1
|
Zahid R, Wang J, Cai Z, Ishtiaq A, Liu M, Ma D, Liang Y, Xu Y. Single chain fragment variable, a new theranostic approach for cardiovascular diseases. Front Immunol 2024; 15:1443290. [PMID: 39735545 PMCID: PMC11671482 DOI: 10.3389/fimmu.2024.1443290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/26/2024] [Indexed: 12/31/2024] Open
Abstract
Cardiovascular diseases (CVDs) remain a significant global health challenge, leading to substantial morbidity and mortality. Despite recent advancements in CVD management, pharmaceutical treatments often suffer from poor pharmacokinetics and high toxicity. With the rapid progress of modern molecular biology and immunology, however, single-chain fragment variable (scFv) molecule engineering has emerged as a promising theranostic tool to offer specificity and versatility in targeting CVD-related antigens. To represent the latest development on the potential of scFv in the context of CVDs, this review summarized the new mechanism of action and applications as therapeutic, as well as diagnostic agents. Furthermore, the advantages of scFv, including its small size, ease of modification, and ability to be engineered for enhanced affinity and specificity, are also described. Finally, such challenges as immunogenicity, stability, and scalability, alongside strategies to overcome these hurdles, are deeply scrutinized to provide safer and more effective strategies for the diagnosis and treatment of the incurable CVDs.
Collapse
Affiliation(s)
- Rukhshan Zahid
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Juncheng Wang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Zecheng Cai
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Ayesha Ishtiaq
- College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Meng Liu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Dan Ma
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Yan Liang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| | - Yuekang Xu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, Anhui, China
| |
Collapse
|
2
|
Zhang Y, Ren Y, Zhou T, Qian Z, Bao Z. Vav family exchange factors: Potential regulator in atherosclerosis. Biochem Biophys Rep 2024; 40:101878. [PMID: 39649800 PMCID: PMC11625217 DOI: 10.1016/j.bbrep.2024.101878] [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: 08/02/2024] [Revised: 10/26/2024] [Accepted: 11/14/2024] [Indexed: 12/11/2024] Open
Abstract
The Vav family of guanosine nucleotide exchange factors (GEFs) regulates the phosphorylation of tyrosinase, influencing various physiological and pathological processes by modulating the binding of Rho GTPases to GDP/GTP. Recent research has highlighted the critical role of Vav family activation in tumorigenesis, neurological disorders, immune-related dysfunctions, and other diseases. This review offers a comprehensive overview of the structure and function of Vav proteins and their significant impact on the pathophysiology of atherosclerosis. In addition, we pay attention to the development of diagnostic and therapeutic targets centered around Vav proteins.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, 214002, China
| | - Yongwei Ren
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, 214002, China
| | - Tao Zhou
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, 214002, China
| | - Zhengtao Qian
- Department of Clinical Laboratory, Changshu Medicine Examination Institute, Changshu, 215500, China
| | - Zhengyang Bao
- Department of Internal Medicine, Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, 214002, China
| |
Collapse
|
3
|
Schaale D, Laspa Z, Balmes A, Sigle M, Dicenta-Baunach V, Hochuli R, Fu X, Serafimov K, Castor T, Harm T, Müller KAL, Rohlfing AK, Laufer S, Schäffer TE, Lämmerhofer M, Gawaz M. Hemin promotes platelet activation and plasma membrane disintegration regulated by the subtilisin-like proprotein convertase furin. FASEB J 2024; 38:e70155. [PMID: 39530531 DOI: 10.1096/fj.202400863rr] [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: 04/17/2024] [Revised: 10/14/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Platelet activation plays a critical role in thrombosis and hemostasis. Several pathophysiological situations lead to hemolysis, resulting in the liberation of free ferric iron-containing hemin. Hemin has been shown to activate platelets and induce thrombo-inflammation. Classical antiplatelet therapy failed to prevent hemin-induced platelet activation. Thus, the aim of the present study was to characterize the mechanism of hemin-induced platelet death (ferroptosis). We evaluated the in vitro effect of hemin on platelet activation, signaling, oxylipins, and plasma membrane destruction using light transmission aggregometry, ex vivo thrombus formation, multiparametric flow cytometry, micro-UHPLC mass spectrometry for oxylipin profiling, and scanning ion conductance microscopy (SICM). We found that hemin induces platelet cell death indicated by increased ROS levels, phosphatidyl serine (PS) exposure, and loss of mitochondrial membrane potential (ΔΨm). Further, hemin causes lipid peroxidation and generation of distinct oxylipins, which strongly affects plasma membrane integrity leading to generation of platelet-derived microvesicles. Interestingly, hemin-dependent platelet death (ferroptosis) is specifically regulated by the subtilisin-like proprotein convertase furin. In summary, platelet undergo a non-apoptotic cell death mediated by furin. Inhibition of furin may offer a therapeutic strategy to control hemin-induced thrombosis and thrombo-inflammation at a site of hemolysis.
Collapse
Affiliation(s)
- David Schaale
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Zoi Laspa
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Aylin Balmes
- Institute of Applied Physics, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Manuel Sigle
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Valerie Dicenta-Baunach
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Ravi Hochuli
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Xiaoqing Fu
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical and Medicinal Chemistry, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Kristian Serafimov
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical and Medicinal Chemistry, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Tatsiana Castor
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Tobias Harm
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Karin Anne Lydia Müller
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Anne-Katrin Rohlfing
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Stefan Laufer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical and Medicinal Chemistry, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Tilman E Schäffer
- Institute of Applied Physics, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Michael Lämmerhofer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical and Medicinal Chemistry, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
4
|
Striesow J, Nasri Z, von Woedtke T, Bekeschus S, Wende K. Epilipidomics reveals lipid fatty acid and headgroup modification in gas plasma-oxidized biomembranes. Redox Biol 2024; 77:103343. [PMID: 39366067 PMCID: PMC11483335 DOI: 10.1016/j.redox.2024.103343] [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: 07/24/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 10/06/2024] Open
Abstract
Lipids, possessing unsaturated fatty acid chains and polar regions with nucleophilic heteroatoms, represent suitable oxidation targets for autologous and heterologous reactive species. Lipid peroxidation products (LPPs) are highly heterogeneous, including hydroperoxides, alkenals, chlorination, or glycation. Accordingly, delineation of lipid targets, species type, resulting products, and oxidation level remains challenging. To this end, liposomal biomimetic models incorporating a phosphatidylcholine, -ethanolamine, and a sphingomyelin were used to deconvolute effects on a single lipid scale to predict potential modification product outcomes. To introduce oxidative modifications, gas plasma technology, a powerful pro-oxidant tool to promote LPP formation by forming highly abundant reactive species in the gas and liquid phases, was employed to liposomes. The plasma parameters (gas type/combination) were modified to modulate the resulting species-profile and LPP formation by enriching specific reactive species types over others. HR-LC-MS (Münzel and et al., 2017) [2] was employed for LPP identification. Moreover, the heavy oxygen isotope 18O was used to trace O2-incorporation into LPPs, providing first information on the plasma-mediated lipid peroxidation mechanism. We found that combination of lipid class and gas composition predetermined the type of attack: admixture of O2 to the plasma and the presence of nitrogen atoms with free electrons in the molecule lead to chlorination of the amide bond and headgroup. Here, atomic oxygen driven formation of hypochlorite is the major reactive species. In contrast, POPC yields mainly to LPPs with oxidation of the oleic acid tail, especially truncations, epoxidation, and hydroperoxide formation. Here, singlet oxygen is assumingly the major driver. 18O labelling revealed that gas phase derived reactive species are dominantly incorporated into the LPPs, supporting previous findings on gas-liquid interface chemistry. In summary, we here provided the first insights into gas plasma-mediated lipid peroxidation, which, employed in more complex cell and tissue models, may support identifying mechanisms of actions in plasma medicine.
Collapse
Affiliation(s)
- Johanna Striesow
- Leibniz Institute for Plasma Science and Technology (INP), a member of the Leibniz Health Technologies Research Alliance, Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Zahra Nasri
- Leibniz Institute for Plasma Science and Technology (INP), a member of the Leibniz Health Technologies Research Alliance, Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Thomas von Woedtke
- Leibniz Institute for Plasma Science and Technology (INP), a member of the Leibniz Health Technologies Research Alliance, Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany; Institute for Hygiene and Environmental Medicine, Greifswald University Medical Center, Sauerbruchstr., 17475, Greifswald, Germany
| | - Sander Bekeschus
- Leibniz Institute for Plasma Science and Technology (INP), a member of the Leibniz Health Technologies Research Alliance, Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany; Department of Dermatology and Venerology, Rostock University Medical Center, Strempelstr. 13, 18057, Rostock, Germany.
| | - Kristian Wende
- Leibniz Institute for Plasma Science and Technology (INP), a member of the Leibniz Health Technologies Research Alliance, Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany.
| |
Collapse
|
5
|
Carpanedo L, Rund KM, Wende LM, Kampschulte N, Schebb NH. LC-HRMS analysis of phospholipids bearing oxylipins. Anal Chim Acta 2024; 1326:343139. [PMID: 39260917 DOI: 10.1016/j.aca.2024.343139] [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: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Several oxylipins including hydroxy- and epoxy-polyunsaturated fatty acids act as lipid mediators. In biological samples they can be present as non-esterified form, but the major part occurs esterified in phospholipids (PL) or other lipids. Esterified oxylipins are quantified indirectly after alkaline hydrolysis as non-esterified oxylipins. However, in this indirect analysis the information in which lipid class oxylipins are bound is lost. In this work, an untargeted liquid chromatography high-resolution mass spectrometry (LC-HRMS) method for the direct analysis of PL bearing oxylipins was developed. RESULTS Optimized reversed-phase LC separation achieved a sufficient separation of isobaric and isomeric PL from different lipid classes bearing oxylipin positional isomers. Individual PL species bearing oxylipins were identified based on retention time, precursor ion and characteristic product ions. The bound oxylipin could be characterized based on product ions resulting from the α-cleavage occurring at the hydroxy/epoxy group. PL sn-1/sn-2 isomers were identified based on the neutral loss of the fatty acyl in the sn-2 position. A total of 422 individual oxPL species from 7 different lipid classes i.e., PI, PS, PC, PE, PC-P, PC-O, and PE-P were detected in human serum and cells. This method enabled to determine in which PL class supplemented oxylipins are incorporated in HEK293 cells: 20:4;15OH, 20:4;14Ep, and 20:5;14Ep were mostly bound to PI. 20:4;8Ep and 20:5;8Ep were esterified to PC and PE while other oxylipins were mainly found in PC. SIGNIFICANCE The developed LC-HRMS method enables the comprehensive detection as well as the semi-quantification of isobaric and isomeric PL species bearing oxylipins. With this method, we show that the position of the oxidation has a great impact and directs the incorporation of oxylipins into the different PL classes in human cells.
Collapse
Affiliation(s)
- Laura Carpanedo
- Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Katharina M Rund
- Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Luca M Wende
- Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Nadja Kampschulte
- Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Nils Helge Schebb
- Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany.
| |
Collapse
|
6
|
Pirotton L, de Cartier d’Yves E, Bertrand L, Beauloye C, Horman S. Platelet lipidomics and de novo lipogenesis: impact on health and disease. Curr Opin Hematol 2024; 31:217-223. [PMID: 38727017 PMCID: PMC11296274 DOI: 10.1097/moh.0000000000000820] [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] [Indexed: 08/02/2024]
Abstract
PURPOSE OF REVIEW Lipids play vital roles in platelet structure, signaling, and metabolism. In addition to capturing exogenous lipids, platelets possess the capacity for de novo lipogenesis, regulated by acetyl-coA carboxylase 1 (ACC1). This review aims to cover the critical roles of platelet de novo lipogenesis and lipidome in platelet production, function, and diseases. RECENT FINDINGS Upon platelet activation, approximately 20% of the platelet lipidome undergoes significant modifications, primarily affecting arachidonic acid-containing species. Multiple studies emphasize the impact of de novo lipogenesis, with ACC1 as key player, on platelet functions. Mouse models suggest the importance of the AMPK-ACC1 axis in regulating platelet membrane arachidonic acid content, associated with TXA 2 secretion, and thrombus formation. In human platelets, ACC1 inhibition leads to reduced platelet reactivity. Remodeling of the platelet lipidome, alongside with de novo lipogenesis, is also crucial for platelet biogenesis. Disruptions in the platelet lipidome are observed in various pathological conditions, including cardiovascular and inflammatory diseases, with associations between these alterations and shifts in platelet reactivity highlighted. SUMMARY The platelet lipidome, partially regulated by ACC-driven de novo lipogenesis, is indispensable for platelet production and function. It is implicated in various pathological conditions involving platelets.
Collapse
Affiliation(s)
- Laurence Pirotton
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain)
| | - Emma de Cartier d’Yves
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain)
| | - Luc Bertrand
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain)
| | - Christophe Beauloye
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain)
- Department of Cardiovascular Intensive Care, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Sandrine Horman
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain)
| |
Collapse
|
7
|
Burla B, Oh J, Nowak A, Piraud N, Meyer E, Mei D, Bendt AK, Studt JD, Frey BM, Torta F, Wenk MR, Krayenbuehl PA. Plasma and platelet lipidome changes in Fabry disease. Clin Chim Acta 2024; 562:119833. [PMID: 38955246 DOI: 10.1016/j.cca.2024.119833] [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: 06/14/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Fabry disease (FD) is an X-linked lysosomal storage disorder characterized by the progressive accumulation of globotriaosylceramide (Gb3) leading to systemic manifestations such as chronic kidney disease, cardiomyopathy, and stroke. There is still a need for novel markers for improved FD screening and prognosis. Moreover, the pathological mechanisms in FD, which also include systemic inflammation and fibrosis, are not yet fully understood. METHODS Plasma and platelets were obtained from 11 ERT (enzyme-replacement therapy)-treated symptomatic, 4 asymptomatic FD patients, and 13 healthy participants. A comprehensive targeted lipidomics analysis was conducted quantitating more than 550 lipid species. RESULTS Sphingadiene (18:2;O2)-containing sphingolipid species, including Gb3 and galabiosylceramide (Ga2), were significantly increased in FD patients. Plasma levels of lyso-dihexosylceramides, sphingoid base 1-phosphates (S1P), and GM3 ganglioside were also altered in FD patients, as well as specific plasma ceramide ratios used in cardiovascular disease risk prediction. Gb3 did not increase in patients' platelets but displayed a high inter-individual variability in patients and healthy participants. Platelets accumulated, however, lyso-Gb3, acylcarnitines, C16:0-sphingolipids, and S1P. CONCLUSIONS This study identified lipidome changes in plasma and platelets from FD patients, a possible involvement of platelets in FD, and potential new markers for screening and monitoring of this disease.
Collapse
Affiliation(s)
- Bo Burla
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore.
| | - Jeongah Oh
- Precision Medicine Translational Research Program and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore.
| | - Albina Nowak
- Department of Internal Medicine, Psychiatric University Clinic Zurich, Switzerland; Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, Switzerland.
| | | | - Eduardo Meyer
- Swiss Red Cross (SRC), Zurich-Schlieren, Switzerland
| | - Ding Mei
- Precision Medicine Translational Research Program and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Anne K Bendt
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore
| | - Jan-Dirk Studt
- Division of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Beat M Frey
- Swiss Red Cross (SRC), Zurich-Schlieren, Switzerland
| | - Federico Torta
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore; Precision Medicine Translational Research Program and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Markus R Wenk
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore; Precision Medicine Translational Research Program and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| | - Pierre-Alexandre Krayenbuehl
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, Switzerland; General Practice Brauereistrasse, Uster-Zurich, Switzerland.
| |
Collapse
|
8
|
Li N. Platelets as an inter-player between hyperlipidaemia and atherosclerosis. J Intern Med 2024; 296:39-52. [PMID: 38704820 DOI: 10.1111/joim.13794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Platelet hyperreactivity and hyperlipidaemia contribute significantly to atherosclerosis. Thus, it is desirable to review the platelet-hyperlipidaemia interplay and its impact on atherogenesis. Native low-density lipoprotein (nLDL) and oxidized LDL (oxLDL) are the key proatherosclerotic components of hyperlipidaemia. nLDL binds to the platelet-specific LDL receptor (LDLR) ApoE-R2', whereas oxLDL binds to the platelet-expressed scavenger receptor CD36, lectin-type oxidized LDLR 1 and scavenger receptor class A 1. Ligation of nLDL/oxLDL induces mild platelet activation and may prime platelets for other platelet agonists. Platelets, in turn, can modulate lipoprotein metabolisms. Platelets contribute to LDL oxidation by enhancing the production of reactive oxygen species and LDLR degradation via proprotein convertase subtilisin/kexin type 9 release. Platelet-released platelet factor 4 and transforming growth factor β modulate LDL uptake and foam cell formation. Thus, platelet dysfunction and hyperlipidaemia work in concert to aggravate atherogenesis. Hypolipidemic drugs modulate platelet function, whereas antiplatelet drugs influence lipid metabolism. The research prospects of the platelet-hyperlipidaemia interplay in atherosclerosis are also discussed.
Collapse
Affiliation(s)
- Nailin Li
- Karolinska Institutet, Department of Medicine-Solna, Division of Cardiovascular Medicine, Stockholm, Sweden
| |
Collapse
|
9
|
Yang W, Feng R, Peng G, Wang Z, Cen M, Jing Y, Feng W, Long T, Liu Y, Li Z, Huang K, Chang G. Glycoursodeoxycholic Acid Alleviates Arterial Thrombosis via Suppressing Diacylglycerol Kinases Activity in Platelet. Arterioscler Thromb Vasc Biol 2024; 44:1283-1301. [PMID: 38572646 DOI: 10.1161/atvbaha.124.320728] [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] [Accepted: 03/19/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Glycoursodeoxycholic acid (GUDCA) has been acknowledged for its ability to regulate lipid homeostasis and provide benefits for various metabolic disorders. However, the impact of GUDCA on arterial thrombotic events remains unexplored. The objective of this study is to examine the effects of GUDCA on thrombogenesis and elucidate its underlying mechanisms. METHODS Plasma samples from patients with arterial thrombotic events and diet-induced obese mice were collected to determine the GUDCA concentrations using mass spectrometry. Multiple in vivo murine thrombosis models and in vitro platelet functional assays were conducted to comprehensively evaluate the antithrombotic effects of GUDCA. Moreover, lipidomic analysis was performed to identify the alterations of intraplatelet lipid components following GUDCA treatment. RESULTS Plasma GUDCA level was significantly decreased in patients with arterial thrombotic events and negatively correlated with thrombotic propensity in diet-induced obese mice. GUDCA exhibited prominent suppressing effects on platelet reactivity as evidenced by the attenuation of platelet activation, secretion, aggregation, spreading, and retraction (P<0.05). In vivo, GUDCA administration robustly alleviated thrombogenesis (P<0.05) without affecting hemostasis. Mechanistically, GUDCA inhibited DGK (diacylglycerol kinase) activity, leading to the downregulation of the phosphatidic acid-mediated signaling pathway. Conversely, phosphatidic acid supplementation was sufficient to abolish the antithrombotic effects of GUDCA. More importantly, long-term oral administration of GUDCA normalized the enhanced DGK activity, thereby remarkably alleviating the platelet hyperreactivity as well as the heightened thrombotic tendency in diet-induced obese mice (P<0.05). CONCLUSIONS Our study implicated that GUDCA reduces platelet hyperreactivity and improves thrombotic propensity by inhibiting DGKs activity, which is a potentially effective prophylactic approach and promising therapeutic agent for arterial thrombotic events.
Collapse
Affiliation(s)
- Wenchao Yang
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Ruijia Feng
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Guiyan Peng
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Zhecun Wang
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Meifeng Cen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, China (M.C.)
| | - Yexiang Jing
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Weiqi Feng
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Ting Long
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Yunchong Liu
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Zilun Li
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Kan Huang
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Guangqi Chang
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| |
Collapse
|
10
|
Akther F, Fallahi H, Zhang J, Nguyen NT, Ta HT. Evaluating thrombosis risk and patient-specific treatment strategy using an atherothrombosis-on-chip model. LAB ON A CHIP 2024; 24:2927-2943. [PMID: 38591995 DOI: 10.1039/d4lc00131a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Platelets play an essential role in thrombotic processes. Recent studies suggest a direct link between increased plasma glucose, lipids, and inflammatory cytokines with platelet activation and aggregation, resulting in an increased risk of atherothrombotic events in cardiovascular patients. Antiplatelet therapies are commonly used for the primary prevention of atherosclerosis. Transitioning from a population-based strategy to patient-specific care requires a better understanding of the risks and advantages of antiplatelet therapy for individuals. This proof-of-concept study evaluates the potential to assess an individual's risk of forming atherothrombosis using a dual-channel microfluidic model emulating multiple atherogenic factors in vitro, including high glucose, high cholesterol, and inflammatory cytokines along with stenosis vessel geometry. The model shows precise sensitivity toward increased plasma glucose, cholesterol, and tumour necrosis factor-alpha (TNF-α)-treated groups in thrombus formation. An in vivo-like dose-dependent increment in platelet aggregation is observed in different treated groups, benefiting the evaluation of thrombosis risk in the individual condition. Moreover, the model could help decide the effective dosing of aspirin in multi-factorial complexities. In the high glucose-treated group, a 50 μM dose of aspirin could significantly reduce platelet aggregation, while a 100 μM dose of aspirin was required to reduce platelet aggregation in the glucose-TNF-α-treated group, which proves the model's potentiality as a tailored tool for customised therapy.
Collapse
Affiliation(s)
- Fahima Akther
- Queensland Micro- and Nanotechnology, Griffith University, Nathan Campus, Brisbane, Queensland 4111, Australia.
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Hedieh Fallahi
- Queensland Micro- and Nanotechnology, Griffith University, Nathan Campus, Brisbane, Queensland 4111, Australia.
- School of Environment and Science, Griffith University, Nathan, Queensland 4111, Australia
| | - Jun Zhang
- Queensland Micro- and Nanotechnology, Griffith University, Nathan Campus, Brisbane, Queensland 4111, Australia.
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology, Griffith University, Nathan Campus, Brisbane, Queensland 4111, Australia.
- School of Environment and Science, Griffith University, Nathan, Queensland 4111, Australia
| | - Hang Thu Ta
- Queensland Micro- and Nanotechnology, Griffith University, Nathan Campus, Brisbane, Queensland 4111, Australia.
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland 4072, Australia
- School of Environment and Science, Griffith University, Nathan, Queensland 4111, Australia
| |
Collapse
|
11
|
Harm T, Fu X, Frey M, Dittrich K, Brun A, Castor T, Borst O, Müller KAL, Geisler T, Rath D, Lämmerhofer M, Gawaz MP. Machine learning insights into thrombo-ischemic risks and bleeding events through platelet lysophospholipids and acylcarnitine species. Sci Rep 2024; 14:6089. [PMID: 38480746 PMCID: PMC10937715 DOI: 10.1038/s41598-024-56304-x] [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: 12/29/2023] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
Coronary artery disease (CAD) often leads to adverse events resulting in significant disease burdens. Underlying risk factors often remain inapparent prior to disease incidence and the cardiovascular (CV) risk is not exclusively explained by traditional risk factors. Platelets inherently promote atheroprogression and enhanced platelet functions and distinct platelet lipid species are associated with disease severity in patients with CAD. Lipidomics data were acquired using mass spectrometry and processed alongside clinical data applying machine learning to model estimates of an increased CV risk in a consecutive CAD cohort (n = 595). By training machine learning models on CV risk measurements, stratification of CAD patients resulted in a phenotyping of risk groups. We found that distinct platelet lipids are associated with an increased CV or bleeding risk and independently predict adverse events. Notably, the addition of platelet lipids to conventional risk factors resulted in an increased diagnostic accuracy of patients with adverse CV events. Thus, patients with aberrant platelet lipid signatures and platelet functions are at elevated risk to develop adverse CV events. Machine learning combining platelet lipidome data and common clinical parameters demonstrated an increased diagnostic value in patients with CAD and might improve early risk discrimination and classification for CV events.
Collapse
Affiliation(s)
- Tobias Harm
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Xiaoqing Fu
- Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | - Moritz Frey
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Kristina Dittrich
- Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | - Adrian Brun
- Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | - Tatsiana Castor
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Karin Anne Lydia Müller
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Tobias Geisler
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Dominik Rath
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Michael Lämmerhofer
- Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | - Meinrad Paul Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany.
| |
Collapse
|
12
|
Hajeyah AA, Protty MB, Paul D, Costa D, Omidvar N, Morgan B, Iwasaki Y, McGill B, Jenkins PV, Yousef Z, Allen-Redpath K, Soyama S, Choudhury A, Mitra R, Yaqoob P, Morrissey JH, Collins PW, O'Donnell VB. Phosphatidylthreonine is a procoagulant lipid detected in human blood and elevated in coronary artery disease. J Lipid Res 2024; 65:100484. [PMID: 38103786 PMCID: PMC10809103 DOI: 10.1016/j.jlr.2023.100484] [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: 06/19/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023] Open
Abstract
Aminophospholipids (aPL) such as phosphatidylserine are essential for supporting the activity of coagulation factors, circulating platelets, and blood cells. Phosphatidylthreonine (PT) is an aminophospholipid previously reported in eukaryotic parasites and animal cell cultures, but not yet in human tissues. Here, we evaluated whether PT is present in blood cells and characterized its ability to support coagulation. Several PT molecular species were detected in human blood, washed platelets, extracellular vesicles, and isolated leukocytes from healthy volunteers using liquid chromatography-tandem mass spectrometry. The ability of PT to support coagulation was demonstrated in vitro using biochemical and biophysical assays. In liposomes, PT supported prothrombinase activity in the presence and absence of phosphatidylserine. PT nanodiscs strongly bound FVa and lactadherin (nM affinity) but poorly bound prothrombin and FX, suggesting that PT supports prothrombinase through recruitment of FVa. PT liposomes bearing tissue factor poorly generated thrombin in platelet poor plasma, indicating that PT poorly supports extrinsic tenase activity. On platelet activation, PT is externalized and partially metabolized. Last, PT was significantly higher in platelets and extracellular vesicle from patients with coronary artery disease than in healthy controls. In summary, PT is present in human blood, binds FVa and lactadherin, supports coagulation in vitro through FVa binding, and is elevated in atherosclerotic vascular disease. Our studies reveal a new phospholipid subclass, that contributes to the procoagulant membrane, and may support thrombosis in patients at elevated risk.
Collapse
Affiliation(s)
- Ali A Hajeyah
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom; Department of Biological Sciences, Kuwait University, Safat, Kuwait.
| | - Majd B Protty
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Divyani Paul
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Daniela Costa
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Nader Omidvar
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Bethan Morgan
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Yugo Iwasaki
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Japan
| | - Beth McGill
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | | | - Zaheer Yousef
- University Hospital of Wales, Cardiff, United Kingdom
| | - Keith Allen-Redpath
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Shin Soyama
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | | | - Rito Mitra
- University Hospital of Wales, Cardiff, United Kingdom
| | - Parveen Yaqoob
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - James H Morrissey
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Peter W Collins
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom; University Hospital of Wales, Cardiff, United Kingdom
| | - Valerie B O'Donnell
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom.
| |
Collapse
|
13
|
Chicanne G, Darcourt J, Bertrand-Michel J, Garcia C, Ribes A, Payrastre B. What can we learn from the platelet lipidome? Platelets 2023; 34:2182180. [PMID: 36880158 DOI: 10.1080/09537104.2023.2182180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Besides their proteome, platelets use, in all responses to the environmental cues, a huge and diverse family of hydrophobic and amphipathic small molecules involved in structural, metabolic and signaling functions; the lipids. Studying how platelet lipidome changes modulate platelet function is an old story constantly renewed through the impressive technical advances allowing the discovery of new lipids, functions and metabolic pathways. Technical progress in analytical lipidomic profiling by top-of-the-line approaches such as nuclear magnetic resonance and gas chromatography or liquid chromatography coupled to mass spectrometry enables either large-scale analysis of lipids or targeted lipidomics. With the support of bioinformatics tools and databases, it is now possible to investigate thousands of lipids over a concentration range of several orders of magnitude. The lipidomic landscape of platelets is considered a treasure trove, not only able to expand our knowledge of platelet biology and pathologies but also to bring diagnostic and therapeutic opportunities. The aim of this commentary article is to summarize the advances in the field and to highlight what lipidomics can tell us about platelet biology and pathophysiology.
Collapse
Affiliation(s)
- Gaëtan Chicanne
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France
| | - Jean Darcourt
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France
| | - Justine Bertrand-Michel
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France.,MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
| | - Cédric Garcia
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France.,Laboratory of Haematology, University Hospital of Toulouse, Toulouse, France
| | - Agnès Ribes
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France.,Laboratory of Haematology, University Hospital of Toulouse, Toulouse, France
| | - Bernard Payrastre
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France.,Laboratory of Haematology, University Hospital of Toulouse, Toulouse, France
| |
Collapse
|
14
|
Zhang X, Gong Z, Shen Y, Cai Z, Yang L, Zhang T, Li W, Zhao Y, Zhu S, Liu C, Wang J, Wang X, Qi R, Liu J, Lei X, Wang W, Jiang C, Fu Y, Kong W. Alkaline ceramidase 1-mediated platelet ceramide catabolism mitigates vascular inflammation and abdominal aortic aneurysm formation. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1173-1189. [PMID: 39196139 DOI: 10.1038/s44161-023-00364-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 10/12/2023] [Indexed: 08/29/2024]
Abstract
Abdominal aortic aneurysm (AAA) is a highly lethal vascular disease. The role of platelets in AAA remains incompletely understood. Here we show that platelet ceramides, rather than other phospholipids, were elevated in an angiotensin II (AngII)-induced AAA murine model and in patients with AAA by using targeted lipidomic analysis. Among key ceramide metabolism enzymes, alkaline ceramidase 1 (Acer1) hydrolyzing ceramides were exclusively downregulated in AAA platelets. Platelet-specific Acer1 knockout mice were more susceptible to AAA upon AngII infusion without affecting hemostasis and thrombosis. Mechanistically, Acer1 deficiency in platelets facilitated platelet pro-inflammatory cytokine secretion as well as P-selectin-mediated circulating platelet-leukocyte aggregation and infiltration in aortic walls via the ceramide-p38 MAPK signaling axis. Of note, AngII repressed Acer1 expression in platelets by decreasing HuR-dependent mRNA stability. In conclusion, Acer1-mediated ceramide degradation in platelets exhibited anti-inflammatory effects and ameliorated AAA formation, potentially serving as a therapeutic target for AAA and other inflammatory vascular diseases.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yicong Shen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Zeyu Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Liu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Tao Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, China
| | - Weihao Li
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, China
| | - Yang Zhao
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Shirong Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Cihang Liu
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jin Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Xian Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Ruomei Qi
- Key Laboratory of Geriatrics, Beijing Hospital and Beijing Institute of Geriatrics, Ministry of Health, Beijing, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoguang Lei
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Wengong Wang
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| |
Collapse
|
15
|
Harm T, Dittrich K, Brun A, Fu X, Frey M, Petersen Uribe A, Schwarz FJ, Rohlfing AK, Castor T, Geisler T, Rath D, Lämmerhofer M, Gawaz MP. Large-scale lipidomics profiling reveals characteristic lipid signatures associated with an increased cardiovascular risk. Clin Res Cardiol 2023; 112:1664-1678. [PMID: 37470807 PMCID: PMC10584760 DOI: 10.1007/s00392-023-02260-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/27/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND AND AIMS Patients with cardiovascular disease (CVD) are at high risk to develop adverse events. The distinct risk of developing adverse cardiovascular (CV) events is not solely explained by traditional risk factors. Platelets are essentially involved in progression of CVD including coronary artery disease (CAD) and platelet hyperreactivity leads to development of adverse CV events. Alterations in the platelet lipidome lead to platelet hyperresponsiveness and thus might alter the individual risk profile. In this study, we investigate the platelet lipidome of CAD patients by untargeted lipidomics and elucidate alterations in the lipid composition of patients with adverse CV events. METHODS We characterized the platelet lipidome in a large consecutive CAD cohort (n = 1057) by an untargeted lipidomics approach using liquid chromatography coupled to mass spectrometry. RESULTS The platelet lipidome in this study identified 767 lipids and characteristic changes occurred in patients with adverse CV events. The most prominent upregulated lipids in patients with cardiovascular events primarily belong to the class of phospholipids and fatty acyls. Further, upregulated platelet lipids are associated with an increased cardiovascular or bleeding risk and independently associated with adverse events. In addition, alterations of the platelet lipidome are associated with modulation of in vitro platelet functions. CONCLUSIONS Our results reveal that the composition of the platelet lipidome is altered in CVD patients with an increased cardiovascular risk and distinct platelet lipids may indicate adverse events. Results of this study may contribute to improved risk discrimination and classification for cardiovascular events in patients with CVD. Main findings of this study and hypothetical impact of altered platelet lipid signatures in patients with adverse cardiovascular events on platelet function and clinical outcome. LPE lysophosphatidylethanolamines, CAR acylcarnitines, FA fatty acids.
Collapse
Affiliation(s)
- Tobias Harm
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Kristina Dittrich
- Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | - Adrian Brun
- Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | - Xiaoqing Fu
- Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | - Moritz Frey
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Alvaro Petersen Uribe
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Frederic-Joaquim Schwarz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Anne-Katrin Rohlfing
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Tatsiana Castor
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Tobias Geisler
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Dominik Rath
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Michael Lämmerhofer
- Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | - Meinrad P Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany.
| |
Collapse
|
16
|
Striesow J, Wesche J, McKitterick N, Busch LM, von Woedtke T, Greinacher A, Bekeschus S, Wende K. Gas plasma-induced platelet activation corresponds to reactive species profiles and lipid oxidation. Free Radic Biol Med 2023; 207:212-225. [PMID: 37490986 DOI: 10.1016/j.freeradbiomed.2023.07.024] [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: 06/20/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 07/27/2023]
Abstract
Surgical-induced hemostasis is a critical step in the closure of incisions, which is frequently achieved via electrocauterization and subsequent tissue necrotization. The latter is associated with postoperative complications. Recent in vivo work suggested reactive species-producing gas plasma technology as a pro-homeostatic agent acting via platelet activation. However, it remained elusive how platelet activation is linked to lipid and protein oxidation and the reactive species compositions. A direct relation between the reactive species composition and platelet activation was revealed by assessing the production of several reactive species and by using antioxidants. In addition, platelet lipidome and proteome analysis identified significantly regulated key lipids in the platelet activation pathway, such as diacylglycerols and phosphatidylinositol as well as oxylipins like thromboxanes. Lipid oxidation products mainly derived from phosphatidylethanolamine and phosphatidylserine species were observed at modest levels. In addition, oxidative post-translational modifications were identified on key proteins of the hemostasis machinery. This study provides new insights into oxidation-induced platelet activation in general and suggests a potential role of those processes in gas plasma-mediated hemostasis in particular.
Collapse
Affiliation(s)
- Johanna Striesow
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Jan Wesche
- Institute of Transfusion Medicine, Greifswald University Medical Center, Sauerbruchstr., 17475, Greifswald, Germany
| | - Nicholas McKitterick
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Larissa M Busch
- Interfaculty Institute for Genetics and Functional Genomics, Greifswald University, Felix-Hausdorff-Str. 8, 17475, Greifswald, Germany
| | - Thomas von Woedtke
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany; Institute for Hygiene and Environmental Medicine, Greifswald University Medical Center, Sauerbruchstr., 17475, Greifswald, Germany
| | - Andreas Greinacher
- Institute of Transfusion Medicine, Greifswald University Medical Center, Sauerbruchstr., 17475, Greifswald, Germany
| | - Sander Bekeschus
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany; Clinic and Policlinic for Dermatology and Venerology, Rostock University Medical Center, Strempelstr. 13, 18057, Rostock, Germany.
| | - Kristian Wende
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany.
| |
Collapse
|
17
|
Gawaz M, Geisler T, Borst O. Current concepts and novel targets for antiplatelet therapy. Nat Rev Cardiol 2023; 20:583-599. [PMID: 37016032 DOI: 10.1038/s41569-023-00854-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/06/2023]
Abstract
Platelets have a crucial role in haemostasis and atherothrombosis. Pharmacological control of platelet hyper-reactivity has become a cornerstone in the prevention of thrombo-ischaemic complications in atherosclerotic diseases. Current antiplatelet therapies substantially improve clinical outcomes in patients with coronary artery disease, but at the cost of increased risk of bleeding. Beyond their role in thrombosis, platelets are known to regulate inflammatory (thrombo-inflammatory) and microcirculatory pathways. Therefore, controlling platelet hyper-reactivity might have implications for both tissue inflammation (myocardial ischaemia) and vascular inflammation (vulnerable plaque formation) to prevent atherosclerosis. In this Review, we summarize the pathophysiological role of platelets in acute myocardial ischaemia, vascular inflammation and atherosclerotic progression. Furthermore, we highlight current clinical concepts of antiplatelet therapy that have contributed to improving patient care and have facilitated more individualized therapy. Finally, we discuss novel therapeutic targets and compounds for antiplatelet therapy that are currently in preclinical development, some of which have a more favourable safety profile than currently approved drugs with regard to bleeding risk. These novel antiplatelet targets might offer new strategies to treat cardiovascular disease.
Collapse
Affiliation(s)
- Meinrad Gawaz
- Department of Cardiology and Angiology, Eberhard Karls University of Tübingen, Tübingen, Germany.
| | - Tobias Geisler
- Department of Cardiology and Angiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
18
|
Ravera S, Signorello MG, Panfoli I. Platelet Metabolic Flexibility: A Matter of Substrate and Location. Cells 2023; 12:1802. [PMID: 37443836 PMCID: PMC10340290 DOI: 10.3390/cells12131802] [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: 05/17/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
Platelets are cellular elements that are physiologically involved in hemostasis, inflammation, thrombotic events, and various human diseases. There is a link between the activation of platelets and their metabolism. Platelets possess considerable metabolic versatility. Although the role of platelets in hemostasis and inflammation is known, our current understanding of platelet metabolism in terms of substrate preference is limited. Platelet activation triggers an oxidative metabolism increase to sustain energy requirements better than aerobic glycolysis alone. In addition, platelets possess extra-mitochondrial oxidative phosphorylation, which could be one of the sources of chemical energy required for platelet activation. This review aims to provide an overview of flexible platelet metabolism, focusing on the role of metabolic compartmentalization in substrate preference, since the metabolic flexibility of stimulated platelets could depend on subcellular localization and functional timing. Thus, developing a detailed understanding of the link between platelet activation and metabolic changes is crucial for improving human health.
Collapse
Affiliation(s)
- Silvia Ravera
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy;
| | | | - Isabella Panfoli
- Department of Pharmacy (DIFAR), University of Genoa, 16132 Genoa, Italy;
| |
Collapse
|
19
|
Zhou X, Huang X, Wu C, Ma Y, Li W, Hu J, Li R, Ya F. Sulforaphane attenuates glycoprotein VI-mediated platelet mitochondrial dysfunction through up-regulating the cAMP/PKA signaling pathway in vitro and in vivo. Food Funct 2023; 14:3613-3629. [PMID: 36946998 DOI: 10.1039/d2fo03958c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Platelet mitochondrial dysfunction is crucial for platelet activation, atherosclerosis and thrombosis. Sulforaphane (SFN) is a dietary isothiocyanate enriched in cruciferous vegetables and possesses multiple health benefits including cardiovascular protection. This study aims to investigate whether and how SFN modulates platelet mitochondrial dysfunction and hyperactivity in vitro and in vivo. Using a series of platelet functional assays in human platelets in vitro, we found that SFN at physiological concentrations attenuated oxidative stress-dependent platelet mitochondrial dysfunction (loss of mitochondrial membrane potential), apoptosis (cytochrome c release, caspase 3 activation and phosphatidylserine exposure) and activation induced by glycoprotein VI (GPVI) agonists (e.g., collagen and convulxin). Moreover, 12-week supplementation of SFN-enriched broccoli sprout extract (BSE, 0.06% diet) in C57BL/6J mice also attenuated GPVI-induced platelet mitochondrial dysfunction, apoptosis and hyperreactivity in vivo. Mechanistically, these inhibitory effects of SFN treatment and BSE supplementation were mainly mediated by up-regulating the cAMP/PKA pathway though decreasing phosphodiesterase 3A (PDE3A) activity. Thus, through modulating the PDE3A/cAMP/PKA signaling pathway, and attenuating platelet mitochondrial dysfunction and hyperreactivity, SFN may be a potent cardioprotective agent.
Collapse
Affiliation(s)
- Xinyu Zhou
- Department of Nutrition, School of Public Health, Dali University, Dali, Yunnan Province 671000, China.
| | - Xinhui Huang
- Department of Nutrition, School of Public Health, Dali University, Dali, Yunnan Province 671000, China.
| | - Chunting Wu
- Department of Nutrition, School of Public Health, Dali University, Dali, Yunnan Province 671000, China.
| | - Yongjie Ma
- Department of Nutrition, School of Public Health, Dali University, Dali, Yunnan Province 671000, China.
| | - Weiqi Li
- Department of Nutrition, School of Public Health, Dali University, Dali, Yunnan Province 671000, China.
| | - Jinqiu Hu
- Department of Nutrition, School of Public Health, Dali University, Dali, Yunnan Province 671000, China.
| | - Rong Li
- Department of Nutrition, School of Public Health, Dali University, Dali, Yunnan Province 671000, China.
| | - Fuli Ya
- Department of Nutrition, School of Public Health, Dali University, Dali, Yunnan Province 671000, China.
- Institute of Translational Medicine for Metabolic Diseases, Dali University, Dali, Yunnan Province 671000, China
| |
Collapse
|
20
|
Harm T, Frey M, Dittrich K, Goldschmied A, Rohlfing AK, Fu X, Brun A, Castor T, Rath D, Müller K, Lammerhofer M, Gawaz M. Statin Treatment Is Associated with Alterations in the Platelet Lipidome. Thromb Haemost 2023; 123:585-596. [PMID: 36898406 DOI: 10.1055/s-0043-1764353] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
BACKGROUND Platelets are key players in the pathophysiology of coronary artery disease (CAD) and platelet hyperreactivity leads to increased risk of developing adverse cardiovascular events. Further, significant changes in the platelet lipidome occur in patients with acute coronary syndrome (ACS) and critically regulated lipids lead to platelet hyperresponsiveness. Statin treatment is crucial in the treatment and prevention of patients with CAD by remodeling lipid metabolism. OBJECTIVE In this study, we investigate the platelet lipidome of CAD patients by untargeted lipidomics, highlighting significant changes between statin-treated and naïve patients. METHODS We characterized the platelet lipidome in a CAD cohort (n = 105) by an untargeted lipidomics approach using liquid chromatography coupled to mass spectrometry. RESULTS Among the annotated lipids, 41 lipids were significantly upregulated in statin-treated patients, whereas 6 lipids were downregulated compared to naïve patients. The most prominent upregulated lipids in statin-treated patients belong to the class of triglycerides, cholesteryl esters, palmitic acid, and oxidized phospholipids, whereas mainly glycerophospholipids were downregulated compared to untreated patients. A more pronounced effect of statin treatment on the platelet lipidome was observed in ACS patients. We further highlight a dose-dependent influence on the platelet lipidome. CONCLUSION Our results reveal that the platelet lipidome is altered in CAD patients with statin treatment and upregulated lipids embody mainly characteristic triglycerides, whereas downregulated lipids mostly compromise glycerophospholipids, which may play a role in the pathophysiology of CAD. Results of this study may contribute to the understanding of statin treatment softening the lipid phenotype.
Collapse
Affiliation(s)
- Tobias Harm
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Moritz Frey
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Kristina Dittrich
- Institute of Pharmaceutical Sciences, Pharmaceutical (Bio-)Analysis, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas Goldschmied
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Anne-Katrin Rohlfing
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Xiaoqing Fu
- Institute of Pharmaceutical Sciences, Pharmaceutical (Bio-)Analysis, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Adrian Brun
- Institute of Pharmaceutical Sciences, Pharmaceutical (Bio-)Analysis, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Tatsiana Castor
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Dominik Rath
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Karin Müller
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Michael Lammerhofer
- Institute of Pharmaceutical Sciences, Pharmaceutical (Bio-)Analysis, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
21
|
Classes of Lipid Mediators and Their Effects on Vascular Inflammation in Atherosclerosis. Int J Mol Sci 2023; 24:ijms24021637. [PMID: 36675152 PMCID: PMC9863938 DOI: 10.3390/ijms24021637] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 01/18/2023] Open
Abstract
It is commonly believed that the inactivation of inflammation is mainly due to the decay or cessation of inducers. In reality, in connection with the development of atherosclerosis, spontaneous decay of inducers is not observed. It is now known that lipid mediators originating from polyunsaturated fatty acids (PUFAs), which are important constituents of all cell membranes, can act in the inflamed tissue and bring it to resolution. In fact, PUFAs, such as arachidonic acid (AA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), are precursors to both pro-inflammatory and anti-inflammatory compounds. In this review, we describe the lipid mediators of vascular inflammation and resolution, and their biochemical activity. In addition, we highlight data from the literature that often show a worsening of atherosclerotic disease in subjects deficient in lipid mediators of inflammation resolution, and we also report on the anti-proteasic and anti-thrombotic properties of these same lipid mediators. It should be noted that despite promising data observed in both animal and in vitro studies, contradictory clinical results have been observed for omega-3 PUFAs. Many further studies will be required in order to clarify the observed conflicts, although lifestyle habits such as smoking or other biochemical factors may often influence the normal synthesis of lipid mediators of inflammation resolution.
Collapse
|
22
|
Nagy M, van der Meijden PEJ, Glunz J, Schurgers L, Lutgens E, ten Cate H, Heitmeier S, Spronk HMH. Integrating Mechanisms in Thrombotic Peripheral Arterial Disease. Pharmaceuticals (Basel) 2022; 15:1428. [PMID: 36422558 PMCID: PMC9695058 DOI: 10.3390/ph15111428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 11/10/2023] Open
Abstract
Peripheral arterial disease (PAD), a manifestation of systemic atherosclerosis, is underdiagnosed in the general population. Despite the extensive research performed to unravel its pathophysiology, inadequate knowledge exists, thus preventing the development of new treatments. This review aims to highlight the essential elements of atherosclerosis contributing to the pathophysiology of PAD. Furthermore, emphasis will be placed on the role of thrombo-inflammation, with particular focus on platelet and coagulation activation as well as cell-cell interactions. Additional insight will be then discussed to reveal the contribution of hypercoagulability to the development of vascular diseases such as PAD. Lastly, the current antithrombotic treatments will be discussed, and light will be shed on promising new targets aiming to aid the development of new treatments.
Collapse
Affiliation(s)
- Magdolna Nagy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Paola E. J. van der Meijden
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, 6229 ER Maastricht, The Netherlands
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
| | - Julia Glunz
- Cardiovascular Research, Bayer AG, 42117 Wuppertal, Germany
| | - Leon Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Esther Lutgens
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 10785 Munich, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian’s University, 80539 Munich, Germany
- Experimental Cardiovascular Immunology Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Hugo ten Cate
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, 6229 ER Maastricht, The Netherlands
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
- Department of Internal Medicine, Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
- Center for Thrombosis and Hemostasis, Gutenberg University Mainz, 55122 Mainz, Germany
| | | | - Henri M. H. Spronk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, 6229 ER Maastricht, The Netherlands
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
- Department of Internal Medicine, Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
| |
Collapse
|
23
|
Platelet Redox Imbalance in Hypercholesterolemia: A Big Problem for a Small Cell. Int J Mol Sci 2022; 23:ijms231911446. [PMID: 36232746 PMCID: PMC9570056 DOI: 10.3390/ijms231911446] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 11/17/2022] Open
Abstract
The imbalance between reactive oxygen species (ROS) synthesis and their scavenging by anti-oxidant defences is the common soil of many disorders, including hypercholesterolemia. Platelets, the smallest blood cells, are deeply involved in the pathophysiology of occlusive arterial thrombi associated with myocardial infarction and stroke. A great deal of evidence shows that both increased intraplatelet ROS synthesis and impaired ROS neutralization are implicated in the thrombotic process. Hypercholesterolemia is recognized as cause of atherosclerosis, cerebro- and cardiovascular disease, and, closely related to this, is the widespread acceptance that it strongly contributes to platelet hyperreactivity via direct oxidized LDL (oxLDL)-platelet membrane interaction via scavenger receptors such as CD36 and signaling pathways including Src family kinases (SFK), mitogen-activated protein kinases (MAPK), and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. In turn, activated platelets contribute to oxLDL generation, which ends up propagating platelet activation and thrombus formation through a mechanism mediated by oxidative stress. When evaluating the effect of lipid-lowering therapies on thrombogenesis, a large body of evidence shows that the effects of statins and proprotein convertase subtilisin/kexin type 9 inhibitors are not limited to the reduction of LDL-C but also to the down-regulation of platelet reactivity mainly by mechanisms sensitive to intracellular redox balance. In this review, we will focus on the role of oxidative stress-related mechanisms as a cause of platelet hyperreactivity and the pathophysiological link of the pleiotropism of lipid-lowering agents to the beneficial effects on platelet function.
Collapse
|
24
|
Glabridin, a Bioactive Flavonoid from Licorice, Effectively Inhibits Platelet Activation in Humans and Mice. Int J Mol Sci 2022; 23:ijms231911372. [PMID: 36232674 PMCID: PMC9570097 DOI: 10.3390/ijms231911372] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/16/2022] [Accepted: 09/22/2022] [Indexed: 11/17/2022] Open
Abstract
Platelets are crucial for hemostasis and arterial thrombosis, which may lead to severe cardiovascular diseases (CVDs). Thus, therapeutic agents must be developed to prevent pathological platelet activation. Glabridin, a major bioalkaloid extracted from licorice root, improves metabolic abnormalities (i.e., obesity and diabetes) and protects against CVDs and neuronal disorders. To the best of our knowledge, no studies have focused on glabridin’s effects on platelet activation. Therefore, we investigated these effects in humans and mice. Glabridin exhibited the highest inhibitory effects on collagen-stimulated platelet aggregation and moderate effects on arachidonic-acid-stimulated activation; however, no effects were observed for any other agonists (e.g., thrombin or U46619). Glabridin evidently reduced P-selectin expression, ATP release, and intracellular Ca2+ ([Ca2+]i) mobilization and thromboxane A2 formation; it further reduced the activation of phospholipase C (PLC)γ2/protein kinase C (PKC), phosphoinositide 3-kinase (PI3K)/Akt/glycogen synthase kinase-3β (GSK3β), mitogen-activated protein kinase (MAPK), and NF-κB. In mice, glabridin reduced the mortality rate caused by acute pulmonary thromboembolism without altering bleeding time. Thus, glabridin effectively inhibits the PLCγ2/PKC cascade and prevents the activation of the PI3K/Akt/GSK3β and MAPK pathways; this leads to a reduction in [Ca2+]i mobilization, which eventually inhibits platelet aggregation. Therefore, glabridin may be a promising therapeutic agent for thromboembolic disorders.
Collapse
|
25
|
Guan T, Emschermann F, Schories C, Groga-Bada P, Martus P, Borst O, Gawaz M, Geisler T, Rath D, Chatterjee M. Platelet SR-PSOX/CXCL16-CXCR6 Axis Influences Thrombotic Propensity and Prognosis in Coronary Artery Disease. Int J Mol Sci 2022; 23:ijms231911066. [PMID: 36232370 PMCID: PMC9570123 DOI: 10.3390/ijms231911066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/10/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Platelets express the transmembrane chemokine SR-PSOX/CXCL16, proteolytic cleavage of which generates the sCXCL16 soluble-(s) chemokine. The sCXCL16 engages CXCR6 on platelets to synergistically propagate degranulation, aggregation and thrombotic response. Currently, we have investigated the pro-thrombotic and prognostic association of platelet CXCL16−CXCR6 axis in CAD-(n = 240; CCS n = 62; ACS n = 178) patients. Platelet surface-associated-CXCL16 and CXCR6 surface expression ascertained by flow cytometry correlated significantly with platelet activation markers (CD62P denoting degranulation and PAC-1 binding denoting α2bβ3-integrin activation). Higher platelet CXCL16 surface association (1st quartile vs. 2nd−4th quartiles) corresponded to significantly elevated collagen-induced platelet aggregation assessed by whole blood impedance aggregometry. Platelet-CXCL16 and CXCR6 expression did not alter with dyslipidemia, triglyceride, total cholesterol, or LDL levels, but higher (>median) plasma HDL levels corresponded with decreased platelet-CXCL16 and CXCR6. Although platelet-CXCL16 and CXCR6 expression did not change significantly with or correlate with troponin I levels, they corresponded with higher Creatine Kinase-(CK) activity and progressively deteriorating left ventricular ejection fraction (LVEF) at admission. Elevated-(4th quartile) platelet-CXCL16 (p = 0.023) and CXCR6 (p = 0.030) measured at admission were significantly associated with a worse prognosis. However, after Cox-PH regression analysis, only platelet-CXCL16 was ascertained as an independent predictor for all-cause of mortality. Therefore, the platelet CXCL16−CXCR6 axis may influence thrombotic propensity and prognosis in CAD patients.
Collapse
Affiliation(s)
- Tianyun Guan
- Department of Cardiology and Angiology, University Hospital Tübingen, Otfried Müller Straße 10, 72076 Tübingen, Germany
| | - Frederic Emschermann
- Department of Cardiology and Angiology, University Hospital Tübingen, Otfried Müller Straße 10, 72076 Tübingen, Germany
| | - Christoph Schories
- Department of Cardiology and Angiology, University Hospital Tübingen, Otfried Müller Straße 10, 72076 Tübingen, Germany
| | - Patrick Groga-Bada
- Department of Cardiology and Angiology, University Hospital Tübingen, Otfried Müller Straße 10, 72076 Tübingen, Germany
| | - Peter Martus
- Institute for Clinical Epidemiology and Applied Biostatistics, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology, University Hospital Tübingen, Otfried Müller Straße 10, 72076 Tübingen, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Otfried Müller Straße 10, 72076 Tübingen, Germany
| | - Tobias Geisler
- Department of Cardiology and Angiology, University Hospital Tübingen, Otfried Müller Straße 10, 72076 Tübingen, Germany
| | - Dominik Rath
- Department of Cardiology and Angiology, University Hospital Tübingen, Otfried Müller Straße 10, 72076 Tübingen, Germany
- Correspondence: (D.R.); (M.C.); Tel.: +49-7071-2974944 (M.C.)
| | - Madhumita Chatterjee
- Department of Cardiology and Angiology, University Hospital Tübingen, Otfried Müller Straße 10, 72076 Tübingen, Germany
- Department of Pharmacology, Experimental Therapy and Toxicology, University Hospital Tübingen, Wilhelmstrasse 56, 72074 Tübingen, Germany
- Correspondence: (D.R.); (M.C.); Tel.: +49-7071-2974944 (M.C.)
| |
Collapse
|
26
|
Manke MC, Ahrends R, Borst O. Platelet lipid metabolism in vascular thrombo-inflammation. Pharmacol Ther 2022; 237:108258. [DOI: 10.1016/j.pharmthera.2022.108258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/12/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022]
|
27
|
Zhang M, Meng L, Chen Y, Li X, Shi L. CYP2C19 polymorphisms and lipoproteins associated with clopidogrel resistance in children with Kawasaki disease in China: A prospective study. Front Cardiovasc Med 2022; 9:925518. [PMID: 36072880 PMCID: PMC9441694 DOI: 10.3389/fcvm.2022.925518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
Background CYP2C19 genetic variation and clinical factors have been proved to be related with clopidogrel resistance (CR) in adults, while the presence of CR in children with Kawasaki disease (KD) was seldom reported. Our objective was to evaluate KD patients’ response to clopidogrel treatment and determine whether CYP2C19 gene polymorphisms and laboratory indicators are associated with CR in this population. Methods This was a prospective and single-center study. We recruited children with KD hospitalized in the cardiology department at the Children’s Hospital Capital Institute of Pediatrics between January 2019 and October 2021, and the distribution of the CYP2C19 gene polymorphisms was assessed. According to the light transmission aggregometry (LTA) test results, KD patients who were treated with clopidogrel were divided into CR group and non-CR (NCR) group. We also analyzed the influence of CYP2C19 gene polymorphisms and laboratory indicators on CR in children with KD. Results (1) A total of 346 children with KD were evaluated for the genotypic and phenotypic distributions of CYP2C19. Loss-of-function (LOF) mutated allele was included in 56.9% of CYP2C19 genotypes, and their corresponding phenotypes were intermediate metabolizers (46.2%) and poor metabolizers (10.7%). (2) The incidence of CR in this study population was 31.4%. The multivariate logistic regression showed that carrying CYP2C19 LOF allele (OR, 3.922; 95%CI, 1.504–10.282; P = 0.005) and high levels of low-density lipoprotein (OR, 1.675; 95%CI, 1.069–2.623; P = 0.024) were independent risk factor for CR, while low levels of high-density lipoprotein (OR, 0.120; 95%CI, 0.020, 0.734; P = 0.022) was an independent protective factor for CR. The area under the receiver operator characteristic curve of the multivariate logistic regression model (including high-density lipoprotein, low-density lipoprotein, and CYP2C19 LOF allele carriers) for predicting CR was 0.769 (95% CI, 0.674–0.863; P < 0.001). The sensitivity and specificity were 70.3 and 74.0%, respectively. Conclusion Carrying CYP2C19 LOF allele, low levels of high-density lipoprotein, and high levels of low-density lipoprotein were independent risk factors for CR in children with KD in China. This may benefit pediatricians in choosing appropriate individualized antiplatelet therapy.
Collapse
Affiliation(s)
- Mingming Zhang
- Department of Cardiology, Children’s Hospital Capital Institute of Pediatrics, Beijing, China
| | - Li Meng
- Department of Cardiology, Children’s Hospital Capital Institute of Pediatrics, Beijing, China
- Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, China
| | - Yeshi Chen
- Department of Cardiology, Children’s Hospital Capital Institute of Pediatrics, Beijing, China
- Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, China
| | - Xiaohui Li
- Department of Cardiology, Children’s Hospital Capital Institute of Pediatrics, Beijing, China
- Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, China
- *Correspondence: Xiaohui Li,
| | - Lin Shi
- Department of Cardiology, Children’s Hospital Capital Institute of Pediatrics, Beijing, China
| |
Collapse
|
28
|
Oxidised Low-Density Lipoprotein-Induced Platelet Hyperactivity—Receptors and Signalling Mechanisms. Int J Mol Sci 2022; 23:ijms23169199. [PMID: 36012465 PMCID: PMC9409144 DOI: 10.3390/ijms23169199] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/26/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
Dyslipidaemia leads to proatherogenic oxidative lipid stress that promotes vascular inflammation and thrombosis, the pathologies that underpin myocardial infarction, stroke, and deep vein thrombosis. These prothrombotic states are driven, at least in part, by platelet hyperactivity, and they are concurrent with the appearancxe of oxidatively modified low-density lipoproteins (LDL) in the circulation. Modified LDL are heterogenous in nature but, in a general sense, constitute a prototype circulating transporter for a plethora of oxidised lipid epitopes that act as danger-associated molecular patterns. It is well-established that oxidatively modified LDL promote platelet activation and arterial thrombosis through a number of constitutively expressed scavenger receptors, which transduce atherogenic lipid stress to a complex array of proactivatory signalling pathways in the platelets. Stimulation of these signalling events underlie the ability of modified LDL to induce platelet activation and blunt platelet inhibitory pathways, as well as promote platelet-mediated coagulation. Accumulating evidence from patients at risk of arterial thrombosis and experimental animal models of disease suggest that oxidised LDL represents a tangible link between the dyslipidaemic environment and increased platelet activation. The aim of this review is to summarise recent advances in our understanding of the pro-thrombotic signalling events induced in platelets by modified LDL ligation, describe the contribution of individual platelet scavenger receptors, and highlight potential future challenges of targeting these pathways.
Collapse
|
29
|
Schuurman AR, Léopold V, Pereverzeva L, Chouchane O, Reijnders TDY, Brabander JD, Douma RA, Weeghel MV, Wever E, Schomaker BV, Vaz FM, Wiersinga WJ, Veer CV, Poll TVD. The Platelet Lipidome Is Altered in Patients with COVID-19 and Correlates with Platelet Reactivity. Thromb Haemost 2022; 122:1683-1692. [PMID: 35850149 PMCID: PMC9512584 DOI: 10.1055/s-0042-1749438] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
BACKGROUND Activated platelets have been implicated in the proinflammatory and prothrombotic phenotype of coronavirus disease 2019 (COVID-19). While it is increasingly recognized that lipids have important structural and signaling roles in platelets, the lipidomic landscape of platelets during infection has remained unexplored. OBJECTIVE To investigate the platelet lipidome of patients hospitalized for COVID-19. METHODS We performed untargeted lipidomics in platelets of 25 patients hospitalized for COVID-19 and 23 noninfectious controls with similar age and sex characteristics, and with comparable comorbidities. RESULTS Twenty-five percent of the 1,650 annotated lipids were significantly different between the groups. The significantly altered part of the platelet lipidome mostly comprised lipids that were less abundant in patients with COVID-19 (20.4% down, 4.6% up, 75% unchanged). Platelets from COVID-19 patients showed decreased levels of membrane plasmalogens, and a distinct decrease of long-chain, unsaturated triacylglycerols. Conversely, platelets from patients with COVID-19 displayed class-wide higher abundances of bis(monoacylglycero)phosphate and its biosynthetic precursor lysophosphatidylglycerol. Levels of these classes positively correlated with ex vivo platelet reactivity-as measured by P-selectin expression after PAR1 activation-irrespective of disease state. CONCLUSION Taken together, this investigation provides the first exploration of the profound impact of infection on the human platelet lipidome, and reveals associations between the lipid composition of platelets and their reactivity. These results warrant further lipidomic research in other infections and disease states involving platelet pathophysiology.
Collapse
Affiliation(s)
- Alex R Schuurman
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Valentine Léopold
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Anesthesia and Intensive Care, Hôpital Lariboisière, INSERM U942S MASCOT, Université de Paris, Paris, France
| | - Liza Pereverzeva
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Osoul Chouchane
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Tom D Y Reijnders
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Justin de Brabander
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Renée A Douma
- Department of Internal Medicine, Flevo Hospital, Almere, The Netherlands
| | - Michel van Weeghel
- Departments of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Core Facility Metabolomics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Eric Wever
- Departments of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Core Facility Metabolomics, Amsterdam UMC, Amsterdam, The Netherlands.,Department of Epidemiology & Data Science, Bioinformatics Laboratory, Amsterdam Public Health Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Bauke V Schomaker
- Departments of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Core Facility Metabolomics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Frédéric M Vaz
- Departments of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Core Facility Metabolomics, Amsterdam UMC, Amsterdam, The Netherlands.,Department of Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Willem Joost Wiersinga
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, The Netherlands.,Division of Infectious Diseases, Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Cornelis Van't Veer
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, The Netherlands.,Division of Infectious Diseases, Amsterdam University Medical Centers - Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Chaudhary PK, Kim S, Kim S. An Insight into Recent Advances on Platelet Function in Health and Disease. Int J Mol Sci 2022; 23:ijms23116022. [PMID: 35682700 PMCID: PMC9181192 DOI: 10.3390/ijms23116022] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/22/2022] [Accepted: 05/24/2022] [Indexed: 12/04/2022] Open
Abstract
Platelets play a variety of roles in vascular biology and are best recognized as primary hemostasis and thrombosis mediators. Platelets have a large number of receptors and secretory molecules that are required for platelet functionality. Upon activation, platelets release multiple substances that have the ability to influence both physiological and pathophysiological processes including inflammation, tissue regeneration and repair, cancer progression, and spreading. The involvement of platelets in the progression and seriousness of a variety of disorders other than thrombosis is still being discovered, especially in the areas of inflammation and the immunological response. This review represents an integrated summary of recent advances on the function of platelets in pathophysiology that connects hemostasis, inflammation, and immunological response in health and disease and suggests that antiplatelet treatment might be used for more than only thrombosis.
Collapse
|
31
|
Ventura R, Martínez-Ruiz I, Hernández-Alvarez MI. Phospholipid Membrane Transport and Associated Diseases. Biomedicines 2022; 10:biomedicines10051201. [PMID: 35625937 PMCID: PMC9138374 DOI: 10.3390/biomedicines10051201] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 11/16/2022] Open
Abstract
Phospholipids are the basic structure block of eukaryotic membranes, in both the outer and inner membranes, which delimit cell organelles. Phospholipids can also be damaged by oxidative stress produced by mitochondria, for instance, becoming oxidized phospholipids. These damaged phospholipids have been related to prevalent diseases such as atherosclerosis or non-alcoholic steatohepatitis (NASH) because they alter gene expression and induce cellular stress and apoptosis. One of the main sites of phospholipid synthesis is the endoplasmic reticulum (ER). ER association with other organelles through membrane contact sites (MCS) provides a close apposition for lipid transport. Additionally, an important advance in this small cytosolic gap are lipid transfer proteins (LTPs), which accelerate and modulate the distribution of phospholipids in other organelles. In this regard, LTPs can be established as an essential point within phospholipid circulation, as relevant data show impaired phospholipid transport when LTPs are defected. This review will focus on phospholipid function, metabolism, non-vesicular transport, and associated diseases.
Collapse
Affiliation(s)
- Raúl Ventura
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (R.V.); (I.M.-R.)
| | - Inma Martínez-Ruiz
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (R.V.); (I.M.-R.)
| | - María Isabel Hernández-Alvarez
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (R.V.); (I.M.-R.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
- IBUB Universitat de Barcelona—Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
- Correspondence:
| |
Collapse
|
32
|
Protty MB, Jenkins PV, Collins PW, O'Donnell VB. The role of procoagulant phospholipids on the surface of circulating blood cells in thrombosis and haemostasis. Open Biol 2022; 12:210318. [PMID: 35440201 PMCID: PMC9019515 DOI: 10.1098/rsob.210318] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/21/2022] [Indexed: 01/09/2023] Open
Abstract
Phospholipids (PLs) are found in all cell types and are required for structural support and cell activation signalling pathways. In resting cells, PLs are asymmetrically distributed throughout the plasma membrane with native procoagulant aminophospholipids (aPLs) being actively maintained in the inner leaflet of the membrane. Upon platelet activation, aPLs rapidly externalize to the outer leaflet and are essential for supporting the coagulation cascade by providing binding sites for factors in the cell-based model. More recent work has uncovered a role for enzymatically oxidized PLs (eoxPLs) in facilitating coagulation, working in concert with native aPLs. Despite this, the role of aPLs and eoxPLs in thrombo-inflammatory conditions, such as arterial and venous thrombosis, has not been fully elucidated. In this review, we describe the biochemical structures, distribution and regulation of aPL externalization and summarize the literature on eoxPL generation in circulating blood cells. We focus on the currently understood role of these lipids in mediating coagulation reactions in vitro, in vivo and in human thrombotic disease. Finally, we highlight gaps in our understanding in how these lipids vary in health and disease, which may place them as future therapeutic targets for the management of thrombo-inflammatory conditions.
Collapse
Affiliation(s)
- Majd B. Protty
- Systems Immunity Research Institute, Cardiff University, Cardiff CF14 4XN, UK
| | - P. Vince Jenkins
- Systems Immunity Research Institute, Cardiff University, Cardiff CF14 4XN, UK
| | - Peter W. Collins
- Systems Immunity Research Institute, Cardiff University, Cardiff CF14 4XN, UK
| | | |
Collapse
|
33
|
Tyagi T, Jain K, Gu SX, Qiu M, Gu VW, Melchinger H, Rinder H, Martin KA, Gardiner EE, Lee AI, Ho Tang W, Hwa J. A guide to molecular and functional investigations of platelets to bridge basic and clinical sciences. NATURE CARDIOVASCULAR RESEARCH 2022; 1:223-237. [PMID: 37502132 PMCID: PMC10373053 DOI: 10.1038/s44161-022-00021-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 01/17/2022] [Indexed: 07/29/2023]
Abstract
Platelets have been shown to be associated with pathophysiological process beyond thrombosis, demonstrating critical additional roles in homeostatic processes, such as immune regulation, and vascular remodeling. Platelets themselves can have multiple functional states and can communicate and regulate other cells including immune cells and vascular smooth muscle cells, to serve such diverse functions. Although traditional platelet functional assays are informative and reliable, they are limited in their ability to unravel platelet phenotypic heterogeneity and interactions. Developments in methods such as electron microscopy, flow cytometry, mass spectrometry, and 'omics' studies, have led to new insights. In this Review, we focus on advances in platelet biology and function, with an emphasis on current and promising methodologies. We also discuss technical and biological challenges in platelet investigations. Using coronavirus disease 2019 (COVID-19) as an example, we further describe the translational relevance of these approaches and the possible 'bench-to-bedside' utility in patient diagnosis and care.
Collapse
Affiliation(s)
- Tarun Tyagi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Kanika Jain
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Sean X Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
- Department of Laboratory Medicine, Yale University School of Medicine, Yale New Haven Hospital, New Haven, CT, USA
| | - Miaoyun Qiu
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Vivian W Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Hannah Melchinger
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Henry Rinder
- Department of Laboratory Medicine, Yale University School of Medicine, Yale New Haven Hospital, New Haven, CT, USA
| | - Kathleen A Martin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Elizabeth E Gardiner
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Alfred I Lee
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Wai Ho Tang
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
34
|
lnc-MRGPRF-6:1 Promotes M1 Polarization of Macrophage and Inflammatory Response through the TLR4-MyD88-MAPK Pathway. Mediators Inflamm 2022; 2022:6979117. [PMID: 35125964 PMCID: PMC8816599 DOI: 10.1155/2022/6979117] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/02/2021] [Accepted: 12/21/2021] [Indexed: 11/17/2022] Open
Abstract
Background. Macrophage-mediated inflammation plays an essential role in the development of atherosclerosis (AS). Long noncoding RNAs (lncRNAs), as crucial regulators, participate in this process. We identified that lnc-MRGPRF-6:1 was significantly upregulated in the plasma exosomes of coronary atherosclerotic disease (CAD) patients in a preliminary work. In the present study, we aim to assess the role of lnc-MRGPRF-6:1 in macrophage-mediated inflammatory process of AS. Methods. The correlation between lnc-MRGPRF-6:1 and inflammatory factors was estimated firstly in plasma exosomes of CAD patients. Subsequently, we established lnc-MRGPRF-6:1 knockout macrophage model via the CRISPR/Cas9 system. We then investigated the regulatory effects of lnc-MRGPRF-6:1 on macrophage polarization and foam cell formation. Eventually, transcriptome analysis by RNA sequencing was carried out to explore the contribution of differential genes and signaling pathways in this process. Results. lnc-MRGPRF-6:1 was highly expressed in the plasma exosomes of CAD patients and was positively correlated with the expression of inflammatory cytokines in plasma. lnc-MRGPRF-6:1 inhibition significantly reduced the formation of foam cells. The expression of lnc-MRGPRF-6:1 was upregulated in M1 macrophage, and lnc-MRGPRF-6:1 knockout decreased the polarization of M1 macrophage. lnc-MRGPRF-6:1 regulates macrophage polarization via the TLR4-MyD88-MAPK signaling pathway. Conclusions. lnc-MRGPRF-6:1 knockdown can inhibit M1 polarization of macrophage and inflammatory response through the TLR4-MyD88-MAPK signaling pathway. lnc-MRGPRF-6:1 is a vital regulator in macrophage-mediated inflammatory process of AS.
Collapse
|
35
|
Isomer-selective analysis of inositol phosphates with differential isotope labelling by phosphate methylation using liquid chromatography with tandem mass spectrometry. Anal Chim Acta 2022; 1191:339286. [PMID: 35033253 DOI: 10.1016/j.aca.2021.339286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/08/2021] [Accepted: 11/14/2021] [Indexed: 11/21/2022]
Abstract
Inositol phosphates belong to a family of structurally diverse signaling molecules playing crucial role in Ca2+ release from intracellular storage vesicles. There are many possibilities of phosphorylation, including their degree and position. Inositol (1,4,5) trisphosphate has been well recognized as the most important second messenger among this family. It remains a challenge to analyse the entire inositol phosphate metabolite family due to its structural complexity, high polarity, and high phosphate density. In this study, we have established an improved UHPLC-ESI-MS/MS method based on a differential isotope labelling methylation strategy. An SPE extraction kit composed of TiO2 and PTFE filter was employed for sample preparation which provided good extraction performance. Samples were methylated (light label) to neutralize the phosphate groups and give better performance in liquid chromatography. Regioisomers and inositol phosphates differing in their number of phosphate residues were successfully separated after optimization on a core-shell cholesterylether-bonded RP-type column (Cosmocore 2.6Cholester) using methanol as organic modifier. Triple quadrupole MS detection was based on selected reaction monitoring (SRM) acquisition with characteristic fragments. Stable isotope labeling methylation was performed to generate internal standards (heavy label). Limits of quantification from 0.32 to 0.89 pmol on column was achieved. This method was validated to be suitable for inositol phosphate profiling in biological samples. After application in cultured HeLa cells, NIST SRM1950 plasma, and human platelets, distinct inositol profiles were obtained. This newly established method exhibited improved analytical performance, holding the potential to advance the understanding of inositol phosphate signaling.
Collapse
|
36
|
Atypical Roles of the Chemokine Receptor ACKR3/CXCR7 in Platelet Pathophysiology. Cells 2022; 11:cells11020213. [PMID: 35053329 PMCID: PMC8773869 DOI: 10.3390/cells11020213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 12/23/2022] Open
Abstract
The manifold actions of the pro-inflammatory and regenerative chemokine CXCL12/SDF-1α are executed through the canonical GProteinCoupledReceptor CXCR4, and the non-canonical ACKR3/CXCR7. Platelets express CXCR4, ACKR3/CXCR7, and are a vital source of CXCL12/SDF-1α themselves. In recent years, a regulatory impact of the CXCL12-CXCR4-CXCR7 axis on platelet biogenesis, i.e., megakaryopoiesis, thrombotic and thrombo-inflammatory actions have been revealed through experimental and clinical studies. Platelet surface expression of ACKR3/CXCR7 is significantly enhanced following myocardial infarction (MI) in acute coronary syndrome (ACS) patients, and is also associated with improved functional recovery and prognosis. The therapeutic implications of ACKR3/CXCR7 in myocardial regeneration and improved recovery following an ischemic episode, are well documented. Cardiomyocytes, cardiac-fibroblasts, endothelial lining of the blood vessels perfusing the heart, besides infiltrating platelets and monocytes, all express ACKR3/CXCR7. This review recapitulates ligand induced differential trafficking of platelet CXCR4-ACKR3/CXCR7 affecting their surface availability, and in regulating thrombo-inflammatory platelet functions and survival through CXCR4 or ACKR3/CXCR7. It emphasizes the pro-thrombotic influence of CXCL12/SDF-1α exerted through CXCR4, as opposed to the anti-thrombotic impact of ACKR3/CXCR7. Offering an innovative translational perspective, this review also discusses the advantages and challenges of utilizing ACKR3/CXCR7 as a potential anti-thrombotic strategy in platelet-associated cardiovascular disorders, particularly in coronary artery disease (CAD) patients post-MI.
Collapse
|
37
|
Platelet ACKR3/CXCR7 Favors Anti-Platelet Lipids over an Atherothrombotic Lipidome and Regulates Thrombo-inflammation. Blood 2021; 139:1722-1742. [PMID: 34905596 DOI: 10.1182/blood.2021013097] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 11/30/2021] [Indexed: 11/20/2022] Open
Abstract
Platelet ACKR3/CXCR7 surface expression is enhanced and influences prognosis in coronary artery disease-(CAD) patients, who exhibit a distinct atherothrombotic platelet lipidome. Current investigation validates the potential of ACKR3/CXCR7 in regulating thrombo-inflammatory response, through its impact on the platelet lipidome. CAD patients-(n=230) with enhanced platelet-ACKR3/CXCR7 expression exhibited reduced aggregation. Pharmacological CXCR7-agonist-(VUF11207) significantly reduced pro-thrombotic platelet response in blood from ACS patients-(n=11) ex vivo. CXCR7-agonist administration reduced thrombotic functions and thrombo-inflammatory platelet-leukocyte interactions post myocardial infarction-(MI) and arterial injury in vivo. ACKR3/CXCR7-ligation did not affect surface availability of GPIbα, GPV, GPVI, GPIX, αv-integrin, β3-integrin, coagulation profile-(APTT, PT), bleeding time, plasma-dependent thrombin generation-(thrombinoscopy) or clot formation-(thromboelastography), but counteracted activation-induced phosphatidylserine exposure and procoagulant platelet-assisted thrombin generation. Targeted-(micro-UHPLC-ESI-QTrap-MS/MS) and untargeted-(UHPLC-ESI-QTOF-MS/MS) lipidomics analysis revealed that ACKR3/CXCR7-ligation favored generation of anti-thrombotic lipids-(dihomo-γ-linolenic acid-DGLA, 12-hydroxyeicosatrienoic acid-12-HETrE) over cyclooxygenase-COX-1-(thromboxane-TxA2), or 12-lipoxygenase-LOX-(12-HETE) metabolized pro-thrombotic, and phospholipase derived atherogenic-(lysophosphatidylcholine-LPC) lipids, in healthy subjects and CAD patients, contrary to anti-platelet therapy. Through 12-HETrE, ACKR3/CXCR7-ligation coordinated with Gαs-coupled prostacyclin receptor-(IP) to trigger cAMP-PKA mediated platelet inhibition. ACKR3/CXCR7-ligation reduced generation of lipid agonists-(arachidonic acid-AA,TxA2), lipid signaling intermediates-(lyophosphatidylinositol-LPI, diacylglycerol-DG), which affected calcium mobilization, intracellular signaling, consequently platelet interaction with physiological matrices and thrombo-inflammatory secretion-(IL1β,IFN-γ,TGF-β,IL-8), emphasizing its functional dichotomy from pro-thrombotic CXCR4. Moreover, CXCR7-agonist regulated heparin-induced thrombocytopenia-(HIT)-sera/IgG-induced platelet and neutrophil activation, heparin induced platelet aggregation-(HIPA), generation of COX-1-(TxA2), 12-LOX-(12-HETE) derived thrombo-inflammatory lipids, platelet-neutrophil aggregate formation, and thrombo-inflammatory secretion (sCD40L, IL-1β, IFN-γ, TNF-α, sP-selectin, IL-8, tissue factor-TF) ex vivo. Therefore, ACKR3/CXCR7 may offer a novel therapeutic strategy in acute/chronic thrombo-inflammation exaggerated cardiovascular pathologies, and CAD.
Collapse
|
38
|
Leiva O, AbdelHameid D, Connors JM, Cannon CP, Bhatt DL. Common Pathophysiology in Cancer, Atrial Fibrillation, Atherosclerosis, and Thrombosis: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2021; 3:619-634. [PMID: 34988471 PMCID: PMC8702799 DOI: 10.1016/j.jaccao.2021.08.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease and cancer are the 2 leading causes of death worldwide. Emerging evidence suggests common mechanisms between cancer and cardiovascular disease, including atrial fibrillation and atherosclerosis. With advances in cancer therapies, screening, and diagnostics, cancer-specific survival and outcomes have improved. This increase in survival has led to the coincidence of cardiovascular disease, including atrial fibrillation and atherosclerosis, as patients with cancer live longer. Additionally, cancer and cardiovascular disease share several risk factors and underlying pathophysiologic mechanisms, including inflammation, cancer-related factors including treatment effects, and alterations in platelet function. Patients with cancer are at increased risk for bleeding and thrombosis compared with the general population. Although optimal antithrombotic therapy, including agent choice and duration, has been extensively studied in the general population, this area remains understudied in patients with cancer despite their altered thrombotic and bleeding risk. Future investigation, including incorporation of cancer-specific characteristics to traditional thrombotic and bleeding risk scores, clinical trials in the cancer population, and the development of novel antithrombotic and anti-inflammatory strategies on the basis of shared pathophysiologic mechanisms, is warranted to improve outcomes in this patient population.
Collapse
Key Words
- AF, atrial fibrillation
- CAD, coronary artery disease
- CHIP, clonal hematopoiesis of indeterminate potential
- CI, confidence interval
- CLEC-2, C-type lectin-like receptor 2
- HR, hazard ratio
- IL, interleukin
- MI, myocardial infarction
- PCI, percutaneous coronary intervention
- ROS, reactive oxygen species
- TKI, tyrosine kinase inhibitor
- VTE, venous thromboembolism
- arrhythmia
- risk factor
- thrombosis
Collapse
Affiliation(s)
- Orly Leiva
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Duaa AbdelHameid
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jean M. Connors
- Division of Hematology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Christopher P. Cannon
- Brigham and Women’s Hospital Heart & Vascular Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Deepak L. Bhatt
- Brigham and Women’s Hospital Heart & Vascular Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
39
|
Seifert J, von Eysmondt H, Chatterjee M, Gawaz M, Schäffer TE. Effect of Oxidized LDL on Platelet Shape, Spreading, and Migration Investigated with Deep Learning Platelet Morphometry. Cells 2021; 10:2932. [PMID: 34831155 PMCID: PMC8616354 DOI: 10.3390/cells10112932] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/15/2021] [Accepted: 10/22/2021] [Indexed: 12/17/2022] Open
Abstract
Platelets are functionally versatile blood cells involved in thrombosis, hemostasis, atherosclerosis, and immune response. Platelet interaction with the immediate microenvironment in blood, vasculature, and tissues alters platelet morphology. The quantification of platelet morphodynamics by geometrical parameters (morphometry) can provide important insights into how platelets sense and respond to stimulatory cues in their vicinity. However, the extraction of platelet shapes from phase contrast microscopy images by conventional image processing is difficult. Here, we used a convolutional neural network (CNN) to develop a deep-learning-based approach for the unbiased extraction of information on platelet morphodynamics by phase contrast microscopy. We then investigated the effect of normal and oxidized low-density lipoproteins (LDL, oxLDL) on platelet morphodynamics, spreading, and haptotactic migration. Exposure of platelets to oxLDL led to a decreased spreading area and rate on fibrinogen, accompanied by increased formation of filopodia and impaired formation of lamellipodia. Haptotactic platelet migration was affected by both LDL and oxLDL in terms of decreased migration velocity and reduced directional persistence. Our results demonstrate the use of deep learning in investigating platelet morphodynamics and reveal differential effects of LDL and oxLDL on platelet morphology and platelet-matrix interaction.
Collapse
Affiliation(s)
- Jan Seifert
- Institute of Applied Physics, University of Tübingen, 72076 Tübingen, Germany; (J.S.); (H.v.E.)
| | - Hendrik von Eysmondt
- Institute of Applied Physics, University of Tübingen, 72076 Tübingen, Germany; (J.S.); (H.v.E.)
| | - Madhumita Chatterjee
- Department of Cardiology and Angiology, University of Tübingen, 72076 Tübingen, Germany; (M.C.); (M.G.)
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University of Tübingen, 72076 Tübingen, Germany; (M.C.); (M.G.)
| | - Tilman E. Schäffer
- Institute of Applied Physics, University of Tübingen, 72076 Tübingen, Germany; (J.S.); (H.v.E.)
| |
Collapse
|
40
|
Platelet-Derived PCSK9 Is Associated with LDL Metabolism and Modulates Atherothrombotic Mechanisms in Coronary Artery Disease. Int J Mol Sci 2021; 22:ijms222011179. [PMID: 34681838 PMCID: PMC8538687 DOI: 10.3390/ijms222011179] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 01/03/2023] Open
Abstract
Platelets play a significant role in atherothrombosis. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is critically involved in the regulation of LDL metabolism and interacts with platelet function. The effect of PCSK9 in platelet function is poorly understood. The authors of this article sought to characterize platelets as a major source of PCSK9 and PCSK9's role in atherothrombosis. In a large cohort of patients with coronary artery disease (CAD), platelet count, platelet reactivity, and platelet-derived PCSK9 release were analyzed. The role of platelet PCSK9 on platelet and monocyte function was investigated in vitro. Platelet count and hyper-reactivity correlated with plasma LDL in CAD. The circulating platelets express on their surface and release substantial amounts of PCSK9. Release of PCSK9 augmented platelet-dependent thrombosis, monocyte migration, and differentiation into macrophages/foam cells. Platelets and PCSK9 accumulated in tissue derived from atherosclerotic carotid arteries in areas of macrophages. PCSK9 inhibition reduced platelet activation and platelet-dependent thrombo-inflammation. The authors identified platelets as a source of PCSK9 in CAD, which may have an impact on LDL metabolism. Furthermore, platelet-derived PCSK9 contributes to atherothrombosis, and inhibition of PCSK9 attenuates thrombo-inflammation, which may contribute to the reported beneficial clinical effects.
Collapse
|
41
|
Abstract
Scanning ion conductance microscopy (SICM) has emerged as a versatile tool for studies of interfaces in biology and materials science with notable utility in biophysical and electrochemical measurements. The heart of the SICM is a nanometer-scale electrolyte filled glass pipette that serves as a scanning probe. In the initial conception, manipulations of ion currents through the tip of the pipette and appropriate positioning hardware provided a route to recording micro- and nanoscopic mapping of the topography of surfaces. Subsequent advances in instrumentation, probe design, and methods significantly increased opportunities for SICM beyond recording topography. Hybridization of SICM with coincident characterization techniques such as optical microscopy and faradaic electrodes have brought SICM to the forefront as a tool for nanoscale chemical measurement for a wide range of applications. Modern approaches to SICM realize an important tool in analytical, bioanalytical, biophysical, and materials measurements, where significant opportunities remain for further exploration. In this review, we chronicle the development of SICM from the perspective of both the development of instrumentation and methods and the breadth of measurements performed.
Collapse
Affiliation(s)
- Cheng Zhu
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, United States
| | - Kaixiang Huang
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, United States
| | - Natasha P Siepser
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, United States
| | - Lane A Baker
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, United States
| |
Collapse
|
42
|
Surendran A, Atefi N, Zhang H, Aliani M, Ravandi A. Defining Acute Coronary Syndrome through Metabolomics. Metabolites 2021; 11:685. [PMID: 34677400 PMCID: PMC8540033 DOI: 10.3390/metabo11100685] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/19/2021] [Accepted: 09/25/2021] [Indexed: 02/06/2023] Open
Abstract
As an emerging platform technology, metabolomics offers new insights into the pathomechanisms associated with complex disease conditions, including cardiovascular diseases. It also facilitates assessing the risk of developing the disease before its clinical manifestation. For this reason, metabolomics is of growing interest for understanding the pathogenesis of acute coronary syndromes (ACS), finding new biomarkers of ACS, and its associated risk management. Metabolomics-based studies in ACS have already demonstrated immense potential for biomarker discovery and mechanistic insights by identifying metabolomic signatures (e.g., branched-chain amino acids, acylcarnitines, lysophosphatidylcholines) associated with disease progression. Herein, we discuss the various metabolomics approaches and the challenges involved in metabolic profiling, focusing on ACS. Special attention has been paid to the clinical studies of metabolomics and lipidomics in ACS, with an emphasis on ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Arun Surendran
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital, Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; (A.S.); (N.A.); (H.Z.)
- Mass Spectrometry and Proteomics Core Facility, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Negar Atefi
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital, Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; (A.S.); (N.A.); (H.Z.)
| | - Hannah Zhang
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital, Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; (A.S.); (N.A.); (H.Z.)
| | - Michel Aliani
- Faculty of Agricultural and Food Sciences, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada;
| | - Amir Ravandi
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital, Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; (A.S.); (N.A.); (H.Z.)
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
- Section of Cardiology, Department of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| |
Collapse
|
43
|
Villaseñor A, Godzien J, Barker-Tejeda TC, Gonzalez-Riano C, López-López Á, Dudzik D, Gradillas A, Barbas C. Analytical approaches for studying oxygenated lipids in the search of potential biomarkers by LC-MS. Trends Analyt Chem 2021. [DOI: 10.1016/j.trac.2021.116367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
44
|
Reveals of candidate active ingredients in Justicia and its anti-thrombotic action of mechanism based on network pharmacology approach and experimental validation. Sci Rep 2021; 11:17187. [PMID: 34433871 PMCID: PMC8387432 DOI: 10.1038/s41598-021-96683-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/13/2021] [Indexed: 11/08/2022] Open
Abstract
Thrombotic diseases seriously threaten human life. Justicia, as a common Chinese medicine, is usually used for anti-inflammatory treatment, and further studies have found that it has an inhibitory effect on platelet aggregation. Therefore, it can be inferred that Justicia can be used as a therapeutic drug for thrombosis. This work aims to reveal the pharmacological mechanism of the anti-thrombotic effect of Justicia through network pharmacology combined with wet experimental verification. During the analysis, 461 compound targets were predicted from various databases and 881 thrombus-related targets were collected. Then, herb-compound-target network and protein-protein interaction network of disease and prediction targets were constructed and cluster analysis was applied to further explore the connection between the targets. In addition, Gene Ontology (GO) and pathway (KEGG) enrichment were used to further determine the association between target proteins and diseases. Finally, the expression of hub target proteins of the core component and the anti-thrombotic effect of Justicia's core compounds were verified by experiments. In conclusion, the core bioactive components, especially justicidin D, can reduce thrombosis by regulating F2, MMP9, CXCL12, MET, RAC1, PDE5A, and ABCB1. The combination of network pharmacology and the experimental research strategies proposed in this paper provides a comprehensive method for systematically exploring the therapeutic mechanism of multi-component medicine.
Collapse
|
45
|
Coenen DM, Heinzmann ACA, Oggero S, Albers HJ, Nagy M, Hagué P, Kuijpers MJE, Vanderwinden JM, van der Meer AD, Perretti M, Koenen RR, Cosemans JMEM. Inhibition of Phosphodiesterase 3A by Cilostazol Dampens Proinflammatory Platelet Functions. Cells 2021; 10:1998. [PMID: 34440764 PMCID: PMC8392606 DOI: 10.3390/cells10081998] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE platelets possess not only haemostatic but also inflammatory properties, which combined are thought to play a detrimental role in thromboinflammatory diseases such as acute coronary syndromes and stroke. Phosphodiesterase (PDE) 3 and -5 inhibitors have demonstrated efficacy in secondary prevention of arterial thrombosis, partially mediated by their antiplatelet action. Yet it is unclear whether such inhibitors also affect platelets' inflammatory functions. Here, we aimed to examine the effect of the PDE3A inhibitor cilostazol and the PDE5 inhibitor tadalafil on platelet function in various aspects of thromboinflammation. Approach and results: cilostazol, but not tadalafil, delayed ex vivo platelet-dependent fibrin formation under whole blood flow over type I collagen at 1000 s-1. Similar results were obtained with blood from Pde3a deficient mice, indicating that cilostazol effects are mediated via PDE3A. Interestingly, cilostazol specifically reduced the release of phosphatidylserine-positive extracellular vesicles (EVs) from human platelets while not affecting total EV release. Both cilostazol and tadalafil reduced the interaction of human platelets with inflamed endothelium under arterial flow and the release of the chemokines CCL5 and CXCL4 from platelets. Moreover, cilostazol, but not tadalafil, reduced monocyte recruitment and platelet-monocyte interaction in vitro. CONCLUSIONS this study demonstrated yet unrecognised roles for platelet PDE3A and platelet PDE5 in platelet procoagulant and proinflammatory responses.
Collapse
Affiliation(s)
- Daniëlle M. Coenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands; (D.M.C.); (A.C.A.H.); (M.N.); (M.J.E.K.); (R.R.K.)
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Alexandra C. A. Heinzmann
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands; (D.M.C.); (A.C.A.H.); (M.N.); (M.J.E.K.); (R.R.K.)
| | - Silvia Oggero
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK; (S.O.); (M.P.)
| | - Hugo J. Albers
- BIOS Lab-on-a-Chip Group, Technical Medical Centre, MESA+ Institute for Nanotechnology, University of Twente, 7522 NB Enschede, The Netherlands;
- Applied Stem Cell Technologies Group, Technical Medical Centre, University of Twente, 7522 NB Enschede, The Netherlands;
| | - Magdolna Nagy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands; (D.M.C.); (A.C.A.H.); (M.N.); (M.J.E.K.); (R.R.K.)
| | - Perrine Hagué
- Laboratory of Neurophysiology, Faculty of Medicine, Université Libre de Bruxelles, B-1070 Brussels, Belgium; (P.H.); (J.-M.V.)
| | - Marijke J. E. Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands; (D.M.C.); (A.C.A.H.); (M.N.); (M.J.E.K.); (R.R.K.)
| | - Jean-Marie Vanderwinden
- Laboratory of Neurophysiology, Faculty of Medicine, Université Libre de Bruxelles, B-1070 Brussels, Belgium; (P.H.); (J.-M.V.)
| | - Andries D. van der Meer
- Applied Stem Cell Technologies Group, Technical Medical Centre, University of Twente, 7522 NB Enschede, The Netherlands;
| | - Mauro Perretti
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK; (S.O.); (M.P.)
| | - Rory R. Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands; (D.M.C.); (A.C.A.H.); (M.N.); (M.J.E.K.); (R.R.K.)
| | - Judith M. E. M. Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands; (D.M.C.); (A.C.A.H.); (M.N.); (M.J.E.K.); (R.R.K.)
| |
Collapse
|
46
|
Cebo M, Calderón Castro C, Schlotterbeck J, Gawaz M, Chatterjee M, Lämmerhofer M. Untargeted UHPLC-ESI-QTOF-MS/MS analysis with targeted feature extraction at precursor and fragment level for profiling of the platelet lipidome with ex vivo thrombin-activation. J Pharm Biomed Anal 2021; 205:114301. [PMID: 34391135 DOI: 10.1016/j.jpba.2021.114301] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 01/17/2023]
Abstract
Lipids play a major role in platelet signaling and activation. In this study, we analyzed the platelet lipidome in an untargeted manner by reversed-phase UHPLC for lipid species separation coupled to high-resolution QTOF-MS/MS in data-independent acquisition (DIA) mode with sequential window acquisition of all theoretical fragment ion mass spectra (SWATH) for compound detection. Lipid identification and peak picking was supported by the characteristic regular elution pattern of lipids differing in carbon and double bond numbers. It was primarily based on post-acquisition targeted feature extraction from the SWATH data. Multiple extracted ion chromatograms (EICs) from SWATH data of diagnostic ions on MS1 and MS2 level from both positive and negative ion mode allowed to distinguish between poorly resolved isomeric lipids based on their distinct fragment ions, which were used for relative quantification at a molecular lipid species level. It supports assay specificity for relative lipid quantitation via multiple quantifiably ions unlike to data-dependent acquisition methods which rely on precursor ions only. This approach was used to analyze human platelet samples. 457 lipids were annotated. Concentrations of lipids were estimated by stable isotope-labelled lipid class-specific internal standards as surrogate calibrants. Heatmaps of lipid concentrations in dependence on carbon and double bond numbers for the distinct lipid classes revealed a snapshot of the platelet lipidome in the resting state with lipid species distributions within classes supporting some functional interpretations. As expected, activation of the platelets by thrombin has led to significant alterations in the platelet lipidome as proven by univariate (volcano plot) and multivariate (PLS-DA) statistics. Several lipids were significantly up-regulated (lysophosphatidylinositols, oxylipins such as thromboxane B2 (TXB2), hydroxyheptadecatrienoic acid (HHT), hydroxyeicosatetraenoic acid (HETE), hydroxyoctadecadienoic acid (HODE), sphingoid-bases, (very) long chain saturated fatty acids) or down-regulated (lysophosphatidylethanolamines, polyunsaturated fatty acids, phosphatidylinositols). Several of them are well known as biomarkers of platelet activation while others may provide some further insights into pathways of platelet activation and platelet metabolism.
Collapse
Affiliation(s)
- Malgorzata Cebo
- University of Tübingen, Institute of Pharmaceutical Sciences, Pharmaceutical (Bio)Analysis, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | | | - Jörg Schlotterbeck
- University of Tübingen, Institute of Pharmaceutical Sciences, Pharmaceutical (Bio)Analysis, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Otfried-Müller-Strasse 10, 72076, Tübingen, Germany
| | - Madhumita Chatterjee
- Department of Cardiology and Angiology, University Hospital Tübingen, Otfried-Müller-Strasse 10, 72076, Tübingen, Germany
| | - Michael Lämmerhofer
- University of Tübingen, Institute of Pharmaceutical Sciences, Pharmaceutical (Bio)Analysis, Auf der Morgenstelle 8, 72076, Tübingen, Germany.
| |
Collapse
|
47
|
Harm T, Bild A, Dittrich K, Goldschmied A, Nestele J, Chatterjee M, Fu X, Kolb K, Castor T, Borst O, Geisler T, Rath D, LäMmerhofer M, Gawaz M. Acute coronary syndrome is associated with a substantial change in the platelet lipidome. Cardiovasc Res 2021; 118:1904-1916. [PMID: 34323932 DOI: 10.1093/cvr/cvab238] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/21/2021] [Indexed: 11/14/2022] Open
Abstract
AIMS Platelets play a key role in the pathophysiology of coronary artery disease (CAD) and patients with enhanced platelet activation are at increased risk to develop adverse cardiovascular events. Beyond reliable cardiovascular risk factors such as dyslipoproteinaemia, significant changes of platelet lipids occur in patients with CAD. In this study, we investigate the platelet lipidome by untargeted liquid chromatography-mass spectrometry, highlighting significant changes between acute coronary syndrome (ACS) and chronic coronary syndrome (CCS) patients. Additionally, we classify the platelet lipidome, spotlighting specific glycerophospholipids as key players in ACS patients. Furthermore, we examine the impact of significantly altered lipids in ACS on platelet-dependent thrombus formation and aggregation. METHODS AND RESULTS In this consecutive study, we characterized the platelet lipidome in a CAD cohort (n = 139) and showed significant changes of lipids between patients with ACS and CCS. We found that among 928 lipids, 7 platelet glycerophospholipids were significantly up-regulated in ACS, whereas 25 lipids were down-regulated compared to CCS. The most prominent up-regulated lipid in ACS, PC18:0 (PC 10:0-8:0), promoted platelet activation and ex vivo platelet-dependent thrombus formation. CONCLUSIONS Our results reveal that the platelet lipidome is altered in ACS and up-regulated lipids embody primarily glycerophospholipids. Alterations of the platelet lipidome, especially of medium chain lipids, may play a role in the pathophysiology of ACS.
Collapse
Affiliation(s)
- Tobias Harm
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Alexander Bild
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Kristina Dittrich
- Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Andreas Goldschmied
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Jeremy Nestele
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Madhumita Chatterjee
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Xiaoqing Fu
- Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Kyra Kolb
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Tatsiana Castor
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Tobias Geisler
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Dominik Rath
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Michael LäMmerhofer
- Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| |
Collapse
|
48
|
McGurk KA, Keavney BD, Nicolaou A. Circulating ceramides as biomarkers of cardiovascular disease: Evidence from phenotypic and genomic studies. Atherosclerosis 2021; 327:18-30. [PMID: 34004484 DOI: 10.1016/j.atherosclerosis.2021.04.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/25/2021] [Accepted: 04/30/2021] [Indexed: 12/20/2022]
Abstract
There is a need for new biomarkers of atherosclerotic cardiovascular disease (ACVD), the main cause of death globally. Ceramides, a class of potent bioactive lipid mediators, have signalling roles in apoptosis, cellular stress and inflammation. Recent studies have highlighted circulating ceramides as novel biomarkers of coronary artery disease, type-2 diabetes and insulin resistance. Ceramides are highly regulated by enzymatic reactions throughout the body in terms of their activity and metabolism, including production, degradation and transport. The genetic studies that have been completed to date on the main ceramide species found in circulation are described, highlighting the importance of DNA variants in genes involved in ceramide biosynthesis as key influencers of heritable, circulating ceramide levels. We also review studies of disease associations with ceramides and discuss mechanistic insights deriving from recent genomic studies. The signalling activities of ceramides in vascular inflammation and apoptosis, associations between circulating ceramides and coronary artery disease risk, type-2 diabetes and insulin resistance, and the potential importance of ceramides with regard to ACVD risk factors, such as blood pressure, lipoproteins and lifestyle factors, are also discussed.
Collapse
Affiliation(s)
- Kathryn A McGurk
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, UK; Laboratory for Lipidomics and Lipid Research, Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, UK
| | - Bernard D Keavney
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, UK; Manchester Heart Centre, Manchester University NHS Foundation Trust, UK
| | - Anna Nicolaou
- Laboratory for Lipidomics and Lipid Research, Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, UK; Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
49
|
Sun Y, Huang S, Wan C, Ruan Q, Xie X, Wei D, Li G, Lin S, Li H, Wu S. Knockdown of lncRNA ENST00000609755.1 Confers Protection Against Early oxLDL-Induced Coronary Heart Disease. Front Cardiovasc Med 2021; 8:650212. [PMID: 34095248 PMCID: PMC8175657 DOI: 10.3389/fcvm.2021.650212] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/12/2021] [Indexed: 12/24/2022] Open
Abstract
Background: This study investigated the association between long non-coding RNAs (lncRNAs) and coronary heart disease (CHD) and further elucidated the potential biological roles of lncRNAs in CHD pathogenesis. Methods: A case-control study (590 patients and 590 controls) was conducted from February 2017 and March 2019 in Fuzhou, China. Environmental factors were investigated using questionnaires and physical examinations. Five representative lncRNAs were screened using lncRNA microarray (peripheral blood in 5 cases and 5 controls) and further verified by quantitative real-time polymerase chain reaction (peripheral blood leukocyte in 100 cases and 100 controls). Oxidized low-density lipoprotein (oxLDL) was used to induce a human coronary artery endothelial cell (HCAECs) injury model, and loss of function was used to elucidate the role of lncRNA ENST00000609755.1 (lnc-MICALL2-2) in oxLDL-induced HCAECs injury. Results: A total of 320 lncRNAs were found dysregulated in CHD patients (fold change> 2, p < 0.05). The results of a discovery microarray, population verification and HCAEC experiments suggested the lnc-MICALL2-2 is upregulated in CHD subjects and in an oxLDL-induced HCAECs injury model. Conversely, lnc-MICALL2-2 inhibition in vitro attenuated the effects of oxLDL on HCAECs morphology, proliferation, and apoptosis. Conclusion: Elevated expression of lnc-MICALL2-2 is an independent risk factor for CHD, and knockdown subsequently confers protection against early pathological processes of oxLDL-induced CHD.
Collapse
Affiliation(s)
- Yi Sun
- School of Public Health, Fujian Medical University, Fuzhou, China
| | - Shuna Huang
- Department of Clinical Research and Translation Center Office, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Chunyu Wan
- School of Public Health, Fujian Medical University, Fuzhou, China
| | - Qishuang Ruan
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoxu Xie
- School of Public Health, Fujian Medical University, Fuzhou, China
| | - Donghong Wei
- School of Public Health, Fujian Medical University, Fuzhou, China
| | - Guobo Li
- School of Public Health, Fujian Medical University, Fuzhou, China
| | - Shaowei Lin
- School of Public Health, Fujian Medical University, Fuzhou, China
| | - Huangyuan Li
- School of Public Health, Fujian Medical University, Fuzhou, China
| | - Siying Wu
- School of Public Health, Fujian Medical University, Fuzhou, China
| |
Collapse
|
50
|
Borén J, Chapman MJ, Krauss RM, Packard CJ, Bentzon JF, Binder CJ, Daemen MJ, Demer LL, Hegele RA, Nicholls SJ, Nordestgaard BG, Watts GF, Bruckert E, Fazio S, Ference BA, Graham I, Horton JD, Landmesser U, Laufs U, Masana L, Pasterkamp G, Raal FJ, Ray KK, Schunkert H, Taskinen MR, van de Sluis B, Wiklund O, Tokgozoglu L, Catapano AL, Ginsberg HN. Low-density lipoproteins cause atherosclerotic cardiovascular disease: pathophysiological, genetic, and therapeutic insights: a consensus statement from the European Atherosclerosis Society Consensus Panel. Eur Heart J 2021; 41:2313-2330. [PMID: 32052833 PMCID: PMC7308544 DOI: 10.1093/eurheartj/ehz962] [Citation(s) in RCA: 780] [Impact Index Per Article: 195.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 11/10/2019] [Accepted: 01/08/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract
Collapse
Affiliation(s)
- Jan Borén
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - M John Chapman
- Endocrinology-Metabolism Division, Pitié-Salpêtrière University Hospital, Sorbonne University, Paris, France.,National Institute for Health and Medical Research (INSERM), Paris, France
| | - Ronald M Krauss
- Department of Atherosclerosis Research, Children's Hospital Oakland Research Institute and UCSF, Oakland, CA 94609, USA
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Jacob F Bentzon
- Department of Clinical Medicine, Heart Diseases, Aarhus University, Aarhus, Denmark.,Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Mat J Daemen
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Linda L Demer
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Physiology, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Robert A Hegele
- Department of Medicine, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Stephen J Nicholls
- Monash Cardiovascular Research Centre, Monash University, Melbourne, Australia
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, University of Copenhagen, Denmark
| | - Gerald F Watts
- School of Medicine, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Australia.,Department of Cardiology, Lipid Disorders Clinic, Royal Perth Hospital, Perth, Australia
| | - Eric Bruckert
- INSERM UMRS1166, Department of Endocrinology-Metabolism, ICAN - Institute of CardioMetabolism and Nutrition, AP-HP, Hopital de la Pitie, Paris, France
| | - Sergio Fazio
- Departments of Medicine, Physiology and Pharmacology, Knight Cardiovascular Institute, Center of Preventive Cardiology, Oregon Health & Science University, Portland, OR, USA
| | - Brian A Ference
- Centre for Naturally Randomized Trials, University of Cambridge, Cambridge, UK.,Institute for Advanced Studies, University of Bristol, Bristol, UK.,MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | | | - Jay D Horton
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ulf Landmesser
- Department of Cardiology, Charité - University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Liebigstraße 20, Leipzig, Germany
| | - Luis Masana
- Research Unit of Lipids and Atherosclerosis, IISPV, CIBERDEM, University Rovira i Virgili, C. Sant Llorenç 21, Reus 43201, Spain
| | - Gerard Pasterkamp
- Laboratory of Clinical Chemistry, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frederick J Raal
- Carbohydrate and Lipid Metabolism Research Unit, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Kausik K Ray
- Department of Primary Care and Public Health, Imperial Centre for Cardiovascular Disease Prevention, Imperial College London, London, UK
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Faculty of Medicine, Technische Universität München, Lazarettstr, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Marja-Riitta Taskinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Bart van de Sluis
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Olov Wiklund
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lale Tokgozoglu
- Department of Cardiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, and IRCCS MultiMedica, Milan, Italy
| | - Henry N Ginsberg
- Department of Medicine, Irving Institute for Clinical and Translational Research, Columbia University, New York, NY, USA
| |
Collapse
|