1
|
Garofano K, Mariani V, Rashid K, Suwunnakorn S, Sidahmed A, Horvath A, Maggirwar SB, O'Brien TJ, Perera MA, Whalen M, Lee NH. Transcriptomic and functional characterization of megakaryocytic-derived platelet-like particles: impaired aggregation and prominent anti-tumor effects. Platelets 2025; 36:2449344. [PMID: 39812346 DOI: 10.1080/09537104.2024.2449344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025]
Abstract
Platelet-like particles (PLPs), derived from megakaryocytic cell lines MEG-01 and K-562, are widely used as a surrogate to study platelet formation and function. We demonstrate by RNA-Seq that PLPs are transcriptionally distinct from platelets. Expression of key genes in signaling pathways promoting platelet activation/aggregation, such as the PI3K/AKT, protein kinase A, phospholipase C, and α-adrenergic and GP6 receptor pathways, was missing or under-expressed in PLPs. Functionally, PLPs do not aggregate following epinephrine, collagen, or ADP stimulation. While PLPs aggregated in response to thrombin, they did not display enhanced expression of surface markers P-selectin and activated α2bβ3, in contrast to platelets. We have previously demonstrated that platelets physically couple to MDA-PCa-2b and RC77T/E prostate cancer (PCa) cells via specific ligand-receptor interactions, leading to platelet-stimulated cell invasiveness and apoptotic resistance, and reciprocal cell-induced platelet aggregation. In contrast, PLP interactions with PCa cells inhibited both cell invasion and apoptotic resistance while failing to promote PLP aggregation. Moreover, PLPs reduced platelet-PCa cell interactions and antagonized platelet-stimulated oncogenic effects in PCa cells. RNA-Seq analysis identified candidate ligand-transmembrane protein combinations involved in anti-tumorigenic signaling of PLPs to PCa cells. Antibody neutralization of the TIMP3-MMP15 and VEGFB-FGFR1 signaling axes reversed PLP-mediated anti-invasion and apoptotic sensitization, respectively. In summary, PLPs lack many transcriptomic, molecular and functional features of platelets and possess novel anti-tumorigenic properties. These findings indicate that PLPs may have a potential therapeutic role in targeting and disrupting the oncogenic signaling between platelets and cancer cells, offering a new avenue for anti-cancer strategies.
Collapse
Affiliation(s)
- Kaitlin Garofano
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Vera Mariani
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Kameron Rashid
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Sumanun Suwunnakorn
- Department of Microbiology Immunology and Tropical Medicine, The George Washington University, Washington, DC, USA
| | - Alfateh Sidahmed
- Department of Medicine, George Washington University, Washington, DC, USA
| | - Anelia Horvath
- Department of Biochemistry and Molecular Medicine, George Washington University, Washington, DC, USA
| | - Sanjay B Maggirwar
- Department of Microbiology Immunology and Tropical Medicine, The George Washington University, Washington, DC, USA
| | - Travis J O'Brien
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Minoli A Perera
- Department of Pharmacology and Center for Pharmacogenomics, Northwestern University, Chicago, IL, USAand
| | - Michael Whalen
- GW Cancer Center, George Washington University, Washington, DC, USA
| | - Norman H Lee
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
- GW Cancer Center, George Washington University, Washington, DC, USA
| |
Collapse
|
2
|
Sveshnikova AN, Panteleev MA. Signal Transduction and Transformation by the Platelet Activation Cascade: Systems Biology Insights. Hamostaseologie 2025; 45:49-62. [PMID: 39970901 DOI: 10.1055/a-2486-6758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025] Open
Abstract
Binding of platelet activators to their receptors initiates a signal transduction network, where intracellular signal is filtered, amplified, and transformed. Computational systems biology methods could be a powerful tool to address and analyze dynamics and regulation of the crucial steps in this cascade. Here we review these approaches and show the logic of their use for a relatively simple case of SFLLRN-induced procoagulant activity. Use of a typical model is employed to track signaling events along the main axis, from the binding of the peptide to PAR1 receptor down to the mPTP opening. Temporal dynamics, concentration dependence, formation of calcium oscillations and their deciphering, and role of stochasticity are quantified for all essential signaling molecules and their complexes. The initial step-wise activation stimulus is transformed to a peak at the early stages, then to oscillation calcium spikes, and then back to a peak shape. The model can show how both amplitude and width of the peak encode the information about the activation level, and show the principle of decoding calcium oscillations via integration of the calcium signal by the mitochondria. Use of stochastic algorithms can reveal that the complexes of Gq, in particular the complex of phospholipase C with Gq, which are the limiting steps in the cascade with their numbers not exceeding several molecules per platelet at any given time; it is them that cause stochastic appearance of the signals downstream. Application of reduction techniques to simplify the system is demonstrated.
Collapse
Affiliation(s)
- Anastasia N Sveshnikova
- Cell Biology, Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
- Molecular Hemostasis, Center for Theoretical Problems of Physicochemical Pharmacology, Moscow, Russian Federation
- Physics Faculty, M. V. Lomonosov Moscow State University, Moscow, Russian Federation
| | - Mikhail Aleksandrovich Panteleev
- Cell Biology, Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
- Molecular Hemostasis, Center for Theoretical Problems of Physicochemical Pharmacology, Moscow, Russian Federation
- Physics Faculty, M. V. Lomonosov Moscow State University, Moscow, Russian Federation
| |
Collapse
|
3
|
Zhang T, Zhang M, Guo L, Liu D, Zhang K, Bi C, Zhang P, Wang J, Fan Y, He Q, Chang ACY, Zhang J. Angiopoietin-like protein 2 inhibits thrombus formation. Mol Cell Biochem 2025; 480:1169-1181. [PMID: 38880861 PMCID: PMC11835982 DOI: 10.1007/s11010-024-05034-9] [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: 03/02/2024] [Accepted: 05/10/2024] [Indexed: 06/18/2024]
Abstract
Acute myocardial infarction is mainly caused by a lack of blood flood in the coronary artery. Angiopoietin-like protein 2 (ANGPTL2) induces platelet activation and thrombus formation in vitro through binding with immunoglobulin-like receptor B, an immunoglobulin superfamily receptor. However, the mechanism by which it regulates platelet function in vivo remains unclear. In this study, we investigated the role of ANGPTL2 during thrombosis in relationship with ST-segment elevation myocardial infarction (STEMI) with spontaneous recanalization (SR). In a cohort of 276 male and female patients, we measured plasma ANGPTL2 protein levels. Using male Angptl2-knockout and wild-type mice, we examined the inhibitory effect of Angptl2 on thrombosis and platelet activation both in vivo and ex vivo. We found that plasma and platelet ANGPTL2 levels were elevated in patients with STEMI with SR compared to those in non-SR (NSR) patients, and was an independent predictor of SR. Angptl2 deficiency accelerated mesenteric artery thrombosis induced by FeCl3 in Angptl2-/- compared to WT animals, promoted platelet granule secretion and aggregation induced by thrombin and collogen while purified ANGPTL2 protein supplementation reversed collagen-induced platelet aggregation. Angptl2 deficiency also increased platelet spreading on immobilized fibrinogen and clot contraction. In collagen-stimulated Angptl2-/- platelets, Src homology region 2 domain-containing phosphatase (Shp)1-Y564 and Shp2-Y580 phosphorylation were attenuated while Src, Syk, and Phospholipase Cγ2 (PLCγ2) phosphorylation increased. Our results demonstrate that ANGPTL2 negatively regulated thrombus formation by activating ITIM which can suppress ITAM signaling pathway. This new knowledge provides a new perspective for designing future antiplatelet aggregation therapies.
Collapse
Affiliation(s)
- Tiantian Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Mingliang Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Lingyu Guo
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Dongsheng Liu
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Kandi Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Changlong Bi
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Peng Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jin Wang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yuqi Fan
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Qing He
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Alex C Y Chang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Junfeng Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.
| |
Collapse
|
4
|
Yan M, Wang Z, Qiu Z, Cui Y, Xiang Q. Platelet signaling in immune landscape: comprehensive mechanism and clinical therapy. Biomark Res 2024; 12:164. [PMID: 39736771 DOI: 10.1186/s40364-024-00700-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 11/28/2024] [Indexed: 01/01/2025] Open
Abstract
Platelets are essential for blood clotting and maintaining normal hemostasis. In pathological conditions, platelets are increasingly recognized as crucial regulatory factors in various immune-mediated inflammatory diseases. Resting platelets are induced by various factors such as immune complexes through Fc receptors, platelet-targeting autoantibodies and other platelet-activating stimuli. Platelet activation in immunological processes involves the release of immune activation stimuli, antigen presentation and interaction with immune cells. Platelets participate in both the innate immune system (neutrophils, monocytes/macrophages, dendritic cells (DCs) and Natural Killer (NK) cells and the adaptive immune system (T and B cells). Clinical therapeutic strategies include targeting platelet activation, platelet-immune cell interaction and platelet-endothelial cell interaction, which display positive development prospects. Understanding the mechanisms of platelets in immunity is important, and developing targeted modulations of these mechanisms will pave the way for promising therapeutic strategies.
Collapse
Affiliation(s)
- Mengyao Yan
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
| | - Zhe Wang
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
| | - Zhiwei Qiu
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
| | - Yimin Cui
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China.
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China.
| | - Qian Xiang
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China.
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
5
|
Ye Y, Leng M, Chai S, Yang L, Ren L, Wan W, Wang H, Li L, Li C, Meng Z. Antiplatelet effects of the CEACAM1-derived peptide QDTT. Platelets 2024; 35:2308635. [PMID: 38345065 DOI: 10.1080/09537104.2024.2308635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/17/2024] [Indexed: 02/15/2024]
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) restricts platelet activation via platelet collagen receptor GPVI/FcRγ-chain. In this study, screening against collagen-induced platelet aggregation was performed to identify functional CEACAM1 extracellular domain fragments. CEACAM1 fragments, including Ala-substituted peptides, were synthesized. Platelet assays were conducted on healthy donor samples for aggregation, cytotoxicity, adhesion, spreading, and secretion. Mice were used for tail bleeding and FeCl3-induced thrombosis experiments. Clot retraction was assessed using platelet-rich plasma. Extracellular segments of CEACAM1 and A1 domain-derived peptide QDTT were identified, while N, A2, and B domains showed no involvement. QDTT inhibited platelet aggregation. Ala substitution for essential amino acids (Asp139, Thr141, Tyr142, Trp144, and Trp145) in the QDTT sequence abrogated collagen-induced aggregation inhibition. QDTT also suppressed platelet secretion and "inside-out" GP IIb/IIIa activation by convulxin, along with inhibiting PI3K/Akt pathways. QDTT curtailed FeCl3-induced mesenteric thrombosis without significantly prolonging bleeding time, implying the potential of CEACAM1 A1 domain against platelet activation without raising bleeding risk, thus paving the way for novel antiplatelet drugs.
Collapse
Affiliation(s)
- Yujia Ye
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Min Leng
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Shengjie Chai
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Lihong Yang
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Longcheng Ren
- Cardiovascular Department, Tengchong Hospital of Traditional Chinese Medicine, Tengchong, PR China
| | - Wen Wan
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Huawei Wang
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Longjun Li
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Chaozhong Li
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Zhaohui Meng
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| |
Collapse
|
6
|
Makarov MS, Borovkova NV, Storozheva MV, Ponomarev IN. Effect of In Vitro Low-Impulse Laser Irradiation on Morphofunctional Properties of Human Platelets. Bull Exp Biol Med 2024:10.1007/s10517-024-06297-4. [PMID: 39579297 DOI: 10.1007/s10517-024-06297-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Indexed: 11/25/2024]
Abstract
We studied morphofunctional features of human platelets exposed in vitro to low-pulse laser radiation (LPLR) with different wavelengths. Platelet-rich plasma (PRP) was irradiated at wavelengths of 635, 488, and 355 nm. LPLR with λ=635 nm and 1 W power after 10 min caused degranulation of many platelets, formation of small platelet conglomerates, and accelerated platelet adhesion on glass. LPLR with λ=635 nm and 2 mW and LPLR with λ=488 nm did not cause significant changes in the morphofunctional platelet status. LPLR with λ=355 nm induced massive platelets' deformation, sharp decrease in their morphofunctional status without activation.
Collapse
Affiliation(s)
- M S Makarov
- Sklifosovsky Research Institute for Emergency Medicine, Moscow, Russia.
| | - N V Borovkova
- Sklifosovsky Research Institute for Emergency Medicine, Moscow, Russia
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - M V Storozheva
- Sklifosovsky Research Institute for Emergency Medicine, Moscow, Russia
| | - I N Ponomarev
- Sklifosovsky Research Institute for Emergency Medicine, Moscow, Russia
| |
Collapse
|
7
|
Pérez-Guàrdia L, Lafabrie E, Diedhiou N, Spiegelhalter C, Laporte J, Böhm J. A Gain-of-Function Mutation in the Ca 2+ Channel ORAI1 Causes Stormorken Syndrome with Tubular Aggregates in Mice. Cells 2024; 13:1829. [PMID: 39594579 PMCID: PMC11592465 DOI: 10.3390/cells13221829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/15/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Store-operated Ca2+ entry (SOCE) controls Ca2+ homeostasis and mediates multiple Ca2+-dependent signaling pathways and cellular processes. It relies on the concerted activity of the reticular Ca2+ sensor STIM1 and the plasma membrane Ca2+ channel ORAI1. STIM1 and ORAI1 gain-of-function (GoF) mutations induce SOCE overactivity and excessive Ca2+ influx, leading to tubular aggregate myopathy (TAM) and Stormorken syndrome (STRMK), two overlapping disorders characterized by muscle weakness and a variable occurrence of multi-systemic anomalies affecting spleen, skin, and platelets. To date, different STIM1 mouse models exist, but only a single ORAI1 mouse model with muscle-specific TAM/STRMK phenotype has been described, precluding a comparative analysis of the physiopathology in all affected tissues. Here, we generated and characterized mice harboring a prevalent ORAI1 TAM/STRMK mutation and we provide phenotypic, physiological, biochemical, and functional data. Examination of Orai1V109M/+ mice revealed smaller size, spleen enlargement, reduced muscle force, and decreased platelet numbers. Morphological analyses of muscle sections evidenced the presence of tubular aggregates, the histopathological hallmark on biopsies from TAM/STRMK patients absent in all reported STIM1 models. Overall, Orai1V109M/+ mice reliably recapitulate the human disorder and highlight the primary physiological defects caused by ORAI1 gain-of-function mutations. They also provide the possibility to investigate the formation of tubular aggregates and to develop a common therapy for different TAM/STRMK forms.
Collapse
MESH Headings
- Animals
- ORAI1 Protein/metabolism
- ORAI1 Protein/genetics
- Gain of Function Mutation/genetics
- Mice
- Blood Platelet Disorders/genetics
- Blood Platelet Disorders/pathology
- Blood Platelet Disorders/metabolism
- Stromal Interaction Molecule 1/genetics
- Stromal Interaction Molecule 1/metabolism
- Erythrocytes, Abnormal/metabolism
- Erythrocytes, Abnormal/pathology
- Ichthyosis/genetics
- Ichthyosis/pathology
- Ichthyosis/metabolism
- Myopathies, Structural, Congenital/genetics
- Myopathies, Structural, Congenital/pathology
- Myopathies, Structural, Congenital/metabolism
- Dyslexia/genetics
- Dyslexia/metabolism
- Dyslexia/pathology
- Disease Models, Animal
- Spleen/pathology
- Spleen/metabolism
- Spleen/abnormalities
- Syndactyly/genetics
- Syndactyly/pathology
- Syndactyly/metabolism
- Miosis/genetics
- Miosis/metabolism
- Miosis/pathology
- Muscle Weakness/genetics
- Muscle Weakness/pathology
- Muscle Weakness/metabolism
- Calcium/metabolism
- Humans
- Phenotype
- Mice, Inbred C57BL
- Migraine Disorders
- Muscle Fatigue
Collapse
Affiliation(s)
| | | | | | | | | | - Johann Böhm
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U1258, CNRS UMR7104, Université de Strasbourg, 67404 Illkirch, France
| |
Collapse
|
8
|
Gaspar RS, Silva França ÁR, Oliveira PVS, Silva Diniz-Filho JF, Teixeira L, Valadão IC, Debbas V, Costa Dos Santos C, Massafera MP, Bustos SO, Rebelo Alencar LM, Ronsein GE, Laurindo FRM. Endothelial protein disulfide isomerase A1 enhances membrane stiffness and platelet-endothelium interaction in hyperglycemia via SLC3A2 and LAMC1. J Thromb Haemost 2024; 22:3305-3321. [PMID: 39128656 DOI: 10.1016/j.jtha.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Diabetes carries an increased risk of cardiovascular disease and thromboembolic events. Upon endothelial dysfunction, platelets bind to endothelial cells to precipitate thrombus formation; however, it is unclear which surface proteins regulate platelet-endothelium interaction. We and others have shown that peri/epicellular protein disulfide isomerase A1 (pecPDI) influences the adhesion and migration of vascular cells. OBJECTIVES We investigated whether pecPDI regulates adhesion-related molecules on the surface of endothelial cells and platelets that influence the binding of these cells in hyperglycemia. METHODS Immunofluorescence was used to assess platelet-endothelium interaction in vitro, cytoskeleton reorganization, and focal adhesions. Hydrogen peroxide production was assessed via Amplex Red assays (ThermoFisher Scientific). Cell biophysics was assessed using atomic force microscopy. Secreted proteins of interest were identified through proteomics (secretomics), and targets were knocked down using small interfering RNA. Protein disulfide isomerase A1 (PDI) contribution was assessed using whole-cell PDI or pecPDI inhibitors or small interfering RNA. RESULTS Platelets of healthy donors adhered more onto hyperglycemic human umbilical vein endothelial cells (HUVECs). Endothelial, but not platelet, pecPDI regulated this effect. Hyperglycemic HUVECs showed marked cytoskeleton reorganization, increased H2O2 production, and elongated focal adhesions. Indeed, hyperglycemic HUVECs were stiffer compared with normoglycemic cells. PDI and pecPDI inhibition reversed the abovementioned processes in hyperglycemic cells. A secretomics analysis revealed 8 proteins secreted in a PDI-dependent manner by hyperglycemic cells. Among these, we showed that genetic deletion of LAMC1 and SLC3A2 decreased platelet-endothelium interaction and did not potentiate the effects of PDI inhibitors. CONCLUSION Endothelial pecPDI regulates platelet-endothelium interaction in hyperglycemia through adhesion-related proteins and alterations in endothelial membrane biophysics.
Collapse
Affiliation(s)
- Renato S Gaspar
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coração (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
| | - Álefe Roger Silva França
- Federal University of Maranhão, Physics Department, Laboratory of Biophysics and Nanosystems, São Luís, Brazil
| | - Percillia Victoria Santos Oliveira
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coração (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | - Livia Teixeira
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coração (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Iuri Cordeiro Valadão
- Laboratorio de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Victor Debbas
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coração (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Clenilton Costa Dos Santos
- Federal University of Maranhão, Physics Department, Laboratory of Biophysics and Nanosystems, São Luís, Brazil
| | | | - Silvina Odete Bustos
- Center for Translational Research in Oncology (LIM24), Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Francisco R M Laurindo
- Laboratorio de Biologia Vascular (LVascBio), LIM-64, Instituto do Coração (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
9
|
Srivastava P, Jha S, Singh SK, Vyas H, Sethupathi P, Nair RS, Ramachandran K, Rana B, Kumar S, Rana A. Protease activated receptor-1 regulates mixed lineage kinase-3 to drive triple-negative breast cancer tumorigenesis. Cancer Lett 2024; 603:217200. [PMID: 39222677 DOI: 10.1016/j.canlet.2024.217200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Triple-negative breast cancer (TNBC) is difficult to treat breast cancer subtype due to lack or insignificant expressions of targetable estrogen receptor (ER) and human epidermal growth factor receptor 2 (HER2). Therefore, finding a targetable protein or signaling pathway in TNBC would impact patient care. Here, we report that a member of the Mixed Lineage Kinase (MLK) family, MLK3, is an effector of G-protein-coupled protease-activated receptors 1 (PAR1) and targeting MLK3 by a small-molecule inhibitor prevented PAR1-mediated TNBC tumorigenesis. In silico and immunohistochemistry analysis of human breast tumors showed overexpression of PAR1 and MLK3 in TNBC tumors. Treating α-thrombin and PAR1 agonist increased MLK3 and JNK activities and induced cell migration in TNBC cells. The PAR1 positive/high (PAR1+/hi) population of TNBC cells showed aggressive tumor phenotype with increased MLK3 signaling. Moreover, combined inhibition of the PAR1 and MLK3 mitigated the TNBC tumor burden in preclinical TNBC models. Our data suggests that activation of the PAR1-MLK3 axis promotes TNBC tumorigenesis. Therefore, combinatorial therapy targeting MLK3 and PAR1 could effectively reduce TNBC tumor burden.
Collapse
Affiliation(s)
- Piush Srivastava
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Saket Jha
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Sunil Kumar Singh
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Harsh Vyas
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Periannan Sethupathi
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Rakesh Sathish Nair
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Kheerthivasan Ramachandran
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Basabi Rana
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA; University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL, 60612, USA; Research Unit, Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
| | - Sandeep Kumar
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA; University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| | - Ajay Rana
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA; University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL, 60612, USA; Research Unit, Jesse Brown VA Medical Center, Chicago, IL, 60612, USA.
| |
Collapse
|
10
|
Sonay AY, Larney BEM, Apfelbaum E, Grimm J. In vivo imaging of ferroptosis through nanodynamic changes in lipid membranes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620222. [PMID: 39484558 PMCID: PMC11527342 DOI: 10.1101/2024.10.25.620222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Ferroptosis emerged as a cell death modality for drug resistant cancer cells, but there are currently no available biomarkers for imaging ferroptosis based therapies. To address this gab, we evaluated the nanodynamic changes in lipid membranes occurring during cell death to explore potential targeting opportunities to image cell death. We nano-sized gaps at late stages of ferroptosis can serve as entry points for dyes that can bind to cellular structures. These changes were accompanied with cellular signaling components similar to platelet activation, with phosphatidyl serine emerging on the surface of the cells and therefore as a potential target for imaging of programed cell death, including ferroptosis. Taking advantage of these changes in cell membrane dynamics, we employed a novel tumor-seeking dye CJ215 that can label apoptotic cells as recently described by us. We show that CJ215 accumulates in ferroptotic cells both in vitro and in vivo by binding to phosphatidyl serine, a process that is prevented by inhibition of ferroptosis. Since phosphatidyl serine exposure also occurs during apoptosis, CJ215 can serve to image both apoptosis and ferroptosis based therapy.
Collapse
Affiliation(s)
- Ali Yasin Sonay
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | | | - Elana Apfelbaum
- Pharmacology Program, Weill Cornell Medical College; New York, NY USA
| | - Jan Grimm
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
- Pharmacology Program, Weill Cornell Medical College; New York, NY USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center; New York, NY, USA
- Department of Radiology, Weill, Cornell Medical Center; New York, NY, USA
| |
Collapse
|
11
|
Hong F, Mollica MY, Golla K, De Silva E, Sniadecki NJ, López JA, Kim H. Filamin A regulates platelet shape change and contractile force generation via phosphorylation of the myosin light chain. Biochem J 2024; 481:1395-1410. [PMID: 39189664 PMCID: PMC11555712 DOI: 10.1042/bcj20240114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 08/13/2024] [Accepted: 08/23/2024] [Indexed: 08/28/2024]
Abstract
Platelets are critical mediators of hemostasis and thrombosis. Platelets circulate as discs in their resting form but change shape rapidly upon activation by vascular damage and/or soluble agonists such as thrombin. Platelet shape change is driven by a dynamic remodeling of the actin cytoskeleton. Actin filaments interact with the protein myosin, which is phosphorylated on the myosin light chain (MLC) upon platelet activation. Actin-myosin interactions trigger contraction of the actin cytoskeleton, which drives platelet spreading and contractile force generation. Filamin A (FLNA) is an actin cross-linking protein that stabilizes the attachment between subcortical actin filaments and the cell membrane. In addition, FLNA binds multiple proteins and serves as a critical intracellular signaling scaffold. Here, we used platelets from mice with a megakaryocyte/platelet-specific deletion of FLNA to investigate the role of FLNA in regulating platelet shape change. Relative to controls, FLNA-null platelets exhibited defects in stress fiber formation, contractile force generation, and MLC phosphorylation in response to thrombin stimulation. Blockade of Rho kinase (ROCK) and protein kinase C (PKC) with the inhibitors Y27632 and bisindolylmaleimide (BIM), respectively, also attenuated MLC phosphorylation; our data further indicate that ROCK and PKC promote MLC phosphorylation through independent pathways. Notably, the activity of both ROCK and PKC was diminished in the FLNA-deficient platelets. We conclude that FLNA regulates thrombin-induced MLC phosphorylation and platelet contraction, in a ROCK- and PKC-dependent manner.
Collapse
Affiliation(s)
- Felix Hong
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Molly Y. Mollica
- Bloodworks Northwest Research Institute, Seattle, WA, U.S.A
- School of Medicine, Division of Hematology, University of Washington, Seattle, WA, U.S.A
- Department of Mechanical Engineering, University of Maryland, Baltimore County, Baltimore, MD, U.S.A
| | - Kalyan Golla
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Enoli De Silva
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Nathan J. Sniadecki
- Department of Mechanical Engineering, University of Washington, Seattle, WA, U.S.A
- Department of Bioengineering, University of Washington, Seattle, WA, U.S.A
- Department of Lab Medicine and Pathology, University of Washington, Seattle, WA, U.S.A
| | - José A. López
- Bloodworks Northwest Research Institute, Seattle, WA, U.S.A
- School of Medicine, Division of Hematology, University of Washington, Seattle, WA, U.S.A
| | - Hugh Kim
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
12
|
Stabile J, Fürstenau CR. Platelets isolation and ectonucleotidase assay: Revealing functional aspects of the communication between the vasculature and the immune system. J Immunol Methods 2024; 533:113746. [PMID: 39181235 DOI: 10.1016/j.jim.2024.113746] [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: 02/10/2024] [Revised: 08/17/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
Platelets are enucleated fragments of cells with a diversity of internal granules. They are responsible for functions related to hemostasis, coagulation, and inflammation. The activation of these processes depends on a cascade coordinated by cytokines, chemokines, and components of purinergic signaling, such as ATP, ADP, and adenosine. Platelets express distinct components of the purinergic system: P2X1, P2Y1, PY12, and P2Y14 receptors; and the ectonucleotidases NTPDase, NPP, and 5NTE (ecto-5'-nucleotidase). Except for P2Y14, which has not yet exhibited a known function, all other components relate to the biological processes mentioned before. Platelets are known to display specific responses to microorganisms, being capable of recognizing pathogen-associated molecular patterns (PAMPs), engulfing certain classes of viruses, and participating in NETosis. Platelet function dysregulation implicates various pathophysiological processes, including cardiovascular diseases (CVDs) and infections. In COVID-19 patients, platelets exhibit altered purinergic signaling and increased activation, contributing to inflammation. Excessive platelet activation can lead to complications from thrombosis, which can affect the circulation of vital organs. Therefore, controlling the activation is necessary to end the inflammatory process and restore homeostasis. Ectonucleotidases, capable of hydrolyzing ATP, ADP, and AMP, are of fundamental importance in activating platelets, promising pharmacological targets for clinical use as cardiovascular protective drugs. In this review, we revisit platelet biology, the purinergic receptors and ectonucleotidases on their surface, and their importance in platelet activity. Additionally, we describe methods for isolating platelets in humans and murine, as well as the main techniques for detecting the activity of ectonucleotidases in platelets. Considering the multitude of functions revealed by platelets and their potential use as potent bioreactors able to secrete and present molecules involved in the communication of the vasculature with the immune system, it is crucial to deeply understand platelet biology and purinergic signaling participation to contribute to the developing of therapeutic strategies in diseases of the cardiovascular, inflammatory, and immune systems.
Collapse
Affiliation(s)
- Jeferson Stabile
- Laboratory of Vascular Biochemistry, Center for Natural and Human Sciences, Federal University of ABC, Santo André, SP, Brazil
| | - Cristina Ribas Fürstenau
- Laboratory of Vascular Biochemistry, Center for Natural and Human Sciences, Federal University of ABC, Santo André, SP, Brazil.
| |
Collapse
|
13
|
Montecino-Garrido H, Trostchansky A, Espinosa-Parrilla Y, Palomo I, Fuentes E. How Protein Depletion Balances Thrombosis and Bleeding Risk in the Context of Platelet's Activatory and Negative Signaling. Int J Mol Sci 2024; 25:10000. [PMID: 39337488 PMCID: PMC11432290 DOI: 10.3390/ijms251810000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/28/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Platelets are small cell fragments that play a crucial role in hemostasis, requiring fast response times and fine signaling pathway regulation. For this regulation, platelets require a balance between two pathway types: the activatory and negative signaling pathways. Activatory signaling mediators are positive responses that enhance stimuli initiated by a receptor in the platelet membrane. Negative signaling regulates and controls the responses downstream of the same receptors to roll back or even avoid spontaneous thrombotic events. Several blood-related pathologies can be observed when these processes are unregulated, such as massive bleeding in activatory signaling inhibition or thrombotic events for negative signaling inhibition. The study of each protein and metabolite in isolation does not help to understand the role of the protein or how it can be contrasted; however, understanding the balance between active and negative signaling could help develop effective therapies to prevent thrombotic events and bleeding disorders.
Collapse
Affiliation(s)
- Hector Montecino-Garrido
- Centro de Estudios en Alimentos Procesados (CEAP), ANID-Regional, Gore Maule R0912001, Talca 3480094, Chile
| | - Andrés Trostchansky
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
| | - Yolanda Espinosa-Parrilla
- Interuniversity Center for Healthy Aging (CIES), Centro Asistencial, Docente e Investigación-CADI-UMAG, Escuela de Medicina, Universidad de Magallanes, Punta Arenas 6210427, Chile
| | - Iván Palomo
- Thrombosis and Healthy Aging Research Center, Interuniversity Center for Healthy Aging (CIES), Interuniversity Network of Healthy Aging in Latin America and Caribbean (RIES-LAC), Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Talca 3480094, Chile
| | - Eduardo Fuentes
- Thrombosis and Healthy Aging Research Center, Interuniversity Center for Healthy Aging (CIES), Interuniversity Network of Healthy Aging in Latin America and Caribbean (RIES-LAC), Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Talca 3480094, Chile
| |
Collapse
|
14
|
Slotabec L, Seale B, Wang H, Wen C, Filho F, Rouhi N, Adenawoola MI, Li J. Platelets at the intersection of inflammation and coagulation in the APC-mediated response to myocardial ischemia/reperfusion injury. FASEB J 2024; 38:e23890. [PMID: 39143722 PMCID: PMC11373610 DOI: 10.1096/fj.202401128r] [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: 05/21/2024] [Revised: 07/23/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024]
Abstract
Thromboinflammation is a complex pathology associated with inflammation and coagulation. In cases of cardiovascular disease, in particular ischemia-reperfusion injury, thromboinflammation is a common complication. Increased understanding of thromboinflammation depends on an improved concept of the mechanisms of cells and proteins at the axis of coagulation and inflammation. Among these elements are activated protein C and platelets. This review summarizes the complex interactions of activated protein C and platelets regulating thromboinflammation in cardiovascular disease. By unraveling the pathways of platelets and APC in the inflammatory and coagulation cascades, this review summarizes the role of these vital mediators in the development and perpetuation of heart disease and the thromboinflammation-driven complications of cardiovascular disease. Furthermore, this review emphasizes the significance of the counteracting effects of platelets and APC and their combined role in disease states.
Collapse
Affiliation(s)
- Lily Slotabec
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| | - Blaise Seale
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Hao Wang
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Changhong Wen
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Fernanda Filho
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Nadiyeh Rouhi
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Michael I Adenawoola
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Ji Li
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
15
|
Muttiah B, Ng SL, Lokanathan Y, Ng MH, Law JX. Beyond Blood Clotting: The Many Roles of Platelet-Derived Extracellular Vesicles. Biomedicines 2024; 12:1850. [PMID: 39200314 PMCID: PMC11351396 DOI: 10.3390/biomedicines12081850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/23/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Platelet-derived extracellular vesicles (pEVs) are emerging as pivotal players in numerous physiological and pathological processes, extending beyond their traditional roles in hemostasis and thrombosis. As one of the most abundant vesicle types in human blood, pEVs transport a diverse array of bioactive molecules, including growth factors, cytokines, and clotting factors, facilitating crucial intercellular communication, immune regulation, and tissue healing. The unique ability of pEVs to traverse tissue barriers and their biocompatibility position them as promising candidates for targeted drug delivery and regenerative medicine applications. Recent studies have underscored their involvement in cancer progression, viral infections, wound healing, osteoarthritis, sepsis, cardiovascular diseases, rheumatoid arthritis, and atherothrombosis. For instance, pEVs promote tumor progression and metastasis, enhance tissue repair, and contribute to thrombo-inflammation in diseases such as COVID-19. Despite their potential, challenges remain, including the need for standardized isolation techniques and a comprehensive understanding of their mechanisms of action. Current research efforts are focused on leveraging pEVs for innovative anti-cancer treatments, advanced drug delivery systems, regenerative therapies, and as biomarkers for disease diagnosis and monitoring. This review highlights the necessity of overcoming technical hurdles, refining isolation methods, and establishing standardized protocols to fully unlock the therapeutic potential of pEVs. By understanding the diverse functions and applications of pEVs, we can advance their use in clinical settings, ultimately revolutionizing treatment strategies across various medical fields and improving patient outcomes.
Collapse
Affiliation(s)
- Barathan Muttiah
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Sook Luan Ng
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Yogeswaran Lokanathan
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Min Hwei Ng
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Jia Xian Law
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| |
Collapse
|
16
|
Corken A, Wahl EC, Sikes JD, Thakali KM. Western Diet Modifies Platelet Activation Profiles in Male Mice. Int J Mol Sci 2024; 25:8019. [PMID: 39125586 PMCID: PMC11311362 DOI: 10.3390/ijms25158019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
The correlation between obesity and cardiovascular disease has long been understood, yet scant investigations endeavored to determine the impact of an obesogenic diet on platelet activation or function. As platelets drive clot formation, the terminus of cardiovascular events, we aimed to elucidate the longitudinal effect of an obesogenic diet on platelet phenotype by assessing markers of platelet activation using flow cytometry. Male, weanling mice were fed either a Western diet (30% kcal sucrose, 40% kcal fat, 8.0% sodium) or Control diet (7% kcal sucrose, 10% kcal fat, 0.24% sodium). At 12, 16 and 20 weeks on diets, platelets were collected and stained to visualize glycoprotein Ibα (GPIbα), P-selectin and the conformationally active state of αIIbβ3 (a platelet specific integrin) after collagen stimulation. At all time points, a Western diet reduced GPIbα and αIIbβ3 expression in platelets broadly while P-selectin levels were unaffected. However, P-selectin was diminished by a Western diet in the GPIbα- subpopulation. Thus, a Western diet persistently primed platelets towards a blunted activation response as indicated by reduced active αIIbβ3 and P-selectin surface expression. This study provides a first look at the influence of diet on platelet activation and revealed that platelet activation is susceptible to dietary intervention.
Collapse
Affiliation(s)
- Adam Corken
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.C.); (E.C.W.); (J.D.S.)
- Arkansas Children’s Nutrition Center, Arkansas Children’s Research Institute, Little Rock, AR 72202, USA
| | - Elizabeth C. Wahl
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.C.); (E.C.W.); (J.D.S.)
| | - James D. Sikes
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.C.); (E.C.W.); (J.D.S.)
| | - Keshari M. Thakali
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.C.); (E.C.W.); (J.D.S.)
- Arkansas Children’s Nutrition Center, Arkansas Children’s Research Institute, Little Rock, AR 72202, USA
| |
Collapse
|
17
|
Nappi F. P2Y12 Receptor Inhibitor for Antiaggregant Therapies: From Molecular Pathway to Clinical Application. Int J Mol Sci 2024; 25:7575. [PMID: 39062819 PMCID: PMC11277343 DOI: 10.3390/ijms25147575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Platelets play a significant role in hemostasis, forming plugs at sites of vascular injury to limit blood loss. However, if platelet activation is not controlled, it can lead to thrombotic events, such as myocardial infarction and stroke. To prevent this, antiplatelet agents are used in clinical settings to limit platelet activation in patients at risk of arterial thrombotic events. However, their use can be associated with a significant risk of bleeding. An enhanced comprehension of platelet signaling mechanisms should facilitate the identification of safer targets for antiplatelet therapy. Over the past decade, our comprehension of the breadth and intricacy of signaling pathways that orchestrate platelet activation has expanded exponentially. Several recent studies have provided further insight into the regulation of platelet signaling events and identified novel targets against which to develop novel antiplatelet agents. Antiplatelet drugs are essential in managing atherothrombotic vascular disease. The current antiplatelet therapy in clinical practice is limited in terms of safety and efficacy. Novel compounds have been developed in response to patient variability and resistance to aspirin and/or clopidogrel. Recent studies based on randomized controlled trials and systematic reviews have definitively demonstrated the role of antiplatelet therapy in reducing the risk of cardiovascular events. Antiplatelet therapy is the recommended course of action for patients with established atherosclerosis. These studies compared monotherapy with a P2Y12 inhibitor versus aspirin for secondary prevention. However, in patients undergoing percutaneous coronary intervention, it is still unclear whether the efficacy of P2Y12 inhibitor monotherapy after a short course of dual antiplatelet therapy depends on the type of P2Y12 inhibitor. This paper focuses on the advanced-stage evaluation of several promising antiplatelet drugs.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| |
Collapse
|
18
|
O'Donoghue L, Smolenski A. Roles of G proteins and their GTPase-activating proteins in platelets. Biosci Rep 2024; 44:BSR20231420. [PMID: 38808367 PMCID: PMC11139668 DOI: 10.1042/bsr20231420] [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: 11/17/2023] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/30/2024] Open
Abstract
Platelets are small anucleate blood cells supporting vascular function. They circulate in a quiescent state monitoring the vasculature for injuries. Platelets adhere to injury sites and can be rapidly activated to secrete granules and to form platelet/platelet aggregates. These responses are controlled by signalling networks that include G proteins and their regulatory guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). Recent proteomics studies have revealed the complete spectrum of G proteins, GEFs, and GAPs present in platelets. Some of these proteins are specific for platelets and very few have been characterised in detail. GEFs and GAPs play a major role in setting local levels of active GTP-bound G proteins in response to activating and inhibitory signals encountered by platelets. Thus, GEFs and GAPs are highly regulated themselves and appear to integrate G protein regulation with other cellular processes. This review focuses on GAPs of small G proteins of the Arf, Rab, Ras, and Rho families, as well as of heterotrimeric G proteins found in platelets.
Collapse
Affiliation(s)
- Lorna O'Donoghue
- UCD School of Medicine, University College Dublin, UCD Conway Institute, Belfield, Dublin 4, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, 123 St. Stephen’s Green 123, Dublin 2, Ireland
| | - Albert Smolenski
- UCD School of Medicine, University College Dublin, UCD Conway Institute, Belfield, Dublin 4, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, 123 St. Stephen’s Green 123, Dublin 2, Ireland
| |
Collapse
|
19
|
Kazemi N, Bordbar A, Bavarsad SS, Ghasemi P, Bakhshi M, Rezaeeyan H. Molecular Insights into the Relationship Between Platelet Activation and Endothelial Dysfunction: Molecular Approaches and Clinical Practice. Mol Biotechnol 2024; 66:932-947. [PMID: 38184492 DOI: 10.1007/s12033-023-01010-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 11/27/2023] [Indexed: 01/08/2024]
Abstract
Platelets are one of the coagulation cells. When platelet activation occurs, many mediators are released and affect endothelial cells (ECs) and lead to endothelial dysfunction (ED). ED plays an important role in the pathogenesis of many diseases, including cardiovascular disease (CVD). Platelet are of important factors in ED. The release of mediators by platelets causes the stimulation of inflammatory pathways, oxidative stress, and apoptosis, which ultimately result in ED.On the other hand, platelet activation in CVD patients can be associated with a bad prognosis. Platelet activation can increase the level of markers such as p-selectin in the serum. Also, in this study, we have discussed the role of platelet as a diagnostic factor, as well as its use as a treatment option. In addition, we discussed some of the molecular pathways that are used to target platelet activation.
Collapse
Affiliation(s)
- Niloufar Kazemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran
| | - Armin Bordbar
- Department of Cardiology, Musavi Hospital, School of Medicine, Zanjan University of Medical Science, Zanjan, Iran
| | | | - Parisa Ghasemi
- Research Committee, Medical School, Arak University of Medical Sciences, Arak, Iran
| | - Maryam Bakhshi
- Islamic Azad University of Najaf Abad, Affiliated Hospitals, Isfahan, Iran
| | - Hadi Rezaeeyan
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran.
| |
Collapse
|
20
|
Balykina A, Naida L, Kirkgöz K, Nikolaev VO, Fock E, Belyakov M, Whaley A, Whaley A, Shpakova V, Rukoyatkina N, Gambaryan S. Antiplatelet Effects of Flavonoid Aglycones Are Mediated by Activation of Cyclic Nucleotide-Dependent Protein Kinases. Int J Mol Sci 2024; 25:4864. [PMID: 38732081 PMCID: PMC11084604 DOI: 10.3390/ijms25094864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Flavonoid aglycones are secondary plant metabolites that exhibit a broad spectrum of pharmacological activities, including anti-inflammatory, antioxidant, anticancer, and antiplatelet effects. However, the precise molecular mechanisms underlying their inhibitory effect on platelet activation remain poorly understood. In this study, we applied flow cytometry to analyze the effects of six flavonoid aglycones (luteolin, myricetin, quercetin, eriodictyol, kaempferol, and apigenin) on platelet activation, phosphatidylserine externalization, formation of reactive oxygen species, and intracellular esterase activity. We found that these compounds significantly inhibit thrombin-induced platelet activation and decrease formation of reactive oxygen species in activated platelets. The tested aglycones did not affect platelet viability, apoptosis induction, or procoagulant platelet formation. Notably, luteolin, myricetin, quercetin, and apigenin increased thrombin-induced thromboxane synthase activity, which was analyzed by a spectrofluorimetric method. Our results obtained from Western blot analysis and liquid chromatography-tandem mass spectrometry demonstrated that the antiplatelet properties of the studied phytochemicals are mediated by activation of cyclic nucleotide-dependent signaling pathways. Specifically, we established by using Förster resonance energy transfer that the molecular mechanisms are, at least partly, associated with the inhibition of phosphodiesterases 2 and/or 5. These findings underscore the therapeutic potential of flavonoid aglycones for clinical application as antiplatelet agents.
Collapse
Affiliation(s)
- Anna Balykina
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg 194223, Russia; (A.B.); (E.F.); (A.W.); (N.R.)
- Faculty of General Medicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Lidia Naida
- Institute of Biomedical Systems and Biotechnologies, Peter the Great Saint Petersburg Polytechnic University, Saint Petersburg 195251, Russia;
| | - Kürsat Kirkgöz
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (K.K.); (V.O.N.)
| | - Viacheslav O. Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (K.K.); (V.O.N.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Ekaterina Fock
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg 194223, Russia; (A.B.); (E.F.); (A.W.); (N.R.)
| | - Michael Belyakov
- Research Institute of Hygiene, Occupational Pathology and Human Ecology, Saint Petersburg 188663, Russia;
| | - Anastasiia Whaley
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg 194223, Russia; (A.B.); (E.F.); (A.W.); (N.R.)
- Department of Pharmacognosy, Saint Petersburg State Chemical and Pharmaceutical University, Saint Petersburg 197022, Russia;
| | - Andrei Whaley
- Department of Pharmacognosy, Saint Petersburg State Chemical and Pharmaceutical University, Saint Petersburg 197022, Russia;
| | - Valentina Shpakova
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading RG6 6AS, UK;
| | - Natalia Rukoyatkina
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg 194223, Russia; (A.B.); (E.F.); (A.W.); (N.R.)
| | - Stepan Gambaryan
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg 194223, Russia; (A.B.); (E.F.); (A.W.); (N.R.)
| |
Collapse
|
21
|
Silva-Rojas R, Pérez-Guàrdia L, Simon A, Djeddi S, Treves S, Ribes A, Silva-Hernández L, Tard C, Laporte J, Böhm J. ORAI1 inhibition as an efficient preclinical therapy for tubular aggregate myopathy and Stormorken syndrome. JCI Insight 2024; 9:e174866. [PMID: 38516893 PMCID: PMC11063934 DOI: 10.1172/jci.insight.174866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 02/14/2024] [Indexed: 03/23/2024] Open
Abstract
Tubular aggregate myopathy (TAM) and Stormorken syndrome (STRMK) are clinically overlapping disorders characterized by childhood-onset muscle weakness and a variable occurrence of multisystemic signs, including short stature, thrombocytopenia, and hyposplenism. TAM/STRMK is caused by gain-of-function mutations in the Ca2+ sensor STIM1 or the Ca2+ channel ORAI1, both of which regulate Ca2+ homeostasis through the ubiquitous store-operated Ca2+ entry (SOCE) mechanism. Functional experiments in cells have demonstrated that the TAM/STRMK mutations induce SOCE overactivation, resulting in excessive influx of extracellular Ca2+. There is currently no treatment for TAM/STRMK, but SOCE is amenable to manipulation. Here, we crossed Stim1R304W/+ mice harboring the most common TAM/STRMK mutation with Orai1R93W/+ mice carrying an ORAI1 mutation partially obstructing Ca2+ influx. Compared with Stim1R304W/+ littermates, Stim1R304W/+Orai1R93W/+ offspring showed a normalization of bone architecture, spleen histology, and muscle morphology; an increase of thrombocytes; and improved muscle contraction and relaxation kinetics. Accordingly, comparative RNA-Seq detected more than 1,200 dysregulated genes in Stim1R304W/+ muscle and revealed a major restoration of gene expression in Stim1R304W/+Orai1R93W/+ mice. Altogether, we provide physiological, morphological, functional, and molecular data highlighting the therapeutic potential of ORAI1 inhibition to rescue the multisystemic TAM/STRMK signs, and we identified myostatin as a promising biomarker for TAM/STRMK in humans and mice.
Collapse
Affiliation(s)
- Roberto Silva-Rojas
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U1258, CNRS UMR7104, University of Strasbourg, Illkirch, France
| | - Laura Pérez-Guàrdia
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U1258, CNRS UMR7104, University of Strasbourg, Illkirch, France
| | - Alix Simon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U1258, CNRS UMR7104, University of Strasbourg, Illkirch, France
| | - Sarah Djeddi
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U1258, CNRS UMR7104, University of Strasbourg, Illkirch, France
| | - Susan Treves
- Departments of Neurology and Biomedicine, Basel University Hospital, Basel, Switzerland
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Agnès Ribes
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France
- Laboratory of Hematology, University Hospital of Toulouse, Toulouse, France
| | - Lorenzo Silva-Hernández
- Neurology Service, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Madrid, Spain
| | - Céline Tard
- University Lille, Inserm, CHU Lille, U1172 Lille Neuroscience & Cognition, Center for Rare Neuromuscular Diseases Nord/Est/Ile-de-France, Lille, France
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U1258, CNRS UMR7104, University of Strasbourg, Illkirch, France
| | - Johann Böhm
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U1258, CNRS UMR7104, University of Strasbourg, Illkirch, France
| |
Collapse
|
22
|
Manikanta K, Paul M, Sandesha VD, Mahalingam SS, Ramesh TN, Harishkumar K, Koundinya SS, Naveen S, Kemparaju K, Girish KS. Oxidative Stress-Induced Platelet Apoptosis/Activation: Alleviation by Purified Curcumin via ASK1-JNK/p-38 Pathway. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:417-430. [PMID: 38648762 DOI: 10.1134/s0006297924030039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/26/2023] [Accepted: 01/29/2024] [Indexed: 04/25/2024]
Abstract
Platelets are known for their indispensable role in hemostasis and thrombosis. However, alteration in platelet function due to oxidative stress is known to mediate various health complications, including cardiovascular diseases and other health complications. To date, several synthetic molecules have displayed antiplatelet activity; however, their uses are associated with bleeding and other adverse effects. The commercially available curcumin is generally a mixture of three curcuminoids: curcumin, demethoxycurcumin, and bisdemethoxycurcumin. Although crude curcumin is known to inhibit platelet aggregation, the effect of purified curcumin on platelet apoptosis, activation, and aggregation remains unclear. Therefore, in this study, curcumin was purified from a crude curcumin mixture and the effects of this preparation on the oxidative stress-induced platelet apoptosis and activation was evaluated. 2,2'-Azobis(2-methylpropionamidine) dihydrochloride (AAPH) compound was used as an inducer of oxidative stress. Purified curcumin restored AAPH-induced platelet apoptotic markers like reactive oxygen species, intracellular calcium level, mitochondrial membrane potential, cardiolipin peroxidation, cytochrome c release from mitochondria to the cytosol, and phosphatidyl serine externalization. Further, it inhibited the agonist-induced platelet activation and aggregation, demonstrating its antiplatelet activity. Western blot analysis confirms protective effect of the purified curcumin against oxidative stress-induced platelet apoptosis and activation via downregulation of MAPKs protein activation, including ASK1, JNK, and p-38. Together, these results suggest that the purified curcumin could be a potential therapeutic bioactive molecule to treat the oxidative stress-induced platelet activation, apoptosis, and associated complications.
Collapse
Affiliation(s)
- Kurnegala Manikanta
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, 570006, India
| | - Manoj Paul
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, 570006, India
| | | | - Shanmuga S Mahalingam
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Thimmasandra Narayan Ramesh
- Department of Studies and Research in Chemistry, University College of Science, Tumkur University, Tumakuru, 572103, India
| | | | - Shashank S Koundinya
- All India Institute of Medical Science, Sri Aurobindo Marg, Ansari Nagar, East, New Delhi, 110029, India
| | - Shivanna Naveen
- Applied Nutrition Discipline, Defense Food Research Laboratory, Mysuru, 570011, India
| | - Kempaiah Kemparaju
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, 570006, India.
| | - Kesturu S Girish
- Department of Studies and Research in Biochemistry, Tumkur University, Tumakuru, 572103, India.
| |
Collapse
|
23
|
Provenzale I, Solari FA, Schönichen C, Brouns SLN, Fernández DI, Kuijpers MJE, van der Meijden PEJ, Gibbins JM, Sickmann A, Jones C, Heemskerk JWM. Endothelium-mediated regulation of platelet activation: Involvement of multiple protein kinases. FASEB J 2024; 38:e23468. [PMID: 38334433 DOI: 10.1096/fj.202300360rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 01/11/2024] [Accepted: 01/23/2024] [Indexed: 02/10/2024]
Abstract
The endothelial regulation of platelet activity is incompletely understood. Here we describe novel approaches to find molecular pathways implicated on the platelet-endothelium interaction. Using high-shear whole-blood microfluidics, employing coagulant or non-coagulant conditions at physiological temperature, we observed that the presence of human umbilical vein endothelial cells (HUVEC) strongly suppressed platelet adhesion and activation, via the collagen receptor glycoprotein VI (GPVI) and the PAR receptors for thrombin. Real-time monitoring of the cytosolic Ca2+ rises in the platelets indicated no major improvement of inhibition by prostacyclin or nitric oxide. Similarly under stasis, exposure of isolated platelets to HUVEC reduced the Ca2+ responses by collagen-related peptide (CRP-XL, GPVI agonist) and thrombin (PAR agonist). We then analyzed the label-free phosphoproteome of platelets (three donors), exposed to HUVEC, CRP-XL, and/or thrombin. High-resolution mass spectrometry gave 5463 phosphopeptides, corresponding to 1472 proteins, with good correlation between biological and technical replicates (R > .86). Stringent filtering steps revealed 26 regulatory pathways (Reactome) and 143 regulated kinase substrates (PhosphoSitePlus), giving a set of protein phosphorylation sites that was differentially (44) or similarly (110) regulated by HUVEC or agonist exposure. The differential regulation was confirmed by stable-isotope analysis of platelets from two additional donors. Substrate analysis indicated major roles of poorly studied protein kinase classes (MAPK, CDK, DYRK, STK, PKC members). Collectively, these results reveal a resetting of the protein phosphorylation profile in platelets exposed to endothelium or to conventional agonists and to endothelium-promoted activity of a multi-kinase network, beyond classical prostacyclin and nitric oxide actors, that may contribute to platelet inhibition.
Collapse
Affiliation(s)
- Isabella Provenzale
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Fiorella A Solari
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Claudia Schönichen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Center for Thrombosis and Haemostasis, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sanne L N Brouns
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Delia I Fernández
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Marijke J E Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Paola E J van der Meijden
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
- Medizinische Fakultät, Medizinische Proteom-Center, Ruhr-Universität Bochum, Bochum, Germany
- Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, UK
| | - Chris Jones
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Synapse Research Institute Maastricht, Maastricht, The Netherlands
| |
Collapse
|
24
|
Jooss NJ, Diender MG, Fernández DI, Huang J, Heubel-Moenen FCJ, van der Veer A, Kuijpers MJE, Poulter NS, Henskens YMC, Te Loo M, Heemskerk JWM. Restraining of glycoprotein VI- and integrin α2β1-dependent thrombus formation by platelet PECAM1. Cell Mol Life Sci 2024; 81:44. [PMID: 38236412 PMCID: PMC10796532 DOI: 10.1007/s00018-023-05058-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/15/2023] [Accepted: 11/21/2023] [Indexed: 01/19/2024]
Abstract
The platelet receptors, glycoprotein VI (GPVI) and integrin α2β1 jointly control collagen-dependent thrombus formation via protein tyrosine kinases. It is unresolved to which extent the ITIM (immunoreceptor tyrosine-based inhibitory motif) receptor PECAM1 and its downstream acting protein tyrosine phosphatase PTPN11 interfere in this process. Here, we hypothesized that integrin α2β1 has a co-regulatory role in the PECAM1- and PTPN11-dependent restraint of thrombus formation. We investigated platelet activation under flow on collagens with a different GPVI dependency and using integrin α2β1 blockage. Blood was obtained from healthy subjects and from patients with Noonan syndrome with a gain-of-function mutation of PTPN11 and variable bleeding phenotype. On collagens with decreasing GPVI activity (types I, III, IV), the surface-dependent inhibition of PECAM1 did not alter thrombus parameters using control blood. Blockage of α2β1 generally reduced thrombus parameters, most effectively on collagen IV. Strikingly, simultaneous inhibition of PECAM1 and α2β1 led to a restoration of thrombus formation, indicating that the suppressing signaling effect of PECAM1 is masked by the platelet-adhesive receptor α2β1. Blood from 4 out of 6 Noonan patients showed subnormal thrombus formation on collagen IV. In these patients, effects of α2β1 blockage were counterbalanced by PECAM1 inhibition to a normal phenotype. In summary, we conclude that the suppression of GPVI-dependent thrombus formation by either PECAM1 or a gain-of-function of PTPN11 can be overruled by α2β1 engagement.
Collapse
Affiliation(s)
- Natalie J Jooss
- Department of Biochemistry, Maastricht University, Maastricht, The Netherlands
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Molecular Haematology Unit, University of Oxford, Headington, OX3 9DS, UK
| | - Marije G Diender
- Department of Pediatric Hematology, Amalia Children's Hospital, Radboud UMC, Nijmegen, The Netherlands
| | - Delia I Fernández
- Department of Biochemistry, Maastricht University, Maastricht, The Netherlands
- Platelet Proteomics Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - Jingnan Huang
- Department of Biochemistry, Maastricht University, Maastricht, The Netherlands
- Platelet Proteomics Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - Floor C J Heubel-Moenen
- Department of Internal Medicine, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Arian van der Veer
- Department of Pediatric Hematology, Amalia Children's Hospital, Radboud UMC, Nijmegen, The Netherlands
- Department of Pediatric Hematology, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | - Natalie S Poulter
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Nottingham, Midlands, UK
| | - Yvonne M C Henskens
- Central Diagnostic Laboratory, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Maroeska Te Loo
- Department of Pediatric Hematology, Amalia Children's Hospital, Radboud UMC, Nijmegen, The Netherlands
| | - Johan W M Heemskerk
- Department of Biochemistry, Maastricht University, Maastricht, The Netherlands.
- Synapse Research Institute Maastricht, Kon. Emmaplein 7, 6217 KD, Maastricht, The Netherlands.
| |
Collapse
|
25
|
Ye Y, Yang L, Leng M, Wang Q, Wu J, Wan W, Wang H, Li L, Peng Y, Chai S, Meng Z. Luteolin inhibits GPVI-mediated platelet activation, oxidative stress, and thrombosis. Front Pharmacol 2023; 14:1255069. [PMID: 38026984 PMCID: PMC10644720 DOI: 10.3389/fphar.2023.1255069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction: Luteolin inhibits platelet activation and thrombus formation, but the mechanisms are unclear. This study investigated the effects of luteolin on GPVI-mediated platelet activation in vitro and explored the effect of luteolin on thrombosis, coagulation, and platelet production in vivo. Methods: Washed human platelets were used for aggregation, membrane protein expression, ATP, Ca2+, and LDH release, platelet adhesion/spreading, and clot retraction experiments. Washed human platelets were used to detect collagen and convulxin-induced reactive oxygen species production and endogenous antioxidant effects. C57BL/6 male mice were used for ferric chloride-induced mesenteric thrombosis, collagen-epinephrine induced acute pulmonary embolism, tail bleeding, coagulation function, and luteolin toxicity experiments. The interaction between luteolin and GPVI was analyzed using solid phase binding assay and surface plasmon resonance (SPR). Results: Luteolin inhibited collagen- and convulxin-mediated platelet aggregation, adhesion, and release. Luteolin inhibited collagen- and convulxin-induced platelet ROS production and increased platelet endogenous antioxidant capacity. Luteolin reduced convulxin-induced activation of ITAM and MAPK signaling molecules. Molecular docking simulation showed that luteolin forms hydrogen bonds with GPVI. The solid phase binding assay showed that luteolin inhibited the interaction between collagen and GPVI. Surface plasmon resonance showed that luteolin bonded GPVI. Luteolin inhibited integrin αIIbβ3-mediated platelet activation. Luteolin inhibited mesenteric artery thrombosis and collagen- adrenergic-induced pulmonary thrombosis in mice. Luteolin decreased oxidative stress in vivo. Luteolin did not affect coagulation, hemostasis, or platelet production in mice. Discussion: Luteolin may be an effective and safe antiplatelet agent target for GPVI. A new mechanism (decreased oxidative stress) for the anti-platelet activity of luteolin has been identified.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Zhaohui Meng
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
26
|
Cooper ID, Kyriakidou Y, Edwards K, Petagine L, Seyfried TN, Duraj T, Soto-Mota A, Scarborough A, Jacome SL, Brookler K, Borgognoni V, Novaes V, Al-Faour R, Elliott BT. Ketosis Suppression and Ageing (KetoSAge): The Effects of Suppressing Ketosis in Long Term Keto-Adapted Non-Athletic Females. Int J Mol Sci 2023; 24:15621. [PMID: 37958602 PMCID: PMC10650498 DOI: 10.3390/ijms242115621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Most studies on ketosis have focused on short-term effects, male athletes, or weight loss. Hereby, we studied the effects of short-term ketosis suppression in healthy women on long-standing ketosis. Ten lean (BMI 20.5 ± 1.4), metabolically healthy, pre-menopausal women (age 32.3 ± 8.9) maintaining nutritional ketosis (NK) for > 1 year (3.9 years ± 2.3) underwent three 21-day phases: nutritional ketosis (NK; P1), suppressed ketosis (SuK; P2), and returned to NK (P3). Adherence to each phase was confirmed with daily capillary D-beta-hydroxybutyrate (BHB) tests (P1 = 1.9 ± 0.7; P2 = 0.1 ± 0.1; and P3 = 1.9 ± 0.6 pmol/L). Ageing biomarkers and anthropometrics were evaluated at the end of each phase. Ketosis suppression significantly increased: insulin, 1.78-fold from 33.60 (± 8.63) to 59.80 (± 14.69) pmol/L (p = 0.0002); IGF1, 1.83-fold from 149.30 (± 32.96) to 273.40 (± 85.66) µg/L (p = 0.0045); glucose, 1.17-fold from 78.6 (± 9.5) to 92.2 (± 10.6) mg/dL (p = 0.0088); respiratory quotient (RQ), 1.09-fold 0.66 (± 0.05) to 0.72 (± 0.06; p = 0.0427); and PAI-1, 13.34 (± 6.85) to 16.69 (± 6.26) ng/mL (p = 0.0428). VEGF, EGF, and monocyte chemotactic protein also significantly increased, indicating a pro-inflammatory shift. Sustained ketosis showed no adverse health effects, and may mitigate hyperinsulinemia without impairing metabolic flexibility in metabolically healthy women.
Collapse
Affiliation(s)
- Isabella D. Cooper
- Ageing Biology and Age-Related Diseases, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (Y.K.); (L.P.); (A.S.); (S.L.J.); (V.B.); (V.N.); (R.A.-F.); (B.T.E.)
| | - Yvoni Kyriakidou
- Ageing Biology and Age-Related Diseases, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (Y.K.); (L.P.); (A.S.); (S.L.J.); (V.B.); (V.N.); (R.A.-F.); (B.T.E.)
| | - Kurtis Edwards
- Cancer Biomarkers and Mechanisms Group, School of Life Sciences, University of Westminster, London W1W 6UW, UK;
| | - Lucy Petagine
- Ageing Biology and Age-Related Diseases, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (Y.K.); (L.P.); (A.S.); (S.L.J.); (V.B.); (V.N.); (R.A.-F.); (B.T.E.)
| | - Thomas N. Seyfried
- Biology Department, Boston College, Chestnut Hill, MA 02467, USA; (T.N.S.); (T.D.)
| | - Tomas Duraj
- Biology Department, Boston College, Chestnut Hill, MA 02467, USA; (T.N.S.); (T.D.)
| | - Adrian Soto-Mota
- Metabolic Diseases Research Unit, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City 14080, Mexico;
- Tecnologico de Monterrey, School of Medicine, Mexico City 14380, Mexico
| | - Andrew Scarborough
- Ageing Biology and Age-Related Diseases, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (Y.K.); (L.P.); (A.S.); (S.L.J.); (V.B.); (V.N.); (R.A.-F.); (B.T.E.)
| | - Sandra L. Jacome
- Ageing Biology and Age-Related Diseases, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (Y.K.); (L.P.); (A.S.); (S.L.J.); (V.B.); (V.N.); (R.A.-F.); (B.T.E.)
| | - Kenneth Brookler
- Retired former Research Collaborator, Aerospace Medicine and Vestibular Research Laboratory, Mayo Clinic, Scottsdale, AZ 85259, USA;
| | - Valentina Borgognoni
- Ageing Biology and Age-Related Diseases, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (Y.K.); (L.P.); (A.S.); (S.L.J.); (V.B.); (V.N.); (R.A.-F.); (B.T.E.)
| | - Vanusa Novaes
- Ageing Biology and Age-Related Diseases, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (Y.K.); (L.P.); (A.S.); (S.L.J.); (V.B.); (V.N.); (R.A.-F.); (B.T.E.)
| | - Rima Al-Faour
- Ageing Biology and Age-Related Diseases, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (Y.K.); (L.P.); (A.S.); (S.L.J.); (V.B.); (V.N.); (R.A.-F.); (B.T.E.)
| | - Bradley T. Elliott
- Ageing Biology and Age-Related Diseases, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (Y.K.); (L.P.); (A.S.); (S.L.J.); (V.B.); (V.N.); (R.A.-F.); (B.T.E.)
| |
Collapse
|
27
|
AlOuda SK, Sasikumar P, AlThunayan T, Alaajam F, Khan S, Sahli KA, Abohassan MS, Pollitt A, Jung SM, Gibbins JM. Role of heat shock protein 47 in platelet glycoprotein VI dimerization and signaling. Res Pract Thromb Haemost 2023; 7:102177. [PMID: 37767064 PMCID: PMC10520510 DOI: 10.1016/j.rpth.2023.102177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/23/2022] [Accepted: 07/21/2023] [Indexed: 09/29/2023] Open
Abstract
Background Heat shock protein 47 (HSP47) is an intracellular chaperone protein with an indispensable role in collagen biosynthesis in collagen-secreting cells. This chaperone has also been shown to be released and present on the surface of platelets. The inhibition of HSP47 in human platelets or its ablation in mouse platelets reduces platelet function in response to collagen and the glycoprotein (GP) VI collagen receptor agonist CRP-XL. Objectives In this study, we sought, through experiments, to explore cellular distribution, trafficking, and influence on GPVI interactions to understand how HSP47 modulates collagen receptor signaling. Methods HSP47-deficient mouse platelets and SMIH- treated human platelets were used to study the role of HSP47 in collagen mediated responses and signaling. Results Using subcellular fractionation analysis and immunofluorescence microscopy, HSP47 was found to be localized to the platelet-dense tubular system. Following platelet stimulation, HSP47 mobilization to the cell surface was shown to be dependent on actin polymerization, a feature common to other dense tubular system resident platelet proteins that are released to the cell surface during activation. In this location, HSP47 was found to contribute to platelet adhesion to collagen or CRP-XL but not to GFOGER peptide (an integrin α2β1-binding sequence within collagens), indicating selective effects of HSP47 on GPVI function. Dimerization of GPVI on the platelet surface increases its affinity for collagen. GPVI dimerization was reduced following HSP47 inhibition, as was collagen and CRP-XL-mediated signaling. Conclusion The present study identifies a role for cell surface-localized HSP47 in modulating platelet responses to collagen through dimerization of GPVI, thereby enhancing platelet signaling and activation.
Collapse
Affiliation(s)
- Sarah K. AlOuda
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Parvathy Sasikumar
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
- Centre for Haematology, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Taysseer AlThunayan
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Fahd Alaajam
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Gizan, Saudi Arabia
| | - Sabeeya Khan
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Khaled A. Sahli
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
- General Directorate of Medical Services, Ministry of Interior, Riyadh, Kingdom of Saudi Arabia
| | - Mohammed S. Abohassan
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Alice Pollitt
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Stephanie M. Jung
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Jonathan M. Gibbins
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| |
Collapse
|
28
|
Nock S, Karim E, Unsworth AJ. Pim Kinases: Important Regulators of Cardiovascular Disease. Int J Mol Sci 2023; 24:11582. [PMID: 37511341 PMCID: PMC10380471 DOI: 10.3390/ijms241411582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Pim Kinases; Pim-1, Pim-2, and Pim-3, are a family of constitutively active serine/threonine kinases, widely associated with cell survival, proliferation, and migration. Historically considered to be functionally redundant, independent roles for the individual isoforms have been described. Whilst most established for their role in cancer progression, there is increasing evidence for wider pathological roles of Pim kinases within the context of cardiovascular disease, including inflammation, thrombosis, and cardiac injury. The Pim kinase isoforms have widespread expression in cardiovascular tissues, including the heart, coronary artery, aorta, and blood, and have been demonstrated to be upregulated in several co-morbidities/risk factors for cardiovascular disease. Pim kinase inhibition may thus be a desirable therapeutic for a multi-targeted approach to treat cardiovascular disease and some of the associated risk factors. In this review, we discuss what is known about Pim kinase expression and activity in cells of the cardiovascular system, identify areas where the role of Pim kinase has yet to be fully explored and characterised and review the suitability of targeting Pim kinase for the prevention and treatment of cardiovascular events in high-risk individuals.
Collapse
Affiliation(s)
| | | | - Amanda J. Unsworth
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester M1 5GD, UK
| |
Collapse
|
29
|
Graca FA, Stephan A, Minden-Birkenmaier BA, Shirinifard A, Wang YD, Demontis F, Labelle M. Platelet-derived chemokines promote skeletal muscle regeneration by guiding neutrophil recruitment to injured muscles. Nat Commun 2023; 14:2900. [PMID: 37217480 DOI: 10.1038/s41467-023-38624-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 05/09/2023] [Indexed: 05/24/2023] Open
Abstract
Skeletal muscle regeneration involves coordinated interactions between different cell types. Injection of platelet-rich plasma is circumstantially considered an aid to muscle repair but whether platelets promote regeneration beyond their role in hemostasis remains unexplored. Here, we find that signaling via platelet-released chemokines is an early event necessary for muscle repair in mice. Platelet depletion reduces the levels of the platelet-secreted neutrophil chemoattractants CXCL5 and CXCL7/PPBP. Consequently, early-phase neutrophil infiltration to injured muscles is impaired whereas later inflammation is exacerbated. Consistent with this model, neutrophil infiltration to injured muscles is compromised in male mice with Cxcl7-knockout platelets. Moreover, neo-angiogenesis and the re-establishment of myofiber size and muscle strength occurs optimally in control mice post-injury but not in Cxcl7ko mice and in neutrophil-depleted mice. Altogether, these findings indicate that platelet-secreted CXCL7 promotes regeneration by recruiting neutrophils to injured muscles, and that this signaling axis could be utilized therapeutically to boost muscle regeneration.
Collapse
Affiliation(s)
- Flavia A Graca
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Anna Stephan
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Benjamin A Minden-Birkenmaier
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Oncology, Division of Molecular Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Abbas Shirinifard
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yong-Dong Wang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Fabio Demontis
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | - Myriam Labelle
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
- Department of Oncology, Division of Molecular Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
30
|
Garcia C, Dejean S, Savy N, Bordet JC, Series J, Cadot S, Ribes A, Voisin S, Rugeri L, Payrastre B, Sié P. Multicolor flow cytometry in clinical samples for platelet signaling assessment. Res Pract Thromb Haemost 2023; 7:100180. [PMID: 37538502 PMCID: PMC10394564 DOI: 10.1016/j.rpth.2023.100180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 05/01/2023] [Accepted: 05/05/2023] [Indexed: 08/05/2023] Open
Abstract
Background Availability of multichannel cytometers and specific commercial antibodies makes flow cytometry a new option to simultaneously assess multiple intracellular platelet signaling pathways for clinical purposes, in small volume of blood or low platelet count. Objectives To describe a multicolor flow cytometry with fluorescent barcoding technique for screening signaling pathways downstream membrane receptors of major platelet agonists (adenosine diphosphate, thrombin, thromboxane, and collagen). Methods By comparison with immunoblotting, we first selected the target phosphoproteins, AKT, P38MAPK, LIMK, and SPL76; the times of stimulation; and phosphoflow barcoding conditions. We then performed a clinical study on whole blood of patients without evidence of blood platelet disorder on standard biological screening, consulting for trivial or occasionally provoked bleeds without familial antecedent (bleeding of unknown origin, n = 23) or type-1 von Willebrand disease (n = 9). In addition, we included a small group of patients with definite platelet disorders (Glanzmann thrombasthenia, δ-storage pool deficiency, and immune glycoprotein VI-related disease with granule secretion defect). Results The range, kinetics, and distribution of fluorescence intensity were established for each agonist-target protein combination. Principal component analysis indicates a correlation in response to a target phosphoprotein (AKT and P38MAPK) to different agonists but no correlation in the response of different target phosphoproteins to the same agonist. The heterogeneity of individual responses in the whole population displayed was analyzed using clustering algorithm. Patients with platelet storage pool deficiency were positioned as lowest responders on the heatmap. Conclusion In complement of functional tests, this study introduces a new approach for rapid platelet signaling profiling in clinical practice.
Collapse
Affiliation(s)
- Cedric Garcia
- CHU de Toulouse, Laboratoire d’Hématologie, Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires INSERM U1048, Université de Toulouse, Toulouse, France
| | - Sebastien Dejean
- Université Paul Sabatier Toulouse III, Institut de Mathématiques, CNRS UMR 5219, Toulouse, France
| | - Nicolas Savy
- Université Paul Sabatier Toulouse III, Institut de Mathématiques, CNRS UMR 5219, Toulouse, France
| | - Jean-Claude Bordet
- Laboratoire d’Hématologie, Hospices Civiles de Lyon, Lyon, France
- EA 4609-Hémostase et Cancer, Université Claude Bernard Lyon 1, Lyon, France
| | - Jennifer Series
- Institut des Maladies Métaboliques et Cardiovasculaires INSERM U1048, Université de Toulouse, Toulouse, France
| | - Sarah Cadot
- Institut des Maladies Métaboliques et Cardiovasculaires INSERM U1048, Université de Toulouse, Toulouse, France
| | - Agnès Ribes
- CHU de Toulouse, Laboratoire d’Hématologie, Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires INSERM U1048, Université de Toulouse, Toulouse, France
- Faculté de Médecine, Université Paul Sabatier Toulouse III, Toulouse, France
| | - Sophie Voisin
- CHU de Toulouse, Laboratoire d’Hématologie, Toulouse, France
| | - Lucia Rugeri
- Laboratoire d’Hématologie, Hospices Civiles de Lyon, Lyon, France
- Hospices Civils de Lyon, Unité d’Hémostase clinique, Bron, France
| | - Bernard Payrastre
- CHU de Toulouse, Laboratoire d’Hématologie, Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires INSERM U1048, Université de Toulouse, Toulouse, France
- Faculté de Médecine, Université Paul Sabatier Toulouse III, Toulouse, France
| | - Pierre Sié
- CHU de Toulouse, Laboratoire d’Hématologie, Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires INSERM U1048, Université de Toulouse, Toulouse, France
- Université Paul Sabatier Toulouse III, Faculté de Pharmacie, Toulouse, France
| |
Collapse
|
31
|
Hernández-García S, Flores-García M, Maldonado-Vega M, Hernández G, Meneses-Melo F, López-Vanegas NC, Calderón-Salinas JV. Adaptive changes in redox response and decreased platelet aggregation in lead-exposed workers. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 100:104134. [PMID: 37116628 DOI: 10.1016/j.etap.2023.104134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 03/11/2023] [Accepted: 04/24/2023] [Indexed: 05/06/2023]
Abstract
Chronic lead exposure can generate pro-oxidative and pro-inflammatory conditions in the blood, related to high platelet activation and aggregation, altering cell functions. We studied ADP-stimulated aggregation and the oxidant/antioxidant system of platelets from chronically lead-exposed workers and non-exposed workers. Platelet aggregation was low in lead-exposed workers (62 vs. 97%), who had normal platelet counts and showed no clinical manifestations of hemostatic failure. ADP-activated platelets from lead-exposed workers failed to increase superoxide release (3.3 vs. 6.6 µmol/g protein), had low NADPH concentration (60 vs. 92 nmol/mg protein), high concentration of hydrogen peroxide (224 vs. 129 nmol/mg protein) and high plasma PGE2 concentration (287 vs. 79 pg/mL). Altogether, those conditions, on the one hand, could account for the low platelet aggregation and, on the other, indicate an adaptive mechanism for the oxidative status of platelets and anti-aggregating molecules to prevent thrombotic problems in the pro-oxidant and pro-inflammatory environment of chronic lead exposure.
Collapse
Affiliation(s)
- Sandra Hernández-García
- Biochemistry Department, Centro de Investigación y de Estudios Avanzados-IPN (Cinvestav), Mexico City, Mexico
| | - Mirthala Flores-García
- Molecular Biology Department, Instituto Nacional de Cardiología "Dr. Ignacio Chávez", Mexico City, Mexico
| | - María Maldonado-Vega
- Planning, Teaching and Research Department, Hospital Regional de Alta Especialidad del Bajío. León, Guanajuato, Mexico
| | - Gerardo Hernández
- Section Methodology of Science, Centro de Investigación y de Estudios Avanzados-IPN (Cinvestav), Mexico City, Mexico
| | | | | | | |
Collapse
|
32
|
Li J, Liang Y. Associations Between Mean Platelet Volume and Risk of Deep Vein Thrombosis: A Mendelian Randomization Study and a Retrospective Study. Int J Gen Med 2023; 16:515-524. [PMID: 36789132 PMCID: PMC9922513 DOI: 10.2147/ijgm.s401059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 01/18/2023] [Indexed: 02/11/2023] Open
Abstract
Objective In this study, it was intended to explore the causal association between mean platelet volume (MPV) and risk of deep vein thrombosis (DVT) using a two-sample Mendelian randomization (MR) analysis and a retrospective study. Methods This study applied two-sample MR analysis to estimate the causal association between MPV and risk of DVT. Twenty-one single nucleotide polymorphisms (SNPs) were selected as genetic variants from two independent consortiums cohorts (p-value <5×10-8, linkage disequilibrium r2<0.1). Inverse variance weighted (IVW), MR-egger method and weighted median were performed. A retrospective study was also conducted to verify the associations identified from the MR study. Results The MR analysis demonstrated that genetically predicted higher MPV was associated with significantly lower risk of DVT (OR 0.982, 95% CI = 0.967-0.998, P = 0.023), with the consistent result in weighted median and MR-Egger. There was no directional horizontal pleiotropy in the method of MR-Egger regression (intercept=2.9e-04, P = 0.194). There was no single SNP was found to strongly drive the combined causal effect in the leave-one-out sensitivity analysis. Additionally, the similar result was observed in the retrospective study. Conclusion This study suggested that MPV was negatively associated with the risk of DVT. More basic researches are needed in the future to explore its specific mechanism.
Collapse
Affiliation(s)
- Jianhong Li
- Department of Radiology, Foshan Hospital of Traditional Chinese Medicine, Foshan, People’s Republic of China
| | - Yingna Liang
- Department of Gynaecology and Obstetrics, Guangzhou Baiyun First People’s Hospital (Baiyun District Maternal and Child Health Hospital), Guangzhou, Guangdong Province, People’s Republic of China,Correspondence: Yingna Liang, Department of Gynaecology and Obstetrics, Guangzhou Baiyun First People’s Hospital (Baiyun District Maternal and Child Health Hospital), Guangzhou, Guangdong Province, People’s Republic of China, Email
| |
Collapse
|
33
|
Semenza GL. Regulation of Erythropoiesis by the Hypoxia-Inducible Factor Pathway: Effects of Genetic and Pharmacological Perturbations. Annu Rev Med 2023; 74:307-319. [PMID: 35773226 DOI: 10.1146/annurev-med-042921-102602] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Red blood cells transport O2 from the lungs to body tissues. Hypoxia stimulates kidney cells to secrete erythropoietin (EPO), which increases red cell mass. Hypoxia-inducible factors (HIFs) mediate EPO gene transcriptional activation. HIF-α subunits are subject to O2-dependent prolyl hydroxylation and then bound by the von Hippel-Lindau protein (VHL), which triggers their ubiquitination and proteasomal degradation. Mutations in the genes encoding EPO, EPO receptor, HIF-2α, prolyl hydroxylase domain protein 2 (PHD2), or VHL cause familial erythrocytosis. In addition to O2, α-ketoglutarate is a substrate for PHD2, and analogs of α-ketoglutarate inhibit hydroxylase activity. In phase III clinical trials evaluating the treatment of anemia in chronic kidney disease, HIF prolyl hydroxylase inhibitors were as efficacious as darbepoetin alfa in stimulating erythropoiesis. However, safety concerns have arisen that are focused on thromboembolism, which is also a phenotypic manifestation of VHL or HIF-2α mutation, suggesting that these events are on-target effects of HIF prolyl hydroxylase inhibitors.
Collapse
Affiliation(s)
- Gregg L Semenza
- McKusick-Nathans Department of Genetic Medicine and Vascular Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA;
| |
Collapse
|
34
|
Yin B, Zhang S, Huang Y, Long Y, Chen Y, Zhao S, Zhou A, Cao M, Yin X, Luo D. The antithrombosis effect of dehydroandrographolide succinate: in vitro and in vivo studies. PHARMACEUTICAL BIOLOGY 2022; 60:175-184. [PMID: 35014931 PMCID: PMC8757605 DOI: 10.1080/13880209.2021.2021948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 11/18/2021] [Accepted: 12/19/2021] [Indexed: 06/14/2023]
Abstract
CONTEXT Dehydroandrographolide succinate (DAS) is mainly used in the clinical treatment of various infectious diseases. Its potential effects on platelet aggregation and blood coagulation systems have not been reported systematically. OBJECTIVE To explore whether DAS exerts an antithrombotic effect and its internal mechanism. MATERIALS AND METHODS Human blood samples and Sprague-Dawley (SD) rats divided into control, aspirin (30 mg/kg), and DAS groups (200, 400 and 600 mg/kg) were used to measure the platelet aggregation rate, coagulation function, coagulation factor activity, and contents of thromboxane B2 (TXB2) and 6-keto-prostaglandin F1α (6-keto-PGF1α). The histopathology of the SD rat gastric mucosa was also observed. All rats were administered intragastric or intraperitoneal injections once a day for 3 consecutive days. RESULTS Compared to control group, DAS significantly inhibited the platelet aggregation rate (ED50 = 386.9 mg/kg) by decreasing TXB2 levels (1531.95 ± 649.90 pg/mL to 511.08 ± 411.82 pg/mL) and activating antithrombin III (AT-III) (103.22 ± 16.22% to 146.46 ± 8.96%) (p < 0.05). In addition, DAS significantly enhanced the coagulation factors FV (304.12 ± 79.65% to 443.44 ± 75.04%), FVII (324.19 ± 48.03% to 790.66 ± 225.56%), FVIII (524.79 ± 115.47% to 679.92 ± 143.34%), FX (34.90 ± 7.40% to 102.76 ± 29.41%) and FXI (38.12 ± 10.33% to 65.47 ± 34.08%), increased the content of Fg (2.18 ± 0.39 to 3.61 ± 0.37 g/L), shorten the PT (10.42 ± 0.44 to 9.22 ± 0.21 s), APTT (16.43 ± 1.4 to 14.07 ± 0.75 s) and TT time (37.04 ± 2.13 to 32.68 ± 1.29 s) (p < 0.05), while the aspirin group showed no such effect on these items but showed reduced activity of FII (89.21 ± 21.72% to 61.83 ± 8.95%) and FVIII (524.79 ± 115.47% to 306.60 ± 29.96%) (p < 0.05). Histopathological changes showed aspirin-induced gastric mucosa haemorrhage and the protective effect of DAS in the gastric mucosa. CONCLUSIONS DAS is more suitable than aspirin in thromboprophylaxis treatment, which provides a reliable theoretical and experimental basis for its clinical application.
Collapse
Affiliation(s)
- Bowen Yin
- Clinical Laboratory, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Shuhua Zhang
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Yuxi Huang
- Dalian Key Laboratory of Marine Animal Disease Control and Prevention, College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - Yuanzhu Long
- Nanchang Maternal and Child Health Care Family Planning Service Centre, Nanchang, China
| | - Yiguo Chen
- Clinical Laboratory, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Shiyun Zhao
- Chinese Medicine Research Institute, Academy of Jiangxi Provincial Traditional Chinese Medicine, Nanchang
| | - Aiqun Zhou
- Clinical Laboratory, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Minghua Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Xiaoming Yin
- Clinical Laboratory, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, China
| |
Collapse
|
35
|
Platelet-Neutrophil Association in NETs-Rich Areas in the Retrieved AIS Patient Thrombi. Int J Mol Sci 2022; 23:ijms232214477. [PMID: 36430952 PMCID: PMC9694992 DOI: 10.3390/ijms232214477] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/23/2022] Open
Abstract
Histological structure of thrombi is a strong determinant of the outcome of vascular recanalization therapy, the only treatment option for acute ischemic stroke (AIS) patients. A total of 21 AIS patients from this study after undergoing non-enhanced CT scan and multimodal MRI were treated with mechanical stent-based and manual aspiration thrombectomy, and thromboembolic retrieved from a cerebral artery. Complementary histopathological and imaging analyses were performed to understand their composition with a specific focus on fibrin, von Willebrand factor, and neutrophil extracellular traps (NETs). Though distinct RBC-rich and platelet-rich areas were found, AIS patient thrombi were overwhelmingly platelet-rich, with 90% of thrombi containing <40% total RBC-rich contents (1.5 to 37%). Structurally, RBC-rich areas were simple, consisting of tightly packed RBCs in thin fibrin meshwork with sparsely populated nucleated cells and lacked any substantial von Willebrand factor (VWF). Platelet-rich areas were structurally more complex with thick fibrin meshwork associated with VWF. Plenty of leukocytes populated the platelet-rich areas, particularly in the periphery and border areas between platelet-rich and RBC-rich areas. Platelet-rich areas showed abundant activated neutrophils (myeloperoxidase+ and neutrophil-elastase+) containing citrullinated histone-decorated DNA. Citrullinated histone-decorated DNA also accumulated extracellularly, pointing to NETosis by the activated neutrophils. Notably, NETs-containing areas showed strong reactivity to VWF, platelets, and high-mobility group box 1 (HMGB1), signifying a close interplay between these components.
Collapse
|
36
|
Ormazabal P, Rodriguez L, Paredes A, Morales G, Fuentes E, Palomo I. Antiplatelet activity of Lampaya medicinalis Phil. in human platelets. NFS JOURNAL 2022. [DOI: 10.1016/j.nfs.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
37
|
Roweth HG, Malloy MW, Goreczny GJ, Becker IC, Guo Q, Mittendorf EA, Italiano JE, McAllister SS, Battinelli EM. Pro-inflammatory megakaryocyte gene expression in murine models of breast cancer. SCIENCE ADVANCES 2022; 8:eabo5224. [PMID: 36223471 PMCID: PMC9555784 DOI: 10.1126/sciadv.abo5224] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 08/24/2022] [Indexed: 06/16/2023]
Abstract
Despite abundant research demonstrating that platelets can promote tumor cell metastasis, whether primary tumors affect platelet-producing megakaryocytes remains understudied. In this study, we used a spontaneous murine model of breast cancer to show that tumor burden reduced megakaryocyte number and size and disrupted polyploidization. Single-cell RNA sequencing demonstrated that megakaryocytes from tumor-bearing mice exhibit a pro-inflammatory phenotype, epitomized by increased Ctsg, Lcn2, S100a8, and S100a9 transcripts. Protein S100A8/A9 and lipocalin-2 levels were also increased in platelets, suggesting that tumor-induced alterations to megakaryocytes are passed on to their platelet progeny, which promoted in vitro tumor cell invasion and tumor cell lung colonization to a greater extent than platelets from wild-type animals. Our study is the first to demonstrate breast cancer-induced alterations in megakaryocytes, leading to qualitative changes in platelet content that may feedback to promote tumor metastasis.
Collapse
Affiliation(s)
- Harvey G. Roweth
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Michael W. Malloy
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Gregory J. Goreczny
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Isabelle C. Becker
- Harvard Medical School, Boston, MA 02115, USA
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Qiuchen Guo
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Elizabeth A. Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Breast Oncology Program, Dana-Farber/Brigham and Women’s Cancer Center, Boston, MA 02215, USA
- Ludwig Centre for Cancer Research at Harvard, Harvard Medical School, Boston, MA 02215, USA
| | - Joseph E. Italiano
- Harvard Medical School, Boston, MA 02115, USA
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Sandra S. McAllister
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Elisabeth M. Battinelli
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
38
|
Chen SH, Tsai SC, Lu HC. Platelets as a Gauge of Liver Disease Kinetics? Int J Mol Sci 2022; 23:11460. [PMID: 36232759 PMCID: PMC9569526 DOI: 10.3390/ijms231911460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/22/2022] [Accepted: 09/25/2022] [Indexed: 11/18/2022] Open
Abstract
A multitude of laboratory and clinical interferences influence the utility of platelet-based diagnostic indices, including immature platelet fraction, in longitudinal monitoring and prognostication of patients with chronic liver disease (CLD). The complex yet highly regulated molecular basis of platelet production and clearance kinetics becomes dysregulated in liver pathogenesis. These underlying molecular mechanisms, including premature platelet clearance and bone marrow suppression in parallel with the progressive (e.g., treatment-naïve) or regressive (e.g., on-treatment and off-treatment) disease courses, involved in CLDs, may further confound the changes in platelet-liver correlations over time. Platelet count and function are commonly and secondarily altered in vivo in CLDs. However, the precise characterization of platelet functions during cirrhosis, including in vitro platelet aggregation, has proven challenging due to interferences such as thrombocytopenia. A flow cytometric approach may help monitor the unstably rebalanced hyper- and hypoaggregable states in patients with cirrhosis at risk of hyperaggregable, prothrombotic, or bleeding events. Studies have attempted to stratify patients with cirrhosis by substages and prognosis through the use of novel indices such as the ratio of in vitro endogenous platelet aggregation to platelet count. This review attempts to highlight clinical and laboratory precautions in the context of platelet-assisted CLD monitoring.
Collapse
Affiliation(s)
- Sheng-Hung Chen
- Department of Medicine, China Medical University, No. 91, Xueshi Road, Taichung 404333, Taiwan
- Center for Digestive Medicine, Department of Internal Medicine, China Medical University Hospital, No. 2, Yude Road, Taichung 404327, Taiwan
| | - Shih-Chang Tsai
- Department of Biological Science and Technology, China Medical University, Taichung 404333, Taiwan
| | - Hsiu-Chen Lu
- Department of Education, China Medical University Hospital, Taichung 404327, Taiwan
| |
Collapse
|
39
|
Jain K, Tyagi T, Du J, Hu X, Patell K, Martin KA, Hwa J. Unfolded Protein Response Differentially Modulates the Platelet Phenotype. Circ Res 2022; 131:290-307. [PMID: 35862006 PMCID: PMC9357223 DOI: 10.1161/circresaha.121.320530] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 07/06/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Unfolded protein response (UPR) is a multifaceted signaling cascade that alleviates protein misfolding. Although well studied in nucleated cells, UPR in absence of transcriptional regulation has not been described. Intricately associated with cardiovascular diseases, platelets, despite being anucleate, respond rapidly to stressors in blood. We investigate the UPR in anucleate platelets and explore its role, if any, on platelet physiology and function. METHODS Human and mouse platelets were studied using a combination of ex vivo and in vivo experiments. Platelet lineage-specific knockout mice were generated independently for each of the 3 UPR pathways, PERK (protein kinase RNA [PKR]-like endoplasmic reticulum kinase), XBP1 (X-binding protein), and ATF6 (activating transcription factor 6). Diabetes patients were prospectively recruited, and platelets were evaluated for activation of UPR under chronic pathophysiological disease conditions. RESULTS Tunicamycin induced the IRE1α (inositol-requiring enzyme-1alpha)-XBP1 pathway in human and mouse platelets, while oxidative stress predominantly activated the PERK pathway. PERK deletion significantly increased platelet aggregation and apoptosis and phosphorylation of PLCγ2, PLCβ3, and p38 MAPK. Deficiency of XBP1 increased platelet aggregation, with higher PLCβ3 and PKCδ activation. ATF6 deletion mediated a relatively modest effect on platelet phenotype with increased PKA (protein kinase A). Platelets from diabetes patients exhibited a positive correlation between disease severity, platelet activation, and protein aggregation, with only IRE1α-XBP1 activation. Moreover, IRE1α inhibition increased platelet aggregation, while clinically approved chemical chaperone, sodium 4-phenylbutyrate reduced the platelet hyperactivation. CONCLUSIONS We show for the first time, that UPR activation occurs in platelets and can be independent of genomic regulation, with selective induction being specific to the source and severity of stress. Each UPR pathway plays a key role and can differentially modulate the platelet activation pathways and phenotype. Targeting the specific arms of UPR may provide a new antiplatelet strategy to mitigate thrombotic risk in diabetes and other cardiovascular diseases.
Collapse
Affiliation(s)
- Kanika Jain
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 George Street, Room 759, New Haven, CT 06511
| | - Tarun Tyagi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 George Street, Room 759, New Haven, CT 06511
| | - Jing Du
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 George Street, Room 759, New Haven, CT 06511
| | - Xiaoyue Hu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 George Street, Room 759, New Haven, CT 06511
| | - Kanchi Patell
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 George Street, Room 759, New Haven, CT 06511
| | - Kathleen A. Martin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 George Street, Room 759, New Haven, CT 06511
| | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 George Street, Room 759, New Haven, CT 06511
| |
Collapse
|
40
|
Kriek N, Nock SH, Sage T, Khalifa B, Bye AP, Mitchell JL, Thomson S, McLaughlin MG, Jones S, Gibbins JM, Unsworth AJ. Cucurbitacins Elicit Anti-Platelet Activity via Perturbation of the Cytoskeleton and Integrin Function. Thromb Haemost 2022; 122:1115-1129. [PMID: 35253142 PMCID: PMC9385249 DOI: 10.1055/a-1788-5322] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 11/06/2021] [Indexed: 11/23/2022]
Abstract
Cucurbitacins are dietary compounds that have been shown to elicit a range of anti-tumour, anti-inflammatory and anti-atherosclerotic activities. Originally identified as signal transducer and activator of transcription, STAT, inhibitors, a variety of mechanisms of action have since been described, including dysregulation of the actin cytoskeleton and disruption of integrin function. Integrin outside-in signalling and cytoskeletal rearrangements are critical for the propagation of stable thrombus formation and clot retraction following platelet adhesion at the site of vessel damage. The effects of cucurbitacins on platelet function and thrombus formation are unknown. We report for the first time anti-platelet and anti-thrombotic effects of cucurbitacins B, E and I in human platelets. Treatment of platelets with cucurbitacins resulted in attenuation of platelet aggregation, secretion and fibrinogen binding following stimulation by platelet agonists. Cucurbitacins were also found to potently inhibit other integrin- and cytoskeleton-mediated events, including adhesion, spreading and clot retraction. Further investigation of cytoskeletal dynamics found treatment with cucurbitacins altered cofilin phosphorylation, enhanced activation and increased F actin polymerisation and microtubule assembly. Disruption to cytoskeletal dynamics has been previously shown to impair integrin activation, platelet spreading and clot retraction. Anti-platelet properties of cucurbitacins were found to extend to a disruption of stable thrombus formation, with an increase in thrombi instability and de-aggregation under flow. Our research identifies novel, anti-platelet and anti-thrombotic actions of cucurbitacins that appear to be linked to dysregulation of cytoskeletal dynamics and integrin function.
Collapse
Affiliation(s)
- Neline Kriek
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Sophie H. Nock
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Tanya Sage
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Badrija Khalifa
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Alexander P. Bye
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Joanne L. Mitchell
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Steven Thomson
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Mark G. McLaughlin
- Department of Chemistry, Lancaster University, Lancaster, United Kingdom
| | - Sarah Jones
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Jonathan M. Gibbins
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Amanda J. Unsworth
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| |
Collapse
|
41
|
ACKR3 regulates platelet activation and ischemia-reperfusion tissue injury. Nat Commun 2022; 13:1823. [PMID: 35383158 PMCID: PMC8983782 DOI: 10.1038/s41467-022-29341-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/25/2022] [Indexed: 12/15/2022] Open
Abstract
Platelet activation plays a critical role in thrombosis. Inhibition of platelet activation is a cornerstone in treatment of acute organ ischemia. Platelet ACKR3 surface expression is independently associated with all-cause mortality in CAD patients. In a novel genetic mouse strain, we show that megakaryocyte/platelet-specific deletion of ACKR3 results in enhanced platelet activation and thrombosis in vitro and in vivo. Further, we performed ischemia/reperfusion experiments (transient LAD-ligation and tMCAO) in mice to assess the impact of genetic ACKR3 deficiency in platelets on tissue injury in ischemic myocardium and brain. Loss of platelet ACKR3 enhances tissue injury in ischemic myocardium and brain and aggravates tissue inflammation. Activation of platelet-ACKR3 via specific ACKR3 agonists inhibits platelet activation and thrombus formation and attenuates tissue injury in ischemic myocardium and brain. Here we demonstrate that ACKR3 is a critical regulator of platelet activation, thrombus formation and organ injury following ischemia/reperfusion. ACKR3 is a critical regulator of platelet-mediated thrombosis and organ injury following ischemia/reperfusion. Platelet ACKR3 surface expression is independently associated with all-cause mortality in patients with cardiovascular diseases.
Collapse
|
42
|
Veuthey L, Aliotta A, Bertaggia Calderara D, Pereira Portela C, Alberio L. Mechanisms Underlying Dichotomous Procoagulant COAT Platelet Generation-A Conceptual Review Summarizing Current Knowledge. Int J Mol Sci 2022; 23:2536. [PMID: 35269679 PMCID: PMC8910683 DOI: 10.3390/ijms23052536] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 12/23/2022] Open
Abstract
Procoagulant platelets are a subtype of activated platelets that sustains thrombin generation in order to consolidate the clot and stop bleeding. This aspect of platelet activation is gaining more and more recognition and interest. In fact, next to aggregating platelets, procoagulant platelets are key regulators of thrombus formation. Imbalance of both subpopulations can lead to undesired thrombotic or bleeding events. COAT platelets derive from a common pro-aggregatory phenotype in cells capable of accumulating enough cytosolic calcium to trigger specific pathways that mediate the loss of their aggregating properties and the development of new adhesive and procoagulant characteristics. Complex cascades of signaling events are involved and this may explain why an inter-individual variability exists in procoagulant potential. Nowadays, we know the key agonists and mediators underlying the generation of a procoagulant platelet response. However, we still lack insight into the actual mechanisms controlling this dichotomous pattern (i.e., procoagulant versus aggregating phenotype). In this review, we describe the phenotypic characteristics of procoagulant COAT platelets, we detail the current knowledge on the mechanisms of the procoagulant response, and discuss possible drivers of this dichotomous diversification, in particular addressing the impact of the platelet environment during in vivo thrombus formation.
Collapse
Affiliation(s)
| | | | | | | | - Lorenzo Alberio
- Hemostasis and Platelet Research Laboratory, Division of Hematology and Central Hematology Laboratory, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), CH-1010 Lausanne, Switzerland; (L.V.); (A.A.); (D.B.C.); (C.P.P.)
| |
Collapse
|
43
|
Platelet Membrane: An Outstanding Factor in Cancer Metastasis. MEMBRANES 2022; 12:membranes12020182. [PMID: 35207103 PMCID: PMC8875259 DOI: 10.3390/membranes12020182] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/02/2022]
Abstract
In addition to being biological barriers where the internalization or release of biomolecules is decided, cell membranes are contact structures between the interior and exterior of the cell. Here, the processes of cell signaling mediated by receptors, ions, hormones, cytokines, enzymes, growth factors, extracellular matrix (ECM), and vesicles begin. They triggering several responses from the cell membrane that include rearranging its components according to the immediate needs of the cell, for example, in the membrane of platelets, the formation of filopodia and lamellipodia as a tissue repair response. In cancer, the cancer cells must adapt to the new tumor microenvironment (TME) and acquire capacities in the cell membrane to transform their shape, such as in the case of epithelial−mesenchymal transition (EMT) in the metastatic process. The cancer cells must also attract allies in this challenging process, such as platelets, fibroblasts associated with cancer (CAF), stromal cells, adipocytes, and the extracellular matrix itself, which limits tumor growth. The platelets are enucleated cells with fairly interesting growth factors, proangiogenic factors, cytokines, mRNA, and proteins, which support the development of a tumor microenvironment and support the metastatic process. This review will discuss the different actions that platelet membranes and cancer cell membranes carry out during their relationship in the tumor microenvironment and metastasis.
Collapse
|
44
|
Nührenberg TG, Stöckle J, Marini F, Zurek M, Grüning BA, Benes V, Hein L, Neumann FJ, Stratz C, Cederqvist M, Hochholzer W. Impact of high platelet turnover on the platelet transcriptome: Results from platelet RNA-sequencing in patients with sepsis. PLoS One 2022; 17:e0260222. [PMID: 35085240 PMCID: PMC8794123 DOI: 10.1371/journal.pone.0260222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 11/04/2021] [Indexed: 12/13/2022] Open
Abstract
Background
Sepsis is associated with high platelet turnover and elevated levels of immature platelets. Changes in the platelet transcriptome and the specific impact of immature platelets on the platelet transcriptome remain unclear. Thus, this study sought to address whether and how elevated levels of immature platelets affect the platelet transcriptome in patients with sepsis.
Methods
Blood samples were obtained from patients with sepsis requiring vasopressor therapy (n = 8) and from a control group of patients with stable coronary artery disease and otherwise similar demographic characteristics (n = 8). Immature platelet fraction (IPF) was determined on a Sysmex XE 2100 analyser and platelet function was tested by impedance aggregometry. RNA from leukocyte-depleted platelets was used for transcriptome analysis by Next Generation Sequencing integrating the use of unique molecular identifiers.
Results
IPF (median [interquartile range]) was significantly elevated in sepsis patients (6.4 [5.3–8.7] % vs. 3.6 [2.6–4.6] %, p = 0.005). Platelet function testing revealed no differences in adenosine diphosphate- or thrombin receptor activating peptide-induced platelet aggregation between control and sepsis patients. Putative circular RNA transcripts were decreased in platelets from septic patients. Leukocyte contamination defined by CD45 abundance levels in RNA-sequencing was absent in both groups. Principal component analysis of transcripts showed only partial overlap of clustering with IPF levels. RNA sequencing showed up-regulation of 524 and down-regulation of 118 genes in platelets from sepsis patients compared to controls. Upregulated genes were mostly related to catabolic processes and protein translation. Comparison to published platelet transcriptomes showed a large overlap of changes observed in sepsis and COVID-19 but not with reticulated platelets from healthy donors.
Conclusions
Patients with sepsis appear to have a less degraded platelet transcriptome as indicated by increased levels of immature platelets and decreased levels of putative circular RNA transcripts. The present data suggests that increased protein translation is a characteristic mechanism of systemic inflammation.
Collapse
Affiliation(s)
- Thomas G. Nührenberg
- Klinik für Kardiologie und Angiologie II, Universitätsklinik Freiburg, Universitäts-Herzzentrum Campus Bad Krozingen, Bad Krozingen, Germany
- Institut für experimentelle und klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
- * E-mail:
| | - Jasmin Stöckle
- Klinik für Kardiologie und Angiologie II, Universitätsklinik Freiburg, Universitäts-Herzzentrum Campus Bad Krozingen, Bad Krozingen, Germany
| | - Federico Marini
- Institut für Medizinische Biometrie, Epidemiologie und Informatik, Universitätsmedizin der Johannes-Gutenberg-Universität Mainz, Mainz, Germany
| | - Mark Zurek
- Klinik für Kardiologie und Angiologie II, Universitätsklinik Freiburg, Universitäts-Herzzentrum Campus Bad Krozingen, Bad Krozingen, Germany
| | - Björn A. Grüning
- Institut für Informatik, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Vladimir Benes
- Genomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Lutz Hein
- Institut für experimentelle und klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Franz-Josef Neumann
- Klinik für Kardiologie und Angiologie II, Universitätsklinik Freiburg, Universitäts-Herzzentrum Campus Bad Krozingen, Bad Krozingen, Germany
| | - Christian Stratz
- Klinik für Kardiologie und Angiologie II, Universitätsklinik Freiburg, Universitäts-Herzzentrum Campus Bad Krozingen, Bad Krozingen, Germany
| | - Marco Cederqvist
- Klinik für Kardiologie und Angiologie II, Universitätsklinik Freiburg, Universitäts-Herzzentrum Campus Bad Krozingen, Bad Krozingen, Germany
| | - Willibald Hochholzer
- Klinik für Kardiologie und Angiologie II, Universitätsklinik Freiburg, Universitäts-Herzzentrum Campus Bad Krozingen, Bad Krozingen, Germany
| |
Collapse
|
45
|
Nguyen N, Thurgood P, Sekar NC, Chen S, Pirogova E, Peter K, Baratchi S, Khoshmanesh K. Microfluidic models of the human circulatory system: versatile platforms for exploring mechanobiology and disease modeling. Biophys Rev 2021; 13:769-786. [PMID: 34777617 DOI: 10.1007/s12551-021-00815-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023] Open
Abstract
The human circulatory system is a marvelous fluidic system, which is very sensitive to biophysical and biochemical cues. The current animal and cell culture models do not recapitulate the functional properties of the human circulatory system, limiting our ability to fully understand the complex biological processes underlying the dysfunction of this multifaceted system. In this review, we discuss the unique ability of microfluidic systems to recapitulate the biophysical, biochemical, and functional properties of the human circulatory system. We also describe the remarkable capacity of microfluidic technologies for exploring the complex mechanobiology of the cardiovascular system, mechanistic studying of cardiovascular diseases, and screening cardiovascular drugs with the additional benefit of reducing the need for animal models. We also discuss opportunities for further advancement in this exciting field.
Collapse
Affiliation(s)
- Ngan Nguyen
- School of Engineering, RMIT University, Melbourne, Australia
| | - Peter Thurgood
- School of Engineering, RMIT University, Melbourne, Australia
| | - Nadia Chandra Sekar
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | - Sheng Chen
- School of Engineering, RMIT University, Melbourne, Australia
| | - Elena Pirogova
- School of Engineering, RMIT University, Melbourne, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Department of Cardiometabolic Health, The University of Melbourne, Parkville, Australia
| | - Sara Baratchi
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | | |
Collapse
|
46
|
Abstract
Significance: Since protein disulfide isomerase (PDI) was first described in 1963, researchers have shown conclusively that PDI and sibling proteins are quintessential for thrombus formation. PDI, endoplasmic reticulum protein (ERp)5, ERp57, and ERp72 are released from platelets and vascular cells and interact with integrin αIIbβ3 on the outer surface of platelets. Recent Advances: At the cell surface they influence protein folding and function, propagating thrombosis and maintaining hemostasis. TMX1, which is a transmembrane thiol isomerase, is the first family member shown to negatively regulate platelets. Targets of thiol isomerases have been identified, including integrin α2β1, Von Willebrand Factor, GpIbα, nicotinamide adenine dinucleotide phosphate oxidase (Nox)-1, Nox-2, and tissue factor, all of which are pro-thrombotic, and several of which are on the cell surface. In spite of this, PDI can paradoxically catalyze the delivery of nitric oxide to platelets, which decrease thrombus formation. Critical Issues: Although the overall effect of PDI is to positively regulate platelet activation, it is still unclear how thiol isomerases function in pro-thrombotic states, such as obesity, diabetes, and cancer. In parallel, there has been a surge in the development of novel thiol isomerase inhibitors, which display selectivity, potency and modulate thrombosis and hemostasis. The availability of selective thiol isomerase inhibitors has culminated in clinical trials, with promising outcomes for the prevention of cancer-associated thrombosis. Future Directions: Altogether, thiol isomerases are perceived as an orchestrating force that regulates thrombus development. In the current review, we will explore the history of PDI in cardiovascular biology, detail known mechanisms of action, and summarize known thiol isomerase inhibitors.
Collapse
Affiliation(s)
- Renato Simões Gaspar
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| |
Collapse
|
47
|
García-Villaseñor E, Bojalil-Álvarez L, Murrieta-Álvarez I, Cantero-Fortiz Y, Ruiz-Delgado GJ, Ruiz-Argüelles GJ. Primary Thrombophilia XVI: A Look at the Genotype of the Sticky Platelet Syndrome Phenotype. Clin Appl Thromb Hemost 2021; 27:10760296211044212. [PMID: 34617458 PMCID: PMC8674482 DOI: 10.1177/10760296211044212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The sticky platelet syndrome (SPS) was described by Mammen in 1983. Since then,
scientists in several countries have identified the condition and published
cases or series of patients, thus enabling the description of the prevalence of
the inherited condition, its salient clinical features, and the treatment of the
disease. The diagnosis of the SPS phenotype requires fresh blood samples and
special equipment which is not available in all coagulation laboratories. In the
era of molecular biology, up to now it has not been possible to define a clear
association of the SPS phenotype with a specific molecular marker. Some
molecular changes which have been described in platelet proteins in some persons
with the phenotype of the SPS are here discussed. Nowadays, the SPS phenotype
may be considered as a risk factor for thrombosis and most cases of the SPS
developing vaso-occlussive episodes are the result of its coexistence with other
thrombosis-prone conditions, some of the inherited and some of them acquired,
thus leading to the concept of multifactorial thrombophilia. Ignoring all these
evidence-based concepts is inappropriate, same as stating that the SPS is a
nonentity simply because not all laboratories are endowed with adequate
equipment to support the diagnosis.
Collapse
Affiliation(s)
- Elizabeth García-Villaseñor
- Centro de Hematología y Medicina Interna de Puebla, Puebla, México.,3972Benemérita Universidad Autónoma de Puebla, Puebla, México
| | - Lorena Bojalil-Álvarez
- Centro de Hematología y Medicina Interna de Puebla, Puebla, México.,27861Universidad Popular Autónoma del Estado de Puebla, Puebla, México
| | - Iván Murrieta-Álvarez
- Centro de Hematología y Medicina Interna de Puebla, Puebla, México.,27861Universidad Popular Autónoma del Estado de Puebla, Puebla, México
| | - Yahveth Cantero-Fortiz
- Centro de Hematología y Medicina Interna de Puebla, Puebla, México.,27806Universidad de las Américas Puebla, Puebla, México
| | - Guillermo J Ruiz-Delgado
- Centro de Hematología y Medicina Interna de Puebla, Puebla, México.,27861Universidad Popular Autónoma del Estado de Puebla, Puebla, México.,56079Laboratorios Clínicos de Puebla, Puebla, México
| | - Guillermo J Ruiz-Argüelles
- Centro de Hematología y Medicina Interna de Puebla, Puebla, México.,27861Universidad Popular Autónoma del Estado de Puebla, Puebla, México.,56079Laboratorios Clínicos de Puebla, Puebla, México
| |
Collapse
|
48
|
Molecular Proteomics and Signalling of Human Platelets in Health and Disease. Int J Mol Sci 2021; 22:ijms22189860. [PMID: 34576024 PMCID: PMC8468031 DOI: 10.3390/ijms22189860] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 12/21/2022] Open
Abstract
Platelets are small anucleate blood cells that play vital roles in haemostasis and thrombosis, besides other physiological and pathophysiological processes. These roles are tightly regulated by a complex network of signalling pathways. Mass spectrometry-based proteomic techniques are contributing not only to the identification and quantification of new platelet proteins, but also reveal post-translational modifications of these molecules, such as acetylation, glycosylation and phosphorylation. Moreover, target proteomic analysis of platelets can provide molecular biomarkers for genetic aberrations with established or non-established links to platelet dysfunctions. In this report, we review 67 reports regarding platelet proteomic analysis and signalling on a molecular base. Collectively, these provide detailed insight into the: (i) technical developments and limitations of the assessment of platelet (sub)proteomes; (ii) molecular protein changes upon ageing of platelets; (iii) complexity of platelet signalling pathways and functions in response to collagen, rhodocytin, thrombin, thromboxane A2 and ADP; (iv) proteomic effects of endothelial-derived mediators such as prostacyclin and the anti-platelet drug aspirin; and (v) molecular protein changes in platelets from patients with congenital disorders or cardiovascular disease. However, sample sizes are still low and the roles of differentially expressed proteins are often unknown. Based on the practical and technical possibilities and limitations, we provide a perspective for further improvements of the platelet proteomic field.
Collapse
|
49
|
Pallini C, Pike JA, O'Shea C, Andrews RK, Gardiner EE, Watson SP, Poulter NS. Immobilized collagen prevents shedding and induces sustained GPVI clustering and signaling in platelets. Platelets 2021; 32:59-73. [PMID: 33455536 DOI: 10.1080/09537104.2020.1849607] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Collagen, the most thrombogenic constituent of blood vessel walls, activates platelets through glycoprotein VI (GPVI). In suspension, following platelet activation by collagen, GPVI is cleaved by A Disintegrin And Metalloproteinase (ADAM)10 and ADAM17. In this study, we use single-molecule localization microscopy and a 2-level DBSCAN-based clustering tool to show that GPVI remains clustered along immobilized collagen fibers for at least 3 hours in the absence of significant shedding. Tyrosine phosphorylation of spleen tyrosine kinase (Syk) and Linker of Activated T cells (LAT), and elevation of intracellular Ca2+, are sustained over this period. Syk, but not Src kinase-dependent signaling is required to maintain clustering of the collagen integrin α2β1, whilst neither is required for GPVI. We propose that clustering of GPVI on immobilized collagen protects GPVI from shedding in order to maintain sustained Src and Syk-kinases dependent signaling, activation of integrin α2β1, and continued adhesion.
Collapse
Affiliation(s)
- Chiara Pallini
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham , Midlands, UK
| | - Jeremy A Pike
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK
| | - Christopher O'Shea
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK
| | - Robert K Andrews
- Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, Australian National University , Canberra, Australia
| | - Elizabeth E Gardiner
- Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, Australian National University , Canberra, Australia
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham , Midlands, UK
| | - Natalie S Poulter
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham , Midlands, UK
| |
Collapse
|
50
|
Zhang S, Gui X, Ding Y, Tong H, Ju W, Li Y, Li Z, Zeng L, Xu K, Qiao J. Matrine Impairs Platelet Function and Thrombosis and Inhibits ROS Production. Front Pharmacol 2021; 12:717725. [PMID: 34366869 PMCID: PMC8339414 DOI: 10.3389/fphar.2021.717725] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/15/2021] [Indexed: 12/13/2022] Open
Abstract
Matrine is a naturally occurring alkaloid and possesses a wide range of pharmacological properties, such as anti-cancer, anti-oxidant, anti-inflammatory effects. However, whether it affects platelet function and thrombosis remains unclear. This study aims to evaluate the effect of matrine on platelet function and thrombus formation. Human platelets were treated with matrine (0–1 mg/ml) for 1 h at 37°C followed by measuring platelet aggregation, granule secretion, receptor expression by flow cytometry, spreading and clot retraction. In addition, matrine (10 mg/kg) was injected intraperitoneally into mice to measure tail bleeding time, arterial and venous thrombus formation. Matrine dose-dependently inhibited platelet aggregation and ATP release in response to either collagen-related peptide (Collagen-related peptide, 0.1 μg/ml) or thrombin (0.04 U/mL) stimulation without altering the expression of P-selectin, glycoprotein Ibα, GPVI, or αIIbβ3. In addition, matrine-treated platelets presented significantly decreased spreading on fibrinogen or collagen and clot retraction along with reduced phosphorylation of c-Src. Moreover, matrine administration significantly impaired the in vivo hemostatic function of platelets, arterial and venous thrombus formation. Furthermore, in platelets stimulated with CRP or thrombin, matrine significantly reduced Reactive oxygen species generation, inhibited the phosphorylation level of ERK1/2 (Thr202/Tyr204), p38 (Thr180/Tyr182) and AKT (Thr308/Ser473) as well as increased VASP phosphorylation (Ser239) and intracellular cGMP level. In conclusion, matrine inhibits platelet function, arterial and venous thrombosis, possibly involving inhibition of ROS generation, suggesting that matrine might be used as an antiplatelet agent for treating thrombotic or cardiovascular diseases.
Collapse
Affiliation(s)
- Sixuan Zhang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Xiang Gui
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Yangyang Ding
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Huan Tong
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Wen Ju
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Yue Li
- School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Zhenyu Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Lingyu Zeng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China.,School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| |
Collapse
|