1
|
Hackenbroch C, Cimen T, Gross C, Rubenzucker S, Burkard P, Groß N, Ahrends R, Girbl T, Stegner D. Neutrophil recruitment depends on platelet-derived leukotriene B4. Blood Adv 2025; 9:2226-2230. [PMID: 40048739 DOI: 10.1182/bloodadvances.2024014947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/15/2025] [Indexed: 04/30/2025] Open
Affiliation(s)
- Christian Hackenbroch
- Julius-Maximilians University of Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany
- University Hospital Würzburg, Institute of Experimental Biomedicine I, Würzburg, Germany
| | - Tugce Cimen
- Julius-Maximilians University of Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany
| | - Carina Gross
- Julius-Maximilians University of Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany
- University Hospital Würzburg, Institute of Experimental Biomedicine I, Würzburg, Germany
| | - Stefanie Rubenzucker
- Department of Analytical Chemistry, University of Vienna, Vienna, Austria
- Vienna Doctoral School in Chemistry, University of Vienna, Vienna, Austria
| | - Philipp Burkard
- Julius-Maximilians University of Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany
- University Hospital Würzburg, Institute of Experimental Biomedicine I, Würzburg, Germany
| | - Niklas Groß
- Julius-Maximilians University of Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany
- University Hospital Würzburg, Institute of Experimental Biomedicine I, Würzburg, Germany
| | - Robert Ahrends
- Department of Analytical Chemistry, University of Vienna, Vienna, Austria
| | - Tamara Girbl
- Julius-Maximilians University of Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany
| | - David Stegner
- Julius-Maximilians University of Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany
- University Hospital Würzburg, Institute of Experimental Biomedicine I, Würzburg, Germany
| |
Collapse
|
2
|
Chen Z, Xu L, Yuan Y, Zhang S, Xue R. Metabolic crosstalk between platelets and cancer: Mechanisms, functions, and therapeutic potential. Semin Cancer Biol 2025; 110:65-82. [PMID: 39954752 DOI: 10.1016/j.semcancer.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
Platelets, traditionally regarded as passive mediators of hemostasis, are now recognized as pivotal regulators in the tumor microenvironment, establishing metabolic feedback loops with tumor and immune cells. Tumor-derived signals trigger platelet activation, which induces rapid metabolic reprogramming, particularly glycolysis, to support activation-dependent functions such as granule secretion, morphological changes, and aggregation. Beyond self-regulation, platelets influence the metabolic processes of adjacent cells. Through direct mitochondrial transfer, platelets reprogram tumor and immune cells, promoting oxidative phosphorylation. Additionally, platelet-derived cytokines, granules, and extracellular vesicles drive metabolic alterations in immune cells, fostering suppressive phenotypes that facilitate tumor progression. This review examines three critical aspects: (1) the distinctive metabolic features of platelets, particularly under tumor-induced activation; (2) the metabolic crosstalk between activated platelets and other cellular components; and (3) the therapeutic potential of targeting platelet metabolism to disrupt tumor-promoting networks. By elucidating platelet metabolism, this review highlights its essential role in tumor biology and its therapeutic implications.
Collapse
Affiliation(s)
- Zhixue Chen
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lin Xu
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yejv Yuan
- The First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232001, China
| | - Si Zhang
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Ruyi Xue
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
3
|
Loroch S, Panagiotidis E, Klewe K, Swieringa F, Heemskerk JWM, Lerch JP, Greinacher A, Walter U, Jurk K, John T, Barkovits K, Dandekar T, Marcus K, Balkenhol J. Middle-throughput LC-MS-based Platelet Proteomics with Minute Sample Amounts Using Semiautomated Positive Pressure FASP in 384-Well Format. Thromb Haemost 2025. [PMID: 39814053 DOI: 10.1055/a-2516-1812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Comprehensive characterization of platelets requires various functional assays and analytical techniques, including omics disciplines, each demanding a separate aliquot of the given sample. Consequently, sample material for each assay is often highly limited, necessitating the downscaling of methods to work with just a few micrograms of platelet protein.Here, we present a novel sample preparation platform for proteomics analysis using only 3 μg of purified platelet protein, corresponding to 2 × 106 platelets, which can be obtained from approximately 2 to 8 μL of blood from a healthy individual (1.5 × 105-4.5 × 105 platelets/μL) or approximately 100 μL of blood from a patient with severe thrombocytopenia (<2 × 104 platelets/µL).Using this platform, we detected a significant fraction of key players in the platelet activation cascade and, most importantly, identified 36 clinically relevant platelet disease markers even with a non-state-of-the art instrument. This makes LC-MS-based proteomics a highly attractive alternative to conventional assays, which often require milliliters of blood. Our platform transitions from our previously established 96-well proteomics workflow (PF96), which has been successfully employed in numerous platelet proteomics studies, into the 384-well format. This transition is accompanied by (1) a more than two-fold increase in sensitivity, (2) improved reproducibility, (3) a four-fold increase in throughput, allowing 1,536 samples to be processed per lab worker per week, and (4) reduced sample preparation costs.Thus, LC-MS-based platelet proteomics offers a compelling alternative to immunoaffinity assays (which depend on antibody availability and quality), as well as to genomic assays (which can only reveal genotypes). In summary, in conjunction with recent advances in LC-MS instrumentation, our platform represents a highly valuable tool for rapid phenotyping of platelets in research with extraordinary potential for future employment in companion or routine diagnostics.
Collapse
Affiliation(s)
- Stefan Loroch
- Medical Proteome-Center (MPC), Medical Faculty, Ruhr-University Bochum, Bochum, Germany
- Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Fa. Dr. Loroch/QC|MS, Biomedizin Zentrum, Dortmund, Dortmund, Germany
- Department of Bioanalytics, Leibniz-Institute for Analytical Sciences - ISAS - e.V., Dortmund Germany
| | - Eleftherios Panagiotidis
- Department of Bioanalytics, Leibniz-Institute for Analytical Sciences - ISAS - e.V., Dortmund Germany
| | - Kristoffer Klewe
- Fa. Dr. Loroch/QC|MS, Biomedizin Zentrum, Dortmund, Dortmund, Germany
| | - Frauke Swieringa
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Synapse Research Institute, Platelet Pathophysiology, Maastricht, the Netherlands
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Synapse Research Institute, Platelet Pathophysiology, Maastricht, the Netherlands
| | - Jan-Paul Lerch
- Fakultät für Mathematik, Universität Bielefeld, Bielefeld, Germany
| | - Andreas Greinacher
- Institut für Immunologie und Transfusionsmedizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Ulrich Walter
- Center for Thrombosis and Hemostasis (CTH), Univeristy Medical Center Mainz, Mainz, Germany
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis (CTH), Univeristy Medical Center Mainz, Mainz, Germany
| | - Tobias John
- Medical Proteome-Center (MPC), Medical Faculty, Ruhr-University Bochum, Bochum, Germany
- Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Katalin Barkovits
- Medical Proteome-Center (MPC), Medical Faculty, Ruhr-University Bochum, Bochum, Germany
- Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Thomas Dandekar
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Katrin Marcus
- Medical Proteome-Center (MPC), Medical Faculty, Ruhr-University Bochum, Bochum, Germany
- Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Johannes Balkenhol
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| |
Collapse
|
4
|
Benkhoff M, Barcik M, Mourikis P, Dahlmanns J, Kahmann P, Wollnitzke P, Hering M, Huckenbeck T, Hoppe J, Semleit N, Deister-Jonas J, Zako S, Seel J, Coman C, Barth M, Cramer M, Helten C, Wildeis L, Hu H, Al-Kassis G, Metzen D, Hesse J, Weber J, Dannenberg L, Akhyari P, Lichtenberg A, Quast C, Gerdes N, Zeus T, Borst O, Kelm M, Petzold T, Ahrends R, Levkau B, Polzin A. Targeting Sphingosine-1-Phosphate Signaling to Prevent the Progression of Aortic Valve Disease. Circulation 2025; 151:333-347. [PMID: 39429140 DOI: 10.1161/circulationaha.123.067270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 10/01/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND Aortic valve disease (AVD) is associated with high mortality and morbidity. To date, there is no pharmacological therapy available to prevent AVD progression. Because valve calcification is the hallmark of AVD and S1P (sphingosine-1-phosphate) plays an important role in osteogenic signaling, we examined the role of S1P signaling in aortic stenosis disease. METHODS AVD progression and its consequences for cardiac function were examined in a murine wire injury-induced AVD model with and without pharmacological and genetic modulation of S1P production, degradation, and receptor signaling. S1P was measured by liquid chromatography-mass spectrometry. Calcification of human valvular interstitial cells and their response to biomechanical stress were analyzed in the context of S1P signaling. Human explanted aortic valves from patients undergoing aortic valve replacement and cardiovascular magnetic resonance imaging were analyzed for S1P by liquid chromatography-mass spectrometry. RESULTS Raising S1P concentrations in mice with injury-induced AVD by pharmacological inhibition of its sole degrading enzyme S1P lyase vastly enhanced AVD progression and impaired cardiac function resembling human disease. In contrast, low S1P levels caused by SphK1 (sphingosine kinase 1) deficiency potently attenuated AVD progression. We found S1P/S1PR2 (S1P receptor 2) signaling to be responsible for the adverse S1P effect because S1PR2-deficient mice were protected against AVD progression and its deterioration by high S1P. It is important to note that pharmacological S1PR2 inhibition administered after wire injury successfully prevented AVD development. Mechanistically, biomechanical stretch stimulated S1P production by SphK1 in human valvular interstitial cells as measured by C17-S1P generation, whereas S1P/S1PR2 signaling induced their osteoblastic differentiation and calcification through osteogenic RUNX2/OPG signaling and the GSK3β-Wnt-β-catenin pathway. In patients with AVD, stenotic valves exposed to high wall shear stress had higher S1P content and increased SphK1 expression. CONCLUSIONS Increased systemic or local S1P levels lead to increased valvular calcification. S1PR2 antagonists and SphK1 inhibitors may offer feasible pharmacological approaches to human AVD in prophylactic, disease-modifying or relapse-preventing manners.
Collapse
Affiliation(s)
- Marcel Benkhoff
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
- Institute of Analytical Chemistry, University of Vienna, Vienna, Austria (M. Benkhoff, C.C., R.A.)
| | - Maike Barcik
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
| | - Philipp Mourikis
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
| | - Jana Dahlmanns
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
| | - Paulina Kahmann
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
| | - Philipp Wollnitzke
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany (P.W., J.H., N.S., J.D.-J., B.L.)
| | - Moritz Hering
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
| | - Tim Huckenbeck
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
| | - Julia Hoppe
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany (P.W., J.H., N.S., J.D.-J., B.L.)
| | - Nina Semleit
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany (P.W., J.H., N.S., J.D.-J., B.L.)
| | - Jennifer Deister-Jonas
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany (P.W., J.H., N.S., J.D.-J., B.L.)
| | - Saif Zako
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
| | - Jasmin Seel
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
| | - Cristina Coman
- Institute of Analytical Chemistry, University of Vienna, Vienna, Austria (M. Benkhoff, C.C., R.A.)
| | - Mareike Barth
- Department of Cardiac Surgery, University Hospital Aachen, RWTH Aachen University, Germany (M. Barth, P.A.)
| | - Mareike Cramer
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
| | - Carolin Helten
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
| | - Laura Wildeis
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
| | - Hao Hu
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
| | - Gabrielle Al-Kassis
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
| | - Daniel Metzen
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
| | - Julia Hesse
- Department of Molecular Cardiology, Heinrich Heine University Düsseldorf, Germany (J.H.)
| | - Jessica Weber
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
| | - Lisa Dannenberg
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
| | - Payam Akhyari
- Department of Cardiac Surgery, University Hospital Aachen, RWTH Aachen University, Germany (M. Barth, P.A.)
| | - Artur Lichtenberg
- Department of Cardiac Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany (A.L.)
| | - Christine Quast
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
| | - Norbert Gerdes
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany (N.G., M.K., A.P.)
| | - Tobias Zeus
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
| | - Oliver Borst
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Tübingen, Germany (O.B.)
- Department of Cardiology and Angiology, University of Tübingen, Tübingen, Germany (O.B.)
| | - Malte Kelm
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany (N.G., M.K., A.P.)
| | - Tobias Petzold
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany (T.P.)
- DZHK (German Centre for Cardiovascular Research), Partner site Berlin, Germany (T.P.)
| | - Robert Ahrends
- Institute of Analytical Chemistry, University of Vienna, Vienna, Austria (M. Benkhoff, C.C., R.A.)
| | - Bodo Levkau
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany (P.W., J.H., N.S., J.D.-J., B.L.)
| | - Amin Polzin
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany (M. Benkhoff, M. Barcik, P.M., J.D., P.K., M.H., T.H., S.Z., J.S., M.C., C.H., L.W., H.H., G.A.-K., D.M., J.W., L.D., C.Q., N.G., T.Z., M.K., A.P.)
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany (N.G., M.K., A.P.)
- National Heart and Lung Institute, Imperial College London, London, United Kingdom (A.P.)
| |
Collapse
|
5
|
Wittenhofer P, Kiesewetter L, Schmitz OJ, Meckelmann SW. Investigation of the Cholesterol Biosynthesis by Heart-Cut Liquid Chromatography and Mass Spectrometric Detection. J Chromatogr A 2024; 1738:465475. [PMID: 39488880 DOI: 10.1016/j.chroma.2024.465475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
The biosynthesis and homeostasis of cholesterol are essential for cellular function. Cholesterol is a major lipid with multiple roles in membrane stability, signaling, or as a precursor for other molecules. Because of the structural similarity of the sterols involved in the biosynthesis, their accurate identification and quantification is still challenging. Moreover, the huge difference in the concentration of cholesterol and its precursors can cause interferences during the detection. To overcome these problems, a heart-cut liquid chromatographic method was developed by evaluating 38 different columns to achieve optimal separation. The method efficiently separates all sterol biosynthesis intermediates, with detection limits in the low nmol/L-range and an upper limit of quantification of 9 mmol/L for cholesterol by using triple quadrupole mass spectrometric detection. Investigation of lung carcinoma cells treated with statins demonstrated the capability to detect a biological response, showing inhibition of sterol synthesis. This technique offers a robust tool for studying cholesterol biosynthesis and its role in disease.
Collapse
Affiliation(s)
- Pia Wittenhofer
- Applied Analytical Chemistry, University of Duisburg-Essen, Universitaetsstrasse 5, 45141 Essen, Germany
| | - Laura Kiesewetter
- Applied Analytical Chemistry, University of Duisburg-Essen, Universitaetsstrasse 5, 45141 Essen, Germany
| | - Oliver J Schmitz
- Applied Analytical Chemistry, University of Duisburg-Essen, Universitaetsstrasse 5, 45141 Essen, Germany
| | - Sven W Meckelmann
- Applied Analytical Chemistry, University of Duisburg-Essen, Universitaetsstrasse 5, 45141 Essen, Germany.
| |
Collapse
|
6
|
Pirotton L, de Cartier d’Yves E, Bertrand L, Beauloye C, Horman S. Platelet lipidomics and de novo lipogenesis: impact on health and disease. Curr Opin Hematol 2024; 31:217-223. [PMID: 38727017 PMCID: PMC11296274 DOI: 10.1097/moh.0000000000000820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
PURPOSE OF REVIEW Lipids play vital roles in platelet structure, signaling, and metabolism. In addition to capturing exogenous lipids, platelets possess the capacity for de novo lipogenesis, regulated by acetyl-coA carboxylase 1 (ACC1). This review aims to cover the critical roles of platelet de novo lipogenesis and lipidome in platelet production, function, and diseases. RECENT FINDINGS Upon platelet activation, approximately 20% of the platelet lipidome undergoes significant modifications, primarily affecting arachidonic acid-containing species. Multiple studies emphasize the impact of de novo lipogenesis, with ACC1 as key player, on platelet functions. Mouse models suggest the importance of the AMPK-ACC1 axis in regulating platelet membrane arachidonic acid content, associated with TXA 2 secretion, and thrombus formation. In human platelets, ACC1 inhibition leads to reduced platelet reactivity. Remodeling of the platelet lipidome, alongside with de novo lipogenesis, is also crucial for platelet biogenesis. Disruptions in the platelet lipidome are observed in various pathological conditions, including cardiovascular and inflammatory diseases, with associations between these alterations and shifts in platelet reactivity highlighted. SUMMARY The platelet lipidome, partially regulated by ACC-driven de novo lipogenesis, is indispensable for platelet production and function. It is implicated in various pathological conditions involving platelets.
Collapse
Affiliation(s)
- Laurence Pirotton
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain)
| | - Emma de Cartier d’Yves
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain)
| | - Luc Bertrand
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain)
| | - Christophe Beauloye
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain)
- Department of Cardiovascular Intensive Care, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Sandrine Horman
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain)
| |
Collapse
|
7
|
Affiliation(s)
- Julie B Larsen
- Thrombosis and Haemostasis Research Unit, Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
8
|
Rubenzucker S, Manke MC, Lehmann R, Assinger A, Borst O, Ahrends R. A Targeted, Bioinert LC-MS/MS Method for Sensitive, Comprehensive Analysis of Signaling Lipids. Anal Chem 2024; 96:9643-9652. [PMID: 38795073 PMCID: PMC11170558 DOI: 10.1021/acs.analchem.4c01388] [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] [Received: 03/14/2024] [Revised: 05/08/2024] [Accepted: 05/15/2024] [Indexed: 05/27/2024]
Abstract
Signaling lipids are key players in cellular processes. Despite their importance, no method currently allows their comprehensive monitoring in one analytical run. Challenges include a wide dynamic range, isomeric and isobaric species, and unwanted interaction along the separation path. Herein, we present a sensitive and robust targeted liquid chromatography-mass spectrometry (LC-MS/MS) approach to overcome these challenges, covering a broad panel of 17 different signaling lipid classes. It involves a simple one-phase sample extraction and lipid analysis using bioinert reversed-phase liquid chromatography coupled to targeted mass spectrometry. The workflow shows excellent sensitivity and repeatability in different biological matrices, enabling the sensitive and robust monitoring of 388 lipids in a single run of only 20 min. To benchmark our workflow, we characterized the human plasma signaling lipidome, quantifying 307 endogenous molecular lipid species. Furthermore, we investigated the signaling lipidome during platelet activation, identifying numerous regulations along important lipid signaling pathways. This highlights the potential of the presented method to investigate signaling lipids in complex biological systems, enabling unprecedentedly comprehensive analysis and direct insight into signaling pathways.
Collapse
Affiliation(s)
- Stefanie Rubenzucker
- Department
of Analytical Chemistry, University of Vienna, 1090 Vienna, Austria
- Vienna
Doctoral School in Chemistry, University
of Vienna, 1090 Vienna, Austria
| | - Mailin-Christin Manke
- DFG
Heisenberg Group Cardiovascular Thromboinflammation and Translational
Thrombocardiology, University of Tübingen, 72076 Tübingen, Germany
- Department
of Cardiology and Angiology, University
of Tübingen, 72076 Tübingen, Germany
| | - Rainer Lehmann
- Institute
for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic
Laboratory Medicine, University Hospital
Tübingen, 72076 Tübingen, Germany
| | - Alice Assinger
- Department
of Vascular Biology and Thrombosis Research, Centre of Physiology
and Pharmacology, Medical University of
Vienna, 1090 Vienna, Austria
| | - Oliver Borst
- DFG
Heisenberg Group Cardiovascular Thromboinflammation and Translational
Thrombocardiology, University of Tübingen, 72076 Tübingen, Germany
- Department
of Cardiology and Angiology, University
of Tübingen, 72076 Tübingen, Germany
| | - Robert Ahrends
- Department
of Analytical Chemistry, University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
9
|
Yang W, Feng R, Peng G, Wang Z, Cen M, Jing Y, Feng W, Long T, Liu Y, Li Z, Huang K, Chang G. Glycoursodeoxycholic Acid Alleviates Arterial Thrombosis via Suppressing Diacylglycerol Kinases Activity in Platelet. Arterioscler Thromb Vasc Biol 2024; 44:1283-1301. [PMID: 38572646 DOI: 10.1161/atvbaha.124.320728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Glycoursodeoxycholic acid (GUDCA) has been acknowledged for its ability to regulate lipid homeostasis and provide benefits for various metabolic disorders. However, the impact of GUDCA on arterial thrombotic events remains unexplored. The objective of this study is to examine the effects of GUDCA on thrombogenesis and elucidate its underlying mechanisms. METHODS Plasma samples from patients with arterial thrombotic events and diet-induced obese mice were collected to determine the GUDCA concentrations using mass spectrometry. Multiple in vivo murine thrombosis models and in vitro platelet functional assays were conducted to comprehensively evaluate the antithrombotic effects of GUDCA. Moreover, lipidomic analysis was performed to identify the alterations of intraplatelet lipid components following GUDCA treatment. RESULTS Plasma GUDCA level was significantly decreased in patients with arterial thrombotic events and negatively correlated with thrombotic propensity in diet-induced obese mice. GUDCA exhibited prominent suppressing effects on platelet reactivity as evidenced by the attenuation of platelet activation, secretion, aggregation, spreading, and retraction (P<0.05). In vivo, GUDCA administration robustly alleviated thrombogenesis (P<0.05) without affecting hemostasis. Mechanistically, GUDCA inhibited DGK (diacylglycerol kinase) activity, leading to the downregulation of the phosphatidic acid-mediated signaling pathway. Conversely, phosphatidic acid supplementation was sufficient to abolish the antithrombotic effects of GUDCA. More importantly, long-term oral administration of GUDCA normalized the enhanced DGK activity, thereby remarkably alleviating the platelet hyperreactivity as well as the heightened thrombotic tendency in diet-induced obese mice (P<0.05). CONCLUSIONS Our study implicated that GUDCA reduces platelet hyperreactivity and improves thrombotic propensity by inhibiting DGKs activity, which is a potentially effective prophylactic approach and promising therapeutic agent for arterial thrombotic events.
Collapse
Affiliation(s)
- Wenchao Yang
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Ruijia Feng
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Guiyan Peng
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Zhecun Wang
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Meifeng Cen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, China (M.C.)
| | - Yexiang Jing
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Weiqi Feng
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Ting Long
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Yunchong Liu
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Zilun Li
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Kan Huang
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Guangqi Chang
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| |
Collapse
|
10
|
Chouchane O, Schuurman AR, Reijnders TDY, Peters-Sengers H, Butler JM, Uhel F, Schultz MJ, Bonten MJ, Cremer OL, Calfee CS, Matthay MA, Langley RJ, Alipanah-Lechner N, Kingsmore SF, Rogers A, van Weeghel M, Vaz FM, van der Poll T. The Plasma Lipidomic Landscape in Patients with Sepsis due to Community-acquired Pneumonia. Am J Respir Crit Care Med 2024; 209:973-986. [PMID: 38240721 PMCID: PMC12039242 DOI: 10.1164/rccm.202308-1321oc] [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: 08/01/2023] [Accepted: 01/18/2024] [Indexed: 04/16/2024] Open
Abstract
Rationale: The plasma lipidome has the potential to reflect many facets of the host status during severe infection. Previous work is limited to specific lipid groups or was focused on lipids as prognosticators.Objectives: To map the plasma lipidome during sepsis due to community-acquired pneumonia (CAP) and determine the disease specificity and associations with clinical features.Methods: We analyzed 1,833 lipid species across 33 classes in 169 patients admitted to the ICU with sepsis due to CAP, 51 noninfected ICU patients, and 48 outpatient controls. In a paired analysis, we reanalyzed patients still in the ICU 4 days after admission (n = 82).Measurements and Main Results: A total of 58% of plasma lipids were significantly lower in patients with CAP-attributable sepsis compared with outpatient controls (6% higher, 36% not different). We found strong lipid class-specific associations with disease severity, validated across two external cohorts, and inflammatory biomarkers, in which triacylglycerols, cholesterol esters, and lysophospholipids exhibited the strongest associations. A total of 36% of lipids increased over time, and stratification by survival revealed diverging lipid recovery, which was confirmed in an external cohort; specifically, a 10% increase in cholesterol ester levels was related to a lower odds ratio (0.84; P = 0.006) for 30-day mortality (absolute mortality, 18 of 82). Comparison with noninfected ICU patients delineated a substantial common illness response (57.5%) and a distinct lipidomic signal for patients with CAP-attributable sepsis (37%).Conclusions: Patients with sepsis due to CAP exhibit a time-dependent and partially disease-specific shift in their plasma lipidome that correlates with disease severity and systemic inflammation and is associated with higher mortality.
Collapse
Affiliation(s)
| | | | | | | | | | - Fabrice Uhel
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche S1151, Centre National de la Recherche Scientifique Unité Mixte de Recherche S8253, Institut Necker-Enfants Malades, Université Paris Cité, Paris, France
- Médecine Intensive Réanimation, Assistance Publique-Hôpitaux de Paris, Hôpital Louis Mourier, DMU ESPRIT, Colombes, France
| | - Marcus J Schultz
- Department of Intensive Care Medicine
- Laboratory of Experimental Intensive Care and Anesthesiology
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Marc J Bonten
- Department of Medical Microbiology
- Julius Center for Health Sciences and Primary Care, and
| | - Olaf L Cremer
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Carolyn S Calfee
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Michael A Matthay
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Raymond J Langley
- Department of Pharmacology, University of South Alabama College of Medicine, Mobile, Alabama
| | | | - Stephen F Kingsmore
- Rady Children's Institute for Genomic Medicine, Rady Children's Hospital, San Diego, California
| | - Angela Rogers
- Division of Pulmonary and Critical Care, Department of Medicine, Stanford, California; and
| | - Michel van Weeghel
- Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Emma Children's Hospital
- Core Facility Metabolomics, and
- Inborn Errors of Metabolism Program, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers-Location Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Frédéric M Vaz
- Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Emma Children's Hospital
- Core Facility Metabolomics, and
- Inborn Errors of Metabolism Program, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers-Location Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine
- Division of Infectious Diseases
| |
Collapse
|
11
|
Harm T, Fu X, Frey M, Dittrich K, Brun A, Castor T, Borst O, Müller KAL, Geisler T, Rath D, Lämmerhofer M, Gawaz MP. Machine learning insights into thrombo-ischemic risks and bleeding events through platelet lysophospholipids and acylcarnitine species. Sci Rep 2024; 14:6089. [PMID: 38480746 PMCID: PMC10937715 DOI: 10.1038/s41598-024-56304-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
Coronary artery disease (CAD) often leads to adverse events resulting in significant disease burdens. Underlying risk factors often remain inapparent prior to disease incidence and the cardiovascular (CV) risk is not exclusively explained by traditional risk factors. Platelets inherently promote atheroprogression and enhanced platelet functions and distinct platelet lipid species are associated with disease severity in patients with CAD. Lipidomics data were acquired using mass spectrometry and processed alongside clinical data applying machine learning to model estimates of an increased CV risk in a consecutive CAD cohort (n = 595). By training machine learning models on CV risk measurements, stratification of CAD patients resulted in a phenotyping of risk groups. We found that distinct platelet lipids are associated with an increased CV or bleeding risk and independently predict adverse events. Notably, the addition of platelet lipids to conventional risk factors resulted in an increased diagnostic accuracy of patients with adverse CV events. Thus, patients with aberrant platelet lipid signatures and platelet functions are at elevated risk to develop adverse CV events. Machine learning combining platelet lipidome data and common clinical parameters demonstrated an increased diagnostic value in patients with CAD and might improve early risk discrimination and classification for CV events.
Collapse
Affiliation(s)
- Tobias Harm
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Xiaoqing Fu
- Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | - Moritz Frey
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Kristina Dittrich
- Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | - Adrian Brun
- Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | - Tatsiana Castor
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Karin Anne Lydia Müller
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Tobias Geisler
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Dominik Rath
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany
| | - Michael Lämmerhofer
- Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | - Meinrad Paul Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076, Tübingen, Germany.
| |
Collapse
|
12
|
Hajeyah AA, Protty MB, Paul D, Costa D, Omidvar N, Morgan B, Iwasaki Y, McGill B, Jenkins PV, Yousef Z, Allen-Redpath K, Soyama S, Choudhury A, Mitra R, Yaqoob P, Morrissey JH, Collins PW, O'Donnell VB. Phosphatidylthreonine is a procoagulant lipid detected in human blood and elevated in coronary artery disease. J Lipid Res 2024; 65:100484. [PMID: 38103786 PMCID: PMC10809103 DOI: 10.1016/j.jlr.2023.100484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023] Open
Abstract
Aminophospholipids (aPL) such as phosphatidylserine are essential for supporting the activity of coagulation factors, circulating platelets, and blood cells. Phosphatidylthreonine (PT) is an aminophospholipid previously reported in eukaryotic parasites and animal cell cultures, but not yet in human tissues. Here, we evaluated whether PT is present in blood cells and characterized its ability to support coagulation. Several PT molecular species were detected in human blood, washed platelets, extracellular vesicles, and isolated leukocytes from healthy volunteers using liquid chromatography-tandem mass spectrometry. The ability of PT to support coagulation was demonstrated in vitro using biochemical and biophysical assays. In liposomes, PT supported prothrombinase activity in the presence and absence of phosphatidylserine. PT nanodiscs strongly bound FVa and lactadherin (nM affinity) but poorly bound prothrombin and FX, suggesting that PT supports prothrombinase through recruitment of FVa. PT liposomes bearing tissue factor poorly generated thrombin in platelet poor plasma, indicating that PT poorly supports extrinsic tenase activity. On platelet activation, PT is externalized and partially metabolized. Last, PT was significantly higher in platelets and extracellular vesicle from patients with coronary artery disease than in healthy controls. In summary, PT is present in human blood, binds FVa and lactadherin, supports coagulation in vitro through FVa binding, and is elevated in atherosclerotic vascular disease. Our studies reveal a new phospholipid subclass, that contributes to the procoagulant membrane, and may support thrombosis in patients at elevated risk.
Collapse
Affiliation(s)
- Ali A Hajeyah
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom; Department of Biological Sciences, Kuwait University, Safat, Kuwait.
| | - Majd B Protty
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Divyani Paul
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Daniela Costa
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Nader Omidvar
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Bethan Morgan
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Yugo Iwasaki
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Japan
| | - Beth McGill
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | | | - Zaheer Yousef
- University Hospital of Wales, Cardiff, United Kingdom
| | - Keith Allen-Redpath
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Shin Soyama
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | | | - Rito Mitra
- University Hospital of Wales, Cardiff, United Kingdom
| | - Parveen Yaqoob
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - James H Morrissey
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Peter W Collins
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom; University Hospital of Wales, Cardiff, United Kingdom
| | - Valerie B O'Donnell
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom.
| |
Collapse
|
13
|
Chicanne G, Darcourt J, Bertrand-Michel J, Garcia C, Ribes A, Payrastre B. What can we learn from the platelet lipidome? Platelets 2023; 34:2182180. [PMID: 36880158 DOI: 10.1080/09537104.2023.2182180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Besides their proteome, platelets use, in all responses to the environmental cues, a huge and diverse family of hydrophobic and amphipathic small molecules involved in structural, metabolic and signaling functions; the lipids. Studying how platelet lipidome changes modulate platelet function is an old story constantly renewed through the impressive technical advances allowing the discovery of new lipids, functions and metabolic pathways. Technical progress in analytical lipidomic profiling by top-of-the-line approaches such as nuclear magnetic resonance and gas chromatography or liquid chromatography coupled to mass spectrometry enables either large-scale analysis of lipids or targeted lipidomics. With the support of bioinformatics tools and databases, it is now possible to investigate thousands of lipids over a concentration range of several orders of magnitude. The lipidomic landscape of platelets is considered a treasure trove, not only able to expand our knowledge of platelet biology and pathologies but also to bring diagnostic and therapeutic opportunities. The aim of this commentary article is to summarize the advances in the field and to highlight what lipidomics can tell us about platelet biology and pathophysiology.
Collapse
Affiliation(s)
- Gaëtan Chicanne
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France
| | - Jean Darcourt
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France
| | - Justine Bertrand-Michel
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France.,MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
| | - Cédric Garcia
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France.,Laboratory of Haematology, University Hospital of Toulouse, Toulouse, France
| | - Agnès Ribes
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France.,Laboratory of Haematology, University Hospital of Toulouse, Toulouse, France
| | - Bernard Payrastre
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France.,Laboratory of Haematology, University Hospital of Toulouse, Toulouse, France
| |
Collapse
|
14
|
Janker L, Schuster D, Bortel P, Hagn G, Meier-Menches SM, Mohr T, Mader JC, Slany A, Bileck A, Brunmair J, Madl C, Unger L, Hennlich B, Weitmayr B, Del Favero G, Pils D, Pukrop T, Pfisterer N, Feichtenschlager T, Gerner C. Multiomics-empowered Deep Phenotyping of Ulcerative Colitis Identifies Biomarker Signatures Reporting Functional Remission States. J Crohns Colitis 2023; 17:1514-1527. [PMID: 36961872 PMCID: PMC10588787 DOI: 10.1093/ecco-jcc/jjad052] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Indexed: 03/25/2023]
Abstract
INTRODUCTION Ulcerative colitis [UC] is a chronic disease with rising incidence and unclear aetiology. Deep molecular phenotyping by multiomics analyses may provide novel insights into disease processes and characteristic features of remission states. METHODS UC pathomechanisms were assessed by proteome profiling of human tissue specimens, obtained from five distinct colon locations for each of the 12 patients included in the study. Systemic disease-associated alterations were evaluated thanks to a cross-sectional setting of mass spectrometry-based multiomics analyses comprising proteins, metabolites, and eicosanoids of plasma obtained from UC patients during acute episodes and upon remission, in comparison with healthy controls. RESULTS Tissue proteome profiling indicated colitis-associated activation of neutrophils, macrophages, B and T cells, fibroblasts, endothelial cells and platelets, and hypoxic stress, and suggested a general downregulation of mitochondrial proteins accompanying the establishment of apparent wound healing-promoting activities including scar formation. Whereas pro-inflammatory proteins were apparently upregulated by immune cells, the colitis-associated epithelial cells, fibroblasts, endothelial cells, and platelets seemed to predominantly contribute anti-inflammatory and wound healing-promoting proteins. Blood plasma proteomics indicated chronic inflammation and platelet activation, whereas plasma metabolomics identified disease-associated deregulations of gut and gut microbiome-derived metabolites. Upon remission several, but not all, molecular candidate biomarker levels recovered back to normal. CONCLUSION The findings may indicate that microvascular damage and platelet deregulation hardly resolve upon remission, but apparently persist as disease-associated molecular signatures. This study presents local and systemic molecular alterations integrated in a model for UC pathomechanisms, potentially supporting the assessment of disease and remission states in UC patients.
Collapse
Affiliation(s)
- Lukas Janker
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Dina Schuster
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Patricia Bortel
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Gerhard Hagn
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Samuel M Meier-Menches
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Joint Metabolome Facility, University of Vienna, Vienna, Austria
| | - Thomas Mohr
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Joint Metabolome Facility, University of Vienna, Vienna, Austria
| | - Johanna C Mader
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Astrid Slany
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Andrea Bileck
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Joint Metabolome Facility, University of Vienna, Vienna, Austria
| | - Julia Brunmair
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Christian Madl
- Institute of Pathology and Microbiology, Krankenanstalt Rudolfstiftung, Vienna, Austria
| | - Lukas Unger
- Division of General Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Barbara Hennlich
- Institute of Pathology and Microbiology, Krankenanstalt Rudolfstiftung, Vienna, Austria
| | - Barbara Weitmayr
- Institute of Pathology and Microbiology, Krankenanstalt Rudolfstiftung, Vienna, Austria
| | - Giorgia Del Favero
- Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Dietmar Pils
- Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| | - Tobias Pukrop
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Nikolaus Pfisterer
- Institute of Pathology and Microbiology, Krankenanstalt Rudolfstiftung, Vienna, Austria
| | | | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Joint Metabolome Facility, University of Vienna, Vienna, Austria
| |
Collapse
|
15
|
Kopczynski D, Hoffmann N, Troppmair N, Coman C, Ekroos K, Kreutz MR, Liebisch G, Schwudke D, Ahrends R. LipidSpace: Simple Exploration, Reanalysis, and Quality Control of Large-Scale Lipidomics Studies. Anal Chem 2023; 95:15236-15244. [PMID: 37792961 PMCID: PMC10585661 DOI: 10.1021/acs.analchem.3c02449] [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] [Received: 06/06/2023] [Accepted: 08/09/2023] [Indexed: 10/06/2023]
Abstract
Lipid analysis gained significant importance due to the enormous range of lipid functions, e.g., energy storage, signaling, or structural components. Whole lipidomes can be quantitatively studied in-depth thanks to recent analytical advancements. However, the systematic comparison of thousands of distinct lipidomes remains challenging. We introduce LipidSpace, a standalone tool for analyzing lipidomes by assessing their structural and quantitative differences. A graph-based comparison of lipid structures is the basis for calculating structural space models and subsequently computing lipidome similarities. When adding study variables such as body weight or health condition, LipidSpace can determine lipid subsets across all lipidomes that describe these study variables well by utilizing machine-learning approaches. The user-friendly GUI offers four built-in tutorials and interactive visual interfaces with pdf export. Many supported data formats allow an efficient (re)analysis of data sets from different sources. An integrated interactive workflow guides the user through the quality control steps. We used this suite to reanalyze and combine already published data sets (e.g., one with about 2500 samples and 576 lipids in one run) and made additional discoveries to the published conclusions with the potential to fill gaps in the current lipid biology understanding. LipidSpace is available for Windows or Linux (https://lifs-tools.org).
Collapse
Affiliation(s)
- Dominik Kopczynski
- Institute
of Analytical Chemistry, University of Vienna, Vienna 1070, Austria
| | - Nils Hoffmann
- Forschungszentrum
Jülich GmbH, Institute for Bio- and Geosciences (IBG-5), Jülich 52428, Germany
| | - Nina Troppmair
- Institute
of Analytical Chemistry, University of Vienna, Vienna 1070, Austria
| | - Cristina Coman
- Institute
of Analytical Chemistry, University of Vienna, Vienna 1070, Austria
| | - Kim Ekroos
- Lipidomics
Consulting Ltd., Esbo 02230, Finland
| | - Michael R. Kreutz
- Leibniz
Group “Dendritic Organelles and Synaptic Function” University
Medical Center Hamburg-Eppendorf, Center
for Molecular Neurobiology, ZMNH, Hamburg 20251, Germany
- RG
Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg 39118, Germany
| | - Gerhard Liebisch
- Institute
of Clinical Chemistry and Laboratory Medicine, University of Regensburg, Regensburg 93053, Germany
| | - Dominik Schwudke
- German
Center for Infection Research (DZIF), Site
Hamburg-Lübeck-Borstel-Riems, Hamburg 22297, Germany
- Airway
Research Center North (ARCN), German Center
for Lung Research (DZL), Grosshansdorf 22927, Germany
- Bioanalytical
Chemistry, Research Center Borstel, Borstel 23845, Germany
| | - Robert Ahrends
- Institute
of Analytical Chemistry, University of Vienna, Vienna 1070, Austria
| |
Collapse
|
16
|
Striesow J, Wesche J, McKitterick N, Busch LM, von Woedtke T, Greinacher A, Bekeschus S, Wende K. Gas plasma-induced platelet activation corresponds to reactive species profiles and lipid oxidation. Free Radic Biol Med 2023; 207:212-225. [PMID: 37490986 DOI: 10.1016/j.freeradbiomed.2023.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 07/27/2023]
Abstract
Surgical-induced hemostasis is a critical step in the closure of incisions, which is frequently achieved via electrocauterization and subsequent tissue necrotization. The latter is associated with postoperative complications. Recent in vivo work suggested reactive species-producing gas plasma technology as a pro-homeostatic agent acting via platelet activation. However, it remained elusive how platelet activation is linked to lipid and protein oxidation and the reactive species compositions. A direct relation between the reactive species composition and platelet activation was revealed by assessing the production of several reactive species and by using antioxidants. In addition, platelet lipidome and proteome analysis identified significantly regulated key lipids in the platelet activation pathway, such as diacylglycerols and phosphatidylinositol as well as oxylipins like thromboxanes. Lipid oxidation products mainly derived from phosphatidylethanolamine and phosphatidylserine species were observed at modest levels. In addition, oxidative post-translational modifications were identified on key proteins of the hemostasis machinery. This study provides new insights into oxidation-induced platelet activation in general and suggests a potential role of those processes in gas plasma-mediated hemostasis in particular.
Collapse
Affiliation(s)
- Johanna Striesow
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Jan Wesche
- Institute of Transfusion Medicine, Greifswald University Medical Center, Sauerbruchstr., 17475, Greifswald, Germany
| | - Nicholas McKitterick
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Larissa M Busch
- Interfaculty Institute for Genetics and Functional Genomics, Greifswald University, Felix-Hausdorff-Str. 8, 17475, Greifswald, Germany
| | - Thomas von Woedtke
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany; Institute for Hygiene and Environmental Medicine, Greifswald University Medical Center, Sauerbruchstr., 17475, Greifswald, Germany
| | - Andreas Greinacher
- Institute of Transfusion Medicine, Greifswald University Medical Center, Sauerbruchstr., 17475, Greifswald, Germany
| | - Sander Bekeschus
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany; Clinic and Policlinic for Dermatology and Venerology, Rostock University Medical Center, Strempelstr. 13, 18057, Rostock, Germany.
| | - Kristian Wende
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany.
| |
Collapse
|
17
|
Karwen T, Kolczynska‐Matysiak K, Gross C, Löffler MC, Friedrich M, Loza‐Valdes A, Schmitz W, Wit M, Dziaczkowski F, Belykh A, Trujillo‐Viera J, El‐Merahbi R, Deppermann C, Nawaz S, Hastoy B, Demczuk A, Erk M, Wieckowski MR, Rorsman P, Heinze KG, Stegner D, Nieswandt B, Sumara G. Platelet-derived lipids promote insulin secretion of pancreatic β cells. EMBO Mol Med 2023; 15:e16858. [PMID: 37490001 PMCID: PMC10493578 DOI: 10.15252/emmm.202216858] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/26/2023] Open
Abstract
Hyperreactive platelets are commonly observed in diabetic patients indicating a potential link between glucose homeostasis and platelet reactivity. This raises the possibility that platelets may play a role in the regulation of metabolism. Pancreatic β cells are the central regulators of systemic glucose homeostasis. Here, we show that factor(s) derived from β cells stimulate platelet activity and platelets selectively localize to the vascular endothelium of pancreatic islets. Both depletion of platelets and ablation of major platelet adhesion or activation pathways consistently resulted in impaired glucose tolerance and decreased circulating insulin levels. Furthermore, we found platelet-derived lipid classes to promote insulin secretion and identified 20-Hydroxyeicosatetraenoic acid (20-HETE) as the main factor promoting β cells function. Finally, we demonstrate that the levels of platelet-derived 20-HETE decline with age and that this parallels with reduced impact of platelets on β cell function. Our findings identify an unexpected function of platelets in the regulation of insulin secretion and glucose metabolism, which promotes metabolic fitness in young individuals.
Collapse
Affiliation(s)
- Till Karwen
- Rudolf Virchow Center for Integrative and Translational BioimagingJulius‐Maximilians University of WürzburgWürzburgGermany
| | | | - Carina Gross
- Institute of Experimental Biomedicine IUniversity Hospital WürzburgWürzburgGermany
| | - Mona C Löffler
- Rudolf Virchow Center for Integrative and Translational BioimagingJulius‐Maximilians University of WürzburgWürzburgGermany
| | - Mike Friedrich
- Rudolf Virchow Center for Integrative and Translational BioimagingJulius‐Maximilians University of WürzburgWürzburgGermany
| | - Angel Loza‐Valdes
- Nencki Institute of Experimental BiologyPolish Academy of SciencesWarszawaPoland
| | - Werner Schmitz
- Theodor Boveri Institute, BiocenterUniversity of WürzburgWürzburgGermany
| | - Magdalena Wit
- Nencki Institute of Experimental BiologyPolish Academy of SciencesWarszawaPoland
| | - Filip Dziaczkowski
- Nencki Institute of Experimental BiologyPolish Academy of SciencesWarszawaPoland
| | - Andrei Belykh
- Nencki Institute of Experimental BiologyPolish Academy of SciencesWarszawaPoland
| | - Jonathan Trujillo‐Viera
- Rudolf Virchow Center for Integrative and Translational BioimagingJulius‐Maximilians University of WürzburgWürzburgGermany
| | - Rabih El‐Merahbi
- Rudolf Virchow Center for Integrative and Translational BioimagingJulius‐Maximilians University of WürzburgWürzburgGermany
| | - Carsten Deppermann
- Rudolf Virchow Center for Integrative and Translational BioimagingJulius‐Maximilians University of WürzburgWürzburgGermany
- Center for Thrombosis and HemostasisUniversity Medical Center of the Johannes Gutenberg‐UniversityMainzGermany
| | - Sameena Nawaz
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and MetabolismChurchill HospitalOxfordUK
| | - Benoit Hastoy
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and MetabolismChurchill HospitalOxfordUK
| | - Agnieszka Demczuk
- Nencki Institute of Experimental BiologyPolish Academy of SciencesWarszawaPoland
| | - Manuela Erk
- Rudolf Virchow Center for Integrative and Translational BioimagingJulius‐Maximilians University of WürzburgWürzburgGermany
| | - Mariusz R Wieckowski
- Nencki Institute of Experimental BiologyPolish Academy of SciencesWarszawaPoland
| | - Patrik Rorsman
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and MetabolismChurchill HospitalOxfordUK
- Department of Physiology, Institute of Neuroscience and PhysiologyUniversity of GöteborgGöteborgSweden
- Oxford National Institute for Health Research, Biomedical Research CentreChurchill HospitalOxfordUK
| | - Katrin G Heinze
- Rudolf Virchow Center for Integrative and Translational BioimagingJulius‐Maximilians University of WürzburgWürzburgGermany
| | - David Stegner
- Rudolf Virchow Center for Integrative and Translational BioimagingJulius‐Maximilians University of WürzburgWürzburgGermany
- Institute of Experimental Biomedicine IUniversity Hospital WürzburgWürzburgGermany
| | - Bernhard Nieswandt
- Rudolf Virchow Center for Integrative and Translational BioimagingJulius‐Maximilians University of WürzburgWürzburgGermany
- Institute of Experimental Biomedicine IUniversity Hospital WürzburgWürzburgGermany
| | - Grzegorz Sumara
- Rudolf Virchow Center for Integrative and Translational BioimagingJulius‐Maximilians University of WürzburgWürzburgGermany
- Nencki Institute of Experimental BiologyPolish Academy of SciencesWarszawaPoland
| |
Collapse
|
18
|
Gawaz M, Geisler T, Borst O. Current concepts and novel targets for antiplatelet therapy. Nat Rev Cardiol 2023; 20:583-599. [PMID: 37016032 DOI: 10.1038/s41569-023-00854-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/06/2023]
Abstract
Platelets have a crucial role in haemostasis and atherothrombosis. Pharmacological control of platelet hyper-reactivity has become a cornerstone in the prevention of thrombo-ischaemic complications in atherosclerotic diseases. Current antiplatelet therapies substantially improve clinical outcomes in patients with coronary artery disease, but at the cost of increased risk of bleeding. Beyond their role in thrombosis, platelets are known to regulate inflammatory (thrombo-inflammatory) and microcirculatory pathways. Therefore, controlling platelet hyper-reactivity might have implications for both tissue inflammation (myocardial ischaemia) and vascular inflammation (vulnerable plaque formation) to prevent atherosclerosis. In this Review, we summarize the pathophysiological role of platelets in acute myocardial ischaemia, vascular inflammation and atherosclerotic progression. Furthermore, we highlight current clinical concepts of antiplatelet therapy that have contributed to improving patient care and have facilitated more individualized therapy. Finally, we discuss novel therapeutic targets and compounds for antiplatelet therapy that are currently in preclinical development, some of which have a more favourable safety profile than currently approved drugs with regard to bleeding risk. These novel antiplatelet targets might offer new strategies to treat cardiovascular disease.
Collapse
Affiliation(s)
- Meinrad Gawaz
- Department of Cardiology and Angiology, Eberhard Karls University of Tübingen, Tübingen, Germany.
| | - Tobias Geisler
- Department of Cardiology and Angiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
19
|
Duchez AC, Fauteux-Daniel S, Ebermeyer T, Heestermans M, Arthaud CA, Eyraud MA, Prier A, Audoux E, Portais JC, Bertrand-Michel J, Garraud O, Hamzeh-Cognasse H, Boilard E, Cognasse F. Lipidomic analysis of differently prepared platelet concentrates in additive solution during storage. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2023; 21:409-421. [PMID: 36346879 PMCID: PMC10497391 DOI: 10.2450/2022.0144-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Structural and biochemical changes in stored platelets are influenced by collection and processing methods. Lesions may appear during platelet concentrate storage, some of which may be involved in adverse transfusion reactions. The preparation and storage of platelet concentrates (PC) may modify and even damage the lipid mediator content. The aim of this study was to investigate the lipidomic profile identified in the supernatants of PCs according to processing and storage conditions, both after leukocyte filtration and contained in platelet additive solution (PAS), comparing single donor apheresis (SDA) products with pooled buffy coat (BC) products. MATERIALS AND METHODS We investigated the accumulation of various lipid mediators including lysophospholipids (LP) and eicosanoids in SDA and BC products stored for 0-5 days. All products were processed following French Blood Establishment (EFS) procedures in accordance with EDQM/GTS European Standards. Both SDA and BC were leukocyte reduced and conserved in 35% autologous donor plasma and 65% platelet additive solution. Lipidomic analysis was performed on PC supernatants using LS/MS spectrometry. RESULTS Our data demonstrate that lysophosphatidylcholine (LPC) levels were higher in BCs compared to SDAs, with no difference in lysophosphatidic acid (LPA) expression between the two preparation methods. Results for other eicosanoids showed greater similarity; indeed, no clear pattern emerged from analysis of eicosanoids in terms of storage time and process. In general, we observed longitudinal lipid mediator modulation for both SDAs and BCs, particularly at later time points. DISCUSSION The expression of LPC and some eicosanoids in BCs could be used as novel biomarkers of PC quality. Future studies are needed to explore their impact on adverse transfusion reactions.
Collapse
Affiliation(s)
- Anne-Claire Duchez
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- University of Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 SAINBIOSE, Saint-Étienne, France
| | - Sébastien Fauteux-Daniel
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- University of Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 SAINBIOSE, Saint-Étienne, France
| | - Theo Ebermeyer
- University of Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 SAINBIOSE, Saint-Étienne, France
| | - Marco Heestermans
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- University of Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 SAINBIOSE, Saint-Étienne, France
| | - Charles-Antoine Arthaud
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- University of Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 SAINBIOSE, Saint-Étienne, France
| | - Marie-Ange Eyraud
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- University of Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 SAINBIOSE, Saint-Étienne, France
| | - Amélie Prier
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- University of Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 SAINBIOSE, Saint-Étienne, France
| | - Estelle Audoux
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- University of Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 SAINBIOSE, Saint-Étienne, France
| | - Jean-Charles Portais
- MetaToul-Lipidomic MetaboHUB Core Facility, Inserm, U1048, Toulouse France
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Justine Bertrand-Michel
- MetaToul-Lipidomic MetaboHUB Core Facility, Inserm, U1048, Toulouse France
- INSERM UMR 1214, ToNIC: Toulouse NeuroImaging Center, Toulouse, France
| | - Olivier Garraud
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- University of Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 SAINBIOSE, Saint-Étienne, France
| | - Hind Hamzeh-Cognasse
- University of Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 SAINBIOSE, Saint-Étienne, France
| | - Eric Boilard
- Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Canada
- Université Laval and Centre de recherche ARThrite, Québec, Canada
| | - Fabrice Cognasse
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- University of Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 SAINBIOSE, Saint-Étienne, France
| |
Collapse
|
20
|
de Jonckheere B, Kollotzek F, Münzer P, Göb V, Fischer M, Mott K, Coman C, Troppmair NN, Manke MC, Zdanyte M, Harm T, Sigle M, Kopczynski D, Bileck A, Gerner C, Hoffmann N, Heinzmann D, Assinger A, Gawaz M, Stegner D, Schulze H, Borst O, Ahrends R. Critical shifts in lipid metabolism promote megakaryocyte differentiation and proplatelet formation. NATURE CARDIOVASCULAR RESEARCH 2023; 2:835-852. [PMID: 38075556 PMCID: PMC7615361 DOI: 10.1038/s44161-023-00325-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2024]
Abstract
During megakaryopoiesis, megakaryocytes (MK) undergo cellular morphological changes with strong modification of membrane composition and lipid signaling. Here we adopt a lipid-centric multiomics approach to create a quantitative map of the MK lipidome during maturation and proplatelet formation. Data reveal that MK differentiation is driven by an increased fatty acyl import and de novo lipid synthesis, resulting in an anionic membrane phenotype. Pharmacological perturbation of fatty acid import and phospholipid synthesis blocked membrane remodeling and directly reduced MK polyploidization and proplatelet formation resulting in thrombocytopenia. The anionic lipid shift during megakaryopoiesis was paralleled by lipid-dependent relocalization of the scaffold protein CKIP-1 and recruitment of the kinase CK2α to the plasma membrane, which seems to be essential for sufficient platelet biogenesis. Overall, this study provides a framework to understand how the MK lipidome is altered during maturation and the impact of MK membrane lipid remodeling on MK kinase signaling involved in thrombopoiesis.
Collapse
Affiliation(s)
- Bianca de Jonckheere
- Institute of Analytical Chemistry, University of Vienna, Austria
- Vienna Doctoral School in Chemistry, University of Vienna, Austria
| | - Ferdinand Kollotzek
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Germany
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Patrick Münzer
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Germany
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Vanessa Göb
- Institute for Experimental Biomedicine, University Hospital Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany
| | - Melina Fischer
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Germany
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Kristina Mott
- Institute for Experimental Biomedicine, University Hospital Würzburg, Germany
| | - Cristina Coman
- Institute of Analytical Chemistry, University of Vienna, Austria
| | - Nina Nicole Troppmair
- Institute of Analytical Chemistry, University of Vienna, Austria
- Vienna Doctoral School in Chemistry, University of Vienna, Austria
| | - Mailin-Christin Manke
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Germany
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Monika Zdanyte
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Germany
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Tobias Harm
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Manuel Sigle
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | | | - Andrea Bileck
- Institute of Analytical Chemistry, University of Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University of Vienna, Austria
| | - Christopher Gerner
- Institute of Analytical Chemistry, University of Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University of Vienna, Austria
| | - Nils Hoffmann
- Institute of Analytical Chemistry, University of Vienna, Austria
- Forschungszentrum Jülich GmbH, Institute for Bio- and Geosciences (IBG-5) Jülich, Germany
| | - David Heinzmann
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Alice Assinger
- Institute of Physiology, Centre of Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - David Stegner
- Institute for Experimental Biomedicine, University Hospital Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany
| | - Harald Schulze
- Institute for Experimental Biomedicine, University Hospital Würzburg, Germany
| | - Oliver Borst
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Germany
- Department of Cardiology and Angiology, University of Tübingen, Germany
| | - Robert Ahrends
- Institute of Analytical Chemistry, University of Vienna, Austria
| |
Collapse
|
21
|
Ravera S, Signorello MG, Panfoli I. Platelet Metabolic Flexibility: A Matter of Substrate and Location. Cells 2023; 12:1802. [PMID: 37443836 PMCID: PMC10340290 DOI: 10.3390/cells12131802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
Platelets are cellular elements that are physiologically involved in hemostasis, inflammation, thrombotic events, and various human diseases. There is a link between the activation of platelets and their metabolism. Platelets possess considerable metabolic versatility. Although the role of platelets in hemostasis and inflammation is known, our current understanding of platelet metabolism in terms of substrate preference is limited. Platelet activation triggers an oxidative metabolism increase to sustain energy requirements better than aerobic glycolysis alone. In addition, platelets possess extra-mitochondrial oxidative phosphorylation, which could be one of the sources of chemical energy required for platelet activation. This review aims to provide an overview of flexible platelet metabolism, focusing on the role of metabolic compartmentalization in substrate preference, since the metabolic flexibility of stimulated platelets could depend on subcellular localization and functional timing. Thus, developing a detailed understanding of the link between platelet activation and metabolic changes is crucial for improving human health.
Collapse
Affiliation(s)
- Silvia Ravera
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy;
| | | | - Isabella Panfoli
- Department of Pharmacy (DIFAR), University of Genoa, 16132 Genoa, Italy;
| |
Collapse
|
22
|
Manke MC, Roslan A, Walker B, Münzer P, Kollotzek F, Peng B, Mencl S, Coman C, Szepanowski RD, Schulze H, Lieberman AP, Lang F, Gawaz M, Kleinschnitz C, Lukowski R, Ahrends R, Bobe R, Borst O. Niemann-Pick C1 protein regulates platelet membrane-associated calcium ion signaling in thrombo-occlusive diseases in mice. J Thromb Haemost 2023; 21:1957-1966. [PMID: 37054918 DOI: 10.1016/j.jtha.2023.03.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/14/2023] [Accepted: 03/28/2023] [Indexed: 04/15/2023]
Abstract
BACKGROUND Pathophysiologic platelet activation leads to thrombo-occlusive diseases such as myocardial infarction or ischemic stroke. Niemann-Pick C1 protein (NPC1) is involved in the regulation of lysosomal lipid trafficking and calcium ion (Ca2+) signaling, and its genetic mutation causes a lysosomal storage disorder. Lipids and Ca2+ are key players in the complex orchestration of platelet activation. OBJECTIVES The present study aimed to determine the impact of NPC1 on Ca2+ mobilization during platelet activation in thrombo-occlusive diseases. METHODS Using MK/platelet-specific knockout mice of Npc1 (Npc1Pf4∆/Pf4∆), ex vivo and in vitro approaches as well as in vivo models of thrombosis, we investigated the effect of Npc1 on platelet function and thrombus formation. RESULTS We showed that Npc1Pf4∆/Pf4∆ platelets display increased sphingosine levels and a locally impaired membrane-associated and SERCA3-dependent Ca2+ mobilisation compared to platelets from wildtype littermates (Npc1lox/lox). Further, we observed decreased platelet. CONCLUSION Our findings highlight that NPC1 regulates membrane-associated and SERCA3-dependent Ca2+ mobilization during platelet activation and that MK/platelet-specific ablation of Npc1 protects against experimental models of arterial thrombosis and myocardial or cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Mailin-Christin Manke
- DFG Heisenberg Group Thrombocardiology; Department of Cardiology, Angiology and Cardiovascular Medicine, University of Tübingen, Germany
| | - Anna Roslan
- Department of Pharmacology, Toxicology and Clinical Pharmacy, University of Tübingen, Germany
| | | | - Patrick Münzer
- DFG Heisenberg Group Thrombocardiology; Department of Cardiology, Angiology and Cardiovascular Medicine, University of Tübingen, Germany
| | - Ferdinand Kollotzek
- DFG Heisenberg Group Thrombocardiology; Department of Cardiology, Angiology and Cardiovascular Medicine, University of Tübingen, Germany
| | - Bing Peng
- Leibniz-Institut für Analytische Wissenschaften-ISAS, Dortmund, Germany; Division of Rheumatology, Department of Medicine Solna, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Stine Mencl
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Cristina Coman
- Department of Analytical Chemistry, University of Vienna, Austria
| | - Rebecca D Szepanowski
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Harald Schulze
- Institute of Experimental Biomedicine, University Hospital Würzburg, Germany
| | | | - Florian Lang
- Department of Physiology, University of Tübingen, Germany
| | | | - Christoph Kleinschnitz
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, University of Tübingen, Germany
| | - Robert Ahrends
- Leibniz-Institut für Analytische Wissenschaften-ISAS, Dortmund, Germany; Department of Analytical Chemistry, University of Vienna, Austria
| | - Régis Bobe
- HITh, UMR_S1176, INSERM, Université Paris-Saclay, France
| | - Oliver Borst
- DFG Heisenberg Group Thrombocardiology; Department of Cardiology, Angiology and Cardiovascular Medicine, University of Tübingen, Germany.
| |
Collapse
|
23
|
Hormann FL, Sommer S, Heiles S. Formation and Tandem Mass Spectrometry of Doubly Charged Lipid-Metal Ion Complexes. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023. [PMID: 37315187 DOI: 10.1021/jasms.3c00126] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Phospholipids are major components of most eukaryotic cell membranes. Changes in metabolic states are often accompanied by phospholipid structure variations. The structural changes of phospholipids are the hallmark of disease states, or specific lipid structures have been associated with distinct organisms. Prime examples are microorganisms that synthesize phospholipids with, for example, different branched chain fatty acids. Assignment and relative quantitation of structural isomers of phospholipids that arise from attachment of different fatty acids to the glycerophospholipid backbone are difficult with routine tandem mass spectrometry or with liquid chromatography without authentic standards. In this work, we report on the observation that all investigated phospholipid classes form doubly charged lipid-metal ion complexes during electrospray ionization (ESI) and show that these complexes can be used to assign lipid classes and fatty acid moieties, distinguish isomers of branched chain fatty acids, and relatively quantify these isomers in positive-ion mode. Use of water free methanol and addition of divalent metal salts (100 mol %) to ESI spray solutions afford highly abundant doubly charged lipid-metal ion complexes (up to 70 times of protonated compounds). Higher-energy collisional dissociation and collision-induced dissociation of doubly charged complexes yield a diverse set of lipid class-dependent fragment ions. In common for all lipid classes is the liberation of fatty acid-metal adducts that yield fragment ions from the fatty acid hydrocarbon chain upon activation. This ability is used to pinpoint sites of branching in saturated fatty acids and is showcased for free fatty acids as well as glycerophospholipids. The analytical utility of doubly charged phospholipid-metal ion complexes is demonstrated by distinguishing fatty acid branching-site isomers in phospholipid mixtures and relatively quantifying the corresponding isomeric compounds.
Collapse
Affiliation(s)
- Felix-Levin Hormann
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Otto-Hahn-Straße 6b, 44139 Dortmund, Germany
- Lipidomics, Faculty of Chemistry, University of Duisburg-Essen, Universitätsstrasse 5, 45141 Essen, Germany
| | - Simon Sommer
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Sven Heiles
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Otto-Hahn-Straße 6b, 44139 Dortmund, Germany
- Lipidomics, Faculty of Chemistry, University of Duisburg-Essen, Universitätsstrasse 5, 45141 Essen, Germany
| |
Collapse
|
24
|
Polzin A, Dannenberg L, Benkhoff M, Barcik M, Helten C, Mourikis P, Ahlbrecht S, Wildeis L, Ziese J, Zikeli D, Metzen D, Hu H, Baensch L, Schröder NH, Keul P, Weske S, Wollnitzke P, Duse D, Saffak S, Cramer M, Bönner F, Müller T, Gräler MH, Zeus T, Kelm M, Levkau B. Revealing concealed cardioprotection by platelet Mfsd2b-released S1P in human and murine myocardial infarction. Nat Commun 2023; 14:2404. [PMID: 37100836 PMCID: PMC10133218 DOI: 10.1038/s41467-023-38069-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/13/2023] [Indexed: 04/28/2023] Open
Abstract
Antiplatelet medication is standard of care in acute myocardial infarction (AMI). However, it may have obscured beneficial properties of the activated platelet secretome. We identify platelets as major source of a sphingosine-1-phosphate (S1P) burst during AMI, and find its magnitude to favorably associate with cardiovascular mortality and infarct size in STEMI patients over 12 months. Experimentally, administration of supernatant from activated platelets reduces infarct size in murine AMI, which is blunted in platelets deficient for S1P export (Mfsd2b) or production (Sphk1) and in mice deficient for cardiomyocyte S1P receptor 1 (S1P1). Our study reveals an exploitable therapeutic window in antiplatelet therapy in AMI as the GPIIb/IIIa antagonist tirofiban preserves S1P release and cardioprotection, whereas the P2Y12 antagonist cangrelor does not. Here, we report that platelet-mediated intrinsic cardioprotection is an exciting therapeutic paradigm reaching beyond AMI, the benefits of which may need to be considered in all antiplatelet therapies.
Collapse
Affiliation(s)
- Amin Polzin
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital, Düsseldorf, Germany
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Lisa Dannenberg
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Marcel Benkhoff
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Maike Barcik
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Carolin Helten
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Mourikis
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Samantha Ahlbrecht
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Laura Wildeis
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Justus Ziese
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Dorothee Zikeli
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Daniel Metzen
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Hao Hu
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Leonard Baensch
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Nathalie H Schröder
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Petra Keul
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sarah Weske
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Wollnitzke
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Dragos Duse
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Süreyya Saffak
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Mareike Cramer
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Florian Bönner
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital, Düsseldorf, Germany
| | - Tina Müller
- Department of Anesthesiology and Intensive Care, University Hospital Jena, Jena, Germany
| | - Markus H Gräler
- Department of Anesthesiology and Intensive Care, University Hospital Jena, Jena, Germany
| | - Tobias Zeus
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital, Düsseldorf, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital, Düsseldorf, Germany
| | - Bodo Levkau
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
25
|
Heimerl S, Höring M, Kopczynski D, Sigruener A, Hart C, Burkhardt R, Black A, Ahrends R, Liebisch G. Quantification of bulk lipid species in human platelets and their thrombin-induced release. Sci Rep 2023; 13:6154. [PMID: 37061580 PMCID: PMC10105721 DOI: 10.1038/s41598-023-33076-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 04/06/2023] [Indexed: 04/17/2023] Open
Abstract
Lipids play a central role in platelet physiology. Changes in the lipidome have already been described for basal and activated platelets. However, quantitative lipidomic data of platelet activation, including the released complex lipids, are unavailable. Here we describe an easy-to-use protocol based on flow-injection mass spectrometry for the quantitative analysis of bulk lipid species in basal and activated human platelets and their lipid release after thrombin activation. We provide lipid species concentrations of 12 healthy human donors, including cholesteryl ester (CE), ceramide (Cer), free cholesterol (FC), hexosylceramide (HexCer), lysophosphatidylcholine (LPC), lysophosphatidylethanolamine (LPE), phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylinositol (PI), phosphatidylserine (PS), sphingomyelin (SM) and triglycerides (TG). The assay exhibited good technical repeatability (CVs < 5% for major lipid species in platelets). Except for CE and TG, the inter-donor variability of the majority of lipid species concentrations in platelets was < 30% CV. Balancing of concentrations revealed the generation of LPC and loss of TG. Changes in lipid species concentrations indicate phospholipase-mediated release of arachidonic acid mainly from PC, PI, and PE but not from PS. Thrombin induced lipid release was mainly composed of FC, PS, PC, LPC, CE, and TG. The similarity of the released lipidome with that of plasma implicates that lipid release may originate from the open-canalicular system (OCS). The repository of lipid species concentrations determined with this standardized platelet release assay contribute to elucidating the physiological role of platelet lipids and provide a basis for investigating the platelet lipidome in patients with hemorrhagic or thrombotic disorders.
Collapse
Affiliation(s)
- Susanne Heimerl
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93042, Regensburg, Germany
| | - Marcus Höring
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93042, Regensburg, Germany
| | - Dominik Kopczynski
- Department of Analytical Chemistry, University of Vienna, Vienna, Austria
| | - Alexander Sigruener
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93042, Regensburg, Germany
| | - Christina Hart
- Department of Hematology and Oncology, Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93042, Regensburg, Germany
| | - Anne Black
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93042, Regensburg, Germany
| | - Robert Ahrends
- Department of Analytical Chemistry, University of Vienna, Vienna, Austria
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93042, Regensburg, Germany.
| |
Collapse
|
26
|
Solari FA, Krahn D, Swieringa F, Verhelst S, Rassaf T, Tasdogan A, Zahedi RP, Lorenz K, Renné T, Heemskerk JWM, Sickmann A. Multi-omics approaches to study platelet mechanisms. Curr Opin Chem Biol 2023; 73:102253. [PMID: 36689818 DOI: 10.1016/j.cbpa.2022.102253] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/17/2022] [Accepted: 11/27/2022] [Indexed: 01/22/2023]
Abstract
Platelets are small anucleate cell fragments (2-4 μm in diameter) in the blood, which play an essential role in thrombosis and hemostasis. Genetic or acquired platelet dysfunctions are linked to bleeding, increased risk of thromboembolic events and cardiovascular diseases. Advanced proteomic approaches may pave the way to a better understanding of the roles of platelets in hemostasis, and pathophysiological processes such as inflammation, metastatic spread and thrombosis. Further insights into the molecular biology of platelets are crucial to aid drug development and identify diagnostic markers of platelet activation. Platelet activation is known to be an extremely rapid process and involves multiple post-translational mechanisms at sub second time scale, including proteolysis and phosphorylation. Multi-omics technologies and biochemical approaches can be exploited to precisely probe and define these posttranslational pathways. Notably, the absence of a nucleus in platelets significantly reduces the number of present proteins, simplifying mass spectrometry-based proteomics and metabolomics approaches.
Collapse
Affiliation(s)
- Fiorella A Solari
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., 44143, Dortmund, Germany
| | - Daniel Krahn
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., 44143, Dortmund, Germany
| | - Frauke Swieringa
- Synapse Research Institute Maastricht, 6217 KD, Maastricht, the Netherlands
| | - Steven Verhelst
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., 44143, Dortmund, Germany; Department of Cellular and Molecular Medicine, KU Leuven, University of Leuven, Leuven, Belgium
| | - Tienush Rassaf
- Clinic for Cardiology and Angiology, University Hospital Essen, Essen, Germany
| | - Alpaslan Tasdogan
- Department of Dermatology, University Hospital Essen & German Cancer Consortium, Partner Site, Essen, Germany
| | - Rene P Zahedi
- Department of Internal Medicine, University of Manitoba, Canada
| | - Kristina Lorenz
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., 44143, Dortmund, Germany; Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Thomas Renné
- Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., 44143, Dortmund, Germany; Medizinische Fakultät, Ruhr-Universität Bochum, Bochum, Germany; Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, United Kingdom.
| |
Collapse
|
27
|
Harm T, Frey M, Dittrich K, Goldschmied A, Rohlfing AK, Fu X, Brun A, Castor T, Rath D, Müller K, Lammerhofer M, Gawaz M. Statin Treatment Is Associated with Alterations in the Platelet Lipidome. Thromb Haemost 2023; 123:585-596. [PMID: 36898406 DOI: 10.1055/s-0043-1764353] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
BACKGROUND Platelets are key players in the pathophysiology of coronary artery disease (CAD) and platelet hyperreactivity leads to increased risk of developing adverse cardiovascular events. Further, significant changes in the platelet lipidome occur in patients with acute coronary syndrome (ACS) and critically regulated lipids lead to platelet hyperresponsiveness. Statin treatment is crucial in the treatment and prevention of patients with CAD by remodeling lipid metabolism. OBJECTIVE In this study, we investigate the platelet lipidome of CAD patients by untargeted lipidomics, highlighting significant changes between statin-treated and naïve patients. METHODS We characterized the platelet lipidome in a CAD cohort (n = 105) by an untargeted lipidomics approach using liquid chromatography coupled to mass spectrometry. RESULTS Among the annotated lipids, 41 lipids were significantly upregulated in statin-treated patients, whereas 6 lipids were downregulated compared to naïve patients. The most prominent upregulated lipids in statin-treated patients belong to the class of triglycerides, cholesteryl esters, palmitic acid, and oxidized phospholipids, whereas mainly glycerophospholipids were downregulated compared to untreated patients. A more pronounced effect of statin treatment on the platelet lipidome was observed in ACS patients. We further highlight a dose-dependent influence on the platelet lipidome. CONCLUSION Our results reveal that the platelet lipidome is altered in CAD patients with statin treatment and upregulated lipids embody mainly characteristic triglycerides, whereas downregulated lipids mostly compromise glycerophospholipids, which may play a role in the pathophysiology of CAD. Results of this study may contribute to the understanding of statin treatment softening the lipid phenotype.
Collapse
Affiliation(s)
- Tobias Harm
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Moritz Frey
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Kristina Dittrich
- Institute of Pharmaceutical Sciences, Pharmaceutical (Bio-)Analysis, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas Goldschmied
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Anne-Katrin Rohlfing
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Xiaoqing Fu
- Institute of Pharmaceutical Sciences, Pharmaceutical (Bio-)Analysis, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Adrian Brun
- Institute of Pharmaceutical Sciences, Pharmaceutical (Bio-)Analysis, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Tatsiana Castor
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Dominik Rath
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Karin Müller
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Michael Lammerhofer
- Institute of Pharmaceutical Sciences, Pharmaceutical (Bio-)Analysis, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
28
|
Duchez AC, Fauteux-Daniel S, Sut C, Ebermeyer T, Heestermans M, Arthaud CA, Eyraud MA, Prier A, Audoux E, Bertrand-Michel J, Payrastre B, Garraud O, Boilard E, Hamzeh-Cognasse H, Cognasse F. Bioactive lipids as biomarkers of adverse reactions associated with apheresis platelet concentrate transfusion. Front Immunol 2023; 14:1031968. [PMID: 37138863 PMCID: PMC10149858 DOI: 10.3389/fimmu.2023.1031968] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 03/31/2023] [Indexed: 05/05/2023] Open
Abstract
Platelet concentrate (PC) transfusion seeks to provide haemostasis in patients presenting severe central thrombocytopenia or severe bleeding. PCs may induce adverse reactions (AR) that can occasionally be severe (SAR). PCs contain active biomolecules such as cytokines and lipid mediators. The processing and storage of PCs creates so-called structural and biochemical storage lesions that accumulate when blood products reach their shelf life. We sought to investigate lipid mediators as bioactive molecules of interest during storage and review associations with adverse reactions post-transfusion. To facilitate understanding, we focused on single donor apheresis (SDA) PCs with approximately 31.8% of PCs being delivered in our setting. Indeed, pooled PCs are the most widely transfused products, but the study of a single donor lipid mediator is easier to interpret. We are investigating key lipid mediators involved in AR. Adverse reactions were closely monitored in accordance with current national and regional haemovigilance protocols. Residual PCs were analysed post-transfusion in a series of observations, both with and without severe reactions in recipients. A decrease in the lysophosphatidylcholine species to produce the lysophosphatidic acid species has been observed during storage and in the case of AR. Lysophosphatidic acid increased with primarily platelet-inhibitor lipids. Anti-inflammatory platelet-induced inhibition lipids were weakly expressed in cases of severe adverse reactions. We therefore propose that a decrease in lysophosphatidylcholine and an increase in lysophosphatidic acid can prospectively predict serious adverse transfusion reactions.
Collapse
Affiliation(s)
- Anne-Claire Duchez
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- SAINBIOSE, INSERM, University of Saint-Etienne, Saint-Étienne, France
- *Correspondence: Anne-Claire Duchez,
| | - Sébastien Fauteux-Daniel
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- SAINBIOSE, INSERM, University of Saint-Etienne, Saint-Étienne, France
| | - Caroline Sut
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
| | - Theo Ebermeyer
- SAINBIOSE, INSERM, University of Saint-Etienne, Saint-Étienne, France
| | - Marco Heestermans
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- SAINBIOSE, INSERM, University of Saint-Etienne, Saint-Étienne, France
| | - Charles-Antoine Arthaud
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- SAINBIOSE, INSERM, University of Saint-Etienne, Saint-Étienne, France
| | - Marie-Ange Eyraud
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- SAINBIOSE, INSERM, University of Saint-Etienne, Saint-Étienne, France
| | - Amélie Prier
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- SAINBIOSE, INSERM, University of Saint-Etienne, Saint-Étienne, France
| | - Estelle Audoux
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- SAINBIOSE, INSERM, University of Saint-Etienne, Saint-Étienne, France
| | - Justine Bertrand-Michel
- MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
- I2MC, Université de Toulouse, Inserm, Université Toulouse III – Paul Sabatier (UPS), Toulouse, France
| | - Bernard Payrastre
- I2MC, Université de Toulouse, Inserm, Université Toulouse III – Paul Sabatier (UPS), Toulouse, France
- INSERM UMR, ToNIC: Toulouse NeuroImaging Centre, Toulouse, France
| | - Olivier Garraud
- SAINBIOSE, INSERM, University of Saint-Etienne, Saint-Étienne, France
| | - Eric Boilard
- Department of Infectious Diseases and Immunity, Centre de recherche du CHU de Québec, Québec, QC, Canada
- Université Laval and Centre de recherche ARThrite, Québec, QC, Canada
| | | | - Fabrice Cognasse
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- SAINBIOSE, INSERM, University of Saint-Etienne, Saint-Étienne, France
| |
Collapse
|
29
|
Sun B, Jiang S, Li M, Zhang Y, Zhou Y, Wei X, Wang H, Si N, Bian B, Zhao H. Lipidomics combined with transcriptomic and mass spectrometry imaging analysis of the Asiatic toad (Bufo gargarizans) during metamorphosis and bufadienolide accumulation. Chin Med 2022; 17:123. [PMID: 36333760 PMCID: PMC9636624 DOI: 10.1186/s13020-022-00676-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/15/2022] [Indexed: 11/06/2022] Open
Abstract
Background To adapt to life on land, Asiatic toads (Bufo gargarizans) must remodel their bodies and refine their chemical defenses in water. The full scope of the mechanisms underlying these processes has yet to be revealed. Bufadienolides (BDs) are chemical defense substances secreted by toads when they are in danger, and they have high medicinal value in treating heart failure, cancer, and hepatitis. However, the artificial breeding of toads to increase BDs has been unsuccessful due to the high mortality of toad larvae during metamorphosis. Method Toad larvae at different growth stages were selected to study the changes in the metamorphosis process under the same growth conditions. The differences of tadpoles were explored, including body remodeling, energy metabolism, synthesis and regulation of BDs, through lipidomic technology, transcriptomic technology, and mass spectrometry imaging technology during metamorphosis. Results During metamorphosis, tadpoles underwent significant changes in lipid metabolism due to body remodeling to adapt to terrestrial life, which involved ketosis, lipogenesis, cholesterol metabolism, and fatty acid oxidation. The accumulation trend of BDs was observed. “Pentose phosphate pathway” and “Aromatase activity” may be the critical pathway and GO term in BD synthesis, involving 16 genes predominantly expressed in the liver. The involved genes were mainly expressed in the liver, consistent with the synthetic site observed by mass spectrometry imaging. Conclusion Together, our findings presented the changes in the toad larvae during metamorphosis and highlighted the accumulation process of BDs as well as the regulatory pathways and synthetic site, providing research and theoretical basis for future development of the toad resources. Supplementary Information The online version contains supplementary material available at 10.1186/s13020-022-00676-7.
Collapse
|
30
|
Lawanprasert A, Pimcharoen S, Sumner SE, Watson CT, Manning KB, Kirimanjeswara GS, Medina SH. Heparin-Peptide Nanogranules for Thrombosis-Actuated Anticoagulation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203751. [PMID: 36192159 PMCID: PMC9671832 DOI: 10.1002/smll.202203751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/25/2022] [Indexed: 06/16/2023]
Abstract
Despite nearly a century of clinical use as a blood thinner, heparin's rapid serum clearance and potential to induce severe bleeding events continue to urge the development of more effective controlled delivery strategies. Subcutaneous depots that steadily release the anticoagulant into circulation represent a promising approach to reducing overdose frequency, sustaining therapeutic concentrations of heparin in plasma, and prolonging anticoagulant activity in a safe and effective manner. Subcutaneously deliverable heparin-peptide nanogranules that allow for long-lasting heparin bioavailability in the circulatory system, while enabling on-demand activation of heparin's anticoagulant effects in the thrombus microenvironment, are reported. Biophysical studies demonstrate this responsive behavior is due to the sequestration of heparin within self-assembling peptide nanofibrils and its mechanically actuated decoupling to elicit antithrombotic effects at the clotting site. In vivo studies show these unique properties converge to allow subcutaneous nanogranule depots to extend heparin serum concentrations for an order of magnitude longer than standard dosing regimens while enabling prolonged and controlled anticoagulant activity. This biohybrid delivery system demonstrates a potentially scalable platform for the development of safer, easier to administer, and more effective antithrombotic nanotechnologies.
Collapse
Affiliation(s)
- Atip Lawanprasert
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, 16802-4400, USA
| | - Sopida Pimcharoen
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, 16802-4400, USA
| | - Sarah E Sumner
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, 16802-4400, USA
| | - Connor T Watson
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, 16802-4400, USA
| | - Keefe B Manning
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, 16802-4400, USA
| | - Girish S Kirimanjeswara
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA, 16802-4400, USA
- Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA, 16802-4400, USA
- Center for Molecular Immunology and Infectious Disease, Pennsylvania State University, University Park, PA, 16802-4400, USA
| | - Scott H Medina
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, 16802-4400, USA
- Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802-4400, USA
| |
Collapse
|
31
|
Manke MC, Ahrends R, Borst O. Platelet lipid metabolism in vascular thrombo-inflammation. Pharmacol Ther 2022; 237:108258. [DOI: 10.1016/j.pharmthera.2022.108258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/12/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022]
|
32
|
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: 0] [Impact Index Per Article: 0] [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
|
33
|
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
|
34
|
Ariëns RA, Hunt BJ, Agbani EO, Ahnström J, Ahrends R, Alikhan R, Assinger A, Bagoly Z, Balduini A, Barbon E, Barrett CD, Batty P, Carneiro JDA, Chan W, de Maat M, de Wit K, Denis C, Ellis MH, Eslick R, Fu H, Hayward CPM, Ho‐Tin‐Noé B, Klok F, Kumar R, Leiderman K, Litvinov RI, Mackman N, McQuilten Z, Neal MD, Parker WAE, Preston RJS, Rayes J, Rezaie AR, Roberts LN, Rocca B, Shapiro S, Siegal DM, Sousa LP, Suzuki‐Inoue K, Zafar T, Zhou J. Illustrated State-of-the-Art Capsules of the ISTH 2022 Congress. Res Pract Thromb Haemost 2022; 6:e12747. [PMID: 35814801 PMCID: PMC9257378 DOI: 10.1002/rth2.12747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
The ISTH London 2022 Congress is the first held (mostly) face-to-face again since the COVID-19 pandemic took the world by surprise in 2020. For 2 years we met virtually, but this year's in-person format will allow the ever-so-important and quintessential creativity and networking to flow again. What a pleasure and joy to be able to see everyone! Importantly, all conference proceedings are also streamed (and available recorded) online for those unable to travel on this occasion. This ensures no one misses out. The 2022 scientific program highlights new developments in hemophilia and its treatment, acquired and other inherited bleeding disorders, thromboinflammation, platelets and coagulation, clot structure and composition, fibrinolysis, vascular biology, venous thromboembolism, women's health, arterial thrombosis, pediatrics, COVID-related thrombosis, vaccine-induced thrombocytopenia with thrombosis, and omics and diagnostics. These areas are elegantly reviewed in this Illustrated Review article. The Illustrated Review is a highlight of the ISTH Congress. The format lends itself very well to explaining the science, and the collection of beautiful graphical summaries of recent developments in the field are stunning and self-explanatory. This clever and effective way to communicate research is revolutionary and different from traditional formats. We hope you enjoy this article and will be inspired by its content to generate new research ideas.
Collapse
Affiliation(s)
| | | | - Ejaife O. Agbani
- Department of Physiology and Pharmacology, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | | | - Robert Ahrends
- Institute of Analytical ChemistryUniversity of ViennaViennaAustria
| | - Raza Alikhan
- Haemostasis & ThrombosisUniversity Hospital of WalesCardiffUK
| | | | - Zsuzsa Bagoly
- Faculty of Medicine, Department of Laboratory Medicine, Division of Clinical Laboratory Sciences and ELKH‐DE Neurodegenerative and Cerebrovascular Research GroupUniversity of DebrecenDebrecenHungary
| | | | - Elena Barbon
- San Raffaele Telethon Institute for Gene TherapyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Christopher D. Barrett
- Division of Acute Care Surgery and Surgical Critical Care, Department of SurgeryUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Koch Institute, Center for Precision Cancer MedicineMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
- Division of Surgical Critical Care, Department of Surgery, Boston University Medical CenterBoston University School of MedicineBostonMassachusettsUSA
| | | | | | - Wee Shian Chan
- University of British ColumbiaVancouverBritish ColumbiaCanada
| | - Moniek de Maat
- Department of HematologyErasmus MCRotterdamThe Netherlands
| | - Kerstin de Wit
- Queen’s University and McMaster UniversityKingstonONCanada
| | | | - Martin H. Ellis
- Hematology Institute and Blood Bank, Meir Medical Center and Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Renee Eslick
- Haematology DepartmentCanberra HospitalGarranAustralian Capital TerritoryAustralia
| | - Hongxia Fu
- Division of Hematology, Department of MedicineUniversity of WashingtonSeattleWashingtonUSA
| | | | | | - Frederikus A. Klok
- Department of Medicine – Thrombosis and HemostasisLeiden University Medical CenterLeidenThe Netherlands
| | - Riten Kumar
- Dana Farber/Boston Children’s Cancer and Blood Disorders CenterBostonMassachusettsUSA
| | | | - Rustem I. Litvinov
- Department of Cell and Developmental BiologyUniversity of Pennsylvania School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Nigel Mackman
- UNC Blood Research Center, Division of Hematology, Department of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | | | - Matthew D. Neal
- Trauma and Transfusion Medicine Research Center, Department of SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - William A. E. Parker
- Cardiovascular Research Unit, Northern General HospitalUniversity of SheffieldSheffieldUK
| | - Roger J. S. Preston
- Irish Centre for Vascular Biology, Department of Pharmacy & Biomolecular SciencesRoyal College of Surgeons in IrelandDublin 2Ireland
| | | | - Alireza R. Rezaie
- Cardiovascular Biology Research ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Lara N. Roberts
- King’s Thrombosis Centre, Department of Haematological MedicineKing’s College Hospital NHS Foundation TrustLondonUK
| | - Bianca Rocca
- Department of Safety and Bioethics, Section of PharmacologyCatholic University School of MedicineRomeItaly
| | - Susan Shapiro
- Oxford University Hospitals NHS Foundation TrustOxfordUK
- Radcliffe Department of MedicineOxford UniversityOxfordUK
| | - Deborah M. Siegal
- Ottawa Hospital Research Institute and University of OttawaOttawaOntarioCanada
| | - Lirlândia P. Sousa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de FarmáciaUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Katsue Suzuki‐Inoue
- Department of Clinical and Laboratory MedicineUniversity of YamanashiYamanashiJapan
| | - Tahira Zafar
- Frontier Medical CollegeAbbotabadPakistan
- Hemophilia Treatment CenterRawalpindiPakistan
| | - Jiaxi Zhou
- Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| |
Collapse
|
35
|
Margraf A, Perretti M. Immune Cell Plasticity in Inflammation: Insights into Description and Regulation of Immune Cell Phenotypes. Cells 2022; 11:cells11111824. [PMID: 35681519 PMCID: PMC9180515 DOI: 10.3390/cells11111824] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 05/28/2022] [Accepted: 05/30/2022] [Indexed: 02/01/2023] Open
Abstract
Inflammation is a life-saving immune reaction occurring in response to invading pathogens. Nonetheless, inflammation can also occur in an uncontrolled, unrestricted manner, leading to chronic disease and organ damage. Mechanisms triggering an inflammatory response, hindering such a response, or leading to its resolution are well-studied but so far insufficiently elucidated with regard to precise therapeutic interventions. Notably, as an immune reaction evolves, requirements and environments for immune cells change, and thus cellular phenotypes adapt and shift, leading to the appearance of distinct cellular subpopulations with new functional features. In this article, we aim to highlight properties of, and overarching regulatory factors involved in, the occurrence of immune cell phenotypes with a special focus on neutrophils, macrophages and platelets. Additionally, we point out implications for both diagnostics and therapeutics in inflammation research.
Collapse
|
36
|
Yu X, Zheng Q, He Y, Yu D, Chang G, Chen C, Bi L, Lv J, Zhao M, Lin X, Zhu L. Associations of Gut Microbiota and Fatty Metabolism With Immune Thrombocytopenia. Front Med (Lausanne) 2022; 9:810612. [PMID: 35665326 PMCID: PMC9160917 DOI: 10.3389/fmed.2022.810612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 04/27/2022] [Indexed: 11/30/2022] Open
Abstract
Objective To determine whether gut microbiota, fatty metabolism and cytokines were associated with immune thrombocytopenia (ITP). Methods In total, 29 preliminarily diagnosed ITP patients and 33 healthy volunteers were enrolled. Fecal bacterial were analyzed based on 16S rRNA sequencing. Plasma cytokines and motabolites were analyzed using flow cytometry and liquid chromatography-mass spectrometry (LC-MS), respectively. Results Bacteroides, Phascolarctobacterium, and Lactobacillus were enriched at the genus level in ITP patients, while Ruminococcaceae UCG-002, Eubacterium coprostanoligeues, Megamonas, and Lachnospiraceae NC2004 were depleted. At the phylum level, the relative abundance of Proteobacteria and Chloroflexi increased in ITP patients, while Firmicutes, Actinobacteria, and the Firmicutes/Bacteroidetes ratio decreased. Plasma levels of 5-hydroxyeicosatetraenoic acid (5-HETE), 6-trans-12-epi-leukotriene B4 (6t,12e-LTB4), and resolvin D2 (RvD2) were upregulated, and stachydrine, dowicide A, dodecanoylcarnitine were downregulated in ITP patients. Furthermore, RvD2 is positively correlated with order Bacteroidetes VC2.1 Bac22, 5-HETE is positively correlated with genus Azospirillum, and 6t,12e-LTB4 is positively correlated with genus Cupriavidus. In addition, stachydrine is positively correlated with family Planococcaceae, dowicide A is positively correlated with class MVP-15, and dodecanoylcarnitine is positively correlated with order WCHB1-41. Plasma levels of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) were upregulated in ITP patients. Conclusion Our study revealed a relationship between microbiota and fatty metabolism in ITP. Gut microbiota may participate in the pathogenesis of ITP through affecting cytokine secretion, interfering with fatty metabolism.
Collapse
Affiliation(s)
- Xiaomin Yu
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qingyun Zheng
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Yun He
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Dandan Yu
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guolin Chang
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Cheng Chen
- Department of Hematopathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Laixi Bi
- Department of Hematopathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jia Lv
- Department of Pathology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Misheng Zhao
- Department of Clinical Laboratory, Wenzhou People’s Hospital, Wenzhou, China
- *Correspondence: Misheng Zhao,
| | - Xiangyang Lin
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Xiangyang Lin,
| | - Liqing Zhu
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Liqing Zhu,
| |
Collapse
|
37
|
Tyagi T, Jain K, Gu SX, Qiu M, Gu VW, Melchinger H, Rinder H, Martin KA, Gardiner EE, Lee AI, Ho Tang W, Hwa J. A guide to molecular and functional investigations of platelets to bridge basic and clinical sciences. NATURE CARDIOVASCULAR RESEARCH 2022; 1:223-237. [PMID: 37502132 PMCID: PMC10373053 DOI: 10.1038/s44161-022-00021-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 01/17/2022] [Indexed: 07/29/2023]
Abstract
Platelets have been shown to be associated with pathophysiological process beyond thrombosis, demonstrating critical additional roles in homeostatic processes, such as immune regulation, and vascular remodeling. Platelets themselves can have multiple functional states and can communicate and regulate other cells including immune cells and vascular smooth muscle cells, to serve such diverse functions. Although traditional platelet functional assays are informative and reliable, they are limited in their ability to unravel platelet phenotypic heterogeneity and interactions. Developments in methods such as electron microscopy, flow cytometry, mass spectrometry, and 'omics' studies, have led to new insights. In this Review, we focus on advances in platelet biology and function, with an emphasis on current and promising methodologies. We also discuss technical and biological challenges in platelet investigations. Using coronavirus disease 2019 (COVID-19) as an example, we further describe the translational relevance of these approaches and the possible 'bench-to-bedside' utility in patient diagnosis and care.
Collapse
Affiliation(s)
- Tarun Tyagi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Kanika Jain
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Sean X Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
- Department of Laboratory Medicine, Yale University School of Medicine, Yale New Haven Hospital, New Haven, CT, USA
| | - Miaoyun Qiu
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Vivian W Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Hannah Melchinger
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Henry Rinder
- Department of Laboratory Medicine, Yale University School of Medicine, Yale New Haven Hospital, New Haven, CT, USA
| | - Kathleen A Martin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Elizabeth E Gardiner
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Alfred I Lee
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Wai Ho Tang
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
38
|
CRACking the Molecular Regulatory Mechanism of SOCE during Platelet Activation in Thrombo-Occlusive Diseases. Cells 2022; 11:cells11040619. [PMID: 35203269 PMCID: PMC8870035 DOI: 10.3390/cells11040619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/31/2022] [Accepted: 02/09/2022] [Indexed: 11/16/2022] Open
Abstract
Thrombo-occlusive diseases such as myocardial infarction, ischemic stroke and deep vein thrombosis with subsequent pulmonary embolism still represent a major health burden worldwide. Besides the cells of the vasculature or other hematopoietic cells, platelets are primarily responsible for the development and progression of an occluding thrombus. The activation and function of platelets crucially depend on free cytosolic calcium (Ca2+) as second messenger, which modulates platelet secretion, aggregation and thrombus formation. Ca2+ is elevated upon platelet activation by release of Ca2+ from intracellular stores thus triggering of the subsequent store-operated Ca2+ entry (SOCE), which is facilitated by Ca2+ release-activated channels (CRACs). In general, CRACs are assembled by the pore-forming unit Orai in the plasma membrane and the Ca2+-sensing stromal interaction molecule (STIM) in the endoplasmic reticulum after the depletion of internal Ca2+ stores. In the last few years, there is a growing body of the literature demonstrating the importance of STIM and Orai-mediated mechanism in thrombo-occlusive disorders. Thus, this review provides an overview of the recent understanding of STIM and Orai signaling in platelet function and its implication in the development and progression of ischemic thrombo-occlusive disorders. Moreover, potential pharmacological implications of STIM and Orai signaling in platelets are anticipated and discussed in the end.
Collapse
|
39
|
Schoeny H, Rampler E, Binh Chu D, Schoeberl A, Galvez L, Blaukopf M, Kosma P, Koellensperger G. Achieving Absolute Molar Lipid Concentrations: A Phospholipidomics Cross-Validation Study. Anal Chem 2022; 94:1618-1625. [PMID: 35025205 PMCID: PMC8792901 DOI: 10.1021/acs.analchem.1c03743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/05/2022] [Indexed: 01/28/2023]
Abstract
Standardization is essential in lipidomics and part of a huge community effort. However, with the still ongoing lack of reference materials, benchmarking quantification is hampered. Here, we propose traceable lipid class quantification as an important layer for the validation of quantitative lipidomics workflows. 31P nuclear magnetic resonance (NMR) and inductively coupled plasma (ICP)-mass spectrometry (MS) can use certified species-unspecific standards to validate shotgun or liquid chromatography (LC)-MS-based lipidomics approaches. We further introduce a novel lipid class quantification strategy based on lipid class separation and mass spectrometry using an all ion fragmentation (AIF) approach. Class-specific fragments, measured over a mass range typical for the lipid classes, are integrated to assess the lipid class concentration. The concept proved particularly interesting as low absolute limits of detection in the fmol range were achieved and LC-MS platforms are widely used in the field of lipidomics, while the accessibility of NMR and ICP-MS is limited. Using completely independent calibration strategies, the introduced validation scheme comprised the quantitative assessment of the complete phospholipid sub-ome, next to the individual lipid classes. Komagataella phaffii served as a prime example, showcasing mass balances and supporting the value of benchmarks for quantification at the lipid species level.
Collapse
Affiliation(s)
- Harald Schoeny
- Department
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
| | - Evelyn Rampler
- Department
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
- Vienna
Metabolomics Center (VIME), University of
Vienna, Althanstraße
14, 1090 Vienna, Austria
- Chemistry
Meets Microbiology, Althanstraße
14, 1090 Vienna, Austria
| | - Dinh Binh Chu
- Department
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
- School
of Chemical Engineering, Hanoi University
of Science and Technology, 1 Dai Co Viet, Hai Ba Trung, Hanoi 100000, Vietnam
| | - Anna Schoeberl
- Department
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
| | - Luis Galvez
- Department
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
| | - Markus Blaukopf
- Department
of Chemistry, University of Natural Resources
and Life Sciences Vienna, 1190 Vienna, Austria
| | - Paul Kosma
- Department
of Chemistry, University of Natural Resources
and Life Sciences Vienna, 1190 Vienna, Austria
| | - Gunda Koellensperger
- Department
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
- Vienna
Metabolomics Center (VIME), University of
Vienna, Althanstraße
14, 1090 Vienna, Austria
- Chemistry
Meets Microbiology, Althanstraße
14, 1090 Vienna, Austria
| |
Collapse
|
40
|
Turnbull RE, Sander KN, Turnbull J, Barrett DA, Goodall AH. Profiling oxylipins released from human platelets activated through the GPVI collagen receptor. Prostaglandins Other Lipid Mediat 2021; 158:106607. [PMID: 34942378 DOI: 10.1016/j.prostaglandins.2021.106607] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022]
Abstract
In addition to haemostasis, platelets are involved in pathological processes, often driven by material released upon activation. Interaction between collagen and glycoprotein VI (GPVI) is a primary platelet stimulus that liberates arachidonic acid and linoleic acid from membrane phospholipids. These are oxidised by cyclooxygenase-1 (COX-1) and 12-lipoxygenase (12-LOX) to eicosanoids and other oxylipins with various biological properties. Using liquid chromatography-tandem mass spectrometry we found that GPVI-stimulated platelets released significant levels of ten oxylipins; the well documented TxA2 and 12-HETE, PGD2 and PGE2, as well as 8-, 9-, 11-, and 15-HETE, 9- and 13-HODE.1 Levels of oxylipins released from washed platelets mirrored those from platelets stimulated in the presence of plasma, indicating generation from intracellular, rather than exogenous AA/LA. Inhibition of COX-1 with aspirin, as expected, completely abolished production of TxA2 and PGD/E2, but also significantly inhibited the release of 11-HETE (89 ± 3%) and 9-HODE (74 ± 6%), and reduced 15-HETE and 13-HODE by ∼33 %. Inhibition of 12-LOX by either esculetin or ML355 inhibited the release of all oxylipins apart from 15-HETE. These findings suggest routes to modify the production of bioactive molecules released by activated platelets.
Collapse
Affiliation(s)
- Robert E Turnbull
- Department of Cardiovascular Sciences, University of Leicester and NIHR Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, UK
| | - Katrin N Sander
- Centre for Analytical Bioscience, Advanced Materials and Healthcare Division, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - James Turnbull
- Centre for Analytical Bioscience, Advanced Materials and Healthcare Division, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - David A Barrett
- Centre for Analytical Bioscience, Advanced Materials and Healthcare Division, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Alison H Goodall
- Department of Cardiovascular Sciences, University of Leicester and NIHR Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, UK.
| |
Collapse
|
41
|
Platelet ACKR3/CXCR7 Favors Anti-Platelet Lipids over an Atherothrombotic Lipidome and Regulates Thrombo-inflammation. Blood 2021; 139:1722-1742. [PMID: 34905596 DOI: 10.1182/blood.2021013097] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 11/30/2021] [Indexed: 11/20/2022] Open
Abstract
Platelet ACKR3/CXCR7 surface expression is enhanced and influences prognosis in coronary artery disease-(CAD) patients, who exhibit a distinct atherothrombotic platelet lipidome. Current investigation validates the potential of ACKR3/CXCR7 in regulating thrombo-inflammatory response, through its impact on the platelet lipidome. CAD patients-(n=230) with enhanced platelet-ACKR3/CXCR7 expression exhibited reduced aggregation. Pharmacological CXCR7-agonist-(VUF11207) significantly reduced pro-thrombotic platelet response in blood from ACS patients-(n=11) ex vivo. CXCR7-agonist administration reduced thrombotic functions and thrombo-inflammatory platelet-leukocyte interactions post myocardial infarction-(MI) and arterial injury in vivo. ACKR3/CXCR7-ligation did not affect surface availability of GPIbα, GPV, GPVI, GPIX, αv-integrin, β3-integrin, coagulation profile-(APTT, PT), bleeding time, plasma-dependent thrombin generation-(thrombinoscopy) or clot formation-(thromboelastography), but counteracted activation-induced phosphatidylserine exposure and procoagulant platelet-assisted thrombin generation. Targeted-(micro-UHPLC-ESI-QTrap-MS/MS) and untargeted-(UHPLC-ESI-QTOF-MS/MS) lipidomics analysis revealed that ACKR3/CXCR7-ligation favored generation of anti-thrombotic lipids-(dihomo-γ-linolenic acid-DGLA, 12-hydroxyeicosatrienoic acid-12-HETrE) over cyclooxygenase-COX-1-(thromboxane-TxA2), or 12-lipoxygenase-LOX-(12-HETE) metabolized pro-thrombotic, and phospholipase derived atherogenic-(lysophosphatidylcholine-LPC) lipids, in healthy subjects and CAD patients, contrary to anti-platelet therapy. Through 12-HETrE, ACKR3/CXCR7-ligation coordinated with Gαs-coupled prostacyclin receptor-(IP) to trigger cAMP-PKA mediated platelet inhibition. ACKR3/CXCR7-ligation reduced generation of lipid agonists-(arachidonic acid-AA,TxA2), lipid signaling intermediates-(lyophosphatidylinositol-LPI, diacylglycerol-DG), which affected calcium mobilization, intracellular signaling, consequently platelet interaction with physiological matrices and thrombo-inflammatory secretion-(IL1β,IFN-γ,TGF-β,IL-8), emphasizing its functional dichotomy from pro-thrombotic CXCR4. Moreover, CXCR7-agonist regulated heparin-induced thrombocytopenia-(HIT)-sera/IgG-induced platelet and neutrophil activation, heparin induced platelet aggregation-(HIPA), generation of COX-1-(TxA2), 12-LOX-(12-HETE) derived thrombo-inflammatory lipids, platelet-neutrophil aggregate formation, and thrombo-inflammatory secretion (sCD40L, IL-1β, IFN-γ, TNF-α, sP-selectin, IL-8, tissue factor-TF) ex vivo. Therefore, ACKR3/CXCR7 may offer a novel therapeutic strategy in acute/chronic thrombo-inflammation exaggerated cardiovascular pathologies, and CAD.
Collapse
|
42
|
Sweedo A, Wise LM, Roka-Moiia Y, Arce FT, Saavedra SS, Sheriff J, Bluestein D, Slepian MJ, Purdy JG. Shear-Mediated Platelet Activation is Accompanied by Unique Alterations in Platelet Release of Lipids. Cell Mol Bioeng 2021; 14:597-612. [PMID: 34900013 PMCID: PMC8630256 DOI: 10.1007/s12195-021-00692-x] [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: 01/21/2021] [Accepted: 07/16/2021] [Indexed: 10/20/2022] Open
Abstract
INTRODUCTION Platelet activation by mechanical means such as shear stress exposure, is a vital driver of thrombotic risk in implantable blood-contacting devices used in the treatment of heart failure. Lipids are essential in platelets activation and have been studied following biochemical activation. However, little is known regarding lipid alterations occurring with mechanical shear-mediated platelet activation. METHODS Here, we determined if shear-activation of platelets induced lipidome changes that differ from those associated with biochemically-mediated platelet activation. We performed high-resolution lipidomic analysis on purified platelets from four healthy human donors. For each donor, we compared the lipidome of platelets that were non-activated or activated by shear, ADP, or thrombin treatment. RESULTS We found that shear activation altered cell-associated lipids and led to the release of lipids into the extracellular environment. Shear-activated platelets released 21 phospholipids and sphingomyelins at levels statistically higher than platelets activated by biochemical stimulation. CONCLUSIONS We conclude that shear-mediated activation of platelets alters the basal platelet lipidome. Further, these alterations differ and are unique in comparison to the lipidome of biochemically activated platelets. Many of the released phospholipids contained an arachidonic acid tail or were phosphatidylserine lipids, which have known procoagulant properties. Our findings suggest that lipids released by shear-activated platelets may contribute to altered thrombosis in patients with implanted cardiovascular therapeutic devices. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s12195-021-00692-x.
Collapse
Affiliation(s)
- Alice Sweedo
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ USA
| | - Lisa M. Wise
- Department of Immunobiology, University of Arizona, 1656 E. Mabel Street, PO Box 245221, Tucson, AZ 85724 USA
- BIO5 Institute, University of Arizona, Tucson, AZ USA
| | - Yana Roka-Moiia
- Department of Medicine, Sarver Heart Center, University of Arizona, Tucson, AZ USA
| | - Fernando Teran Arce
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ USA
- Department of Medicine, Sarver Heart Center, University of Arizona, Tucson, AZ USA
| | - S. Scott Saavedra
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ USA
- BIO5 Institute, University of Arizona, Tucson, AZ USA
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ USA
| | - Jawaad Sheriff
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY USA
| | - Danny Bluestein
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY USA
| | - Marvin J. Slepian
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ USA
- BIO5 Institute, University of Arizona, Tucson, AZ USA
- Department of Medicine, Sarver Heart Center, University of Arizona, Tucson, AZ USA
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY USA
- Department of Material Sciences and Engineering, University of Arizona, Tucson, AZ USA
| | - John G. Purdy
- Department of Immunobiology, University of Arizona, 1656 E. Mabel Street, PO Box 245221, Tucson, AZ 85724 USA
- BIO5 Institute, University of Arizona, Tucson, AZ USA
| |
Collapse
|
43
|
Borgmeyer M, Coman C, Has C, Schött HF, Li T, Westhoff P, Cheung YFH, Hoffmann N, Yuanxiang P, Behnisch T, Gomes GM, Dumenieu M, Schweizer M, Chocholoušková M, Holčapek M, Mikhaylova M, Kreutz MR, Ahrends R. Multiomics of synaptic junctions reveals altered lipid metabolism and signaling following environmental enrichment. Cell Rep 2021; 37:109797. [PMID: 34610315 DOI: 10.1016/j.celrep.2021.109797] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 04/12/2021] [Accepted: 09/15/2021] [Indexed: 12/30/2022] Open
Abstract
Membrane lipids and their metabolism have key functions in neurotransmission. Here we provide a quantitative lipid inventory of mouse and rat synaptic junctions. To this end, we developed a multiomics extraction and analysis workflow to probe the interplay of proteins and lipids in synaptic signal transduction from the same sample. Based on this workflow, we generate hypotheses about novel mechanisms underlying complex changes in synaptic connectivity elicited by environmental stimuli. As a proof of principle, this approach reveals that in mice exposed to an enriched environment, reduced endocannabinoid synthesis and signaling is linked to increased surface expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) in a subset of Cannabinoid-receptor 1 positive synapses. This mechanism regulates synaptic strength in an input-specific manner. Thus, we establish a compartment-specific multiomics workflow that is suitable to extract information from complex lipid and protein networks involved in synaptic function and plasticity.
Collapse
Affiliation(s)
- Maximilian Borgmeyer
- Leibniz Group 'Dendritic Organelles and Synaptic Function,' University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology, ZMNH, 20251 Hamburg, Germany; RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Cristina Coman
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44227 Dortmund, Germany; Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Wien, Austria
| | - Canan Has
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44227 Dortmund, Germany
| | - Hans-Frieder Schött
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44227 Dortmund, Germany
| | - Tingting Li
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44227 Dortmund, Germany
| | - Philipp Westhoff
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44227 Dortmund, Germany
| | - Yam F H Cheung
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44227 Dortmund, Germany
| | - Nils Hoffmann
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44227 Dortmund, Germany
| | - PingAn Yuanxiang
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Thomas Behnisch
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Guilherme M Gomes
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Mael Dumenieu
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Michaela Schweizer
- Morphology and Electron Microscopy, University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology, ZMNH, 20251 Hamburg, Germany
| | - Michaela Chocholoušková
- University of Pardubice, Department of Analytical Chemistry, CZ-532 10 Pardubice, Czech Republic
| | - Michal Holčapek
- University of Pardubice, Department of Analytical Chemistry, CZ-532 10 Pardubice, Czech Republic
| | - Marina Mikhaylova
- Emmy Noether Group 'Neuronal Protein Transport,' University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology, ZMNH, 20251 Hamburg, Germany; AG Optobiology, Institute for Biology, Humboldt Universität zu Berlin, 10115 Berlin, Germany
| | - Michael R Kreutz
- Leibniz Group 'Dendritic Organelles and Synaptic Function,' University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology, ZMNH, 20251 Hamburg, Germany; RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany; Center for Behavioral Brain Sciences, 30120 Magdeburg, Germany.
| | - Robert Ahrends
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44227 Dortmund, Germany; Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Wien, Austria.
| |
Collapse
|
44
|
Cebo M, Calderón Castro C, Schlotterbeck J, Gawaz M, Chatterjee M, Lämmerhofer M. Untargeted UHPLC-ESI-QTOF-MS/MS analysis with targeted feature extraction at precursor and fragment level for profiling of the platelet lipidome with ex vivo thrombin-activation. J Pharm Biomed Anal 2021; 205:114301. [PMID: 34391135 DOI: 10.1016/j.jpba.2021.114301] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 01/17/2023]
Abstract
Lipids play a major role in platelet signaling and activation. In this study, we analyzed the platelet lipidome in an untargeted manner by reversed-phase UHPLC for lipid species separation coupled to high-resolution QTOF-MS/MS in data-independent acquisition (DIA) mode with sequential window acquisition of all theoretical fragment ion mass spectra (SWATH) for compound detection. Lipid identification and peak picking was supported by the characteristic regular elution pattern of lipids differing in carbon and double bond numbers. It was primarily based on post-acquisition targeted feature extraction from the SWATH data. Multiple extracted ion chromatograms (EICs) from SWATH data of diagnostic ions on MS1 and MS2 level from both positive and negative ion mode allowed to distinguish between poorly resolved isomeric lipids based on their distinct fragment ions, which were used for relative quantification at a molecular lipid species level. It supports assay specificity for relative lipid quantitation via multiple quantifiably ions unlike to data-dependent acquisition methods which rely on precursor ions only. This approach was used to analyze human platelet samples. 457 lipids were annotated. Concentrations of lipids were estimated by stable isotope-labelled lipid class-specific internal standards as surrogate calibrants. Heatmaps of lipid concentrations in dependence on carbon and double bond numbers for the distinct lipid classes revealed a snapshot of the platelet lipidome in the resting state with lipid species distributions within classes supporting some functional interpretations. As expected, activation of the platelets by thrombin has led to significant alterations in the platelet lipidome as proven by univariate (volcano plot) and multivariate (PLS-DA) statistics. Several lipids were significantly up-regulated (lysophosphatidylinositols, oxylipins such as thromboxane B2 (TXB2), hydroxyheptadecatrienoic acid (HHT), hydroxyeicosatetraenoic acid (HETE), hydroxyoctadecadienoic acid (HODE), sphingoid-bases, (very) long chain saturated fatty acids) or down-regulated (lysophosphatidylethanolamines, polyunsaturated fatty acids, phosphatidylinositols). Several of them are well known as biomarkers of platelet activation while others may provide some further insights into pathways of platelet activation and platelet metabolism.
Collapse
Affiliation(s)
- Malgorzata Cebo
- University of Tübingen, Institute of Pharmaceutical Sciences, Pharmaceutical (Bio)Analysis, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | | | - Jörg Schlotterbeck
- University of Tübingen, Institute of Pharmaceutical Sciences, Pharmaceutical (Bio)Analysis, Auf der Morgenstelle 8, 72076, Tübingen, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Otfried-Müller-Strasse 10, 72076, Tübingen, Germany
| | - Madhumita Chatterjee
- Department of Cardiology and Angiology, University Hospital Tübingen, Otfried-Müller-Strasse 10, 72076, Tübingen, Germany
| | - Michael Lämmerhofer
- University of Tübingen, Institute of Pharmaceutical Sciences, Pharmaceutical (Bio)Analysis, Auf der Morgenstelle 8, 72076, Tübingen, Germany.
| |
Collapse
|
45
|
Harm T, Bild A, Dittrich K, Goldschmied A, Nestele J, Chatterjee M, Fu X, Kolb K, Castor T, Borst O, Geisler T, Rath D, LäMmerhofer M, Gawaz M. Acute coronary syndrome is associated with a substantial change in the platelet lipidome. Cardiovasc Res 2021; 118:1904-1916. [PMID: 34323932 DOI: 10.1093/cvr/cvab238] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/21/2021] [Indexed: 11/14/2022] Open
Abstract
AIMS Platelets play a key role in the pathophysiology of coronary artery disease (CAD) and patients with enhanced platelet activation are at increased risk to develop adverse cardiovascular events. Beyond reliable cardiovascular risk factors such as dyslipoproteinaemia, significant changes of platelet lipids occur in patients with CAD. In this study, we investigate the platelet lipidome by untargeted liquid chromatography-mass spectrometry, highlighting significant changes between acute coronary syndrome (ACS) and chronic coronary syndrome (CCS) patients. Additionally, we classify the platelet lipidome, spotlighting specific glycerophospholipids as key players in ACS patients. Furthermore, we examine the impact of significantly altered lipids in ACS on platelet-dependent thrombus formation and aggregation. METHODS AND RESULTS In this consecutive study, we characterized the platelet lipidome in a CAD cohort (n = 139) and showed significant changes of lipids between patients with ACS and CCS. We found that among 928 lipids, 7 platelet glycerophospholipids were significantly up-regulated in ACS, whereas 25 lipids were down-regulated compared to CCS. The most prominent up-regulated lipid in ACS, PC18:0 (PC 10:0-8:0), promoted platelet activation and ex vivo platelet-dependent thrombus formation. CONCLUSIONS Our results reveal that the platelet lipidome is altered in ACS and up-regulated lipids embody primarily glycerophospholipids. Alterations of the platelet lipidome, especially of medium chain lipids, may play a role in the pathophysiology of ACS.
Collapse
Affiliation(s)
- Tobias Harm
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Alexander Bild
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Kristina Dittrich
- Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Andreas Goldschmied
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Jeremy Nestele
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Madhumita Chatterjee
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Xiaoqing Fu
- Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Kyra Kolb
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Tatsiana Castor
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Tobias Geisler
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Dominik Rath
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Michael LäMmerhofer
- Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| |
Collapse
|
46
|
Sas KM, Lin J, Wang CH, Zhang H, Saha J, Rajendiran TM, Soni T, Nair V, Eichinger F, Kretzler M, Brosius FC, Michailidis G, Pennathur S. Renin-angiotensin system inhibition reverses the altered triacylglycerol metabolic network in diabetic kidney disease. Metabolomics 2021; 17:65. [PMID: 34219205 PMCID: PMC8312633 DOI: 10.1007/s11306-021-01816-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/24/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Dyslipidemia is a significant risk factor for progression of diabetic kidney disease (DKD). Determining the changes in individual lipids and lipid networks across a spectrum of DKD severity may identify lipids that are pathogenic to DKD progression. METHODS We performed untargeted lipidomic analysis of kidney cortex tissue from diabetic db/db and db/db eNOS-/- mice along with non-diabetic littermate controls. A subset of mice were treated with the renin-angiotensin system (RAS) inhibitors, lisinopril and losartan, which improves the DKD phenotype in the db/db eNOS-/- mouse model. RESULTS Of the three independent variables in this study, diabetes had the largest impact on overall lipid levels in the kidney cortex, while eNOS expression and RAS inhibition had smaller impacts on kidney lipid levels. Kidney lipid network architecture, particularly of networks involving glycerolipids such as triacylglycerols, was substantially disrupted by worsening kidney disease in the db/db eNOS-/- mice compared to the db/db mice, a feature that was reversed with RAS inhibition. This was associated with decreased expression of the stearoyl-CoA desaturases, Scd1 and Scd2, with RAS inhibition. CONCLUSIONS In addition to the known salutary effect of RAS inhibition on DKD progression, our results suggest a previously unrecognized role for RAS inhibition on the kidney triacylglycerol lipid metabolic network.
Collapse
Affiliation(s)
- Kelli M Sas
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 5309 Brehm Center, 1000 Wall St., Ann Arbor, Michigan, 48105, USA
| | - Jiahe Lin
- Department of Statistics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Chih-Hong Wang
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 5309 Brehm Center, 1000 Wall St., Ann Arbor, Michigan, 48105, USA
| | - Hongyu Zhang
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 5309 Brehm Center, 1000 Wall St., Ann Arbor, Michigan, 48105, USA
| | - Jharna Saha
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 5309 Brehm Center, 1000 Wall St., Ann Arbor, Michigan, 48105, USA
| | - Thekkelnaycke M Rajendiran
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, 48109, USA
- Michigan Regional Comprehensive Metabolomics Resource Core, Ann Arbor, Michigan, 48105, USA
| | - Tanu Soni
- Michigan Regional Comprehensive Metabolomics Resource Core, Ann Arbor, Michigan, 48105, USA
| | - Viji Nair
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 5309 Brehm Center, 1000 Wall St., Ann Arbor, Michigan, 48105, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Felix Eichinger
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 5309 Brehm Center, 1000 Wall St., Ann Arbor, Michigan, 48105, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 5309 Brehm Center, 1000 Wall St., Ann Arbor, Michigan, 48105, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Frank C Brosius
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 5309 Brehm Center, 1000 Wall St., Ann Arbor, Michigan, 48105, USA
- Division of Nephrology, Department of Medicine, University of Arizona, Tucson, Arizona, 85724, USA
| | - George Michailidis
- Department of Statistics and Computer and Information Sciences, University of Florida, Gainesville, Florida, 32611, USA
| | - Subramaniam Pennathur
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 5309 Brehm Center, 1000 Wall St., Ann Arbor, Michigan, 48105, USA.
- Michigan Regional Comprehensive Metabolomics Resource Core, Ann Arbor, Michigan, 48105, USA.
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, 48109, USA.
| |
Collapse
|
47
|
Manke MC, Geue S, Coman C, Peng B, Kollotzek F, Münzer P, Walker B, Huber SM, Rath D, Sickmann A, Stegner D, Duerschmied D, Lang F, Nieswandt B, Gawaz M, Ahrends R, Borst O. ANXA7 Regulates Platelet Lipid Metabolism and Ca 2+ Release in Arterial Thrombosis. Circ Res 2021; 129:494-507. [PMID: 34176316 DOI: 10.1161/circresaha.121.319207] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Mailin-Christin Manke
- Department of Cardiology, Angiology and Cardiovascular Medicine (M.-C.M., S.G., F.K., P.M., B.W., D.R., M.G., O.B.), University of Tübingen, Germany.,DFG Heisenberg Group Thrombocardiology (M.-C.M., F.K., P.M., O.B.)
| | - Sascha Geue
- Department of Cardiology, Angiology and Cardiovascular Medicine (M.-C.M., S.G., F.K., P.M., B.W., D.R., M.G., O.B.), University of Tübingen, Germany
| | - Cristina Coman
- Department of Analytical Chemistry, University of Vienna, Austria (C.C., R.A.)
| | - Bing Peng
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden (B.P.).,Leibniz-Institut für Analytische Wissenschaften - ISAS, Dortmund, Germany (B.P., A.S., R.A.)
| | - Ferdinand Kollotzek
- Department of Cardiology, Angiology and Cardiovascular Medicine (M.-C.M., S.G., F.K., P.M., B.W., D.R., M.G., O.B.), University of Tübingen, Germany.,DFG Heisenberg Group Thrombocardiology (M.-C.M., F.K., P.M., O.B.)
| | - Patrick Münzer
- Department of Cardiology, Angiology and Cardiovascular Medicine (M.-C.M., S.G., F.K., P.M., B.W., D.R., M.G., O.B.), University of Tübingen, Germany.,DFG Heisenberg Group Thrombocardiology (M.-C.M., F.K., P.M., O.B.)
| | - Britta Walker
- Department of Cardiology, Angiology and Cardiovascular Medicine (M.-C.M., S.G., F.K., P.M., B.W., D.R., M.G., O.B.), University of Tübingen, Germany
| | - Stephan M Huber
- Department of Radiation Oncology (S.M.H.), University of Tübingen, Germany
| | - Dominik Rath
- Department of Cardiology, Angiology and Cardiovascular Medicine (M.-C.M., S.G., F.K., P.M., B.W., D.R., M.G., O.B.), University of Tübingen, Germany
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften - ISAS, Dortmund, Germany (B.P., A.S., R.A.)
| | - David Stegner
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Germany (D.S., B.N.)
| | - Daniel Duerschmied
- Heart Center, Faculty of Medicine, University of Freiburg, Germany (D.D.)
| | - Florian Lang
- Department of Physiology (F.L.), University of Tübingen, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Germany (D.S., B.N.)
| | - Meinrad Gawaz
- Department of Cardiology, Angiology and Cardiovascular Medicine (M.-C.M., S.G., F.K., P.M., B.W., D.R., M.G., O.B.), University of Tübingen, Germany
| | - Robert Ahrends
- Department of Analytical Chemistry, University of Vienna, Austria (C.C., R.A.).,Leibniz-Institut für Analytische Wissenschaften - ISAS, Dortmund, Germany (B.P., A.S., R.A.)
| | - Oliver Borst
- Department of Cardiology, Angiology and Cardiovascular Medicine (M.-C.M., S.G., F.K., P.M., B.W., D.R., M.G., O.B.), University of Tübingen, Germany.,DFG Heisenberg Group Thrombocardiology (M.-C.M., F.K., P.M., O.B.)
| |
Collapse
|
48
|
Chandrakanthan M, Nguyen TQ, Hasan Z, Muralidharan S, Vu TM, Li AWL, Le UTN, Thi Thuy Ha H, Baik SH, Tan SH, Foo JC, Wenk MR, Cazenave-Gassiot A, Torta F, Ong WY, Chan MYY, Nguyen LN. Deletion of Mfsd2b impairs thrombotic functions of platelets. Nat Commun 2021; 12:2286. [PMID: 33863882 PMCID: PMC8052357 DOI: 10.1038/s41467-021-22642-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 03/23/2021] [Indexed: 11/09/2022] Open
Abstract
We recently discovered that Mfsd2b, which is the S1P exporter found in blood cells. Here, we report that Mfsd2b is critical for the release of all S1P species in both resting and activated platelets. We show that resting platelets store S1P in the cytoplasm. After activation, this S1P pool is delivered to the plasma membrane, where Mfsd2b is predominantly localized for export. Employing knockout mice of Mfsd2b, we reveal that platelets contribute a minor amount of plasma S1P. Nevertheless, Mfsd2b deletion in whole body or platelets impairs platelet morphology and functions. In particular, Mfsd2b knockout mice show significantly reduced thrombus formation. We show that loss of Mfsd2b affects intrinsic platelet functions as part of remarkable sphingolipid accumulation. These findings indicate that accumulation of sphingolipids including S1P by deletion of Mfsd2b strongly impairs platelet functions, which suggests that the transporter may be a target for the prevention of thrombotic disorders.
Collapse
Affiliation(s)
- Madhuvanthi Chandrakanthan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Toan Quoc Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zafrul Hasan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sneha Muralidharan
- Department of Medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Thiet Minh Vu
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City, Vietnam
| | - Aaron Wei Liang Li
- Department of Medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Uyen Thanh Nha Le
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hoa Thi Thuy Ha
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sang-Ha Baik
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sock Hwee Tan
- Department of Medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Juat Chin Foo
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Markus R Wenk
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Amaury Cazenave-Gassiot
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Federico Torta
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Wei Yi Ong
- Department of Anatomy, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mark Yan Yee Chan
- Department of Medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Long N Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore. .,Cardiovascular Disease Research (CVD) Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Immunology Program Research Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
49
|
Influence of Antiplatelet Agents on the Lipid Composition of Platelet Plasma Membrane: A Lipidomics Approach with Ticagrelor and Its Active Metabolite. Int J Mol Sci 2021; 22:ijms22031432. [PMID: 33572690 PMCID: PMC7866994 DOI: 10.3390/ijms22031432] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/21/2021] [Accepted: 01/23/2021] [Indexed: 12/18/2022] Open
Abstract
Lipids contained in the plasma membrane of platelets play an important role in platelet function. Modifications in the lipid composition can fluidify or rigidify the environment around embedded receptors, in order to facilitate the access of the receptor by the drug. However, data concerning the lipid composition of platelet plasma membrane need to be updated. In addition, data on the impact of drugs on plasma membrane composition, in particular antiplatelet agents, remain sparse. After isolation of platelet plasma membrane, we assessed, using lipidomics, the effect of ticagrelor, a P2Y12 antagonist, and its active metabolite on the lipid composition of these plasma membranes. We describe the exact lipid composition of plasma membrane, including all sub-species. Ticagrelor and its active metabolite significantly increased cholesterol and phosphatidylcholine ether with short saturated acyl chains 16:0/16:0, and decreased phosphatidylcholine, suggesting overall rigidification of the membrane. Furthermore, ticagrelor and its active metabolite decreased some arachidonylated plasmalogens, suggesting a decrease in availability of arachidonic acid from the membrane phospholipids for synthesis of biologically active mediators. To conclude, ticagrelor and its active metabolite seem to influence the lipid environment of receptors embedded in the lipid bilayer and modify the behavior of the plasma membrane.
Collapse
|
50
|
Claus RA, Graeler MH. Sphingolipidomics in Translational Sepsis Research-Biomedical Considerations and Perspectives. Front Med (Lausanne) 2021; 7:616578. [PMID: 33553212 PMCID: PMC7854573 DOI: 10.3389/fmed.2020.616578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022] Open
Abstract
Scientific Background: Sphingolipids are a highly diverse group of lipids with respect to physicochemical properties controlling either structure, distribution, or function, all of them regulating cellular response in health and disease. Mass spectrometry, on the other hand, is an analytical technique characterizing ionized molecules or fragments thereof by mass-to-charge ratios, which has been prosperingly developed for rapid and reliable qualitative and quantitative identification of lipid species. Parallel to best performance of in-depth chromatographical separation of lipid classes, preconditions of precise quantitation of unique molecular species by preprocessing of biological samples have to be fulfilled. As a consequence, “lipid profiles” across model systems and human individuals, esp. complex (clinical) samples, have become eminent over the last couple of years due to sensitivity, specificity, and discriminatory capability. Therefore, it is significance to consider the entire experimental strategy from sample collection and preparation, data acquisition, analysis, and interpretation. Areas Covered: In this review, we outline considerations with clinical (i.e., human) samples with special emphasis on sample handling, specific physicochemical properties, target measurements, and resulting profiling of sphingolipids in biomedicine and translational research to maximize sensitivity and specificity as well as to provide robust and reproducible results. A brief commentary is also provided regarding new insights of “clinical sphingolipidomics” in translational sepsis research. Expert Opinion: The role of mass spectrometry of sphingolipids and related species (“sphingolipidomics”) to investigate cellular and compartment-specific response to stress, e.g., in generalized infection and sepsis, is on the rise and the ability to integrate multiple datasets from diverse classes of biomolecules by mass spectrometry measurements and metabolomics will be crucial to fostering our understanding of human health as well as response to disease and treatment.
Collapse
Affiliation(s)
- Ralf A Claus
- Department for Anesthesiology and Intensive Care Medicine, Sepsis Research, Jena University Hospital, Jena, Germany
| | - Markus H Graeler
- Department for Anesthesiology and Intensive Care Medicine, Sepsis Research, Jena University Hospital, Jena, Germany.,Center for Sepsis Care & Control, Jena University Hospital, Jena, Germany.,Center for Molecular Biomedicine (CMB), Jena University Hospital, Jena, Germany
| |
Collapse
|