1
|
Chen PHB, Li XL, Baskin JM. Synthetic Lipid Biology. Chem Rev 2025; 125:2502-2560. [PMID: 39805091 PMCID: PMC11969270 DOI: 10.1021/acs.chemrev.4c00761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Cells contain thousands of different lipids. Their rapid and redundant metabolism, dynamic movement, and many interactions with other biomolecules have justly earned lipids a reputation as a vexing class of molecules to understand. Further, as the cell's hydrophobic metabolites, lipids assemble into supramolecular structures─most commonly bilayers, or membranes─from which they carry out myriad biological functions. Motivated by this daunting complexity, researchers across disciplines are bringing order to the seeming chaos of biological lipids and membranes. Here, we formalize these efforts as "synthetic lipid biology". Inspired by the idea, central to synthetic biology, that our abilities to understand and build biological systems are intimately connected, we organize studies and approaches across numerous fields to create, manipulate, and analyze lipids and biomembranes. These include construction of lipids and membranes from scratch using chemical and chemoenzymatic synthesis, editing of pre-existing membranes using optogenetics and protein engineering, detection of lipid metabolism and transport using bioorthogonal chemistry, and probing of lipid-protein interactions and membrane biophysical properties. What emerges is a portrait of an incipient field where chemists, biologists, physicists, and engineers work together in proximity─like lipids themselves─to build a clearer description of the properties, behaviors, and functions of lipids and membranes.
Collapse
Affiliation(s)
- Po-Hsun Brian Chen
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853, United States
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Xiang-Ling Li
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853, United States
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Jeremy M Baskin
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853, United States
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
2
|
Allemand F, Yesylevskyy S, Lagoutte-Renosi J, Davani S, Ramseyer C. Nanodomains enriched in arachidonic acid promote P2Y12 receptor oligomerization in the platelet plasma membrane. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2025; 1867:184402. [PMID: 39557210 DOI: 10.1016/j.bbamem.2024.184402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/16/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024]
Abstract
P2Y12 receptors on the platelet plasma membrane are targeted by several antiplatelets drugs. Although oligomerization and functioning of P2Y12 receptors depend on the membrane environment, little is known about their preferred membrane localization and the role of surrounding lipid composition, especially the arachidonic acids (ARA), which are abundant in platelets. Coarse-grained molecular dynamics simulations of platelet plasma membrane based on the lipidomics data were used to investigate the P2Y12 lipid environment and the involvement of ARA in its oligomerization in platelet plasma membranes. The platelet plasma membrane contains two types of lipids nanodomains: ordered, enriched in SM and cholesterol, and disordered, enriched in ARA-containing lipids. P2Y12 receptors prefer to localize in these ARA-rich domains and induce the sorting of the ARA-containing lipids in their vicinity. This ARA-rich environment promotes the oligomerization of P2Y12 receptors and stabilizes the protein-protein interfaces of oligomers. As summary, oligomerization of P2Y12 receptors is promoted in ARA-rich nano-domains of the platelet plasma membrane.
Collapse
Affiliation(s)
- Florentin Allemand
- Université de Franche-Comté, SINERGIES, F-25000 Besançon, France; Université de Franche-Comté, CNRS, Chrono-environnement, F-25000 Besançon, France.
| | - Semen Yesylevskyy
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, CZ-166 10 Prague, Czech Republic; Receptor.AI Inc., 20-22 Wenlock Road, London N1 7GU, United Kingdom; Department of Physical Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 12, 771 46 Olomouc, Czech Republic; Department of Physics of Biological Systems, Institute of Physics of the National Academy of Sciences of Ukraine, Prospect Nauky 46, 03028 Kyiv, Ukraine
| | - Jennifer Lagoutte-Renosi
- Université de Franche-Comté, CHU Besançon, SINERGIES, Service de Pharmacologie Clinique et Toxicologie, F-25000 Besançon, France
| | - Siamak Davani
- Université de Franche-Comté, CHU Besançon, SINERGIES, Service de Pharmacologie Clinique et Toxicologie, F-25000 Besançon, France
| | - Christophe Ramseyer
- Université de Franche-Comté, CNRS, Chrono-environnement, F-25000 Besançon, France
| |
Collapse
|
3
|
Xue R, Li M, Zhang G, Zhang W, Han L, Bo T, Zhong H, Yao D, Deng Y, Chen S, Zhang S. GSDME-mediated pyroptosis contributes to chemotherapy-induced platelet hyperactivity and thrombotic potential. Blood 2024; 144:2652-2665. [PMID: 39378585 DOI: 10.1182/blood.2023023179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 07/05/2024] [Accepted: 07/23/2024] [Indexed: 10/10/2024] Open
Abstract
Thrombotic complications due to platelet hyperreactivity are a major cause of death in patients undergoing chemotherapy. However, the underlying mechanisms are not fully understood. Herein, using human platelets and platelets from mice lacking gasdermin E (GSDME), we show that GSDME is functionally expressed in anucleate platelets, and that GSDME-mediated pyroptosis, a newly identified form of cell death in mammalian nucleated cells, contributes to platelet hyperactivity in cisplatin-based chemotherapy. Cisplatin or etoposide activates caspase-3 to cleave GSDME, thereby releasing the N-terminal fragment of GSDME (GSDME-N) toward the platelet plasma membrane, subsequently forming membrane pores and facilitating platelet granule release. This eventually promotes platelet hyperactivity and thrombotic potential. We identified flotillin-2, a scaffold protein, as a GSDME-N interactor that recruits GSDME-N to the platelet membrane. Loss of GSDME protects mice from cisplatin-induced platelet hyperactivity. Our results provide evidence that targeting GSDME-mediated pyroptosis could reduce thrombotic potential in chemotherapy.
Collapse
Affiliation(s)
- Ruyi Xue
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Min Li
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ge Zhang
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wei Zhang
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Liping Han
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Blood Transfusion, Huashan Hospital, Fudan University, Shanghai, China
| | - Tao Bo
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Haoxuan Zhong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Dingjin Yao
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yiran Deng
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - She Chen
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Si Zhang
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Komatsuya K, Ishikawa M, Kikuchi N, Hirabayashi T, Taguchi R, Yamamoto N, Arai M, Kasahara K. Integrin-Dependent Transient Density Increase in Detergent-Resistant Membrane Rafts in Platelets Activated by Thrombin. Biomedicines 2023; 12:69. [PMID: 38255176 PMCID: PMC10813660 DOI: 10.3390/biomedicines12010069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
Platelet lipid rafts are critical membrane domains for adhesion, aggregation, and clot retraction. Lipid rafts are isolated as a detergent-resistant membrane fraction via sucrose density gradient centrifugation. The platelet detergent-resistant membrane shifted to a higher density on the sucrose density gradient upon thrombin stimulation. The shift peaked at 1 min and returned to the control level at 60 min. During this time, platelets underwent clot retraction and spreading on a fibronectin-coated glass strip. Thrombin induced the transient tyrosine phosphorylation of several proteins in the detergent-resistant membrane raft fraction and the transient translocation of fibrin and myosin to the detergent-resistant membrane raft fraction. The level of phosphatidylserine (36:1) was increased and the level of phosphatidylserine (38:4) was decreased in the detergent-resistant membrane raft fraction via the thrombin stimulation. Furthermore, Glanzmann's thrombasthenia integrin αIIbβ3-deficient platelets underwent no detergent-resistant membrane shift to a higher density upon thrombin stimulation. As the phosphorylation of the myosin regulatory light chain on Ser19 was at a high level in Glanzmann's thrombasthenia resting platelets, thrombin caused no further phosphorylation of the myosin regulatory light chain on Ser19 or clot retraction. These observations suggest that the fibrin-integrin αIIbβ3-myosin axis and compositional change of phosphatidylserine species may be required for the platelet detergent-resistant membrane shift to a higher density upon stimulation with thrombin.
Collapse
Affiliation(s)
- Keisuke Komatsuya
- Biomembrane Group, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (K.K.); (N.K.); (T.H.); (N.Y.)
| | - Masaki Ishikawa
- Laboratory of Clinical Omics Research, Department of Applied Genomics, Kazusa DNA Research Institute, Kisarazu, Chiba 292-0818, Japan;
| | - Norihito Kikuchi
- Biomembrane Group, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (K.K.); (N.K.); (T.H.); (N.Y.)
| | - Tetsuya Hirabayashi
- Biomembrane Group, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (K.K.); (N.K.); (T.H.); (N.Y.)
| | - Ryo Taguchi
- Department of Metabolome, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Naomasa Yamamoto
- Biomembrane Group, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (K.K.); (N.K.); (T.H.); (N.Y.)
| | - Morio Arai
- Biomembrane Group, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (K.K.); (N.K.); (T.H.); (N.Y.)
- Sado General Hospital, Niigata 952-1209, Japan
| | - Kohji Kasahara
- Biomembrane Group, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (K.K.); (N.K.); (T.H.); (N.Y.)
| |
Collapse
|
5
|
Arjoonsingh A, Haines JM, Hwang JK, Guess SC, Wardrop KJ. Evaluation of platelet additive solution for prolonging storage of functional canine platelet concentrate. J Vet Emerg Crit Care (San Antonio) 2023; 33:656-664. [PMID: 37639315 DOI: 10.1111/vec.13335] [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: 04/29/2021] [Revised: 04/07/2022] [Accepted: 06/23/2022] [Indexed: 08/29/2023]
Abstract
OBJECTIVE To assess storage lesion development, platelet function, and bacterial growth in canine platelet concentrates (PCs) stored in a platelet additive solution (PAS) or a plasma control at 4°C for 21 days. DESIGN Prospective, ex vivo, experimental controlled study. SETTING University veterinary teaching hospital. ANIMALS Ten units of canine PCs collected from blood bank donations. INTERVENTIONS The PCs were separated into 2 bags, 1 containing 100% plasma and the other containing 35% plasma and 65% of a PAS (Plasma-Lyte A), and stored at 4°C for 21 days. At days 0, 7, 14, and 21, PCs were analyzed for the presence of swirling, aggregate formation, platelet counts, platelet indices, glucose, lactate, lactate dehydrogenase, Pvco2 , Pvo2 , aggregation via light aggregometry, activation percentages using flow cytometry, and bacterial growth. MEASUREMENTS AND MAIN RESULTS Cold-stored PCs in both PAS and plasma control maintained mean pH >6.8 and mean lactate <9.0 mmol/L over 21 days, with no difference in glucose utilization. Swirl was maintained in both solutions for most days (76/80 combined total samples), with no difference in aggregate formation between solutions. The Pvco2 was higher in plasma on all days (P < 0.001), with no difference in Pvo2 . Platelet indices did not reflect significant storage lesion development in either solution. Lactate dehydrogenase did not differ between solutions but did increase from day 7 to day 21. Mean maximal aggregation percentage was reduced overall but with no significant difference between solutions. The only observed difference in mean activation percentage between solutions was in PAS on day 7, which was significantly higher than plasma (P < 0.05). No bacterial growth occurred during storage. CONCLUSIONS Cold storage in PAS and plasma allowed PCs to be stored for up to 21 days with minimal storage lesion development, maintenance of platelet function, limited platelet activation, and no bacterial growth within stored bags.
Collapse
Affiliation(s)
- Avin Arjoonsingh
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Jillian M Haines
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Julianne K Hwang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Sarah C Guess
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - K Jane Wardrop
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| |
Collapse
|
6
|
Ji H, Li Y, Su B, Zhao W, Kizhakkedathu JN, Zhao C. Advances in Enhancing Hemocompatibility of Hemodialysis Hollow-Fiber Membranes. ADVANCED FIBER MATERIALS 2023; 5:1-43. [PMID: 37361105 PMCID: PMC10068248 DOI: 10.1007/s42765-023-00277-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 02/19/2023] [Indexed: 06/28/2023]
Abstract
Hemodialysis, the most common modality of renal replacement therapy, is critically required to remove uremic toxins from the blood of patients with end-stage kidney disease. However, the chronic inflammation, oxidative stress as well as thrombosis induced by the long-term contact of hemoincompatible hollow-fiber membranes (HFMs) contribute to the increase in cardiovascular diseases and mortality in this patient population. This review first retrospectively analyzes the current clinical and laboratory research progress in improving the hemocompatibility of HFMs. Details on different HFMs currently in clinical use and their design are described. Subsequently, we elaborate on the adverse interactions between blood and HFMs, involving protein adsorption, platelet adhesion and activation, and the activation of immune and coagulation systems, and the focus is on how to improve the hemocompatibility of HFMs in these aspects. Finally, challenges and future perspectives for improving the hemocompatibility of HFMs are also discussed to promote the development and clinical application of new hemocompatible HFMs. Graphical Abstract
Collapse
Affiliation(s)
- Haifeng Ji
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065 People’s Republic of China
- Department of Pathology and Lab Medicine & Center for Blood Research & Life Science Institute, 2350 Health Sciences Mall, Life Sciences Centre, The School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Yupei Li
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, 610041 China
- Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, 610207 China
| | - Baihai Su
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, 610041 China
| | - Weifeng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065 People’s Republic of China
| | - Jayachandran N. Kizhakkedathu
- Department of Pathology and Lab Medicine & Center for Blood Research & Life Science Institute, 2350 Health Sciences Mall, Life Sciences Centre, The School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Changsheng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065 People’s Republic of China
| |
Collapse
|
7
|
Tantiwong C, Dunster JL, Cavill R, Tomlinson MG, Wierling C, Heemskerk JWM, Gibbins JM. An agent-based approach for modelling and simulation of glycoprotein VI receptor diffusion, localisation and dimerisation in platelet lipid rafts. Sci Rep 2023; 13:3906. [PMID: 36890261 PMCID: PMC9994409 DOI: 10.1038/s41598-023-30884-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 03/02/2023] [Indexed: 03/10/2023] Open
Abstract
Receptor diffusion plays an essential role in cellular signalling via the plasma membrane microenvironment and receptor interactions, but the regulation is not well understood. To aid in understanding of the key determinants of receptor diffusion and signalling, we developed agent-based models (ABMs) to explore the extent of dimerisation of the platelet- and megakaryocyte-specific receptor for collagen glycoprotein VI (GPVI). This approach assessed the importance of glycolipid enriched raft-like domains within the plasma membrane that lower receptor diffusivity. Our model simulations demonstrated that GPVI dimers preferentially concentrate in confined domains and, if diffusivity within domains is decreased relative to outside of domains, dimerisation rates are increased. While an increased amount of confined domains resulted in further dimerisation, merging of domains, which may occur upon membrane rearrangements, was without effect. Modelling of the proportion of the cell membrane which constitutes lipid rafts indicated that dimerisation levels could not be explained by these alone. Crowding of receptors by other membrane proteins was also an important determinant of GPVI dimerisation. Together, these results demonstrate the value of ABM approaches in exploring the interactions on a cell surface, guiding the experimentation for new therapeutic avenues.
Collapse
Affiliation(s)
- Chukiat Tantiwong
- School of Biological Sciences, University of Reading, Reading, UK.,Department of Biochemistry, CARIM, Maastricht University, Maastricht, The Netherlands
| | - Joanne L Dunster
- School of Biological Sciences, University of Reading, Reading, UK
| | - Rachel Cavill
- Department of Data Science and Knowledge Engineering, Maastricht University, Maastricht, The Netherlands
| | | | | | - Johan W M Heemskerk
- Department of Biochemistry, CARIM, Maastricht University, Maastricht, The Netherlands.,Synapse Research Institute, Maastricht, The Netherlands
| | | |
Collapse
|
8
|
Platelet Lipidome Fingerprint: New Assistance to Characterize Platelet Dysfunction in Obesity. Int J Mol Sci 2022; 23:ijms23158326. [PMID: 35955459 PMCID: PMC9369067 DOI: 10.3390/ijms23158326] [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: 06/30/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 11/27/2022] Open
Abstract
Obesity is associated with a pro-inflammatory and pro-thrombotic state that supports atherosclerosis progression and platelet hyper-reactivity. During the last decade, the platelet lipidome has been considered a treasure trove, as it is a source of biomarkers for preventing and treating different pathologies. The goal of the present study was to determine the lipid profile of platelets from non-diabetic, severely obese patients compared with their age- and sex-matched lean controls. Lipids from washed platelets were isolated and major phospholipids, sphingolipids and neutral lipids were analyzed either by gas chromatography or by liquid chromatography coupled to mass spectrometry. Despite a significant increase in obese patient’s plasma triglycerides, there were no significant differences in the levels of triglycerides in platelets among the two groups. In contrast, total platelet cholesterol was significantly decreased in the obese group. The profiling of phospholipids showed that phosphatidylcholine and phosphatidylethanolamine contents were significantly reduced in platelets from obese patients. On the other hand, no significant differences were found in the sphingomyelin and ceramide levels, although there was also a tendency for reduced levels in the obese group. The outline of the glycerophospholipid and sphingolipid molecular species (fatty-acyl profiles) was similar in the two groups. In summary, these lipidomics data indicate that platelets from obese patients have a unique lipid fingerprint that may guide further studies and provide mechanistic-driven perspectives related to the hyperactivate state of platelets in obesity.
Collapse
|
9
|
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: 19] [Impact Index Per Article: 6.3] [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
|
10
|
Li D, Li Y, Yang S, Yu Z, Xing Y, Wu M. Mechanism and Potential Target of Blood-Activating Chinese Botanical Drugs Combined With Anti-Platelet Drugs: Prevention and Treatment of Atherosclerotic Cardiovascular Diseases. Front Pharmacol 2022; 13:811422. [PMID: 35721128 PMCID: PMC9204194 DOI: 10.3389/fphar.2022.811422] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 04/25/2022] [Indexed: 11/14/2022] Open
Abstract
Atherosclerotic cardiovascular diseases (ASCVDs) are the most important diseases that endanger people’s health, leading to high morbidity and mortality worldwide. In addition, various thrombotic events secondary to cardiovascular and cerebrovascular diseases need must be considered seriously. Therefore, the development of novel anti-platelet drugs with high efficiency, and fewer adverse effects has become a research focus for preventing of cardiovascular diseases (CVDs). Blood-activation and stasis-removal from circulation have been widely considered as principles for treating syndromes related to CVDs. Blood-activating Chinese (BAC botanical drugs, as members of traditional Chinese medicine (TCM), have shown to improve hemodynamics and hemorheology, and inhibit thrombosis and atherosclerosis. Modern medical research has identified that a combination of BAC botanical drugs and anti-platelet drugs, such as aspirin or clopidogrel, not only enhances the anti-platelet effects, but also reduces the risk of bleeding and protects the vascular endothelium. The anti-platelet mechanism of Blood-activating Chinese (BAC) botanical drugs and their compounds is not clear; therefore, their potential targets need to be explored. With the continuous development of bioinformatics and “omics” technology, some unconventional applications of BAC botanical drugs have been discovered. In this review, we will focus on the related targets and signaling pathways of anti-atherosclerotic treatments involving a combination of BAC botanical drugs and anti-platelet drugs reported in recent years.
Collapse
Affiliation(s)
- Dan Li
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yujuan Li
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shengjie Yang
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zongliang Yu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanwei Xing
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Wu
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
11
|
Platelet Membrane: An Outstanding Factor in Cancer Metastasis. MEMBRANES 2022; 12:membranes12020182. [PMID: 35207103 PMCID: PMC8875259 DOI: 10.3390/membranes12020182] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/02/2022]
Abstract
In addition to being biological barriers where the internalization or release of biomolecules is decided, cell membranes are contact structures between the interior and exterior of the cell. Here, the processes of cell signaling mediated by receptors, ions, hormones, cytokines, enzymes, growth factors, extracellular matrix (ECM), and vesicles begin. They triggering several responses from the cell membrane that include rearranging its components according to the immediate needs of the cell, for example, in the membrane of platelets, the formation of filopodia and lamellipodia as a tissue repair response. In cancer, the cancer cells must adapt to the new tumor microenvironment (TME) and acquire capacities in the cell membrane to transform their shape, such as in the case of epithelial−mesenchymal transition (EMT) in the metastatic process. The cancer cells must also attract allies in this challenging process, such as platelets, fibroblasts associated with cancer (CAF), stromal cells, adipocytes, and the extracellular matrix itself, which limits tumor growth. The platelets are enucleated cells with fairly interesting growth factors, proangiogenic factors, cytokines, mRNA, and proteins, which support the development of a tumor microenvironment and support the metastatic process. This review will discuss the different actions that platelet membranes and cancer cell membranes carry out during their relationship in the tumor microenvironment and metastasis.
Collapse
|
12
|
Belleville-Rolland T, Leuci A, Mansour A, Decouture B, Martin F, Poirault-Chassac S, Rouaud M, Guerineau H, Dizier B, Pidard D, Gaussem P, Bachelot-Loza C. Role of Membrane Lipid Rafts in MRP4 (ABCC4) Dependent Regulation of the cAMP Pathway in Blood Platelets. Thromb Haemost 2021; 121:1628-1636. [PMID: 33851387 DOI: 10.1055/a-1481-2663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Platelet cytosolic cyclic adenosine monophosphate (cAMP) levels are balanced by synthesis, degradation, and efflux. Efflux can occur via multidrug resistant protein-4 (MRP4; ABCC4) present on dense granule and/or plasma membranes. As lipid rafts have been shown to interfere on cAMP homeostasis, we evaluated the relationships between the distribution and activity of MRP4 in lipid rafts and cAMP efflux. METHODS Platelet activation and cAMP homeostasis were analyzed in human and wild-type or MRP4-deleted mouse platelets in the presence of methyl-β-cyclodextrin (MßCD) to disrupt lipid rafts, and of activators of the cAMP signalling pathways. Human platelet MRP4 and effector proteins of the cAMP pathway were analyzed by immunoblots in lipid rafts isolated by differential centrifugation. RESULTS MßCD dose dependently inhibited human and mouse platelet aggregation without affecting per se cAMP levels. An additive inhibitory effect existed between the adenylate cyclase (AC) activator forskolin and MßCD that was accompanied by an overincrease of cAMP, and which was significantly enhanced upon MRP4 deletion. Finally, an efflux of cAMP out of resting platelets incubated with prostaglandin E1 (PGE1) was observed that was partly dependent on MRP4. Lipid rafts contained a small fraction (≈15%) of MRP4 and most of the inhibitory G-protein Gi, whereas Gs protein, AC3, and phosphodiesterases PDE2 and PDE3A were all present as only trace amounts. CONCLUSION Our results are in favour of part of MRP4 present at the platelet surface, including in lipid rafts. Lipid raft integrity is necessary for cAMP signalling regulation, although MRP4 and most players of cAMP homeostasis are essentially located outside rafts.
Collapse
Affiliation(s)
- Tiphaine Belleville-Rolland
- Service d'hématologie biologique, AH-HP, Hopital Européen Georges Pompidou, Paris, France
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Alexandre Leuci
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Alexandre Mansour
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Benoit Decouture
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Fanny Martin
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | | | - Margot Rouaud
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Hippolyte Guerineau
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Blandine Dizier
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Dominique Pidard
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Pascale Gaussem
- Service d'hématologie biologique, AH-HP, Hopital Européen Georges Pompidou, Paris, France
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | | |
Collapse
|
13
|
Angelis I, Moussis V, Tsoukatos DC, Tsikaris V. Multidrug Resistance Protein 4 (MRP4/ABCC4): A Suspected Efflux Transporter for Human's Platelet Activation. Protein Pept Lett 2021; 28:983-995. [PMID: 33964863 DOI: 10.2174/0929866528666210505120659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/10/2021] [Accepted: 03/16/2021] [Indexed: 11/22/2022]
Abstract
The main role of platelets is to contribute to hemostasis. However, under pathophysiological conditions, platelet activation may lead to thrombotic events of cardiovascular diseases. Thus, anti-thrombotic treatment is important in patients with cardiovascular disease. This review focuses on a platelet receptor, a transmembrane protein, the Multidrug Resistance Protein 4, MRP4, which contributes to platelet activation by extruding endogenous molecules responsible for their activation and accumulation. The regulation of the intracellular concentration levels of these molecules by MRP4 turned to make the protein suspicious and, at the same time, an interesting regulatory factor of normal platelet function. Especially, the possible role of MRP4 in the excretion of xenobiotic and antiplatelet drugs such as aspirin is discussed, thus imparting platelet aspirin tolerance and correlating the protein with the ineffectiveness of aspirin antiplatelet therapy. Based on the above, this review finally underlines that the development of a highly selective and targeted strategy for platelet MRP4 inhibition will also lead to inhibition of platelet activation and accumulation.
Collapse
Affiliation(s)
- Ioannis Angelis
- Department of Chemistry, Organic Chemistry & Biochemistry, University of Ioannina, Ioannina. Greece
| | - Vassilios Moussis
- Department of Chemistry, Organic Chemistry & Biochemistry, University of Ioannina, Ioannina. Greece
| | - Demokritos C Tsoukatos
- Department of Chemistry, Organic Chemistry & Biochemistry, University of Ioannina, Ioannina. Greece
| | - Vassilios Tsikaris
- Department of Chemistry, Organic Chemistry & Biochemistry, University of Ioannina, Ioannina. Greece
| |
Collapse
|
14
|
Roka-Moiia Y, Ammann KR, Miller-Gutierrez S, Sweedo A, Palomares D, Italiano J, Sheriff J, Bluestein D, Slepian MJ. Shear-mediated platelet activation in the free flow II: Evolving mechanobiological mechanisms reveal an identifiable signature of activation and a bi-directional platelet dyscrasia with thrombotic and bleeding features. J Biomech 2021; 123:110415. [PMID: 34052772 DOI: 10.1016/j.jbiomech.2021.110415] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 03/03/2021] [Indexed: 01/17/2023]
Abstract
Shear-mediated platelet activation (SMPA) in the "free flow" is the net result of a range of cell mechanobiological mechanisms. Previously, we outlined three main groups of mechanisms including: 1) mechano-destruction - i.e. additive platelet (membrane) damage; 2) mechano-activation - i.e. activation of shear-sensitive ion channels and pores; and 3) mechano-transduction - i.e. "outside-in" signaling via a range of transducers. Here, we report on recent advances since our original report which describes additional features of SMPA. A clear "signature" of SMPA has been defined, allowing differentiation from biochemically-mediated activation. Notably, SMPA is characterized by mitochondrial dysfunction, platelet membrane eversion, externalization of anionic phospholipids, and increased thrombin generation on the platelet surface. However, SMPA does not lead to integrin αIIbβ3 activation or P-selectin exposure due to platelet degranulation, as is commonly observed in biochemical activation. Rather, downregulation of GPIb, αIIbβ3, and P-selectin surface expression is evident. Furthermore, SMPA is accompanied by a decrease in overall platelet size coupled with a concomitant, progressive increase in microparticle generation. Shear-ejected microparticles are highly enriched in GPIb and αIIbβ3. These observations indicate the enhanced diffusion, migration, or otherwise dispersion of platelet adhesion receptors to membrane zones, which are ultimately shed as receptor-rich PDMPs. The pathophysiological consequence of this progressive shear accumulation phenomenon is an associated dyscrasia of remaining platelets - being both reduced in size and less activatable via biochemical means - a tendency to favor bleeding, while concomitantly shed microparticles are highly prothrombotic and increase the tendency for thrombosis in both local and systemic milieu. These mechanisms and observations offer direct clinical utility in allowing measurement and guidance of the net balance of platelet driven events in patients with implanted cardiovascular therapeutic devices.
Collapse
Affiliation(s)
- Yana Roka-Moiia
- Departments of Medicine and Biomedical Engineering, Sarver Heart Center, University of Arizona, Tucson, AZ 85721, United States; Arizona Center for Accelerated Biomedical Innovation, University of Arizona, Tucson, AZ 85721, United States
| | - Kaitlyn R Ammann
- Departments of Medicine and Biomedical Engineering, Sarver Heart Center, University of Arizona, Tucson, AZ 85721, United States; Arizona Center for Accelerated Biomedical Innovation, University of Arizona, Tucson, AZ 85721, United States
| | - Samuel Miller-Gutierrez
- Departments of Medicine and Biomedical Engineering, Sarver Heart Center, University of Arizona, Tucson, AZ 85721, United States; Arizona Center for Accelerated Biomedical Innovation, University of Arizona, Tucson, AZ 85721, United States
| | - Alice Sweedo
- Departments of Medicine and Biomedical Engineering, Sarver Heart Center, University of Arizona, Tucson, AZ 85721, United States; Arizona Center for Accelerated Biomedical Innovation, University of Arizona, Tucson, AZ 85721, United States
| | - Daniel Palomares
- Departments of Medicine and Biomedical Engineering, Sarver Heart Center, University of Arizona, Tucson, AZ 85721, United States; Arizona Center for Accelerated Biomedical Innovation, University of Arizona, Tucson, AZ 85721, United States
| | - Joseph Italiano
- Department of Medicine, Harvard Medical School, Boston, MA 02115, United States
| | - Jawaad Sheriff
- Department of Biomedical Engineering, Stony Brook University, NY 11794, United States
| | - Danny Bluestein
- Department of Biomedical Engineering, Stony Brook University, NY 11794, United States
| | - Marvin J Slepian
- Departments of Medicine and Biomedical Engineering, Sarver Heart Center, University of Arizona, Tucson, AZ 85721, United States; Department of Biomedical Engineering, Stony Brook University, NY 11794, United States; Arizona Center for Accelerated Biomedical Innovation, University of Arizona, Tucson, AZ 85721, United States.
| |
Collapse
|
15
|
van Geffen JP, Swieringa F, van Kuijk K, Tullemans BME, Solari FA, Peng B, Clemetson KJ, Farndale RW, Dubois LJ, Sickmann A, Zahedi RP, Ahrends R, Biessen EAL, Sluimer JC, Heemskerk JWM, Kuijpers MJE. Mild hyperlipidemia in mice aggravates platelet responsiveness in thrombus formation and exploration of platelet proteome and lipidome. Sci Rep 2020; 10:21407. [PMID: 33293576 PMCID: PMC7722935 DOI: 10.1038/s41598-020-78522-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/23/2020] [Indexed: 01/21/2023] Open
Abstract
Hyperlipidemia is a well-established risk factor for cardiovascular diseases. Millions of people worldwide display mildly elevated levels of plasma lipids and cholesterol linked to diet and life-style. While the prothrombotic risk of severe hyperlipidemia has been established, the effects of moderate hyperlipidemia are less clear. Here, we studied platelet activation and arterial thrombus formation in Apoe-/- and Ldlr-/- mice fed a normal chow diet, resulting in mildly increased plasma cholesterol. In blood from both knockout mice, collagen-dependent thrombus and fibrin formation under flow were enhanced. These effects did not increase in severe hyperlipidemic blood from aged mice and upon feeding a high-fat diet (Apoe-/- mice). Bone marrow from wild-type or Ldlr-/- mice was transplanted into irradiated Ldlr-/- recipients. Markedly, thrombus formation was enhanced in blood from chimeric mice, suggesting that the hyperlipidemic environment altered the wild-type platelets, rather than the genetic modification. The platelet proteome revealed high similarity between the three genotypes, without clear indication for a common protein-based gain-of-function. The platelet lipidome revealed an altered lipid profile in mildly hyperlipidemic mice. In conclusion, in Apoe-/- and Ldlr-/- mice, modest elevation in plasma and platelet cholesterol increased platelet responsiveness in thrombus formation and ensuing fibrin formation, resulting in a prothrombotic phenotype.
Collapse
Affiliation(s)
- Johanna P van Geffen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Frauke Swieringa
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.,Leibniz Institut für Analytische Wissenschaften - ISAS- e.V, Dortmund, Germany
| | - Kim van Kuijk
- Department of Pathology, CARIM, Maastricht University, Maastricht, The Netherlands
| | - Bibian M E Tullemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Fiorella A Solari
- Leibniz Institut für Analytische Wissenschaften - ISAS- e.V, Dortmund, Germany
| | - Bing Peng
- Leibniz Institut für Analytische Wissenschaften - ISAS- e.V, Dortmund, Germany
| | - Kenneth J Clemetson
- Department of Haematology, Inselspital, University of Bern, Bern, Switzerland
| | | | - Ludwig J Dubois
- The M-Lab, Department of Precision Medicine, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands
| | - Albert Sickmann
- Leibniz Institut für Analytische Wissenschaften - ISAS- e.V, Dortmund, Germany
| | - René P Zahedi
- Leibniz Institut für Analytische Wissenschaften - ISAS- e.V, Dortmund, Germany.,Segal Cancer Proteomics Centre, Jewish General Hospital, McGill University, Montreal, Canada
| | - Robert Ahrends
- Leibniz Institut für Analytische Wissenschaften - ISAS- e.V, Dortmund, Germany.,Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Wien, Austria
| | - Erik A L Biessen
- Department of Pathology, CARIM, Maastricht University, Maastricht, The Netherlands.,Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | - Judith C Sluimer
- Department of Pathology, CARIM, Maastricht University, Maastricht, The Netherlands.,BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Marijke J E Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
| |
Collapse
|
16
|
Cholesterol-Rich Microdomains Contribute to PAR1 Signaling in Platelets Despite a Weak Localization of the Receptor in These Microdomains. Int J Mol Sci 2020; 21:ijms21218065. [PMID: 33138025 PMCID: PMC7663584 DOI: 10.3390/ijms21218065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/22/2020] [Accepted: 10/27/2020] [Indexed: 01/03/2023] Open
Abstract
Platelet protease-activated receptor 1 (PAR1) is a cell surface G-protein-coupled receptor (GPCR) that acts as a thrombin receptor promoting platelet aggregation. Targeting the PAR1 pathway by vorapaxar, a PAR1 antagonist, leads to a reduction in ischemic events in cardiovascular patients with a history of myocardial infarction or with peripheral arterial disease. In platelets, specialized microdomains highly enriched in cholesterol act as modulators of the activity of several GPCRs and play a pivotal role in the signaling pathway. However, their involvement in platelet PAR1 function remains incompletely characterized. In this context, we aimed to investigate whether activation of PAR1 in human platelets requires its localization in the membrane cholesterol-rich microdomains. Using confocal microscopy, biochemical isolation, and proteomics approaches, we found that PAR1 was not localized in cholesterol-rich microdomains in resting platelets, and only a small fraction of the receptor relocated to the microdomains following its activation. Vorapaxar treatment increased the level of PAR1 at the platelet surface, possibly by reducing its endocytosis, while its colocalization with cholesterol-rich microdomains remained weak. Consistent with a cholesterol-dependent activation of Akt and p38 MAP kinase in thrombin receptor-activating peptide (TRAP)-activated platelets, the proteomic data of cholesterol-rich microdomains isolated from TRAP-activated platelets showed the recruitment of proteins contributing to these signaling pathways. In conclusion, contrary to endothelial cells, we found that PAR1 was only weakly present in cholesterol-rich microdomains in human platelets but used these microdomains for efficient activation of downstream signaling pathways following TRAP activation.
Collapse
|
17
|
Kessler T, Schunkert H, von Hundelshausen P. Novel Approaches to Fine-Tune Therapeutic Targeting of Platelets in Atherosclerosis: A Critical Appraisal. Thromb Haemost 2020; 120:1492-1504. [PMID: 32772352 DOI: 10.1055/s-0040-1714352] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The pathogenesis of atherosclerotic vascular disease is driven by a multitude of risk factors intertwining metabolic and inflammatory pathways. Increasing knowledge about platelet biology sheds light on how platelets take part in these processes from early to later stages of plaque development. Recent insights from experimental studies and mouse models substantiate platelets as initiators and amplifiers in atherogenic leukocyte recruitment. These studies are complemented by results from genetics studies shedding light on novel molecular mechanisms which provide an interesting prospect as novel targets. For instance, experimental studies provide further details how platelet-decorated von Willebrand factor tethered to activated endothelial cells plays a role in atherogenic monocyte recruitment. Novel aspects of platelets as atherogenic inductors of neutrophil extracellular traps and particularities in signaling pathways such as cyclic guanosine monophosphate and the inhibitory adaptor molecule SHB23/LNK associating platelets with atherogenesis are shared. In summary, it was our intention to balance insights from recent experimental data that support a plausible role for platelets in atherogenesis against a paucity of clinical evidence needed to validate this concept in humans.
Collapse
Affiliation(s)
- Thorsten Kessler
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany
| | - Philipp von Hundelshausen
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany.,Institut für Prophylaxe und Epidemiologie der Kreislaufkrankheiten, Klinikum der Universität, Ludwig-Maximilians-Universität, Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
18
|
Komatsuya K, Kaneko K, Kasahara K. Function of Platelet Glycosphingolipid Microdomains/Lipid Rafts. Int J Mol Sci 2020; 21:ijms21155539. [PMID: 32748854 PMCID: PMC7432685 DOI: 10.3390/ijms21155539] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 01/09/2023] Open
Abstract
Lipid rafts are dynamic assemblies of glycosphingolipids, sphingomyelin, cholesterol, and specific proteins which are stabilized into platforms involved in the regulation of vital cellular processes. The rafts at the cell surface play important functions in signal transduction. Recent reports have demonstrated that lipid rafts are spatially and compositionally heterogeneous in the single-cell membrane. In this review, we summarize our recent data on living platelets using two specific probes of raft components: lysenin as a probe of sphingomyelin-rich rafts and BCθ as a probe of cholesterol-rich rafts. Sphingomyelin-rich rafts that are spatially and functionally distinct from the cholesterol-rich rafts were found at spreading platelets. Fibrin is translocated to sphingomyelin-rich rafts and platelet sphingomyelin-rich rafts act as platforms where extracellular fibrin and intracellular actomyosin join to promote clot retraction. On the other hand, the collagen receptor glycoprotein VI is known to be translocated to cholesterol-rich rafts during platelet adhesion to collagen. Furthermore, the functional roles of platelet glycosphingolipids and platelet raft-binding proteins including G protein-coupled receptors, stomatin, prohibitin, flotillin, and HflK/C-domain protein family, tetraspanin family, and calcium channels are discussed.
Collapse
|
19
|
Li M, Li J, Chen J, Liu Y, Cheng X, Yang F, Gu N. Platelet Membrane Biomimetic Magnetic Nanocarriers for Targeted Delivery and in Situ Generation of Nitric Oxide in Early Ischemic Stroke. ACS NANO 2020; 14:2024-2035. [PMID: 31927980 DOI: 10.1021/acsnano.9b08587] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Early diagnosis and treatment of acute ischemic stroke poses a significant challenge due to its suddenness and short therapeutic time window. Human endogenous cells derived biomimetic drug carriers have provided new options for stroke theranostics since these cells have higher biosafety and targeting abilities than artificial carriers. Inspired by natural platelets (PLTs) and their role in targeting adhesion to the damaged blood vessel during thrombus formation, we fabricated a biomimetic nanocarrier comprising a PLT membrane envelope loaded with l-arginine and γ-Fe2O3 magnetic nanoparticles (PAMNs) for thrombus-targeted delivery of l-arginine and in situ generation of nitric oxide (NO). Results demonstrate that the engineered 200 nm PAMNs inherit the natural properties of the PLT membrane and achieve rapid targeting to ischemic stroke lesions under the guidance of an external magnetic field. Subsequent to the release of l-arginine at the thrombus site, endothelial cells produce NO, which promotes vasodilation to disrupt the local PLT aggregation. Rapid targeting of PAMNs to stroke lesions as well as in situ generation of NO prompts vasodilation, recovery of blood flow, and reperfusion of the stroke microvascular. Thus, these PLT membrane derived nanocarriers are diagnostically beneficial for localizing stroke lesions and a promising modality for executing therapies.
Collapse
Affiliation(s)
- Mingxi Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering , Southeast University , Nanjing 210096 , China
| | - Jing Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering , Southeast University , Nanjing 210096 , China
| | - Jinpeng Chen
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Microbiology and Immunology, Medical School , Southeast University , Nanjing 210009 , China
| | - Yang Liu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering , Southeast University , Nanjing 210096 , China
| | - Xiao Cheng
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering , Southeast University , Nanjing 210096 , China
| | - Fang Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering , Southeast University , Nanjing 210096 , China
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering , Southeast University , Nanjing 210096 , China
| |
Collapse
|
20
|
Green SM, Padula MP, Marks DC, Johnson L. The Lipid Composition of Platelets and the Impact of Storage: An Overview. Transfus Med Rev 2020; 34:108-116. [PMID: 31987597 DOI: 10.1016/j.tmrv.2019.12.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/01/2019] [Accepted: 12/07/2019] [Indexed: 02/07/2023]
Abstract
Lipids and bioactive lipid mediators are essential for platelet function. The lipid profile of platelets is highly dynamic due to free exchange of lipids with the plasma, release of extracellular vesicles, and both enzymatic and nonenzymatic lipid conversion. The lipidome of platelets changes in response to activation to accommodate the functional requirements of platelets, particularly for maintenance of hemostasis. Furthermore, when stored at room temperature as a component for transfusion, the lipid profile of platelets is altered. Although there is a growing interest in alternate storage conditions, such as refrigeration and cryopreservation, few contemporary studies have examined the impact of these storage modes on the lipid profile. However, evidence exists that bioactive lipid mediators produced over the storage of blood products may have functional implications once these products are transfused. As such, there is a need to determine the changes occurring to the lipid profile of these products over storage. This review outlines the role of lipids in platelets and discusses the current state of lipidomics for studying platelet components for transfusion in an effort to highlight the necessity for additional transfusion-focused investigations.
Collapse
Affiliation(s)
- Sarah M Green
- Research & Development, Australian Red Cross Blood Service, Alexandria, NSW, Australia; School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Matthew P Padula
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Denese C Marks
- Research & Development, Australian Red Cross Blood Service, Alexandria, NSW, Australia; Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Lacey Johnson
- Research & Development, Australian Red Cross Blood Service, Alexandria, NSW, Australia.
| |
Collapse
|
21
|
Stivala S, Sorrentino S, Gobbato S, Bonetti NR, Camici GG, Lüscher TF, Medalia O, Beer JH. Glycoprotein Ib clustering in platelets can be inhibited by α-linolenic acid as revealed by cryo-electron tomography. Haematologica 2019; 105:1660-1666. [PMID: 31439672 PMCID: PMC7271563 DOI: 10.3324/haematol.2019.220988] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 08/14/2019] [Indexed: 11/26/2022] Open
Abstract
Platelet adhesion to the sub-endothelial matrix and damaged endothelium occurs through a multi-step process mediated in the initial phase by glycoprotein Ib binding to von Willebrand factor (vWF), which leads to the subsequent formation of a platelet plug. The plant-derived ω-3 fatty acid α-linolenic acid is an abundant alternative to fish-derived n-3 fatty acids and has anti-inflammatory and antithrombotic properties. In this study, we investigated the impact of α-linolenic acid on human platelet binding to vWF under high-shear flow conditions (mimicking blood flow in stenosed arteries). Pre-incubation of fresh human blood from healthy donors with α-linolenic acid at dietary relevant concentrations reduced platelet binding and rolling on vWF-coated microchannels at a shear rate of 100 dyn/cm2. Depletion of membrane cholesterol by incubation of platelet-rich plasma with methyl-β cyclodextrin abrogated platelet rolling on vWF. Analysis of glycoprotein Ib by applying cryo-electron tomography to intact platelets revealed local clusters of glycoprotein Ib complexes upon exposure to shear force: the formation of these complexes could be prevented by treatment with α-linolenic acid. This study provides novel findings on the rapid local rearrangement of glycoprotein Ib complexes in response to high-shear flow and highlights the mechanism of in vitro inhibition of platelet binding to and rolling on vWF by α-linolenic acid.
Collapse
Affiliation(s)
- Simona Stivala
- Laboratory for Platelet Research, Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Simona Sorrentino
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Sara Gobbato
- Laboratory for Platelet Research, Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Nicole R Bonetti
- Laboratory for Platelet Research, Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland.,Internal Medicine, Cantonal Hospital of Baden, Baden, Switzerland
| | - Giovanni G Camici
- Laboratory for Platelet Research, Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland.,University Heart Center, University Hospital Zurich, Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Thomas F Lüscher
- Laboratory for Platelet Research, Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Zurich, Switzerland.,Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University, Beer-Sheva, Israel
| | - Jürg H Beer
- Laboratory for Platelet Research, Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland .,Internal Medicine, Cantonal Hospital of Baden, Baden, Switzerland
| |
Collapse
|
22
|
Ma X, Jia C, Fu D, Chu M, Ding X, Wu X, Guo X, Pei J, Bao P, Liang C, Yan P. Analysis of Hematological Traits in Polled Yak by Genome-Wide Association Studies Using Individual SNPs and Haplotypes. Genes (Basel) 2019; 10:E463. [PMID: 31212963 PMCID: PMC6627507 DOI: 10.3390/genes10060463] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/11/2019] [Accepted: 06/12/2019] [Indexed: 12/21/2022] Open
Abstract
Yak (Bos grunniens) is an important domestic animal living in high-altitude plateaus. Due to inadequate disease prevention, each year, the yak industry suffers significant economic losses. The identification of causal genes that affect blood- and immunity-related cells could provide preliminary reference guidelines for the prevention of diseases in the population of yaks. The genome-wide association studies (GWASs) utilizing a single-marker or haplotype method were employed to analyze 15 hematological traits in the genome of 315 unrelated yaks. Single-marker GWASs identified a total of 43 significant SNPs, including 35 suggestive and eight genome-wide significant SNPs, associated with nine traits. Haplotype analysis detected nine significant haplotype blocks, including two genome-wide and seven suggestive blocks, associated with seven traits. The study provides data on the genetic variability of hematological traits in the yak. Five essential genes (GPLD1, EDNRA,APOB, HIST1H1E, and HIST1H2BI) were identified, which affect the HCT, HGB, RBC, PDW, PLT, and RDWSD traits and can serve as candidate genes for regulating hematological traits. The results provide a valuable reference to be used in the analysis of blood properties and immune diseases in the yak.
Collapse
Affiliation(s)
- Xiaoming Ma
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China.
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China.
| | - Congjun Jia
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China.
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China.
| | - Donghai Fu
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China.
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China.
| | - Min Chu
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China.
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China.
| | - Xuezhi Ding
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China.
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China.
| | - Xiaoyun Wu
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China.
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China.
| | - Xian Guo
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China.
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China.
| | - Jie Pei
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China.
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China.
| | - Pengjia Bao
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China.
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China.
| | - Chunnian Liang
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China.
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China.
| | - Ping Yan
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China.
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Lanzhou 730050, China.
| |
Collapse
|
23
|
Martyanov AA, Kaneva VN, Panteleev MA, Sveshnikova AN. [CLEC-2 induced signalling in blood platelets]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2019; 64:387-396. [PMID: 30378555 DOI: 10.18097/pbmc20186405387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Platelet activating receptor CLEC-2 has been identified on platelet surface a decade ago. The only confirmed endogenous CLEC-2 agonist is podoplanin. Podoplanin is a transmembrane protein expressed by lymphatic endothelial cells, reticular fibroblastic cells in lymph nodes, kidney podocytes and by cells of certain tumors. CLEC-2 and podoplanin are involved in the processes of embryonic development (blood-lymph vessel separation and angiogenesis), maintaining of vascular integrity of small vessels during inflammation and prevention of blood-lymphatic mixing in high endothelial venules. However, CLEC-2 and podoplanin are contributing to tumor methastasis progression, Salmonella sepsis, deep-vein thrombosis. CLEC-2 signalling cascade includes tyrosine-kinases (Syk, SFK, Btk) as well as adapter LAT and phospholipase Cg2, which induces calcium signalling. CLEC-2, podoplanin and proteins, participating in CLEC-2 signalling cascade, are perspective targets for antithrombotic therapy.
Collapse
Affiliation(s)
- A A Martyanov
- Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia; Center for Theoretical Problems of Physicochemical Pharmacology, Moscow, Russia
| | - V N Kaneva
- Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia; Rogachev National Scientific and Practical Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - M A Panteleev
- Center for Theoretical Problems of Physicochemical Pharmacology, Moscow, Russia; Rogachev National Scientific and Practical Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - A N Sveshnikova
- Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia; Center for Theoretical Problems of Physicochemical Pharmacology, Moscow, Russia
| |
Collapse
|
24
|
A Novel Discovery: Holistic Efficacy at the Special Organ Level of Pungent Flavored Compounds from Pungent Traditional Chinese Medicine. Int J Mol Sci 2019; 20:ijms20030752. [PMID: 30754631 PMCID: PMC6387020 DOI: 10.3390/ijms20030752] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 12/25/2022] Open
Abstract
Pungent traditional Chinese medicines (TCMs) play a vital role in the clinical treatment of hepatobiliary disease, gastrointestinal diseases, cardiovascular diseases, diabetes, skin diseases and so on. Pungent TCMs have a vastness of pungent flavored (with pungent taste or smell) compounds. To elucidate the molecular mechanism of pungent flavored compounds in treating cardiovascular diseases (CVDs) and liver diseases, five pungent TCMs with the action of blood-activating and stasis-resolving (BASR) were selected. Here, an integrated systems pharmacology approach is presented for illustrating the molecular correlations between pungent flavored compounds and their holistic efficacy at the special organ level. First, we identified target proteins that are associated with pungent flavored compounds and found that these targets were functionally related to CVDs and liver diseases. Then, based on the phenotype that directly links human genes to the body parts they affect, we clustered target modules associated with pungent flavored compounds into liver and heart organs. We applied systems-based analysis to introduce a pungent flavored compound-target-pathway-organ network that clarifies mechanisms of pungent substances treating cardiovascular diseases and liver diseases by acting on the heart/liver organ. The systems pharmacology also suggests a novel systematic strategy for rational drug development from pungent TCMs in treating cardiovascular disease and associated liver diseases.
Collapse
|
25
|
Izquierdo I, Barrachina MN, Hermida-Nogueira L, Casas V, Eble JA, Carrascal M, Abián J, García Á. Platelet membrane lipid rafts protein composition varies following GPVI and CLEC-2 receptors activation. J Proteomics 2019; 195:88-97. [PMID: 30677554 DOI: 10.1016/j.jprot.2019.01.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/03/2019] [Accepted: 01/20/2019] [Indexed: 12/11/2022]
Abstract
Lipid rafts are membrane microdomains that have been proposed to play an important role in several platelet-signalling cascades, including those mediated by the receptors Glycoprotein VI (GPVI), and C-type lectin domain family 1 member B (CLEC-2), both involved in thrombus formation. We have performed a LC-MS/MS proteomic analysis of lipid rafts isolated from platelets activated through GPVI and CLEC-2 as well as from resting platelets. Our aim was to determine the magnitude of changes in lipid rafts protein composition and to elucidate the relevance of these alterations in platelet function. A number of relevant signalling proteins were found enriched in lipid rafts following platelet activation (such as the tyrosine protein kinases Fyn, Lyn and Yes; the G proteins G(i) and G(z); and cAMP protein kinase). Interestingly, our results indicate that the relative enrichment of lipid rafts in these signalling proteins may not be a consequence of protein translocation to these domains upon platelet stimulation, but the result of a massive loss in cytoskeletal proteins after platelet activation. Thus, this study may help to better understand the effects of platelet activation in the reorganization of lipid rafts and set the basis for further proteomic studies of these membrane microdomains in platelets. SIGNIFICANCE: We performed the first proteomic comparative analysis of lipid rafts- protein composition in platelets activated through GPVI and CLEC-2 receptors and in resting state. We identified a number of signalling proteins essential for platelet activation relatively enriched in platelets activated through both receptors, and we show that lipid rafts reorganization upon platelet activation leads to a loss in cytoskeletal proteins, highly associated to these domains in resting platelets.
Collapse
Affiliation(s)
- Irene Izquierdo
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - María N Barrachina
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Lidia Hermida-Nogueira
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Vanessa Casas
- CSIC/UAB Proteomics Laboratory, IIBB-CSIC-IDIBAPS, Barcelona, Spain
| | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | | | - Joaquín Abián
- CSIC/UAB Proteomics Laboratory, IIBB-CSIC-IDIBAPS, Barcelona, Spain
| | - Ángel García
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain.
| |
Collapse
|
26
|
The relation between ABO blood types and clinical and platelet function parameters in patients who underwent percutaneous coronary intervention. Coron Artery Dis 2019; 30:51-58. [DOI: 10.1097/mca.0000000000000676] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
27
|
Pollet H, Conrard L, Cloos AS, Tyteca D. Plasma Membrane Lipid Domains as Platforms for Vesicle Biogenesis and Shedding? Biomolecules 2018; 8:E94. [PMID: 30223513 PMCID: PMC6164003 DOI: 10.3390/biom8030094] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 09/03/2018] [Accepted: 09/04/2018] [Indexed: 12/18/2022] Open
Abstract
Extracellular vesicles (EVs) contribute to several pathophysiological processes and appear as emerging targets for disease diagnosis and therapy. However, successful translation from bench to bedside requires deeper understanding of EVs, in particular their diversity, composition, biogenesis and shedding mechanisms. In this review, we focus on plasma membrane-derived microvesicles (MVs), far less appreciated than exosomes. We integrate documented mechanisms involved in MV biogenesis and shedding, focusing on the red blood cell as a model. We then provide a perspective for the relevance of plasma membrane lipid composition and biophysical properties in microvesiculation on red blood cells but also platelets, immune and nervous cells as well as tumor cells. Although only a few data are available in this respect, most of them appear to converge to the idea that modulation of plasma membrane lipid content, transversal asymmetry and lateral heterogeneity in lipid domains may play a significant role in the vesiculation process. We suggest that lipid domains may represent platforms for inclusion/exclusion of membrane lipids and proteins into MVs and that MVs could originate from distinct domains during physiological processes and disease evolution.
Collapse
Affiliation(s)
- Hélène Pollet
- CELL Unit, de Duve Institute & Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, B-1200 Brussels, Belgium.
| | - Louise Conrard
- CELL Unit, de Duve Institute & Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, B-1200 Brussels, Belgium.
| | - Anne-Sophie Cloos
- CELL Unit, de Duve Institute & Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, B-1200 Brussels, Belgium.
| | - Donatienne Tyteca
- CELL Unit, de Duve Institute & Université Catholique de Louvain, UCL B1.75.05, Avenue Hippocrate, 75, B-1200 Brussels, Belgium.
| |
Collapse
|
28
|
Garcia C, Montée N, Faccini J, Series J, Meilhac O, Cantero AV, Le Faouder P, Elbaz M, Payrastre B, Vindis C. Acute coronary syndrome remodels the antiplatelet aggregation properties of HDL particle subclasses. J Thromb Haemost 2018; 16:933-945. [PMID: 29543379 DOI: 10.1111/jth.14003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Indexed: 01/29/2023]
Abstract
Essentials HDL subclasses were studied in acute coronary syndrome (ACS). HDL2 from ACS patients have better antiplatelet potency than HDL from non ACS subjects. ACS remodels the antiplatelet properties of HDL subclasses. Oxidized polyunsaturated fatty acids content of HDL is modified by ACS. SUMMARY Background Although HDLs have antithrombotic effects by reducing platelet activation, the relationship between HDL levels and the risk of acute coronary syndrome (ACS) is unclear, as HDL particles are heterogeneous in composition and biological properties. Objective To characterize the effects of HDL2 and HDL3 subclasses from ACS patients and non-coronary artery disease (CAD) subjects on platelet activation. Methods We measured platelet aggregation and ex vivo thrombus formation, analyzed signaling pathways by flow cytometry, and performed a targeted lipidomics analysis on HDL subclasses. Results Analysis of human platelet aggregation in suspension, adhesion on von Willebrand factor and thrombus formation on collagen under arterial shear demonstrated that HDL2 from ACS patients had higher antiplatelet potency than HDL3 from ACS patients and HDL from non-CAD subjects. HDL binding to scavenger receptor class B type I was essential for this effect. A lipidomics analysis revealed that HDL2 from ACS patients had more oxidized polyunsaturated fatty acids (PUFAs). An inverse correlation between the concentrations of 9-hydroxyoctadecadienoic acid (9-HODE), 13-hydroxyoctadecadienoic acid (13-HODE), the eicosapentaenoic acid metabolite 18-hydroxyeicosapentaenoic acid (18-HEPE) and hydroxyeicosatetraenoic acid isomers in HDL2 and platelet aggregation was observed. This relationship was further demonstrated by the direct inhibitory effects of 18-HEPE, 9-HODE, 13-HODE, 17-hydroxydocosahexaenoic acid and 14-hydroxydocosahexaenoic acid on collagen-related peptide-induced platelet aggregation, indicating that oxidized PUFAs contribute to the antithrombotic effect of ACS HDL2. Conclusions Our data shed new light on the antiplatelet effects of HDL subclasses, and suggest physiological adaptation through the modulation of HDL properties in ACS patients that may limit their platelet-dependent thrombotic risk.
Collapse
Affiliation(s)
- C Garcia
- Laboratory of Hematology, CHU Toulouse, Toulouse, France
| | - N Montée
- Institute of Metabolic and Cardiovascular Diseases/I2MC, INSERM, UMR-1048 and University Toulouse 3, Toulouse, France
- INSERM, UMR-1188, Diabète Athérothrombose Thérapies Réunion Océan Indien, Université de la Réunion, Saint Denis, France
| | - J Faccini
- Institute of Metabolic and Cardiovascular Diseases/I2MC, INSERM, UMR-1048 and University Toulouse 3, Toulouse, France
| | - J Series
- Laboratory of Hematology, CHU Toulouse, Toulouse, France
| | - O Meilhac
- INSERM, UMR-1188, Diabète Athérothrombose Thérapies Réunion Océan Indien, Université de la Réunion, Saint Denis, France
| | - A-V Cantero
- Institute of Metabolic and Cardiovascular Diseases/I2MC, INSERM, UMR-1048 and University Toulouse 3, Toulouse, France
- Laboratory of Biochemistry, CHU Toulouse, Toulouse, France
| | - P Le Faouder
- MetaToul-Lipidomic Core Facility, MetaboHUB, INSERM, UMR-1048, Toulouse, France
| | - M Elbaz
- Institute of Metabolic and Cardiovascular Diseases/I2MC, INSERM, UMR-1048 and University Toulouse 3, Toulouse, France
- Department of Cardiology, CHU Toulouse, Toulouse, France
| | - B Payrastre
- Laboratory of Hematology, CHU Toulouse, Toulouse, France
- Institute of Metabolic and Cardiovascular Diseases/I2MC, INSERM, UMR-1048 and University Toulouse 3, Toulouse, France
| | - C Vindis
- Laboratory of Hematology, CHU Toulouse, Toulouse, France
- Institute of Metabolic and Cardiovascular Diseases/I2MC, INSERM, UMR-1048 and University Toulouse 3, Toulouse, France
| |
Collapse
|
29
|
Secretory phospholipase A 2 modified HDL rapidly and potently suppresses platelet activation. Sci Rep 2017; 7:8030. [PMID: 28808297 PMCID: PMC5556053 DOI: 10.1038/s41598-017-08136-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 07/05/2017] [Indexed: 12/16/2022] Open
Abstract
Levels of secretory phospholipases A2 (sPLA2) highly increase under acute and chronic inflammatory conditions. sPLA2 is mainly associated with high-density lipoproteins (HDL) and generates bioactive lysophospholipids implicated in acute and chronic inflammatory processes. Unexpectedly, pharmacological inhibition of sPLA2 in patients with acute coronary syndrome was associated with an increased risk of myocardial infarction and stroke. Given that platelets are key players in thrombosis and inflammation, we hypothesized that sPLA2-induced hydrolysis of HDL-associated phospholipids (sPLA2-HDL) generates modified HDL particles that affect platelet function. We observed that sPLA2-HDL potently and rapidly inhibited platelet aggregation induced by several agonists, P-selectin expression, GPIIb/IIIa activation and superoxide production, whereas native HDL showed little effects. sPLA2-HDL suppressed the agonist-induced rise of intracellular Ca2+ levels and phosphorylation of Akt and ERK1/2, which trigger key steps in promoting platelet activation. Importantly, sPLA2 in the absence of HDL showed no effects, whereas enrichment of HDL with lysophosphatidylcholines containing saturated fatty acids (the main sPLA2 products) mimicked sPLA2-HDL activities. Our findings suggest that sPLA2 generates lysophosphatidylcholine-enriched HDL particles that modulate platelet function under inflammatory conditions.
Collapse
|
30
|
Saha AK, Osmulski P, Dallo SF, Gaczynska M, Huang THM, Ramasubramanian AK. Cholesterol Regulates Monocyte Rolling through CD44 Distribution. Biophys J 2017; 112:1481-1488. [PMID: 28402890 DOI: 10.1016/j.bpj.2017.02.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 01/16/2017] [Accepted: 02/08/2017] [Indexed: 10/19/2022] Open
Abstract
Cholesterol is an important risk factor of atherosclerosis, due to its active uptake by monocytes/macrophages. Monocyte recruitment from flowing blood to atherosclerotic foci is the key first step in the development of atherosclerosis. Cholesterol content alters cell membrane stiffness, and lateral lipid and protein diffusion. We hypothesized that cholesterol content will modulate the recruitment of monocytes to inflamed endothelial surface by altering the dynamics of adhesion receptors. We depleted or enriched the cellular cholesterol levels using methyl-β-cyclodextran in freshly isolated human monocytes. We investigated the effect of these changes on the mechanics of monocyte rolling on E-selectin surfaces at 1 dyn/cm2 in microchannels. Using imaging flow cytometry and atomic force microscopy, we characterized the distribution of lipid rafts and the E-selectin counterreceptor CD44 on the monocyte surface. We observed that lower levels of cholesterol resulted in the uniform, CD44-mediated rolling of monocytes on the E-selectin-coated surfaces. We also observed that cells depleted of cholesterol had higher membrane fluidity, and more uniform distribution of CD44 counterreceptor, which resulted in smooth motion of the cells compared to cells enriched with cholesterol. This work demonstrates that cholesterol can modulate monocyte adhesion by regulating the receptor mobility, and our results provide insights into the biophysical regulation of inflammation for the better understanding of diseases like atherosclerosis and hypercholesterolemia.
Collapse
Affiliation(s)
- Amit K Saha
- Department of Biomedical Engineering, The University of Texas at San Antonio, San Antonio, Texas
| | - Pawel Osmulski
- Department of Molecular Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Shatha F Dallo
- Department of Biomedical Engineering, The University of Texas at San Antonio, San Antonio, Texas
| | - Maria Gaczynska
- Department of Molecular Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Tim H-M Huang
- Department of Molecular Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Anand K Ramasubramanian
- Department of Biomedical Engineering, The University of Texas at San Antonio, San Antonio, Texas; Department of Biomedical, Chemical and Materials Engineering, San José State University, San José, California.
| |
Collapse
|
31
|
SDF-1α/CXCR4 Signaling in Lipid Rafts Induces Platelet Aggregation via PI3 Kinase-Dependent Akt Phosphorylation. PLoS One 2017; 12:e0169609. [PMID: 28072855 PMCID: PMC5224795 DOI: 10.1371/journal.pone.0169609] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 12/18/2016] [Indexed: 01/05/2023] Open
Abstract
Stromal cell-derived factor-1α (SDF-1α)-induced platelet aggregation is mediated through its G protein-coupled receptor CXCR4 and phosphatidylinositol 3 kinase (PI3K). Here, we demonstrate that SDF-1α induces phosphorylation of Akt at Thr308 and Ser473 in human platelets. SDF-1α-induced platelet aggregation and Akt phosphorylation are inhibited by pretreatment with the CXCR4 antagonist AMD3100 or the PI3K inhibitor LY294002. SDF-1α also induces the phosphorylation of PDK1 at Ser241 (an upstream activator of Akt), GSK3β at Ser9 (a downstream substrate of Akt), and myosin light chain at Ser19 (a downstream element of the Akt signaling pathway). SDF-1α-induced platelet aggregation is inhibited by pretreatment with the Akt inhibitor MK-2206 in a dose-dependent manner. Furthermore, SDF-1α-induced platelet aggregation and Akt phosphorylation are inhibited by pretreatment with the raft-disrupting agent methyl-β-cyclodextrin. Sucrose density gradient analysis shows that 35% of CXCR4, 93% of the heterotrimeric G proteins Gαi-1, 91% of Gαi-2, 50% of Gβ and 4.0% of PI3Kβ, and 4.5% of Akt2 are localized in the detergent-resistant membrane raft fraction. These findings suggest that SDF-1α/CXCR4 signaling in lipid rafts induces platelet aggregation via PI3K-dependent Akt phosphorylation.
Collapse
|
32
|
Izquierdo I, García Á. Platelet proteomics applied to the search for novel antiplatelet therapeutic targets. Expert Rev Proteomics 2016; 13:993-1006. [DOI: 10.1080/14789450.2016.1246188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
33
|
Hegde S, Cancelas JA. Dissecting the metabolic pathways controlling platelet survival in vivo: are our platelets what they eat? Transfusion 2016; 56:1928-31. [PMID: 27500916 DOI: 10.1111/trf.13684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 05/09/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Shailaja Hegde
- Hoxworth Blood Center, University of Cincinnati College of Medicine.,Division of Experimental Hematology, Cincinnati Children's Research Foundation, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Jose A Cancelas
- Hoxworth Blood Center, University of Cincinnati College of Medicine.,Division of Experimental Hematology, Cincinnati Children's Research Foundation, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
34
|
Rabani V, Davani S, Gambert-Nicot S, Meneveau N, Montange D. Comparative lipidomics and proteomics analysis of platelet lipid rafts using different detergents. Platelets 2016; 27:634-641. [DOI: 10.3109/09537104.2016.1174203] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
35
|
Rabani V, Montange D, Davani S. Interactive protein network of FXIII-A1 in lipid rafts of activated and non-activated platelets. Platelets 2016; 27:598-602. [PMID: 27540960 DOI: 10.3109/09537104.2016.1153621] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Lipid-rafts are defined as membrane microdomains enriched in cholesterol and glycosphingolipids within platelet plasma membrane. Lipid raft-mediated clot retraction requires factor XIII and other interacting proteins. The aim of this study was to investigate the proteins that interact with factor XIII in raft and non-raft domains of activated and non-activated platelet plasma membrane. By lipidomics analysis, we identified cholesterol- and sphingomyelin-enriched areas as lipid rafts. Platelets were activated by thrombin. Proteomics analysis provided an overview of the pathways in which proteins of rafts and non-rafts participated in the interaction network of FXIII-A1, a catalytic subunit of FXIII. "Platelet activation" was the principal pathway among KEGG pathways for proteins of rafts, both before and after activation. Network analysis showed four types of interactions (activation, binding, reaction, and catalysis) in raft and non-raft domains in interactive network of FXIII-A1. FXIII-A1 interactions with other proteins in raft domains and their role in homeostasis highlight the specialization of the raft domain in clot retraction via the Factor XIII protein network.
Collapse
Affiliation(s)
- Vahideh Rabani
- a EA 3920 - Université de Franche Comté , Besançon , France
| | - Damien Montange
- a EA 3920 - Université de Franche Comté , Besançon , France.,b Laboratoire de Pharmacologie Clinique et Toxicologie , CHU de Besançon , France
| | - Siamak Davani
- a EA 3920 - Université de Franche Comté , Besançon , France.,b Laboratoire de Pharmacologie Clinique et Toxicologie , CHU de Besançon , France
| |
Collapse
|
36
|
Belleville-Rolland T, Sassi Y, Decouture B, Dreano E, Hulot JS, Gaussem P, Bachelot-Loza C. MRP4 (ABCC4) as a potential pharmacologic target for cardiovascular disease. Pharmacol Res 2016; 107:381-389. [PMID: 27063943 DOI: 10.1016/j.phrs.2016.04.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 04/02/2016] [Indexed: 01/13/2023]
Abstract
This review focuses on multidrug resistance protein 4 (MRP4 or ABCC4) that has recently been shown to play a role in cAMP homeostasis, a key-pathway in vascular biology and in platelet functions. In vascular system, recent data provide evidence that inhibition of MRP4 prevents human coronary artery smooth muscle cell proliferation in vitro and in vivo, as well as human pulmonary artery smooth muscle cell proliferation in vitro and pulmonary hypertension in mice in vivo. In the heart, MRP4 silencing in adult rat ventricular myocytes results in an increase in intracellular cAMP levels leading to enhanced cardiomyocyte contractility. However, a prolonged inhibition of MRP4 can promote cardiac hypertrophy. In addition, secreted cAMP, through its metabolite adenosine, prevents adrenergically induced cardiac hypertrophy and fibrosis. Finally, MRP4 inhibition in platelets induces a moderate thrombopathy. The localization of MRP4 underlines the emerging concept of cAMP compartmentalization in platelets, which is a major regulatory mechanism in other cells. cAMP storage in platelet dense granules might limit the cAMP cytosolic concentration upon adenylate cyclase activation, a necessary step to induce platelet activation. In this review, we discuss the therapeutic potential of direct pharmacological inhibition of MRP4 in atherothrombotic disease, via its vasodilating and antiplatelet effects.
Collapse
Affiliation(s)
- Tiphaine Belleville-Rolland
- Inserm UMR-S1140, Faculté de Pharmacie, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; AP-HP, Hôpital Européen Georges Pompidou, Service dhématologie biologique, Paris, France
| | - Yassine Sassi
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benoit Decouture
- Inserm UMR-S1140, Faculté de Pharmacie, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Elise Dreano
- Inserm UMR-S1140, Faculté de Pharmacie, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jean-Sébastien Hulot
- AP-HP, Institute of Cardiometabolism and Nutrition (ICAN), Pitié-Salpêtrière Hospital, F-75013 Paris, France; Sorbonne Universités, UPMC Univ. Paris 06, France
| | - Pascale Gaussem
- Inserm UMR-S1140, Faculté de Pharmacie, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; AP-HP, Hôpital Européen Georges Pompidou, Service dhématologie biologique, Paris, France
| | - Christilla Bachelot-Loza
- Inserm UMR-S1140, Faculté de Pharmacie, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
37
|
Lhermusier T, Severin S, Van Rothem J, Garcia C, Bertrand-Michel J, Le Faouder P, Hechler B, Broccardo C, Couvert P, Chimini G, Sié P, Payrastre B. ATP-binding cassette transporter 1 (ABCA1) deficiency decreases platelet reactivity and reduces thromboxane A2 production independently of hematopoietic ABCA1. J Thromb Haemost 2016; 14:585-95. [PMID: 26749169 DOI: 10.1111/jth.13247] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Indexed: 02/03/2023]
Abstract
UNLABELLED ESSENTIALS: The role of ATP-binding cassette transporter 1 (ABCA1) in platelet functions is poorly characterized. We studied the impact of ABCA1 deficiency on platelet responses in a mouse model and two Tangier patients. ABCA1-deficient platelets exhibit reduced positive feedback loop mechanisms. This reduced reactivity is dependent on external environment and independent of hematopoietic ABCA1. SUMMARY BACKGROUND The ATP-binding cassette transporter ABCA1 is required for the conversion of apolipoprotein A-1 to high-density lipoprotein (HDL), and its defect causes Tangier disease, a rare disorder characterized by an absence of HDL and accumulation of cholesterol in peripheral tissues. The role of ABCA1 in platelet functions remains poorly characterized. OBJECTIVE To determine the role of ABCA1 in platelet functions and to clarify controversies concerning its implication in processes as fundamental as platelet phosphatidylserine exposure and control of platelet membrane lipid composition. METHODS AND RESULTS We studied the impact of ABCA1 deficiency on platelet responses in a mouse model and in two Tangier patients. We show that platelets in ABCA1-deficient mice are slightly larger in size and exhibit aggregation and secretion defects in response to low concentrations of thrombin and collagen. These platelets have normal cholesterol and major phospholipid composition, granule morphology, or calcium-induced phosphatidylserine exposure. Interestingly, ABCA1-deficient platelets display a reduction in positive feedback loop mechanisms, particularly in thromboxane A2 (TXA2) production. Hematopoietic chimera mice demonstrated that defective eicosanoids production, particularly TXA2, was primarily dependent on external environment and not on the hematopoietic ABCA1. Decreased aggregation and production of TXA2 and eicosanoids were also observed in platelets from Tangier patients. CONCLUSIONS Absence of ABCA1 and low HDL level induce reduction of platelet reactivity by decreasing positive feedback loops, particularly TXA2 production through a hematopoietic ABCA1-independent mechanism.
Collapse
Affiliation(s)
- T Lhermusier
- Inserm, U1048 et Université Toulouse 3, I2MC, Toulouse, France
- Département de Cardiologie, CHU de Toulouse, Toulouse, France
| | - S Severin
- Inserm, U1048 et Université Toulouse 3, I2MC, Toulouse, France
| | - J Van Rothem
- Inserm, U1048 et Université Toulouse 3, I2MC, Toulouse, France
- Département de Cardiologie, CHU de Toulouse, Toulouse, France
| | - C Garcia
- Laboratoire d'Hématologie, CHU de Toulouse, Toulouse, France
| | | | - P Le Faouder
- I2MC, Lipidomic Core Facility-MetaToul, Toulouse, France
| | - B Hechler
- Inserm UMR-S949, Etablissement Français du Sang-Alsace, Strasbourg, France
| | | | - P Couvert
- Service de Biochimie Endocrinienne et Oncologique, Hôpital Pitié Salpêtrière, Paris, France
| | - G Chimini
- CIML, Parc Scientifique de Luminy, Case 906, Marseille Luminy, France
| | - P Sié
- Inserm, U1048 et Université Toulouse 3, I2MC, Toulouse, France
- Laboratoire d'Hématologie, CHU de Toulouse, Toulouse, France
| | - B Payrastre
- Inserm, U1048 et Université Toulouse 3, I2MC, Toulouse, France
- Laboratoire d'Hématologie, CHU de Toulouse, Toulouse, France
| |
Collapse
|
38
|
Qu D, Huang H, DI J, Gao K, Lu Z, Zheng J. Structure, functional regulation and signaling properties of Rap2B. Oncol Lett 2016; 11:2339-2346. [PMID: 27073477 DOI: 10.3892/ol.2016.4261] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Accepted: 12/17/2015] [Indexed: 12/16/2022] Open
Abstract
The Ras family small guanosine 5'-triphosphate (GTP)-binding protein Rap2B is is a member of the Ras oncogene family and a novel target of p53 that regulates the p53-mediated pro-survival function of cells. The Rap2B protein shares ~90% homology with Rap2A, and its sequence is 70% identical to other members of the Rap family such as RaplA and RaplB. As a result, Rap2B has been theorized to have similar signaling effectors to the GTPase-binding protein Rap, which mediates various biological functions, including the regulation of sterile 20/mitogen-activated proteins. Since its identification in the early 1990s, Rap2B has elicited a considerable interest. Numerous studies indicate that Rap2B exerts specific biological functions, including binding and stimulating phospholipase C-ε and interferon-γ. In addition, downregulation of Rap2B affects the growth of melanoma cells. The present review summarizes the possible effectors and biological functions of Rap2B. Increasing evidence clearly supports the association between Rap2B function and tumor development. Therefore, it is conceivable that anticancer drugs targeting Rap2B may be generated as novel therapies against cancer.
Collapse
Affiliation(s)
- Debao Qu
- Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China; Department of Radiotherapy, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Hui Huang
- Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Jiehui DI
- Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Keyu Gao
- Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Zheng Lu
- Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
39
|
Manne BK, Badolia R, Dangelmaier CA, Kunapuli SP. C-type lectin like receptor 2 (CLEC-2) signals independently of lipid raft microdomains in platelets. Biochem Pharmacol 2014; 93:163-70. [PMID: 25462818 DOI: 10.1016/j.bcp.2014.11.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 11/05/2014] [Accepted: 11/12/2014] [Indexed: 10/24/2022]
Abstract
C-type lectin like receptor 2 (CLEC-2) has been reported to activate platelets through a lipid raft-dependent manner. Secreted ADP potentiates CLEC-2-mediated platelet aggregation. We have investigated whether the decrease in CLEC-2-mediated platelet aggregation, previously reported in platelets with disrupted rafts, is a result of the loss of agonist potentiation by ADP. We disrupted platelet lipid rafts with methyl-β-cyclodextrin (MβCD) and measured signaling events downstream of CLEC-2 activation. Lipid raft disruption decreases platelet aggregation induced by CLEC-2 agonists. The inhibition of platelet aggregation by the disruption of lipid rafts was rescued by the exogenous addition of epinephrine but not 2-methylthioadenosine diphosphate (2MeSADP), which suggests that lipid raft disruption effects P2Y12-mediated Gi activation but not Gz. Phosphorylation of Syk (Y525/526) and PLCγ2 (Y759), were not affected by raft disruption in CLEC-2 agonist-stimulated platelets. Furthermore, tyrosine phosphorylation of the CLEC-2 hemi-ITAM was not effected when MβCD disrupts lipid rafts. Lipid rafts do not directly contribute to CLEC-2 receptor activation in platelets. The effects of disruption of lipid rafts in in vitro assays can be attributed to inhibition of ADP feedback that potentiates CLEC-2 signaling.
Collapse
Affiliation(s)
- Bhanu Kanth Manne
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA; Sol Sherry Thrombosis Research Center and Temple University School of Medicine, Philadelphia, PA, USA
| | - Rachit Badolia
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA; Sol Sherry Thrombosis Research Center and Temple University School of Medicine, Philadelphia, PA, USA
| | - Carol A Dangelmaier
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA; Sol Sherry Thrombosis Research Center and Temple University School of Medicine, Philadelphia, PA, USA
| | - Satya P Kunapuli
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA; Sol Sherry Thrombosis Research Center and Temple University School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
40
|
Moscardó A, Vallés J, Latorre A, Santos M. The association of thromboxane A2
receptor with lipid rafts is a determinant for platelet functional responses. FEBS Lett 2014; 588:3154-9. [DOI: 10.1016/j.febslet.2014.06.057] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 06/24/2014] [Indexed: 10/25/2022]
|
41
|
Clot retraction is mediated by factor XIII-dependent fibrin-αIIbβ3-myosin axis in platelet sphingomyelin-rich membrane rafts. Blood 2013; 122:3340-8. [PMID: 24002447 DOI: 10.1182/blood-2013-04-491290] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Membrane rafts are spatially and functionally heterogenous in the cell membrane. We observed that lysenin-positive sphingomyelin (SM)-rich rafts are identified histochemically in the central region of adhered platelets where fibrin and myosin are colocalized on activation by thrombin. The clot retraction of SM-depleted platelets from SM synthase knockout mouse was delayed significantly, suggesting that platelet SM-rich rafts are involved in clot retraction. We found that fibrin converted by thrombin translocated immediately in platelet detergent-resistant membrane (DRM) rafts but that from Glanzmann's thrombasthenic platelets failed. The fibrinogen γ-chain C-terminal (residues 144-411) fusion protein translocated to platelet DRM rafts on thrombin activation, but its mutant that was replaced by A398A399 at factor XIII crosslinking sites (Q398Q399) was inhibited. Furthermore, fibrin translocation to DRM rafts was impaired in factor XIII A subunit-deficient mouse platelets, which show impaired clot retraction. In the cytoplasm, myosin translocated concomitantly with fibrin translocation into the DRM raft of thrombin-stimulated platelets. Furthermore, the disruption of SM-rich rafts by methyl-β-cyclodextrin impaired myosin activation and clot retraction. Thus, we propose that clot retraction takes place in SM-rich rafts where a fibrin-αIIbβ3-myosin complex is formed as a primary axis to promote platelet contraction.
Collapse
|
42
|
Shiraishi M, Tamura K, Egoshi M, Miyamoto A. Cholesterol enrichment of rabbit platelets enhances the Ca(2+) entry pathway induced by platelet-derived secondary feedback agonists. Life Sci 2013; 92:838-44. [PMID: 23499558 DOI: 10.1016/j.lfs.2013.02.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 02/18/2013] [Accepted: 02/25/2013] [Indexed: 01/15/2023]
Abstract
AIMS Hypersensitivity of platelets due to increased platelet cholesterol levels has been reported in hypercholesterolemia. However, the signaling pathways linking increased platelet reactivity and cholesterol contents are not fully understood. This study aims to determine the direct effect of cholesterol enrichment of platelets on the pathways including Ca(2+) mobilization and secondary feedback agonists such as adenosine diphosphate (ADP) and thromboxane A2 (TXA2). MAIN METHODS In vitro cholesterol enrichment of rabbit platelets was performed by incubation with cholesterol complexed with methyl-β-cyclodextrin. Ca(2+) mobilization was monitored using platelets loaded with fura-PE3/AM, a fluorescent calcium indicator. Released ATP and TXB2 from platelets were measured by a luciferin-luciferase ATP assay system and a TXB2 ELISA Kit, respectively. KEY FINDINGS Cholesterol enrichment of rabbit platelets significantly enhanced Ca(2+) mobilization induced by thrombin, accompanying an augmented Ca(2+) entry. The augmentation of Ca(2+) entry by cholesterol enrichment was significantly suppressed by treatment with inhibitors for secondary feedback agonists. In cholesterol-enriched platelets, the amount of released ATP or TXB2 induced by thrombin was not significantly altered in comparison with control platelets, whereas an increase in [Ca(2+)]i induced by ADP or U46619, a TXA2 mimetic, was significantly enhanced. SIGNIFICANCE These results suggest that cholesterol enrichment of rabbit platelets results in enhanced Ca(2+) mobilization via ADP/TXA2-dependent augmentation of the Ca(2+) entry pathway. The results reveal a novel mechanism by which platelet hypersensitivity is regulated by cholesterol contents.
Collapse
Affiliation(s)
- Mitsuya Shiraishi
- Department of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | | | | | | |
Collapse
|
43
|
Goggs R, Poole AW. Platelet signaling-a primer. J Vet Emerg Crit Care (San Antonio) 2012; 22:5-29. [PMID: 22316389 DOI: 10.1111/j.1476-4431.2011.00704.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 11/25/2011] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To review the receptors and signal transduction pathways involved in platelet plug formation and to highlight links between platelets, leukocytes, endothelium, and the coagulation system. DATA SOURCES Original studies, review articles, and book chapters in the human and veterinary medical fields. DATA SYNTHESIS Platelets express numerous surface receptors. Critical among these are glycoprotein VI, the glycoprotein Ib-IX-V complex, integrin α(IIb) β(3) , and the G-protein-coupled receptors for thrombin, ADP, and thromboxane. Activation of these receptors leads to various important functional events, in particular activation of the principal adhesion receptor α(IIb) β(3) . Integrin activation allows binding of ligands such as fibrinogen, mediating platelet-platelet interaction in the process of aggregation. Signals activated by these receptors also couple to 3 other important functional events, secretion of granule contents, change in cell shape through cytoskeletal rearrangement, and procoagulant membrane expression. These processes generate a stable thrombus to limit blood loss and promote restoration of endothelial integrity. CONCLUSIONS Improvements in our understanding of how platelets operate through their signaling networks are critical for diagnosis of unusual primary hemostatic disorders and for rational antithrombotic drug design.
Collapse
Affiliation(s)
- Robert Goggs
- School of Physiology and Pharmacology, Faculty of Medical and Veterinary Sciences, University of Bristol, UK.
| | | |
Collapse
|
44
|
Peramo A, Diaz JA. Physical characterization of mouse deep vein thrombosis derived microparticles by differential filtration with nanopore filters. MEMBRANES 2011; 2:1-15. [PMID: 24957960 PMCID: PMC4021882 DOI: 10.3390/membranes2010001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 12/16/2011] [Accepted: 12/22/2011] [Indexed: 11/16/2022]
Abstract
With the objective of making advancements in the area of pro-thrombotic microparticle characterization in cardiovascular biology, we present a novel method to separate blood circulating microparticles using a membrane-based, nanopore filtration system. In this qualitative study, electron microscopy observations of these pro-thrombotic mouse microparticles, as well as mouse platelets and leukocytes obtained using a mouse inferior vena cava ligation model of deep-vein thrombosis are presented. In particular, we present mouse microparticle morphology and microstructure using SEM and TEM indicating that they appear to be mostly spherical with diameters in the 100 to 350 nm range. The nanopore filtration technique presented is focused on the development of novel methodologies to isolate and characterize blood circulating microparticles that can be used in conjunction with other methodologies. We believe that determination of microparticle size and structure is a critical step for the development of reliable assays with clinical or research application in thrombosis and it will contribute to the field of nanomedicine in thrombosis.
Collapse
Affiliation(s)
- Antonio Peramo
- Department of Oral and Maxillofacial Surgery, 1150 W Medical Drive, MSRBII A560, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Jose A Diaz
- Department of Surgery, Section of Vascular Surgery, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
45
|
Investigation of interaction of human platelet membrane components with anticoagulant drugs Abciximab and Eptifibatide. Folia Histochem Cytobiol 2011; 48:687-92. [PMID: 21478117 DOI: 10.2478/v10042-010-0090-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abciximab (Abci) and eptifibatide (Epti) are antiaggregate drugs which may reduce thrombotic complications in acute coronary syndromes. The aim of this work was the investigation of the interaction between the phospholipid-GPIIb/IIIa glycoprotein complex and Abci or Epti, and the influence of these drugs on the phospholipid ratio in the platelet membrane. The interaction between the phospholipid-GPIIb/IIIa glycoprotein complex and antiaggregate drugs were investigated using the Surface Plasmon Resonance Imaging technique (SPRI). Phospholipids phosphatidylinositol (PI), phosphatidylserine (PS), phosphatidylethanolamine (PE), phosphatidylcholine (PC) and sphingomyelin (SM) were first immobilized onto the gold chip surface. The phospholipid ratio in the platelet membrane was determined by the HPLC. Only PI, PS, PE and PC were determined. Human platelets treated 'in vitro' with Abci or Epti exhibit changes in the phospholipid ratio in the platelet membrane. The ratio of PS decreases and PC rises. The SPRI distinctly shows interactions between phospholipids and glycoprotein GPIIb/IIIa, and between the phospholipid-glycoprotein GPIIb/IIIa complex and Abci or Epti. The interaction between phospholipids and glycoprotein GPIIb/IIIa is growing in the sequence: PI<<SM<PE<PC<PS. The interaction between phospholipid-glycoprotein GPIIb/IIIa complex and Abci/Epti is growing in the sequence: PS<PI<PC<PE<SM. SPRI was proved to be excellent tool for observation of such interactions.
Collapse
|
46
|
Nagy B, Jin J, Ashby B, Reilly MP, Kunapuli SP. Contribution of the P2Y12 receptor-mediated pathway to platelet hyperreactivity in hypercholesterolemia. J Thromb Haemost 2011; 9:810-9. [PMID: 21261805 PMCID: PMC3071452 DOI: 10.1111/j.1538-7836.2011.04217.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND In hypercholesterolemia, platelets demonstrate increased reactivity and promote the development of cardiovascular disease. OBJECTIVE This study was carried out to investigate the contribution of the ADP receptor P2Y12-mediated pathway to platelet hyperreactivity due to hypercholesterolemia. METHODS Low-density lipoprotein receptor-deficient mice and C57Bl/6 wild-type mice were fed on normal chow and high-fat (Western or Paigen) diets for 8 weeks to generate differently elevated cholesterol levels. P2Y12 receptor-induced functional responses via G(i) signaling were studied ex vivo when washed murine platelets were activated by 2MeSADP and PAR4 agonist AYPGKF in the presence and absence of indomethacin. Platelet aggregation and secretion, α(IIb)β(3) receptor activation and the phosphorylation of extracellular signal-regulated protein kinase (ERK) and Akt were analyzed. RESULTS Plasma cholesterol levels ranged from 69 ± 10 to 1011 ± 185 mg dL(-1) depending on diet in mice with different genotypes. Agonist-dependent aggregation, dense and α-granule secretion and JON/A binding were gradually and significantly (P < 0.05) augmented at low agonist concentration in correlation with the increasing plasma cholesterol levels, even if elevated thromboxane generation was blocked. These functional responses were induced via increased levels of G(i) -mediated ERK and Akt phosphorylation in hypercholesterolemic mice vs. normocholesterolemic animals. In addition, blocking of the P2Y12 receptor by AR-C69931MX (Cangrelor) resulted in strongly reduced platelet aggregation in mice with elevated cholesterol levels compared with normocholesterolemic controls. CONCLUSIONS These data revealed that the P2Y12 receptor pathway was substantially involved in platelet hyperreactivity associated with mild and severe hypercholesterolemia.
Collapse
Affiliation(s)
- Béla Nagy
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA
| | - Jianguo Jin
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA, USA
| | - Barrie Ashby
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, USA
| | - Michael P. Reilly
- Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA, USA
| | - Satya P. Kunapuli
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, USA
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
47
|
|
48
|
Korporaal SJA, Meurs I, Hauer AD, Hildebrand RB, Hoekstra M, Cate HT, Praticò D, Akkerman JWN, Van Berkel TJC, Kuiper J, Van Eck M. Deletion of the high-density lipoprotein receptor scavenger receptor BI in mice modulates thrombosis susceptibility and indirectly affects platelet function by elevation of plasma free cholesterol. Arterioscler Thromb Vasc Biol 2010; 31:34-42. [PMID: 21051668 DOI: 10.1161/atvbaha.110.210252] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Scavenger receptor BI (SR-BI) is a cell surface receptor that promotes the selective uptake of cholesteryl esters from high-density lipoprotein (HDL) by the liver. In mice, SR-BI deficiency results in increased plasma HDL cholesterol levels and enhanced susceptibility to atherosclerosis. The aim of this study was to investigate the role of SR-BI deficiency on platelet function. METHODS AND RESULTS SR-BI-deficient mice were thrombocytopenic, and their platelets were abnormally large, probably because of an increased cholesterol content. The FeCl(3) acute injury model to study arterial thrombosis susceptibility showed that SR-BI wild-type mice developed total arterial occlusion after 24±2 minutes. In SR-BI-deficient mice, however, the time to occlusion was reduced to 13±1 minutes (P=0.02). Correspondingly, in SR-BI-deficient mice, platelets circulated in an activated state and showed increased adherence to immobilized fibrinogen. In contrast, platelet-specific disruption of SR-BI by bone marrow transplantation in wild-type mice did not alter plasma cholesterol levels or affect platelet count, size, cholesterol content, or reactivity, suggesting that changes in plasma cholesterol levels were responsible for the altered responsiveness of platelets in SR-BI-deficient mice. CONCLUSIONS The function of SR-BI in HDL cholesterol homeostasis and prevention of atherosclerosis is indirectly also essential for maintaining normal platelet function and prevention of thrombosis.
Collapse
Affiliation(s)
- Suzanne J A Korporaal
- Leiden/Amsterdam Center for Drug Research, Division of Biopharmaceutics, Gorlaeus Laboratories, Einsteinweg 55, 2333 CC, Leiden, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Ruebsaamen K, Liebisch G, Boettcher A, Schmitz G. Lipidomic analysis of platelet senescence. Transfusion 2010; 50:1665-76. [DOI: 10.1111/j.1537-2995.2010.02584.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
50
|
Horstman LL, Jy W, Ahn YS, Zivadinov R, Maghzi AH, Etemadifar M, Steven Alexander J, Minagar A. Role of platelets in neuroinflammation: a wide-angle perspective. J Neuroinflammation 2010; 7:10. [PMID: 20128908 PMCID: PMC2829540 DOI: 10.1186/1742-2094-7-10] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Accepted: 02/03/2010] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVES This review summarizes recent developments in platelet biology relevant to neuroinflammatory disorders. Multiple sclerosis (MS) is taken as the "Poster Child" of these disorders but the implications are wide. The role of platelets in inflammation is well appreciated in the cardiovascular and cancer research communities but appears to be relatively neglected in neurological research. ORGANIZATION After a brief introduction to platelets, topics covered include the matrix metalloproteinases, platelet chemokines, cytokines and growth factors, the recent finding of platelet PPAR receptors and Toll-like receptors, complement, bioactive lipids, and other agents/functions likely to be relevant in neuroinflammatory diseases. Each section cites literature linking the topic to areas of active research in MS or other disorders, including especially Alzheimer's disease. CONCLUSION The final section summarizes evidence of platelet involvement in MS. The general conclusion is that platelets may be key players in MS and related disorders, and warrant more attention in neurological research.
Collapse
Affiliation(s)
- Lawrence L Horstman
- Wallace Coulter Platelet Laboratory, Division of Hematology and Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Wenche Jy
- Wallace Coulter Platelet Laboratory, Division of Hematology and Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Yeon S Ahn
- Wallace Coulter Platelet Laboratory, Division of Hematology and Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, The Jacobs Neurological Institute, Department of Neurology, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo NY, USA
| | - Amir H Maghzi
- Department of Neurology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Masoud Etemadifar
- Department of Neurology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - J Steven Alexander
- Department of Cellular and Molecular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Alireza Minagar
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| |
Collapse
|