1
|
Zhao A, Zhang G, Wei H, Yan X, Gan J, Jiang X. Heat shock proteins in cerebral ischemia-reperfusion injury: Mechanisms and therapeutic implications. Exp Neurol 2025; 390:115284. [PMID: 40318821 DOI: 10.1016/j.expneurol.2025.115284] [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: 03/03/2025] [Revised: 04/24/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) remains a significant challenge in ischemic stroke treatment. Heat shock proteins (HSPs), a cadre of molecular chaperones, have emerged as pivotal regulators in this pathological cascade. This review synthesizes the latest research on HSPs in CIRI from 2013 to 2024 focusing on their multifaceted roles and therapeutic potential. We explore the diverse cellular functions of HSPs, including regulation of oxidative stress, apoptosis, necroptosis, ferroptosis, autophagy, neuroinflammation, and blood-brain barrier integrity. Key HSPs, such as HSP90, HSP70, HSP32, HSP60, HSP47, and small HSPs, are investigated for their specific mechanisms of action in CIRI. Potential therapeutic strategies targeting HSPs, including HSP inhibitors, traditional Chinese medicine components, and gene therapy, are discussed. This review provides a comprehensive understanding of HSPs in CIRI and offers insights into the development of innovative neuroprotective treatments.
Collapse
Affiliation(s)
- Anliu Zhao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Guangming Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Huayuan Wei
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xu Yan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiali Gan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
2
|
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
|
3
|
Shi Y, He Y, Li Y, Zhang M, Liu Y, Wang H, Shen Z, Zhao X, Wang R, Ma T, Yang P, Chen J. Downregulation of heat shock protein 47 caused lysosomal dysfunction leading to excessive chondrocyte apoptosis. Exp Cell Res 2024; 443:114294. [PMID: 39447624 DOI: 10.1016/j.yexcr.2024.114294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 10/19/2024] [Accepted: 10/20/2024] [Indexed: 10/26/2024]
Abstract
Heat shock protein 47 (HSP47) is a collagen-specific chaperone present in several regions of the endoplasmic reticulum and cytoplasm. Elevated HSP47 expression in cells causes various cancers and fibrotic disorders. However, the consequences of HSP47 downregulation leading to chondrocyte death, as well as the underlying pathways, remain largely unclear. This study presents the first experimental evidence of the localization of HSP47 on lysosomes. Additionally, it successfully designed and generated shRNA HSP47 target sequences to suppress the expression of HSP47 in ATDC5 chondrocytes using lentiviral vectors. By employing a chondrocyte model that has undergone stable downregulation of HSP47, we observed that HSP47 downregulation in chondrocytes, disturbs the acidic homeostatic environment of chondrocyte lysosomes, causes hydrolytic enzyme activity dysregulation, impairs the lysosome-mediated autophagy-lysosome pathway, and causes abnormal expression of lysosomal morphology, number, and functional effector proteins. This implies the significance of the presence of HSP47 in maintaining proper lysosomal function. Significantly, the inhibitor CA-074 Me, which can restore the dysfunction of lysosomes, successfully reversed the negative effects of HSP47 on the autophagy-lysosomal pathway and partially reduced the occurrence of excessive cell death in chondrocytes. This suggests that maintaining proper lysosomal function is crucial for preventing HSP47-induced apoptosis in chondrocytes. The existence of HSP47 is crucial for preserving optimal lysosomal function and autophagic flux, while also inhibiting excessive apoptosis in ATDC5 chondrocytes.
Collapse
Affiliation(s)
- Yawen Shi
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, 710061, China
| | - Ying He
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, 710061, China
| | - Yanan Li
- School of Energy and Power Engineering, Xi'an Jiaotong University, Key Laboratory of Thermo-Fluid Science and Engineering, Ministry of Education, Xi'an, Shaanxi, 710049, China
| | - Meng Zhang
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, 710061, China
| | - Yinan Liu
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, 710061, China
| | - Hui Wang
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, 710061, China
| | - Zhiran Shen
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, 710061, China
| | - Xiaoru Zhao
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, 710061, China
| | - Rui Wang
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, 710061, China; Xi'an No. 3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, 710018, China
| | - Tianyou Ma
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, 710061, China
| | - Pinglin Yang
- Department of Orthopaedic Surgery, Second Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China.
| | - Jinghong Chen
- School of Public Health, Xi'an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases in the Education Ministry and Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
4
|
Khan ES, Däinghaus T. HSP47 in human diseases: Navigating pathophysiology, diagnosis and therapy. Clin Transl Med 2024; 14:e1755. [PMID: 39135385 PMCID: PMC11319607 DOI: 10.1002/ctm2.1755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 08/16/2024] Open
Abstract
Heat shock protein 47 (HSP47) is a chaperone protein responsible for regulating collagen maturation and transport, directly impacting collagen synthesis levels. Aberrant HSP47 expression or malfunction has been associated with collagen-related disorders, most notably fibrosis. Recent reports have uncovered new functions of HSP47 in various cellular processes. Hsp47 dysregulation in these alternative roles has been linked to various diseases, such as cancer, autoimmune and neurodegenerative disorders, thereby highlighting its potential as both a diagnostic biomarker and a therapeutic target. In this review, we discuss the pathophysiological roles of HSP47 in human diseases, its potential as a diagnostic tool, clinical screening techniques and its role as a target for therapeutic interventions.
Collapse
Affiliation(s)
- Essak. S. Khan
- Posttranscriptional Gene RegulationCancer Research and Experimental HemostasisUniversity Medical Center Mainz (UMCM)MainzGermany
- Center for Thrombosis and Hemostasis (CTH)UMCMMainzGermany
- German Consortium for Translational Cancer Research (DKTK)DKFZ Frankfurt‐MainzFrankfurt am MainGermany
| | - Tobias Däinghaus
- Posttranscriptional Gene RegulationCancer Research and Experimental HemostasisUniversity Medical Center Mainz (UMCM)MainzGermany
- Center for Thrombosis and Hemostasis (CTH)UMCMMainzGermany
| |
Collapse
|
5
|
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
|
6
|
Kelliher S, Gamba S, Weiss L, Shen Z, Marchetti M, Schieppati F, Scaife C, Madden S, Bennett K, Fortune A, Maung S, Fay M, Ní Áinle F, Maguire P, Falanga A, Kevane B, Krishnan A. Platelet proteo-transcriptomic profiling validates mediators of thrombosis and proteostasis in patients with myeloproliferative neoplasms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563619. [PMID: 37961700 PMCID: PMC10634751 DOI: 10.1101/2023.10.23.563619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Patients with chronic Myeloproliferative Neoplasms (MPN) including polycythemia vera (PV) and essential thrombocythemia (ET) exhibit unique clinical features, such as a tendency toward thrombosis and hemorrhage, and risk of disease progression to secondary bone marrow fibrosis and/or acute leukemia. Although an increase in blood cell lineage counts (quantitative features) contribute to these morbid sequelae, the significant qualitative abnormalities of myeloid cells that contribute to vascular risk are not well understood. Here, we address this critical knowledge gap via a comprehensive and untargeted profiling of the platelet proteome in a large (n= 140) cohort of patients (from two independent sites) with an established diagnosis of PV and ET (and complement prior work on the MPN platelet transcriptome from a third site). We discover distinct MPN platelet protein expression and confirm key molecular impairments associated with proteostasis and thrombosis mechanisms of potential relevance to MPN pathology. Specifically, we validate expression of high-priority candidate markers from the platelet transcriptome at the platelet proteome (e.g., calreticulin (CALR), Fc gamma receptor (FcγRIIA) and galectin-1 (LGALS1) pointing to their likely significance in the proinflammatory, prothrombotic and profibrotic phenotypes in patients with MPN. Together, our proteo-transcriptomic study identifies the peripherally-derived platelet molecular profile as a potential window into MPN pathophysiology and demonstrates the value of integrative multi-omic approaches in gaining a better understanding of the complex molecular dynamics of disease.
Collapse
Affiliation(s)
- Sarah Kelliher
- School of Medicine, University College Dublin, Dublin, Ireland
- Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
| | - Sara Gamba
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Luisa Weiss
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Zhu Shen
- Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Marina Marchetti
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Francesca Schieppati
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Caitriona Scaife
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Stephen Madden
- Data Science Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Kathleen Bennett
- School of Population Health, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Anne Fortune
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Su Maung
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Michael Fay
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Fionnuala Ní Áinle
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
- School of Medicine, Royal College of Surgeons in Ireland
| | - Patricia Maguire
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Institute for Discovery, University College Dublin, Dublin, Ireland
| | - Anna Falanga
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
- University of Milano-Bicocca, Department of Medicine and Surgery, Monza, Italy
| | - Barry Kevane
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
| | - Anandi Krishnan
- Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Rutgers University, Piscataway, NJ
- Stanford Cancer Institute, Stanford, CA, USA
| |
Collapse
|
7
|
Grover SP, Mackman N, Bendapudi PK. Heat shock protein 47 and venous thrombosis: letting sleeping bears lie. J Thromb Haemost 2023; 21:2648-2652. [PMID: 37473845 DOI: 10.1016/j.jtha.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/22/2023]
Affiliation(s)
- Steven P Grover
- University of North Carolina Blood Research Center, The University of North Carolina at Chapel Hill, North Carolina, USA; Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, North Carolina, USA.
| | - Nigel Mackman
- University of North Carolina Blood Research Center, The University of North Carolina at Chapel Hill, North Carolina, USA; Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, North Carolina, USA
| | - Pavan K Bendapudi
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA; Division of Hematology and Blood Transfusion Service, Massachusetts General Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA; Center for the Development of Therapeutics, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
8
|
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
|
9
|
Thienel M, Müller-Reif JB, Zhang Z, Ehreiser V, Huth J, Shchurovska K, Kilani B, Schweizer L, Geyer PE, Zwiebel M, Novotny J, Lüsebrink E, Little G, Orban M, Nicolai L, El Nemr S, Titova A, Spannagl M, Kindberg J, Evans AL, Mach O, Vogel M, Tiedt S, Ormanns S, Kessler B, Dueck A, Friebe A, Jørgensen PG, Majzoub-Altweck M, Blutke A, Polzin A, Stark K, Kääb S, Maier D, Gibbins JM, Limper U, Frobert O, Mann M, Massberg S, Petzold T. Immobility-associated thromboprotection is conserved across mammalian species from bear to human. Science 2023; 380:178-187. [PMID: 37053338 DOI: 10.1126/science.abo5044] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/10/2023] [Indexed: 04/15/2023]
Abstract
Venous thromboembolism (VTE) comprising deep venous thrombosis and pulmonary embolism is a major cause of morbidity and mortality. Short-term immobility-related conditions are a major risk factor for the development of VTE. Paradoxically, long-term immobilized free-ranging hibernating brown bears and paralyzed spinal cord injury (SCI) patients are protected from VTE. We aimed to identify mechanisms of immobility-associated VTE protection in a cross-species approach. Mass spectrometry-based proteomics revealed an antithrombotic signature in platelets of hibernating brown bears with heat shock protein 47 (HSP47) as the most substantially reduced protein. HSP47 down-regulation or ablation attenuated immune cell activation and neutrophil extracellular trap formation, contributing to thromboprotection in bears, SCI patients, and mice. This cross-species conserved platelet signature may give rise to antithrombotic therapeutics and prognostic markers beyond immobility-associated VTE.
Collapse
Affiliation(s)
- Manuela Thienel
- Department of Cardiology, University Hospital, LMU Munich, 81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| | - Johannes B Müller-Reif
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
- Omicera Diagnostics, 82152 Martinsried, Germany
| | - Zhe Zhang
- Department of Cardiology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Vincent Ehreiser
- Department of Cardiology, University Hospital, LMU Munich, 81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| | - Judith Huth
- Department of Cardiology, University Hospital, LMU Munich, 81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| | - Khrystyna Shchurovska
- Department of Cardiology, University Hospital, LMU Munich, 81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| | - Badr Kilani
- Department of Cardiology, University Hospital, LMU Munich, 81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| | - Lisa Schweizer
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Philipp E Geyer
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
- Omicera Diagnostics, 82152 Martinsried, Germany
| | - Maximilian Zwiebel
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Julia Novotny
- Department of Cardiology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Enzo Lüsebrink
- Department of Cardiology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Gemma Little
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, Health and Life Sciences Building, University of Reading, RG6 6UR, UK
| | - Martin Orban
- Department of Cardiology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Leo Nicolai
- Department of Cardiology, University Hospital, LMU Munich, 81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| | - Shaza El Nemr
- Department of Cardiology, University Hospital, LMU Munich, 81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| | - Anna Titova
- Department of Cardiology, University Hospital, LMU Munich, 81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| | - Michael Spannagl
- Anesthesiology and Transfusion Medicine, Cell Therapeutics and Hemostaseology, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| | - Jonas Kindberg
- Norwegian Institute for Nature Research, 7034 Trondheim, Norway
- Scandinavian Brown Bear Research Project, Tackåsen 2, SE-79498 Orsa, Sweden
- Department of Wildlife, Fish and Environmental Studies, Swedish University of Agricultural Sciences, SE-90183 Umeå, Sweden
| | - Alina L Evans
- Department of Forestry and Wildlife Management, Faculty of Applied Ecology and Agricultural Sciences, Inland Norway University of Applied Sciences, 2480 Koppang, Norway
| | - Orpheus Mach
- Zentrum für Rückenmarkverletzte mit Neuro-Urologie, BG Unfallklinik Murnau, 82418 Murnau am Staffelsee, Germany
| | - Matthias Vogel
- Zentrum für Rückenmarkverletzte mit Neuro-Urologie, BG Unfallklinik Murnau, 82418 Murnau am Staffelsee, Germany
| | - Steffen Tiedt
- Neurologische Klinik und Poliklinik, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany
| | - Steffen Ormanns
- Pathologisches Institut, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany
| | - Barbara Kessler
- Gene Center, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Anne Dueck
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
- Institute of Pharmacology and Toxicology, Technical University of Munich, 80802 Munich, Germany
| | - Andrea Friebe
- Norwegian Institute for Nature Research, 7034 Trondheim, Norway
- Scandinavian Brown Bear Research Project, Tackåsen 2, SE-79498 Orsa, Sweden
| | - Peter Godsk Jørgensen
- Herlev and Gentofte University Hospital, Borgmester Ib Juuls Vej 1, DK-2730, Herlev, Copenhagen, Denmark
| | - Monir Majzoub-Altweck
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, LMU Munich, 80539 Munich, Germany
| | - Andreas Blutke
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, LMU Munich, 80539 Munich, Germany
| | - Amin Polzin
- Division of Cardiology, Pulmonology, and Vascular Medicine, Heinrich Heine University Medical Center Dusseldorf, 40225 Dusseldorf, Germany
| | - Konstantin Stark
- Department of Cardiology, University Hospital, LMU Munich, 81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| | - Stefan Kääb
- Department of Cardiology, University Hospital, LMU Munich, 81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| | - Doris Maier
- Zentrum für Rückenmarkverletzte mit Neuro-Urologie, BG Unfallklinik Murnau, 82418 Murnau am Staffelsee, Germany
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, Health and Life Sciences Building, University of Reading, RG6 6UR, UK
| | - Ulrich Limper
- Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany
- Department of Anesthesiology and Intensive Care Medicine, Merheim Medical Center, Hospitals of Cologne, University of Witten/Herdecke, 51109 Cologne, Germany
| | - Ole Frobert
- Faculty of Health, Department of Cardiology, Örebro University, 701 85 Örebro, Sweden
- Department of Clinical Medicine, Faculty of Health, Aarhus University, 8000 Aarhus, Denmark
- Department of Clinical Pharmacology, Aarhus University Hospital, 8000 Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, 8000 Aarhus, Denmark
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Steffen Massberg
- Department of Cardiology, University Hospital, LMU Munich, 81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| | - Tobias Petzold
- Department of Cardiology, University Hospital, LMU Munich, 81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| |
Collapse
|
10
|
Correll VL, Otto JJ, Risi CM, Main BP, Boutros PC, Kislinger T, Galkin VE, Nyalwidhe JO, Semmes OJ, Yang L. Optimization of small extracellular vesicle isolation from expressed prostatic secretions in urine for in-depth proteomic analysis. J Extracell Vesicles 2022; 11:e12184. [PMID: 35119778 PMCID: PMC8815402 DOI: 10.1002/jev2.12184] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 11/22/2021] [Accepted: 12/22/2021] [Indexed: 01/23/2023] Open
Abstract
The isolation and subsequent molecular analysis of extracellular vesicles (EVs) derived from patient samples is a widely used strategy to understand vesicle biology and to facilitate biomarker discovery. Expressed prostatic secretions in urine are a tumor proximal fluid that has received significant attention as a source of potential prostate cancer (PCa) biomarkers for use in liquid biopsy protocols. Standard EV isolation methods like differential ultracentrifugation (dUC) co-isolate protein contaminants that mask lower-abundance proteins in typical mass spectrometry (MS) protocols. Further complicating the analysis of expressed prostatic secretions, uromodulin, also known as Tamm-Horsfall protein (THP), is present at high concentrations in urine. THP can form polymers that entrap EVs during purification, reducing yield. Disruption of THP polymer networks with dithiothreitol (DTT) can release trapped EVs, but smaller THP fibres co-isolate with EVs during subsequent ultracentrifugation. To resolve these challenges, we describe here a dUC method that incorporates THP polymer reduction and alkaline washing to improve EV isolation and deplete both THP and other common protein contaminants. When applied to human expressed prostatic secretions in urine, we achieved relative enrichment of known prostate and prostate cancer-associated EV-resident proteins. Our approach provides a promising strategy for global proteomic analyses of urinary EVs.
Collapse
Affiliation(s)
- Vanessa L. Correll
- Leroy T. Canoles Jr. Cancer Research CenterEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - Joseph J. Otto
- Leroy T. Canoles Jr. Cancer Research CenterEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - Cristina M. Risi
- Department of Physiological SciencesEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - Brian P. Main
- Leroy T. Canoles Jr. Cancer Research CenterEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - Paul C. Boutros
- Department of Medical BiophysicsUniversity of TorontoTorontoCanada
- Department of Pharmacology and ToxicologyUniversity of TorontoTorontoCanada
- Department of Human GeneticsUniversity of CaliforniaLos AngelesCaliforniaUSA
- Department of UrologyUniversity of CaliforniaLos AngelesCaliforniaUSA
- Institute for Precision HealthUniversity of CaliforniaLos AngelesCaliforniaUSA
- Jonsson Comprehensive Cancer CenterUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Thomas Kislinger
- Department of Medical BiophysicsUniversity of TorontoTorontoCanada
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoCanada
| | - Vitold E. Galkin
- Department of Physiological SciencesEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - Julius O. Nyalwidhe
- Leroy T. Canoles Jr. Cancer Research CenterEastern Virginia Medical SchoolNorfolkVirginiaUSA
- Department of Microbiology and Molecular Cell BiologyEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - O. John Semmes
- Leroy T. Canoles Jr. Cancer Research CenterEastern Virginia Medical SchoolNorfolkVirginiaUSA
- Department of Microbiology and Molecular Cell BiologyEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - Lifang Yang
- Leroy T. Canoles Jr. Cancer Research CenterEastern Virginia Medical SchoolNorfolkVirginiaUSA
- Department of Microbiology and Molecular Cell BiologyEastern Virginia Medical SchoolNorfolkVirginiaUSA
| |
Collapse
|
11
|
Wu S, Liang C, Xie X, Huang H, Fu J, Wang C, Su Z, Wang Y, Qu X, Li J, Liu J. Hsp47 Inhibitor Col003 Attenuates Collagen-Induced Platelet Activation and Cerebral Ischemic-Reperfusion Injury in Rats. Front Pharmacol 2022; 12:792263. [PMID: 35082674 PMCID: PMC8784769 DOI: 10.3389/fphar.2021.792263] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
Ischemic stroke is a major type of stroke worldwide currently without effective treatment, although antiplatelet therapy is an existing option for it. In previous studies, heat shock protein 47 (Hsp47) was found to be expressed on the surface of human and mice platelets and to strengthen the interaction between platelets and collagen. In recent years, Col003 was discovered to inhibit the interaction of Hsp47 with collagen. We evaluated whether the Hsp47 inhibitor Col003 is a promising therapeutic agent for ischemic stroke. Here, we first verified that Hsp47 is also expressed on the surface of rat platelets, and its inhibitor Col003 significantly inhibited thrombus formation in the FeCl3-induced rat carotid arterial thrombus model. Both Col003 and clopidogrel did not alter the bleeding time or coagulation parameters, while aspirin increased the tail-bleeding time (p < 0.05). The low cytotoxicity level of Col003 to rat platelets and human liver cells was similar to those of aspirin and clopidogrel. Col003 inhibited collagen-induced platelet aggregation, adhesion, [Ca2+]i mobilization, P-selectin expression, reactive oxygen species production and the downstream signal pathway of collagen receptors. The results of the middle cerebral artery occlusion model indicated that Col003 has a protective effect against cerebral ischemic-reperfusion injury in rats. The Hsp47 inhibitor Col003 exerted antiplatelet effect and protective effect against brain damage induced by ischemic stroke through the inhibition of glycoprotein VI (GPVI)and mitogen-activated protein kinase (MAPK) signaling events, which might yield a new antiplatelet agent and strategy to treat ischemic stroke.
Collapse
Affiliation(s)
- Shuang Wu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chengwei Liang
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaoyun Xie
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Haiping Huang
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jinfeng Fu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Cilan Wang
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhiheng Su
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Youqiong Wang
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Xiang Qu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jinpin Li
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jingli Liu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
12
|
Kellici TF, Pilka ES, Bodkin MJ. Small-molecule modulators of serine protease inhibitor proteins (serpins). Drug Discov Today 2020; 26:442-454. [PMID: 33259801 DOI: 10.1016/j.drudis.2020.11.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 10/11/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023]
Abstract
Serine protease inhibitors (serpins) are a large family of proteins that regulate and control crucial physiological processes, such as inflammation, coagulation, thrombosis and thrombolysis, and immune responses. The extraordinary impact that these proteins have on numerous crucial pathways makes them an attractive target for drug discovery. In this review, we discuss recent advances in research on small-molecule modulators of serpins, examine their mode of action, analyse the structural data from crystallised protein-ligand complexes, and highlight the potential obstacles and possible therapeutic perspectives. The application of in silico methods for rational drug discovery is also summarised. In addition, we stress the need for continued research in this field.
Collapse
|
13
|
Stainer AR, Sasikumar P, Bye AP, Unsworth AJ, Holbrook LM, Tindall M, Lovegrove JA, Gibbins JM. The Metabolites of the Dietary Flavonoid Quercetin Possess Potent Antithrombotic Activity, and Interact with Aspirin to Enhance Antiplatelet Effects. TH OPEN 2019; 3:e244-e258. [PMID: 31367693 PMCID: PMC6667742 DOI: 10.1055/s-0039-1694028] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 06/24/2019] [Indexed: 01/09/2023] Open
Abstract
Quercetin, a dietary flavonoid, has been reported to possess antiplatelet activity. However, its extensive metabolism following ingestion has resulted in difficulty elucidating precise mechanisms of action. In this study, we aimed to characterize the antiplatelet mechanisms of two methylated metabolites of quercetin-isorhamnetin and tamarixetin-and explore potential interactions with aspirin. Isorhamnetin and tamarixetin inhibited human platelet aggregation, and suppressed activatory processes including granule secretion, integrin αIIbβ3 function, calcium mobilization, and spleen tyrosine kinase (Syk)/linker for activation of T cells (LAT) phosphorylation downstream of glycoprotein VI with similar potency to quercetin. All three flavonoids attenuated thrombus formation in an in vitro microfluidic model, and isoquercetin, a 3-O-glucoside of quercetin, inhibited thrombosis in a murine laser injury model. Isorhamnetin, tamarixetin, and quercetin enhanced the antiplatelet effects of aspirin more-than-additively in a plate-based aggregometry assay, reducing aspirin IC 50 values by an order of magnitude, with this synergy maintained in a whole blood test of platelet function. Our data provide mechanistic evidence for the antiplatelet activity of two quercetin metabolites, isorhamnetin and tamarixetin, and suggest a potential antithrombotic role for these flavonoids. In combination with their interactions with aspirin, this may represent a novel avenue of investigation for the development of new antithrombotic strategies and management of current therapies.
Collapse
Affiliation(s)
- Alexander R Stainer
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Parvathy Sasikumar
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom.,Centre for Haematology, Imperial College London, London, United Kingdom
| | - Alexander P Bye
- 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.,School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom
| | - Lisa M Holbrook
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom.,School of Cardiovascular Medicine and Sciences, King's College London, London, United Kingdom
| | - Marcus Tindall
- Department of Mathematics and Statistics, University of Reading, Reading, United Kingdom
| | - Julie A Lovegrove
- Department of Food and Nutritional Sciences, Hugh Sinclair Unit of Human Nutrition, 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
|
14
|
Ito S, Nagata K. Roles of the endoplasmic reticulum-resident, collagen-specific molecular chaperone Hsp47 in vertebrate cells and human disease. J Biol Chem 2018; 294:2133-2141. [PMID: 30541925 DOI: 10.1074/jbc.tm118.002812] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Heat shock protein 47 (Hsp47) is an endoplasmic reticulum (ER)-resident molecular chaperone essential for correct folding of procollagen in mammalian cells. In this Review, we discuss the role and function of Hsp47 in vertebrate cells and its role in connective tissue disorders. Hsp47 binds to collagenous (Gly-Xaa-Arg) repeats within triple-helical procollagen in the ER and can prevent its local unfolding or aggregate formation, resulting in accelerating triple-helix formation of procollagen. Hsp47 pH-dependently dissociates from procollagen in the cis-Golgi or ER-Golgi intermediate compartment and is then transported back to the ER. Although Hsp47 belongs to the serine protease inhibitor (serpin) superfamily, it does not possess serine protease inhibitory activity. Whereas general molecular chaperones such as Hsp70 and Hsp90 exhibit broad substrate specificity, Hsp47 has narrower specificity mainly for procollagens. However, other Hsp47-interacting proteins have been recently reported, suggesting a much broader role for Hsp47 in the cell that warrants further investigation. Other ER-resident stress proteins, such as binding immunoglobulin protein (BiP), are induced by ER stress, whereas Hsp47 is induced only by heat shock. Constitutive expression of Hsp47 is always correlated with expression of various collagen types, and disruption of the Hsp47 gene in mice causes embryonic lethality due to impaired basement membrane and collagen fibril formation. Increased Hsp47 expression is associated with collagen-related disorders such as fibrosis, characterized by abnormal collagen accumulation, highlighting Hsp47's potential as a clinically relevant therapeutic target.
Collapse
Affiliation(s)
| | - Kazuhiro Nagata
- From the Institute for Protein Dynamics, .,Department of Molecular Biosciences, Faculty of Life Sciences, and.,CREST, Japan Science and Technology Agency, Kyoto Sangyo University, Kyoto 603-8555, Japan
| |
Collapse
|
15
|
Unsworth AJ, Bye AP, Kriek N, Sage T, Osborne AA, Donaghy D, Gibbins JM. Cobimetinib and trametinib inhibit platelet MEK but do not cause platelet dysfunction. Platelets 2018; 30:762-772. [PMID: 30252580 PMCID: PMC6594423 DOI: 10.1080/09537104.2018.1514107] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The MEK inhibitors cobimetinib and trametinib are used in combination with BRAF inhibitors to treat metastatic melanoma but increase rates of hemorrhage relative to BRAF inhibitors alone. Platelets express several members of the MAPK signalling cascade including MEK1 and MEK2 and ERK1 and ERK2 but their role in platelet function and haemostasis is ambiguous as previous reports have been contradictory. It is therefore unclear if MEK inhibitors might be causing platelet dysfunction and contributing to increased hemorrhage. In the present study we performed pharmacological characterisation of cobimetinib and trametinib in vitro to investigate potential for MEK inhibitors to cause platelet dysfunction. We report that whilst both cobimetinib and trametinib are potent inhibitors of platelet MEK activity, treatment with trametinib did not alter platelet function. Treatment with cobimetinib results in inhibition of platelet aggregation, integrin activation, alpha-granule secretion and adhesion but only at suprapharmacological concentrations. We identified that the inhibitory effects of high concentrations of cobimetinib are associated with off-target inhibition on Akt and PKC. Neither inhibitor caused any alteration in thrombus formation on collagen under flow conditions in vitro. Our findings demonstrate that platelets are able to function normally when MEK activity is fully inhibited, indicating MEK activity is dispensable for normal platelet function. We conclude that the MEK inhibitors cobimetinib and trametinib do not induce platelet dysfunction and are therefore unlikely to contribute to increased incidence of bleeding reported during MEK inhibitor therapy.
Collapse
Affiliation(s)
- Amanda J Unsworth
- a Institute for Cardiovascular and Metabolic Research , University of Reading, School of Biological Sciences , Reading , UK
| | - Alexander P Bye
- a Institute for Cardiovascular and Metabolic Research , University of Reading, School of Biological Sciences , Reading , UK
| | - Neline Kriek
- a Institute for Cardiovascular and Metabolic Research , University of Reading, School of Biological Sciences , Reading , UK
| | - Tanya Sage
- a Institute for Cardiovascular and Metabolic Research , University of Reading, School of Biological Sciences , Reading , UK
| | - Ashley A Osborne
- a Institute for Cardiovascular and Metabolic Research , University of Reading, School of Biological Sciences , Reading , UK
| | - Dillon Donaghy
- b Department of Microbiology Immunology and Pathology , Colorado State University , Fort Collins , CO , USA
| | - Jonathan M Gibbins
- a Institute for Cardiovascular and Metabolic Research , University of Reading, School of Biological Sciences , Reading , UK
| |
Collapse
|
16
|
Sasikumar P, AlOuda KS, Kaiser WJ, Holbrook LM, Kriek N, Unsworth AJ, Bye AP, Sage T, Ushioda R, Nagata K, Farndale RW, Gibbins JM. The chaperone protein HSP47: a platelet collagen binding protein that contributes to thrombosis and hemostasis. J Thromb Haemost 2018; 16:946-959. [PMID: 29512284 PMCID: PMC6434988 DOI: 10.1111/jth.13998] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Indexed: 11/30/2022]
Abstract
Essentials Heat shock protein 47 (HSP47), a collagen specific chaperone is present on the platelet surface. Collagen mediated platelet function was reduced following blockade or deletion of HSP47. GPVI receptor regulated signalling was reduced in HSP47 deficient platelets. Platelet HSP47 tethers to exposed collagen thus modulating thrombosis and hemostasis. SUMMARY Objective Heat shock protein 47 (HSP47) is an intracellular chaperone protein that is vital for collagen biosynthesis in collagen secreting cells. This protein has also been shown to be present on the surface of platelets. Given the importance of collagen and its interactions with platelets in triggering hemostasis and thrombosis, in this study we sought to characterize the role of HSP47 in these cells. Methods and Results The deletion of HSP47 in mouse platelets or its inhibition in human platelets reduced their function in response to collagen and the GPVI agonist (CRP-XL), but responses to thrombin were unaltered. In the absence of functional HSP47, the interaction of collagen with platelets was reduced, and this was associated with reduced GPVI-collagen binding, signalling and platelet activation. Thrombus formation on collagen, under arterial flow conditions, was also decreased following the inhibition or deletion of HSP47, in the presence or absence of eptifibatide, consistent with a role for HSP47 in enhancing platelet adhesion to collagen. Platelet adhesion under flow to von Willebrand factor was unaltered following HSP47 inhibition. Laser-induced thrombosis in cremaster muscle arterioles was reduced and bleeding time was prolonged in HSP47-deficient mice or following inhibition of HSP47. Conclusions Our study demonstrates the presence of HSP47 on the platelet surface, where it interacts with collagen, stabilizes platelet adhesion and increases collagen-mediated signalling and therefore thrombus formation and hemostasis.
Collapse
Affiliation(s)
- P. Sasikumar
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - K. S. AlOuda
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - W. J. Kaiser
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - L. M. Holbrook
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - N. Kriek
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - A. J. Unsworth
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - A. P. Bye
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - T. Sage
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - R. Ushioda
- Laboratory of Molecular and Cellular BiologyFaculty of Life SciencesKyoto Sangyo UniversityKyotoJapan
| | - K. Nagata
- Laboratory of Molecular and Cellular BiologyFaculty of Life SciencesKyoto Sangyo UniversityKyotoJapan
| | - R. W. Farndale
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | - J. M. Gibbins
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| |
Collapse
|
17
|
Rigg RA, McCarty OJT, Aslan JE. Heat Shock Protein 70 (Hsp70) in the Regulation of Platelet Function. REGULATION OF HEAT SHOCK PROTEIN RESPONSES 2018. [DOI: 10.1007/978-3-319-74715-6_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
18
|
Lopes Pires ME, Clarke SR, Marcondes S, Gibbins JM. Lipopolysaccharide potentiates platelet responses via toll-like receptor 4-stimulated Akt-Erk-PLA2 signalling. PLoS One 2017; 12:e0186981. [PMID: 29136006 PMCID: PMC5685579 DOI: 10.1371/journal.pone.0186981] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 10/11/2017] [Indexed: 02/06/2023] Open
Abstract
Lipopolysaccharide (LPS) from the cell envelope of Gram-negative bacteria is a principal cause of the symptoms of sepsis. LPS has been reported to modulate the function of platelets although the underlying mechanisms of LPS action in these cells remain unclear. Platelets express the Toll-like receptor 4 (TLR4) which serves as a receptor for LPS, although the potential role of TLR4 and associated cell signalling in controlling platelet responses to LPS has not been extensively explored. In this study, we therefore investigated the actions of LPS prepared from different strains of Escherichia coli on platelet function, the underlying signalling mechanisms, and the potential role of TLR4 in orchestrating these. We report that LPS increased the aggregation of washed platelets stimulated by thromboxane (U46619) or GPVI collagen receptor agonists, effects that were prevented by a TLR4 antagonist. Associated with this, LPS enhanced fibrinogen binding, P-selectin exposure and reactive oxygen species (ROS) release. Increase of ROS was found to be important for the actions of LPS on platelets, since these were inhibited in the presence of superoxide dismutase or catalase. The effects of LPS were associated with phosphorylation of Akt, ERK1/2 and PLA2 in stimulated platelets, and inhibitors of PI3-kinase, Akt and ERK1/2 reduced significantly LPS enhanced platelet function and associated ROS production. Furthermore, inhibition of platelet cyclooxygenase or the thromboxane receptor, revealed an important role for thromboxane A2. We therefore conclude that LPS increases human platelet activation through a TLR4-PI3K-Akt-ERK1/2-PLA2 -dependent pathway that is dependent on ROS and TXA2 formation.
Collapse
Affiliation(s)
- Maria E. Lopes Pires
- School of Biological Science, Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, Berkshire, United Kingdom
| | - Simon R. Clarke
- School of Biological Science, Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, Berkshire, United Kingdom
| | - Sisi Marcondes
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Jonathan M. Gibbins
- School of Biological Science, Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, Berkshire, United Kingdom
| |
Collapse
|
19
|
Unsworth AJ, Kriek N, Bye AP, Naran K, Sage T, Flora GD, Gibbins JM. PPARγ agonists negatively regulate αIIbβ3 integrin outside-in signaling and platelet function through up-regulation of protein kinase A activity. J Thromb Haemost 2017; 15:356-369. [PMID: 27896950 PMCID: PMC5396324 DOI: 10.1111/jth.13578] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Indexed: 12/31/2022]
Abstract
Essentials peroxisome proliferator-activated receptor γ (PPARγ) agonists inhibit platelet function. PPARγ agonists negatively regulate outside-in signaling via integrin αIIbβ3. PPARγ agonists disrupt the interaction of Gα13 with integrin β3. This is attributed to an upregulation of protein kinase A activity. SUMMARY Background Agonists for the peroxisome proliferator-activated receptor (PPARγ) have been shown to have inhibitory effects on platelet activity following stimulation by GPVI and GPCR agonists. Objectives Profound effects on thrombus formation led us to suspect a role for PPARγ agonists in the regulation of integrin αIIbβ3 mediated signaling. Both GPVI and GPCR signaling pathways lead to αIIbβ3 activation, and signaling through αIIbβ3 plays a critical role in platelet function and normal hemostasis. Methods The effects of PPARγ agonists on the regulation of αIIbβ3 outside-in signaling was determined by monitoring the ability of platelets to adhere and spread on fibrinogen and undergo clot retraction. Effects on signaling components downstream of αIIbβ3 activation were also determined following adhesion to fibrinogen by Western blotting. Results Treatment of platelets with PPARγ agonists inhibited platelet adhesion and spreading on fibrinogen and diminished clot retraction. A reduction in phosphorylation of several components of αIIbβ3 signaling, including the integrin β3 subunit, Syk, PLCγ2, focal adhesion kinase (FAK) and Akt, was also observed as a result of reduced interaction of the integrin β3 subunit with Gα13. Studies of VASP phosphorylation revealed that this was because of an increase in PKA activity following treatment with PPARγ receptor agonists. Conclusions This study provides further evidence for antiplatelet actions of PPARγ agonists, identifies a negative regulatory role for PPARγ agonists in the control of integrin αIIbβ3 outside-in signaling, and provides a molecular basis by which the PPARγ agonists negatively regulate platelet activation and thrombus formation.
Collapse
Affiliation(s)
- A. J. Unsworth
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - N. Kriek
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - A. P. Bye
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - K. Naran
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - T. Sage
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - G. D. Flora
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - J. M. Gibbins
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| |
Collapse
|
20
|
New molecular insights into modulation of platelet reactivity in aspirin-treated patients using a network-based approach. Hum Genet 2016; 135:403-414. [DOI: 10.1007/s00439-016-1642-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/23/2016] [Indexed: 10/22/2022]
|
21
|
Marcone S, Dervin F, Fitzgerald DJ. Proteomic signatures of antiplatelet drugs: new approaches to exploring drug effects. J Thromb Haemost 2015; 13 Suppl 1:S323-31. [PMID: 26149042 DOI: 10.1111/jth.12943] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Antiplatelet agents represent the mainstay of acute coronary syndrome (ACS) therapy to prevent ischemic events and to improve safety in patients undergoing percutaneous coronary intervention. However, despite the availability of several drugs and the use of dual antiplatelet therapy, the pharmacological response is highly variable with a subset of patients continuing to experience recurrent thrombotic events, revealing a wide variability in platelet response to antiplatelet drugs. Several factors may explain this, including genetic variation and environmental factors. Here we look at the application of proteomic analysis, an approach that provides an integrated readout of these diverse influences.
Collapse
Affiliation(s)
- S Marcone
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - F Dervin
- School of Biomedical and Biomolecular Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - D J Fitzgerald
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
22
|
Burkhart JM, Gambaryan S, Watson SP, Jurk K, Walter U, Sickmann A, Heemskerk JWM, Zahedi RP. What can proteomics tell us about platelets? Circ Res 2014; 114:1204-19. [PMID: 24677239 DOI: 10.1161/circresaha.114.301598] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
More than 130 years ago, it was recognized that platelets are key mediators of hemostasis. Nowadays, it is established that platelets participate in additional physiological processes and contribute to the genesis and progression of cardiovascular diseases. Recent data indicate that the platelet proteome, defined as the complete set of expressed proteins, comprises >5000 proteins and is highly similar between different healthy individuals. Owing to their anucleate nature, platelets have limited protein synthesis. By implication, in patients experiencing platelet disorders, platelet (dys)function is almost completely attributable to alterations in protein expression and dynamic differences in post-translational modifications. Modern platelet proteomics approaches can reveal (1) quantitative changes in the abundance of thousands of proteins, (2) post-translational modifications, (3) protein-protein interactions, and (4) protein localization, while requiring only small blood donations in the range of a few milliliters. Consequently, platelet proteomics will represent an invaluable tool for characterizing the fundamental processes that affect platelet homeostasis and thus determine the roles of platelets in health and disease. In this article we provide a critical overview on the achievements, the current possibilities, and the future perspectives of platelet proteomics to study patients experiencing cardiovascular, inflammatory, and bleeding disorders.
Collapse
Affiliation(s)
- Julia M Burkhart
- From the Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany (J.M.B., A.S., R.P.Z); Institut für Klinische Biochemie und Pathobiochemie, Universitätsklinikum Würzburg, Würzburg, Germany (S.G.); Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia (S.G.); Centre for Cardiovascular Sciences, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (S.P.W.); Center for Thrombosis and Hemostasis, Universitätsklinikum der Johannes Gutenberg-Universität Mainz, Mainz, Germany (K.J., U.W.); Medizinisches Proteom Center, Ruhr Universität Bochum, Bochum, Germany (A.S.); Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, Scotland, United Kingdom (A.S.); and Department of Biochemistry, CARIM, Maastricht University, Maastricht, The Netherlands (J.W.M.H.)
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Zufferey A, Schvartz D, Nolli S, Reny JL, Sanchez JC, Fontana P. Characterization of the platelet granule proteome: evidence of the presence of MHC1 in alpha-granules. J Proteomics 2014; 101:130-40. [PMID: 24549006 DOI: 10.1016/j.jprot.2014.02.008] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 01/28/2014] [Accepted: 02/04/2014] [Indexed: 10/25/2022]
Abstract
UNLABELLED In the present study, we performed an extensive qualitative characterization of the platelet granule proteome using subcellular fractionation followed by mass spectrometry analysis and functional annotation. Eight-hundred-and-twenty-seven proteins were identified, most of them being associated to granules and to the granule's secretory machinery. Functional pathway analysis revealed 30 pathways, including the major histocompatibility complex class 1 (MHC I) presenting antigen pathway. This pathway was of particular interest for its potential interrelation between platelets and the immune system. Key proteins belonging to this metabolic route such as β-2-microglobulin, 26S protease regulatory subunit 10B from the proteasome and proteins 1 and 2 of the transporter associated with antigen processing were shown to co-localize with von Willebrand factor in resting platelets and to be located on the plasma membrane when platelets were activated. Key proteins of the MHC1 antigen-presenting pathway are located in platelet alpha-granules. These results suggest a possible functional role of platelet granules in platelet-related immune modulation. BIOLOGICAL SIGNIFICANCE In this study, we described the largest dataset related to platelet granule proteins. We performed a functional pathway analysis that evidenced several expected granule-related pathways. We also highlighted the "Antigen processing and presentation" pathway that has drawn our attention. Using immunofluorescence technique, we confirmed the presence of several key proteins for antigen presentation in platelet granules. This study suggests a putative functional role of MHC1 and platelet granules in the immune modulation.
Collapse
Affiliation(s)
- Anne Zufferey
- Division of Angiology and Haemostasis, Department of Medical Specialisations, Faculty of Medicine and University Hospitals of Geneva, Geneva, Switzerland; Biomedical Proteomics Research Group, Department of Human Protein Sciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Domitille Schvartz
- Biomedical Proteomics Research Group, Department of Human Protein Sciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Séverine Nolli
- Division of Angiology and Haemostasis, Department of Medical Specialisations, Faculty of Medicine and University Hospitals of Geneva, Geneva, Switzerland; Biomedical Proteomics Research Group, Department of Human Protein Sciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jean-Luc Reny
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Division of Internal Medicine, and Rehabilitation, Trois-Chêne Hospital, Faculty of Medicine and University Hospitals of Geneva, Geneva, Switzerland
| | - Jean-Charles Sanchez
- Biomedical Proteomics Research Group, Department of Human Protein Sciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Pierre Fontana
- Division of Angiology and Haemostasis, Department of Medical Specialisations, Faculty of Medicine and University Hospitals of Geneva, Geneva, Switzerland; Biomedical Proteomics Research Group, Department of Human Protein Sciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
24
|
Quantitative profiling of the rat heart myoblast secretome reveals differential responses to hypoxia and re-oxygenation stress. J Proteomics 2014; 98:138-49. [DOI: 10.1016/j.jprot.2013.12.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/25/2013] [Accepted: 12/28/2013] [Indexed: 11/18/2022]
|
25
|
Prudent M, Tissot JD, Lion N. Proteomics of blood and derived products: what’s next? Expert Rev Proteomics 2014; 8:717-37. [DOI: 10.1586/epr.11.58] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
26
|
|
27
|
Abstract
Statins are widely prescribed cholesterol-lowering drugs that are a first-line treatment of coronary artery disease and atherosclerosis, reducing the incidence of thrombotic events such as myocardial infarction and stroke. Statins have been shown to reduce platelet activation, although the mechanism(s) through which this occurs is unclear. Because several of the characteristic effects of statins on platelets are shared with those elicited by the inhibitory platelet adhesion receptor PECAM-1 (platelet endothelial cell adhesion molecule-1), we investigated a potential connection between the influence of statins on platelet function and PECAM-1 signaling. Statins were found to inhibit a range of platelet functional responses and thrombus formation in vitro and in vivo. Notably, these effects of statins on platelet function in vitro and in vivo were diminished in PECAM-1(-/-) platelets. Activation of PECAM-1 signaling results in its tyrosine phosphorylation, the recruitment and activation of tyrosine phosphatase SHP-2, the subsequent binding of phosphoinositol 3-kinase (PI3K), and diminished PI3K signaling. Statins resulted in the stimulation of these events, leading to the inhibition of Akt activation. Together, these data provide evidence for a fundamental role of PECAM-1 in the inhibitory effects of statins on platelet activation, which may explain some of the pleiotropic actions of these drugs.
Collapse
|
28
|
Eberini I, Wait R, Calabresi L, Sensi C, Miller I, Gianazza E. A proteomic portrait of atherosclerosis. J Proteomics 2013; 82:92-112. [DOI: 10.1016/j.jprot.2013.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 02/11/2013] [Accepted: 02/13/2013] [Indexed: 01/11/2023]
|
29
|
Wright B, Stanley RG, Kaiser WJ, Gibbins JM. The integration of proteomics and systems approaches to map regulatory mechanisms underpinning platelet function. Proteomics Clin Appl 2013. [DOI: 10.1002/prca.201200095] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Bernice Wright
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences; University of Reading; Reading; Berkshire; UK
| | - Ronald G. Stanley
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences; University of Reading; Reading; Berkshire; UK
| | - William J. Kaiser
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences; University of Reading; Reading; Berkshire; UK
| | - Jonathan M. Gibbins
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences; University of Reading; Reading; Berkshire; UK
| |
Collapse
|
30
|
|
31
|
Vaiyapuri S, Hutchinson EG, Ali MS, Dannoura A, Stanley RG, Harrison RA, Bicknell AB, Gibbins JM. Rhinocetin, a venom-derived integrin-specific antagonist inhibits collagen-induced platelet and endothelial cell functions. J Biol Chem 2012; 287:26235-44. [PMID: 22689571 PMCID: PMC3406708 DOI: 10.1074/jbc.m112.381483] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Snaclecs are small non-enzymatic proteins present in viper venoms reported to modulate hemostasis of victims through effects on platelets, vascular endothelial, and smooth muscle cells. In this study, we have isolated and functionally characterized a snaclec that we named "rhinocetin" from the venom of West African gaboon viper, Bitis gabonica rhinoceros. Rhinocetin was shown to comprise α and β chains with the molecular masses of 13.5 and 13 kDa, respectively. Sequence and immunoblot analysis of rhinocetin confirmed this to be a novel snaclec. Rhinocetin inhibited collagen-stimulated activation of human platelets in a dose-dependent manner but displayed no inhibitory effects on glycoprotein VI (collagen receptor) selective agonist, CRP-XL-, ADP-, or thrombin-induced platelet activation. Rhinocetin antagonized the binding of monoclonal antibodies against the α2 subunit of integrin α2β1 to platelets and coimmunoprecipitation analysis confirmed integrin α2β1 as a target for this venom protein. Rhinocetin inhibited a range of collagen-induced platelet functions such as fibrinogen binding, calcium mobilization, granule secretion, aggregation, and thrombus formation. It also inhibited integrin α2β1-dependent functions of human endothelial cells. Together, our data suggest rhinocetin to be a modulator of integrin α2β1 function and thus may provide valuable insights into the role of this integrin in physiological and pathophysiological scenarios, including hemostasis, thrombosis, and envenomation.
Collapse
Affiliation(s)
- Sakthivel Vaiyapuri
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading RG6 6UB, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Zufferey A, Fontana P, Reny JL, Nolli S, Sanchez JC. Platelet proteomics. MASS SPECTROMETRY REVIEWS 2012; 31:331-351. [PMID: 22009795 DOI: 10.1002/mas.20345] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 06/10/2011] [Accepted: 06/10/2011] [Indexed: 05/31/2023]
Abstract
Platelets are small cell fragments, produced by megakaryocytes, in the bone marrow. They play an important role in hemostasis and diverse thrombotic disorders. They are therefore primary targets of antithrombotic therapies. They are implicated in several pathophysiological pathways, such as inflammation or wound repair. In blood circulation, platelets are activated by several pathways including subendothelial matrix and thrombin, triggering the formation of the platelet plug. Studying their proteome is a powerful approach to understand their biology and function. However, particular attention must be paid to different experimental parameters, such as platelet quality and purity. Several technologies are involved during the platelet proteome processing, yielding information on protein identification, characterization, localization, and quantification. Recent technical improvements in proteomics combined with inter-disciplinary strategies, such as metabolomic, transcriptomics, and bioinformatics, will help to understand platelets biological mechanisms. Therefore, a comprehensive analysis of the platelet proteome under different environmental conditions may contribute to elucidate complex processes relevant to platelet function regarding bleeding disorders or platelet hyperreactivity and identify new targets for antiplatelet therapy.
Collapse
Affiliation(s)
- Anne Zufferey
- Division of Angiology and Haemostasis, Department of Internal Medicine, Faculty of Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | | | | | | | | |
Collapse
|
33
|
Abstract
Platelets pose unique challenges to cell biologists due to their lack of nucleus and low levels of messenger RNA. Platelets cannot be cultured in great abundance or manipulated using common recombinant DNA technologies. As a result, platelet research has lagged behind that of nucleated cells. The advent of mass spectrometry and its application to protein biochemistry brought with it great hopes for the platelet community that are now being realized. This technology is ideally suited for identifying low-abundance proteins, protein-protein interactions, and post-translational modifications in complex protein mixtures. Over the past 10 years, proteomics has delivered in many ways, providing platelet biologists with a comprehensive list of proteins expressed in platelets, information on post-translational modifications, protein interactions and sub-cellular localization. Several novel and important platelet membrane proteins, including CLEC-2, CD148, G6b-B, G6f, and Hsp47, have been identified using proteomics-based approaches. New, more sensitive instrumentation and novel approaches are making it increasingly possible to identify ever lower amounts of proteins. In this chapter we highlight some of the major achievements of platelet proteomics to date, discussing challenges and how they were overcome. We also discuss new frontiers and applications of proteomics to platelets and microparticles in health and disease, as we strive to better understand the molecular mechanisms underlying the platelet response to vascular injury.
Collapse
Affiliation(s)
- Yotis Senis
- Centre for Cardiovascular Sciences, Institute of Biomedical Research, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, UK.
| | | |
Collapse
|
34
|
Deckmyn H, De Meyer SF, Broos K, Vanhoorelbeke K. Inhibitors of the interactions between collagen and its receptors on platelets. Handb Exp Pharmacol 2012:311-337. [PMID: 22918737 DOI: 10.1007/978-3-642-29423-5_13] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
At sites of vascular injury, collagen-mediated platelet adhesion and activation have long been known as one of the first events in platelet-dependent thrombus formation. Studying patients with bleeding disorders that are caused by defective platelet adhesion to collagen resulted in the identification of several platelet collagen receptors, with glycoprotein VI and integrin α2β1 being the most important ones. Subsequent development of specific collagen receptor knockout mice and various inhibitors of platelet binding to collagen have further proven the role of these receptors in haemostasis and thrombosis. The search for clinically applicable inhibitors for use as antithrombotic drug has led to the identification of inhibitory antibodies, soluble receptor fragments, peptides, collagen-mimetics and proteins from snake venoms or haematophagous animals. In experimental settings, these inhibitors have a good antithrombotic effect, with little prolongation of bleeding times, suggesting a larger therapeutic window than currently available antiplatelet drugs. However, at present, none of the collagen receptor blockers are in clinical development yet.
Collapse
Affiliation(s)
- Hans Deckmyn
- Laboratory for Thrombosis Research, KU Leuven campus Kortrijk, Kortrijk, Belgium.
| | | | | | | |
Collapse
|
35
|
Wright B, Stanley RG, Kaiser WJ, Mills DJ, Gibbins JM. Analysis of protein networks in resting and collagen receptor (GPVI)-stimulated platelet sub-proteomes. Proteomics 2011; 11:4588-92. [DOI: 10.1002/pmic.201100410] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2011] [Revised: 09/12/2011] [Accepted: 09/14/2011] [Indexed: 02/04/2023]
|
36
|
Egidi MG, Rinalducci S, Marrocco C, Vaglio S, Zolla L. Proteomic analysis of plasma derived from platelet buffy coats during storage at room temperature. An application of ProteoMiner™ technology. Platelets 2011; 22:252-69. [PMID: 21405958 DOI: 10.3109/09537104.2010.550348] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The present study was aimed at revealing new insights into the analysis of storage-related processes occurring in the supernatants of platelet concentrates (PCs) derived from pooled buffy coats suspended in whole plasma. To reduce the dynamic range of plasma protein concentrations and access low-abundance proteins, we made use of a solid-phase combinatorial peptide ligand library, known under the trade name of ProteoMiner™. Afterwards, two-dimensional electrophoresis (2-DE) was coupled with mass spectrometry (MS) to reveal changes in proteomic profiles. Several storage-induced protein alterations were identified including changes to major plasma proteins. In particular, a precursor of the secretory form of clusterin was shown to accumulate during storage of PC supernatants, together with platelet-derived tropomyosin, suggesting a progressive loss of platelet integrity. Platelet-released proteins following activation have also been detected (alpha-1-B-glycoprotein, kininogen-1, and serpin proteinase inhibitor 8). Moreover, specific protein fragments (vitronectin, plakoglobin, hornerin, and apolipoprotein A-IV) were found to be modulated upon storage, possibly indicating a time-dependent buffy-coat PC deterioration. Globally, our findings provided the disclosure of unique proteins in PC supernatants with respect to previous studies conducted in similar experimental conditions, suggesting ProteoMiner enrichment technology to be a possible complementary tool in the identification of diagnostically relevant proteins as age/quality biomarkers of therapeutic products.
Collapse
Affiliation(s)
- Maria Giulia Egidi
- Department of Environmental Sciences, University of Tuscia, Viterbo, Italy
| | | | | | | | | |
Collapse
|
37
|
Májek P, Reicheltová Z, Stikarová J, Suttnar J, Sobotková A, Dyr JE. Proteome changes in platelets activated by arachidonic acid, collagen, and thrombin. Proteome Sci 2010. [PMID: 21073729 DOI: 10.1186/1477-5956-8-56.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Platelets are small anucleated blood particles that play a key role in the control of bleeding. Platelets need to be activated to perform their functions and participate in hemostasis. The process of activation is accompanied by vast protein reorganization and posttranslational modifications. The goal of this study was to identify changes in proteins in platelets activated by different agonists. Platelets were activated by three different agonists - arachidonic acid, collagen, and thrombin. 2D SDS-PAGE (pI 4-7) was used to separate platelet proteins. Proteomes of activated and resting platelets were compared with each other by Progenesis SameSpots statistical software; and proteins were identified by nanoLC-MS/MS. RESULTS 190 spots were found to be significantly different. Of these, 180 spots were successfully identified and correspond to 144 different proteins. Five proteins were found that had not previously been identified in platelets: protein CDV3 homolog, protein ETHE1, protein LZIC, FGFR1 oncogene partner 2, and guanine nucleotide-binding protein subunit beta-5. Using spot expression profile analysis, we found two proteins (WD repeat-containing protein 1 and mitochondrial glycerol-3-phosphate dehydrogenase) that may be part of thrombin specific activation or signal transduction pathway(s). CONCLUSIONS Our results, characterizing the differences within proteins in both activated (by various agonists) and resting platelets, can thus contribute to the basic knowledge of platelets and to the understanding of the function and development of new antiplatelet drugs.
Collapse
Affiliation(s)
- Pavel Májek
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic.
| | | | | | | | | | | |
Collapse
|
38
|
Májek P, Reicheltová Z, Stikarová J, Suttnar J, Sobotková A, Dyr JE. Proteome changes in platelets activated by arachidonic acid, collagen, and thrombin. Proteome Sci 2010; 8:56. [PMID: 21073729 PMCID: PMC2996359 DOI: 10.1186/1477-5956-8-56] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Accepted: 11/12/2010] [Indexed: 12/27/2022] Open
Abstract
Background Platelets are small anucleated blood particles that play a key role in the control of bleeding. Platelets need to be activated to perform their functions and participate in hemostasis. The process of activation is accompanied by vast protein reorganization and posttranslational modifications. The goal of this study was to identify changes in proteins in platelets activated by different agonists. Platelets were activated by three different agonists - arachidonic acid, collagen, and thrombin. 2D SDS-PAGE (pI 4-7) was used to separate platelet proteins. Proteomes of activated and resting platelets were compared with each other by Progenesis SameSpots statistical software; and proteins were identified by nanoLC-MS/MS. Results 190 spots were found to be significantly different. Of these, 180 spots were successfully identified and correspond to 144 different proteins. Five proteins were found that had not previously been identified in platelets: protein CDV3 homolog, protein ETHE1, protein LZIC, FGFR1 oncogene partner 2, and guanine nucleotide-binding protein subunit beta-5. Using spot expression profile analysis, we found two proteins (WD repeat-containing protein 1 and mitochondrial glycerol-3-phosphate dehydrogenase) that may be part of thrombin specific activation or signal transduction pathway(s). Conclusions Our results, characterizing the differences within proteins in both activated (by various agonists) and resting platelets, can thus contribute to the basic knowledge of platelets and to the understanding of the function and development of new antiplatelet drugs.
Collapse
Affiliation(s)
- Pavel Májek
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic.
| | | | | | | | | | | |
Collapse
|
39
|
Martinez-Pinna R, Barbas C, Blanco-Colio LM, Tunon J, Ramos-Mozo P, Lopez JA, Meilhac O, Michel JB, Egido J, Martin-Ventura JL. Proteomic and metabolomic profiles in atherothrombotic vascular disease. Curr Atheroscler Rep 2010; 12:202-8. [PMID: 20425260 DOI: 10.1007/s11883-010-0102-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Atherothrombosis remains a major cause of morbidity and mortality in the western world. The underlying processes associated with clinical expression of atherothrombosis include oxidative stress and proteolysis in relation to neovascularisation and intraplaque hemorrhages, leading to immuno-inflammatory response, cell death, and extracellular matrix breakdown. The complex biological multifactorial nature of atherothrombosis requires the development of novel technologies that allow the analysis of cellular and molecular processes responsible for the transition to disease phenotypes and the discovery of new diagnostic and prognostic biomarkers. In the present article, we have reviewed recent advances in the application of proteomic and metabolomic techniques to the study of atherothrombosis. We have focused on recent studies analyzing cells involved in hemo-thrombus formation (platelets, red blood cells, and polymorphonuclear cells), as well as tissues, tissue-conditioned media, and plasma of atherothrombotic patients. In the future, the application of these high-throughput technologies, along with imaging techniques, in systems biology approaches will help to individualize medicine.
Collapse
Affiliation(s)
- Roxana Martinez-Pinna
- Vascular Research Laboratory, Instituto de Investigacion Sanitaria, Fundación Jimenez Diaz, Autonoma University, Av. Reyes Católicos 2, 28040, Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|