1
|
Carvalho LDA, Alves VS, Coutinho-Silva R, Savio LEB. G protein-coupled purinergic P2Y receptors in infectious diseases. Pharmacol Ther 2025; 267:108796. [PMID: 39814144 DOI: 10.1016/j.pharmthera.2025.108796] [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: 09/09/2024] [Revised: 10/24/2024] [Accepted: 01/03/2025] [Indexed: 01/18/2025]
Abstract
The purinergic P2Y receptors comprise eight G-coupled receptor (GPCR) subtypes already identified (P2Y1, P2Y2, P2Y4, P2Y6, P2Y11, P2Y12-14). P2Y receptor physiological agonists are extracellular purine and pyrimidine nucleotides such as ATP (Adenosine triphosphate), ADP (Adenosine diphosphate), UTP (Uridine triphosphate), UDP (Uridine diphosphate), and UDP-glucose. These receptors are expressed in almost all cells. P2Y receptors are found in immune cells, such as macrophages, neutrophils, mast cells, dendritic cells, and lymphocytes. P2Y receptors play essential roles in inflammation and are involved in several cell processes, including efferocytosis, phagocytosis, chemotaxis, degranulation, killing pathogens, cytokine production, and platelet aggregation. These processes underpin immune responses against pathogens. Therefore, here we discuss P2Y receptor pharmacology and mechanisms triggered by the activation of these receptors in virus, bacteria, and parasite infections. In addition, we highlight the therapeutical potential of P2Y receptors for developing new pharmacological strategies to modulate inflammation and disease outcomes in pathogen infections.
Collapse
Affiliation(s)
- Letícia de Almeida Carvalho
- Laboratório de Neuroimunologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vinícius Santos Alves
- Laboratório de Neuroimunologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Robson Coutinho-Silva
- Laboratório de Imunofisiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Luiz Eduardo Baggio Savio
- Laboratório de Neuroimunologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
2
|
Sophocleous RA, Curtis SJ, Curtis BL, Ooi L, Sluyter R. P2Y 1 and P2Y 12 Receptors Mediate Aggregation of Dog and Cat Platelets: A Comparison to Human Platelets. Int J Mol Sci 2025; 26:1206. [PMID: 39940972 PMCID: PMC11818226 DOI: 10.3390/ijms26031206] [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: 12/03/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Thrombosis is one of the most prevalent and serious health issues amongst humans. A key component of thrombotic events is the activation and aggregation of platelets, of which the P2Y1 and P2Y12 receptors play a crucial role in this process. Despite a breadth of knowledge on thrombosis and its mechanisms and treatment in various disorders in humans, there is less of an understanding of the expression and exact role of these receptors in companion animals such as dogs and cats. Therefore, this study aimed to investigate P2Y1 and P2Y12 receptors on dog and cat platelets in platelet-rich plasma and compare them to human platelets. Immunoblotting revealed the presence of P2Y1 and P2Y12 receptor proteins on dog and cat platelets, although relative amounts of each receptor appeared to contrast those of human platelets, with increased amounts of P2Y1 compared to P2Y12 receptors in dogs and cats. Using a modified 384-well plate aggregation assay, designed for use with small volumes, the human P2Y1 and P2Y12 receptor agonists adenosine 5'-diphosphate and 2-methylthio-adenosine 5'-diphosphate caused aggregation of dog and cat platelets. This aggregation was near-completely inhibited by the selective P2Y12 antagonist ticagrelor. Aggregation of dog and cat platelets was partly inhibited by the human P2Y1 receptor antagonist MRS2179. The agonist and antagonist responses in dog and cat platelets were like those of human platelets. In contrast, the aggregation of dog platelets in the absence of added nucleotides was two-fold greater than that of cats and humans. This study indicates that platelets of cats and dogs possess functional P2Y1 and P2Y12 receptors that can be inhibited by human antagonists. The data presented suggest differing roles or responses of the platelet P2Y receptors in dogs and cats compared to humans but also highlight the potential of using currently available P2Y1 or P2Y12 antiplatelet drugs such as ticagrelor for the treatment of thrombosis in these companion animals.
Collapse
Affiliation(s)
- Reece A. Sophocleous
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (R.A.S.); (L.O.)
| | - Stephen J. Curtis
- Your Village Vet Balgownie, Balgownie, NSW 2519, Australia; (S.J.C.); (B.L.C.)
| | - Belinda L. Curtis
- Your Village Vet Balgownie, Balgownie, NSW 2519, Australia; (S.J.C.); (B.L.C.)
| | - Lezanne Ooi
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (R.A.S.); (L.O.)
| | - Ronald Sluyter
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (R.A.S.); (L.O.)
| |
Collapse
|
3
|
Chaudhary PK, Kim S, Kunapuli SP, Kim S. Distinct Role of GRK3 in Platelet Activation by Desensitization of G Protein-Coupled Receptors. Thromb Haemost 2024. [PMID: 39419098 DOI: 10.1055/a-2442-9031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
BACKGROUND Many platelet agonists mediate their cellular effects through G protein-coupled receptors (GPCRs) to induce platelet activation, and GPCR kinases (GRKs) have been demonstrated to have crucial roles in most GPCR functions in other cell types. Here, we investigated the functional role of GRK3 and the molecular basis for the regulation of GPCR desensitization by GRK3 in platelets. METHODS We used mice lacking GRK3 as well as β-arrestin2, which has been shown to be important in GPCR function in platelets. RESULTS Platelet aggregation and dense granule secretion induced by 2-MeSADP, U46619, thrombin, and AYPGKF were significantly potentiated in both GRK3 -/- and β-arrestin2 -/- platelets compared with wild-type (WT) platelets, whereas non-GPCR agonist collagen-induced platelet aggregation and secretion were not affected. We have previously shown that GRK6 is not involved in the regulation of Gq-coupled 5HT2A and Gz-coupled α2A adrenergic receptors. Interestingly, in contrast to GRK6, platelet aggregation induced by costimulation of serotonin and epinephrine, which activate 5-HT2A and α2A adrenergic receptors, respectively, was significantly potentiated in GRK3 -/- platelets, suggesting that GRK3 is involved in general GPCR regulation. In addition, platelet aggregation in response to the second challenge of adenosine diphosphate was restored in GRK3 -/- platelets, whereas restimulation of the agonist failed to induce aggregation in WT platelets, confirming that GRK3 contributes to general GPCR desensitization. Furthermore, 2-MeSADP- and AYPGKF-induced AKT and ERK phosphorylation were significantly potentiated in GRK3 -/- platelets. Finally, GRK3 -/- mice showed shorter tail bleeding times compared with WT, indicating that GRK3 -/- mice is more susceptible to hemostasis. CONCLUSION GRK3 plays a crucial role in the regulation of platelet activation through general GPCR desensitization in platelets.
Collapse
Affiliation(s)
- Preeti K Chaudhary
- Laboratory of Veterinary Pathology and Platelet Signaling, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Sanggu Kim
- Laboratory of Veterinary Pathology and Platelet Signaling, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Satya P Kunapuli
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Soochong Kim
- Laboratory of Veterinary Pathology and Platelet Signaling, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
4
|
Sándor L, Donka T, Baráth B, Jávor P, Jász DK, Perényi D, Babik B, Varga E, Török L, Hartmann P. Mitochondrial dysfunction in platelets from severe trauma patients - A prospective case-control study. Injury 2024; 55 Suppl 3:111481. [PMID: 39300624 DOI: 10.1016/j.injury.2024.111481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/13/2024] [Accepted: 02/25/2024] [Indexed: 09/22/2024]
Abstract
INTRODUCTION Trauma-induced coagulopathy (TIC) refers to an abnormal coagulation process, an imbalance between coagulation and fibrinolysis due to several pathological factors, such as haemorrhage and tissue injury. Platelet activation and subsequent clot formation are associated with mitochondrial activity, suggesting a possible role for mitochondria in TIC. Comprehensive studies of mitochondrial dysfunction in platelets from severe trauma patients have not yet been performed. METHODS In this prospective case-control study, patients with severe trauma (ISS≥16) had venous blood samples taken at arrival to the Emergency Unit of a Level 1 Trauma Centre. Mitochondrial functional measurements (Oxygraph-2k, Oroboros) were performed to determine oxygen consumption in different respiratory states, the H2O2 production and extramitochondrial Ca2+ movements. In addition, standard laboratory and coagulation tests, viscoelastometry (ClotPro) and aggregometry (Multiplate) were performed. Measurements data were compared with age and sex matched healthy control patients. RESULTS Severe trauma patients (n = 113) with a median age of 38 years (IQR, 20-51), a median ISS of 28 (IQR, 20-48) met our inclusion criteria. Oxidative phosphorylation in platelet mitochondria from severe trauma patients significantly decreased compared to controls (34.7 ± 8.8 pmol/s/mL vs. 48.0 ± 19.7 pmol/s/mL). The mitochondrial H2O2 production significantly increased and greater endogenous Ca2+ release was found in the polytrauma group. Consistent with these results, clotting time (CT) increased while maximum clot firmness (MCF) decreased with the EX-test and FIB-test in severe trauma samples. Multiplate aggregometry showed significantly decreased ADP-test (38 ± 12 AUC vs. 112 ± 14 AUC) and ASPI test (78 ± 22 AUC vs. 84 ± 28 AUC) also tended to decrease in mitochondria of polytrauma patients as compared with controls. Significant strong correlation has been demonstrated between mitochondrial OxPhos and MCF while it was negatively correlated with ISS (R2=0.448, P˂0.05), INR, CT and lactate level of patients. CONCLUSIONS The present study revealed that severe trauma is associated with platelet mitochondrial dysfunction resulting in reduced ATP synthesis and impaired extramitochondrial Ca2+ movement. These factors are required for platelet activation, recruitment and clot stability likely thus, platelet mitochondrial dysfunction contributes to the development of TIC.
Collapse
Affiliation(s)
- Lilla Sándor
- Department of Traumatology, University of Szeged, Semmelweis str 6., Szeged, 6725, Hungary
| | - Tibor Donka
- National Academy of Scientist Education, Pacsirta str 31., Szeged, 6724, Hungary
| | - Bálint Baráth
- Department of Traumatology, University of Szeged, Semmelweis str 6., Szeged, 6725, Hungary
| | - Péter Jávor
- Department of Traumatology, University of Szeged, Semmelweis str 6., Szeged, 6725, Hungary
| | - Dávid Kurszán Jász
- National Academy of Scientist Education, Pacsirta str 31., Szeged, 6724, Hungary
| | - Domonkos Perényi
- National Academy of Scientist Education, Pacsirta str 31., Szeged, 6724, Hungary
| | - Barna Babik
- Department of Anaesthesiology and Intensive Therapy, University of Szeged, Semmelweis str 6., Szeged, 6725, Hungary
| | - Endre Varga
- Department of Traumatology, University of Szeged, Semmelweis str 6., Szeged, 6725, Hungary
| | - László Török
- Department of Traumatology, University of Szeged, Semmelweis str 6., Szeged, 6725, Hungary; Department of Sports Medicine, University of Szeged, Semmelweis str 6., Szeged, 6725, Hungary
| | - Petra Hartmann
- Department of Traumatology, University of Szeged, Semmelweis str 6., Szeged, 6725, Hungary.
| |
Collapse
|
5
|
Han JH, Yoon I, Jeon HJ. Microfluidic System-Based Quantitative Analysis of Platelet Function through Speckle Size Measurement. Biomolecules 2024; 14:612. [PMID: 38927016 PMCID: PMC11201690 DOI: 10.3390/biom14060612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/18/2024] [Accepted: 05/19/2024] [Indexed: 06/28/2024] Open
Abstract
Platelets play essential roles in the formation of blood clots by clumping with coagulation factors at the site of vascular injury to stop bleeding; therefore, a reduction in the platelet number or disorder in their function causes bleeding risk. In our research, we developed a method to assess platelet aggregation using an optical approach within a microfluidic chip's channel by evaluating the size of laser speckles. These speckles, associated with slowed blood flow in the microfluidic channel, had a baseline size of 28.54 ± 0.72 µm in whole blood. Removing platelets from the sample led to a notable decrease in speckle size to 27.04 ± 1.23 µm. Moreover, the addition of an ADP-containing agonist, which activates platelets, resulted in an increased speckle size of 32.89 ± 1.69 µm. This finding may provide a simple optical method via microfluidics that could be utilized to assess platelet functionality in diagnosing bleeding disorders and potentially in monitoring therapies that target platelets.
Collapse
Affiliation(s)
- Jong Hyeok Han
- Department of Mechanical and Biomedical Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
- Department of Smart Health Science and Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Inkwon Yoon
- Department of Mechanical and Biomedical Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
- Department of Smart Health Science and Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hee-Jae Jeon
- Department of Mechanical and Biomedical Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
- Department of Smart Health Science and Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
6
|
Pan D, Ladds G, Rahman KM, Pitchford SC. Exploring bias in platelet P2Y 1 signalling: Host defence versus haemostasis. Br J Pharmacol 2024; 181:580-592. [PMID: 37442808 PMCID: PMC10952580 DOI: 10.1111/bph.16191] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/21/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Platelets are necessary for maintaining haemostasis. Separately, platelets are important for the propagation of inflammation during the host immune response against infection. The activation of platelets also causes inappropriate inflammation in various disease pathologies, often in the absence of changes to haemostasis. The separate functions of platelets during inflammation compared with haemostasis are therefore varied and this will be reflected in distinct pathways of activation. The activation of platelets by the nucleotide adenosine diphosphate (ADP) acting on P2Y1 and P2Y12 receptors is important for the development of platelet thrombi during haemostasis. However, P2Y1 stimulation of platelets is also important during the inflammatory response and paradoxically in scenarios where no changes to haemostasis and platelet aggregation occur. In these events, Rho-GTPase signalling, rather than the canonical phospholipase Cβ (PLCβ) signalling pathway, is necessary. We describe our current understanding of these differences, reflecting on recent advances in knowledge of P2Y1 structure, and the possibility of biased agonism occurring from activation via other endogenous nucleotides compared with ADP. Knowledge arising from these different pathways of P2Y1 stimulation of platelets during inflammation compared with haemostasis may help therapeutic control of platelet function during inflammation or infection, while preserving essential haemostasis. LINKED ARTICLES: This article is part of a themed issue on Platelet purinergic receptor and non-thrombotic disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.4/issuetoc.
Collapse
Affiliation(s)
- Dingxin Pan
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical ScienceKing's College LondonLondonUK
| | - Graham Ladds
- Department of PharmacologyUniversity of CambridgeCambridgeUK
| | - Khondaker Miraz Rahman
- Chemical Biology Group, Institute of Pharmaceutical ScienceKing's College LondonLondonUK
| | - Simon C. Pitchford
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical ScienceKing's College LondonLondonUK
| |
Collapse
|
7
|
Dangelmaier C, Kunapuli SP. Evidence for a PI3-kinase independent pathway in the regulation of Rap1b activation downstream of the P2Y12 receptor in platelets. Platelets 2022; 33:1301-1306. [PMID: 35514261 PMCID: PMC9547944 DOI: 10.1080/09537104.2022.2071855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/26/2022] [Accepted: 04/14/2022] [Indexed: 10/18/2022]
Abstract
Platelet activation by adenosine diphosphate (ADP) is mediated through two G-protein-coupled receptors, P2Y1 and P2Y12, which signal through Gq and Gi, respectively. P2Y1 stimulation leads to phospholipase C activation and an increase in cytosolic calcium necessary for CalDAG-GEF1 activation. Engagement of P2Y12 inhibits adenylate cyclase, which reduces cAMP, and activation of PI3-kinase, which inhibits RASA3 resulting in sustained activated Rap1b. In this study we activated human platelets with 2-MeSADP in the presence of LY294002, a PI3-kinase inhibitor, AR-C69931MX, a P2Y12 antagonist or MRS2179, a P2Y1 antagonist. We measured the phosphorylation of Akt on Ser473 as an indicator of PI3-kinase activity. As previously shown, LY294002 and ARC69931MX abolished 2MeSADP-induced Akt phosphorylation. MRS2179 reduced ADP-induced Akt phosphorylation but did not abolish it. Rap1b activation, however, was only reduced, but not ablated, using LY294002 and was completely inhibited by ARC69931MX or MRS2179. Furthermore, 2MeSADP-induced Rap1b activation was abolished in either P2Y1 or P2Y12 null platelets. These data suggest that ADP-induced Rap1b activation requires both P2Y1 and P2Y12. In addition, although stimulation of P2Y12 results in PI3-kinase activation leading to Akt phosphorylation and Rap1b activation, Rap1b activation can occur independently of PI3-kinase downstream of P2Y12. Thus, we propose that the P2Y12 receptor can regulate Rap1b, possibly through RASA3, in a pathway independent of PI3-kinase.
Collapse
Affiliation(s)
- Carol Dangelmaier
- Sol Sherry Thrombosis Research Center and the Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Satya P Kunapuli
- Sol Sherry Thrombosis Research Center and the Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
8
|
Liu Z, Avila C, Malone LE, Gnatenko DV, Sheriff J, Zhu W, Bahou WF. Age-restricted functional and developmental differences of neonatal platelets. J Thromb Haemost 2022; 20:2632-2645. [PMID: 35962592 PMCID: PMC10953828 DOI: 10.1111/jth.15847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Developmental ontogeny of neonatal thrombopoiesis retains characteristics that are distinct from adults although molecular mechanisms remain unestablished. METHODS We applied multiparameter quantitative platelet responses with integrated ribosome profiling/transcriptomic studies to better define gene/pathway perturbations regulating the neonatal-to-adult transition. A bioinformatics pipeline was developed to identify stable, neonatal-restricted platelet biomarkers for clinical application. RESULTS Cord blood (CB) platelets retained the capacity for linear agonist-receptor coupling linked to phosphatidylserine (PS) exposure and α-granule release, although a restricted block in cross-agonist activation pathways was evident. Functional immaturity of synergistic signaling pathways was due to younger ontogenetic age and singular underdevelopment of the protein secretory gene network, with reciprocal expansion of developmental pathways (E2F, G2M checkpoint, c-Myc) important for megakaryocytopoiesis. Genetic perturbations regulating vesicle transport and fusion (TOM1L1, VAMP3, SNAP23, and DNM1L) and PS exposure and procoagulant activity (CLCN3) were the most significant, providing a molecular explanation for globally attenuated responses. Integrated transcriptomic and ribosomal footprints identified highly abundant (ribosome-protected) DEFA3 (encoding human defensin neutrophil peptide 3) and HBG1 as stable biomarkers of neonatal thrombopoiesis. Studies comparing CB- or adult-derived megakaryocytopoiesis confirmed inducible and abundant DEFA3 antigenic expression in CB megakaryocytes, ~3.5-fold greater than in leukocytes (the most abundant source in humans). An initial feasibility cohort of at-risk pregnancies manifested by maternal/fetal hemorrhage (chimerism) were applied for detection and validation of platelet HBG1 and DEFA3 as neonatal thrombopoiesis markers, most consistent for HBG1, which displayed gestational age-dependent expression. CONCLUSIONS These studies establish an ontogenetically divergent stage of neonatal thrombopoiesis, and provide initial feasibility studies to track disordered fetal-to-adult megakaryocytopoiesis in vivo.
Collapse
Affiliation(s)
- Zhaoyan Liu
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York, USA
| | - Cecilia Avila
- Department of Obstetrics and Gynecology, Stony Brook University, Stony Brook, New York, USA
| | - Lisa E. Malone
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Dmitri V. Gnatenko
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Scientific Review, National Institutes of Health, Bethesda, Maryland, USA
| | - Jawaad Sheriff
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Wei Zhu
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York, USA
| | - Wadie F. Bahou
- Department of Obstetrics and Gynecology, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
9
|
Coumarin Derivatives Inhibit ADP-Induced Platelet Activation and Aggregation. Molecules 2022; 27:molecules27134054. [PMID: 35807298 PMCID: PMC9268609 DOI: 10.3390/molecules27134054] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/16/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
Coumarin was first discovered in Tonka bean and then widely in other plants. Coumarin has an anticoagulant effect, and its derivative, warfarin, is a vitamin K analogue that inhibits the synthesis of clotting factors and is more widely used in the clinical treatment of endovascular embolism. At present, many artificial chemical synthesis methods can be used to modify the structure of coumarin to develop many effective drugs with low toxicity. In this study, we investigated the effects of six coumarin derivatives on the platelet aggregation induced by adenosine diphosphate (ADP). We found that the six coumarin derivatives inhibited the active form of GPIIb/IIIa on platelets and hence inhibit platelet aggregation. We found that 7-hydroxy-3-phenyl 4H-chromen-4-one (7-hydroxyflavone) had the most severe effect. In addition, we further analyzed the downstream signal transduction of the ADP receptor, including the release of calcium ions and the regulation of cAMP, which were inhibited by the six coumarin derivatives selected in this study. These results suggest that coumarin derivatives inhibit coagulation by inhibiting the synthesis of coagulation factors and they may also inhibit platelet aggregation.
Collapse
|
10
|
Xu Y, Yu G, Nie R, Wu Z. Microfluidic systems toward blood hemostasis monitoring and thrombosis diagnosis: From design principles to micro/nano fabrication technologies. VIEW 2022. [DOI: 10.1002/viw.20200183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Yi Xu
- Soft Intelligence Lab State Key Laboratory of Digital Manufacturing Equipment and Technology School of Mechanical Science and Engineering Huazhong University of Science and Technology Wuhan China
| | - Guang Yu
- Experimental Medicine Center Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Ruqiong Nie
- Department of Cardiology Sun Yat‐Sen Memorial Hospital Sun Yat‐Sen University Guangzhou China
| | - Zhigang Wu
- Soft Intelligence Lab State Key Laboratory of Digital Manufacturing Equipment and Technology School of Mechanical Science and Engineering Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
11
|
La Rose AM, Bazioti V, Hoogerland JA, Svendsen AF, Groenen AG, van Faassen M, Rutten MGS, Kloosterhuis NJ, Dethmers-Ausema B, Nijland JH, Mithieux G, Rajas F, Kuipers F, Lukens MV, Soehnlein O, Oosterveer MH, Westerterp M. Hepatocyte-specific glucose-6-phosphatase deficiency disturbs platelet aggregation and decreases blood monocytes upon fasting-induced hypoglycemia. Mol Metab 2021; 53:101265. [PMID: 34091064 PMCID: PMC8243524 DOI: 10.1016/j.molmet.2021.101265] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/20/2021] [Accepted: 05/31/2021] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Glycogen storage disease type 1a (GSD Ia) is a rare inherited metabolic disorder caused by mutations in the glucose-6-phosphatase (G6PC1) gene. When untreated, GSD Ia leads to severe fasting-induced hypoglycemia. Although current intensive dietary management aims to prevent hypoglycemia, patients still experience hypoglycemic events. Poor glycemic control in GSD Ia is associated with hypertriglyceridemia, hepatocellular adenoma and carcinoma, and also with an increased bleeding tendency of unknown origin. METHODS To evaluate the effect of glycemic control on leukocyte levels and coagulation in GSD Ia, we employed hepatocyte-specific G6pc1 deficient (L-G6pc-/-) mice under fed or fasted conditions, to match good or poor glycemic control in GSD Ia, respectively. RESULTS We found that fasting-induced hypoglycemia in L-G6pc-/- mice decreased blood leukocytes, specifically proinflammatory Ly6Chi monocytes, compared to controls. Refeeding reversed this decrease. The decrease in Ly6Chi monocytes was accompanied by an increase in plasma corticosterone levels and was prevented by the glucocorticoid receptor antagonist mifepristone. Further, fasting-induced hypoglycemia in L-G6pc-/- mice prolonged bleeding time in the tail vein bleeding assay, with reversal by refeeding. This could not be explained by changes in coagulation factors V, VII, or VIII, or von Willebrand factor. While the prothrombin and activated partial thromboplastin time as well as total platelet counts were not affected by fasting-induced hypoglycemia in L-G6pc-/- mice, ADP-induced platelet aggregation was disturbed. CONCLUSIONS These studies reveal a relationship between fasting-induced hypoglycemia, decreased blood monocytes, and disturbed platelet aggregation in L-G6pc-/- mice. While disturbed platelet aggregation likely accounts for the bleeding phenotype in GSD Ia, elevated plasma corticosterone decreases the levels of proinflammatory monocytes. These studies highlight the necessity of maintaining good glycemic control in GSD Ia.
Collapse
Affiliation(s)
- Anouk M La Rose
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Venetia Bazioti
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Joanne A Hoogerland
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Arthur F Svendsen
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anouk G Groenen
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Martijn van Faassen
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Martijn G S Rutten
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Niels J Kloosterhuis
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Bertien Dethmers-Ausema
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - J Hendrik Nijland
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Gilles Mithieux
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Fabienne Rajas
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Folkert Kuipers
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Michaël V Lukens
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Oliver Soehnlein
- Institute for Experimental Pathology (ExPat), Center for Molecular Biology of Inflammation (ZBME), University of Münster, Münster, Germany; Department of Physiology and Pharmacology (FyFa), Karolinska Institutet, Stockholm, Sweden
| | - Maaike H Oosterveer
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marit Westerterp
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
12
|
Ablat N, Ablimit M, Abudoukadier A, Kadeer B, Yang L. Investigating the hemostatic effect of medicinal plant Arnebia euchroma (Royle) I.M.Johnst extract in a mouse model. JOURNAL OF ETHNOPHARMACOLOGY 2021; 278:114306. [PMID: 34111535 DOI: 10.1016/j.jep.2021.114306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 05/26/2021] [Accepted: 06/03/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Arnebia euchroma (Royle) I.M.Johnst (AE) has been reported to be a potentially useful medicinal herb for the treatment of several circulatory diseases in traditional Chinese medicine. It shows effects such as "cooling of the blood," promotion of blood circulation, detoxification, and rash clearance. AIM OF THE STUDY To explore the hemostatic effect of the ethyl acetate extract of AE in mice. MATERIALS AND METHODS In this study, we explored the effects of AE on bleeding time, blood coagulation time, platelet count, and blood coagulation parameters in normal Kunming mice. Different doses of the AE extract (5, 10, and 20 g kg-1·day-1) were administered to mice for 14 days. Sodium carboxymethyl cellulose (CMC-Na at 0.5%) and Yunnan Baiyao (0.8 g kg-1·day-1) were administered as negative and positive control treatments, respectively. Bleeding time, blood coagulation time, platelet count, blood platelet aggregation, blood platelet adhesion to fibrinogen, platelet factor 4 (PF-4) secretions from blood platelets, and blood coagulation parameters including prothrombin time (PT), activated partial thromboplastin time (aPTT), thrombin time (TT), and fibrinogen (FIB) levels were measured on day 15 of administration. RESULTS Bleeding and blood coagulation time were significantly lower and TT was shorter in the AE extract-treated groups than in the control groups. Furthermore, FIB levels and platelet count were higher, whereas blood platelet aggregation, blood platelet adhesion to fibrinogen, and PF-4 secretion from blood platelets were more obvious in the AE extract-treated groups than in the control group. However, no significant differences were detected for PT and aPTT between the extract-treated and control groups. CONCLUSIONS The ethyl acetate extract of AE showed potential hemostasis effects in mice by shortening the bleeding and coagulation time. In addition, the extract increased platelet count and induced blood platelet aggregation, blood platelet adhesion to fibrinogen, PF-4 secretion from blood platelets, and FIB level, while it shortened TT.
Collapse
Affiliation(s)
- Nuramatjan Ablat
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China; School of Medicine, Huanghuai University, Henan Province, 463000, China.
| | - Mihray Ablimit
- Xinjiang Uygur Autonomous Region Shache County Dunbag Township Health Center, 844700, China.
| | - Abudoureheman Abudoukadier
- Department of Cardiology, Urumqi City Friendship Hospital, Xinjiang Uygur Autonomous Region, Urumqi, 830049, China.
| | - Buhaiqiemu Kadeer
- Department of Gynecology, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, 830000, China.
| | - Lei Yang
- Key Laboratory of Cardiovascular and Cerebrovascular Diseases, School of Medicine, Huanghuai University, Henan Province, 463000, China.
| |
Collapse
|
13
|
Praetorius H. The bacteria and the host: a story of purinergic signaling in urinary tract infections. Am J Physiol Cell Physiol 2021; 321:C134-C146. [PMID: 33979212 DOI: 10.1152/ajpcell.00054.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The local environment forces a selection of bacteria that might invade the urinary tract, allowing only the most virulent to access the kidney. Quite similar to the diet in setting the stage for the gut microbiome, renal function determines the conditions for bacteria-host interaction in the urinary tract. In the kidney, the term local environment or microenvironment is completely justified because the environment literally changes within a few micrometers. The precise composition of the urine is a function of the epithelium lining the microdomain, and the microenvironment in the kidney shows more variation in the content of nutrients, ion composition, osmolality, and pH than any other site of bacteria-host interaction. This review will cover some of the aspects of bacterial-host interaction in this unique setting and how uropathogenic bacteria can alter the condition for bacteria-host interaction. There will be a particular focus on the recent findings regarding how bacteria specifically trigger host paracrine signaling, via release of extracellular ATP and activation of P2 purinergic receptors. These finding will be discussed from the perspective of severe urinary tract infections, including pyelonephritis and urosepsis.
Collapse
|
14
|
Huang B, Qian Y, Xie S, Ye X, Chen H, Chen Z, Zhang L, Xu J, Hu H, Ma S, Héroux P, Wang D, Shen HM, Wu Y, Xia D. Ticagrelor inhibits the NLRP3 inflammasome to protect against inflammatory disease independent of the P2Y 12 signaling pathway. Cell Mol Immunol 2021; 18:1278-1289. [PMID: 32523112 PMCID: PMC8093290 DOI: 10.1038/s41423-020-0444-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 04/12/2020] [Indexed: 12/16/2022] Open
Abstract
Ticagrelor is the first reversibly binding oral P2Y12 receptor antagonist to inhibit platelet activation and has been approved by the Food and Drug Administration for the treatment of coronary artery disease. At present, the other pharmacological functions of ticagrelor remain poorly understood. The NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome plays a critical role in the innate immune system, but its excessive activation also contributes to the pathogenesis of complex diseases. In this study, we systematically examined the effects of ticagrelor on the NLRP3 inflammasome and found that ticagrelor inhibits NLRP3 inflammasome activation in macrophages independent of its classic inhibitory effect on the P2Y12 signaling pathway. Further mechanistic studies demonstrate that ticagrelor attenuates the oligomerization of apoptosis-associated speck-like protein containing a CARD (ASC) by blocking chloride efflux, an effect achieved through the degradation of chloride intracellular channel proteins (CLICs) and blockade of the translocation of CLICs to the plasma membrane. Moreover, experiments on lipopolysaccharide-induced sepsis and alum-induced peritonitis in mice confirmed that ticagrelor mitigates the severity of systemic inflammation independent of P2Y12 receptor antagonism. Importantly, oral administration of ticagrelor rapidly and strongly inhibited NLRP3 inflammasome activation in peripheral blood mononuclear cells from patients with acute coronary syndrome. Overall, our study reveals a novel pharmacological function of ticagrelor in addition to its classic antiplatelet properties, which suggests that ticagrelor may serve as a potential therapeutic agent for use in NLRP3-associated diseases.
Collapse
Affiliation(s)
- Bo Huang
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yufeng Qian
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shujun Xie
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Translational Medicine Research Center, and Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xianhua Ye
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hanwen Chen
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhifeng Chen
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lihuan Zhang
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinming Xu
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hu Hu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Shenglin Ma
- Department of Oncology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Paul Héroux
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
| | - Di Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yihua Wu
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Dajing Xia
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
15
|
Alarabi AB, Karim ZA, Hinojos V, Lozano PA, Hernandez KR, Montes Ramirez JE, Ali HEA, Khasawneh FT, Alshbool FZ. The G-protein βγ subunits regulate platelet function. Life Sci 2020; 262:118481. [PMID: 32971104 DOI: 10.1016/j.lfs.2020.118481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 10/23/2022]
Abstract
AIMS G-protein coupled receptors (GPCRs) tightly regulate platelet function by interacting with various physiological agonists. An essential mediator of GPCR signaling is the G protein αβγ heterotrimers, in which the βγ subunits are central players in downstream signaling. Herein, we investigated the role of Gβγ subunits in platelet function, hemostasis and thrombogenesis. METHODS To achieve this goal, platelets from both mice and humans were employed in the context of a small molecule inhibitor of Gβγ, namely gallein. We used an aggregometer to examine aggregation and dense granules secretion. We also used flow cytometry for P-selectin and PAC1 to determine the impact of inhibiting Gβγ on α -granule secretion and αIIbβ3 activation. Clot retraction and the platelet spreading assay were used to examine Gβγ role in outside-in platelet signaling, whereas Western blot was employed to examine its role in Akt activation. Finally, we used the bleeding time assay and the FeCl3-induced carotid-artery injury thrombosis model to determine Gβγ contribution to in vivo platelet function. RESULTS We observed that gallein inhibits platelet aggregation and secretion in response to agonist stimulation, in both mouse and human platelets. Furthermore, gallein also exerted inhibitory effects on integrin αIIbβ3 activation, clot retraction, platelet spreading and Akt activation/phosphorylation. Finally, gallein's inhibitory effects manifested in vivo, as documented by its ability to modulate physiological hemostasis and delay thrombus formation. CONCLUSION Our findings demonstrate, for the first time, that Gβγ subunits directly regulate GPCR-dependent platelet function, in vitro and in vivo. Moreover, these data highlight Gβγ as a novel therapeutic target for managing thrombotic disorders.
Collapse
Affiliation(s)
- Ahmed B Alarabi
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| | - Zubair A Karim
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, USA
| | - Victoria Hinojos
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, USA
| | - Patricia A Lozano
- Department of Pharmacy Practice, Irma Lerma Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| | - Keziah R Hernandez
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, USA
| | - Jean E Montes Ramirez
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, USA
| | - Hamdy E A Ali
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| | - Fadi T Khasawneh
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| | - Fatima Z Alshbool
- Department of Pharmacy Practice, Irma Lerma Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA.
| |
Collapse
|
16
|
Characterisation of the secreted apyrase family of Heligmosomoides polygyrus. Int J Parasitol 2020; 51:39-48. [PMID: 32931780 DOI: 10.1016/j.ijpara.2020.07.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/16/2020] [Accepted: 07/21/2020] [Indexed: 12/21/2022]
Abstract
Apyrases are a recurrent feature of secretomes from numerous species of parasitic nematodes. Here we characterise the five apyrases secreted by Heligmosomoides polygyrus, a natural parasite of mice and a widely used laboratory model for intestinal nematode infection. All five enzymes are closely related to soluble calcium-activated nucleotidases described in a variety of organisms, and distinct from the CD39 family of ecto-nucleotidases. Expression is maximal in adult worms and restricted to adults and L4s. Recombinant apyrases were produced and purified from Pichia pastoris. The five enzymes showed very similar biochemical properties, with strict calcium dependence and a broad substrate specificity, catalysing the hydrolysis of all nucleoside tri- and diphosphates, with no activity against nucleoside monophosphates. Natural infection of mice provoked very low antibodies to any enzyme, but immunisation with an apyrase cocktail showed partial protection against reinfection, with reduced egg output and parasite recovery. The most likely role for nematode secreted apyrases is hydrolysis of extracellular ATP, which acts as an alarmin for cellular release of IL-33 and initiation of type 2 immunity.
Collapse
|
17
|
Prevention of P2 Receptor-Dependent Thrombocyte Activation by Pore-Forming Bacterial Toxins Improves Outcome in A Murine Model of Urosepsis. Int J Mol Sci 2020; 21:ijms21165652. [PMID: 32781764 PMCID: PMC7460651 DOI: 10.3390/ijms21165652] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 12/31/2022] Open
Abstract
Urosepsis is a potentially life-threatening, systemic reaction to uropathogenic bacteria entering the bloodstream of the host. One of the hallmarks of sepsis is early thrombocyte activation with a following fall in circulating thrombocytes as a result of intravascular aggregation and sequestering of thrombocytes in the major organs. Development of a thrombocytopenic state is associated with a poorer outcome of sepsis. Uropathogenic Escherichia coli frequently produce the pore-forming, virulence factor α-haemolysin (HlyA), of which the biological effects are mediated by ATP release and subsequent activation of P2 receptors. Thus, we speculated that inhibition of thrombocyte P2Y1 and P2Y12 receptors might ameliorate the septic response to HlyA-producing E. coli. The study combined in vitro measurements of toxin-induced thrombocyte activation assessed as increased membrane abundance of P-selectin, fibronectin and CD63 and data from in vivo murine model of sepsis-induced by HlyA-producing E. coli under infusion of P2Y1 and P2Y12 antagonists. Our data show that the P2Y1 receptor antagonist almost abolishes thrombocyte activation by pore-forming bacterial toxins. Inhibition of P2Y1, by constant infusion of MRS2500, markedly increased the survival in mice with induced sepsis. Moreover, MRS2500 partially prevented the sepsis-induced depletion of circulating thrombocytes and dampened the sepsis-associated increase in proinflammatory cytokines. In contrast, P2Y12 receptor inhibition had only a marginal effect in vivo and in vitro. Taken together, inhibition of the P2Y1 receptor gives a subtle dampening of the thrombocyte activation and the cytokine response to bacteraemia, which may explain the improved survival observed by P2Y1 receptor antagonists.
Collapse
|
18
|
Zebrafish for thrombocytopoiesis- and hemostasis-related researches and disorders. BLOOD SCIENCE 2020; 2:44-49. [PMID: 35402814 PMCID: PMC8975081 DOI: 10.1097/bs9.0000000000000043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 03/05/2020] [Indexed: 11/30/2022] Open
Abstract
Platelets play vital roles in hemostasis, inflammation, and vascular biology. Platelets are also active participants in the immune responses. As vertebrates, zebrafish have a highly conserved hematopoietic system in the developmental, cellular, functional, biochemical, and genetic levels with mammals. Thrombocytes in zebrafish are functional homologs of mammalian platelets. Here, we summarized thrombocyte development, function, and related research techniques in zebrafish, and reviewed available zebrafish models of platelet-associated disorders, including congenital amegakaryocytic thrombocytopenia, inherited thrombocytopenia, essential thrombocythemia, and blood coagulation disorders such as gray platelet syndrome. These elegant zebrafish models and methods are crucial for understanding the molecular and genetic mechanisms of thrombocyte development and function, and provide deep insights into related human disease pathophysiology and drug development.
Collapse
|
19
|
Lordan R, Tsoupras A, Zabetakis I. Platelet activation and prothrombotic mediators at the nexus of inflammation and atherosclerosis: Potential role of antiplatelet agents. Blood Rev 2020; 45:100694. [PMID: 32340775 DOI: 10.1016/j.blre.2020.100694] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 03/22/2020] [Accepted: 04/07/2020] [Indexed: 12/20/2022]
Abstract
Platelets are central to inflammation-related manifestations of cardiovascular diseases (CVD) such as atherosclerosis. Platelet-activating factor (PAF), thrombin, thromboxane A2 (TxA2), and adenosine diphosphate (ADP) are some of the key agonists of platelet activation that are at the intersection between a plethora of inflammatory pathways that modulate pro-inflammatory and coagulation processes. The aim of this article is to review the role of platelets and the relationship between their structure, function, and the interactions of their constituents in systemic inflammation and atherosclerosis. Antiplatelet therapies are discussed with a view to primary prevention of CVD by the clinical reduction of platelet reactivity and inflammation. Current antiplatelet therapies are effective in reducing cardiovascular risk but increase bleeding risk. Novel therapeutic antiplatelet approaches beyond current pharmacological modalities that do not increase the risk of bleeding require further investigation. There is potential for specifically designed nutraceuticals that may become safer alternatives to pharmacological antiplatelet agents for the primary prevention of CVD but there is serious concern over their efficacy and regulation, which requires considerably more research.
Collapse
Affiliation(s)
- Ronan Lordan
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute (HRI), University of Limerick, Limerick, Ireland; Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-5158, USA.
| | - Alexandros Tsoupras
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| | - Ioannis Zabetakis
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| |
Collapse
|
20
|
Johnsen N, Christensen MG, Hamilton AD, Praetorius H. HlyA cause platelet activation and neutrophil/ monocyte interaction during urosepsis. FASEB J 2020. [DOI: 10.1096/fasebj.2020.34.s1.05088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
21
|
Wypych D, Pomorski P. Calcium Signaling in Glioma Cells: The Role of Nucleotide Receptors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1202:67-86. [PMID: 32034709 DOI: 10.1007/978-3-030-30651-9_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Calcium signaling is probably one of the evolutionary oldest and the most common way by which the signal can be transmitted from the cell environment to the cytoplasmic calcium binding effectors. Calcium signal is fast and due to diversity of calcium binding proteins it may have a very broad effect on cell behavior. Being a crucial player in neuronal transmission it is also very important for glia physiology. It is responsible for the cross-talk between neurons and astrocytes, for microglia activation and motility. Changes in calcium signaling are also crucial for the behavior of transformed glioma cells. The present chapter summarizes molecular mechanisms of calcium signal formation present in glial cells with a strong emphasis on extracellular nucleotide-evoked signaling pathways. Some aspects of glioma C6 signaling such as the cross-talk between P2Y1 and P2Y12 nucleotide receptors in calcium signal generation will be discussed in-depth, to show complexity of machinery engaged in formation of this signal. Moreover, possible mechanisms of modulation of the calcium signal in diverse environments there will be presented herein. Finally, the possible role of calcium signal in glioma motility is also discussed. This is a very important issue, since glioma cells, contrary to the vast majority of neoplastic cells, cannot spread in the body with the bloodstream and, at least in early stages of tumor development, may expand only by means of sheer motility.
Collapse
Affiliation(s)
- Dorota Wypych
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Paweł Pomorski
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
22
|
Li J, Ge Y, Huang JX, Strømgaard K, Zhang X, Xiong XF. Heterotrimeric G Proteins as Therapeutic Targets in Drug Discovery. J Med Chem 2019; 63:5013-5030. [PMID: 31841625 DOI: 10.1021/acs.jmedchem.9b01452] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Heterotrimeric G proteins are molecular switches in GPCR signaling pathways and regulate a plethora of physiological and pathological processes. GPCRs are efficient drug targets, and more than 30% of the drugs in use target them. However, selectively targeting an individual GPCR may be undesirable in various multifactorial diseases in which multiple receptors are involved. In addition, abnormal activation or expression of G proteins is frequently associated with diseases. Furthermore, G proteins harboring mutations often result in malignant diseases. Thus, targeting G proteins instead of GPCRs might provide alternative approaches for combating these diseases. In this review, we discuss the biochemistry of heterotrimeric G proteins, describe the G protein-associated diseases, and summarize the currently known modulators that can regulate the activities of G proteins. The outlook for targeting G proteins to treat diverse diseases is also included in this manuscript.
Collapse
Affiliation(s)
- Jian Li
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, Guangdong, P. R. China
| | - Yang Ge
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, Guangdong, P. R. China
| | - Jun-Xiang Huang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, Guangdong, P. R. China
| | - Kristian Strømgaard
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Xiaolei Zhang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, Guangdong, P. R. China
| | - Xiao-Feng Xiong
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, Guangdong, P. R. China
| |
Collapse
|
23
|
Norris JW, Watson JL, Tablin F, Kozikowski TA, Knych HK. Pharmacokinetics and competitive pharmacodynamics of ADP-induced platelet activation after oral administration of clopidogrel to horses. Am J Vet Res 2019; 80:505-512. [PMID: 31034271 DOI: 10.2460/ajvr.80.5.505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To determine pharmacokinetics and pharmacodynamics after oral administration of a single dose of clopidogrel to horses. ANIMALS 6 healthy adult horses. PROCEDURES Blood samples were collected before and at various times up to 24 hours after oral administration of clopidogrel (2 mg/kg). Reactivity of platelets from each blood sample was determined by optical aggregometry and phosphorylation of vasodilator-stimulated phosphoprotein (VASP). Concentrations of clopidogrel and the clopidogrel active metabolite derivative (CAMD) were measured in each blood sample by use of liquid chromatography-tandem mass spectrometry, and pharmacokinetic parameters were determined with a noncompartmental model. RESULTS Compared with results for preadministration samples, platelet aggregation in response to 12.5μM ADP decreased significantly within 4 hours after clopidogrel administration for 5 of 6 horses. After 24 hours, platelet aggregation was identical to that measured before administration. Platelet aggregation in response to 25μM ADP was identical between samples obtained before and after administration. Phosphorylation of VASP in response to ADP (20μM) and prostaglandin E1 (3.3μM) was also unchanged by administration of clopidogrel. Time to maximum concentration of clopidogrel and CAMD was 0.54 and 0.71 hours, respectively, and calculated terminal-phase half-life of clopidogrel and CAMD was 1.81 and 0.97 hours, respectively. CONCLUSIONS AND CLINICAL RELEVANCE Clopidogrel or CAMD caused competitive inhibition of ADP-induced platelet aggregation during the first 24 hours after clopidogrel administration. Because CAMD was rapidly eliminated from horses, clopidogrel administration may be needed more frequently than in other species in which clopidogrel causes irreversible platelet inhibition. (Am J Vet Res 2019;80:505-512).
Collapse
|
24
|
P2X 1 receptor blockers reduce the number of circulating thrombocytes and the overall survival of urosepsis with haemolysin-producing Escherichia coli. Purinergic Signal 2019; 15:265-276. [PMID: 31129780 DOI: 10.1007/s11302-019-09658-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 05/06/2019] [Indexed: 01/14/2023] Open
Abstract
Urosepsis is a severe condition often caused by Escherichia coli that spontaneously have ascended the urinary tract to the kidneys causing pyelonephritis and potentially bacteraemia. The number of sepsis cases has been steadily increasing over the last decades, and there are still no specific, molecular supportive therapies for sepsis to supplement antibiotic treatment. P2X1 receptors are expressed by a number of immune cells including thrombocytes, which presently have been established as an important player in the acute immune response to bacterial infections. P2X1 receptor-deficient mice have been shown to be relatively protected against urosepsis, with markedly reduced levels of circulating proinflammatory cytokines and intravascular coagulation. However, here we show that continuous intravenous infusion with P2X1 receptor antagonist markedly accelerates development of a septic response to induced bacteraemia with uropathogenic E. coli. Mice exposed to the P2X1 receptor antagonists die very early with haematuria, substantially elevated plasma levels of proinflammatory cytokines, massive intravascular coagulation and a concomitant reduction in circulating thrombocytes. Interestingly, infusion of P2X1 receptor antagonists causes a marked acute reduction in circulating thrombocytes and a higher number of bacteria in the blood. These data support the notion that the number of functional thrombocytes is important for the acute defence against bacteria in the circulation and that the P2X1 receptor potentially could be essential for this response.
Collapse
|
25
|
Johnsen N, Hamilton ADM, Greve AS, Christensen MG, Therkildsen JR, Wehmöller J, Skals M, Praetorius HA. α-Haemolysin production, as a single factor, causes fulminant sepsis in a model of Escherichia coli-induced bacteraemia. Cell Microbiol 2019; 21:e13017. [PMID: 30761726 DOI: 10.1111/cmi.13017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 02/08/2019] [Accepted: 02/10/2019] [Indexed: 12/14/2022]
Abstract
α-Haemolysin (HlyA) from uropathogenic Escherichia coli has been demonstrated to be a significant virulence factor for ascending urinary tract infections. Once the E. coli reach the well-vascularised kidneys, there is a high risk of bacteraemia and a subsequent septic host response. Despite this, HlyA has the potential to accelerate the host response both directly and via its ability to facilitate adenosine triphosphate release from cells. It has not been settled whether HlyA aggravates bacteraemia into a septic state. To address this, we used an E. coli strain in a model of acute urosepsis that was either transfected with a plasmid containing the full HlyA operon or one with deletion in the HlyA gene. Here, we show that HlyA accelerates the host response to E. coli in the circulation. Mice exposed to HlyA-producing E. coli showed massively increased proinflammatory cytokines, a substantial fall in circulating thrombocytes, extensive haematuria, and intravascular haemolysis. This was not seen in mice exposed to either E. coli that do not secrete HlyA or vehicle controls. Consistent with the massive host response to the bacteria, the mice exposed to HlyA-producing E. coli died exceedingly early, whereas mice exposed to E. coli without HlyA production and vehicle controls survived the entire observation period. These data allow us to conclude that HlyA is a virulence factor that accelerates a state of bacteraemia into fulminant sepsis in a mouse model.
Collapse
Affiliation(s)
- Nanna Johnsen
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | | | | | | | | | - Julia Wehmöller
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Marianne Skals
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | | |
Collapse
|
26
|
Le Minh G, Peshkova AD, Andrianova IA, Weisel JW, Litvinov RI. Differential sensitivity of various markers of platelet activation with adenosine diphosphate. BIONANOSCIENCE 2019; 9:53-58. [PMID: 31534882 PMCID: PMC6750022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A number of techniques have been available to assess platelet activation, but their relative sensitivity is unknown and their usage is variable and not based on any rational criteria. Here, we compared the ability of several techniques based on morphological and biochemical markers to detect the first signs of ADP-induced platelet activation. Scanning electron microscopy of platelets was performed in parallel with flow cytometry to quantify the surface expression of P-selectin (marked by labeled anti-CD62P antibodies), active αIIbβ3-intergrin (assessed by the binding of labeled fibrinogen) and phosphatidylserine (assessed by the binding of labeled Annexin V). When expressed as a fraction of activated platelets, shape changes were the most sensitive to a low ADP concentration compared to the biochemical markers in the following order of sensitivity: morphological changes>fibrinogen binding capacity>P-selectin expression> phosphatidylserine exposure. These results suggest the greater sensitivity of platelet microscopy and the importance of its combination with flow cytometry used to detect surface expression of the molecular markers of platelet activation.
Collapse
Affiliation(s)
- Giang Le Minh
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Alina D. Peshkova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Izabella A. Andrianova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - John W. Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rustem I. Litvinov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
27
|
Cardenas EI, Gonzalez R, Breaux K, Da Q, Gutierrez BA, Ramos MA, Cardenas RA, Burns AR, Rumbaut RE, Adachi R. Munc18-2, but not Munc18-1 or Munc18-3, regulates platelet exocytosis, hemostasis, and thrombosis. J Biol Chem 2019; 294:4784-4792. [PMID: 30696774 DOI: 10.1074/jbc.ra118.006922] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/24/2019] [Indexed: 12/17/2022] Open
Abstract
Platelet degranulation, a form of regulated exocytosis, is crucial for hemostasis and thrombosis. Exocytosis in platelets is mediated by SNARE proteins, and in most mammalian cells this process is controlled by Munc18 (mammalian homolog of Caenorhabditis elegans uncoordinated gene 18) proteins. Platelets express all Munc18 paralogs (Munc18-1, -2, and -3), but their roles in platelet secretion and function have not been fully characterized. Using Munc18-1, -2, and -3 conditional knockout mice, here we deleted expression of these proteins in platelets and assessed granule exocytosis. We measured products secreted by each type of platelet granule and analyzed EM platelet profiles by design-based stereology. We observed that the removal of Munc18-2 ablates the release of alpha, dense, and lysosomal granules from platelets, but we found no exocytic role for Munc18-1 or -3 in platelets. In vitro, Munc18-2-deficient platelets exhibited defective aggregation at low doses of collagen and impaired thrombus formation under shear stress. In vivo, megakaryocyte-specific Munc18-2 conditional knockout mice had a severe hemostatic defect and prolonged arterial and venous bleeding times. They were also protected against arterial thrombosis in a chemically induced model of arterial injury. Taken together, our results indicate that Munc18-2, but not Munc18-1 or Munc18-3, is essential for regulated exocytosis in platelets and platelet participation in thrombosis and hemostasis.
Collapse
Affiliation(s)
- Eduardo I Cardenas
- From the Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030.,the Escuela de Ingenieria y Ciencias, Instituto Tecnologico de Monterrey, Monterrey, Nuevo Leon 64849, Mexico
| | - Ricardo Gonzalez
- From the Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030.,the Escuela de Ingenieria y Ciencias, Instituto Tecnologico de Monterrey, Monterrey, Nuevo Leon 64849, Mexico
| | - Keegan Breaux
- From the Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Qi Da
- the Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas 77030.,the Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| | - Berenice A Gutierrez
- From the Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030.,the Escuela de Ingenieria y Ciencias, Instituto Tecnologico de Monterrey, Monterrey, Nuevo Leon 64849, Mexico
| | - Marco A Ramos
- From the Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Rodolfo A Cardenas
- From the Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030.,the Escuela de Medicina y Ciencias de la Salud, Instituto Tecnologico de Monterrey, Monterrey, Nuevo León 64710, México, and
| | - Alan R Burns
- the College of Optometry, University of Houston, Houston, Texas 77204
| | - Rolando E Rumbaut
- the Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas 77030.,the Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| | - Roberto Adachi
- From the Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030,
| |
Collapse
|
28
|
|
29
|
|
30
|
Le Minh G, Peshkova AD, Andrianova IA, Weisel JW, Litvinov RI. Differential Sensitivity of Various Markers of Platelet Activation with Adenosine Diphosphate. BIONANOSCIENCE 2018. [DOI: 10.1007/s12668-018-0586-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
31
|
Individual variations in platelet reactivity towards ADP, epinephrine, collagen and nitric oxide, and the association to arterial function in young, healthy adults. Thromb Res 2018; 174:5-12. [PMID: 30543988 DOI: 10.1016/j.thromres.2018.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/13/2018] [Accepted: 12/05/2018] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Platelet aggregation and secretion can be induced by a large number of endogenous activators, such as collagen, adenosine diphosphate (ADP) and epinephrine. Conversely, the blood vessel endothelium constitutively release platelet inhibitors including nitric oxide (NO) and prostacyclin. NO and prostacyclin are also well-known vasodilators and contribute to alterations in local blood flow and systemic blood pressure. MATERIALS AND METHODS In this study we investigated individual variations in platelet reactivity and arterial functions including blood pressure and flow-mediated vasodilation (FMD) in 43 young, healthy individuals participating in the Lifestyle, Biomarkers and Atherosclerosis (LBA) study. Platelet aggregation and dense granule secretion were measured simultaneously by light transmission and luminescence. FMD was measured with ultrasound. RESULTS The platelet function assay showed inter-individual differences in platelet reactivity. Specifically, a sub-group of individuals had platelets with an increased response to low concentrations of ADP and epinephrine, but not collagen. When the NO-donor S-nitroso-N-acetyl-DL-penicillamine (SNAP) was combined with high doses of these platelet activators, the results indicated for sub-groups of NO-sensitive and NO-insensitive platelets. The individuals with NO-sensitive platelets in response to SNAP in combination with collagen had a higher capacity of FMD of the arteria brachialis. CONCLUSIONS Platelet reactivity towards ADP, epinephrine and NO differs between young, healthy individuals. Some individuals have a more effective response towards NO, both in the aspect of platelet inhibition ex vivo, as well as vasodilation in vivo.
Collapse
|
32
|
Tomaiuolo M, Brass LF, Stalker TJ. Regulation of Platelet Activation and Coagulation and Its Role in Vascular Injury and Arterial Thrombosis. Interv Cardiol Clin 2018; 6:1-12. [PMID: 27886814 DOI: 10.1016/j.iccl.2016.08.001] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Hemostasis requires tightly regulated interaction of the coagulation system, platelets, blood cells, and vessel wall components at a site of vascular injury. Dysregulation of this response may result in excessive bleeding if the response is impaired, and pathologic thrombosis with vessel occlusion and tissue ischemia if the response is robust. Studies have elucidated the major molecular signaling pathways responsible for platelet activation and aggregation. Antithrombotic agents targeting these pathways are in clinical use. This review summarizes research examining mechanisms by which these multiple platelet signaling pathways are integrated at a site of vascular injury to produce an optimal hemostatic response.
Collapse
Affiliation(s)
- Maurizio Tomaiuolo
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 421 Curie Boulevard, Philadelphia, PA, 19104, USA
| | - Lawrence F Brass
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 421 Curie Boulevard, Philadelphia, PA, 19104, USA
| | - Timothy J Stalker
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 421 Curie Boulevard, Philadelphia, PA, 19104, USA.
| |
Collapse
|
33
|
Targeted metabolomic approach in men with carotid plaque. PLoS One 2018; 13:e0200547. [PMID: 30011297 PMCID: PMC6047792 DOI: 10.1371/journal.pone.0200547] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/28/2018] [Indexed: 12/22/2022] Open
Abstract
Background The aim of the study was to analyse the presence of several metabolites related to atherosclerosis in the plasma of patients with unstable carotid plaque and in the plasma of healthy subjects. Materials and methods We included 20 patients who had undergone carotid endarterectomy and 20 healthy subjects as a control group. All the subjects recruited were male. We used a metabolomic approach with liquid chromatography coupled to mass spectrometry to evaluate plasma metabolite levels in the metabolic pathway involved in the progression of atherosclerotic plaque. Results We observed that circulating levels of 20-HETE were significantly higher in patients with atheroma plaque than in healthy subjects (p = 0.018). No differences were found with regard to the other metabolites analysed. We also conducted a random forest analysis and found that 20-HETE was the main differentiator in the list of selected metabolites. In addition, plasma levels of 20-HETE correlated positively with body mass index (r = 0.427, p = 0.007) and diastolic blood pressure (r = 0.365, p = 0.028). Conclusion This study confirms that of all the molecules studied only 20-HETE is related to carotid plaque. Further studies are needed to compare patients with stable carotid plaque vs. patients with unstable carotid plaque in order to confirm that 20-HETE could be a potential factor related to carotid plaque.
Collapse
|
34
|
Koessler J, Trulley VN, Bosch A, Weber K, Koessler A, Boeck M, Kobsar A. The role of agonist-induced activation and inhibition for the regulation of purinergic receptor expression in human platelets. Thromb Res 2018; 168:40-46. [PMID: 29902630 DOI: 10.1016/j.thromres.2018.05.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/09/2018] [Accepted: 05/29/2018] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Adenosine diphosphate (ADP) as physiological activator of human platelets mediates its effects via three purinergic receptors: P2Y1, P2Y12 and P2X1. The inhibition of P2Y12 is used pharmacologically to suppress aggregation underlining the physiological significance of this receptor. Since the regulation of purinergic receptor expression has not thoroughly been investigated yet, this study analyzed the content of purinergic receptors on the platelet surface membrane upon activation and inhibition. MATERIALS AND METHODS The surface expression of purinergic receptors was measured by flow cytometry using two different polyclonal antibodies as basal values and after incubation with thrombin receptor activating peptide (TRAP-6) or with inhibitors DEA/NO, MAHMA/NO or Prostaglandin E1 (PGE1). Western blot analysis was used to confirm inhibitory effects. RESULTS Both investigated antibodies revealed a significant increase of purinergic receptor expression upon TRAP-6 stimulation. The NO donors, DEA/NO and MAHMA/NO, did not influence basal or TRAP-6 stimulated values. PGE1 did not affect basal receptor expression, but diminished TRAP-6 stimulated purinergic receptor expression in a dose-dependent manner. CONCLUSIONS In summary, TRAP-6 induced platelet activation leads to an elevation of purinergic receptor expression. In contrast to other surface ligands, this effect is not suppressed by cGMP-mediated inhibition, but almost completely abrogated by enhanced cAMP-mediated signaling as induced by PGE1.
Collapse
Affiliation(s)
- Juergen Koessler
- Institute of Clinical Transfusion Medicine and Haemotherapy, University of Wuerzburg, Oberduerrbacher Straße 6, D-97080 Wuerzburg, Germany.
| | - Valerie-Noelle Trulley
- Institute of Clinical Transfusion Medicine and Haemotherapy, University of Wuerzburg, Oberduerrbacher Straße 6, D-97080 Wuerzburg, Germany.
| | - Andrea Bosch
- Institute of Clinical Transfusion Medicine and Haemotherapy, University of Wuerzburg, Oberduerrbacher Straße 6, D-97080 Wuerzburg, Germany.
| | - Katja Weber
- Institute of Clinical Transfusion Medicine and Haemotherapy, University of Wuerzburg, Oberduerrbacher Straße 6, D-97080 Wuerzburg, Germany.
| | - Angela Koessler
- Institute of Clinical Transfusion Medicine and Haemotherapy, University of Wuerzburg, Oberduerrbacher Straße 6, D-97080 Wuerzburg, Germany.
| | - Markus Boeck
- Institute of Clinical Transfusion Medicine and Haemotherapy, University of Wuerzburg, Oberduerrbacher Straße 6, D-97080 Wuerzburg, Germany.
| | - Anna Kobsar
- Institute of Clinical Transfusion Medicine and Haemotherapy, University of Wuerzburg, Oberduerrbacher Straße 6, D-97080 Wuerzburg, Germany.
| |
Collapse
|
35
|
Salem M, Tremblay A, Pelletier J, Robaye B, Sévigny J. P2Y 6 Receptors Regulate CXCL10 Expression and Secretion in Mouse Intestinal Epithelial Cells. Front Pharmacol 2018. [PMID: 29541027 PMCID: PMC5835513 DOI: 10.3389/fphar.2018.00149] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In this study, we investigated the role of extracellular nucleotides in chemokine (KC, MIP-2, MCP-1, and CXCL10) expression and secretion by murine primary intestinal epithelial cells (IECs) with a focus on P2Y6 receptors. qRT-PCR experiments showed that P2Y6 was the dominant nucleotide receptor expressed in mouse IEC. In addition, the P2Y6 ligand UDP induced expression and secretion of CXCL10. For the other studies, we took advantage of mice deficient in P2Y6 (P2ry6-/-). Similar expression levels of P2Y1, P2Y2, P2X2, P2X4, and A2A were detected in P2ry6-/- and WT IEC. Agonists of TLR3 (poly(I:C)), TLR4 (LPS), P2Y1, and P2Y2 increased the expression and secretion of CXCL10 more prominently in P2ry6-/- IEC than in WT IEC. CXCL10 expression and secretion induced by poly(I:C) in both P2ry6-/- and WT IEC were inhibited by general P2 antagonists (suramin and Reactive-Blue-2), by apyrase, and by specific antagonists of P2Y1, P2Y2, P2Y6 (only in WT), and P2X4. Neither adenosine nor an A2A antagonist had an effect on CXCL10 expression and secretion. Macrophage chemotaxis was induced by the supernatant of poly(I:C)-treated IEC which was consistent with the level of CXCL10 secreted. Finally, the non-nucleotide agonist FGF2 induced MMP9 mRNA expression also at a higher level in P2ry6-/- IEC than in WT IEC. In conclusion, extracellular nucleotides regulate CXCL10 expression and secretion by IEC. In the absence of P2Y6, these effects are modulated by other P2 receptors also present on IEC. These data suggest that the presence of P2Y6 regulates chemokine secretion and may also regulate IEC homeostasis.
Collapse
Affiliation(s)
- Mabrouka Salem
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine, Université Laval, Québec City, QC, Canada.,Centre de Recherche du CHU de Québec - Université Laval, Québec City, QC, Canada
| | - Alain Tremblay
- Centre de Recherche du CHU de Québec - Université Laval, Québec City, QC, Canada
| | - Julie Pelletier
- Centre de Recherche du CHU de Québec - Université Laval, Québec City, QC, Canada
| | - Bernard Robaye
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles, Gosselies, Belgium
| | - Jean Sévigny
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine, Université Laval, Québec City, QC, Canada.,Centre de Recherche du CHU de Québec - Université Laval, Québec City, QC, Canada
| |
Collapse
|
36
|
Bijak M, Szelenberger R, Dziedzic A, Saluk-Bijak J. Inhibitory Effect of Flavonolignans on the P2Y12 Pathway in Blood Platelets. Molecules 2018; 23:molecules23020374. [PMID: 29439388 PMCID: PMC6017715 DOI: 10.3390/molecules23020374] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/05/2018] [Accepted: 02/07/2018] [Indexed: 12/27/2022] Open
Abstract
Adenosine diphosphate (ADP) is the major platelet agonist, which is important in the shape changes, stability, and growth of the thrombus. Platelet activation by ADP is associated with the G protein-coupled receptors P2Y1 and P2Y12. The pharmacologic blockade of the P2Y12 receptor significantly reduces the risk of peripheral artery disease, myocardial infarction, ischemic stroke, and vascular death. Recent studies demonstrated the inhibition of ADP-induced blood platelet activation by three major compounds of the flavonolignans group: silybin, silychristin, and silydianin. For this reason, the aim of the current work was to verify the effects of silybin, silychristin, and silydianin on ADP-induced physiological platelets responses, as well as mechanisms of P2Y12-dependent intracellular signal transduction. We evaluated the effect of tested flavonolignans on ADP-induced blood platelets’ aggregation in platelet-rich plasma (PRP) (using light transmission aggregometry), adhesion to fibrinogen (using the static method), and the secretion of PF-4 (using the ELISA method). Additionally, using the double labeled flow cytometry method, we estimated platelet vasodilator-stimulated phosphoprotein (VASP) phosphorylation. We demonstrated a dose-dependent reduction of blood platelets’ ability to perform ADP-induced aggregation, adhere to fibrinogen, and secrete PF-4 in samples treated with flavonolignans. Additionally, we observed that all of the tested flavonolignans were able to increase VASP phosphorylation in blood platelets samples, which is correlated with P2Y12 receptor inhibition. All of these analyses show that silychristin and silybin have the strongest inhibitory effect on blood platelet activation by ADP, while silydianin also inhibits the ADP pathway, but to a lesser extent. The results obtained in this study clearly demonstrate that silybin, silychristin, and silydianin have inhibitory properties against the P2Y12 receptor and block ADP-induced blood platelet activation.
Collapse
Affiliation(s)
- Michal Bijak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland.
| | - Rafal Szelenberger
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland.
| | - Angela Dziedzic
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland.
| | - Joanna Saluk-Bijak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland.
| |
Collapse
|
37
|
Koessler J, Schwarz M, Weber K, Etzel J, Koessler A, Boeck M, Kobsar A. The role of adenosine diphosphate mediated platelet responsiveness for the stability of platelet integrity in citrated whole blood under ex vivo conditions. PLoS One 2017; 12:e0188193. [PMID: 29155852 PMCID: PMC5695795 DOI: 10.1371/journal.pone.0188193] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 11/02/2017] [Indexed: 11/19/2022] Open
Abstract
Background Platelets are important for effective hemostasis and considered to be involved in pathophysiological processes, e.g. in cardiovascular diseases. Platelets provided for research or for therapeutic use are frequently separated from citrated whole blood (WB) stored for different periods of time. Although functionally intact platelets are required, the stability of platelet integrity, e.g. adenosine diphosphate (ADP) mediated responsiveness, has never been thoroughly investigated in citrated WB under ex vivo conditions. Objectives Platelet integrity was evaluated at different time points in citrated WB units, collected from healthy donors and stored for 5 days at ambient temperature. The analysis included the measurement of activation markers, of induced light transmission aggregometry and of purinergic receptor expression or function. Inhibitory pathways were explored by determination of basal vasodilator-stimulated phosphoprotein (VASP)-phosphorylation, intracellular cyclic nucleotide levels and the content of phosphodiesterase 5A. Fresh peripheral blood (PB) samples served as controls. Results On day 5 of storage, thrombin receptor activating peptide-6 (TRAP-6) stimulated CD62P expression and fibrinogen binding were comparable to PB samples. ADP induced aggregation continuously decreased during storage. Purinergic receptor expression remained unchanged, whereas the P2Y1 activity progressively declined in contrast to preserved P2Y12 and P2X1 function. Inhibitory pathways were unaffected except for a slight elevation of VASP phosphorylation at Ser239 on day 5. Conclusion After 5 days of storage in citrated WB, platelet responsiveness to TRAP-6 is sufficiently maintained. However, ADP-mediated platelet integrity is more sensitive to deterioration, especially after storage for more than 2 days. Decreasing ADP-induced aggregation is particularly caused by the impairment of the purinergic receptor P2Y1 activity. These characteristics should be considered in the use of platelets from stored citrated WB for experimental or therapeutic issues.
Collapse
Affiliation(s)
- Juergen Koessler
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Wuerzburg, Germany
- * E-mail:
| | - Michaela Schwarz
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Katja Weber
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Julia Etzel
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Angela Koessler
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Markus Boeck
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Anna Kobsar
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
38
|
Study on the Mg-Li-Zn ternary alloy system with improved mechanical properties, good degradation performance and different responses to cells. Acta Biomater 2017; 62:418-433. [PMID: 28823717 DOI: 10.1016/j.actbio.2017.08.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/22/2017] [Accepted: 08/16/2017] [Indexed: 01/04/2023]
Abstract
Novel Mg-(3.5, 6.5wt%)Li-(0.5, 2, 4wt%)Zn ternary alloys were developed as new kinds of biodegradable metallic materials with potential for stent application. Their mechanical properties, degradation behavior, cytocompatibility and hemocompatibility were studied. These potential biomaterials showed higher ultimate tensile strength than previously reported binary Mg-Li alloys and ternary Mg-Li-X (X=Al, Y, Ce, Sc, Mn and Ag) alloys. Among the alloys studied, the Mg-3.5Li-2Zn and Mg-6.5Li-2Zn alloys exhibited comparable corrosion resistance in Hank's solution to pure magnesium and better corrosion resistance in a cell culture medium than pure magnesium. Corrosion products observed on the corroded surface were composed of Mg(OH)2, MgCO3 and Ca-free Mg/P inorganics and Ca/P inorganics. In vitro cytotoxicity assay revealed different behaviors of Human Umbilical Vein Endothelial Cells (HUVECs) and Human Aorta Vascular Smooth Muscle Cells (VSMCs) to material extracts. HUVECs showed increasing nitric oxide (NO) release and tolerable toxicity, whereas VSMCs exhibited limited decreasing viability with time. Platelet adhesion, hemolysis and coagulation tests of these Mg-Li-Zn alloys showed different degrees of activation behavior, in which the hemolysis of the Mg-3.5Li-2Zn alloy was lower than 5%. These results indicated the potential of the Mg-Li-Zn alloys as good candidate materials for cardiovascular stent applications. STATEMENT OF SIGNIFICANCE Mg-Li alloys are promising as absorbable metallic biomaterials, which however have not received significant attention since the low strength, controversial corrosion performance and the doubts in Li toxicity. The Mg-Li-Zn alloy in the present study revealed much improved mechanical properties higher than most reported binary Mg-Li and ternary Mg-Li-X alloys, with superior corrosion resistance in cell culture media. Surprisingly, the addition of Li and Zn showed increased nitric oxide release. The present study indicates good potential of Mg-Li-Zn alloy as absorbable cardiovascular stent material.
Collapse
|
39
|
Bethi Y, Shewade DG, Dutta TK, Gitanjali B. Prevalence and predictors of potential drug-drug interactions in patients of internal medicine wards of a tertiary care hospital in India. Eur J Hosp Pharm 2017; 25:317-321. [PMID: 31157049 DOI: 10.1136/ejhpharm-2017-001272] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/07/2017] [Accepted: 06/12/2017] [Indexed: 11/03/2022] Open
Abstract
Background Drug-drug interactions are a major source of adverse drug events (ADEs). Polypharmacy, age and the number of comorbid conditions are important predictors of adverse drug interactions. ADEs account for up to 5% of hospital admissions per year and an increase in the length of hospital stay. Objective To find the prevalence and predictors of potential drug-drug interactions (pDDIs) in patients admitted to the wards of an internal medicine department of a tertiary care hospital. Method Patients admitted to internal medicine wards with prescriptions having more than one drug were selected. Demographic details including age, gender, number of comorbid conditions, number of drugs prescribed and the disease for which the patient was admitted were recorded in a case record form. Interactions were checked using Micromedex DrugReax software. Results A total of 939 patients were recruited for this study based on inclusion criteria. 433 prescriptions (46%) had one or more pDDIs, with a range of 1-13 drug interactions per prescription. A total of 1395 drug interactions were found, with 866 moderate drug interactions (62%), 435 major interactions (31.1%) and 89 minor interactions (6.3%). During the study period only three contraindicated drug combinations (0.2%) were recorded. A significant association (p<0.01) was found between the number of pDDIs and predictors, age and number of drugs. Conclusion A total of 433 prescriptions (46%) had one or more pDDIs. Older patients and those prescribed >6 drugs are at major risk for occurrence of pDDIs. Moderate severity interactions were the highest number followed by major severity interactions.
Collapse
Affiliation(s)
- Yugandhar Bethi
- Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Deepak Gopal Shewade
- Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Tarun Kumar Dutta
- Department of Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Batmanabane Gitanjali
- Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| |
Collapse
|
40
|
Nonhuman primate model of polytraumatic hemorrhagic shock recapitulates early platelet dysfunction observed following severe injury in humans. J Trauma Acute Care Surg 2017; 82:461-469. [PMID: 28225526 DOI: 10.1097/ta.0000000000001343] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Platelet dysfunction has been described as an early component of trauma-induced coagulopathy. The platelet component of trauma-induced coagulopathy remains to be fully elucidated and translatable animal models are required to facilitate mechanistic investigations. We sought to determine if the early platelet dysfunction described in trauma patients could be recapitulated in a nonhuman primate model of polytraumatic hemorrhagic shock. METHODS Twenty-four male rhesus macaques weighting 7 to 14 kg were subjected to 60 minutes (min) of severe pressure-targeted controlled hemorrhagic shock (HS) with and without other injuries. After 60 min, resuscitation with 0.9% NaCl and whole blood was initiated. Platelet counts and platelet aggregation assays were performed at baseline (BSLN), end of shock (EOS; T = 60 min), end of resuscitation (EOR; T = 180 min), and T = 360 min on overall cohort. Results are reported as mean ± standard deviation (SD) or median (interquartile range). Statistical analysis was conducted using Spearmen correlation, one-way analysis of variance, two-way repeated-measures analysis of variance, paired t-test or Wilcoxon nonparametric test, with p < 0.05 considered significant. RESULTS Platelet count in all injury cohorts decreased over time, but no animals developed thrombocytopenia. Correlations were observed between platelet aggregation and platelet count for all agonists: adenosine diphosphate, thrombin recognition-activating peptide-6, collagen, and arachidonic acid. Overall, compared to BSLN, platelet aggregation decreased for all agonist at EOS, EOR, and T = 360 min. When normalized to platelet count, platelet aggregation in response to agonist thrombin recognition-activating peptide-6 demonstrated no change from BSLN at subsequent time points. Aggregation to adenosine diphosphate was significantly less at EOR but not EOS or T = 360 min compared to BSLN. Platelet aggregation to collagen and arachidonic acid was not significantly different at EOS compared to BSLN but was significantly less at EOR and T = 360 min. CONCLUSION Nonhuman primates manifest early platelet dysfunction in response to polytraumatic hemorrhagic shock, consistent with that reported in severely injured human patients. Nonhuman primate models potentially are translationally valuable for understanding the mechanisms and pathophysiology of trauma-induced platelet dysfunction.
Collapse
|
41
|
Qin C, Zhou J, Gao Y, Lai W, Yang C, Cai Y, Chen S, Du C. Critical Role of P2Y12 Receptor in Regulation of Th17 Differentiation and Experimental Autoimmune Encephalomyelitis Pathogenesis. THE JOURNAL OF IMMUNOLOGY 2017; 199:72-81. [DOI: 10.4049/jimmunol.1601549] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 04/24/2017] [Indexed: 11/19/2022]
|
42
|
Liu M, Yao M, Wang H, Xu L, Zheng Y, Huang B, Ni H, Xu S, Zhou X, Lian Q. P2Y 12 receptor-mediated activation of spinal microglia and p38MAPK pathway contribute to cancer-induced bone pain. J Pain Res 2017; 10:417-426. [PMID: 28243146 PMCID: PMC5317303 DOI: 10.2147/jpr.s124326] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Background Cancer-induced bone pain (CIBP) is one of the most challenging clinical problems due to a lack of understanding the mechanisms. Recent evidence has demonstrated that activation of microglial G-protein-coupled P2Y12 receptor (P2Y12R) and proinflammatory cytokine production play an important role in neuropathic pain generation and maintenance. However, whether P2Y12R is involved in CIBP remains unknown. Methods The purpose of this study was to investigate the role of P2Y12R in CIBP and its molecular mechanisms. Using the bone cancer model inoculated with Walker 256 tumor cells into the left tibia of Sprague Dawley rat, we blocked spinal P2Y12R through intrathecal administration of its selective antagonist MRS2395 (400 pmol/µL, 15 µL). Results We found that not only the ionized calcium-binding adapter molecule 1 (Iba-1)-positive microglia in the ipsilateral spinal cord but also mechanical allodynia was significantly inhibited. Furthermore, it decreased the phosphorylation of p38 mitogen-activated protein kinase (p38 MAPK) and the production of proinflammatory cytokines interleukin-1β (IL-1β) and interleukin-6 (IL-6), whereas it increased tumor necrosis factor-α (TNF-α). Conclusion Taken together, our present results suggest that microglial P2Y12R in the spinal cord may contribute to CIBP by the activation of spinal microglia and p38MAPK pathway, thus identifying a potential therapeutic target for the treatment of CIBP.
Collapse
Affiliation(s)
- Mingjuan Liu
- Department of Anesthesiology and Pain Medicine, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing
| | - Ming Yao
- Department of Anesthesiology and Pain Medicine, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing; Department of Anesthesiology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Hanqi Wang
- Department of Anesthesiology and Pain Medicine, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing
| | - Longsheng Xu
- Department of Anesthesiology and Pain Medicine, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing
| | - Ying Zheng
- Department of Anesthesiology and Pain Medicine, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing
| | - Bing Huang
- Department of Anesthesiology and Pain Medicine, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing
| | - Huadong Ni
- Department of Anesthesiology and Pain Medicine, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing
| | - Shijie Xu
- Department of Anesthesiology and Pain Medicine, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing
| | - Xuyan Zhou
- Department of Anesthesiology and Pain Medicine, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing
| | - Qingquan Lian
- Department of Anesthesiology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| |
Collapse
|
43
|
Abstract
INTRODUCTION Despite advances in antiplatelet therapy, the optimum antithrombotic regimen during percutaneous coronary intervention (PCI) remains to be determined. Cangrelor is an intravenous, reversibly-binding platelet P2Y12 receptor antagonist with ultra-rapid onset and offset of action that is approved in Europe and United States for use in patients undergoing PCI. This article describes the background for the development of cangrelor, the biology, pharmacology and clinical evidence supporting its use, and its likely position in the future. AREAS COVERED The role of the platelet P2Y12 receptor in platelet biology and the implications of this for atherothrombotic disease are described. Currently unmet needs in antithrombotic management during and after PCI are discussed followed by a description of the chemistry, pharmacokinetics and pharmacodynamics of cangrelor, including its interactions with oral thienopyridines. Subsequently, the clinical trial evidence supporting its adoption into clinical practice is reviewed, including the evidence indicating its superiority over a strategy based on clopidogrel treatment alone. Expert commentary: The current status and future potential of cangrelor is discussed, including a view of its place in current clinical practice.
Collapse
Affiliation(s)
- Robert F Storey
- a Department of Infection, Immunity and Cardiovascular Disease , University of Sheffield , Sheffield , UK
| | - Akanksha Sinha
- a Department of Infection, Immunity and Cardiovascular Disease , University of Sheffield , Sheffield , UK
| |
Collapse
|
44
|
Cupini LM, Bari M, Battista N, Argirò G, Finazzi-Agrò A, Calabresi P, Maccarrone M. Biochemical Changes in Endocannabinoid System are Expressed in Platelets of Female but not Male Migraineurs. Cephalalgia 2016; 26:277-81. [PMID: 16472333 DOI: 10.1111/j.1468-2982.2005.01031.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The endogenous cannabinoid anandamide (AEA) plays important roles in modulating pain. Head pain is an almost universal human experience, yet primary headache disorders, such as migraine without aura (MoA) or episodic tension-type headache (ETTH), can represent a serious threat to well-being when frequent and disabling. We assessed the discriminating role of endocannabinoids among patients with ETTH or MoA, and control subjects. We measured the activity of AEA hydrolase and AEA transporter, and the level of cannabinoid receptors in peripheral platelets from MoA, ETTH and healthy controls. Sixty-nine headache patients and 36 controls were selected. Diagnosis of headache type was made according to the International Headache Society criteria. We observed significant sex differences concerning AEA membrane transporter and fatty acid amide hydrolase activity in all groups. An increase in the activity of AEA hydrolase and AEA transporter was found in female but not male migraineurs. Cannabinoid receptors were the same in all groups. Here we show that the endocannabinoid system in human platelets is altered in female but not male migraneurs. Our results suggest that in migraineur women an increased AEA degradation by platelets, and hence a reduced concentration of AEA in blood, might reduce the pain threshold and possibly explain the prevalence of migraine in women. The involvement of the endocannabinoid system in migraine is new and broadens our knowledge of this widespread and multifactorial disease.
Collapse
Affiliation(s)
- L M Cupini
- Centro Cefalee, Clinica Neurologica, Ospedale S. Eugenio, Roma, Italy.
| | | | | | | | | | | | | |
Collapse
|
45
|
Peterson JF, Field JR, Unertl KM, Schildcrout JS, Johnson DC, Shi Y, Danciu I, Cleator JH, Pulley JM, McPherson JA, Denny JC, Laposata M, Roden DM, Johnson KB. Physician response to implementation of genotype-tailored antiplatelet therapy. Clin Pharmacol Ther 2016; 100:67-74. [PMID: 26693963 DOI: 10.1002/cpt.331] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/20/2015] [Accepted: 12/17/2015] [Indexed: 01/07/2023]
Abstract
Physician responses to genomic information are vital to the success of precision medicine initiatives. We prospectively studied a pharmacogenomics implementation program for the propensity of clinicians to select antiplatelet therapy based on CYP2C19 loss-of-function variants in stented patients. Among 2,676 patients, 514 (19.2%) were found to have a CYP2C19 variant affecting clopidogrel metabolism. For the majority (93.6%) of the cohort, cardiologists received active and direct notification of CYP2C19 status. Over 12 months, 57.6% of poor metabolizers and 33.2% of intermediate metabolizers received alternatives to clopidogrel. CYP2C19 variant status was the most influential factor impacting the prescribing decision (hazard ratio [HR] in poor metabolizers 8.1, 95% confidence interval [CI] [5.4, 12.2] and HR 5.0, 95% CI [4.0, 6.3] in intermediate metabolizers), followed by patient age and type of stent implanted. We conclude that cardiologists tailored antiplatelet therapy for a minority of patients with a CYP2C19 variant and considered both genomic and nongenomic risks in their clinical decision-making.
Collapse
Affiliation(s)
- J F Peterson
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - J R Field
- Institute of Clinical and Translational Research, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - K M Unertl
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - J S Schildcrout
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - D C Johnson
- Department of Pharmacy, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Y Shi
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - I Danciu
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Institute of Clinical and Translational Research, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - J H Cleator
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - J M Pulley
- Institute of Clinical and Translational Research, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - J A McPherson
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - J C Denny
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - M Laposata
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - D M Roden
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - K B Johnson
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
46
|
Koessler J, Hermann S, Weber K, Koessler A, Kuhn S, Boeck M, Kobsar A. Role of Purinergic Receptor Expression and Function for Reduced Responsiveness to Adenosine Diphosphate in Washed Human Platelets. PLoS One 2016; 11:e0147370. [PMID: 26808867 PMCID: PMC4725951 DOI: 10.1371/journal.pone.0147370] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/04/2016] [Indexed: 01/09/2023] Open
Abstract
Background Washing of platelets is an important procedure commonly used for experimental studies, e.g. in cardiovascular research. As a known phenomenon, responsiveness to adenosine diphosphate (ADP) is reduced in washed platelets, although underlying molecular mechanisms—potentially interfering with experimental results—have not been thoroughly studied. Objectives Since ADP mediates its effects via three purinergic receptors P2Y1, P2X1 and P2Y12, their surface expression and function were investigated in washed platelets and, for comparison, in platelet-rich-plasma (PRP) at different time points for up to 2 hours after preparation. Results In contrast to PRP, flow cytometric analysis of surface expression in washed platelets revealed an increase of all receptors during the first 60 minutes after preparation followed by a significant reduction, which points to an initial preactivation of platelets and consecutive degeneration. The activity of the P2X1 receptor (measured by selectively induced calcium flux) was substantially maintained in both PRP and washed platelets. P2Y12 function (determined by flow cytometry as platelet reactivity index) was partially reduced after platelet washing compared to PRP, but remained stable in course of ongoing storage. However, the function of the P2Y1 receptor (measured by selectively induced calcium flux) continuously declined after preparation of washed platelets. Conclusion In conclusion, decreasing ADP responsiveness in washed platelets is particularly caused by impaired activity of the P2Y1 receptor associated with disturbed calcium regulation, which has to be considered in the design of experimental studies addressing ADP mediated platelet function.
Collapse
Affiliation(s)
- Juergen Koessler
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Wuerzburg, Germany
- * E-mail:
| | - Stephanie Hermann
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Katja Weber
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Angela Koessler
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Sabine Kuhn
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Markus Boeck
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Anna Kobsar
- Institute of Transfusion Medicine and Haemotherapy, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
47
|
Burnstock G. Blood cells: an historical account of the roles of purinergic signalling. Purinergic Signal 2015; 11:411-34. [PMID: 26260710 PMCID: PMC4648797 DOI: 10.1007/s11302-015-9462-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 07/23/2015] [Indexed: 12/17/2022] Open
Abstract
The involvement of purinergic signalling in the physiology of erythrocytes, platelets and leukocytes was recognised early. The release of ATP and the expression of purinoceptors and ectonucleotidases on erythrocytes in health and disease are reviewed. The release of ATP and ADP from platelets and the expression and roles of P1, P2Y(1), P2Y(12) and P2X1 receptors on platelets are described. P2Y(1) and P2X(1) receptors mediate changes in platelet shape, while P2Y(12) receptors mediate platelet aggregation. The changes in the role of purinergic signalling in a variety of disease conditions are considered. The successful use of P2Y(12) receptor antagonists, such as clopidogrel and ticagrelor, for the treatment of thrombosis, myocardial infarction and stroke is discussed.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London, NW3 2PF, UK.
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
48
|
Lever RA, Hussain A, Sun BB, Sage SO, Harper AGS. Conventional protein kinase C isoforms differentially regulate ADP- and thrombin-evoked Ca²⁺ signalling in human platelets. Cell Calcium 2015; 58:577-88. [PMID: 26434503 DOI: 10.1016/j.ceca.2015.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 11/15/2022]
Abstract
Rises in cytosolic Ca(2+) concentration ([Ca(2+)]cyt) are central in platelet activation, yet many aspects of the underlying mechanisms are poorly understood. Most studies examine how experimental manipulations affect agonist-evoked rises in [Ca(2+)]cyt, but these only monitor the net effect of manipulations on the processes controlling [Ca(2+)]cyt (Ca(2+) buffering, sequestration, release, entry and removal), and cannot resolve the source of the Ca(2+) or the transporters or channels affected. To investigate the effects of protein kinase C (PKC) on platelet Ca(2+) signalling, we here monitor Ca(2+) flux around the platelet by measuring net Ca(2+) fluxes to or from the extracellular space and the intracellular Ca(2+) stores, which act as the major sources and sinks for Ca(2+) influx into and efflux from the cytosol, as well as monitoring the cytosolic Na(+) concentration ([Na(+)]cyt), which influences platelet Ca(2+) fluxes via Na(+)/Ca(2+) exchange. The intracellular store Ca(2+) concentration ([Ca(2+)]st) was monitored using Fluo-5N, the extracellular Ca(2+) concentration ([Ca(2+)]ext) was monitored using Fluo-4 whilst [Ca(2+)]cyt and [Na(+)]cyt were monitored using Fura-2 and SFBI, respectively. PKC inhibition using Ro-31-8220 or bisindolylmaleimide I potentiated ADP- and thrombin-evoked rises in [Ca(2+)]cyt in the absence of extracellular Ca(2+). PKC inhibition potentiated ADP-evoked but reduced thrombin-evoked intracellular Ca(2+) release and Ca(2+) removal into the extracellular medium. SERCA inhibition using thapsigargin and 2,5-di(tert-butyl) l,4-benzohydroquinone abolished the effect of PKC inhibitors on ADP-evoked changes in [Ca(2+)]cyt but only reduced the effect on thrombin-evoked responses. Thrombin evokes substantial rises in [Na(+)]cyt which would be expected to reduce Ca(2+) removal via the Na(+)/Ca(2+) exchanger (NCX). Thrombin-evoked rises in [Na(+)]cyt were potentiated by PKC inhibition, an effect which was not due to altered changes in non-selective cation permeability of the plasma membrane as assessed by Mn(2+) quench of Fura-2 fluorescence. PKC inhibition was without effect on thrombin-evoked rises in [Ca(2+)]cyt following SERCA inhibition and either removal of extracellular Na(+) or inhibition of Na(+)/K(+)-ATPase activity by removal of extracellular K(+) or treatment with digoxin. These data suggest that PKC limits ADP-evoked rises in [Ca(2+)]cyt by acceleration of SERCA activity, whilst rises in [Ca(2+)]cyt evoked by the stronger platelet activator thrombin are limited by PKC through acceleration of both SERCA and Na(+)/K(+)-ATPase activity, with the latter limiting the effect of thrombin on rises in [Na(+)]cyt and so forward mode NCX activity. The use of selective PKC inhibitors indicated that conventional and not novel PKC isoforms are responsible for the inhibition of agonist-evoked Ca(2+) signalling.
Collapse
Affiliation(s)
- Robert A Lever
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom
| | - Azhar Hussain
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom
| | - Benjamin B Sun
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom
| | - Stewart O Sage
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom
| | - Alan G S Harper
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom; Institute for Science and Technology in Medicine, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Hartshill, Stoke-on-Trent ST4 7QB, United Kingdom.
| |
Collapse
|
49
|
Clopidogrel reduces the inflammatory response of lung in a rat model of decompression sickness. Respir Physiol Neurobiol 2015; 211:9-16. [DOI: 10.1016/j.resp.2015.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 02/24/2015] [Accepted: 02/24/2015] [Indexed: 02/08/2023]
|
50
|
Bhavanasi D, Badolia R, Manne BK, Janapati S, Dangelmaier CT, Mazharian A, Jin J, Kim S, Zhang X, Chen X, Senis YA, Kunapuli SP. Cross talk between serine/threonine and tyrosine kinases regulates ADP-induced thromboxane generation in platelets. Thromb Haemost 2015; 114:558-68. [PMID: 25947062 DOI: 10.1160/th14-09-0775] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 03/26/2015] [Indexed: 11/05/2022]
Abstract
ADP-induced thromboxane generation depends on Src family kinases (SFKs) and is enhanced with pan-protein kinase C (PKC) inhibitors, but it is not clear how these two events are linked. The aim of the current study is to investigate the role of Y311 phosphorylated PKCδ in regulating ADP-induced platelet activation. In the current study, we employed various inhibitors and murine platelets from mice deficient in specific molecules to evaluate the role of PKCδ in ADP-induced platelet responses. We show that, upon stimulation of platelets with 2MeSADP, Y311 on PKCδ is phosphorylated in a P2Y1/Gq and Lyn-dependent manner. By using PKCδ and Lyn knockout murine platelets, we also show that tyrosine phosphorylated PKCδ plays a functional role in mediating 2MeSADP-induced thromboxane generation. 2MeSADP-induced PKCδ Y311 phosphorylation and thromboxane generation were potentiated in human platelets pre-treated with either a pan-PKC inhibitor, GF109203X or a PKC α/β inhibitor and in PKC α or β knockout murine platelets compared to controls. Furthermore, we show that PKC α/β inhibition potentiates the activity of SFK, which further hyper-phosphorylates PKCδ and potentiates thromboxane generation. These results show for the first time that tyrosine phosphorylated PKCδ regulates ADP-induced thromboxane generation independent of its catalytic activity and that classical PKC isoforms α/β regulate the tyrosine phosphorylation on PKCδ and subsequent thromboxane generation through tyrosine kinase, Lyn, in platelets.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Satya P Kunapuli
- Satya P. Kunapuli PhD, Department of Physiology and Sol Sherry Thrombosis Center,, Temple University School of Medicine,, 3420 North Broad street, MRB 414, Philadelphia PA, 19140, USA, Tel.: +1 215 707 4615, Fax: +1 215 707 6944, E-mail:
| |
Collapse
|