1
|
Jang J, Yu H, Oh EB, Park JW, Kim S, Kim T, Sohn J, Jin BR, Chang TS. Targeting NADPH Oxidase with APX-115: Suppression of Platelet Activation and Thrombotic Response. Antioxid Redox Signal 2025. [PMID: 40183134 DOI: 10.1089/ars.2024.0695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Aims: NADPH oxidase (NOX)-derived reactive oxygen species (ROS) are critical for platelet activation and thrombus formation. We hypothesized that inhibiting NOX-mediated ROS production with a pan-NOX inhibitor, APX-115, could effectively suppress platelet activation and thrombus formation, potentially serving as a novel antiplatelet therapeutic. This study aimed to explore the effects of APX-115 on human platelet functional responses and ROS-mediated signaling pathways. Results: APX-115 inhibited intracellular and extracellular ROS production in collagen-stimulated platelets, suppressing aggregation, P-selectin exposure, and ATP release. By preserving protein tyrosine phosphatase activity, APX-115 reduced tyrosine phosphorylation-dependent pathways inhibition, including spleen tyrosine kinase, LAT, Vav1, Bruton's tyrosine kinase, and phospholipase Cγ2, leading to decreased PKC activation and calcium mobilization. APX-115 also suppressed collagen-induced integrin αIIbβ3 activation, accompanied by elevated cGMP and vasodilator-stimulated phosphoprotein phosphorylation levels. In addition, APX-115 reduced p38 MAPK and ERK5 activation, leading to diminished phospholipase A2 phosphorylation, thromboxane production, and the exposure of procoagulant phosphatidylserine. These inhibitory effects extended to thrombus development caused by platelet adherence under shear and arterial thrombosis without prolonging bleeding time in murine models. Innovation: This study is the first to demonstrate that APX-115 inhibits NOX-mediated ROS production, platelet activation, and thrombus formation. By uncovering its effects on collagen receptor glycoprotein VI-mediated pathways, the work highlights the promise of APX-115 as an antiplatelet and antithrombotic agent. Conclusion: Our findings highlight the therapeutic potential of APX-115 in treating thrombotic and cardiovascular disorders by targeting NOX-mediated ROS production to mitigate platelet hyperreactivity and thrombus formation. Antioxid. Redox Signal. 00, 000-000. [Figure: see text].
Collapse
Affiliation(s)
- Joara Jang
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Hyunseong Yu
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Eun Bee Oh
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Ji Won Park
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Solee Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Taeryeong Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Jisue Sohn
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Bo-Ram Jin
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tong-Shin Chang
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
2
|
Beura SK, Panigrahi AR, Yadav P, Kulkarni PP, Lakhanpal V, Singh B, Singh SK. Role of Thrombosis in Neurodegenerative Diseases: An Intricate Mechanism of Neurovascular Complications. Mol Neurobiol 2025; 62:4802-4836. [PMID: 39482419 DOI: 10.1007/s12035-024-04589-4] [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: 04/11/2024] [Accepted: 10/23/2024] [Indexed: 11/03/2024]
Abstract
Thrombosis, the formation of blood clots in arteries or veins, poses a significant health risk by disrupting the blood flow. It can potentially lead to major cardiovascular complications such as acute myocardial infarction or ischemic stroke (arterial thrombosis) and deep vein thrombosis or pulmonary embolism (venous thrombosis). Nevertheless, over the course of several decades, researchers have observed an association between different cardiovascular events and neurodegenerative diseases, which progressively harm and impair parts of the nervous system, particularly the brain. Furthermore, thrombotic complications have been identified in numerous clinical instances of neurodegenerative diseases, particularly Alzheimer's disease, Parkinson's disease, multiple sclerosis, and Huntington's disease. Substantial research indicates that endothelial dysfunction, vascular inflammation, coagulation abnormalities, and platelet hyperactivation are commonly observed in these conditions, collectively contributing to an increased risk of thrombosis. Thrombosis can, in turn, contribute to the onset, pathogenesis, and severity of these neurological disorders. Hence, this concise review comprehensively explores the correlation between cardiovascular diseases and neurodegenerative diseases, elucidating the cellular and molecular mechanisms of thrombosis in these neurodegenerative diseases. Additionally, a detailed discussion is provided on the commonly employed antithrombotic medications in the context of these neuronal diseases.
Collapse
Affiliation(s)
- Samir Kumar Beura
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401
| | | | - Pooja Yadav
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401
| | - Paresh P Kulkarni
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Vikas Lakhanpal
- Department of Neurology, All India Institute of Medical Sciences, Bathinda, Punjab, India, 151001
| | - Bhupinder Singh
- Department of Cardiology, All India Institute of Medical Sciences, Bathinda, Punjab, India, 151001
| | - Sunil Kumar Singh
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401.
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401.
| |
Collapse
|
3
|
Zhao Z, Song H, Qi M, Liu Y, Zhang Y, Li S, Zhang H, Sun Y, Sun Y, Gao Z. Brain targeted polymeric micelles as drug carriers for ischaemic stroke treatment. J Drug Target 2025; 33:232-248. [PMID: 39403962 DOI: 10.1080/1061186x.2024.2417190] [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: 08/24/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
Ischaemic stroke is a central nervous system disease with high morbidity, recurrence and mortality rates. Thrombolytic and neuroprotective therapies are the main therapeutic strategies for ischaemic stroke, however, the poor delivery efficiency of thrombolytic and neuroprotective drugs to the brain limits their clinical application. So far, the development of nanomedicine has brought opportunities for the above challenges, which can not only realise the effective accumulation of drugs in the target site, but also improve the pharmacokinetic behaviour of the drugs. Among the most rapidly developing nanoparticles, micelles gradually emerging as an effective strategy for ischaemic stroke treatment due to their own unique advantages. This review provided an overview of targeted and response-release micelles based on the physicochemical properties of the ischaemic stroke microenvironment, summarised the targeting strategies for delivering micellar formulations to the thrombus, blood-brain barrier, and brain parenchyma, and finally described the potentials and challenges of polymeric micelles in the treatment of ischaemic stroke.
Collapse
Affiliation(s)
- Zirui Zhao
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Huijia Song
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Mengge Qi
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yurong Liu
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yanchao Zhang
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Shuo Li
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Huimin Zhang
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yongjun Sun
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yanping Sun
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Zibin Gao
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| |
Collapse
|
4
|
Marta-Enguita J, Machado FJD, Orbe J, Muñoz R. Thrombus composition and its implication in ischemic stroke assessment and revascularization treatments. Neurologia 2025; 40:77-88. [PMID: 39716574 DOI: 10.1016/j.nrleng.2024.12.001] [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: 07/19/2022] [Accepted: 11/01/2022] [Indexed: 12/25/2024] Open
Abstract
INTRODUCTION Since mechanical thrombectomy has allowed ischaemic stroke thrombus retrieval, the exhaustive study of this material has enabled better understanding of the potential physiopathological processes involved in thrombus formation. DEVELOPMENT Thrombotic pathways involved in the different vascular beds share common mechanisms, causing difficulties in the identification of specific patterns associated with stroke aetiology. However, other factors such as clot formation time, associated inflammatory status, or activation of additional immune and coagulation pathways (neutrophil extracellular trap [NET] delivery, platelet aggregation, endothelial activation, and von Willebrand Factor release) have been described as determinants in thrombus characteristics. Thus, variable proportions of fibrin-/platelet-rich and erythrocyte-rich areas are closely interrelated within the thrombus, frequently associated with a protective outer shell with high concentrations of fibrin, NETs, and von Willebrand Factor. The presence of these components, as well as their distribution and interrelationships, have been shown to have effects on the thrombus' resistance to revascularisation treatments. Understanding of these pathways has enabled the development of adjuvant therapies capable of enhancing current fibrinolytic drugs and/or increasing the efficacy of endovascular treatments. CONCLUSION Understanding of thrombus components and mechanisms involved in thrombus formation represent a potential pathway for the development of ischaemic stroke therapeutics with promising perspectives.
Collapse
Affiliation(s)
- Juan Marta-Enguita
- Servicio de Neurología, Hospital Universitario Navarra, Pamplona, Navarra, Spain; Servicio de Neurología, Hospital Universitario Donostia, San Sebastián, Guipúzcoa, Spain; Laboratorio Aterotrombosis, CIMA-Universidad de Navarra, IdiSNA, Pamplona, Navarra, Spain; RICORS-ICTUS, ISCIII, Madrid, Spain.
| | - Florencio J D Machado
- Laboratorio Aterotrombosis, CIMA-Universidad de Navarra, IdiSNA, Pamplona, Navarra, Spain
| | - Josune Orbe
- Laboratorio Aterotrombosis, CIMA-Universidad de Navarra, IdiSNA, Pamplona, Navarra, Spain; RICORS-ICTUS, ISCIII, Madrid, Spain
| | - Roberto Muñoz
- Servicio de Neurología, Hospital Universitario Navarra, Pamplona, Navarra, Spain; RICORS-ICTUS, ISCIII, Madrid, Spain
| |
Collapse
|
5
|
Upadhyay RK, Kumar K, Vishwakarma VK, Singh N, Narang R, Parakh N, Yadav M, Yadav S, Kumar S, Goyal A, Yadav HN. Delineating the NOX-Mediated Promising Therapeutic Strategies for the Management of Various Cardiovascular Disorders: A Comprehensive Review. Curr Vasc Pharmacol 2025; 23:12-30. [PMID: 39313896 DOI: 10.2174/0115701611308870240910115023] [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: 03/06/2024] [Revised: 04/21/2024] [Accepted: 08/22/2024] [Indexed: 09/25/2024]
Abstract
Cardiovascular disorders (CVDs) are reported to occur with very high rates of incidence and exhibit high morbidity and mortality rates across the globe. Therefore, research is focused on searching for novel therapeutic targets involving multiple pathophysiological mechanisms. Oxidative stress plays a critical role in the development and progression of various CVDs, such as hypertension, pulmonary hypertension, heart failure, arrhythmia, atherosclerosis, ischemia- reperfusion injury, and myocardial infarction. Among multiple pathways generating reactive oxygen species (ROS), Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases of the NOX family as the major source of ROS generation and plays an intricate role in the development and progression of CVDs. Therefore, exploring the role of different NADPH oxidase isoforms in various cardiovascular pathologies has attracted attention to current cardiovascular research. Focusing on NADPH oxidases to reduce oxidative stress in managing diverse CVDs may offer unique therapeutic approaches to prevent and treat various heart conditions. The current review article highlights the role of different NADPH oxidase isoforms in the pathophysiology of various CVDs. Moreover, the focus is also to emphasize different experimental studies that utilized various NADPH oxidase isoform modulators to manage other disorders. The present review article considers new avenues for researchers/scientists working in the field of cardiovascular pharmacology utilizing NADPH oxidase isoform modulators.
Collapse
Affiliation(s)
- Rohit Kumar Upadhyay
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Kuldeep Kumar
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | | | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, (Punjab)-147002-India
| | - Rajiv Narang
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Neeraj Parakh
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Mayank Yadav
- Department of CTVS, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sangeeta Yadav
- Department of Pharmacology, Dr. B.R. Ambedkar Centre for Biomedical Research, New Delhi, 110085-India
| | - Sachin Kumar
- Department of Medical Laboratory Technology, School of Allied Health Sciences, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi, 110017, India
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, 280406, India
| | - Harlokesh Narayan Yadav
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| |
Collapse
|
6
|
Chai X, Pan F, Wang Q, Wang X, Li X, Qi D, Yi Z, Liu H, Zhang J, Zhang Y, Pan Y, Liu Y, Wang G. Identification, screening, and comprehensive evaluation of novel thrombin inhibitory peptides from the hirudo produced using pepsin. Front Pharmacol 2024; 15:1460053. [PMID: 39640485 PMCID: PMC11617586 DOI: 10.3389/fphar.2024.1460053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/30/2024] [Indexed: 12/07/2024] Open
Abstract
Purpose The inhibition of thrombin has proven to be an efficacious therapeutic approach for managing cardiovascular disease (CVD), with widespread implementation in clinical settings. Oral ingestion of peptides and protein drugs is influenced by gastrointestinal digestive enzymes. We aimed to evaluate the thrombin inhibitory properties of hirudo hydrolysates (HHS) produced by pepsin and propose a comprehensive approach to screen and evaluate thrombin inhibitors. Methods We evaluated the in vitro inhibitory properties of the hirudo extract, both before and after hydrolysis with pepsin, toward thrombin. We screened for the most potent thrombin inhibitory peptide (TIP) using nano liquid chromatography-tandem mass spectrometry (Nano LC-MS/MS) coupled with in silico analysis. Next, we employed the thrombin inhibition activity IC50 to investigate the interaction between TIP and thrombin, and conducted in vitro evaluations of its anticoagulant effects (APTT, TT, PT), as well as its ability to inhibit platelet aggregation. Furthermore, we utilized UV-Vis spectroscopy to explore structural changes in thrombin upon binding with TIP and employed molecular dynamics simulations to delve deeper into the potential atomic-level interaction modes between thrombin and TIP. Results The retention rate of thrombin inhibition for HHS was found to be between 60% and 75%. A total of 90 peptides from the HHS were identified using LC-MS/MS combined with de novo sequencing. Asn-Asp-Leu-Trp-Asp-Gln-Gly-Leu-Val-Ser-Gln-Asp-Leu (NDLWDQGLVSQDL, P1) was identified as the most potent thrombin inhibitory peptide after in silico screening (molecular docking and ADMET). Then, the in vitro study revealed that P1 had a high inhibitory effect on thrombin (IC50: 2,425.5 ± 109.7 μM). P1 exhibited a dose-dependent prolongation of the thrombin time (TT) and a reduction in platelet aggregation rate. Both UV-Vis spectroscopy and molecular dynamics simulations demonstrated that P1 binds effectively to thrombin. Conclusion Overall, the results suggested that HHS provides new insights for searching and evaluating potential antithrombotic compounds. The obtained P1 can be structurally optimized for in-depth evaluation in animal and cellular experiments.
Collapse
Affiliation(s)
- Xiaoyu Chai
- Department of Chemistry of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Fulu Pan
- Department of Chemistry of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Qianqian Wang
- Department of Chemistry of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyu Wang
- Department of Chemistry of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xueyan Li
- Department of Chemistry of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Dongying Qi
- Department of Chemistry of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zirong Yi
- Department of Chemistry of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Huan Liu
- Department of Chemistry of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Zhang
- Institute of Information on Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yiming Zhang
- Institute of Information on Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanli Pan
- Institute of Information on Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yang Liu
- Department of Chemistry of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Guopeng Wang
- Zhongcai Health (Beijing) Biological Technology Development Co., Ltd., Beijing, China
| |
Collapse
|
7
|
Oh EB, Shin HJ, Yu H, Jang J, Park JW, Chang TS. NADPH oxidase 1/4 dual inhibitor setanaxib suppresses platelet activation and thrombus formation. Life Sci 2024; 357:123061. [PMID: 39293714 DOI: 10.1016/j.lfs.2024.123061] [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: 07/06/2024] [Revised: 09/07/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
AIMS The production of reactive oxygen species (ROS) by NADPH oxidase (NOX) is able to induce platelet activation, making NOX a promising target for antiplatelet therapy. In this study, we examined the effects of setanaxib, a dual NOX1/4 inhibitor, on human platelet function and ROS-related signaling pathways. MATERIALS AND METHODS In collagen-stimulated human platelets, aggregometry, assessment of ROS and Ca2+, immunoblotting, ELISA, flow cytometry, platelet adhesion assay, and assessment of mouse arterial thrombosis were performed in this study. KEY FINDINGS Setanaxib inhibited both intracellular and extracellular ROS production in collagen-activated platelets. Additionally, setanaxib significantly inhibited collagen-induced platelet aggregation, P-selectin exposure from α-granule release, and ATP release from dense granules. Setanaxib blocked the specific tyrosine phosphorylation-mediated activation of Syk, LAT, Vav1, and Btk within collagen receptor signaling pathways, leading to reduced activation of PLCγ2, PKC, and Ca2+ mobilization. Setanaxib also inhibited collagen-induced activation of integrin αIIbβ3, which is linked to increased cGMP levels and VASP phosphorylation. Furthermore, setanaxib suppressed collagen-induced p38 MAPK activation, resulting in decreased phosphorylation of cytosolic PLA2 and reduced TXA2 generation. Setanaxib also inhibited ERK5 activation, affecting the exposure of procoagulant phosphatidylserine. Setanaxib reduced thrombus formation under shear conditions by preventing platelet adhesion to collagen. Finally, in vivo administration of setanaxib in animal models led to the inhibition of arterial thrombosis. SIGNIFICANCE This study is the first to show that setanaxib suppresses ROS generation, platelet activation, and collagen-induced thrombus formation, suggesting its potential use in treating thrombotic or cardiovascular diseases.
Collapse
Affiliation(s)
- Eun Bee Oh
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| | - Hye Ji Shin
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| | - Hyunseong Yu
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| | - Joara Jang
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| | - Ji Won Park
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| | - Tong-Shin Chang
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
8
|
Kumar Beura S, Yadav P, Ramachandra Panigrahi A, Sahoo G, Kumar Singh S. Impact of 6-hydroxydopamine on agonist-induced human platelet functional parameters: An explanation for platelet impairment in Parkinson's disease. Neuroscience 2024; 559:237-248. [PMID: 39260561 DOI: 10.1016/j.neuroscience.2024.09.015] [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: 05/02/2024] [Revised: 08/30/2024] [Accepted: 09/05/2024] [Indexed: 09/13/2024]
Abstract
Parkinson's disease (PD) is the second-most prevalent neurodegenerative disease worldwide, which worsens with advancing age. It is a common movement disorder and is often associated with several vascular diseases with decreased stroke frequency. Circulating platelets substantially regulate vascular complications, including stroke, and share striking similarities with PD neurons. Although structural alterations in platelets are well-documented in PD, their functional parameters remain unclear. This study aimed to investigate the functional abnormalities in platelets associated with PD by evaluating key functional aspects such as adhesion, activation, secretion, aggregation, and clot retraction. To achieve this, we treated human blood platelets with 6-hydroxydopamine or 6-OHDA, that selectively destroys dopaminergic neurons, thereby creating an in vitro experimental model that closely resembles the pathogenic environment in PD, and examine its impact on platelet functions. In our study, platelet adhesion was assessed and further evaluated by a microplate reader, activation and secretion by a flow cytometer, aggregation by aggregometer, and clot retraction by Sonoclot. Phase-contrast and confocal microscopic studies further verified the results from the above experiments. Our findings showed that 6-OHDA treatment significantly inhibited thrombin (a platelet agonist)-induced functions, including adhesion, activation, aggregation, secretion, and clot retraction in human-washed platelets. In summary, this research provides pioneering evidence that 6-OHDA induces abnormal platelet functions, shedding light on the previously unexplored processes by which 6-OHDA affects platelet activity.
Collapse
Affiliation(s)
- Samir Kumar Beura
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | - Pooja Yadav
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | | | - Gaurahari Sahoo
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India
| | - Sunil Kumar Singh
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India; Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab 151401, India.
| |
Collapse
|
9
|
Guo J, He J, Xu S, Chen X, Zhu Z, Ji X, Wu D. Phosphatidylserine: A Novel Target for Ischemic Stroke Treatment. Biomolecules 2024; 14:1293. [PMID: 39456225 PMCID: PMC11506168 DOI: 10.3390/biom14101293] [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: 07/24/2024] [Revised: 09/12/2024] [Accepted: 09/22/2024] [Indexed: 10/28/2024] Open
Abstract
Over the past 40 years, research has heavily emphasized stroke treatments that directly target ischemic cascades after stroke onset. Much attention has focused on studying neuroprotective drugs targeting one aspect of the ischemic cascade. However, the single-target therapeutic approach resulted in minimal clinical benefit and poor outcomes in patients. Considering the ischemic cascade is a multifaceted and complex pathophysiological process with many interrelated pathways, the spotlight is now shifting towards the development of neuroprotective drugs that affect multiple aspects of the ischemic cascade. Phosphatidylserine (PS), known as the "eat-me" signal, is a promising candidate. PS is involved in many pathophysiological changes in the central nervous system after stroke onset, including apoptosis, inflammation, coagulation, and neuronal regeneration. Moreover, PS might also exert various roles in different phases after stroke onset. In this review, we describe the synthesis, regulation, and function of PS under physiological conditions. Furthermore, we also summarize the different roles of PS after stroke onset. More importantly, we also discuss several treatment strategies that target PS. We aim to advocate a novel stroke care strategy by targeting PS through a translational perspective.
Collapse
Affiliation(s)
- Jiaqi Guo
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China; (J.G.); (J.H.); (X.C.); (Z.Z.)
| | - Jiachen He
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China; (J.G.); (J.H.); (X.C.); (Z.Z.)
| | - Shuaili Xu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China;
| | - Xi Chen
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China; (J.G.); (J.H.); (X.C.); (Z.Z.)
| | - Zhanwei Zhu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China; (J.G.); (J.H.); (X.C.); (Z.Z.)
| | - Xunming Ji
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China; (J.G.); (J.H.); (X.C.); (Z.Z.)
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China;
| | - Di Wu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China; (J.G.); (J.H.); (X.C.); (Z.Z.)
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China;
| |
Collapse
|
10
|
Zeller-Hahn J, Bittl M, Kuhn S, Koessler A, Weber K, Koessler J, Kobsar A. Influence of short-term refrigeration on collagen-dependent signalling mechanisms in stored platelets. Cell Signal 2024; 122:111306. [PMID: 39048036 DOI: 10.1016/j.cellsig.2024.111306] [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: 05/29/2024] [Revised: 07/06/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
Platelet concentrates (PC) are used to treat patients with thrombocytopenia and hemorrhage, but there is still the demand to find the optimal strategy for temperature-dependent storage of PC. Recently, we could show that cold storage for 1 h (short-term refrigeration) is sufficient to induce enhanced platelet responsiveness. The aim of this study was to investigate effects of cold storage on collagen-dependent activating signalling pathways in platelets from apheresis-derived PC (APC). APC on day 1 or day 2 of storage, were either continuously kept at room temperature (RT, 22 °C), or for comparison, additionally kept at cold temperature (CT, 4 °C) for 1 h. CD62P expression was determined by flow cytometry. Western Blot technique was used to analyze collagen-induced phosphorylation of p38, ERK1/2 or Akt/PKB and its inhibition by prostaglandin E1 (PGE1) or nitric monoxide donor. Adhesion of platelets on collagen-coated surfaces and intracellular phosphorylation of vasodilator-stimulated phosphoprotein (VASP) was visualized by immune fluorescence microscopy. CD62P expression was increased after short-term refrigeration. CT exposition for 1 h induced an elevation of basal ERK1/2 phosphorylation and an alleviation of PGE1- or DEA/NO-suppressed ERK1/2 phosphorylation in APC on day 1 and 2 of storage. Similar, but more moderate effects were observable for p38 phosphorylation. Akt/PKB phosphorylation was increased only in APC on day 2. Refrigeration for 1 h promoted platelet adhesion and reduced basal VASP phosphorylation in adherent platelets. The attenuation of inhibitory signalling in short-term refrigerated stored platelets is associated with enhanced reactivity of activating signalling pathways, especially ERK1/2. Functionally, these processes correlate with increased adhesion of refrigerated platelets on collagen-coated surfaces. The results help to further optimize temperature-dependent strategies for platelet storage.
Collapse
Affiliation(s)
- Julia Zeller-Hahn
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Oberduerrbacher Straße 6, D-97080 Wuerzburg, Germany.
| | - Marius Bittl
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Oberduerrbacher Straße 6, D-97080 Wuerzburg, Germany.
| | - Sabine Kuhn
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Oberduerrbacher Straße 6, D-97080 Wuerzburg, Germany.
| | - Angela Koessler
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Oberduerrbacher Straße 6, D-97080 Wuerzburg, Germany.
| | - Katja Weber
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Oberduerrbacher Straße 6, D-97080 Wuerzburg, Germany.
| | - Juergen Koessler
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Oberduerrbacher Straße 6, D-97080 Wuerzburg, Germany.
| | - Anna Kobsar
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Oberduerrbacher Straße 6, D-97080 Wuerzburg, Germany.
| |
Collapse
|
11
|
Baxter RM, Harper MT. Dissecting the roles of dynamin and clathrin in platelet pinocytosis. Biochem Biophys Res Commun 2024; 725:150250. [PMID: 38870846 DOI: 10.1016/j.bbrc.2024.150250] [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: 05/16/2024] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024]
Abstract
Platelets endocytose many molecules from their environment. However, this process of pinocytosis in platelets is poorly understood. Key endocytic regulators such as dynamin, clathrin, CDC42 and Arf6 are expressed in platelets but their roles in pinocytosis is not known. Stimulated platelets form two subpopulations of pro-aggregatory and procoagulant platelets. The effect of stimulation on pinocytosis is also poorly understood. In this study, washed human platelets were treated with a range of endocytosis inhibitors and stimulated using different activators. The rate of pinocytosis was assessed using pHrodo green, a pH-sensitive 10 kDa dextran. In unstimulated platelets, pHrodo fluorescence increased over time and accumulated as intracellular puncta indicating constituently active pinocytosis. Stimulated platelets (both pro-aggregatory and procoagulant) had an elevated pinocytosis rate compared to unstimulated platelets. Dynamin inhibition blocked pinocytosis in unstimulated, pro-aggregatory and procoagulant platelets indicating that most platelet pinocytosis is dynamin dependent. Although pinocytosis was clathrin-independent in unstimulated and procoagulant populations, clathrin partially contributed to pinocytosis in pro-aggregatory platelets.
Collapse
Affiliation(s)
- Ruby M Baxter
- Department of Pharmacology, University of Cambridge, United Kingdom
| | - Matthew T Harper
- Department of Pharmacology, University of Cambridge, United Kingdom.
| |
Collapse
|
12
|
Safdar A, Wang P, Muhaymin A, Nie G, Li S. From bench to bedside: Platelet biomimetic nanoparticles as a promising carriers for personalized drug delivery. J Control Release 2024; 373:128-144. [PMID: 38977134 DOI: 10.1016/j.jconrel.2024.07.013] [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: 03/30/2024] [Revised: 06/24/2024] [Accepted: 07/05/2024] [Indexed: 07/10/2024]
Abstract
In recent decades, there has been a burgeoning interest in cell membrane coating strategies as innovative approach for targeted delivery systems in biomedical applications. Platelet membrane-coated nanoparticles (PNPs), in particular, are gaining interest as a new route for targeted therapy due to their advantages over conventional drug therapies. Their stepwise approach blends the capabilities of the natural platelet membrane (PM) with the adaptable nature of manufactured nanomaterials, resulting in a synergistic combination that enhances drug delivery and enables the development of innovative therapeutics. In this context, we present an overview of the latest advancements in designing PNPs with various structures tailored for precise drug delivery. Initially, we describe the types, preparation methods, delivery mechanisms, and specific advantages of PNPs. Next, we focus on three critical applications of PNPs in diseases: vascular disease therapy, cancer treatment, and management of infectious diseases. This review presents our knowledge of PNPs, summarizes their advancements in targeted therapies and discusses the promising potential for clinical translation of PNPs.
Collapse
Affiliation(s)
- Ammara Safdar
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Peina Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Department of Histology and Embryology, College of Basic Medical Sciences, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China.
| | - Abdul Muhaymin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| | - Suping Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| |
Collapse
|
13
|
Botros L, Qayyum R. Association of platelet distribution width with all-cause and cause-specific mortality in US adults. Int J Cardiol 2024; 407:132100. [PMID: 38663809 DOI: 10.1016/j.ijcard.2024.132100] [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: 12/18/2023] [Revised: 03/31/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND Platelet distribution width (PDW) indicates heterogeneity in circulating platelet sizes. Studies reporting PDW association with mortality were limited by small sample sizes. Therefore, we examined the relationship between PDW and all-cause and cause-specific mortality in a large representative cohort. METHODS The NHANES III data were linked to mortality files to examine the association between PDW and mortality. We excluded participants <18 years old and had a history of myocardial infarction. Since the hazards violated the proportionality assumption, we used piece-wise spline with 5-year time intervals in Cox models without and with adjustment for age, gender, race, smoking history, diabetes mellitus, hypertension, eGFR and total cholesterol. RESULTS Of 15,688 participants, 53.2% were females, 36.2% had a history of hypertension, and 6368(40.6%) died during follow-up (range 0 to 31 years). The mean (SD) age of the participants was 47(20) years, platelet count was 275.0(71.7) 109/L, and PDW 16.5(0.5). In multivariable analyses, PDW was associated with all-cause mortality at 0-5 years (HR = 1.44; 95%CI = 1.21, 1.72; P < 0.001) and at 5-10 years (HR = 1.23; 95%CI =1.03, 1.46; P = 0.02). Similarly, PDW association was significant for the first 0-5 years in cardiovascular mortality (HR = 1.58, 95%CI = 1.10, 2.25; P = 0.013) and for cancer mortality (HR = 1.48 (1.15, 95%CI = 1.15, 1.91, P = 0.003). For other-cause mortality, PDW remained significantly associated for 0-5 years (HR = 1.35, 95%CI =1.05, 1.74; P = 0.02) and for 5-10 years (HR = 1.38, 95%CI = 1.05, 1.83; P = 0.023). CONCLUSIONS PDW is an independent, but time-dependent, predictor of all-cause, cardiovascular, cancer and other-cause mortality up to 5 years. The mechanisms underlying this association need further study.
Collapse
Affiliation(s)
- Liza Botros
- Department of Medicine, Eastern Virginia Medical School, Norfolk, USA; Department of Pulmonary Disease, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Rehan Qayyum
- Department of Medicine, Eastern Virginia Medical School, Norfolk, USA.
| |
Collapse
|
14
|
Nappi F. P2Y12 Receptor Inhibitor for Antiaggregant Therapies: From Molecular Pathway to Clinical Application. Int J Mol Sci 2024; 25:7575. [PMID: 39062819 PMCID: PMC11277343 DOI: 10.3390/ijms25147575] [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: 06/09/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Platelets play a significant role in hemostasis, forming plugs at sites of vascular injury to limit blood loss. However, if platelet activation is not controlled, it can lead to thrombotic events, such as myocardial infarction and stroke. To prevent this, antiplatelet agents are used in clinical settings to limit platelet activation in patients at risk of arterial thrombotic events. However, their use can be associated with a significant risk of bleeding. An enhanced comprehension of platelet signaling mechanisms should facilitate the identification of safer targets for antiplatelet therapy. Over the past decade, our comprehension of the breadth and intricacy of signaling pathways that orchestrate platelet activation has expanded exponentially. Several recent studies have provided further insight into the regulation of platelet signaling events and identified novel targets against which to develop novel antiplatelet agents. Antiplatelet drugs are essential in managing atherothrombotic vascular disease. The current antiplatelet therapy in clinical practice is limited in terms of safety and efficacy. Novel compounds have been developed in response to patient variability and resistance to aspirin and/or clopidogrel. Recent studies based on randomized controlled trials and systematic reviews have definitively demonstrated the role of antiplatelet therapy in reducing the risk of cardiovascular events. Antiplatelet therapy is the recommended course of action for patients with established atherosclerosis. These studies compared monotherapy with a P2Y12 inhibitor versus aspirin for secondary prevention. However, in patients undergoing percutaneous coronary intervention, it is still unclear whether the efficacy of P2Y12 inhibitor monotherapy after a short course of dual antiplatelet therapy depends on the type of P2Y12 inhibitor. This paper focuses on the advanced-stage evaluation of several promising antiplatelet drugs.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| |
Collapse
|
15
|
Martinez-Vargas M, Courson J, Gardea L, Sen M, Yee A, Rumbaut R, Cruz MA. The impact of von Willebrand factor on fibrin formation and structure unveiled with type 3 von Willebrand disease plasma. Blood Coagul Fibrinolysis 2024; 35:256-264. [PMID: 38973517 PMCID: PMC11232922 DOI: 10.1097/mbc.0000000000001309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
Normally, von Willebrand factor (VWF) remains inactive unless its A1A2 domains undergo a shear stress-triggered conformational change. We demonstrated the capacity of a recombinant A2 domain of VWF to bind and to affect fibrin formation, altering the fibrin clot structure. The data indicated that VWF contains an additional binding site for fibrin in the A2 domain that plays a role in the incorporation of VWF to the polymerizing fibrin. This study is to examine the hypothesis that active plasma VWF directly influence fibrin polymerization and the structure of fibrin clots. The study used healthy and type 3 von Willebrand disease (VWD) plasma, purified plasma VWF, fibrin polymerization assays, confocal microscopy and scanning electron microscopy. The exposed A2 domain in active VWF harbors additional binding sites for fibrinogen, and significantly potentiates fibrin formation (P < 0.02). Antibody against the A2 domain of VWF significantly decreased the initial rate of change of fibrin formation (P < 0.002). Clot analyses revealed a significant difference in porosity between normal and type 3 VWD plasma (P < 0.008), further supported by scanning electron microscopy, which demonstrated thicker fibrin fibers in the presence of plasma VWF (P < 0.0003). Confocal immunofluorescence microscopy showed punctate VWF staining along fibrin fibrils, providing visual evidence of the integration of plasma VWF into the fibrin matrix. The study with type 3 VWD plasma supports the hypothesis that plasma VWF directly influences fibrin polymerization and clot structure. In addition, a conformational change in the A1A2 domains exposes a hidden fibrin(ogen) binding site, indicating that plasma VWF determines the fibrin clot structure.
Collapse
Affiliation(s)
- Marina Martinez-Vargas
- Section of Cardiovascular Research
- Department of Medicine
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center
| | - Justin Courson
- Department of Medicine
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center
| | - Luis Gardea
- Department of Pediatrics, Baylor College of Medicine
| | - Mehmet Sen
- University of Houston, Houston, Texas, USA
| | - Andrew Yee
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center
- Department of Pediatrics, Baylor College of Medicine
| | - Rolando Rumbaut
- Department of Medicine
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center
| | - Miguel A Cruz
- Section of Cardiovascular Research
- Department of Medicine
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center
| |
Collapse
|
16
|
Pretorius E, Kell DB. A Perspective on How Fibrinaloid Microclots and Platelet Pathology May be Applied in Clinical Investigations. Semin Thromb Hemost 2024; 50:537-551. [PMID: 37748515 PMCID: PMC11105946 DOI: 10.1055/s-0043-1774796] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Microscopy imaging has enabled us to establish the presence of fibrin(ogen) amyloid (fibrinaloid) microclots in a range of chronic, inflammatory diseases. Microclots may also be induced by a variety of purified substances, often at very low concentrations. These molecules include bacterial inflammagens, serum amyloid A, and the S1 spike protein of severe acute respiratory syndrome coronavirus 2. Here, we explore which of the properties of these microclots might be used to contribute to differential clinical diagnoses and prognoses of the various diseases with which they may be associated. Such properties include distributions in their size and number before and after the addition of exogenous thrombin, their spectral properties, the diameter of the fibers of which they are made, their resistance to proteolysis by various proteases, their cross-seeding ability, and the concentration dependence of their ability to bind small molecules including fluorogenic amyloid stains. Measuring these microclot parameters, together with microscopy imaging itself, along with methodologies like proteomics and imaging flow cytometry, as well as more conventional assays such as those for cytokines, might open up the possibility of a much finer use of these microclot properties in generative methods for a future where personalized medicine will be standard procedures in all clotting pathology disease diagnoses.
Collapse
Affiliation(s)
- Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Matieland, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Douglas B. Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Matieland, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
17
|
Jiang H, Nechipurenko DY, Panteleev MA, Xu K, Qiao J. Redox regulation of platelet function and thrombosis. J Thromb Haemost 2024; 22:1550-1557. [PMID: 38460839 DOI: 10.1016/j.jtha.2024.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/15/2024] [Accepted: 02/24/2024] [Indexed: 03/11/2024]
Abstract
Platelets are well-known players in several cardiovascular diseases such as atherosclerosis and venous thrombosis. There is increasing evidence demonstrating that reactive oxygen species (ROS) are generated within activated platelets. Nicotinamide adenine dinucleotide phosphate oxidase (NOX) is a major source of ROS generation in platelets. Ligand binding to platelet receptor glycoprotein (GP) VI stimulates intracellular ROS generation consisting of a spleen tyrosine kinase-independent production involving NOX activation and a following spleen tyrosine kinase-dependent generation. In addition to GPVI, stimulation of platelet thrombin receptors (protease-activated receptors [PARs]) can also trigger NOX-derived ROS production. Our recent study found that mitochondria-derived ROS production can be induced by engagement of thrombin receptors but not by GPVI, indicating that mitochondria are another source of PAR-dependent ROS generation apart from NOX. However, mitochondria are not involved in GPVI-dependent ROS generation. Once generated, the intracellular ROS are also involved in modulating platelet function and thrombus formation; therefore, the site-specific targeting of ROS production or clearance of excess ROS within platelets is a potential intervention and treatment option for thrombotic events. In this review, we will summarize the signaling pathways involving regulation of platelet ROS production and their role in platelet function and thrombosis, with a focus on GPVI- and PAR-dependent platelet responses.
Collapse
Affiliation(s)
- Huimin Jiang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Dmitry Yu Nechipurenko
- Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia; Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Science, Moscow, Russia; Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Mikhail A Panteleev
- Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia; Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Science, Moscow, Russia; Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.
| |
Collapse
|
18
|
Nicolai L, Pekayvaz K, Massberg S. Platelets: Orchestrators of immunity in host defense and beyond. Immunity 2024; 57:957-972. [PMID: 38749398 DOI: 10.1016/j.immuni.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 04/06/2024] [Accepted: 04/12/2024] [Indexed: 06/05/2024]
Abstract
Platelets prevent blood loss during vascular injury and contribute to thrombus formation in cardiovascular disease. Beyond these classical roles, platelets are critical for the host immune response. They guard the vasculature against pathogens via specialized receptors, intracellular signaling cascades, and effector functions. Platelets also skew inflammatory responses by instructing innate immune cells, support adaptive immunosurveillance, and influence antibody production and T cell polarization. Concomitantly, platelets contribute to tissue reconstitution and maintain vascular function after inflammatory challenges. However, dysregulated activation of these multitalented cells exacerbates immunopathology with ensuing microvascular clotting, excessive inflammation, and elevated risk of macrovascular thrombosis. This dichotomy underscores the critical importance of precisely defining and potentially modulating platelet function in immunity.
Collapse
Affiliation(s)
- Leo Nicolai
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| | - Kami Pekayvaz
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
19
|
Song C, Zhang L. Intelligent Design of Antithrombotic Peptide Targeting Collagen. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:9661-9668. [PMID: 38664943 DOI: 10.1021/acs.langmuir.4c00543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Binding of blood components to collagen was proved to be a key step in thrombus formation. Intelligent Design of Protein Matcher (IDProMat), a neural network model, was then developed based on the principle of seq2seq to design an antithrombotic peptide targeting collagen. The encoding and decoding of peptide sequence data and the interaction patterns of peptide chains at the interface were studied, and then, IDProMat was applied to the design of peptides to cover collagen. The 99.3% decrease in seq2seq loss and 58.3% decrease in MLP loss demonstrated that IDProMat learned the interaction patterns between residues at the binding interface. An efficient peptide, LRWNSYY, was then designed using this model. Validations on its binding on collagen and its inhibition of platelet adhesion were obtained using docking, MD simulations, and experimental approaches.
Collapse
Affiliation(s)
- Changwei Song
- Department of Biochemical Engineering and Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, People's Republic of China
| | - Lin Zhang
- Department of Biochemical Engineering and Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, People's Republic of China
| |
Collapse
|
20
|
Wang A, Yue K, Yan X, Zhong W, Zhang G, Wang L, Zhang H, Zhang X. Inhibition of platelet adhesion to exposed subendothelial collagen by steric hindrance with blocking peptide nanoparticles. Colloids Surf B Biointerfaces 2024; 237:113866. [PMID: 38520952 DOI: 10.1016/j.colsurfb.2024.113866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/04/2024] [Accepted: 03/19/2024] [Indexed: 03/25/2024]
Abstract
The inhibition of platelet adhesion to collagen in exposed vessels represents an innovative approach to the treatment of atherosclerosis and thrombosis. This study aimed to engineer peptide-based nanoparticles that prevent platelet binding to subendothelial collagen by engaging with collagen with high affinity. We examined the interactions between integrin α2/ glycoprotein VI/ von Willebrand factor A3 domain and collagen, as well as between the synthesized peptide nanoparticles and collagen, utilizing molecular dynamics simulations and empirical assays. Our findings indicated that the bond between von Willebrand factor and collagen was more robust. Specifically, the sequences SITTIDV, VDVMQRE, and YLTSEMH in von Willebrand factor were identified as essential for its attachment to collagen. Based on these sequences, three peptide nanoparticles were synthesized (BPa: Capric-GNNQQNYK-SITTIDV, BPb: Capric-GNNQQNYK-VDVMQRE, BPc: Capric-GNNQQNYK-YLTSEMH), each displaying significant affinity towards collagen. Of these, the BPa nanoparticles exhibited the most potent interaction with collagen, leading to a 75% reduction in platelet adhesion.
Collapse
Affiliation(s)
- Anqi Wang
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Kai Yue
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China; Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province 528399, China.
| | - Xiaotong Yan
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Weishen Zhong
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Genpei Zhang
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Lei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
| | - Hua Zhang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xinxin Zhang
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China; Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province 528399, China
| |
Collapse
|
21
|
Fang L, Hu Y, Lin Z, Ren Y, Liu X, Gong J. Instant mucus dressing of PEO reinforced by chitosan nanofiber scaffold for open wound healing. Int J Biol Macromol 2024; 263:130512. [PMID: 38423418 DOI: 10.1016/j.ijbiomac.2024.130512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024]
Abstract
Dressings seamlessly attached to the open wound bed are necessary for fully unleashing the dressing healing ability, as leaving the voids beneath the dressing poses infection hazards. The present study prepared an instant mucus dressing (IMD) of polyethylene oxide (PEO) reinforced by chitosan (CS) nanofiber scaffold, which formed by immersing PEO/CS nanofiber mat in water. The PEO/CS nanofiber mat were fabricated by the solution blow spinning (SBS) method using PEO and CS mixed solutions. Attenuated total reflection Fourier transform infrared spectroscopy (FTIR-ATR), transmission electron microscope (TEM), X-ray photoelectron spectroscopy (XPS), X-ray diffraction (XRD) and differential scan calorimetry (DSC) analyses indicate that PEO macromolecules formed the most of nanofiber shell due to their lower surface tension while CS macromolecules stayed mainly inside the fiber as the core. When such a PEO/CS nanofiber mat was immersed in water, PEO swelled to form mucus dressing reinforced by CS inside the nanofiber, which was fluidic and able to fully fill the voids on the wound. In vivo rat experiment verified that the dressing significantly accelerated the open wound healing through seamlessly attaching of mucus to the open wound and providing moist environment. The dressings exhibit good platelets and whole blood cells adhesion properties, excellent hemostasis function and no cytotoxicity. This instant mucus dressing provided a new perspective for manufacturing high performance open wound dressings.
Collapse
Affiliation(s)
- Lei Fang
- School of Textile Science and Engineering, Tiangong University, 399 Binshui West Road, Tianjin 300387, China.
| | - Yanling Hu
- Traumatology Department, Affiliated Hospital of Qingdao University, 59 Haier Road, Qingdao 266071, China.
| | - Zhihao Lin
- Traumatology Department, Affiliated Hospital of Qingdao University, 59 Haier Road, Qingdao 266071, China
| | - Yanfei Ren
- College of Textiles & Clothing, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Xiuming Liu
- School of Textile Science and Engineering, Tiangong University, 399 Binshui West Road, Tianjin 300387, China.
| | - Jixian Gong
- School of Textile Science and Engineering, Tiangong University, 399 Binshui West Road, Tianjin 300387, China.
| |
Collapse
|
22
|
Amadio P, Sandrini L, Zarà M, Barbieri SS, Ieraci A. NADPH-oxidases as potential pharmacological targets for thrombosis and depression comorbidity. Redox Biol 2024; 70:103060. [PMID: 38310682 PMCID: PMC10848036 DOI: 10.1016/j.redox.2024.103060] [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: 12/08/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/06/2024] Open
Abstract
There is a complex interrelationship between the nervous system and the cardiovascular system. Comorbidities of cardiovascular diseases (CVD) with mental disorders, and vice versa, are prevalent. Adults with mental disorders such as anxiety and depression have a higher risk of developing CVD, and people with CVD have an increased risk of being diagnosed with mental disorders. Oxidative stress is one of the many pathways associated with the pathophysiology of brain and cardiovascular disease. Nicotinamide adenine dinucleotide phosphate oxidase (NOX) is one of the major generators of reactive oxygen species (ROS) in mammalian cells, as it is the enzyme that specifically produces superoxide. This review summarizes recent findings on the consequences of NOX activation in thrombosis and depression. It also discusses the therapeutic effects and pharmacological strategies of NOX inhibitors in CVD and brain disorders. A better comprehension of these processes could facilitate the development of new therapeutic approaches for the prevention and treatment of the comorbidity of thrombosis and depression.
Collapse
Affiliation(s)
- Patrizia Amadio
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| | - Leonardo Sandrini
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| | - Marta Zarà
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| | - Silvia S Barbieri
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy.
| | - Alessandro Ieraci
- Department of Theoretical and Applied Sciences, eCampus University, 22060, Novedrate (CO), Italy; Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy.
| |
Collapse
|
23
|
Boncler M, Bartczak K, Rozalski M. Potential for modulation of platelet function via adenosine receptors during inflammation. Br J Pharmacol 2024; 181:547-563. [PMID: 37218380 DOI: 10.1111/bph.16146] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/15/2023] [Accepted: 05/15/2023] [Indexed: 05/24/2023] Open
Abstract
Traditionally, platelets are known to play an important role in haemostasis and thrombosis; however, they serve also as important modulators of inflammation and immunity. Platelets secrete adhesion molecules and cytokines, interact with leukocytes and endothelium, and express toll-like receptors involved in a direct interaction with pathogens. Platelets express A2A and A2B subtypes of receptors for adenosine. The activation of these receptors leads to an increase in cAMP concentration in the cytoplasm, thereby resulting in inhibited secretion of pro-inflammatory mediators and reduced cell activation. Therefore, platelet adenosine receptors could be a potential target for inhibiting platelet activation and thus down-regulating inflammation or immunity. The biological effects of adenosine are short-lasting, because the compound is rapidly metabolized; hence, its lability has triggered efforts to synthesize new, longer-lasting adenosine analogues. In this article, we have reviewed the literature regarding the pharmacological potential of adenosine and other agonists of A2A and A2B receptors to affect platelet function during inflammation. LINKED ARTICLES: This article is part of a themed issue on Platelet purinergic receptor and non-thrombotic disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.4/issuetoc.
Collapse
Affiliation(s)
- Magdalena Boncler
- Department of Haemostasis and Haemostatic Disorders, Chair of Biomedical Sciences, Medical University of Lodz, Lodz, Poland
| | - Kinga Bartczak
- Department of Haemostasis and Haemostatic Disorders, Chair of Biomedical Sciences, Medical University of Lodz, Lodz, Poland
| | - Marcin Rozalski
- Department of Haemostasis and Haemostatic Disorders, Chair of Biomedical Sciences, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
24
|
Drysdale A, Blanco-Lopez M, White SJ, Unsworth AJ, Jones S. Differential Proteoglycan Expression in Atherosclerosis Alters Platelet Adhesion and Activation. Int J Mol Sci 2024; 25:950. [PMID: 38256024 PMCID: PMC10815981 DOI: 10.3390/ijms25020950] [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: 12/07/2023] [Revised: 01/06/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Proteoglycans are differentially expressed in different atherosclerotic plaque phenotypes, with biglycan and decorin characteristic of ruptured plaques and versican and hyaluronan more prominent in eroded plaques. Following plaque disruption, the exposure of extracellular matrix (ECM) proteins triggers platelet adhesion and thrombus formation. In this study, the impact of differential plaque composition on platelet function and thrombus formation was investigated. Platelet adhesion, activation and thrombus formation under different shear stress conditions were assessed in response to individual proteoglycans and composites representing different plaque phenotypes. The results demonstrated that all the proteoglycans tested mediated platelet adhesion but not platelet activation, and the extent of adhesion observed was significantly lower than that observed with type I and type III collagens. Thrombus formation upon the rupture and erosion ECM composites was significantly reduced (p < 0.05) compared to relevant collagen alone, indicating that proteoglycans negatively regulate platelet collagen responses. This was supported by results demonstrating that the addition of soluble biglycan or decorin to whole blood markedly reduced thrombus formation on type I collagen (p < 0.05). Interestingly, thrombus formation upon the erosion composite displayed aspirin sensitivity, whereas the rupture composite was intensive to aspirin, having implications for current antiplatelet therapy regimes. In conclusion, differential platelet responses and antiplatelet efficacy are observed on ECM composites phenotypic of plaque rupture and erosion. Proteoglycans inhibit thrombus formation and may offer a novel plaque-specific approach to limit arterial thrombosis.
Collapse
Affiliation(s)
- Amelia Drysdale
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (M.B.-L.); (A.J.U.)
| | - Maria Blanco-Lopez
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (M.B.-L.); (A.J.U.)
| | - Stephen J. White
- Faculty of Medical Sciences, The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK;
| | - Amanda J. Unsworth
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (M.B.-L.); (A.J.U.)
| | - Sarah Jones
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (M.B.-L.); (A.J.U.)
| |
Collapse
|
25
|
Choi JH, Kim K. Polyhexamethylene Guanidine Phosphate Enhanced Procoagulant Activity through Oxidative-Stress-Mediated Phosphatidylserine Exposure in Platelets. TOXICS 2024; 12:50. [PMID: 38251006 PMCID: PMC10820372 DOI: 10.3390/toxics12010050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/23/2024]
Abstract
Polyhexamethylene guanidine phosphate (PHMG-p) is a common biocidal disinfectant that is widely used in industry and household products. However, PHMG-p was misused as a humidifier disinfectant (HD) in South Korea, which had fatal health effects. Various health problems including cardiovascular diseases were observed in HD-exposed groups. However, the potential underlying mechanism of HD-associated cardiovascular diseases is poorly understood. Here, we examined the procoagulant activity of platelets caused by PHMG-p and clarified the underlying mechanism. PHMG-p enhanced phosphatidylserine (PS) exposure through alteration of phospholipid transporters, scramblase, and flippase. Intracellular calcium elevation, intracellular ATP depletion, and caspase-3 activation appeared to underlie phospholipid transporter dysregulation caused by PHMG-p, which was mediated by oxidative stress and mitochondrial dysfunction. Notably, antioxidant enzyme catalase and calcium chelator EGTA reversed PHMG-p-induced PS exposure and thrombin generation, confirming the contributive role of oxidative stress and intracellular calcium in the procoagulant effects of PHMG-p. These series of events led to procoagulant activation of platelets, which was revealed as enhanced thrombin generation. Collectively, PHMG-p triggered procoagulant activation of platelets, which may promote prothrombotic risks and cardiovascular diseases. These findings improve our understanding of HD-associated cardiovascular diseases.
Collapse
Affiliation(s)
| | - Keunyoung Kim
- College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea;
| |
Collapse
|
26
|
Ajanel A, Middleton EA. Alterations in the megakaryocyte transcriptome impacts platelet function in sepsis and COVID-19 infection. Thromb Res 2023; 231:247-254. [PMID: 37258336 PMCID: PMC10198739 DOI: 10.1016/j.thromres.2023.05.015] [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: 01/04/2023] [Revised: 04/04/2023] [Accepted: 05/16/2023] [Indexed: 06/02/2023]
Abstract
Platelets and their parent cell, the megakaryocyte (MK), are increasingly recognized for their roles during infection and inflammation. The MK residing in the bone marrow or arising from precursors trafficked to other organs for development go on to form platelets through thrombopoiesis. Infection, by direct and indirect mechanisms, can alter the transcriptional profile of MKs. The altered environment, whether mediated by inflammatory cytokines or other signaling mechanisms results in an altered platelet transcriptome. Platelets released into the circulation, in turn, interact with each other, circulating leukocytes and endothelial cells and contribute to the clearance of pathogens or the potentiation of pathophysiology through such mechanisms as immunothrombosis. In this article we hope to identify key contributions that explore the impact of an altered transcriptomic landscape during severe, systemic response to infection broadly defined as sepsis, and viral infections, including SARS-CoV2. We include current publications that outline the role of MKs from bone-marrow and extra-medullary sites as well as the circulating platelet. The underlying diseases result in thrombotic complications that exacerbate organ dysfunction and mortality. Understanding the impact of platelets on the pathophysiology of disease may drive therapeutic advances to improve the morbidity and mortality of these deadly afflictions.
Collapse
Affiliation(s)
- Abigail Ajanel
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Elizabeth A Middleton
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA; Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
27
|
Nardin M, Verdoia M, Cao D, Nardin S, Kedhi E, Galasso G, van ‘t Hof AWJ, Condorelli G, De Luca G. Platelets and the Atherosclerotic Process: An Overview of New Markers of Platelet Activation and Reactivity, and Their Implications in Primary and Secondary Prevention. J Clin Med 2023; 12:6074. [PMID: 37763014 PMCID: PMC10531614 DOI: 10.3390/jcm12186074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
The key role played by platelets in the atherosclerosis physiopathology, especially in the acute setting, is ascertained: they are the main actors during thrombus formation and, thus, one of the major investigated elements related to atherothrombotic process involving coronary arteries. Platelets have been studied from different points of view, according with the technology advances and the improvement in the hemostasis knowledge achieved in the last years. Morphology and reactivity constitute the first aspects investigated related to platelets with a significant body of evidence published linking a number of their values and markers to coronary artery disease and cardiovascular events. Recently, the impact of genetics on platelet activation has been explored with promising findings as additional instrument for patient risk stratification; however, this deserves further confirmations. Moreover, the interplay between immune system and platelets has been partially elucidated in the last years, providing intriguing elements that will be basic components for future research to better understand platelet regulation and improve cardiovascular outcome of patients.
Collapse
Affiliation(s)
- Matteo Nardin
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy
- Third Medicine Division, Department of Medicine, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Monica Verdoia
- Division of Cardiology, Ospedale degli Infermi, ASL Biella, 13875 Biella, Italy
- Department of Translational Medicine, Eastern Piedmont University, 28100 Novara, Italy
| | - Davide Cao
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy
- Department of Cardiology, Humanitas Gavazzeni Hospital, 24125 Bergamo, Italy
| | - Simone Nardin
- U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- Department of Internal Medicine and Medical Sciences, School of Medicine, University of Genova, 16126 Genova, Italy
| | - Elvin Kedhi
- Division of Cardiology, Hopital Erasmus, Universitè Libre de Bruxelles, 1050 Bruxelles, Belgium
| | - Gennaro Galasso
- Division of Cardiology, Ospedale Ruggi D’Aragona, Università di Salerno, 84084 Salerno, Italy
| | - Arnoud W. J. van ‘t Hof
- Department of Cardiology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), 6229 ER Maastricht, The Netherlands
- Department of Cardiology, Zuyderland Medical Center, 6419 PC Heerlen, The Netherlands
| | - Gianluigi Condorelli
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy
- Department of Cardiovascular Medicine, IRCCS-Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Giuseppe De Luca
- Division of Cardiology, AOU “Policlinico G. Martino”, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
- Division of Cardiology, IRCCS Hospital Galeazzi-Sant’Ambrogio, 20157 Milan, Italy
| |
Collapse
|
28
|
Wang L, Liu Y, Tian R, Zuo W, Qian H, Wang L, Yang X, Liu Z, Zhang S. What do we know about platelets in myocardial ischemia-reperfusion injury and why is it important? Thromb Res 2023; 229:114-126. [PMID: 37437517 DOI: 10.1016/j.thromres.2023.06.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/22/2023] [Accepted: 06/23/2023] [Indexed: 07/14/2023]
Abstract
Myocardial ischemia-reperfusion injury (MIRI), the joint result of ischemic injury and reperfusion injury, is associated with poor outcomes in patients with acute myocardial infarction undergoing primary percutaneous coronary intervention. Accumulating evidence demonstrates that activated platelets directly contribute to the pathogenesis of MIRI through participating in the formation of microthrombi, interaction with leukocytes, secretion of active substances, constriction of microvasculature, and activation of spinal afferent nerves. The molecular mechanisms underlying the above detrimental effects of activated platelets include the homotypic and heterotypic interactions through surface receptors, transduction of intracellular signals, and secretion of active substances. Revealing the roles of platelet activation in MIRI and the associated mechanisms would provide potential targets/strategies for the clinical evaluation and treatment of MIRI. Further studies are needed to characterize the temporal (ischemia phase vs. reperfusion phase) and spatial (systemic vs. local) distributions of platelet activation in MIRI by multi-omics strategies. To improve the likelihood of translating novel cardioprotective interventions into clinical practice, basic researches maximally replicating the complexity of clinical scenarios would be necessary.
Collapse
Affiliation(s)
- Lun Wang
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Yifan Liu
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Ran Tian
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Wei Zuo
- Department of Pharmacy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Hao Qian
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Liang Wang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Xinglin Yang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Zhenyu Liu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China.
| | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
29
|
Xie C, Zhang L. Design and characterization of antithrombotic ClEKnsTy-Au nanoparticles as diagnostic and therapeutic reagents. Phys Chem Chem Phys 2023. [PMID: 37466214 DOI: 10.1039/d3cp01000g] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Thrombosis can cause various cardiovascular diseases, which seriously endanger human life. Development of diagnostic and therapeutic reagents for thrombosis at an early stage would be helpful for the improvement of treatment and the reduction of mortality. In the present study, based on an antithrombotic peptide lEKnsTy (lowercase letters represent D-amino acid residues), a diagnostic and therapeutic reagent targeting collagen and the early stage of thrombosis was proposed, where cysteine was introduced into the amino terminus of lEKnsTy to prepare ClEKnsTy, followed by coupling with AuNPs to prepare nanoconjugate AuNP-Cl. The binding of AuNP-Cl on the collagen surface was then confirmed by the molecular dynamics simulations of the binding of ClEKnsTy on collagen, and the experimental results of the binding of AuNP-Cl on collagen. The inhibition of platelet adhesion on the collagen surface by AuNP-Cl was also confirmed. Moreover, the good imaging ability of AuNP-Cl was confirmed by dark-field microscopy. These results indicated that AuNP-Cl was a potential effective diagnostic and therapeutic reagent targeting collagen, which would be helpful for the research and development of multifunctional antithrombotic reagents.
Collapse
Affiliation(s)
- Chen Xie
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, People's Republic of China.
| | - Lin Zhang
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, People's Republic of China.
| |
Collapse
|
30
|
De Vrij EL, Bouma HR, Henning RH, Cooper ST. Hibernation and hemostasis. Front Physiol 2023; 14:1207003. [PMID: 37435313 PMCID: PMC10331295 DOI: 10.3389/fphys.2023.1207003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 06/12/2023] [Indexed: 07/13/2023] Open
Abstract
Hibernating mammals have developed many physiological adaptations to accommodate their decreased metabolism, body temperature, heart rate and prolonged immobility without suffering organ injury. During hibernation, the animals must suppress blood clotting to survive prolonged periods of immobility and decreased blood flow that could otherwise lead to the formation of potentially lethal clots. Conversely, upon arousal hibernators must be able to quickly restore normal clotting activity to avoid bleeding. Studies in multiple species of hibernating mammals have shown reversible decreases in circulating platelets, cells involved in hemostasis, as well as in protein coagulation factors during torpor. Hibernator platelets themselves also have adaptations that allow them to survive in the cold, while those from non-hibernating mammals undergo lesions during cold exposure that lead to their rapid clearance from circulation when re-transfused. While platelets lack a nucleus with DNA, they contain RNA and other organelles including mitochondria, in which metabolic adaptations may play a role in hibernator's platelet resistance to cold induced lesions. Finally, the breakdown of clots, fibrinolysis, is accelerated during torpor. Collectively, these reversible physiological and metabolic adaptations allow hibernating mammals to survive low blood flow, low body temperature, and immobility without the formation of clots during torpor, yet have normal hemostasis when not hibernating. In this review we summarize blood clotting changes and the underlying mechanisms in multiple species of hibernating mammals. We also discuss possible medical applications to improve cold preservation of platelets and antithrombotic therapy.
Collapse
Affiliation(s)
- Edwin L. De Vrij
- Department of Plastic Surgery, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, Netherlands
| | - Hjalmar R. Bouma
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, Netherlands
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Robert H. Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, Netherlands
| | - Scott T. Cooper
- Biology Department, University of Wisconsin-La Crosse, La Crosse, WI, United States
| |
Collapse
|
31
|
Schurr Y, Reil L, Spindler M, Nieswandt B, Machesky LM, Bender M. The WASH-complex subunit Strumpellin regulates integrin αIIbβ3 trafficking in murine platelets. Sci Rep 2023; 13:9526. [PMID: 37308549 PMCID: PMC10260982 DOI: 10.1038/s41598-023-36387-8] [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: 02/03/2023] [Accepted: 06/02/2023] [Indexed: 06/14/2023] Open
Abstract
The platelet specific integrin αIIbβ3 mediates platelet adhesion, aggregation and plays a central role in thrombosis and hemostasis. In resting platelets, αIIbβ3 is expressed on the membrane surface and in intracellular compartments. Upon activation, the number of surface-expressed αIIbβ3 is increased by the translocation of internal granule pools to the plasma membrane. The WASH complex is the major endosomal actin polymerization-promoting complex and has been implicated in the generation of actin networks involved in endocytic trafficking of integrins in other cell types. The role of the WASH complex and its subunit Strumpellin in platelet function is still unknown. Here, we report that Strumpellin-deficient murine platelets display an approximately 20% reduction in integrin αIIbβ3 surface expression. While exposure of the internal αIIbβ3 pool after platelet activation was unaffected, the uptake of the αIIbβ3 ligand fibrinogen was delayed. The number of platelet α-granules was slightly but significantly increased in Strumpellin-deficient platelets. Quantitative proteome analysis of isolated αIIbβ3-positive vesicular structures revealed an enrichment of protein markers, which are associated with the endoplasmic reticulum, Golgi complex and early endosomes in Strumpellin-deficient platelets. These results point to a so far unidentified role of the WASH complex subunit Strumpellin in integrin αIIbβ3 trafficking in murine platelets.
Collapse
Affiliation(s)
- Yvonne Schurr
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Lucy Reil
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Markus Spindler
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Laura M Machesky
- Department of Biochemistry, University of Cambridge, Sanger Building, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Markus Bender
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, Josef-Schneider-Str. 2, 97080, Würzburg, Germany.
| |
Collapse
|
32
|
Barakzie A, Jansen AG, ten Cate H, de Maat MP. Coagulation biomarkers for ischemic stroke. Res Pract Thromb Haemost 2023; 7:100160. [PMID: 37274178 PMCID: PMC10236221 DOI: 10.1016/j.rpth.2023.100160] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/24/2023] [Accepted: 03/25/2023] [Indexed: 06/06/2023] Open
Abstract
A State of the Art lecture titled "coagulation biomarkers for ischemic stroke" was presented at the International Society on Thrombosis and Haemostasis (ISTH) Congress in 2022. Ischemic stroke (IS) is a common disease with major morbidity and mortality. It is a challenge to determine which patients are at risk for IS or have poor clinical outcome after IS. An imbalance of coagulation markers may contribute to the progression and prognosis of IS. Therefore, we now discuss studies on the association of selected coagulation biomarkers from the hemostasis, inflammation, and immunothrombosis systems with the risk of IS, stroke severity at the acute phase, and clinical outcome after treatment. We report on coagulation biomarker-induced risk of IS, stroke severity, and outcomes following IS derived from prospective population studies, case-control studies, and acute-phase IS studies. We found indications that many coagulation and inflammation biomarkers are associated with IS, but it is early to conclude that any of these biomarkers can be applied in a therapeutic setting to predict patients at risk of IS, stroke severity at the acute phase, and clinical outcome after treatment. The strongest evidence for a role in IS was found for beta-thromboglobulin, von Willebrand factor, factor VIII, fibrinogen, thrombin-activatable fibrinolysis inhibitor, D-dimer, and neutrophil extracellular traps, and therefore, they are promising candidates. Further research and validation in large-size populations using well-defined study designs are warranted. Finally, we provide a selection of recent data relevant to this subject that was presented at the 2022 ISTH Congress.
Collapse
Affiliation(s)
- Aarazo Barakzie
- Department of Hematology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - A.J. Gerard Jansen
- Department of Hematology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Hugo ten Cate
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, The Netherlands
- Thrombosis Expertise Center and Department of Internal Medicine, Maastricht University Medical Centre, The Netherlands
| | - Moniek P.M. de Maat
- Department of Hematology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
33
|
Krishnan N, Peng FX, Mohapatra A, Fang RH, Zhang L. Genetically engineered cellular nanoparticles for biomedical applications. Biomaterials 2023; 296:122065. [PMID: 36841215 PMCID: PMC10542936 DOI: 10.1016/j.biomaterials.2023.122065] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/14/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023]
Abstract
In recent years, nanoparticles derived from cellular membranes have been increasingly explored for the prevention and treatment of human disease. With their flexible design and ability to interface effectively with the surrounding environment, these biomimetic nanoparticles can outperform their traditional synthetic counterparts. As their popularity has increased, researchers have developed novel ways to modify the nanoparticle surface to introduce new or enhanced capabilities. Moving beyond naturally occurring materials derived from wild-type cells, genetic manipulation has proven to be a robust and flexible method by which nanoformulations with augmented functionalities can be generated. In this review, an overview of genetic engineering approaches to express novel surface proteins is provided, followed by a discussion on the various biomedical applications of genetically modified cellular nanoparticles.
Collapse
Affiliation(s)
- Nishta Krishnan
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Fei-Xing Peng
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Animesh Mohapatra
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Ronnie H Fang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
34
|
Lin KH, Li JY, Chen RJ, Chen TY, Hsu SH, Wang HH, Peng HY, Sun YY, Lu WJ. Paclitaxel exerts antiplatelet and antithrombotic activities: Additional benefit from use of paclitaxel-coated balloons and -eluting stents in coronary revascularization and prevention of in-stent restenosis. Thromb Res 2023; 225:63-72. [PMID: 37030187 DOI: 10.1016/j.thromres.2023.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 03/12/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
INTRODUCTION Paclitaxel is a microtubule-stabilizing drug used to treat several types of cancer, including ovarian and breast cancer. Because of its antiproliferative effect on vascular smooth muscle cells, balloons and stents are coated with paclitaxel for use in coronary revascularization and prevention of in-stent restenosis (ISR). However, mechanisms underlying ISR are complicated. Platelet activation is one of the major causes of ISR after percutaneous coronary intervention. Although the antiplatelet activity of paclitaxel was noted in rabbit platelets, the effect of paclitaxel on platelets remains unclear. This study investigated whether paclitaxel exhibits antiplatelet activity in human platelets. METHODS AND RESULTS Paclitaxel inhibited platelet aggregation induced by collagen but not that induced by thrombin, arachidonic acid, or U46619, suggesting that paclitaxel is more sensitive to the inhibition of collagen-induced platelet activation. Moreover, paclitaxel blocked collagen receptor glycoprotein (GP) VI downstream signaling molecules, including Lyn, Fyn, PLCγ2, PKC, Akt, and MAPKs. However, paclitaxel did not directly bind to GPVI and cause GPVI shedding, as detected by surface plasmon resonance and flow cytometry, respectively, indicating that paclitaxel may interfere with GPVI downstream signaling molecules, such as Lyn and Fyn. Paclitaxel also prevented granule release and GPIIbIIIa activation induced by collagen and low convulxin doses. Moreover, paclitaxel attenuated pulmonary thrombosis and delayed platelet thrombus formation in mesenteric microvessels without significantly affecting hemostasis. CONCLUSION Paclitaxel exerts antiplatelet and antithrombotic effects. Thus, paclitaxel may provide additional benefits beyond its antiproliferative effect when used in drug-coated balloons and drug-eluting stents for coronary revascularization and prevention of ISR.
Collapse
|
35
|
Jooss NJ, Henskens YMC, Watson SP, Farndale RW, Gawaz MP, Jandrot-Perrus M, Poulter NS, Heemskerk JWM. Pharmacological Inhibition of Glycoprotein VI- and Integrin α2β1-Induced Thrombus Formation Modulated by the Collagen Type. Thromb Haemost 2023; 123:597-612. [PMID: 36807826 DOI: 10.1055/s-0043-1761463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
BACKGROUND In secondary cardiovascular disease prevention, treatments blocking platelet-derived secondary mediators pose a risk of bleeding. Pharmacological interference of the interaction of platelets with exposed vascular collagens is an attractive alternative, with clinical trials ongoing. Antagonists of the collagen receptors, glycoprotein VI (GPVI), and integrin α2β1, include recombinant GPVI-Fc dimer construct Revacept, 9O12 mAb based on the GPVI-blocking reagent Glenzocimab, Syk tyrosine-kinase inhibitor PRT-060318, and anti-α2β1 mAb 6F1. No direct comparison has been made of the antithrombic potential of these drugs. METHODS Using a multiparameter whole-blood microfluidic assay, we compared the effects of Revacept, 9O12-Fab, PRT-060318, or 6F1 mAb intervention with vascular collagens and collagen-related substrates with varying dependencies on GPVI and α2β1. To inform on Revacept binding to collagen, we used fluorescent-labelled anti-GPVI nanobody-28. RESULTS AND CONCLUSION In this first comparison of four inhibitors of platelet-collagen interactions with antithrombotic potential, we find that at arterial shear rate: (1) the thrombus-inhibiting effect of Revacept was restricted to highly GPVI-activating surfaces; (2) 9O12-Fab consistently but partly inhibited thrombus size on all surfaces; (3) effects of GPVI-directed interventions were surpassed by Syk inhibition; and (4) α2β1-directed intervention with 6F1 mAb was strongest for collagens where Revacept and 9O12-Fab were limitedly effective. Our data hence reveal a distinct pharmacological profile for GPVI-binding competition (Revacept), GPVI receptor blockage (9O12-Fab), GPVI signaling (PRT-060318), and α2β1 blockage (6F1 mAb) in flow-dependent thrombus formation, depending on the platelet-activating potential of the collagen substrate. This work thus points to additive antithrombotic action mechanisms of the investigated drugs.
Collapse
Affiliation(s)
- Natalie J Jooss
- Department of Biochemistry, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands.,Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Yvonne M C Henskens
- Central Diagnostic Laboratory, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Midlands, United Kingdom
| | - Richard W Farndale
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.,CambCol Laboratories, Ely, United Kingdom
| | - Meinrad P Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Martine Jandrot-Perrus
- UMR_S1148, Laboratory for Vascular Translational Science, INSERM, University Paris Cité, Paris, France
| | - Natalie S Poulter
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Midlands, United Kingdom
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands.,Synapse Research Institute, Maastricht, The Netherlands
| |
Collapse
|
36
|
Fang L, Yu S, Tian X, Fu W, Su L, Chen Z, Yan C, He J, Hong J, Lian W, Liu G, Zhang Y, Zhou J, Hu L. Severe fever with thrombocytopenia syndrome virus replicates in platelets and enhances platelet activation. J Thromb Haemost 2023; 21:1336-1351. [PMID: 36792011 DOI: 10.1016/j.jtha.2023.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023]
Abstract
BACKGROUND Severe fever with thrombocytopenia syndrome (SFTS) virus (SFTSV) infection causes an emerging hemorrhagic fever in East Asia with a high mortality rate. Thrombocytopenia is a consistent feature of SFTS illness, but the mechanism remains elusive. OBJECTIVES We aimed to better understand the role of platelets in the pathophysiology of SFTSV infection, including the development of thrombocytopenia. METHODS Using platelets from healthy volunteers and patients with SFTS, we evaluated the functional changes in platelets against SFTSV infection. We investigated the direct effect of glycoprotein VI on platelet-SFTSV interaction by quantitative real-time PCR, molecular docking, surface plasmon resonance spectrometry, flow cytometry, western blot, and platelet functional studies in vitro. Interactions of SFTSV and platelet-SFTSV complexes with macrophages were also determined by scanning electron microscope, quantitative real-time PCR, and flow cytometry. RESULTS This study is the first to demonstrate that platelets are capable of harboring and producing SFTSV particles. Structural and functional studies found that SFTSVs bind platelet glycoprotein VI to potentiate platelet activation, including platelet aggregation, adenosine triphosphate release, spreading, clot retraction, coagulation, phosphatidylserine exposure, thrombus formation, and adherence. In vitro mechanistic studies highlighted that the interaction of platelets with human THP-1 cells promoted SFTSV clearance and suppressed cytokine production in macrophages. However, unwanted SFTSV replication in macrophages reciprocally aggravated SFTSV persistence in the circulation, which may contribute to thrombocytopenia and other complications during SFTSV infection. CONCLUSION These findings together highlighted the pathophysiological role of platelets in initial intrinsic defense against SFTSV infections, as well as intertwined processes with host immunity, which can also lead to thrombocytopenia and poor prognosis.
Collapse
Affiliation(s)
- Lei Fang
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Zhejiang Provincial Centers for Disease Control and Prevention, Hangzhou, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Sicong Yu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaoxu Tian
- Department of Cardiology, Cardiovascular Center, Henan Key Laboratory of Hereditary, Cardiovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wanrong Fu
- Department of Cardiology, Cardiovascular Center, Henan Key Laboratory of Hereditary, Cardiovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lingxuan Su
- Zhejiang Provincial Centers for Disease Control and Prevention, Hangzhou, China
| | - Zhi Chen
- National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Hangzhou, China
| | - Chunlan Yan
- Department of Biophysics, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ji He
- Blood Center of Zhejiang Province, Hangzhou, China
| | - Jin Hong
- Department of Cardiology, Cardiovascular Center, Henan Key Laboratory of Hereditary, Cardiovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenwen Lian
- National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Hangzhou, China
| | - Gangqiong Liu
- Department of Cardiology, Cardiovascular Center, Henan Key Laboratory of Hereditary, Cardiovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yanjun Zhang
- Zhejiang Provincial Centers for Disease Control and Prevention, Hangzhou, China.
| | - Jiancang Zhou
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China.
| | - Liang Hu
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Department of Cardiology, Cardiovascular Center, Henan Key Laboratory of Hereditary, Cardiovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
37
|
Ayyoub S, Orriols R, Oliver E, Ceide OT. Thrombosis Models: An Overview of Common In Vivo and In Vitro Models of Thrombosis. Int J Mol Sci 2023; 24:2569. [PMID: 36768891 PMCID: PMC9917341 DOI: 10.3390/ijms24032569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 02/03/2023] Open
Abstract
Occlusions in the blood vessels caused by blood clots, referred to as thrombosis, and the subsequent outcomes are leading causes of morbidity and mortality worldwide. In vitro and in vivo models of thrombosis have advanced our understanding of the complex pathways involved in its development and allowed the evaluation of different therapeutic approaches for its management. This review summarizes different commonly used approaches to induce thrombosis in vivo and in vitro, without detailing the protocols for each technique or the mechanism of thrombus development. For ease of flow, a schematic illustration of the models mentioned in the review is shown below. Considering the number of available approaches, we emphasize the importance of standardizing thrombosis models in research per study aim and application, as different pathophysiological mechanisms are involved in each model, and they exert varying responses to the same carried tests. For the time being, the selection of the appropriate model depends on several factors, including the available settings and research facilities, the aim of the research and its application, and the researchers' experience and ability to perform surgical interventions if needed.
Collapse
Affiliation(s)
- Sana Ayyoub
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
| | - Ramon Orriols
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
| | - Eduardo Oliver
- Centro de Investigaciones Biologicas Margarita Salas (CIB-CSIC), 28040 Madrid, Spain
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Olga Tura Ceide
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| |
Collapse
|
38
|
Alimardani V, Rahiminezhad Z, DehghanKhold M, Farahavar G, Jafari M, Abedi M, Moradi L, Niroumand U, Ashfaq M, Abolmaali SS, Yousefi G. Nanotechnology-based cell-mediated delivery systems for cancer therapy and diagnosis. Drug Deliv Transl Res 2023; 13:189-221. [PMID: 36074253 DOI: 10.1007/s13346-022-01211-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2022] [Indexed: 12/13/2022]
Abstract
The global prevalence of cancer is increasing, necessitating new additions to traditional treatments and diagnoses to address shortcomings such as ineffectiveness, complications, and high cost. In this context, nano and microparticulate carriers stand out due to their unique properties such as controlled release, higher bioavailability, and lower toxicity. Despite their popularity, they face several challenges including rapid liver uptake, low chemical stability in blood circulation, immunogenicity concerns, and acute adverse effects. Cell-mediated delivery systems are important topics to research because of their biocompatibility, biodegradability, prolonged delivery, high loading capacity, and targeted drug delivery capabilities. To date, a variety of cells including blood, immune, cancer, and stem cells, sperm, and bacteria have been combined with nanoparticles to develop efficient targeted cancer delivery or diagnosis systems. The review paper aimed to provide an overview of the potential applications of cell-based delivery systems in cancer therapy and diagnosis.
Collapse
Affiliation(s)
- Vahid Alimardani
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Rahiminezhad
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahvash DehghanKhold
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ghazal Farahavar
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahboobeh Jafari
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Abedi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Leila Moradi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Uranous Niroumand
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Ashfaq
- University Centre for Research & Development (UCRD), Chandigarh University, Gharaun, Mohali, 140413, Punjab, India. .,Department of Biotechnology, Chandigarh University, Gharaun, Mohali, 140413, Punjab, India.
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran. .,Center for Drug Delivery in Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Gholamhossein Yousefi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran. .,Center for Drug Delivery in Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
39
|
Wang H, Zhang B, Zhong X, Qin D, Li Z. Mechanism Research of Platelet Core Marker Prediction and Molecular Recognition in Cardiovascular Events. Comb Chem High Throughput Screen 2023; 26:103-115. [PMID: 35345996 DOI: 10.2174/1386207325666220328091748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/17/2022] [Accepted: 01/27/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Thrombosis triggered by platelet activation plays a vital role in the pathogenesis of cardiovascular and cerebrovascular diseases. OBJECTIVE This study aims to find platelet combined biomarkers for cardiovascular diseases and investigate the possibility of Concanavalin A (ConA) acting on platelets as a new pharmacological target. METHODS High-throughput Technology and bioinformatics analysis were combined and groups of microarray chip gene expression profiles for acute myocardial infarction (AMI) and sickle cell disease (SCD) were obtained using GEO database screening. R language limma package was used to obtain differentially expressed genes (DEGs). GO, KEGG, and other databases were utilized to perform the enrichment analysis of DEGs' functions, pathways, etc. PPI network was constructed using STRING database and Cytoscape software, and MCC algorithm was used to obtain the 200 core genes of the two groups of DEGs. Core targets were confirmed by constructing an intersection area screening. A type of molecular probe, ConA, was molecularly docked with the above core targets on the Zdock, HEX, and 3D-DOCK servers. RESULTS We found six core markers, CD34, SOCS2, ABL1, MTOR, VEGFA, and SMURF1, which were simultaneously related to both diseases, and the docking effect showed that VEGFA is the best-performing. CONCLUSION VEGFA is most likely to reduce its expression by binding to ConA, which could affect the downstream regulation of the PI3K/Akt signaling pathway during platelet activation. Some other core targets also have the opportunity to interact with ConA to affect platelet-activated thrombosis and trigger changes in cardiovascular events.
Collapse
Affiliation(s)
- Hongdan Wang
- Chongqing Engineering Research Center of Medical Electronics and Information Technology, College of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
| | - Bingyu Zhang
- Chongqing Engineering Research Center of Medical Electronics and Information Technology, College of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
| | - Xianhua Zhong
- Chongqing Engineering Research Center of Medical Electronics and Information Technology, College of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
| | - Dui Qin
- Chongqing Engineering Research Center of Medical Electronics and Information Technology, College of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
| | - Zhangyong Li
- Chongqing Engineering Research Center of Medical Electronics and Information Technology, College of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing 400065, China
| |
Collapse
|
40
|
Yu W, Ilyas I, Hu X, Xu S, Yu H. Therapeutic potential of paeoniflorin in atherosclerosis: A cellular action and mechanism-based perspective. Front Immunol 2022; 13:1072007. [PMID: 36618414 PMCID: PMC9811007 DOI: 10.3389/fimmu.2022.1072007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Epidemiological studies have shown that the incidence, prevalence and mortality of atherosclerotic cardiovascular disease (ASCVD) are increasing globally. Atherosclerosis is characterized as a chronic inflammatory disease which involves inflammation and immune dysfunction. P. lactiflora Pall. is a plant origin traditional medicine that has been widely used for the treatment of various diseases for more than a millennium in China, Japan and Korean. Paeoniflorin is a bioactive monomer extracted from P. lactiflora Pall. with anti-atherosclerosis effects. In this article, we comprehensively reviewed the potential therapeutic effects and molecular mechanism whereby paeoniflorin protects against atherosclerosis from the unique angle of inflammation and immune-related pathway dysfunction in vascular endothelial cells, smooth muscle cells, monocytes, macrophages, platelets and mast cells. Paeoniflorin, with multiple protective effects in atherosclerosis, has the potential to be used as a promising therapeutic agent for the treatment of atherosclerosis and its complications. We conclude with a detailed discussion of the challenges and future perspective of paeoniflorin in translational cardiovascular medicine.
Collapse
Affiliation(s)
- Wei Yu
- School of Materials Science and Engineering, Hefei University of Technology, Hefei, Anhui, China,Center for Drug Research and Development, Anhui Renovo Pharmaceutical Co., Ltd, Center for Drug Research and Development, Hefei, Anhui, China
| | - Iqra Ilyas
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xuerui Hu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Suowen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Hui Yu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Materials Laminating Fabrication and Interfacial Controlling Technology, Hebei University of Technology, Tianjin, China,*Correspondence: Hui Yu,
| |
Collapse
|
41
|
Mubeen S, Domingo-Fernández D, Díaz del Ser S, Solanki DM, Kodamullil AT, Hofmann-Apitius M, Hopp MT, Imhof D. Exploring the Complex Network of Heme-Triggered Effects on the Blood Coagulation System. J Clin Med 2022; 11:jcm11195975. [PMID: 36233841 PMCID: PMC9572022 DOI: 10.3390/jcm11195975] [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: 09/20/2022] [Revised: 10/06/2022] [Accepted: 10/08/2022] [Indexed: 11/16/2022] Open
Abstract
Excess labile heme, occurring under hemolytic conditions, displays a versatile modulator in the blood coagulation system. As such, heme provokes prothrombotic states, either by binding to plasma proteins or through interaction with participating cell types. However, despite several independent reports on these effects, apparently contradictory observations and significant knowledge gaps characterize this relationship, which hampers a complete understanding of heme-driven coagulopathies and the development of suitable and specific treatment options. Thus, the computational exploration of the complex network of heme-triggered effects in the blood coagulation system is presented herein. Combining hemostasis- and heme-specific terminology, the knowledge available thus far was curated and modeled in a mechanistic interactome. Further, these data were incorporated in the earlier established heme knowledge graph, "HemeKG", to better comprehend the knowledge surrounding heme biology. Finally, a pathway enrichment analysis of these data provided deep insights into so far unknown links and novel experimental targets within the blood coagulation cascade and platelet activation pathways for further investigation of the prothrombotic nature of heme. In summary, this study allows, for the first time, a detailed network analysis of the effects of heme in the blood coagulation system.
Collapse
Affiliation(s)
- Sarah Mubeen
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany
| | - Daniel Domingo-Fernández
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany
- Enveda Biosciences, Inc., San Francisco, CA 94080, USA
| | - Sara Díaz del Ser
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany
- Polytechnic University of Madrid, E-28040 Madrid, Spain
| | - Dhwani M. Solanki
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Alpha T. Kodamullil
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany
- Causality Biomodels, Kinfra Hi-Tech Park, Kalamassery, Cochin 683503, Kerala, India
| | - Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, D-53757 Sankt Augustin, Germany
| | - Marie-T. Hopp
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
- Correspondence: (M.-T.H.); (D.I.); Tel.: +49-228-73-5231 (M.-T.H.); +49-228-73-5254 (D.I.)
| | - Diana Imhof
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
- Correspondence: (M.-T.H.); (D.I.); Tel.: +49-228-73-5231 (M.-T.H.); +49-228-73-5254 (D.I.)
| |
Collapse
|
42
|
Siahaan AMP, Tandean S, Nainggolan BWM. Spontaneous epidural hematoma induced by rivaroxaban: A case report and review of the literature. Surg Neurol Int 2022; 13:420. [PMID: 36324933 PMCID: PMC9610455 DOI: 10.25259/sni_608_2022] [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] [Received: 07/07/2022] [Accepted: 09/03/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Trauma is the most frequent reason for epidural bleeding. However, numerous investigation had discovered that anticoagulants such as rivaroxaban could cause epidural hematoma. Here, we present a case of epidural hematoma in young man who got rivaroxaban as treatment of deep vein thrombosis. Case Description: A 27-year-old male with a history of deep vein thrombosis and one month of rivaroxaban medication presented with seizure and loss of consciousness following a severe headache. A CT scan of the head revealed epidural bleeding, and emergency blood clot removal was performed. As a reversal, prothrombin complex was utilized. Conclusion: Rivaroxaban has the potential to cause an epidural hemorrhage. Reversal anticoagulant should be administered before doing emergency surgery.
Collapse
Affiliation(s)
| | - Steven Tandean
- Department of Neurosurgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | | |
Collapse
|
43
|
Artesunate as a glycoprotein VI antagonist for preventing platelet activation and thrombus formation. Biomed Pharmacother 2022; 153:113531. [DOI: 10.1016/j.biopha.2022.113531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 11/22/2022] Open
|
44
|
Sun S, Qiao B, Han Y, Wang B, Wei S, Chen Y. Posttranslational modifications of platelet adhesion receptors. Pharmacol Res 2022; 183:106413. [PMID: 36007773 DOI: 10.1016/j.phrs.2022.106413] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/11/2022] [Accepted: 08/19/2022] [Indexed: 10/15/2022]
Abstract
Platelets play a key role in normal hemostasis, whereas pathological platelet adhesion is involved in various cardiovascular events. The underlying cause in cardiovascular events involves plaque rupture leading to subsequent platelet adhesion, activation, release, and eventual thrombosis. Traditional antithrombotic drugs often target the signal transduction process of platelet adhesion receptors by influencing the synthesis of some key molecules, and their effects are limited. Posttranslational modifications (PTMs) of platelet adhesion receptors increase the functional diversity of the receptors and affect platelet physiological and pathological processes. Antithrombotic drugs targeting PTMs of platelet adhesion receptors may represent a new therapeutic idea. In this review, various PTMs, including phosphorylation, glycosylation, ubiquitination, nitrosylation, methylation, lipidation, and proteolysis, of three platelet adhesion receptors, glycoprotein Ib-IX-V (GPIb-IX-V), glycoprotein VI (GPVI), and integrin αIIbβ3, are reviewed. It is important to comprehensively understand the PTMs process of platelet adhesion receptors.
Collapse
Affiliation(s)
- Shukun Sun
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Bao Qiao
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yu Han
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Bailu Wang
- Clinical Trial Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Shujian Wei
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China.
| | - Yuguo Chen
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
45
|
Navarro S, Starke A, Heemskerk JWM, Kuijpers MJE, Stegner D, Nieswandt B. Targeting of a Conserved Epitope in Mouse and Human GPVI Differently Affects Receptor Function. Int J Mol Sci 2022; 23:8610. [PMID: 35955743 PMCID: PMC9369317 DOI: 10.3390/ijms23158610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 12/04/2022] Open
Abstract
Glycoprotein (GP) VI is the major platelet collagen receptor and a promising anti-thrombotic target. This was first demonstrated in mice using the rat monoclonal antibody JAQ1, which completely blocks the Collagen-Related Peptide (CRP)-binding site on mouse GPVI and efficiently inhibits mouse platelet adhesion, activation and aggregation on collagen. Here, we show for the first time that JAQ1 cross-reacts with human GPVI (huGPVI), but not with GPVI in other tested species, including rat, rabbit, guinea pig, swine, and dog. We further demonstrate that JAQ1 differently modulates mouse and human GPVI function. Similar to its effects on mouse GPVI (mGPVI), JAQ1 inhibits CRP-induced activation in human platelets, whereas, in stark contrast to mouse GPVI, it does not inhibit the adhesion, activation or aggregate formation of human platelets on collagen, but causes instead an increased response. This effect was also seen with platelets from newly generated human GPVI knockin mice (hGP6tg/tg). These results indicate that the binding of JAQ1 to a structurally conserved epitope in GPVI differently affects its function in human and mouse platelets.
Collapse
Affiliation(s)
- Stefano Navarro
- Institute of Experimental Biomedicine, University Hospital Würzburg and Rudolf Virchow Center for Integrative and Translational Bioimaging, Josef-Schneider-Straße 2, 97080 Würzburg, Germany; (S.N.); (A.S.); (D.S.)
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands; (J.W.M.H.); (M.J.E.K.)
| | - Andreas Starke
- Institute of Experimental Biomedicine, University Hospital Würzburg and Rudolf Virchow Center for Integrative and Translational Bioimaging, Josef-Schneider-Straße 2, 97080 Würzburg, Germany; (S.N.); (A.S.); (D.S.)
| | - Johan W. M. Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands; (J.W.M.H.); (M.J.E.K.)
- Synapse Research Institute, Kon. Emmaplein 7, 6214 AC Maastricht, The Netherlands
| | - Marijke J. E. Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands; (J.W.M.H.); (M.J.E.K.)
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center+, Professor Debyelaan 25, 6229 HX Maastricht, The Netherlands
| | - David Stegner
- Institute of Experimental Biomedicine, University Hospital Würzburg and Rudolf Virchow Center for Integrative and Translational Bioimaging, Josef-Schneider-Straße 2, 97080 Würzburg, Germany; (S.N.); (A.S.); (D.S.)
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital Würzburg and Rudolf Virchow Center for Integrative and Translational Bioimaging, Josef-Schneider-Straße 2, 97080 Würzburg, Germany; (S.N.); (A.S.); (D.S.)
| |
Collapse
|
46
|
Zhang G, Pan Y, Cheng H, Gong S, Chu Q, Chen P. Theaflavin: a natural candidate to restrain thrombosis. Food Funct 2022; 13:7572-7581. [PMID: 35815842 DOI: 10.1039/d2fo00152g] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Many clinical studies have demonstrated the beneficial effects of black tea on cardiovascular diseases. However, the antiplatelet and antithrombotic activities of theaflavin (TF-1) remain unknown. In this study, we aimed to investigate the beneficial effects of TF-1 on platelet activation and thrombosis formation both in vitro and in vivo. Firstly, the in vitro antiplatelet activity of TF-1 was analyzed using platelets isolated from human blood via aggregometry, flow cytometry, the ELISA kit, western blot and fluorescence microscopy. Subsequently, the in vivo analysis of the hemostatic state and thrombosis formation was carried out in C57BL/6 mice based on the tail bleeding time and an FeCl3-induced arterial thrombus model. The results showed that TF-1 could prominently inhibit platelet aggregation in a dose-dependent manner, and attenuate P-selectin expression, fibrinogen binding, spreading and thromboxane A2 (TxA2) formation. Western blot analysis showed that TF-1 potently inhibited spleen tyrosine kinase (Syk) and Akt (ser473/474) phosphorylation. The in vivo data further confirmed the inhibition of platelet activation by TF-1 with a prolonged arterial occlusion time (from 15.0 ± 1.1 minutes to 40.0 ± 5.4 minutes). All the results indicated that TF-1 is a powerful inhibitor of platelet activation and thrombosis formation in C57BL/6 mice, and could be developed as a novel food-based inhibitor of thrombotic disorders.
Collapse
Affiliation(s)
- Gang Zhang
- Department of Tea Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China.
| | - Yani Pan
- Department of Tea Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China.
| | - Hao Cheng
- Department of Tea Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China.
| | - Shuying Gong
- Department of Tea Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China.
| | - Qiang Chu
- Department of Tea Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China.
| | - Ping Chen
- Department of Tea Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China.
| |
Collapse
|
47
|
Effects of Momordica charantia exosomes on platelet activation, adhesion, and aggregation. Blood Coagul Fibrinolysis 2022; 33:372-380. [PMID: 35834718 DOI: 10.1097/mbc.0000000000001151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The platelets play a crucial role in the progression of multiple medical conditions, such as stroke and tumor metastasis, where antiplatelet therapy may be a boon for treating these diseases. In this study, we have attempted to study the effects of extracted Momordica charantia exosomes (MCEs) on platelet activation, adhesion, and aggregation. Adult platelets isolated from healthy individuals were dose-dependently treated with MCEs (0.1, 40, and 200 μg/ml). We performed flow cytometry to detect the expression of platelet activation protein marker-activated GP IIb/IIIa (PAC-1) and P-selectin (CD62P). Platelet adhesion was analyzed through fluorescence labeling assays. The effect of MCEs on platelet-mediated cell migration of HCT116 cells was observed by transwell. Furthermore, the MCAO model of Sprague-Dawley rats was used to observe the effect of MCEs (200, 400, and 800 μg/kg) on platelet aggregation and maximum thrombotic agglutination in vivo. The results showed that 200 μg/ml MCEs exerted the most pronounced effect on platelet activation, adhesion, and aggregation. Experiments on animals showed that MCEs significantly inhibited platelet aggregation and attenuated the maximum thrombus agglutination. We concluded that MCEs inhibited platelet activation, adhesion, aggregation, and platelet-mediated migration of HCT116 cells, indicating the potential role MCEs may play in the treatment of stroke and tumor metastasis.
Collapse
|
48
|
Coumarin Derivatives Inhibit ADP-Induced Platelet Activation and Aggregation. Molecules 2022; 27:molecules27134054. [PMID: 35807298 PMCID: PMC9268609 DOI: 10.3390/molecules27134054] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/16/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
Coumarin was first discovered in Tonka bean and then widely in other plants. Coumarin has an anticoagulant effect, and its derivative, warfarin, is a vitamin K analogue that inhibits the synthesis of clotting factors and is more widely used in the clinical treatment of endovascular embolism. At present, many artificial chemical synthesis methods can be used to modify the structure of coumarin to develop many effective drugs with low toxicity. In this study, we investigated the effects of six coumarin derivatives on the platelet aggregation induced by adenosine diphosphate (ADP). We found that the six coumarin derivatives inhibited the active form of GPIIb/IIIa on platelets and hence inhibit platelet aggregation. We found that 7-hydroxy-3-phenyl 4H-chromen-4-one (7-hydroxyflavone) had the most severe effect. In addition, we further analyzed the downstream signal transduction of the ADP receptor, including the release of calcium ions and the regulation of cAMP, which were inhibited by the six coumarin derivatives selected in this study. These results suggest that coumarin derivatives inhibit coagulation by inhibiting the synthesis of coagulation factors and they may also inhibit platelet aggregation.
Collapse
|
49
|
Zheng S, Hao T, Zhang L. Development of the Antithrombotic Peptide LEKNSTY Targeting the Collagen Surface: I. Design and Validation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:7107-7113. [PMID: 35622988 DOI: 10.1021/acs.langmuir.2c00586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Exposed collagen on the diseased vessel wall is crucial for arterial thrombosis. The currently developed antithrombotic drugs mostly target blood components such as platelets and suffer from the risk of bleeding. Therefore, anticollagen therapy of covering the collagen surface was proposed as an alternative in our previous study, and an antithrombotic peptide LWWNSYY was designed and validated. However, its application was hindered due to the poor water solubility. In the present study, in order to develop a novel antithrombotic peptide with enhanced water solubility, redesigning of LWWNSYY to LEKNSTY using the EK pattern was proposed. Improved solubility was obtained for LEKNSTY. Moreover, the binding of LEKNSTY on the collagen surface was confirmed by molecular docking, molecular dynamics simulations, and experimental validation. A Kd of 0.91 ± 0.44 μM was observed. The effective inhibition of platelet adhesion on the collagen surface by LEKNSTY was demonstrated at an IC50 of 2.48 ± 0.59 μg/mL. Therefore, the successful design of the antithrombotic peptide LEKNSTY was confirmed, which would facilitate the research into the interface involving thrombus and the development of antithrombotic agents.
Collapse
Affiliation(s)
- Si Zheng
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, People's Republic of China
| | - Tanyi Hao
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, People's Republic of China
| | - Lin Zhang
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, People's Republic of China
| |
Collapse
|
50
|
Ji X, Tian L, Yao S, Han F, Niu S, Qu C. A Systematic Review of Body Fluids Biomarkers Associated With Early Neurological Deterioration Following Acute Ischemic Stroke. Front Aging Neurosci 2022; 14:918473. [PMID: 35711907 PMCID: PMC9196239 DOI: 10.3389/fnagi.2022.918473] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/10/2022] [Indexed: 11/17/2022] Open
Abstract
Biomarkers are objectively measured biological properties of normal and pathological processes. Early neurological deterioration (END) refers to the deterioration of neurological function in a short time after the onset of acute ischemic stroke (AIS) and is associated with adverse outcomes. Although multiple biomarkers have been found to predict END, there are currently no suitable biomarkers to be applied in routine stroke care. According to the Preferred Reporting Items for Systematic Review standards, we present a systematic review, concentrating on body fluids biomarkers that have shown potential to be transferred into clinical practice. We also describe newly reported body fluids biomarkers that can supply different insights into the mechanism of END. In our review, 40 scientific papers were included. Depending on the various mechanisms, sources or physicochemical characteristics of body fluids biomarkers, we classified related biomarkers as inflammation, protease, coagulation, metabolism, oxidative stress, and excitatory neurotoxicity. The body fluids biomarkers whose related articles are limited or mechanisms are unknown are categorized as other biomarkers. The inflammation-related biomarkers, such as neutrophil-to-lymphocyte ratio and hypersensitive C-reactive protein, play a crucial role among the mentioned biomarkers. Considering the vast heterogeneity of stroke progression, using a single body fluids biomarker may not accurately predict the risk of stroke progression, and it is necessary to combine multiple biomarkers (panels, scores, or indices) to improve their capacity to estimate END.
Collapse
Affiliation(s)
- Xiaotan Ji
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, China
- Department of Neurology, Jining No. 1 People’s Hospital, Jining, China
| | - Long Tian
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shumei Yao
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Fengyue Han
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Shenna Niu
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Chuanqiang Qu
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, China
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Chuanqiang Qu,
| |
Collapse
|