1
|
Nock SH, Hutchinson JL, Blanco-Lopez M, Naseem K, Jones S, Mundell SJ, Unsworth AJ. Constitutive surface expression of the thromboxane A2 receptor is Pim kinase-dependent. J Thromb Haemost 2025; 23:293-305. [PMID: 39798965 DOI: 10.1016/j.jtha.2024.09.013] [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: 05/15/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND The thromboxane A2 receptor (TPαR) plays an important role in the amplification of platelet responses during thrombosis. Receptor activity is regulated by internalization and receptor desensitization. The mechanism by which constitutive surface expression of the TPαR is regulated is unknown. Recently, it has been demonstrated that proviral insertion in murine lymphoma (Pim) kinase inhibitors reduce platelet functional responses in a TPαR-dependent manner. OBJECTIVES To investigate whether Pim kinases regulate constitutive TPαR surface expression. METHODS TPαR surface expression was measured in platelets, and human embryonic kidney 293T (HEK293T) cells transfected with tagged TPαRs in the presence and absence of Pim kinase inhibitors using flow cytometry and confocal microscopy. TPαR-dependent calcium flux was assessed using Fluo-4 AM. Site prediction modeling and site-directed mutagenesis were used to identify the TPαR PIM kinase phosphorylation site. RESULTS Surface expression of TPαR and calcium responses to U46619 were reduced in platelets and HEK293T cells following Pim kinase inhibition. Overexpression of kinase-dead Pim-1 also reduced TPαR surface expression on HEK293T cells. Reduced surface expression of the TPαR was found to be mediated by increased receptor internalization in a dynamin and β-arrestin-dependent manner. Four putative Pim kinase phosphorylation sites in the TPαR were mutated, and serine 57 in the first intracellular loop of TPαR was identified to be a novel regulatory site important for maintaining TPαR surface expression and thromboxane A2-dependent functional responses. CONCLUSION Pim kinase inhibition may offer a novel therapeutic approach to limit cellular responses to thromboxane A2, independent of cyclooxygenase inhibition and direct antagonism of the receptor.
Collapse
Affiliation(s)
- Sophie H Nock
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - James L Hutchinson
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Maria Blanco-Lopez
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Khalid Naseem
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Sarah Jones
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Stuart J Mundell
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Amanda J Unsworth
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom; Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom.
| |
Collapse
|
2
|
Patrignani P, De Michele A, Tacconelli S. New antiplatelet approach: inhibiting Pim kinase to reduce constitutive surface expression of thromboxane A 2 receptor. J Thromb Haemost 2025; 23:43-46. [PMID: 39798970 DOI: 10.1016/j.jtha.2024.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 01/15/2025]
Affiliation(s)
- Paola Patrignani
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), and Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University Medical School, Chieti, Italy.
| | - Alessandra De Michele
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), and Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University Medical School, Chieti, Italy
| | - Stefania Tacconelli
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), and Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University Medical School, Chieti, Italy
| |
Collapse
|
3
|
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
|
4
|
Nock SH, Blanco-Lopez MR, Stephenson-Deakin C, Jones S, Unsworth AJ. Pim Kinase Inhibition Disrupts CXCR4 Signalling in Megakaryocytes and Platelets by Reducing Receptor Availability at the Surface. Int J Mol Sci 2024; 25:7606. [PMID: 39062849 PMCID: PMC11276893 DOI: 10.3390/ijms25147606] [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: 05/20/2024] [Revised: 06/06/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
A key step in platelet production is the migration of megakaryocytes to the vascular sinusoids within the bone marrow. This homing is mediated by the chemokine CXCL12 and its receptor CXCR4. CXCR4 is also a positive regulator of platelet activation and thrombosis. Pim-1 kinase has been shown to regulate CXCR4 signalling in other cell types, and we have previously described how Pim kinase inhibitors attenuate platelet aggregation to CXCL12. However, the mechanism by which Pim-1 regulates CXCR4 signalling in platelets and megakaryocytes has yet to be elucidated. Using human platelets, murine bone marrow-derived megakaryocytes, and the megakaryocyte cell line MEG-01, we demonstrate that pharmacological Pim kinase inhibition leads to reduced megakaryocyte and platelet function responses to CXCL12, including reduced megakaryocyte migration and platelet granule secretion. Attenuation of CXCL12 signalling was found to be attributed to the reduced surface expression of CXCR4. The decrease in CXCR4 surface levels was found to be mediated by rapid receptor internalisation, in the absence of agonist stimulation. We demonstrate that pharmacological Pim kinase inhibition disrupts megakaryocyte and platelet function by reducing constitutive CXCR4 surface expression, decreasing the number of receptors available for agonist stimulation and signalling. These findings have implications for the development and use of Pim kinase inhibitors for the treatment of conditions associated with elevated circulating levels of CXCL12/SDF1α and increased thrombotic risk.
Collapse
Affiliation(s)
- Sophie H. Nock
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester M1 5GD, UK
| | - Maria R. Blanco-Lopez
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester M1 5GD, UK
| | - Chloe Stephenson-Deakin
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester M1 5GD, UK
| | - Sarah Jones
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester M1 5GD, UK
| | - Amanda J. Unsworth
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester M1 5GD, UK
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 3AA, UK
| |
Collapse
|
5
|
Drysdale A, Blanco-Lopez M, White SJ, Unsworth AJ, Jones S. Differential Proteoglycan Expression in Atherosclerosis Alters Platelet Adhesion and Activation. Int J Mol Sci 2024; 25:950. [PMID: 38256024 PMCID: PMC10815981 DOI: 10.3390/ijms25020950] [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: 12/07/2023] [Revised: 01/06/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Proteoglycans are differentially expressed in different atherosclerotic plaque phenotypes, with biglycan and decorin characteristic of ruptured plaques and versican and hyaluronan more prominent in eroded plaques. Following plaque disruption, the exposure of extracellular matrix (ECM) proteins triggers platelet adhesion and thrombus formation. In this study, the impact of differential plaque composition on platelet function and thrombus formation was investigated. Platelet adhesion, activation and thrombus formation under different shear stress conditions were assessed in response to individual proteoglycans and composites representing different plaque phenotypes. The results demonstrated that all the proteoglycans tested mediated platelet adhesion but not platelet activation, and the extent of adhesion observed was significantly lower than that observed with type I and type III collagens. Thrombus formation upon the rupture and erosion ECM composites was significantly reduced (p < 0.05) compared to relevant collagen alone, indicating that proteoglycans negatively regulate platelet collagen responses. This was supported by results demonstrating that the addition of soluble biglycan or decorin to whole blood markedly reduced thrombus formation on type I collagen (p < 0.05). Interestingly, thrombus formation upon the erosion composite displayed aspirin sensitivity, whereas the rupture composite was intensive to aspirin, having implications for current antiplatelet therapy regimes. In conclusion, differential platelet responses and antiplatelet efficacy are observed on ECM composites phenotypic of plaque rupture and erosion. Proteoglycans inhibit thrombus formation and may offer a novel plaque-specific approach to limit arterial thrombosis.
Collapse
Affiliation(s)
- Amelia Drysdale
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (M.B.-L.); (A.J.U.)
| | - Maria Blanco-Lopez
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (M.B.-L.); (A.J.U.)
| | - Stephen J. White
- Faculty of Medical Sciences, The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK;
| | - Amanda J. Unsworth
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (M.B.-L.); (A.J.U.)
| | - Sarah Jones
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (M.B.-L.); (A.J.U.)
| |
Collapse
|
6
|
Abstract
PURPOSE OF REVIEW Models of arterial thrombus formation represent a vital experimental tool to investigate platelet function and test novel antithrombotic drugs. This review highlights some of the recent advances in modelling thrombus formation in vitro and suggests potential future directions. RECENT FINDINGS Microfluidic devices and the availability of commercial chips in addition to enhanced accessibility of 3D printing has facilitated a rapid surge in the development of novel in-vitro thrombosis models. These include progression towards more sophisticated, 'vessel on a chip' models which incorporate vascular endothelial cells and smooth muscle cells. Other approaches include the addition of branches to the traditional single channel to yield an occlusive model; and developments in the adhesive coating of microfluidic chambers to better mimic the thrombogenic surface exposed following plaque rupture. Future developments in the drive to create more biologically relevant chambers could see a move towards the use of human placental vessels, perfused ex-vivo. However, further work is required to determine the feasibility and validity of this approach. SUMMARY Recent advances in thrombus formation models have significantly improved the pathophysiological relevance of in-vitro flow chambers to better reflect the in-vivo environment and provide a more translational platform to test novel antithrombotics.
Collapse
Affiliation(s)
- Amelia Drysdale
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
| | | | | |
Collapse
|
7
|
Li Z, Zhang J, Ma Z, Zhao G, He X, Yu X, Fu Q, Wu N, Ding Z, Sun H, Zhang X, Zhu Y, Chen L, He J. Endothelial YAP Mediates Hyperglycemia-Induced Platelet Hyperactivity and Arterial Thrombosis. Arterioscler Thromb Vasc Biol 2024; 44:254-270. [PMID: 37916416 DOI: 10.1161/atvbaha.123.319835] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 10/11/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Hyperglycemia-a symptom that characterizes diabetes-is highly associated with atherothrombotic complications. However, the underlying mechanism by which hyperglycemia fuels platelet activation and arterial thrombus formation is still not fully understood. METHODS The profiles of polyunsaturated fatty acid metabolites in the plasma of patients with diabetes and healthy controls were determined with targeted metabolomics. FeCl3-induced carotid injury model was used to assess arterial thrombus formation in mice with endothelial cell (EC)-specific YAP (yes-associated protein) deletion or overexpression. Flow cytometry and clot retraction assay were used to evaluate platelet activation. RNA sequencing and multiple biochemical analyses were conducted to unravel the underlying mechanism. RESULTS The plasma PGE2 (prostaglandin E2) concentration was elevated in patients with diabetes with thrombotic complications and positively correlated with platelet activation. The PGE2 synthetases COX-2 (cyclooxygenase-2) and mPGES-1 (microsomal prostaglandin E synthase-1) were found to be highly expressed in ECs but not in other type of vessel cells in arteries from both patients with diabetes and hyperglycemic mice, compared with nondiabetic individuals and control mice, respectively. A combination of RNA sequencing and ingenuity pathway analyses indicated the involvement of YAP signaling. EC-specific deletion of YAP limited platelet activation and arterial thrombosis in hyperglycemic mice, whereas EC-specific overexpression of YAP in mice mimicked the prothrombotic state of diabetes, without affecting hemostasis. Mechanistically, we found that hyperglycemia/high glucose-induced endothelial YAP nuclear translocation and subsequently transcriptional expression of COX-2 and mPGES-1 contributed to the elevation of PGE2 and platelet activation. Blockade of EP3 (prostaglandin E receptor 3) activation by oral administration of DG-041 reversed the hyperactivity of platelets and delayed thrombus formation in both EC-specific YAP-overexpressing and hyperglycemic mice. CONCLUSIONS Collectively, our data suggest that hyperglycemia-induced endothelial YAP activation aggravates platelet activation and arterial thrombus formation via PGE2/EP3 signaling. Targeting EP3 with DG-041 might be therapeutic for diabetes-related thrombosis.
Collapse
Affiliation(s)
- Zhiyu Li
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| | - Jiachen Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| | - Zejun Ma
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
- National Humanities Center Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology (Z.M., H.S., L.C.), Tianjin Medical University, China
| | - Guobing Zhao
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| | - Xue He
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| | - Xuefang Yu
- Departments of Cardiology (X.Y.), Tianjin Medical University General Hospital, China
| | - Qiang Fu
- Cardiovascular Surgery (Q.F., N.W.), Tianjin Medical University General Hospital, China
| | - Naishi Wu
- Cardiovascular Surgery (Q.F., N.W.), Tianjin Medical University General Hospital, China
| | - Zhongren Ding
- School of Pharmacy (Z.D.), Tianjin Medical University, China
| | - Haipeng Sun
- National Humanities Center Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology (Z.M., H.S., L.C.), Tianjin Medical University, China
| | - Xu Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| | - Yi Zhu
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| | - Liming Chen
- National Humanities Center Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology (Z.M., H.S., L.C.), Tianjin Medical University, China
| | - Jinlong He
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Rovati G, Contursi A, Bruno A, Tacconelli S, Ballerini P, Patrignani P. Antiplatelet Agents Affecting GPCR Signaling Implicated in Tumor Metastasis. Cells 2022; 11:725. [PMID: 35203374 PMCID: PMC8870128 DOI: 10.3390/cells11040725] [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: 01/17/2022] [Revised: 02/10/2022] [Accepted: 02/16/2022] [Indexed: 11/16/2022] Open
Abstract
Metastasis requires that cancer cells survive in the circulation, colonize distant organs, and grow. Despite platelets being central contributors to hemostasis, leukocyte trafficking during inflammation, and vessel stability maintenance, there is significant evidence to support their essential role in supporting metastasis through different mechanisms. In addition to their direct interaction with cancer cells, thus forming heteroaggregates such as leukocytes, platelets release molecules that are necessary to promote a disseminating phenotype in cancer cells via the induction of an epithelial-mesenchymal-like transition. Therefore, agents that affect platelet activation can potentially restrain these prometastatic mechanisms. Although the primary adhesion of platelets to cancer cells is mainly independent of G protein-mediated signaling, soluble mediators released from platelets, such as ADP, thromboxane (TX) A2, and prostaglandin (PG) E2, act through G protein-coupled receptors (GPCRs) to cause the activation of more additional platelets and drive metastatic signaling pathways in cancer cells. In this review, we examine the contribution of the GPCRs of platelets and cancer cells in the development of cancer metastasis. Finally, the possible use of agents affecting GPCR signaling pathways as antimetastatic agents is discussed.
Collapse
Affiliation(s)
- Gianenrico Rovati
- Department of Pharmaceutical Sciences, University of Milan, 20122 Milan, Italy;
| | - Annalisa Contursi
- Laboratory of Systems Pharmacology and Translational Therapies, Center for Advanced Studies and Technology (CAST), School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy; (A.C.); (A.B.); (S.T.); (P.B.)
- Department of Neuroscience, Imaging and Clinical Science, School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy
| | - Annalisa Bruno
- Laboratory of Systems Pharmacology and Translational Therapies, Center for Advanced Studies and Technology (CAST), School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy; (A.C.); (A.B.); (S.T.); (P.B.)
- Department of Neuroscience, Imaging and Clinical Science, School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy
| | - Stefania Tacconelli
- Laboratory of Systems Pharmacology and Translational Therapies, Center for Advanced Studies and Technology (CAST), School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy; (A.C.); (A.B.); (S.T.); (P.B.)
- Department of Neuroscience, Imaging and Clinical Science, School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy
| | - Patrizia Ballerini
- Laboratory of Systems Pharmacology and Translational Therapies, Center for Advanced Studies and Technology (CAST), School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy; (A.C.); (A.B.); (S.T.); (P.B.)
- Department of Innovative Technologies in Medicine and Dentistry, “G. d’Annunzio” University, 66100 Chieti, Italy
| | - Paola Patrignani
- Laboratory of Systems Pharmacology and Translational Therapies, Center for Advanced Studies and Technology (CAST), School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy; (A.C.); (A.B.); (S.T.); (P.B.)
- Department of Neuroscience, Imaging and Clinical Science, School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy
| |
Collapse
|
11
|
Meng Y, Sun J, Zheng Y, Zhang G, Yu T, Piao H. Platelets: The Emerging Clinical Diagnostics and Therapy Selection of Cancer Liquid Biopsies. Onco Targets Ther 2021; 14:3417-3428. [PMID: 34079287 PMCID: PMC8164876 DOI: 10.2147/ott.s311907] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/16/2021] [Indexed: 12/11/2022] Open
Abstract
Due to the inherent molecular heterogeneity of metastatic tumours and the dynamic evolution ability of tumour genomes, tumour tissues obtained through biopsy and other methods cannot capture all of the features of tumour genomes. A new diagnostic concept called “liquid biopsy” has received widespread attention in recent years. Liquid biopsy has changed the clinical practice of oncology and is widely used to guide targeted drug utilization, monitor disease progression and track drug resistance. The latest research subject in liquid biopsy is platelets. Platelets originate from multifunctional haematopoietic stem cells in the bone marrow haematopoietic system. They are small cells from the cytoplasm of bone marrow megakaryocytes. Their main physiological functions are to participate in the processes of physiological haemostasis and coagulation. Tumour cells transfer biomolecules (such as RNA) to platelets through direct contact and release of exosomes, which changes the platelet precursor RNA. Under the stimulation of tumour cells and the tumour microenvironment, platelet precursor mRNA is spliced into mature RNA and converted into functional protein to respond to external stimuli, forming tumour-educated platelets (TEPs). The detection of TEPs in the peripheral blood of patients is expected to be used in clinical tumour diagnosis. This emerging liquid biopsy method can replace and supplement the current tumour detection methods. Further research on the role of platelets in tumour diagnosis will help provide a novel theoretical basis for clinical tumour diagnosis.
Collapse
Affiliation(s)
- Yiming Meng
- Department of Central Laboratory, Cancer Hospital of China Medical University, Liaoning province Cancer Hospital, Shenyang, 110042, People's Republic of China
| | - Jing Sun
- Department of Biobank, Cancer Hospital of China Medical University, Liaoning Province Cancer Hospital, Shenyang, 110042, People's Republic of China
| | - Yang Zheng
- Department of Clinical Laboratory, Cancer Hospital of China Medical University, Liaoning Province Cancer Hospital, Shenyang, 110042, People's Republic of China
| | - Guirong Zhang
- Department of Central Laboratory, Cancer Hospital of China Medical University, Liaoning province Cancer Hospital, Shenyang, 110042, People's Republic of China
| | - Tao Yu
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Province Cancer Hospital, Shenyang, 110042, People's Republic of China
| | - Haozhe Piao
- Department of Central Laboratory, Cancer Hospital of China Medical University, Liaoning province Cancer Hospital, Shenyang, 110042, People's Republic of China.,Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Province Cancer Hospital, Shenyang, 110042, People's Republic of China
| |
Collapse
|