays an important role in intracranial atherosclerotic stenosis (ICAS). The platelet-to-lymphocyte ratio (PLR) has recently emerged as a potential inflammatory biomarker. This study aimed to explore the association of the PLR with ICAS in a Chinese Han population. METHODS A total of 2134 participants (518 with ICAS, 1616 without ICAS) were enrolled in this study. ICAS was defined as atherosclerotic stenosis >50% or the occlusion of several main intracranial arteries. Multivariable logistic regression analyses were used to assess the association of the PLR with ICAS. Additional subgroup analyses were performed according to age (<60 vs. ≥ 60 years) and acute ischemic stroke. RESULTS Multivariate regression analysis showed that a high PLR was associated with a higher risk of ICAS in all participants (P < 0.001). Compared with the lowest quartile, the fourth PLR quartile was significantly associated with ICAS (OR 1.705, 95% confidence interval 1.278-2.275, P < 0.001). In the subgroups stratified by age, an association between the PLR and ICAS was found in the late-life group (P < 0.001), but not in the mid-life group (P = 0.650). In the subgroups stratified by acute ischemic stroke, the relationship between an elevated PLR and a higher risk of ICAS remained unchanged (stroke group, P < 0.001; non-stroke group, P = 0.027). CONCLUSIONS An elevated PLR was associated with a higher risk of ICAS in a Chinese Han population. The PLR might serve as a potential biomarker for ICAS in the elderly population.
Collapse
|
11
|
Hu ZB, Zhong QQ, Lu ZX, Zhu F. Association of platelet-to-white blood cell ratio and platelet-to-neutrophil ratio with the risk of fatal stroke occurrence in middle-aged to older Chinese. BMC Geriatr 2022; 22:430. [PMID: 35581556 PMCID: PMC9112464 DOI: 10.1186/s12877-022-03134-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/05/2022] [Indexed: 11/10/2022] Open
Abstract
Background White blood cell (WBC) and neutrophil (NEUT) counts, which are commonly inflammatory markers, have been related to an increased risk of fatal stroke. However, it is unclear whether platelet-to-white blood cell ratio (PWR) and platelet-to-neutrophil ratio (PNR) are related to the risk of fatal stroke in middle-aged to older populations. Method In total, 27,811 participants without a stroke history at baseline were included and followed up for a mean of 14.3 years (standard deviation = 3.2), and 838 stroke deaths were recorded. The Cox proportional hazards regression was used to assess the relationships between the PWR and the PNR and the risk of fatal strokes. Results Compared to the 1st quartile, an increased risk of fatal all stroke showed among the participants in the highest quartiles of both the WBC (adjusted hazard ratio (aHR) = 1.35, 95% confidence interval (CI) 1.09–1.66) and the NEUT (aHR = 1.45, 95% CI 1.18–1.79). The restricted cubic splines showed decreased trends in associations of the PWR and the PNR with the risk of fatal all stroke. A decreased risk of fatal all stroke showed in those with the highest quartiles for both the PWR (aHR = 0.73, 95% CI 0.53–1.00) and the PNR (aHR = 0.74, 95% CI 0.54–1.01). The participants with the 2nd, the 3rd and the 4th change quartiles for the PWR and the PNR had weak decreasing trends for the risk of fatal all stroke, compared to those in the 1st change quartile, and the significant associations were observed in those with an increase of 20% for the PWR with the risk of fatal haemarragic stroke (aHR = 0.47, 95% CI 0.22–0.95) and a decrease of 20% for the PNR with the risk of fatal all stroke (aHR = 1.33, 95% CI 0.99–1.79), compared to those with stable dynamic changes. Conclusions Higher neutrophil count and platelet-to-neutrophil ratio were associated with a contrary risk of fatal stroke, with an increased for the former and a decreased for the later. A potentially chronic inflammation should be paid close attention to stroke occurrence in relatively healthy middle-aged to older populations. Supplementary Information The online version contains supplementary material available at 10.1186/s12877-022-03134-z.
Collapse
Affiliation(s)
- Zhi-Bing Hu
- Department of Internal Medicine and Central Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Qiong-Qiong Zhong
- Department of Internal Medicine and Central Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou, China.,Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Jinan, China
| | - Ze-Xiong Lu
- Department of Internal Medicine, Sanya Central Hospital, Sanya, China
| | - Feng Zhu
- Department of Internal Medicine and Central Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou, China.
| |
Collapse
|
12
|
Prognostic value of the combination of TIMI risk score and mean platelet volume to lymphocyte count ratio in patients with acute coronary syndrome. COR ET VASA 2021. [DOI: 10.33678/cor.2021.095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
13
|
Baidildinova G, Nagy M, Jurk K, Wild PS, Ten Cate H, van der Meijden PEJ. Soluble Platelet Release Factors as Biomarkers for Cardiovascular Disease. Front Cardiovasc Med 2021; 8:684920. [PMID: 34235190 PMCID: PMC8255615 DOI: 10.3389/fcvm.2021.684920] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/24/2021] [Indexed: 12/19/2022] Open
Abstract
Platelets are the main players in thrombotic diseases, where activated platelets not only mediate thrombus formation but also are involved in multiple interactions with vascular cells, inflammatory components, and the coagulation system. Although in vitro reactivity of platelets provides information on the function of circulating platelets, it is not a full reflection of the in vivo activation state, which may be relevant for thrombotic risk assessment in various disease conditions. Therefore, studying release markers of activated platelets in plasma is of interest. While this type of study has been done for decades, there are several new discoveries that highlight the need for a critical assessment of the available tests and indications for platelet release products. First, new insights have shown that platelets are not only prominent players in arterial vascular disease, but also in venous thromboembolism and atrial fibrillation. Second, knowledge of the platelet proteome has dramatically expanded over the past years, which contributed to an increasing array of tests for proteins released and shed from platelets upon activation. Identification of changes in the level of plasma biomarkers associated with upcoming thromboembolic events allows timely and individualized adjustment of the treatment strategy to prevent disease aggravation. Therefore, biomarkers of platelet activation may become a valuable instrument for acute event prognosis. In this narrative review based on a systematic search of the literature, we summarize the process of platelet activation and release products, discuss the clinical context in which platelet release products have been measured as well as the potential clinical relevance.
Collapse
Affiliation(s)
- Gaukhar Baidildinova
- Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands.,Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Magdolna Nagy
- Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site RhineMain, Mainz, Germany
| | - Philipp S Wild
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site RhineMain, Mainz, Germany.,Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Hugo Ten Cate
- Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands.,Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center, Maastricht, Netherlands
| | - Paola E J van der Meijden
- Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands.,Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center, Maastricht, Netherlands
| |
Collapse
|
14
|
Different types of cell death in vascular diseases. Mol Biol Rep 2021; 48:4687-4702. [PMID: 34013393 DOI: 10.1007/s11033-021-06402-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/08/2021] [Indexed: 10/21/2022]
Abstract
In a mature organism, tissue homeostasis is regulated by cell division and cell demise as the two major physiological procedures. There is increasing evidence that deregulation of these processes is important in the pathogenicity of main diseases, including myocardial infarction, stroke, atherosclerosis, and inflammatory diseases. Therefore, there are ongoing efforts to discover modulating factors of the cell cycle and cell demise planners aiming at shaping innovative therapeutically modalities to the therapy of such diseases. Although the life of a cell is terminated by several modes of action, a few cell deaths exist-some of which resemble apoptosis and/or necrosis, and most of them are different from one another-that contribute to a wide range of functions to either support or disrupt the homoeostasis. Even in normal physiological conditions, cell life is severe within the cardiovascular system. Cells are persistently undergoing stretch, contraction, injurious metabolic byproducts, and hemodynamic forces, and a few of cells sustain decade-long lifetimes. The duration of vascular disease causes further exposure of vascular cells to a novel range of offences, most of which induce cell death. There is growing evidence on consequences of direct damage to a cell, as well as on responses of adjacent and infiltrating cells, which also have an effect on the pathology. In this study, by focusing on different pathways of cell death in different vascular diseases, an attempt is made to open a new perspective on the therapeutic goals associated with cell death in these diseases.
Collapse
|
15
|
Mariscal A, Zamora C, Magallares B, Salman-Monte TC, Ortiz MÀ, Díaz-Torné C, Castellví I, Corominas H, Vidal S. Phenotypic and Functional Consequences of PLT Binding to Monocytes and Its Association with Clinical Features in SLE. Int J Mol Sci 2021; 22:4719. [PMID: 33947017 PMCID: PMC8125177 DOI: 10.3390/ijms22094719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/21/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022] Open
Abstract
Platelets (PLTs) can modulate the immune system through the release of soluble mediators or through interaction with immune cells. Monocytes are the main immune cells that bind with PLTs, and this interaction is increased in several inflammatory and autoimmune conditions, including systemic lupus erythematosus (SLE). Our aim was to characterize the phenotypic and functional consequences of PLT binding to monocytes in healthy donors (HD) and in SLE and to relate it to the pathogenesis of SLE. We analyzed the phenotypic and functional features of monocytes with non-activated and activated bound PLTs by flow cytometry. We observed that monocytes with bound PLTs and especially those with activated PLTs have an up-regulated HLA-DR, CD86, CD54, CD16 and CD64 expression. Monocytes with bound PLTs also have an increased capacity for phagocytosis, though not for efferocytosis. In addition, monocytes with bound PLTs have increased IL-10, but not TNF-α, secretion. The altered phenotypic and functional features are comparable in SLE and HD monocytes and in bound PLTs. However, the percentages of monocytes with bound PLTs are significantly higher in SLE patients and are associated with undetectable levels of anti-dsDNA antibodies and hematuria, and with normal C3 and albumin/creatinine levels. Our results suggest that PLTs have a modulatory influence on monocytes and that this effect may be highlighted by an increased binding of PLTs to monocytes in autoimmune conditions.
Collapse
Affiliation(s)
- Anaís Mariscal
- Immunology Department, Hospital de la Santa Creu I Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain;
| | - Carlos Zamora
- Laboratory of Inflammatory Diseases, Hospital de la Santa Creu I Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (C.Z.); (M.À.O.)
| | - Berta Magallares
- Rheumatology Department, Hospital de la Santa Creu I Sant Pau, 08041 Barcelona, Spain; (B.M.); (C.D.-T.); (I.C.); (H.C.)
| | | | - Mª Àngels Ortiz
- Laboratory of Inflammatory Diseases, Hospital de la Santa Creu I Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (C.Z.); (M.À.O.)
| | - Cesar Díaz-Torné
- Rheumatology Department, Hospital de la Santa Creu I Sant Pau, 08041 Barcelona, Spain; (B.M.); (C.D.-T.); (I.C.); (H.C.)
| | - Iván Castellví
- Rheumatology Department, Hospital de la Santa Creu I Sant Pau, 08041 Barcelona, Spain; (B.M.); (C.D.-T.); (I.C.); (H.C.)
| | - Héctor Corominas
- Rheumatology Department, Hospital de la Santa Creu I Sant Pau, 08041 Barcelona, Spain; (B.M.); (C.D.-T.); (I.C.); (H.C.)
| | - Silvia Vidal
- Laboratory of Inflammatory Diseases, Hospital de la Santa Creu I Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (C.Z.); (M.À.O.)
| |
Collapse
|
16
|
Zarmehri B, Shahi B, Rahmani S, Dehghan Tafti F, Foroughian M. Association of platelet count and mean platelet volume (MPV) index with types of stroke. CASPIAN JOURNAL OF INTERNAL MEDICINE 2021; 11:398-402. [PMID: 33680381 PMCID: PMC7911760 DOI: 10.22088/cjim.11.4.398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Background: Stroke is known to be the third most prominent cause of death in the developing countries and the most common debilitating neurologic disease. This study aimed to investigate the association of platelet count (PC) and mean platelet volume (MPV) index with various stroke types. Methods: This cross-sectional study was carried out on patients over the age of 18 years who presented with signs and symptoms of the first acute stroke. Exclusion criteria were underlying chronic liver or renal disease and the time more than 6 hours from symptom initiation, hematological and infectious disorders in patients. After recording of demographic data, a complete blood cell count (CBC) test was performed. Results: From 150 patients, who enrolled in the study, 54.7% of patients were males. The initial brain CT scan was normal in 13 (8.7%) patients and showed evidence of brain infarction and intracranial hemorrhage in 84 (56%) and 53 (35.3%) patients respectively. Patients with intracranial hemorrhage had significantly higher mean of MPV index than the patients with normal brain-CT scan and patients with evidence of brain infarction (p<0.001). Conclusion: The MVP index can be a predictor of the type of hemorrhagic or ischemic finding in emergency CT scan in stroke patients. This relationship may help to better understand the physiopathologic role of platelets in the development of stroke (hemorrhagic or ischemic), but will not replace cerebral computed tomography to diagnose the type of stroke, or it may not initiate treatment for hemorrhagic stroke.
Collapse
Affiliation(s)
- Bahram Zarmehri
- Department of Emergency Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Behzad Shahi
- Department of Emergency Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shaghayegh Rahmani
- Department of Emergency Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mahdi Foroughian
- Department of Emergency Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
17
|
Gąsecka A, Rogula S, Szarpak Ł, Filipiak KJ. LDL-Cholesterol and Platelets: Insights into Their Interactions in Atherosclerosis. Life (Basel) 2021; 11:39. [PMID: 33440673 PMCID: PMC7826814 DOI: 10.3390/life11010039] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 12/23/2022] Open
Abstract
Atherosclerosis and its complications, including acute coronary syndromes, are the major cause of death worldwide. The two most important pathophysiological mechanisms underlying atherosclerosis include increased platelet activation and increased low-density lipoproteins (LDL) concentration. In contrast to LDL, oxidized (ox)-LDL have direct pro-thrombotic properties by functional interactions with platelets, leading to platelet activation and favoring thrombus formation. In this review, we summarize the currently available evidence on the interactions between LDL-cholesterol and platelets, which are based on (i) the presence of ox-LDL-binding sites on platelets, (ii) generation of ox-LDL by platelets and (iii) the role of activated platelets and ox-LDL in atherosclerosis. In addition, we elaborate on the clinical implications of these interactions, including development of the new therapeutic possibilities. The ability to understand and modulate mechanisms governing interactions between LDL-cholesterol and platelets may offer new treatment strategies for atherosclerosis prevention.
Collapse
Affiliation(s)
- Aleksandra Gąsecka
- Department of Cardiology, Medical University of Warsaw, 02-091 Warsaw, Poland; (S.R.); (K.J.F.)
| | - Sylwester Rogula
- Department of Cardiology, Medical University of Warsaw, 02-091 Warsaw, Poland; (S.R.); (K.J.F.)
| | - Łukasz Szarpak
- Bialystok Oncology Center, 15-027, Bialystok, Poland;
- Maria Sklodowska-Curie Medical Academy in Warsaw, 03-411 Warsaw, Poland
| | - Krzysztof J. Filipiak
- Department of Cardiology, Medical University of Warsaw, 02-091 Warsaw, Poland; (S.R.); (K.J.F.)
| |
Collapse
|
18
|
Cankurt T, Celik İE, Ozturk S, Maden O. Inflammatory Conditions in Acute Coronary Syndrome Patients Treated with Percutaneous Coronary Intervention of Saphenous Vein Graft. Int J Angiol 2020; 29:237-244. [PMID: 33268974 PMCID: PMC7690989 DOI: 10.1055/s-0040-1714751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The study aimed to evaluate the inflammatory blood parameters in acute coronary syndrome (ACS) patients with a history of coronary artery bypass graft (CABG) and treated with percutaneous coronary intervention (PCI) of saphenous vein graft (SVG). A total of 347 patients who underwent urgent SVG PCI with the diagnosis of ACS were included in the study. After the application of exclusion criteria, 79 patients were allocated into two groups, namely, successful PCI ( n = 59) and unsuccessful PCI ( n = 20), and included in the statistical analysis. Neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) levels were significantly higher in patients with unsuccessful SVG PCI. In the logistic regression analysis, PLR, C-reactive protein, and diabetes mellitus emerged as independent factors associated with unsuccessful SVG PCI. The area under the curve for PLR was 0.70 (95% confidence interval: 0.55-0.85, p = 0.006). The cut-off value of PLR (128.99) was associated with 70.0% sensitivity and 69.5% specificity. Elevated inflammatory status is associated with unsuccessful PCI of SVG in ACS patients. Increased PLR levels on admission is an independent predictor of this situation. This cheap and simple marker can help us to predict unsuccessful SVG PCI in ACS patients.
Collapse
Affiliation(s)
- Tayyar Cankurt
- Cardiology Clinic, Amasya University Sabuncuoglu Sereefeddin State Hospital, Ankara, Turkey
| | - İbrahim E. Celik
- Department of Cardiology, University of Health Sciences, Ankara Education and Research Hospital, Ankara, Turkey
| | - Selcuk Ozturk
- Department of Cardiology, University of Health Sciences, Ankara Education and Research Hospital, Ankara, Turkey
| | - Orhan Maden
- Department of Cardiology, University of Health Sciences, Ankara Bilkent City Hospital, Ankara, Turkey
| |
Collapse
|
19
|
Zhao Y, Zheng K, Guan B, Guo M, Song L, Gao J, Qu H, Wang Y, Shi D, Zhang Y. DLDTI: a learning-based framework for drug-target interaction identification using neural networks and network representation. J Transl Med 2020; 18:434. [PMID: 33187537 PMCID: PMC7666529 DOI: 10.1186/s12967-020-02602-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/01/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Drug repositioning, the strategy of unveiling novel targets of existing drugs could reduce costs and accelerate the pace of drug development. To elucidate the novel molecular mechanism of known drugs, considering the long time and high cost of experimental determination, the efficient and feasible computational methods to predict the potential associations between drugs and targets are of great aid. METHODS A novel calculation model for drug-target interaction (DTI) prediction based on network representation learning and convolutional neural networks, called DLDTI, was generated. The proposed approach simultaneously fused the topology of complex networks and diverse information from heterogeneous data sources, and coped with the noisy, incomplete, and high-dimensional nature of large-scale biological data by learning the low-dimensional and rich depth features of drugs and proteins. The low-dimensional feature vectors were used to train DLDTI to obtain the optimal mapping space and to infer new DTIs by ranking candidates according to their proximity to the optimal mapping space. More specifically, based on the results from the DLDTI, we experimentally validated the predicted targets of tetramethylpyrazine (TMPZ) on atherosclerosis progression in vivo. RESULTS The experimental results showed that the DLDTI model achieved promising performance under fivefold cross-validations with AUC values of 0.9172, which was higher than the methods using different classifiers or different feature combination methods mentioned in this paper. For the validation study of TMPZ on atherosclerosis, a total of 288 targets were identified and 190 of them were involved in platelet activation. The pathway analysis indicated signaling pathways, namely PI3K/Akt, cAMP and calcium pathways might be the potential targets. Effects and molecular mechanism of TMPZ on atherosclerosis were experimentally confirmed in animal models. CONCLUSIONS DLDTI model can serve as a useful tool to provide promising DTI candidates for experimental validation. Based on the predicted results of DLDTI model, we found TMPZ could attenuate atherosclerosis by inhibiting signal transductions in platelets. The source code and datasets explored in this work are available at https://github.com/CUMTzackGit/DLDTI .
Collapse
Affiliation(s)
- Yihan Zhao
- Department of Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Kai Zheng
- School of Computer Science and Engineering, Central South University, Changsha, China
| | - Baoyi Guan
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Cardiovascular Diseases Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mengmeng Guo
- Institute of Cardiovascular Sciences, Health Science Center, Peking University, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Lei Song
- Department of Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Gao
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Cardiovascular Diseases Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hua Qu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Cardiovascular Diseases Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuhui Wang
- Institute of Cardiovascular Sciences, Health Science Center, Peking University, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Dazhuo Shi
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Cardiovascular Diseases Center, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Ying Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Cardiovascular Diseases Center, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
20
|
Rodriguez BA, Bhan A, Beswick A, Elwood PC, Niiranen TJ, Salomaa V, Trégouët DA, Morange PE, Civelek M, Ben-Shlomo Y, Schlaeger T, Chen MH, Johnson AD, Johnson AD. A Platelet Function Modulator of Thrombin Activation Is Causally Linked to Cardiovascular Disease and Affects PAR4 Receptor Signaling. Am J Hum Genet 2020; 107:211-221. [PMID: 32649856 DOI: 10.1016/j.ajhg.2020.06.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 06/03/2020] [Indexed: 12/17/2022] Open
Abstract
Dual antiplatelet therapy reduces ischemic events in cardiovascular disease, but it increases bleeding risk. Thrombin receptors PAR1 and PAR4 are drug targets, but the role of thrombin in platelet aggregation remains largely unexplored in large populations. We performed a genome-wide association study (GWAS) of platelet aggregation in response to full-length thrombin, followed by clinical association analyses, Mendelian randomization, and functional characterization including iPSC-derived megakaryocyte and platelet experiments. We identified a single sentinel variant in the GRK5 locus (rs10886430-G, p = 3.0 × 10-42) associated with increased thrombin-induced platelet aggregation (β = 0.70, SE = 0.05). We show that disruption of platelet GRK5 expression by rs10886430-G is associated with enhanced platelet reactivity. The proposed mechanism of a GATA1-driven megakaryocyte enhancer is confirmed in allele-specific experiments. Utilizing further data, we demonstrate that the allelic effect is highly platelet- and thrombin-specific and not likely due to effects on thrombin levels. The variant is associated with increased risk of cardiovascular disease outcomes in UK BioBank, most strongly with pulmonary embolism. The variant associates with increased risk of stroke in the MEGASTROKE, UK BioBank, and FinnGen studies. Mendelian randomization analyses in independent samples support a causal role for rs10886430-G in increasing risk for stroke, pulmonary embolism, and venous thromboembolism through its effect on thrombin-induced platelet reactivity. We demonstrate that G protein-coupled receptor kinase 5 (GRK5) promotes platelet activation specifically via PAR4 receptor signaling. GRK5 inhibitors in development for the treatment of heart failure and cancer could have platelet off-target deleterious effects. Common variants in GRK5 may modify clinical outcomes with PAR4 inhibitors, and upregulation of GRK5 activity or signaling in platelets may have therapeutic benefits.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Andrew D Johnson
- National Heart, Lung, and Blood Institute, Division of Intramural Research, Population Sciences Branch, The Framingham Heart Study, Framingham, MA 01702, USA.
| |
Collapse
|
21
|
Platelet-to-lymphocyte ratio is a predictor of long-term mortality in patients with acute coronary syndrome. ADVANCES IN INTERVENTIONAL CARDIOLOGY 2020; 16:170-176. [PMID: 32636901 PMCID: PMC7333190 DOI: 10.5114/aic.2020.95859] [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: 02/18/2020] [Accepted: 04/27/2020] [Indexed: 01/23/2023] Open
Abstract
Introduction Atherosclerosis is a chronic inflammatory process and inflammation is an important component of acute coronary syndrome (ACS). Platelet-to-lymphocyte ratio (PLR) is a useful parameter showing the degree of the inflammatory response. Aim To explore the association between PLR and long-term mortality in patients with ACS. Material and methods A total of 538 patients who had a diagnosis of ACS between January 2012 and August 2013 were followed up to 60 months. On admission, blood sampling to calculate PLR and detailed clinical data were obtained. Results In total, 538 patients with a mean age of 61.5 ±13.1 years (69% male) were enrolled in the study. Median follow-up was 79 months (IQR: 74–83 months). Patients were divided into 3 tertiles based on PLR levels. Five-year mortality of the patients was significantly higher among patients in the upper PLR tertile when compared with the lower and middle PLR tertile groups (55 (30.7%) vs. 27 (15.0%) and 34 (19.0%); p < 0.001, p = 0.010 respectively). In the Cox regression analysis, a high level of PLR was an independent predictor of 5-year mortality (OR = 1.005, 95% CI: 1.001–1.008, p = 0.004). Kaplan-Meier analysis according to the long-term mortality-free survival revealed the higher occurrence of mortality in the third PLR tertile group compared to the first (p < 0.001) and second tertiles (p = 0.009). Conclusions PLR, which is an easily calculated and universally available marker, may be useful in long-term risk classification of patients presenting with ACS.
Collapse
|
22
|
Chen X, Shao M, Zhang T, Zhang W, Meng Y, Zhang H, Hai H, Li G. Prognostic value of the combination of GRACE risk score and mean platelet volume to lymphocyte count ratio in patients with ST-segment elevation myocardial infarction after percutaneous coronary intervention. Exp Ther Med 2020; 19:3664-3674. [PMID: 32346430 PMCID: PMC7185188 DOI: 10.3892/etm.2020.8626] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 12/17/2019] [Indexed: 12/16/2022] Open
Abstract
The Global Registry of Acute Coronary Events (GRACE) risk score and the mean platelet volume to lymphocyte count ratio (MPVLR) can be used independently to predict adverse outcomes in patients with acute coronary syndromes. However, the level of MPVLR in relation to the GRACE score, and whether a combination of these methods can better predict the clinical adverse outcome of patients with ST-segment elevation myocardial infarction (STEMI), have not been previously examined. Therefore, the aim of the present study was to investigate whether the combination of GRACE risk score and MPVLR is a good predictor of a 30-day major adverse cardiovascular events (MACE) in patients with STEMI. A total of 464 patients with STEMI undergoing percutaneous coronary intervention (PCI) were enrolled, and divided into four groups based on the optimal cut-off values for GRACE score and MPVLR. GRACE score and MPVLR levels were separately recorded during admission. Spearman's rank correlation analysis showed a positive correlation between GRACE score and MPVLR (ρ=0.304; P<0.001). Both GRACE score [hazard ratio (HR), 1.706; 95% CI, 1.435-3.058; P<0.001] and MPVLR level (HR, 1.668; 95% CI, 1.202-2.170; P<0.001) were found to be independent predictors of a 30-day MACE. Additionally, the high MPVLR + high GRACE score group of patients had an HR of 2.455 (95% CI, 1.736-3.188) for a 30-day MACE, when using the low MPVLR + low GRACE score group as a reference. Based on the area under the curve, MPVLR combined with GRACE scores achieved an improved performance in differentiating angiographic no-reflow during a 30-day MACE, compared with individual MPVLR and GRACE scores. Therefore, the present results suggested that the GRACE score may be positively correlated with MPVLR and that their combination accurately predicted the occurrence of short-term MACE in patients with STEMI after PCI.
Collapse
Affiliation(s)
- Xinsen Chen
- Department of Emergency, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| | - Meng Shao
- Department of Pathophysiology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| | - Tian Zhang
- Department of Emergency, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| | - Wei Zhang
- Department of Emergency, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| | - Youbao Meng
- Department of Emergency, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| | - Hongyan Zhang
- Department of Emergency, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| | - Hua Hai
- Department of Emergency, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| | - Guihua Li
- Department of Emergency, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| |
Collapse
|
23
|
Bukhari IA, Habib SS, Alnahedh A, Almutairi F, Alkahtani L, Alareek LA, Assiri GA. Relationship of Body Adiposity with Platelet Function in Obese and Non-obese Individuals. Cureus 2020; 12:e6815. [PMID: 32133271 PMCID: PMC7049893 DOI: 10.7759/cureus.6815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background Adiposity is firmly linked to a higher incidence of various cardiovascular and metabolic morbidities, including diabetes, hypertension, and thromboembolism. This research study was aimed to verify the association of increased adiposity and hyperreactivity of platelets in obese and non-obese individuals. Methods This cross-sectional study was conducted on 42 subjects aged 18 years and above. Subjects were divided into obese and non-obese groups based on their body mass index (BMI). The data was collected through self-administered questionnaires. All participants underwent body composition analysis. Blood samples were collected from all subjects and taken to the Pharmacology Department for the preparation of platelet-rich plasma (PRP) and poor platelet plasma (PPP). Platelet aggregation was induced by arachidonic acid and was monitored with a Bio/Data multichannel aggregation profiler (Bio/Data Corp., Horsham, PA, USA). Results Significant differences were observed in most parameters, such as fat mass, body fat percentage, free fat mass (FFM), the percentage of trunk fat, total body water, waist-hip ratio (WHR), and basal metabolic rate (BMR) of obese and non-obese subjects. The average percent of platelet aggregation in obese and non-obese subjects was 56.33 ± 15.62 and 59.38 ± 12.62, respectively. The average area under the curve (AUC) for platelet aggregation for both groups was 339.33 ± 191.55 and 342 ± 146.68, respectively. Platelet function was not significantly different and didn’t positively correlate with most parameters of the body composition, except WHR, which positively correlated with AUC for platelet function. Conclusion There was no significant direct correlation between adiposity and platelet activation in obese subjects. However, a significant positive correlation of AUC for platelet aggregation with WHR was observed (resistance (r)-value: 0.307, p < 0.05). These findings suggest that WHR could be an effective determinant to assess the risk of thromboembolism in obese individuals.
Collapse
Affiliation(s)
- Ishfaq A Bukhari
- Pharmacology, College of Medicine, King Saud University, Riyadh, SAU
| | - Syed S Habib
- Physiology Department, College of Medicine and King Saud University Medical City, King Saud University, Riyadh, SAU
| | - Alaa Alnahedh
- Pharmacology Section, College of Medicine and King Saud University Medical City, King Saud University, Riyadh, SAU
| | - Futoon Almutairi
- Pharmacology Section, College of Medicine and King Saud University Medical City, King Saud University, Riyadh, SAU
| | - Lama Alkahtani
- Pharmacology Section, College of Medicine and King Saud University Medical City, King Saud University, Riyadh, SAU
| | - Latefa A Alareek
- Pharmacology Section, College of Medicine and King Saud University Medical City, King Saud University, Riyadh, SAU
| | - Ghadah A Assiri
- Clinical Pharmacology Department, College of Pharmacy, King Saud University, Riyadh, SAU
| |
Collapse
|
24
|
Chen SY, Lin YS, Cheng YF, Wang H, Niu XT, Zhang WL. Mean Platelet Volume-To-Lymphocyte Ratio Predicts Poor Functional Outcomes Among Ischemic Stroke Patients Treated With Intravenous Thrombolysis. Front Neurol 2020; 10:1274. [PMID: 31920909 PMCID: PMC6914757 DOI: 10.3389/fneur.2019.01274] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 11/18/2019] [Indexed: 12/22/2022] Open
Abstract
Background and Purpose: According to previous studies, the mean platelet volume-to-lymphocyte ratio (MPVLR) represents a novel marker of a poor short-term prognosis in patients with a myocardial infarction who underwent primary percutaneous coronary intervention. We aimed to evaluate the association between MPVLR and clinical outcomes of patients with acute ischemic stroke who were treated with intravenous thrombolysis. Methods: Two hundred forty-one patients with ischemic stroke receiving intravenous thrombolysis were prospectively enrolled in this study. Blood samples for MPVLR were obtained at admission and at 18-24 h after treatment with intravenous thrombolysis. A poor functional outcome was defined as a modified Rankin scale score of 3-6 at 3 months after stroke. Results: At admission, the area under the curve of MPVLR to predict poor functional outcomes at 3 months was 0.613 [95% confidence interval (CI), 0.541-0.686; P = 0.003), and the best predictive MPVLR value was 5.8. Patients with an MPVLR ≥5.8 had a 3.141-fold increased risk of a poor outcome at 3 months (95% CI, 1.491-6.615; P = 0.003) compared to patients with an MPVLR <5.8. At 18-24 h after treatment with intravenous thrombolysis, the area under the curve of MPVLR to predict a poor outcome at 3 months was 0.697 (95% CI, 0.630-0.765, P < 0.001), and the best predictive MPVLR value was 6.9. The inclusion of MPVLR as a continuous (odds ratio, 1.145; 95% CI, 1.044-1.256, P = 0.004) and categorical variable (odds ratio, 6.555; 95% CI, 2.986-14.393, P < 0.001) was independently associated with poor outcomes at 3 months. Conclusions: Both the values of MPVLR at admission and 18-24 h after intravenous thrombolysis were independently associated with poor functional outcomes. MPVLR may serve as an activity marker for a poor prognosis in patients with acute ischemic stroke receiving intravenous thrombolysis.
Collapse
Affiliation(s)
- Si-Yan Chen
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuan-Shao Lin
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi-Fan Cheng
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hong Wang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiao-Ting Niu
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wan-Li Zhang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
25
|
Arauna D, Furrianca M, Espinosa-Parrilla Y, Fuentes E, Alarcón M, Palomo I. Natural Bioactive Compounds As Protectors Of Mitochondrial Dysfunction In Cardiovascular Diseases And Aging. Molecules 2019; 24:molecules24234259. [PMID: 31766727 PMCID: PMC6930637 DOI: 10.3390/molecules24234259] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/05/2019] [Accepted: 09/08/2019] [Indexed: 01/04/2023] Open
Abstract
Diet, particularly the Mediterranean diet, has been considered as a protective factor against the development of cardiovascular diseases, the main cause of death in the world. Aging is one of the major risk factors for cardiovascular diseases, which have an oxidative pathophysiological component, being the mitochondria one of the key organelles in the regulation of oxidative stress. Certain natural bioactive compounds have the ability to regulate oxidative phosphorylation, the production of reactive oxygen species and the expression of mitochondrial proteins; but their efficacy within the mitochondrial physiopathology of cardiovascular diseases has not been clarified yet. The following review has the purpose of evaluating several natural compounds with evidence of mitochondrial effect in cardiovascular disease models, ascertaining the main cellular mechanisms and their potential use as functional foods for prevention of cardiovascular disease and healthy aging.
Collapse
Affiliation(s)
- Diego Arauna
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Center on Aging, Universidad de Talca, Talca 3460000, Chile; (D.A.); (M.A.)
| | - María Furrianca
- Thematic Task Force on Aging, CUECH Research Network, Santiago 8320000, Chile; (M.F.); (Y.E.-P.)
- Departamento de enfermería, Universidad de Magallanes, Punta Arenas 6200000, Chile
| | - Yolanda Espinosa-Parrilla
- Thematic Task Force on Aging, CUECH Research Network, Santiago 8320000, Chile; (M.F.); (Y.E.-P.)
- Laboratory of Molecular Medicine —LMM, Center for Education, Healthcare and Investigation—CADI, Universidad de Magallanes, Punta Arenas 6200000, Chile
- School of Medicine, Universidad de Magallanes, Punta Arenas 6200000, Chile
| | - Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Center on Aging, Universidad de Talca, Talca 3460000, Chile; (D.A.); (M.A.)
- Thematic Task Force on Aging, CUECH Research Network, Santiago 8320000, Chile; (M.F.); (Y.E.-P.)
- Correspondence: (E.F.); (I.P.)
| | - Marcelo Alarcón
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Center on Aging, Universidad de Talca, Talca 3460000, Chile; (D.A.); (M.A.)
- Thematic Task Force on Aging, CUECH Research Network, Santiago 8320000, Chile; (M.F.); (Y.E.-P.)
| | - Iván Palomo
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Center on Aging, Universidad de Talca, Talca 3460000, Chile; (D.A.); (M.A.)
- Thematic Task Force on Aging, CUECH Research Network, Santiago 8320000, Chile; (M.F.); (Y.E.-P.)
- Correspondence: (E.F.); (I.P.)
| |
Collapse
|
26
|
Zhang H, Tang W, Wang S, Zhang J, Fan X. Tetramethylpyrazine Inhibits Platelet Adhesion and Inflammatory Response in Vascular Endothelial Cells by Inhibiting P38 MAPK and NF-κB Signaling Pathways. Inflammation 2019; 43:286-297. [DOI: 10.1007/s10753-019-01119-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
27
|
Abstract
Some vitamins have beneficial effects on cardiovascular diseases, normalizing platelet function and preventing their excess activation. Anti-platelet vitamins can act directly through inhibitory biochemical pathways in platelets or indirectly by preventing damage to the endothelium or low-density lipoprotein from oxidation. As a rule, each vitamin alone is a weak inhibitor of platelet aggregation. However, in combination, they may act synergistically or enhance the effects of endogenous anti-platelet compounds, such as prostacyclin or nitric oxide, and appear to have a sufficient anti-thrombotic effect. This review will focus on vitamins, which inhibit platelet activation and the mechanisms of their action. The relationship between the vitamins that inhibit platelet aggregation and vascular diseases is examined.
Collapse
Affiliation(s)
- Gennadi Kobzar
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| |
Collapse
|
28
|
Sonia D'Souza C, Li Z, Luke Maxwell D, Trusler O, Murphy M, Crewther S, Peter K, Orian JM. Platelets Drive Inflammation and Target Gray Matter and the Retina in Autoimmune-Mediated Encephalomyelitis. J Neuropathol Exp Neurol 2019; 77:567-576. [PMID: 29757405 DOI: 10.1093/jnen/nly032] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Despite growing evidence for platelets as active players in infection and immunity, it remains unresolved whether platelets contribute to, or are key elements in the development of neuroinflammation. Using the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis, we identified platelet accumulation in the circulation by 7-day postinduction (dpi), ahead of clinical onset which occurs at 13-14 dpi. By inducing platelet depletion between 7 and 16 dpi, we demonstrate an association between platelet accumulation in the spinal cord and disease development. Additionally, we provide evidence for platelet infiltration in the white and gray matter parenchyma, but with different outcomes. Thus, while in white matter platelets are clearly associated with lesions, in gray matter large-scale platelet infiltration and expression of the platelet-specific molecule PF4 are detectable prior to T cell entry. In the retina, platelet accumulation also precedes clinical onset and is associated with significant increase in retinal thickness in experimental relative to control animals. Platelet accumulation increases over the disease course in this tissue, but without subsequent T cell infiltration. These findings provide definitive confirmation that platelet accumulation is key to EAE pathophysiology. Furthermore, they suggest an undescribed and, most importantly, therapeutically targetable mechanism of neuronal damage.
Collapse
Affiliation(s)
| | - Zenjiang Li
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science
| | - Dain Luke Maxwell
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science
| | - Oliver Trusler
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science
| | - Melanie Murphy
- Department of Psychology and Counselling, La Trobe University, Melbourne, Victoria, Australia
| | - Sheila Crewther
- Department of Psychology and Counselling, La Trobe University, Melbourne, Victoria, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | |
Collapse
|
29
|
İyigün T, Kyaruzi MM, Timur B, Satılmışoğlu MH, İyigün M, Kaya M. The Predictive Effects of Clinical Hematological Changes on Saphenous Graft Patency after Coronary Artery Surgery. Braz J Cardiovasc Surg 2019; 34:297-304. [PMID: 31310468 PMCID: PMC6629236 DOI: 10.21470/1678-9741-2018-0211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Objectives To investigate the association between clinical hematologic parameters and
saphenous vein graft failure after on-pump coronary artery bypass
surgery. Methods A total of 1950 consecutive patients underwent isolated on-pump coronary
artery surgery between November 2010 and February 2013. Of these, 284
patients met our inclusion criteria; their preoperative clinical
hematological parameters were retrospectively obtained for this cohort
study. And of them, 109 patients underwent conventional coronary angiography
after graft failure was revealed by coronary computed tomography
angiography. The primary endpoint was to catch at least one saphenous vein
graft stenosis or occlusion following the coronary angiogram. We then
analyzed risk factors for graft failure. In sequential or T grafts, each
segment was analyzed as a separate graft. Results In logistic regression analysis, older age, platelet distribution width, and
diabetes mellitus were identified as independent predictors of saphenous
vein graft failure (P<0.). In contrast, preserved
ejection fraction value favored graft patency
(P<0.001). Conclusion Increased platelet distribution width is easily measurable and can be used as
a simple and valuable marker in the prediction of saphenous vein graft
failure.
Collapse
Affiliation(s)
- Taner İyigün
- Cardiovascular Surgery Training and Research Hospital Department of Cardiovascular Surgery Istanbul Turkey Department of Cardiovascular Surgery, Istanbul Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Mugisha Markior Kyaruzi
- Cardiovascular Surgery Training and Research Hospital Department of Cardiovascular Surgery Istanbul Turkey Department of Cardiovascular Surgery, Istanbul Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Barış Timur
- Cardiovascular Surgery Training and Research Hospital Department of Cardiovascular Surgery Istanbul Turkey Department of Cardiovascular Surgery, Istanbul Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Muhammed Hulusi Satılmışoğlu
- Cardiovascular Surgery Training and Research Hospital Department of Cardiology Istanbul Turkey Department of Cardiology, Istanbul Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Müzeyyen İyigün
- Acıbadem University Department of Anesthesiology and Reanimation Istanbul Turkey Department of Anesthesiology and Reanimation, Acıbadem University, Istanbul, Turkey
| | - Mehmet Kaya
- Cardiovascular Surgery Training and Research Hospital Department of Cardiovascular Surgery Istanbul Turkey Department of Cardiovascular Surgery, Istanbul Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
30
|
Gao J, Liu Y, Li YM. Review of risk factors, treatment, and prevention of saphenous vein graft disease after coronary artery bypass grafting. J Int Med Res 2018; 46:4907-4919. [PMID: 30178686 PMCID: PMC6300967 DOI: 10.1177/0300060518792445] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Saphenous vein graft disease (SVGD) is a type of vascular disease that may develop after coronary artery bypass grafting (CABG). SVGD seriously affects the short-term and long-term effects of CABG and increases the incidence of major adverse cardiovascular events. It is very important to identify patients at greatest risk and carry out prevention and treatment measures to determine the risk factors for SVGD. Many factors contribute to SVGD when the vein is grafted into an arterial environment, such as surgery-related factors, smoking, diabetes mellitus, hyperlipidemia, and others. In this review, we discuss the risk factors for SVGD, current surgical and pharmacologic therapies with which to manage SVGD, and the prevention of SVGD.
Collapse
Affiliation(s)
- Jing Gao
- 1 Logistics University of Chinese People's Armed Police Forces, Dongli District, Tianjin, P. R. China.,2 Cardiovascular Institute, Tianjin Chest Hospital, Jinnan District, Tianjin, P. R. China
| | - Yin Liu
- 3 Department of Cardiology, Tianjin Chest Hospital, Jinnan District, Tianjin, P. R. China
| | - Yu-Ming Li
- 1 Logistics University of Chinese People's Armed Police Forces, Dongli District, Tianjin, P. R. China
| |
Collapse
|
31
|
Haynes A, Linden MD, Robey E, Naylor LH, Ainslie PN, Cox KL, Lautenschlager NT, Green DJ. Beneficial impacts of regular exercise on platelet function in sedentary older adults: evidence from a randomized 6-mo walking trial. J Appl Physiol (1985) 2018; 125:401-408. [PMID: 29648514 DOI: 10.1152/japplphysiol.00079.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Platelet activation, including the formation of monocyte platelet aggregates (MPAs), contributes to atherosclerosis, thrombus formation, and acute coronary syndromes. Regular participation in exercise can lower cardiovascular risk, but little is known regarding the impact of exercise training on platelet function. We investigated the effect of 6 mo of walking exercise on platelet function in sedentary older adults without significant cardiovascular disease. Twenty-seven participants were randomly allocated to 6 mo of either: no-exercise ( n = 13) or 3 × 50 min/wk of supervised center-based walking ( n = 14). Circulating and agonist-induced MPAs were assessed using flow cytometry before [ month 0 (0M)] and after [ month 6 (6M)] the intervention. Circulating MPAs increased from 0M (3.7 ± 1.0%) to 6M (4.7 ± 1.6%) in the no-exercise group ( P = 0.009), whereas a nonsignificant decrease was observed in the walking group (0M 4.3 ± 1.7 vs. 6M 3.7 ± 1.2 %, P = 0.052). The change in MPAs between groups was significant ( P = 0.001). There were no differences between groups in platelet responses to agonists across the interventions (all P > 0.05). Collectively, these data suggest that the absence of regular exercise may increase MPAs, which are cellular mediators involved in atherosclerosis, while regular walking inhibits such increases. The thrombotic function of platelets appears to be relatively unaltered by exercise training. This study provides novel data related to the cardioprotective effects associated with participation in exercise. NEW & NOTEWORTHY Monocyte-platelet aggregates contribute to atherosclerosis and exercise can lower cardiovascular risk. This is the first study to discover that a lack of regular physical activity is associated with increased monocyte-platelet aggregates over a 6-mo intervention period. In contrast, walking exercise inhibits increased monocyte-platelet aggregates in the circulation. This study highlights a novel pathway by which regular participation in exercise exerts its cardioprotective effects.
Collapse
Affiliation(s)
- Andrew Haynes
- School of Human Sciences (Exercise and Sport Science), The University of Western Australia , Crawley, Western Australia
| | - Matthew D Linden
- School of Biomedical Sciences, The University of Western Australia , Crawley, Western Australia
| | - Elisa Robey
- School of Human Sciences (Exercise and Sport Science), The University of Western Australia , Crawley, Western Australia
| | - Louise H Naylor
- School of Human Sciences (Exercise and Sport Science), The University of Western Australia , Crawley, Western Australia
| | - Philip N Ainslie
- Centre for Heart, Lund and Vascular Health, School of Health and Exercise Science, The University of British Columbia , Kelowna, British Columbia , Canada
| | - Kay L Cox
- School of Human Sciences (Exercise and Sport Science), The University of Western Australia , Crawley, Western Australia.,School of Medicine (Royal Perth Hospital Unit), The University of Western Australia , Crawley, Western Australia
| | - Nicola T Lautenschlager
- Academic Unit for Psychiatry of Old Age, Department of Psychiatry, The University of Melbourne , Victoria , Australia.,NorthWestern Mental Health, Melbourne Health, Parkville, Victoria , Australia.,School of Clinical Neurosciences and the Western Australia Centre for Health and Ageing, The University of Western Australia , Crawley, Western Australia
| | - Daniel J Green
- School of Human Sciences (Exercise and Sport Science), The University of Western Australia , Crawley, Western Australia.,Principal Research Fellow, National Health and Medical Research Council , Australia
| |
Collapse
|
32
|
Blekkenhorst LC, Bondonno NP, Liu AH, Ward NC, Prince RL, Lewis JR, Devine A, Croft KD, Hodgson JM, Bondonno CP. Nitrate, the oral microbiome, and cardiovascular health: a systematic literature review of human and animal studies. Am J Clin Nutr 2018; 107:504-522. [PMID: 29635489 DOI: 10.1093/ajcn/nqx046] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/21/2017] [Indexed: 12/25/2022] Open
Abstract
Background Dietary nitrate is an important source of nitric oxide (NO), a molecule critical for cardiovascular health. Nitrate is sequentially reduced to NO through an enterosalivary nitrate-nitrite-NO pathway that involves the oral microbiome. This pathway is considered an important adjunct pathway to the classical l-arginine-NO synthase pathway. Objective The objective of this study was to systematically assess the evidence for dietary nitrate intake and improved cardiovascular health from both human and animal studies. Design A systematic literature search was performed according to PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines by using key search terms in Medline and EMBASE databases and defined inclusion and exclusion criteria. Results Thirty-seven articles on humans and 14 articles on animals were included from 12,541 screened references. Data on the effects of dietary nitrate on blood pressure, endothelial function, ischemic reperfusion injury, arterial stiffness, platelet function, and cerebral blood flow in both human and animal models were identified. Beneficial effects of nitrate on vascular health have predominantly been observed in healthy human populations, whereas effects in populations at risk of cardiovascular disease are less clear. Few studies have investigated the long-term effects of dietary nitrate on cardiovascular disease clinical endpoints. In animal studies, there is evidence that nitrate improves blood pressure and endothelial function, particularly in animal models with reduced NO bioavailability. Nitrate dose seems to be a critical factor because there is evidence of cross-talk between the 2 pathways of NO production. Conclusions Evidence for a beneficial effect in humans at risk of cardiovascular disease is limited. Furthermore, there is a need to investigate the long-term effects of dietary nitrate on cardiovascular disease clinical endpoints. Further animal studies are required to elucidate the mechanisms behind the observed effects.
Collapse
Affiliation(s)
- Lauren C Blekkenhorst
- Medical School, Royal Perth Hospital Unit, University of Western Australia, Perth, Western Australia, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Nicola P Bondonno
- Medical School, Royal Perth Hospital Unit, University of Western Australia, Perth, Western Australia, Australia
| | - Alex H Liu
- Medical School, Royal Perth Hospital Unit, University of Western Australia, Perth, Western Australia, Australia
| | - Natalie C Ward
- Medical School, Royal Perth Hospital Unit, University of Western Australia, Perth, Western Australia, Australia.,School of Biomedical Sciences and Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Richard L Prince
- Medical School, Queen Elizabeth Medical Center Unit, University of Western Australia, Nedlands, Western Australia, Australia
| | - Joshua R Lewis
- Medical School, Queen Elizabeth Medical Center Unit, University of Western Australia, Nedlands, Western Australia, Australia
| | - Amanda Devine
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Kevin D Croft
- Medical School, Royal Perth Hospital Unit, University of Western Australia, Perth, Western Australia, Australia
| | - Jonathan M Hodgson
- Medical School, Royal Perth Hospital Unit, University of Western Australia, Perth, Western Australia, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Catherine P Bondonno
- Medical School, Royal Perth Hospital Unit, University of Western Australia, Perth, Western Australia, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| |
Collapse
|
33
|
Jin R, Xiao AY, Song Z, Yu S, Li J, Cui MZ, Li G. Platelet CD40 Mediates Leukocyte Recruitment and Neointima Formation after Arterial Denudation Injury in Atherosclerosis-Prone Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:252-263. [PMID: 29037856 PMCID: PMC5745524 DOI: 10.1016/j.ajpath.2017.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 09/06/2017] [Accepted: 09/21/2017] [Indexed: 12/31/2022]
Abstract
The role of platelets in the development of thrombosis and abrupt closure after angioplasty is well recognized. However, the direct impact of platelets on neointima formation after arterial injury remains undetermined. Herein, we show that neointima formation after carotid artery wire injury reduces markedly in CD40-/- apolipoprotein E-deficient (apoE-/-) mice but only slightly in CD40 ligand-/-apoE-/- mice, compared with apoE-/- mice. Wild-type and CD40-deficient platelets were isolated from blood of apoE-/- and CD40-/-apoE-/- mice, respectively. The i.v. injection of thrombin-activated platelets into CD40-/-apoE-/- mice was performed every 5 days, starting at 2 days before wire injury. Injection of wild-type platelets promoted neointima formation, which was associated with increased inflammation by stimulating leukocyte recruitment via up-regulation of circulating platelet surface P-selectin expression and the formation of platelet-leukocyte aggregates. It was also associated with further promoting the luminal deposition of platelet-derived regulated on activation normal T cell expressed and secreted/chemokine (C-C motif) ligand 5 and expression of monocyte chemoattractant protein-1 and vascular cell adhesion molecule 1 in wire-injured carotid arteries. Remarkably, all these inflammatory actions by activated platelets were abrogated by lack of CD40 on injected platelets. Moreover, injection of wild-type platelets inhibited endothelial recovery in wire-injured carotid arteries, but this effect was also abrogated by lack of CD40 on injected platelets. Results suggest that platelet CD40 plays a pivotal role in neointima formation after arterial injury and might represent an attractive target to prevent restenosis after vascular interventions.
Collapse
Affiliation(s)
- Rong Jin
- Department of Neurosurgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana; Department of Neurosurgery, the Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Adam Y Xiao
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Zifang Song
- Department of Neurosurgery, the Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Shiyong Yu
- Department of Neurosurgery, the Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Jarvis Li
- Caddo Magnet High School, Shreveport, Louisiana
| | - Mei-Zhen Cui
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee
| | - Guohong Li
- Department of Neurosurgery, Louisiana State University Health Sciences Center, Shreveport, Louisiana; Department of Neurosurgery, the Pennsylvania State University College of Medicine, Hershey, Pennsylvania; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana.
| |
Collapse
|
34
|
Yao Y, Chen Y, Adili R, McKeown T, Chen P, Zhu G, Li D, Ling W, Ni H, Yang Y. Plant-based Food Cyanidin-3-Glucoside Modulates Human Platelet Glycoprotein VI Signaling and Inhibits Platelet Activation and Thrombus Formation. J Nutr 2017; 147:1917-1925. [PMID: 28855423 DOI: 10.3945/jn.116.245944] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 01/19/2017] [Accepted: 07/19/2017] [Indexed: 11/14/2022] Open
Abstract
Background: Platelets play an important role in hemostasis, thrombosis, and atherosclerosis. Glycoprotein VI (GPVI) is a major platelet receptor that interacts with exposed collagen on injured vessel walls. Our previous studies have shown that anthocyanins (a type of natural plant pigment) attenuate platelet function; however, whether anthocyanins affect collagen-induced GPVI signaling remains unknown.Objective: The objective of this study was to explore the effects of cyanidin-3-glucoside (Cy-3-g, one of the major bioactive compounds in anthocyanins) on platelet activation and thrombosis and the GPVI signaling pathway.Methods: Platelets from healthy men and women were isolated and incubated with different concentrations (0, 0.5, 5, and 50 μM) of Cy-3-g. The expression of activated integrin αIIbβ3, P-selectin, CD63, and CD40L, fibrinogen binding to platelets, and platelet aggregation were evaluated in vitro. Platelet adhesion and aggregation in whole blood under flow conditions were assessed in collagen-coated perfusion chambers. Thrombosis and hemostasis were assessed in 3-4-wk-old male C57BL/6J mice through FeCl3-induced intravital microscopy and tail bleeding time. The effect of Cy-3-g on collagen-induced human platelet GPVI signaling was explored with Western blot.Results: Cy-3-g attenuated platelet function in a dose-dependent manner. The 0.5-μM dose of Cy-3-g inhibited (P < 0.05) human platelet adhesion and aggregation to collagen at both venous (-54.02%) and arterial (-22.90%) shear stresses. The 5-μM dose inhibited (P < 0.05) collagen-induced human platelet activation (PAC-1: -48.21%, P-selectin: -50.63%), secretion (CD63: -73.89%, CD40L: -43.70%), fibrinogen binding (-56.79%), and aggregation (-17.81%). The 5-μM dose attenuated (P < 0.01) thrombus growth (-66.67%) without prolonging bleeding time in mice. The 50-μM dose downregulated (P < 0.05) collagen-induced GPVI signaling in human platelets and significantly decreased phosphorylation of Syk-linker for activation of T cells (LAT)-SLP76 (Syk: -39.08%, LAT: -32.25%, SLP76: -40.00%) and the expression of Lyn (-31.89%), Fyn (-36.27%), and phospholipase C-γ2 (-39.08%).Conclusions: Cy-3-g inhibits human platelet activation, aggregation, secretion, and thrombus formation, and downregulates the collagen-GPVI signaling pathway. Supplementation of Cy-3-g may have protective effects against atherothrombosis.
Collapse
Affiliation(s)
- Yanling Yao
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, People's Republic of China
| | - Yanqiu Chen
- Guangzhou Women and Children's Medical Centre, Guangzhou, People's Republic of China
| | - Reheman Adili
- Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Departments of
| | - Thomas McKeown
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Departments of
| | - Pingguo Chen
- Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Departments of
| | - Guangheng Zhu
- Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Departments of
| | - Dan Li
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, People's Republic of China
| | - Wenhua Ling
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, People's Republic of China
| | - Heyu Ni
- Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Ontario, Canada; .,Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Departments of.,Laboratory Medicine and Pathobiology.,Physiology, and.,Medicine, University of Toronto, Toronto, Ontario, Canada; and.,Canadian Blood Services, Toronto, Ontario, Canada
| | - Yan Yang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China; .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, People's Republic of China
| |
Collapse
|
35
|
Contursi A, Sacco A, Grande R, Dovizio M, Patrignani P. Platelets as crucial partners for tumor metastasis: from mechanistic aspects to pharmacological targeting. Cell Mol Life Sci 2017; 74:3491-3507. [PMID: 28488110 PMCID: PMC11107532 DOI: 10.1007/s00018-017-2536-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 05/02/2017] [Accepted: 05/03/2017] [Indexed: 02/08/2023]
Abstract
Platelets are anucleated cells that circulate in the blood as sentinels of tissue integrity. In fact, they are rich in a plethora of proteins and other factors stored in different granules which they selectively release upon stimulation. Moreover, platelets synthesize a vast number of lipids and release various types of vesicles, including exosomes which are rich in genetic material. Platelets possess a central function to interact with other cell types, including inflammatory cells and cancer cells. Recent findings have enlightened the capacity of platelets to induce changes in the phenotype of cancer cells which acquire invasiveness thus enhancing their metastatic potential. Thus, it has been hypothesized that targeting the platelet may represent a novel strategy to prevent the development and progression of cancer. This is supported by the efficacy of the antiplatelet agent low-dose aspirin. Studies are ongoing to verify whether other antiplatelet agents share the anticancer effectiveness of aspirin.
Collapse
Affiliation(s)
- Annalisa Contursi
- Section of Cardiovascular and Pharmacological Sciences, Department of Neuroscience, Imaging and Clinical Science, and CeSI-MeT (Centro Scienze dell' Invecchiamento e Medicina Traslazionale), "G. d'Annunzio" University, Via dei Vestini 31, 66100, Chieti, Italy
| | - Angela Sacco
- Section of Cardiovascular and Pharmacological Sciences, Department of Neuroscience, Imaging and Clinical Science, and CeSI-MeT (Centro Scienze dell' Invecchiamento e Medicina Traslazionale), "G. d'Annunzio" University, Via dei Vestini 31, 66100, Chieti, Italy
| | - Rosalia Grande
- Section of Cardiovascular and Pharmacological Sciences, Department of Neuroscience, Imaging and Clinical Science, and CeSI-MeT (Centro Scienze dell' Invecchiamento e Medicina Traslazionale), "G. d'Annunzio" University, Via dei Vestini 31, 66100, Chieti, Italy
| | - Melania Dovizio
- Section of Cardiovascular and Pharmacological Sciences, Department of Neuroscience, Imaging and Clinical Science, and CeSI-MeT (Centro Scienze dell' Invecchiamento e Medicina Traslazionale), "G. d'Annunzio" University, Via dei Vestini 31, 66100, Chieti, Italy
| | - Paola Patrignani
- Section of Cardiovascular and Pharmacological Sciences, Department of Neuroscience, Imaging and Clinical Science, and CeSI-MeT (Centro Scienze dell' Invecchiamento e Medicina Traslazionale), "G. d'Annunzio" University, Via dei Vestini 31, 66100, Chieti, Italy.
| |
Collapse
|
36
|
Twigg HL, Crystal R, Currier J, Ridker P, Berliner N, Kiem HP, Rutherford G, Zou S, Glynn S, Wong R, Peprah E, Engelgau M, Creazzo T, Colombini-Hatch S, Caler E. Refining Current Scientific Priorities and Identifying New Scientific Gaps in HIV-Related Heart, Lung, Blood, and Sleep Research. AIDS Res Hum Retroviruses 2017; 33:889-897. [PMID: 28530113 DOI: 10.1089/aid.2017.0026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The National Heart, Lung, and Blood Institute (NHLBI) AIDS Program's goal is to provide direction and support for research and training programs in areas of HIV-related heart, lung, blood, and sleep (HLBS) diseases. To better define NHLBI current HIV-related scientific priorities and with the goal of identifying new scientific priorities and gaps in HIV-related HLBS research, a wide group of investigators gathered for a scientific NHLBI HIV Working Group on December 14-15, 2015, in Bethesda, MD. The core objectives of the Working Group included discussions on: (1) HIV-related HLBS comorbidities in the antiretroviral era; (2) HIV cure; (3) HIV prevention; and (4) mechanisms to implement new scientific discoveries in an efficient and timely manner so as to have the most impact on people living with HIV. The 2015 Working Group represented an opportunity for the NHLBI to obtain expert advice on HIV/AIDS scientific priorities and approaches over the next decade.
Collapse
Affiliation(s)
- Homer L. Twigg
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Indiana University Medical Center, Indianapolis, Indiana
| | - Ronald Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Judith Currier
- Department of Medicine, University of California, Los Angeles, California
| | - Paul Ridker
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Nancy Berliner
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Hans-Peter Kiem
- Department of Medicine, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - George Rutherford
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California
| | - Shimian Zou
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Simone Glynn
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Renee Wong
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Emmanuel Peprah
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Michael Engelgau
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Tony Creazzo
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Sandra Colombini-Hatch
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Elisabet Caler
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
37
|
Quan W, Chen Z, Yang X, Li J, Li X, Weng Y, Li Y, Zhang X. Mean platelet volume/platelet count ratio as a predictor of 90-day outcome in large artery atherosclerosis stroke patients. Int J Neurosci 2017; 127:1019-1027. [PMID: 28270030 DOI: 10.1080/00207454.2017.1296438] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Weiwei Quan
- Department of Neurology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhibo Chen
- Department of Neurology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xuezhi Yang
- Department of Neurology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jia Li
- Department of Neurology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiang Li
- Department of Neurology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiyun Weng
- Department of Neurology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Youyu Li
- Department of Emergency Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xu Zhang
- Department of Neurology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
38
|
Lim B, Yao Y, Huang ALI, Yap ML, Flierl U, Palasubramaniam J, Zaldivia MT, Wang X, Peter K. A Unique Recombinant Fluoroprobe Targeting Activated Platelets Allows In Vivo Detection of Arterial Thrombosis and Pulmonary Embolism Using a Novel Three-Dimensional Fluorescence Emission Computed Tomography (FLECT) Technology. Am J Cancer Res 2017; 7:1047-1061. [PMID: 28435447 PMCID: PMC5399575 DOI: 10.7150/thno.18099] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/17/2017] [Indexed: 01/21/2023] Open
Abstract
Progress in pharmaceutical development is highly-dependent on preclinical in vivo animal studies. Small animal imaging is invaluable for the identification of new disease markers and the evaluation of drug efficacy. Here, we report for the first time the use of a three-dimensional fluorescence bioimager called FLuorescence Emission Computed Tomography (FLECT) for the detection of a novel recombinant fluoroprobe that is safe, easily prepared on a large scale and stably stored prior to scan. This novel fluoroprobe (Targ-Cy7) comprises a single-chain antibody-fragment (scFvTarg), which binds exclusively to activated-platelets, conjugated to a near-infrared (NIR) dye, Cy7, for detection. Upon mouse carotid artery injury, the injected fluoroprobe circulates and binds within the platelet-rich thrombus. This specific in vivo binding of the fluoroprobe to the thrombus, compared to its non-targeting control-fluoroprobe, is detected by the FLECT imager. The analyzed FLECT image quantifies the NIR signal and localizes it to the site of vascular injury. The detected fluorescence is further verified using a two-dimensional IVIS® Lumina scanner, where significant NIR fluorescence is detected in vivo at the thrombotic site, and ex vivo, at the injured carotid artery. Furthermore, fluorescence levels in various organs have also been quantified for biodistribution, with the highest fluoroprobe uptake shown to be in the injured artery. Subsequently, this live animal imaging technique is successfully employed to monitor the response of the induced thrombus to treatment over time. This demonstrates the potential of using longitudinal FLECT scanning to examine the efficacy of candidate drugs in preclinical settings. Besides intravascular thrombosis, we have shown that this non-invasive FLECT-imaging can also detect in vivo pulmonary embolism. Overall, this report describes a novel fluorescence-based preclinical imaging modality that uses an easy-to-prepare and non-radioactive recombinant fluoroprobe. This represents a unique tool to study mechanisms of thromboembolic diseases and it will strongly facilitate the in vivo testing of antithrombotic drugs. Furthermore, the non-radiation nature, low-cost, high sensitivity, and the rapid advancement of optical scanning technologies make this fluorescence imaging an attractive development for future clinical applications.
Collapse
|
39
|
Kim SJ, Jenne CN. Role of platelets in neutrophil extracellular trap (NET) production and tissue injury. Semin Immunol 2016; 28:546-554. [PMID: 27876233 DOI: 10.1016/j.smim.2016.10.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 10/07/2016] [Accepted: 10/12/2016] [Indexed: 12/13/2022]
Abstract
In addition to their well-known role as the cellular mediator of thrombosis, numerous studies have identified key roles for platelets during various disease processes. Importantly, platelets play a critical role in the host immune response, directly interacting with, and eliminating pathogens, from the blood stream. In addition to pathogen clearance, platelets also contribute to leukocyte recruitment at sites of infection and inflammation, and modulate leukocyte activity. Platelet interaction with activated neutrophils is a potent inducer of neutrophil extracellular trap (NET). NETs consist of a diffuse, sticky web of extracellular DNA, nuclear and granular proteins, and serve to ensnare and kill pathogens. In addition to catching pathogens, the cytotoxic molecules and proteases on NETs have the potential to inflict significant tissue damage. Additionally, NET components have been suggested to be key activators of infection-induced coagulopathy. These critical roles, at the interface between hemostasis and immunity, highlight the need for balance in the platelet response; too little platelet activity results in bleeding and immune deficit, too much leads to tissue pathogenesis. In this review, we highlight recent advances in our understanding of the role platelets play in inflammation, the link between platelets and NETs and the role platelets play in disease pathogenesis.
Collapse
Affiliation(s)
- Seok-Joo Kim
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, 3330 Hospital Drive NW, T2N 4N1, Calgary, AB, Canada
| | - Craig N Jenne
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, 3330 Hospital Drive NW, T2N 4N1, Calgary, AB, Canada.
| |
Collapse
|
40
|
Increased Platelet-leukocyte Aggregates Are Associated With Myocardial No-reflow in Patients With ST Elevation Myocardial Infarction. Am J Med Sci 2016; 352:261-6. [DOI: 10.1016/j.amjms.2016.05.034] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/08/2016] [Accepted: 05/17/2016] [Indexed: 12/15/2022]
|
41
|
Badimon L, Suades R, Fuentes E, Palomo I, Padró T. Role of Platelet-Derived Microvesicles As Crosstalk Mediators in Atherothrombosis and Future Pharmacology Targets: A Link between Inflammation, Atherosclerosis, and Thrombosis. Front Pharmacol 2016; 7:293. [PMID: 27630570 PMCID: PMC5005978 DOI: 10.3389/fphar.2016.00293] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/22/2016] [Indexed: 12/24/2022] Open
Abstract
Reports in the last decade have suggested that the role of platelets in atherosclerosis and its thrombotic complications may be mediated, in part, by local secretion of platelet-derived microvesicles (pMVs), small cell blebs released during the platelet activation process. MVs are the most abundant cell-derived microvesicle subtype in the circulation. High concentrations of circulating MVs have been reported in patients with atherosclerosis, acute vascular syndromes, and/or diabetes mellitus, suggesting a potential correlation between the quantity of microvesicles and the clinical severity of the atherosclerotic disease. pMVs are considered to be biomarkers of disease but new information indicates that pMVs are also involved in signaling functions. pMVs evoke or promote haemostatic and inflammatory responses, neovascularization, cell survival, and apoptosis, processes involved in the pathophysiology of cardiovascular disease. This review is focused on the complex cross-talk between platelet-derived microvesicles, inflammatory cells and vascular elements and their relevance in the development of the atherosclerotic disease and its clinical outcomes, providing an updated state-of-the art of pMV involvement in atherothrombosis and pMV potential use as therapeutic agent influencing cardiovascular biomedicine in the future.
Collapse
Affiliation(s)
- Lina Badimon
- Cardiovascular Research Center, Consejo Superior de Investigaciones Científicas - Institut Català de Ciències Cardiovasculars, Institut d'Investigació Biomèdica Sant Pau, Hospital Santa Creu i Sant PauBarcelona, Spain; Cardiovascular Research Chair, Universitat Autònoma de BarcelonaBarcelona, Spain
| | - Rosa Suades
- Cardiovascular Research Center, Consejo Superior de Investigaciones Científicas - Institut Català de Ciències Cardiovasculars, Institut d'Investigació Biomèdica Sant Pau, Hospital Santa Creu i Sant Pau Barcelona, Spain
| | - Eduardo Fuentes
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging, Universidad de TalcaTalca, Chile; Centro de Estudios en Alimentos Procesados, Conicyt-RegionalGore-Maule, Talca, Chile
| | - Iván Palomo
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging, Universidad de TalcaTalca, Chile; Centro de Estudios en Alimentos Procesados, Conicyt-RegionalGore-Maule, Talca, Chile
| | - Teresa Padró
- Cardiovascular Research Center, Consejo Superior de Investigaciones Científicas - Institut Català de Ciències Cardiovasculars, Institut d'Investigació Biomèdica Sant Pau, Hospital Santa Creu i Sant Pau Barcelona, Spain
| |
Collapse
|
42
|
Twarock S, Bagheri S, Bagheri S, Hohlfeld T. Platelet-vessel wall interactions and drug effects. Pharmacol Ther 2016; 167:74-84. [PMID: 27492900 DOI: 10.1016/j.pharmthera.2016.07.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/16/2016] [Indexed: 01/07/2023]
Affiliation(s)
- Sören Twarock
- Institut für Pharmakologie und Klinische Pharmakologie, Heinrich-Heine-Universität Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Saghar Bagheri
- Institut für Pharmakologie und Klinische Pharmakologie, Heinrich-Heine-Universität Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Sayeh Bagheri
- Institut für Pharmakologie und Klinische Pharmakologie, Heinrich-Heine-Universität Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Thomas Hohlfeld
- Institut für Pharmakologie und Klinische Pharmakologie, Heinrich-Heine-Universität Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany.
| |
Collapse
|
43
|
Alhawiti N, Burbury KL, Kwa FA, O'Malley CJ, Shuttleworth P, Alzard M, Hamadi A, Grigg AP, Jackson DE. The tyrosine kinase inhibitor, nilotinib potentiates a prothrombotic state. Thromb Res 2016; 145:54-64. [PMID: 27494773 DOI: 10.1016/j.thromres.2016.07.019] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 07/28/2016] [Accepted: 07/29/2016] [Indexed: 12/31/2022]
Abstract
Tyrosine kinase inhibitors (TKI) such as imatinib, nilotinib and dasatinib are now established as highly effective frontline therapies for chronic myeloid leukaemia (CML). Disease control is achieved in the majority of patients and survival is excellent such that recent focus has been on toxicities of these agents. Cumulative data have reported an excess of serious vascular complications, including arterial thrombosis and peripheral arterial occlusive disease, in patients receiving nilotinib in comparison with other TKIs, with resultant interest in delineating the pathophysiology and implications for rationale cardiovascular risk modification. To address this issue, we studied the effects of imatinib, nilotinib and dasatinib on platelet function and thrombus formation in human and mouse models using in vitro, ex vivo and in vivo approaches. In vitro studies demonstrated that dasatinib and imatinib but not nilotinib inhibited ADP, CRP, and collagen-induced platelet aggregation and moreover, that nilotinib potentiated PAR-1-mediated alpha granule release. Pretreatment of wild-type C57BL/6 mice with nilotinib but not imatinib or dasatinib, significantly increased thrombus growth and stability, on type I collagen under ex vivo arterial flow conditions and increased thrombus growth and stability following FeCl3-induced vascular injury of mesenteric arterioles and carotid artery injury in vivo. Whole blood from nilotinib-treated CML patients, demonstrated increased platelet adhesion ex vivo under flow, increased plasma soluble P- and E-selectin, sICAM-1, sVCAM-1, TNF-alpha, IL-6 levels and endogenous thrombin potential (ETP) levels in vivo, despite being on daily low-dose aspirin. These results demonstrate that nilotinib can potentiate platelet and endothelial activation and platelet thrombus formation ex vivo and in vivo.
Collapse
Affiliation(s)
- Naif Alhawiti
- Thrombosis and Vascular Diseases Laboratory, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Kate L Burbury
- Haematology Department, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Faith A Kwa
- Thrombosis and Vascular Diseases Laboratory, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Cindy J O'Malley
- Thrombosis and Vascular Diseases Laboratory, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Peter Shuttleworth
- Department of Clinical Haematology, Austin Health, Heidelberg, Victoria, Australia
| | - Mohamad Alzard
- Thrombosis and Vascular Diseases Laboratory, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Abdullah Hamadi
- Thrombosis and Vascular Diseases Laboratory, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Andrew P Grigg
- Department of Clinical Haematology, Austin Health, Heidelberg, Victoria, Australia
| | - Denise E Jackson
- Thrombosis and Vascular Diseases Laboratory, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia.
| |
Collapse
|
44
|
Ahmadi A, Stone GW, Leipsic J, Shaw LJ, Villines TC, Kern MJ, Hecht H, Erlinge D, Ben-Yehuda O, Maehara A, Arbustini E, Serruys P, Garcia-Garcia HM, Narula J. Prognostic Determinants of Coronary Atherosclerosis in Stable Ischemic Heart Disease. Circ Res 2016; 119:317-29. [DOI: 10.1161/circresaha.116.308952] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 06/10/2016] [Indexed: 01/10/2023]
Abstract
Risk stratification in patients with stable ischemic heart disease is essential to guide treatment decisions. In this regard, whether coronary anatomy, physiology, or plaque morphology is the best determinant of prognosis (and driver an effective therapeutic risk reduction) remains one of the greatest ongoing debates in cardiology. In the present report, we review the evidence for each of these characteristics and explore potential algorithms that may enable a practical diagnostic and therapeutic strategy for the management of patients with stable ischemic heart disease.
Collapse
Affiliation(s)
- Amir Ahmadi
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY (A.A., H.H., J.N.); Columbia University Medical Center, Cardiovascular Research Foundation, New York, NY (G.W.S., A.M.); University of British Columbia, Vancouver, British Columbia, Canada (A.A., J.L.); Emory University School of Medicine, Atlanta, GA (L.J.S.); Walter Reed National Military Medical Center, Bethesda, MD (T.C.V.); University of California Irvine (M.J.K.); Lund University, Sweden (D.E.); University of
| | - Gregg W. Stone
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY (A.A., H.H., J.N.); Columbia University Medical Center, Cardiovascular Research Foundation, New York, NY (G.W.S., A.M.); University of British Columbia, Vancouver, British Columbia, Canada (A.A., J.L.); Emory University School of Medicine, Atlanta, GA (L.J.S.); Walter Reed National Military Medical Center, Bethesda, MD (T.C.V.); University of California Irvine (M.J.K.); Lund University, Sweden (D.E.); University of
| | - Jonathon Leipsic
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY (A.A., H.H., J.N.); Columbia University Medical Center, Cardiovascular Research Foundation, New York, NY (G.W.S., A.M.); University of British Columbia, Vancouver, British Columbia, Canada (A.A., J.L.); Emory University School of Medicine, Atlanta, GA (L.J.S.); Walter Reed National Military Medical Center, Bethesda, MD (T.C.V.); University of California Irvine (M.J.K.); Lund University, Sweden (D.E.); University of
| | - Leslee J. Shaw
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY (A.A., H.H., J.N.); Columbia University Medical Center, Cardiovascular Research Foundation, New York, NY (G.W.S., A.M.); University of British Columbia, Vancouver, British Columbia, Canada (A.A., J.L.); Emory University School of Medicine, Atlanta, GA (L.J.S.); Walter Reed National Military Medical Center, Bethesda, MD (T.C.V.); University of California Irvine (M.J.K.); Lund University, Sweden (D.E.); University of
| | - Todd C. Villines
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY (A.A., H.H., J.N.); Columbia University Medical Center, Cardiovascular Research Foundation, New York, NY (G.W.S., A.M.); University of British Columbia, Vancouver, British Columbia, Canada (A.A., J.L.); Emory University School of Medicine, Atlanta, GA (L.J.S.); Walter Reed National Military Medical Center, Bethesda, MD (T.C.V.); University of California Irvine (M.J.K.); Lund University, Sweden (D.E.); University of
| | - Morton J. Kern
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY (A.A., H.H., J.N.); Columbia University Medical Center, Cardiovascular Research Foundation, New York, NY (G.W.S., A.M.); University of British Columbia, Vancouver, British Columbia, Canada (A.A., J.L.); Emory University School of Medicine, Atlanta, GA (L.J.S.); Walter Reed National Military Medical Center, Bethesda, MD (T.C.V.); University of California Irvine (M.J.K.); Lund University, Sweden (D.E.); University of
| | - Harvey Hecht
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY (A.A., H.H., J.N.); Columbia University Medical Center, Cardiovascular Research Foundation, New York, NY (G.W.S., A.M.); University of British Columbia, Vancouver, British Columbia, Canada (A.A., J.L.); Emory University School of Medicine, Atlanta, GA (L.J.S.); Walter Reed National Military Medical Center, Bethesda, MD (T.C.V.); University of California Irvine (M.J.K.); Lund University, Sweden (D.E.); University of
| | - David Erlinge
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY (A.A., H.H., J.N.); Columbia University Medical Center, Cardiovascular Research Foundation, New York, NY (G.W.S., A.M.); University of British Columbia, Vancouver, British Columbia, Canada (A.A., J.L.); Emory University School of Medicine, Atlanta, GA (L.J.S.); Walter Reed National Military Medical Center, Bethesda, MD (T.C.V.); University of California Irvine (M.J.K.); Lund University, Sweden (D.E.); University of
| | - Ori Ben-Yehuda
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY (A.A., H.H., J.N.); Columbia University Medical Center, Cardiovascular Research Foundation, New York, NY (G.W.S., A.M.); University of British Columbia, Vancouver, British Columbia, Canada (A.A., J.L.); Emory University School of Medicine, Atlanta, GA (L.J.S.); Walter Reed National Military Medical Center, Bethesda, MD (T.C.V.); University of California Irvine (M.J.K.); Lund University, Sweden (D.E.); University of
| | - Akiko Maehara
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY (A.A., H.H., J.N.); Columbia University Medical Center, Cardiovascular Research Foundation, New York, NY (G.W.S., A.M.); University of British Columbia, Vancouver, British Columbia, Canada (A.A., J.L.); Emory University School of Medicine, Atlanta, GA (L.J.S.); Walter Reed National Military Medical Center, Bethesda, MD (T.C.V.); University of California Irvine (M.J.K.); Lund University, Sweden (D.E.); University of
| | - Eloisa Arbustini
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY (A.A., H.H., J.N.); Columbia University Medical Center, Cardiovascular Research Foundation, New York, NY (G.W.S., A.M.); University of British Columbia, Vancouver, British Columbia, Canada (A.A., J.L.); Emory University School of Medicine, Atlanta, GA (L.J.S.); Walter Reed National Military Medical Center, Bethesda, MD (T.C.V.); University of California Irvine (M.J.K.); Lund University, Sweden (D.E.); University of
| | - Patrick Serruys
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY (A.A., H.H., J.N.); Columbia University Medical Center, Cardiovascular Research Foundation, New York, NY (G.W.S., A.M.); University of British Columbia, Vancouver, British Columbia, Canada (A.A., J.L.); Emory University School of Medicine, Atlanta, GA (L.J.S.); Walter Reed National Military Medical Center, Bethesda, MD (T.C.V.); University of California Irvine (M.J.K.); Lund University, Sweden (D.E.); University of
| | - Hector M. Garcia-Garcia
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY (A.A., H.H., J.N.); Columbia University Medical Center, Cardiovascular Research Foundation, New York, NY (G.W.S., A.M.); University of British Columbia, Vancouver, British Columbia, Canada (A.A., J.L.); Emory University School of Medicine, Atlanta, GA (L.J.S.); Walter Reed National Military Medical Center, Bethesda, MD (T.C.V.); University of California Irvine (M.J.K.); Lund University, Sweden (D.E.); University of
| | - Jagat Narula
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY (A.A., H.H., J.N.); Columbia University Medical Center, Cardiovascular Research Foundation, New York, NY (G.W.S., A.M.); University of British Columbia, Vancouver, British Columbia, Canada (A.A., J.L.); Emory University School of Medicine, Atlanta, GA (L.J.S.); Walter Reed National Military Medical Center, Bethesda, MD (T.C.V.); University of California Irvine (M.J.K.); Lund University, Sweden (D.E.); University of
| |
Collapse
|
45
|
Fuentes E, Rojas A, Palomo I. NF-κB signaling pathway as target for antiplatelet activity. Blood Rev 2016; 30:309-15. [PMID: 27075489 DOI: 10.1016/j.blre.2016.03.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 02/26/2016] [Accepted: 03/08/2016] [Indexed: 02/07/2023]
Abstract
In different nucleated cells, NF-κB has long been considered a prototypical proinflammatory signaling pathway with the expression of proinflammatory genes. Although platelets lack a nucleus, a number of functional transcription factors are involved in activated platelets, such as NF-κB. In platelet activation NF-κB regulation events include IKKβ phosphorylation, IκBα degradation, and p65 phosphorylation. Multiple pathways contribute to platelet activation and NF-κB is a common pathway in this activation. Therefore, in platelet activation the modulation of NF-κB pathway could be a potential new target in the treatment of inflammation-related vascular disease therapy (antiplatelet and antithrombotic activities).
Collapse
Affiliation(s)
- Eduardo Fuentes
- Laboratory of Hematology and Immunology, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001, Talca, Chile.
| | - Armando Rojas
- Biomedical Research Laboratories, Medicine Faculty, Catholic University of Maule, Talca, Chile
| | - Iván Palomo
- Laboratory of Hematology and Immunology, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001, Talca, Chile.
| |
Collapse
|
46
|
Platelet-to-White Blood Cell Ratio: A Prognostic Predictor for 90-Day Outcomes in Ischemic Stroke Patients with Intravenous Thrombolysis. J Stroke Cerebrovasc Dis 2016; 25:2430-8. [PMID: 27372596 DOI: 10.1016/j.jstrokecerebrovasdis.2016.06.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 05/16/2016] [Accepted: 06/08/2016] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND This study is aimed to investigate the relationship between platelet-to-white blood cell ratio (PWR) and 90-day outcomes in acute stroke patients with intravenous thrombolysis (IVT). MATERIALS AND METHODS A retrospective analysis was performed on 168 patients receiving IVT for acute ischemic stroke. Complete blood count evaluation was conducted at admission before IVT. A modified Rankin Scale (mRS) score of 3-6 at 90 days was considered an unfavorable outcome. RESULTS A total of 168 patients were included from 2013 to 2015. The mean age of the sample was 64.6 (±12.3) years, and 23.2% were women. The median baseline National Institutes of Health Stroke Scale score was 7.5 (interquartile range [IQR] 8.0) and the 90-day mRS score was 2 (IQR 2). In our multivariate logistic regression model, a PWR greater than 23.52 (odds ratio .454, 95% confidence interval: .212-.973, P < .050) was a predictor of 90-day outcomes. In addition, there was a significant difference in the PWR values of patients between favorable outcome and unfavorable outcome in the large-artery atherosclerosis subtype (28.241 ± 11.581 and 21.899 ± 9.107, respectively; P = .005). CONCLUSIONS The PWR at admission predicts 90-day outcomes in ischemic stroke patients with IVT. With the easy and routine use of hemogram analysis, the PWR should be investigated in further prospective randomized controlled trials of acute stroke.
Collapse
|
47
|
Effects of Endovascular Interventions on vWF and Fb Levels in Type 2 Diabetic Patients with Peripheral Artery Disease. Ann Vasc Surg 2016; 33:159-66. [DOI: 10.1016/j.avsg.2015.11.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 10/20/2015] [Accepted: 11/15/2015] [Indexed: 01/15/2023]
|
48
|
Fuentes E, Palomo I, Rojas A. Cross-talk between platelet and tumor microenvironment: Role of multiligand/RAGE axis in platelet activation. Blood Rev 2016; 30:213-21. [PMID: 26723842 DOI: 10.1016/j.blre.2015.11.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 10/02/2015] [Accepted: 11/30/2015] [Indexed: 02/07/2023]
Affiliation(s)
- Eduardo Fuentes
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule R09I2001, Talca, Chile.
| | - Iván Palomo
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule R09I2001, Talca, Chile
| | - Armando Rojas
- Biomedical Research Laboratories, Medicine Faculty, Catholic University of Maule, Talca, Chile.
| |
Collapse
|
49
|
Wu YW, Goubran H, Seghatchian J, Burnouf T. Smart blood cell and microvesicle-based Trojan horse drug delivery: Merging expertise in blood transfusion and biomedical engineering in the field of nanomedicine. Transfus Apher Sci 2016; 54:309-18. [PMID: 27179926 DOI: 10.1016/j.transci.2016.04.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Therapeutic and diagnostic applications of nanomedicine are playing increasingly important roles in human health. Various types of synthetic nanoparticles, including liposomes, micelles, and other nanotherapeutic platforms and conjugates, are being engineered to encapsulate or carry drugs for treating diseases such as cancer, cardiovascular disorders, neurodegeneration, and inflammations. Nanocarriers are designed to increase the half-life of drugs, decrease their toxicity and, ideally, target pathological sites. Developing smart carriers with the capacity to deliver drugs specifically to the microenvironment of diseased cells with minimum systemic toxicity is the goal. Blood cells, and potentially also the liposome-like micro- and nano-vesicles they generate, may be regarded as ideally suited to perform such specific targeting with minimum immunogenic risks. Blood cell membranes are "decorated" with complex physiological receptors capable of targeting and communicating with other cells and tissues and delivering their content to the surrounding pathological microenvironment. Blood cells, such as erythrocytes, have been developed as permeable carriers to release drugs to diseased tissues or act as biofactory allowing enzymatic degradation of a pathological substrate. Interestingly, attempts are also being made to improve the targeting capacity of synthetic nanoparticles by "decorating" their surface with blood cell membrane receptor-like biochemical structures. Research is needed to further explore the benefits that blood cell-derived microvesicles, as a Trojan horse delivery systems, can bring to the arsenal of therapeutic micro- and nanotechnologies. This short review focuses on the therapeutic roles that red blood cells and platelets can play as smart drug-delivery systems, and highlights the benefits that blood transfusion expertise can bring to this exciting and novel biomedical engineering field.
Collapse
Affiliation(s)
- Yu-Wen Wu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Hadi Goubran
- Saskatoon Cancer Centre and College of Medicine, University of Saskatchewan, Saskatoon, Canada.
| | - Jerard Seghatchian
- International Consultancy in Blood Components Quality/Safety Improvement, Audit/Inspection and DDR Strategies, London, UK.
| | - Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
50
|
Yayla Ç, Canpolat U, Akyel A, Yayla KG, Yilmaz S, Açikgöz SK, Özcan F, Turak O, Doğan M, Yeter E, Aydoğdu S. Association Between Platelet to Lymphocyte Ratio and Saphenous Vein Graft Disease. Angiology 2016; 67:133-138. [PMID: 25818104 DOI: 10.1177/0003319715578258] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Atherosclerosis plays an important role in saphenous vein graft disease (SVGD). Previous trials showed that inflammatory blood cells play a role in this process. The platelet to lymphocyte ratio (PLR) has been proposed as a novel predictor for cardiovascular risk and indicator of atherosclerosis. The aim of this study was to assess the relationship between SVGD and PLR. A total of 220 patients with SVG were enrolled (n = 87 with SVGD and n = 133 with patent SVG). A ≥ 50% stenosis within the SVG was defined as clinically significant. Median PLR (P < .001) and mean platelet volume (MPV; P = .043) were significantly higher in patients with SVGD. Also, PLR showed significantly positive correlation with age of SVG (P < .05). Median age of SVGs was also higher in the SVGD group (P = .025). In multivariate logistic regression analyses, the PLR and MPV were independent predictors of SVGD. Using a cutoff level of 106.3, the PLR predicted SVGD with a sensitivity of 87.4% and a specificity of 80.3%. To the best of our knowledge, this study showed, for the first time, that PLR was independently associated with SVGD. Both PLR and MPV might predict SVGD.
Collapse
Affiliation(s)
- Çağrı Yayla
- Cardiology Clinic, Türkiye Yüksek Ihtisas Training and Research Hospital, Ankara, Turkey
| | - Uğur Canpolat
- Cardiology Clinic, Türkiye Yüksek Ihtisas Training and Research Hospital, Ankara, Turkey
| | - Ahmet Akyel
- Cardiology Clinic, Dışkapı Yıldırım Beyazıt Training and Research Hospital, Ankara, Turkey
| | - Kadriye Gayretli Yayla
- Cardiology Clinic, Dışkapı Yıldırım Beyazıt Training and Research Hospital, Ankara, Turkey
| | - Samet Yilmaz
- Cardiology Clinic, Türkiye Yüksek Ihtisas Training and Research Hospital, Ankara, Turkey
| | - Sadık Kadri Açikgöz
- Cardiology Clinic, Türkiye Yüksek Ihtisas Training and Research Hospital, Ankara, Turkey
| | - Fırat Özcan
- Cardiology Clinic, Türkiye Yüksek Ihtisas Training and Research Hospital, Ankara, Turkey
| | - Osman Turak
- Cardiology Clinic, Türkiye Yüksek Ihtisas Training and Research Hospital, Ankara, Turkey
| | - Mehmet Doğan
- Cardiology Clinic, Dışkapı Yıldırım Beyazıt Training and Research Hospital, Ankara, Turkey
| | - Ekrem Yeter
- Cardiology Clinic, Dışkapı Yıldırım Beyazıt Training and Research Hospital, Ankara, Turkey
| | - Sinan Aydoğdu
- Cardiology Clinic, Türkiye Yüksek Ihtisas Training and Research Hospital, Ankara, Turkey
| |
Collapse
|